aboutsummaryrefslogtreecommitdiff
path: root/gnqa/paper2_eval/data/responses/gpt4o/de_diabetes_responses.json
blob: 61ee90fd9e7c531c12f719ac3b2c16bc8749eba1 (plain)
1
2
3
4
5
6
7
8
9
10
11
12
13
14
15
16
17
18
19
20
21
22
23
24
25
26
27
28
29
30
31
32
33
34
35
36
37
38
39
40
41
42
43
44
45
46
47
48
49
50
51
52
53
54
55
56
57
58
59
60
61
62
63
64
65
66
67
68
69
70
71
72
73
74
75
76
77
78
79
80
81
82
83
84
85
86
87
88
89
90
91
92
93
94
95
96
97
98
99
100
101
102
103
104
105
106
107
108
109
110
111
112
113
114
115
116
117
118
119
120
121
122
123
124
125
126
127
128
129
130
131
132
133
134
135
136
137
138
139
140
141
142
143
144
145
146
147
148
149
150
151
152
153
154
155
156
157
158
159
160
161
162
163
164
165
166
167
168
169
170
171
172
173
174
175
176
177
178
179
180
181
182
183
184
185
186
187
188
189
190
191
192
193
194
195
196
197
198
199
200
201
202
203
204
205
206
207
208
209
210
211
212
213
214
215
216
217
218
219
220
221
222
223
224
225
226
227
228
229
230
231
232
233
234
235
236
237
238
239
240
241
242
243
244
245
246
247
248
249
250
251
252
253
254
255
256
257
258
259
260
261
262
263
264
265
266
267
268
269
270
271
272
273
274
275
276
277
278
279
280
281
282
283
284
285
286
287
288
289
290
291
292
293
294
295
296
297
298
299
300
301
302
303
304
305
306
307
308
309
310
311
312
313
314
315
316
317
318
319
320
321
322
323
324
325
326
327
328
329
330
331
332
333
334
335
336
337
338
339
340
341
342
343
344
345
346
347
348
349
350
351
352
353
354
355
356
357
358
359
360
361
362
363
364
365
366
367
368
369
370
371
372
373
374
375
376
377
378
379
380
381
382
383
384
385
386
387
388
389
390
391
392
393
394
395
396
397
398
399
400
401
402
403
404
405
406
407
408
409
410
411
412
413
414
415
416
417
418
419
420
421
422
423
424
425
426
427
428
429
430
431
432
433
434
435
436
437
438
439
440
441
442
443
444
445
446
447
448
449
450
451
452
453
454
455
456
457
458
459
460
461
462
463
464
465
466
467
468
469
470
471
472
473
474
475
476
477
478
479
480
481
482
483
484
485
486
487
488
489
490
491
492
493
494
495
496
497
498
499
500
501
502
503
504
505
506
507
508
509
510
511
512
513
514
515
516
517
518
519
520
521
522
523
524
525
526
527
528
529
530
531
532
533
534
535
536
537
538
539
540
541
542
543
544
545
546
547
548
549
550
551
552
553
554
555
556
557
558
559
560
561
562
563
564
565
566
567
568
569
570
571
572
573
574
575
576
577
578
579
580
581
582
583
584
585
586
587
588
589
590
591
592
593
594
595
596
597
598
599
600
601
602
603
604
605
606
607
608
609
610
611
612
613
614
615
616
617
618
619
620
621
622
623
624
625
626
627
628
629
630
631
632
633
634
635
636
637
638
639
640
641
642
643
644
645
646
647
648
649
650
651
652
653
654
655
656
657
658
659
660
661
662
663
664
665
666
667
668
669
670
671
672
673
674
675
676
677
678
679
680
681
682
683
684
685
686
687
688
689
690
691
692
693
694
695
696
697
698
699
700
701
702
703
704
705
706
707
708
709
710
711
712
713
714
715
716
717
718
719
720
721
722
723
724
725
726
727
728
729
730
731
732
733
734
735
736
737
738
739
740
741
742
743
744
745
746
747
748
749
750
751
752
753
754
755
756
757
758
759
760
761
762
763
764
765
766
767
768
769
770
771
772
773
774
775
776
777
778
779
780
781
782
783
784
785
786
787
788
789
790
791
792
793
794
795
796
797
798
799
800
801
802
803
804
805
806
807
808
809
810
811
812
813
814
815
816
817
818
819
820
821
822
823
824
825
826
827
828
829
830
831
832
833
834
835
836
837
838
839
840
841
842
843
844
845
846
847
848
849
850
851
852
853
854
855
856
857
858
859
860
861
862
863
864
865
866
867
868
869
870
871
872
873
874
875
876
877
878
879
880
881
882
883
884
885
886
887
888
889
890
891
892
893
894
895
896
897
898
899
900
901
902
903
904
905
906
907
908
909
910
911
912
913
914
915
916
917
918
919
920
921
922
923
924
925
926
927
928
929
930
931
932
933
934
935
936
937
938
939
940
941
942
943
944
945
946
947
948
949
950
951
952
953
954
955
956
957
958
959
960
961
962
963
964
965
966
967
968
969
970
971
972
973
974
975
976
977
978
979
980
981
982
983
984
985
986
987
988
989
990
991
992
993
994
995
996
997
998
999
1000
1001
1002
1003
1004
1005
1006
1007
1008
1009
1010
1011
1012
1013
1014
1015
1016
1017
1018
1019
1020
1021
1022
1023
1024
1025
1026
1027
1028
1029
1030
1031
1032
1033
1034
1035
1036
1037
1038
1039
1040
1041
1042
1043
1044
1045
1046
1047
1048
1049
1050
1051
1052
1053
1054
1055
1056
1057
1058
1059
1060
1061
1062
1063
1064
1065
1066
1067
1068
1069
1070
1071
1072
1073
1074
1075
1076
1077
1078
1079
1080
1081
1082
1083
1084
1085
1086
1087
1088
1089
1090
1091
1092
1093
1094
1095
1096
1097
1098
1099
1100
1101
1102
1103
1104
1105
1106
1107
1108
1109
1110
1111
1112
1113
1114
1115
1116
1117
1118
1119
1120
1121
1122
1123
1124
1125
1126
1127
1128
1129
1130
1131
1132
1133
1134
1135
1136
1137
1138
1139
1140
1141
1142
1143
1144
1145
1146
1147
1148
1149
1150
1151
1152
1153
1154
1155
1156
1157
1158
1159
1160
1161
1162
1163
1164
1165
1166
1167
1168
1169
1170
1171
1172
1173
1174
1175
1176
1177
1178
1179
1180
1181
1182
1183
1184
1185
1186
1187
1188
1189
1190
1191
1192
1193
1194
1195
1196
1197
1198
1199
1200
1201
1202
1203
1204
1205
1206
1207
1208
1209
1210
1211
1212
1213
1214
1215
1216
1217
1218
1219
1220
1221
1222
1223
1224
1225
1226
1227
1228
1229
1230
1231
1232
1233
1234
1235
1236
1237
1238
1239
1240
1241
1242
1243
1244
1245
1246
1247
1248
1249
1250
1251
1252
1253
1254
1255
1256
1257
1258
1259
1260
1261
1262
1263
1264
1265
1266
1267
1268
1269
1270
1271
1272
1273
1274
1275
1276
1277
1278
1279
1280
1281
1282
1283
1284
1285
1286
1287
1288
1289
1290
1291
1292
1293
1294
1295
1296
1297
1298
1299
1300
1301
1302
1303
1304
1305
1306
1307
1308
1309
1310
1311
1312
1313
1314
1315
1316
1317
1318
1319
1320
1321
1322
1323
1324
1325
1326
1327
1328
1329
1330
1331
1332
1333
1334
1335
1336
1337
1338
1339
1340
1341
1342
1343
1344
1345
1346
1347
1348
1349
1350
1351
1352
1353
1354
1355
1356
1357
1358
1359
1360
1361
1362
1363
1364
1365
1366
1367
1368
1369
1370
1371
1372
1373
1374
1375
1376
1377
1378
1379
1380
1381
1382
1383
1384
1385
1386
1387
1388
1389
1390
1391
1392
1393
1394
1395
1396
1397
1398
1399
1400
1401
1402
1403
1404
1405
1406
1407
1408
1409
1410
1411
1412
1413
1414
1415
1416
1417
1418
1419
1420
1421
1422
1423
1424
1425
1426
1427
1428
1429
1430
1431
1432
1433
1434
1435
1436
1437
1438
1439
1440
1441
1442
1443
1444
1445
1446
1447
1448
1449
1450
1451
1452
1453
1454
1455
1456
1457
1458
1459
1460
1461
1462
1463
1464
1465
1466
1467
1468
1469
1470
1471
1472
1473
1474
1475
1476
1477
1478
1479
1480
1481
1482
1483
1484
1485
1486
1487
1488
1489
1490
1491
1492
1493
1494
1495
1496
1497
1498
1499
1500
1501
1502
1503
1504
1505
1506
1507
1508
1509
1510
1511
1512
1513
1514
1515
1516
1517
1518
1519
1520
1521
1522
1523
1524
1525
1526
1527
1528
1529
1530
1531
1532
1533
1534
1535
1536
1537
1538
1539
1540
1541
1542
1543
1544
1545
1546
1547
1548
1549
1550
1551
1552
1553
1554
1555
1556
1557
1558
1559
1560
1561
1562
1563
1564
1565
1566
1567
1568
1569
1570
1571
1572
1573
1574
1575
1576
1577
1578
1579
1580
1581
1582
1583
1584
1585
1586
1587
1588
1589
1590
1591
1592
1593
1594
1595
1596
1597
1598
1599
1600
1601
1602
1603
1604
1605
1606
1607
1608
1609
1610
1611
1612
1613
1614
1615
1616
1617
1618
1619
1620
1621
1622
1623
1624
1625
1626
1627
1628
1629
1630
1631
1632
1633
1634
1635
1636
1637
1638
1639
1640
1641
1642
1643
1644
1645
1646
1647
1648
1649
1650
1651
1652
1653
1654
1655
1656
1657
1658
1659
1660
1661
1662
1663
1664
1665
1666
1667
1668
1669
1670
1671
1672
1673
1674
1675
1676
1677
1678
1679
1680
1681
1682
1683
1684
1685
1686
1687
1688
1689
1690
1691
1692
1693
1694
1695
1696
1697
1698
1699
1700
1701
1702
1703
1704
1705
1706
1707
1708
1709
1710
1711
1712
1713
1714
1715
1716
1717
1718
1719
1720
1721
1722
1723
1724
1725
1726
1727
1728
1729
1730
1731
1732
1733
1734
1735
1736
1737
1738
1739
1740
1741
1742
1743
1744
1745
1746
1747
1748
1749
1750
1751
1752
1753
1754
1755
1756
1757
1758
1759
1760
1761
1762
1763
1764
1765
1766
1767
1768
1769
1770
1771
1772
1773
1774
1775
1776
1777
1778
1779
1780
1781
1782
1783
1784
1785
1786
1787
1788
1789
1790
1791
1792
1793
1794
1795
1796
1797
1798
1799
1800
1801
1802
1803
1804
1805
1806
1807
1808
1809
1810
1811
1812
1813
1814
1815
1816
1817
1818
1819
1820
1821
1822
1823
1824
1825
1826
1827
1828
1829
1830
1831
1832
1833
1834
1835
1836
1837
1838
1839
1840
1841
1842
1843
1844
1845
1846
1847
1848
1849
1850
1851
1852
1853
1854
1855
1856
1857
1858
1859
1860
1861
1862
1863
1864
1865
1866
1867
1868
1869
1870
1871
1872
1873
1874
1875
1876
1877
1878
1879
1880
1881
1882
1883
1884
1885
1886
1887
1888
1889
1890
1891
1892
1893
1894
1895
1896
1897
1898
1899
1900
1901
1902
1903
1904
1905
1906
1907
1908
1909
1910
1911
1912
1913
1914
1915
1916
1917
1918
1919
1920
1921
1922
1923
1924
1925
1926
1927
1928
1929
1930
1931
1932
1933
1934
1935
1936
1937
1938
1939
1940
1941
1942
1943
1944
1945
1946
1947
1948
1949
1950
1951
1952
1953
1954
1955
1956
1957
1958
1959
1960
1961
1962
1963
1964
1965
1966
1967
1968
1969
1970
1971
1972
1973
1974
1975
1976
1977
1978
1979
1980
1981
1982
1983
1984
1985
1986
1987
1988
1989
1990
1991
1992
1993
1994
1995
1996
1997
1998
1999
2000
2001
2002
2003
2004
2005
2006
2007
2008
2009
2010
2011
2012
2013
2014
2015
2016
2017
2018
2019
2020
2021
2022
2023
2024
2025
2026
2027
2028
2029
2030
2031
2032
2033
2034
2035
2036
2037
2038
2039
2040
2041
2042
2043
2044
2045
2046
2047
2048
2049
2050
2051
2052
2053
2054
2055
2056
2057
2058
2059
2060
2061
2062
2063
2064
2065
2066
2067
2068
2069
2070
2071
2072
2073
2074
2075
2076
2077
2078
2079
2080
2081
2082
2083
2084
2085
2086
2087
2088
2089
2090
2091
2092
2093
2094
2095
2096
2097
2098
2099
2100
2101
2102
2103
2104
2105
2106
2107
2108
2109
2110
2111
2112
2113
2114
2115
2116
2117
2118
2119
2120
2121
2122
2123
2124
2125
2126
2127
2128
2129
2130
2131
2132
2133
2134
2135
2136
2137
2138
2139
2140
2141
2142
2143
2144
2145
2146
2147
2148
2149
2150
2151
2152
2153
2154
2155
2156
2157
2158
2159
2160
2161
2162
2163
2164
2165
2166
2167
2168
2169
2170
2171
2172
2173
2174
2175
2176
2177
2178
2179
2180
2181
2182
2183
2184
2185
2186
2187
2188
2189
2190
2191
2192
2193
2194
2195
2196
2197
2198
2199
2200
2201
2202
2203
2204
2205
2206
2207
2208
2209
2210
2211
2212
2213
2214
2215
2216
2217
2218
2219
2220
2221
2222
2223
2224
2225
2226
2227
2228
2229
2230
2231
2232
2233
2234
2235
2236
2237
2238
2239
2240
2241
2242
2243
2244
2245
2246
2247
2248
2249
2250
2251
2252
2253
2254
2255
2256
2257
2258
2259
2260
2261
2262
2263
2264
2265
2266
2267
2268
2269
2270
2271
2272
2273
2274
2275
2276
2277
2278
2279
2280
2281
2282
2283
2284
2285
2286
2287
2288
2289
2290
2291
2292
2293
2294
2295
2296
2297
2298
2299
2300
2301
2302
2303
2304
2305
2306
2307
2308
2309
2310
2311
2312
2313
2314
2315
2316
2317
2318
2319
2320
2321
2322
2323
2324
2325
2326
2327
2328
2329
2330
2331
2332
2333
2334
2335
2336
2337
2338
2339
2340
2341
2342
2343
2344
2345
2346
2347
2348
2349
2350
2351
2352
2353
2354
2355
2356
2357
2358
2359
2360
2361
2362
2363
2364
2365
2366
2367
2368
2369
2370
2371
2372
2373
2374
2375
2376
2377
2378
2379
2380
2381
2382
2383
2384
2385
2386
2387
2388
2389
2390
2391
2392
2393
2394
2395
2396
2397
2398
2399
2400
2401
2402
2403
2404
2405
2406
2407
2408
2409
2410
2411
2412
2413
2414
2415
2416
2417
2418
2419
2420
2421
2422
2423
2424
2425
2426
2427
2428
2429
2430
2431
2432
2433
2434
2435
2436
2437
2438
2439
2440
2441
2442
2443
2444
2445
2446
2447
2448
2449
2450
2451
2452
2453
2454
2455
2456
2457
2458
2459
2460
2461
2462
2463
2464
2465
2466
2467
2468
2469
2470
2471
2472
2473
2474
2475
2476
2477
2478
2479
2480
2481
2482
2483
2484
2485
2486
2487
2488
2489
2490
2491
2492
2493
2494
2495
2496
2497
2498
2499
2500
2501
2502
2503
2504
2505
2506
2507
2508
2509
2510
2511
2512
2513
2514
2515
2516
2517
2518
2519
2520
2521
2522
2523
2524
2525
2526
2527
2528
2529
2530
2531
2532
2533
2534
2535
2536
2537
2538
2539
2540
2541
2542
2543
2544
2545
2546
2547
2548
2549
2550
2551
2552
2553
2554
2555
2556
2557
2558
2559
2560
2561
2562
2563
2564
2565
2566
2567
2568
2569
2570
2571
2572
2573
2574
2575
2576
2577
2578
2579
2580
2581
2582
2583
2584
2585
2586
2587
2588
2589
2590
2591
2592
2593
2594
2595
2596
2597
2598
2599
2600
2601
2602
2603
2604
2605
2606
2607
2608
2609
2610
2611
2612
2613
2614
2615
2616
2617
2618
2619
2620
2621
2622
2623
2624
2625
2626
2627
2628
2629
2630
2631
2632
2633
2634
2635
2636
2637
2638
2639
2640
2641
2642
2643
2644
2645
2646
2647
2648
2649
2650
2651
2652
2653
2654
2655
2656
2657
2658
2659
2660
2661
2662
2663
2664
2665
2666
2667
2668
2669
2670
2671
2672
2673
2674
2675
2676
2677
2678
2679
2680
2681
2682
2683
2684
2685
2686
2687
2688
2689
2690
2691
2692
2693
2694
2695
2696
2697
2698
2699
2700
2701
2702
2703
2704
2705
2706
2707
2708
2709
2710
2711
2712
2713
2714
2715
2716
2717
2718
2719
2720
2721
2722
2723
2724
2725
2726
2727
2728
2729
2730
2731
2732
2733
2734
2735
2736
2737
2738
2739
2740
2741
2742
2743
2744
2745
2746
2747
2748
2749
2750
2751
2752
2753
2754
2755
2756
2757
2758
2759
2760
2761
2762
2763
2764
2765
2766
2767
2768
2769
2770
2771
2772
2773
2774
2775
2776
2777
2778
2779
2780
2781
2782
2783
2784
2785
2786
2787
2788
2789
2790
2791
2792
2793
2794
2795
2796
2797
2798
2799
2800
2801
2802
2803
2804
2805
2806
2807
2808
2809
2810
2811
2812
2813
2814
2815
2816
2817
2818
2819
2820
2821
2822
2823
2824
2825
2826
2827
2828
2829
2830
2831
2832
2833
2834
2835
2836
2837
2838
2839
2840
2841
2842
2843
2844
2845
2846
2847
2848
2849
2850
2851
2852
2853
2854
2855
2856
2857
2858
2859
2860
2861
2862
2863
2864
2865
2866
2867
2868
2869
2870
2871
2872
2873
2874
2875
2876
2877
2878
2879
2880
2881
2882
2883
2884
2885
2886
2887
2888
2889
2890
2891
2892
2893
2894
2895
2896
2897
2898
2899
2900
2901
2902
2903
2904
2905
2906
2907
2908
2909
2910
2911
2912
2913
2914
2915
2916
2917
2918
2919
2920
2921
2922
2923
2924
2925
2926
2927
2928
2929
2930
2931
2932
2933
2934
2935
2936
2937
2938
2939
2940
2941
2942
2943
2944
2945
2946
2947
2948
2949
2950
2951
2952
2953
2954
2955
2956
2957
2958
2959
2960
2961
2962
2963
2964
2965
2966
2967
2968
2969
2970
2971
2972
2973
2974
2975
2976
2977
2978
2979
2980
2981
2982
2983
2984
2985
2986
2987
2988
2989
2990
2991
2992
2993
2994
2995
2996
2997
2998
2999
3000
3001
3002
3003
3004
3005
3006
3007
3008
3009
3010
3011
3012
3013
3014
3015
3016
3017
3018
3019
3020
3021
3022
3023
3024
3025
3026
3027
3028
3029
3030
3031
3032
3033
3034
3035
3036
3037
3038
3039
3040
3041
3042
3043
3044
3045
3046
3047
3048
3049
3050
3051
3052
3053
3054
3055
3056
3057
3058
3059
3060
3061
3062
3063
3064
3065
3066
3067
3068
3069
3070
3071
3072
3073
3074
3075
3076
3077
3078
3079
3080
3081
3082
3083
3084
3085
3086
3087
3088
3089
3090
3091
3092
3093
3094
3095
3096
3097
3098
3099
3100
3101
3102
3103
3104
3105
3106
3107
3108
3109
3110
3111
3112
3113
3114
3115
3116
3117
3118
3119
3120
3121
3122
3123
3124
3125
3126
3127
3128
3129
3130
3131
3132
3133
3134
3135
3136
3137
3138
3139
3140
3141
3142
3143
3144
3145
3146
3147
3148
3149
3150
3151
3152
3153
3154
3155
3156
3157
3158
3159
3160
3161
3162
3163
3164
3165
3166
3167
3168
3169
3170
3171
3172
3173
3174
3175
3176
3177
3178
3179
3180
3181
3182
3183
3184
3185
3186
3187
3188
3189
3190
3191
3192
3193
3194
3195
3196
3197
3198
3199
3200
3201
3202
3203
3204
3205
3206
3207
3208
3209
3210
3211
3212
3213
3214
3215
3216
3217
3218
3219
3220
3221
3222
{
  "1": {
    "results": {
      "completion": {
        "id": "chatcmpl-AIHIPLyXp5Go74Qys43ojpQ0czAzb",
        "choices": [
          {
            "finish_reason": "stop",
            "index": 0,
            "message": {
              "content": "Recent advancements in multi-omics approaches, including proteomics and metabolomics, have significantly enhanced our understanding of the pathogenesis of Type 2 diabetes (T2D). These approaches allow for a comprehensive analysis of the molecular components involved in T2D, providing insights into the disease's complexity and heterogeneity.\n\n1. **Integration of Omics Data**: The integration of data from proteomics, genomics, and transcriptomics helps in understanding the pathophysiology of T2D and the varied responses to different glucose-lowering therapies. This holistic view is crucial for identifying the molecular underpinnings of T2D and tailoring personalized treatment strategies [1].\n\n2. **Metabolomics Insights**: Metabolomics studies have identified specific metabolites, such as branched-chain and aromatic amino acids, that are associated with obesity and T2D. These findings suggest that metabolic alterations play a significant role in the development of T2D, offering potential targets for therapeutic intervention [1].\n\n3. **Complex Interactions**: T2D is caused by complex interactions between genetic and environmental factors. Metabolomics has helped elucidate these interactions by identifying metabolic footprints associated with T2D, thus moving from mere information gathering to actionable knowledge [2], [3].\n\n4. **Biomarker Discovery**: Advances in omics technologies have led to the discovery of numerous candidate biomarkers for T2D. Although only a small proportion of these biomarkers have been thoroughly investigated, they hold promise for improving the prediction and management of T2D [9].\n\n5. **Beyond Traditional Models**: The use of metabolomics has expanded our understanding of T2D beyond traditional models focused on the pancreas, liver, and muscle. This broader perspective is essential for developing more effective prevention and treatment strategies [10].\n\nOverall, these advancements in multi-omics approaches provide a more detailed and nuanced understanding of T2D pathogenesis, paving the way for improved diagnostic and therapeutic options.",
              "role": "assistant"
            }
          }
        ],
        "created": 1728919753,
        "model": "gpt-4o-2024-08-06",
        "object": "chat.completion",
        "system_fingerprint": "fp_a20a4ee344",
        "usage": {
          "completion_tokens": 394,
          "prompt_tokens": 1432,
          "total_tokens": 1826
        }
      },
      "search_results": {
        "vector_search_results": [
          {
            "id": "012b6e5f-ab45-53aa-a392-45a46916e752",
            "score": 0.7536256166473412,
            "metadata": {
              "text": "proteomics, genomics, and transcriptomics) are based on the study of constituents of the cell or body in a collective way. The   ndings made with use of these approaches are being integrated to better understand the pathophysiology of type 2 diabetes and the heterogeneity of responses to di  erent glucose-lowering therapies. Findings from studies that used metabolomics and lipidomics showed that increases in branched-chain and aromatic aminoacids were associated with obesity and type 2 diabetes.",
              "title": "2014 - Pathophysiology and treatment of type 2 diabetes.pdf",
              "version": "v0",
              "chunk_order": 51,
              "document_id": "ab9288ab-e3ad-58f1-b5ba-183ee17ce4bd",
              "extraction_id": "8b15673a-deaf-5e34-945c-ea2a1365552d",
              "associatedQuery": "How do recent advancements in multi-omics approaches, including proteomics and metabolomics, contribute to our understanding of Type 2 diabetes pathogenesis?"
            }
          },
          {
            "id": "aaf89eb0-09a8-517d-b8ae-4e76a8211be6",
            "score": 0.7278114557266235,
            "metadata": {
              "text": "Metabolomics Applied to Diabetes Research\nMoving From Information to Knowledge\nJames R. Bain, Robert D. Stevens, Brett R. Wenner, Olga Ilkayeva, Deborah M. Muoio, and\nChristopher B. Newgard\nType 2 diabetes is caused by a complex set of\ninteractions between genetic and environmentalfactors. Recent work has shown that human type2 diabetes is a constellation of disorders associ-\nated with polymorphisms in a wide array of genes, witheach individual gene accounting for /H110211% of disease risk",
              "title": "2009 - Metabolomics Applied to Diabetes Research.pdf",
              "version": "v0",
              "chunk_order": 0,
              "document_id": "a6ae2fb6-88ae-588f-a98d-b6092f886ed9",
              "extraction_id": "380e9a2e-8f9f-5f9e-ba20-3695b1c60fda",
              "associatedQuery": "How do recent advancements in multi-omics approaches, including proteomics and metabolomics, contribute to our understanding of Type 2 diabetes pathogenesis?"
            }
          },
          {
            "id": "6919bc75-2637-5359-9c05-96d192be8c4e",
            "score": 0.7128690481185913,
            "metadata": {
              "text": "between protein signals and type 2 diabetes incidence.\nActa Diabetol. doi: 10.1007/s00592-012-0376-3\n82. Bain JR, Stevens RD, Wenner BR, Ilkayeva O, Muoio DM,\nNewgard CB (2009) Metabolomics applied to diabetes re-search: moving from information to knowledge. Diabetes 58:\n2429 244383. Suhre K, Meisinger C, Dring A et al (2011) Metabolic footprint of\ndiabetes: a multiplatform metabolomics study in an epidemiological\nsetting. PLoS One 5:e13953",
              "title": "2014 - The potential of novel biomarkers to improve risk prediction of type 2 diabetes.pdf",
              "version": "v0",
              "chunk_order": 148,
              "document_id": "2bc2f4be-378f-5ced-8288-e2a132a94540",
              "extraction_id": "75485c9d-6c66-52fe-8fb1-e6d2440a7f49",
              "associatedQuery": "How do recent advancements in multi-omics approaches, including proteomics and metabolomics, contribute to our understanding of Type 2 diabetes pathogenesis?"
            }
          },
          {
            "id": "93455356-fe0b-58f4-9ae7-58f932d33560",
            "score": 0.6947442293167114,
            "metadata": {
              "text": "The future: genetics, epigenetics, and omics\nAlthough understanding of the genetics of type 2 diabetes has advanced rapidly, much remains unknown. How genes interact with the environment to cause progressive loss of -cell function is unclear. Environmental factors and hyperglycaemia could contribute to epigenetic changes in DNA and histones, thereby modifying gene expression in organs implicated in the pathogenesis and progression of type 2 diabetes, including in  cells.\n82,83",
              "title": "2014 - Pathophysiology and treatment of type 2 diabetes.pdf",
              "version": "v0",
              "chunk_order": 49,
              "document_id": "ab9288ab-e3ad-58f1-b5ba-183ee17ce4bd",
              "extraction_id": "8b15673a-deaf-5e34-945c-ea2a1365552d",
              "associatedQuery": "How do recent advancements in multi-omics approaches, including proteomics and metabolomics, contribute to our understanding of Type 2 diabetes pathogenesis?"
            }
          },
          {
            "id": "cfc35db4-346c-55fd-b0bc-fa3cac307731",
            "score": 0.6932104229927063,
            "metadata": {
              "text": "potential to make far-reaching contributions to our understanding of molecular\nbasis of T2D and the development of novel strategies for patient care.\n2.1 Introduction\nType 2 diabetes (T2D) is a common, chronic disorder whose prevalence is increas-ing rapidly across the globe. Like other complex diseases, T2D represents achallenge for genetic studies aiming to uncover the underlying pathophysiological\nmechanisms. It is predicted that T2D will affect 592 million individuals by 2035",
              "title": "2016 - Genome-Wide Association Studies of Type 2 Diabetes.pdf",
              "version": "v0",
              "chunk_order": 3,
              "document_id": "185aad8a-6a5b-5b18-81c4-ef251edef5e7",
              "extraction_id": "7cec13b8-d349-5ea4-b866-17fc760d364c",
              "associatedQuery": "How do recent advancements in multi-omics approaches, including proteomics and metabolomics, contribute to our understanding of Type 2 diabetes pathogenesis?"
            }
          },
          {
            "id": "3b5c1a49-cb11-57ef-9046-e3c8f7af589e",
            "score": 0.692622322283846,
            "metadata": {
              "text": "inthepathogenesisoftype2diabetesandmetabolism, Current\nOpinion in Clinical Nutrition and Metabolic Care ,vol.10,no .4,\npp .420426,2007 .\n[110] M.C.Cornelis,E.J.T.Tchetgen,L.Liangetal.,Gene-environ-\nment interactions in genome-wide association studies: a com-\nparative study of tests applied to empirical studies of type 2\ndiabetes, American Journal of Epidemiology ,v o l.17 5,no .3,p p .\n191202,2012.\n[111] M.L.Metzker,Sequencingtechnologiesthenextgeneration,\nNature Reviews Genetics ,vol.11,no.1,pp.3146,2010.",
              "title": "2013 - Variants of Insulin-Signaling Inhibitor Genes.pdf",
              "version": "v0",
              "chunk_order": 164,
              "document_id": "d43a59e8-fe3b-503a-863b-235af8790f2a",
              "extraction_id": "f258a3c5-02d6-5f8f-a989-27f6c795145c",
              "associatedQuery": "How do recent advancements in multi-omics approaches, including proteomics and metabolomics, contribute to our understanding of Type 2 diabetes pathogenesis?"
            }
          },
          {
            "id": "b74d0bb9-eb0d-59bb-8a37-d3425d5591a2",
            "score": 0.6839994192123413,
            "metadata": {
              "text": "meta-ana lysis provides insight intothegenetic architecture oftype2diabetes susceptibility. NatGenet.\n2014; 46:234 244. https://doi.or g/10.103 8/ng.2897 PMID: 24509480\n26. Morris AP,Voight BF,Teslovich TM,Ferreira T,Segr A-V, Steinthorsdot tirV,etal.Large-sc aleassoci-\nation analysis provide sinsights intothegenetic architecture andpathophysi ology oftype2diabetes.\nNatGenet. 2012; 44:981 990. https://doi.or g/10.103 8/ng.2383 PMID: 228859 22",
              "title": "2021 - A genome-wide association study identifies 5 loci associated with frozen shoulder and implicates diabetes as a causal risk factor.pdf",
              "version": "v0",
              "chunk_order": 93,
              "document_id": "8276e137-4591-51bd-9351-f4d27d3b35da",
              "extraction_id": "2052d37d-f778-53e2-a2f9-9e4311e8a953",
              "associatedQuery": "How do recent advancements in multi-omics approaches, including proteomics and metabolomics, contribute to our understanding of Type 2 diabetes pathogenesis?"
            }
          },
          {
            "id": "ead10261-182f-5ab1-9af0-ce8a17677d4a",
            "score": 0.6823235750198364,
            "metadata": {
              "text": "monitoring and preventing progression to costly co-morbidities.\nThe principal concept of metabolomics being able to find some\nmetabolites differing in a control and a type 2 diabetic group is\nestablished. It is not our goal here to show this once again. The\nquestions we ask are rather How well are different approaches\nsuited to attain this goal? and What are optimal settings under\nwhich such studies can be successful?. Others have already\ninvestigated these questions before [16,17,18]. However, we",
              "title": "2010 - Metabolic Footprint of Diabetes A Multiplatform.pdf",
              "version": "v0",
              "chunk_order": 18,
              "document_id": "b199607e-293e-56e8-88c8-e0716d1ee9eb",
              "extraction_id": "97b6d492-9139-50ec-9685-53a803f5c995",
              "associatedQuery": "How do recent advancements in multi-omics approaches, including proteomics and metabolomics, contribute to our understanding of Type 2 diabetes pathogenesis?"
            }
          },
          {
            "id": "4971b4de-b190-56b5-b7b6-64b2c8e2a565",
            "score": 0.6784495115280151,
            "metadata": {
              "text": "Owing to current advances in -omics technologies, such as\ngenomics, transcriptomics, proteomics and metabolomics, the\nnumber of candidate biomarkers keeps growing; however,\nonly a small proportion of these has been investigated withreference to their potential to improve the prediction of type 2\ndiabetes.\nGenetic variants\nThe heritability of glycaemic traits and type 2 diabetes is high\n[40], and the large genome-wide association studies published\nto date since the first in 2007, based on up to >10\n5study",
              "title": "2014 - The potential of novel biomarkers to improve risk prediction of type 2 diabetes.pdf",
              "version": "v0",
              "chunk_order": 53,
              "document_id": "2bc2f4be-378f-5ced-8288-e2a132a94540",
              "extraction_id": "496d9615-7530-530c-bea1-62fe63ea54ca",
              "associatedQuery": "How do recent advancements in multi-omics approaches, including proteomics and metabolomics, contribute to our understanding of Type 2 diabetes pathogenesis?"
            }
          },
          {
            "id": "01a2230a-b91d-57b6-b138-7aae805f4383",
            "score": 0.6783756017684937,
            "metadata": {
              "text": "have improved our understanding of the complexity of \nT2DM pathophysiology, beyond the classic triumvirate of \n-cell, skeletal muscle and liver87. However, the ability of \nthese biomarkers to predict future risk of T2DM beyond \nanthropometric measures, lifestyle factors and fasting \nlevels of glucose and lipids is still debatable87.\nWithin the past 7years, a complementary, novel set of \nT2DM biomarkers has largely been generated by metabo-\nlomic studies, which systematically analyse metabolites",
              "title": "2018 - Global aetiology and epidemiology of type 2 diabetes mellitus and its complications.pdf",
              "version": "v0",
              "chunk_order": 67,
              "document_id": "8bc8f3d4-968f-5252-ab4c-832b92e9ec0d",
              "extraction_id": "751ccb98-2846-5ca7-8ab8-2684100c28fa",
              "associatedQuery": "How do recent advancements in multi-omics approaches, including proteomics and metabolomics, contribute to our understanding of Type 2 diabetes pathogenesis?"
            }
          }
        ],
        "kg_search_results": null
      }
    }
  },
  "2": {
    "results": {
      "completion": {
        "id": "chatcmpl-AIHIcyJRqSPUlYLtzZ5hVN5aLL9iw",
        "choices": [
          {
            "finish_reason": "stop",
            "index": 0,
            "message": {
              "content": "The latest meta-analyses of large-scale genome-wide association studies (GWAS) have identified several novel loci associated with diabetes. Specifically, the DIAbetes Genetics Replication and Meta-analysis (DIAGRAM) Consortium, using genotype data from almost 150,000 individuals predominantly of European descent, identified 10 novel type 2 diabetes (T2D) susceptibility loci that reached genome-wide significance [8].",
              "role": "assistant"
            }
          }
        ],
        "created": 1728919766,
        "model": "gpt-4o-2024-08-06",
        "object": "chat.completion",
        "system_fingerprint": "fp_a20a4ee344",
        "usage": {
          "completion_tokens": 82,
          "prompt_tokens": 1620,
          "total_tokens": 1702
        }
      },
      "search_results": {
        "vector_search_results": [
          {
            "id": "0c0634ba-c437-52d3-b3a9-caa5eda120c6",
            "score": 0.7532689571380615,
            "metadata": {
              "text": "wide association study identi es novel risk loci for type 2 diabetes. Nature\n(2007) 445:881 5. doi: 10.1038/nature05616\n27. Scott LJ, Mohlke KL, Bonnycastle LL, Willer CJ, Li Y, Duren WL, et al. A\ngenome-wide association study of type 2 diabetes in Finns detects multiple\nsusceptibility variants. Science (2007) 316:1341 5. doi: 10.1126/science.1142382\n28. Fuchsberger C, Flannick J, Teslovich TM, Mahajan A, Agarwala V, Gaulton\nKJ, et al. The genetic architecture of type 2 diabetes. Nature (2016) 536:41 7.",
              "title": "2020 - Functional Genomics in Pancreatic \u03b2 Cells Recent Advances in Gene Deletion and Genome Editing Technologies for Diabetes Research.pdf",
              "version": "v0",
              "chunk_order": 162,
              "document_id": "51350055-d53c-5692-ab53-337b8a8bafd6",
              "extraction_id": "5f148509-8a55-5e9c-8c68-e327f519c1c9",
              "associatedQuery": "What novel diabetic loci have been identified through the latest meta-analyses of large-scale genome-wide association studies (GWAS)?"
            }
          },
          {
            "id": "1ab64c6e-e930-597e-bc12-ed540eabcf46",
            "score": 0.7524292020541918,
            "metadata": {
              "text": "novel loci for type 1 diabetes. Diabetes 58:290295. DOI: https://doi.org/10.2337/db08-1022, PMID: \n18840781\nHuang J, Ellinghaus D, Franke A, Howie B, Li Y . 2012. 1000 Genomes- based imputation identifies novel and \nrefined associations for the Wellcome Trust Case Control Consortium phase 1 Data. European Journal of \nHuman Genetics 20:801805. DOI: https://doi.org/10.1038/ejhg.2012.3, PMID: 22293688\nHundhausen C, Roth A, Whalen E, Chen J, Schneider A, Long SA, Wei S, Rawlings R, Kinsman M, Evanko SP ,",
              "title": "2022 - A genome-wide functional genomics approach uncovers genetic determinants of immune phenotypes in type 1 diabetes.pdf",
              "version": "v0",
              "chunk_order": 167,
              "document_id": "368e0215-393e-5bec-a87c-e976adaa3ca5",
              "extraction_id": "692b342f-5d48-5046-84f9-37f1cf4275b5",
              "associatedQuery": "What novel diabetic loci have been identified through the latest meta-analyses of large-scale genome-wide association studies (GWAS)?"
            }
          },
          {
            "id": "46ac5572-ac56-5f29-b7bf-49a1e29d3936",
            "score": 0.7521799802780151,
            "metadata": {
              "text": "general population, these loci show limited effect in DKD,\nespecially in individuals with type 1 diabetes [ 6]. Genome-\nwide association studies (GWAS) have previously identified ahandful of genetic loci for DKD at the genome-wide signifi-\ncance level ( p<510\n8)[711]. Recently, a meta-analysis of\nGWAS, including up to 19,406 individuals with type 1 diabetes\nfrom the Diabetic Nephropathy Collaborative Research",
              "title": "2022 - Genome-wide meta-analysis and omics integration identifies novel genes associated with diabetic kidney disease.pdf",
              "version": "v0",
              "chunk_order": 11,
              "document_id": "b9194555-5fdb-549e-9edb-d108132a7dd1",
              "extraction_id": "d7e0e5ad-bad5-5b14-896e-45702d6605f9",
              "associatedQuery": "What novel diabetic loci have been identified through the latest meta-analyses of large-scale genome-wide association studies (GWAS)?"
            }
          },
          {
            "id": "6d5d4c24-5bc8-539a-9faa-8b2370f8c87a",
            "score": 0.7515840530395508,
            "metadata": {
              "text": "Table 2.1 Major published T2D GWAS and meta-analyses\nStudyEthnicity/\norigin NcasesaN\ncontrolsaNovel loci identiedGWAS or\nmeta-analysis\ndiscoveryapproach GWAS arrayReference\npanel forimputationT2D phenotype\ndenition/otherspecs\nDiabetes Gene\nDiscovery Group\n(Sladek et al. 2007 ),\nNatureEuropean 694 645 SLC30A8 ,HHEX /IDE GWA Illumina 300k +  Family history of\nT2D, AAO <45\nyears, BMI <30\nkg/m\n2\nFinlandUS Investi-gation of NIDDMGenetics (FUSION)(Scott et al. 2007a ),\nScienceEuropean 1161 1174 CDKN2A/2B ,",
              "title": "2016 - Genome-Wide Association Studies of Type 2 Diabetes.pdf",
              "version": "v0",
              "chunk_order": 27,
              "document_id": "185aad8a-6a5b-5b18-81c4-ef251edef5e7",
              "extraction_id": "a620eedf-5d5b-506f-97f5-c25dbe0493c0",
              "associatedQuery": "What novel diabetic loci have been identified through the latest meta-analyses of large-scale genome-wide association studies (GWAS)?"
            }
          },
          {
            "id": "54da57b3-e577-5c00-a7d5-6f569a41d28b",
            "score": 0.7501821517944336,
            "metadata": {
              "text": "scale gene-centric meta-analysis across 39 studies identifies type 2diabetes loci. Am J Hum Genet. 2012;90(3):410 25.\n13. Haiman C, Fesinmeyer M, Spencer K, Buzkova P, V oruganti V ,\nWan P, et al. Consistent directions ofeffect for established type 2\ndiabetes risk variants across populations: the Population Architectureusing Genomics and Epidemiology (PAGE) Consortium. Diabetes.\n2012;61(6):1642 7.In the most complete trans-ethnic T2D GWAS",
              "title": "2012 - Recent Developments in the Genetic and Genomic Basis of Type 2 Diabetes.pdf",
              "version": "v0",
              "chunk_order": 62,
              "document_id": "7d051350-d939-5183-be22-742727573a75",
              "extraction_id": "1213249d-8ed3-5d13-9137-f11b87a7a78b",
              "associatedQuery": "What novel diabetic loci have been identified through the latest meta-analyses of large-scale genome-wide association studies (GWAS)?"
            }
          },
          {
            "id": "0cf52952-0d83-58ed-b402-05dd2f085841",
            "score": 0.7471976280212402,
            "metadata": {
              "text": "9. Sladek R, Rocheleau G, Rung J, Dina C, Shen L, et al. (2007) A genome-wide\nassociation study identifies novel risk loci for type 2 diabetes. Nature 445:881885.\n10. Zeggini E, Scott LJ, Saxena R, Voight BF, Marchini JL, et al. (2008) Meta-\nanalysis of genome-wide association data and large-scale replication identifies\nadditional susceptibility loci for type 2 diabetes. Nat Genet 40: 638645.11. Altshuler D, Daly MJ, Lander ES (2008) Genetic mapping in human disease.\nScience 322: 881888.",
              "title": "2010 - Liver and Adipose Expression Associated SNPs.pdf",
              "version": "v0",
              "chunk_order": 149,
              "document_id": "ebeef1bf-341d-5aa1-807b-1f23186cf2bc",
              "extraction_id": "35ce49d5-7af3-5f24-927c-f800e8ae024d",
              "associatedQuery": "What novel diabetic loci have been identified through the latest meta-analyses of large-scale genome-wide association studies (GWAS)?"
            }
          },
          {
            "id": "2a91a466-c271-5368-b0a1-cf15e6478bb1",
            "score": 0.7458800077438354,
            "metadata": {
              "text": "scale ongoing efforts to localize and characterize T2D susceptibility\ngenes using genome-wide association study (GWAS) approaches. To\ndate, the GWAS method has achieved substantial success in localizing\nnovel T2D susceptibility loci and loci for T2D-related glycemic traits\n(about 90 loci), obesity loci (~90), and loci for metabolic syndrome or\nits components (~50 loci), e.g. reviews: [4,20,28,29,41,47,51,64,65,67] .\nHowever, common variants identi ed by GWAS explain only about",
              "title": "2016 - Transcriptomics in type 2 diabetes Bridging the gap between genotype and phenotype.pdf",
              "version": "v0",
              "chunk_order": 17,
              "document_id": "98e49a13-9887-5b27-879b-0816a3da1c1d",
              "extraction_id": "71934c29-338d-57a2-8f45-e3e795e0ec9b",
              "associatedQuery": "What novel diabetic loci have been identified through the latest meta-analyses of large-scale genome-wide association studies (GWAS)?"
            }
          },
          {
            "id": "de3b49f1-9dcc-5056-8232-b76e5f985736",
            "score": 0.7458185705054672,
            "metadata": {
              "text": "T2D GWA meta-analysis performed by the DIAbetes Genet-ics Replication and Meta-analysis (DIAGRAM) Consortium\n[6]. Using genotype data from almost 150,000 individuals,\npredominantly of European descent, the consortium was ableto define 10 novel T2D-susceptibility loci to genome-wide\nsignificance, and to highlight several hundreds more that,\nwhilst failing to reach the stringent criteria typically regardedas proof, are nonetheless highly likely to reflect genuine",
              "title": "2012 - Recent Developments in the Genetic and Genomic Basis of Type 2 Diabetes.pdf",
              "version": "v0",
              "chunk_order": 13,
              "document_id": "7d051350-d939-5183-be22-742727573a75",
              "extraction_id": "924d35c5-0ee8-53a7-9fdf-9309a27ce9ae",
              "associatedQuery": "What novel diabetic loci have been identified through the latest meta-analyses of large-scale genome-wide association studies (GWAS)?"
            }
          },
          {
            "id": "72622bca-2fce-5732-9c8b-2909d231d09d",
            "score": 0.745085597038269,
            "metadata": {
              "text": "18. Sladek R, Rocheleau G, Rung J, Dina C, Shen L, Serre D, et al. \nA genome-wide association study identifies novel risk loci for type 2 diabetes. Nature 2007;445:881-885.\n19. Scott LJ, Mohlke KL, Bonnycastle LL, Willer CJ, Li Y, Duren \nWL, et al. A genome-wide association study of type 2 diabetes \nin Finns detects multiple susceptibility variants. Science 2007; \n316:1341-1345.\n20. Diabetes Genetics Initiative of Broad Institute of Harvard and \nMIT , Lund University, and Novartis Institutes of BioMedical",
              "title": "2012 - Finding Genetic Risk Factors of Gestational Diabetes.pdf",
              "version": "v0",
              "chunk_order": 42,
              "document_id": "81d6ccba-6203-5879-b206-b8711d1ff35c",
              "extraction_id": "e7bf3f2d-8180-5a84-965c-8289f107a718",
              "associatedQuery": "What novel diabetic loci have been identified through the latest meta-analyses of large-scale genome-wide association studies (GWAS)?"
            }
          },
          {
            "id": "5af0c2b9-9957-5c8f-b8ae-c115e365576f",
            "score": 0.7448982818394391,
            "metadata": {
              "text": "additive, dominant, and recessive) and did not adjust for mul -\ntiple comparisons. The third study is the largest GWAS con -\nducted to date and is a meta-analysis of two GWASs, Genetics \nof Kidneys in Diabetes (GoKinD) and Epidemiology of Dia -\nbetes Interventions and Complications (EDIC) studies [24]. \nThis study by Grassi et al. [24] involved 2,829 European sub -\njects with T1DM. The most significant variant was rs476141 \nlocated in a long non-coding RNA ( LOC339529 ) in chromo -",
              "title": "2015 - Genetic Studies on Diabetic Microvascular Complications.pdf",
              "version": "v0",
              "chunk_order": 28,
              "document_id": "1df9d9a8-0fb0-5a03-9749-9471b4b2b2f3",
              "extraction_id": "d3335459-5fec-5104-932f-f4fd7566edf7",
              "associatedQuery": "What novel diabetic loci have been identified through the latest meta-analyses of large-scale genome-wide association studies (GWAS)?"
            }
          }
        ],
        "kg_search_results": null
      }
    }
  },
  "3": {
    "results": {
      "completion": {
        "id": "chatcmpl-AIHIljZhr1AUuC7qfsdHZaKkRKz2A",
        "choices": [
          {
            "finish_reason": "stop",
            "index": 0,
            "message": {
              "content": "Epigenetic modifications, such as DNA methylation and histone modification, play significant roles in influencing the expression of diabetes-related genes. \n\n1. **DNA Methylation**: This modification can lead to the silencing of key genes involved in diabetes. For example, increased DNA methylation of the promoter of the PPARGC1A gene, which is crucial for regulating mitochondrial genes, has been observed in diabetic islets. This methylation can modulate diabetes by altering gene expression [1]. Additionally, altered DNA methylation patterns can change the expression of inflammatory and pathological genes, contributing to the development of diabetic complications [2].\n\n2. **Histone Modifications**: These modifications can affect chromatin structure and gene expression. In the context of diabetes, histone post-translational modifications (PTMs) have been associated with the regulation of genes susceptible to type 1 diabetes [3]. Changes in chromatin histone modifications can regulate pathological gene expression in diabetes, influencing the disease's progression [2].\n\nOverall, both DNA methylation and histone modifications can lead to the repression or activation of genes that are critical in the pathogenesis and progression of diabetes, affecting insulin production, inflammation, and other metabolic processes.",
              "role": "assistant"
            }
          }
        ],
        "created": 1728919775,
        "model": "gpt-4o-2024-08-06",
        "object": "chat.completion",
        "system_fingerprint": "fp_6b68a8204b",
        "usage": {
          "completion_tokens": 247,
          "prompt_tokens": 1468,
          "total_tokens": 1715
        }
      },
      "search_results": {
        "vector_search_results": [
          {
            "id": "eb133825-7500-5160-b39a-298961323f9c",
            "score": 0.7901671764304395,
            "metadata": {
              "text": "diabetes due to epigenetic silencing of Pdx1, a key transcription factor that regulates insulin gene 301 \nexpression and beta cell differentiation. Both hi stone modifications a nd DNA methylation were 302 \nimplicated (111). In another study, it was shown th at, in diabetic islets , there was increased DNA 303 \nmethylation of the promoter of PPAR-gamma co-activator 1  gene ( PPARGC1A ), a factor that 304 \nplays a key role in regulating mitochondrial ge nes and in the modulation of diabetes (87). 305",
              "title": "2010 - The Role of Epigenetics in the Pathology of Diabetic Complications.pdf",
              "version": "v0",
              "chunk_order": 63,
              "document_id": "766edfd5-4756-51bf-b636-c94b041d030c",
              "extraction_id": "77eb6a3d-2e3b-5304-873f-4fe14ec290d1",
              "associatedQuery": "How do epigenetic modifications, such as DNA methylation and histone modification, influence the expression of diabetes-related genes?"
            }
          },
          {
            "id": "a97f140f-63b1-5963-9c38-d90f59f58ced",
            "score": 0.786042356075119,
            "metadata": {
              "text": "altered DNA methylation (DNA-me) at  various genes in target cells  all of which over time can 1009 \nresult in changes to the expr ession patterns of inflammatory, sclerotic and other pathological 1010 \ngenes and the ultimate developm ent of diabetic complications. 1011 \n 1012 \nFigure 2: Model for epigenetic regulation of pa thological gene expressi on in diabetes via 1013 \nchanges in chromatin histone modifications. Post translational modifications on the N- 1014",
              "title": "2010 - The Role of Epigenetics in the Pathology of Diabetic Complications.pdf",
              "version": "v0",
              "chunk_order": 206,
              "document_id": "766edfd5-4756-51bf-b636-c94b041d030c",
              "extraction_id": "21de4c95-4171-52bb-a867-2df5336c3c71",
              "associatedQuery": "How do epigenetic modifications, such as DNA methylation and histone modification, influence the expression of diabetes-related genes?"
            }
          },
          {
            "id": "41899c3d-64db-556a-882a-4e39b964c6d5",
            "score": 0.7817012564807093,
            "metadata": {
              "text": "Dependent Demethylation of Regulatory Elements Correlates with Chromatin State and Improved Cell Function. Cell Metab.\n2015 ,22, 619632. [CrossRef]\n228. Zhang, H.; Pollin, T.I. Epigenetics Variation and Pathogenesis in Diabetes. Curr. Diab. Rep. 2018 ,18, 121. [CrossRef]\n229. Miao, F.; Chen, Z.; Zhang, L.; Liu, Z.; Wu, X.; Yuan, Y.-C.; Natarajan, R. Proles of epigenetic histone post-translational\nmodications at type 1 diabetes susceptible genes. J. Biol. Chem. 2012 ,287, 1633516345. [CrossRef]",
              "title": "2021 - Epigenetics of Aging and Aging-Associated Diseases.pdf",
              "version": "v0",
              "chunk_order": 271,
              "document_id": "70945353-4808-539a-80f9-5632c27913e5",
              "extraction_id": "3d7cb780-5f0a-5500-8176-4c2055cac9dc",
              "associatedQuery": "How do epigenetic modifications, such as DNA methylation and histone modification, influence the expression of diabetes-related genes?"
            }
          },
          {
            "id": "6f647f65-0c70-5abf-8944-e2b1ade8ee1d",
            "score": 0.7764175765638824,
            "metadata": {
              "text": "Epigenetic Mechanisms in Diabetic Complications     14 \nDNA methylation at prom oter CpG islands has been associ ated with gene repression and 292 \nis a well studied epigenetic mark in the c ontext of tumor suppressor genes and cancer (129). 293 \nHowever, much less is known a bout DNA methylation in diabetes . A recent report has shown 294 \nthat the insulin promoter DNA was methylated in mouse embryonic stem cells and only becomes 295",
              "title": "2010 - The Role of Epigenetics in the Pathology of Diabetic Complications.pdf",
              "version": "v0",
              "chunk_order": 61,
              "document_id": "766edfd5-4756-51bf-b636-c94b041d030c",
              "extraction_id": "77eb6a3d-2e3b-5304-873f-4fe14ec290d1",
              "associatedQuery": "How do epigenetic modifications, such as DNA methylation and histone modification, influence the expression of diabetes-related genes?"
            }
          },
          {
            "id": "883de652-2a30-5587-89bb-474facc861fe",
            "score": 0.7744899026897524,
            "metadata": {
              "text": "Epigenetics: deciphering its role in diabetes and \nits chronic complications. Clin. Exp. Pharmacol. \nPhysiol.  38, 401409 (2011).\n61. Cooper, M.E. & El-Osta, A. Epigenetics: \nmechanisms and implications for diabetic complications. Circ. Res.  107, 14031413 \n(2010).\n62. Miao, F. etal. Profiles of epigenetic histone post-\ntranslational modifications at type1 diabetes \nsusceptible genes. J.Biol. Chem.  287,  \n1633516345 (2012).\n63. Sapienza, C. etal. DNA methylation profiling",
              "title": "2014 - Diabetic nephropathy\u2014emerging epigenetic mechanisms.pdf",
              "version": "v0",
              "chunk_order": 160,
              "document_id": "be05127e-1be8-5573-b571-51a11c3b2be2",
              "extraction_id": "cbbe696b-8541-537a-ac5f-77b82cdb8201",
              "associatedQuery": "How do epigenetic modifications, such as DNA methylation and histone modification, influence the expression of diabetes-related genes?"
            }
          },
          {
            "id": "796ed77e-4539-543b-a392-5736392f93ba",
            "score": 0.7697410812896816,
            "metadata": {
              "text": "Emerging evidence shows that epigenetic mecha-nisms in chromatin including histone PTMs,\nDNAme, and miRNAs also might play key roles in\nthe etiology of diabetes and DN. The persistence ofepigenetic modi cations triggered by diabetic stim-\nuli could be one of the key mechanisms underlying\nmetabolic memory. A role for several HMTs and thecorresponding histone PTMs has been shown in the\nexpression of brotic and in ammatory genes asso-",
              "title": "2013 - Epigenetic Modifications in the Pathogenesis of Diabetic Nephropathy.pdf",
              "version": "v0",
              "chunk_order": 104,
              "document_id": "9cffb997-a205-5f72-89a6-945df5b9af28",
              "extraction_id": "7680731d-0b98-5f45-85f9-d06883504dd1",
              "associatedQuery": "How do epigenetic modifications, such as DNA methylation and histone modification, influence the expression of diabetes-related genes?"
            }
          },
          {
            "id": "3f3fb648-0a87-5d2b-82c8-da1f3caf91b0",
            "score": 0.7688729992173019,
            "metadata": {
              "text": "inflammation-related epigenetic modifications: focus on DNA\nmethylation. Exerc Immunol Rev. 2015;21:26 41.\n17. Milagro FI, Mansego ML, De Miguel C, Martinez JA. Dietary\nfactors, epigenetic modifications and obesity outcomes: progresses\nand perspectives. Mol Aspects Med. 2013;34(4):782 812.\n18. Caramori ML, Kim Y , Goldfine AB, et al. Differential gene expres-\nsion in diabetic nephropathy in individuals with type 1 diabetes. J\nClin Endocrinol Metab. 2015;100(6):E876 82.",
              "title": "2016  - Epigenetic Mechanisms in Diabetic Kidney Disease.pdf",
              "version": "v0",
              "chunk_order": 71,
              "document_id": "6f773bda-0b8f-5da2-a9b5-e6c013d75050",
              "extraction_id": "767d65c7-b99d-5427-8f5a-4afa10669e11",
              "associatedQuery": "How do epigenetic modifications, such as DNA methylation and histone modification, influence the expression of diabetes-related genes?"
            }
          },
          {
            "id": "aaeb4ad0-7848-554e-8ec1-2b5a094d3112",
            "score": 0.7677191724693532,
            "metadata": {
              "text": "elevated glucose level is not the only factor that leads to mal-\nadaptive epigenetic modifications in diabetes. DNA methyla-\ntion can also be influenced by reactive oxygen species, both\ndirectly through oxidative m odification DNA preventing\nmethylation and indirectly through its effects on methylation\nwriting/erasing enzymes [ 15]. Many other factors including\nhypoxia, inflammation, cytokines and growth factors, drugs,\nnutrition and even physical activity can modify epigenetic",
              "title": "2016  - Epigenetic Mechanisms in Diabetic Kidney Disease.pdf",
              "version": "v0",
              "chunk_order": 31,
              "document_id": "6f773bda-0b8f-5da2-a9b5-e6c013d75050",
              "extraction_id": "7a924f08-78ef-528a-8f9e-7bc12b004ff2",
              "associatedQuery": "How do epigenetic modifications, such as DNA methylation and histone modification, influence the expression of diabetes-related genes?"
            }
          },
          {
            "id": "c51c94d1-c182-5e77-8a14-6af868d66ee1",
            "score": 0.7632846691182058,
            "metadata": {
              "text": "1306 1313.\n31. Miao F, et al.; DCCT/EDIC Research Group (2014) Evaluating the role of epigenetic\nhistone modifications in the metabolic memory of type 1 diabetes. Diabetes 63(5):\n1748 1762.\n32. Reddy MA, Tak Park J, Natarajan R (2013) Epigenetic modifications in the patho-\ngenesis of diabetic nephropathy. Semin Nephrol 33(4):341 353.\n33. Bell CG, et al. (2010) Genome-wide DNA methylation analysis for diabetic nephrop-\nathy in type 1 diabetes mellitus. BMC Med Genomics 3:33.",
              "title": "2016 - Epigenomic profiling reveals an association betweenpersistence of DNA methylation and metabolicmemory in the DCCTEDIC type 1 diabetes cohor.pdf",
              "version": "v0",
              "chunk_order": 158,
              "document_id": "4b44425c-00c2-504f-be3c-34c002951cc2",
              "extraction_id": "745c11f0-789f-5f0a-9f19-69af42a19c75",
              "associatedQuery": "How do epigenetic modifications, such as DNA methylation and histone modification, influence the expression of diabetes-related genes?"
            }
          },
          {
            "id": "1c0b9205-340f-568f-990f-785c66154732",
            "score": 0.7595745551459362,
            "metadata": {
              "text": "ing that environment and diet may influence epigenetic mod-ifications that predispose individuals to diabetes [ 46]. Aber-\nrant DNAme has also been reported in the reduced expression\nof genes involved in diabetes and metabolism, and DNAme\nvariations have also been noted near diabetes susceptibility\ngenes and enhancers [ 15,47].\nGenomic DNA from diabetic patients with nephropa-\nthy relative to those without displayed differential meth-\nylation at several genes, including UNC13B , which had",
              "title": "2015 - Epigenetic mechanisms in diabetic complications and metabolic memory.pdf",
              "version": "v0",
              "chunk_order": 40,
              "document_id": "470f1f94-792d-5273-a88f-7e06084951c5",
              "extraction_id": "44d96546-84c3-51f1-85f9-22790a91d105",
              "associatedQuery": "How do epigenetic modifications, such as DNA methylation and histone modification, influence the expression of diabetes-related genes?"
            }
          }
        ],
        "kg_search_results": null
      }
    }
  },
  "4": {
    "results": {
      "completion": {
        "id": "chatcmpl-AIHItZX0vwpceBtjbHWMD13xwSdHl",
        "choices": [
          {
            "finish_reason": "stop",
            "index": 0,
            "message": {
              "content": "The gut microbiome plays a significant role in modulating host genetic predispositions to diabetes through various mechanisms. The interactions between host genetics, metabolism, and the immune system are crucial in shaping the microbiome and influencing the predisposition to diseases like diabetes [1]. \n\nResearch suggests that the gut microbiota affects numerous biological functions throughout the body, and its characterization has become a major research area in biomedicine. Studies have shown that gut bacteria play a fundamental role in diseases such as obesity and diabetes [6]. Dysbiosis, or an imbalance in the gut microbiota, is associated with obesity and type 2 diabetes (T2D), indicating that the composition of the gut microbiome can influence metabolic diseases [6].\n\nMoreover, genetic control of the gut microbiota composition has been observed, suggesting that host genetics can influence the microbiome, which in turn can modulate metabolic traits and potentially affect the risk of developing diabetes [10]. This implies a bidirectional relationship where genetics can shape the microbiome, and the microbiome can influence the expression of genetic predispositions to diabetes.\n\nIn summary, the gut microbiome interacts with host genetics and metabolism, potentially modulating the risk of diabetes by influencing metabolic processes and immune responses [1], [6], [10].",
              "role": "assistant"
            }
          }
        ],
        "created": 1728919783,
        "model": "gpt-4o-2024-08-06",
        "object": "chat.completion",
        "system_fingerprint": "fp_a20a4ee344",
        "usage": {
          "completion_tokens": 254,
          "prompt_tokens": 1527,
          "total_tokens": 1781
        }
      },
      "search_results": {
        "vector_search_results": [
          {
            "id": "d79a5c86-df6a-5b3d-93b4-a26f47b47e83",
            "score": 0.7613752768784431,
            "metadata": {
              "text": "diabetes?\n Is altered gut epithelial function and integrity important in the pathoge nesis of type 1 diabetes, and if so, what is the mechanism(s) and relation to\ndysbiosis and how do we demonstrate impaired function in humans?\n How important are the interactions between host genetics, metab olism and the immune system in shaping the microbiome and predilection to\ndisease?",
              "title": "2014 - The intestinal microbiome in type 1 diabetes.pdf",
              "version": "v0",
              "chunk_order": 93,
              "document_id": "138189d1-a16e-5c76-9b19-bd6877e7ee6d",
              "extraction_id": "b5ecc19b-eae0-51de-8e87-e5d01060e5be",
              "associatedQuery": "Can you elaborate on the role of the gut microbiome in modulating host genetic predispositions to diabetes?"
            }
          },
          {
            "id": "6cef232c-d7c6-5968-ad74-2903b688793a",
            "score": 0.7209897373926952,
            "metadata": {
              "text": "the gut, which might trigger an inflammatory response and play arole in the development of diabetes.\nIn conclusion, our data suggest that the levels of glucose tolerance or\nseverity of diabetes should be considered while linking microbiota with\nobesity and other metabolic diseases in humans. It is especially\nimportant for developing the strategies to modify the gut microbiota inorder to control metabolic diseases, since obesity and diabetes mightbe associated with different bacterial populations.\nMethods",
              "title": "2010 - Gut Microbiota in Human Adults with Type 2 Diabetes.pdf",
              "version": "v0",
              "chunk_order": 51,
              "document_id": "27aaf82e-944d-55b3-8b6d-cc43bcdb3eab",
              "extraction_id": "0eb4bb40-b16c-5203-8c83-dac0695d43a2",
              "associatedQuery": "Can you elaborate on the role of the gut microbiome in modulating host genetic predispositions to diabetes?"
            }
          },
          {
            "id": "89360f80-d048-5c02-a61d-6d56a99eedcd",
            "score": 0.6944796625089859,
            "metadata": {
              "text": "2011;342:d35.\n[68]  Hara  N,  Alkanani  AK,  Ir  D,  Robertson  CE,  Wagner  BD,\nFrank  DN,  et  al.  The  role  of  the  intestinal  microbiota  in\ntype  1  diabetes.  Clin  Immunol  2013;146:1129.\n[69]  Beyan  H,  Wen  L,  Leslie  RD.  Guts,  germs,  and  meals:  the\norigin  of  type  1  diabetes.  Curr  Diab  Rep  2012;12:45662.\n[70]  Atkinson  MA,  Chervonsky  A.  Does  the  gut  microbiota  have\na  role  in  type  1  diabetes?  Early  evidence  from  humans  and",
              "title": "2014 - Diabetes in Europe An update.pdf",
              "version": "v0",
              "chunk_order": 124,
              "document_id": "81e1fc53-6768-590f-9b47-9a5105b6ddb5",
              "extraction_id": "5c27f434-3a7c-5ec9-80fc-6399dd3570c3",
              "associatedQuery": "Can you elaborate on the role of the gut microbiome in modulating host genetic predispositions to diabetes?"
            }
          },
          {
            "id": "e7e8ef7b-bad0-54bc-814d-d947ea04756b",
            "score": 0.6925423924599664,
            "metadata": {
              "text": "diabetes. ISME J. 5,8291 (2011).\n30. Brown, C. T. et al. Gut microbiome metagenomics analysis suggests a\nfunctional model for the development of autoimmunity for type 1 diabetes.PLoS ONE 6,e25792 (2011).\n31. Endesfelder, D. et al. Compromised gut microbiota networks in children with\nanti-islet cell autoimmunity. Diabetes 63,2006 2014 (2014).\n32. Kostic, A. D. et al. The dynamics of the human infant gut microbiome in\ndevelopment and in progression toward type 1 diabetes. Cell Host Microbe 17,\n260273 (2015).",
              "title": "2016 - Integrated multi-omics of the human gut microbiome in a case study of familial type 1 diabetes.pdf",
              "version": "v0",
              "chunk_order": 161,
              "document_id": "f0405966-38bf-5a04-aa2c-1474b11362bb",
              "extraction_id": "092a9b75-9985-5876-a650-59bc3f0d10fb",
              "associatedQuery": "Can you elaborate on the role of the gut microbiome in modulating host genetic predispositions to diabetes?"
            }
          },
          {
            "id": "da881999-9d70-560f-91b3-eda465b7a639",
            "score": 0.6895030325947397,
            "metadata": {
              "text": "661678 (2007).\n4. Scott, L. J. et al. A genome-wide association study of type 2 diabetes in Finns\ndetects multiple susceptibility variants. Science 316, 13411345 (2007).\n5. Musso, G., Gambino, R. & Cassader, M. Interactions between gut microbiota and\nhost metabolism predisposing to obesity and diabetes. Annu. Rev. Med. 62,\n361380 (2011).\n6. Eckburg, P. B. et al. Diversity of the human intestinal microbial flora. Science 308,\n16351638 (2005).",
              "title": "2012 - A metagenome-wide association study of gut microbiota in type 2 diabetes.pdf",
              "version": "v0",
              "chunk_order": 91,
              "document_id": "0c088ef3-83a7-5a5e-8308-011cf4b25924",
              "extraction_id": "0a4d545f-0682-5ce1-b38c-88b5fdb4add3",
              "associatedQuery": "Can you elaborate on the role of the gut microbiome in modulating host genetic predispositions to diabetes?"
            }
          },
          {
            "id": "2589b0db-190e-5847-aef0-0bc3b415fb94",
            "score": 0.6874836683273364,
            "metadata": {
              "text": "The gut microbiota affects numerous biological functionsthroughout the body and its characterisation has becomea major research area in biomedicine. Recent studieshave suggested that gut bacteria play a fundamental rolein diseases such as obesity, diabetes and cardiovasculardisease. Data are accumulating in animal models andhumans suggesting that obesity and type 2 diabetes(T2D) are associated with a profound dysbiosis. Firsthuman metagenome-wide association studiesdemonstrated highly signi cant",
              "title": "2014 - Microbiota and diabetes an evolving relationship.pdf",
              "version": "v0",
              "chunk_order": 2,
              "document_id": "4bbbe579-1d9e-50b8-9403-b50bc3282c8f",
              "extraction_id": "44b12386-be75-5141-a5a0-77ab97136863",
              "associatedQuery": "Can you elaborate on the role of the gut microbiome in modulating host genetic predispositions to diabetes?"
            }
          },
          {
            "id": "a5d5d05b-a824-5b8f-a774-b0b9ec5d0182",
            "score": 0.6868209429490016,
            "metadata": {
              "text": "18 Burcelin R. Regulation of metabolism: a cross talk between gut microbiota and its\nhuman host. Physiology (Bethesda) 2012;27:300 7.\n19 Breen DM, Rasmussen BA, Cote CD, et al . Nutrient-sensing mechanisms in the gut\nas therapeutic targets for diabetes. Diabetes 2013;62:3005 13.\n20 Karlsson F, Tremaroli V, Nielsen J, et al . Assessing the human gut microbiota in\nmetabolic diseases. Diabetes 2013;62:3341 9.\n21 Backhed F, Ding H, Wang T, et al . The gut microbiota as an environmental factor",
              "title": "2014 - Microbiota and diabetes an evolving relationship.pdf",
              "version": "v0",
              "chunk_order": 112,
              "document_id": "4bbbe579-1d9e-50b8-9403-b50bc3282c8f",
              "extraction_id": "223f3f31-fb62-5f0d-ac8a-5a6deb1191d2",
              "associatedQuery": "Can you elaborate on the role of the gut microbiome in modulating host genetic predispositions to diabetes?"
            }
          },
          {
            "id": "63e887b3-0db0-547d-a81c-716909ead0b6",
            "score": 0.6848769971801653,
            "metadata": {
              "text": "interactions play a role in human obesity, insulin resistance and type 2 diabetes? Obes Rev 2011; 12: 27281.\n47 Kootte RS, Vrieze A, Holleman F, et al. The therapeutic potential of \nmanipulating gut microbiota in obesity and type 2 diabetes mellitus. Diabetes Obes Metab 2012; 14: 11220.\n48 Qin J, Li Y , Cai Z, et al. A metagenome-wide association study of \ngut microbiota in type 2 diabetes. Nature 2012; 490: 5560.\n49 Karlsson FH, Tremaroli V, Nookaew I, et al. Gut metagenome in",
              "title": "2014 - Pathophysiology and treatment of type 2 diabetes.pdf",
              "version": "v0",
              "chunk_order": 172,
              "document_id": "ab9288ab-e3ad-58f1-b5ba-183ee17ce4bd",
              "extraction_id": "3754ce7f-9671-5636-a4e6-849fb672366a",
              "associatedQuery": "Can you elaborate on the role of the gut microbiome in modulating host genetic predispositions to diabetes?"
            }
          },
          {
            "id": "d9bc6a49-c40e-520f-9e2d-afa05829416f",
            "score": 0.6843650717300092,
            "metadata": {
              "text": "Other factors\nInterest in the role of the gut microbiome in the devel -\nopment of T2DM has exploded in the past few years, \nand variation in the diversity and composition of the gut \nmicrobiota has been tied to T2DM100. For example, levels \nof butyrate-producing bacteria are decreased in the gut \nmicrobiota of patients with T2DM compared with that \nof healthy individuals101. In addition, evidence suggests \nthat ambient air pollution is an emerging risk factor for",
              "title": "2018 - Global aetiology and epidemiology of type 2 diabetes mellitus and its complications.pdf",
              "version": "v0",
              "chunk_order": 82,
              "document_id": "8bc8f3d4-968f-5252-ab4c-832b92e9ec0d",
              "extraction_id": "736476e2-62be-52c5-b4a2-ee7cd7666a6f",
              "associatedQuery": "Can you elaborate on the role of the gut microbiome in modulating host genetic predispositions to diabetes?"
            }
          },
          {
            "id": "b0aa9c89-a8f4-5388-97ed-5d6556c565e7",
            "score": 0.6784937573377051,
            "metadata": {
              "text": "52. Parks, B.W., et al., Genetic control of obesity and gut microbiota composition in response to high -fat, high -sucrose \ndiet in mice.  Cell Metab, 2013. 17(1): p. 141 -52. \n53. Org, E., et al., Genetic and environmental c ontrol of host -gut microbiota interactions.  Genome Res, 2015. 25(10): \np. 1558 -69. \n54. McKnite, A.M., et al., Murine gut microbiota is defined by host genetics and modulates variation of metabolic traits.  \nPLoS One, 2012. 7(6): p. e39191.",
              "title": "2019 - Systems genetics approaches to probe gene function.pdf",
              "version": "v0",
              "chunk_order": 516,
              "document_id": "1cd18d9c-0fd1-52e3-b0cf-c5e3ad0ff683",
              "extraction_id": "5ab39f63-c4e0-56b8-b6ed-26df7bee89af",
              "associatedQuery": "Can you elaborate on the role of the gut microbiome in modulating host genetic predispositions to diabetes?"
            }
          }
        ],
        "kg_search_results": null
      }
    }
  },
  "5": {
    "results": {
      "completion": {
        "id": "chatcmpl-AIHJ0Y0u83bg8S6UfGkVEYfllY6XI",
        "choices": [
          {
            "finish_reason": "stop",
            "index": 0,
            "message": {
              "content": "Machine learning algorithms show promise in integrating genomic data to predict individual risk and progression of diabetes, but there are challenges and limitations to consider.\n\n1. Genomic data is considered to yield better patient-centric outcomes than traditional tabular data for predicting diabetic illnesses [1]. This suggests that machine learning models that incorporate genomic data may provide more accurate predictions.\n\n2. Machine learning has been applied to integrate various types of data, including genomic and epigenomic biomarkers, to determine type 2 diabetic status. This approach has revealed connections between diabetic classification and other biological functions, indicating the potential of machine learning in this area [5].\n\n3. The integration of physiological, biochemical, genetic, and epigenetic features with machine learning algorithms has shown potential for more informative diagnostics and personalized treatment approaches for diabetes [8].\n\n4. However, there are limitations, such as the need for larger sample sizes and extensive training to achieve considerable accuracy when using polygenic scores-based approaches with genomic data [4].\n\nOverall, while machine learning algorithms have demonstrated potential in integrating genomic data for diabetes prediction, further research and development are needed to overcome current limitations and improve accuracy and applicability in clinical settings.",
              "role": "assistant"
            }
          }
        ],
        "created": 1728919790,
        "model": "gpt-4o-2024-08-06",
        "object": "chat.completion",
        "system_fingerprint": "fp_6b68a8204b",
        "usage": {
          "completion_tokens": 231,
          "prompt_tokens": 1226,
          "total_tokens": 1457
        }
      },
      "search_results": {
        "vector_search_results": [
          {
            "id": "a1e1110f-551a-5481-bbd6-0495d8effd38",
            "score": 0.755035593805577,
            "metadata": {
              "text": "All the mentioned models rely on tabular datasets such as PIMA and ECG signals [ 47]\nin classifying the records with possible diabetic illnesses. The current study considers that\ngenomic data yields a better patient-centric outcome than tabular data.\n2.3. Genomics for Type 2 Diabetes\nMany research studies have been carried out on genetic-based illness prediction.\nIncorporating machine learning approaches with genetic-based illness prediction could",
              "title": "2022 - Using Recurrent Neural Networks for Predicting Type-2 Diabetes from Genomic and Tabular Data.pdf",
              "version": "v0",
              "chunk_order": 60,
              "document_id": "be0e50e0-3de8-53c5-8126-a0b618647f80",
              "extraction_id": "6b4157fa-dcf0-5b70-b508-38ffb5fcda8d",
              "associatedQuery": "How effective are machine learning algorithms in integrating genomic data to predict individual risk and progression of diabetes?"
            }
          },
          {
            "id": "73df0579-3c65-590b-9293-e0dc8fa32863",
            "score": 0.723041968164328,
            "metadata": {
              "text": "-\nchondrially rich, provides a direct connection between physiological dysfunction observed in the heart and the impact of altered genomic profiles in the mitochondrion and nucleus. Machine-learning, which at current has been applied to very few genetic applications, may play a significant role in defining the epigenome of those with diabetes mellitus, likely unveiling genes and molecular pathways first impacted by the pathology.\nThe challenges ofmachine learning intheclinical setting",
              "title": "2019 - Machine-learning to stratify diabetic patients using novel cardiac biomarkers and integrative genomics.pdf",
              "version": "v0",
              "chunk_order": 146,
              "document_id": "332ac2ec-accc-5370-a4d2-6fec9ce7e072",
              "extraction_id": "aff84b9e-3855-5960-accd-dcac6b362346",
              "associatedQuery": "How effective are machine learning algorithms in integrating genomic data to predict individual risk and progression of diabetes?"
            }
          },
          {
            "id": "d0503557-1135-531d-9316-d3be3f620f3f",
            "score": 0.7097123438291357,
            "metadata": {
              "text": "15. Ali, M.M.; Paul, B.K.; Ahmed, K.; Bui, F.M.; Quinn, J.M.W.; Moni, M.A. Heart disease prediction using supervised machine\nlearning algorithms: Performance analysis and comparison. Comput. Biol. Med. 2021 ,136, 104672. [CrossRef]\n16. Bell, C.G.; Teschendorff, A.E.; Rakyan, V .K.; Maxwell, A.P .; Beck, S.; Savage, D.A. Genome-wide DNA methylation analysis for\ndiabetic nephropathy in type 1 diabetes mellitus. BMC Med. Genom. 2010 ,3, 33. [CrossRef]",
              "title": "2022 - Using Recurrent Neural Networks for Predicting Type-2 Diabetes from Genomic and Tabular Data.pdf",
              "version": "v0",
              "chunk_order": 245,
              "document_id": "be0e50e0-3de8-53c5-8126-a0b618647f80",
              "extraction_id": "a500eb31-13d8-5a0f-adfc-d260189a7555",
              "associatedQuery": "How effective are machine learning algorithms in integrating genomic data to predict individual risk and progression of diabetes?"
            }
          },
          {
            "id": "4a73b79f-f3bb-5341-865d-c6162c2f4b98",
            "score": 0.7002751409843493,
            "metadata": {
              "text": "Diagnostics 2022 ,12, 3067 6 of 30\nTable 1. Various existing models for diabetes prediction.\nApproach Type of Data Applicability Limitations\npolygenic scores-based\napproach\n[12]Genomic DataUsed in the evaluation of clinical\ntrials and illness screening\nmechanismsThe polygenic score approach needs\nlarger samples and tremendous\ntraining for considerable Accuracy.\nSingular Value\nDecomposition\n[13]Genomic Data\nTabular Data\nThe image they are\nusedThey are used in ranking the feature",
              "title": "2022 - Using Recurrent Neural Networks for Predicting Type-2 Diabetes from Genomic and Tabular Data.pdf",
              "version": "v0",
              "chunk_order": 50,
              "document_id": "be0e50e0-3de8-53c5-8126-a0b618647f80",
              "extraction_id": "a0ebb8e0-1414-52f4-aa8d-9bde3a9f26c2",
              "associatedQuery": "How effective are machine learning algorithms in integrating genomic data to predict individual risk and progression of diabetes?"
            }
          },
          {
            "id": "9fc6ebb2-02a9-5bc3-9623-6cd353ada65e",
            "score": 0.700267034753143,
            "metadata": {
              "text": "In the current study, machine-learning was used as a \npredictive tool to integrate cardiac physiological, bio\n-\nchemical, genomic, and epigenomic biomarker data in a patient-matched fashion and enable determination of type 2 diabetic status. In 50 patients, machine-learning algorithms revealed the interconnectedness between dia\n-\nbetic classification, mitochondrial function, and methyla -",
              "title": "2019 - Machine-learning to stratify diabetic patients using novel cardiac biomarkers and integrative genomics.pdf",
              "version": "v0",
              "chunk_order": 22,
              "document_id": "332ac2ec-accc-5370-a4d2-6fec9ce7e072",
              "extraction_id": "8d323598-fdf7-56cf-8290-be85929f0eaf",
              "associatedQuery": "How effective are machine learning algorithms in integrating genomic data to predict individual risk and progression of diabetes?"
            }
          },
          {
            "id": "c15a7933-675c-5790-9165-9fef8c091920",
            "score": 0.6963416699955857,
            "metadata": {
              "text": "Diabetes mellitus is a multifaceted disease, consisting \nof systemic comorbidities which necessitate a variety of treatment modalities and stratify those affected with the disease [5]. Before the implementation of machine-learning algorithms in medicine, linear statistical models have highlighted measures, such as HbA1c, as diagnos\n-\ntic staples for the evaluation of diabetes mellitus onset and progression [6]. By exploring these previously pub\n-",
              "title": "2019 - Machine-learning to stratify diabetic patients using novel cardiac biomarkers and integrative genomics.pdf",
              "version": "v0",
              "chunk_order": 14,
              "document_id": "332ac2ec-accc-5370-a4d2-6fec9ce7e072",
              "extraction_id": "8d323598-fdf7-56cf-8290-be85929f0eaf",
              "associatedQuery": "How effective are machine learning algorithms in integrating genomic data to predict individual risk and progression of diabetes?"
            }
          },
          {
            "id": "d3d65022-c072-5880-8d27-a95b285e77cd",
            "score": 0.6921416108523851,
            "metadata": {
              "text": "tool that combines both genetic and clinical featur es in order to identify diabetic \nnephropathy in patients with T2D [81].  Leung et al . compared several machine \nlearning methods that include partial least square regression, classification and \nregression tree, the C5.0 Decision Tree, Random For est, naive Bayes, neural networks \nand support vector machines [82].  The dataset used  consists of both genetic (Single \nNucleotide Polymorphisms - SNPs) and clinical data.    Age, age of diagnosis, systolic",
              "title": "2017 - Machine Learning and Data Mining Methods in Diabetes Research.pdf",
              "version": "v0",
              "chunk_order": 75,
              "document_id": "e2dcbb80-5ad7-5441-b170-9b46607445b0",
              "extraction_id": "20ba070b-900d-5213-9b38-d53492e48532",
              "associatedQuery": "How effective are machine learning algorithms in integrating genomic data to predict individual risk and progression of diabetes?"
            }
          },
          {
            "id": "d8b7be7f-3f83-5f1d-897b-da01d2a7baaf",
            "score": 0.6870492287248845,
            "metadata": {
              "text": "-\nylation status and total nuclear methylation provided the best predictive measures for assessing type 2 diabetes mellitus. The incorporation of physiological, biochemical, genetic, and epigenetic features with machine-learning algorithms exemplifies the potential for more informa\n-\ntive diagnostics in the future, as well as personalized approaches to generalized treatment modalities (Fig.6). \nDiscussion\nMachine-learning can be applied as a systems biol -",
              "title": "2019 - Machine-learning to stratify diabetic patients using novel cardiac biomarkers and integrative genomics.pdf",
              "version": "v0",
              "chunk_order": 123,
              "document_id": "332ac2ec-accc-5370-a4d2-6fec9ce7e072",
              "extraction_id": "7079e9da-e08b-5e9f-ad3d-4709915aa9e0",
              "associatedQuery": "How effective are machine learning algorithms in integrating genomic data to predict individual risk and progression of diabetes?"
            }
          },
          {
            "id": "83b5d15e-5c0e-5abd-aa88-1affe9148052",
            "score": 0.6858820710813409,
            "metadata": {
              "text": "-\ntures is likely to occur, enhancing the diagnostic potential for the individual diabetic or prediabetic patient. Indeed, this is the advantage of using machine-learning models, in that they continue to learn and develop more accurate predictions as the number of features and sampled popu\n-\nlation grows.\nConclusions\nOur work highlights the importance of identifying bio -",
              "title": "2019 - Machine-learning to stratify diabetic patients using novel cardiac biomarkers and integrative genomics.pdf",
              "version": "v0",
              "chunk_order": 156,
              "document_id": "332ac2ec-accc-5370-a4d2-6fec9ce7e072",
              "extraction_id": "493e5840-f65b-5245-8f07-126e1d9eedc3",
              "associatedQuery": "How effective are machine learning algorithms in integrating genomic data to predict individual risk and progression of diabetes?"
            }
          },
          {
            "id": "065cb845-8ff9-5919-8e1f-7d2604a52e34",
            "score": 0.681487638861679,
            "metadata": {
              "text": "10 Meigs JB, Shrader P, Sullivan LM et al. Genotype score in addition to common risk factors for prediction of Type 2 diabetes. N. Engl. J. Med. 359, 22082219 (2008).\n11 Scheuner MT, Sieverding P, Shekelle PG. Delivery of genomic medicine for common chronic adult diseases: a systematic review. JAMA  299, 13201334 (2008).\n\t Systematic\treview\tof\tearly\tresearch\tinto\tgenomic\tmedicine \t\nadoption\tin\tthe\tclinical\tcare\tof\tcommon\tchronic\tdiseases. \t\nOutlines\tboth\tphysician\tand\tpatient\tperspectives\ttowards",
              "title": "2014 - Do physicians think genomic medicine will be useful for patient care.pdf",
              "version": "v0",
              "chunk_order": 95,
              "document_id": "5418b59c-465c-5b1e-aee1-52ca7a1ead52",
              "extraction_id": "5feb39eb-3945-5a31-9d03-7b83766df1e1",
              "associatedQuery": "How effective are machine learning algorithms in integrating genomic data to predict individual risk and progression of diabetes?"
            }
          }
        ],
        "kg_search_results": null
      }
    }
  },
  "6": {
    "results": {
      "completion": {
        "id": "chatcmpl-AIHJ7kKFoNZYhA6ZvKYEgyC7wipHg",
        "choices": [
          {
            "finish_reason": "stop",
            "index": 0,
            "message": {
              "content": "Recent findings highlight the significant role of long non-coding RNAs (lncRNAs) in the regulation of insulin secretion and sensitivity, with several implications for understanding and potentially treating diabetes. \n\n1. **Regulation of Islet Function**: LncRNAs have been shown to regulate the development and function of pancreatic islets, which are crucial for insulin secretion. For instance, the lncRNA H19 is involved in this regulatory process [1]. This suggests that lncRNAs could be critical in maintaining normal insulin secretion and could be targets for therapeutic intervention in diabetes.\n\n2. **Impact on Insulin Synthesis and Secretion**: Specific lncRNAs, such as Meg3, have been found to affect insulin synthesis and secretion in pancreatic beta cells [4]. This indicates that lncRNAs play a direct role in the cellular mechanisms that control insulin production, which is essential for maintaining glucose homeostasis.\n\n3. **Therapeutic Potential**: Due to their specific functions in regulating cellular pathways, lncRNAs are considered promising therapeutic targets. Their expression patterns in tissues often correlate with the progression of diabetes, making them potential biomarkers for diagnosis and prognosis [3].\n\n4. **Association with Insulin Resistance and Diabetes**: Altered levels of lncRNAs are closely associated with the onset and progression of insulin resistance and diabetes [5]. This association underscores the potential of lncRNAs as targets for interventions aimed at improving insulin sensitivity and managing diabetes.\n\n5. **Research and Clinical Implications**: The diverse roles of lncRNAs in insulin resistance and diabetes suggest their importance in future research for diagnosis, prognosis, and therapy of the disease [2]. This calls for further investigations and collaborations among researchers, clinicians, and patients to fully understand and harness the potential of lncRNAs in diabetes management.\n\nOverall, these findings imply that lncRNAs are integral to the regulation of insulin secretion and sensitivity, offering new avenues for research and treatment strategies in diabetes.",
              "role": "assistant"
            }
          }
        ],
        "created": 1728919797,
        "model": "gpt-4o-2024-08-06",
        "object": "chat.completion",
        "system_fingerprint": "fp_a20a4ee344",
        "usage": {
          "completion_tokens": 393,
          "prompt_tokens": 1190,
          "total_tokens": 1583
        }
      },
      "search_results": {
        "vector_search_results": [
          {
            "id": "66b05301-179b-597c-bb68-e6fd0e0d1d5a",
            "score": 0.7479288578033447,
            "metadata": {
              "text": "NAs to be mapped to diabetic susceptible loci [49 52], \nall suggesting towards critical roles of lncRNAs in insulin resistance, diabetes, and its associated complications.\nLncRNAs asregulators ofislet function\nThe pancreatic islet is an important central node to researchers to understand the pathophysiology of diabe-tes [53]. The possible regulation of islet development and function by lncRNAs was first demonstrated by Ding etal., where the lncRNA, H19 (Fig. 4), was shown to be involved",
              "title": "2018 - Lnc\u2011ing non\u2011coding RNAs with metabolism and diabetes roles.pdf",
              "version": "v0",
              "chunk_order": 42,
              "document_id": "019efefb-65db-55f5-a3a7-4f224473f51f",
              "extraction_id": "96a78d74-ac6d-513e-a5a7-b22ef95ea041",
              "associatedQuery": "What are the implications of recent findings on the role of long non-coding RNAs (lncRNAs) in the regulation of insulin secretion and sensitivity?"
            }
          },
          {
            "id": "e85449e5-801e-5431-80e1-521699d18780",
            "score": 0.7348612108909375,
            "metadata": {
              "text": "this would require further investiga-tions, both invivo and invitro and critical networking among researchers, clinicians, and patients. Nevertheless, the implications of lncRNAs in diverse facets of insulin resistance and diabetes are indicative of their roles in the diagnosis, prognosis, and therapy of this disease in future.",
              "title": "2018 - Lnc\u2011ing non\u2011coding RNAs with metabolism and diabetes roles.pdf",
              "version": "v0",
              "chunk_order": 90,
              "document_id": "019efefb-65db-55f5-a3a7-4f224473f51f",
              "extraction_id": "14656f4f-b0bd-5f4f-a67a-aeb902f24757",
              "associatedQuery": "What are the implications of recent findings on the role of long non-coding RNAs (lncRNAs) in the regulation of insulin secretion and sensitivity?"
            }
          },
          {
            "id": "2d9e043b-a3fa-52dc-9a4e-71ed49f9ec1d",
            "score": 0.7105234861373901,
            "metadata": {
              "text": "To conclude, it would be apt to state that lncRNAs are widely implicated in diverse domains of cell metabolism and their altered expression is associated with diabetes and its complications. Although originally thought to be non-functional, lncRNA genes transcribe into lncRNAs that exert important and specific functions in regulating cellular pathways. Due to this specificity, lncRNAs are considered better therapeutic targets. In addition, their expression patterns in tissues quite follow the progress of",
              "title": "2018 - Lnc\u2011ing non\u2011coding RNAs with metabolism and diabetes roles.pdf",
              "version": "v0",
              "chunk_order": 88,
              "document_id": "019efefb-65db-55f5-a3a7-4f224473f51f",
              "extraction_id": "14656f4f-b0bd-5f4f-a67a-aeb902f24757",
              "associatedQuery": "What are the implications of recent findings on the role of long non-coding RNAs (lncRNAs) in the regulation of insulin secretion and sensitivity?"
            }
          },
          {
            "id": "a0146183-d255-5eae-85eb-adaf007d1b32",
            "score": 0.7006916164527447,
            "metadata": {
              "text": "58. You L, Wang N, Yin D etal (2016) Downregulation of long noncoding RNA Meg3 affects insulin synthesis and secretion in mouse pancreatic beta cells. J Cell Physiol 231:852862\n 59. Arnes L, Akerman I, Balderes DA, Ferrer J, Sussel L (2016) betalinc1 encodes a long noncoding RNA that regulates islet beta-cell formation and function. Genes Dev 30:502507\n 60. Akerman I, Tu Z, Beucher A etal (2017) Human pancreatic beta cell lncRNAs control cell-specific regulatory networks. Cell Metab 25:400411",
              "title": "2018 - Lnc\u2011ing non\u2011coding RNAs with metabolism and diabetes roles.pdf",
              "version": "v0",
              "chunk_order": 114,
              "document_id": "019efefb-65db-55f5-a3a7-4f224473f51f",
              "extraction_id": "8bbfb009-87b7-54ae-8465-8796db8c271a",
              "associatedQuery": "What are the implications of recent findings on the role of long non-coding RNAs (lncRNAs) in the regulation of insulin secretion and sensitivity?"
            }
          },
          {
            "id": "b3c5f734-aa0d-5da9-bdb9-e330e6c02e00",
            "score": 0.6984752236887959,
            "metadata": {
              "text": "of lncRNAs in the development and function of metabolic tissues, and therefore, their altered levels are closely asso-ciated with the onset and progression of insulin resistance and diabetes.\nRoles oflncRNAs indiabetic complications\nApart from being involved in major metabolic tissues dur -",
              "title": "2018 - Lnc\u2011ing non\u2011coding RNAs with metabolism and diabetes roles.pdf",
              "version": "v0",
              "chunk_order": 66,
              "document_id": "019efefb-65db-55f5-a3a7-4f224473f51f",
              "extraction_id": "d7b2d258-d566-5552-8308-4ac35953884d",
              "associatedQuery": "What are the implications of recent findings on the role of long non-coding RNAs (lncRNAs) in the regulation of insulin secretion and sensitivity?"
            }
          },
          {
            "id": "b774bf7b-4546-56d2-ae7b-7bc2c9f2fb08",
            "score": 0.6904380527269263,
            "metadata": {
              "text": "tion among researchers ( Knoll et al., 2015 ). As an important post-transcriptional pathogenesis of diabetes,\nlncRNAs and their associated orchestrated networks are implicated in mediating complex pathological\nmechanisms of diabetes ( Kato et al., 2016; Liu et al., 2014 ). To delineate the inuence of lncRNAs and\n172 iScience 19, 162176, September 27, 2019",
              "title": "2019 - Development and Genome Sequencing.pdf",
              "version": "v0",
              "chunk_order": 105,
              "document_id": "18820c9e-f7ae-57ae-897d-0d9c3f616b6a",
              "extraction_id": "d971dced-935c-566b-a4a2-11bcf99b9c84",
              "associatedQuery": "What are the implications of recent findings on the role of long non-coding RNAs (lncRNAs) in the regulation of insulin secretion and sensitivity?"
            }
          },
          {
            "id": "c8d55dea-0656-527e-93bd-9624cec8f3c9",
            "score": 0.6839436481032594,
            "metadata": {
              "text": "in transgenerational transmission of gestational diabetes mellitus which leads to impaired islet structure and func-tion [ 54]. To understand the roles of lncRNAs in regu-\nlating pancreatic function, several research groups have profiled lncRNA expression in mouse and human pancre-atic islets [55, 56]. Transcriptome analysis in pancreatic \n-cells of type 2 diabetes patients identified tissue-specific and dynamically regulated abnormally expressed lncR -",
              "title": "2018 - Lnc\u2011ing non\u2011coding RNAs with metabolism and diabetes roles.pdf",
              "version": "v0",
              "chunk_order": 43,
              "document_id": "019efefb-65db-55f5-a3a7-4f224473f51f",
              "extraction_id": "96a78d74-ac6d-513e-a5a7-b22ef95ea041",
              "associatedQuery": "What are the implications of recent findings on the role of long non-coding RNAs (lncRNAs) in the regulation of insulin secretion and sensitivity?"
            }
          },
          {
            "id": "e5669569-f9ba-5797-b468-3a1980addc0a",
            "score": 0.6827709674835205,
            "metadata": {
              "text": "1831 Lnc-ing non- coding RNAs withmetabolism anddiabetes: roles oflncRNAs  \n1 3\nendocrine hormones, insulin and glucagon, where insulin \nis the anabolic master regulator which controls periph -",
              "title": "2018 - Lnc\u2011ing non\u2011coding RNAs with metabolism and diabetes roles.pdf",
              "version": "v0",
              "chunk_order": 39,
              "document_id": "019efefb-65db-55f5-a3a7-4f224473f51f",
              "extraction_id": "96a78d74-ac6d-513e-a5a7-b22ef95ea041",
              "associatedQuery": "What are the implications of recent findings on the role of long non-coding RNAs (lncRNAs) in the regulation of insulin secretion and sensitivity?"
            }
          },
          {
            "id": "9ca17d26-cc06-5afe-a7dd-3f80b1b99da0",
            "score": 0.6780256628990173,
            "metadata": {
              "text": "Vol.:(0123456789)1 3Cellular and Molecular Life Sciences (2018) 75:18271837 \nhttps://doi.org/10.1007/s00018-018-2760-9\nREVIEW\nLncing noncoding RNAs withmetabolism anddiabetes: roles \noflncRNAs\nNehaGoyal1,2 DeveshKesharwani1,2 MalabikaDatta1,2 \nReceived: 18 September 2017 / Revised: 29 December 2017 / Accepted: 24 January 2018 / Published online: 31 January 2018 \n Springer International Publishing AG, part of Springer Nature 2018\nAbstract",
              "title": "2018 - Lnc\u2011ing non\u2011coding RNAs with metabolism and diabetes roles.pdf",
              "version": "v0",
              "chunk_order": 0,
              "document_id": "019efefb-65db-55f5-a3a7-4f224473f51f",
              "extraction_id": "efc73cf6-99c6-5272-9bb0-7bd6a34633f0",
              "associatedQuery": "What are the implications of recent findings on the role of long non-coding RNAs (lncRNAs) in the regulation of insulin secretion and sensitivity?"
            }
          },
          {
            "id": "45d35985-9183-55f0-8b51-41df27cd7677",
            "score": 0.6768291198262313,
            "metadata": {
              "text": "(2013). A novel mechanism regulating insulin secretion involving Herpud1 inmice. Diabetologia 56, 15691576 .\nZhao, X.Y., and Lin, J.D. (2015). Long noncoding RNAs: a new regulatory code\nin metabolic control. Trends Biochem. Sci. 40, 586596 .\n1806 Cell Reports 17, 17951806, November 8, 2016",
              "title": "2016 - A genetic screen identifies hypothalamic Fgf15 as a regulator of glucagon secretion.pdf",
              "version": "v0",
              "chunk_order": 137,
              "document_id": "288adb9b-a547-5e61-8593-1b2ab36271d3",
              "extraction_id": "a847f5f4-0c56-5678-9e1e-93b9b5b294f2",
              "associatedQuery": "What are the implications of recent findings on the role of long non-coding RNAs (lncRNAs) in the regulation of insulin secretion and sensitivity?"
            }
          }
        ],
        "kg_search_results": null
      }
    }
  },
  "7": {
    "results": {
      "completion": {
        "id": "chatcmpl-AIHJKDULZO7wnGlvKQDCFnpw8Cumo",
        "choices": [
          {
            "finish_reason": "stop",
            "index": 0,
            "message": {
              "content": "Post-translational modifications (PTMs) of proteins play a significant role in regulating key signaling pathways involved in glucose homeostasis. Here are some ways PTMs affect these pathways:\n\n1. **Protein Kinases and Glucose Homeostasis**: The PI3K/AKT, MAPK, and AMPK signaling pathways are crucial for glucose homeostasis, and these pathways are regulated by protein kinases, which can be modulated by PTMs such as phosphorylation [1].\n\n2. **Histone Modifications**: Histone post-translational modifications are involved in the regulation of genes associated with diabetes pathogenesis, including those related to insulin and islet-specific transcription factors. These modifications can influence gene expression and thereby affect glucose metabolism [3].\n\n3. **N-glycosylation**: The glycosylation of glucose transporter 2 is an example of a PTM that promotes insulin secretion, which is vital for maintaining glucose levels and suppressing diabetes [6].\n\n4. **Sirtuins and Deacetylation**: The sirtuin family, particularly SIRT1, is involved in regulating factors related to metabolism and insulin secretion. Sirtuins are deacetylases, and their activity represents a form of PTM that can influence glucose homeostasis [10].\n\nThese examples illustrate how PTMs can modulate signaling pathways and protein functions, ultimately impacting glucose homeostasis and related metabolic processes.",
              "role": "assistant"
            }
          }
        ],
        "created": 1728919810,
        "model": "gpt-4o-2024-08-06",
        "object": "chat.completion",
        "system_fingerprint": "fp_6b68a8204b",
        "usage": {
          "completion_tokens": 286,
          "prompt_tokens": 1455,
          "total_tokens": 1741
        }
      },
      "search_results": {
        "vector_search_results": [
          {
            "id": "64482aec-5688-5431-adda-b8f1de92a183",
            "score": 0.7271759510040283,
            "metadata": {
              "text": "regulates glucose-induced biological responses in pancreatic \nbeta-cells. Diabetes. 2008;57:2708-17.\n29. Schultze SM, Hemmings BA, Niessen M, Tschopp O. \nPI3K/AKT, MAPK and AMPK signalling: protein kinases \nin glucose homeostasis. Expert Rev Mol Med. 2012;14:e1.\n30. White MF. IRS proteins and the common path to diabetes. \nAm J Physiol Endocrinol Metab. 2002;283:E413-22.\n31. Erener S, Marwaha A, Tan R, Panagiotopoulos C, Kieffer \nTJ. Profiling of circulating microRNAs in children with",
              "title": "2018 - MicroRNA profiling and their pathways in South African.pdf",
              "version": "v0",
              "chunk_order": 97,
              "document_id": "b6bb090d-7176-59db-af04-582aa1d5cf10",
              "extraction_id": "14577d73-d320-54dd-93f2-c55f986bc8bc",
              "associatedQuery": "How do post-translational modifications of proteins affect key signaling pathways involved in glucose homeostasis?"
            }
          },
          {
            "id": "b604dabf-3dc2-5d01-9cc4-6e9f916c464a",
            "score": 0.7032889723777771,
            "metadata": {
              "text": "pathological processes involved in glucose metabolism \nby post transcriptional regulation of gene expression. \nParticular microRNAs can regulate cell function271, \nexposing key regulatory signalling pathways involved in \nrestoration of cell mass, and provide a promising strat \negy for improving insulin secretion and cell health in \nT2DM. Identification of novel insulin secretagogues \nthat act directly on cells and enteroendocrine Kcells \nand Lcells in the intestine are under investigation, and",
              "title": "2015 - Type 2 diabetes mellitus.pdf",
              "version": "v0",
              "chunk_order": 163,
              "document_id": "415516ba-5365-501b-84ce-0789045862f8",
              "extraction_id": "617b6fab-a5e5-59b7-a593-a0477e6bf9fe",
              "associatedQuery": "How do post-translational modifications of proteins affect key signaling pathways involved in glucose homeostasis?"
            }
          },
          {
            "id": "e1f984ac-aa42-5eb4-92cb-303886f6f1db",
            "score": 0.7016167640686035,
            "metadata": {
              "text": "can result in diabetes and its complications including DN.\nSeveral studies show that key histone post-  translational \nmodifications are involved in the regulation of genes \nassociated with the pathogenesis of diabetes, such as \ninsulin and islet-specific transcription factors.48,60 Inaddi -\ntion, several groups are examining the role of histone \npost-translational modifications in adipocytes related to \ntype2 diabetes, obesity and the metabolic syndrome.48,60",
              "title": "2014 - Diabetic nephropathy\u2014emerging epigenetic mechanisms.pdf",
              "version": "v0",
              "chunk_order": 53,
              "document_id": "be05127e-1be8-5573-b571-51a11c3b2be2",
              "extraction_id": "bf537fe8-5508-5355-a656-b4053febe0e5",
              "associatedQuery": "How do post-translational modifications of proteins affect key signaling pathways involved in glucose homeostasis?"
            }
          },
          {
            "id": "7b6e89ec-b690-5ff1-b24d-3ed6744f3486",
            "score": 0.6925344054380426,
            "metadata": {
              "text": "cascade of protein kinases and regulatory proteins of which IRS-1 and IRS-2 \nare most important. This causes suppression of glucose release from liver \nand kidney/ translocation of glucose transporters in muscle and adipose \ntissue to increase their glucose uptake, and inhibition of release of FF A into \nthe circulation due to suppression of the activity of hormone-sensitive lipase \nand a simultaneous increase in their clearance from the circulation. Although",
              "title": "2004 - Diabetes Genes a.pdf",
              "version": "v0",
              "chunk_order": 178,
              "document_id": "805ad0d5-6a3d-5e5f-b1ba-121e10c6daaa",
              "extraction_id": "06912a59-fdd6-5731-af8f-6c98ff1ace5c",
              "associatedQuery": "How do post-translational modifications of proteins affect key signaling pathways involved in glucose homeostasis?"
            }
          },
          {
            "id": "9a8edd2d-c06a-559e-8397-beaaa84705b7",
            "score": 0.6861554384231567,
            "metadata": {
              "text": "Magnan C, Postic C, Prip-Buus C, Vasseur-Cognet M (2008) The\ntranscription factor COUP-TFII is negatively regulated by insulin and\nglucose via Foxo1- and ChREBP-controlled pathways. Mol Cell Biol 28:\n65686579Rodgers JT, Lerin C, Haas W, Gygi SP, Spiegelman BM, Puigserver P (2005)\nNutrient control of glucose homeostasis through a complex ofPGC-1alpha and SIRT1. Nature 434: 113118\nSchwer B, Verdin E (2008) Conserved metabolic regulatory functions of\nsirtuins. Cell Metab 7:104112",
              "title": "2011 - CREB and ChREBP oppositely regulate SIRT1 expression in response to energy availability.pdf",
              "version": "v0",
              "chunk_order": 90,
              "document_id": "32538f01-9eaf-5f9b-8615-ec47cc4ca8e2",
              "extraction_id": "4217906f-87c5-54b0-95a5-7c26dc08afce",
              "associatedQuery": "How do post-translational modifications of proteins affect key signaling pathways involved in glucose homeostasis?"
            }
          },
          {
            "id": "7d522337-e875-55eb-9b67-4718e5db8ffd",
            "score": 0.6820673942565918,
            "metadata": {
              "text": "of glucose transporter 2 glycosylation promotes insulin secretion in suppressing diabetes. Cell 123:1307 1321. PMID: 16377570\n47. Whitaker GM, Lynn FC, McIntosh CH, Accili EA (2012) Regulation of GIP and GLP1 receptor cell sur-\nface expression by N-glycosylation and receptor heteromerization. PLoS One 7: e32675. doi: 10.1371/\njournal.pone.0032675 PMID: 22412906\n48. Johswich A, Longuet C, Pawling J, Abdel Rahman A, Ryczko M, et al. (2014) N-glycan remodeling on",
              "title": "2015 - Transcript Expression Data from Human.pdf",
              "version": "v0",
              "chunk_order": 175,
              "document_id": "2b30d4f3-9ec3-574f-9a36-709b0e09c3f2",
              "extraction_id": "867d0b1b-16a1-53ea-b014-3c204b9001a5",
              "associatedQuery": "How do post-translational modifications of proteins affect key signaling pathways involved in glucose homeostasis?"
            }
          },
          {
            "id": "1edee360-5de0-51c9-bf8d-7c2e2f23a682",
            "score": 0.6818087697029114,
            "metadata": {
              "text": "strate 1), Pde3b (phosphodiesterase 3B), Hk2 (hexokinase 2), Foxo1\n(forkhead box O1), Socs6 (suppressor of cytokine signaling 6), and Ogt\n(O-linked N-acetylglucosamine (GlcNAc) transferase). Impaired insulinsignaling is well known to negatively in uence glucose and lipid\nmetabolism [62]. In adipose tissue, insulin stimulates glucose uptake\nby inducing translocation of GLUT4 to the cell surface, it increasesglycolysis rate by stimulating hexokinases ( Hk2) and suppresses\nlipolysis ( Acaca and Prkaa1 )[63].",
              "title": "2018 - A computational biology approach of a genome-wide screen.pdf",
              "version": "v0",
              "chunk_order": 98,
              "document_id": "ae727c80-a0a3-52f4-9e2f-b93a539558ad",
              "extraction_id": "ab4f6ea7-767f-5783-9e1a-8570eaabe96c",
              "associatedQuery": "How do post-translational modifications of proteins affect key signaling pathways involved in glucose homeostasis?"
            }
          },
          {
            "id": "43a104b3-f34b-5f52-86ff-fd7d45827f32",
            "score": 0.6812280416488647,
            "metadata": {
              "text": "signalling pathways by reducing insulin induced tyro \nsine phosphorylation of IRS1 and IRS2 (REF. 161) and by \nincreasing degradation of IRS1 (REF. 162). Recent studies \nhave demonstrated that the p85 regulatory subunit of \nPI3K interacts with XBP1s (the spliced, transcription \nally active isoform of XBP1) and promotes the trans \nlocation of XBP1s into the nucleus to initiate the ER \nstress response163.Diabetic complications\nDiabetic microvascular complications are closely related",
              "title": "2015 - Type 2 diabetes mellitus.pdf",
              "version": "v0",
              "chunk_order": 88,
              "document_id": "415516ba-5365-501b-84ce-0789045862f8",
              "extraction_id": "e4e89eba-6032-5781-83f4-8d47ab5b3825",
              "associatedQuery": "How do post-translational modifications of proteins affect key signaling pathways involved in glucose homeostasis?"
            }
          },
          {
            "id": "3e08ef82-888b-58a0-9a80-3547ab4bd516",
            "score": 0.6800271677987092,
            "metadata": {
              "text": "activated protein kinase. J Biol Chem. 2007;282:9777 -88. \n[44] Chakrabarti S, Davidge ST. High glucose -induced oxidative stress alters estrogen effects on ERalpha and \nERbeta in human endothelial cells: reversal by AMPK activator. J Steroid Biochem Mol Biol. 2009;117:99 -106. \n[45] Mortuza R, Chen S, Feng B, Sen S, Chakrabarti S. High glucose induced alteration of SIRTs in endothelial \ncells causes ra pid aging in a p300 and FOXO regulated pathway. PLoS One. 2013;8:e54514.",
              "title": "2015 - Cellular and Molecular Biology of Aging Endothelial Cells.pdf",
              "version": "v0",
              "chunk_order": 153,
              "document_id": "815d7f3e-e219-502f-aba0-57a68ae787d3",
              "extraction_id": "283e34bb-6e2b-5aa9-85c5-2584b669f122",
              "associatedQuery": "How do post-translational modifications of proteins affect key signaling pathways involved in glucose homeostasis?"
            }
          },
          {
            "id": "cf4f3239-dd62-5eef-b5fc-85f4780e3f48",
            "score": 0.6797555685043335,
            "metadata": {
              "text": "Epigenetic Mechanisms in Diabetic Complications     17 \nInterestingly, the sirtuin (SIRT) family of deacetylases, specifically SIRT1, has been found to 360 \nregulate several factors involved in metabolism, adipogenesis a nd insulin secretion (86). HATs 361 \nand HDACs can also modulate NF- B transcriptional activity (4, 44) resulting in changes in 362",
              "title": "2010 - The Role of Epigenetics in the Pathology of Diabetic Complications.pdf",
              "version": "v0",
              "chunk_order": 76,
              "document_id": "766edfd5-4756-51bf-b636-c94b041d030c",
              "extraction_id": "41ac576d-b850-5ee8-9753-ba9b060ba798",
              "associatedQuery": "How do post-translational modifications of proteins affect key signaling pathways involved in glucose homeostasis?"
            }
          }
        ],
        "kg_search_results": null
      }
    }
  },
  "8": {
    "results": {
      "completion": {
        "id": "chatcmpl-AIHJRHE43F2P7lugdT3xuuPfMvKGY",
        "choices": [
          {
            "finish_reason": "stop",
            "index": 0,
            "message": {
              "content": "Studying the genetic basis of syndromic forms of diabetes, such as Wolfram Syndrome, has provided several insights:\n\n1. **Genotype-Phenotype Correlation**: Research has highlighted the importance of understanding genotype-phenotype correlations in Wolfram Syndrome. This involves identifying specific genetic mutations and understanding how they manifest in clinical symptoms, which can aid in more accurate diagnosis and management of the syndrome [1], [6].\n\n2. **Genetic Mutations**: Wolfram Syndrome is primarily caused by mutations in the WFS1 gene, which encodes the wolframin ER transmembrane glycoprotein. These mutations lead to a range of symptoms including diabetes mellitus, optic atrophy, and deafness. Some severe cases are associated with dominant heterozygous variants [4].\n\n3. **Early Onset and Symptoms**: The syndrome is characterized by early onset of diabetes mellitus, typically around the age of 6, followed by other symptoms such as optic atrophy and sensorineural deafness in later years [5].\n\n4. **Potential for Broader Implications**: Insights from studying Wolfram Syndrome have implications for basic science and clinical practice. They emphasize the need for accurate clinical descriptions and early recognition of symptoms, which can improve patient outcomes and inform treatment strategies [6].\n\n5. **Risk Assessment**: There is ongoing research to determine if heterozygotes for Wolfram Syndrome are at risk for maturity-onset diabetes, which could have implications for understanding genetic risk factors in broader populations [2].\n\nThese insights not only enhance our understanding of Wolfram Syndrome but also contribute to the broader field of genetic research in diabetes, potentially informing personalized medicine approaches for more common forms of the disease.",
              "role": "assistant"
            }
          }
        ],
        "created": 1728919817,
        "model": "gpt-4o-2024-08-06",
        "object": "chat.completion",
        "system_fingerprint": "fp_a20a4ee344",
        "usage": {
          "completion_tokens": 346,
          "prompt_tokens": 1377,
          "total_tokens": 1723
        }
      },
      "search_results": {
        "vector_search_results": [
          {
            "id": "d0465c0a-e45a-5fff-a9b3-40ad191963a3",
            "score": 0.7042259187765696,
            "metadata": {
              "text": "WFS1 and genotype-phenotype correlation in Wolfram syndrome. Am J\nMed Genet A. 2007;143A(14):1605 12.\n61. McCarthy MI. Painting a new picture of personalised medicine for diabetes.\nDiabetologia. 2017;60(5):793 9.\n62. Fuchsberger C, Flannick J, Teslovich TM, et al. The genetic architecture of\ntype 2 diabetes. Nature. 2016;536(7614):41 7.\n63. Patch AM, Flanagan SE, Boustred C, Hattersley AT, Ellard S. Mutations in the\nABCC8 gene encoding the SUR1 subunit of the KATP channel cause",
              "title": "2017 - Spectrum of mutations in monogenic diabetes genes identified from high-throughput DNA sequencing of 6888 individuals.pdf",
              "version": "v0",
              "chunk_order": 146,
              "document_id": "18a8a000-69ed-5d34-b13f-f5ae016d1067",
              "extraction_id": "0f16b510-caa9-521f-8d87-e225f52de9f5",
              "associatedQuery": "What insights have been gained from studying the genetic basis of syndromic forms of diabetes, such as Wolfram Syndrome and Alstr\u00f6m Syndrome?"
            }
          },
          {
            "id": "f4b3a7bd-4a5c-5c95-aa11-936340eecb8b",
            "score": 0.6707017225445343,
            "metadata": {
              "text": "enable physicians to ameliorate some of the complications\nthat so devastate the lives of these patients.\nThree questions need answers from further studies: is\nthere really a lack of diabetic complications in Wolfram\nsyndrome patients compared with other diabetics? What\nis the nature of the neurodegeneration and its relation to\ndiabetes mellitus? Are heterozygotes for Wolfram\nsyndrome at risk of maturity-onset diabetes?\nThis paper is dedicated to the memory of Robin Smith, a Wolfram",
              "title": "1995 - Neurodegeneration and diabetes UK nationwide study of Wolfram syndrome.pdf",
              "version": "v0",
              "chunk_order": 54,
              "document_id": "1fa2280b-868e-571f-830e-bebdd874fc9d",
              "extraction_id": "744f3821-fc61-58d1-8107-17d5674fe1d8",
              "associatedQuery": "What insights have been gained from studying the genetic basis of syndromic forms of diabetes, such as Wolfram Syndrome and Alstr\u00f6m Syndrome?"
            }
          },
          {
            "id": "e4240b4f-b19f-5f8c-b9ce-e0b60cf4aceb",
            "score": 0.6695254048563664,
            "metadata": {
              "text": "Monogenic and syndromic forms account for only a small,though highly informative, proportion of cases of nonau-toimmune diabetes. The challenge for medical science liesin bringing equivalent mechanistic insights and transla-tional benets to the hundreds of millions of peoplealready affected by, or at risk of, more common, typicalforms of diabetes. For type 2 diabetes, there is abundantevidence that individual susceptibility is inuenced byboth the combination of genetic variation at multiple sitesand a",
              "title": "2008 - Learning From Molecular Genetics.pdf",
              "version": "v0",
              "chunk_order": 48,
              "document_id": "3aa0aea1-fe53-519b-b367-79fa1c79aa01",
              "extraction_id": "979b0578-b02a-526e-b3b4-aa7fec3eeb91",
              "associatedQuery": "What insights have been gained from studying the genetic basis of syndromic forms of diabetes, such as Wolfram Syndrome and Alstr\u00f6m Syndrome?"
            }
          },
          {
            "id": "3efa9423-0651-5096-8ccd-50a0f906e30e",
            "score": 0.6586581664877587,
            "metadata": {
              "text": "responding to two causative genes have been identified to date. \nWolfram syndrome 1 (WS1), characterized by diabetes insipidus, \nDM, optic atrophy, and deafness, is a rare autosomal recessive \ndisease caused by variants in wolframin ER transmembrane gly-\ncoprotein (WFS1). Severe cases with dominant heterozygous vari-\nants are also reported (92). Often, patients first manifestation \nis DM at an average age of 6 years. Though most WS1 patients",
              "title": "2021 -  Monogenic diabetes a gateway to precision medicine.pdf",
              "version": "v0",
              "chunk_order": 68,
              "document_id": "0b6ff786-6a7b-5d24-ba5e-7a61fee7757f",
              "extraction_id": "f01be500-1e96-57ca-b164-1b97017ec44d",
              "associatedQuery": "What insights have been gained from studying the genetic basis of syndromic forms of diabetes, such as Wolfram Syndrome and Alstr\u00f6m Syndrome?"
            }
          },
          {
            "id": "1ae4ea48-5dbd-5502-aa74-a1ef1527a493",
            "score": 0.6534469517139764,
            "metadata": {
              "text": "finding study to describe the natural history, complications,\nprevalence, and inheritance of the syndrome.\nWe identified 45 patients with Wolfram syndrome&mdash;a\nprevalence of one per 770000. Non-autoimmune, insulin-\ndeficient diabetes mellitus presented at a median age of 6\nyears, followed by optic atrophy (11 years). Cranial\ndiabetes insipidus occurred in 33 patients (73%) with\nsensorineural deafness (28, 62%) in the second decade;\nrenal-tract abnormalities (26, 58%) presented in the third",
              "title": "1995 - Neurodegeneration and diabetes UK nationwide study of Wolfram syndrome.pdf",
              "version": "v0",
              "chunk_order": 1,
              "document_id": "1fa2280b-868e-571f-830e-bebdd874fc9d",
              "extraction_id": "782db3c2-ab52-59df-b798-c5b2492c8946",
              "associatedQuery": "What insights have been gained from studying the genetic basis of syndromic forms of diabetes, such as Wolfram Syndrome and Alstr\u00f6m Syndrome?"
            }
          },
          {
            "id": "e0234ab8-850f-585c-9de2-edaa3ac615fd",
            "score": 0.6526280836054115,
            "metadata": {
              "text": "Wolfram patients have a mitochondrial genome\nabnormality, but this has not yet been shown. The\ndifferential diagnosis indicates the importance of accurate\nclinical descriptions when presenting cases of the\nsyndrome.\nOur study has implications for basic science and\npractice: more accurate characterisation of the syndrome\nwill allow assessment of genotype/phenotype correlations;\nand earlier recognition of diabetes insipidus,\ngastrointestinal dysfunction, and central apnoeas should",
              "title": "1995 - Neurodegeneration and diabetes UK nationwide study of Wolfram syndrome.pdf",
              "version": "v0",
              "chunk_order": 53,
              "document_id": "1fa2280b-868e-571f-830e-bebdd874fc9d",
              "extraction_id": "744f3821-fc61-58d1-8107-17d5674fe1d8",
              "associatedQuery": "What insights have been gained from studying the genetic basis of syndromic forms of diabetes, such as Wolfram Syndrome and Alstr\u00f6m Syndrome?"
            }
          },
          {
            "id": "85de5eb6-8034-5ebb-8526-1de0a173b2ae",
            "score": 0.6522817806089458,
            "metadata": {
              "text": "onset diabetes of the young, multiple causes of neonatal DM, and syndromic diabetes such as Wolfram syndrome and \nlipodystrophy. We also review methods of prioritizing patients undergoing genetic testing, and highlight existing challenges \nfacing sequence data interpretation that can be addressed by forming collaborations of expertise and by pooling cases.Monogenic diabetes: a gateway to precision medicine \nin diabetes\nHaichen Zhang,1 Kevin Colclough,2 Anna L. Gloyn,3,4 and Toni I. Pollin1",
              "title": "2021 -  Monogenic diabetes a gateway to precision medicine.pdf",
              "version": "v0",
              "chunk_order": 14,
              "document_id": "0b6ff786-6a7b-5d24-ba5e-7a61fee7757f",
              "extraction_id": "5a39ee4b-ba00-56d6-ba6c-0edeac3b4f2e",
              "associatedQuery": "What insights have been gained from studying the genetic basis of syndromic forms of diabetes, such as Wolfram Syndrome and Alstr\u00f6m Syndrome?"
            }
          },
          {
            "id": "0b2437f9-1aff-5e6a-bca6-cc17d96c5d56",
            "score": 0.6496991917580633,
            "metadata": {
              "text": "WFS1 mutations underlie a genetic syndrome \nof neonatal/infancy-onset diabetes, congenital \nsensorineural deafness, and congenital cataracts. \nDiabetes . 2017;66(7):20442053.\n 93. Rigoli L, Di Bella C. Wolfram syndrome 1 \nand Wolfram syndrome 2. Curr Opin Pediatr. \n2012;24(4):512517 .\n 94. Bansal V, et al. Identification of a missense vari-\nant in the WFS1 gene that causes a mild form of \nWolfram syndrome and is associated with risk for \ntype 2 diabetes in Ashkenazi Jewish individuals.",
              "title": "2021 -  Monogenic diabetes a gateway to precision medicine.pdf",
              "version": "v0",
              "chunk_order": 156,
              "document_id": "0b6ff786-6a7b-5d24-ba5e-7a61fee7757f",
              "extraction_id": "31b3b484-d769-5b88-9633-45ca3508be8f",
              "associatedQuery": "What insights have been gained from studying the genetic basis of syndromic forms of diabetes, such as Wolfram Syndrome and Alstr\u00f6m Syndrome?"
            }
          },
          {
            "id": "8fbdd64f-d761-5e8d-989c-7e4bf376b1ec",
            "score": 0.6475472257272261,
            "metadata": {
              "text": "established. It has been corroborated by a series of obser-vations that include ethnic differences, familial aggrega-tion, twin studies, admixture studies, linkage studies, monogenic cases (e.g., MODY), mitochondrial cases of diabetes, and a constantly growing number of molecular markers  \n[5] . On the other hand, the genetics of the meta-\nbolic syndrome remains complex  [6] . It is highly unlikely \nthat  a  single gene will account for a substantial portion",
              "title": "2010 - Family History of Diabetes and Prevalence.pdf",
              "version": "v0",
              "chunk_order": 11,
              "document_id": "f16c4c6e-bb5f-5d4a-9945-8af4d0df19f4",
              "extraction_id": "59b6bf24-b9f7-53bc-b809-5ce97d5e9898",
              "associatedQuery": "What insights have been gained from studying the genetic basis of syndromic forms of diabetes, such as Wolfram Syndrome and Alstr\u00f6m Syndrome?"
            }
          },
          {
            "id": "bd8d61dd-e027-5a43-8b00-1e7e9e748ea3",
            "score": 0.6464164449696677,
            "metadata": {
              "text": "diabetes (0.5% carrier frequency) compared to controls (0.035%). One individual with early onset diabetes was\nhomozygous for a rare pathogenic missense variant in the WFS1 gene but did not have the additional phenotypes\nassociated with Wolfram syndrome.\nConclusion: Targeted sequencing of genes linked with monogenic diabetes can identify disease-relevant mutations in\nindividuals diagnosed with type 2 diabetes not suspected of having monogenic forms of the disease. Our data suggests",
              "title": "2017 - Spectrum of mutations in monogenic diabetes genes identified from high-throughput DNA sequencing of 6888 individuals.pdf",
              "version": "v0",
              "chunk_order": 4,
              "document_id": "18a8a000-69ed-5d34-b13f-f5ae016d1067",
              "extraction_id": "36a362bd-1d4a-5e47-a00f-5ae6d3a0626f",
              "associatedQuery": "What insights have been gained from studying the genetic basis of syndromic forms of diabetes, such as Wolfram Syndrome and Alstr\u00f6m Syndrome?"
            }
          }
        ],
        "kg_search_results": null
      }
    }
  },
  "9": {
    "results": {
      "completion": {
        "id": "chatcmpl-AIHJckmqoH46qp1BDk6zYdLg1Akw5",
        "choices": [
          {
            "finish_reason": "stop",
            "index": 0,
            "message": {
              "content": "The study of monozygotic twins discordant for diabetes provides valuable insights into the etiology of the disease by highlighting the roles of both genetic and epigenetic factors. Since monozygotic twins share identical genomes, any differences in disease manifestation between them can often be attributed to non-genetic factors, such as epigenetic modifications or environmental influences.\n\n1. **Genetic Component**: The high concordance rates for diabetes in monozygotic twins, compared to dizygotic twins, underscore a significant genetic component to the disease [1], [6]. This suggests that genetic predispositions, particularly those affecting insulin sensitivity and secretion, play a crucial role in the development of diabetes [1].\n\n2. **Epigenetic Influences**: Despite identical genetic makeup, monozygotic twins can exhibit differences in disease susceptibility due to epigenetic variations. These variations can arise from environmental factors and gene-environment interactions, which are crucial in understanding the etiology of diabetes [4]. Epigenetic differences, such as DNA methylation patterns, have been observed in monozygotic twins and may contribute to discordance in disease states [9].\n\n3. **Environmental and Lifestyle Factors**: The discordance in diabetes among monozygotic twins also points to the influence of environmental factors and lifestyle choices, such as diet and physical activity, which can modify epigenetic marks and affect disease outcomes [6].\n\nIn summary, the study of monozygotic twins discordant for diabetes highlights that while genetic predispositions are significant, epigenetic modifications and environmental factors also play critical roles in the disease's etiology. This understanding can help in developing more targeted prevention and treatment strategies that consider both genetic and non-genetic factors.",
              "role": "assistant"
            }
          }
        ],
        "created": 1728919828,
        "model": "gpt-4o-2024-08-06",
        "object": "chat.completion",
        "system_fingerprint": "fp_6b68a8204b",
        "usage": {
          "completion_tokens": 351,
          "prompt_tokens": 1453,
          "total_tokens": 1804
        }
      },
      "search_results": {
        "vector_search_results": [
          {
            "id": "6623716f-8275-5f17-be27-a8241560a645",
            "score": 0.7489541472251293,
            "metadata": {
              "text": "Studies of twins also provide compelling evidence for a genetic component to \nT2D. Estimates for concordance rates range from 0.29 to 1.00 in monozygotic (MZ) twins, while in dizygotic (DZ) twins the range is 0.100.43 [57, 58, 6164]. \nThe high levels of heritability observed for insulin sensitivity and insulin secretion [6567] further reinforce the role of genetics in diabetes and indicate the primary genetic lesions for diabetes are likely to localize to genes in beta-cell-centric pathways.",
              "title": "2011 - Interaction Between Exercise and Genetics.pdf",
              "version": "v0",
              "chunk_order": 65,
              "document_id": "c36db75e-4b76-540d-9efb-d0e156e61541",
              "extraction_id": "861346c7-0fcf-5cae-ace6-a012a370d297",
              "associatedQuery": "How do genetic and epigenetic differences between monozygotic twins discordant for diabetes inform our understanding of its etiology?"
            }
          },
          {
            "id": "15e73a17-cae0-5dc1-8cad-5805809ae872",
            "score": 0.7349158245425486,
            "metadata": {
              "text": "It is therefore intriguing that A1C levels are signicantly\ncorrelated in monozygotic twins whether they are concor-\ndant for type 1 diabetes or not (4): in a discordant twin pairone twin is treated with insulin, whereas the other oneisnt, and thus this degree of correlation suggests thatgenetic contributors to A1C may be detectable despite thesuperimposition of a strong environmental modier. Rig-orous estimates of heritability of treated A1C, however,\nare not available.",
              "title": "2010 - A Genome-Wide Association Study of Treated A1C.pdf",
              "version": "v0",
              "chunk_order": 7,
              "document_id": "4de9f054-4a02-5b6a-905d-420744075755",
              "extraction_id": "cce780d7-60c0-5cb3-976f-15e9808cab59",
              "associatedQuery": "How do genetic and epigenetic differences between monozygotic twins discordant for diabetes inform our understanding of its etiology?"
            }
          },
          {
            "id": "c57caee6-9b8f-5cb5-ab53-337dda1d92a7",
            "score": 0.7254222958776649,
            "metadata": {
              "text": "Concordance rate for type II diabetes mellitus in monozy-gotic twins: actuarial analysis. Diabetologia 42:146150\n3. Lehtovirta M, Kaprio J, Forsblom C, Eriksson J, Tuomilehto\nJ, Groop L (2000) Insulin sensitivity and insulin secretionin monozygotic and dizygotic twins. Diabetologia43:285293\n4. Florez JC, Hirschhorn J, Altshuler D (2003) The inherited\nbasis of diabetes mellitus: implications for the genetic anal-ysis of complex traits. Annu Rev Genomics Hum Genet4:257291",
              "title": "2004 - Common polymorphisms of the PPAR-\u03b32 (Pro12Ala) and PGC-1\u03b1 (Gly482Ser) genes are associated with the conversion from impaired glucose tolerance to type 2 diabetes in the STOP-NIDDM trial.pdf",
              "version": "v0",
              "chunk_order": 73,
              "document_id": "07b6ab4b-1aff-5d0e-aa98-d49a66b7b5d1",
              "extraction_id": "feb52f56-db94-5e03-90a8-af3bf38d087e",
              "associatedQuery": "How do genetic and epigenetic differences between monozygotic twins discordant for diabetes inform our understanding of its etiology?"
            }
          },
          {
            "id": "bcfe4c03-f0bb-540d-b529-adda644998a6",
            "score": 0.7199152016791354,
            "metadata": {
              "text": "disease susceptibility is not explained by genetics alone; environ-\nmental factors, gene by environment interactions, and epigenetic\ninuences are likely to play important roles in the etiology of T1D\n[5,6] . Monozygotic (MZ) twin pairs, discordant for T1D, represent\nan ideal system to test susceptibility factors not attributable to\ngenetic variation, especially epigenetic variation, since the ge-\nnomes of the twins are identical. The ascertainment of disease-",
              "title": "2016 - Hypomethylation within gene promoter regions and type 1 diabetes.pdf",
              "version": "v0",
              "chunk_order": 10,
              "document_id": "3d0f1bab-738c-5ba3-8fd9-206b3455704d",
              "extraction_id": "bc569d05-fc39-5487-95e7-63b0d7bf9b7e",
              "associatedQuery": "How do genetic and epigenetic differences between monozygotic twins discordant for diabetes inform our understanding of its etiology?"
            }
          },
          {
            "id": "4e393756-1b48-56f7-a3e7-33d15a55dc73",
            "score": 0.7184282876934713,
            "metadata": {
              "text": "epigenetic differences among monozygotic twins. A critical\nquestion is whether epigenetic marks are transmitted intactfrom parent to offspring and whether DNAm is allele-\nspecific and covaries with allele-specific gene expression.\nFor example, can we develop an epigenetic transmissiontest comparable to the transmission disequilibrium test used\nin genetic epidemiology? Finally, and most excitingly, we",
              "title": "2010 - Genome-scale approaches to the epigenetics of common.pdf",
              "version": "v0",
              "chunk_order": 80,
              "document_id": "3f21702c-3fdf-50b5-8060-04a9a1ce09ae",
              "extraction_id": "8881623e-fe7a-53bd-b703-2e8bf6a5c240",
              "associatedQuery": "How do genetic and epigenetic differences between monozygotic twins discordant for diabetes inform our understanding of its etiology?"
            }
          },
          {
            "id": "a4c3abc2-635c-5ace-ad39-c678a505fa48",
            "score": 0.7131724699530092,
            "metadata": {
              "text": "their dietary and physical activity habits (Maes\net al, 1997 ).\nThere is also ample evidence that diabetes\nhas a substantial genetic component. The con-\ncordance of type 2 diabetes in monozygotictwins ranges between 50 and 70% compared to\n2037% in dizygotic twins (Kaprio et al, 1992 ;\nNewman et al, 1987 ; Poulsen et al 1999). Further\nevidence comes from studies that compare therisk in offspring with a family history of type\n2 diabetes with offspring without such a fam-",
              "title": "2010 - Candidate Gene and Genome-Wide Association Studies in Behavioral Medicine.pdf",
              "version": "v0",
              "chunk_order": 4221,
              "document_id": "17637a6f-804e-50e4-9cf5-37318e17f15c",
              "extraction_id": "2778ece8-df84-58d2-9002-e036f0d007dc",
              "associatedQuery": "How do genetic and epigenetic differences between monozygotic twins discordant for diabetes inform our understanding of its etiology?"
            }
          },
          {
            "id": "f295f1d3-43d5-5eef-a39f-a8b91c47500f",
            "score": 0.7123925432690977,
            "metadata": {
              "text": "monozygotic and dizygotic Danish twin pairs withinsulin dependent diabetes mellitus. Bmj 1997: 314:1575 1579.\n30. R\nEDONDO MJ, R EWERS M, Y UL et al. Genetic deter-\nmination of islet cell autoimmunity in monozygotictwin, dizygotic twin, and non-twin siblings of patientswith type 1 diabetes: prospective twin study. Bmj 1999:318: 698 702.\n31. L\nEVY-M ARCHAL C, P ATTERSON C, G REEN A. Variation",
              "title": "2003 -Genetic epidemiology of type 1 diabetes.pdf",
              "version": "v0",
              "chunk_order": 122,
              "document_id": "cbc7f2d3-3f65-50ba-b281-96dd1c77f2c0",
              "extraction_id": "592fd011-4dfb-5a78-8973-482e35541468",
              "associatedQuery": "How do genetic and epigenetic differences between monozygotic twins discordant for diabetes inform our understanding of its etiology?"
            }
          },
          {
            "id": "0cd29c12-48e8-5f9f-9744-6b8acfaae0c7",
            "score": 0.7090156847825352,
            "metadata": {
              "text": "Studies in twins have demonstrated that 5070 % in the body mass index (BMI) variance may be explained by genetics (   Allison et al., 1996   ), and T2DM concordance was reported ranging from 1737 % in dizygotic to 5070 % in monozygotic twins (   Kaprio et al., 1992   ;    Medici et al., 1999   ;    Poulsen et al., 1999   ). In addition, family and adoption studies have reported heritability ranging from 2060 % for obesity (   Rice et al., 1999   ;    Stunkard et al., 1986   ) and 3070 % for T2DM (   Meigs",
              "title": "2011 - Lifestyle and Genetics in Obesity and type 2 Diabetes.pdf",
              "version": "v0",
              "chunk_order": 14,
              "document_id": "a16d3328-039c-530a-bfe5-f6f80ecf2ad0",
              "extraction_id": "551087b1-8e80-5a7b-839a-304f566a6417",
              "associatedQuery": "How do genetic and epigenetic differences between monozygotic twins discordant for diabetes inform our understanding of its etiology?"
            }
          },
          {
            "id": "3a9e7574-8914-5a96-86b6-b7b87a89b894",
            "score": 0.7063656203310993,
            "metadata": {
              "text": "Monozygotic twins exhibit numerous epigenetic differences: clues to twindiscordance? Schizophr Bull 29: 169178.\n8. Oates NA, van Vliet J, Duffy DL, Kroes HY, Martin NG, et al. (2006) Increased\nDNA methylation at the AXIN1 gene in a monozygotic twin from a pair\ndiscordant for a caudal duplication anomaly. Am J Hum Genet 79: 155162.\n9. Kuratomi G, Iwamoto K, Bundo M, Kusumi I, Kato N, et al. (2008) Aberrant\nDNA methylation associated with bipolar disorder identified from discordant",
              "title": "2013 - Continuous Aging of the Human DNA Methylome.pdf",
              "version": "v0",
              "chunk_order": 106,
              "document_id": "34783678-d4c2-566f-9b5b-0db18a72e86d",
              "extraction_id": "a0916b04-3463-5247-94da-0c97fd5da20d",
              "associatedQuery": "How do genetic and epigenetic differences between monozygotic twins discordant for diabetes inform our understanding of its etiology?"
            }
          },
          {
            "id": "af680560-47c6-5556-bb80-c7584d762f66",
            "score": 0.7053382782537412,
            "metadata": {
              "text": "5\nE/C128orts to estimate the heritability of T2D by a comparison of the concordance rates in\nmono- and dizygotic twins have varied greatly as a result of di/C128erences in ascertainment\nscheme, diagnostic criteria and follow-up duration.69Concordance for diabetes is\ngenerally higher in identical twins (supporting a genetic basis for disease), although the\nextremely high concordance rates in some early studies6were undoubtedly inated by\nascertainment bias.\nEvidence from population studies",
              "title": "2001 - The genetics of type 2 diabetes.pdf",
              "version": "v0",
              "chunk_order": 8,
              "document_id": "8ab06972-1c6c-5d68-a270-65fb0af0917b",
              "extraction_id": "737e4fe2-91ba-50c5-8f64-1149944fb60c",
              "associatedQuery": "How do genetic and epigenetic differences between monozygotic twins discordant for diabetes inform our understanding of its etiology?"
            }
          }
        ],
        "kg_search_results": null
      }
    }
  },
  "10": {
    "results": {
      "completion": {
        "id": "chatcmpl-AIHJknEcr96E1ybbJw2DE0EMMQI1v",
        "choices": [
          {
            "finish_reason": "stop",
            "index": 0,
            "message": {
              "content": "Recent studies on the interaction between genetic variants and environmental factors in diabetes development have identified several potential therapeutic targets. These include:\n\n1. **Primary Regulators of Insulin Secretion and Action**: Several type 2 diabetes (T2D) risk variants have been identified as primary regulators of insulin secretion, insulin action, and pancreatic islet transcription factors. This suggests that targeting these pathways could be a potential therapeutic strategy [6].\n\n2. **Specific Genetic Variants**: Newly discovered single nucleotide variants (SNVs) allow for better characterization of abnormalities in early insulin processing and secretion. Genes such as TCF7L2, SLC30A8, and C2CD4B have been highlighted as potential targets due to their roles in these processes [6].\n\n3. **Gene-Environment Interactions**: The interaction between genetic susceptibility and environmental factors such as physical activity and dietary fat has been shown to modify the risk of glucose homeostasis and T2D. This indicates that interventions targeting these environmental factors could potentially mitigate the genetic risk [7].\n\nThese findings underscore the importance of considering both genetic and environmental factors in developing therapeutic strategies for diabetes.",
              "role": "assistant"
            }
          }
        ],
        "created": 1728919836,
        "model": "gpt-4o-2024-08-06",
        "object": "chat.completion",
        "system_fingerprint": "fp_6b68a8204b",
        "usage": {
          "completion_tokens": 231,
          "prompt_tokens": 1238,
          "total_tokens": 1469
        }
      },
      "search_results": {
        "vector_search_results": [
          {
            "id": "b092c8b9-edb1-55fb-ae16-c67e3298946e",
            "score": 0.7191900443548004,
            "metadata": {
              "text": "that genetic studies will ultimately identify key genetic elements that help determine susceptibility to diabetes,disease progression, and responsiveness to specific therapies, as well as help identify novel targets for futureintervention. A substantial number of genetic loci, gene polymorphisms, and mutations have already beenreported as having variable degrees of association with one or other type of diabetes (type 1, type 2, maturityonset diabetes of the young [MODY]), while others appear to be involved",
              "title": "2008 - Glossary of Genetics Genomics Terms.pdf",
              "version": "v0",
              "chunk_order": 3,
              "document_id": "c66d2572-071d-5aaf-829c-b3ca6cf6d697",
              "extraction_id": "53e868dd-b318-5cf3-8b2e-98a548aab7cf",
              "associatedQuery": "What potential therapeutic targets have been identified through recent studies on the interaction between genetic variants and environmental factors in diabetes development?"
            }
          },
          {
            "id": "55f842a4-506a-5992-9b6e-47c81aee6809",
            "score": 0.7178966234860804,
            "metadata": {
              "text": "ponse to thiazolidinedione therapy and candidate genes \n[100103]. Results from pharmacogenetic studies could \npotentially provide physicians with a powerful tool to \nadjust therapy appropriately for those individuals carry\ning variants known to affect a given medication. Distefano \nand Watanabe have recently reviewed the pharmaco\ngenetics of diabetes [104].\nGenegene and geneenvironment interactions are also \nlikely to be helpful to the clinician in making therapeutic",
              "title": "2011 - Inherited destiny Genetics and gestational diabetes mellitus.pdf",
              "version": "v0",
              "chunk_order": 65,
              "document_id": "6d341cd2-ae56-5807-9aff-39298efc4d06",
              "extraction_id": "48c3e4a4-db23-5fca-9c46-775e80894655",
              "associatedQuery": "What potential therapeutic targets have been identified through recent studies on the interaction between genetic variants and environmental factors in diabetes development?"
            }
          },
          {
            "id": "728c47bb-e8e2-5359-9ff5-9ad9b13f999c",
            "score": 0.7136673160593114,
            "metadata": {
              "text": "Genomics of T2D\nDiet, lifestyle, environment, and even genetic variation influence an individuals response to disease therapy. Like GWAS which identify genetic variants conferring risk for a disease, studies have been carried out for iden -\ntifying genetic variants responsible for patient differ -",
              "title": "2015 - Genetics, genomics and personalized medicine in Type 2 Diabetes.pdf",
              "version": "v0",
              "chunk_order": 38,
              "document_id": "d8b85c3e-62f3-5e67-99b0-d0a2f225aff0",
              "extraction_id": "52a000e5-d790-55f2-9eac-14554d426173",
              "associatedQuery": "What potential therapeutic targets have been identified through recent studies on the interaction between genetic variants and environmental factors in diabetes development?"
            }
          },
          {
            "id": "15872da6-8175-5db6-b741-10ae3cf85088",
            "score": 0.7108629385408409,
            "metadata": {
              "text": "ease caused by interactions between multiple genetic\nand environmental factors. Significant progress has been\nmade in understanding the genetic architecture of T2D\nover the past 10 years [1]. A number of genome-wide as-\nsociation studies in diverse human populations have\nidentified more than 60 common variants and loci asso-\nciated with risk for T2D [2]. These studies have also\nrevealed a significant overlap between traits and pheno-\ntypes of monogenic diabetes with related common",
              "title": "2017 - Spectrum of mutations in monogenic diabetes genes identified from high-throughput DNA sequencing of 6888 individuals.pdf",
              "version": "v0",
              "chunk_order": 10,
              "document_id": "18a8a000-69ed-5d34-b13f-f5ae016d1067",
              "extraction_id": "b24927c4-ee83-51a8-b431-b43be7d3b678",
              "associatedQuery": "What potential therapeutic targets have been identified through recent studies on the interaction between genetic variants and environmental factors in diabetes development?"
            }
          },
          {
            "id": "53fd1ea0-5ca7-5066-bb07-e7469c640e22",
            "score": 0.7107654041508094,
            "metadata": {
              "text": "21582171 (2014).\n 29. Wood, A. R. et al. A genome-wide association study of IVGTT-based measures of first-phase insulin secretion refines the underlying physiology of \ntype 2 diabetes variants. Diabetes  66, 22962309 (2017). 30. Pickrell, J. K. Joint analysis of functional genomic data and genome- \nwide association studies of 18 human traits. Am. J. Hum. Genet. 94,  \n559573 (2014).\n 31. Plenge, R. M., Scolnick, E. M. & Altshuler, D. Validating therapeutic targets",
              "title": "2018 - Fine-mapping type 2 diabetes loci to single-variant resolution using high-density imputation and islet-specific epigenome maps.pdf",
              "version": "v0",
              "chunk_order": 97,
              "document_id": "ab2868dd-62f6-5350-994c-fcea4328e8a3",
              "extraction_id": "9190d1c1-41a4-5af3-a570-7fea6a15e71a",
              "associatedQuery": "What potential therapeutic targets have been identified through recent studies on the interaction between genetic variants and environmental factors in diabetes development?"
            }
          },
          {
            "id": "027f0c97-d38d-551d-add3-4a759a406895",
            "score": 0.7100871032528355,
            "metadata": {
              "text": "by GWASs [ 16,28,29]. A wide variety of network-based approaches have been applied to investigate the\nextent to which the genetics of T2D predisposition converge on a restricted set of biological pathways.\nSeveral T2D risk variants have been identied as primary regulators of insulin secretion, insulin\naction, and pancreatic islet transcription factors. [ 10,16]. The newly discovered SNVs allow the better\ncharacterization of abnormalities in early insulin processing and secretion. TCF7L2 ,SLC30A8 ,C2CD4B ,",
              "title": "2018 - High-Throughput Approaches onto Uncover (Epi)Genomic Architecture of Type 2 Diabetes.pdf",
              "version": "v0",
              "chunk_order": 26,
              "document_id": "1cb0c4ac-c1fe-55c2-919c-52cd5018c00d",
              "extraction_id": "455b92f7-6156-5735-8586-29a66af0f9e5",
              "associatedQuery": "What potential therapeutic targets have been identified through recent studies on the interaction between genetic variants and environmental factors in diabetes development?"
            }
          },
          {
            "id": "155260c5-ba90-540f-8d48-bafece83fa47",
            "score": 0.709456205368047,
            "metadata": {
              "text": "[10] , many environmental factors  [11] , and the interac-\ntions among those genetic and environmental factors. Physical activity and dietary fat have been reported to be important modifiers of the associations between glucose homeostasis and well-known candidate genes for T2DM  \n[12]  and there is reason to believe that a significant pro-\nportion of the susceptibility genes identified by GWASs will interact with these environmental factors to influ-ence the disease risk. Florez et al.",
              "title": "2010 - Evidence of Interaction between Type 2 Diabetes.pdf",
              "version": "v0",
              "chunk_order": 9,
              "document_id": "1a33b1d1-23ee-5b33-b42d-c745c8210166",
              "extraction_id": "d2de4ed1-897b-5e5b-bc29-c03310096d64",
              "associatedQuery": "What potential therapeutic targets have been identified through recent studies on the interaction between genetic variants and environmental factors in diabetes development?"
            }
          },
          {
            "id": "3d00ac57-9828-5146-a895-9840de9af5f7",
            "score": 0.7094038302061773,
            "metadata": {
              "text": "interactions suggest a way by which genetic risk may beameliorated, these environmental factors are of great relevanceto public health, and are the focus of a growing number of studies\n[7].\nEnvironmental factors, such as diet and lifestyle, are important\nin the onset, development and progression of T2D and its related\nphenotypes [8,9]. The interactions of environmental factors with",
              "title": "2013 - Genome-Wide Contribution of Genotype by Environment Interaction.pdf",
              "version": "v0",
              "chunk_order": 13,
              "document_id": "8c310d76-0a3b-574c-9859-859258870ee5",
              "extraction_id": "f3975a2c-8a66-582e-a4b8-868b1f4722d4",
              "associatedQuery": "What potential therapeutic targets have been identified through recent studies on the interaction between genetic variants and environmental factors in diabetes development?"
            }
          },
          {
            "id": "518d294f-67c5-5870-9f28-3cb4dfa81e42",
            "score": 0.7045328860918147,
            "metadata": {
              "text": "cases. J Am Med Assoc. 1956;161:1628 30.\n3. Duncan LE, Keller MC. A critical review of the first 10 years of\ncandidate gene-by-environment interaction research in psychiatry.\nAm J Psychiatry. 2011;168:1041 9.\n4. Brito EC et al. Previously associated type 2 diabetes variants may\ninteract with physical activity to modify the risk of impaired glu-\ncose regulation and type 2 diabetes: a study of 16,003 Swedish\nadults. Diabetes. 2009;58:1411 8.",
              "title": "2016 - Putting the Genome in Context Gene-Environment Interactions.pdf",
              "version": "v0",
              "chunk_order": 112,
              "document_id": "ea43bb66-b6fe-5682-8f48-90568c080401",
              "extraction_id": "cb5c4aab-77ed-58cd-98b8-9e1ba64eb9cf",
              "associatedQuery": "What potential therapeutic targets have been identified through recent studies on the interaction between genetic variants and environmental factors in diabetes development?"
            }
          },
          {
            "id": "6b83f0af-1145-5679-9dae-0f645771d25d",
            "score": 0.7044464945793202,
            "metadata": {
              "text": "this occurs. Findings to date, however, indicate\nthat behavioral changes can substantially\nmitigate diabetogenic and obesogenic effects\nof individual or multiple risk alleles, which\nhas much broader clinical and public health\nimplications.We have seen considerable progress in our\nunderstanding of the role that both environ-\nment and genetics play in the development of\nT2D. Recent work suggests that the adverse\neffect of some established T2D-associated\nloci may be greatly attenuated by appropriate",
              "title": "2012 - Gene-Environment Interactions in the Development of Type 2 Diabetes.pdf",
              "version": "v0",
              "chunk_order": 77,
              "document_id": "ea9601ed-ad83-506e-b1b7-e7211671ff73",
              "extraction_id": "89bf4316-d0cc-5310-a45e-1dd8b8aefe1b",
              "associatedQuery": "What potential therapeutic targets have been identified through recent studies on the interaction between genetic variants and environmental factors in diabetes development?"
            }
          }
        ],
        "kg_search_results": null
      }
    }
  },
  "11": {
    "results": {
      "completion": {
        "id": "chatcmpl-AIHJu3dzRIlHnqdmlSb6lzwzfrntr",
        "choices": [
          {
            "finish_reason": "stop",
            "index": 0,
            "message": {
              "content": "Rare variants identified through whole-genome sequencing contribute to the heritability of Type 2 diabetes by potentially explaining some of the \"missing heritability\" that common variants identified through genome-wide association studies (GWAS) do not account for. While GWAS have identified many common variants associated with Type 2 diabetes, these explain only a fraction of the heritability of the disease [4]. The missing heritability could be located in low-frequency and rare variants, particularly in noncoding regions of the genome [1]. However, studies have shown that rare coding variants, especially when clustered in a small number of genes, are unlikely to account for much of the missing heritability [10]. Instead, if rare coding variants are significant, they are likely scattered across many genes [10]. Therefore, while rare variants may contribute to the heritability of Type 2 diabetes, their exact role and impact remain to be fully elucidated, and larger multi-population studies are needed to reliably identify rare variants exclusively associated with Type 2 diabetes [6].",
              "role": "assistant"
            }
          }
        ],
        "created": 1728919846,
        "model": "gpt-4o-2024-08-06",
        "object": "chat.completion",
        "system_fingerprint": "fp_6b68a8204b",
        "usage": {
          "completion_tokens": 213,
          "prompt_tokens": 1205,
          "total_tokens": 1418
        }
      },
      "search_results": {
        "vector_search_results": [
          {
            "id": "935ff4d1-9840-5fab-8ad8-82e668319e91",
            "score": 0.7924776920702824,
            "metadata": {
              "text": "and rare coding variants do not account for much of theheritability of type 2 diabetes. Under this scenario, themissing heritability could be located in common orlow-frequency and rare variants in noncoding regionsof the genome. Recent studies that jointly modeled dia-betes or obesity risk as a function of genetic relatednessacross all of the GWAS SNPs have suggested that much\nof the heritability of these traits can be explained by",
              "title": "2013 -Whole-Exome Sequencing of 2,000 Danish Individuals.pdf",
              "version": "v0",
              "chunk_order": 110,
              "document_id": "2f2f6ff1-8f05-510d-bcf6-b1860fd9350c",
              "extraction_id": "01778b74-61b2-5f64-be8e-775c79af171d",
              "associatedQuery": "How do rare variants identified through whole-genome sequencing contribute to the heritability of Type 2 diabetes?"
            }
          },
          {
            "id": "b6905dfe-c622-58cd-b9ac-2cdcedada7e4",
            "score": 0.7733684547731772,
            "metadata": {
              "text": "T2D heritability.\n3. Uncovering the Signicance of Rare-Coding and Non-Coding Genetic Variants in the Etiology\nof Type 2 Diabetes\nAs previously stated, GWASs have uncovered many new genetic associations that are relevant to\nT2D, but GWAS ndings represent common and mid-frequency genetic variations, thus excluding\nrare frequency variants and also cumulative effect of many variants with small effect sizes. Missing\nheritability refers to the portion of genetic variance that cannot be explained by all signicant",
              "title": "2018 - High-Throughput Approaches onto Uncover (Epi)Genomic Architecture of Type 2 Diabetes.pdf",
              "version": "v0",
              "chunk_order": 34,
              "document_id": "1cb0c4ac-c1fe-55c2-919c-52cd5018c00d",
              "extraction_id": "0f2a8ab2-1666-50c0-b0b0-2a37e1f6917f",
              "associatedQuery": "How do rare variants identified through whole-genome sequencing contribute to the heritability of Type 2 diabetes?"
            }
          },
          {
            "id": "0ab59821-8bf4-50d3-92e7-b0dd593883a8",
            "score": 0.7672769794679619,
            "metadata": {
              "text": "could be accounted for by low-frequency and rare variants\nof moderate effect in a small number of genes. Our whole-exome sequencing study has explicitly addressed thisquestion. Additionally, we did not examine whether thereare fewer than 20 genes involved in type 2 diabetes butrather looked at whether rare coding variants in fewerthan 20 genes account for much of the heritability. In\nsuch a model, any number of other genes that do not",
              "title": "2013 -Whole-Exome Sequencing of 2,000 Danish Individuals.pdf",
              "version": "v0",
              "chunk_order": 101,
              "document_id": "2f2f6ff1-8f05-510d-bcf6-b1860fd9350c",
              "extraction_id": "01778b74-61b2-5f64-be8e-775c79af171d",
              "associatedQuery": "How do rare variants identified through whole-genome sequencing contribute to the heritability of Type 2 diabetes?"
            }
          },
          {
            "id": "e266cecd-e881-5c64-8ce3-4894cbe47db5",
            "score": 0.7656311988830621,
            "metadata": {
              "text": "contribute to individual risk, has been long debated. Genome-wide association studies have identified scores of common \nvariants associated with type 2 diabetes, but in aggregate, these explain only a fraction of the heritability of this disease. \nHere, to test the hypothesis that lower-frequency variants explain much of the remainder, the GoT2D and T2D-GENES \nconsortia performed whole-genome sequencing in 2,657 European individuals with and without diabetes, and exome",
              "title": "2016 - The genetic architecture of type 2 diabetes.pdf",
              "version": "v0",
              "chunk_order": 12,
              "document_id": "d7e2a9de-46f1-5191-9cb0-dd68eb9f365a",
              "extraction_id": "c266fa33-e779-514b-9337-636a69c6e6a4",
              "associatedQuery": "How do rare variants identified through whole-genome sequencing contribute to the heritability of Type 2 diabetes?"
            }
          },
          {
            "id": "aacfbc09-f4ec-5b71-a4a8-efd43cf8b6db",
            "score": 0.7605376982368425,
            "metadata": {
              "text": "One common disease that has been subjected to intense\ngenetic study is type 2 diabetes.\n32The heritability of type 2\ndiabetes has been estimated to be around 30%.3335\nThrough GWASs, 63 loci have been reproducibly associ-ated with type 2 diabetes.\n36However, as for other complex\ntraits, the associated SNPs can only account for <20% of\nthe heritability estimated from family studies.36\nHere, we seek to evaluate the role that rare coding vari-",
              "title": "2013 -Whole-Exome Sequencing of 2,000 Danish Individuals.pdf",
              "version": "v0",
              "chunk_order": 18,
              "document_id": "2f2f6ff1-8f05-510d-bcf6-b1860fd9350c",
              "extraction_id": "89a75bbb-f0f6-5391-98fd-56631343a38e",
              "associatedQuery": "How do rare variants identified through whole-genome sequencing contribute to the heritability of Type 2 diabetes?"
            }
          },
          {
            "id": "0977e7bd-4c4c-5c6c-a4d1-3b5f6fda03c5",
            "score": 0.7549468053893237,
            "metadata": {
              "text": "prevalence of T2D. These authors found rare variants that were not detected previously in population\nstudies, but none of them were associated with T2D [ 49]. Larger multi-population studies and more\nadvanced study methods are needed to reliably identify rare variants that are exclusively associated\nwith T2D to eventually uncover missing T2D heritability.\n3.2. Genetic Variants in Familial Studies of Type 2 Diabetes\nThe development of T2D is driven by the combined effect of environmental factors and a",
              "title": "2018 - High-Throughput Approaches onto Uncover (Epi)Genomic Architecture of Type 2 Diabetes.pdf",
              "version": "v0",
              "chunk_order": 50,
              "document_id": "1cb0c4ac-c1fe-55c2-919c-52cd5018c00d",
              "extraction_id": "524594ab-31ca-5f5c-8126-7c58060bb73e",
              "associatedQuery": "How do rare variants identified through whole-genome sequencing contribute to the heritability of Type 2 diabetes?"
            }
          },
          {
            "id": "9a3d06ce-e86f-511f-82ac-97e486618e47",
            "score": 0.7514202370816429,
            "metadata": {
              "text": "variance in disease risk that can be accounted for bythe 63 previously identied associations with commonvariants.\nOur empirical and simulation results are compatible\nwith a variety of different genetic architectures for type2 diabetes. First, if rare coding variants are responsiblefor the majority of the heritability of the trait, the variants\nare most likely scattered across many ( >20) different",
              "title": "2013 -Whole-Exome Sequencing of 2,000 Danish Individuals.pdf",
              "version": "v0",
              "chunk_order": 107,
              "document_id": "2f2f6ff1-8f05-510d-bcf6-b1860fd9350c",
              "extraction_id": "01778b74-61b2-5f64-be8e-775c79af171d",
              "associatedQuery": "How do rare variants identified through whole-genome sequencing contribute to the heritability of Type 2 diabetes?"
            }
          },
          {
            "id": "451c2da6-3fd5-53f4-a58e-32b4f1d2cbbd",
            "score": 0.7468359772162465,
            "metadata": {
              "text": "Genome-wide association studies (GWAS) have been helpful in identifying a large number of genetic variants conferring risk to T2D. However, only close to 10% heritability is explained by these variants. Other genetic variants, particularly those which are rare but with significant effects need to be identified.",
              "title": "2015 - Genetics, genomics and personalized medicine in Type 2 Diabetes.pdf",
              "version": "v0",
              "chunk_order": 120,
              "document_id": "d8b85c3e-62f3-5e67-99b0-d0a2f225aff0",
              "extraction_id": "5cd40b2d-72d6-5386-be94-b4e8188e4114",
              "associatedQuery": "How do rare variants identified through whole-genome sequencing contribute to the heritability of Type 2 diabetes?"
            }
          },
          {
            "id": "40f471a6-3615-52f3-a306-9f3568680409",
            "score": 0.7453516429243513,
            "metadata": {
              "text": "and rare sequence variants associated with elevated \nor reduced risk of type 2 diabetes. Nat. Genet. 46, \n294298 (2014).\n168. Lek, M. etal. Analysis of protein-coding genetic \nvariation in 60,706 humans. Nature 536, 285291 \n(2016).169. Xue, A. etal. Genome-wide association analyses \nidentify 143 risk variants and putative regulatory \nmechanisms for type 2 diabetes. Nat. Commun. 9, \n2941 (2018).\n170. Huyghe, J. R. etal. Exome array analysis identifies",
              "title": "2020 - Insights into pancreatic islet cell dysfunction from type 2 diabetes mellitus genetics..pdf",
              "version": "v0",
              "chunk_order": 168,
              "document_id": "2a386c81-8f24-5993-8e48-0e89d7fb4fec",
              "extraction_id": "36db7673-abd7-55b1-9caf-b66498e19e78",
              "associatedQuery": "How do rare variants identified through whole-genome sequencing contribute to the heritability of Type 2 diabetes?"
            }
          },
          {
            "id": "a5469aca-198e-56f5-ab92-16fd00c5e0fc",
            "score": 0.7422869512692748,
            "metadata": {
              "text": "diabetes. In particular, our study suggests that when clus-tered in a small number of genes, rare coding variants ofmoderate to strong effect are unlikely to account formuch of the missing heritability. Rather, if rare coding var-iants are an important factor in type 2 diabetes risk, theyare most likely scattered across many genes. Our resultshave important implications for the design and interpreta-\ntion of future medical resequencing studies.\nSubjects and Methods\nStudy Populations",
              "title": "2013 -Whole-Exome Sequencing of 2,000 Danish Individuals.pdf",
              "version": "v0",
              "chunk_order": 20,
              "document_id": "2f2f6ff1-8f05-510d-bcf6-b1860fd9350c",
              "extraction_id": "89a75bbb-f0f6-5391-98fd-56631343a38e",
              "associatedQuery": "How do rare variants identified through whole-genome sequencing contribute to the heritability of Type 2 diabetes?"
            }
          }
        ],
        "kg_search_results": null
      }
    }
  },
  "12": {
    "results": {
      "completion": {
        "id": "chatcmpl-AIHK1iKM6Po4mTFBDwGSbImYGY94p",
        "choices": [
          {
            "finish_reason": "stop",
            "index": 0,
            "message": {
              "content": "The latest findings on the role of non-coding RNAs in the pathogenesis of diabetes highlight several key aspects:\n\n1. **Role of lncRNAs in Diabetes**: Long non-coding RNAs (lncRNAs) are implicated in mediating complex pathological mechanisms of diabetes. They are involved in post-transcriptional regulation and are associated with orchestrated networks that influence diabetes pathogenesis [5]. LncRNAs are considered better therapeutic targets due to their specific functions in regulating cellular pathways and their expression patterns that correlate with the progression of diabetes [7].\n\n2. **Epigenetic Influence**: Non-coding RNAs, including microRNAs and lncRNAs, can influence epigenetic mechanisms. They can promote the expression of pathological genes through post-transcriptional and post-translational mechanisms, contributing to metabolic memory and sustained gene expression in diabetic conditions [4].\n\n3. **Regulation of Islet Function**: LncRNAs have been shown to regulate pancreatic islet function, which is central to understanding diabetes pathophysiology. For instance, the lncRNA H19 has been implicated in islet development and function [8].\n\n4. **MicroRNAs in Disease**: MicroRNAs (miRs) play critical roles in various diseases, including diabetes, by influencing proliferation, differentiation, and development [2].\n\nThese findings underscore the importance of non-coding RNAs as regulatory players in diabetes and its complications, offering potential avenues for therapeutic intervention.",
              "role": "assistant"
            }
          }
        ],
        "created": 1728919853,
        "model": "gpt-4o-2024-08-06",
        "object": "chat.completion",
        "system_fingerprint": "fp_a20a4ee344",
        "usage": {
          "completion_tokens": 295,
          "prompt_tokens": 1346,
          "total_tokens": 1641
        }
      },
      "search_results": {
        "vector_search_results": [
          {
            "id": "1eb14f0c-3d81-53bc-91c8-98acf2e014b7",
            "score": 0.719383716583252,
            "metadata": {
              "text": "13\nDe Rosa et al.\nType 2 Diabetes and CVD\nFrontiers in Endocrinology | www.frontiersin.org January 2018 | Volume 9 | Article 2176. Fatica A, Bozzoni I. Long non-coding RNAs: new players in cell differentia-\ntion and development. Nat Rev Genet (2014) 15:721. doi:10.1038/nrg3606 \n177. Wang KC, Chang HY . Molecular mechanisms of long noncoding RNAs. Mol Cell (2011) 43:90414. doi:10.1016/j.molcel.2011.08.018 \n178. Esteller M. Non-coding RNAs in human disease. Nat Rev Genet (2011) \n12:86174. doi:10.1038/nrg3074",
              "title": "2018 - Type 2 Diabetes Mellitus and Cardiovascular Disease Genetic and Epigenetic Links.pdf",
              "version": "v0",
              "chunk_order": 186,
              "document_id": "3e82a2e5-4b2c-59c0-99cd-f3b06d8dabf2",
              "extraction_id": "097b0feb-4ffa-5ad6-8140-5e404e83c80b",
              "associatedQuery": "What are the latest findings on the role of non-coding RNAs in the pathogenesis of diabetes?"
            }
          },
          {
            "id": "92a20945-b038-52a4-8cc8-ffb70e6f7559",
            "score": 0.7168975045146058,
            "metadata": {
              "text": "Epigenetic Mechanisms in Diabetic Complications     16 \nother non-coding RNAs can also in teract with transcriptional co -regulators and thereby further 337 \ninfluence epigenetics and tran scriptional regulation (82, 104). 338 \n Recent findings have demonstrated  a critical role for miRs in various diseases. They have 339 \nbeen found to play key roles in proliferation, di fferentiation, development, and in cancer, where 340",
              "title": "2010 - The Role of Epigenetics in the Pathology of Diabetic Complications.pdf",
              "version": "v0",
              "chunk_order": 71,
              "document_id": "766edfd5-4756-51bf-b636-c94b041d030c",
              "extraction_id": "b1d2c95c-d639-5c75-8c52-278f1e187675",
              "associatedQuery": "What are the latest findings on the role of non-coding RNAs in the pathogenesis of diabetes?"
            }
          },
          {
            "id": "9c11148d-9f7a-5d84-aa05-2b67e7a8f1f3",
            "score": 0.7107552117559452,
            "metadata": {
              "text": "Beltrami, C., Angelini, T.G., Emanueli, C., 2015. Noncoding RNAs in diabetes vascular\ncomplications. J. Mol. Cell. Cardiol. 89, 42 50.https://doi.org/10.1016/j.yjmcc.\n2014.12.014 .\nBrookheart, R.T., Michel, C.I., Listenberger, L.L., et al., 2009. The non-coding RNA gadd7\nis a regulator of lipid-induced oxidative and endoplasmic reticulum stress. J. Biol.Chem. 284, 7446 7454. https://doi.org/10.1074/jbc.M806209200 .\nCarter, G., Miladinovic, B., Patel, A.A., et al., 2015. Circulating long noncoding RNA",
              "title": "2018 - Pilot genome-wide association study identifying novel risk loci for type 2.pdf",
              "version": "v0",
              "chunk_order": 70,
              "document_id": "e2c6283c-d95f-574a-9dab-345a708d388c",
              "extraction_id": "a166bf29-6be5-54ff-b869-9d4ff087d1e5",
              "associatedQuery": "What are the latest findings on the role of non-coding RNAs in the pathogenesis of diabetes?"
            }
          },
          {
            "id": "19d9d3a6-c982-5c57-a16c-226b8aa76ed5",
            "score": 0.7056097388267517,
            "metadata": {
              "text": "Noncoding RNAs that are induced by diabetic conditions can also promote \ntheexpression of pathological genes via various post-transcriptional and \npost-translational mechanisms\n These epigenetic mechanisms and noncoding RNAs can lead to persistently \nopen chromatin structures at pathological genes and sustained gene \nexpression, which can also be a mechanism for metabolic memory\n Key epigenetic regulators, microRNAs and long noncoding RNAs could serve",
              "title": "2014 - Diabetic nephropathy\u2014emerging epigenetic mechanisms.pdf",
              "version": "v0",
              "chunk_order": 13,
              "document_id": "be05127e-1be8-5573-b571-51a11c3b2be2",
              "extraction_id": "79ce3e1f-1c6a-51dc-b5ad-848173af4e69",
              "associatedQuery": "What are the latest findings on the role of non-coding RNAs in the pathogenesis of diabetes?"
            }
          },
          {
            "id": "b774bf7b-4546-56d2-ae7b-7bc2c9f2fb08",
            "score": 0.696410080683532,
            "metadata": {
              "text": "tion among researchers ( Knoll et al., 2015 ). As an important post-transcriptional pathogenesis of diabetes,\nlncRNAs and their associated orchestrated networks are implicated in mediating complex pathological\nmechanisms of diabetes ( Kato et al., 2016; Liu et al., 2014 ). To delineate the inuence of lncRNAs and\n172 iScience 19, 162176, September 27, 2019",
              "title": "2019 - Development and Genome Sequencing.pdf",
              "version": "v0",
              "chunk_order": 105,
              "document_id": "18820c9e-f7ae-57ae-897d-0d9c3f616b6a",
              "extraction_id": "d971dced-935c-566b-a4a2-11bcf99b9c84",
              "associatedQuery": "What are the latest findings on the role of non-coding RNAs in the pathogenesis of diabetes?"
            }
          },
          {
            "id": "94eed8ea-cc78-52d0-a188-442380512b85",
            "score": 0.687047004699707,
            "metadata": {
              "text": "coding RNAs [18]. A number of indirect lines of evi-dence point to the involvement of epigenetic changes indiabetic nephropathy. Murine models of disease progres-sion displaying temporal variation in gene expressionhave indicated these supra-sequence devices may beinvolved in the pathogenesis [19]. Gene expressionchanges reflect dynamic alterations in gene transcription\nand also messenger RNA stabi lity, which may be influ-",
              "title": "2010 - Genome-wide DNA methylation analysis for diabetic nephropathy in type 1 diabetes mellitus.pdf",
              "version": "v0",
              "chunk_order": 12,
              "document_id": "23f0ee09-5536-5f63-bf15-bce1894b5fed",
              "extraction_id": "f9500ec9-0600-5e2c-b64e-b062fb7a7552",
              "associatedQuery": "What are the latest findings on the role of non-coding RNAs in the pathogenesis of diabetes?"
            }
          },
          {
            "id": "2d9e043b-a3fa-52dc-9a4e-71ed49f9ec1d",
            "score": 0.6849800944328308,
            "metadata": {
              "text": "To conclude, it would be apt to state that lncRNAs are widely implicated in diverse domains of cell metabolism and their altered expression is associated with diabetes and its complications. Although originally thought to be non-functional, lncRNA genes transcribe into lncRNAs that exert important and specific functions in regulating cellular pathways. Due to this specificity, lncRNAs are considered better therapeutic targets. In addition, their expression patterns in tissues quite follow the progress of",
              "title": "2018 - Lnc\u2011ing non\u2011coding RNAs with metabolism and diabetes roles.pdf",
              "version": "v0",
              "chunk_order": 88,
              "document_id": "019efefb-65db-55f5-a3a7-4f224473f51f",
              "extraction_id": "14656f4f-b0bd-5f4f-a67a-aeb902f24757",
              "associatedQuery": "What are the latest findings on the role of non-coding RNAs in the pathogenesis of diabetes?"
            }
          },
          {
            "id": "66b05301-179b-597c-bb68-e6fd0e0d1d5a",
            "score": 0.6833786368370056,
            "metadata": {
              "text": "NAs to be mapped to diabetic susceptible loci [49 52], \nall suggesting towards critical roles of lncRNAs in insulin resistance, diabetes, and its associated complications.\nLncRNAs asregulators ofislet function\nThe pancreatic islet is an important central node to researchers to understand the pathophysiology of diabe-tes [53]. The possible regulation of islet development and function by lncRNAs was first demonstrated by Ding etal., where the lncRNA, H19 (Fig. 4), was shown to be involved",
              "title": "2018 - Lnc\u2011ing non\u2011coding RNAs with metabolism and diabetes roles.pdf",
              "version": "v0",
              "chunk_order": 42,
              "document_id": "019efefb-65db-55f5-a3a7-4f224473f51f",
              "extraction_id": "96a78d74-ac6d-513e-a5a7-b22ef95ea041",
              "associatedQuery": "What are the latest findings on the role of non-coding RNAs in the pathogenesis of diabetes?"
            }
          },
          {
            "id": "4a8a2861-62b9-520c-8833-45fb8bd3ffd7",
            "score": 0.6825283358974269,
            "metadata": {
              "text": "expected to rise due to the increasing incidence of diabetes, which necessitates the need for exploration of new molecular aspects of DR to expand the current scope of therapy.\nIn the last two decades, the rapid advent of high-throughput genomic technology has made it evident that \nmore than 97% of the human genome is comprised of non-protein-coding elements, such as non-coding RNAs \n(ncRNAs)\n6. Although significant research has been conducted in annotating the transcripts that arise from these",
              "title": "2018 - MALAT1 An Epigenetic Regulator of Inflammation in Diabetic Retinopathy.pdf",
              "version": "v0",
              "chunk_order": 6,
              "document_id": "cd4dd3bc-bcea-5670-a40f-bc95c319f3ed",
              "extraction_id": "2d8abaf8-9f48-5b9a-b50e-897fd4751b7b",
              "associatedQuery": "What are the latest findings on the role of non-coding RNAs in the pathogenesis of diabetes?"
            }
          },
          {
            "id": "25d3616b-1ba4-59ce-a11b-38d108d5b387",
            "score": 0.6798065304756165,
            "metadata": {
              "text": "regulation, control of mRNA decay, and sequestration of transcription factors. Although the underlying causes that define \nthe diabetic phenotype are extremely intricate, most of the studies in the last decades were mostly centered on protein-coding \ngenes. However, current opinion in the recent past has authenticated the contributions of diverse lncRNAs as critical regula -\ntory players during the manifestation of diabetes. The current review will highlight the importance of lncRNAs in regulating",
              "title": "2018 - Lnc\u2011ing non\u2011coding RNAs with metabolism and diabetes roles.pdf",
              "version": "v0",
              "chunk_order": 2,
              "document_id": "019efefb-65db-55f5-a3a7-4f224473f51f",
              "extraction_id": "efc73cf6-99c6-5272-9bb0-7bd6a34633f0",
              "associatedQuery": "What are the latest findings on the role of non-coding RNAs in the pathogenesis of diabetes?"
            }
          }
        ],
        "kg_search_results": null
      }
    }
  },
  "13": {
    "results": {
      "completion": {
        "id": "chatcmpl-AIHKAjqtg6gr5hkyEsdT3wwz3yXTB",
        "choices": [
          {
            "finish_reason": "stop",
            "index": 0,
            "message": {
              "content": "The interaction between multiple polygenic risk scores (PRS) can improve the prediction of Type 1 and Type 2 diabetes by combining information from various genetic loci associated with these diseases. This approach allows for a more comprehensive assessment of an individual's genetic risk. Specifically, combining information from common risk polymorphisms has been shown to improve disease prediction for Type 2 diabetes [3]. Additionally, partitioning polygenic scores according to factors of disease heterogeneity and mapping genetic loci to different immune-cell subtypes can enhance the predictive power of PRS, particularly for Type 2 diabetes [9]. These strategies leverage the aggregation of genetic risk from multiple sources, thereby capturing a larger proportion of the genetic variance underlying these traits and improving early diagnosis, intervention, and prevention efforts [4].",
              "role": "assistant"
            }
          }
        ],
        "created": 1728919862,
        "model": "gpt-4o-2024-08-06",
        "object": "chat.completion",
        "system_fingerprint": "fp_6b68a8204b",
        "usage": {
          "completion_tokens": 154,
          "prompt_tokens": 1370,
          "total_tokens": 1524
        }
      },
      "search_results": {
        "vector_search_results": [
          {
            "id": "748c1d81-0c27-515a-8bf1-12e717645e66",
            "score": 0.7702065706253052,
            "metadata": {
              "text": "review of polygenic risk scores for type 1 and type 2 diabetes. Int J Mol \nSci. 2020;21(5):1703.\n 48. Khera AV, Chaffin M, Aragam KG, Haas ME, Roselli C, Choi SH, et al. \nGenome wide polygenic scores for common diseases identify \nindividuals with risk equivalent to monogenic mutations. Nat Genet. \n2018;50:121924.\n 49. Ding Y, Hou K, Burch KS, Lapinska S, Priv F, Vilhjalmsson B, et al. Large \nuncertainty in individual polygenic risk score estimation impacts PRS",
              "title": "2022 - Development and validation of a trans-ancestry polygenic risk score for type 2 diabetes in diverse populations.pdf",
              "version": "v0",
              "chunk_order": 176,
              "document_id": "4ece243f-acda-569d-b75d-37539260dcb3",
              "extraction_id": "02701cd5-d2ce-560c-b5a9-e694fecdb3c2",
              "associatedQuery": "How does the interaction between multiple polygenic risk scores (PRS) improve the prediction of Type 1 and Type 2 diabetes?"
            }
          },
          {
            "id": "2c09a46a-20d0-54b4-abcb-608fef7c7f80",
            "score": 0.7527217864990234,
            "metadata": {
              "text": "(GWAS), polygenic risk scores (PRS) have shown promise to complement established clinical risk factors and inter \nvention paradigms, and improve early diagnosis and prevention of T2D. However, to date, T2D PRS have been most \nwidely developed and validated in individuals of European descent. Comprehensive assessment of T2D PRS in non\nEuropean populations is critical for equitable deployment of PRS to clinical practice that benefits global populations.",
              "title": "2022 - Development and validation of a trans-ancestry polygenic risk score for type 2 diabetes in diverse populations.pdf",
              "version": "v0",
              "chunk_order": 3,
              "document_id": "4ece243f-acda-569d-b75d-37539260dcb3",
              "extraction_id": "f6f0c89d-5c35-5889-8619-a3914e5d2c7e",
              "associatedQuery": "How does the interaction between multiple polygenic risk scores (PRS) improve the prediction of Type 1 and Type 2 diabetes?"
            }
          },
          {
            "id": "3b9e0030-8bf9-5d63-9813-3cf18e98be3b",
            "score": 0.7513685822486877,
            "metadata": {
              "text": "prediction of type 2 diabetes. N. Engl. J. Med. 359, 22082219 (2008).\n 45. Weedon, M. N. et al. Combining information from common type 2 diabetes \nrisk polymorphisms improves disease prediction. PLoS. Med. 3, e374 (2006).\n 46. Euesden, J., Lewis, C. M. & OReilly, P . F. PRSice: Polygenic Risk Score \nsoftware. Bioinformatics  31, 14661468 (2015).\n 47. Gatineau, M. et al. Adult obesity and type 2 diabetes (Public Health England,",
              "title": "2018 - Fine-mapping type 2 diabetes loci to single-variant resolution using high-density imputation and islet-specific epigenome maps.pdf",
              "version": "v0",
              "chunk_order": 102,
              "document_id": "ab2868dd-62f6-5350-994c-fcea4328e8a3",
              "extraction_id": "9190d1c1-41a4-5af3-a570-7fea6a15e71a",
              "associatedQuery": "How does the interaction between multiple polygenic risk scores (PRS) improve the prediction of Type 1 and Type 2 diabetes?"
            }
          },
          {
            "id": "1677b3ee-7d95-5e10-a6dd-d80b4bb87b29",
            "score": 0.7487341388682768,
            "metadata": {
              "text": "(GWAS) in diverse populations have identified hundreds \nof genetic loci associated with T2D [79]. Polygenic risk \nscores (PRS), which aggregate the genetic risk of individ -\nual alleles across the genome, are thus promising to pre -\ndict future T2D occurrence and improve early diagnosis, \nintervention, and prevention of T2D [1015]. However, \nto date, T2D PRS were most widely developed and vali -\ndated in individuals of European descent. Given that the \npredictive performance of PRS often attenuates in non-",
              "title": "2022 - Development and validation of a trans-ancestry polygenic risk score for type 2 diabetes in diverse populations.pdf",
              "version": "v0",
              "chunk_order": 12,
              "document_id": "4ece243f-acda-569d-b75d-37539260dcb3",
              "extraction_id": "17c49e58-c89a-5495-b17f-adcade90a4c6",
              "associatedQuery": "How does the interaction between multiple polygenic risk scores (PRS) improve the prediction of Type 1 and Type 2 diabetes?"
            }
          },
          {
            "id": "a374d88e-458e-5252-8b3a-5ca162fa6982",
            "score": 0.7476893217748821,
            "metadata": {
              "text": "in advance. Polygenic Risk Scores (PRS) were proposed by Duncan L. et al. [ 8] for risk\nanalysis using the sum of the weight of each risk-associated locus of genomic sequence\nobtained from the corresponding evidence. These weights are assessed from the regression\ncoefcient associated with each locus. These combined genetics features and correlation\nmatrices would signicantly assist the entire eld of genomics study [ 9]. These studies on",
              "title": "2022 - Using Recurrent Neural Networks for Predicting Type-2 Diabetes from Genomic and Tabular Data.pdf",
              "version": "v0",
              "chunk_order": 17,
              "document_id": "be0e50e0-3de8-53c5-8126-a0b618647f80",
              "extraction_id": "3c30b33b-8928-5cee-9c37-c70642fff75c",
              "associatedQuery": "How does the interaction between multiple polygenic risk scores (PRS) improve the prediction of Type 1 and Type 2 diabetes?"
            }
          },
          {
            "id": "a551335d-c3ed-5d12-a611-9991d192cc1e",
            "score": 0.7324631214141846,
            "metadata": {
              "text": "performance.\nConclusions: By integrating T2D GWAS from multiple populations, we developed and validated a transancestry PRS, \nand demonstrated its potential as a meaningful index of risk among diverse patients in clinical settings. Our efforts \nrepresent the first step towards the implementation of the T2D PRS into routine healthcare.\nKeywords: Polygenic risk score, Type 2 diabetes, Diverse populations, Clinical implementation",
              "title": "2022 - Development and validation of a trans-ancestry polygenic risk score for type 2 diabetes in diverse populations.pdf",
              "version": "v0",
              "chunk_order": 20,
              "document_id": "4ece243f-acda-569d-b75d-37539260dcb3",
              "extraction_id": "17c49e58-c89a-5495-b17f-adcade90a4c6",
              "associatedQuery": "How does the interaction between multiple polygenic risk scores (PRS) improve the prediction of Type 1 and Type 2 diabetes?"
            }
          },
          {
            "id": "bcce1092-32ea-5f65-bc10-4dc1a2dac53a",
            "score": 0.7306916438708781,
            "metadata": {
              "text": "Owing to their small effect sizes, SNP associations have very little clinical applicability for risk prediction.  \nA polygenic risk score (PRS) attempts to estimate the combined risk from multiple SNPs that have been associated with a certain trait with genome-wide sig-nificance. By accounting for a large proportion of the \ngenetic variance underlying a trait, the overall effect size",
              "title": "2021 - Genetics and genomics of arrhythmic.pdf",
              "version": "v0",
              "chunk_order": 81,
              "document_id": "462ed035-e4fb-5847-a92d-927f05a2b58b",
              "extraction_id": "ada410d0-6b91-5959-b834-cc3389e29c5f",
              "associatedQuery": "How does the interaction between multiple polygenic risk scores (PRS) improve the prediction of Type 1 and Type 2 diabetes?"
            }
          },
          {
            "id": "635180f9-540f-5533-9d61-c5cfe14657fa",
            "score": 0.7235454320907593,
            "metadata": {
              "text": "8.Padilla-Mart nez, F., Collin, F., Kwasniewski, M., and Kretow-\nski, A. (2020). Systematic review of polygenic risk scores for\ntype 1 and type 2 diabetes. Int. J. Mol. Sci. 21, 1703 .\n9.Rao, A., and Knowles, J. (2019). Polygenic risk scores in coro-\nnary artery disease. Curr. Opin. Cardiol. 34, 435440 .\n10.Dikilitas, O., Schaid, D.J., Kosel, M.L., Carroll, R.J., Chute,\nC.G., Denny, J.A., Fedotov, A., Feng, Q., Hakonarson, H., Jar-vik, G.P., et al. (2020). Predictive utility of polygenic risk scores",
              "title": "2022 - Stability of polygenic scores across.pdf",
              "version": "v0",
              "chunk_order": 131,
              "document_id": "30af2d38-7941-5d0a-9da1-a8ad2dc22329",
              "extraction_id": "a548bb25-cbff-5466-b932-afe160bfbe32",
              "associatedQuery": "How does the interaction between multiple polygenic risk scores (PRS) improve the prediction of Type 1 and Type 2 diabetes?"
            }
          },
          {
            "id": "fd7ccb09-2768-5ceb-8b29-9b29cdef57a8",
            "score": 0.7130113840103149,
            "metadata": {
              "text": "partitioned polygenic scores according to factors of disease heteroge-\nneity, as successfully demonstrated for type 2 diabetes (32). Another\nstrategy could be the mapping of statistically associated genetic loci to\ndifferent immune-cell subtypes according to gene expression patterns\nderived from single-cell RNA sequencing (33).\nAutoimmune PRS, possibly in combination with other genetic and\nnongenetic predictors, may be of importance to manage the risk of",
              "title": "2022 - Coming of Age Human Genomics.pdf",
              "version": "v0",
              "chunk_order": 36,
              "document_id": "45506895-eef1-57f4-8ca1-79fe23a2493f",
              "extraction_id": "d2add072-cb41-54f8-9583-9616b11e4ae3",
              "associatedQuery": "How does the interaction between multiple polygenic risk scores (PRS) improve the prediction of Type 1 and Type 2 diabetes?"
            }
          },
          {
            "id": "cc476583-54c8-5607-95bd-d06ae875dfb8",
            "score": 0.705722591451484,
            "metadata": {
              "text": "genome-wide polygenic risk scores (PRSs) for four lipid traits. We validated ( n= 4271) and subsequently tested\nassociations of these scores with 3-year lipid changes in adolescents ( n= 620), carotid intima-media thickness (cIMT)\nin adult women ( n= 781), dyslipidemia ( n= 7723), and coronary heart disease (CHD) ( n= 2374 cases and 6246\ncontrols) in type 2 diabetes (T2D) patients.\n(Continued on next page)",
              "title": "2021- Development of genome-wide polygenic risk scores for lipid traits and clinical applications for dyslipidemia, subclinical atherosclerosis, and diabetes cardiovascular complications among East Asians.pdf",
              "version": "v0",
              "chunk_order": 3,
              "document_id": "ce8040c7-157f-54c5-b28b-3224e8871415",
              "extraction_id": "5f2ac528-4965-5d5e-86d0-8862032bb7b9",
              "associatedQuery": "How does the interaction between multiple polygenic risk scores (PRS) improve the prediction of Type 1 and Type 2 diabetes?"
            }
          }
        ],
        "kg_search_results": null
      }
    }
  },
  "14": {
    "results": {
      "completion": {
        "id": "chatcmpl-AIHKFuXAocol6QH0B6QHJlkuJdiDC",
        "choices": [
          {
            "finish_reason": "stop",
            "index": 0,
            "message": {
              "content": "Recent single-cell RNA-sequencing studies have provided significant mechanistic insights into beta-cell failure pathways. These insights include:\n\n1. **De-differentiation Signatures**: Single-cell analyses of human islet cells have revealed de-differentiation signatures, suggesting that beta cells may lose their specialized functions and revert to a more progenitor-like state, which contributes to their dysfunction in diabetes [1].\n\n2. **Transcriptional Regulation**: Advances in single-cell genomic profiling have enhanced our understanding of transcriptional regulation in non-beta cell types, which may play crucial roles in the hallmark features of beta-cell insufficiency and dysfunction in type 2 diabetes (T2D) [2].\n\n3. **ER Stress and Heterogeneity**: Single-cell transcriptomic analyses have identified subpopulations of beta cells experiencing endoplasmic reticulum (ER) stress. This stress is implicated in the dysfunction of both alpha and beta cells, contributing to diabetes pathogenesis [8].\n\nThese findings highlight the complexity of beta-cell failure and underscore the importance of single-cell technologies in unraveling the molecular mechanisms underlying diabetes.",
              "role": "assistant"
            }
          }
        ],
        "created": 1728919867,
        "model": "gpt-4o-2024-08-06",
        "object": "chat.completion",
        "system_fingerprint": "fp_6b68a8204b",
        "usage": {
          "completion_tokens": 223,
          "prompt_tokens": 1528,
          "total_tokens": 1751
        }
      },
      "search_results": {
        "vector_search_results": [
          {
            "id": "b7812a7a-5504-57ca-8755-969dee45717e",
            "score": 0.7292243838310242,
            "metadata": {
              "text": "Tang X, Huang Y, Lei J, Luo H, Zhu X (2019) The single-cell sequenc-\ning: new developments and medical applications. Cell Biosci \n9:53. https ://doi.org/10.1186/s1357 8-019-0314-y\nTeo AKK etal (2018) Single-cell analyses of human islet cells reveal \nde-differentiation signatures. Cell Death Discov 4:14. https ://doi.\norg/10.1038/s4142 0-017-0014-5\nTheis FJ, Lickert H (2019) A map of beta-cell differentiation pathways \nsupports cell therapies for diabetes. Nature 569:342343. https  ://",
              "title": "2020 - Advances of single?cell genomics and epigenomics in human disease.pdf",
              "version": "v0",
              "chunk_order": 158,
              "document_id": "afe53f5a-3962-520f-be55-9df5bfdaad70",
              "extraction_id": "7f7a7f30-2e4e-50aa-bbcb-9f211c371e38",
              "associatedQuery": "What are the mechanistic insights into the beta-cell failure pathways gleaned from recent single-cell RNA-sequencing studies?"
            }
          },
          {
            "id": "d5c2a32a-b869-59c1-8a63-45ab620669de",
            "score": 0.7201639413833618,
            "metadata": {
              "text": "4. PRECISE CELLULAR GENOMICS\nElucidating the molecular mechanisms that lead to beta cell\ndysfunction and T2D pathogenesis has been a major focus of diabetes\nresearch for decades. However, advances in single cell genomic\nproling techniques have led to greater understanding of non-beta cell\ntype transcriptional regulation and suggest that they may play\nimportant roles in hallmark features of beta cell insuf ciency and",
              "title": "2019 - (Epi)genomic heterogeneity of pancreatic islet function and failure in type 2 diabetes.pdf",
              "version": "v0",
              "chunk_order": 55,
              "document_id": "b9bc63a5-e366-5685-bd7a-4732a8eeffb7",
              "extraction_id": "7a2a9981-4096-5049-a717-3e69eb609777",
              "associatedQuery": "What are the mechanistic insights into the beta-cell failure pathways gleaned from recent single-cell RNA-sequencing studies?"
            }
          },
          {
            "id": "ab373b7e-8c0b-59d8-9408-3e09ac76761e",
            "score": 0.6964799761772156,
            "metadata": {
              "text": "53. Eliasson L, Esguerra JL (2014) Role of non-coding RNAs in pancreatic beta-cell development and physiology. Acta Physiol \n(Oxf) 211:273284\n 54. Ding GL, Wang FF, Shu J etal (2012) Transgenerational glucose \nintolerance with Igf2/H19 epigenetic alterations in mouse islet induced by intrauterine hyperglycemia. Diabetes 61:11331142\n 55. Ku GM, Kim H, Vaughn IW etal (2012) Research resource: RNA-Seq reveals unique features of the pancreatic beta-cell tran-scriptome. Mol Endocrinol 26:17831792",
              "title": "2018 - Lnc\u2011ing non\u2011coding RNAs with metabolism and diabetes roles.pdf",
              "version": "v0",
              "chunk_order": 112,
              "document_id": "019efefb-65db-55f5-a3a7-4f224473f51f",
              "extraction_id": "8bbfb009-87b7-54ae-8465-8796db8c271a",
              "associatedQuery": "What are the mechanistic insights into the beta-cell failure pathways gleaned from recent single-cell RNA-sequencing studies?"
            }
          },
          {
            "id": "a2adc65b-035b-568f-a0ae-9f7821ef45bc",
            "score": 0.6789516997962189,
            "metadata": {
              "text": "understand each cell type s genomic architecture and better charac-\nterize their roles in islet resilience and failure. Experimental manipu-\nlation of the regulatory elements and/or the target genes identi ed by\n(epi)genomic approaches described above and modeling the putativepathways and processes they implicate in human islet cell lines (e.g.,\nEndoC-\nbH1-H3) is essential to progress from correlation to causation.\nSimilarly, transitioning from themouse (C57BL/6) to multiple mouse",
              "title": "2019 - (Epi)genomic heterogeneity of pancreatic islet function and failure in type 2 diabetes.pdf",
              "version": "v0",
              "chunk_order": 97,
              "document_id": "b9bc63a5-e366-5685-bd7a-4732a8eeffb7",
              "extraction_id": "117cc1a5-d236-56b2-a69d-9c0a2fb9053d",
              "associatedQuery": "What are the mechanistic insights into the beta-cell failure pathways gleaned from recent single-cell RNA-sequencing studies?"
            }
          },
          {
            "id": "887e1f7e-5044-5be8-a506-588ca7afa004",
            "score": 0.6767109632492065,
            "metadata": {
              "text": "therapeutic pathways for beta cell regeneration. An integrative analysis of whole-exome andRNA-sequencing data was employed to extensively characterize the genomic and molecularlandscape of insulinomas relative to normal beta cells. Here, we show at the pathway levelthat the majority of the insulinomas display mutations, copy number variants and/or dys-regulation of epigenetic modifying genes, most prominently in the polycomb and trithoraxfamilies. Importantly, these processes are coupled to co-expression",
              "title": "2017 - Insights into beta cell regeneration for diabetes via integration of molecular landscapes in human insulinomas.pdf",
              "version": "v0",
              "chunk_order": 2,
              "document_id": "6cf1eb8d-a91e-58a2-b6f4-29653678d0d3",
              "extraction_id": "dee54186-e75e-5ed2-818d-cd6f4370b153",
              "associatedQuery": "What are the mechanistic insights into the beta-cell failure pathways gleaned from recent single-cell RNA-sequencing studies?"
            }
          },
          {
            "id": "4bfcfbd6-f45e-553d-a043-a12e7abeff61",
            "score": 0.6755803823471069,
            "metadata": {
              "text": "gesting that changes in alpha cell identity may ultimately lead to theirdysfunction. Analysis of normal and T2D islet single cells with\nsimultaneous RNA-seq and patch clamping (patch-seq) also revealed\nsubpopulations of alpha cells with varying enrichment for ER stressresponse genes (e.g., DDIT3, XBP1, PPP1R15A )[30]. Interestingly, this\ntranscriptomic heterogeneity was consistent in normal and T2D islets",
              "title": "2019 - (Epi)genomic heterogeneity of pancreatic islet function and failure in type 2 diabetes.pdf",
              "version": "v0",
              "chunk_order": 58,
              "document_id": "b9bc63a5-e366-5685-bd7a-4732a8eeffb7",
              "extraction_id": "7a2a9981-4096-5049-a717-3e69eb609777",
              "associatedQuery": "What are the mechanistic insights into the beta-cell failure pathways gleaned from recent single-cell RNA-sequencing studies?"
            }
          },
          {
            "id": "d32d6338-6cda-5f58-999d-2b4287ee4a77",
            "score": 0.668185830116272,
            "metadata": {
              "text": "RNA-seq analysis: a tutorial. Mol Syst Biol 15:e8746. https ://doi.org/10.15252 /msb.20188 746\nMa L, Zheng J (2018) Single-cell gene expression analysis reveals \n-cell dysfunction and deficit mechanisms in type 2 diabe-tes. BMC Bioinform 19:515. https ://doi.org/10.1186/s1285  \n9-018-2519-1\nMacaulay IC, Ponting CP, Voet T (2017) Single-cell multiom-\nics: multiple measurements from single cells. Trends Genet 33:155168. https ://doi.org/10.1016/j.tig.2016.12.003",
              "title": "2020 - Advances of single?cell genomics and epigenomics in human disease.pdf",
              "version": "v0",
              "chunk_order": 140,
              "document_id": "afe53f5a-3962-520f-be55-9df5bfdaad70",
              "extraction_id": "10e4029f-0324-55c9-8fe8-023a924d1732",
              "associatedQuery": "What are the mechanistic insights into the beta-cell failure pathways gleaned from recent single-cell RNA-sequencing studies?"
            }
          },
          {
            "id": "ef0b8934-2af1-5848-88f9-ff5a2e4f3cc1",
            "score": 0.6666788260127561,
            "metadata": {
              "text": "peak current. Prior single cell transcriptomic analyses have also notedsubpopulations of ER-stressed beta cells [31,32] which implicates the\ndysfunction of both alpha and beta cells in diabetes pathogenesis.Similarly, the integrity of beta and alpha cell functions seem to beReview\nS18MOLECULAR METABOLISM 27 (2019) S15 eS24/C2112019 Published by Elsevier GmbH. This is an open access article under the CC BY-NC-ND license ( http://creativecommons.org/licenses/by-nc-nd/4.0/ ).\nwww.molecularmetabolism.com",
              "title": "2019 - (Epi)genomic heterogeneity of pancreatic islet function and failure in type 2 diabetes.pdf",
              "version": "v0",
              "chunk_order": 60,
              "document_id": "b9bc63a5-e366-5685-bd7a-4732a8eeffb7",
              "extraction_id": "7a2a9981-4096-5049-a717-3e69eb609777",
              "associatedQuery": "What are the mechanistic insights into the beta-cell failure pathways gleaned from recent single-cell RNA-sequencing studies?"
            }
          },
          {
            "id": "46ed97d7-7b3e-5be2-a409-04a37d105ef2",
            "score": 0.6642862558364868,
            "metadata": {
              "text": "to understanding human development using single-cell tran-scriptomics. Development 144:1584. https ://doi.org/10.1242/dev.15045 8\nCamp JG, Wollny D, Treutlein B (2018) Single-cell genomics to guide \nhuman stem cell and tissue engineering. Nat Methods 15:661667. https ://doi.org/10.1038/s4159 2-018-0113-0\nCarrano AC, Mulas F, Zeng C, Sander M (2017) Interrogating islets \nin health and disease with single-cell technologies. Mol Metab \n6:9911001. https ://doi.org/10.1016/j.molme  t.2017.04.012",
              "title": "2020 - Advances of single?cell genomics and epigenomics in human disease.pdf",
              "version": "v0",
              "chunk_order": 117,
              "document_id": "afe53f5a-3962-520f-be55-9df5bfdaad70",
              "extraction_id": "f740892a-7817-58b0-bec4-8648086b2353",
              "associatedQuery": "What are the mechanistic insights into the beta-cell failure pathways gleaned from recent single-cell RNA-sequencing studies?"
            }
          },
          {
            "id": "f06bcc81-6ef9-5874-8ef9-6bcb3c34b0d0",
            "score": 0.6579534011469308,
            "metadata": {
              "text": "Advances ofsingle -cell genomics andepigenomics inhuman disease: whereare we now?  \n1 3\nBrissova etal. 2018; Tritschler etal. 2017). Moreover, an \nincrease in hyperglycaemia has been associated with a loss of beta-cell mass, function and organization and is the cell type most frequently studied for insulin resistance (Carrano etal. 2017; Lawlor etal. 2017b; Segerstolpe etal. 2016; \nTheis and Lickert 2019; Tritschler etal. 2017).\nNotably, single-cell transcriptome profiling has been",
              "title": "2020 - Advances of single?cell genomics and epigenomics in human disease.pdf",
              "version": "v0",
              "chunk_order": 62,
              "document_id": "afe53f5a-3962-520f-be55-9df5bfdaad70",
              "extraction_id": "65471d38-cd13-5de2-8c19-1eb72d24d6f5",
              "associatedQuery": "What are the mechanistic insights into the beta-cell failure pathways gleaned from recent single-cell RNA-sequencing studies?"
            }
          }
        ],
        "kg_search_results": null
      }
    }
  },
  "15": {
    "results": {
      "completion": {
        "id": "chatcmpl-AIHKMXGqZPQ6g67pYsMpeLJDEH7VZ",
        "choices": [
          {
            "finish_reason": "stop",
            "index": 0,
            "message": {
              "content": "The epigenetic landscape of key metabolic tissues shows several changes when comparing diabetic individuals to non-diabetic individuals:\n\n1. **DNA Methylation Changes**: In diabetic individuals, increased DNA methylation has been observed in the promoter region of the PPARGC1A gene in both islets and skeletal muscle [3]. This suggests a potential mechanism by which gene expression related to metabolism is altered in diabetes.\n\n2. **Histone Modifications**: There are disruptions in histone methylation patterns in diabetic states. While healthy individuals maintain stable histone methylation patterns, these can be disrupted in diabetes, indicating changes in the epigenome associated with inflammation and metabolic memory [2].\n\n3. **Impact on Gene Expression**: Epigenetic modifications, such as DNA methylation, have been linked to reduced expression of genes involved in diabetes and metabolism. Variations in DNA methylation have been noted near diabetes susceptibility genes and enhancers [6].\n\n4. **Tissue-Wide Epigenetic Changes**: Diabetes mellitus, characterized by high glucose stress, leads to epigenetic changes across most tissues impacted by the disease, including the cardiovascular system and immune system [7].\n\n5. **Adipose Tissue**: In subjects with type 2 diabetes, altered DNA methylation and differential expression of genes influencing metabolism and inflammation have been observed in adipose tissue [9].\n\nThese findings collectively suggest that diabetes is associated with specific epigenetic alterations across various metabolic tissues, which may contribute to the pathophysiology of the disease.",
              "role": "assistant"
            }
          }
        ],
        "created": 1728919874,
        "model": "gpt-4o-2024-08-06",
        "object": "chat.completion",
        "system_fingerprint": "fp_a20a4ee344",
        "usage": {
          "completion_tokens": 309,
          "prompt_tokens": 1337,
          "total_tokens": 1646
        }
      },
      "search_results": {
        "vector_search_results": [
          {
            "id": "d198d71b-34ac-5402-b166-b476c15e2eef",
            "score": 0.7108756648450297,
            "metadata": {
              "text": "To date, the overwhelming majority of studies including and assessing\ngenetic variation have pro led the steady state patterns of epigeneticmodi cations and gene expression in islets or their constituent cell\ntypes. Others have compared how these steady state measures differ\nbetween T2D and non-diabetic (ND) individuals [13,16,40 e44]. Sur-\nprisingly, these studies, especially transcriptome analyses, haveidenti ed only modest alterations despite clear phenotypic differences",
              "title": "2019 - (Epi)genomic heterogeneity of pancreatic islet function and failure in type 2 diabetes.pdf",
              "version": "v0",
              "chunk_order": 69,
              "document_id": "b9bc63a5-e366-5685-bd7a-4732a8eeffb7",
              "extraction_id": "043ee0bf-ec42-57dd-aa0e-4f4f5aac2437",
              "associatedQuery": "How does the epigenetic landscape of key metabolic tissues change in diabetic versus non-diabetic individuals?"
            }
          },
          {
            "id": "d032460f-1bab-5710-8a79-872651f9c36e",
            "score": 0.6921367416218828,
            "metadata": {
              "text": "T1D and resulting complications (99). These epig enomic profiling studies suggest that, while a 415 \nreasonably stable histone methylation pattern is maintained in  healthy individuals over time in a 416 \ncell-type specific setting, this pa ttern can be disrupted in a dis ease state. Moreover, they also 417 \nprovide a glimpse of the inflammatory cell epig enome under the diabetic state and suggest that 418 \nnew information about diabetes, its complicatio ns and metabolic memory can be obtained by 419",
              "title": "2010 - The Role of Epigenetics in the Pathology of Diabetic Complications.pdf",
              "version": "v0",
              "chunk_order": 88,
              "document_id": "766edfd5-4756-51bf-b636-c94b041d030c",
              "extraction_id": "efbaf00f-0cb1-531f-a9fd-2844670ec92c",
              "associatedQuery": "How does the epigenetic landscape of key metabolic tissues change in diabetic versus non-diabetic individuals?"
            }
          },
          {
            "id": "a6efcd76-42f1-5d4c-bcfa-f5e63ac9eb4c",
            "score": 0.6888615674459012,
            "metadata": {
              "text": "hyperglycaemia, epigenetic changes have also been noted in\nother experimental settings of hyperglycaemia. For example,\nincreased DNA methylation has been described for the promoter\nregion of the peroxisome proliferator-activated receptor-\ng(PPAR g)\ncoactivator-1 agene (PPARGC1A) in diabetic islets ( Ling et al., 2008 ).\nSimilar hypermethylation in the promoter region of the PPARGC1A\ngene has been noted in the skeletal muscle from diabetic patients,",
              "title": "2015 - Maternal diabetes, gestational diabetes and the role of epigenetics in their long term effects on offspring.pdf",
              "version": "v0",
              "chunk_order": 116,
              "document_id": "3e92bd8e-fbf7-5bc4-9395-0a6dd0b0934e",
              "extraction_id": "daf2d7fd-e789-5ceb-9984-d95656b5dd91",
              "associatedQuery": "How does the epigenetic landscape of key metabolic tissues change in diabetic versus non-diabetic individuals?"
            }
          },
          {
            "id": "29bdfc8d-cb06-5ce5-8db6-f92b2f106b7d",
            "score": 0.6861002819418571,
            "metadata": {
              "text": "and correlated with mitochondrial content ( Barr /C18es et al., 2009 ).\nEpigenetic changes have also been suggested to be responsible forthe legacy effect of reduced risk of vascular complications after a\nperiod of sustained tight glucose control, or metabolic memory of\ntransient hyperglycaemia and increased risk of diabetic vascular\ninjury ( Pirola et al., 2010 ). Histone methylation variations have\nbeen noted in monocytes cultured in high glucose, as well as blood",
              "title": "2015 - Maternal diabetes, gestational diabetes and the role of epigenetics in their long term effects on offspring.pdf",
              "version": "v0",
              "chunk_order": 117,
              "document_id": "3e92bd8e-fbf7-5bc4-9395-0a6dd0b0934e",
              "extraction_id": "daf2d7fd-e789-5ceb-9984-d95656b5dd91",
              "associatedQuery": "How does the epigenetic landscape of key metabolic tissues change in diabetic versus non-diabetic individuals?"
            }
          },
          {
            "id": "cf4f3239-dd62-5eef-b5fc-85f4780e3f48",
            "score": 0.6848712751346079,
            "metadata": {
              "text": "Epigenetic Mechanisms in Diabetic Complications     17 \nInterestingly, the sirtuin (SIRT) family of deacetylases, specifically SIRT1, has been found to 360 \nregulate several factors involved in metabolism, adipogenesis a nd insulin secretion (86). HATs 361 \nand HDACs can also modulate NF- B transcriptional activity (4, 44) resulting in changes in 362",
              "title": "2010 - The Role of Epigenetics in the Pathology of Diabetic Complications.pdf",
              "version": "v0",
              "chunk_order": 76,
              "document_id": "766edfd5-4756-51bf-b636-c94b041d030c",
              "extraction_id": "41ac576d-b850-5ee8-9753-ba9b060ba798",
              "associatedQuery": "How does the epigenetic landscape of key metabolic tissues change in diabetic versus non-diabetic individuals?"
            }
          },
          {
            "id": "1c0b9205-340f-568f-990f-785c66154732",
            "score": 0.6813801120889132,
            "metadata": {
              "text": "ing that environment and diet may influence epigenetic mod-ifications that predispose individuals to diabetes [ 46]. Aber-\nrant DNAme has also been reported in the reduced expression\nof genes involved in diabetes and metabolism, and DNAme\nvariations have also been noted near diabetes susceptibility\ngenes and enhancers [ 15,47].\nGenomic DNA from diabetic patients with nephropa-\nthy relative to those without displayed differential meth-\nylation at several genes, including UNC13B , which had",
              "title": "2015 - Epigenetic mechanisms in diabetic complications and metabolic memory.pdf",
              "version": "v0",
              "chunk_order": 40,
              "document_id": "470f1f94-792d-5273-a88f-7e06084951c5",
              "extraction_id": "44d96546-84c3-51f1-85f9-22790a91d105",
              "associatedQuery": "How does the epigenetic landscape of key metabolic tissues change in diabetic versus non-diabetic individuals?"
            }
          },
          {
            "id": "64f12ea8-a441-5fb5-a80e-1acbfb596a1e",
            "score": 0.680983085573729,
            "metadata": {
              "text": "of diabetes mellitus on the body is a high glucose stressed condition, altering substrate metabolism and causing systemic inflammation [60]. Due to this environmental change, researchers have shown how epigenetic changes occur across most, if not all, tissues that are impacted by diabetes mellitus [49, 61].\nIn the cardiovascular system, the heart, circulatory \nsystem, and regulating immune system are all tran\n-",
              "title": "2019 - Machine-learning to stratify diabetic patients using novel cardiac biomarkers and integrative genomics.pdf",
              "version": "v0",
              "chunk_order": 144,
              "document_id": "332ac2ec-accc-5370-a4d2-6fec9ce7e072",
              "extraction_id": "aff84b9e-3855-5960-accd-dcac6b362346",
              "associatedQuery": "How does the epigenetic landscape of key metabolic tissues change in diabetic versus non-diabetic individuals?"
            }
          },
          {
            "id": "8f447303-efd0-5564-9213-2cdc8fce12dd",
            "score": 0.6785546136763935,
            "metadata": {
              "text": "nephropathy. Exp. Physiol.  98, 934945 (2013).\n48. Reddy, M.A., Tak Park, J. & Natarajan, R. \nEpigenetic modifications in the pathogenesis \nofdiabetic nephropathy. Semin. Nephrol.  33, \n341353 (2013).\n49. Li, S.L. etal. Enhanced proatherogenic \nresponses in macrophages and vascular smooth \nmuscle cells derived from diabetic db/db  mice. \nDiabetes  55, 26112619 (2006).\n50. El-Osta, A. etal. Transient high glucose causes \npersistent epigenetic changes and altered gene",
              "title": "2014 - Diabetic nephropathy\u2014emerging epigenetic mechanisms.pdf",
              "version": "v0",
              "chunk_order": 156,
              "document_id": "be05127e-1be8-5573-b571-51a11c3b2be2",
              "extraction_id": "cbbe696b-8541-537a-ac5f-77b82cdb8201",
              "associatedQuery": "How does the epigenetic landscape of key metabolic tissues change in diabetic versus non-diabetic individuals?"
            }
          },
          {
            "id": "7aafd7e0-2446-57a4-bb1e-1f77b4f4559e",
            "score": 0.6759020283280606,
            "metadata": {
              "text": "exhibit decreased plasticity of genome-wide muscle DNA methylation by high-fatoverfeeding. Diabetologia 2014;57:1154-1158.\n53. Nilsson E, Jansson PA, Perfilyev A, et al. Altered DNA methylation and differential\nexpression of genes influencing metabolism and inflammation in adipose tissue\nfrom subjects with type 2 diabetes. Diabetes 2014;63:2962-2976.\n54. Aslibekyan S, Demerath EW, Mendelson M, et al. Epigenome-wide study identifies",
              "title": "2016 - NIH working group report using genomic information to guide weight management  From universal.pdf",
              "version": "v0",
              "chunk_order": 125,
              "document_id": "0ee28c8a-3618-559e-be0a-30f2579a0d1f",
              "extraction_id": "68f800e3-8a0c-50ee-ba8b-ceb361f14fa5",
              "associatedQuery": "How does the epigenetic landscape of key metabolic tissues change in diabetic versus non-diabetic individuals?"
            }
          },
          {
            "id": "cf775ed3-457e-52a7-b54b-5c1b5c179819",
            "score": 0.6755989387274433,
            "metadata": {
              "text": "etal. Hyperglycemia induces a dynamic cooperativity of histone methylase \nand demethylase enzymes associated with gene-activating epigenetic marks that coexist on the lysine tail. Diabetes  (2009) 58:122936. doi:10.2337/\ndb08-1666 \n111. Keating S, Plutzky J, El-Osta A. Epigenetic changes in diabetic and cardio-vascular risk. Circ Res  (2016) 118:170622. doi:10.1161/CIRCRESAHA.  \n116.306819 \n112. Paneni F, Volpe M, Lscher TF, Cosentino F. SIRT1, p66(Shc), and Set7/9 in",
              "title": "2018 - Type 2 Diabetes Mellitus and Cardiovascular Disease Genetic and Epigenetic Links.pdf",
              "version": "v0",
              "chunk_order": 150,
              "document_id": "3e82a2e5-4b2c-59c0-99cd-f3b06d8dabf2",
              "extraction_id": "9e010393-b98f-5f6c-a62d-fc0646ba8667",
              "associatedQuery": "How does the epigenetic landscape of key metabolic tissues change in diabetic versus non-diabetic individuals?"
            }
          }
        ],
        "kg_search_results": null
      }
    }
  },
  "16": {
    "results": {
      "completion": {
        "id": "chatcmpl-AIHKSpSdna9OyEUtDVRTMkIkEtBS5",
        "choices": [
          {
            "finish_reason": "stop",
            "index": 0,
            "message": {
              "content": "Recent advancements in leveraging CRISPR-based approaches to correct monogenic forms of diabetes in vivo include the use of CRISPR-mediated homology-directed repair (HDR) to correct specific genetic mutations associated with diabetes. For instance, CRISPR technology has been used to correct point mutations in patient-derived induced pluripotent stem cells (iPSCs) targeting diabetes-related gene defects. The most efficient method employed in iPSCs is CRISPR/Cas9-based HDR, where a Cas9-mediated cut is generated adjacent to the site of interest, and a homologous donor template with the intended nucleotide change is recombined by HDR [9]. Additionally, there has been a successful correction of a variant in the Wolfram syndrome 1 (WFS1) gene using CRISPR-mediated HDR, which improved insulin secretion in iPSC-differentiated beta-like cells [3]. These advancements highlight the potential of CRISPR-based genome editing to correct monogenic forms of diabetes by targeting specific genetic mutations in vivo.",
              "role": "assistant"
            }
          }
        ],
        "created": 1728919880,
        "model": "gpt-4o-2024-08-06",
        "object": "chat.completion",
        "system_fingerprint": "fp_a20a4ee344",
        "usage": {
          "completion_tokens": 204,
          "prompt_tokens": 1367,
          "total_tokens": 1571
        }
      },
      "search_results": {
        "vector_search_results": [
          {
            "id": "f42c0f84-d2a8-5bf9-89c2-3dd182bfb235",
            "score": 0.72869388632142,
            "metadata": {
              "text": "A variety of cellular and animal models have been developed and applied\nover the past few years to experimentally manipulate cis-regulatory\nelements and their target gene function as it related to beta cell/isletfunction, glucose homeostasis, and T2D pathogenesis. CRISPR/Cas9 hasrevolutionized our ability to modify genomes and epigenomes almost at\nwill. Unsurprisingly, CRISPR (epi)genome editing tools can and have\nbeen used to target putative T2D target genes [54] orcis-REs[55] in beta",
              "title": "2019 - (Epi)genomic heterogeneity of pancreatic islet function and failure in type 2 diabetes.pdf",
              "version": "v0",
              "chunk_order": 79,
              "document_id": "b9bc63a5-e366-5685-bd7a-4732a8eeffb7",
              "extraction_id": "57736895-897e-54e5-a735-aadcbd77cb63",
              "associatedQuery": "What recent advancements have been made in leveraging CRISPR-based approaches to correct monogenic forms of diabetes in vivo?"
            }
          },
          {
            "id": "2af36592-3e59-583c-a9c7-d612175f4afc",
            "score": 0.723567528937313,
            "metadata": {
              "text": "(276279). Through CRISPR-mediated HDR and base editing, it\nis possible to correct the vast majority of genetic variants, if notall. Conversion of GWAS-identi ed non-coding variants has not\nbeen conducted/documented in the diabetes eld, but it seems\ninevitable that such work will be carried out in the near futureHu et al. Genome Editing of Pancreatic Beta Cells\nFrontiers in Endocrinology | www.frontiersin.org October 2020 | Volume 11 | Article 576632 11",
              "title": "2020 - Functional Genomics in Pancreatic \u03b2 Cells Recent Advances in Gene Deletion and Genome Editing Technologies for Diabetes Research.pdf",
              "version": "v0",
              "chunk_order": 140,
              "document_id": "51350055-d53c-5692-ab53-337b8a8bafd6",
              "extraction_id": "ab61a462-21d3-50dc-afb3-3e1cdeb15b1f",
              "associatedQuery": "What recent advancements have been made in leveraging CRISPR-based approaches to correct monogenic forms of diabetes in vivo?"
            }
          },
          {
            "id": "75b937b2-1e0b-5d63-b542-618ad91bbd1f",
            "score": 0.7071702516529192,
            "metadata": {
              "text": "Cas9 editing to restore insulin production in differentiated iPSCcells that mimicked neonatal diabetes ( 251,252). Likewise, Shi\net al. converted a patient-speci c mutation in GATA6 gene and\nshowed that the mutation involved (GATA6\nR456C) has a similar\neffect to GATA6 knockout ( 21). Most recently, correction of a\nvariant in the Wolfram syndrome 1 ( WFS1 ) gene by CRISPR-\nmediated HDR improved insulin secretion in iPSC-differentiatedb-like cells ( 253). Studies on GWAS identi ed genetic variants",
              "title": "2020 - Functional Genomics in Pancreatic \u03b2 Cells Recent Advances in Gene Deletion and Genome Editing Technologies for Diabetes Research.pdf",
              "version": "v0",
              "chunk_order": 127,
              "document_id": "51350055-d53c-5692-ab53-337b8a8bafd6",
              "extraction_id": "ab61a462-21d3-50dc-afb3-3e1cdeb15b1f",
              "associatedQuery": "What recent advancements have been made in leveraging CRISPR-based approaches to correct monogenic forms of diabetes in vivo?"
            }
          },
          {
            "id": "1f114642-3f77-5346-89e8-394c433f66ff",
            "score": 0.6914571935806998,
            "metadata": {
              "text": "in response to various stimuli including glucose aftertransplantation in an immunocompromised mouse model\n(230,231). However, the use of iPSC is controversial and there\nare some concerns over genetic and epigenetic variations iniPSCs which might affect cell function after differentiation ( 275).\nManipulation of hESC/iPSC cells via CRISPR-Cas9\ntechnology provides a platform for the correction of genomic\nmutations not only in diabetes but in other disease elds as well",
              "title": "2020 - Functional Genomics in Pancreatic \u03b2 Cells Recent Advances in Gene Deletion and Genome Editing Technologies for Diabetes Research.pdf",
              "version": "v0",
              "chunk_order": 139,
              "document_id": "51350055-d53c-5692-ab53-337b8a8bafd6",
              "extraction_id": "ab61a462-21d3-50dc-afb3-3e1cdeb15b1f",
              "associatedQuery": "What recent advancements have been made in leveraging CRISPR-based approaches to correct monogenic forms of diabetes in vivo?"
            }
          },
          {
            "id": "df30dab3-a490-5497-a079-2741f9039f87",
            "score": 0.6890247278824299,
            "metadata": {
              "text": "hPSCs [48,49] for correcting the COL7A1 [50] anda1-antitrypsin\ngenes [51]. Given the superior cutting ef ciency, CRISPR/Cas9 is\nincreasingly becoming the favored choice for genome editing inhPSCs [16,52] .\n3.2. Employing hPSCs and genome editing tools to study diabetes\nand metabolic syndromes\nIn general, the strategy to carry out in vitro disease modeling of dia-",
              "title": "2016 - Dissecting diabetes metabolic disease.pdf",
              "version": "v0",
              "chunk_order": 40,
              "document_id": "eee2f79d-e093-52fb-871a-798fd859235e",
              "extraction_id": "998a92ba-e7fc-5553-b629-7b5797fbfafe",
              "associatedQuery": "What recent advancements have been made in leveraging CRISPR-based approaches to correct monogenic forms of diabetes in vivo?"
            }
          },
          {
            "id": "eadf2320-de70-5499-ade0-7aa9930ac091",
            "score": 0.6886285953896413,
            "metadata": {
              "text": "Due to its simplicity and adaptability, CRISPR has rapidly\nbecome the most popular genome editing tool available for the\nmammalian genome ( 50,63). Because NHEJ DNA repair often\nintroduces unwanted indels at the Cas9 cutting site, CRISPR hasbeen used to knock-out genes by introducing frameshiftmutations, resulting in protein depletion ( 156,157). In the\ndiabetes eld, CRISPR has also been adopted to study several\ngenes in bcell lines and in human ES-derived bcells ( 21,151,",
              "title": "2020 - Functional Genomics in Pancreatic \u03b2 Cells Recent Advances in Gene Deletion and Genome Editing Technologies for Diabetes Research.pdf",
              "version": "v0",
              "chunk_order": 76,
              "document_id": "51350055-d53c-5692-ab53-337b8a8bafd6",
              "extraction_id": "fe5bf2df-2eda-5ef0-8aad-79bbc5b898d6",
              "associatedQuery": "What recent advancements have been made in leveraging CRISPR-based approaches to correct monogenic forms of diabetes in vivo?"
            }
          },
          {
            "id": "57b9550d-0258-5a87-be57-976f471e5763",
            "score": 0.685957871193632,
            "metadata": {
              "text": "RNP and single strand edDNA (ssDNA) donor which carriesdesired changes such as insertion of loxP site ( 255,259265).\nUsing CRISPR-Cas9, leptin and leptin receptor knockout mice\nhave been established as tools in diabetes and obesity research ( 160,255,256). Knock-in mouse models have also been established via\nHDR to achieve cell-speci c deletion of the gene ( 266).\nGenome Editing: Clinical Application\nin Diabetes\nAn important goal in genetic research is to identify the genetic",
              "title": "2020 - Functional Genomics in Pancreatic \u03b2 Cells Recent Advances in Gene Deletion and Genome Editing Technologies for Diabetes Research.pdf",
              "version": "v0",
              "chunk_order": 133,
              "document_id": "51350055-d53c-5692-ab53-337b8a8bafd6",
              "extraction_id": "ab61a462-21d3-50dc-afb3-3e1cdeb15b1f",
              "associatedQuery": "What recent advancements have been made in leveraging CRISPR-based approaches to correct monogenic forms of diabetes in vivo?"
            }
          },
          {
            "id": "1859f32b-8f5c-5c3c-9f4d-54193d37645d",
            "score": 0.6847404241561938,
            "metadata": {
              "text": "to how CRISPR/Cas9 technology may nd clinical application in patients with diabetes.\nKeywords: genome editing, beta cell, genome-wide association studies, maturity onset of diabetes of the young,\nstem cells, mouse models\nINTRODUCTION\nType 2 diabetes (T2D) affects an estimated 425 million people worldwide, a number predicted to rise\nto 629 million by 2045 ( 1). The disease usually involves insulin resistance but is ultimately the result",
              "title": "2020 - Functional Genomics in Pancreatic \u03b2 Cells Recent Advances in Gene Deletion and Genome Editing Technologies for Diabetes Research.pdf",
              "version": "v0",
              "chunk_order": 3,
              "document_id": "51350055-d53c-5692-ab53-337b8a8bafd6",
              "extraction_id": "5f8a0ddd-a0c7-5151-9b6a-e0980bb94aa6",
              "associatedQuery": "What recent advancements have been made in leveraging CRISPR-based approaches to correct monogenic forms of diabetes in vivo?"
            }
          },
          {
            "id": "99ccc9a2-865f-5d11-9b08-b26261d02fc9",
            "score": 0.682744264602666,
            "metadata": {
              "text": "samples ( 236).\nCRISPR technology has been used recently to correct point\nmutations in patient-derived iPSCs to target diabetes-relatedgene defects. To date, the most ef cient method used in iPSC\nis CRISPR/Cas9-based homology-directed repair (HDR). Here, a\nCas9-mediated cut is generated adjacent to the site of interest. A\nhomologous donor template with the intended nucleotidechange containing silent mutations in the gRNA sequence(167) can then be recombined by HDR. This approach has",
              "title": "2020 - Functional Genomics in Pancreatic \u03b2 Cells Recent Advances in Gene Deletion and Genome Editing Technologies for Diabetes Research.pdf",
              "version": "v0",
              "chunk_order": 117,
              "document_id": "51350055-d53c-5692-ab53-337b8a8bafd6",
              "extraction_id": "0a3e3095-4789-505a-96b7-123a05078e95",
              "associatedQuery": "What recent advancements have been made in leveraging CRISPR-based approaches to correct monogenic forms of diabetes in vivo?"
            }
          },
          {
            "id": "83053df5-47ac-59da-9c30-69740a64372d",
            "score": 0.679858367434644,
            "metadata": {
              "text": "free IPSCs from Human Pancreatic Cells Using the CRISPR-Cas9 System.\nJ Vis Exp JoVE (2017). doi: 10.3791/56260\n277. Millette K, Georgia S. Gene Editing and Human Pluripotent Stem Cells:\nTools for Advancing Diabetes Disease Modeling and Beta-Cell Development.\nCurr Diabetes Rep (2017) 17:116. doi: 10.1007/s11892-017-0947-3Hu et al. Genome Editing of Pancreatic Beta Cells\nFrontiers in Endocrinology | www.frontiersin.org October 2020 | Volume 11 | Article 576632 19",
              "title": "2020 - Functional Genomics in Pancreatic \u03b2 Cells Recent Advances in Gene Deletion and Genome Editing Technologies for Diabetes Research.pdf",
              "version": "v0",
              "chunk_order": 280,
              "document_id": "51350055-d53c-5692-ab53-337b8a8bafd6",
              "extraction_id": "a36cee80-5961-55e5-8ea4-8d4e1bc501a9",
              "associatedQuery": "What recent advancements have been made in leveraging CRISPR-based approaches to correct monogenic forms of diabetes in vivo?"
            }
          }
        ],
        "kg_search_results": null
      }
    }
  },
  "17": {
    "results": {
      "completion": {
        "id": "chatcmpl-AIHKYN37xsXdGCjQ8Ms8PgKZ10CIR",
        "choices": [
          {
            "finish_reason": "stop",
            "index": 0,
            "message": {
              "content": "Genome-wide association studies (GWAS) integrate with multi-omics data to elucidate the complex genetic architectures of diabetes by combining genetic, epigenetic, transcriptomic, and phenotypic information. This integration helps identify genes and novel metabolic pathway targets that are crucial for understanding mechanistic relationships with insulin resistance and pancreatic islet failure [1]. Additionally, complementary systems-level data, such as protein-protein interactions and gene expression, provide insights into the mechanisms underlying the pathogenesis of complex traits like type 2 diabetes (T2D) [8]. This multi-omics approach allows for a more comprehensive understanding of the genome-to-phenome correlation in T2D, which is essential for examining the disease's complex genetic architecture [9].",
              "role": "assistant"
            }
          }
        ],
        "created": 1728919886,
        "model": "gpt-4o-2024-08-06",
        "object": "chat.completion",
        "system_fingerprint": "fp_6b68a8204b",
        "usage": {
          "completion_tokens": 149,
          "prompt_tokens": 1144,
          "total_tokens": 1293
        }
      },
      "search_results": {
        "vector_search_results": [
          {
            "id": "7302a27a-6e56-589d-a579-635f25fc46a3",
            "score": 0.7181355953216553,
            "metadata": {
              "text": "The integration of genetic, epigenetic, transcriptomic and phenotypic information allows to identify\ngenes and novel metabolic pathway targets that deserve further attention to elucidate mechanistic\nrelationships with insulin resistance and pancreatic islet failure. Although the GWASs and EWASs\nshed light onto (epi)genomic landscape of T2D to a great extent, these methods have still explicit\nlimitations to conquer, such as sample size, small effect size, low allele frequency, genetic heterogeneity",
              "title": "2018 - High-Throughput Approaches onto Uncover (Epi)Genomic Architecture of Type 2 Diabetes.pdf",
              "version": "v0",
              "chunk_order": 121,
              "document_id": "1cb0c4ac-c1fe-55c2-919c-52cd5018c00d",
              "extraction_id": "978df5a8-acb4-53d3-b351-66a3bc613c78",
              "associatedQuery": "How do genome-wide association studies (GWAS) integrate with multi-omics data to elucidate the complex genetic architectures of diabetes?"
            }
          },
          {
            "id": "4d780759-36bb-5295-a63a-16dab6aeab8c",
            "score": 0.7155009508132935,
            "metadata": {
              "text": "map of the human genome, spurred larger multi-institutional programs (e.g., 1000 Genomes Projects,\nEncyclopedia of DNA Elements [ENCODE], and Roadmap Epigenomics), that have the goal of tracking\ngenomic and epigenomic changes across multiple populations [ 8]. Aforementioned studies enabled\nGWASs for complex diseases such as T2D. DNA amplication, Sanger sequencing, and microarray\nstudies have shed light on the genetics of diabetes but have only provided a limited amount of data. An",
              "title": "2018 - High-Throughput Approaches onto Uncover (Epi)Genomic Architecture of Type 2 Diabetes.pdf",
              "version": "v0",
              "chunk_order": 10,
              "document_id": "1cb0c4ac-c1fe-55c2-919c-52cd5018c00d",
              "extraction_id": "aba850e8-8c0d-5256-b2ba-fa1dfc221114",
              "associatedQuery": "How do genome-wide association studies (GWAS) integrate with multi-omics data to elucidate the complex genetic architectures of diabetes?"
            }
          },
          {
            "id": "ac4d8521-b492-59b5-9978-891f5a5ce0c5",
            "score": 0.7094894859042614,
            "metadata": {
              "text": "Abstract\nWhile genome-wide association studies (GWAS) and candidate gene approaches have identified many genetic variants that\ncontribute to disease risk as main effects, the impact of genotype by environment (GxE) interactions remains rather under-\nsurveyed. To explore the importance of GxE interactions for diabetes-related traits, a tool for Genome-wide Complex Trait",
              "title": "2013 - Genome-Wide Contribution of Genotype by Environment Interaction.pdf",
              "version": "v0",
              "chunk_order": 2,
              "document_id": "8c310d76-0a3b-574c-9859-859258870ee5",
              "extraction_id": "f3975a2c-8a66-582e-a4b8-868b1f4722d4",
              "associatedQuery": "How do genome-wide association studies (GWAS) integrate with multi-omics data to elucidate the complex genetic architectures of diabetes?"
            }
          },
          {
            "id": "81fb2df2-4154-58a7-b217-b07153a6c921",
            "score": 0.694022536277771,
            "metadata": {
              "text": "The advancement that has taken place in Genome-Wide Association Studies (GWAS)\nholds tremendous information related to various gene patterns associated with divergent\nillnesses that are complex and challenging to perform reductive analysis from a single locus,\nas stated by Cho Ys [6] and Coron [7]. The evolution of GWAS has focused on integrating\ndata related to multi-locus across the gene that would assist in predicting complex illnesses",
              "title": "2022 - Using Recurrent Neural Networks for Predicting Type-2 Diabetes from Genomic and Tabular Data.pdf",
              "version": "v0",
              "chunk_order": 16,
              "document_id": "be0e50e0-3de8-53c5-8126-a0b618647f80",
              "extraction_id": "3c30b33b-8928-5cee-9c37-c70642fff75c",
              "associatedQuery": "How do genome-wide association studies (GWAS) integrate with multi-omics data to elucidate the complex genetic architectures of diabetes?"
            }
          },
          {
            "id": "263ea999-9662-5518-a606-939f69d09f90",
            "score": 0.688255786895752,
            "metadata": {
              "text": "1. Genome-wide association studies (GW AS) have made considerable progress in identifying genetic risk \nfactors and in providing evidence for more in-depth understanding of the biological and pathological pathways underlying T2D. A recent study performed a meta-analysis of T2D across 32 GW AS of European ancestry par -\nticipants and identified 243 genome-wide significant loci (403 distinct genetic variants) associated with T2D risk",
              "title": "2020 - Genome-wide association analysis of type 2 diabetes in the EPIC-InterAct study.pdf",
              "version": "v0",
              "chunk_order": 4,
              "document_id": "5dd7d700-03db-595d-b1a5-beca77f9579e",
              "extraction_id": "2c601441-443d-5c47-95bb-6343378dd5dc",
              "associatedQuery": "How do genome-wide association studies (GWAS) integrate with multi-omics data to elucidate the complex genetic architectures of diabetes?"
            }
          },
          {
            "id": "c807fc8b-966e-56a9-91ce-07b9baf940d9",
            "score": 0.6872100234031677,
            "metadata": {
              "text": "1. Introduction\nGenome wide association studies (GWAS) of type 2 diabetes\nmellitus and relevant endophenotypes have shed new light on the\ncomplex etiology of the disease and underscored the multiple\nmolecular mechanisms involved in the pathogenic processes\nleading to hyperglycemia [1]. Even though these studies have\nsuccessfully mapped many diabetes risk genetic loci that could not\nbe detected by linkage analysis, the risk single nucleotide poly-",
              "title": "2017 - Genomic regulation of type 2 diabetes endophenotypes Contribution.pdf",
              "version": "v0",
              "chunk_order": 12,
              "document_id": "fef1ae33-b3af-50ea-909c-f1b57f7fe981",
              "extraction_id": "3dc37987-5204-5414-92ee-9d97af221261",
              "associatedQuery": "How do genome-wide association studies (GWAS) integrate with multi-omics data to elucidate the complex genetic architectures of diabetes?"
            }
          },
          {
            "id": "ef027493-6063-5abd-9ee7-0c9a37379317",
            "score": 0.6856701970100403,
            "metadata": {
              "text": "how they will continue to expand our understanding of the\ngenetic risk factors and underlying biology of diabetes.\nKeywords Genotyping .Genome-wide association .\nSequencing .Imputation .Exome .Genome .\nFine-mapping .Diabetes .Quantitative traits .Metabochip .\nSingle nucleotide polymorphism\nIntroduction\nGWA studies have made progress toward understanding the\ninherited basis of type 1 and type 2 diabetes by detecting\ndisease-associated DNA variants, usually with allele fre-",
              "title": "2012 - What will Diabetes Genomes Tell Us.pdf",
              "version": "v0",
              "chunk_order": 2,
              "document_id": "38b3b7ab-d13e-5986-9a3a-54abe8a3e1e9",
              "extraction_id": "50a110f8-e91d-5985-9fe9-62a373a58c9d",
              "associatedQuery": "How do genome-wide association studies (GWAS) integrate with multi-omics data to elucidate the complex genetic architectures of diabetes?"
            }
          },
          {
            "id": "869d46b4-e379-54f8-bd71-143d9f31fa93",
            "score": 0.6849029251990971,
            "metadata": {
              "text": "complementary systems level data such as that related to protein-\nprotein interactions and to and gene expression can provideinsights into the mechanisms underlying pathogenesis of complextraits [2224]. Here, we have combined these approaches towarddeciphering genome to phenome correlation in T2D ( Figure 1 ).\nGiven that T2D GWAS genes do not directly relate to disease",
              "title": "2013 - Systems Biology Approach Reveals Genome to Phenome Correlation in Type 2 Diabetes.pdf",
              "version": "v0",
              "chunk_order": 24,
              "document_id": "ea7c2799-c259-5d0e-b40b-ecebe0a9fc9f",
              "extraction_id": "8dd91a24-2ac7-57b3-9cb3-f8ac74b1885c",
              "associatedQuery": "How do genome-wide association studies (GWAS) integrate with multi-omics data to elucidate the complex genetic architectures of diabetes?"
            }
          },
          {
            "id": "b92b959c-2f31-5177-8a21-627f3ee81b6c",
            "score": 0.6839315483610912,
            "metadata": {
              "text": "phenotypes [2,6]. The recently accomplished deep sequencing of\nhuman exomes has indeed suggested that rare variations\ncontribute substantially to human phenotypic variation and\ndisease susceptibility [73]. Availability of post-GWASs era data\nfor T2D will be crucial in examining genome to phenomecorrelation in greater details. Emerging methods in pathway-wide\nanalysis and integrative network based analysis of genetic\nassociation data in complex disorders will further help accelerate",
              "title": "2013 - Systems Biology Approach Reveals Genome to Phenome Correlation in Type 2 Diabetes.pdf",
              "version": "v0",
              "chunk_order": 90,
              "document_id": "ea7c2799-c259-5d0e-b40b-ecebe0a9fc9f",
              "extraction_id": "f6926cab-e00d-5972-a815-2ecc9f8c35d5",
              "associatedQuery": "How do genome-wide association studies (GWAS) integrate with multi-omics data to elucidate the complex genetic architectures of diabetes?"
            }
          },
          {
            "id": "7fd80e84-ec0c-564c-8e8b-278b8c622abb",
            "score": 0.6818853210279167,
            "metadata": {
              "text": "Abstract\nGenome-wide association studies (GWASs) have discovered association of several loci with Type 2 diabetes (T2D), a\ncommon complex disease characterized by impaired insulin secretion by pancreatic bcells and insulin signaling in target\ntissues. However, effect of genetic risk variants on continuous glycemic measures in nondiabetic subjects mainly elucidatesperturbation of insulin secretion. Also, the disease associated genes do not clearly converge on functional categories",
              "title": "2013 - Systems Biology Approach Reveals Genome to Phenome Correlation in Type 2 Diabetes.pdf",
              "version": "v0",
              "chunk_order": 1,
              "document_id": "ea7c2799-c259-5d0e-b40b-ecebe0a9fc9f",
              "extraction_id": "9369222f-e125-58c0-8f2b-cf5daa867f77",
              "associatedQuery": "How do genome-wide association studies (GWAS) integrate with multi-omics data to elucidate the complex genetic architectures of diabetes?"
            }
          }
        ],
        "kg_search_results": null
      }
    }
  },
  "18": {
    "results": {
      "completion": {
        "id": "chatcmpl-AIHKdF53rZo0tRRSpImOeG4mHUbkt",
        "choices": [
          {
            "finish_reason": "stop",
            "index": 0,
            "message": {
              "content": "Genomic imprinting has a significant impact on the susceptibility and progression of diabetes. Imprinting can influence the expression of genes involved in metabolic processes, which are crucial in the development of diabetes. For instance, changes in imprinting status at specific loci, such as the KCNQ1 locus, have been linked to type 2 diabetes susceptibility, indicating that temporal changes in imprinting can affect the function of pancreatic islets and contribute to diabetes risk [6]. Additionally, imprinting defects have been associated with specific forms of diabetes, such as transient neonatal diabetes, suggesting that imprinted genes play a role in the disease's onset and progression [4]. Furthermore, the effects of maternal diabetes on the offspring's epigenome, including alterations in DNA methylation profiles, highlight the role of imprinting in the intergenerational transmission of diabetes risk [3], [7]. These epigenetic changes can lead to a permanent programming of the developing offspring, increasing the risk of diabetes in subsequent generations [8]. Overall, genomic imprinting is a critical factor in understanding the genetic and epigenetic mechanisms underlying diabetes susceptibility and progression.",
              "role": "assistant"
            }
          }
        ],
        "created": 1728919891,
        "model": "gpt-4o-2024-08-06",
        "object": "chat.completion",
        "system_fingerprint": "fp_6b68a8204b",
        "usage": {
          "completion_tokens": 225,
          "prompt_tokens": 1415,
          "total_tokens": 1640
        }
      },
      "search_results": {
        "vector_search_results": [
          {
            "id": "10776283-4b6d-544c-89ac-0225c65bec1e",
            "score": 0.7016704260307309,
            "metadata": {
              "text": "maternal diabetes reduces the precision of gene regulation in exposed individuals. Loss of precision in embry-onic gene regulation may include changes to the epigenome via deregulated expression of chromatin-modify-ing factors. Unraveling the mechanisms underlying such epigenetic modications in diabetic pregnancies willhelp to understand how teratogenic insults compromise embryonic development and possibly provide ave-nues for therapeutic intervention. Birth Defects Research (Part A) 88:601611, 2010.",
              "title": "2010 - Neural tube defect genes and maternal diabetes during pregnancy.pdf",
              "version": "v0",
              "chunk_order": 4,
              "document_id": "aa74b552-7e06-5596-8dec-298c40ad558c",
              "extraction_id": "a9352adc-46d0-5947-a70d-940a7686008d",
              "associatedQuery": "What is the impact of genomic imprinting on the susceptibility and progression of diabetes?,"
            }
          },
          {
            "id": "dc64e623-a130-5814-b54a-dd5f787f10d5",
            "score": 0.6944549083709717,
            "metadata": {
              "text": "and metabolic imprinting: the ongoing effects of maternal hyper-glycemia. Diabetes Care 30:2287 2292\n9. Clausen TD, Mathiesen ER, Hansen T et al (2008) High prevalence\nof type 2 diabetes and pre-diabetes in adult offspring of women withgestational diabetes mellitus or type 1 diabetes: the role of intrauter-\nine hyperglycemia. Diabetes Care 31:340 346\n10. Solomon CG, Willett WC, Carey VJ et al (1997) A prospective\nstudy of pregravid determinants of gestational diabetes mellitus.\nJAMA 278:1078 1083",
              "title": "2018 - Genetic variants of gestational diabetes mellitus a study of 112 SNPs among 8722 women in two independent populations.pdf",
              "version": "v0",
              "chunk_order": 92,
              "document_id": "3b301dd1-17bd-5632-9a96-d6294c6d7650",
              "extraction_id": "6ca1166c-ba51-5437-b325-5299e3e8fcef",
              "associatedQuery": "What is the impact of genomic imprinting on the susceptibility and progression of diabetes?,"
            }
          },
          {
            "id": "5495230d-c26d-5633-90e8-028912e5298a",
            "score": 0.6944146959309192,
            "metadata": {
              "text": "M. Gestational diabetes alters offspring DNA methylation profiles in human and rat: Identification of key \npathways involved in endocrine system disorders, insulin signaling, diabetes signaling, and ILK signaling. \nEndocriniology 2015;156:2222 -38. \n[33] Murphy SK, Huang Z, Hoyo  C. Differentially methylated regions of imprinted genes in prenatal, \nperinatal and postnatal human tissues. PLOS ONE 2012;7:e40924.",
              "title": "2017 - Genome-wide DNA methylation variation in maternal and cord blood of gestational diabetes population.pdf",
              "version": "v0",
              "chunk_order": 79,
              "document_id": "e02a2e19-3527-5466-b8d6-69e62f657698",
              "extraction_id": "971ff653-c42a-5366-ae2b-080df9aa679f",
              "associatedQuery": "What is the impact of genomic imprinting on the susceptibility and progression of diabetes?,"
            }
          },
          {
            "id": "4ecf5607-8d58-5908-aa1b-4416af202e69",
            "score": 0.6941716877869277,
            "metadata": {
              "text": "12. Kim JK, Samaranayake M, Pradhan S. Epigenetic mechanisms in\nmammals. Cell Mol Life Sci. 2009;66:596-612.\n13. Horsthemke B, Buiting K. Genomic imprinting and imprinting\ndefects in humans. Adv Genet. 2008;61:225-246.\n14. Iacobuzio-Donahue CA. Epigenetic Changes in Cancer. Annu\nRev Pathol. 2009;4:229-249.\n15. Temple IK. Imprinting in human disease with special reference\nto transient neonatal diabetes and Beckwith-Wiedemann syn-\ndrome. Endocr Dev. 2007;12:113-123.",
              "title": "2010 - Autism Spectrum Disorders and Epigenetics.pdf",
              "version": "v0",
              "chunk_order": 115,
              "document_id": "6b435185-b16c-5b05-826b-eb98ca7bf806",
              "extraction_id": "dcc77767-4641-5969-b3c1-4ea96a644a74",
              "associatedQuery": "What is the impact of genomic imprinting on the susceptibility and progression of diabetes?,"
            }
          },
          {
            "id": "a5412cf9-367c-518e-bb4f-77d8deb00a32",
            "score": 0.6896819081807708,
            "metadata": {
              "text": "and Knowler W C. Intrauterine exposure to diabetes conveys risks for type 2 diabetes and obesity: A study \nof discordant sibships. Diabetes 2000;49:2208 -11. \n[11] Feil R and Fraga  MF. Epigenetics and the environment: Emerging patterns and implications. Nature  \nReviews Genetics 2012;13:97 -109. \n[12] Recillas -Targa F. DNA Methylation, Chromatin boundaries, and mechanisms of genomic imprinting. \nArchives of Medical Research 2002;33:428 -38.",
              "title": "2017 - Genome-wide DNA methylation variation in maternal and cord blood of gestational diabetes population.pdf",
              "version": "v0",
              "chunk_order": 69,
              "document_id": "e02a2e19-3527-5466-b8d6-69e62f657698",
              "extraction_id": "a17ed56f-20d4-56be-9aec-ac0b4943d19a",
              "associatedQuery": "What is the impact of genomic imprinting on the susceptibility and progression of diabetes?,"
            }
          },
          {
            "id": "9814f4a0-2701-5920-bfd7-df5e1f3b134e",
            "score": 0.6849690675735474,
            "metadata": {
              "text": "53. T ravers,M.E. etal.  Insights into the molecular \nmechanism for type2 diabetes susceptibility at the \nKCNQ1  locus from temporal changes in imprinting \nstatus in human islets. Diabetes 62, 987992 (2013).\n54. Gulli,G., Ferrannini,E., Stern,M., Haffner,S. \n&DeFronzo,R.A. The metabolic profile of NIDDM \nisfully established in glucose-tolerant offspring of \ntwoMexican-American NIDDM parents. Diabetes 41, \n15751586 (1992).\nPRIMER\nNATURE REVIEWS | DISEASE PRIMERS   VOLUME 1 | 2015  | 17",
              "title": "2015 - Type 2 diabetes mellitus.pdf",
              "version": "v0",
              "chunk_order": 211,
              "document_id": "415516ba-5365-501b-84ce-0789045862f8",
              "extraction_id": "bbe952b1-6cc2-56a8-b5e8-5ca6b44b4316",
              "associatedQuery": "What is the impact of genomic imprinting on the susceptibility and progression of diabetes?,"
            }
          },
          {
            "id": "4f7b210f-26f7-5726-baff-8d469b2cc3df",
            "score": 0.6846446990966797,
            "metadata": {
              "text": "Gaudet, D., Hivert, M.F., Brisson, D., Bouchard, L., 2013 Sep. Gestational diabetesmellitus epigenetically affects genes predominantly involved in metabolic dis-\neases. Epigenetics 8 (9), 935 e943.\nSalbaum, J.M., Kappen, C., 2012 Oct. Responses of the embryonic epigenome to\nmaternal diabetes. Birth Defects Res. A Clin. Mol. Teratol. 94 (10), 770 e781.\nSalbe, A.D., Lindsay, R.S., Collins, C.B., Tataranni, P.A., Krakoff, J., Bunt, J.C., 2007 Feb.",
              "title": "2015 - Maternal diabetes, gestational diabetes and the role of epigenetics in their long term effects on offspring.pdf",
              "version": "v0",
              "chunk_order": 242,
              "document_id": "3e92bd8e-fbf7-5bc4-9395-0a6dd0b0934e",
              "extraction_id": "e7e97f1e-d947-5b94-b2a9-5ac4b443628c",
              "associatedQuery": "What is the impact of genomic imprinting on the susceptibility and progression of diabetes?,"
            }
          },
          {
            "id": "8267bc80-1791-5e21-b228-053cba0629fd",
            "score": 0.6832183192242206,
            "metadata": {
              "text": "environment are probably mediated by a permanent program-ming of the developing offspring, e.g. by the mechanism ofimprinting. Of interest, the increased risk of diabetes continuesinto subsequent generations, suggesting the changes also affectthe germ cell line [143]. \nConclusions \nThere is little doubt that some animal models of diabetes have",
              "title": "2005  - Animal models of diabetes mellitus.pdf",
              "version": "v0",
              "chunk_order": 119,
              "document_id": "2fd381ac-2898-5a8c-af93-bcc86e7dec14",
              "extraction_id": "f7b36272-9780-52e8-9cb3-62d1c6c8c3b6",
              "associatedQuery": "What is the impact of genomic imprinting on the susceptibility and progression of diabetes?,"
            }
          },
          {
            "id": "4bb50efe-65b0-5c3c-9f58-03b423c93c0d",
            "score": 0.6831094622612,
            "metadata": {
              "text": "tal diabetes and later onset diabetes: a case of inher -\nited insulin resistance. Arch. Dis. Child.  72:5657.\n 6. Temple, I.K., et al. 1995. An imprinted gene(s) for \ndiabetes? Nat. Genet.  9:110112.\n 7. Temple, I.K., et al. 1996. Further evidence for an \nimprinted gene for neonatal diabetes localised to chro -",
              "title": "2004 - Impaired glucose homeostasis in transgenic mice expressing the human transient neonatal diabetes mellitus locus.pdf",
              "version": "v0",
              "chunk_order": 111,
              "document_id": "268bc8e3-7787-5bc0-8f7d-fffe20194dca",
              "extraction_id": "f68a90b3-5e03-57f4-8cb6-252e3a3fa132",
              "associatedQuery": "What is the impact of genomic imprinting on the susceptibility and progression of diabetes?,"
            }
          },
          {
            "id": "f703ae7e-5f64-52ee-860e-7b91b3066477",
            "score": 0.6825436760511218,
            "metadata": {
              "text": "1994; Martinez-Frias et al., 1998). The underlying mecha-nisms are not well understood, but are thought to\ninvolve various responses of the embryonic genome to\nthe adverse intrauterine environment (Greene, 2001;Loeken, 2008).\nTo explore how conditions of maternal diabetes affect\ngene expression in the embryo, we recently conducted\nexpression proling experiments on embryos from dia-betic dams compared to embryos from normal dams(Pavlinkova et al., 2009). We were able to demonstrate",
              "title": "2010 - Neural tube defect genes and maternal diabetes during pregnancy.pdf",
              "version": "v0",
              "chunk_order": 8,
              "document_id": "aa74b552-7e06-5596-8dec-298c40ad558c",
              "extraction_id": "a9352adc-46d0-5947-a70d-940a7686008d",
              "associatedQuery": "What is the impact of genomic imprinting on the susceptibility and progression of diabetes?,"
            }
          }
        ],
        "kg_search_results": null
      }
    }
  },
  "19": {
    "results": {
      "completion": {
        "id": "chatcmpl-AIHKkTED9VE0du8urGhS0MeefXMR7",
        "choices": [
          {
            "finish_reason": "stop",
            "index": 0,
            "message": {
              "content": "Longitudinal genomics studies are crucial for understanding gene-environment interactions in diabetes onset and management because they allow researchers to observe how genetic variations interact with environmental factors over time. This approach helps in identifying temporal patterns and causal relationships that are not possible to discern from cross-sectional studies [2]. By incorporating genotype measurements into longitudinal studies, researchers can gain significant insights into the genetic basis of diseases like diabetes, which can lead to better prediction, targeted prevention, and stratified treatment of type 2 diabetes (T2D) [3]. Additionally, these studies can help stratify T2D into subclasses that can be treated more effectively by understanding gene-lifestyle interactions [8]. Overall, longitudinal genomics studies provide a comprehensive framework to explore how genetic predispositions and environmental exposures collectively influence diabetes-related traits and disease progression.",
              "role": "assistant"
            }
          }
        ],
        "created": 1728919898,
        "model": "gpt-4o-2024-08-06",
        "object": "chat.completion",
        "system_fingerprint": "fp_6b68a8204b",
        "usage": {
          "completion_tokens": 163,
          "prompt_tokens": 1342,
          "total_tokens": 1505
        }
      },
      "search_results": {
        "vector_search_results": [
          {
            "id": "ee24ad01-f93a-55c4-8c2c-9dea6a6a84d5",
            "score": 0.7031167922546404,
            "metadata": {
              "text": "genome-wide association scans on type 2 dia-betes (Lango et al, 2008 ; van Hoek et al, 2008 ).\nBoth studies found a similar predictive value\nshowing only a marginal improvement in the\nprediction of type 2 diabetes beyond classicalclinical characteristics.\nThus, despite overwhelming signicances and\nrepeated replications, the explained variance andpredictive value of the currently identied sus-\nceptibility loci is too low to be clinically useful.\n5 GeneEnvironment Interactions\nin Obesity and Diabetes",
              "title": "2010 - Candidate Gene and Genome-Wide Association Studies in Behavioral Medicine.pdf",
              "version": "v0",
              "chunk_order": 4330,
              "document_id": "17637a6f-804e-50e4-9cf5-37318e17f15c",
              "extraction_id": "3bf3c6a7-de03-5114-bad8-d53fd76d0fba",
              "associatedQuery": "How do longitudinal genomics studies help in understanding gene-environment interactions in diabetes onset and management?"
            }
          },
          {
            "id": "de2af111-7fad-5dc1-baae-4742ccc8ba0d",
            "score": 0.7029099045449565,
            "metadata": {
              "text": "actions between genetic variation and environmental exposures\nand medical therapies has important implications for the predic-\ntion, targeted prevention, and s tratified treatment of T2D and\nmany other diseases.\nThe literature on gene-e nvironment interactions in\ndiabetes-related traits is extensive, but few studies are accom-\npanied by adequate replication data or compelling mechanistic\nexplanations. Moreover, most studies are cross-sectional,\nfrom which temporal patterns and causal effects cannot be",
              "title": "2016 - Putting the Genome in Context Gene-Environment Interactions.pdf",
              "version": "v0",
              "chunk_order": 8,
              "document_id": "ea43bb66-b6fe-5682-8f48-90568c080401",
              "extraction_id": "08acfe03-73b3-5533-b8e4-9caa031d33dd",
              "associatedQuery": "How do longitudinal genomics studies help in understanding gene-environment interactions in diabetes onset and management?"
            }
          },
          {
            "id": "e07d8080-aba7-5216-8a75-e078201b8c0a",
            "score": 0.7002737313427437,
            "metadata": {
              "text": "ined for a range of disorders, from diabetes, cancer and in  ammatory bowel disease to \ndepression. We refute the contention that incorporating the measurement of genotype into longitudinal-epidemiological studies is wasteful or unlikely to yield signi  cant \nbene  ts.\n2008 Genetic effects on environmental vulnerability to disease. Wiley, Chichester (Novartis Foundation Symposium) p 128142\nSlow progress understanding the genetic basis of many common diseases has been",
              "title": "2008 - Genetic Effects on Environmental Vulnerability to Disease Novartis Foundation Symposium 293.pdf",
              "version": "v0",
              "chunk_order": 986,
              "document_id": "5d65e407-34e5-5c1c-b394-989b7a09b57d",
              "extraction_id": "cfc4760c-755e-5693-8d7b-4332fb6c45e5",
              "associatedQuery": "How do longitudinal genomics studies help in understanding gene-environment interactions in diabetes onset and management?"
            }
          },
          {
            "id": "e76c1d0c-33b7-5d9e-958f-fce6adfe81aa",
            "score": 0.6990422010421803,
            "metadata": {
              "text": "In principle, each of these loci provides an opportunity to define \nthe genetic architecture and pathophysiology of these traits.\nThe earliest successes for genetic discovery in diabetes and \nobesity arose from the study of monogenic and syndromic \nforms of disease, for which the segregation of rare, but highly \npenetrant, alleles could be tracked using family-based linkage \napproaches that are well suited to that setting. Maturity-onset \ndiabetes of the young, for example, accounts for ~12% of cases",
              "title": "2012 - The Genetic and Epigenetic Basis of Type 2 Diabetes and Obesity.pdf",
              "version": "v0",
              "chunk_order": 15,
              "document_id": "d74ac751-712b-5970-98e6-bd348adc1dee",
              "extraction_id": "50bde36d-2968-5eaa-9713-924e73383427",
              "associatedQuery": "How do longitudinal genomics studies help in understanding gene-environment interactions in diabetes onset and management?"
            }
          },
          {
            "id": "30728ec3-882c-5bb0-8f41-4c74dfafdf13",
            "score": 0.6983946145537738,
            "metadata": {
              "text": "wide GxE interactions in explaining the variance of diabetes-related traits.\nCitation: Zheng J-S, Arnett DK, Lee Y-C, Shen J, Parnell LD, et al. (2013) Genome-Wide Contribution of Genotype by Environment Interaction to Variation of\nDiabetes-Related Traits. PLoS ONE 8(10): e77442. doi:10.1371/journal.pone.0077442\nEditor: Maria Eugenia Saez, CAEBi, Spain\nReceived April 10, 2013; Accepted September 3, 2013; Published October 28, 2013",
              "title": "2013 - Genome-Wide Contribution of Genotype by Environment Interaction.pdf",
              "version": "v0",
              "chunk_order": 6,
              "document_id": "8c310d76-0a3b-574c-9859-859258870ee5",
              "extraction_id": "f3975a2c-8a66-582e-a4b8-868b1f4722d4",
              "associatedQuery": "How do longitudinal genomics studies help in understanding gene-environment interactions in diabetes onset and management?"
            }
          },
          {
            "id": "f7ed49ac-f617-5c13-851e-98d1583e020f",
            "score": 0.690583825111394,
            "metadata": {
              "text": "data sharing to advance complex disease research. \nNat. Rev. Genet. 17, 535549 (2016).\n82. Franks,P .W., Pearson,E. & Florez,J.C. Gene-\nenvironment and gene-treatment interactions in \ntype2 diabetes: progress, pitfalls, and prospects. \nDiabetes Care 36, 14131421 (2013).\n83. Hagberg,J.M., Jenkins,N.T . & Spangenburg,E. \nExercise training, genetics and type2 diabetes-\nrelated phenotypes. Acta Physiol. 205, 456471 \n(2012).\n84. Langenberg,C.  etal.  Gene-lifestyle interaction and",
              "title": "2018 - Global aetiology and epidemiology of type 2 diabetes mellitus and its complications.pdf",
              "version": "v0",
              "chunk_order": 138,
              "document_id": "8bc8f3d4-968f-5252-ab4c-832b92e9ec0d",
              "extraction_id": "512ae4b5-27c8-509c-87ad-abd64d4295a6",
              "associatedQuery": "How do longitudinal genomics studies help in understanding gene-environment interactions in diabetes onset and management?"
            }
          },
          {
            "id": "151c185f-3300-5518-810c-3fb0d6715f2c",
            "score": 0.6864032336233131,
            "metadata": {
              "text": "Genomics and geneenvironment interactions\nEven though many cases of T2DM could be prevented \nby maintaining a healthy body weight and adhering to a \nhealthy lifestyle, some individuals with prediabetes mel -\nlitus are more susceptible to T2DM than others, which \nsuggests that individual differences in response to life -\nstyle interventions exist76. Substantial evidence from \ntwin and family studies has suggested a genetic basis \nof T2DM77. Over the past decade, successive waves of",
              "title": "2018 - Global aetiology and epidemiology of type 2 diabetes mellitus and its complications.pdf",
              "version": "v0",
              "chunk_order": 57,
              "document_id": "8bc8f3d4-968f-5252-ab4c-832b92e9ec0d",
              "extraction_id": "df2a8699-692f-5f25-94b3-508f9ed2f210",
              "associatedQuery": "How do longitudinal genomics studies help in understanding gene-environment interactions in diabetes onset and management?"
            }
          },
          {
            "id": "cc98a5b9-131e-5b60-919e-82e86b7a37a7",
            "score": 0.6857945101506102,
            "metadata": {
              "text": "DNA variation with disease processes in a range of settings, from cell\nlines to human populations, and major advances have been made in\ncoupling these complex datasets with information about extrinsic\nenvironmental exposures including drug prescription in ways that allowthe logical interrogation of gene-drug and gene-lifestyle interactions.\nDoing so may teach us about disease etiology and help stratify type 2\ndiabetes (T2D) into subclasses that can be treated more effectively, with",
              "title": "2016 - Putting the Genome in Context Gene-Environment Interactions.pdf",
              "version": "v0",
              "chunk_order": 102,
              "document_id": "ea43bb66-b6fe-5682-8f48-90568c080401",
              "extraction_id": "c362793d-c70f-5225-afe5-88098042daef",
              "associatedQuery": "How do longitudinal genomics studies help in understanding gene-environment interactions in diabetes onset and management?"
            }
          },
          {
            "id": "a94c609e-4816-5e10-96fd-ba8d79218405",
            "score": 0.6852905154228259,
            "metadata": {
              "text": "fuel subsequent functional and clinical translation studies.\nThis is important, because diabetes medicine may rely increas-\ningly on genomic stratification of patient populations and\ndisease phenotype, for which gene-environment interaction\nstudies might prove highly informative.\nHow Are Gene-Environment Interactions Defined?\nThe term gene-environment interaction has different meanings\nto different biomedical re searchers (see Supplement 1for\nglossary of terms used). However, here, we focus on the",
              "title": "2016 - Putting the Genome in Context Gene-Environment Interactions.pdf",
              "version": "v0",
              "chunk_order": 11,
              "document_id": "ea43bb66-b6fe-5682-8f48-90568c080401",
              "extraction_id": "08acfe03-73b3-5533-b8e4-9caa031d33dd",
              "associatedQuery": "How do longitudinal genomics studies help in understanding gene-environment interactions in diabetes onset and management?"
            }
          },
          {
            "id": "1d13cf78-3215-5873-b910-cbcac141779b",
            "score": 0.6837809274327215,
            "metadata": {
              "text": "Nutrients 2014, 6 5362 \n \n48. Cornelis, M.C.; Hu, F.B. Gene -enviroment interactions in the development of type 2 diabetes: \nRecent progress and continuing challenges. Annu. Rev. Nutr. 2012, 32, 245259. \n49. Lee, Y.C.; Lai, C.Q.; Ordovas, J.M.; Parnell, L.D. A database of gene -enviroment interactions \npertaining to blood lipid traits, cardiovascular disease and type 2 diabetes. J. Data Mining \nGenomics Proteomics 2011, 2, 106, doi:10.4172/2153- 0602.1000106.",
              "title": "2014 - Nutrigenetics and Nutrigenomics Insights into Diabetes Etiopathogenesis.pdf",
              "version": "v0",
              "chunk_order": 181,
              "document_id": "ce4f171c-494c-53f2-a770-c3edd3561c40",
              "extraction_id": "232f9536-eeac-5739-a57d-770cf5b32947",
              "associatedQuery": "How do longitudinal genomics studies help in understanding gene-environment interactions in diabetes onset and management?"
            }
          }
        ],
        "kg_search_results": null
      }
    }
  },
  "20": {
    "results": {
      "completion": {
        "id": "chatcmpl-AIHKoCrJvacxorigznvNb5BV4LGGI",
        "choices": [
          {
            "finish_reason": "stop",
            "index": 0,
            "message": {
              "content": "Recent integrative genomics approaches, such as the combination of single-cell RNA sequencing and epigenomic profiling, have significantly advanced our understanding of cellular heterogeneity and gene regulatory networks in pancreatic beta cells under diabetic conditions. These approaches have allowed researchers to delve deeper into the transcriptional regulation of non-beta cell types, which may play crucial roles in the hallmark features of beta cell insufficiency and dysfunction associated with Type 2 Diabetes (T2D) [1]. \n\nSingle-cell RNA sequencing has been particularly instrumental in high-throughput diabetes research by enabling the sequencing of individual cells from human pancreatic islets. This is important given the heterogeneity within the islets of Langerhans, which consist of various cell types. By tracking genetic changes in individual cells, researchers can better understand the complex cellular landscape and the specific contributions of different cell types to diabetes pathogenesis [2].\n\nFurthermore, epigenomic profiling adds another layer of complexity by revealing how epigenetic changes can modulate gene expression without altering the DNA sequence. These changes are crucial for maintaining the secretory capacity, survival, and functional identity of pancreatic islets, as well as their response to insulin [8]. The integration of these genomic and epigenomic data helps identify regulatory elements and pathways that could be targeted for therapeutic interventions, moving from correlation to causation in understanding diabetes [10].",
              "role": "assistant"
            }
          }
        ],
        "created": 1728919902,
        "model": "gpt-4o-2024-08-06",
        "object": "chat.completion",
        "system_fingerprint": "fp_6b68a8204b",
        "usage": {
          "completion_tokens": 271,
          "prompt_tokens": 1500,
          "total_tokens": 1771
        }
      },
      "search_results": {
        "vector_search_results": [
          {
            "id": "d5c2a32a-b869-59c1-8a63-45ab620669de",
            "score": 0.7126145958900452,
            "metadata": {
              "text": "4. PRECISE CELLULAR GENOMICS\nElucidating the molecular mechanisms that lead to beta cell\ndysfunction and T2D pathogenesis has been a major focus of diabetes\nresearch for decades. However, advances in single cell genomic\nproling techniques have led to greater understanding of non-beta cell\ntype transcriptional regulation and suggest that they may play\nimportant roles in hallmark features of beta cell insuf ciency and",
              "title": "2019 - (Epi)genomic heterogeneity of pancreatic islet function and failure in type 2 diabetes.pdf",
              "version": "v0",
              "chunk_order": 55,
              "document_id": "b9bc63a5-e366-5685-bd7a-4732a8eeffb7",
              "extraction_id": "7a2a9981-4096-5049-a717-3e69eb609777",
              "associatedQuery": "How have recent integrative genomics approaches, such as the use of single-cell RNA sequencing combined with epigenomic profiling, advanced our understanding of cellular heterogeneity and gene regulatory networks in pancreatic beta cells under diabetic conditions?"
            }
          },
          {
            "id": "1c659cb4-085b-55b9-be3c-6332c36cbeba",
            "score": 0.6997478008270264,
            "metadata": {
              "text": "Genes 2018 ,9, 374 7 of 19\n4. Single-Cell RNA-seq as a Novel Approach in High-Throughput Type 2 Diabetes Research\nIslets of Langerhans are heterogeneous structures that consist of different cell types. Further\nresearch is needed to track genetic changes in individual pancreatic islet cells and in sorted cell\npopulations. The massive development of NGS allowed the sequencing of single cells from human\npancreatic islets. Considering the cell-type heterogeneity within Langerhans islets, such an approach",
              "title": "2018 - High-Throughput Approaches onto Uncover (Epi)Genomic Architecture of Type 2 Diabetes.pdf",
              "version": "v0",
              "chunk_order": 56,
              "document_id": "1cb0c4ac-c1fe-55c2-919c-52cd5018c00d",
              "extraction_id": "52e8a636-ced9-5c14-a7e5-0c30b7f05107",
              "associatedQuery": "How have recent integrative genomics approaches, such as the use of single-cell RNA sequencing combined with epigenomic profiling, advanced our understanding of cellular heterogeneity and gene regulatory networks in pancreatic beta cells under diabetic conditions?"
            }
          },
          {
            "id": "f06bcc81-6ef9-5874-8ef9-6bcb3c34b0d0",
            "score": 0.6991837233061711,
            "metadata": {
              "text": "Advances ofsingle -cell genomics andepigenomics inhuman disease: whereare we now?  \n1 3\nBrissova etal. 2018; Tritschler etal. 2017). Moreover, an \nincrease in hyperglycaemia has been associated with a loss of beta-cell mass, function and organization and is the cell type most frequently studied for insulin resistance (Carrano etal. 2017; Lawlor etal. 2017b; Segerstolpe etal. 2016; \nTheis and Lickert 2019; Tritschler etal. 2017).\nNotably, single-cell transcriptome profiling has been",
              "title": "2020 - Advances of single?cell genomics and epigenomics in human disease.pdf",
              "version": "v0",
              "chunk_order": 62,
              "document_id": "afe53f5a-3962-520f-be55-9df5bfdaad70",
              "extraction_id": "65471d38-cd13-5de2-8c19-1eb72d24d6f5",
              "associatedQuery": "How have recent integrative genomics approaches, such as the use of single-cell RNA sequencing combined with epigenomic profiling, advanced our understanding of cellular heterogeneity and gene regulatory networks in pancreatic beta cells under diabetic conditions?"
            }
          },
          {
            "id": "b7812a7a-5504-57ca-8755-969dee45717e",
            "score": 0.6979928016662598,
            "metadata": {
              "text": "Tang X, Huang Y, Lei J, Luo H, Zhu X (2019) The single-cell sequenc-\ning: new developments and medical applications. Cell Biosci \n9:53. https ://doi.org/10.1186/s1357 8-019-0314-y\nTeo AKK etal (2018) Single-cell analyses of human islet cells reveal \nde-differentiation signatures. Cell Death Discov 4:14. https ://doi.\norg/10.1038/s4142 0-017-0014-5\nTheis FJ, Lickert H (2019) A map of beta-cell differentiation pathways \nsupports cell therapies for diabetes. Nature 569:342343. https  ://",
              "title": "2020 - Advances of single?cell genomics and epigenomics in human disease.pdf",
              "version": "v0",
              "chunk_order": 158,
              "document_id": "afe53f5a-3962-520f-be55-9df5bfdaad70",
              "extraction_id": "7f7a7f30-2e4e-50aa-bbcb-9f211c371e38",
              "associatedQuery": "How have recent integrative genomics approaches, such as the use of single-cell RNA sequencing combined with epigenomic profiling, advanced our understanding of cellular heterogeneity and gene regulatory networks in pancreatic beta cells under diabetic conditions?"
            }
          },
          {
            "id": "ab373b7e-8c0b-59d8-9408-3e09ac76761e",
            "score": 0.6955804824829102,
            "metadata": {
              "text": "53. Eliasson L, Esguerra JL (2014) Role of non-coding RNAs in pancreatic beta-cell development and physiology. Acta Physiol \n(Oxf) 211:273284\n 54. Ding GL, Wang FF, Shu J etal (2012) Transgenerational glucose \nintolerance with Igf2/H19 epigenetic alterations in mouse islet induced by intrauterine hyperglycemia. Diabetes 61:11331142\n 55. Ku GM, Kim H, Vaughn IW etal (2012) Research resource: RNA-Seq reveals unique features of the pancreatic beta-cell tran-scriptome. Mol Endocrinol 26:17831792",
              "title": "2018 - Lnc\u2011ing non\u2011coding RNAs with metabolism and diabetes roles.pdf",
              "version": "v0",
              "chunk_order": 112,
              "document_id": "019efefb-65db-55f5-a3a7-4f224473f51f",
              "extraction_id": "8bbfb009-87b7-54ae-8465-8796db8c271a",
              "associatedQuery": "How have recent integrative genomics approaches, such as the use of single-cell RNA sequencing combined with epigenomic profiling, advanced our understanding of cellular heterogeneity and gene regulatory networks in pancreatic beta cells under diabetic conditions?"
            }
          },
          {
            "id": "7a5c8fad-97c5-59d2-8e5e-ee72d3dc2362",
            "score": 0.6935632594152429,
            "metadata": {
              "text": "24. Nica, A. C. et al. Cell-type, allelic, and genetic signatures in the human\npancreatic beta cell transcriptome. Genome Res. 23, 1554 1562 (2013).\n25. Takane, K. K., Bender, A. & Stewart, A. F. Speci c targeting and sorting of\npuried human beta cells: de ning the human beta cell transcriptome. ADA\nScienti c Sessions, San Francisco (2014).\n26. Langfelder, P. & Horvath, S. WGCNA: an R package for weighted correlation\nnetwork analysis. BMC Bioinformatics 9, 559 (2008).",
              "title": "2017 - Insights into beta cell regeneration for diabetes via integration of molecular landscapes in human insulinomas.pdf",
              "version": "v0",
              "chunk_order": 203,
              "document_id": "6cf1eb8d-a91e-58a2-b6f4-29653678d0d3",
              "extraction_id": "bdf327a6-decb-5c7a-a981-a7969206b455",
              "associatedQuery": "How have recent integrative genomics approaches, such as the use of single-cell RNA sequencing combined with epigenomic profiling, advanced our understanding of cellular heterogeneity and gene regulatory networks in pancreatic beta cells under diabetic conditions?"
            }
          },
          {
            "id": "b7c1d2be-88c5-5f33-b812-b05e842f1647",
            "score": 0.6914374048374549,
            "metadata": {
              "text": "5. Genome-Wide Proling of Epigenetic Changes in Pancreatic Islets and Peripheral Tissues\nEpigenetic data added another layer of complexity to our understanding of the genomic bases\nof T2D. Given that a variable epigenetic pattern can modulate the link between the SNP and trait,\nconsideration of this interplay is critically important. Molecular epigenetics involves changes in\ngene function that occur without a change in the nucleotide sequence via DNA methylation, histone",
              "title": "2018 - High-Throughput Approaches onto Uncover (Epi)Genomic Architecture of Type 2 Diabetes.pdf",
              "version": "v0",
              "chunk_order": 60,
              "document_id": "1cb0c4ac-c1fe-55c2-919c-52cd5018c00d",
              "extraction_id": "52e8a636-ced9-5c14-a7e5-0c30b7f05107",
              "associatedQuery": "How have recent integrative genomics approaches, such as the use of single-cell RNA sequencing combined with epigenomic profiling, advanced our understanding of cellular heterogeneity and gene regulatory networks in pancreatic beta cells under diabetic conditions?"
            }
          },
          {
            "id": "11a5527b-8d22-5e69-8a84-6d9180517d81",
            "score": 0.6867216042710319,
            "metadata": {
              "text": "and model organisms. The combination of data from high-throughput approaches and association\nstudies has provided compelling evidence that some epigenetic markers contribute to the risk of\nT2D [ 57,58]. Epigenetic alterations have been shown to affect the expression of genes that are crucial\nfor maintaining pancreatic islet secretory capacity, survival, and functional identity and the proper\nresponse to insulin in peripheral tissues [ 59,60]. Furthermore, several epigenetic signatures, such",
              "title": "2018 - High-Throughput Approaches onto Uncover (Epi)Genomic Architecture of Type 2 Diabetes.pdf",
              "version": "v0",
              "chunk_order": 64,
              "document_id": "1cb0c4ac-c1fe-55c2-919c-52cd5018c00d",
              "extraction_id": "52e8a636-ced9-5c14-a7e5-0c30b7f05107",
              "associatedQuery": "How have recent integrative genomics approaches, such as the use of single-cell RNA sequencing combined with epigenomic profiling, advanced our understanding of cellular heterogeneity and gene regulatory networks in pancreatic beta cells under diabetic conditions?"
            }
          },
          {
            "id": "db06230d-31c0-5947-8c1c-f58c48b6f439",
            "score": 0.6851639339230112,
            "metadata": {
              "text": "Epigenomic approaches: applications in diabetic\ncomplications research\nEpigenetic studies in human disease have been greatly accel-\nerated as a result of advances in whole-genome and epige-\nnome profiling technologies as well as bioinformatics andgenomic data analysis platforms [ 99,100]. DNAme is\nanalysed using bisulfite conversion of genomic DNA, immu-\nnoprecipitation of methylated DNA, followed byhybridisation to arrays or next-generation sequencing to ob-",
              "title": "2015 - Epigenetic mechanisms in diabetic complications and metabolic memory.pdf",
              "version": "v0",
              "chunk_order": 96,
              "document_id": "470f1f94-792d-5273-a88f-7e06084951c5",
              "extraction_id": "312b1856-e1b1-5ae7-8cba-370becf5f7cb",
              "associatedQuery": "How have recent integrative genomics approaches, such as the use of single-cell RNA sequencing combined with epigenomic profiling, advanced our understanding of cellular heterogeneity and gene regulatory networks in pancreatic beta cells under diabetic conditions?"
            }
          },
          {
            "id": "a2adc65b-035b-568f-a0ae-9f7821ef45bc",
            "score": 0.6835848877677401,
            "metadata": {
              "text": "understand each cell type s genomic architecture and better charac-\nterize their roles in islet resilience and failure. Experimental manipu-\nlation of the regulatory elements and/or the target genes identi ed by\n(epi)genomic approaches described above and modeling the putativepathways and processes they implicate in human islet cell lines (e.g.,\nEndoC-\nbH1-H3) is essential to progress from correlation to causation.\nSimilarly, transitioning from themouse (C57BL/6) to multiple mouse",
              "title": "2019 - (Epi)genomic heterogeneity of pancreatic islet function and failure in type 2 diabetes.pdf",
              "version": "v0",
              "chunk_order": 97,
              "document_id": "b9bc63a5-e366-5685-bd7a-4732a8eeffb7",
              "extraction_id": "117cc1a5-d236-56b2-a69d-9c0a2fb9053d",
              "associatedQuery": "How have recent integrative genomics approaches, such as the use of single-cell RNA sequencing combined with epigenomic profiling, advanced our understanding of cellular heterogeneity and gene regulatory networks in pancreatic beta cells under diabetic conditions?"
            }
          }
        ],
        "kg_search_results": null
      }
    }
  }
}