aboutsummaryrefslogtreecommitdiff
path: root/gnqa/paper2_eval/data/responses/gpt4o/cs_diabetes_responses.json
blob: 870d3c05d272fc4ff1b6997daeddc6d194682e3e (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-AIFp3LBsYqGcLUvIYyVnKoJE32qw3",
        "choices": [
          {
            "finish_reason": "stop",
            "index": 0,
            "message": {
              "content": "Genetic mutations in the insulin gene can significantly affect glucose metabolism in several ways:\n\n1. Mutations that result in mutant insulin or the inability to convert proinsulin to insulin can lead to glucose intolerance. This is because the proper conversion and function of insulin are crucial for maintaining normal glucose levels in the blood [1].\n\n2. Insulin gene mutations can cause early-onset diabetes, with marked differences in clinical presentation and metabolic status. These mutations can lead to the retention of insulin in the endoplasmic reticulum, causing stress and potentially leading to beta-cell apoptosis, which reduces insulin production and secretion [2], [10].\n\n3. Insulin-deficient conditions, such as those caused by certain mutations, can lead to severe hyperglycemia, as insulin is a central hormone for maintaining glucose homeostasis. Without sufficient insulin, glucose cannot be effectively taken up by cells, leading to elevated blood glucose levels [5].\n\n4. Some mutations may result in abnormal proinsulin folding, leading to an accumulation of misfolded proteins in the endoplasmic reticulum. This accumulation can cause endoplasmic reticulum stress and beta-cell apoptosis, further impairing insulin production and secretion [10].\n\nOverall, genetic mutations in the insulin gene can disrupt normal insulin synthesis and function, leading to impaired glucose metabolism and conditions such as diabetes.",
              "role": "assistant"
            }
          }
        ],
        "created": 1728914089,
        "model": "gpt-4o-2024-08-06",
        "object": "chat.completion",
        "system_fingerprint": "fp_6b68a8204b",
        "usage": {
          "completion_tokens": 266,
          "prompt_tokens": 1393,
          "total_tokens": 1659
        }
      },
      "search_results": {
        "vector_search_results": [
          {
            "id": "111f4875-7ccd-502f-bd5b-5d4ee88e5af6",
            "score": 0.7227188944816589,
            "metadata": {
              "text": "Mutations that result in mutant insulin or the \ninability to convert proinsulin to insulin result in gl ucose \nintolerance in some of these cases. Genetic defects \nin the insulin receptor or in the signal transduction \npathway of insulin have been demonstrated to result in \nhyperinsulinemia and modest hyperglycemia to severe \ndiabetes[1]. \nDisease of the exocrine pancreas\nDamage of the  cells of the pancreas due to diffused \ninjury of the pancreas can cause diabetes. This damage",
              "title": "2015 - Diabetes mellitus The epidemic of the century.pdf",
              "version": "v0",
              "chunk_order": 54,
              "document_id": "e114dd28-fd39-56df-bdeb-8806474a6c10",
              "extraction_id": "7d87ee73-2cc1-576c-8c0d-eb58479177b8",
              "associatedQuery": "How do genetic mutations in the insulin gene affect glucose metabolism?"
            }
          },
          {
            "id": "3f7cc31a-dc57-568a-a3b0-602280c56428",
            "score": 0.7041979432106018,
            "metadata": {
              "text": "A, et al. Insulin gene mutations resulting in early-onset diabetes:\nmarked differences in clinical presentation, metabolic status, and\npathogenic effect through endoplasmic reticulum retention.\nDiabetes. 2010;59:653 61.\n21. Steele AM, Shields BM, Wensley KJ, Colclough K, Ellard S,\nHattersley AT. Prevalence of vascular complications among pa-\ntients with glucokinase mutations and prolonged, mild hyperglyce-\nmia. JAMA. 2014;311:279 86.22. Chakera AJ, Spyer G, Vincent N, Ellard S, Hattersley AT, Dunne FP.",
              "title": "2019 - How Recent Advances in Genomics Improve Precision Diagnosis.pdf",
              "version": "v0",
              "chunk_order": 94,
              "document_id": "42b64375-06af-5e09-9ae6-6bd0ecb782c7",
              "extraction_id": "ebb48c39-f48a-5dce-a4dc-fcd34e6f17e1",
              "associatedQuery": "How do genetic mutations in the insulin gene affect glucose metabolism?"
            }
          },
          {
            "id": "3cb068b3-0761-5fd8-91bd-92744fa9ca9a",
            "score": 0.6915489045247112,
            "metadata": {
              "text": "presumed glucose toxicity (34). The finding that a mutation\nof a single nucleotide in the gene encoding the glucokinase\nenzyme can result in NIDDM lends credibility to the hypoth-\nesis that inherited defects in insulin production contribute to\nNIDDM (6). Increased insulin demand of obesity and insulin\nresistance is accompanied by enhanced insulin biosynthesis,",
              "title": "1994 - Isolation of the Human LIMTHomeodomain Gene Islet-1.pdf",
              "version": "v0",
              "chunk_order": 58,
              "document_id": "c77b101b-9f78-5090-8be9-6f98d9380466",
              "extraction_id": "b3b6cf5d-8cc3-5559-af5f-36780a303792",
              "associatedQuery": "How do genetic mutations in the insulin gene affect glucose metabolism?"
            }
          },
          {
            "id": "5d64fd7b-32d2-55ac-8586-c7ba9172a9ef",
            "score": 0.6910686492919922,
            "metadata": {
              "text": "insulin synthesis and function while mutations in the insulin gene ( INS)\nobviously affect the key hormone made by pancreatic beta cells [62].\nATP synthesis defect (mitochondrial diabetes) and mutations in ATP-\nsensitive potassium channel subunits (channel-building Kir6.2 [po-\ntassium inwardly-rectifying channel, subfamily J, member 11;KCNJ11 ] and regulatory SUR1 [ATP-binding cassette transporter\nsubfamily C member 8], ABCC8 ) all affect insulin secretion [63].",
              "title": "2016 - Dissecting diabetes metabolic disease.pdf",
              "version": "v0",
              "chunk_order": 47,
              "document_id": "eee2f79d-e093-52fb-871a-798fd859235e",
              "extraction_id": "998a92ba-e7fc-5553-b629-7b5797fbfafe",
              "associatedQuery": "How do genetic mutations in the insulin gene affect glucose metabolism?"
            }
          },
          {
            "id": "551f3603-6a4c-51e6-b568-7ae2fc9e7b33",
            "score": 0.6834791302680969,
            "metadata": {
              "text": "Insulin gene mutations \nInsulin is synthesized in 13-cells of the islets of Langerhans and is a \ncentral honnone that maintains glucose homeostasis. Insulin-deficient mice \ndie shortly after birth due to severe hyperglycemia.53 All cell types of the \nendocrine pancreas are present in insulin deficient mice suggesting that \ninsulin is not required for development and differentiation of the endocrine \npancreas. 53 \nNaturally occurring mutations in the insulin gene that result in the",
              "title": "2004 - Diabetes Genes a.pdf",
              "version": "v0",
              "chunk_order": 1118,
              "document_id": "805ad0d5-6a3d-5e5f-b1ba-121e10c6daaa",
              "extraction_id": "ce0307a5-fae4-5b6d-9786-10619e49e075",
              "associatedQuery": "How do genetic mutations in the insulin gene affect glucose metabolism?"
            }
          },
          {
            "id": "2126e367-c1aa-56ae-aff4-0ba7e7070a22",
            "score": 0.6803162097930908,
            "metadata": {
              "text": "Theprevalenceofgeneticmutationsaffectingthestructure\noftheinsulinmoleculeinthegeneralpopulationisunknown.\nUptothepresent,onlythosepatientsmanifestingthemutant\ninsulinsyndrome(5-8,36)withunusualorfamilialTypeII\ndiabeteshavebeenscreenedanddiscovered.Thus,mutantin-\nsulinspecieswithnormalorrelativelywell-preservedbinding\nandbiologicalactivitycharacteristics,andthereforenormal\nmetabolicclearances,areunlikelytobediscoveredbythisap-\nproachsincehyperinsulinemiawillbeabsentorsubtle.Future",
              "title": "1986 - Diabetes due to secretion of a structurally abnormal insulin.pdf",
              "version": "v0",
              "chunk_order": 56,
              "document_id": "5b884835-4cf4-5e80-a762-36582271e63e",
              "extraction_id": "ffdee7b8-ff45-57bf-973a-ca03ba595d23",
              "associatedQuery": "How do genetic mutations in the insulin gene affect glucose metabolism?"
            }
          },
          {
            "id": "487ca988-cce2-5b92-a05f-2e1cd11efea3",
            "score": 0.6698050498962402,
            "metadata": {
              "text": "at various steps, resulting in an impaired insulin action and potential \ndevelopment of extreme insulin resistant clinical conditions. Many mutations \nhave been identified in the insulin receptor gene. These mutations may lead \nto: \n Decreased insulin receptor biosynthesis \n Premature chain termination in extracellular or intracellular domain \n Accelerated receptor degradation \n Defect in the receptor transport to plasma membranes \n Decreased insulin binding affinity \n Impaired tyrosine kinase activity",
              "title": "2004 - Diabetes Genes a.pdf",
              "version": "v0",
              "chunk_order": 1336,
              "document_id": "805ad0d5-6a3d-5e5f-b1ba-121e10c6daaa",
              "extraction_id": "508fd29c-5cf8-52bc-8bf2-5cebb1833cb1",
              "associatedQuery": "How do genetic mutations in the insulin gene affect glucose metabolism?"
            }
          },
          {
            "id": "fa07c029-ad6e-5768-97da-a4bc5aa4e44f",
            "score": 0.6691384514909524,
            "metadata": {
              "text": "15. Steiner DF, Tager HS, Chan SJ, et al . Lessons learned from\nmolecular biology of insulin-gene mutations. Diabetes Care\n1990; 13: 600609.\n16. Vionnet N, Stoffel M, Takeda J, et al . Nonsense mutation in the\nglucokinase gene causes early-onset non-insulin-dependent\ndiabetes mellitus. Nature 1992; 356 : 721722.\n17. Sakagashira S, Sanke T, Hanabusa T, et al . Missense mutation of\namylin gene (S20G) in Japanese NIDDM patients. Diabetes\n1996; 45: 12791281.",
              "title": "2001 - Genomic variation in pancreatic ion channel genes in Japanese type 2 diabetic patients.pdf",
              "version": "v0",
              "chunk_order": 40,
              "document_id": "bff7795f-c109-5d7f-871d-ef1f4400a2c6",
              "extraction_id": "2d845f58-a674-5e08-a68b-e8cfea8a9b84",
              "associatedQuery": "How do genetic mutations in the insulin gene affect glucose metabolism?"
            }
          },
          {
            "id": "644810c4-af08-5c60-b333-8c97ddadae8b",
            "score": 0.6649872064590454,
            "metadata": {
              "text": "vating mutations in the gene encoding\nKir6.2 alter fetal and postnatal growthand also cause neonatal diabetes. J\nClin Endocrinol Metab 2006; 91(7):\n27822788.\n93. Stoy J, Edghill EL, Flanagan SE, et al.\nInsulin gene mutations as a cause\nof permanent neonatal diabetes. Proc\nNatl Acad Sci U S A 2007; 104(38):\n1504015044.\n94. Pulizzi N, Lyssenko V, Jonsson A, et al.\nInteraction between prenatal growth\nand high-risk genotypes in the devel-opment of type 2 diabetes. Diabetolo-\ngia2009; 52(5): 825829.",
              "title": "2011 - Genome-wide association studies (GWAS) impact.pdf",
              "version": "v0",
              "chunk_order": 131,
              "document_id": "086c6869-7c70-5364-9269-760267fb458d",
              "extraction_id": "a4721e1b-273b-5042-bc20-cf5602f0955a",
              "associatedQuery": "How do genetic mutations in the insulin gene affect glucose metabolism?"
            }
          },
          {
            "id": "ac0df77e-c676-552b-b742-1591cb18fbbb",
            "score": 0.6630228161811829,
            "metadata": {
              "text": "(Edghill et al., 2008; Garin et al., 2010; Stoy et al., 2007). Hyperglycemia occurs due to decreased insulin biosynthe-sis, in which most of the reported missense heterozygous mutations are expected to cause an abnormal proinsulin folding. An accumulation of the misfolded protein in the en-doplasmic reticulum (ER) consequently occurs, resulting in ER stress and betacell apoptosis (Liu, Hodish, Rhodes, & Arvan, 2007). Our identified de novo novel variant in INS is expected to result in aberrant proinsulin",
              "title": "2019 - The clinical and genetic characteristics of permanent neonatal diabetes  PNDM  in the state of Qatar.pdf",
              "version": "v0",
              "chunk_order": 123,
              "document_id": "90268a68-3926-535e-952e-735e206eb3ab",
              "extraction_id": "d340209d-0773-5cfc-b0d1-e714f2ebe9f2",
              "associatedQuery": "How do genetic mutations in the insulin gene affect glucose metabolism?"
            }
          }
        ],
        "kg_search_results": null
      }
    }
  },
  "2": {
    "results": {
      "completion": {
        "id": "chatcmpl-AIFpDYOJMy59ARMmvejZqYaNW81X4",
        "choices": [
          {
            "finish_reason": "stop",
            "index": 0,
            "message": {
              "content": "The most common genetic loci associated with an increased risk of Type 2 diabetes include:\n\n1. **TCF7L2** [2], [5], [7]\n2. **KCNJ11** [2], [5]\n3. **PPARG** [2], [5]\n4. **CDKAL1** [2], [5], [7]\n5. **CDKN2A/CDKN2B** [2], [5], [7]\n6. **IGF2BP2** [2], [5], [7]\n7. **SLC30A8** [2], [5], [7]\n8. **FTO** [2], [5]\n9. **HHEX** [2], [5], [7]\n\nThese loci have been identified and replicated in multiple studies as being associated with an increased susceptibility to Type 2 diabetes.",
              "role": "assistant"
            }
          }
        ],
        "created": 1728914099,
        "model": "gpt-4o-2024-08-06",
        "object": "chat.completion",
        "system_fingerprint": "fp_6b68a8204b",
        "usage": {
          "completion_tokens": 185,
          "prompt_tokens": 1627,
          "total_tokens": 1812
        }
      },
      "search_results": {
        "vector_search_results": [
          {
            "id": "47558743-2803-51a7-856b-8f6606712d08",
            "score": 0.7865780846383527,
            "metadata": {
              "text": "novel risk loci for type 2 diabetes. Nature 2007, 445(7130) :881-885.5. Gaulton KJ, Willer CJ, Li Y, Scott LJ, Conneely KN, Jackson AU, Duren WL,\nChines PS, Narisu N, Bonnycastle LL, et al:Comprehensive association\nstudy of type 2 diabetes and related quantitative traits with 222\ncandidate genes. Diabetes 2008, 57(11) :3136-3144.\n6. Hu C, Zhang R, Wang C, Wang J, Ma X, Lu J, Qin W, Hou X, Bao Y, Xiang K,\net al:PPARG, KCNJ11, CDKAL1, CDKN2A-CDKN2B, IDE-KIF11-HHEX,",
              "title": "2012 - Genome-wide meta-analysis of genetic susceptible genes for Type 2 Diabetes.pdf",
              "version": "v0",
              "chunk_order": 67,
              "document_id": "f5096148-3f85-57c1-8414-2f240ea42068",
              "extraction_id": "e61efd89-f844-5c3a-98b9-1a827b58b507",
              "associatedQuery": "What are the most common genetic loci associated with an increased risk of Type 2 diabetes?"
            }
          },
          {
            "id": "aa72551a-ac0c-5d7d-8057-34f229f68eb1",
            "score": 0.7847404479980469,
            "metadata": {
              "text": "ly associated with type 2 diabetes: TCF7L2, KCNJ11,  \nand PPARG .\n5-7 However, in 2007, a number of novel \ngenetic variants ( CDKAL1, IGF2BP2,  the locus on \nchromosome 9 close to CDKN2A/CDKN2B, FTO, \nHHEX, SLC30A8,  and WFS1)8-14 were shown to in -\ncrease susceptibility to type 2 diabetes in repro -\nducible studies. Furthermore, a recent meta-analy -\nsis identified six novel variants ( JAZF1, CDC123/\nCAMK1D, TSPAN8/LGR5, THADA, ADAMTS9, and NOTCH2 ) that are associated with type 2 dia -\nbetes.\n15",
              "title": "2008 - Clinical Risk Factors, DNA Variants.pdf",
              "version": "v0",
              "chunk_order": 10,
              "document_id": "0018610a-9c86-5e2d-a27d-f66cf4f8519d",
              "extraction_id": "2643b341-8c50-5cea-af36-86a8b070a80e",
              "associatedQuery": "What are the most common genetic loci associated with an increased risk of Type 2 diabetes?"
            }
          },
          {
            "id": "225792f4-c56b-5139-8bec-d5d1d393a6b2",
            "score": 0.7845839986453071,
            "metadata": {
              "text": "2009. There are now at least 19 loci containing genes that increase\nrisk of T2D, including PPARG [27], KCNJ11 [27], KCNQ1 [28,29],\nPLoS Genetics | www.plosgenetics.org 1 February 2010 | Volume 6 | Issue 2 | e1000847",
              "title": "2010 - A Genome-Wide Association Study Identifies.pdf",
              "version": "v0",
              "chunk_order": 13,
              "document_id": "0301881d-40dd-5343-b22e-927d58c2cb2a",
              "extraction_id": "f5b0ecdc-fdf2-5ac3-bebb-9c9ff5863935",
              "associatedQuery": "What are the most common genetic loci associated with an increased risk of Type 2 diabetes?"
            }
          },
          {
            "id": "8b718138-167a-50b0-afb7-4b507abc05ff",
            "score": 0.7839787006378174,
            "metadata": {
              "text": "et al. Association between type 2 diabetes loci and measures of fatness. PLoS One 5,\ne8541 (2010).\n22 Ng, M. C., Park, K. S., Oh, B., Tam, C. H., Cho, Y. M., Shin, H. D. et al. Implication of\ngenetic variants near TCF7L2, SLC30A8, HHEX, CDKAL1, CDKN2A/B, IGF2BP2, and\nFTO in type 2 diabetes and obesity in 6,719 Asians. Diabetes 57,22262233 (2008).\n23 Thorsby, P. M., Midthjell, K., Gjerlaugsen, N., Holmen, J., Hanssen, K. F., Birkeland, K. I.",
              "title": "2012 - Association between type 2 diabetes genetic susceptibility loci and visceral and subcutaneous fat area as determined by computed tomography.pdf",
              "version": "v0",
              "chunk_order": 72,
              "document_id": "b86d3101-f383-520b-8360-7d80bc7ec6fa",
              "extraction_id": "e0bbfc0e-ae79-568c-b704-96febad87d6f",
              "associatedQuery": "What are the most common genetic loci associated with an increased risk of Type 2 diabetes?"
            }
          },
          {
            "id": "e3cbe02b-9a3c-5b66-a5fb-d9d75b5db3f9",
            "score": 0.7799915307757718,
            "metadata": {
              "text": "Genome-wide association studies validated these old culprits of T2D and expanded them to include\nhundreds of single-nucleotide variants (SNVs) that represent more than 150 genomic loci that are\nassociated with T2D, insulin secretion, and insulin resistance [ 11]. Besides TCF7L2 ,PP ARG , and\nKCNJ11 loci, the most replicated T2D susceptibility variants identied in GWASs were found in and\naround CDKN2A/2B ,IGF2BP2 ,SLC30A8 ,CDKAL1 and FTO genes [ 1215]. The variants that are most",
              "title": "2018 - High-Throughput Approaches onto Uncover (Epi)Genomic Architecture of Type 2 Diabetes.pdf",
              "version": "v0",
              "chunk_order": 15,
              "document_id": "1cb0c4ac-c1fe-55c2-919c-52cd5018c00d",
              "extraction_id": "aba850e8-8c0d-5256-b2ba-fa1dfc221114",
              "associatedQuery": "What are the most common genetic loci associated with an increased risk of Type 2 diabetes?"
            }
          },
          {
            "id": "f3ce8455-f123-5840-8a50-da7885c7e18d",
            "score": 0.7798925042152405,
            "metadata": {
              "text": "Meta-analysis of genome-wide association data and large-scale replication identifies additional susceptibility loci for type 2 diabetes. Nat Genet 2008;40:638-45.\n20. Dupuis J, Langenberg C, Prokopenko \nI, et al. New genetic loci implicated in fasting glucose homeostasis and their im -\npact on type 2 diabetes risk. Nat Genet 2010;42:105-16.\n21. Qi L, Cornelis MC, Kraft P, et al. Ge -\nnetic variants at 2q24 are associated with susceptibility to type 2 diabetes. Hum Mol Genet 2010;19:2706-15.",
              "title": "2010 - Genomics, Type 2 Diabetes, and Obesity.pdf",
              "version": "v0",
              "chunk_order": 104,
              "document_id": "3d629777-f1b6-5450-94ef-56736e5a4e10",
              "extraction_id": "8a28c11f-e0d2-526b-ac85-2f2fbf054fc5",
              "associatedQuery": "What are the most common genetic loci associated with an increased risk of Type 2 diabetes?"
            }
          },
          {
            "id": "dfba6b2e-1531-5ac4-a41d-aa4a6d76d7e0",
            "score": 0.7754033207893372,
            "metadata": {
              "text": "multiple loci associated with susceptibility to type 2 diabetes, includ-\ningTCF7L2 (transcription factor 7-like 2), which had been originally\nidentied by a large-scale association mapping prompted by prior\nevidence of linkage in that area2,SLC30A8 (solute carrier family 30\nmember 8), HHEX (haematopoietically expressed homeobox),\nCDKAL1 (CDK5 regulatory subunit associated protein 1-like 1),\nCDKN2A/B (cyclin-dependent kinase inhibitor 2A/B) and IGF2BP2\n(insulin-like growth factor 2 mRNA-binding protein 2)37.",
              "title": "2008 - SNPs in KCNQ1 are associated with susceptibility to type 2 diabetes.pdf",
              "version": "v0",
              "chunk_order": 5,
              "document_id": "78702b1e-0f14-5757-b967-9bcb7852f6ac",
              "extraction_id": "706cb4a1-57c4-5b63-9d4e-4a7ea027a8f1",
              "associatedQuery": "What are the most common genetic loci associated with an increased risk of Type 2 diabetes?"
            }
          },
          {
            "id": "78d81651-7215-596a-b128-37e429dc7edb",
            "score": 0.7752246856689453,
            "metadata": {
              "text": "associated with susceptibility to type 2 diabetes mellitus. Nat Genet 2008; 40: 109297 .\n74 Unoki H, Takahashi A, Kawaguchi T, et al. SNPs in KCNQ1 are \nassociated with susceptibility to type 2 diabetes in East Asian and European populations. Nat Genet 2008; 40: 1098102.\n75 Lyssenko V, Lupi R, Marchetti P, et al. Mechanisms by which \ncommon variants in the TCF7L2 gene increase risk of type 2 diabetes. J Clin Invest 2007; 117: 215563. \n76 Lyssenko V, Jonsson A, Almgren P, et al. Clinical risk factors, DNA",
              "title": "2010 - Diabetes in Asia.pdf",
              "version": "v0",
              "chunk_order": 128,
              "document_id": "0be842b8-7f69-503b-baed-c336e5c834d6",
              "extraction_id": "11faf4fe-7b71-562e-9901-c428ab20b285",
              "associatedQuery": "What are the most common genetic loci associated with an increased risk of Type 2 diabetes?"
            }
          },
          {
            "id": "b0d3a09d-36a3-5c6e-a110-3fccddaa74b7",
            "score": 0.7739214531755884,
            "metadata": {
              "text": "type 2 diabetes or the inability to replicate linkage withdened loci. However, at least one susceptibility gene, namelyCAPN10, was found using a genome-wide scan approach [3].\nObesity is the greatest risk factor for type 2 diabetes\nmellitus, as it is known to induce insulin resistance via variousmechanisms ( TNF \n \n release, free fatty acids, etc.). Both",
              "title": "2003 - Genome-wide screen in obese pedigrees with type 2 diabetes.pdf",
              "version": "v0",
              "chunk_order": 5,
              "document_id": "335a3c08-14d3-5511-ab84-340e64c6f993",
              "extraction_id": "1110f7b4-ab5a-5b41-b37d-a992b29cb20c",
              "associatedQuery": "What are the most common genetic loci associated with an increased risk of Type 2 diabetes?"
            }
          },
          {
            "id": "8469faae-c6c9-5fd4-8437-870eef394dd1",
            "score": 0.7739064962975443,
            "metadata": {
              "text": "Clinical Risk Factors, DNA Variants, and the Development of Type 2 Diabetes\nn engl j med 359;21 www.nejm.org november 20, 2008 2231MPP subjects (P = 0.001) and from 0.79 to 0.83 in \nthe Botnia subjects (P = 0.006).\nOf the 16 loci that have been associated with \ntype 2 diabetes previously,8-15 we showed that 11 \n TCF7L2, PPARG, FTO, KCNJ11, NOTCH2, WFS1, \nCDKAL1, IGF2BP2, SLC30A8, JAZF1, and HHEX   \nwere associated with an enhanced risk of future",
              "title": "2008 - Clinical Risk Factors, DNA Variants.pdf",
              "version": "v0",
              "chunk_order": 95,
              "document_id": "0018610a-9c86-5e2d-a27d-f66cf4f8519d",
              "extraction_id": "0aae948a-50f9-568a-b0dc-5960a2d2ceaa",
              "associatedQuery": "What are the most common genetic loci associated with an increased risk of Type 2 diabetes?"
            }
          }
        ],
        "kg_search_results": null
      }
    }
  },
  "3": {
    "results": {
      "completion": {
        "id": "chatcmpl-AIFpJNprqmrM6nedwSTz4Aw1PacbM",
        "choices": [
          {
            "finish_reason": "stop",
            "index": 0,
            "message": {
              "content": "Genome-wide association studies (GWAS) help in identifying diabetes-related genes by examining associations between a large number of single-nucleotide polymorphisms (SNPs) and the disease, such as type 2 diabetes mellitus (T2DM) [1]. GWAS have successfully identified genetic loci associated with T2DM, providing insights into the genetic risk factors and biological pathways underlying the disease [2], [4]. These studies have identified hundreds of SNPs associated with T2DM, which can uncover previously unexpected disease pathways and contribute significantly to our understanding of the genetics of diabetes [5], [9]. Additionally, meta-analyses of GWAS data have improved the ability to detect new loci by combining findings from multiple studies, thereby increasing statistical power [2], [10]. Despite challenges such as the modest effect sizes of individual variants and stringent significance thresholds, GWAS remain a powerful tool for identifying genetic loci for complex diseases like diabetes [7], [10].",
              "role": "assistant"
            }
          }
        ],
        "created": 1728914105,
        "model": "gpt-4o-2024-08-06",
        "object": "chat.completion",
        "system_fingerprint": "fp_6b68a8204b",
        "usage": {
          "completion_tokens": 194,
          "prompt_tokens": 1163,
          "total_tokens": 1357
        }
      },
      "search_results": {
        "vector_search_results": [
          {
            "id": "b6827ec6-aa43-53e3-8d00-19e802bc3010",
            "score": 0.7417203408992395,
            "metadata": {
              "text": "BMC Medical Genomics  2009, 2:72 http://www.biomedcentral.com/1755-8794/2/72\nPage 2 of 8\n(page number not for citation purposes)Background\nGenome-wide association study (GWAS) offers unbiased\nways to examine association of more than a million singlenucleotide polymorphisms (SNPs) with disease [1]. Sev-eral GWAS have indentified novel genomic regions influ-encing risk for type 2 diabetes mellitus (T2DM) [2-6].However, the challenge remains to prioritize SNPs from",
              "title": "2009 - Prioritizing genes for follow-up from genome wide association studies using information on gene expression in tissues relevant for type 2 diabetes mellitus.pdf",
              "version": "v0",
              "chunk_order": 8,
              "document_id": "4b1a56e7-6821-5504-b6da-27dcdf57c6a5",
              "extraction_id": "e2b46a32-6616-55ad-8511-31ee8f9cce45",
              "associatedQuery": "How does genome-wide association studies (GWAS) help in identifying diabetes-related genes?"
            }
          },
          {
            "id": "9abaf02e-eee2-504d-be20-d589cb9a3164",
            "score": 0.740890405800653,
            "metadata": {
              "text": "GWAS have successfully identified genetic loci associ-\nated with a variety of conditions such as type 2 diabetes2\nand coronary disease.35The large number of statistical\ntests required in GWAS poses a special challenge because\nfew studies that have DNA and high-quality phenotypedata are sufficiently large to provide adequate statisticalpower for detecting small to modest effect sizes.\n6Meta-\nanalyses combining previously published findings have im-proved the ability to detect new loci.",
              "title": "2009 - Cohorts for Heart and Aging Research in Genomic.pdf",
              "version": "v0",
              "chunk_order": 14,
              "document_id": "9534989a-a5a5-52d8-95b8-0ad2926f228c",
              "extraction_id": "746e7837-d0f3-5a73-bfef-adfd748e35d6",
              "associatedQuery": "How does genome-wide association studies (GWAS) help in identifying diabetes-related genes?"
            }
          },
          {
            "id": "a1e3ca85-6fd1-5364-87c5-442c3f96ba74",
            "score": 0.7397869294691324,
            "metadata": {
              "text": "diabetes mellitus6,7. However, the traditional GWAS ignored a\nlarge number of loci with moderate effects, because of the strin-gent signi cance thresholds used.\nGene-based analysis takes a gene as a basic unit for\nassociation analysis. As this method can combine genetic\ninformation given by all the SNPs in a gene to obtain moreinformative results\n8, it is being used as a novel method com-\nplementing SNP-based GWAS to identify disease susceptibilitygenes. Notably, this method can increase our chance of nd-",
              "title": "2014 - Identification of novel risk genes associated with type 1 diabetes mellitus.pdf",
              "version": "v0",
              "chunk_order": 10,
              "document_id": "97fe33b0-a6c7-59b6-bd34-05528e77293f",
              "extraction_id": "4b1681f4-4088-5b15-a704-040e35e31080",
              "associatedQuery": "How does genome-wide association studies (GWAS) help in identifying diabetes-related genes?"
            }
          },
          {
            "id": "263ea999-9662-5518-a606-939f69d09f90",
            "score": 0.7390662651975833,
            "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 does genome-wide association studies (GWAS) help in identifying diabetes-related genes?"
            }
          },
          {
            "id": "53c3668c-95f8-5fb9-b978-e4c03ddfa40f",
            "score": 0.7377198714697428,
            "metadata": {
              "text": "that a genome-wide approach could uncover \npreviously unexpected disease pathways.\nIn early 2007, GW AS provided by far the \nbiggest increment to date in our knowledge of \nthe genetics of this common health problem.\nSix new gene regions identified\nTogether, the six recent GW AS papers \nprovide convincing evidence for six new \ngene regions involved in type\n \n2 diabetes1621; \na seventh publication describes how one \nof these variants alters BMI and represents by far the best example of an association",
              "title": "2007 - Genome\u2013wide association studies provide new insights into type 2 diabetes aetiology..pdf",
              "version": "v0",
              "chunk_order": 12,
              "document_id": "2ad9b6c6-56ed-5ba6-ad88-c1a6777f5196",
              "extraction_id": "aa94128a-99f6-59f3-b5fa-33ac97b858d5",
              "associatedQuery": "How does genome-wide association studies (GWAS) help in identifying diabetes-related genes?"
            }
          },
          {
            "id": "7fd80e84-ec0c-564c-8e8b-278b8c622abb",
            "score": 0.734627821009525,
            "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 does genome-wide association studies (GWAS) help in identifying diabetes-related genes?"
            }
          },
          {
            "id": "9afcf9a9-3abf-5441-a711-55e25f1ef9b7",
            "score": 0.733059688392859,
            "metadata": {
              "text": "mechanisms of DR remain poorly understood.\nA genome-wide association study (GWAS) is a powerful tool\nto identify genetic loci for complex diseases, and a large number\nof genetic loci for the susceptibility to various diseases, such astype 2 diabetes, have been successfully identified through GWAS\n(69). GWAS for DR have been performed, but most of the studies\nonly reported suggestive signals with no replication ( 5)b e c a u s e\nof their limited sample sizes. Recently, several loci with genome-",
              "title": "2021 - Genome-wide association studies identify two novel loci.pdf",
              "version": "v0",
              "chunk_order": 15,
              "document_id": "7131256d-7d55-597d-aac5-a62956736923",
              "extraction_id": "fc9812ae-7b35-5dac-af9b-6d60f4faaa54",
              "associatedQuery": "How does genome-wide association studies (GWAS) help in identifying diabetes-related genes?"
            }
          },
          {
            "id": "ad7955f2-824c-59f8-8357-6ee201756ec9",
            "score": 0.7320249894864858,
            "metadata": {
              "text": "kidney disease, several loci have been identi ed and validated,\nbut the results were quite heterogenic across different popula-\ntions and depended on the type of diabetes and stage of disease.\nThe major bene t of GWAS results is to be found in the in-\ncreased understanding of disease mechanism and identi ca-\ntion of novel pathways and possibly new therapeutic targets.Follow-up studies are important in order to identify variants\nwith speci c biological effect and may provide important",
              "title": "2015 - Genome-wide studies to identify risk factors for kidney disease.pdf",
              "version": "v0",
              "chunk_order": 77,
              "document_id": "3e696b99-6306-5429-bce9-8d04a2471b2d",
              "extraction_id": "92bd58f8-6770-5c1c-8202-19b08bd57df8",
              "associatedQuery": "How does genome-wide association studies (GWAS) help in identifying diabetes-related genes?"
            }
          },
          {
            "id": "5488da5b-5efa-55cd-92c3-a0d77e587fce",
            "score": 0.7298565128402866,
            "metadata": {
              "text": "Abstract\nGenome-wide association studies (GWASs) have identified hundreds of single nucleotide polymorphisms (SNPs) associated with type 2 diabetes (T2D) and coronary artery disease (CAD), respectively. Nevertheless, these studies were generally per -",
              "title": "2020 - Identification of novel functional CpG-SNPs associated with type 2 diabetes and coronary artery disease..pdf",
              "version": "v0",
              "chunk_order": 1,
              "document_id": "f0385a45-ad3e-5813-ab1f-b3e227d5164b",
              "extraction_id": "2341dbc6-8084-5d51-a52e-f8f667b79bbb",
              "associatedQuery": "How does genome-wide association studies (GWAS) help in identifying diabetes-related genes?"
            }
          },
          {
            "id": "7f17fa56-1b7a-5d51-a111-3c74b31a5821",
            "score": 0.7289577343749328,
            "metadata": {
              "text": "linkage or association data. But, none of these studies include in\nthe analysis existing data from GWAs.\nFinally, a recent study identied additional susceptibility loci\nfor type 2 diabetes by performing a meta-analysis of three\npublished GWAs.21As acknowledged by the authors, GWAs\nare limited by the modest effect sizes of individual common\nvariants and the need for stringent statistical thresholds. Thus,\nby combining data involving 10,128 samples, the authors found",
              "title": "2009 - Gene prioritization based on biological plausibility over genome wide association studies renders new loci associated with type 2 diabetes.pdf",
              "version": "v0",
              "chunk_order": 63,
              "document_id": "0fd2b5c8-9bda-5cc8-adb4-231d3842d50f",
              "extraction_id": "0c5401ea-2a43-5578-af0b-6ad1e818fa42",
              "associatedQuery": "How does genome-wide association studies (GWAS) help in identifying diabetes-related genes?"
            }
          }
        ],
        "kg_search_results": null
      }
    }
  },
  "4": {
    "results": {
      "completion": {
        "id": "chatcmpl-AIFpPxGF7qh3CvkVYK1PXW0J3bHPe",
        "choices": [
          {
            "finish_reason": "stop",
            "index": 0,
            "message": {
              "content": "The HLA region plays a significant role in the genetic predisposition to Type 1 diabetes. The major genetic susceptibility determinants for Type 1 diabetes have been mapped to the MHC class II genes, specifically HLA-DQB1 and HLA-DRB1 [7]. These genes are part of the highly polymorphic HLA class II genes, which play the most important single role in susceptibility to Type 1 diabetes [3]. The class II genes encoding HLA-DR and HLA-DQ, along with one or more additional genes within the HLA region, confer most of the genetic risk for Type 1 diabetes [4]. The contribution of HLA genes to Type 1 diabetes susceptibility accounts for about 44% of the genetic risk [6]. However, variation at these loci alone cannot explain all of the genetic association and linkage of the MHC with Type 1 diabetes, indicating that other genes within the MHC region may also affect Type 1 diabetes risk [3].",
              "role": "assistant"
            }
          }
        ],
        "created": 1728914111,
        "model": "gpt-4o-2024-08-06",
        "object": "chat.completion",
        "system_fingerprint": "fp_6b68a8204b",
        "usage": {
          "completion_tokens": 201,
          "prompt_tokens": 1519,
          "total_tokens": 1720
        }
      },
      "search_results": {
        "vector_search_results": [
          {
            "id": "f654b4ad-31d8-528c-800d-000a0f56438a",
            "score": 0.7919327020645142,
            "metadata": {
              "text": "conferred by specic alleles, genotypes, and haplotypes ofthe HLA class II (and class I) genes. There are currentlyabout 50 non-HLA region loci that also affect the type 1diabetes risk. Many of the assumed functions of thenon-HLA genes of interest suggest that variants at theseloci act in concert on the adaptive and innate immunesystems to initiate, magnify, and perpetuate /H9252-cell destruc-",
              "title": "2010 - Genetics of Type 1 Diabetes What\u2019s Next.pdf",
              "version": "v0",
              "chunk_order": 118,
              "document_id": "261cbb40-ed6b-554c-a70d-db6b9f14cf74",
              "extraction_id": "c584fa40-ecb0-57e0-8bd1-6761ae3b4d27",
              "associatedQuery": "What is the role of the HLA region in the genetic predisposition to Type 1 diabetes?"
            }
          },
          {
            "id": "8480cb40-fb38-5f5e-98c5-41eb2b3bdad2",
            "score": 0.7839834094047546,
            "metadata": {
              "text": "II HLA gene associated with type 1 diabetes maps to the 240-kbregion near HLA-B. Diabetes 49: 22172221, 2000.\n303. Nejentsev S, Howson JM, Walker NM, Szeszko J, Field SF.\nLocalization of type 1 diabetes susceptibility to the MHC class Igenes HLA-B and HLA-A. Nature 450: 887892, 2007.\n304. Nejentsev S, Walker N, Riches D, Egholm M, Todd JA. Rare\nvariants of IFIH1, a gene implicated in antiviral responses, protectagainst type 1 diabetes. Science 324: 387389, 2009.",
              "title": "2011 - Type 1 Diabetes Etiology, Immunology.pdf",
              "version": "v0",
              "chunk_order": 535,
              "document_id": "3c9823cd-3615-53b6-96c8-b7d2123d3eb0",
              "extraction_id": "36ba6e4a-46c7-5a84-89ca-2b6e2a3c2a2c",
              "associatedQuery": "What is the role of the HLA region in the genetic predisposition to Type 1 diabetes?"
            }
          },
          {
            "id": "5157af28-d09f-5bbb-8984-61da49bed642",
            "score": 0.7792037725448608,
            "metadata": {
              "text": "Although the highly polymorphic HLA class II genesclearly play the most important single role in susceptibilityto type 1 diabetes, variation at these loci alone cannotexplain all of the evidence of genetic association andlinkage of the MHC with type 1 diabetes. To better denegenes within the MHC that may affect type 1 diabetes riskand would therefore merit further studies, the T1DGCundertook a comprehensive study of the genetics of theclassic 4-Mb MHC region. More than 3,000 SNPs and 66microsatellite",
              "title": "2010 - Genetics of Type 1 Diabetes What\u2019s Next.pdf",
              "version": "v0",
              "chunk_order": 21,
              "document_id": "261cbb40-ed6b-554c-a70d-db6b9f14cf74",
              "extraction_id": "508a0315-d23a-506d-bf66-b5c355576f96",
              "associatedQuery": "What is the role of the HLA region in the genetic predisposition to Type 1 diabetes?"
            }
          },
          {
            "id": "5c650a7a-98f1-5119-b66f-5a93db18faec",
            "score": 0.77757328748703,
            "metadata": {
              "text": "age to type 1 diabetes in the HLA region and suggestive evidence at a small number of other regions in the genome. In general, the emerging picture from linkage studies is that the class II genes encoding HLA-DR and HLA-DQ, as well as one or more additional genes within the HLA re -\ngion, confer most of the genetic risk for type 1 dia -\nbetes. Genes outside the HLA region also con -\ntribute to the risk of type 1 diabetes, but their individual contributions are much smaller than that of HLA.",
              "title": "2009 - Genetics of Type 1A Diabetes.pdf",
              "version": "v0",
              "chunk_order": 16,
              "document_id": "7a98f456-6c43-5e9e-b404-31122159eab8",
              "extraction_id": "79b78db5-d77a-51bd-84ff-14b558534df8",
              "associatedQuery": "What is the role of the HLA region in the genetic predisposition to Type 1 diabetes?"
            }
          },
          {
            "id": "0fa5241d-e039-55b3-ba8c-aa14d0125967",
            "score": 0.7742921113967896,
            "metadata": {
              "text": "Benkalha and Polychronakos, 2008 ). Other genetic loci ( Table 1) are\nbelieved to in uence population-level risk for T1D, although it is poorly\nunderstood how these non-HLA loci contribute to disease susceptibility\n(Ram et al., 2016a ).\n2.1. Human leukocyte antigen (HLA)\nThe association between T1D and the HLA complex was rst de-\nmonstrated in 1973 following observation of an increased frequency ofHL-W15 (HLA antigen) in T1D patients compared to controls ( Singal",
              "title": "2018 - The genetic architecture of type 1 diabetes mellitus.pdf",
              "version": "v0",
              "chunk_order": 18,
              "document_id": "341261db-b38a-5bd2-8d8d-fc04a0b3da30",
              "extraction_id": "65402f67-0066-5d04-98c6-5e7b16087495",
              "associatedQuery": "What is the role of the HLA region in the genetic predisposition to Type 1 diabetes?"
            }
          },
          {
            "id": "f159c8b5-357c-57f6-98e4-5d5436f59925",
            "score": 0.7736279964447021,
            "metadata": {
              "text": "cyte Antigen (HLA) gene region in immune regulation, and ready availability of\nserologic markers, led investigators to discover the association between certainHLAalleles and T1D in the early 1970s (33,130,158). The global importance of\ntheHLAonT1Dhassincebeenconrmedingenome-widescansforlinkage:All\nsuchscansperformedtodateshowamajorlocusatthe HLA(28,32,36,78,119).\nThefractionofallgeneticrisk,whichcanbeattributedtothecontributionof HLA\ngenes to T1D susceptibility, is about 44%, with a \nSof3.4 (160).",
              "title": "2003 - The Inherited Basis of Diabetes Mellitus.pdf",
              "version": "v0",
              "chunk_order": 26,
              "document_id": "7b85b290-d711-55d5-9b1e-b06e4d6f14a2",
              "extraction_id": "548d7a8d-5314-53f3-b646-108a4b1ac2f2",
              "associatedQuery": "What is the role of the HLA region in the genetic predisposition to Type 1 diabetes?"
            }
          },
          {
            "id": "277e2627-b99d-5b35-ae45-1fbaa2bf0710",
            "score": 0.7721036076545715,
            "metadata": {
              "text": "The major histocompatibility complex (MHC) on chromosome 6\nis associated with susceptibility to more common diseases than\nany other region of the human genome, including almost all dis-\norders classified as autoimmune. In type 1 diabetes the major\ngenetic susceptibility determinants have been mapped to the\nMHC class II genes HLA-DQB1 andHLA-DRB1 (refs 13), but\nthese genes cannot completely explain the association between\ntype 1 diabetes and the MHC region411.Owing to the regions",
              "title": "2007 - Localization of type 1 diabetes susceptibility to the MHC Class 1 Genes.pdf",
              "version": "v0",
              "chunk_order": 1,
              "document_id": "3887995f-fa61-5472-b0a2-90b7b39592c2",
              "extraction_id": "0ed848eb-6e55-5fab-82d0-f10b3c83d98f",
              "associatedQuery": "What is the role of the HLA region in the genetic predisposition to Type 1 diabetes?"
            }
          },
          {
            "id": "69fb55b3-37a8-5fb6-9916-2ab5be15a0a8",
            "score": 0.7700294256210327,
            "metadata": {
              "text": "The HLA class I A locus a ects susceptibility to type 1 diabetes. Hum. Immunol. 63,\n657 664. pii). https://doi.org/S0198885902004214 .\nNoble, J.A., Valdes, A.M., Cook, M., Klitz, W., Thomson, G., Erlich, H.A., 1996. The role of\nHLA class II genes in insulin-dependent diabetes mellitus: molecular analysis of 180\nCaucasian, multiplex families. Am. J. Hum. Genet. 59, 1134 1148 .\nNoble, J.A., Valdes, A.M., Thomson, G., Erlich, H.A., 2000. The HLA class II locus DPB1",
              "title": "2018 - The genetic architecture of type 1 diabetes mellitus.pdf",
              "version": "v0",
              "chunk_order": 147,
              "document_id": "341261db-b38a-5bd2-8d8d-fc04a0b3da30",
              "extraction_id": "dd2e688a-2f06-5911-9eb8-13c5d935885e",
              "associatedQuery": "What is the role of the HLA region in the genetic predisposition to Type 1 diabetes?"
            }
          },
          {
            "id": "3ccbc6c4-a2a3-53ab-b904-c4d5875e2e2c",
            "score": 0.7676813006401062,
            "metadata": {
              "text": "to type 1diabetes susceptibility, including within the MHC itself.Currently, there are over 50 non-HLA regions that signi-cantly affect the risk for type 1 diabetes (http://www.t1dbase.org). Many of these regions contain interesting,but previously unrecognized, candidate genes. A few re-gions contain genes of unknown function or no knownannotated genes, suggesting roles for long-distance generegulatory effects, noncoding RNAs, or unknown mecha-nisms. Against a background of ever-improving knowledgeof the",
              "title": "2010 - Genetics of Type 1 Diabetes What\u2019s Next.pdf",
              "version": "v0",
              "chunk_order": 2,
              "document_id": "261cbb40-ed6b-554c-a70d-db6b9f14cf74",
              "extraction_id": "3e3ff61a-86ee-5e2a-9b14-9aa3b06937ff",
              "associatedQuery": "What is the role of the HLA region in the genetic predisposition to Type 1 diabetes?"
            }
          },
          {
            "id": "2b1f2a05-4693-595c-94c0-fea40e19539c",
            "score": 0.7664471864700317,
            "metadata": {
              "text": "the 240-kb region near HLA-B. Diabetes 49,22172221 (2000).\n6. Lie, B. A. et al. The predisposition to type 1 diabetes linked to the human leukocyte\nantigen complex includes at least one non-class II gene. Am. J. Hum. Genet. 64,\n793800 (1999).\n7. Valdes, A. M. et al. Extended DR3 D6S273-HLA-B haplotypes are associated\nwith increased susceptibility to type 1 diabetes in US Caucasians. Tissue Antigens\n65,115119 (2005).\n8. Valdes, A. M., Erlich, H. A. & Noble, J. A. Human leukocyte antigen class I B and C",
              "title": "2007 - Localization of type 1 diabetes susceptibility to the MHC Class 1 Genes.pdf",
              "version": "v0",
              "chunk_order": 49,
              "document_id": "3887995f-fa61-5472-b0a2-90b7b39592c2",
              "extraction_id": "92a54171-9f94-51ea-83cb-11698b1f0c21",
              "associatedQuery": "What is the role of the HLA region in the genetic predisposition to Type 1 diabetes?"
            }
          }
        ],
        "kg_search_results": null
      }
    }
  },
  "5": {
    "results": {
      "completion": {
        "id": "chatcmpl-AIFpUuEUTWxzzcta8xK3fjxfSUNPx",
        "choices": [
          {
            "finish_reason": "stop",
            "index": 0,
            "message": {
              "content": "Genetic differences contribute to variations in diabetes prevalence among different populations in several ways:\n\n1. **Genetic Heterogeneity**: Different genes may be responsible for the development of type 2 diabetes mellitus (T2DM) in different populations, indicating genetic heterogeneity. Even within the same ethnic group, different genes might be responsible for different subtypes of diabetes, such as those with predominating failure in insulin secretion or insulin resistance [3].\n\n2. **Ethnicity-Specific Genetic Variants**: The pathogenesis of T2DM involves genetic variants in candidate genes, and these variants can show ethnicity-specific differences. The interactions between genes involved in insulin signaling and secretory pathways play a crucial role in determining an individual's susceptibility to T2DM [4].\n\n3. **Single Nucleotide Polymorphisms (SNPs) and Haplotypes**: Minute genetic differences, such as SNPs and haplotypes, are used to explain patterns of disease between populations. These genetic markers are understood as the basis for biological differences between populations, which can affect diabetes prevalence [5].\n\n4. **Risk Allele Frequencies**: Human populations exhibit substantial variation in risk allele frequencies, which contributes to different susceptibilities to diseases, including diabetes, among populations [7].\n\n5. **Insulin Secretion Capacity**: In some populations, such as the Asian population, genetic influences on the prevalence of type 2 diabetes are mainly related to insulin secretion capacity. This suggests that genes associated with insulin secretion capacity are particularly relevant in contributing to ethnic differences in diabetes prevalence [10].\n\nOverall, genetic differences, including variations in specific genes, SNPs, and risk allele frequencies, contribute to the observed variations in diabetes prevalence among different populations. These genetic factors interact with environmental and lifestyle factors to influence diabetes risk.",
              "role": "assistant"
            }
          }
        ],
        "created": 1728914116,
        "model": "gpt-4o-2024-08-06",
        "object": "chat.completion",
        "system_fingerprint": "fp_6b68a8204b",
        "usage": {
          "completion_tokens": 361,
          "prompt_tokens": 1133,
          "total_tokens": 1494
        }
      },
      "search_results": {
        "vector_search_results": [
          {
            "id": "49748fe8-4351-5cd1-8367-957a160a59d9",
            "score": 0.7598252522893328,
            "metadata": {
              "text": "of diabetes when compared to the native population while not necessar-ily different from populations where they origi-nate from. Risk factors for diabetes appear to be similar between populations, mostly insulin resistance, obesity, and sedentary lifestyle with possible genetic differences contributing to the increased susceptibility. Some data suggest a greater prevalence of microvascular complica-",
              "title": "2017 - diabetes-mellitus-in-developing-countries-and-underserved-commun-2017.pdf",
              "version": "v0",
              "chunk_order": 2135,
              "document_id": "8a9451b9-d7e8-5417-b6a5-5fd1b791cc4d",
              "extraction_id": "61fb4dd8-1428-5add-8c41-9ec2459ffd5a",
              "associatedQuery": "How do genetic differences contribute to variations in diabetes prevalence among different populations?"
            }
          },
          {
            "id": "80ad1f9c-4f67-5a68-9446-1f692b23f324",
            "score": 0.7463788167721416,
            "metadata": {
              "text": "nants of type 2 diabetes between immigrant and native populations. Some studies in South Asian (Indian) populations suggest that genetic differ-ences may exist [ \n17 ,  30 ], but larger studies are \nneeded to get better insight into this issue.  \n    Prevalence Estimates \n The prevalence of diabetes in minorities is affected by ethnicity and country of residence. In one study in the UK [ \n59 ], standardized preva-",
              "title": "2017 - diabetes-mellitus-in-developing-countries-and-underserved-commun-2017.pdf",
              "version": "v0",
              "chunk_order": 2155,
              "document_id": "8a9451b9-d7e8-5417-b6a5-5fd1b791cc4d",
              "extraction_id": "090365f1-32e0-5adc-b589-b9331e0630a0",
              "associatedQuery": "How do genetic differences contribute to variations in diabetes prevalence among different populations?"
            }
          },
          {
            "id": "5fd9c60a-410f-5782-90a9-03d377a5f72b",
            "score": 0.7383460182444477,
            "metadata": {
              "text": "majority of cases it is difficult to replicate the findingsin other populations. One of the major problems in thesearch for genes responsible for common forms ofdiabetes is the genetic heterogeneity of the diseasewith different genes responsible for the developmentof T2DM in different populations. Furthermore, evenwithin the same ethnic group, different genes may beresponsible for different subtypes of diabetes (for in-stance with predominating failure in insulin secretionor insulin resistance). This is",
              "title": "2005 - Type 2 diabetes mellitus from genes to disease.pdf",
              "version": "v0",
              "chunk_order": 40,
              "document_id": "52687a38-6a4b-51d2-aafa-812c76981dfe",
              "extraction_id": "73278198-67af-5556-9414-86580dd07c48",
              "associatedQuery": "How do genetic differences contribute to variations in diabetes prevalence among different populations?"
            }
          },
          {
            "id": "d02a16ce-c62e-537d-9d32-266018c70415",
            "score": 0.7162386392426089,
            "metadata": {
              "text": "across different races or populations but show ethnicity-\nspecific differences. The pathogenesis of T2D involves \ngenetic variants in the candidate genes. The interactions \nbetween the genes involved in insulin signaling and secre -\ntory pathways are believed to play an important role in \ndetermining an individuals susceptibility towards T2D. \nTherefore, the present study was initiated to examine the \ndifferences, if any, in the contribution of polymorphisms",
              "title": "2016 - Association of genetic variants in INS (rs689), INSR (rs1799816) and PP1G.G (rs1799999) with type 2 diabetes (T2D) a case\u2013control study in three ethnic groups from N.pdf",
              "version": "v0",
              "chunk_order": 5,
              "document_id": "5fe7c5f4-a209-56be-8504-c08073335c3b",
              "extraction_id": "4cbd4dfc-da8e-5432-b844-5f70d6f3811d",
              "associatedQuery": "How do genetic differences contribute to variations in diabetes prevalence among different populations?"
            }
          },
          {
            "id": "684d1e26-b78a-5dde-b405-a79ee28087c3",
            "score": 0.714162013356746,
            "metadata": {
              "text": "That is, the minute genetic differences discernable with SNPs, patterns of single nu-cleotides (A,G,T ,C), and other mutation analysis technologies are now used to explainpatterns of disease between populations, which are in turn understood as the basisfor biological differences between the populations themselves. The case of diabetesgenetics research affords a more nuanced look at what is labeled genetic determinism.It is evident in diabetes research that SNPs and haplotypes, (an inherited pattern of\n99",
              "title": "2007 - Bioethnic Conscription Genes, Race.pdf",
              "version": "v0",
              "chunk_order": 51,
              "document_id": "d90126d9-fd87-5b38-87f7-08415f690836",
              "extraction_id": "95f0e6f8-da7d-5997-ab8a-a1aad020c706",
              "associatedQuery": "How do genetic differences contribute to variations in diabetes prevalence among different populations?"
            }
          },
          {
            "id": "8445ab0a-2287-5537-ab3a-cb058205e944",
            "score": 0.7116393871092431,
            "metadata": {
              "text": "-\ntion for disease classification. This genetic component may be specifically important when understanding the pathogenesis of diabetes in ethnic groups, when BMI [14, 15] and HbA1c [16] show distinct differences between ethnicities. Though applying patient-matched, genomic information is currently unrealistic for disease diagnosis, it may hold the key for revealing commonalities across ethnic and demographic groups when classifying diabetic onset, progression, and severity.",
              "title": "2019 - Machine-learning to stratify diabetic patients using novel cardiac biomarkers and integrative genomics.pdf",
              "version": "v0",
              "chunk_order": 21,
              "document_id": "332ac2ec-accc-5370-a4d2-6fec9ce7e072",
              "extraction_id": "8d323598-fdf7-56cf-8290-be85929f0eaf",
              "associatedQuery": "How do genetic differences contribute to variations in diabetes prevalence among different populations?"
            }
          },
          {
            "id": "10c1db42-f724-5885-99e0-7637dfce63ca",
            "score": 0.7018723697028273,
            "metadata": {
              "text": "particularly useful for understanding differences in dis-ease prevalence and drug response among differentpopulations. There is ample evidence that human popu-lations have different susceptibility to diseases, exhibit-ing substantial variation in risk allele frequencies [1].For example, genetic predisposition to asthma differsamong the differentially-admixed Hispanic populations\nof the United States, with the highest prevalence\nobserved in Puerto Ricans. Ge netic variants responsible",
              "title": "2011 - Dating the age of admixture via wavelet.pdf",
              "version": "v0",
              "chunk_order": 2,
              "document_id": "786cebc5-c3cc-586e-bdc0-e7bee67edc19",
              "extraction_id": "a5c137e5-84d2-5d75-8191-fa6b0be3d39e",
              "associatedQuery": "How do genetic differences contribute to variations in diabetes prevalence among different populations?"
            }
          },
          {
            "id": "d29cdd31-d214-52cf-b236-be4de1182b26",
            "score": 0.7005400866431026,
            "metadata": {
              "text": "populations and across countries. World-wide differences in prevalence of theforms of diabetes necessitates inclusion\nof currently understudied populationsfor the development of precision diag-nostics and therapeutics. As a result, theprecise subtype of diabetes a particularindividual is diagnosed with may vary indifferent populations based on subtypefrequency or genetic or dietary or life-style differences.\nThe communication strategy used by\nthe interventionalist and the patient s",
              "title": "2020 - Precision Medicine in Diabetes.pdf",
              "version": "v0",
              "chunk_order": 182,
              "document_id": "0ad5b2de-d782-5d43-b294-bff5c7befd2d",
              "extraction_id": "9dc25bb6-787b-5e7a-af5d-d1353d122959",
              "associatedQuery": "How do genetic differences contribute to variations in diabetes prevalence among different populations?"
            }
          },
          {
            "id": "6fd138d2-6960-55fd-b656-05f4e84a0c6d",
            "score": 0.6991221696400024,
            "metadata": {
              "text": "were  positively  associated  with  country  level  income  [49].\nHowever,  the  drivers  for  the  observed  pattern  with  geographi-\ncal  differences  and  varying  time  trends  are  still  unclear.\nSusceptibility  to  type  1  diabetes  denitely  has  a  strong  genetic\ncomponent  (HLA  genotype)  [50],  but  the  heterogeneity  of  type\n1  diabetes  cannot  be  explained  solely  by  the  prevalence  of\nsusceptibility  genes  [5153] .  Thus,  the  reasons  for  changes  in",
              "title": "2014 - Diabetes in Europe An update.pdf",
              "version": "v0",
              "chunk_order": 35,
              "document_id": "81e1fc53-6768-590f-9b47-9a5105b6ddb5",
              "extraction_id": "fa58324a-e5b7-538e-9cbb-0549887a2154",
              "associatedQuery": "How do genetic differences contribute to variations in diabetes prevalence among different populations?"
            }
          },
          {
            "id": "2771c343-be7b-51a2-a598-235647357416",
            "score": 0.6972634611882842,
            "metadata": {
              "text": "twice higher than that of 2010  [3] .\n  The genetic influences on the prevalence of type 2 diabetes i n the Asian population are \nmainly related to insulin secretion capacity  [4] ; other genes involved in the risk of type 2 \ndiabetes are not substantially different in other ethnic groups   [5] . The most relevant genes \ncontributing to ethnic differences are associated with insulin secretion capacity, and they are",
              "title": "2016 - TRPV1 Gene Polymorphisms Are Associated with Type 2 Diabetes by Their Interaction with Fat Consumption in the Korean Genome Epidemiology Study.pdf",
              "version": "v0",
              "chunk_order": 9,
              "document_id": "521db985-2ce8-56c3-aed7-b38ef41cce45",
              "extraction_id": "8276c974-f60b-5f59-943d-94a635160d1d",
              "associatedQuery": "How do genetic differences contribute to variations in diabetes prevalence among different populations?"
            }
          }
        ],
        "kg_search_results": null
      }
    }
  },
  "6": {
    "results": {
      "completion": {
        "id": "chatcmpl-AIFpdRa9QE6LvKot0urXoNDlpAF5x",
        "choices": [
          {
            "finish_reason": "stop",
            "index": 0,
            "message": {
              "content": "The PPAR-gamma gene encodes a transcription factor that plays a crucial role in regulating insulin sensitivity and lipid metabolism, which are important in the context of diabetes [4]. PPAR-gamma influences insulin sensitivity partly by modulating circulating adiponectin levels, with PPAR-gamma agonists known to increase these levels [1]. This gene is a target for the thiazolidinedione class of oral antidiabetic agents, which are used to improve insulin sensitivity in patients with type 2 diabetes [4].\n\nVariants of the PPAR-gamma gene can significantly impact the risk and progression of type 2 diabetes. The Pro12Ala variant, for example, is a well-established genetic variant that modulates insulin sensitivity and increases the risk of type 2 diabetes [1], [4]. Other pathogenic PPARG variants, such as R194Q, A417V, R212W, P387S, M203I, and T356R, have been identified in patients with partial lipodystrophy and are associated with an increased risk of type 2 diabetes [7]. These variants can affect the gene's function in adipocyte differentiation and metabolism, thereby influencing the development of insulin resistance and type 2 diabetes [10].",
              "role": "assistant"
            }
          }
        ],
        "created": 1728914125,
        "model": "gpt-4o-2024-08-06",
        "object": "chat.completion",
        "system_fingerprint": "fp_e5e4913e83",
        "usage": {
          "completion_tokens": 250,
          "prompt_tokens": 1427,
          "total_tokens": 1677
        }
      },
      "search_results": {
        "vector_search_results": [
          {
            "id": "6f046969-4e26-5dee-a310-cf32dc1f799c",
            "score": 0.741815999638082,
            "metadata": {
              "text": "The transcription factor peroxisome-proliferator-\nactivated receptor gamma (PPAR g) is known to\ninuence insulin sensitivity, and acts partly via amodulation of the circulating adiponectin level\n(PPAR gagonists increase the adiponectin level)\n(Ref. 38). The PPAR gP12A SNP is a well-\nestablished genetic variant that modulates\ninsulin sensitivity and the risk of type 2 diabetes\n(Ref. 39). In a Chinese family study, Yang et al.demonstrated a genetic interaction between the",
              "title": "2006 - Adiponectin, type 2 diabetes and the metabolic syndrome.pdf",
              "version": "v0",
              "chunk_order": 35,
              "document_id": "6a46f7cf-e75b-5b72-b77b-7e0cc03f92d8",
              "extraction_id": "4647b43a-e4a0-5e8a-9cf5-6bf33cd6e672",
              "associatedQuery": "What is the function of the PPAR-gamma gene in diabetes, and how do its variants impact the disease?"
            }
          },
          {
            "id": "c909cc5c-6fdb-5646-8332-973a92ac9486",
            "score": 0.7338819066356489,
            "metadata": {
              "text": "intricate regulation of PPAR signaling to pave the way to tailored therapies in patients with insulin resistance and T2D.\nKeywords PPARG genetic variants .Dominant-negative isoforms .Post-tranlational modifications .Adipose tissue\ndysfunctions .Drug responsiveness .Type 2 diabetes\nIntroduction\nPeroxisome proliferator activated receptor gamma (PPAR )\nis a ligand-activated transcription factor belonging to the nu-",
              "title": "2021 - PPAR\u03b3 and Diabetes Beyond the Genome and Towards Personalized Medicine.pdf",
              "version": "v0",
              "chunk_order": 5,
              "document_id": "4ea83190-476d-5090-a461-abde1adccbc5",
              "extraction_id": "2d610953-ea5c-5c01-ad19-60c607383da4",
              "associatedQuery": "What is the function of the PPAR-gamma gene in diabetes, and how do its variants impact the disease?"
            }
          },
          {
            "id": "c3ac7ed2-1b42-5c87-9104-b6da2e33b30b",
            "score": 0.7244109295484176,
            "metadata": {
              "text": "2 . A widespread \nGly482Ser polymorphism of PGC1 -    (known as  PPARGC1  ), a \ntranscriptional coactivator of a series of nuclear receptors includ-ing  PPARG  , has been associated with a 1.34 genotype relative risk \nof T2DM  [93] . In this study, a test for interaction with the Pro12Ala variant in  PPARG   gave no indication for additive effects \non diabetes status. \n Other genes have been shown to be implicated in the genetic",
              "title": "2010 - The Genetics of Type 2 Diabetes.pdf",
              "version": "v0",
              "chunk_order": 91,
              "document_id": "a1d211d4-279e-51d7-b2b2-33bc2763d089",
              "extraction_id": "1df8f645-85c4-5832-8142-09bacafcd01d",
              "associatedQuery": "What is the function of the PPAR-gamma gene in diabetes, and how do its variants impact the disease?"
            }
          },
          {
            "id": "02a160ba-95ee-5aa9-bc45-445b4706715b",
            "score": 0.721716182129612,
            "metadata": {
              "text": "PPARG Peroxisome proliferator-activated receptor- gene. This gene is located on chromosome 3p25, and has been studied as a candidate genefor type 2 diabetes based on its role in adipocyte and lipid metabolism. The Pro12Ala variant in particular has been associated with adecrease in insulin sensitivity and a several-fold increased risk of type 2 diabetes. PPAR is a target for the thiazolidinedione class of oralantidiabetic agents",
              "title": "2008 - Glossary of Genetics Genomics Terms.pdf",
              "version": "v0",
              "chunk_order": 32,
              "document_id": "c66d2572-071d-5aaf-829c-b3ca6cf6d697",
              "extraction_id": "f8b79de5-3e0c-5495-b6c2-8a3be6138223",
              "associatedQuery": "What is the function of the PPAR-gamma gene in diabetes, and how do its variants impact the disease?"
            }
          },
          {
            "id": "4e415210-bf41-542f-841c-4bb17622d2e6",
            "score": 0.720844106623883,
            "metadata": {
              "text": "Genetic variation in the peroxisome proliferator-activated receptor (PPAR) and peroxisome proliferator-activated receptor gamma co-activator 1 (PGC1) gene families and type 2 diabetes. Ann Hum Genet 78:2332\nVimaleswaran KS, Radha V, Ghosh S, Majumder PP, Deepa R, Babu \nHN etal (2005) Peroxisome proliferator-activated receptor-gamma \nco-activator-1alpha (PGC-1alpha) gene polymorphisms and their \nrelationship to type 2 diabetes in Asian Indians. Diabetic Med 22:15161521",
              "title": "2018 - Association of PGC-1\u03b1 gene with type 2 diabetes in three unrelated endogamous groups of North-West India (Punjab) a case-control and meta-analysis study.pdf",
              "version": "v0",
              "chunk_order": 116,
              "document_id": "2faa21d2-146e-568a-b881-63201819e99a",
              "extraction_id": "94ee1317-d606-5921-8175-a86da2fa95d6",
              "associatedQuery": "What is the function of the PPAR-gamma gene in diabetes, and how do its variants impact the disease?"
            }
          },
          {
            "id": "8d7fb270-e23f-5d89-b75c-50b8fbd22fe8",
            "score": 0.7202155160775486,
            "metadata": {
              "text": "Dali-Youcef N, et al. The Pro12Ala PPARgamma2 variant deter-\nmines metabolism at the gene-environment interface. Cell Metab.\n2009;9:88 98.\n53. Agostini M, Schoenmakers E, Mitchell C, Szatmari I, Savage D,\nSmith A, et al. Non-DNA binding, dominant-negative, human\nPPARgamma mutations cause lipodystrophic insulin resistance.\nCell Metab. 2006;4:303 11.\n54. Agostini M, Gurnell M, Savage DB, Wood EM, Smith AG,\nRajanayagam O, et al. Tyrosine agonists reverse the molecular",
              "title": "2021 - PPAR\u03b3 and Diabetes Beyond the Genome and Towards Personalized Medicine.pdf",
              "version": "v0",
              "chunk_order": 136,
              "document_id": "4ea83190-476d-5090-a461-abde1adccbc5",
              "extraction_id": "02cdfa1b-cc8f-5141-bde0-1079d252c6e8",
              "associatedQuery": "What is the function of the PPAR-gamma gene in diabetes, and how do its variants impact the disease?"
            }
          },
          {
            "id": "9f62a8cf-a14f-5989-a899-cf1f525905bf",
            "score": 0.719792144310944,
            "metadata": {
              "text": "associated with a marked increase in T2D risk in the general\npopulation, schematized in Fig. 1. The latter systematically\ntested all the possible PPAR protein variants by using a\nlarge-scale pooled functional assay based on a human macro-\nphage cell line. Using these in vitro data to train a classifier by\nsupervised machine learning, they identified six pathogenic\nPPARG variants (R194Q, A417V, R212W, P387S, M203I,\nand T356R) in patients with partial lipodystrophy [ 109].",
              "title": "2021 - PPAR\u03b3 and Diabetes Beyond the Genome and Towards Personalized Medicine.pdf",
              "version": "v0",
              "chunk_order": 53,
              "document_id": "4ea83190-476d-5090-a461-abde1adccbc5",
              "extraction_id": "4bdd6cdb-1f2a-585f-b08e-392a54c6dad8",
              "associatedQuery": "What is the function of the PPAR-gamma gene in diabetes, and how do its variants impact the disease?"
            }
          },
          {
            "id": "818c1d6b-c1c1-570d-9e7a-87449fae279a",
            "score": 0.7187401056289724,
            "metadata": {
              "text": "lipid metabolism, as well as insulin sensitivity and inflammatory pathways. These pleiotropic functions confer great relevance to\nPPAR in physiological regulation of whole-body metabolism, as well as in the etiology of metabolic disorders. Accordingly,\nPPARG gene mutations, nucleotide variations, and post-translational modifications have been associated with adipose tissue\ndisorders and the related risk of insulin resistance and type 2 diabetes (T2D). Moreover, PPAR alternative splicing isoforms",
              "title": "2021 - PPAR\u03b3 and Diabetes Beyond the Genome and Towards Personalized Medicine.pdf",
              "version": "v0",
              "chunk_order": 2,
              "document_id": "4ea83190-476d-5090-a461-abde1adccbc5",
              "extraction_id": "2d610953-ea5c-5c01-ad19-60c607383da4",
              "associatedQuery": "What is the function of the PPAR-gamma gene in diabetes, and how do its variants impact the disease?"
            }
          },
          {
            "id": "793e2430-fa2b-513a-a4ab-0c85a167de3f",
            "score": 0.7186153102746433,
            "metadata": {
              "text": "the PPARgamma locus. Diabetes 2001;50:686 689\n12. Kahara T, Takamura T, Hayakawa T, et al.\nPPARgamma gene polymorphism is as-sociated with exercise-mediated changes\nof insulin resistance in healthy men. Me-\ntabolism 2003;52:209 212\n13. Franks PW, Luan J, Browne PO, et al.\nDoes peroxisome proliferator-activated\nreceptor gamma genotype (Pro12ala)\nmodify the association of physical activityand dietary fat with fasting insulin level?\nMetabolism 2004;53:11 16\n14. Memisoglu A, Hu FB, Hankinson SE, et al.",
              "title": "2013 - Gene-Environment and Gene-Treatment.pdf",
              "version": "v0",
              "chunk_order": 98,
              "document_id": "fe958fb1-5408-56ec-b102-ccf07b4bac2d",
              "extraction_id": "a6b92963-2cf0-51a4-8686-ce3a7515d443",
              "associatedQuery": "What is the function of the PPAR-gamma gene in diabetes, and how do its variants impact the disease?"
            }
          },
          {
            "id": "7c375d6d-672d-594c-a56e-7391ed3e9daa",
            "score": 0.7166055227337651,
            "metadata": {
              "text": "30. Majithia, A. R. et al. Rare variants in PPARG  with decreased activity in \nadipocyte differentiation are associated with increased risk of type 2 diabetes. \nProc Natl Acad Sci USA 111, 1312713132 (2014).\n 31. Majithia, A. R. et al. Prospective functional classification of all  \npossible missense variants in PPARG . Nat. Genet.  48, 15701575 (2016).\n 32. Claussnitzer, M. et al. Leveraging cross-species transcription factor binding",
              "title": "2018 - Refining the accuracy of validated target identification through coding variant.pdf",
              "version": "v0",
              "chunk_order": 117,
              "document_id": "3362e616-f824-55fa-9b4d-3ee8dcf52ac0",
              "extraction_id": "d96545e5-f3a0-5765-9b06-27a41219d3b9",
              "associatedQuery": "What is the function of the PPAR-gamma gene in diabetes, and how do its variants impact the disease?"
            }
          }
        ],
        "kg_search_results": null
      }
    }
  },
  "7": {
    "results": {
      "completion": {
        "id": "chatcmpl-AIFppDyOUKllFXSAk1UvPBBd5ythq",
        "choices": [
          {
            "finish_reason": "stop",
            "index": 0,
            "message": {
              "content": "CRISPR/Cas9 technology can be used to study or treat genetic forms of diabetes in several ways:\n\n1. **Genome and Epigenome Editing**: CRISPR/Cas9 has revolutionized the ability to modify genomes and epigenomes, allowing researchers to target specific genes associated with Type 2 Diabetes (T2D) and manipulate cis-regulatory elements related to beta cell function and glucose homeostasis [1].\n\n2. **Gene Knockout Studies**: CRISPR has been used to knock out genes by introducing frameshift mutations, which result in protein depletion. This approach has been adopted to study several genes in beta cell lines and human embryonic stem cell-derived beta cells, providing insights into the genetic basis of diabetes [4].\n\n3. **Correction of Point Mutations**: 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 for this is CRISPR/Cas9-based homology-directed repair (HDR), which involves generating a Cas9-mediated cut near the mutation site and using a homologous donor template to introduce the intended nucleotide change [5].\n\n4. **Disease Modeling**: CRISPR/Cas9 can be employed in human pluripotent stem cells (hPSCs) to model diabetes and metabolic syndromes in vitro. This allows researchers to study the disease mechanisms and test potential treatments in a controlled environment [3].\n\n5. **Animal Models**: CRISPR/Cas9 has been used to create knockout and knock-in mouse models, such as leptin and leptin receptor knockout mice, which serve as tools for diabetes and obesity research. These models help in understanding the genetic factors contributing to diabetes and testing therapeutic interventions [7].\n\nOverall, CRISPR/Cas9 technology provides a powerful platform for both studying the genetic underpinnings of diabetes and developing potential gene therapies to treat the disease.",
              "role": "assistant"
            }
          }
        ],
        "created": 1728914137,
        "model": "gpt-4o-2024-08-06",
        "object": "chat.completion",
        "system_fingerprint": "fp_6b68a8204b",
        "usage": {
          "completion_tokens": 396,
          "prompt_tokens": 1358,
          "total_tokens": 1754
        }
      },
      "search_results": {
        "vector_search_results": [
          {
            "id": "f42c0f84-d2a8-5bf9-89c2-3dd182bfb235",
            "score": 0.7536211609840393,
            "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": "How can CRISPR/Cas9 technology be used to study or treat genetic forms of diabetes?"
            }
          },
          {
            "id": "1859f32b-8f5c-5c3c-9f4d-54193d37645d",
            "score": 0.7519766836180293,
            "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": "How can CRISPR/Cas9 technology be used to study or treat genetic forms of diabetes?"
            }
          },
          {
            "id": "df30dab3-a490-5497-a079-2741f9039f87",
            "score": 0.7391794919967651,
            "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": "How can CRISPR/Cas9 technology be used to study or treat genetic forms of diabetes?"
            }
          },
          {
            "id": "eadf2320-de70-5499-ade0-7aa9930ac091",
            "score": 0.734539270401001,
            "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": "How can CRISPR/Cas9 technology be used to study or treat genetic forms of diabetes?"
            }
          },
          {
            "id": "99ccc9a2-865f-5d11-9b08-b26261d02fc9",
            "score": 0.7254999443590687,
            "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": "How can CRISPR/Cas9 technology be used to study or treat genetic forms of diabetes?"
            }
          },
          {
            "id": "1f114642-3f77-5346-89e8-394c433f66ff",
            "score": 0.722271203994751,
            "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": "How can CRISPR/Cas9 technology be used to study or treat genetic forms of diabetes?"
            }
          },
          {
            "id": "57b9550d-0258-5a87-be57-976f471e5763",
            "score": 0.7202445101883729,
            "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": "How can CRISPR/Cas9 technology be used to study or treat genetic forms of diabetes?"
            }
          },
          {
            "id": "4b170851-2dbd-5c06-9e3a-188d30a00170",
            "score": 0.7056117233269392,
            "metadata": {
              "text": "CRISPR-Cas9 epigenome editing enables high-throughput screening for functionalregulatory elements in the human genome. Nature Biotechnology 35(6):561 e568.\n[58] Hodson, D.J., Mitchell, R.K., Marselli, L., Pullen, T.J., Gimeno Brias, S.,\nSemplici, F., et al., 2014. ADCY5 couples glucose to insulin secretion in humanislets. Diabetes 63(9):3009 e3021 .\n[59] Zhou, Y., Park, S.-Y., Su, J., Bailey, K., Ottosson-Laakso, E., Shcherbina, L.,",
              "title": "2019 - (Epi)genomic heterogeneity of pancreatic islet function and failure in type 2 diabetes.pdf",
              "version": "v0",
              "chunk_order": 135,
              "document_id": "b9bc63a5-e366-5685-bd7a-4732a8eeffb7",
              "extraction_id": "4e73f54b-d265-594d-9fc1-9535a2d84672",
              "associatedQuery": "How can CRISPR/Cas9 technology be used to study or treat genetic forms of diabetes?"
            }
          },
          {
            "id": "83053df5-47ac-59da-9c30-69740a64372d",
            "score": 0.6952319145202637,
            "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": "How can CRISPR/Cas9 technology be used to study or treat genetic forms of diabetes?"
            }
          },
          {
            "id": "6f0adc7f-54ce-5a70-a2ea-153e074ccbdf",
            "score": 0.6797996163368225,
            "metadata": {
              "text": "DNA donors as templates, it is possible the nCas9-RT will beable to convert all variants at once. This new technique, however,is still in early development, and its editing ef ciency and side-\neffects remain to be seen.FUTURE PROSPECTIVES\nRecent technological developments around CRISPR-Cas9 and itsderivative technologies, combined with advances in humancellular models, should accelerate our understanding of theinterplay between diabetes risk-associated genetic variants and",
              "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": 144,
              "document_id": "51350055-d53c-5692-ab53-337b8a8bafd6",
              "extraction_id": "62d513ed-2dca-5f45-9da2-d847f92fc931",
              "associatedQuery": "How can CRISPR/Cas9 technology be used to study or treat genetic forms of diabetes?"
            }
          }
        ],
        "kg_search_results": null
      }
    }
  },
  "8": {
    "results": {
      "completion": {
        "id": "chatcmpl-AIFq1suHUy36hH4x2WAKqkv1fOq53",
        "choices": [
          {
            "finish_reason": "stop",
            "index": 0,
            "message": {
              "content": "Genetic polymorphisms in the GLUT4 gene have been studied to determine their significance in relation to Type 2 diabetes. However, according to the provided context, none of the identified polymorphisms in the GLUT4 gene have been linked to or found to be associated with Type 2 diabetes across various populations [1]. This suggests that, despite investigations, there is no established connection between GLUT4 gene polymorphisms and the susceptibility to Type 2 diabetes.",
              "role": "assistant"
            }
          }
        ],
        "created": 1728914149,
        "model": "gpt-4o-2024-08-06",
        "object": "chat.completion",
        "system_fingerprint": "fp_6b68a8204b",
        "usage": {
          "completion_tokens": 93,
          "prompt_tokens": 1423,
          "total_tokens": 1516
        }
      },
      "search_results": {
        "vector_search_results": [
          {
            "id": "bdc8f1de-8c90-5e28-8a21-a5bb0182cfe1",
            "score": 0.7754814296386922,
            "metadata": {
              "text": "Effectors \nGlucose transporters. A number of polymorphisms have been identified in \nthe GLUT4 gene. None of them have been linked to or found to be \nassociated with type 2 diabetes in a variety of populations. 5960 Interestingly, \nan association was found between a polymorphism in the human GLUT! \ngene and type 2 diabetes60 that was significant for obese women. Regulation \nof GLUT4 protein expression in diabetes occurs in a strongly tissue-specific",
              "title": "2004 - Diabetes Genes a.pdf",
              "version": "v0",
              "chunk_order": 434,
              "document_id": "805ad0d5-6a3d-5e5f-b1ba-121e10c6daaa",
              "extraction_id": "0734af87-4854-5a0f-b10c-2ea89376cb87",
              "associatedQuery": "What is the significance of genetic polymorphisms in the GLUT4 gene for Type 2 diabetes?"
            }
          },
          {
            "id": "bc93539a-df5f-5720-a4ce-0345fe4b66d4",
            "score": 0.7438287444318837,
            "metadata": {
              "text": "M,XiangKS,etal.1996.Geneticcontri-bution of polymorphism of the GLUT1and GLUT4 genes to the susceptibilityto type 2 (non-insulin-dependent) dia-betes mellitus in different populations.Acta Diabetologica 33:19397\n141. Poulsen P, Kyvik KO, Vaag A, Beck-\nNielsen H. 1999. Heritability of type II(non-insulin-dependent) diabetes melli-tus and abnormal glucose toleranceapopulation-basedtwinstudy. Diabetolo-\ngia42:13945\n142. Pugliese A, Zeller M, Fernandez AJ,",
              "title": "2003 - The Inherited Basis of Diabetes Mellitus.pdf",
              "version": "v0",
              "chunk_order": 239,
              "document_id": "7b85b290-d711-55d5-9b1e-b06e4d6f14a2",
              "extraction_id": "78e2a11a-4e89-5d14-b076-ef24c92b35b2",
              "associatedQuery": "What is the significance of genetic polymorphisms in the GLUT4 gene for Type 2 diabetes?"
            }
          },
          {
            "id": "0a7e6fb3-bf85-5440-adeb-c66fca9d170a",
            "score": 0.7292735737995176,
            "metadata": {
              "text": "A mutation in the Glut2 glucose transporter gene of a diabetic patientabolishes transport activity. J Biol Chem 269: 1776517767, 1994.\n36.Patel P, Bell GI, Cook JT, Turner RC, Wainscoat JS. Multiple\nrestriction fragment length polymorphisms at the GLUT2 locus: GLUT2haplotypes for genetic analysis of type 2 (non-insulin-dependent) diabetesmellitus. Diabetologia 34: 817821, 1991.\n37.Pereira MA, FitzerGerald SJ, Gregg EW, Joswiak ML, Ryan WJ,\nSuminski RR, Utter AC, Zmuda JM. A collection of Physical Activity",
              "title": "2007 - Physical activity modifies the effect of SNPs in the SLC2A2 (GLUT2).pdf",
              "version": "v0",
              "chunk_order": 133,
              "document_id": "6f5ced46-b777-563a-b644-432f4e7e2644",
              "extraction_id": "276a7b90-6325-59c8-b8b2-77f855aa2553",
              "associatedQuery": "What is the significance of genetic polymorphisms in the GLUT4 gene for Type 2 diabetes?"
            }
          },
          {
            "id": "a4973968-2510-5f08-8252-f2be85be3c42",
            "score": 0.7235005992351904,
            "metadata": {
              "text": "NootherrecentassociationsofpolymorphismswithT2Dhavebeenreplicated\nto date (Table 5). However, a recent meta-analysis (106) identied some earlyreproducibilityofanassociationbetweenvariationin GLUT1andT2D,originally\nreportedin1988(104).Itislikelythatthisassociationhasnotbeenpursuedfurtherfor several reasons, but one possibility is a study that reported the rejection oflinkageto GLUT1athighlevelsofsignicance(46).However,linkagehaslimited",
              "title": "2003 - The Inherited Basis of Diabetes Mellitus.pdf",
              "version": "v0",
              "chunk_order": 155,
              "document_id": "7b85b290-d711-55d5-9b1e-b06e4d6f14a2",
              "extraction_id": "51702d4a-735b-5bc4-98a4-d26bf1e58b40",
              "associatedQuery": "What is the significance of genetic polymorphisms in the GLUT4 gene for Type 2 diabetes?"
            }
          },
          {
            "id": "4bab532c-8b73-54b8-905a-d7b070af1da8",
            "score": 0.7182047274938725,
            "metadata": {
              "text": "mechanism by which type 2 diabetes is influenced remains to\nbe identified.\nThere have been several attempts to clarify the role of the\npolymorphism in SLC30A8 in the development of type 2\ndiabetes and the focus has been set on insulin secretion dueto the importance of ZnT-8 for insulin storage in the granulaof pancreatic cells.\nThe results are controversial, but there appears to be an\nassociation between the risk variant of rs13266634 and\nreduced insulin secretion. Interestingly, decreased insulin",
              "title": "2009 - Zinc and Diabetes - clinical links and molecular mechanisms.pdf",
              "version": "v0",
              "chunk_order": 132,
              "document_id": "72ab8458-928b-56b9-9547-1ba4b59dfab9",
              "extraction_id": "a482defd-8d6a-5966-8ec1-5aa7e49c14f1",
              "associatedQuery": "What is the significance of genetic polymorphisms in the GLUT4 gene for Type 2 diabetes?"
            }
          },
          {
            "id": "42eee55f-adfa-5a04-b3f0-a592b3b08a2c",
            "score": 0.7140612772666659,
            "metadata": {
              "text": "glucose tolerance, suggesting a r ole for this polymorphism in the\nonset of GDM as well as type 2 diabetes mellitus ( 17). The switch on\nIRS-1 of the amino acid GLY972 Arg (rs1801278) impairs insulinsecretion, and a study on 1306 GDM patients and 1973 pregnantwomen without GDM found a signi cant association between the\npresence of this polymorphism and the risk of GDM ( 18).\nIntriguing results were generated by a study on the genetic",
              "title": "2020 - Genetics and Epigenetics New Insight on Gestational Diabetes Mellitus.pdf",
              "version": "v0",
              "chunk_order": 25,
              "document_id": "301a7093-a33a-57c9-8979-58146c57ced1",
              "extraction_id": "7d315f2c-43f0-587a-9370-e0f205d6c611",
              "associatedQuery": "What is the significance of genetic polymorphisms in the GLUT4 gene for Type 2 diabetes?"
            }
          },
          {
            "id": "65c8f702-eee5-550e-bd63-78892b158c93",
            "score": 0.710313873868355,
            "metadata": {
              "text": "tients the EUGENE2 study. Diabetologia 2008;51:816 820\n32. Kirchhoff K, Machicao F, Haupt A, et al. Polymorphisms in the TCF7L2,\nCDKAL1 and SLC30A8 genes are associated with impaired proinsulinconversion. Diabetologia 2008;51:597 601\n33. Nicolson TJ, Bellomo EA, Wijesekara N, et al. Insulin storage and glucose\nhomeostasis in mice null for the granule zinc transporter ZnT8 and studies\nof the type 2 diabetes-associated variants. Diabetes 2009;58:2070 2083",
              "title": "2012 - Reduced Insulin Exocytosis in Human Pancreatic b-Cells.pdf",
              "version": "v0",
              "chunk_order": 98,
              "document_id": "689e4fcc-99db-5798-8f1d-68c41d4638db",
              "extraction_id": "e6e7fc9f-e4a4-5d51-9070-01ce34cffcd3",
              "associatedQuery": "What is the significance of genetic polymorphisms in the GLUT4 gene for Type 2 diabetes?"
            }
          },
          {
            "id": "a6ef60db-d564-5f55-a31a-db893879ab14",
            "score": 0.7092558492117108,
            "metadata": {
              "text": "is markedly reduced in glucose-unresponsive islets from ani-mal models of type 2 diabetes (51). In a previous study in PimaIndians, we found that ~5% of this population carries a mis-sense polymorphism in exon 3 of the GLUT2 gene (52), but\nthis polymorphism was not associated with the residual fast-ing plasma insulin concentration in the present study.Despite the fact that GLUT2 is an attractive candidate, it",
              "title": "2000 - A High Fasting Plasma Insulin Concentration.pdf",
              "version": "v0",
              "chunk_order": 96,
              "document_id": "77375e8f-ca8e-5bbb-b310-910dd82ade9c",
              "extraction_id": "6aefb64e-b732-5742-90a4-f2aa43c8b866",
              "associatedQuery": "What is the significance of genetic polymorphisms in the GLUT4 gene for Type 2 diabetes?"
            }
          },
          {
            "id": "b91a2e1b-eb4d-5e1b-a85c-46a8f394603c",
            "score": 0.7054600295150275,
            "metadata": {
              "text": "polymorphisms in 24 DNA samples. Common variants were thengenotyped in 760 type 2 diabetic patients and 641 nondiabetic sub-jects. Genetic associations with diabetes-related phenotypes werealso analyzed.\nResults: Nine polymorphisms were identified, and four common poly-\nmorphisms [g. /H110021500C /H11022G, g./H110021062G /H11022C, g./H11002994C/H11022T, g./H11001408C/H11022A\n(Leu72Met)] were genotyped in a larger study. The genotype distri-butions of these four common polymorphisms in type 2 diabetes pa-",
              "title": "2006 - Polymorphisms in the Ghrelin Gene Are Associated with Serum High-Density Lipoprotein.pdf",
              "version": "v0",
              "chunk_order": 3,
              "document_id": "8ad4e1b5-9c29-5b20-bed8-afbf4a14862a",
              "extraction_id": "00f6985d-f69b-50e3-b673-0ec508e6c025",
              "associatedQuery": "What is the significance of genetic polymorphisms in the GLUT4 gene for Type 2 diabetes?"
            }
          },
          {
            "id": "277f37e3-ee45-5619-b051-33d5ba95bd07",
            "score": 0.7024430809949005,
            "metadata": {
              "text": "in turn, result in a defective or poorly expressed glucagonprotein and lead to decreased insulin secretion and conse-\nquently hyperglycaemia [ 48].\nThe current study identified, for the first time, several type\n2 diabetes-associated risk alleles associated with a higher riskof GDM, namely rs7957197 ( HNF1A ), rs10814916 ( GLIS3 ),\nrs3802177 ( SLC30A8 ) and rs7041847 ( GLIS3 ). These SNPs",
              "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": 69,
              "document_id": "3b301dd1-17bd-5632-9a96-d6294c6d7650",
              "extraction_id": "a228ec1a-de5b-5e0c-b24f-db8249be4053",
              "associatedQuery": "What is the significance of genetic polymorphisms in the GLUT4 gene for Type 2 diabetes?"
            }
          }
        ],
        "kg_search_results": null
      }
    }
  },
  "9": {
    "results": {
      "completion": {
        "id": "chatcmpl-AIFq5UdPTHMiHxXb8m3RyqvBs55fY",
        "choices": [
          {
            "finish_reason": "stop",
            "index": 0,
            "message": {
              "content": "MicroRNAs (miRNAs) regulate gene expression related to diabetes by acting at the post-transcriptional level to control their target genes. They are involved in several crucial pathways associated with diabetes, including insulin secretion, cholesterol biosynthesis, fat metabolism, and adipogenesis [2]. miRNAs also play significant roles in pancreatic islet development, beta-cell dysfunction, insulin synthesis and secretion, and insulin resistance, which are key factors in the pathology of both Type 1 and Type 2 Diabetes Mellitus (T1DM and T2DM) [6]. Additionally, specific miRNAs have been implicated in the pathogenesis of diabetic complications, such as diabetic nephropathy, where miRNAs like miR-192, miR-216a, miR-217, and miR-377 are up-regulated [2]. These miRNAs can modulate the actions of growth factors and inflammatory factors, further influencing diabetic complications [5].",
              "role": "assistant"
            }
          }
        ],
        "created": 1728914153,
        "model": "gpt-4o-2024-08-06",
        "object": "chat.completion",
        "system_fingerprint": "fp_6b68a8204b",
        "usage": {
          "completion_tokens": 188,
          "prompt_tokens": 1439,
          "total_tokens": 1627
        }
      },
      "search_results": {
        "vector_search_results": [
          {
            "id": "c2fa8cbd-5f7f-5086-90ec-d1e5e6df0ee9",
            "score": 0.736937733061589,
            "metadata": {
              "text": "MicroRNAs (miRNA) ar e single -stranded, small RNA molecules that act at the post -\ntranscriptional standard to regulate their target or source genes. Many biological processes \nare regulated by this Micro RNA. Since its discovery about two decades ago. It is correlated \nwith a com prehensive set of diseases and described by numerous miRNAs, including T2DM \nand cardiovascular diseases. Specifically, with respect to T2DM, micro RNA plays a",
              "title": "2023 - Genetic Link Between Type 2 Diabetes Mellitus and Cardiovascular Disease.pdf",
              "version": "v0",
              "chunk_order": 45,
              "document_id": "c54f9f64-7e6d-5186-a1de-d487ba9d19b8",
              "extraction_id": "2211fc04-119d-534b-8de8-dfa4d1bfbf09",
              "associatedQuery": "How do microRNAs regulate gene expression related to diabetes?"
            }
          },
          {
            "id": "3cd8facc-0c2c-5a48-9f7c-cbd5685d914a",
            "score": 0.7289735997946791,
            "metadata": {
              "text": "they can act as oncogenes or tumor suppressors  (8, 29, 72). miRs are associated with the 341 \nregulation of genes relevant to insulin secre tion, cholesterol biosynthesis, fat metabolism and 342 \nadipogenesis, crucial pathways in the pathogene sis of diabetes (53, 114, 115). miRs have also 343 \nbeen implicated in TGF-  signaling related to th e pathogenesis of diabetic nephropathy with key 344 \nmiRs such as miR-192, miR-216a, miR-217 and miR-377 being up-regula ted in glomerular 345",
              "title": "2010 - The Role of Epigenetics in the Pathology of Diabetic Complications.pdf",
              "version": "v0",
              "chunk_order": 72,
              "document_id": "766edfd5-4756-51bf-b636-c94b041d030c",
              "extraction_id": "b1d2c95c-d639-5c75-8c52-278f1e187675",
              "associatedQuery": "How do microRNAs regulate gene expression related to diabetes?"
            }
          },
          {
            "id": "f35c5082-c877-5cdf-9ba8-a91dd72da2e8",
            "score": 0.7277096238482499,
            "metadata": {
              "text": "Lim LP, Lau NC, Garrett-Engele P, Grimson A, Schelter JM et al\n(2005) Microarray analysis shows that some microRNAs down-regulate large numbers of target mRNAs. Nature 433:769773\nLovis P, Roggli E, Laybutt DR, Gattesco S, Yang JY et al (2008)\nAlterations in microRNA expression contribute to fatty acid-induced pancreatic beta-cell dysfunction. Diabetes 57:27282736\nNadler ST, Stoehr JP, Schueler KL, Tanimoto G, Yandell BS et al",
              "title": "2009 - Obesity and genetics regulate microRNAs in islets, liver, and adipose of diabetic mice.pdf",
              "version": "v0",
              "chunk_order": 100,
              "document_id": "c3d2aced-4550-553f-abed-0d3a7ac1414f",
              "extraction_id": "7d22ecdf-dd9f-53e9-aa2b-df81bd03c3bc",
              "associatedQuery": "How do microRNAs regulate gene expression related to diabetes?"
            }
          },
          {
            "id": "abbcafb6-f502-5648-a9a4-196466452564",
            "score": 0.7271047126463582,
            "metadata": {
              "text": "Abstract\nRecent advances in the understanding of the genetics of type 2 diabetes (T2D) susceptibility have focused attention on the\nregulation of transcriptional activity within the pancreatic beta-cell. MicroRNAs (miRNAs) represent an important\ncomponent of regulatory control, and have proven roles in the development of human disease and control of glucose",
              "title": "2013 - The miRNA Profile of Human Pancreatic Islets and BetaCells and Relationship to Type 2 Diabetes Pathogenesis.pdf",
              "version": "v0",
              "chunk_order": 2,
              "document_id": "05e7f076-6b4a-5ab0-b4d0-28e4b6eeef8f",
              "extraction_id": "65ad21df-f728-54b6-b329-9ed8793c33ce",
              "associatedQuery": "How do microRNAs regulate gene expression related to diabetes?"
            }
          },
          {
            "id": "8347a530-d264-5d7a-81f6-704f8ed7bf57",
            "score": 0.7260474293175475,
            "metadata": {
              "text": "evidence demonstrates that miRNAs and lncRNAs can alsoregulate the expression of genes and modulate the actions of\ngrowth factors and inflammatory factors related to diabetic\ncomplications [ 8]. These reports have been described in sev-\neral reviews [ 8,8791] and are only briefly discussed here.\nNumerous recent reports have demonstrated abnormal ex-\npression of various miRNAs in renal, vascular and retinal cellsunder diabetic conditions, and in vivo models of related",
              "title": "2015 - Epigenetic mechanisms in diabetic complications and metabolic memory.pdf",
              "version": "v0",
              "chunk_order": 88,
              "document_id": "470f1f94-792d-5273-a88f-7e06084951c5",
              "extraction_id": "593dfb70-8b55-5a74-abd5-446394a0bd23",
              "associatedQuery": "How do microRNAs regulate gene expression related to diabetes?"
            }
          },
          {
            "id": "f0bb404a-2062-584e-850d-cf49a1e0b4a7",
            "score": 0.7203913282998289,
            "metadata": {
              "text": "In addition, miRNAs have been shown to be involved in T2DM. For example, miRNAs play major roles \nin pancreatic islet development,  cell dysfunction, insulin synthesis and secretion and insulin  \nresistance [148] . Studies based on miRNA microarray analysis have identified many different miRNAs \ninvolved in the pathology of both T1DM and T2DM; these miRNAs include mi R-375, miR -29, miR -9, \nmiR-124a, miR -195, miR -222, miR -126, miR -133a, miR -296, miR -96, miR -34a, miR -146b, miR -657,",
              "title": "2014 - Nutrigenetics and Nutrigenomics Insights into Diabetes Etiopathogenesis.pdf",
              "version": "v0",
              "chunk_order": 147,
              "document_id": "ce4f171c-494c-53f2-a770-c3edd3561c40",
              "extraction_id": "0cb154ce-660d-54fa-a31f-0391434a5470",
              "associatedQuery": "How do microRNAs regulate gene expression related to diabetes?"
            }
          },
          {
            "id": "a9695ed0-6f3d-5e79-ab99-514119637e0b",
            "score": 0.7115685435370103,
            "metadata": {
              "text": "26. He Y , Ding Y , Liang B, Lin J, Kim TK, Yu H, Hang H, \nWang K. A Systematic Study of Dysregulated MicroRNA in \nType 2 Diabetes Mellitus. Int J Mol Sci. 2017:18.\n27. Dias S, Hemmings S, Muller C, Louw J, Pheiffer C. MicroRNA Expression Varies according to Glucose \nTolerance, Measurement Platform, and Biological Source. \nBiomed Res Int. 2017;2017:1080157.\n28. El Ouaamari A, Baroukh N, Martens GA, Lebrun P, Pipeleers D, van Obberghen E. miR-375 targets \n3'-phosphoinositide-dependent protein kinase-1 and",
              "title": "2018 - MicroRNA profiling and their pathways in South African.pdf",
              "version": "v0",
              "chunk_order": 96,
              "document_id": "b6bb090d-7176-59db-af04-582aa1d5cf10",
              "extraction_id": "14577d73-d320-54dd-93f2-c55f986bc8bc",
              "associatedQuery": "How do microRNAs regulate gene expression related to diabetes?"
            }
          },
          {
            "id": "1d9d150b-27f9-55f7-8111-1f6de79a78bc",
            "score": 0.7088165630320882,
            "metadata": {
              "text": "nucleotide RNA molecules that potentially regulate the\nexpression of thousands of genes. To understand therelationship between miRNA regulation and obesity-\ninduced diabetes, we quantitatively proled approximately220 miRNAs in pancreatic islets, adipose tissue, and liver\nfrom diabetes-resistant (B6) and diabetes-susceptible\n(BTBR) mice. More than half of the miRNAs proled wereexpressed in all three tissues, with many miRNAs in each\ntissue showing signicant changes in response to genetic",
              "title": "2009 - Obesity and genetics regulate microRNAs in islets, liver, and adipose of diabetic mice.pdf",
              "version": "v0",
              "chunk_order": 2,
              "document_id": "c3d2aced-4550-553f-abed-0d3a7ac1414f",
              "extraction_id": "42c407dd-9f88-57b3-b47b-e467c486e3a4",
              "associatedQuery": "How do microRNAs regulate gene expression related to diabetes?"
            }
          },
          {
            "id": "5bf6de7b-8b41-5a32-a513-843f0f71c640",
            "score": 0.70811406277251,
            "metadata": {
              "text": "11. Bartel DP. MicroRNAs: genomics, biogenesis, mechanism, and\nfunction. Cell. 2004;116(2):281 97.\n12. Pirola L, Balcerczyk A, Tothill RW, et al. Genome-wide analysis\ndistinguishes hyperglycemia regulated epigenetic signatures of pri-\nmary vascular cells. Genome Res. 2011;21(10):1601 15.\n13.Cooper ME, El-Osta A. Epigenetics: mechanisms and implications\nfor diabetic complications. Circ Res. 2010;107(12):1403 13.Thispaper also provides a review of evidence pertaining to the role",
              "title": "2016  - Epigenetic Mechanisms in Diabetic Kidney Disease.pdf",
              "version": "v0",
              "chunk_order": 69,
              "document_id": "6f773bda-0b8f-5da2-a9b5-e6c013d75050",
              "extraction_id": "767d65c7-b99d-5427-8f5a-4afa10669e11",
              "associatedQuery": "How do microRNAs regulate gene expression related to diabetes?"
            }
          },
          {
            "id": "01d78f49-9996-58ea-b076-e352ff22461c",
            "score": 0.7069891023059317,
            "metadata": {
              "text": "128. Diao X, Shen E, Wang X, Hu B. Differentially expressed microRNAs and their target genes in the hearts of streptozotocin-induced diabetic mice. Mol Med Rep (2011) 4:63340. doi:10.3892/mmr.2011.489 \n129. La Sala L, Cattaneo M, De Nigris V , Pujadas G, Testa R, Bonfigli AR,  \net al. Oscillating glucose induces microRNA-185 and impairs an efficient antioxidant response in human endothelial cells. Cardiovasc Diabetol  (2016) \n15:71. doi:10.1186/s12933-016-0390-9",
              "title": "2018 - Type 2 Diabetes Mellitus and Cardiovascular Disease Genetic and Epigenetic Links.pdf",
              "version": "v0",
              "chunk_order": 159,
              "document_id": "3e82a2e5-4b2c-59c0-99cd-f3b06d8dabf2",
              "extraction_id": "9e010393-b98f-5f6c-a62d-fc0646ba8667",
              "associatedQuery": "How do microRNAs regulate gene expression related to diabetes?"
            }
          }
        ],
        "kg_search_results": null
      }
    }
  },
  "10": {
    "results": {
      "completion": {
        "id": "chatcmpl-AIFqHVwVjpSqrvqjfDgFColFbW31Q",
        "choices": [
          {
            "finish_reason": "stop",
            "index": 0,
            "message": {
              "content": "Studying the genetic basis of MODY (Maturity Onset Diabetes of the Young) has provided several important insights:\n\n1. **Genetic Heterogeneity**: MODY is caused by mutations in multiple genes, with at least 13 known genes implicated. The most prevalent mutations occur in the genes HNF1A, GCK, and HNF4A [3]. This genetic diversity leads to different subtypes of MODY, each with distinct clinical characteristics such as age of onset, pattern of hyperglycemia, response to treatment, and associated extrapancreatic manifestations [3].\n\n2. **Inheritance Pattern**: MODY is inherited in an autosomal dominant manner, which means that it can be passed down through families. This inheritance pattern allows for the collection of multigenerational pedigrees, making MODY an attractive model for genetic studies [2].\n\n3. **Clinical Presentation**: MODY typically presents in young adults, often before the age of 25, and is characterized by primary insulin secretion defects. It is not related to obesity or autoimmune processes, distinguishing it from other forms of diabetes like type 1 and type 2 diabetes [5].\n\n4. **Pathogenic Mechanisms**: Despite advances in understanding the molecular pathogenesis of MODY, there remain unknown genetic determinants in many patients with a MODY-like phenotype, suggesting additional locus heterogeneity and new pathogenic mechanisms yet to be discovered [4].\n\n5. **Impact on Treatment and Diagnosis**: Genetic testing for MODY can lead to more accurate diagnoses and tailored treatment plans. Many patients with MODY are currently undiagnosed or misdiagnosed with type 1 or type 2 diabetes, highlighting the importance of genetic testing in identifying this condition [7].\n\nThese insights underscore the complexity and variability of MODY, as well as the importance of genetic research in improving diagnosis and treatment strategies for this form of diabetes.",
              "role": "assistant"
            }
          }
        ],
        "created": 1728914165,
        "model": "gpt-4o-2024-08-06",
        "object": "chat.completion",
        "system_fingerprint": "fp_a20a4ee344",
        "usage": {
          "completion_tokens": 383,
          "prompt_tokens": 1343,
          "total_tokens": 1726
        }
      },
      "search_results": {
        "vector_search_results": [
          {
            "id": "7d0ed0e8-6967-5dbe-b57e-f1e3a5821fd0",
            "score": 0.8096703418794584,
            "metadata": {
              "text": "studying the highly familial MODY form of young - onset diabetes or other rare forms of monogenic diabetes.  \n  Table 12.2    The different subtypes of maturity - onset diabetes of the young ( MODY ). \n   MODY \ntype     Gene \nlocus     Gene name     Year of \ndiscovery     Distribution     Onset of \ndiabetes     Primary \ndefect     Severity of \ndiabetes     Complications     OMIM  \n  MODY1    20q     HNF4A   ( TCF14  )    1996    Rare (2  3%)    Adolescence/",
              "title": "2010 - The Genetics of Type 2 Diabetes.pdf",
              "version": "v0",
              "chunk_order": 38,
              "document_id": "a1d211d4-279e-51d7-b2b2-33bc2763d089",
              "extraction_id": "6c0f6484-fb94-5583-8c4a-f707983ff29f",
              "associatedQuery": "What insights have been gained from studying the genetic basis of MODY (Maturity Onset Diabetes of the Young)?"
            }
          },
          {
            "id": "e3ec46a7-b0c9-567d-ba4a-e1c4f1341364",
            "score": 0.8007671118239841,
            "metadata": {
              "text": "penetrance and early - onset diabetes, allows the collection of multigenerational pedigrees, making MODY an attractive model for genetic studies. MODY usually develops in thin young adults (usually before 25 years of age; in childhood, adolescence or young adulthood), and is associated with primary insulin - secretion defects  [4,5] . The prevalence of MODY is estimated to be less than 1  2% of patients with T2DM, although it could represent as many as 5% of European cases of diabetes  [4,25] . MODY is not",
              "title": "2010 - The Genetics of Type 2 Diabetes.pdf",
              "version": "v0",
              "chunk_order": 31,
              "document_id": "a1d211d4-279e-51d7-b2b2-33bc2763d089",
              "extraction_id": "6c0f6484-fb94-5583-8c4a-f707983ff29f",
              "associatedQuery": "What insights have been gained from studying the genetic basis of MODY (Maturity Onset Diabetes of the Young)?"
            }
          },
          {
            "id": "72000563-bea1-562e-b5d6-ea2c82f74d53",
            "score": 0.7927499289959641,
            "metadata": {
              "text": "[2] . Mutations in 13 genes are known \nto cause MODY; the most prevalent are  HNF1A  ,  GCK   \nand  HNF4A   [3, 4]  . The MODY subtypes differ in age of \nonset of diabetes, the pattern of hyperglycemia, response to treatment, and associated extrapancreatic manifesta-tions  \n[5] . As compared to type 2 diabetes, the clinical  Key Words \n Best practice  Genetic testing  Healthcare providers  Interview study  Maturity onset diabetes of the young \n Abstract",
              "title": "2015 - Current and Best Practices of Genetic Testing for Maturity.pdf",
              "version": "v0",
              "chunk_order": 3,
              "document_id": "076f1140-8992-536f-832b-65ebdb9232a2",
              "extraction_id": "c5c209f5-e4dc-5eb2-a9f0-536a686efa96",
              "associatedQuery": "What insights have been gained from studying the genetic basis of MODY (Maturity Onset Diabetes of the Young)?"
            }
          },
          {
            "id": "b1c43f5d-53c4-58e5-ac10-a90ecdd9d576",
            "score": 0.7827710379821695,
            "metadata": {
              "text": "causal for MODY , although genetic or functional evidence of\nobvious pathogenicity is not fully compelling (Table 1).\nDespite these important advances in understanding the mo-\nlecular pathogenesis of MODY , the genetic determinants in\nmany patients with young-onset diabetes resembling a\nMODY-like phenotype remain unknown, suggesting addi-\ntional locus heterogeneity and new pathogenic mechanismsto be yet discovered. This has particularly been observed in",
              "title": "2019 - How Recent Advances in Genomics Improve Precision Diagnosis.pdf",
              "version": "v0",
              "chunk_order": 42,
              "document_id": "42b64375-06af-5e09-9ae6-6bd0ecb782c7",
              "extraction_id": "7117f141-8841-5c11-ba1a-85039181b393",
              "associatedQuery": "What insights have been gained from studying the genetic basis of MODY (Maturity Onset Diabetes of the Young)?"
            }
          },
          {
            "id": "1555d1c2-53e4-5f7f-8411-7bb11d990eed",
            "score": 0.7714792583715256,
            "metadata": {
              "text": "MODY Maturity Onset Diabetes of the Young. This is an uncommon form of diabetes, inherited as an autosomal dominant condition, and displaysa slow onset of symptoms. It generally presents before 25 years of age, is not related to obesity, and appears to have no autoi mmune\nbasis. Multiple forms of MODY have been characterised based on mutations affecting different genes involved in the control of -cellfunction, and display different degrees of disease severity\nContinued over page",
              "title": "2008 - Glossary of Genetics Genomics Terms.pdf",
              "version": "v0",
              "chunk_order": 24,
              "document_id": "c66d2572-071d-5aaf-829c-b3ca6cf6d697",
              "extraction_id": "4668a316-6c87-5039-b55b-47fe0b8fbc71",
              "associatedQuery": "What insights have been gained from studying the genetic basis of MODY (Maturity Onset Diabetes of the Young)?"
            }
          },
          {
            "id": "25e3417d-4e7e-595c-bec6-6f6e3d697ab4",
            "score": 0.7695330381393487,
            "metadata": {
              "text": "Genetic Testing for MODY  Public Health Genomics 2015;18:5259 \nDOI: 10.1159/00036796359  1 Singh R, Pearson ER: The importance of mak-\ning a genetic diagnosis of diabetes. Can J Dia-betes 2006; \n  30:   183190. \n  2 Ledermann HM: Is maturity onset diabetes at \nyoung age (MODY) more common in Europe than previously assumed? Lancet 1995; \n  345:  \n 648.",
              "title": "2015 - Current and Best Practices of Genetic Testing for Maturity.pdf",
              "version": "v0",
              "chunk_order": 106,
              "document_id": "076f1140-8992-536f-832b-65ebdb9232a2",
              "extraction_id": "3c09962c-226d-5271-a5c8-14f6327a079b",
              "associatedQuery": "What insights have been gained from studying the genetic basis of MODY (Maturity Onset Diabetes of the Young)?"
            }
          },
          {
            "id": "e479acca-9418-552b-98ae-edb6eb74ee6f",
            "score": 0.767810651762246,
            "metadata": {
              "text": "Genetic Testing for MODY  Public Health Genomics 2015;18:5259 \nDOI: 10.1159/00036796353symptoms present often at a relatively young age in pa-\ntients without overweight, who have a positive family his-tory. As compared to type 1 diabetes, progression may be less severe, and the required dosage of insulin low.\n  Many patients with MODY are currently undiagnosed \nor misdiagnosed with type 1 or 2 diabetes mellitus  [4] . In",
              "title": "2015 - Current and Best Practices of Genetic Testing for Maturity.pdf",
              "version": "v0",
              "chunk_order": 10,
              "document_id": "076f1140-8992-536f-832b-65ebdb9232a2",
              "extraction_id": "039d8c39-8fe3-5183-b952-3e8a25510b86",
              "associatedQuery": "What insights have been gained from studying the genetic basis of MODY (Maturity Onset Diabetes of the Young)?"
            }
          },
          {
            "id": "b964fb31-cf7f-5d5d-9d73-d737daa96b8d",
            "score": 0.7649612426757867,
            "metadata": {
              "text": "in 1992, through familial linkage analysis of French pedigreeswith early-onset, non-auto-immune, non-obese diabetes thatwas also called maturity-onset diabetes of the young (MODY)\n(Froguel et al., 1992 ). Mutations in GCK (encoding glucokinase)\nwere shown to cause a relatively benign form of MODY. Inciden-tally, it was the rst time that the direct causative effect of rela-\ntive insulin deciency was demonstrated in T2D, when insulin",
              "title": "2015 - Rare and Common Genetic Events in Type 2 Diabetes.pdf",
              "version": "v0",
              "chunk_order": 6,
              "document_id": "641771c3-048c-5afb-a41c-05a883e99408",
              "extraction_id": "587c2476-a12a-5e32-b0cf-ada54ee4a1db",
              "associatedQuery": "What insights have been gained from studying the genetic basis of MODY (Maturity Onset Diabetes of the Young)?"
            }
          },
          {
            "id": "847efd79-3919-5ec0-b5b3-9934cdb29c39",
            "score": 0.7617053247514575,
            "metadata": {
              "text": "gene studies were under powered. However, studies of \nmonogenic forms of diabetes, specifically maturity onset \ndiabetes of the young 2 (MODY2), provided some of the \nfirst insights into the contribution of genetic variation to \nhyperglycemia observed during pregnancy and fetal \noutcomes. MODY2 is an autosomal dominant form of \nMODY due to mutations in glucokinase ( GCK ) [2527]. Table 1. Characteristics and treatment modalities of different forms of diabetes mellitus\nCharacteristics Treatment modalities",
              "title": "2011 - Inherited destiny Genetics and gestational diabetes mellitus.pdf",
              "version": "v0",
              "chunk_order": 19,
              "document_id": "6d341cd2-ae56-5807-9aff-39298efc4d06",
              "extraction_id": "318e6c28-16d2-5d59-b047-cfdc6b2e7301",
              "associatedQuery": "What insights have been gained from studying the genetic basis of MODY (Maturity Onset Diabetes of the Young)?"
            }
          },
          {
            "id": "77d42dce-1bb6-577f-95f4-f8c7ece85c19",
            "score": 0.7578971385955865,
            "metadata": {
              "text": "is variable, underlining that this disorder is genetically heterogeneous. \nTable 1. Definition of MODY \n Impaired glucose tolerance \n Age of onset <25 years \n Autosomal-dominant inheritance \nUsing genetic linkage and candidate gene approaches, mutations in \ngenes on chromosomes 2, 7, 12, 13, 19, and 20 have been linked to MODY \nand collectively may represent up to 3% of all patients with type 2 diabetes \n(Table 2). The gene on chromosome 7 (MODY2) encodes the glycolytic",
              "title": "2004 - Diabetes Genes a.pdf",
              "version": "v0",
              "chunk_order": 1054,
              "document_id": "805ad0d5-6a3d-5e5f-b1ba-121e10c6daaa",
              "extraction_id": "d4fcf425-4ac9-5918-973a-fe6c422b19bc",
              "associatedQuery": "What insights have been gained from studying the genetic basis of MODY (Maturity Onset Diabetes of the Young)?"
            }
          }
        ],
        "kg_search_results": null
      }
    }
  },
  "11": {
    "results": {
      "completion": {
        "id": "chatcmpl-AIFqUmWTKdcimZ6Y2TYtW6SieUkPG",
        "choices": [
          {
            "finish_reason": "stop",
            "index": 0,
            "message": {
              "content": "The genes most commonly associated with an increased risk of developing diabetes, particularly type 2 diabetes, include:\n\n1. **CDKAL1, CDKN2A, CDKN2B** - These genes are linked to reduced insulin secretion via reduced beta-cell mass [1].\n2. **MTNR1B, TCF7L2, KCNJ11** - These genes are associated with beta-cell dysfunction [1].\n3. **FTO** - This gene is related to increased insulin resistance associated with obesity [1].\n4. **IRS1, PPARG** - These genes are related to increased insulin resistance unrelated to obesity [1].\n5. **IGF2BP2, HHEX, SLC30A8, WFS1** - These genes have been shown to increase susceptibility to type 2 diabetes in reproducible studies [3].\n6. **JAZF1, CDC123/CAMK1D, TSPAN8/LGR5, THADA, ADAMTS9, NOTCH2** - These are additional variants identified in a recent meta-analysis as being associated with type 2 diabetes [3].\n7. **KCNQ1** - This gene is associated with susceptibility to type 2 diabetes in East Asian and European populations [6].\n\nThese genes have been identified through various genome-wide association studies (GWAS) and other genetic research efforts.",
              "role": "assistant"
            }
          }
        ],
        "created": 1728914178,
        "model": "gpt-4o-2024-08-06",
        "object": "chat.completion",
        "system_fingerprint": "fp_a20a4ee344",
        "usage": {
          "completion_tokens": 282,
          "prompt_tokens": 1557,
          "total_tokens": 1839
        }
      },
      "search_results": {
        "vector_search_results": [
          {
            "id": "47e8bd94-fd61-57f2-b1d0-cc139d71936a",
            "score": 0.767777144908905,
            "metadata": {
              "text": "of Diabetes \n Results of several genome-wide association stud-\nies (GWAS) have linked the following common gene variants with a 1520% increased risk of diabetes: reduced insulin secretion via reduce beta-cell mass (CDKAL1, CDKN2A, CDKN2B) and beta-cell dysfunction (MTNR1B, TCF7L2, KCNJ11) and increased insulin resistance related to obesity (FTO) and unrelated to obesity (IRS1, PPARG) [ \n11 ]. While most of the early studies",
              "title": "2017 - diabetes-mellitus-in-developing-countries-and-underserved-commun-2017.pdf",
              "version": "v0",
              "chunk_order": 2429,
              "document_id": "8a9451b9-d7e8-5417-b6a5-5fd1b791cc4d",
              "extraction_id": "d7bd898b-1d46-557a-b065-f94fc5310b2a",
              "associatedQuery": "What genes are most commonly associated with an increased risk of developing diabetes?"
            }
          },
          {
            "id": "437a7129-63b8-5f34-8273-2eef9535e987",
            "score": 0.7428935170173645,
            "metadata": {
              "text": "gene are associated with NIDDM in Caucasians. Diabetes 1996 , 45, 825-831. \n46.  Tarasov, A.I.; Nicolson, T.J. ; Riveline, J.P.; Taneja, T.K. ; Baldwin, S.A.; Baldwin, J.M.; \nCharpentier, G.; Gautier, J.F. ; Froguel, P.; Vaxillaire, M.; et al.  A rare mutation in ABCC8/SUR1 \nleading to altered ATP-sensitive K+ channel activ ity and beta-cell glucose sensing is associated \nwith type 2 diabetes in adults. Diabetes 2008 , 57, 1595-1604.",
              "title": "2010 - Pharmacogenetics of Anti-Diabetes Drugs.pdf",
              "version": "v0",
              "chunk_order": 184,
              "document_id": "ffeebaf9-ff76-5751-9b8b-7a2a4a4f1dc3",
              "extraction_id": "73e1aaff-7ef6-5ca2-9c94-23f5674a4f88",
              "associatedQuery": "What genes are most commonly associated with an increased risk of developing diabetes?"
            }
          },
          {
            "id": "aa72551a-ac0c-5d7d-8057-34f229f68eb1",
            "score": 0.7374500036239624,
            "metadata": {
              "text": "ly associated with type 2 diabetes: TCF7L2, KCNJ11,  \nand PPARG .\n5-7 However, in 2007, a number of novel \ngenetic variants ( CDKAL1, IGF2BP2,  the locus on \nchromosome 9 close to CDKN2A/CDKN2B, FTO, \nHHEX, SLC30A8,  and WFS1)8-14 were shown to in -\ncrease susceptibility to type 2 diabetes in repro -\nducible studies. Furthermore, a recent meta-analy -\nsis identified six novel variants ( JAZF1, CDC123/\nCAMK1D, TSPAN8/LGR5, THADA, ADAMTS9, and NOTCH2 ) that are associated with type 2 dia -\nbetes.\n15",
              "title": "2008 - Clinical Risk Factors, DNA Variants.pdf",
              "version": "v0",
              "chunk_order": 10,
              "document_id": "0018610a-9c86-5e2d-a27d-f66cf4f8519d",
              "extraction_id": "2643b341-8c50-5cea-af36-86a8b070a80e",
              "associatedQuery": "What genes are most commonly associated with an increased risk of developing diabetes?"
            }
          },
          {
            "id": "461b6f32-4dd8-5dc1-b69f-134f949fc021",
            "score": 0.7339475154876709,
            "metadata": {
              "text": "CDKAL1 in  uences insulin response and risk of type 2 diabetes. Nat Genet 2007; 39: 77075.\n69 Wu Y , Li H, Loos RJ, et al. Common variants in CDKAL1, CDKN2A/\nB, IGF2BP2, SLC30A8, and HHEX/IDE genes are associated with type 2 diabetes and impaired fasting glucose in a Chinese Han population. Diabetes 2008; 57: 283442.\n70 Sandhu MS, Weedon MN, Fawcett KA, et al. Common variants in \nWFS1 confer risk of type 2 diabetes. Nat Genet 2007; 39: 95153.",
              "title": "2010 - Diabetes in Asia.pdf",
              "version": "v0",
              "chunk_order": 126,
              "document_id": "0be842b8-7f69-503b-baed-c336e5c834d6",
              "extraction_id": "11faf4fe-7b71-562e-9901-c428ab20b285",
              "associatedQuery": "What genes are most commonly associated with an increased risk of developing diabetes?"
            }
          },
          {
            "id": "263dc0cb-dfa0-5ee2-b927-f9a196294d46",
            "score": 0.7332150936126709,
            "metadata": {
              "text": "Genes signifying increased risk for both type 1 and type 2 dia-betes have been identified. Genomewide association studies have identified over 50 loci associated with an increased genetic risk of type 1 diabetes. Several T1D candidate genes for increased risk of developing type 1 diabetes have been sug-gested or identified within these regions, but the molecular basis by which they contribute to islet cell inflammation and beta cell destruction is not fully understood.\n12 Also, several",
              "title": "2015 -precision-medicine-for-managing-diabetes.pdf",
              "version": "v0",
              "chunk_order": 27,
              "document_id": "80949bab-d085-5f61-b98a-4bee043bc4e2",
              "extraction_id": "f53ccf4e-f47f-5b44-8b41-f7068efc8be3",
              "associatedQuery": "What genes are most commonly associated with an increased risk of developing diabetes?"
            }
          },
          {
            "id": "78d81651-7215-596a-b128-37e429dc7edb",
            "score": 0.7301425933837891,
            "metadata": {
              "text": "associated with susceptibility to type 2 diabetes mellitus. Nat Genet 2008; 40: 109297 .\n74 Unoki H, Takahashi A, Kawaguchi T, et al. SNPs in KCNQ1 are \nassociated with susceptibility to type 2 diabetes in East Asian and European populations. Nat Genet 2008; 40: 1098102.\n75 Lyssenko V, Lupi R, Marchetti P, et al. Mechanisms by which \ncommon variants in the TCF7L2 gene increase risk of type 2 diabetes. J Clin Invest 2007; 117: 215563. \n76 Lyssenko V, Jonsson A, Almgren P, et al. Clinical risk factors, DNA",
              "title": "2010 - Diabetes in Asia.pdf",
              "version": "v0",
              "chunk_order": 128,
              "document_id": "0be842b8-7f69-503b-baed-c336e5c834d6",
              "extraction_id": "11faf4fe-7b71-562e-9901-c428ab20b285",
              "associatedQuery": "What genes are most commonly associated with an increased risk of developing diabetes?"
            }
          },
          {
            "id": "b0d3a09d-36a3-5c6e-a110-3fccddaa74b7",
            "score": 0.7266103245797854,
            "metadata": {
              "text": "type 2 diabetes or the inability to replicate linkage withdened loci. However, at least one susceptibility gene, namelyCAPN10, was found using a genome-wide scan approach [3].\nObesity is the greatest risk factor for type 2 diabetes\nmellitus, as it is known to induce insulin resistance via variousmechanisms ( TNF \n \n release, free fatty acids, etc.). Both",
              "title": "2003 - Genome-wide screen in obese pedigrees with type 2 diabetes.pdf",
              "version": "v0",
              "chunk_order": 5,
              "document_id": "335a3c08-14d3-5511-ab84-340e64c6f993",
              "extraction_id": "1110f7b4-ab5a-5b41-b37d-a992b29cb20c",
              "associatedQuery": "What genes are most commonly associated with an increased risk of developing diabetes?"
            }
          },
          {
            "id": "e6c0f12d-8136-5a16-b77c-88dd17c3a212",
            "score": 0.7217490242869257,
            "metadata": {
              "text": "50 most cases of type 2 diabetes are thought to be \ndue to genetic variations that are more common but exert less e  ect. In early studies, genetic variants in the peroxisome proliferator-activated receptor- gene (PPARG)\n51 and the ATP-sensitive potassium channel \nKir62 (KCNJ11) were reproducibly associated with type 2 diabetes.\n52 In Asian populations, the protective e  ect of \nthe PPARG*A12Ala allele on insulin resistance and risk of type 2 diabetes was not consistently seen.\n53",
              "title": "2010 - Diabetes in Asia.pdf",
              "version": "v0",
              "chunk_order": 19,
              "document_id": "0be842b8-7f69-503b-baed-c336e5c834d6",
              "extraction_id": "e99fe157-eda9-5e56-9ec9-8f428de2a161",
              "associatedQuery": "What genes are most commonly associated with an increased risk of developing diabetes?"
            }
          },
          {
            "id": "d632d486-4e04-5c2d-9cf0-9d614453cab3",
            "score": 0.7215678691864014,
            "metadata": {
              "text": "49. Cornelis MC, Qi L, Zhang C, et al. Joint e ects of common genetic\nvariants on the risk for type 2 diabetes in U.S. men and women ofEuropean ancestry. Ann Intern Med . 2009;150:541 550(in eng).\n50. Hu C, Zhang R, Wang C, et al. PPARG, KCNJ11, CDKAL1,\nCDKN2A-CDKN2B, IDE-KIF11-HHEX, IGF2BP2 and SLC30A8are associated with type 2 diabetes in a Chinese population. PLoS\nOne. 2009;4:e7643 (in eng).\n51. Lin X, Song K, Lim N, et al. Risk prediction of prevalent diabetes in",
              "title": "2018 - Quantitative Relationship Between Cumulative Risk Alleles Based.pdf",
              "version": "v0",
              "chunk_order": 183,
              "document_id": "d585896e-1c32-51cb-827d-e4fd3b3943f3",
              "extraction_id": "6db9f25e-36fd-51c0-be36-6dfacd963b1b",
              "associatedQuery": "What genes are most commonly associated with an increased risk of developing diabetes?"
            }
          },
          {
            "id": "e1ba568f-cc08-549a-9c87-a23285c3b5dc",
            "score": 0.7215024994908287,
            "metadata": {
              "text": "46. Sladek R, Rocheleau G, Rung J et al (2007) A genome-wide asso-\nciation study identifies novel risk loci for type 2 diabetes. Nature\n445:881 885\n47. Lauenborg J, Grarup N, Damm P et al (2009) Common type 2\ndiabetes risk gene variants associate with gestational diabetes. J\nClin Endocrinol Metab 94:145 150\n48. Florez JC, Jablonski KA, Bayley N et al (2006) TCF7L2 polymor-\nphisms and progression to diabetes in the Diabetes Prevention\nProgram. N Engl J Med 355:241 250",
              "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": 106,
              "document_id": "3b301dd1-17bd-5632-9a96-d6294c6d7650",
              "extraction_id": "f6de8981-a79b-5817-b688-a20f76bff86c",
              "associatedQuery": "What genes are most commonly associated with an increased risk of developing diabetes?"
            }
          }
        ],
        "kg_search_results": null
      }
    }
  },
  "12": {
    "results": {
      "completion": {
        "id": "chatcmpl-AIFqbEtJpAtIpQor3Q5twvd1eoH61",
        "choices": [
          {
            "finish_reason": "stop",
            "index": 0,
            "message": {
              "content": "Genetic testing can help predict a person's risk for diabetes in several ways:\n\n1. **Tailored Interventions**: Knowing an individual's genotype can allow for the development of personalized lifestyle intervention programs aimed at preventing or significantly delaying the onset of type 2 diabetes [1].\n\n2. **Role of Genetic Factors**: Genetic factors play a role in determining an individual's risk of developing diabetes, suggesting that genetic testing can help identify those at higher risk [2].\n\n3. **Genetic Risk Scores**: A genotype risk score can predict type 2 diabetes from a young age, as demonstrated in studies like the CARDIA study [6]. This score can help identify individuals who are at increased risk due to their genetic makeup.\n\n4. **Heritability and Risk Assessment**: Type 2 diabetes is heritable, and genetic testing can help identify individuals with a familial risk, which is increased by a factor of 2 to 6 compared to those without familial diabetes [7].\n\n5. **Improved Prediction and Stratification**: Genetic testing offers the potential for improved prediction and stratification of patients according to their risk, which can aid in selecting possible therapeutic targets [8].\n\n6. **Identification of Genetic Variants**: By genotyping specific single nucleotide polymorphisms (SNPs) associated with diabetes, genetic testing can improve the ability to detect who will ultimately develop the disease [9].\n\nOverall, genetic testing provides valuable insights into an individual's risk for diabetes, enabling more targeted prevention and management strategies.",
              "role": "assistant"
            }
          }
        ],
        "created": 1728914185,
        "model": "gpt-4o-2024-08-06",
        "object": "chat.completion",
        "system_fingerprint": "fp_6b68a8204b",
        "usage": {
          "completion_tokens": 303,
          "prompt_tokens": 1059,
          "total_tokens": 1362
        }
      },
      "search_results": {
        "vector_search_results": [
          {
            "id": "28d6dfa3-28eb-537b-ad53-7d312f20fc88",
            "score": 0.7473591566085815,
            "metadata": {
              "text": "genetic knowledge beyond its use for predic-tion of the individuals type 2 diabetes risk?One major advantage of knowing an at-riskpersons genotype could be to offer an individ-ually tailored lifestyle intervention program to\nprevent or, at least, to significantly retard the",
              "title": "2009 - Pathomechanisms of Type 2 Diabetes Genes.pdf",
              "version": "v0",
              "chunk_order": 161,
              "document_id": "cf8ec75c-8ffe-5baa-830d-ac7a4a5964bd",
              "extraction_id": "9c49d40d-91d3-5f0d-8eaa-b3efa49ac200",
              "associatedQuery": "How can genetic testing help predict a person's risk for diabetes?"
            }
          },
          {
            "id": "54ff4672-bf7f-5158-b228-ca3d45e0cb0d",
            "score": 0.7451751232147217,
            "metadata": {
              "text": "Genetic factors appear to play a role in determining an individuals risk of developing diabetes. It is hoped",
              "title": "2008 - Glossary of Genetics Genomics Terms.pdf",
              "version": "v0",
              "chunk_order": 2,
              "document_id": "c66d2572-071d-5aaf-829c-b3ca6cf6d697",
              "extraction_id": "53e868dd-b318-5cf3-8b2e-98a548aab7cf",
              "associatedQuery": "How can genetic testing help predict a person's risk for diabetes?"
            }
          },
          {
            "id": "71ebe60b-4807-5b6f-887a-2ab897a46039",
            "score": 0.7436718940734863,
            "metadata": {
              "text": "(35).\nIf genetic tests are not helpful in the\nprediction and prevention of diabetes,they could have a role in discriminatingbetween type 1 and type 2 diabetes. Theepidemic of obesity (36) has made it moredifcult to distinguish diabetes type be-\ncause many children and young adultswith type 1 diabetes are also obese (37).Misclassi cation poses signi cant risks;\nan incorrect diagnosis of type 2 diabetes",
              "title": "2016 - The Application of Genomics.pdf",
              "version": "v0",
              "chunk_order": 19,
              "document_id": "2ec5c9c1-fe53-59ca-b36f-d360dfce0da5",
              "extraction_id": "7aa2ab48-620b-5b30-b2de-103e103579ba",
              "associatedQuery": "How can genetic testing help predict a person's risk for diabetes?"
            }
          },
          {
            "id": "6cf756f6-bc3a-515a-a879-7270f663c516",
            "score": 0.7371379137039185,
            "metadata": {
              "text": "geted at specific genetic mutations, it is likely that accompa-nying diagnostic tests for biomarkers will also become available to confirm whether the target biomarker is present.\nGenomic Analyses for Diabetes Risk",
              "title": "2015 -precision-medicine-for-managing-diabetes.pdf",
              "version": "v0",
              "chunk_order": 26,
              "document_id": "80949bab-d085-5f61-b98a-4bee043bc4e2",
              "extraction_id": "f53ccf4e-f47f-5b44-8b41-f7068efc8be3",
              "associatedQuery": "How can genetic testing help predict a person's risk for diabetes?"
            }
          },
          {
            "id": "59b0a653-0d03-582e-8fb5-009af723b984",
            "score": 0.736946485547617,
            "metadata": {
              "text": "genes improves prediction of type 1 diabetes[published correction appears in Diabetologia. 2015;\n58(1):206]. Diabetologia . 2014; 57(12):2521 2529.\n57. Oram RA, Patel K, Hill A, Shields B, McDonald TJ,\nJones A, Hattersley AT, Weedon MN. A type 1\ndiabetes genetic risk score can aid discrimination\nbetween type 1 and type 2 diabetes in young adults.Diabetes Care . 2016; 39(3):337 344.\n58. Redondo MJ, Oram RA, Steck AK. Genetic risk",
              "title": "2019 - Genetic Risk Scores for Diabetes Diagnosis.pdf",
              "version": "v0",
              "chunk_order": 246,
              "document_id": "8c66aca1-d4ba-534d-a037-4273de340ee1",
              "extraction_id": "ba3abde6-0fac-587f-976e-bd0e08c48ae3",
              "associatedQuery": "How can genetic testing help predict a person's risk for diabetes?"
            }
          },
          {
            "id": "9d44b00e-027f-557f-a851-e870605ea20f",
            "score": 0.7363494634628296,
            "metadata": {
              "text": "10.2337/db13-1663.\n 20. Vassy JL, et al. A genotype risk score predicts type 2 diabetes from young \nadulthood: the CARDIA study. Diabetologia. 2012;55:26042612. doi: \n10.1007/s00125-012-2637-7.\n 21. Vassy JL, et al. Is genetic testing useful to predict type 2 diabe-tes? Best Pract Res Clin Endocrinol Metab. 2012;26:189201. doi: \n10.1016/j.beem.2011.09.002.\n 22. Khera AV, et al. Genome-wide polygenic score to identify a monogenic \nrisk-equivalent for coronary disease. bioRxiv. 2017. doi: 10.1101/218388.",
              "title": "2018 - Human Genetics of Obesity and Type 2 Diabetes Mellitus.pdf",
              "version": "v0",
              "chunk_order": 126,
              "document_id": "2083de31-17c6-5d1e-9aa6-2efc6c1d9ac2",
              "extraction_id": "e9c258eb-26f2-5e33-87a2-7ac5a5b29989",
              "associatedQuery": "How can genetic testing help predict a person's risk for diabetes?"
            }
          },
          {
            "id": "f0ca71ce-f2bb-54f2-a933-dc9c952f1eb8",
            "score": 0.7260829426670955,
            "metadata": {
              "text": "Genotype Score for Prediction of Type 2 Diabetes\nn engl j med 359;21 www.nejm.org november 20, 2008 2209Type 2 diabetes mellitus is a m ajor \nhealth problem worldwide.1 Fortunately, \nits development can be prevented in many \ninstances,2 and persons at risk can be readily \nidentified with the measurement of a few com -\nmon risk factors.3-5 Type 2 diabetes is heritable, \nwith a risk for people with familial diabetes as \ncompared with those without familial diabetes that is increased by a factor of 2 to 6.",
              "title": "2008 - Genotype Score in Addition to Common Risk Factors for Prediction of Type 2 Diabetes.pdf",
              "version": "v0",
              "chunk_order": 9,
              "document_id": "fb502e5b-7094-58aa-9508-103476a9c035",
              "extraction_id": "e0f816e4-3c97-575e-8bbe-0e006c8c8e61",
              "associatedQuery": "How can genetic testing help predict a person's risk for diabetes?"
            }
          },
          {
            "id": "e32de26a-7ad6-51a9-860e-5df0b45d981d",
            "score": 0.7240273356437683,
            "metadata": {
              "text": "risk of type 1 diabetes offers the potential for improved prediction, stratification of patients according to risk, and selection of possible therapeutic targets. As germ-line factors, genetic risk variants are present and amenable to study at all times  be -",
              "title": "2009 - Genetics of Type 1A Diabetes.pdf",
              "version": "v0",
              "chunk_order": 6,
              "document_id": "7a98f456-6c43-5e9e-b404-31122159eab8",
              "extraction_id": "d3fa98dd-b7be-5192-9a7c-71742b1b05e4",
              "associatedQuery": "How can genetic testing help predict a person's risk for diabetes?"
            }
          },
          {
            "id": "b677fe54-5f7e-5d87-a16d-6694578c6f2b",
            "score": 0.7192047834396362,
            "metadata": {
              "text": "offers the opportunity to test whetherknowledge of these genetic loci canimprove our ability to detect who willultimately develop diabetes. To answerthis question, we genotyped 18 well-validated single nucleotide polymorph-isms that had previously been associat-\ned with diabetes in large genetics",
              "title": "2010 - Cardiovascular Disease Risk Factors, Type 2 Diabetes Mellitus, and the Framingham Heart Study.pdf",
              "version": "v0",
              "chunk_order": 50,
              "document_id": "134c506f-f66f-5a17-9e81-1f4c5923fe91",
              "extraction_id": "5763fc63-1abb-5baf-b2ed-ad1b019bdb56",
              "associatedQuery": "How can genetic testing help predict a person's risk for diabetes?"
            }
          },
          {
            "id": "530788ae-3a97-50d6-ad96-5463a3dc75e8",
            "score": 0.7177377343177795,
            "metadata": {
              "text": "Comprehension of Genomic Risk for \nDiabetes  Public Health Genomics 2014;17:95104 \nDOI: 10.1159/000358413101their results in-person from a genetic counselor were able \nto correctly indicate their genomic or lifetime risk score for T2DM and interpret their genomic risk, compared to 50% of participants receiving their results online. This finding aligns with reports that suggest genetic counsel-ing (though limited to reporting of test results in this study) improves patients accuracy of risk perception",
              "title": "2014 -  Impact of Delivery Models on Understanding Genomic Risk for Type 2 Diabetes.pdf",
              "version": "v0",
              "chunk_order": 80,
              "document_id": "b2665466-da66-59f0-8581-a68131e924bf",
              "extraction_id": "aafcb80d-7069-59da-8a21-d6a32f1a8820",
              "associatedQuery": "How can genetic testing help predict a person's risk for diabetes?"
            }
          }
        ],
        "kg_search_results": null
      }
    }
  },
  "13": {
    "results": {
      "completion": {
        "id": "chatcmpl-AIFqiY2VOktGY4xVSkvpvMDbynoMw",
        "choices": [
          {
            "finish_reason": "stop",
            "index": 0,
            "message": {
              "content": "Family genetics play a significant role in the likelihood of developing diabetes. Several studies and observations highlight this connection:\n\n1. Genetic factors are important in determining an individual's risk of developing diabetes [1].\n2. A family history of diabetes, particularly in first-degree relatives such as parents or siblings, is associated with a two- to fourfold increased risk of developing diabetes [3].\n3. Type 2 diabetes is strongly influenced by genetics, as evidenced by high concordance rates in identical twins, with studies showing a 58-75% concordance rate [5]. In some studies, the concordance rate is reported to be nearly 100% [6].\n4. The risk of developing type 2 diabetes is approximately 70% if both parents have the disease and about 40% if one parent is affected [7], [9].\n5. Type 2 diabetes clusters in families, and having a first-degree relative with the disease increases the lifetime risk significantly, up to 40% or more [9].\n\nOverall, these findings underscore the strong genetic component in the susceptibility to diabetes, particularly type 2 diabetes.",
              "role": "assistant"
            }
          }
        ],
        "created": 1728914192,
        "model": "gpt-4o-2024-08-06",
        "object": "chat.completion",
        "system_fingerprint": "fp_a20a4ee344",
        "usage": {
          "completion_tokens": 224,
          "prompt_tokens": 1244,
          "total_tokens": 1468
        }
      },
      "search_results": {
        "vector_search_results": [
          {
            "id": "54ff4672-bf7f-5158-b228-ca3d45e0cb0d",
            "score": 0.755399763584137,
            "metadata": {
              "text": "Genetic factors appear to play a role in determining an individuals risk of developing diabetes. It is hoped",
              "title": "2008 - Glossary of Genetics Genomics Terms.pdf",
              "version": "v0",
              "chunk_order": 2,
              "document_id": "c66d2572-071d-5aaf-829c-b3ca6cf6d697",
              "extraction_id": "53e868dd-b318-5cf3-8b2e-98a548aab7cf",
              "associatedQuery": "What role do family genetics play in the likelihood of getting diabetes?"
            }
          },
          {
            "id": "03dbb574-1b16-5300-af34-08b82263388e",
            "score": 0.7400701642036438,
            "metadata": {
              "text": "Metabolic Syndrome and Family History \nof Diabetes Public Health Genomics 2010;13:353359 357able difference in the odds between these 2 risk levels. \nThis table indicates that, compared with the average fa-milial risk, a moderate or high familial risk of diabetes increases the odds for each single component of the met-a b o l i c  s y n d r o m e .  T h e s e  o d d s  v a r y  f r o m  1 . 1 9  ( 9 5 %  C I :  0.881.61) to 1.53 (95% CI: 1.301.81).\n  C o n c l u s i o n",
              "title": "2010 - Family History of Diabetes and Prevalence.pdf",
              "version": "v0",
              "chunk_order": 51,
              "document_id": "f16c4c6e-bb5f-5d4a-9945-8af4d0df19f4",
              "extraction_id": "b91922c6-7b5b-5fa1-a740-4564ec4cfa36",
              "associatedQuery": "What role do family genetics play in the likelihood of getting diabetes?"
            }
          },
          {
            "id": "13fa34fd-9bf6-5ae5-8a7e-e1998d56d084",
            "score": 0.721138553551523,
            "metadata": {
              "text": "For type 2 diabetes, there have been a few \nstudies utilising a candidate-gene approach as well as genome-wide association studies, although some argue that genetic factors play only a minor role among Caribbean populations [ \n90 ]. A family history of diabetes in any   rst- \ndegree relative (parent, sibling) or in a grandpar-ent is associated with a two- to fourfold increased risk of diabetes [ \n10 ,  91 ]. A family history of dia-",
              "title": "2017 - diabetes-mellitus-in-developing-countries-and-underserved-commun-2017.pdf",
              "version": "v0",
              "chunk_order": 1363,
              "document_id": "8a9451b9-d7e8-5417-b6a5-5fd1b791cc4d",
              "extraction_id": "5ae0e120-7064-5ced-84ff-e74fb0f90047",
              "associatedQuery": "What role do family genetics play in the likelihood of getting diabetes?"
            }
          },
          {
            "id": "527419f1-075d-5d53-a8b5-1685952ecdb0",
            "score": 0.7160833647201452,
            "metadata": {
              "text": "evidenced by a very high positive rate of family history of diabetes, and \ndrastically different prevalence in various ethnic groups. Therefore, there is \nno doubt that type 2 diabetes is a disease with a strong genetic influence. \nHowever, the prediction of the relative contribution of genetic influence and \nnumber of genes involved in the pathogenesis of the disease has changed in \nthe past few years. Initially, enthusiastic searches of diabetes genes were",
              "title": "2004 - Diabetes Genes a.pdf",
              "version": "v0",
              "chunk_order": 638,
              "document_id": "805ad0d5-6a3d-5e5f-b1ba-121e10c6daaa",
              "extraction_id": "40d292c1-03bc-5780-a2ae-9b0fe245f39c",
              "associatedQuery": "What role do family genetics play in the likelihood of getting diabetes?"
            }
          },
          {
            "id": "3a807b66-fcae-5cae-b8ad-83a5c6815221",
            "score": 0.7089239954948425,
            "metadata": {
              "text": "can decrease risk of diabetes.22 Diet may also play a role. High calorie diets, \nincluding those high in fat, and especially saturated fat, have been implicated \nin the development of type 2 diabetes?4-26 Family history is a very strong risk \nfactor for type 2 diabetes. A strong genetic component is suggested by the \n58-75% concordance rates for type 2 diabetes observed in identical twins \n(Table 3).3 \nTable 3. Estimated risk of developing type 2 diabetes by family history \nOne parent with type 2 diabetes",
              "title": "2004 - Diabetes Genes a.pdf",
              "version": "v0",
              "chunk_order": 570,
              "document_id": "805ad0d5-6a3d-5e5f-b1ba-121e10c6daaa",
              "extraction_id": "8e5322e6-a8a2-5d98-b87d-1ba3846d5fe1",
              "associatedQuery": "What role do family genetics play in the likelihood of getting diabetes?"
            }
          },
          {
            "id": "b63c48dd-b954-56d4-bdfa-8ab135e7bf47",
            "score": 0.7072950179997088,
            "metadata": {
              "text": "The fact that type 2 diabetes is a genetic disease is well\nknown to clinicians by how it occurs in families, and by\nthere being ethnic populations who are particularly high risk.\nThe genetic link was clearly shown more than two decades\nago by a famous study of identical twins in the U.K. that\nfound essentially a 100% concordance rate for this disease\nif one twin developed type 2 diabetes, then the other one\ninvariably developed it (9). However, this kind of study",
              "title": "2005 - Pathogenesis of Type 2 Diabetes Mellitus.pdf",
              "version": "v0",
              "chunk_order": 11,
              "document_id": "75b4ae7d-7abf-57b8-bda9-5b022d698ae6",
              "extraction_id": "d62a1716-bd6a-5532-ab22-ee6e7ec4cf37",
              "associatedQuery": "What role do family genetics play in the likelihood of getting diabetes?"
            }
          },
          {
            "id": "ee3d0900-a422-59cd-a6db-308f20052cc0",
            "score": 0.7053035497665405,
            "metadata": {
              "text": "genetic factors play an important role in the susceptibility to T2D. The risk of the disease developing\nat some point of life is ~70% when both parents are diabetic and ~40% when one parent has T2D [ 4].\nFurthermore, latest data show that more than 400 genetic risk variants at 250 loci for T2D have been\nGenes 2018 ,9, 374; doi:10.3390/genes9080374 www.mdpi.com/journal/genes",
              "title": "2018 - High-Throughput Approaches onto Uncover (Epi)Genomic Architecture of Type 2 Diabetes.pdf",
              "version": "v0",
              "chunk_order": 7,
              "document_id": "1cb0c4ac-c1fe-55c2-919c-52cd5018c00d",
              "extraction_id": "f6b9d6b9-a60b-56f5-9727-d90d43efe0ac",
              "associatedQuery": "What role do family genetics play in the likelihood of getting diabetes?"
            }
          },
          {
            "id": "2aa9f009-ae05-5c93-ac3a-58b1f516d844",
            "score": 0.7022915659969124,
            "metadata": {
              "text": "36 Herder C, Roden M. Genetics of type 2 diabetes: pathophysiologic \nand clinical relevance. Eur J Clin Invest 2011; 41: 67992.\n37 Dabelea D, Hanson RL, Lindsay RS, et al. Intrauterine exposure \nto diabetes conveys risks for type 2 diabetes and obesity: a study of discordant sibships. Diabetes 2000; 49: 220811.\n38 Voight BF, Scott LJ, Steinthorsdottir V, et al. Twelve type 2 diabetes \nsusceptibility loci identi  ed through large-scale association analysis. Nat Genet 2010; 42: 57989.",
              "title": "2011 - Type 2 diabetes across generations from pathophysiology to prevention and management.pdf",
              "version": "v0",
              "chunk_order": 151,
              "document_id": "0f49b102-1d7e-5702-af30-35e5f2ed93a6",
              "extraction_id": "baec13ec-c42b-51b4-9974-8ef1c2d10ddc",
              "associatedQuery": "What role do family genetics play in the likelihood of getting diabetes?"
            }
          },
          {
            "id": "353dc970-3106-5bbe-8a58-d65d13e5e6ee",
            "score": 0.7021226705385859,
            "metadata": {
              "text": "long follow-up. Type 2 diabetes and impaired glucose tolerance (IGT) \ncluster in families. Thus, most patients have a positive family history, and \nthe lifetime risk for developing type 2 diabetes is increased up to 40% (more \nthan five times the background rate) by having a first degree relative with the \ndisease. If both parents have type 2 diabetes the risk to the offspring may be \nas high as 70%. Available evidence supports a polygenic mode of \ninheritance with a considerable environmental input. 1",
              "title": "2004 - Diabetes Genes a.pdf",
              "version": "v0",
              "chunk_order": 938,
              "document_id": "805ad0d5-6a3d-5e5f-b1ba-121e10c6daaa",
              "extraction_id": "5a2221e0-dabc-523c-8358-3e43789e8f7a",
              "associatedQuery": "What role do family genetics play in the likelihood of getting diabetes?"
            }
          },
          {
            "id": "6c14eef8-bb27-503a-9523-9e7a16d71021",
            "score": 0.7017937898635864,
            "metadata": {
              "text": "Genetic factors\nType 2 diabetes has a strong genetic component and most Asian patients have a   rst-degree relative with diabetes.\n48,49 Much progress has been made in our \nunderstanding of the genetics of this disease. Importantly, most of the loci originally associated with diabetes in European populations have been replicated in Asian populations. Whereas monogenic forms of diabetes result from rare genetic mutations with large e  ects, \nsuch as those seen in maturity-onset diabetes of young people,",
              "title": "2010 - Diabetes in Asia.pdf",
              "version": "v0",
              "chunk_order": 18,
              "document_id": "0be842b8-7f69-503b-baed-c336e5c834d6",
              "extraction_id": "e99fe157-eda9-5e56-9ec9-8f428de2a161",
              "associatedQuery": "What role do family genetics play in the likelihood of getting diabetes?"
            }
          }
        ],
        "kg_search_results": null
      }
    }
  },
  "14": {
    "results": {
      "completion": {
        "id": "chatcmpl-AIFqrzKmzcOBxhh6XTfMBqYsubXv7",
        "choices": [
          {
            "finish_reason": "stop",
            "index": 0,
            "message": {
              "content": "Yes, lifestyle changes can affect genetic risk factors for diabetes. Evidence from both observational studies and clinical trials suggests that lifestyle factors can modify the genetic effects on the risk of developing type 2 diabetes mellitus (T2DM) [1]. Lifestyle modifications, such as improvements in dietary quality and increased physical activity, have been shown to be particularly effective in preventing and managing type 2 diabetes, even in individuals with a high genetic risk [6]. Furthermore, behavioral changes can substantially mitigate the diabetogenic effects of individual or multiple risk alleles, indicating that the adverse effects of some established T2D-associated loci may be greatly attenuated by appropriate lifestyle changes [9].",
              "role": "assistant"
            }
          }
        ],
        "created": 1728914201,
        "model": "gpt-4o-2024-08-06",
        "object": "chat.completion",
        "system_fingerprint": "fp_a20a4ee344",
        "usage": {
          "completion_tokens": 132,
          "prompt_tokens": 1075,
          "total_tokens": 1207
        }
      },
      "search_results": {
        "vector_search_results": [
          {
            "id": "a1c71566-1d75-551a-8588-9a05436545dc",
            "score": 0.7656337758520989,
            "metadata": {
              "text": "of a given genetic variant is modified by the environ -\nmental milieu (and vice versa). Evidence that lifestyle factors modify the genetic effects on T2DM risk has been \ngenerated from both observational studies and clinical \ntrials82. However, genetic background might also affect \nthe individuals response to lifestyle interventions83. In \naddition, replication data are sparse, and comprehensive, \nlarge-scale studies have failed to provide a compelling",
              "title": "2018 - Global aetiology and epidemiology of type 2 diabetes mellitus and its complications.pdf",
              "version": "v0",
              "chunk_order": 65,
              "document_id": "8bc8f3d4-968f-5252-ab4c-832b92e9ec0d",
              "extraction_id": "751ccb98-2846-5ca7-8ab8-2684100c28fa",
              "associatedQuery": "Can lifestyle changes affect genetic risk factors for diabetes?"
            }
          },
          {
            "id": "fe89ba68-d709-5494-bcdc-82d81e1498d1",
            "score": 0.7589735984802246,
            "metadata": {
              "text": "genetic risk for diabetes may not moti-vate improvements in lifestyle behaviors.Indeed, knowledge of increased geneticrisk for diabetes may decrease motiva-tion to modify behavior in genetic fatal-ists (83).\nDiet recommendations optimized to\nthe individual have been shown to re-duce postprandial glycemic excursionsto a greater extent than standard\napproaches in healthy individuals (84).Meal compositions that induce the most\nfavorable glycemic pro les have been",
              "title": "2020 - Precision Medicine in Diabetes.pdf",
              "version": "v0",
              "chunk_order": 136,
              "document_id": "0ad5b2de-d782-5d43-b294-bff5c7befd2d",
              "extraction_id": "0504a937-6b88-5004-a13e-5e9c3073eaf6",
              "associatedQuery": "Can lifestyle changes affect genetic risk factors for diabetes?"
            }
          },
          {
            "id": "799f3578-a7ac-551f-b84a-b9fb3be53040",
            "score": 0.7469942569732666,
            "metadata": {
              "text": "diabetes regardless of the underlying\ngenetic risk. This contrasts with theextensive epidemiological evidence sug-gesting that the relationship of lifestylewith obesity is dependent on genetic risk(7881); however, with few exceptions\n(e.g., [74]), analyses in large randomizedcontrolled trials have failed to show thatthese same genetic variants modifyweight loss in response to lifestyle in-tervention (82). It is also important to\nrecognize that knowledge of increased",
              "title": "2020 - Precision Medicine in Diabetes.pdf",
              "version": "v0",
              "chunk_order": 135,
              "document_id": "0ad5b2de-d782-5d43-b294-bff5c7befd2d",
              "extraction_id": "0504a937-6b88-5004-a13e-5e9c3073eaf6",
              "associatedQuery": "Can lifestyle changes affect genetic risk factors for diabetes?"
            }
          },
          {
            "id": "54ff4672-bf7f-5158-b228-ca3d45e0cb0d",
            "score": 0.7338041067123413,
            "metadata": {
              "text": "Genetic factors appear to play a role in determining an individuals risk of developing diabetes. It is hoped",
              "title": "2008 - Glossary of Genetics Genomics Terms.pdf",
              "version": "v0",
              "chunk_order": 2,
              "document_id": "c66d2572-071d-5aaf-829c-b3ca6cf6d697",
              "extraction_id": "53e868dd-b318-5cf3-8b2e-98a548aab7cf",
              "associatedQuery": "Can lifestyle changes affect genetic risk factors for diabetes?"
            }
          },
          {
            "id": "be87703d-e7b2-5db5-9983-5412e09a57ba",
            "score": 0.7328479290008545,
            "metadata": {
              "text": "suggested to attenuate its negative e  ect on metabolic pro  le, \nbody weight, and diabetes risk (   Franks et al., 2007   ;    Kilpelainen et al., 2008   ;    Lindi et al., 2002   ;    Ruchat et al., 2010   ) (     \n       Table 1   ). \nThe notion that lifestyle modi  cation can eliminate the increased \nrisk for development of T2DM in subjects with genetic suscepti-bility is also supported by  ndings of    Barwell et al. (2008)    who",
              "title": "2011 - Lifestyle and Genetics in Obesity and type 2 Diabetes.pdf",
              "version": "v0",
              "chunk_order": 52,
              "document_id": "a16d3328-039c-530a-bfe5-f6f80ecf2ad0",
              "extraction_id": "93638ea5-6d1f-5b6a-9629-798804de24dd",
              "associatedQuery": "Can lifestyle changes affect genetic risk factors for diabetes?"
            }
          },
          {
            "id": "89339b65-325f-588f-9f25-761124f0012f",
            "score": 0.7295693916339058,
            "metadata": {
              "text": "proven particularly effective for preven-tion and management of type 2 diabetes.For example, improvement in dietaryquality, in conjunction with other lifestylemodications like increased physical ac-tivity, was shown to be more effectivethan pharmacological treatment in pre-vention of diabetes in individuals at highrisk (1). Further, lifestyle modicationmay mitigate the risk associated with thestrongest known diabetes risk loci (2).While the existence of environmental in-uences on genetic risk (and vice",
              "title": "2010 - Interactions of Dietary Whole-Grain Intake.pdf",
              "version": "v0",
              "chunk_order": 6,
              "document_id": "e4d4a19e-18a0-5a08-9ab7-537f31b7cdc1",
              "extraction_id": "6283c124-b479-5050-86ca-dc42390147a1",
              "associatedQuery": "Can lifestyle changes affect genetic risk factors for diabetes?"
            }
          },
          {
            "id": "fe35615a-6df7-548c-b313-4abca69b1e2d",
            "score": 0.7234389997553531,
            "metadata": {
              "text": "who is lean, genetic risk factors are more\nlikely to be present than in someone who\nis obese and develops the disease or that\nweight loss enhances the genetic risk ofdiabetes.\nGenetic analyses performed in clinical\ntrials involving intensive lifestyle modi -\ncation provide an important adjunct to\nthe epidemiological literature on gene-\nlifestyle interactions in type 2 diabetes.On one hand, a major advantage of ran-\ndomized controlled trials is that interac-",
              "title": "2013 - Gene-Environment and Gene-Treatment.pdf",
              "version": "v0",
              "chunk_order": 52,
              "document_id": "fe958fb1-5408-56ec-b102-ccf07b4bac2d",
              "extraction_id": "ee6a4bf3-6f68-58e7-a96f-c879b5269694",
              "associatedQuery": "Can lifestyle changes affect genetic risk factors for diabetes?"
            }
          },
          {
            "id": "68a382e9-85e0-548c-910e-5f24cb48f9c8",
            "score": 0.7233020663261414,
            "metadata": {
              "text": "Lifestyle behaviors and genetic loci have clear and distinguishable effects on \nT2D risk; however, the pattern of disease occurrence within and between popula-tions that differ in their genetic and environmental underpinnings suggests T2D is caused in part by the interaction between adverse lifestyle behaviors and the genetic profile of an individual. For many, this seems a reasonable assumption, but there is little robust empirical evidence supporting the presence of such interactions.",
              "title": "2011 - Interaction Between Exercise and Genetics.pdf",
              "version": "v0",
              "chunk_order": 10,
              "document_id": "c36db75e-4b76-540d-9efb-d0e156e61541",
              "extraction_id": "ed6dcfee-8273-5512-8fb4-fc51a9c921da",
              "associatedQuery": "Can lifestyle changes affect genetic risk factors for diabetes?"
            }
          },
          {
            "id": "6b83f0af-1145-5679-9dae-0f645771d25d",
            "score": 0.7165352871098956,
            "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": "Can lifestyle changes affect genetic risk factors for diabetes?"
            }
          },
          {
            "id": "1b364e28-08e2-5813-b066-7ce37eeb36cf",
            "score": 0.7156190872192383,
            "metadata": {
              "text": "Susceptibility to obesity and diabetes is deter-\nmined by both genetic and lifestyle factors.Suggestive evidence of genelifestyle interac-\ntion (Box 33.3) in the development of common\ndiseases such as obesity and type 2 diabetes wasrst provided by descriptive epidemiological\nstudies such as migration studies that compare\nthe disease risk between genetically related pop-ulations who live different lifestyles. A classicalexample is the comparison of the risk of obesity",
              "title": "2010 - Candidate Gene and Genome-Wide Association Studies in Behavioral Medicine.pdf",
              "version": "v0",
              "chunk_order": 4331,
              "document_id": "17637a6f-804e-50e4-9cf5-37318e17f15c",
              "extraction_id": "3bf3c6a7-de03-5114-bad8-d53fd76d0fba",
              "associatedQuery": "Can lifestyle changes affect genetic risk factors for diabetes?"
            }
          }
        ],
        "kg_search_results": null
      }
    }
  },
  "15": {
    "results": {
      "completion": {
        "id": "chatcmpl-AIFqw6zPQKQT7tNlJNiyf2dx560ep",
        "choices": [
          {
            "finish_reason": "stop",
            "index": 0,
            "message": {
              "content": "Recent breakthroughs in understanding the genetic causes of diabetes include several key advancements:\n\n1. The number of genetic regions causally implicated in both type 1 and type 2 diabetes has increased to 11 each, marking significant progress in identifying genetic factors associated with these conditions [1].\n\n2. Genome-wide association studies have been particularly successful in uncovering genetic determinants of complex diseases like diabetes. These studies have identified a number of novel disease-associated variants, which have been largely replicated by independent research groups, highlighting their reliability and significance [2].\n\n3. Advances in technology, such as the use of single nucleotide polymorphism (SNP) chips, have enabled the analysis of up to a million genetic variations. This technological leap has greatly enhanced our understanding of the genetic basis of complex, multifactorial forms of diabetes, bringing it closer to the understanding of rarer, single-gene disorders [5].\n\n4. Recent studies have yielded six new replicating gene regions associated with type 2 diabetes, providing new insights into the genetic architecture of the disease and suggesting potential directions for future research [8].\n\nThese breakthroughs represent a significant leap forward in the genetic understanding of diabetes, driven by technological advancements and collaborative research efforts.",
              "role": "assistant"
            }
          }
        ],
        "created": 1728914206,
        "model": "gpt-4o-2024-08-06",
        "object": "chat.completion",
        "system_fingerprint": "fp_e5e4913e83",
        "usage": {
          "completion_tokens": 240,
          "prompt_tokens": 1212,
          "total_tokens": 1452
        }
      },
      "search_results": {
        "vector_search_results": [
          {
            "id": "9250b1a6-26d8-5c38-840f-547a9647e809",
            "score": 0.7339037219343364,
            "metadata": {
              "text": "understanding of the genetic basis of diabetes, and the\nadvances of recent months are arguably the most important\nmade since the role of the HLA region was recognised in type1 diabetes. The number of genetic regions causally implicated\nis now 11 each for type 1 and type 2 diabetes [ 19], and is set\nto rise further. The bewildering pace of new discovery standsin stark contrast to the slow progress that characterised the\nprevious two decades, with a total combined output of three",
              "title": "2007 - Genetic studies of diabetes following the advent of the genome-wide association study where do we go from here.pdf",
              "version": "v0",
              "chunk_order": 1,
              "document_id": "7b96d9b2-6494-5c20-9693-dc146a4e347c",
              "extraction_id": "1a155200-3610-528f-a51d-b2f27562037a",
              "associatedQuery": "What recent breakthroughs have been made in understanding the genetic causes of diabetes?"
            }
          },
          {
            "id": "9d55a0b9-d125-587d-b21e-f4bd55b8de28",
            "score": 0.7038693008500767,
            "metadata": {
              "text": "It has proven to be challenging to isolate the genes underlying the genetic\ncomponents conferring susceptibility to type 1 and type 2 diabetes. Unlike\nprevious approaches, genome-wide association studies have extensively\ndelivered on the promise of uncovering genetic determinants of complexdiseases, with a number of novel disease-associated variants being largelyreplicated by independent groups. This review provides an overview of these\nrecent breakthroughs in the context of type 1 and type 2 diabetes, and",
              "title": "2011 - Genome-wide association studies (GWAS) impact.pdf",
              "version": "v0",
              "chunk_order": 2,
              "document_id": "086c6869-7c70-5364-9269-760267fb458d",
              "extraction_id": "cf06774a-9e13-59fd-9652-d5013ef83387",
              "associatedQuery": "What recent breakthroughs have been made in understanding the genetic causes of diabetes?"
            }
          },
          {
            "id": "0372a2d5-28c0-5369-8f05-18f7124eb4ae",
            "score": 0.6993317187230768,
            "metadata": {
              "text": "The history of diabetes genetics traces human genetic research more broadly.Initially, only a few polymorphic genetic markers were known, and these werestudiedinpopulation-basedassociationstudies.Withthedevelopmentofgenome-wide maps for family-based linkage analysis and of positional cloning, attentionturned to monogenic forms of disease. The application of family-based linkagemethods to common forms of diabetes, however, met with less clear success.More recently, with progress in genome sequencing and",
              "title": "2003 - The Inherited Basis of Diabetes Mellitus.pdf",
              "version": "v0",
              "chunk_order": 22,
              "document_id": "7b85b290-d711-55d5-9b1e-b06e4d6f14a2",
              "extraction_id": "238129d2-439f-5a25-8e86-297e7a69d81c",
              "associatedQuery": "What recent breakthroughs have been made in understanding the genetic causes of diabetes?"
            }
          },
          {
            "id": "5b134bfd-6af3-5189-b144-57bf70c2cf20",
            "score": 0.6979157328605702,
            "metadata": {
              "text": "the elucidation of the wide spectrum of genes that \nplayed a role in the molecular mechanism of diabetes \ndevelopment[142-144]. However , despite the vast flow of \ngenetic information including the identification of many \ngene mutations and a large array of single nucleotide \npolymorphisms (SNPs) in many genes involved in the \nmetabolic pathways that affect blood glucose levels, \nthe exact genetic mechanism of diabetes remains \nelusive[145,146]. Evidently, a major complication is the",
              "title": "2015 - Diabetes mellitus The epidemic of the century.pdf",
              "version": "v0",
              "chunk_order": 78,
              "document_id": "e114dd28-fd39-56df-bdeb-8806474a6c10",
              "extraction_id": "6b04dc27-e7ff-53c8-9021-a3cdb5415059",
              "associatedQuery": "What recent breakthroughs have been made in understanding the genetic causes of diabetes?"
            }
          },
          {
            "id": "3cd5df03-7c2b-585c-a3bb-67dc0e1c615c",
            "score": 0.6874925680462988,
            "metadata": {
              "text": "confirmed genes for type 2 diabetes and six for type 1(Fig. 1). At last, it seems, our understanding of the genetic\nbasis of complex, multifactorial forms of diabetes is catching\nup with that of rarer, single-gene disorders.\nThis leap in knowledge is the result of major advances in\ntechnology plus an improved understanding of patterns of\nhuman genetic variation. Using single nucleotide polymor-\nphism (SNP) chips it is now possible to analyse up to a million",
              "title": "2007 - Genetic studies of diabetes following the advent of the genome-wide association study where do we go from here.pdf",
              "version": "v0",
              "chunk_order": 2,
              "document_id": "7b96d9b2-6494-5c20-9693-dc146a4e347c",
              "extraction_id": "1a155200-3610-528f-a51d-b2f27562037a",
              "associatedQuery": "What recent breakthroughs have been made in understanding the genetic causes of diabetes?"
            }
          },
          {
            "id": "9b04e578-bfe5-5f3c-8556-aac26d6429cc",
            "score": 0.6869953274726917,
            "metadata": {
              "text": "make dissection of the black box of genetics of diabetespossible in the near future, but at this point, apart fromthe pro les that distinguish between type 1 and type 2\ndiabetes and a limited number of speci c variants that\nidentify small subgroups of patients (MODY), genetics has\nnot been successful in further differentiating subclasses ofdiabetes.\nResearch Gaps\nAfter consideration of the known genetic associations\nwith diabetes risk, consensus developed that the eld is",
              "title": "2017 - Differentiation of Diabetes by Pathophysiology.pdf",
              "version": "v0",
              "chunk_order": 30,
              "document_id": "9cfaef1e-fb60-5c2b-94f0-632c89b2eb16",
              "extraction_id": "a9accd40-eb89-5595-bf27-b6b82b49f4d4",
              "associatedQuery": "What recent breakthroughs have been made in understanding the genetic causes of diabetes?"
            }
          },
          {
            "id": "f3c6864c-7c06-5a61-bdda-d5730821c237",
            "score": 0.6792303706132924,
            "metadata": {
              "text": "studies provide new insights into type 2diabetes aetiology. Nat Rev Genet 2007;8:657662\n11. Grant RW, Moore AF, Florez JC. Genetic\narchitecture of type 2 diabetes: recentprogress and clinical implications. Diabe-tes Care 2009;32:11071114\n12. Dupuis J, Langenberg C, Prokopenko I,",
              "title": "2010 - Interactions of Dietary Whole-Grain Intake.pdf",
              "version": "v0",
              "chunk_order": 106,
              "document_id": "e4d4a19e-18a0-5a08-9ab7-537f31b7cdc1",
              "extraction_id": "40190f1d-aad5-5d71-b5ba-78331d5e3abb",
              "associatedQuery": "What recent breakthroughs have been made in understanding the genetic causes of diabetes?"
            }
          },
          {
            "id": "81e7ee8d-adb5-5fd7-a3b1-1f6bfb059974",
            "score": 0.6752230122552518,
            "metadata": {
              "text": "early results have been excellent, yielding six \nnew replicating gene regions.\nHere I discuss the insights into type\n \n2 \ndiabetes genetics that have been provided by \nthese new findings. I consider where diabe -\ntes genetic studies might go from here, and \npresent a perspective that may be applicable \nto other common traits. I also briefly discuss \nthe wider implications that surround the \nidentification of a common gene that predis -\nposes to type",
              "title": "2007 - Genome\u2013wide association studies provide new insights into type 2 diabetes aetiology..pdf",
              "version": "v0",
              "chunk_order": 2,
              "document_id": "2ad9b6c6-56ed-5ba6-ad88-c1a6777f5196",
              "extraction_id": "cd034e2b-72bd-5cda-a456-48cf17ead1bf",
              "associatedQuery": "What recent breakthroughs have been made in understanding the genetic causes of diabetes?"
            }
          },
          {
            "id": "b092c8b9-edb1-55fb-ae16-c67e3298946e",
            "score": 0.6748439863417168,
            "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 recent breakthroughs have been made in understanding the genetic causes of diabetes?"
            }
          },
          {
            "id": "23321ca3-f73d-5542-a6c0-1133c3d3e9e5",
            "score": 0.673834641347252,
            "metadata": {
              "text": "24. Varshney, A. et al. Genetic regulatory signatures underlying islet gene expression and type 2 diabetes. Proc. Natl. Acad. Sci. USA 114,  \n23012306 (2017).\n 25. Thurner, M. et al. Integration of human pancreatic islet genomic data refines regulatory mechanisms at Type 2 diabetes susceptibility loci. eLife  7,  \ne31977 (2018).\n 26. Gaulton, K. J. et al. Genetic fine mapping and genomic annotation defines causal mechanisms at type 2 diabetes susceptibility loci. Nat. Genet.  47, \n14151425 (2015).",
              "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": 95,
              "document_id": "ab2868dd-62f6-5350-994c-fcea4328e8a3",
              "extraction_id": "9190d1c1-41a4-5af3-a570-7fea6a15e71a",
              "associatedQuery": "What recent breakthroughs have been made in understanding the genetic causes of diabetes?"
            }
          }
        ],
        "kg_search_results": null
      }
    }
  },
  "16": {
    "results": {
      "completion": {
        "id": "chatcmpl-AIFr5iGpAfX0NjS78CDO8cOuBpwUj",
        "choices": [
          {
            "finish_reason": "stop",
            "index": 0,
            "message": {
              "content": "Genes influence how our bodies respond to sugar and insulin in several ways:\n\n1. **Insulin Secretion and Resistance**: Genes are directly related to insulin secretion and indirectly influence insulin resistance by interacting with other genes. This interaction supports the idea that environmentally triggered insulin resistance can interact with genetically programmed beta-cell dysfunction, leading to diabetes [1].\n\n2. **Beta-Cell Function**: Many genetic variants primarily affect beta-cell function, development, or survival, rather than insulin sensitivity. This suggests that genetic factors play a significant role in how beta cells respond to sugar and insulin [2].\n\n3. **Expression in Metabolically Relevant Tissues**: Genes affecting insulin sensitivity are often expressed in peripheral insulin-sensitive tissues, such as the liver and adipose tissue, and their expression can be responsive to metabolic status. For instance, a high-fat diet can decrease the expression of several of these genes, indicating a genetic influence on how the body responds to dietary changes [3].\n\n4. **Regulation by Metabolic Conditions**: The expression of certain genes can be altered by conditions such as fasting and feeding, which are known to affect peripheral insulin sensitivity. This suggests that genetic regulation can influence how the body responds to changes in sugar and insulin levels [4].\n\n5. **Genetic Variation and Insulin Response**: Specific genetic variations, such as those in the GIPR gene, can influence glucose and insulin responses to an oral glucose challenge, highlighting the role of genetic differences in individual responses to sugar intake [9].\n\nOverall, genetic factors can influence both the secretion of insulin and the body's sensitivity to it, affecting how we metabolize sugar and respond to dietary changes.",
              "role": "assistant"
            }
          }
        ],
        "created": 1728914215,
        "model": "gpt-4o-2024-08-06",
        "object": "chat.completion",
        "system_fingerprint": "fp_a20a4ee344",
        "usage": {
          "completion_tokens": 334,
          "prompt_tokens": 1215,
          "total_tokens": 1549
        }
      },
      "search_results": {
        "vector_search_results": [
          {
            "id": "3bbf736e-7d8b-5e67-a4bf-e1ae28738bf3",
            "score": 0.6845700112754098,
            "metadata": {
              "text": "genes relate directly to insulin secretion and indirectly, through collaborating with other genes, to insulin resistance. Thisseems to support the epidemiological evidence that environmentally triggered insulin resistance interacts with geneticallyprogrammed bcell dysfunction to precipitate diabetes.\nCitation: Jain P, Vig S, Datta M, Jindel D, Mathur AK, et al. (2013) Systems Biology Approach Reveals Genome to Phenome Correlation in Type 2 Diabetes. PLoS\nONE 8(1): e53522. doi:10.1371/journal.pone.0053522",
              "title": "2013 - Systems Biology Approach Reveals Genome to Phenome Correlation in Type 2 Diabetes.pdf",
              "version": "v0",
              "chunk_order": 5,
              "document_id": "ea7c2799-c259-5d0e-b40b-ecebe0a9fc9f",
              "extraction_id": "9369222f-e125-58c0-8f2b-cf5daa867f77",
              "associatedQuery": "How do genes influence how our bodies respond to sugar and insulin?"
            }
          },
          {
            "id": "ccf2d9af-4dca-5021-9c9d-301f817f80e4",
            "score": 0.6621851526388447,
            "metadata": {
              "text": "have been the subject of most follow-up studies to date.Specifically, we examined acute changes in expression of\nthese genes in response to feeding and fasting and\nlonger term changes in the expression of these genes inresponse to a diet high in fat and sugar, recognized as a\ncritical environmental risk factor for type 2 diabetes.\nIt has been hypothesized that most of the new genetic\nvariants affect -cell function, development or survival\nbut not insulin sensitivity [6]. Consistent with this,",
              "title": "2013 - Diabetes genes identified by genome-wide association studies are regulated in mice by nutritional factors in metabolically relevant tissues and by glucose concentrations in islets.pdf",
              "version": "v0",
              "chunk_order": 47,
              "document_id": "98564dd2-424b-557a-a539-022508283567",
              "extraction_id": "c9f74729-056d-556f-8aa8-e0f7a7bd6b66",
              "associatedQuery": "How do genes influence how our bodies respond to sugar and insulin?"
            }
          },
          {
            "id": "d580609b-d24b-5718-ab63-0e6088c8bfeb",
            "score": 0.6557703428542205,
            "metadata": {
              "text": "or survival. However, we also found evidence that most\nof the genes could have potential roles in other\nmetabolically-relevant tissues. Genes affecting insulinsensitivity may be expected to be expressed in peripheralinsulin sensitive tissues, such as liver and adipose tissue,\nand be responsive to metabolic status. Consumption of a\nhigh fat diet was associated with a tendency for the ex-\npression of several of these genes to be decreased. Simi-larly, many of the genes were regulated by feeding and",
              "title": "2013 - Diabetes genes identified by genome-wide association studies are regulated in mice by nutritional factors in metabolically relevant tissues and by glucose concentrations in islets.pdf",
              "version": "v0",
              "chunk_order": 49,
              "document_id": "98564dd2-424b-557a-a539-022508283567",
              "extraction_id": "c9f74729-056d-556f-8aa8-e0f7a7bd6b66",
              "associatedQuery": "How do genes influence how our bodies respond to sugar and insulin?"
            }
          },
          {
            "id": "3f90af62-9a1d-5ac2-b5ee-a616857b34df",
            "score": 0.6532995311439328,
            "metadata": {
              "text": "secretion versus insulin sensitivity). We also sought todetermine whether any of these genes are regulated by\nconditions known to alter the expression of metabolic-\nally relevant genes. We examined the expression of thesegenes under fasting and non-fasting conditions (e.g. in\nresponse to insulin), which might be altered if they affect\nperipheral insulin sensitivity. Consumption of diets high\nin fats and sugars is associated with risk of developing\ntype 2 diabetes [34] and many genes that are critical for",
              "title": "2013 - Diabetes genes identified by genome-wide association studies are regulated in mice by nutritional factors in metabolically relevant tissues and by glucose concentrations in islets.pdf",
              "version": "v0",
              "chunk_order": 15,
              "document_id": "98564dd2-424b-557a-a539-022508283567",
              "extraction_id": "a9ec4c4f-b038-52d2-90db-7bee1ef1f78c",
              "associatedQuery": "How do genes influence how our bodies respond to sugar and insulin?"
            }
          },
          {
            "id": "c171a147-2cf6-5340-82d4-caa63cdafbbd",
            "score": 0.6383192374325708,
            "metadata": {
              "text": "regulating sugar metabolism. Moreover, genes that were",
              "title": "2015 - Gestational Diabetes Alters Offspring DNA.pdf",
              "version": "v0",
              "chunk_order": 129,
              "document_id": "59b51d61-b2c4-540b-a2fb-4c56badb26c1",
              "extraction_id": "af8de1bb-e71e-514f-a5eb-59f37498028e",
              "associatedQuery": "How do genes influence how our bodies respond to sugar and insulin?"
            }
          },
          {
            "id": "81eb21fb-488a-5b08-b883-cd8780110c66",
            "score": 0.6332761267855103,
            "metadata": {
              "text": "Figure 2: The role of type 2 diabetes genes in insulin secretion\nPancreatic -cell genes associated with type 2 diabetes are in italics. G6P=glucose-6-phosphate. Adapted from Florez JC. Newly identi  ed loci highlight beta cell dysfunction as a key cause of type 2 diabetes: where are the insulin resistance genes? Diabetologia 2008; 51: 110010, by kind permission of the author and Springer Science + Business Media. \nPositive calorie balance\nCycle A++\nCycle B Liver fat\n Insulin suppression of",
              "title": "2010 - Diabetes in Asia.pdf",
              "version": "v0",
              "chunk_order": 80,
              "document_id": "0be842b8-7f69-503b-baed-c336e5c834d6",
              "extraction_id": "510b7c7b-ccbb-5d0d-b654-e1dbcf859cb7",
              "associatedQuery": "How do genes influence how our bodies respond to sugar and insulin?"
            }
          },
          {
            "id": "9b60d258-714a-5e70-b2fa-b0a29fc0d672",
            "score": 0.6329711298363738,
            "metadata": {
              "text": "tive Glis3  expression, which in turn drive increased levels of beta cell \napoptosis and senescence. Genetic susceptibility could be replicated \nby elevated levels of dietary fat. Transcriptional analysis of human \nislets identified the same genetic networks at play. Together, these \nfindings demonstrate both the important role of genetic variation in \nbeta cells for diabetes susceptibility and a mechanism by which the \nWestern diet may contribute to the growing diabetes epidemic.\nRESULTS",
              "title": "2016 - Genetic predisposition for beta cell fragility underlies type 1 and type 2 diabetes.pdf",
              "version": "v0",
              "chunk_order": 15,
              "document_id": "5b239c51-7b4c-58e0-acca-2061593fe317",
              "extraction_id": "af25ae45-0c5f-5492-86d0-734eb0fbac12",
              "associatedQuery": "How do genes influence how our bodies respond to sugar and insulin?"
            }
          },
          {
            "id": "dd3348a8-1f07-5e6d-8ba0-3c6c263c0799",
            "score": 0.6316082101101761,
            "metadata": {
              "text": "associated with fasting proinsulin levels and provides new insights into the \npathophysiology of type 2 diabetes. Diabetes 60, 26242634 (2011).\n65. Saxena, R. etal. Genetic variation in GIPR influences the glucose and insulin responses to an oral glucose challenge. Nat. Genet. 42, 142148 (2010).\n66. Tobacco and Genetics Consortium. Genome-wide meta-analyses identify multiple loci associated with smoking behavior. Nat. Genet. 42, 441447 (2010).",
              "title": "2021 - Interpreting type 1 diabetes risk.pdf",
              "version": "v0",
              "chunk_order": 176,
              "document_id": "9f13ec69-195b-55eb-a549-b3eb3dc0f321",
              "extraction_id": "0bd0fae3-9297-5da2-b3ae-21190af84094",
              "associatedQuery": "How do genes influence how our bodies respond to sugar and insulin?"
            }
          },
          {
            "id": "9080e28b-1c0d-5bfa-8698-7ae677aa64ed",
            "score": 0.6283100468927605,
            "metadata": {
              "text": "38. Saxena R, Hivert M, Langenberg C, Tanaka T, Pankow JS, et al. (2010) Genetic\nvariation in GIPR influences the glucose and insulin responses to an oral glucose\nchallenge. Nat Genet 42: 142148. doi:10.1038/ng.521.\n39. Neale BM, Sham PC (2004) The future of association studies: gene-based\nanalysis and replication. Am J Hum Genet 75: 353362. doi:10.1086/423901.\n40. Saccone SF, Hinrichs AL, Saccone NL, Chase GA, Konvicka K, et al. (2007)",
              "title": "2010 - Common Inherited Variation in Mitochondrial Genes.pdf",
              "version": "v0",
              "chunk_order": 288,
              "document_id": "9a5c8cba-06cb-5280-871f-1bbe128c3dc4",
              "extraction_id": "930831e1-56c3-5ef6-b847-2f25f2567032",
              "associatedQuery": "How do genes influence how our bodies respond to sugar and insulin?"
            }
          },
          {
            "id": "c2f1a416-7f04-55b0-b19b-8a8aa858b801",
            "score": 0.6278498394578802,
            "metadata": {
              "text": "Nature Reviews | EndocrinologyFactors that aect insulin secretion and action\n Body weight\n Level of physical activity Smoking Heavy alcohol consumption Genetic predisposition Geneenvironment interaction\nPositive risk prole Negative risk prole\nNormoglycaemia/uni03B2-cell dysfunction and insulin resistanceAdipose tissue Skeletal muscle LiverInsulin-mediated \nglucose production /uni2191Insulin-mediated glucose uptake /uni2193 Insulin-mediated glucose uptake /uni2193\nHyperglycaemia\n Epigenetics",
              "title": "2018 - Global aetiology and epidemiology of type 2 diabetes mellitus and its complications.pdf",
              "version": "v0",
              "chunk_order": 46,
              "document_id": "8bc8f3d4-968f-5252-ab4c-832b92e9ec0d",
              "extraction_id": "8248ba2b-335c-53ce-afbe-f31b68507443",
              "associatedQuery": "How do genes influence how our bodies respond to sugar and insulin?"
            }
          }
        ],
        "kg_search_results": null
      }
    }
  },
  "17": {
    "results": {
      "completion": {
        "id": "chatcmpl-AIFrBAew5HsqHnMUVkuc9dpSmo0io",
        "choices": [
          {
            "finish_reason": "stop",
            "index": 0,
            "message": {
              "content": "Yes, there are specific genetic markers that can indicate a higher risk for Type 1 versus Type 2 diabetes.\n\nFor Type 1 diabetes, genome-wide association studies have identified over 50 loci associated with an increased genetic risk. Several candidate genes within these regions have been suggested or identified, although the exact molecular mechanisms by which they contribute to islet cell inflammation and beta cell destruction are not fully understood [1].\n\nFor Type 2 diabetes, specific genetic markers have also been identified. Some of the loci associated with an increased risk include TCF7L2, PPARG, FTO, KCNJ11, NOTCH2, WFS1, CDKAL1, IGF2BP2, SLC30A8, JAZF1, and HHEX [9]. Additionally, markers such as TCF7L2 and CAPN10 have been strongly associated with the risk of developing Type 2 diabetes [8].\n\nThese findings indicate that while both types of diabetes have genetic components, the specific markers and loci associated with each type differ, reflecting their distinct pathophysiological mechanisms.",
              "role": "assistant"
            }
          }
        ],
        "created": 1728914221,
        "model": "gpt-4o-2024-08-06",
        "object": "chat.completion",
        "system_fingerprint": "fp_a20a4ee344",
        "usage": {
          "completion_tokens": 223,
          "prompt_tokens": 1479,
          "total_tokens": 1702
        }
      },
      "search_results": {
        "vector_search_results": [
          {
            "id": "263dc0cb-dfa0-5ee2-b927-f9a196294d46",
            "score": 0.7546218183862092,
            "metadata": {
              "text": "Genes signifying increased risk for both type 1 and type 2 dia-betes have been identified. Genomewide association studies have identified over 50 loci associated with an increased genetic risk of type 1 diabetes. Several T1D candidate genes for increased risk of developing type 1 diabetes have been sug-gested or identified within these regions, but the molecular basis by which they contribute to islet cell inflammation and beta cell destruction is not fully understood.\n12 Also, several",
              "title": "2015 -precision-medicine-for-managing-diabetes.pdf",
              "version": "v0",
              "chunk_order": 27,
              "document_id": "80949bab-d085-5f61-b98a-4bee043bc4e2",
              "extraction_id": "f53ccf4e-f47f-5b44-8b41-f7068efc8be3",
              "associatedQuery": "Are there specific genetic markers that can indicate a higher risk for Type 1 versus Type 2 diabetes?"
            }
          },
          {
            "id": "988cae28-e149-5190-8ff0-6ecce8d001bc",
            "score": 0.7422494445890533,
            "metadata": {
              "text": "Genetics of Type 2 Diabetes Chapter 12\n197400 multiallelic markers (short tandem repeats or microsatellites, \nwith a density of   1 marker/10   cmol) allows identi  cation of \npolymorphic markers showing strong allele identity by descent in diabetic family members (i.e. allele sharing in sibships is signi  -\ncantly higher than 50%). Once identi  ed, such susceptibility \ngenes for diabetes may then be positionally cloned in the intervals of linkage.",
              "title": "2010 - The Genetics of Type 2 Diabetes.pdf",
              "version": "v0",
              "chunk_order": 95,
              "document_id": "a1d211d4-279e-51d7-b2b2-33bc2763d089",
              "extraction_id": "2610c9c1-5e75-528e-98d8-c4a543ea2f89",
              "associatedQuery": "Are there specific genetic markers that can indicate a higher risk for Type 1 versus Type 2 diabetes?"
            }
          },
          {
            "id": "ca9e53b7-6e51-5ae6-9ef4-8f2f5f40acb5",
            "score": 0.7348826904629888,
            "metadata": {
              "text": "3. Katsarou, A. etal. Type 1 diabetes mellitus. Nat. Rev. Dis. Primers 3, 17016 (2017).\n4. Onengut-Gumuscu, S. etal. Fine mapping of type 1 diabetes susceptibility loci and evidence for colocalization of causal variants with lymphoid gene enhancers. Nat. Genet. \n47, 381386 (2015).\n5. Barrett, J. C. etal. Genome-wide association study and meta-analysis find that over 40 \nloci affect risk of type 1 diabetes. Nat. Genet. 41, 703707 (2009).",
              "title": "2021 - Interpreting type 1 diabetes risk.pdf",
              "version": "v0",
              "chunk_order": 60,
              "document_id": "9f13ec69-195b-55eb-a549-b3eb3dc0f321",
              "extraction_id": "254be2dd-1b4f-5cf9-af93-dbf3d5867510",
              "associatedQuery": "Are there specific genetic markers that can indicate a higher risk for Type 1 versus Type 2 diabetes?"
            }
          },
          {
            "id": "61f523a8-f466-5148-afba-6400c44ed278",
            "score": 0.7347316145896964,
            "metadata": {
              "text": "Clinical Risk Factors, DNA Variants, and the Development of Type 2 Diabetes\nn engl j med 359;21 www.nejm.org november 20, 2008 2229(Fig. 3). An increase in the BMI and a concomi -\ntant decrease in insulin sensitivity during the \n8-year period were consistent findings, with no differences between subjects at high and low genetic risk (Fig. 3A and 3B). However, subjects with a high genetic risk did not increase their insulin secretion (disposition index) to compen -",
              "title": "2008 - Clinical Risk Factors, DNA Variants.pdf",
              "version": "v0",
              "chunk_order": 77,
              "document_id": "0018610a-9c86-5e2d-a27d-f66cf4f8519d",
              "extraction_id": "640f3749-a2bf-5b6b-adab-72ce7f029a28",
              "associatedQuery": "Are there specific genetic markers that can indicate a higher risk for Type 1 versus Type 2 diabetes?"
            }
          },
          {
            "id": "151d8a78-8aa8-5024-8e15-54fba4f1857b",
            "score": 0.7289990565479112,
            "metadata": {
              "text": "and genetic markers to improve the prediction of type 2 diabetes: theEPIC-Potsdam Study. Diabetes Care . 2009;32:2116 2119 (in eng).\n56. Cauchi S, Meyre D, Durand E, et al. Post genome-wide association\nstudies of novel genes associated with type 2 diabetes show gene-gene interaction and high predictive value. PLoS One . 2008;3(5):\ne2031 .\n57. Lyssenko V, Jonsson A, Almgren P, et al. Clinical risk factors, DNA\nvariants, and the development of type 2 diabetes. N Engl J Med .\n2008;359:2220 2232 (in eng).",
              "title": "2018 - Quantitative Relationship Between Cumulative Risk Alleles Based.pdf",
              "version": "v0",
              "chunk_order": 186,
              "document_id": "d585896e-1c32-51cb-827d-e4fd3b3943f3",
              "extraction_id": "6db9f25e-36fd-51c0-be36-6dfacd963b1b",
              "associatedQuery": "Are there specific genetic markers that can indicate a higher risk for Type 1 versus Type 2 diabetes?"
            }
          },
          {
            "id": "f692be48-b905-5463-8101-22eaf14e6405",
            "score": 0.7246813506267845,
            "metadata": {
              "text": "etically expressed homeobox variant (rs1111875) on type 2 diabetes risk.  Molecular Genetics \nand Metabolism  ,  102 (2), 194199. \n Watanabe, R. M., Black, M. H., Xiang, A. H., Allayee, H., Lawrence, J. M., & Buchanan, T. A. (2007). \nGenetics of gestational diabetes mellitus and type 2 diabetes.  Diabetes Care  ,  30 (Suppl. 2), \nS134S140. \n Williams, M. A., Qiu, C., Dempsey , J. C., & Luthy , D. A. (2003). Familial aggregation of type 2",
              "title": "2011 - Shared Genomics of Type 2 and Gestational Diabetes Mellitus.pdf",
              "version": "v0",
              "chunk_order": 232,
              "document_id": "bef0cabe-0bca-5715-9ffc-0b825744fbcf",
              "extraction_id": "41fefdf5-447e-556e-b95f-c132bdea7c41",
              "associatedQuery": "Are there specific genetic markers that can indicate a higher risk for Type 1 versus Type 2 diabetes?"
            }
          },
          {
            "id": "48c93a37-d0d5-51de-b2d1-5c6122c01ab1",
            "score": 0.7246804021394162,
            "metadata": {
              "text": "markers, genetic markers do not change with disease progression.Dimas and collaborators examined the association of 37 establishedT2D susceptibility loci and indices of proinsulin processing, insulin\nsecretion, and insulin sensitivity in 58,614 nondiabetic subjects [6].\nCluster analysis classi ed the risk loci into ve major categories on the\nbasis of their association with glycemic phenotypes. The rst cluster\nwas characterized by the effects of the risk alleles of PPARG ,KLF14 ,",
              "title": "2019 - Biomarkers for type 2 diabetes.pdf",
              "version": "v0",
              "chunk_order": 66,
              "document_id": "c8ee94fc-f9bc-5a32-9524-9d1d9cf37159",
              "extraction_id": "bc4717c3-d353-5f44-9513-50634f8d5196",
              "associatedQuery": "Are there specific genetic markers that can indicate a higher risk for Type 1 versus Type 2 diabetes?"
            }
          },
          {
            "id": "82debd98-f2fe-51aa-931c-63e11249de7b",
            "score": 0.7228839296906133,
            "metadata": {
              "text": "recently, meta-analysis of GWAS data involving African \nAmerican type 2 diabetes patients identified similar loci \nto the previous studies with the addition of two novel \nloci, HLA-B and INS-IGF[157]. These results provide \nstrong evidence of common genetic determinants \nincluding common specific genes that are linked to \ndiabetes. A small list of specific genetic markers seem \nstrongly associated with the risk of developing type 2 \ndiabetes including the TCF7L2[158] and CAPN10[159,160]",
              "title": "2015 - Diabetes mellitus The epidemic of the century.pdf",
              "version": "v0",
              "chunk_order": 86,
              "document_id": "e114dd28-fd39-56df-bdeb-8806474a6c10",
              "extraction_id": "7cfe9f29-a0ee-56d3-be3b-1b238a43bc07",
              "associatedQuery": "Are there specific genetic markers that can indicate a higher risk for Type 1 versus Type 2 diabetes?"
            }
          },
          {
            "id": "8469faae-c6c9-5fd4-8437-870eef394dd1",
            "score": 0.7217622896857948,
            "metadata": {
              "text": "Clinical Risk Factors, DNA Variants, and the Development of Type 2 Diabetes\nn engl j med 359;21 www.nejm.org november 20, 2008 2231MPP subjects (P = 0.001) and from 0.79 to 0.83 in \nthe Botnia subjects (P = 0.006).\nOf the 16 loci that have been associated with \ntype 2 diabetes previously,8-15 we showed that 11 \n TCF7L2, PPARG, FTO, KCNJ11, NOTCH2, WFS1, \nCDKAL1, IGF2BP2, SLC30A8, JAZF1, and HHEX   \nwere associated with an enhanced risk of future",
              "title": "2008 - Clinical Risk Factors, DNA Variants.pdf",
              "version": "v0",
              "chunk_order": 95,
              "document_id": "0018610a-9c86-5e2d-a27d-f66cf4f8519d",
              "extraction_id": "0aae948a-50f9-568a-b0dc-5960a2d2ceaa",
              "associatedQuery": "Are there specific genetic markers that can indicate a higher risk for Type 1 versus Type 2 diabetes?"
            }
          },
          {
            "id": "387e1774-0250-5c72-b11c-069bdf3ef9ea",
            "score": 0.7195725179228165,
            "metadata": {
              "text": "Clinical Risk Factors, DNA Variants, and the Development of Type 2 Diabetes\nn engl j med 359;21 www.nejm.org november 20, 2008 2227(Fig. 1B), whereas impaired fasting glucose or \nimpaired glucose tolerance developed in 313 of 2039 subjects (15.4%).\nClinical Factors Predicting Incidence  \nof Diabetes\nIn both the MPP and Botnia studies, a family his -\ntory of diabetes, an increased BMI, and increased levels of blood pressure and serum levels of tri -",
              "title": "2008 - Clinical Risk Factors, DNA Variants.pdf",
              "version": "v0",
              "chunk_order": 57,
              "document_id": "0018610a-9c86-5e2d-a27d-f66cf4f8519d",
              "extraction_id": "38bacfcd-d182-5220-b8bc-18f6c74b14a8",
              "associatedQuery": "Are there specific genetic markers that can indicate a higher risk for Type 1 versus Type 2 diabetes?"
            }
          }
        ],
        "kg_search_results": null
      }
    }
  },
  "18": {
    "results": {
      "completion": {
        "id": "chatcmpl-AIFrIc4qRPbtDcHbcrNOicZwU9hKr",
        "choices": [
          {
            "finish_reason": "stop",
            "index": 0,
            "message": {
              "content": "New gene therapies have the potential to cure or treat diabetes through several innovative approaches:\n\n1. **Gene-Based Therapies**: Advances in understanding the biological mechanisms that maintain glucose homeostasis and the molecular defects leading to chronic hyperglycemia could lead to the development of gene-based therapies. These therapies aim to target specific genetic factors involved in diabetes, potentially offering more precise treatment options [3].\n\n2. **In Vivo Gene Therapy**: This approach involves directly inserting a vector containing the desired gene into the patient. It is considered a promising therapeutic strategy for type 1 diabetes, although challenges remain in developing safe and effective vectors [9].\n\n3. **Inducing Islet Neogenesis**: Gene therapy techniques, such as betacellulin gene therapy, have been shown to induce islet neogenesis in the liver and reverse diabetes in mice. This suggests that gene therapy can stimulate the body to produce insulin-producing cells, offering a potential cure for diabetes [10].\n\nThese strategies highlight the potential of gene therapies to address the underlying genetic causes of diabetes and restore normal insulin production and glucose regulation.",
              "role": "assistant"
            }
          }
        ],
        "created": 1728914228,
        "model": "gpt-4o-2024-08-06",
        "object": "chat.completion",
        "system_fingerprint": "fp_6b68a8204b",
        "usage": {
          "completion_tokens": 217,
          "prompt_tokens": 1323,
          "total_tokens": 1540
        }
      },
      "search_results": {
        "vector_search_results": [
          {
            "id": "59cce584-cd38-52d1-bdaa-d5500175eefb",
            "score": 0.6679459810256958,
            "metadata": {
              "text": "unraveling the pathophysiological mechanisms of this disease, identifying \ncandidate diabetic genes, and discovering and testing new therapeutic agents. \nThe classical rodent models of diabetes allow unbiased discovery, while the \nnew models made by genetic manipulation allow testing of the role of \nspecific genes and tissues. Experimental animal models are an irreplaceable \nresource for diabetes research and are hastening the progress towards the \ngoals of better treatment, prevention, and cure.",
              "title": "2004 - Diabetes Genes a.pdf",
              "version": "v0",
              "chunk_order": 773,
              "document_id": "805ad0d5-6a3d-5e5f-b1ba-121e10c6daaa",
              "extraction_id": "a5ae065c-371f-5459-830b-7a34891ca091",
              "associatedQuery": "How can new gene therapies potentially cure or treat diabetes?"
            }
          },
          {
            "id": "d37e62ab-6261-5f14-8423-3b6e2574422e",
            "score": 0.6633472838012757,
            "metadata": {
              "text": "is absence of reliable methods for generating specific celltypes,immunologicalrejectionofthetransplantedcells,anddifficulty in purification of specific lineages [55]. Furtherconcernsincludetheuncontrolledproliferationofthetrans-planted embryonic stem cells into a specific type, once theyaretransplanted[56].Still,despiteofitsmanifoldlimitationsboth scientific and ethical, the application of stem cell tech-nologyholdsimmenseprospectsintreatmentofdiabetes.\n6. Gene Therapy in Diabetes",
              "title": "2015 - Recent Trends in Therapeutic Approaches for Diabetes Management  A Comprehensive Updat.pdf",
              "version": "v0",
              "chunk_order": 62,
              "document_id": "ec4921c2-af14-56cc-aed3-65f8ea236bde",
              "extraction_id": "6b2ac076-ee4b-53b3-b49b-1d15f46e6a98",
              "associatedQuery": "How can new gene therapies potentially cure or treat diabetes?"
            }
          },
          {
            "id": "f4e8a3c8-0b85-5595-8917-933aced8b3ba",
            "score": 0.6623994309425187,
            "metadata": {
              "text": "T ogether, these discoveries will continue to improve our \nunderstanding of the biologic mechanisms that maintain glucose homeostasis, and of still hidden  molecular defects leading to \nchronic hyperglycemia, and could also lead to the development of more speci  cally targeted antidiabetic drugs or even gene -\n based therapies. Moreover, pharmacogenetic testing might then be used to predict, for each patient, the therapeutic response to different classes of drugs. The identi  cation of T2DM genes will",
              "title": "2010 - The Genetics of Type 2 Diabetes.pdf",
              "version": "v0",
              "chunk_order": 258,
              "document_id": "a1d211d4-279e-51d7-b2b2-33bc2763d089",
              "extraction_id": "c4de4c07-4749-5401-bbf3-16988c132852",
              "associatedQuery": "How can new gene therapies potentially cure or treat diabetes?"
            }
          },
          {
            "id": "cb7178a0-7015-555c-801a-cd2d258cf3dc",
            "score": 0.6559618711471558,
            "metadata": {
              "text": "Greatstrideshavebeenmadeclinicallyintheprevention,\ndevelopment,andtreatmentofthediseasebutnotherapeuticmethod have been completely successful till date. With newtechnologies revolutionizing the treatment possibilities, thesearch for an effective medication is not far ahead. Theextensive research leading to the discovery of the pathwaygenes contributing to the development of the disease andthe sequencing of complete genomes have revolutionized\nthe diabetes research. The development of the techniques",
              "title": "2015 - Recent Trends in Therapeutic Approaches for Diabetes Management  A Comprehensive Updat.pdf",
              "version": "v0",
              "chunk_order": 96,
              "document_id": "ec4921c2-af14-56cc-aed3-65f8ea236bde",
              "extraction_id": "48643e77-c5b4-5042-8f08-82c986d9f5b2",
              "associatedQuery": "How can new gene therapies potentially cure or treat diabetes?"
            }
          },
          {
            "id": "d5963c8e-686f-52f5-a6de-b978d5c40e20",
            "score": 0.6545608455466263,
            "metadata": {
              "text": "into different genetic levels of disease categories, from which pre-\nvention or treatment methods could be provided accordingly [ 4].\nFor example, some forms of diabetes are directly related to a change\nin a single gene [ 34]. Some patients who are diagnosed with type 1\ndiabetes can now be tested for one of monogenic diabetes. The\nappropriate treatment for these patients is not injecting insulin, but\ngiving oral sulfonylureas [ 34]. Moreover, it is now well understood",
              "title": "2018 - A theoretical framework for interaction of nursing discipline.pdf",
              "version": "v0",
              "chunk_order": 35,
              "document_id": "4c90f95f-3365-522e-9eb4-9ea002beddb2",
              "extraction_id": "abf78c3a-ad53-5c86-979d-2d9d176a51a4",
              "associatedQuery": "How can new gene therapies potentially cure or treat diabetes?"
            }
          },
          {
            "id": "0b4a495d-fdee-515a-a524-d9415b17f97e",
            "score": 0.6466418504714966,
            "metadata": {
              "text": "pp .430435,2003.\n[58] M. Zalzman, S. Gupta, R. K. Giri et al., Reversal of\nhyperglycemia in mice by using human expandable insulin-\nproducing cells differentiated from fetal liver progenitor cells,Proceedings of the National Academy of Sciences of the United\nStatesofAmerica ,vol.100,no .12,pp .72537258,2003.\n[59] H.-S. Jun and J.-W. Yoon, Approaches for the cure of type 1\ndiabetes by cellular and gene therapy, Current Gene Therapy ,\nvol.5,no.2,pp.249262,2005.",
              "title": "2015 - Recent Trends in Therapeutic Approaches for Diabetes Management  A Comprehensive Updat.pdf",
              "version": "v0",
              "chunk_order": 124,
              "document_id": "ec4921c2-af14-56cc-aed3-65f8ea236bde",
              "extraction_id": "168e94e9-e8c2-547c-878a-1e5306564193",
              "associatedQuery": "How can new gene therapies potentially cure or treat diabetes?"
            }
          },
          {
            "id": "13b73999-262c-50e1-b668-2d5f7ca02067",
            "score": 0.6438010334968567,
            "metadata": {
              "text": "transgenics. It is likely that animal models will play an importantrole in the eventual cure of human diabetes mellitus. \nCompeting interests \nNone declared. \nReferences \n1Sima AAF, Shafrir E, eds.  \nAnimal Models of Diabetes: A Primer. \nAmsterdam: Harwood Academic Publishers, 2000.\n2British Union for the Abolition of Vivisection. Home page. Available\nfrom: http://www.buav.org.\n3Patterson C.  \nEternal Treblinka. Our Treatment of Animals and the\nHolocaust \n. New York: Lantern Books, 2002.\n4Regan T.",
              "title": "2005  - Animal models of diabetes mellitus.pdf",
              "version": "v0",
              "chunk_order": 126,
              "document_id": "2fd381ac-2898-5a8c-af93-bcc86e7dec14",
              "extraction_id": "3dca156c-64c4-577f-b0a6-069de0f31234",
              "associatedQuery": "How can new gene therapies potentially cure or treat diabetes?"
            }
          },
          {
            "id": "1299cc23-f6b0-5801-bead-b46ac90bc3a8",
            "score": 0.6380507946014404,
            "metadata": {
              "text": "Third, this view of diabetes pathogenesis is consistent with\nthe growing portfolio of available therapies. We have agents\nand interventions that can prevent or ameliorate diabetesthrough, for example, beneficial effects on islet function\n(e.g. sulfonylureas), obesity (weight loss), insulin resistance\n(e.g. exercise), fuel partitioning (e.g. thiazolidinediones) andmicrobiome content (metformin, possibly). Just as diabetes\nrisk alleles influence metabolic phenotype through pushing",
              "title": "2017 - Painting a new picture of personalised medicine for diabetes.pdf",
              "version": "v0",
              "chunk_order": 41,
              "document_id": "e226b2b1-0bc4-5d79-b931-ad47f21be045",
              "extraction_id": "1cd3076d-af86-55d7-903c-9065bc640af0",
              "associatedQuery": "How can new gene therapies potentially cure or treat diabetes?"
            }
          },
          {
            "id": "3201da93-5a34-5164-8bf4-c98d32019019",
            "score": 0.6354519724845886,
            "metadata": {
              "text": "aprospectivetherapeuticapproachfortype1diabetes[59].\nThein vivogene therapy is the method of choice as\na therapeutic strategy because it is simpler and the vectorcontaining the desired gene is directly inserted into thepatient, but the development of safe (not toxic to host)and effective vectors remains as a challenging task for genetherapist. Presently, the strategies for in vivotherapy involve",
              "title": "2015 - Recent Trends in Therapeutic Approaches for Diabetes Management  A Comprehensive Updat.pdf",
              "version": "v0",
              "chunk_order": 67,
              "document_id": "ec4921c2-af14-56cc-aed3-65f8ea236bde",
              "extraction_id": "6b2ac076-ee4b-53b3-b49b-1d15f46e6a98",
              "associatedQuery": "How can new gene therapies potentially cure or treat diabetes?"
            }
          },
          {
            "id": "0418b345-7005-5d7d-a79f-570fb61bd14b",
            "score": 0.6338961938681498,
            "metadata": {
              "text": "betacellulin gene therapy induces islet neogenesis in the liver\na n dr e v e r s e sd i a b e t e si nm i c e ,  Nature Medicine ,v o l .9 ,n o .5 ,\npp.596603,2003.\n[73] S. Ferber, A. Halkin, H. Cohen et al., Pancreatic and duode-\nnal homeobox gene 1 induces expression of insulin genes inliver and ameliorates streptozotocin-induced hyperglycemia,\nNature Medicine ,vol.6,no .5,pp .568572,2000.\n[74] P.A.Halban,S.E.Kahn, A.Lernmark,andC.J.Rhodes,Gene\nandcell-replacementtherapyinthetreatmentoftype1diabetes.",
              "title": "2015 - Recent Trends in Therapeutic Approaches for Diabetes Management  A Comprehensive Updat.pdf",
              "version": "v0",
              "chunk_order": 130,
              "document_id": "ec4921c2-af14-56cc-aed3-65f8ea236bde",
              "extraction_id": "168e94e9-e8c2-547c-878a-1e5306564193",
              "associatedQuery": "How can new gene therapies potentially cure or treat diabetes?"
            }
          }
        ],
        "kg_search_results": null
      }
    }
  },
  "19": {
    "results": {
      "completion": {
        "id": "chatcmpl-AIFrQL8Phj0Q41oiXeOOZsabJcULg",
        "choices": [
          {
            "finish_reason": "stop",
            "index": 0,
            "message": {
              "content": "Monogenic and polygenic diabetes are distinct forms of diabetes with different genetic underpinnings:\n\n1. **Monogenic Diabetes**: This form of diabetes results from a mutation in a single gene that is highly penetrant, meaning it has a strong effect on the individual who carries it [1], [6]. Monogenic diabetes is often associated with defects in beta-cell function, leading to a decrease in the number or function of these cells [2]. It is typically characterized by early onset, often before the age of 25, and can include conditions like Maturity-Onset Diabetes of the Young (MODY) [5]. Monogenic diabetes is relatively rare, representing about 2%-5% of diabetes cases [2].\n\n2. **Polygenic Diabetes**: In contrast, polygenic diabetes results from the combined effect of multiple genetic variants, each contributing a small effect, along with environmental and lifestyle factors [1], [6]. This form of diabetes is more common and is the predominant mode of inheritance in type 2 diabetes [7]. The genetic variants involved in polygenic diabetes do not have as strong an effect individually as those in monogenic diabetes, but together they contribute to the disease risk in the presence of other factors like obesity and sedentary lifestyle [3].\n\nIn summary, monogenic diabetes is caused by a single gene mutation with a strong effect, while polygenic diabetes involves multiple genes with smaller effects combined with environmental influences.",
              "role": "assistant"
            }
          }
        ],
        "created": 1728914236,
        "model": "gpt-4o-2024-08-06",
        "object": "chat.completion",
        "system_fingerprint": "fp_6b68a8204b",
        "usage": {
          "completion_tokens": 289,
          "prompt_tokens": 1172,
          "total_tokens": 1461
        }
      },
      "search_results": {
        "vector_search_results": [
          {
            "id": "c38627c6-0dd1-5dec-aeb6-ea1edb924480",
            "score": 0.7862402200698853,
            "metadata": {
              "text": "to improve diagnosis. Monogenic vs. polygenic diabetes  \nMonogenic and polygenic diabetes are traditionally considered distinct, with \nmonogenic diabetes resulting from one highly penetrant variant in one gene in a given \nindividual, and polygenic diabetes resulting from the contribution of several variants with \nsmaller effects in the context of environmental/lifestyle factors.  In T1D, autoimmune \ndysfunction is the prominent mechanism, with variation in the major histocompatibility",
              "title": "2021 - Genomic Medicine in Diabetes Improving the Diagnostic Rate of Monogenic Diabetes.pdf",
              "version": "v0",
              "chunk_order": 79,
              "document_id": "e315a891-ba59-57e9-856b-602544375324",
              "extraction_id": "29df597d-e40d-5bc8-8ee0-89141d8e7fc0",
              "associatedQuery": "What is the difference between monogenic and polygenic diabetes?"
            }
          },
          {
            "id": "d704148a-88c0-58fe-810a-89b767a1f53b",
            "score": 0.7451968789100647,
            "metadata": {
              "text": "represent about 2%-5% of diabetes patients. Mono -\ngenic diabetes results primarily from gene defects that \nlead to a decrease in beta cell number or function. \nMonogenic diabetes genes were identified using linkage \nstudies or code for proteins that directly affected \nglucose homeostasis. The majority of genes responsible \nfor monogenetic diabetes code for either transcription \nfactors that participate in the control of nuclear gene \nexpression or proteins that are located on the cell",
              "title": "2015 - Diabetes mellitus The epidemic of the century.pdf",
              "version": "v0",
              "chunk_order": 102,
              "document_id": "e114dd28-fd39-56df-bdeb-8806474a6c10",
              "extraction_id": "e119acfb-4ad6-515e-a1bb-7796d283befc",
              "associatedQuery": "What is the difference between monogenic and polygenic diabetes?"
            }
          },
          {
            "id": "7bbf950d-cbf8-5221-8ea6-b3571fab4fad",
            "score": 0.7153393030166626,
            "metadata": {
              "text": "diabetic patients inwhom rare, highly\npenetrant mutations ofasingle gene\ncause their diabetes (13). While com -\nmon variants ofthese genes that make a\nsmall contribution topolygenic diabetes\nmay also exist (13), thevariants causing\nmonogenic diabetes have limited util-\nityinpharmacogenetics duetotheir low\nallele frequency. Thevast majority oftype\n2diabetes patients have polygenetic forms\nofthedisease that typically also require a\npermissive environment (e.g., obesity, sed-",
              "title": "2007 - Pharmacogenetics of metformin response a step in the path toward personalized medicine.pdf",
              "version": "v0",
              "chunk_order": 31,
              "document_id": "12344230-0ed1-516f-bf2d-9c6e71ac76b5",
              "extraction_id": "c66bcb9f-15af-5843-9e9c-168e8cf230d0",
              "associatedQuery": "What is the difference between monogenic and polygenic diabetes?"
            }
          },
          {
            "id": "35efeaf0-c6b6-509e-9426-d23c8727164f",
            "score": 0.7151061296463013,
            "metadata": {
              "text": "diabetes exist along more of a continuum than previously appre -\nciated. Therefore, knowledge about monogenic diabetes not only \nprovides opportunities for etiology-based treatment of the minori-\nty of individuals with highly penetrant variants, but also informs \nbroader understanding of diabetes etiology.\nTypes of monogenic diabetes\nMaturity-onset diabetes of the young\nMODY comprises most monogenic diabetes cases, with classical \ncharacteristics of young diagnosis age, family history of diabe -",
              "title": "2021 -  Monogenic diabetes a gateway to precision medicine.pdf",
              "version": "v0",
              "chunk_order": 20,
              "document_id": "0b6ff786-6a7b-5d24-ba5e-7a61fee7757f",
              "extraction_id": "38df3fac-cb86-5e74-b270-1e1e9e12dcdb",
              "associatedQuery": "What is the difference between monogenic and polygenic diabetes?"
            }
          },
          {
            "id": "55e16624-4a02-5fba-bbe7-a07db8559401",
            "score": 0.7041106643742732,
            "metadata": {
              "text": "Monogenic Diabetes \n Monogenic diabetes is a class of diabetes associated with genetic defects in beta - cell function. They are frequently associated with early onset of hyperglycemia (typically before 25 years of age). Three common forms of mono-genic diabetes include maturity - onset diabetes of the",
              "title": "2012 - Systems Biology Approaches to Nutrition.pdf",
              "version": "v0",
              "chunk_order": 9661,
              "document_id": "6955478b-950d-5d29-b24c-3a5ca656f3ae",
              "extraction_id": "8d7fefe4-325f-5c64-9fee-0587c545d5ab",
              "associatedQuery": "What is the difference between monogenic and polygenic diabetes?"
            }
          },
          {
            "id": "854afd34-91ed-5817-b24e-1fd5894261f3",
            "score": 0.7016476810960592,
            "metadata": {
              "text": "HNF4A-MODY  and requires genetic testing to diagnose. Here \nwe will describe monogenic diabetes types, etiologies, diagnosis, \nmanagement, and strategies to improve diagnosis.\nMonogenic versus polygenic diabetes\nMonogenic and polygenic diabetes are traditionally considered \ndistinct, with monogenic diabetes resulting from one highly pene -\ntrant variant in one gene in a given individual and polygenic diabe -\ntes resulting from the contribution of several variants with smaller",
              "title": "2021 -  Monogenic diabetes a gateway to precision medicine.pdf",
              "version": "v0",
              "chunk_order": 6,
              "document_id": "0b6ff786-6a7b-5d24-ba5e-7a61fee7757f",
              "extraction_id": "5a39ee4b-ba00-56d6-ba6c-0edeac3b4f2e",
              "associatedQuery": "What is the difference between monogenic and polygenic diabetes?"
            }
          },
          {
            "id": "a74728c6-2903-5035-afd9-0f6a0f0c295e",
            "score": 0.7016175389289856,
            "metadata": {
              "text": "Monogenic inheritance is caused by mutation of a single gene. There are \nsome well-defined monogenic rodent models. In humans, monogenic obesity \nand diabetes exist as well, but are extremely rare. \nPolygenic inheritance is the result of multiple contributing genes and \nis the predominant mode of inheritance in human type 2 diabetes. Multiple \npolygenic animal models are also available. However, even in monogenic \nanimal models, genetic background plays an important influence. For",
              "title": "2004 - Diabetes Genes a.pdf",
              "version": "v0",
              "chunk_order": 741,
              "document_id": "805ad0d5-6a3d-5e5f-b1ba-121e10c6daaa",
              "extraction_id": "c6bf083c-f045-55e2-9eae-ff96a4ceea4c",
              "associatedQuery": "What is the difference between monogenic and polygenic diabetes?"
            }
          },
          {
            "id": "5b06a49e-7ef9-558a-b402-11866c555cd5",
            "score": 0.6908171772956848,
            "metadata": {
              "text": "(Mendelian) that may also cause type 2 diabetes (Yang & Chan, 2016). More than twenty genes highly expressed in pancreatic cells have been identified within these mono-genic subtypes (AlkortaAranburu et al., 2014). Recently, two national surveys revealed that most patients with mono-genic diabetes are likely to be unrecognized and misdiag-nosed as type 1 or type 2 diabetes (Delvecchio et al., 2017; Johansson et al., 2017). Genetic diagnosis leads to improved treatment, better prediction of disease",
              "title": "2019 - HDAC4 mutations cause diabetes and induce  \u2010cell FoxO1 nuclear exclusion.pdf",
              "version": "v0",
              "chunk_order": 10,
              "document_id": "7e4028b2-d5c1-5ddc-a06d-fd4f767d0f39",
              "extraction_id": "7f53ea65-79ed-5207-9397-68b6d14bc19c",
              "associatedQuery": "What is the difference between monogenic and polygenic diabetes?"
            }
          },
          {
            "id": "fedbf66e-cfd2-52bb-b9db-393d815aade7",
            "score": 0.690174400806427,
            "metadata": {
              "text": "Key words:\ndiabetes, gene, polygenic, monogenic\nIntroduction\nDiabetes is one of the most common metabolic disor -\nders. It is estimated that the number of diabetes pa -\ntients worldwide has already exceeded 200 million\n[92]. This creates a need to understand the etiology ofthe disease, genetic and enviromental factors influ -\nencing development of diabetes. Diabetes is a group\nof metabolic diseases that are characterized by ele -\nvated glucose level. Poorly controlled or undiagnosed",
              "title": "2005 - Type 2 diabetes mellitus from genes to disease.pdf",
              "version": "v0",
              "chunk_order": 5,
              "document_id": "52687a38-6a4b-51d2-aafa-812c76981dfe",
              "extraction_id": "d79047d9-58d0-5440-b63c-e648b5df5538",
              "associatedQuery": "What is the difference between monogenic and polygenic diabetes?"
            }
          },
          {
            "id": "c48920f3-1236-5921-b2a1-f09edba1e7ec",
            "score": 0.689247727394104,
            "metadata": {
              "text": "2 \n 1.1.2 Introduction \nMonogenic diabetes is caused by a single defect in one of over 40 genes1,2. Since \nMODY (maturity onset diabetes of the young) was named by Fajans for the T2D -like \npresentation in young people with an autosomal dominant pattern of inheritance3,4, our \nunderstanding of phenotypic and genetic heterogeneity in monogenic diabetes has \nincreased. The major monogenic diabetes categories are MODY, neon atal diabetes",
              "title": "2021 - Genomic Medicine in Diabetes Improving the Diagnostic Rate of Monogenic Diabetes.pdf",
              "version": "v0",
              "chunk_order": 77,
              "document_id": "e315a891-ba59-57e9-856b-602544375324",
              "extraction_id": "29df597d-e40d-5bc8-8ee0-89141d8e7fc0",
              "associatedQuery": "What is the difference between monogenic and polygenic diabetes?"
            }
          }
        ],
        "kg_search_results": null
      }
    }
  },
  "20": {
    "results": {
      "completion": {
        "id": "chatcmpl-AIFrXDhQoyphIy4Ti77mFez47y0g5",
        "choices": [
          {
            "finish_reason": "stop",
            "index": 0,
            "message": {
              "content": "Studying the DNA of people with diabetes helps scientists find better treatments or cures in several ways:\n\n1. **Identification of Genetic Determinants**: By performing genetic profiling on diabetic patients, scientists can identify genetic determinants that define the targets of current and future therapies. This leads to the development of therapies that are more specific to the genetic makeup of individuals with diabetes [1].\n\n2. **Understanding Disease Mechanisms**: Genetic studies improve our understanding of the biological mechanisms that maintain glucose homeostasis and reveal molecular defects leading to chronic hyperglycemia. This knowledge can lead to the development of more specifically targeted antidiabetic drugs or even gene-based therapies [4].\n\n3. **Pharmacogenetics**: Pharmacogenetic testing can be used to predict therapeutic responses to different classes of drugs for each patient, allowing for more personalized treatment plans [4].\n\n4. **Discovery of New Therapeutic Targets**: A greater understanding of the genetic and epigenetic basis of diabetes can enable the discovery of new therapeutic targets, potentially leading to novel treatments for diabetes and its complications [3].\n\n5. **Stratification of Diabetes Subclasses**: By analyzing DNA variations and their interactions with environmental factors, scientists can stratify type 2 diabetes into subclasses. This stratification allows for more effective treatment strategies tailored to specific genetic and lifestyle interactions [8].\n\n6. **Identification of Key Genetic Elements**: Genetic studies can identify key genetic elements that determine susceptibility to diabetes, disease progression, and responsiveness to specific therapies. This information helps in identifying novel targets for future interventions [9].\n\nOverall, studying the DNA of people with diabetes provides critical insights that drive the development of more effective and personalized treatments.",
              "role": "assistant"
            }
          }
        ],
        "created": 1728914243,
        "model": "gpt-4o-2024-08-06",
        "object": "chat.completion",
        "system_fingerprint": "fp_6b68a8204b",
        "usage": {
          "completion_tokens": 337,
          "prompt_tokens": 1130,
          "total_tokens": 1467
        }
      },
      "search_results": {
        "vector_search_results": [
          {
            "id": "e7d89095-ef66-5c11-982c-879791dd14a4",
            "score": 0.7071230586097814,
            "metadata": {
              "text": "by performing a genetic profile on diabetic patients (pharmacogenetics). \nFurthermore, identification of genetic determinants of diabetic patients will \nbetter define the targets of current and future therapies, and will lead to \ntherapies that are more specific for their genetic constitutes. \nSUMMARY \nWith the advancement of the Human Genome Project, we enter the \nera of a sequence-based biology. Some progress has been made in the",
              "title": "2004 - Diabetes Genes a.pdf",
              "version": "v0",
              "chunk_order": 685,
              "document_id": "805ad0d5-6a3d-5e5f-b1ba-121e10c6daaa",
              "extraction_id": "a19924b0-a834-5100-8b24-6b57dcddb82a",
              "associatedQuery": "How does studying the DNA of people with diabetes help scientists find better treatments or cures?"
            }
          },
          {
            "id": "c21caf96-f04a-551d-92b2-f4ff084d43c8",
            "score": 0.6807519983797087,
            "metadata": {
              "text": "Todate,studiesofdiabeteshaveplayedamajorroleinshapingthinkingabout\nthegeneticanalysisofcomplexdiseases.Basedontrendsingenomicinformationandtechnology,combinedwiththegrowingpublichealthimportanceofdiabetes,diabetes will likely continue to be an important arena in which methods will bepioneeredandlessonslearned.Itiswithgreatenthusiasmthatwelookforwardtothis effort, and with avid curiosity we await to see whether the lessons of todaywill be supported by the data of tomorrow.",
              "title": "2003 - The Inherited Basis of Diabetes Mellitus.pdf",
              "version": "v0",
              "chunk_order": 172,
              "document_id": "7b85b290-d711-55d5-9b1e-b06e4d6f14a2",
              "extraction_id": "2aa8d99c-99d7-55de-aa2a-c24a46ea9058",
              "associatedQuery": "How does studying the DNA of people with diabetes help scientists find better treatments or cures?"
            }
          },
          {
            "id": "fceee048-359b-5854-b45d-5531b9374ce8",
            "score": 0.6783680511493665,
            "metadata": {
              "text": "DNA code. Therefore, greater unders tanding of the epigenetic basis of disease could enable the 576 \ndiscovery new therapeutic targets for the treat ment of numerous human diseases including 577 \ndiabetes and its complications. 578 \n 579 \n580",
              "title": "2010 - The Role of Epigenetics in the Pathology of Diabetic Complications.pdf",
              "version": "v0",
              "chunk_order": 123,
              "document_id": "766edfd5-4756-51bf-b636-c94b041d030c",
              "extraction_id": "919cb859-8f47-5930-8713-090520be523f",
              "associatedQuery": "How does studying the DNA of people with diabetes help scientists find better treatments or cures?"
            }
          },
          {
            "id": "f4e8a3c8-0b85-5595-8917-933aced8b3ba",
            "score": 0.6781570709300941,
            "metadata": {
              "text": "T ogether, these discoveries will continue to improve our \nunderstanding of the biologic mechanisms that maintain glucose homeostasis, and of still hidden  molecular defects leading to \nchronic hyperglycemia, and could also lead to the development of more speci  cally targeted antidiabetic drugs or even gene -\n based therapies. Moreover, pharmacogenetic testing might then be used to predict, for each patient, the therapeutic response to different classes of drugs. The identi  cation of T2DM genes will",
              "title": "2010 - The Genetics of Type 2 Diabetes.pdf",
              "version": "v0",
              "chunk_order": 258,
              "document_id": "a1d211d4-279e-51d7-b2b2-33bc2763d089",
              "extraction_id": "c4de4c07-4749-5401-bbf3-16988c132852",
              "associatedQuery": "How does studying the DNA of people with diabetes help scientists find better treatments or cures?"
            }
          },
          {
            "id": "df21554e-6053-53ae-aae5-e3d1dba1b1f5",
            "score": 0.6774097479161723,
            "metadata": {
              "text": "research will contribute positive ly to the life of people living with T1D . Being able pinpoint \nmutations, and then discover how they contribute to the genetic  cause  of a condition, can help \nto open up path s for pharmaceutical treatments. Currently, m ost treatment strategies for genetic \ndisorders do not alter the underlying genetic mutation;  but are designed to improve particular \nsigns and symptoms associated with the disorder. For instance, T1D  is managed by",
              "title": "2016 - The Genomics of Type 1 Diabetes.pdf",
              "version": "v0",
              "chunk_order": 490,
              "document_id": "4933cdc2-7d36-5181-87c9-63b58498839f",
              "extraction_id": "5e43ab7d-3e2b-551c-9a90-f91e970cb8d7",
              "associatedQuery": "How does studying the DNA of people with diabetes help scientists find better treatments or cures?"
            }
          },
          {
            "id": "db06230d-31c0-5947-8c1c-f58c48b6f439",
            "score": 0.6734455021088717,
            "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 does studying the DNA of people with diabetes help scientists find better treatments or cures?"
            }
          },
          {
            "id": "e0b86e8e-4e1a-5f6b-9b41-e9a4f912790c",
            "score": 0.6734237269506629,
            "metadata": {
              "text": "new therapeutic targets and identify potential diabetic neuropathy\nbiomarkers. The genes identied in the current study conrm datagathered from experimental models of diabetes and provide a\ncomprehensive picture of the expression of multiple targets in asingle human tissue sample.\nOur initial analyses of this data set classied the patient samples\nbased on myelinated bre density and found that two large\ngroups emerged; those with a loss of myelinated bre density\n5500 bres/mm",
              "title": "2011 - The identification of gene expression.pdf",
              "version": "v0",
              "chunk_order": 79,
              "document_id": "61558082-f092-5a1d-abbb-a5a81e8a959b",
              "extraction_id": "e5e4169a-56d8-539f-8ebc-ad44eb75433f",
              "associatedQuery": "How does studying the DNA of people with diabetes help scientists find better treatments or cures?"
            }
          },
          {
            "id": "cc98a5b9-131e-5b60-919e-82e86b7a37a7",
            "score": 0.670618513748823,
            "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 does studying the DNA of people with diabetes help scientists find better treatments or cures?"
            }
          },
          {
            "id": "b092c8b9-edb1-55fb-ae16-c67e3298946e",
            "score": 0.6671704848307094,
            "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": "How does studying the DNA of people with diabetes help scientists find better treatments or cures?"
            }
          },
          {
            "id": "efd7c210-858d-5125-8da9-46862e19a58a",
            "score": 0.6628995343566496,
            "metadata": {
              "text": "the onset and progression of diabetic neuropathy is of primeimportance.\nThe current study takes an important rst step towards this goal\nby identifying specic sets of genes whose expression accurately\nclassies patient samples with regard to diabetic neuropathy pro-\ngression and by analysing their interactions within known cellularpathways. Identifying common elements in these complex net-works will yield novel insights into disease pathogenesis, provide",
              "title": "2011 - The identification of gene expression.pdf",
              "version": "v0",
              "chunk_order": 78,
              "document_id": "61558082-f092-5a1d-abbb-a5a81e8a959b",
              "extraction_id": "e5e4169a-56d8-539f-8ebc-ad44eb75433f",
              "associatedQuery": "How does studying the DNA of people with diabetes help scientists find better treatments or cures?"
            }
          }
        ],
        "kg_search_results": null
      }
    }
  }
}