aboutsummaryrefslogtreecommitdiff
path: root/gnqa/paper2_eval/data/responses/gpt4o/cs_aging_responses.json
blob: 25d5e17301d47094cc6d7284d7b6e12fb4336c4b (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-AIFgRqvOB8PnpNpKMnpdr80oxf2MI",
        "choices": [
          {
            "finish_reason": "stop",
            "index": 0,
            "message": {
              "content": "The main genetic factors that influence aging include:\n\n1. **Genomic Instability**: Aging is associated with the accumulation of DNA damage and the dysregulation of repair mechanisms, leading to genomic instability [9].\n\n2. **Telomere Attrition**: The shortening of telomeres, which are protective caps at the ends of chromosomes, is a significant factor in aging [9], [10].\n\n3. **Epigenetic Alterations**: Changes in epigenetic marks, which regulate gene expression without altering the DNA sequence, play a crucial role in aging [2], [4], [9], [10].\n\n4. **Deregulated Nutrient Sensing**: The pathways that sense and respond to nutrients become deregulated with age, affecting longevity [10].\n\n5. **Mitochondrial Dysfunction**: Mitochondria, the energy-producing organelles in cells, become less efficient with age, contributing to the aging process [10].\n\n6. **Cellular Senescence**: The process by which cells lose the ability to divide and function properly is a hallmark of aging [9], [10].\n\n7. **Loss of Proteostasis**: The ability of cells to maintain protein homeostasis declines with age, leading to the accumulation of damaged proteins [9], [10].\n\n8. **Stem Cell Exhaustion**: The decline in the regenerative capacity of stem cells contributes to aging [10].\n\nThese factors collectively shape the complex genetic landscape of aging, influencing the expression of aging phenotypes and lifespan [7].",
              "role": "assistant"
            }
          }
        ],
        "created": 1728913555,
        "model": "gpt-4o-2024-08-06",
        "object": "chat.completion",
        "system_fingerprint": "fp_6b68a8204b",
        "usage": {
          "completion_tokens": 312,
          "prompt_tokens": 1050,
          "total_tokens": 1362
        }
      },
      "search_results": {
        "vector_search_results": [
          {
            "id": "3117c019-7311-53ae-8ab1-927ca822c709",
            "score": 0.698876044120671,
            "metadata": {
              "text": "It is undisputed that genetic factors influence aging. In a remarkable",
              "title": "2009 - The Human Ageing Genomic Resources online.pdf",
              "version": "v0",
              "chunk_order": 9,
              "document_id": "e43cd3b6-ad8e-5422-ba7c-ceb6e66cc529",
              "extraction_id": "7ada6b55-99c2-5e20-bf96-d153f927256c",
              "associatedQuery": "What are the main genetic factors that influence aging?"
            }
          },
          {
            "id": "a9434032-4a9d-54f8-a7a6-16110d1b3118",
            "score": 0.697221577167511,
            "metadata": {
              "text": "males: what are the molecular and evolutionary causes? Aging Cell. 2007;6:225233. doi:10.1111/j.1474-9726.2007.00279.x\n 63. Benayoun BA, Pollina EA, Brunet A. Epigenetic regulation of ageing: link-\ning environmental inputs to genomic stability. Nat Rev Mol Cell Biol. 2015;16:593610. doi:10.1038/nrm4048\n 64. Sen P, Shah PP, Nativio R, Berger SL. Epigenetic mechanisms of longevity \nand aging. Cell. 2016;166:822839. doi:10.1016/j.cell.2016.07.050",
              "title": "2018 - Sex Differences in Aging Genomic Instability.pdf",
              "version": "v0",
              "chunk_order": 158,
              "document_id": "8cfb5529-7f0c-58fc-b6e4-b3ee800fb72f",
              "extraction_id": "0104338d-cc9c-538f-be29-8343a64da37d",
              "associatedQuery": "What are the main genetic factors that influence aging?"
            }
          },
          {
            "id": "a0672677-71ad-5603-8427-a0648eec407f",
            "score": 0.6917247176170349,
            "metadata": {
              "text": "Clinical Genetics and Genomics of Aging",
              "title": "2020 - Clinical Genetics and Genomics of Aging.pdf",
              "version": "v0",
              "chunk_order": 1,
              "document_id": "62b635c3-040e-512a-b016-6ef295308a1e",
              "extraction_id": "4ea8424f-1cd8-569c-a1df-3f0f54206e70",
              "associatedQuery": "What are the main genetic factors that influence aging?"
            }
          },
          {
            "id": "c1b5a31a-066d-571b-af1f-db746d9d17f6",
            "score": 0.684185266494751,
            "metadata": {
              "text": "standing the cause and mechanisms of aging is imperative in\nassisting to suppress age-related diseases and promote healthylongevity. It is well-known that aging is influenced by a combin-\nation of genetic and environmental factors. Previous twin stud-\nies have shown that the genetic contribution to general human\nlongevity is about 2030% [ 4,5], whereas environmental factors\nin human aging and longevity still account for the largest effect.\nEpigenetic factors influence the regulation of gene expres-",
              "title": "2016 - Progress on the role of DNA methylation in aging.pdf",
              "version": "v0",
              "chunk_order": 4,
              "document_id": "e4cdc02f-4415-5638-aab8-f848b4d64a22",
              "extraction_id": "bcb3c620-b960-5af6-95ea-13215c31672e",
              "associatedQuery": "What are the main genetic factors that influence aging?"
            }
          },
          {
            "id": "e09c33ea-4139-5cc2-9cf5-a40045f26a0c",
            "score": 0.6728764376901522,
            "metadata": {
              "text": "Recent developments on the genetics of aging can be seen as several streams\nof effort. In general, humans show a relatively modest ( <50%) heritability of",
              "title": "2001 - The genetics of aging.pdf",
              "version": "v0",
              "chunk_order": 13,
              "document_id": "aa9a9193-b6f3-5ef8-aefd-e01ec44abb46",
              "extraction_id": "76bae746-eabf-51ed-a01f-d32ecc89c11b",
              "associatedQuery": "What are the main genetic factors that influence aging?"
            }
          },
          {
            "id": "2d0a20b8-4196-5451-9d99-282f82234464",
            "score": 0.6651506823121736,
            "metadata": {
              "text": "effect  genetic  variants  on  human  longevity.  Aging  2,  612620.\nYu,  C.E.,  Seltman,  H.,  Peskind,  E.R.,  Galloway,  N.,  Zhou,  P.X.,  Rosenthal,  E.,  Wijsman,\nE.M.,  Tsuang,  D.W.,  Devlin,  B.,  Schellenberg,  G.D.,  2007.  Comprehensive  analysis\nof  APOE  and  selected  proximate  markers  for  late-onset  Alzheimers  disease:\npatterns  of  linkage  disequilibrium  and  disease/marker  association.  Genomics",
              "title": "2011 - A genome-wide association study confirms APOE as the major gene influencing.pdf",
              "version": "v0",
              "chunk_order": 109,
              "document_id": "63b27b06-db2c-5542-9b1a-cb9ebe64d339",
              "extraction_id": "210aa417-372c-5bf6-b961-e281a1817458",
              "associatedQuery": "What are the main genetic factors that influence aging?"
            }
          },
          {
            "id": "8bcb7ae0-ac45-5b4c-8a4b-626564e8ec11",
            "score": 0.6649871668227227,
            "metadata": {
              "text": "factors shape a complex scenario for which clear answers\nof the regulation of longevity have been dicult to distill.\nWith the discovery of genetic factors underlying aging in\nexperimental laboratory models, forays into the genetic\nregulation of these properties have rapidly expanded,\nuncovering conserved mechanisms across diverse metazoa\nthat inuence expression of aging phenotypes and lifespan.\nYet, the story gets muddled in that these factors are often",
              "title": "2021 - Footprints in the Sand Deep Taxonomic Comparisons in Vertebrate Genomics to Unveil the Genetic Programs of Human Longevity.pdf",
              "version": "v0",
              "chunk_order": 8,
              "document_id": "0dc45abe-ab02-5b07-9916-7093b53323c0",
              "extraction_id": "34223e0e-590c-5f26-b120-b7250cd91b99",
              "associatedQuery": "What are the main genetic factors that influence aging?"
            }
          },
          {
            "id": "786d2756-4c4d-5ac0-8d3d-63f914d51664",
            "score": 0.6641277475204816,
            "metadata": {
              "text": "In addition to aging- and CR-related genes, another\nsource of candidate genes and pathways for drug designare human longevity-associated genes (Barzilai andShuldiner, 2001; Browner et al., 2004; Kenyon, 2010).Dozens of genes have now been associated with humanlongevity (de Magalha es et al., 2009a), although only ahandful of genes have been shown to have consistenteffects across populations.\nMany longevity-associated genes are related to spe-",
              "title": "2012 - Genome-Environment Interactions That Modulate.pdf",
              "version": "v0",
              "chunk_order": 119,
              "document_id": "b1a1997c-e9df-5dc0-9d12-a3977d0c64ec",
              "extraction_id": "d59d7882-333d-5576-86ab-3cfa6354b946",
              "associatedQuery": "What are the main genetic factors that influence aging?"
            }
          },
          {
            "id": "d811de8c-b666-5bb5-b0eb-a9b17fa16a8e",
            "score": 0.6602960026440634,
            "metadata": {
              "text": "tion for decades, the underlying molecular genetic\ncauses of and responses to aging remain an area of\nactive study. Research from model systems hascharacterized a range of physiological and molecular\nphenotypes associated with aging. These include genomic\ninstability caused by accumulation of DNA damage,\ndysregulation of repair mechanisms, and telomere attri-\ntion; epigenetic alterations; dysregulation of transcription;\nloss of proteostasis; cellular senescence; and deregulated",
              "title": "2017 - Genome-wide transcriptomics of aging.pdf",
              "version": "v0",
              "chunk_order": 6,
              "document_id": "1a2a3737-b0a6-58b9-908f-50753241a309",
              "extraction_id": "c7d6d597-a9c7-5db2-888d-5f9297f0af47",
              "associatedQuery": "What are the main genetic factors that influence aging?"
            }
          },
          {
            "id": "081e12f9-359c-5a2c-b740-714d637367d3",
            "score": 0.6599280440850244,
            "metadata": {
              "text": "143\nThe molecular bases of ageing are multi factorial, but there are nine distinctive \nfeatures related to this process, which include genomic instability, telomere shorten-\ning, de-regulated nutrient sensing, mitochondrial dysfunction, cellular senescence, \nstem cell exhaustion, altered cellular senescence, loss of proteostasis and a change \nin the patterns of epigenetic modifications [4, 5].\n Epigenetics andAgeing\nEpigenetics is considered as a dynamic interface between the genome and the envi-",
              "title": "2020 - Clinical Genetics and Genomics of Aging.pdf",
              "version": "v0",
              "chunk_order": 986,
              "document_id": "62b635c3-040e-512a-b016-6ef295308a1e",
              "extraction_id": "517379dd-d351-5e9a-8e78-72e543bb2945",
              "associatedQuery": "What are the main genetic factors that influence aging?"
            }
          }
        ],
        "kg_search_results": null
      }
    }
  },
  "2": {
    "results": {
      "completion": {
        "id": "chatcmpl-AIFgbQ1pPISWJITI2YHN6poeJtTyV",
        "choices": [
          {
            "finish_reason": "stop",
            "index": 0,
            "message": {
              "content": "Genes affect the aging process in humans through complex interactions and pathways. Research has shown that single genes can regulate aging in model organisms, indicating that aging can be genetically manipulated [2]. Hundreds of genes have been identified that modulate longevity in these organisms, and some of these genes and their associated pathways, such as the insulin/IGF1/GH pathway, have been shown to affect longevity across different species, suggesting that some mechanisms of aging are evolutionarily conserved [3]. \n\nIn humans, it has been more challenging to identify specific longevity candidate genes, but studies have shown that certain genes associated with aging in model organisms are evolutionarily conserved and may be relevant to human aging [5]. Additionally, approximately 4% of genes analyzed in a study of postmortem human brain tissue showed significant age-related expression changes, indicating that these genes play central roles in processes like synaptic plasticity, vesicular transport, and mitochondrial function [8]. \n\nOverall, while aging is a complex process involving multiple genes and their interactions with the environment, genetic studies in model organisms provide insights that may be applicable to understanding human aging [9].",
              "role": "assistant"
            }
          }
        ],
        "created": 1728913565,
        "model": "gpt-4o-2024-08-06",
        "object": "chat.completion",
        "system_fingerprint": "fp_6b68a8204b",
        "usage": {
          "completion_tokens": 226,
          "prompt_tokens": 1287,
          "total_tokens": 1513
        }
      },
      "search_results": {
        "vector_search_results": [
          {
            "id": "0ae63c75-df5f-59b0-9561-30d5115f0f74",
            "score": 0.7094772631241535,
            "metadata": {
              "text": "potentially associated with human ageing. For eachgene, a description compiled from the studies that linkthe gene to ageing is provided. It should be noted thatour focus is on genes that might affect the ageingprocess, rather than individual age-related pathologies;\ngenes affecting multiple, even if not all, age-related",
              "title": "2012 - Human Ageing Genomic Resources Integrated.pdf",
              "version": "v0",
              "chunk_order": 20,
              "document_id": "5f554cc7-c94d-5fbd-9567-528499663ed6",
              "extraction_id": "25e9d8a3-54ac-5412-8efb-3b56d93f363f",
              "associatedQuery": "How do genes affect the aging process in humans?"
            }
          },
          {
            "id": "c258bd44-b5b3-5eb5-9c72-60b365f18f81",
            "score": 0.7014725146044777,
            "metadata": {
              "text": "showing that single genes can regulate aging in modelorganisms demonstrate that aging can be geneticallymanipulated (Finch and Ruvkun, 2001; Kenyon, 2010).Hundreds of genes that modulate longevity have nowbeen identified in model organisms (de Magalha es et al.,2009a). In some cases (e.g., in worms), mutations insingle genes can extend lifespan by almost 10-fold (Ayy-adevara et al., 2008). Nonetheless, aging is a complexprocess that derives not from single genes but from theinteractions of multiple genes",
              "title": "2012 - Genome-Environment Interactions That Modulate.pdf",
              "version": "v0",
              "chunk_order": 17,
              "document_id": "b1a1997c-e9df-5dc0-9d12-a3977d0c64ec",
              "extraction_id": "68b12e10-d643-570a-aa63-eda62de81928",
              "associatedQuery": "How do genes affect the aging process in humans?"
            }
          },
          {
            "id": "4d6876c5-9226-587c-8d3e-d4957ee42dba",
            "score": 0.6974861206122577,
            "metadata": {
              "text": "genes (http://genomics.senescence.info/genes/), more than700 genes have been identified that regulate lifespan inmodel organisms (de Magalha es et al., 2009a). Many ofthese genes and their associated pathwayssuch as theinsulin/IGF1/GH pathwayhave been shown to affect lon-gevity across different model organisms (Kenyon, 2010).Therefore, at least some mechanisms of aging are evolu-tionarily conserved and may have potential therapeuticapplications (Baur et al., 2006). For example, evidencesuggests the use of",
              "title": "2012 - Genome-Environment Interactions That Modulate.pdf",
              "version": "v0",
              "chunk_order": 65,
              "document_id": "b1a1997c-e9df-5dc0-9d12-a3977d0c64ec",
              "extraction_id": "a01ca925-4ccf-5863-a162-7bd4c754fe89",
              "associatedQuery": "How do genes affect the aging process in humans?"
            }
          },
          {
            "id": "0671ed95-2909-54ac-baea-b156ee0ea8f8",
            "score": 0.6964022698162645,
            "metadata": {
              "text": "key genes and pathways important in aging; geneticstudies of heritable diseases that cause the appearanceof premature aging in affected people; physiological ex-Introductionperiments that relate the pace of aging to caloric intake;Is aging the final act in the script of developmental biol-and advances in human genetics, as well as cell andogy? The characteristic changes that are part and parcelmolecular biology leading to an understanding of theof aging appear similar to developmentally regulatedbasis of",
              "title": "1999 - Molecular Biology of Aging.pdf",
              "version": "v0",
              "chunk_order": 1,
              "document_id": "6f122d3a-d8ca-598f-8767-c059a941cef3",
              "extraction_id": "66c6e911-2b6e-5d79-9e13-9f0e370c6fa6",
              "associatedQuery": "How do genes affect the aging process in humans?"
            }
          },
          {
            "id": "650300e1-898c-56e2-9358-0bb6625b0073",
            "score": 0.694001967787392,
            "metadata": {
              "text": "shown that genes associated with aging and/or longevity inmodel organisms are evolutionary conserved in terms of havingmore homologues than predicted by chance (Budovsky  \net \n  \nal \n.,2007, 2008) and exhibiting slower molecular evolution rates (de\nMagalhes & Church, 2007). Therefore, it is now clear that atleast some genes identified in model organisms may be relevantto human aging.\nTo allow researchers to focus specifically on human aging,",
              "title": "2009 - The Human Ageing Genomic Resources online.pdf",
              "version": "v0",
              "chunk_order": 16,
              "document_id": "e43cd3b6-ad8e-5422-ba7c-ceb6e66cc529",
              "extraction_id": "52c67b46-63f2-54ae-a78e-e9d54a55f6e4",
              "associatedQuery": "How do genes affect the aging process in humans?"
            }
          },
          {
            "id": "cc78a209-081f-5e0c-877a-36b85d5105a6",
            "score": 0.6935852352627984,
            "metadata": {
              "text": "expression of certain genes have an effect upon longevity.\nAlthough similar aging processes are likely to operateacross multiple species [30], it has been much more diffi-cult to identify longevity candidate genes in human studies[30]. A key question in human aging is to what extent asignature of aging may be detectable across tissues. Until\nnow there has been a lack of large transcriptional profiles\nfrom the same human individuals in multiple tissues. TheMuTHER study provides ins ight into the human aging",
              "title": "2013 - Gene expression changes with age in skin.pdf",
              "version": "v0",
              "chunk_order": 49,
              "document_id": "5c121bbb-57b8-51cc-8461-effa1bfd87b9",
              "extraction_id": "18e9f96b-9af8-59bf-9323-084df9a4f9b6",
              "associatedQuery": "How do genes affect the aging process in humans?"
            }
          },
          {
            "id": "a3859151-a94e-5085-8e7a-3c1d50716319",
            "score": 0.6925199581647007,
            "metadata": {
              "text": "complex.108,109Studies on models such as the yeast Sac-\ncharomyces cerevisiae110the nematode Caenorhabditis\nelegans,111the fly Drosophila melanogaster,112-114the\nmouse Mus musculus,115and humans116show that single\ngene mutations can contribute to the initiation of aging andinduce premature aging syndromes. There are, however, nospecial genes that can cause aging-associated damages. Themanifestation of aging is mostly due to the failure of main-tenance and repair mechanisms.\n117,118",
              "title": "2011 - Clinical aspects and molecular diagnostics of skin aging.pdf",
              "version": "v0",
              "chunk_order": 70,
              "document_id": "e32f8f2c-d3ad-5dae-a393-9bd87c370ebe",
              "extraction_id": "39cc0d86-f175-5fe3-990f-8471c7395ba4",
              "associatedQuery": "How do genes affect the aging process in humans?"
            }
          },
          {
            "id": "d53018ae-0881-5ef4-9c49-48623e8aa342",
            "score": 0.6913864200480938,
            "metadata": {
              "text": "on model organisms [3] or have been confined to specificaging-associated disorders such as progeria syndromes\n[4]. A study of postmortem human brain tissue from 30\nindividuals aged 26 to 106 years [5] showed that approxi-\nmately 4% of approximately 11,000 genes analyzed show\na significant age-related expression change (1.5-fold or\nmore) in individuals aged >40 years. These genes were\nreported to play central roles in synaptic plasticity, vesi-\ncular transport, and mitoch ondrial function. Another",
              "title": "2013 - Gene expression changes with age in skin.pdf",
              "version": "v0",
              "chunk_order": 6,
              "document_id": "5c121bbb-57b8-51cc-8461-effa1bfd87b9",
              "extraction_id": "dfb687b2-f1ff-5e22-8a67-4a1db9ebeb3c",
              "associatedQuery": "How do genes affect the aging process in humans?"
            }
          },
          {
            "id": "a4773f1a-f2d3-5950-a81e-d22357e97a0f",
            "score": 0.6908473144951683,
            "metadata": {
              "text": "of multiple genes with each other and withthe environment. Evidence from animal systems showsa major impact of the environment on aging, yet envi-ronmental manipulations of aging act through genesand proteins, usually by triggering signaling pathwaysand modulating gene expression. In fact, some geneshave been shown in model organisms to have varyingeffects on lifespan depending on diet (Heikkinen et al.,2009). Genes that can regulate aging in model organ-isms cannot be directly applied to humans through",
              "title": "2012 - Genome-Environment Interactions That Modulate.pdf",
              "version": "v0",
              "chunk_order": 18,
              "document_id": "b1a1997c-e9df-5dc0-9d12-a3977d0c64ec",
              "extraction_id": "68b12e10-d643-570a-aa63-eda62de81928",
              "associatedQuery": "How do genes affect the aging process in humans?"
            }
          },
          {
            "id": "91530253-7015-5b7a-bb7b-506ddeb3c3f8",
            "score": 0.6904643546981388,
            "metadata": {
              "text": "[2] L. Partridge, D. Gems, Mechanisms of ageing: public or private? Nat. Rev.\nGenet. 3 (2002) 165 175.\n[3] A.M. Leroi, et al., What evidence is there for the existence of individual\ngenes with antagonistic pleiotropic effects? Mech. Ageing Dev. 126 (2005)421429.\n[4] S.N. Austad, Is aging programmed? Aging Cells 3 (2004) 249 251.\n[5] V.D. Longo, J. Mitteldorf, V.P. Skulachev, Opinion: programmed and\naltruistic ageing, Nat. Rev. Genet. 6 (2005) 866 872.",
              "title": "2007 - Characterization of a bidirectional promoter shared between two human.pdf",
              "version": "v0",
              "chunk_order": 76,
              "document_id": "e9a81a0d-52b4-5ced-be05-130eed317085",
              "extraction_id": "a5581fce-8af9-5131-a94b-8fce7fbf9557",
              "associatedQuery": "How do genes affect the aging process in humans?"
            }
          }
        ],
        "kg_search_results": null
      }
    }
  },
  "3": {
    "results": {
      "completion": {
        "id": "chatcmpl-AIFgiWkzt5opfBd5VTvAKGVKegG8y",
        "choices": [
          {
            "finish_reason": "stop",
            "index": 0,
            "message": {
              "content": "To slow down genetic aging, several lifestyle choices can be beneficial:\n\n1. **Dietary Restriction (DR)**: Reducing food intake without causing malnutrition has been shown to extend lifespan in various organisms. This approach improves healthspan by influencing nutrient-sensing signaling networks and metabolism [1].\n\n2. **Healthy Diet and Physical Exercise**: Engaging in regular physical activity and maintaining a healthy diet can slow down aging and prevent or modify many chronic diseases prevalent in older adults. This contributes to successful aging, allowing individuals to achieve physical, social, and mental well-being [4].\n\n3. **Caloric Restriction**: This is considered a reasonable anti-aging intervention, as it has been shown to slow aging in multiple species, including yeast, nematodes, fruit flies, and rodents [6].\n\n4. **Exercise**: While exercise may not retard aging in all tissues, it can contribute to molecular-level aging retardation, which is beneficial for overall health and longevity [10].\n\nThese lifestyle choices, when combined, can help mitigate the effects of genetic aging and promote a longer, healthier life.",
              "role": "assistant"
            }
          }
        ],
        "created": 1728913572,
        "model": "gpt-4o-2024-08-06",
        "object": "chat.completion",
        "system_fingerprint": "fp_6b68a8204b",
        "usage": {
          "completion_tokens": 222,
          "prompt_tokens": 1158,
          "total_tokens": 1380
        }
      },
      "search_results": {
        "vector_search_results": [
          {
            "id": "7460a40c-8723-5de9-9f2e-c781f4872f1f",
            "score": 0.6213953680215888,
            "metadata": {
              "text": "as diabetes, cancer and neurodegenerative disorders\n[1, 2]. Environmental and genetic interventions can\nameliorate the effects of aging, with nutrition,\nnutrient-sensing signaling networks and metabolism\nplaying evolutionarily conserved roles [1, 3 5]. Diet-\nary restriction (DR), in which food intake is reducedwhile avoiding malnutrition, extends lifespan in di-\nverse model and non-model organisms [3, 6]. DR\ninduces a remarkably broad-spectrum improvement in",
              "title": "2017 - Dietary restriction protects from age-associated DNA methylation and induces epigenetic reprogramming of lipid metabolism.pdf",
              "version": "v0",
              "chunk_order": 4,
              "document_id": "fe573bb0-3d37-55e5-93fa-65b3fbc5f532",
              "extraction_id": "21efa872-9d89-5dee-9dd1-27dcaa1208cf",
              "associatedQuery": "What lifestyle choices can help slow down genetic aging?"
            }
          },
          {
            "id": "d78564d5-d785-554a-bb2c-d71917ccfe19",
            "score": 0.6209702676823817,
            "metadata": {
              "text": "limiting exposure to exogenous genotoxins and by suppressing metabolism  thereby producing fewer reactive species. However, DNA damage, like caloric restriction, can also elicit a protective survival response that promotes longevity and healthy aging. Recently, the use of sirolimus in mice was found to extend their life span and de -\nlay the development of conditions associated with aging, including cancer.\n1 Sirolimus is one of pre -",
              "title": "2009 - DNA Damage, Aging, and Cancer.pdf",
              "version": "v0",
              "chunk_order": 95,
              "document_id": "630c29c7-1dd7-509e-9b6b-b4af98b4ea48",
              "extraction_id": "b03f4297-85f4-5011-8dcf-ec169d3051d3",
              "associatedQuery": "What lifestyle choices can help slow down genetic aging?"
            }
          },
          {
            "id": "4bf7307d-d8a0-5594-b0b5-487fe0f265ca",
            "score": 0.6070101442042038,
            "metadata": {
              "text": "Longev. Heal. 2, 10 (2013).\n7. Kreienkamp Ret al.Doubled lifespan and patient-like pathologies in progeria mice fed high-fat diet. \nAging Cell18, e12852 (2019). [PubMed: 30548460] \n8. Heilbronn LK & Ravussin E Calorie restriction and aging: review of the literature and implications \nfor studies in humans. Am. J. Clin. Nutr. 78, 361369 (2003). [PubMed: 12936916] \n9. Liang Yet al.Calorie restriction is the most reasonable anti-ageing intervention: a meta-analysis of",
              "title": "2021 - Gene-by-environment modulation of lifespan and weight gain in the murine BXD family.pdf",
              "version": "v0",
              "chunk_order": 132,
              "document_id": "4d082da4-fa48-5170-8147-c4fea47a5d4b",
              "extraction_id": "30ba3324-6e19-58c2-9e32-508f827af3e5",
              "associatedQuery": "What lifestyle choices can help slow down genetic aging?"
            }
          },
          {
            "id": "da620f88-db92-5267-af81-d6b548e9f29c",
            "score": 0.6065477548078396,
            "metadata": {
              "text": "can be slowed down to some extent by eating a healthy diet and taking physical exercise, and many of the chronic diseases prevalent in older adults are either preventable or modi  able with healthy lifestyle habits. Thus, older adults \ncan experience successful aging that allows them to achieve physical, social and mental well - being over the life course and to participate in society. \n Much research has been conducted in recent years to",
              "title": "2012 - Systems Biology Approaches to Nutrition.pdf",
              "version": "v0",
              "chunk_order": 7631,
              "document_id": "6955478b-950d-5d29-b24c-3a5ca656f3ae",
              "extraction_id": "791bae8d-8d24-5873-b611-9c289591d11d",
              "associatedQuery": "What lifestyle choices can help slow down genetic aging?"
            }
          },
          {
            "id": "c96b67f8-ad31-50fd-b053-07b127938ef2",
            "score": 0.5996953070254556,
            "metadata": {
              "text": "During the past century, remarkable progress has been \nmade in unveiling the mechanisms of aging. Genetic and molecular pathways that regulate healthspan and lifespan have been identified in various model organisms, provid-ing a rich knowledge base (Longo etal. 2015; Lopez-Otin etal. 2013, 2016; Singh etal. 2019). However, the focus on",
              "title": "2020 - Mitonuclear genomics and aging.pdf",
              "version": "v0",
              "chunk_order": 10,
              "document_id": "e05fdc09-c8d8-5134-a1fd-bf07a1564981",
              "extraction_id": "e6fb876b-e91c-505a-aa16-7b428ec61f10",
              "associatedQuery": "What lifestyle choices can help slow down genetic aging?"
            }
          },
          {
            "id": "a4e0cb76-8950-5471-a3c1-1ed43094fdf3",
            "score": 0.599365091286009,
            "metadata": {
              "text": "13,14\nPrior studies have identified dozens of genetic and environ -\nmental modifiers of chronological or replicative longevity, some of which are now known to function similarly to modulate life span in multicellular eukaryotes.\n15-17 One example of such a con -\nserved longevity intervention is dietary restriction, which has been shown to slow aging in many different species including yeast, nematodes, fruit flies and rodents,\n18,19 and most recently",
              "title": "2011 - A genomic analysis of chronological longevity.pdf",
              "version": "v0",
              "chunk_order": 24,
              "document_id": "a2e69cf7-8475-55f6-8fab-a572c12de9f0",
              "extraction_id": "d7daf4ea-f57a-5f7b-b6f7-afae08c35b45",
              "associatedQuery": "What lifestyle choices can help slow down genetic aging?"
            }
          },
          {
            "id": "1da274d3-c789-5af5-a8b5-72cdc9a01899",
            "score": 0.5979077398153468,
            "metadata": {
              "text": "Genetic studies have shown that aging can be slowed in\nmutants that are defective in a wide range of cellularprocesses (such as mitochondrial function, chromatin regu-\nlation, insulin signaling, tran scriptional regulation, and\ngenome stability). This indicates that aging is a complex\nprocess driven by diverse molecular pathways and biochem-\nical events. As such, a powerful approach to study aging is touse systems biology, which allows a multitude of factors",
              "title": "2004 - A Transcriptional Profile of Aging.pdf",
              "version": "v0",
              "chunk_order": 7,
              "document_id": "4ab656a7-9656-526b-94e1-422875409b44",
              "extraction_id": "b382fe8a-0267-5515-ac4b-07be55420040",
              "associatedQuery": "What lifestyle choices can help slow down genetic aging?"
            }
          },
          {
            "id": "5fc33fac-ab39-5ec1-9fb9-dcaa93a595d3",
            "score": 0.5952795921349406,
            "metadata": {
              "text": "Dietary interventions, including starvation and protein\ndeprivation, can also alter patterns of DNA methyla-\ntion, potentially in a long-lasting manner [42, 43],\nincluding transgenerationally [26, 44].\nDietary, genetic and pharmacological interventions\nthat improve health during aging and extend lifespan\ninduce long-lasting changes in gene expression that\nmediate their effects. Here we have asked if and how\nage-related DNA methylation, transcription and lipid",
              "title": "2017 - Dietary restriction protects from age-associated DNA methylation and induces epigenetic reprogramming of lipid metabolism.pdf",
              "version": "v0",
              "chunk_order": 13,
              "document_id": "fe573bb0-3d37-55e5-93fa-65b3fbc5f532",
              "extraction_id": "6364d669-4b96-5d2f-8ce8-526b065dce72",
              "associatedQuery": "What lifestyle choices can help slow down genetic aging?"
            }
          },
          {
            "id": "321d14fd-f2ae-5904-b502-dae3491cd370",
            "score": 0.5949770034856521,
            "metadata": {
              "text": "in yeast , Drosophila, and C. elegans  is able to slow aging and increase lifespan  [252-255]. Follow -up stud ies \nout of Richard Millers laboratory reproduced these findings in mice fed a diet with rapamycin incorporated  \n[256, 257]. These studies suggested that inhibiting mTOR via rapamycin could delay age-associated diseases \nand extend lifespan in mammals. A subsequent study replicated these  findings by genetically manipulating a",
              "title": "2015 - Cellular and Molecular Biology of Aging Endothelial Cells.pdf",
              "version": "v0",
              "chunk_order": 107,
              "document_id": "815d7f3e-e219-502f-aba0-57a68ae787d3",
              "extraction_id": "86f9502b-7a3a-501f-9053-8af1d37043b4",
              "associatedQuery": "What lifestyle choices can help slow down genetic aging?"
            }
          },
          {
            "id": "4c3d343d-d443-5bb4-a9ef-dd1eecaf9fac",
            "score": 0.5949385343473715,
            "metadata": {
              "text": "appears to retard aging at the molecular level as indi-cated by the gene expression analysis? Most likely,aging retardation at the molecular level by exercise isnot observed in all tissues, including some that maylimit lifespan. For example, if exercise does not reduceaging rates in replicative tissues, then it will not retardage-related tumor onset, which tends to limit maxi-mum lifespan. Another possibility relates to the obser-vation that wheel running decreased to an average 680m/day at 33 mo of age",
              "title": "2003 - Lifelong voluntary exercise in the mouse prevents.pdf",
              "version": "v0",
              "chunk_order": 83,
              "document_id": "24d4f270-f45b-5830-84f9-b1e5bcd3c070",
              "extraction_id": "e6c82594-27ba-5754-a106-69ae8b5e72ae",
              "associatedQuery": "What lifestyle choices can help slow down genetic aging?"
            }
          }
        ],
        "kg_search_results": null
      }
    }
  },
  "4": {
    "results": {
      "completion": {
        "id": "chatcmpl-AIFgqiJDPdSbdoRhIXU84YMtAnqaJ",
        "choices": [
          {
            "finish_reason": "stop",
            "index": 0,
            "message": {
              "content": "Scientists study the genetics of aging in animals using a variety of approaches and model organisms. Here are some key methods and models mentioned in the context:\n\n1. **Animal Model Systems**: Rodents, such as rats and mice, are indispensable for molecular biological studies on aging due to their practical advantages and similarities in aging processes to humans [1]. \n\n2. **Tissue-Specific Studies**: Recent genomic studies have been performed on specialized mammalian tissues, both post-mitotic (e.g., heart, nervous system) and mitotic (e.g., liver), to understand the tissue-specific effects of aging. These studies also explore how caloric restriction affects age-related transcriptional changes, which are tissue- or species-specific [2].\n\n3. **Laboratory Models**: Common laboratory models for studying aging include Caenorhabditis elegans (nematode worm) and Mus musculus (mice), which have shorter lifespans than humans, allowing for more rapid observation of aging processes [5].\n\n4. **Transgenic Mouse Strains and Interventional Studies**: These studies have identified evolutionarily conserved pathways involved in lifespan regulation and common denominators of aging across different organisms [6].\n\nThese methods and models help scientists uncover the genetic and molecular mechanisms underlying aging, providing insights that can be applied to understanding human aging and developing potential interventions.",
              "role": "assistant"
            }
          }
        ],
        "created": 1728913580,
        "model": "gpt-4o-2024-08-06",
        "object": "chat.completion",
        "system_fingerprint": "fp_a20a4ee344",
        "usage": {
          "completion_tokens": 274,
          "prompt_tokens": 1348,
          "total_tokens": 1622
        }
      },
      "search_results": {
        "vector_search_results": [
          {
            "id": "91375d45-be1d-5c54-8d0f-a9b1dded69bb",
            "score": 0.6931690573692322,
            "metadata": {
              "text": "for molecular biological studies on aging. Although material \nfrom humans should be employed where possible, for prac- \ntical reasons animal model systems like rats and mice are \nindispensible. There is evidence that, provided their health sta- \ntus and husbandry is optimal, rodents age much in the same \nway as humans do (Burek 1978). For studying certain funda- \nmental processes, such as the occurrence of various types of \nDNA rearrangement, lower organisms and cell lines can also",
              "title": "1989 - Genetic instability and aging theories, facts, and future perspectives.pdf",
              "version": "v0",
              "chunk_order": 117,
              "document_id": "4d5b1800-b676-5865-a555-09ea740cc14a",
              "extraction_id": "b0531531-f629-512b-9835-24cc870b4ef3",
              "associatedQuery": "How do scientists study the genetics of aging in animals?"
            }
          },
          {
            "id": "a32e8775-583f-5827-a590-b7058b255d26",
            "score": 0.6927486245185969,
            "metadata": {
              "text": "Until now most of the genomic studies of invertebrate\nmodels have been performed on whole animals. Several\nstudies, however, recently performed on specialized\nmammalian tissues, either post-mitotic (heart or nervous\nsystem) or mitotic (liver), show that the effects of aging\nare tissue-specific [19-25]. In addition, effects of caloric\nrestriction on age related transcriptional changes are also\ntissue- or species-specific [19].\nTo better understand the aging process in invertebrate",
              "title": "2006 - Specific age related signatures in Drosophila body parts.pdf",
              "version": "v0",
              "chunk_order": 12,
              "document_id": "24f073af-ef97-5ba3-9923-9a7d958bd411",
              "extraction_id": "efba6890-9b12-567c-b3f0-4e6ff5c6e9c4",
              "associatedQuery": "How do scientists study the genetics of aging in animals?"
            }
          },
          {
            "id": "aba78d88-b097-52fe-8246-66301e39cdd5",
            "score": 0.687142349822246,
            "metadata": {
              "text": "opportunities for assessing the efcacy of interventions onaging.\nWhen considering the advantages and disadvantages of\ndogs as a model for geroscience research, it is useful tonote that the vast majority of mammalian studies on thebasic biology of aging are performed in a relatively small\nnumber of inbred mouse strains. Typical average lifespan\nfor most of these mouse strains is approximately 23 years,",
              "title": "2016 - The dog aging project translational geroscience in companion.pdf",
              "version": "v0",
              "chunk_order": 24,
              "document_id": "e841c6bd-78b8-56e1-b3dd-e2bcc8a0f590",
              "extraction_id": "9c8bc002-4f7d-5c53-9736-70f59a6ee518",
              "associatedQuery": "How do scientists study the genetics of aging in animals?"
            }
          },
          {
            "id": "741dc9f2-2e8e-5fe3-9e6f-806a5a93213b",
            "score": 0.6835690140724182,
            "metadata": {
              "text": "[14] Gerstbrein, B., Stamatas, G., Kollias, N., Driscoll, M. In vivo  spec-\ntrofluorimetry reveals endogenous biomarkers that report health-\nspan and dietary restriction in Caenorhabditis elegans . Aging Cell  \n2005 , 4: 127-137. \n[15] Kennedy, B.K. The genetics of ageing: insight from genome-wide \napproaches in invertebrate model organisms. J. Intern. Med.  2008 , \n263: 142-152. \n[16] Kenyon, C., Chang, J., Gensch, E., Rudner, A., Tabtiang, R. A C.",
              "title": "2009 - MicroRNAs in C. elegans Aging Molecular Insurance for Robustness.pdf",
              "version": "v0",
              "chunk_order": 101,
              "document_id": "dff49223-ac74-5419-a190-a0c7f43a5ee5",
              "extraction_id": "c8d6f90d-a25c-590a-a546-4500df09aa28",
              "associatedQuery": "How do scientists study the genetics of aging in animals?"
            }
          },
          {
            "id": "0916cf4a-a863-5c5d-b687-2ae5fa80bac0",
            "score": 0.6824588775634766,
            "metadata": {
              "text": "the DNA level leads to changes in gross phenotype, we must \nnow look downstream at changes in gene expression associ -\nated with genetic variation, aging, and ARD.\nComparison With Laboratory Models of Aging\nLaboratory models typically used to study aging, such as \nCaenorhabditis  elegans  (nematode worm) and Mus musculus  \n(mice), have drastically shorter life spans than our own \n(~3 wk [ 51] and ~3 y [ 52], respectively, vs a 122 y maxi -\nmum for humans thus far; [ 53]). In some respects, these",
              "title": "2012 - Genomics and Successful Aging Grounds for Renewed.pdf",
              "version": "v0",
              "chunk_order": 47,
              "document_id": "6d2b82c3-4256-562a-9b23-ff7c71e9fd93",
              "extraction_id": "3d18e792-3d83-5cc3-b9ab-309322ecf55d",
              "associatedQuery": "How do scientists study the genetics of aging in animals?"
            }
          },
          {
            "id": "b3e0de69-763f-5f19-aeb7-ea1df79a143b",
            "score": 0.678830087184906,
            "metadata": {
              "text": "ing studies on invertebrate models of aging, long-lived mam-mals, transgenic mouse strains, and interventional studies, have led to the identification of evolutionarily conserved path-\nways involved in life span regulation, as well as common de-\nnominators of aging in different organisms.\n4 In this review, the \npathophysiological roles of these aging mechanisms, including \noxidative stress, mitochondrial dysfunction, impaired resis-",
              "title": "2018 -  Mechanisms of Vascular Aging.pdf",
              "version": "v0",
              "chunk_order": 4,
              "document_id": "659b84b6-63dd-5bb1-80ee-7478ed3c47e3",
              "extraction_id": "bfeb5c38-4fa6-5df5-90ce-63204deba3a8",
              "associatedQuery": "How do scientists study the genetics of aging in animals?"
            }
          },
          {
            "id": "e58a6718-dfef-58f6-9417-4abd793fe74d",
            "score": 0.6787930727005005,
            "metadata": {
              "text": "chain triglyceride oil on life span of genetically heterogeneous mice. J. Gerontol. A. Biol. Sci. \nMed. Sci. 68, 616 (2013). [PubMed: 22451473] \n24. Yuan R, Peters LL & Paigen B Mice as a mammalian model for research on the genetics of aging. \nILAR J. Natl. Res. Counc. Inst. Lab. Anim. Resour. 52, 415 (2011).\n25. Saul MC, Philip VM, Reinholdt LG & Chesler EJ High-diversity mouse populations for complex \ntraits. Trends Genet. 35, 501514 (2019). [PubMed: 31133439]",
              "title": "2021 - Gene-by-environment modulation of lifespan and weight gain in the murine BXD family.pdf",
              "version": "v0",
              "chunk_order": 139,
              "document_id": "4d082da4-fa48-5170-8147-c4fea47a5d4b",
              "extraction_id": "396683f9-b2e3-5942-bec8-f96fa798c341",
              "associatedQuery": "How do scientists study the genetics of aging in animals?"
            }
          },
          {
            "id": "71eb66cb-130c-5183-ba9e-038637582775",
            "score": 0.6782771348953247,
            "metadata": {
              "text": "lowing the discovery of genes and pathways involved inanimal lifespan extension, human research has focusedon the corresponding candidate human genes withgenetic, genomic and epigenetic studies into ageingand longevity. The designs of these studies differwith respect to the selection of naturally occurringphenotypes and the study populations, which includepopulation-based, patient-based, family-based andexposure-based cohorts.\nStudies into human age-related disease phenotypes",
              "title": "2011 - Genomics of human longevity.pdf",
              "version": "v0",
              "chunk_order": 11,
              "document_id": "2e038219-fdaa-506f-9cd3-51379054130e",
              "extraction_id": "89586b79-902d-5e2b-9b8a-b7a8c4971783",
              "associatedQuery": "How do scientists study the genetics of aging in animals?"
            }
          },
          {
            "id": "a0aa0b47-91a6-5f3e-b8a2-9ccdfcd79865",
            "score": 0.6779917074446865,
            "metadata": {
              "text": "Animal studies as stalking horses for human biogerontology.  For\nthe most part, studies on the biology of aging are as difficult and imprac-tical in humans as are studies of health insurance in rodents.  It is fairlyCopyright  National Academy of Sciences. All rights reserved.Cells and Surveys:  Should Biological Measures Be Included in Social Science Research?\nhttp://www.nap.edu/catalog/9995.html",
              "title": "2001 - Demography in the age of genomics.pdf",
              "version": "v0",
              "chunk_order": 1759,
              "document_id": "0f07fa43-feb6-5656-b7e7-b8faa86f5623",
              "extraction_id": "94acf45b-980d-5273-8a09-5d748c94a51b",
              "associatedQuery": "How do scientists study the genetics of aging in animals?"
            }
          },
          {
            "id": "322613d7-921b-5e2e-b410-57ab4acc4130",
            "score": 0.6763045384702373,
            "metadata": {
              "text": "review of the evidence for genotype-dependent eects on lifespan. Ageing Res.\nRev. 11, 254270. doi: 10.1016/j.arr.2011.12.006\nTurturro, A., Witt, W. W., Lewis, S., Hass, B. S., Lipman, R. D., and Hart, R. W.\n(1999). Growth curves and survival characteristics of the animals used in the\nbiomarkers of aging program. J. Gerontol. Ser. Biol. Sci. Med. Sci 54, B492B501.\ndoi: 10.1093/gerona/54.11.b492\nVertti-Quintero, N., Berger, S., Solvas, X. C. I, Statzer, C., Annis, J., Ruppen,",
              "title": "2021 - Lifespan-Associated Gene Expression Signatures of Recombinant BXD Mice Implicates Coro7 and Set in Longevity.pdf",
              "version": "v0",
              "chunk_order": 106,
              "document_id": "6277f22c-f56d-51a7-add1-1fe7674dda74",
              "extraction_id": "e3eb627c-15f4-5713-92a4-e92a891b7136",
              "associatedQuery": "How do scientists study the genetics of aging in animals?"
            }
          }
        ],
        "kg_search_results": null
      }
    }
  },
  "5": {
    "results": {
      "completion": {
        "id": "chatcmpl-AIFgxEMof4wOrO9UyQT5bEPH8q8fk",
        "choices": [
          {
            "finish_reason": "stop",
            "index": 0,
            "message": {
              "content": "Yes, specific genes have been linked to longer lifespans. Research has identified more than 700 genes that regulate lifespan in model organisms, with many of these genes and their associated pathways, such as the insulin/IGF1/GH pathway, shown to affect longevity across different model organisms [2]. Additionally, variants near genes such as CHRNA3/5 and APOE have been found to have age- and sex-related effects on human lifespan [7]. Genome-wide association studies have also identified loci associated with exceptional human longevity [10].",
              "role": "assistant"
            }
          }
        ],
        "created": 1728913587,
        "model": "gpt-4o-2024-08-06",
        "object": "chat.completion",
        "system_fingerprint": "fp_6b68a8204b",
        "usage": {
          "completion_tokens": 109,
          "prompt_tokens": 1456,
          "total_tokens": 1565
        }
      },
      "search_results": {
        "vector_search_results": [
          {
            "id": "9defe0af-80a1-56da-90df-551fd55baa13",
            "score": 0.7708674209792973,
            "metadata": {
              "text": "genes analyzed for their possible association with human lon-gevity (http://genomics.senescence.info/genes/longevity.html).All longevity association studies in humans we could find by thetime of the latest update were added to this list. These includestudies reporting negative results, which we see as essentialsince many genes display population-specific associations withlongevity.\nFig. 1 From the main page of the Human Ageing",
              "title": "2009 - The Human Ageing Genomic Resources online.pdf",
              "version": "v0",
              "chunk_order": 20,
              "document_id": "e43cd3b6-ad8e-5422-ba7c-ceb6e66cc529",
              "extraction_id": "52c67b46-63f2-54ae-a78e-e9d54a55f6e4",
              "associatedQuery": "Are there specific genes that have been linked to longer lifespans?"
            }
          },
          {
            "id": "4d6876c5-9226-587c-8d3e-d4957ee42dba",
            "score": 0.7568767215683043,
            "metadata": {
              "text": "genes (http://genomics.senescence.info/genes/), more than700 genes have been identified that regulate lifespan inmodel organisms (de Magalha es et al., 2009a). Many ofthese genes and their associated pathwayssuch as theinsulin/IGF1/GH pathwayhave been shown to affect lon-gevity across different model organisms (Kenyon, 2010).Therefore, at least some mechanisms of aging are evolu-tionarily conserved and may have potential therapeuticapplications (Baur et al., 2006). For example, evidencesuggests the use of",
              "title": "2012 - Genome-Environment Interactions That Modulate.pdf",
              "version": "v0",
              "chunk_order": 65,
              "document_id": "b1a1997c-e9df-5dc0-9d12-a3977d0c64ec",
              "extraction_id": "a01ca925-4ccf-5863-a162-7bd4c754fe89",
              "associatedQuery": "Are there specific genes that have been linked to longer lifespans?"
            }
          },
          {
            "id": "2f28f34e-bf5e-57d6-8a8c-dd946f574906",
            "score": 0.7510410844038624,
            "metadata": {
              "text": "Exceptional Longevity\nOne approach to identifying genes associated with low mortality is to\nexamine the genes of those who survive to the oldest ages.  Several studieshave examined gene frequencies among centenarians or nonagenariansand compared them with frequencies at younger ages.  Since changes ingene frequencies are more rapid when mortality rates are high, cross-sectional comparisons must be adjusted for differences in mortality amongcohorts.",
              "title": "2001 - Demography in the age of genomics.pdf",
              "version": "v0",
              "chunk_order": 855,
              "document_id": "0f07fa43-feb6-5656-b7e7-b8faa86f5623",
              "extraction_id": "0e71cb8b-4647-56bb-9b5f-d14d0ea2ff50",
              "associatedQuery": "Are there specific genes that have been linked to longer lifespans?"
            }
          },
          {
            "id": "b3e21ac9-8df8-5119-a769-a9da82db78da",
            "score": 0.7497157156806331,
            "metadata": {
              "text": "informed by age-related disease identifies loci for exceptional human longevity. Li H, editor. \nPLoS Genet. 2015. https://doi.org/10.1371/journal.pgen.\n 15. Polderman TJC, Benyamin B, de Leeuw CA, Sullivan PF, van Bochoven A, Visscher PM, \netal. Meta-analysis of the heritability of human traits based on fifty years of twin studies. Nat \nGenet. 2015;47:7029.\n 16. Cellerino A, Ori A.What have we learned on aging from omics studies? Semin Cell Dev Biol. \n2017;70:17789.",
              "title": "2020 - Clinical Genetics and Genomics of Aging.pdf",
              "version": "v0",
              "chunk_order": 167,
              "document_id": "62b635c3-040e-512a-b016-6ef295308a1e",
              "extraction_id": "ead14808-bfb7-5e32-9830-28efaae71151",
              "associatedQuery": "Are there specific genes that have been linked to longer lifespans?"
            }
          },
          {
            "id": "c2234f77-2268-57d0-a227-e931fc4802c1",
            "score": 0.7427925178059914,
            "metadata": {
              "text": "GENOME-WIDE ASSOCIATION STUDY OF LONGEVITY 479\nINCREASES in longevity of the general population world -\nwide are an unprecedented phenomenon with significant \nhealth and social impact. Although environmental factors \nhave led to an increase in life span, there is ample evidence \nthat genetic factors are involved in extreme longevity both \nin humans (17) and in other organisms (8). The protective \ngenetic factors that lead to longevity are likely to involve",
              "title": "2010 - A Meta-analysis of Four Genome-Wide Association Studies.pdf",
              "version": "v0",
              "chunk_order": 12,
              "document_id": "8e452186-a71c-5b62-81b2-7681c87c8e1d",
              "extraction_id": "8bc54e5b-f45f-54f9-9591-1e26dd80b50d",
              "associatedQuery": "Are there specific genes that have been linked to longer lifespans?"
            }
          },
          {
            "id": "cc78a209-081f-5e0c-877a-36b85d5105a6",
            "score": 0.7383499897509832,
            "metadata": {
              "text": "expression of certain genes have an effect upon longevity.\nAlthough similar aging processes are likely to operateacross multiple species [30], it has been much more diffi-cult to identify longevity candidate genes in human studies[30]. A key question in human aging is to what extent asignature of aging may be detectable across tissues. Until\nnow there has been a lack of large transcriptional profiles\nfrom the same human individuals in multiple tissues. TheMuTHER study provides ins ight into the human aging",
              "title": "2013 - Gene expression changes with age in skin.pdf",
              "version": "v0",
              "chunk_order": 49,
              "document_id": "5c121bbb-57b8-51cc-8461-effa1bfd87b9",
              "extraction_id": "18e9f96b-9af8-59bf-9323-084df9a4f9b6",
              "associatedQuery": "Are there specific genes that have been linked to longer lifespans?"
            }
          },
          {
            "id": "726417dd-f626-5197-966d-6a6ad25ff718",
            "score": 0.7361661949058668,
            "metadata": {
              "text": "4. Joshi, P. K. et al. Variants near CHRNA3/5 and APOE have age- and sex-\nrelated effects on human lifespan. Nat. Commun. 7, 11174 (2016).\n5. Pilling, L. C. et al. Human longevity is in uenced by many genetic variants:\nevidence from 75,000 UK Biobank participants. Aging 8, 547560 (2016).\n6. Deelen, J. et al. Genome-wide association meta-analysis of human longevity\nidenti es a novel locus conferring survival beyond 90 years of age. Hum. Mol.\nGenet. 23, 4420 4432 (2014).",
              "title": "2017 - Genome-wide meta-analysis associates HLA.pdf",
              "version": "v0",
              "chunk_order": 123,
              "document_id": "3a565ba9-ee5b-5596-b870-ce8c055cb1f1",
              "extraction_id": "c302507d-508e-5093-a877-1cec20f8440e",
              "associatedQuery": "Are there specific genes that have been linked to longer lifespans?"
            }
          },
          {
            "id": "300f0303-caec-52b9-852b-8e67cec5d326",
            "score": 0.7353308954637722,
            "metadata": {
              "text": "79-91. \n[97] Smith, E.D.; Kennedy, B.K.; Kaeberlein, M. Genome-wide \nidentification of conserved longevity genes in yeast and worms . \nMech. Ageing Dev. , 2007 , 128(1), 106-11. \n[98] Chen, D.; Pan, K.Z.; Palter, J.E.; Kapahi, P. Longevity determined \nby developmental arrest genes in Caenorhabditis elegans.  Aging \nCell, 2007 , 6(4), 525-33. \n[99] Curran, S.P.; Ruvkun, G. Lifespan regulation by evolutionarily \nconserved genes essential for viability . PLoS Genet. , 2007 , 3(4), \ne56.",
              "title": "2012 - Genome-Wide RNAi Longevity Screens in Caenorhabditis elegans.pdf",
              "version": "v0",
              "chunk_order": 160,
              "document_id": "7589fec2-e893-5a4d-9f0c-09abb35858ab",
              "extraction_id": "f160f818-03bf-5b4e-b1f4-bfbd3b0bfb99",
              "associatedQuery": "Are there specific genes that have been linked to longer lifespans?"
            }
          },
          {
            "id": "025a94a9-595e-56f6-8c03-89ccea15a22c",
            "score": 0.7336123148433987,
            "metadata": {
              "text": "9. vB Hjelmborg J, Iachine I, Skytthe A, Vaupel JW, McGue M, et al. (2006)\nGenetic influence on human lifespan and longevity. Hum Genet 119: 312321.doi:10.1007/s00439-006-0144-y.\n10. Sebastiani P, Perls TT (2012) The genetics of extreme longevity: lessons from the\nnew England centenarian study. Front Genet 3: 277. doi:10.3389/fgene.2012.00277.11. Perls TT, Wilmoth J, Levenson R, Drinkwater M, Cohen M, et al. (2002) Life-",
              "title": "2014 - Whole-Genome Sequencing of the World?s Oldest People.pdf",
              "version": "v0",
              "chunk_order": 110,
              "document_id": "d2a5ec28-873a-5ff3-9cf4-dbec3b52dd21",
              "extraction_id": "eed176e2-226d-5638-b10c-4813cfbbd213",
              "associatedQuery": "Are there specific genes that have been linked to longer lifespans?"
            }
          },
          {
            "id": "68e705e1-54a1-578a-98ee-0c76b02ccf79",
            "score": 0.7321038405833352,
            "metadata": {
              "text": "39. Fortney K, Dobriban E, Garagnani P, etal. Genome-wide scan informed by \nage-related disease identifies loci for exceptional human longevity. PLoS Genet. 2015;11:e1005728. doi:10.1371/journal.pgen.1005728\n 40. Beekman M, Nederstigt C, Suchiman HE, et al. Genome-wide asso-\nciation study (GWAS)-identified disease risk alleles do not compromise \nhuman longevity. Proc Natl Acad Sci U S A. 2010;107:1804618049. \ndoi:10.1073/pnas.1003540107",
              "title": "2017 - Four Genome-Wide Association Studies Identify New.pdf",
              "version": "v0",
              "chunk_order": 154,
              "document_id": "c10653f6-b3d7-5b92-9271-ab8fcc7905a7",
              "extraction_id": "52557433-1cf1-5441-b44d-361963a61eca",
              "associatedQuery": "Are there specific genes that have been linked to longer lifespans?"
            }
          }
        ],
        "kg_search_results": null
      }
    }
  },
  "6": {
    "results": {
      "completion": {
        "id": "chatcmpl-AIFh26X5nul0obtiAeqSkHmHNgJoq",
        "choices": [
          {
            "finish_reason": "stop",
            "index": 0,
            "message": {
              "content": "Telomeres affect the aging process in several ways:\n\n1. **Telomere Shortening**: Telomeres are specialized structures that protect the ends of linear chromosomes. They shorten during aging due to the unidirectional activity of DNA polymerase, which leaves a section of DNA unreplicated on the lagging strand [1]. This shortening is a natural part of the aging process and is exacerbated by genotoxic stress, such as oxidative damage [1].\n\n2. **Cellular Senescence and Apoptosis**: The shortening of telomeres can induce processes such as apoptosis (programmed cell death) and cellular senescence (a state where cells stop dividing), which affect the health and lifespan of an individual [2]. When telomeres become critically short, they trigger a DNA damage response, leading to cellular senescence or apoptosis [4].\n\n3. **Genome Stability**: Telomeres ensure the stability of the genome and protect chromosomes from incorrect actions by the DNA repair machinery [3]. When telomeres are too short, they can no longer form protective structures, leading to genome instability and potentially contributing to aging [8].\n\n4. **Cancer Prevention**: Short telomeres limit the number of cell cycles, which is important for preventing the onset of cancer. However, this also contributes to the aging process as cells enter a state of permanent cell cycle arrest (senescence) [7].\n\n5. **Telomerase Activity**: The enzyme telomerase can maintain telomere length, but its activity varies over the lifespan and between cell types, tissues, and species [1]. In most human somatic cells, telomerase activity is limited, which contributes to telomere shortening and aging [4].\n\nOverall, telomere shortening acts as a biological clock that limits cellular replication, contributing to aging and age-related diseases [6].",
              "role": "assistant"
            }
          }
        ],
        "created": 1728913592,
        "model": "gpt-4o-2024-08-06",
        "object": "chat.completion",
        "system_fingerprint": "fp_a20a4ee344",
        "usage": {
          "completion_tokens": 378,
          "prompt_tokens": 1222,
          "total_tokens": 1600
        }
      },
      "search_results": {
        "vector_search_results": [
          {
            "id": "53508a9e-d064-58a3-a4f9-0785470a1462",
            "score": 0.7356500625610352,
            "metadata": {
              "text": "Telomeres are specialized structures that protect the ends of linear chromosomes. They shorten during aging due to the unidirectional activity of DNA polymerase, which leaves a section of DNA unrepli-cated on the lagging strand. Telomeres also are subject to shortening by genotoxic stress, such as oxidative damage (33). Among many eukaryotes, the enzyme telomerase maintains telomere length; but telomerase activity varies over the lifespan and between cell types, tissues, and species (34). In most human",
              "title": "2018 - Sex Differences in Aging Genomic Instability.pdf",
              "version": "v0",
              "chunk_order": 46,
              "document_id": "8cfb5529-7f0c-58fc-b6e4-b3ee800fb72f",
              "extraction_id": "396708f1-aa0a-571e-a8d3-7cb8404e9502",
              "associatedQuery": "How do telomeres affect the aging process?"
            }
          },
          {
            "id": "b532d055-ab02-5326-8eb4-67e7277a92b8",
            "score": 0.73097825050354,
            "metadata": {
              "text": "that shorten their length with progressing age. This shortening of telomeres is the \nresult of the absence of the activity of an enzyme called telomerase, and in turn it \ninduces several processes, such as apoptosis, senescence, or oncogenic transforma-\ntion of somatic cells, affecting the health and lifespan of an individual [42]. Human \ntelomere shortening has been mostly studied in leukocytes and linked not only to \nageing and life expectancy [43] but also to age-related diseases, including cardio-",
              "title": "2020 - Clinical Genetics and Genomics of Aging.pdf",
              "version": "v0",
              "chunk_order": 1721,
              "document_id": "62b635c3-040e-512a-b016-6ef295308a1e",
              "extraction_id": "41b98643-1948-519b-8b27-ab0fa4041048",
              "associatedQuery": "How do telomeres affect the aging process?"
            }
          },
          {
            "id": "65fb74aa-f3c3-5c80-919f-329169db982f",
            "score": 0.7253162264823914,
            "metadata": {
              "text": "nization may directly affect telomere attrition, resulting in accelerated replicative \nsenescence and progeroid phenotypes [180].\nTelomeres are regions constituted by tandem repeats of non-coding DNA \nsequences 5-(TTAGGG)n-3 and a protein complex called shelterin, bound to them. \nThis structure ensures the stability of the genome and protects the chromosomes \nfrom a wrong action of the DNA repair machinery [184] by allowing the formation \nof a chromatin loop called T-Loop [185].",
              "title": "2020 - Clinical Genetics and Genomics of Aging.pdf",
              "version": "v0",
              "chunk_order": 1084,
              "document_id": "62b635c3-040e-512a-b016-6ef295308a1e",
              "extraction_id": "d4afa45a-5efa-577b-822e-7a82c2f6508d",
              "associatedQuery": "How do telomeres affect the aging process?"
            }
          },
          {
            "id": "ab6a6bda-490d-5b7e-a715-3b9b4f89243f",
            "score": 0.7144376252424013,
            "metadata": {
              "text": "Telomeres play a central role in cell fate and aging by adjusting the cellular response to stress and growth stimulation on thebasis of previous cell divisions and DNA damage. At least a few hundred nucleotides of telomere repeats must cap eachchromosome end to avoid activation of DNA repair pathways. Repair of critically short or uncapped telomeres by telomeraseor recombination is limited in most somatic cells and apoptosis or cellular senescence is triggered when too many uncappedtelomeres accumulate.",
              "title": "2008 - Telomeres and Aging.pdf",
              "version": "v0",
              "chunk_order": 2,
              "document_id": "61d9c326-d36e-55c1-a891-335dc943e70f",
              "extraction_id": "55fd2e43-f58e-5d89-8730-7d82d3b6c44f",
              "associatedQuery": "How do telomeres affect the aging process?"
            }
          },
          {
            "id": "80a2162f-6208-5f97-a646-e8803d501f4e",
            "score": 0.7132883413686396,
            "metadata": {
              "text": "ing (84). This process is believed to be the trigger for the aging \nprocess, according to the telomere theory (11, 85, 86). It is further \nsupported by Bodnar etal. who proved that telomere elongation caused by ectopic expression of telomerase avoids the senescence phenotype (87). His work relied on one of the earliest studies \nlinking telomere shortening to aging which was performed",
              "title": "2018 - Genomic Instabilities, Cellular Senescence, and Aging In Vitro, In Vivo and Aging-Like Human Syndromes.pdf",
              "version": "v0",
              "chunk_order": 77,
              "document_id": "7de8d462-8a3c-5625-8cbb-374f3bb46425",
              "extraction_id": "016d8de2-949f-511e-a9e1-d2d5fd2bede5",
              "associatedQuery": "How do telomeres affect the aging process?"
            }
          },
          {
            "id": "f181e6da-58b6-5f26-87a2-355e25388673",
            "score": 0.7043320536613464,
            "metadata": {
              "text": "telomeres, the repetitive sequence at the end of linear chromosomes, has garnered much attention for\nits relation to aging. Telomere repeats serve as an internal clock for cycling cells because each round of\nreplication results in the loss of telomeric DNA in the absence of active telomerase (reviewed in [66]).\nEventually, this loss over cellular generations culminates in telomere crisis and a permanent state of",
              "title": "2018 - Repetitive Fragile Sites Centromere Satellite DNA.pdf",
              "version": "v0",
              "chunk_order": 35,
              "document_id": "262df0d6-ad68-544a-88ed-b4568f305858",
              "extraction_id": "3b0cb0ab-421d-54d7-9816-c6a2e6f1ac68",
              "associatedQuery": "How do telomeres affect the aging process?"
            }
          },
          {
            "id": "6d0cccc5-3ed7-507e-9f7a-6035badacc00",
            "score": 0.6978059771292286,
            "metadata": {
              "text": "and consequently lose telomeric sequences, thereby limiting the number of cell cycles, which is\nimportant for preventing the onset of cancer. Cells perceive critically short telomeres as persistentDNA damage. This activates the DNA damage responses, including cell cycle checkpoints, which\nultimately leads to a permanent cell cycle arrest (cellular senescence). Senescence protects from\ncancer but contributes to the aging process (37).",
              "title": "2016 - Genome Integrity in Aging.pdf",
              "version": "v0",
              "chunk_order": 39,
              "document_id": "85d5fcbb-5385-5a01-8139-d11fc8b1fe3a",
              "extraction_id": "5179130e-5fa6-5979-ba68-270e546e43d7",
              "associatedQuery": "How do telomeres affect the aging process?"
            }
          },
          {
            "id": "72b978c7-44fc-530d-a1d2-eaffaf2c8782",
            "score": 0.6957833585610019,
            "metadata": {
              "text": "When the telomeres shorten, this loop is no longer able to form \nand in turn, the epigenetic regulation is changed to activation of the TPE-OLD genes. This happens before the telomeres reach \nthe critical length that causes activation of DDR, thus leading to \nanother earlier possible effect of telomere shortening on aging (138, 139). Interestingly, a following study by Kim etal. showed \nthat one of the TPE-OLD sensitive genes is hTERT, the core reverse transcriptase component of telomerase (140). This is",
              "title": "2018 - Genomic Instabilities, Cellular Senescence, and Aging In Vitro, In Vivo and Aging-Like Human Syndromes.pdf",
              "version": "v0",
              "chunk_order": 104,
              "document_id": "7de8d462-8a3c-5625-8cbb-374f3bb46425",
              "extraction_id": "9fafad4c-f208-53e0-b2ac-f10569429a5e",
              "associatedQuery": "How do telomeres affect the aging process?"
            }
          },
          {
            "id": "0faa4fb9-efa7-5e92-8fe4-5e28c51dbee4",
            "score": 0.694516658782959,
            "metadata": {
              "text": "to maintain proliferation potential (94). Cells with mutated telomerase exhibited irregular morphology and short telomeres, but these changes did not cause deadly damage and determinate senescence (95). One hypothesis connects aging to telomere \nerosion through the transcription of subtelomeric genes. Genes \nlocated in subtelomeric regions are affected by transcriptional silencing which was found to change in an age-related manner. \nKim et al. (96) found that silencing of genes in subtelomeric",
              "title": "2018 - Genomic Instabilities, Cellular Senescence, and Aging In Vitro, In Vivo and Aging-Like Human Syndromes.pdf",
              "version": "v0",
              "chunk_order": 84,
              "document_id": "7de8d462-8a3c-5625-8cbb-374f3bb46425",
              "extraction_id": "016d8de2-949f-511e-a9e1-d2d5fd2bede5",
              "associatedQuery": "How do telomeres affect the aging process?"
            }
          },
          {
            "id": "b1383516-a23e-5048-9cf3-944b5142e16b",
            "score": 0.6876281108745372,
            "metadata": {
              "text": "evidence implicates telomere shortening in cellularsenescence. Telomeres consist of repetitive nucleotides e q u e n c e s( T T A G G G )a tt h ee n d so fm a m m a l i a nc h r o -mosomes, that preserve chromosome stability andintegrity by preventing deterioration or fusion withneighboring chromosomes (76) (Central Illustration ).JACC VOL. 69, NO. 15, 2017 Paneni et al .\nAPRIL 18, 2017:1952 67 The Aging Cardiovascular System1957",
              "title": "2017 - The Aging Cardiovascular System.pdf",
              "version": "v0",
              "chunk_order": 60,
              "document_id": "d3ff8471-986b-5fa0-b9c4-96eaaa8fce7c",
              "extraction_id": "82798504-5de9-513c-b3df-09968387cd42",
              "associatedQuery": "How do telomeres affect the aging process?"
            }
          }
        ],
        "kg_search_results": null
      }
    }
  },
  "7": {
    "results": {
      "completion": {
        "id": "chatcmpl-AIFhETjzplVDZqcInKYA6bobssz1r",
        "choices": [
          {
            "finish_reason": "stop",
            "index": 0,
            "message": {
              "content": "DNA repair plays a significant role in aging by maintaining the integrity and stability of the nuclear genome. Impairment of DNA repair mechanisms can result in accelerated aging and/or cancer [2]. As organisms age, endogenous sources of genotoxins increase, DNA repair capacity declines, and levels of DNA damage and mutations increase [2]. This accumulation of DNA damage is associated with aging phenotypes, as DNA damage can activate cellular responses that contribute to aging [6]. The DNA damage theory of aging suggests that genomic instability, caused by accumulated DNA damage, plays a causal role in aging [5]. Additionally, the burden of DNA lesions is greater in older mammals compared to younger ones, indicating that DNA repair is crucial for mitigating the effects of aging [5].",
              "role": "assistant"
            }
          }
        ],
        "created": 1728913604,
        "model": "gpt-4o-2024-08-06",
        "object": "chat.completion",
        "system_fingerprint": "fp_6b68a8204b",
        "usage": {
          "completion_tokens": 150,
          "prompt_tokens": 1179,
          "total_tokens": 1329
        }
      },
      "search_results": {
        "vector_search_results": [
          {
            "id": "566bd0c9-262e-543e-8934-1af5fa9edef5",
            "score": 0.7609542096514323,
            "metadata": {
              "text": "Effect of age on DNA repair\nResearch over the past decades suggest that many steps\nin DNA metabolism are altered with age in a variety\nof tissues and animal models (56,57). The relation of DNArepair to aging has been studied by measuring the ability\nof cells from organisms of various life spans to repair\nDNA damage and by experiments that have comparedthe ability of cells from young and old organisms to repair\nDNA damage. Interest was peaked by the original",
              "title": "2007 - Caloric restriction and genomic stability.pdf",
              "version": "v0",
              "chunk_order": 40,
              "document_id": "76c08863-1522-519b-8da6-65a872418fee",
              "extraction_id": "a563be97-fd42-50ba-8a26-3e1ca3b738db",
              "associatedQuery": "What role does DNA repair play in aging?"
            }
          },
          {
            "id": "b8c3720d-f697-5d2f-9728-49b7489d6509",
            "score": 0.7479147911071777,
            "metadata": {
              "text": "BI87CH14_Niedernhofer ARI 18 May 2018 15:1\nSUMMARY POINTS\n1. Evolutionarily conserved DNA repair pathways maintain the integrity and stability of\nthe nuclear genome. Impairment of DNA repair mechanisms results in accelerated agingand/or cancer.\n2. Evidence in humans and model organisms supports the conclusions that with age\n(a) endogenous sources of genotoxins increase, ( b) DNA repair capacity declines, and\n(c) levels of DNA damage and mutations increase.",
              "title": "2018 - Nuclear Genomic Instability.pdf",
              "version": "v0",
              "chunk_order": 180,
              "document_id": "54d28a91-8db6-56b1-baaa-b67274c93a36",
              "extraction_id": "44047f31-85e4-587c-ba58-8c3494fb7d52",
              "associatedQuery": "What role does DNA repair play in aging?"
            }
          },
          {
            "id": "9180d1c5-31b6-533e-bf2e-4b367dc2097d",
            "score": 0.7460238488898381,
            "metadata": {
              "text": "Several lines of evidence suggest that DNA repair capacity might decrease with age. However,it should be noted that measuring DNA repair in tissues is challenging and that the validity ofsurrogate markers of repair capacity is not well established. For example, a reduction in expression\nof DNA repair genes/proteins is not proven to impact DNA repair. Frequently, the reduction in",
              "title": "2018 - Nuclear Genomic Instability.pdf",
              "version": "v0",
              "chunk_order": 75,
              "document_id": "54d28a91-8db6-56b1-baaa-b67274c93a36",
              "extraction_id": "e3e52327-4a23-5003-b418-dafdcdcae82c",
              "associatedQuery": "What role does DNA repair play in aging?"
            }
          },
          {
            "id": "ca253ce9-4661-5ca2-bf17-3a86ef3eff1d",
            "score": 0.7436333894729614,
            "metadata": {
              "text": "improved DNA repair. Finally, there should be a plausible mechanism by which DNA damage\ncan drive aging. Here, we review the evidence currently supporting each of these predictions.\nEVIDENCE THAT DNA DAMAGE INCREASES WITH AGE\nSources of Damage Increase with Age\nThe free radical theory of aging posits that aging is caused primarily by oxidative damage in-\ncurred by ROS that chemically modify critical cellular biomolecules (13). This theory has evolved",
              "title": "2018 - Nuclear Genomic Instability.pdf",
              "version": "v0",
              "chunk_order": 46,
              "document_id": "54d28a91-8db6-56b1-baaa-b67274c93a36",
              "extraction_id": "b934a2a9-a672-5d65-9d0d-bbc36652a148",
              "associatedQuery": "What role does DNA repair play in aging?"
            }
          },
          {
            "id": "494f865d-a7b6-5978-9b02-d5e628952a9d",
            "score": 0.7432990823209846,
            "metadata": {
              "text": "All rights reservedKeywords\nDNA damage, aging, mutations, senescence, DNA damage response, DNA\nrepair\nAbstract\nThe nuclear genome decays as organisms age. Numerous studies demon-\nstrate that the burden of several classes of DNA lesions is greater in older\nmammals than in young mammals. More challenging is proving this is acause rather than a consequence of aging. The DNA damage theory of\naging, which argues that genomic instability plays a causal role in aging,",
              "title": "2018 - Nuclear Genomic Instability.pdf",
              "version": "v0",
              "chunk_order": 2,
              "document_id": "54d28a91-8db6-56b1-baaa-b67274c93a36",
              "extraction_id": "2b406c50-28e1-5b8c-a39d-a26db15f8aaa",
              "associatedQuery": "What role does DNA repair play in aging?"
            }
          },
          {
            "id": "a1370bf9-13f2-5c98-9d9d-9dfead21ebd7",
            "score": 0.7399291461642843,
            "metadata": {
              "text": "repaired; otherwise the genome would soon become saturated with\ndamage and life would cease. There is some evidence that DNA damage\naccumulates with age in some tissues ( Maslov et al., 2013 ), but the exact\nnature of the damage remains unclear. Indeed, even these low levels of\nspontaneous DNA damage may represent a steady state due to continu-\nous repair and induction of new damage. However, DNA damage can\ncause certain aging phenotypes by activating cellular responses, such",
              "title": "2017 - Mutation and catastrophe in the aging genome.pdf",
              "version": "v0",
              "chunk_order": 25,
              "document_id": "7ae205a2-e002-5e8b-bbf1-ea96ab599b37",
              "extraction_id": "eb91e436-a1bb-5d10-b648-07224b9e5bff",
              "associatedQuery": "What role does DNA repair play in aging?"
            }
          },
          {
            "id": "8d2bc107-4d94-5dd8-8f67-b593aecc0478",
            "score": 0.7375595789420483,
            "metadata": {
              "text": "36:1049-1062.\n66. Hasty P, Vijg J: Accelerating aging by mouse reverse genetics:\na rational approach to understanding longevity.   Aging Cell\n2004, 3:55-65.\n67. Bohr VA: Deficient DNA repair in the human progeroid dis-\norder, Werner syndrome.   Mutat Res  2005, 577:252-259.\n68. Nouspikel T, Hanawalt PC: DNA repair in term inally differenti-\nated cells.   DNA Repair  2002, 1:59-75.\n69. Nouspikel T, Hanawalt PC: When parsimony backfires: neglect-\ning DNA repair may doom neurons in Alzheimer's disease.",
              "title": "2007 - Decline of nucleotide excision repair capacity in aging.pdf",
              "version": "v0",
              "chunk_order": 201,
              "document_id": "c9bb2ba2-a001-5c1b-8be8-d1c184924362",
              "extraction_id": "a0e59df7-6a34-5f03-af2e-82bdc0edacb9",
              "associatedQuery": "What role does DNA repair play in aging?"
            }
          },
          {
            "id": "4db748ed-7063-50e5-b42c-cb6fa3ecd9a2",
            "score": 0.7375421524047852,
            "metadata": {
              "text": "DNA repair. In the latterdifficult to arrive at a strict, experimentally useful defini-context, most premature aging syndromes are causedtion of aging. Factors implicated in organismal declineby mutations in genes encoding proteins involved inin genetic models might not play a role in the normalDNA repair ( Karanjawala and Lieber, 2004 ). Accumula-aging processes. A related difficulty is that prematuretion of mutations in critical genes may be one generalaging models fail to recapitulate all aspects of",
              "title": "2005 - DNA Repair, Genome Stability.pdf",
              "version": "v0",
              "chunk_order": 136,
              "document_id": "e658e73b-2494-5fa3-ae39-9f4933bc037b",
              "extraction_id": "5ea2fb27-ddd7-50b4-b318-39ca71f1c7e2",
              "associatedQuery": "What role does DNA repair play in aging?"
            }
          },
          {
            "id": "4521b426-a67e-51e4-bc63-b6da5fab60cf",
            "score": 0.7356736502452893,
            "metadata": {
              "text": "escape the repair process and accumulate in the genome, impacting several processes and\naging [67,145147].\nThere is little evidence of association between DNA repair improvement and life-\ntime expansion [ 148,149], thus, indicating that such mechanism seems to have evolved\nto maintain DNA stabilityand therefore healthonly until reproductive age, without\nany regard for the fate of the individual in old age, both in terms of quality and length of",
              "title": "2023 - Genomic Instability Evolutionary Footprints on Human Health.pdf",
              "version": "v0",
              "chunk_order": 101,
              "document_id": "59dec4a5-f80a-5a82-b55a-b6b1b33b907f",
              "extraction_id": "57e201b2-a357-5cff-9555-49955299669e",
              "associatedQuery": "What role does DNA repair play in aging?"
            }
          },
          {
            "id": "4c627903-8a25-5db0-8a60-1850a924a27b",
            "score": 0.7347921449193834,
            "metadata": {
              "text": "with age, and DNA repairtween different tissues. These differences likely reflectdefects can cause phenotypes resembling prematurefunctional characteristics of those tissues, such as mi-aging. We discuss how cellular DNA damage re-totic rate, transcriptional activity, metabolism, and thesponses may contribute to manifestations of aging.action of specific DNA repair systems.We review Sir2, a factor linking genomic stability, me-Reactive Oxygen Species: An Important Sourcetabolism, and aging. We conclude",
              "title": "2005 - DNA Repair, Genome Stability.pdf",
              "version": "v0",
              "chunk_order": 4,
              "document_id": "e658e73b-2494-5fa3-ae39-9f4933bc037b",
              "extraction_id": "67128b6e-9bd6-53fe-b1e7-d0721db8619d",
              "associatedQuery": "What role does DNA repair play in aging?"
            }
          }
        ],
        "kg_search_results": null
      }
    }
  },
  "8": {
    "results": {
      "completion": {
        "id": "chatcmpl-AIFhM7HonwMIv1KCdMHKw9gGzAYlV",
        "choices": [
          {
            "finish_reason": "stop",
            "index": 0,
            "message": {
              "content": "Yes, genetic research can potentially lead to treatments that slow down aging. Several pieces of evidence from the context support this possibility:\n\n1. The discovery of genetic markers for slow aging in humans suggests that understanding these genes could pave the way for therapeutic interventions for age-related maladies, including cancers, neurodegeneration, and metabolic syndrome [4].\n\n2. Research indicates that manipulating aging-related genes through various means, such as diet, lifestyle, and pharmaceuticals, could dramatically improve human health and lead to the development of drugs against age-related diseases [7].\n\n3. Advances in molecular biology, such as CRISPR/Cas9, are expected to clarify aging processes and identify new potential therapeutic targets, which could be crucial for developing treatments that slow aging [6].\n\n4. The use of senolytic drugs, which target senescent cells, has shown promise in halting biological aging in mice, and trials are underway to test their effectiveness in humans [3].\n\n5. There is a suggestion that interventions targeting DNA methylation and other genetic modifications could prevent age-related diseases and promote longevity, highlighting the potential of genetic research in developing therapeutic strategies against aging [10].\n\nOverall, while the research is still ongoing and some findings are speculative, there is significant potential for genetic research to contribute to treatments that slow down the aging process.",
              "role": "assistant"
            }
          }
        ],
        "created": 1728913612,
        "model": "gpt-4o-2024-08-06",
        "object": "chat.completion",
        "system_fingerprint": "fp_6b68a8204b",
        "usage": {
          "completion_tokens": 263,
          "prompt_tokens": 1182,
          "total_tokens": 1445
        }
      },
      "search_results": {
        "vector_search_results": [
          {
            "id": "9da7c5dc-0deb-577c-bb22-83f987bd76dd",
            "score": 0.7105742269412156,
            "metadata": {
              "text": "raises the possibility of therapies to slow aging.  Therefore the discoveryof a gerontogene with even very rare mutations that increased longevitywould cause speculation about future trends in mortality.    However, thediscovery of such a gene would be relevant only to long-term (and, there-fore, very speculative) projections.\nProspective Epidemiologic Surveys that Include Genetic Information\nSome epidemiologic cohort studies of populations have collected",
              "title": "2001 - Demography in the age of genomics.pdf",
              "version": "v0",
              "chunk_order": 784,
              "document_id": "0f07fa43-feb6-5656-b7e7-b8faa86f5623",
              "extraction_id": "e3014138-3d5b-58bc-a1a5-5ac6f04cac1c",
              "associatedQuery": "Can genetic research lead to treatments that slow down aging?"
            }
          },
          {
            "id": "78733c6a-d870-5154-9128-eb66291fa967",
            "score": 0.7009225248190731,
            "metadata": {
              "text": "need to develop approaches and therapies targeting theaging process and age-related diseases (Butler et al.,2008). Delaying the process of aging, even slightly,would have profound social, medical and economic ben-efits (Olshansky et al., 2006; Butler et al., 2008). Forexample, slowing aging by a mere 7 years would cutmortality of age-related diseases by half at every age.Therefore, the potential benefits from research on thebasic biology and genetics of aging are unparalleled interms of improving quality",
              "title": "2012 - Genome-Environment Interactions That Modulate.pdf",
              "version": "v0",
              "chunk_order": 14,
              "document_id": "b1a1997c-e9df-5dc0-9d12-a3977d0c64ec",
              "extraction_id": "68b12e10-d643-570a-aa63-eda62de81928",
              "associatedQuery": "Can genetic research lead to treatments that slow down aging?"
            }
          },
          {
            "id": "3c636897-c47e-505d-9203-306124b73e0e",
            "score": 0.6825295686721851,
            "metadata": {
              "text": "Interestingly, when senescent cells are abolished either through genetic manipulation or via senolytic\ndrugs, biological aging is signicantly halted in mice [ 53,54]. Therefore, trials are now under way to\ntest the ability of senolytics to postpone age-associated pathologies in humans [ 55]. Notably, multi-\nple drugs are being pursued that either directly or indirectly impact DNA repair or the consequenceof DNA damage.\nFuture Prospects: Developing Interventions through DNA Repair",
              "title": "2020 - Protecting the Aging Genome.pdf",
              "version": "v0",
              "chunk_order": 57,
              "document_id": "bb774030-2570-5596-b2ab-b8f57ff81086",
              "extraction_id": "e5067ce2-69a6-5433-bed4-b95daeaa691e",
              "associatedQuery": "Can genetic research lead to treatments that slow down aging?"
            }
          },
          {
            "id": "43cba086-7f03-529f-bcd0-6483202bf3c7",
            "score": 0.6815123354937958,
            "metadata": {
              "text": "and potentially important genetic markers for slow aging\nhave been found in humans (Suh et al. 2008). Elucidating\nthe function of such genes is believed to enable decipher-\ning the core of the aging process, answer to what extentthe process is conserved, and pave the way for therapeutic\ninterventions of age-related maladies, including cancers,\nneurodegeneration, and metabolic syndrome (Guarente\n2011).\nThe identity of the virtual gerontogenes so far discov-",
              "title": "2012 - Peroxiredoxins, gerontogenes linking.pdf",
              "version": "v0",
              "chunk_order": 5,
              "document_id": "2eaad7ba-b6ae-5382-ba79-84609080b53e",
              "extraction_id": "38ebdc6a-8e8e-5472-a3ed-9a0f06591474",
              "associatedQuery": "Can genetic research lead to treatments that slow down aging?"
            }
          },
          {
            "id": "de7c30f6-cce9-563d-83f4-809f2aab781b",
            "score": 0.67976732970671,
            "metadata": {
              "text": "discover specific genes that directly influence how quickly people age, beyond diseases. If such\ngenes exist, their effects were too small to be detected in this study. The next step will be to\nexpand the study to include more participants, which will hopefully pinpoint further genomic regions\nand help disentangle the biology of ageing and disease.\nDOI: https://doi.org/10.7554/eLife.39856.002",
              "title": "2019 - Genomics of 1 million parent lifespans.pdf",
              "version": "v0",
              "chunk_order": 17,
              "document_id": "f68b939c-847b-5eac-8926-24713ae43478",
              "extraction_id": "ff0adc7c-70ff-5b14-ba7d-a9dda60fac80",
              "associatedQuery": "Can genetic research lead to treatments that slow down aging?"
            }
          },
          {
            "id": "4eb34c07-921b-55bb-98eb-ff013bb2ace0",
            "score": 0.6764809278868523,
            "metadata": {
              "text": "using bulk mRNA or even analyzing single cells (scRNA-seq). In addition, advances in\nmolecular biology and cell culture approaches (for instance Clustered Regularly Interspaced\nShort Palindromic Repeats (CRISPR)/Cas9) will be benecial in clarifying aging-processes\nacross species.\nAn improved understanding of epigenetic mechanisms affecting longevity will be\ndeciding crucial step towards the identication of new potential therapeutic targets. In",
              "title": "2021 - Epigenetics of Aging and Aging-Associated Diseases.pdf",
              "version": "v0",
              "chunk_order": 146,
              "document_id": "70945353-4808-539a-80f9-5632c27913e5",
              "extraction_id": "e2bc9b8e-2349-509b-a148-fbd86f0455f4",
              "associatedQuery": "Can genetic research lead to treatments that slow down aging?"
            }
          },
          {
            "id": "f20fd517-5f05-53ca-93a5-916bc891ad92",
            "score": 0.6719373855665463,
            "metadata": {
              "text": "century. Manipulation of aging-related genes by diet,lifestyle, and pharmaceuticals could dramatically im-prove human health and could be used to develop drugsagainst age-related diseases such as cancer, heart dis-ease, type 2 diabetes, obesity, and neurodegenerativediseases. The hundreds of aging-related genes and genesrelated to CR already identified offer enormous oppor-tunities for target discovery (Fig. 2). Although aging-related genes cannot be modified in humans, under-standing how these can be",
              "title": "2012 - Genome-Environment Interactions That Modulate.pdf",
              "version": "v0",
              "chunk_order": 170,
              "document_id": "b1a1997c-e9df-5dc0-9d12-a3977d0c64ec",
              "extraction_id": "8650652a-1765-563b-a98e-2e9336bcf29a",
              "associatedQuery": "Can genetic research lead to treatments that slow down aging?"
            }
          },
          {
            "id": "265126e3-2a4d-518f-93cf-21a201747eef",
            "score": 0.669631520553646,
            "metadata": {
              "text": "5. Goldman DP, etal. Substantial health and economic returns from delayed aging may warrant \na new focus for medical research. Health Aff (Millwood). 2013;32(10):1698705.\n 6. Esplin ED, Oei L, Snyder MP.Personalized sequencing and the future of medicine: discov-\nery, diagnosis and defeat of disease. Pharmacogenomics. 2014;15(14):177190.\n 7. Marian AJ.Clinical applications of molecular genetic discoveries. Transl Res. 2016;168:614.",
              "title": "2020 - Clinical Genetics and Genomics of Aging.pdf",
              "version": "v0",
              "chunk_order": 1774,
              "document_id": "62b635c3-040e-512a-b016-6ef295308a1e",
              "extraction_id": "822571e2-b05d-5e17-9eaa-431151851111",
              "associatedQuery": "Can genetic research lead to treatments that slow down aging?"
            }
          },
          {
            "id": "afc304d1-dd43-55ec-811d-27ca27fc4e5d",
            "score": 0.6661873459816027,
            "metadata": {
              "text": "a medical intervention), without changing the fundamental rateof organismal aging. Nevertheless, it does seem that manyso-called longevity genes, as well as dietary restriction,\nappear to extend not only life span, but also health span\n(Kauffman et al., 2010; Luo et al., 2010 ). In that regard, it does\nappear that it is possible to experimentally slow the rate of aging.\nStill, in each case, aging does continue on as if there is some",
              "title": "2012 - Aging, Rejuvenation, and Epigenetic.pdf",
              "version": "v0",
              "chunk_order": 13,
              "document_id": "bde26feb-f423-51b0-89ec-6f079bfc8b17",
              "extraction_id": "b9f038dd-97af-51ea-bb32-d73bf66c3dcb",
              "associatedQuery": "Can genetic research lead to treatments that slow down aging?"
            }
          },
          {
            "id": "1c77b8dc-2fd6-5e3d-9cf0-5585e7c9fb57",
            "score": 0.6659484108717209,
            "metadata": {
              "text": "genetic modification. Currently, emerging evidence suggeststhat certain interventions (e.g. CR, dietary supplementation andchemical drugs) can prevent age-related diseases and promote\nlongevity, at least in part, through reversing the aberrant age-\nassociated changes in DNA methylation, suggesting the greatpotential of DNA methylation in therapeutic strategies againstage-related diseases ( Figure 1B ).However, to further understand the roles of DNA methyla-",
              "title": "2016 - Progress on the role of DNA methylation in aging.pdf",
              "version": "v0",
              "chunk_order": 43,
              "document_id": "e4cdc02f-4415-5638-aab8-f848b4d64a22",
              "extraction_id": "8829c724-73ff-582b-ab94-c9f1a906cfd5",
              "associatedQuery": "Can genetic research lead to treatments that slow down aging?"
            }
          }
        ],
        "kg_search_results": null
      }
    }
  },
  "9": {
    "results": {
      "completion": {
        "id": "chatcmpl-AIFhRRoCeBcURczVYxgfGYsJd8loh",
        "choices": [
          {
            "finish_reason": "stop",
            "index": 0,
            "message": {
              "content": "Mitochondrial DNA (mtDNA) influences aging through several mechanisms:\n\n1. **Oxidative Damage**: Mitochondria are crucial for energy production and are highly susceptible to oxidative damage. The accumulation of oxidative lesions in mtDNA is a significant source of age-related damage [1].\n\n2. **Mutations and Lifespan**: Mutations in mtDNA can reduce lifespan. These mutations can aggravate aging and impair brain development, indicating a direct link between mtDNA mutations and the aging process [2].\n\n3. **Mitochondrial Dysfunction**: Aging is associated with mtDNA mutations, which contribute to mitochondrial dysfunction. This dysfunction is linked to age-related diseases and metabolic disorders, further influencing lifespan [4].\n\n4. **Genetic Instability**: The mutation rate for mtDNA is significantly higher than for nuclear DNA. These mutations can compromise mitochondrial functions, such as electron transport and oxidative phosphorylation, leading to declines in ATP levels and increased production of reactive oxygen species, which further damage both nuclear and mitochondrial DNA [9].\n\nOverall, mtDNA influences aging by accumulating mutations and oxidative damage, leading to mitochondrial dysfunction and contributing to age-related physiological decline.",
              "role": "assistant"
            }
          }
        ],
        "created": 1728913617,
        "model": "gpt-4o-2024-08-06",
        "object": "chat.completion",
        "system_fingerprint": "fp_6b68a8204b",
        "usage": {
          "completion_tokens": 239,
          "prompt_tokens": 1385,
          "total_tokens": 1624
        }
      },
      "search_results": {
        "vector_search_results": [
          {
            "id": "9ec5a15f-8232-5e79-a78d-64eeba35747f",
            "score": 0.8005366086799949,
            "metadata": {
              "text": "In addition to nuclear DNA, mitochondrial DNA (mtDNA) also is affected by aging. Alterations in mitochondrial function and mito-chondrial-nuclear signaling occur during aging and have been linked to sex biases in aging and age-related diseases (28). Due to their role in energy production, mitochondria are at high risk of oxida-tive damage. Not surprisingly, accumulation of oxidative lesions is an important source of age-related mtDNA damage (29). In aged Wistar rats brains, DNA oxidation, as measured by",
              "title": "2018 - Sex Differences in Aging Genomic Instability.pdf",
              "version": "v0",
              "chunk_order": 39,
              "document_id": "8cfb5529-7f0c-58fc-b6e4-b3ee800fb72f",
              "extraction_id": "400784cf-bb7d-5bf8-b735-2142ebf7c953",
              "associatedQuery": "How does mitochondrial DNA influence aging?"
            }
          },
          {
            "id": "39019881-9b6d-5111-87ea-71c413bdf4ff",
            "score": 0.7608792412543074,
            "metadata": {
              "text": "mitochondrial DNA mutations can reduce lifespan. Sci Rep. 2014;4:6569.\n20. Ross JM, Stewart JB, Hagstrm E, Bren S, Mourier A, Coppotelli G,\nFreyer C, Lagouge M, Hoffer BJ, Olson L. Germline mitochondrial DNA\nmutations aggravate ageing and can impair brain development. Nature.\n2013;501(7467):412 5.\n21. Sondheimer N, Glatz CE, Tirone JE, Deardorff MA, Krieger AM, Hakonarson H.\nNeutral mitochondrial heteroplasmy and the influence of aging. Hum Mol\nGenet. 2011;20(8):1653 9.",
              "title": "2017 - Independent impacts of aging.pdf",
              "version": "v0",
              "chunk_order": 126,
              "document_id": "d1d0b9ce-f827-5dfb-8e39-d87a9ca52f6d",
              "extraction_id": "1f0b6363-a045-53aa-a124-4cf89e61fc26",
              "associatedQuery": "How does mitochondrial DNA influence aging?"
            }
          },
          {
            "id": "1a9d5c26-f606-5cb5-98ee-4120de3fbd1a",
            "score": 0.7512575611846916,
            "metadata": {
              "text": "102. Zhang R, Wang Y , Ye K, Picard M, Gu Z.Independent impacts of aging on mitochondrial \nDNA quantity and quality in humans. BMC Genomics. 2017;18:890. https://doi.org/10.1186/\ns12864-017-4287-0.\n 103. Norddahl GL, et al. Accumulating mitochondrial DNA mutations drive premature hema-\ntopoietic aging phenotypes distinct from physiological stem cell aging. Cell Stem Cell. \n2011;8:499510. https://doi.org/10.1016/j.stem.2011.03.009.",
              "title": "2020 - Clinical Genetics and Genomics of Aging.pdf",
              "version": "v0",
              "chunk_order": 783,
              "document_id": "62b635c3-040e-512a-b016-6ef295308a1e",
              "extraction_id": "c8db1d28-f6c2-5896-95ec-bb01159ba483",
              "associatedQuery": "How does mitochondrial DNA influence aging?"
            }
          },
          {
            "id": "e0d41918-20fb-53f0-ac63-cd079c6dce1e",
            "score": 0.7381081801089105,
            "metadata": {
              "text": "other studies, the risk for metabolic disorders is highly associated with age-related \ndiseases that affect lifespan, and interestingly these conditions exhibit mitochon-\ndrial dysfunction [73].\nAging is a complex process as a time-dependent progressive loss of physiologi-\ncal integrity, leading to impaired function and increased vulnerability to death [74], \nand as we described above, aging is highly associated with mtDNA mutations; in",
              "title": "2020 - Clinical Genetics and Genomics of Aging.pdf",
              "version": "v0",
              "chunk_order": 530,
              "document_id": "62b635c3-040e-512a-b016-6ef295308a1e",
              "extraction_id": "385c192b-a416-5208-9615-20111ce782aa",
              "associatedQuery": "How does mitochondrial DNA influence aging?"
            }
          },
          {
            "id": "ef3be4e1-4cbc-5b61-a286-9b759df08cd9",
            "score": 0.735212868620116,
            "metadata": {
              "text": "mt, and overall mitonuclear genomic compatibility. \nGiven the uncertainty of mtDNA mutation accumulation in driving the natural aging process, it is plausible that mito -\nchondrial communication may be a significant evolutionarily conserved force that influences lifespan and/or healthspan.\nAcknowledgements Funding was provided by the American Federa-\ntion for Aging Research (AFAR), the National Institute on Aging (T32",
              "title": "2020 - Mitonuclear genomics and aging.pdf",
              "version": "v0",
              "chunk_order": 148,
              "document_id": "e05fdc09-c8d8-5134-a1fd-bf07a1564981",
              "extraction_id": "381cc064-9970-5dcd-b959-c52a8e487fe7",
              "associatedQuery": "How does mitochondrial DNA influence aging?"
            }
          },
          {
            "id": "4206977e-23df-5307-8d8a-cb2ed7b33595",
            "score": 0.7351043601303022,
            "metadata": {
              "text": "abolic regulation through mitochondrial signaling. Am J Physiol Endocrinol Metab. \n2014;306:E58191.\n 74. Zhang R, Wang Y , Ye K, Picard M, Gu Z.Independent impacts of aging on mitochondrial DNA \nquantity and quality in humans. BMC Genomics. 2017;18:890.\n 75. Hebert SL, Lanza IR, Nair KS.Mitochondrial DNA alterations and reduced mitochondrial \nfunction in aging. Mech Ageing Dev. 2010;131:45162.\n 76. Liu D, Li H, Lu J, Bai Y .Tissue-specific implications of mitochondrial alterations in aging.",
              "title": "2020 - Clinical Genetics and Genomics of Aging.pdf",
              "version": "v0",
              "chunk_order": 607,
              "document_id": "62b635c3-040e-512a-b016-6ef295308a1e",
              "extraction_id": "ef9463cd-cf21-527f-ae4a-3df211c78435",
              "associatedQuery": "How does mitochondrial DNA influence aging?"
            }
          },
          {
            "id": "7095cdbb-852e-541e-884b-a9e67c2c790c",
            "score": 0.7326218108124084,
            "metadata": {
              "text": "Sun., N, Youle, R. J. and Finkel, T. (2016). The mitochondrial basis of aging.\nMol. Cell 61, 654-666. doi:10.1016/j.molcel.2016.01.028\nSymer, D. E., Connelly, C., Szak, S. T., Caputo, E. M., Cost, G. J., Parmigiani, G.\nand Boeke, J. D. (2002). Human L1 retrotransposition is associated with genetic\ninstability in vivo. Cell110, 327-338. doi:10.1016/S0092-8674(02)00839-5\nSzabo, L., Morey, R., Palpant, N. J., Wang, P. L., Afari, N., Jiang, C., Parast,",
              "title": "2020 - Transposable elements, circular RNAs and mitochondrial.pdf",
              "version": "v0",
              "chunk_order": 300,
              "document_id": "7bebb41c-ac73-5917-91d3-4f59fbb3266a",
              "extraction_id": "5cbace8d-e538-5531-9311-ea9726ad2f15",
              "associatedQuery": "How does mitochondrial DNA influence aging?"
            }
          },
          {
            "id": "512e09e3-c880-5bed-9071-bfa84d80a5a3",
            "score": 0.7307272014672046,
            "metadata": {
              "text": "than ones that affect mitochondrial DNA12,57,58,71.So,this is an important reason for favouring\nnuclear DNA as the ultimate damage target in natural ageing. Nevertheless, it is conceivable that\nwhen mutations occur in the mitochondrial genome, mutant-protein production could increase\nthe inefficiency of the mitochondrial respiratory chain, thereby resulting in more reactive oxygenspecies, which would then damage nuclear and mitochondrial DNA further.",
              "title": "2004 - Ageing, repetitive genomes and DNA.pdf",
              "version": "v0",
              "chunk_order": 30,
              "document_id": "1772d596-16a3-547a-9f76-2cb658e89746",
              "extraction_id": "9b7b2005-857b-5379-ba5c-3a8c6fd6c891",
              "associatedQuery": "How does mitochondrial DNA influence aging?"
            }
          },
          {
            "id": "2e2de9a7-2e83-5f46-a4b7-08eddcd37baa",
            "score": 0.7305692869764141,
            "metadata": {
              "text": "generation animals as they grow older.Mitochondrial DNAGenetic instability outside of the nuclear genome mightalso contribute to aging (reviewed in Lee et al., 1997;Wallace et al., 1998). The mutation rate for mitochondrialDNA (mtDNA) is 10- to 20-fold greater than for nuclearDNA, and it is believed that mtDNA mutations may com-promise mitochondrial functions in different ways (Fig-ure 4). First, defects in electron transport and oxidativephosphorylation could lead to declines in ATP levelsand the NAD:NADH",
              "title": "1999 - Molecular Biology of Aging.pdf",
              "version": "v0",
              "chunk_order": 64,
              "document_id": "6f122d3a-d8ca-598f-8767-c059a941cef3",
              "extraction_id": "68c55aac-d8fa-5287-a420-2bb83a2c159e",
              "associatedQuery": "How does mitochondrial DNA influence aging?"
            }
          },
          {
            "id": "c35761f9-58e8-526b-94fd-96ed7599cb53",
            "score": 0.7301242190673481,
            "metadata": {
              "text": "of the human aging process(Corral-Debrinski et al., 1992; Soong et al., 1992;Wei etal., 1996b), and it has been demonstrated that certain pointmutations of mitochondrial DNA accumulate in the aginghuman brain (Zhang et al., 1993; Liu et al., 1997). However,thefunctionalimplicationsofthesendingsarecontroversial(Hayashietal.,1994).Tocomplicatethematterfurther,Takaiand co-workers discuss the possibility that the commonage-associated changes in human and mouse",
              "title": "1998 - Neurodegeneration and Aging Role.pdf",
              "version": "v0",
              "chunk_order": 30,
              "document_id": "694c44f3-6025-5a2c-9c72-d9c5f16c8b85",
              "extraction_id": "21db434d-3cf5-5ff1-8257-0941ebe74822",
              "associatedQuery": "How does mitochondrial DNA influence aging?"
            }
          }
        ],
        "kg_search_results": null
      }
    }
  },
  "10": {
    "results": {
      "completion": {
        "id": "chatcmpl-AIFhaX8SGHyXXi1vHCPCMEW3CahGu",
        "choices": [
          {
            "finish_reason": "stop",
            "index": 0,
            "message": {
              "content": "Yes, there are known genetic mutations that cause premature aging. Specifically, mutations in the LMNA gene are associated with Hutchinson-Gilford Progeria Syndrome, a disease characterized by premature aging [4]. Additionally, mutations in the RECQL4 gene are linked to Rothmund-Thomson syndrome, which also involves premature aging [4]. These genetic disorders highlight the connection between genome integrity and premature aging [7].",
              "role": "assistant"
            }
          }
        ],
        "created": 1728913626,
        "model": "gpt-4o-2024-08-06",
        "object": "chat.completion",
        "system_fingerprint": "fp_6b68a8204b",
        "usage": {
          "completion_tokens": 84,
          "prompt_tokens": 1643,
          "total_tokens": 1727
        }
      },
      "search_results": {
        "vector_search_results": [
          {
            "id": "a9f7eda5-1b64-507e-95dd-07c81f2d603b",
            "score": 0.7192796468734741,
            "metadata": {
              "text": "logical phenomena is often facilitated by the \nstudy of genetic mutants, and, in the case of \nhumans, genetic disorders. Accordingly, a search \nwas made, over the years, for genetic disorders \ncharacterized by premature aging. If DNA dam- \nage and repair has anything to do with aging it \nshould be evidenced in such individuals. Martin \n(1978) listed 162 genetic syndromes in humans with some or many signs of premature aging. \nAbout 21 feahares are considered as markers for",
              "title": "1993 - Genomic Damage and Its Repair.pdf",
              "version": "v0",
              "chunk_order": 127,
              "document_id": "d049f302-a130-5ee4-a1b5-5091605d5173",
              "extraction_id": "64063108-0ff2-54e5-9801-bc1c49cbdee4",
              "associatedQuery": "Are there any known genetic mutations that cause premature aging?"
            }
          },
          {
            "id": "882149e3-8186-5577-a2a7-79f2659ff9b4",
            "score": 0.6718193683839645,
            "metadata": {
              "text": "[315] Szilard, L. On the nature of the aging process. Proc. Natl. Acad. Sci. USA\n45:3545; 1959.\n[316] Vijg, J.; Dolle, M. E. Large genome rearrangements as a primary cause of\naging. Mech. Ageing Dev. 123:907915; 2002.\n[317] Vijg, J. Somatic mutations and aging: a re-evaluation. Mutat. Res.\n447:117135; 2000.\n[318] Martin, G. M. Genetic syndromes in Man with potential relevance to the\npathobiology of aging. Birth Defects Orig. Artic. Ser. 14:539; 1978.",
              "title": "2007 - Trends in oxidative aging theories.pdf",
              "version": "v0",
              "chunk_order": 379,
              "document_id": "0d752c1a-706a-5b9e-88ef-ba7c51735c3c",
              "extraction_id": "752c6f1a-0c4d-5419-86cd-687d2aed7817",
              "associatedQuery": "Are there any known genetic mutations that cause premature aging?"
            }
          },
          {
            "id": "da4e59b7-d5b6-5992-9607-f6697c8f5276",
            "score": 0.6711906790733337,
            "metadata": {
              "text": "19\n 6. Milholland B, Suh Y , Vijg J.Mutation and catastrophe in the aging genome. Exp Gerontol. \n2017;94:3440.\n 7. Maslov AY , Ganapathi S, Westerhof M, Quispe-Tintaya W, White RR, Van Houten B, etal. \nDNA damage in normally and prematurely aged mice. Aging Cell. 2013;12:46777.\n 8. Blokzijl F, de Ligt J, Jager M, Sasselli V , Roerink S, Sasaki N, etal. Tissue-specific mutation \naccumulation in human adult stem cells during life. Nature. 2016;538:2604.",
              "title": "2020 - Clinical Genetics and Genomics of Aging.pdf",
              "version": "v0",
              "chunk_order": 164,
              "document_id": "62b635c3-040e-512a-b016-6ef295308a1e",
              "extraction_id": "ead14808-bfb7-5e32-9830-28efaae71151",
              "associatedQuery": "Are there any known genetic mutations that cause premature aging?"
            }
          },
          {
            "id": "4841d806-98b4-513e-94a2-714df6c896f5",
            "score": 0.6635691523551941,
            "metadata": {
              "text": "143 Gonzalo S, Kreienkamp R & Askjaer P (2017) Hutchinson -Gilford Progeria \nSyndrome: A premature aging disease caused by LMNA gene mutations. \nAgeing Res. Rev.  33, 1829. \n144 Lu L, Jin W & Wang LL (2017) Aging in Ro thmund -Thomson syndrome and \nrelated RECQL4 genetic disorders. Ageing Res. Rev.  33, 3035. \n145 de Renty C & Ellis NA (2017) Blooms syndrome: Why not premature aging? \nAgeing Res. Rev.  33, 3651. \n146 Shiloh Y & Lederman HM (2017) Ataxia -telangiectasia (A -T): An emerging",
              "title": "2019 - Towards Understanding Genomic Instability, Mitochondrial.pdf",
              "version": "v0",
              "chunk_order": 137,
              "document_id": "9b34514d-3d0e-52b5-8e5e-2f3c0708fd82",
              "extraction_id": "d620ea24-4422-5636-86f5-0943371a4a18",
              "associatedQuery": "Are there any known genetic mutations that cause premature aging?"
            }
          },
          {
            "id": "fc10c968-3108-5c4b-a49c-cb0feabd18c5",
            "score": 0.6526112945444175,
            "metadata": {
              "text": "genetic disease model of premature aging, In: Harrison,D.E., eds, Genetic Effects on Aging II (Telford Press, Caldwell,NJ), pp. 521542.\n[2] Djawdan, M., Sugiyama, T., Schlaeger, L., Bradley, T.J. and\nRose, M.R. (1996) Metabolic aspects of the trade-off between\nfecundity and longevity in Drosophila melanogaster ,Physiol.\nZool. 69, 11751195.\n[3] Fleming, J.E., Spicer, G.S., Garrison, R.C. and Rose, M.R.",
              "title": "2002 - Pharmacology, Genomics, and the Evolutionary Biology.pdf",
              "version": "v0",
              "chunk_order": 43,
              "document_id": "1bc636a3-6ce0-5fea-b549-0dae90a78f1b",
              "extraction_id": "e501662f-ffca-563b-97a7-b682a5d7f6ba",
              "associatedQuery": "Are there any known genetic mutations that cause premature aging?"
            }
          },
          {
            "id": "eb8b89de-422a-5e9e-9ac8-60af4cd718c2",
            "score": 0.6519961953163147,
            "metadata": {
              "text": "genes of a whole chromosome ineffective, couldbe a main causal factor in aging (Szilard, 1959).According to Maynard Smith, such types of mu-tations do not seem likely to be common enoughto be the main cause of aging. However, at thetime quantitative information on the possible age-related accumulation of different types of muta-tions in various tissues of mammals wascompletely lacking. The question, therefore,whether somatic mutations are a cause of aging,has not been resolved, more than four decadesafter",
              "title": "2002 - Large genome rearrangements as a primary cause of aging.pdf",
              "version": "v0",
              "chunk_order": 12,
              "document_id": "8a8926dc-2360-5a54-b586-8acc34e51c32",
              "extraction_id": "8f1a0875-8179-5d45-abc0-bbd4c9ac8da5",
              "associatedQuery": "Are there any known genetic mutations that cause premature aging?"
            }
          },
          {
            "id": "34e6b3c4-63bf-5198-ab09-2a7200a7c19a",
            "score": 0.6518602578723288,
            "metadata": {
              "text": "features of premature aging (16, 17). Subsequent experiments conrmed that mitochondrial DNA\nmutations and deletions were the driving force behind the observed accelerated aging phenotypes(18).\nTHE LINK BETWEEN NUCLEAR GENOME INTEGRITY\nAND PREMATURE AGING\nThe notion that the majority of currently identied progeria syndromes originate from defects\nin genome maintenance highlights the importance of the condition of DNA in the process of",
              "title": "2016 - Genome Integrity in Aging.pdf",
              "version": "v0",
              "chunk_order": 20,
              "document_id": "85d5fcbb-5385-5a01-8139-d11fc8b1fe3a",
              "extraction_id": "17b26647-4659-5f2d-a9b0-7c122d4b5d1a",
              "associatedQuery": "Are there any known genetic mutations that cause premature aging?"
            }
          },
          {
            "id": "beed04cc-28c7-5dc7-b334-51226a217439",
            "score": 0.6509696453231796,
            "metadata": {
              "text": "Tryggvason K,ZhouZ.Genomicinstability inlaminopathy based\npremature aging,NatMed. 2005;11:780 785.\n13.MisteliT,ScaffidiP.Genomeinstability inprogeria:when\nrepairgetsold,NatMed. 2005;11:718 719.\n14.PereiraS,Bourgeois P,NavarroC,EstevesVieiraV,CauP,De\nSandreGiovannoli A,LvyN.HGPSandrelatedpremature aging\ndisorders: Fromgenomicidentification tothefirsttherapeutic \napproaches, MechAgeingDev.2008;129:449 459.\n15.SmithED,Kudlow BA,FrockRL,KennedyBK.Atypenuclear",
              "title": "2009 - Genomic instability and DNA damage responses in progeria arising.pdf",
              "version": "v0",
              "chunk_order": 83,
              "document_id": "b7d96f9f-8ad4-5f8f-94f9-60404806d478",
              "extraction_id": "72beba0d-8c77-5aa9-82ac-ddf6a19355ac",
              "associatedQuery": "Are there any known genetic mutations that cause premature aging?"
            }
          },
          {
            "id": "badf3a36-1f99-58aa-b80c-725eccf4e8f3",
            "score": 0.6501748561859131,
            "metadata": {
              "text": "Nature Genetics | Volume 55 | February 2023 | 268279 278\nArticle https://doi.org/10.1038/s41588-022-01279-621. Tiwari, V. & Wilson, D. M. 3rd. DNA damage and associated DNA \nrepair defects in disease and premature aging. Am. J. Hum. Genet.  \n105, 237257 (2019).\n22. Tamae, D., Lim, P., Wuenschell, G. E. & Termini, J. Mutagenesis and repair induced by the DNA advanced glycation end product N2-1-(carboxyethyl)-2-deoxyguanosine in human cells. Biochemistry  \n50, 23212329 (2011).",
              "title": "2023 - Genome-wide RNA polymerase stalling.pdf",
              "version": "v0",
              "chunk_order": 119,
              "document_id": "78812a12-8d31-5159-8367-b0d38e5bc84b",
              "extraction_id": "31088092-778f-59e0-a9de-5ec25c241aab",
              "associatedQuery": "Are there any known genetic mutations that cause premature aging?"
            }
          },
          {
            "id": "c35d1f43-c3bd-5cac-ae4d-937be35f1121",
            "score": 0.6501638889312744,
            "metadata": {
              "text": "[36] J.  de  Boer,  J.O.  Andressoo,  J.  de  Wit,  J.  Huijmans,  R.B.  Beems,  H.  van  Steeg,  et  al.,\nPremature  aging  in  mice  decient  in  DNA  repair  and  transcription,  Science\n296  (2002)  12761279.\n[37]  S.M.  Schuh-Huerta,  N.A.  Johnson,  M.P.  Rosen,  B.  Sternfeld,  M.I.  Cedars,  R.A.\nReijo Pera,  Genetic  markers  of  ovarian  follicle  number  and  menopause  in\nwomen  of  multiple  ethnicities,  Hum.  Genet.  131  (2012)  17091724.",
              "title": "2016 - Menopause Genome stability as new paradigm.pdf",
              "version": "v0",
              "chunk_order": 145,
              "document_id": "564dead1-2737-572f-860c-f00de4d0395e",
              "extraction_id": "0855231d-cb95-540c-a3dd-c93729efb34c",
              "associatedQuery": "Are there any known genetic mutations that cause premature aging?"
            }
          }
        ],
        "kg_search_results": null
      }
    }
  },
  "11": {
    "results": {
      "completion": {
        "id": "chatcmpl-AIFhez5FFXsDDkyj8CmiEuE5k6YSr",
        "choices": [
          {
            "finish_reason": "stop",
            "index": 0,
            "message": {
              "content": "Recent discoveries in the genetics of aging include the identification of a number of genes capable of altering the aging process significantly in animal models and even in some humans [2]. Additionally, recent efforts have focused on isolating aging mutants through mutagenesis experiments to determine the mechanistic basis for unusual life spans, leading to the discovery of genes that can either enhance or reduce life span [4]. These findings contribute to a growing understanding of the genetic factors influencing aging and longevity.",
              "role": "assistant"
            }
          }
        ],
        "created": 1728913630,
        "model": "gpt-4o-2024-08-06",
        "object": "chat.completion",
        "system_fingerprint": "fp_a20a4ee344",
        "usage": {
          "completion_tokens": 93,
          "prompt_tokens": 1336,
          "total_tokens": 1429
        }
      },
      "search_results": {
        "vector_search_results": [
          {
            "id": "c96b67f8-ad31-50fd-b053-07b127938ef2",
            "score": 0.7399749314799959,
            "metadata": {
              "text": "During the past century, remarkable progress has been \nmade in unveiling the mechanisms of aging. Genetic and molecular pathways that regulate healthspan and lifespan have been identified in various model organisms, provid-ing a rich knowledge base (Longo etal. 2015; Lopez-Otin etal. 2013, 2016; Singh etal. 2019). However, the focus on",
              "title": "2020 - Mitonuclear genomics and aging.pdf",
              "version": "v0",
              "chunk_order": 10,
              "document_id": "e05fdc09-c8d8-5134-a1fd-bf07a1564981",
              "extraction_id": "e6fb876b-e91c-505a-aa16-7b428ec61f10",
              "associatedQuery": "What recent discoveries have been made about the genetics of aging?"
            }
          },
          {
            "id": "1c4286b6-ede2-568b-9c18-b1e99ede17a6",
            "score": 0.7397668361663818,
            "metadata": {
              "text": "series of recent breakthroughs, a number of genes capable ofaltering the aging process as a whole  or at least to a largedegree  have been identified in animal models and even a fewin humans (Finch & Ruvkun, 2001; de Magalhes, 2005; Kenyon,2005). Furthermore, multiple alleles have been examined fortheir association with human exceptional longevity (Vijg & Suh,2005). This is a fascinating and important area of research, yetthere are now so many genes being associated with aging andlongevity that keeping",
              "title": "2009 - The Human Ageing Genomic Resources online.pdf",
              "version": "v0",
              "chunk_order": 10,
              "document_id": "e43cd3b6-ad8e-5422-ba7c-ceb6e66cc529",
              "extraction_id": "7ada6b55-99c2-5e20-bf96-d153f927256c",
              "associatedQuery": "What recent discoveries have been made about the genetics of aging?"
            }
          },
          {
            "id": "e09c33ea-4139-5cc2-9cf5-a40045f26a0c",
            "score": 0.7192355608712308,
            "metadata": {
              "text": "Recent developments on the genetics of aging can be seen as several streams\nof effort. In general, humans show a relatively modest ( <50%) heritability of",
              "title": "2001 - The genetics of aging.pdf",
              "version": "v0",
              "chunk_order": 13,
              "document_id": "aa9a9193-b6f3-5ef8-aefd-e01ec44abb46",
              "extraction_id": "76bae746-eabf-51ed-a01f-d32ecc89c11b",
              "associatedQuery": "What recent discoveries have been made about the genetics of aging?"
            }
          },
          {
            "id": "f7120061-9773-5f74-9760-5442d49fbaae",
            "score": 0.7119012475013733,
            "metadata": {
              "text": "One approach that has become increasingly common in the characterization of the ge-netics of aging is to isolate aging mutants, usually from mutagenesis experiments, andthen to determine the mechanistic basis for the unusual life span in the mutants. Thisapproach has led to the discovery of genes that can enhance (e.g., Maynard Smith 1958;Lin et al. 1988; reviewed in Guarente and Kenyon 2000, Kim 2007) or reduce life span(e.g., Pearl and Parker 1922). Most of the large-effect mutants affecting aging",
              "title": "2009 - Garland_and_Rose_Experimental_Evolution.pdf",
              "version": "v0",
              "chunk_order": 4645,
              "document_id": "496faa7f-9623-5ab7-9816-7c3755abb3aa",
              "extraction_id": "5aa7f5b9-df70-54ec-a95c-dcaefa3b617f",
              "associatedQuery": "What recent discoveries have been made about the genetics of aging?"
            }
          },
          {
            "id": "d0e74ffd-034d-5e0e-86b6-4cf0de57d774",
            "score": 0.7118854098388321,
            "metadata": {
              "text": "One approach that has become increasingly common in the characterization of the ge-netics of aging is to isolate aging mutants, usually from mutagenesis experiments, andthen to determine the mechanistic basis for the unusual life span in the mutants. Thisapproach has led to the discovery of genes that can enhance (e.g., Maynard Smith 1958;Lin et al. 1988; reviewed in Guarente and Kenyon 2000, Kim 2007) or reduce life span(e.g., Pearl and Parker 1922). Most of the large-effect mutants affecting aging",
              "title": "2009 - Experimental_Evolution.pdf",
              "version": "v0",
              "chunk_order": 4645,
              "document_id": "34821353-1b74-5ee2-ac39-66dd46f145bf",
              "extraction_id": "c9d59e72-f068-58da-be7a-71b2f51a23f3",
              "associatedQuery": "What recent discoveries have been made about the genetics of aging?"
            }
          },
          {
            "id": "217c3592-1622-503f-a140-fd1452083301",
            "score": 0.7038054642793272,
            "metadata": {
              "text": "genetics of aging I. What is aging? Frontiers in Genetics. doi:10.3389/fgene.2012.00134.\nr\nose, Michael \nr\n., Anthony D. Long, Laurence D. Mueller, Cristina L. \nr\nizza, Kennedy C. Matsagas, \nLeeF. Greer, and Bryant \nvilleponteau. 2009. \ne\nvolutionary nutrigenomics. In The future of aging, eds. \nG. M. Fahy, M. D. West, L. S. Coles, and S. B. \nh\narris. Berlin: Springer.\nr\nushton, J. \np\nhillippe. 1995. Race, evolution, and behavior: A life history approach. New Brunswick, NJ: \nTransaction \np\nublishers.",
              "title": "2015 - Great Is Their Sin.pdf",
              "version": "v0",
              "chunk_order": 251,
              "document_id": "e5ae9710-3049-5327-82e4-e6626eb670c2",
              "extraction_id": "44c57701-0d0e-5ef8-afa1-ea3a6c4742d6",
              "associatedQuery": "What recent discoveries have been made about the genetics of aging?"
            }
          },
          {
            "id": "b3e21ac9-8df8-5119-a769-a9da82db78da",
            "score": 0.7030582008923185,
            "metadata": {
              "text": "informed by age-related disease identifies loci for exceptional human longevity. Li H, editor. \nPLoS Genet. 2015. https://doi.org/10.1371/journal.pgen.\n 15. Polderman TJC, Benyamin B, de Leeuw CA, Sullivan PF, van Bochoven A, Visscher PM, \netal. Meta-analysis of the heritability of human traits based on fifty years of twin studies. Nat \nGenet. 2015;47:7029.\n 16. Cellerino A, Ori A.What have we learned on aging from omics studies? Semin Cell Dev Biol. \n2017;70:17789.",
              "title": "2020 - Clinical Genetics and Genomics of Aging.pdf",
              "version": "v0",
              "chunk_order": 167,
              "document_id": "62b635c3-040e-512a-b016-6ef295308a1e",
              "extraction_id": "ead14808-bfb7-5e32-9830-28efaae71151",
              "associatedQuery": "What recent discoveries have been made about the genetics of aging?"
            }
          },
          {
            "id": "fd811aec-6e33-5078-83d5-b68bd59b5a61",
            "score": 0.7030342817306519,
            "metadata": {
              "text": "eries that have inspired thousands of researchers across the world \nto study aging, and we acknowledge the wider significance of the \ncreation of a field that has the potential to transform human health.\nGenetics\nAging is influenced by genetic factors. It may be surprising to know \nthat as recently as the 1970s and 1980s, the concept of modulating \nDownloaded from https://academic.oup.com/biomedgerontology/article/76/7/e85/6145792 by guest on 15 October 2023",
              "title": "2021 - Career Retrospective Tom Johnson?Genetics, Genomics.pdf",
              "version": "v0",
              "chunk_order": 6,
              "document_id": "f3a26f44-f5af-5b2b-aa1c-aec2fd99f17e",
              "extraction_id": "dd4a6239-2e79-5b99-89ef-3e4939b87805",
              "associatedQuery": "What recent discoveries have been made about the genetics of aging?"
            }
          },
          {
            "id": "de7c30f6-cce9-563d-83f4-809f2aab781b",
            "score": 0.7005462050437927,
            "metadata": {
              "text": "discover specific genes that directly influence how quickly people age, beyond diseases. If such\ngenes exist, their effects were too small to be detected in this study. The next step will be to\nexpand the study to include more participants, which will hopefully pinpoint further genomic regions\nand help disentangle the biology of ageing and disease.\nDOI: https://doi.org/10.7554/eLife.39856.002",
              "title": "2019 - Genomics of 1 million parent lifespans.pdf",
              "version": "v0",
              "chunk_order": 17,
              "document_id": "f68b939c-847b-5eac-8926-24713ae43478",
              "extraction_id": "ff0adc7c-70ff-5b14-ba7d-a9dda60fac80",
              "associatedQuery": "What recent discoveries have been made about the genetics of aging?"
            }
          },
          {
            "id": "a9434032-4a9d-54f8-a7a6-16110d1b3118",
            "score": 0.6990343928337097,
            "metadata": {
              "text": "males: what are the molecular and evolutionary causes? Aging Cell. 2007;6:225233. doi:10.1111/j.1474-9726.2007.00279.x\n 63. Benayoun BA, Pollina EA, Brunet A. Epigenetic regulation of ageing: link-\ning environmental inputs to genomic stability. Nat Rev Mol Cell Biol. 2015;16:593610. doi:10.1038/nrm4048\n 64. Sen P, Shah PP, Nativio R, Berger SL. Epigenetic mechanisms of longevity \nand aging. Cell. 2016;166:822839. doi:10.1016/j.cell.2016.07.050",
              "title": "2018 - Sex Differences in Aging Genomic Instability.pdf",
              "version": "v0",
              "chunk_order": 158,
              "document_id": "8cfb5529-7f0c-58fc-b6e4-b3ee800fb72f",
              "extraction_id": "0104338d-cc9c-538f-be29-8343a64da37d",
              "associatedQuery": "What recent discoveries have been made about the genetics of aging?"
            }
          }
        ],
        "kg_search_results": null
      }
    }
  },
  "12": {
    "results": {
      "completion": {
        "id": "chatcmpl-AIFhiQpAn2q80Ivtr6ZFWK3Q6aNNm",
        "choices": [
          {
            "finish_reason": "stop",
            "index": 0,
            "message": {
              "content": "Epigenetic changes affect aging through several mechanisms:\n\n1. **Alterations in Chromatin Structure**: During aging, there are various epigenetic alterations such as the accumulation of histone variants, changes in chromatin accessibility, loss of histones and heterochromatin, and imbalances in activating/repressing histone modifications. These changes can affect transcription and translation processes, impacting cellular function [1].\n\n2. **Epigenetic Drift**: As individuals age, epigenetic changes accumulate, leading to a phenomenon known as epigenetic drift. This drift results in the cumulative loss of gene regulation over time, impairing cellular and tissue function. It is suggested that this disruption may also play a role in the development of age-related diseases, such as cancer [3].\n\n3. **DNA Damage and Chromatin Remodeling**: It is suggested that epigenetic alterations during aging are largely triggered by DNA damage. This damage leads to chromatin remodeling and redistribution of chromatin modifiers, which are recruited away from their normal sites to engage in DNA repair [4].\n\n4. **Increased Expression of Pro-aging Genes**: Epigenetic dysregulation can lead to increased expression of pro-aging genes, such as the cell-cycle inhibitor p16, which drives cell senescence. This contributes to increased transcriptional noise and decreased coordination of gene expression, further contributing to organismal aging [10].\n\n5. **Potential for Reversibility**: Despite these changes, there is potential reversibility in epigenetic modifications, offering opportunities to alter the trajectory of age-related diseases. This highlights the plasticity of aging and the potential for interventions that could slow down the aging process [7].\n\nOverall, epigenetic changes play a crucial role in the aging process by affecting gene expression, cellular function, and the development of age-related diseases.",
              "role": "assistant"
            }
          }
        ],
        "created": 1728913634,
        "model": "gpt-4o-2024-08-06",
        "object": "chat.completion",
        "system_fingerprint": "fp_a20a4ee344",
        "usage": {
          "completion_tokens": 370,
          "prompt_tokens": 904,
          "total_tokens": 1274
        }
      },
      "search_results": {
        "vector_search_results": [
          {
            "id": "e97baf15-d14f-563f-875f-a2a6bd314ad4",
            "score": 0.765143417273266,
            "metadata": {
              "text": "Figure 1. Epigenetics of aging and aging-relate d diseases. During aging, various ep igenetic alterations occur including \naccumulation of histone variants, change s in chromatin accessibility mediated by chromatin remodeling complexes, loss \nof histones and heterochroma tin, imbalance of activating /repressing histone modifications and aberrant expres-\nsion/activity of miRNAs. These deregulations can affect transcrip tion and, subsequently, transl ation, as well as the stabi-",
              "title": "2021 - Epigenetics of Aging and Aging-Associated Diseases.pdf",
              "version": "v0",
              "chunk_order": 150,
              "document_id": "70945353-4808-539a-80f9-5632c27913e5",
              "extraction_id": "8db25d5e-25bd-5873-a53d-3815badbfd32",
              "associatedQuery": "How do epigenetic changes affect aging?"
            }
          },
          {
            "id": "38dc7635-499d-52ba-a35f-f57ba8f8df22",
            "score": 0.7465935349464417,
            "metadata": {
              "text": "ment of 5 years corresponded to a 21% increased risk of\nmortality overall [7]. Thus, predictions of epigenetic\nagemay be an indication of an individual s biological\nstate of aging.\nBeyond these examples of advanced epigenetic aging, a\ncomplementary but unanswered question is whether\nepigenetic clocks can also be slowed. Epigenetic aging\nstudies in humans have not thus far been well suited to\naddress questions of slowed aging, given the lack of\nwell-documented interventions that enhance health or",
              "title": "2017 - Epigenetic aging signatures in mice livers.pdf",
              "version": "v0",
              "chunk_order": 6,
              "document_id": "b20b11a6-1490-51b8-9218-c441a2e65ba7",
              "extraction_id": "8e77571a-60ea-5e53-877d-08169e86d553",
              "associatedQuery": "How do epigenetic changes affect aging?"
            }
          },
          {
            "id": "975250c9-7b4b-5ce5-b907-606ca6c70641",
            "score": 0.743276834487915,
            "metadata": {
              "text": "al., 2005 ).  The epigenetic changes that accumulated with age had a dramatic effect on gene expression, thus the authors propos e that a so-called epigenetic drift accompanies the aging process.   Epigenetic modifications can result in the cumulative loss of gene regulation over time, ultimately impairing cellular and tissue function.  Further, recent data sugge st that epigenetic disruption of tissue specific stem and progenitor cells may play a role in cancer development (Feinberg et al., 2006 ).  The",
              "title": "2008 - GENETIC REGULATION OF HEMATOPOIETIC STEM CELL AGING (3).pdf",
              "version": "v0",
              "chunk_order": 52,
              "document_id": "7412a162-ee3b-5f09-9886-8e9172dd3ee8",
              "extraction_id": "6c8b334f-2e14-5099-85fe-93f9ed6046ad",
              "associatedQuery": "How do epigenetic changes affect aging?"
            }
          },
          {
            "id": "7fad6aa1-d4d9-585c-bdbd-394a9552ec0e",
            "score": 0.7432393584704795,
            "metadata": {
              "text": "epigenetic changes during aging are currentlyunknown (Fig. 3). It has been suggested thatthe epigenetic alterations are largely triggered\nby DNA damage (reviewed in Oberdoerffer\nand Sinclair 2007). In this scenario, randomlyoccurring DNA damage leads to chromatin\nremodeling and to redistribution of chromatin\nmodiers within the genome with modiersbeing recruited away from their normal sites\nso that they can engage in the repair of the",
              "title": "2010 - Higher-order Genome Organization.pdf",
              "version": "v0",
              "chunk_order": 103,
              "document_id": "91339298-860e-57d0-b58d-5a4571b4fc2b",
              "extraction_id": "1a3a302a-4009-5ccf-aafa-f5f5a258ffde",
              "associatedQuery": "How do epigenetic changes affect aging?"
            }
          },
          {
            "id": "b43c1348-b982-59c2-9685-af7bb9fd0c4a",
            "score": 0.7407828790182737,
            "metadata": {
              "text": "Epigenetic Dysregulation with Age",
              "title": "2007 - Aging Hematopoietic Stem Cells Decline in Function and Exhibit Epigenetic Dysregulation.pdf",
              "version": "v0",
              "chunk_order": 150,
              "document_id": "a6fabf0c-e4a5-59f6-82c5-ebabce24fd0a",
              "extraction_id": "42a32702-d380-56e7-8309-6ab1efbcd479",
              "associatedQuery": "How do epigenetic changes affect aging?"
            }
          },
          {
            "id": "33f6a665-bb01-5c9f-9325-0f9acf312b54",
            "score": 0.7407828790182737,
            "metadata": {
              "text": "Epigenetic Dysregulation with Age",
              "title": "2007 - Aging Hematopoietic Stem Cells Decline in Function and Exhibit Epigenetic Dysregulation.pdf",
              "version": "v0",
              "chunk_order": 44,
              "document_id": "a6fabf0c-e4a5-59f6-82c5-ebabce24fd0a",
              "extraction_id": "eb735cbe-b7f5-56cb-8cd6-1f2fb2b990a4",
              "associatedQuery": "How do epigenetic changes affect aging?"
            }
          },
          {
            "id": "0aede05b-f0dd-595a-a11d-acac0970d25d",
            "score": 0.7407643044450478,
            "metadata": {
              "text": "Recently, studying the direct relationship between epigeneticmechanisms and the aging process itself is gaining increasing\nattention. The potential reversibility of these epigenetic\nchanges that occur as a hallmark of aging offers excitingopportunities to alter the trajectory of age-related diseases.\n8\nThis is especially important given the remarkable plasticityof aging.\n9,10In the literature, age-associated epigenetic alter-\nations have been identified by epigenome-wide association",
              "title": "2016 - Epigenetic drift in the aging genome a ten-year.pdf",
              "version": "v0",
              "chunk_order": 9,
              "document_id": "52f09ef3-4e4c-538f-909c-d28eb72d91f3",
              "extraction_id": "2f6d20f0-addc-51e8-979d-1aac7ac26694",
              "associatedQuery": "How do epigenetic changes affect aging?"
            }
          },
          {
            "id": "5e3a0748-9dc0-55b1-ac4d-d8b2291fa297",
            "score": 0.7391665798177299,
            "metadata": {
              "text": "in gene transcription and, as a consequence, translation as well as the stabilization or\ndegradation of molecular factors. While mechanisms underlying aging-related pathologies\nremain to be elucidated in detail, various studies demonstrate an epigenetic component.\nIn fact, the aforementioned epigenetic modications were shown to play essential roles\nin diseases including inammation, cancer, osteoporosis, neurodegenerative diseases,\nand diabetes.",
              "title": "2021 - Epigenetics of Aging and Aging-Associated Diseases.pdf",
              "version": "v0",
              "chunk_order": 144,
              "document_id": "70945353-4808-539a-80f9-5632c27913e5",
              "extraction_id": "e2bc9b8e-2349-509b-a148-fbd86f0455f4",
              "associatedQuery": "How do epigenetic changes affect aging?"
            }
          },
          {
            "id": "c35ad17b-fe97-5ce5-bae1-59fd08201a7b",
            "score": 0.7377009391784668,
            "metadata": {
              "text": "PLoS Biology | www.plosbiology.org August 2007 | Volume 5 | Issue 8 | e201 1759\nEpigenetic Dysregulation with Age",
              "title": "2007 - Aging Hematopoietic Stem Cells Decline in Function and Exhibit Epigenetic Dysregulation.pdf",
              "version": "v0",
              "chunk_order": 105,
              "document_id": "a6fabf0c-e4a5-59f6-82c5-ebabce24fd0a",
              "extraction_id": "0a77e94f-85de-5ab3-b23f-129b08839502",
              "associatedQuery": "How do epigenetic changes affect aging?"
            }
          },
          {
            "id": "dea115e3-3d9b-5d08-a604-ab227fcd1b71",
            "score": 0.7310480097248561,
            "metadata": {
              "text": "and increased expression of proaging genes such as the cell-cycle inhibitor p16, which drives cell senescence. Additional consequences of epigenetic dys-regulation include increased transcriptional noise and decreased coordination of gene expression that contributes to organismal aging.\nCell148, January 20, 2012 2012 Elsevier Inc. 53",
              "title": "2012 - Aging, Rejuvenation, and Epigenetic.pdf",
              "version": "v0",
              "chunk_order": 110,
              "document_id": "bde26feb-f423-51b0-89ec-6f079bfc8b17",
              "extraction_id": "0e274732-b0df-53b8-999b-30b798af92e2",
              "associatedQuery": "How do epigenetic changes affect aging?"
            }
          }
        ],
        "kg_search_results": null
      }
    }
  },
  "13": {
    "results": {
      "completion": {
        "id": "chatcmpl-AIFhpW2QcT6L6LqU3pI7kcz7hsxkv",
        "choices": [
          {
            "finish_reason": "stop",
            "index": 0,
            "message": {
              "content": "The gene FOXO3 plays a significant role in human longevity. Multiple studies have shown a strong association between variations in the FOXO3 gene and increased lifespan. For instance, the FOXO3A genotype has been strongly linked with human longevity, as demonstrated in studies by Willcox et al. (2008) and confirmed in various populations, including German and Southern Italian centenarians [1], [2], [3]. The FOXO3 locus is associated with extreme longevity in humans, particularly among centenarians [5]. Additionally, specific variants at the FOXO3 locus have been identified that respond to cellular stress, which may contribute to their role in promoting longevity [8].",
              "role": "assistant"
            }
          }
        ],
        "created": 1728913641,
        "model": "gpt-4o-2024-08-06",
        "object": "chat.completion",
        "system_fingerprint": "fp_6b68a8204b",
        "usage": {
          "completion_tokens": 138,
          "prompt_tokens": 1654,
          "total_tokens": 1792
        }
      },
      "search_results": {
        "vector_search_results": [
          {
            "id": "77c88648-7807-5606-8793-4389378a82fd",
            "score": 0.7730233073234558,
            "metadata": {
              "text": "27 Willcox, B. J. et al. 2008 FOXO3A genotype is\nstrongly associated with human longevity. Proc. Natl\nAcad. Sci. USA 105, 13 98713 992. ( doi:10.1073/\npnas.0801030105 )\n28 Flachsbart, F., Caliebe, A., Kleindorp, R., Blanche, H.,\nvon Eller-Eberstein, H., Nikolaus, S., Schreiber, S. &\nNebela, A. 2009 Association of FOXO3A variationwith human longevity conrmed in GermanGenomics of human longevity P . E. Slagboom et al. 41",
              "title": "2011 - Genomics of human longevity.pdf",
              "version": "v0",
              "chunk_order": 102,
              "document_id": "2e038219-fdaa-506f-9cd3-51379054130e",
              "extraction_id": "7c183ae5-f10e-5f0c-962e-32135887b3bd",
              "associatedQuery": "What is the role of the gene FOXO3 in longevity?"
            }
          },
          {
            "id": "9c463b71-be3a-5f01-bc6f-d1d29b7a162f",
            "score": 0.7644128343812936,
            "metadata": {
              "text": "3. Willcox BJ, Donlon TA, He Q et al (2008) FOXO3A genotype is\nstrongly associated with human longevity. Proc Natl Acad Sci\nUSA 105(37):1398713992. doi: 10.1073/pnas.0801030105\n4. Anselmi CV, Malovini A, Roncarati R et al (2009) Association of\nthe FOXO3A locus with extreme longevity in a southern Italian\ncentenarian study. Rejuvenation Res 12(2):95104. doi: 10.1089/\nrej.2008.0827\n5. Flachsbart F, Caliebe A, Kleindorp R et al (2009) Association of\nFOXO3A variation with human longevity conrmed in German",
              "title": "2015 - A Chromosome 13 locus is associated with male-specific mortality in mice.pdf",
              "version": "v0",
              "chunk_order": 64,
              "document_id": "ad8f2626-87fb-520e-8cef-ee9a9cc3ab0b",
              "extraction_id": "5cc56e3b-53ab-5299-814d-014e2ed31d2f",
              "associatedQuery": "What is the role of the gene FOXO3 in longevity?"
            }
          },
          {
            "id": "2f98af09-5895-545a-b36f-c05b70beee07",
            "score": 0.7606979751170903,
            "metadata": {
              "text": "are, in fact, part of the same insulin/IGF1/GH pathway(Fig. 1) that modulates lifespan across organisms (Ke-nyon, 2010). A strong association between FOXO3 and\nhuman longevity has been reported (Willcox et al., 2008)and subsequently validated in other populations (forreview, see Kenyon, 2010). FOXO3 was also associatedAGING GENES AS TARGETS FOR DRUG DISCOVERY 95",
              "title": "2012 - Genome-Environment Interactions That Modulate.pdf",
              "version": "v0",
              "chunk_order": 131,
              "document_id": "b1a1997c-e9df-5dc0-9d12-a3977d0c64ec",
              "extraction_id": "d59d7882-333d-5576-86ab-3cfa6354b946",
              "associatedQuery": "What is the role of the gene FOXO3 in longevity?"
            }
          },
          {
            "id": "c6e1f317-e421-5f6b-ab4e-034f1aa94ba1",
            "score": 0.756262748686945,
            "metadata": {
              "text": "Biogerontology 11:28797\n117. Willcox BJ, Donlon TA, He Q, Chen R, Grove JS, et al. 2008. FOXO3A genotype is strongly associated\nwith human longevity. Proc. Natl. Acad. Sci. USA 105:1398792\n118. Soerensen M, Dato S, Christensen K, McGue M, Stevnsner T, et al. 2010. Replication of an association\nof variation in the FOXO3A gene with human longevity using both case-control and longitudinal data.\nAging Cell 9:101017\n119. Mardis ER. 2011. A decades perspective on DNA sequencing technology. Nature 470:198203",
              "title": "2013 - Genome Instability and Aging.pdf",
              "version": "v0",
              "chunk_order": 200,
              "document_id": "71e08916-8cc8-5d96-8c06-4461b972b54d",
              "extraction_id": "3091bce3-8eb6-593d-8a92-ee3570e8e9a9",
              "associatedQuery": "What is the role of the gene FOXO3 in longevity?"
            }
          },
          {
            "id": "34dfec26-9828-56c8-be82-69eb114fa9e3",
            "score": 0.7560266110912939,
            "metadata": {
              "text": "FOXO3 locus is associated with extreme longevity in humans (centenarians) [2, \n58, 59].\nNRF/SKN-1 activates the expression of genes involved in protecting the cell in \nresponse to ROS, toxins, and metabolic changes through mTOR and insulin/IGF \nsignaling, and it is also dysregulated later in life [60, 61]. Increasing the levels of \nL. Garca-Velzquez and C. Arias",
              "title": "2020 - Clinical Genetics and Genomics of Aging.pdf",
              "version": "v0",
              "chunk_order": 95,
              "document_id": "62b635c3-040e-512a-b016-6ef295308a1e",
              "extraction_id": "68deea31-59de-5665-9c97-df57d72d0b52",
              "associatedQuery": "What is the role of the gene FOXO3 in longevity?"
            }
          },
          {
            "id": "6dd65017-bb91-5a1a-9d85-c1c1cfcd5780",
            "score": 0.7414736747741699,
            "metadata": {
              "text": "A. 2003;100:406671. https://doi.org/10.1073/pnas.2628028100.\n 24. van den Akker EB, Deelen J, Slagboom PE, Beekman M. Exome and whole genome \nsequencing in aging and longevity. Adv Exp Med Biol. 2015;847:12739. https://doi.\norg/10.1007/978-1-4939-2404-2_6.\n 25. Flachsbart F, etal. Association of FOXO3A variation with human longevity confirmed in \nGerman centenarians. Proc Natl Acad Sci U S A. 2009;106:27005. https://doi.org/10.1073/\npnas.0809594106.\nA. Garca-Venzor and E. A. Mandujano-Tinoco",
              "title": "2020 - Clinical Genetics and Genomics of Aging.pdf",
              "version": "v0",
              "chunk_order": 750,
              "document_id": "62b635c3-040e-512a-b016-6ef295308a1e",
              "extraction_id": "7555b8ec-cf4e-54a4-b654-6ae7e63d150c",
              "associatedQuery": "What is the role of the gene FOXO3 in longevity?"
            }
          },
          {
            "id": "160acccd-d5c5-5e54-8f88-ada1d413e91b",
            "score": 0.7410400355482136,
            "metadata": {
              "text": "X.L.,  2009.  Genetic  association  of  FOXO1A  and  FOXO3A  with  longevity  trait  in\nHan  Chinese  populations.  Hum.  Mol.  Genet.  18,  48974904.\nLunetta,  K.L.,  DAgostino  Sr.,  R.B.,  Karasik,  D.,  Benjamin,  E.J.,  Guo,  C.Y.,  Govindaraju,\nR.,  Kiel,  D.P.,  Kelly-Hayes,  M.,  Massaro,  J.M.,  Pencina,  M.J.,  Seshadri,  S.,  Murabito,\nJ.M.,  2007.  Genetic  correlates  of  longevity  and  selected  age-related  phenotypes:",
              "title": "2011 - A genome-wide association study confirms APOE as the major gene influencing.pdf",
              "version": "v0",
              "chunk_order": 97,
              "document_id": "63b27b06-db2c-5542-9b1a-cb9ebe64d339",
              "extraction_id": "210aa417-372c-5bf6-b961-e281a1817458",
              "associatedQuery": "What is the role of the gene FOXO3 in longevity?"
            }
          },
          {
            "id": "aceb74e0-8b79-587f-9dd0-e260eeb90ab5",
            "score": 0.732019305229187,
            "metadata": {
              "text": "theFOXO3 locus is not surprising, since this locus was previously\nreported in the longevity GWA study from the CHARGE con-\nsortium\n7, from which many cohorts are included in these meta-\nanalyses. So far, three functional longevity-associated variants\nhave been identi ed at the FOXO3 locus (rs2802292, rs12206094,\nand rs4946935). For all of them, an allele-speci c response to\ncellular stress was observed. Consistently, the longevity-associated\nalleles of all three variants were shown to induce FOXO3",
              "title": "2019 - A meta-analysis of genome-wide association.pdf",
              "version": "v0",
              "chunk_order": 88,
              "document_id": "9d36fc35-9708-5d1a-9514-9ce3469d7591",
              "extraction_id": "68c41fe5-4413-5cfc-846b-a0097f994bcd",
              "associatedQuery": "What is the role of the gene FOXO3 in longevity?"
            }
          },
          {
            "id": "049ee89e-2f05-595b-9112-725976cb4ab3",
            "score": 0.7297561925794976,
            "metadata": {
              "text": "exceptional longevity with no significant genetic contribution. Interestingly, the \nauthors found that FOXO3A, a longevity allele, may not be related to healthy aging \nphenotype [29].\nAging is a complex process usually accompanied by the onset of different dis-\neases like neurodegenerative disorders (Alzheimers disease and Parkinsons dis-\nease), cardiovascular illnesses, and cancer. The study of the genetic basis of these \naging-related diseases is another approach in the study of the genomic basis of",
              "title": "2020 - Clinical Genetics and Genomics of Aging.pdf",
              "version": "v0",
              "chunk_order": 654,
              "document_id": "62b635c3-040e-512a-b016-6ef295308a1e",
              "extraction_id": "bdfc934a-d31b-57e4-9a78-15c719049c4f",
              "associatedQuery": "What is the role of the gene FOXO3 in longevity?"
            }
          },
          {
            "id": "f6636c31-1105-5ea2-9b3b-ae8b21e08bee",
            "score": 0.7286234498023987,
            "metadata": {
              "text": "centenarians. Proc Natl Acad Sci USA 106(8):27002705. doi: 10.\n1073/pnas.0809594106\n6. Li Y, Wang WJ, Cao H et al (2009) Genetic association of\nFOXO1A and FOXO3A with longevity trait in Han Chinese\npopulations. Hum Mol Genet 18(24):48974904. doi: 10.1093/\nhmg/ddp459\n7. Soerensen M, Dato S, Christensen K et al (2010) Replication of\nan association of variation in the FOXO3A gene with human\nlongevity using both case-control and longitudinal data. AgingCell 9(6):10101017. doi: 10.1111/j.1474-9726.2010.00627.x",
              "title": "2015 - A Chromosome 13 locus is associated with male-specific mortality in mice.pdf",
              "version": "v0",
              "chunk_order": 65,
              "document_id": "ad8f2626-87fb-520e-8cef-ee9a9cc3ab0b",
              "extraction_id": "5cc56e3b-53ab-5299-814d-014e2ed31d2f",
              "associatedQuery": "What is the role of the gene FOXO3 in longevity?"
            }
          }
        ],
        "kg_search_results": null
      }
    }
  },
  "14": {
    "results": {
      "completion": {
        "id": "chatcmpl-AIFhuoRML5l0E69TztcoQUZAgCOF2",
        "choices": [
          {
            "finish_reason": "stop",
            "index": 0,
            "message": {
              "content": "The environment interacts with genes to influence aging through several mechanisms:\n\n1. **Signaling Pathways and Gene Expression**: Environmental factors can trigger signaling pathways and modulate gene expression, impacting aging. For example, certain genes have varying effects on lifespan depending on environmental factors like diet [1].\n\n2. **Epigenetic Modifications**: Environmental factors can lead to changes in DNA methylation, acetylation, or deacetylation of histones, which are epigenetic modifications that influence gene expression. These changes can result in cellular damage and accelerated aging, such as the shortening of telomeres [2], [6].\n\n3. **Epigenetics as a Link**: Epigenetic modifications provide a potential link between the environment, disease, and aging. This suggests that specific environmental factors might directly induce specific epigenetic changes, which could be targeted for interventions aimed at improving healthspan or promoting healthy aging [3].\n\n4. **Physiological Capacity and Longevity**: Environmental factors, along with protective genetic alleles, contribute to an individual's physiological capacity, indirectly determining healthy lifespan and longevity. For instance, caloric restriction and smoking have opposite effects on the rate of aging [4].\n\n5. **Plasticity of Aging**: Aging is a plastic process that can be manipulated by both genetic and environmental factors. Understanding these interactions can help identify targets for anti-aging therapies, potentially through diet, lifestyle, and pharmacological interventions [5], [10].\n\nOverall, the interaction between the environment and genes is complex and involves multiple pathways and mechanisms that collectively influence the aging process.",
              "role": "assistant"
            }
          }
        ],
        "created": 1728913646,
        "model": "gpt-4o-2024-08-06",
        "object": "chat.completion",
        "system_fingerprint": "fp_6b68a8204b",
        "usage": {
          "completion_tokens": 321,
          "prompt_tokens": 1106,
          "total_tokens": 1427
        }
      },
      "search_results": {
        "vector_search_results": [
          {
            "id": "a4773f1a-f2d3-5950-a81e-d22357e97a0f",
            "score": 0.7851045387858702,
            "metadata": {
              "text": "of multiple genes with each other and withthe environment. Evidence from animal systems showsa major impact of the environment on aging, yet envi-ronmental manipulations of aging act through genesand proteins, usually by triggering signaling pathwaysand modulating gene expression. In fact, some geneshave been shown in model organisms to have varyingeffects on lifespan depending on diet (Heikkinen et al.,2009). Genes that can regulate aging in model organ-isms cannot be directly applied to humans through",
              "title": "2012 - Genome-Environment Interactions That Modulate.pdf",
              "version": "v0",
              "chunk_order": 18,
              "document_id": "b1a1997c-e9df-5dc0-9d12-a3977d0c64ec",
              "extraction_id": "68b12e10-d643-570a-aa63-eda62de81928",
              "associatedQuery": "How does the environment interact with genes to influence aging?"
            }
          },
          {
            "id": "3d657599-d2c8-518d-aee3-46c0643a88ec",
            "score": 0.7439095087159265,
            "metadata": {
              "text": "Several studies show the influence of the environment on the ageing process [24]. \nEnvironmental factors may affect homeostasis and lead to the development of dis-\neases, thus affecting the quality of life in older age [25]. They also produce cellular \ndamage, which causes an accelerated shortening of the telomeres at the genetic \nlevel, accompanied by changes in DNA methylation, acetylation or deacetylation \nof histones, among others. Altogether, these changes induce an aberrant gene",
              "title": "2020 - Clinical Genetics and Genomics of Aging.pdf",
              "version": "v0",
              "chunk_order": 1707,
              "document_id": "62b635c3-040e-512a-b016-6ef295308a1e",
              "extraction_id": "53f4a434-3d4b-5ae5-a788-9b32c94fc0ab",
              "associatedQuery": "How does the environment interact with genes to influence aging?"
            }
          },
          {
            "id": "49127379-fac4-525a-bf90-5c3bae66860a",
            "score": 0.7258800431561457,
            "metadata": {
              "text": "changes are generated during the aging process. For a long time it\nhas been believed that epigenetic modications occurring during\naging may depend on environmental factors. This idea is attractive\nbecause, if true, epigenetics could provide a link between the\nenvironment, disease and aging. It also opens the possibility of\ntargeted intervention aimed, for example, at improving healthspan\nor healthy aging. Thus, the rst question is whether specic\nenvironmental factors can directly induce specic epigenetic",
              "title": "2009 - The role of epigenetics in aging and age-related diseases.pdf",
              "version": "v0",
              "chunk_order": 63,
              "document_id": "f7b452fc-0115-5582-b0c0-c2829f090e9d",
              "extraction_id": "5a231e39-0927-56e5-970c-e86d69490a69",
              "associatedQuery": "How does the environment interact with genes to influence aging?"
            }
          },
          {
            "id": "7ce9af40-0bf8-58e1-ad7c-cd55ba0a7cf8",
            "score": 0.7208803677296558,
            "metadata": {
              "text": "In addition, environmental factors influence the\norganism s ability to withstand the increase in entropy\nwith aging: for example, caloric restriction and\nsmoking can exert opposite effects on the rate ofaging (Colman et al. 2009 ; Fraser and Shavlik 2001 ).\nBoth protective alleles and a benevolent environment\ncontribute to excess physiological capacity, which in\nturn indirectly determines an individual s healthy life\nspan and longevity (Martin et al. 2007 ). The well-",
              "title": "2011 - How pleiotropic genetics of the musculoskeletal system.pdf",
              "version": "v0",
              "chunk_order": 8,
              "document_id": "ed31486c-a651-5894-bd96-21fbd78f2646",
              "extraction_id": "677eccb2-c51a-59dc-9690-7d02c51fbb7a",
              "associatedQuery": "How does the environment interact with genes to influence aging?"
            }
          },
          {
            "id": "3f37774f-e56b-5350-93e8-371948bf3e23",
            "score": 0.7131877328995574,
            "metadata": {
              "text": "to humans through ge-netic manipulations for numerous legal, ethical, andtechnical reasons. If we could understand how the envi-ronment modulates these aging-related genes, we mightbe able to create antiaging therapies applicable to hu-mans, potentially through diet, lifestyle, and even phar-macological interventions. Therefore, understanding ge-nome-environment interactions in the context of agingcan be a powerful approach to identify attractive targetsfor drug design.",
              "title": "2012 - Genome-Environment Interactions That Modulate.pdf",
              "version": "v0",
              "chunk_order": 19,
              "document_id": "b1a1997c-e9df-5dc0-9d12-a3977d0c64ec",
              "extraction_id": "68b12e10-d643-570a-aa63-eda62de81928",
              "associatedQuery": "How does the environment interact with genes to influence aging?"
            }
          },
          {
            "id": "3466f905-760d-5d0b-a3e1-b39f506e6289",
            "score": 0.7015897035598805,
            "metadata": {
              "text": "ing human life span have been identified [2,3]. At the\nsame time, there is a growing realization that environ-\nmental factors are major contributors to aging and age-\nassociated illness. Epigenetics is the study of chemical\nmodifications of the genome, heritable by cell progeny,\nand it has been an attractive target for studies of aging\nand environmentally influenced disease. Several groups\nhave shown differences in DNA methylation - a covalent",
              "title": "2015 - Age and sun exposure-related widespread.pdf",
              "version": "v0",
              "chunk_order": 4,
              "document_id": "acf06062-9ca8-50be-a543-ef3b34ad6ad3",
              "extraction_id": "aa62f800-8e83-5033-889a-64ff3f453fca",
              "associatedQuery": "How does the environment interact with genes to influence aging?"
            }
          },
          {
            "id": "3c369292-4b9c-5156-a80f-4b3301026f30",
            "score": 0.7009334742466538,
            "metadata": {
              "text": "al., 2009; Stanfel et al., 2009). Many of these genesmodulate the response to environmental signals, such asfood availability, and act in signaling pathways that ifunderstood can be targeted (Fig. 1). The genetic regula-tion of aging is therefore an emerging field with multipleapplications in the human nutrition, cosmetic, and phar-maceutical industries.\nAGING GENES AS TARGETS FOR DRUG DISCOVERY 91",
              "title": "2012 - Genome-Environment Interactions That Modulate.pdf",
              "version": "v0",
              "chunk_order": 67,
              "document_id": "b1a1997c-e9df-5dc0-9d12-a3977d0c64ec",
              "extraction_id": "a01ca925-4ccf-5863-a162-7bd4c754fe89",
              "associatedQuery": "How does the environment interact with genes to influence aging?"
            }
          },
          {
            "id": "c1b5a31a-066d-571b-af1f-db746d9d17f6",
            "score": 0.6962083163137259,
            "metadata": {
              "text": "standing the cause and mechanisms of aging is imperative in\nassisting to suppress age-related diseases and promote healthylongevity. It is well-known that aging is influenced by a combin-\nation of genetic and environmental factors. Previous twin stud-\nies have shown that the genetic contribution to general human\nlongevity is about 2030% [ 4,5], whereas environmental factors\nin human aging and longevity still account for the largest effect.\nEpigenetic factors influence the regulation of gene expres-",
              "title": "2016 - Progress on the role of DNA methylation in aging.pdf",
              "version": "v0",
              "chunk_order": 4,
              "document_id": "e4cdc02f-4415-5638-aab8-f848b4d64a22",
              "extraction_id": "bcb3c620-b960-5af6-95ea-13215c31672e",
              "associatedQuery": "How does the environment interact with genes to influence aging?"
            }
          },
          {
            "id": "90f9e09f-f339-5d59-ae24-fcbdd2ca6ceb",
            "score": 0.6893077677900942,
            "metadata": {
              "text": "known to affect the function of epigenetic regulators, this may be an example of how aging interacts\nwith our genome to inuence AD development.",
              "title": "2018 - Genomics New Light on Alzheimer?s.pdf",
              "version": "v0",
              "chunk_order": 95,
              "document_id": "940593d2-04c3-59b9-a5bf-976febbc6f71",
              "extraction_id": "05bcb479-ca17-57eb-9674-1c2fecb5726c",
              "associatedQuery": "How does the environment interact with genes to influence aging?"
            }
          },
          {
            "id": "c44c36ad-fcca-540a-a4f3-3965e48e3948",
            "score": 0.6884725465211217,
            "metadata": {
              "text": "consequently the incidence of age-related diseasessuch as heart disease, cancer, and neurodegenerativediseases, is projected to increase considerably in thecoming decades. Findings from model organisms haverevealed that aging is a surprisingly plastic processthat can be manipulated by both genetic and environ-mental factors. Here we review a broad range of find-ings in model organisms, from environmental to ge-netic manipulations of aging, with a focus on thosewith underlying gene-environment interactions",
              "title": "2012 - Genome-Environment Interactions That Modulate.pdf",
              "version": "v0",
              "chunk_order": 4,
              "document_id": "b1a1997c-e9df-5dc0-9d12-a3977d0c64ec",
              "extraction_id": "df213743-7428-59be-ba19-2563f8ce5c70",
              "associatedQuery": "How does the environment interact with genes to influence aging?"
            }
          }
        ],
        "kg_search_results": null
      }
    }
  },
  "15": {
    "results": {
      "completion": {
        "id": "chatcmpl-AIFi4Qsa1GjY5azJi3IYJdr8DLXln",
        "choices": [
          {
            "finish_reason": "stop",
            "index": 0,
            "message": {
              "content": "Senescent cells are cells that have stopped dividing and have entered a state of permanent growth arrest. They are characterized by an altered metabolism and the secretion of pro-inflammatory factors, a phenomenon known as the senescence-associated secretory phenotype (SASP) [1], [3]. These cells accumulate in tissues over time and are resistant to apoptosis, meaning they are not easily cleared from the body [9].\n\nSenescent cells contribute to aging and age-related diseases in several ways. They impact the tissue environment by secreting inflammatory cytokines, proteases, and growth factors, which can lead to chronic inflammation and tissue dysfunction [3], [4]. This chronic inflammation is a significant factor in the development of age-related degenerative diseases [1], [4]. Additionally, senescent cells can alter the tissue microenvironment, promoting the degeneration of organs and stem cell niches, and potentially stimulating cancer cell growth [6].\n\nThe accumulation of senescent cells is associated with various age-related pathologies, such as atherosclerosis, osteoarthritis, and Alzheimer's disease [5], [9]. Recent studies have shown that clearing senescent cells can prevent or delay tissue dysfunction and extend health span, highlighting their causative role in aging [5].",
              "role": "assistant"
            }
          }
        ],
        "created": 1728913656,
        "model": "gpt-4o-2024-08-06",
        "object": "chat.completion",
        "system_fingerprint": "fp_a20a4ee344",
        "usage": {
          "completion_tokens": 249,
          "prompt_tokens": 1323,
          "total_tokens": 1572
        }
      },
      "search_results": {
        "vector_search_results": [
          {
            "id": "2f35de05-41ee-5471-870d-a4e663cf32f6",
            "score": 0.6795354271488873,
            "metadata": {
              "text": "senescence, exhausting the ability for a tissue to regenerate after injury, impacting mitochondrial function,and inducing protein aggregation. Senescent cells have altered metabolism, and they can secreteproinammatory factors and alter the local tissue environment, thereby contributing to aging andage-related degenerative diseases. In addition, stem cell function can be impacted by DNA damage by bothcell autonomous and nonautonomous mechanisms. Proper function of mitochondria is dependent upongenome",
              "title": "2018 - Nuclear Genomic Instability.pdf",
              "version": "v0",
              "chunk_order": 124,
              "document_id": "54d28a91-8db6-56b1-baaa-b67274c93a36",
              "extraction_id": "4b00515d-e599-5ce1-84e3-012d7efe1a30",
              "associatedQuery": "What are senescent cells and how do they contribute to aging?"
            }
          },
          {
            "id": "1efa76cb-2289-5dd3-9fa5-776083aa5cd5",
            "score": 0.6562108602398132,
            "metadata": {
              "text": "[87] and the accumulation of senescent cells in human\ntissues with age has been implicated as a driver of aging-\nrelated diseases. Indeed, pharmacological approaches\ntargeting senescent cells, like senolytics, are a major and\ntimely area of research that could result in human clin-\nical applications [ 5,88]. It is imperative that we fully\nunderstand and deconstruct cellular senescence in order\nto target aging-related diseases. We hope that CellAge\nwill help researchers understand the role that CS plays",
              "title": "2020 - A multidimensional systems biology.pdf",
              "version": "v0",
              "chunk_order": 153,
              "document_id": "d040bfe3-e409-5b5c-b8f8-f3dd4fc060e3",
              "extraction_id": "95744ef5-34b9-5540-a5e5-01fd580539e6",
              "associatedQuery": "What are senescent cells and how do they contribute to aging?"
            }
          },
          {
            "id": "9faa9b6b-6a97-5979-bf49-8bbdb4bb383d",
            "score": 0.6410194467282955,
            "metadata": {
              "text": "An important source of inflammatory signals in aged organ-\nisms is thought to be the accumulation of senescent cells across\ntissues [ 5,7]. Indeed, accumulating evidence has shown that\nsenescent cells are characterized by a senescence-associatedsecretory phenotype [ 810], which includes a panoply of\npro-inflammatory cytokines, proteases, growth factors and\nmetabolites [ 10,11]. The impact of senescent cells on age-related\ninflammation, and their potential role as a target for pro-",
              "title": "2022 - Functional genomics of inflamm-aging.pdf",
              "version": "v0",
              "chunk_order": 8,
              "document_id": "435dc081-e3d1-52c5-93a1-caa11206422f",
              "extraction_id": "1635dbe1-1dcb-5213-9446-74129d50c5f8",
              "associatedQuery": "What are senescent cells and how do they contribute to aging?"
            }
          },
          {
            "id": "6d4a1a0b-2af3-5cc4-b7c0-a7223ce3edfa",
            "score": 0.6345081140247575,
            "metadata": {
              "text": "senescent cells [150]. SASP factors exert their functions in either an autocrine or a \nparacrine manner and are responsible for the induction of the chronic inflammation \nand cell proliferation that contributes to cell dysfunction and cancer. Thus, the accu-\nmulation of senescent cells in tissue is closely associated with aging-related dis-\neases. Recently, it was determined that senescent fibroblasts significantly increase \nthe expression of HLA-E, which inhibits the receptor NKG2A in killer cells, and",
              "title": "2020 - Clinical Genetics and Genomics of Aging.pdf",
              "version": "v0",
              "chunk_order": 152,
              "document_id": "62b635c3-040e-512a-b016-6ef295308a1e",
              "extraction_id": "6a2a94de-cfc0-50eb-b50e-bf3a0f813c78",
              "associatedQuery": "What are senescent cells and how do they contribute to aging?"
            }
          },
          {
            "id": "45f74737-847a-52c2-a0b9-bf9de335a7ce",
            "score": 0.6322256922721907,
            "metadata": {
              "text": "atherosclerosis, osteoarthritis, sarcopenia, ulcer formation, cancer, and Alzheimer disease, which\nis suggestive of a causative role. However, the most convincing evidence that senescent cells causeaging comes from recent genetic (85) and pharmacologic studies (86) revealing that clearance of\nsenescent cells can prevent or delay tissue dysfunction and extend health span.\nSenescent cells induce autocrine, as well as paracrine, signaling by secretion of proinamma-",
              "title": "2018 - Nuclear Genomic Instability.pdf",
              "version": "v0",
              "chunk_order": 120,
              "document_id": "54d28a91-8db6-56b1-baaa-b67274c93a36",
              "extraction_id": "2b1396d1-ea5d-5708-a6b1-2adf1712c7b4",
              "associatedQuery": "What are senescent cells and how do they contribute to aging?"
            }
          },
          {
            "id": "bd5fffd3-cf7a-5f67-b581-6cb803a48de4",
            "score": 0.627122245602475,
            "metadata": {
              "text": "senescence can deplete both stem (5153) and stromal\n(10,11) cell pools. Moreover, because senescent cellspersist, they have the ability to alter the tissue micro-environment, and can therefore also promote the degen-eration of organs and stem cell niches (14,46). Finally,\nsenescent cells secrete factors such as matrix metallopro-\nteinase-3 (MMP-3), which favors extra-cellular matrixremodeling, promotes defects in epithelial cell dierentia-tion and stimulates cancer cell growth (46,54,55).",
              "title": "2007 - Two faces of p53 aging and tumor suppression.pdf",
              "version": "v0",
              "chunk_order": 39,
              "document_id": "b1ef905a-c145-5270-9110-ae6954ea3d72",
              "extraction_id": "4a95fed4-61db-58e9-96d7-3a9dcf87ef7f",
              "associatedQuery": "What are senescent cells and how do they contribute to aging?"
            }
          },
          {
            "id": "27d74137-3987-571d-87ab-2c12ec66d1f7",
            "score": 0.6243986115218048,
            "metadata": {
              "text": "potential role of senescence in in vivo  aging and disease has been difficult to assess and somewhat controversial  \n[146]. However, recent studies have shown that senescent cells accumulate in normal arterial tissue over the \nlifespan of humans  [147, 148]. Likewise, the accumulation of senescent cells has been reported in diseased \ntissues, such as atherosclerotic plaques  [149] and abdominal aortic aneurysms  [150]. Baker et al.  showed that",
              "title": "2015 - Cellular and Molecular Biology of Aging Endothelial Cells.pdf",
              "version": "v0",
              "chunk_order": 67,
              "document_id": "815d7f3e-e219-502f-aba0-57a68ae787d3",
              "extraction_id": "10f1fcbd-35a6-507d-880f-1f3f303737ea",
              "associatedQuery": "What are senescent cells and how do they contribute to aging?"
            }
          },
          {
            "id": "180adffa-397c-599b-adb3-64a7f464aaaa",
            "score": 0.616710030036568,
            "metadata": {
              "text": "51. Jeyapalan JC, Ferreira M, Sedivy JM, Herbig U. 2007. Accumulation of senescent cells in mitotic tissue\nof aging primates. Mech. Ageing Dev. 128:3644\n52. Boyle J, Kill IR, Parris CN. 2005. Heterogeneity of dimer excision in young and senescent human dermal\nbroblasts. Aging Cell 4:24755\n53. Seluanov A, Mittelman D, Pereira-Smith OM, Wilson JH, Gorbunova V. 2004. DNA end joining\nbecomes less efcient and more error-prone during cellular senescence. PNAS 101:762429",
              "title": "2018 - Nuclear Genomic Instability.pdf",
              "version": "v0",
              "chunk_order": 211,
              "document_id": "54d28a91-8db6-56b1-baaa-b67274c93a36",
              "extraction_id": "029ae7be-b0ab-55f8-84a2-5a74681e454d",
              "associatedQuery": "What are senescent cells and how do they contribute to aging?"
            }
          },
          {
            "id": "93b3cc74-a414-5097-802a-7dc2ad10171d",
            "score": 0.6157262554331877,
            "metadata": {
              "text": "in many accelerated-aging mouse models and in a plethora of human age-associated pathologies,\nincluding osteoporosis, atherosclerosis, glomerular disease, diabetic venous ulcers, chronic ob-structive pulmonary disease and emphysema, osteoarthritis, herniated intervertebral discs, and\nvascular calcication (112). Senescent cells are resistant to apoptosis and accumulate exponen-\ntially with age as a consequence of inefcient clearance. Unlike apoptotic tissues, senescent tissues\n436 VermeijHoeijmakersPothof",
              "title": "2016 - Genome Integrity in Aging.pdf",
              "version": "v0",
              "chunk_order": 84,
              "document_id": "85d5fcbb-5385-5a01-8139-d11fc8b1fe3a",
              "extraction_id": "102fcfb3-b333-5b67-ab94-08033f04ba5c",
              "associatedQuery": "What are senescent cells and how do they contribute to aging?"
            }
          },
          {
            "id": "3593241d-677d-5042-a1e9-dd92760a8c0e",
            "score": 0.6138603321397117,
            "metadata": {
              "text": "wound healing [ 8], and immune clearance [ 9,10]. By\ncontrast, the gradual accumulation and chronic\npersistence of senescent cells with time promotes dele-\nterious effects that are considered to accelerate deterior-\nation and hyperplasia in aging [ 11]. Senescent cells\nsecrete a cocktail of inflammatory and stromal regula-\ntorsdenoted as the senescence-associated secretory\nphenotype, or SASP which adversely impact neighbor-\ning cells, the surrounding extracellular matrix, and other",
              "title": "2020 - A multidimensional systems biology.pdf",
              "version": "v0",
              "chunk_order": 11,
              "document_id": "d040bfe3-e409-5b5c-b8f8-f3dd4fc060e3",
              "extraction_id": "fe4ec57e-6ae7-59c4-b8fa-da73fe77ce96",
              "associatedQuery": "What are senescent cells and how do they contribute to aging?"
            }
          }
        ],
        "kg_search_results": null
      }
    }
  },
  "16": {
    "results": {
      "completion": {
        "id": "chatcmpl-AIFjcxBUYW02ZQfh6Ogs975bGNDi2",
        "choices": [
          {
            "finish_reason": "stop",
            "index": 0,
            "message": {
              "content": "Yes, there are known lifestyle interventions that can positively impact genes related to aging. Dietary interventions, such as dietary restriction (DR) and calorie restriction, have been shown to alter patterns of DNA methylation and induce long-lasting changes in gene expression that improve health during aging and extend lifespan [1], [8]. These interventions can modify the epigenome, which is linked to the biology of aging [5]. Additionally, glucose restriction has been shown to extend human cellular lifespan through SIRT1-mediated epigenetic and genetic mechanisms [7].",
              "role": "assistant"
            }
          }
        ],
        "created": 1728913752,
        "model": "gpt-4o-2024-08-06",
        "object": "chat.completion",
        "system_fingerprint": "fp_6b68a8204b",
        "usage": {
          "completion_tokens": 108,
          "prompt_tokens": 1348,
          "total_tokens": 1456
        }
      },
      "search_results": {
        "vector_search_results": [
          {
            "id": "5fc33fac-ab39-5ec1-9fb9-dcaa93a595d3",
            "score": 0.7159391854983329,
            "metadata": {
              "text": "Dietary interventions, including starvation and protein\ndeprivation, can also alter patterns of DNA methyla-\ntion, potentially in a long-lasting manner [42, 43],\nincluding transgenerationally [26, 44].\nDietary, genetic and pharmacological interventions\nthat improve health during aging and extend lifespan\ninduce long-lasting changes in gene expression that\nmediate their effects. Here we have asked if and how\nage-related DNA methylation, transcription and lipid",
              "title": "2017 - Dietary restriction protects from age-associated DNA methylation and induces epigenetic reprogramming of lipid metabolism.pdf",
              "version": "v0",
              "chunk_order": 13,
              "document_id": "fe573bb0-3d37-55e5-93fa-65b3fbc5f532",
              "extraction_id": "6364d669-4b96-5d2f-8ce8-526b065dce72",
              "associatedQuery": "Are there any known lifestyle interventions that can positively impact genes related to aging?"
            }
          },
          {
            "id": "4bf7307d-d8a0-5594-b0b5-487fe0f265ca",
            "score": 0.700632593054551,
            "metadata": {
              "text": "Longev. Heal. 2, 10 (2013).\n7. Kreienkamp Ret al.Doubled lifespan and patient-like pathologies in progeria mice fed high-fat diet. \nAging Cell18, e12852 (2019). [PubMed: 30548460] \n8. Heilbronn LK & Ravussin E Calorie restriction and aging: review of the literature and implications \nfor studies in humans. Am. J. Clin. Nutr. 78, 361369 (2003). [PubMed: 12936916] \n9. Liang Yet al.Calorie restriction is the most reasonable anti-ageing intervention: a meta-analysis of",
              "title": "2021 - Gene-by-environment modulation of lifespan and weight gain in the murine BXD family.pdf",
              "version": "v0",
              "chunk_order": 132,
              "document_id": "4d082da4-fa48-5170-8147-c4fea47a5d4b",
              "extraction_id": "30ba3324-6e19-58c2-9e32-508f827af3e5",
              "associatedQuery": "Are there any known lifestyle interventions that can positively impact genes related to aging?"
            }
          },
          {
            "id": "afc304d1-dd43-55ec-811d-27ca27fc4e5d",
            "score": 0.69575321934401,
            "metadata": {
              "text": "a medical intervention), without changing the fundamental rateof organismal aging. Nevertheless, it does seem that manyso-called longevity genes, as well as dietary restriction,\nappear to extend not only life span, but also health span\n(Kauffman et al., 2010; Luo et al., 2010 ). In that regard, it does\nappear that it is possible to experimentally slow the rate of aging.\nStill, in each case, aging does continue on as if there is some",
              "title": "2012 - Aging, Rejuvenation, and Epigenetic.pdf",
              "version": "v0",
              "chunk_order": 13,
              "document_id": "bde26feb-f423-51b0-89ec-6f079bfc8b17",
              "extraction_id": "b9f038dd-97af-51ea-bb32-d73bf66c3dcb",
              "associatedQuery": "Are there any known lifestyle interventions that can positively impact genes related to aging?"
            }
          },
          {
            "id": "3fc1603d-dd9e-5bcf-96e6-6b927d344be1",
            "score": 0.6918727571888421,
            "metadata": {
              "text": "As we describe above, a small but growing number ofinterventions has been shown to reproducibly increase\nlifespan in laboratory animals and, in a few cases, to also\ndelay or reverse age-related declines in multiple organsystems. These healthy aging interventions could, in prin-\nciple, be tested to determine whether they also increase\nlifespan and promote healthspan in dogs (Table 1). There\nare several questions that immediately present themselves\nwhen considering the design of a healthy aging interven-",
              "title": "2016 - The dog aging project translational geroscience in companion.pdf",
              "version": "v0",
              "chunk_order": 50,
              "document_id": "e841c6bd-78b8-56e1-b3dd-e2bcc8a0f590",
              "extraction_id": "e433208e-665d-550c-b8e8-c9fb400f1159",
              "associatedQuery": "Are there any known lifestyle interventions that can positively impact genes related to aging?"
            }
          },
          {
            "id": "7ca45b81-3f97-5b1b-9a84-84cfffc4cc08",
            "score": 0.6913794485145646,
            "metadata": {
              "text": "be linked to the biology of stem cell quiescence and self-renewal.\nAlthough genetic and environmental interventions have clearly\nproven to be effective in prolonging life span, we postulate thatthose interventions, as well as the rejuvenating interventions\ndescribed above, are, in fact, acting primarily to modify theepigenome. Consistent with this, genetic interventions directlytargeting the epigenome can extend life span ( Greer et al.,\n2010 ). Studying aging and rejuvenation through the lens of",
              "title": "2012 - Aging, Rejuvenation, and Epigenetic.pdf",
              "version": "v0",
              "chunk_order": 116,
              "document_id": "bde26feb-f423-51b0-89ec-6f079bfc8b17",
              "extraction_id": "915ca931-d49d-5837-97fd-f06c145764d0",
              "associatedQuery": "Are there any known lifestyle interventions that can positively impact genes related to aging?"
            }
          },
          {
            "id": "c96b67f8-ad31-50fd-b053-07b127938ef2",
            "score": 0.6899251732252514,
            "metadata": {
              "text": "During the past century, remarkable progress has been \nmade in unveiling the mechanisms of aging. Genetic and molecular pathways that regulate healthspan and lifespan have been identified in various model organisms, provid-ing a rich knowledge base (Longo etal. 2015; Lopez-Otin etal. 2013, 2016; Singh etal. 2019). However, the focus on",
              "title": "2020 - Mitonuclear genomics and aging.pdf",
              "version": "v0",
              "chunk_order": 10,
              "document_id": "e05fdc09-c8d8-5134-a1fd-bf07a1564981",
              "extraction_id": "e6fb876b-e91c-505a-aa16-7b428ec61f10",
              "associatedQuery": "Are there any known lifestyle interventions that can positively impact genes related to aging?"
            }
          },
          {
            "id": "193d98c7-8d37-5f83-b1b2-84aee242f079",
            "score": 0.6873417515166182,
            "metadata": {
              "text": "205. Li, Y.; Tollefsbol, T.O. p16INK4a Suppression by Glucose Restriction Contributes to Human Cellular Lifespan Extension through\nSIRT1-Mediated Epigenetic and Genetic Mechanisms. PLoS ONE 2011 ,6, e17421. [CrossRef]\n206. Daniel, M.; Tollefsbol, T.O. Epigenetic linkage of aging, cancer and nutrition. J. Exp. Biol. 2015 ,218, 5970. [CrossRef]\n207. Kapahi, P .; Kaeberlein, M.; Hansen, M. Dietary restriction and lifespan: Lessons from invertebrate models. Ageing Res. Rev. 2017 ,\n39, 314. [CrossRef]",
              "title": "2021 - Epigenetics of Aging and Aging-Associated Diseases.pdf",
              "version": "v0",
              "chunk_order": 259,
              "document_id": "70945353-4808-539a-80f9-5632c27913e5",
              "extraction_id": "9770f6f4-b86a-514f-9cce-c23d2963aeae",
              "associatedQuery": "Are there any known lifestyle interventions that can positively impact genes related to aging?"
            }
          },
          {
            "id": "7460a40c-8723-5de9-9f2e-c781f4872f1f",
            "score": 0.6848835553194155,
            "metadata": {
              "text": "as diabetes, cancer and neurodegenerative disorders\n[1, 2]. Environmental and genetic interventions can\nameliorate the effects of aging, with nutrition,\nnutrient-sensing signaling networks and metabolism\nplaying evolutionarily conserved roles [1, 3 5]. Diet-\nary restriction (DR), in which food intake is reducedwhile avoiding malnutrition, extends lifespan in di-\nverse model and non-model organisms [3, 6]. DR\ninduces a remarkably broad-spectrum improvement in",
              "title": "2017 - Dietary restriction protects from age-associated DNA methylation and induces epigenetic reprogramming of lipid metabolism.pdf",
              "version": "v0",
              "chunk_order": 4,
              "document_id": "fe573bb0-3d37-55e5-93fa-65b3fbc5f532",
              "extraction_id": "21efa872-9d89-5dee-9dd1-27dcaa1208cf",
              "associatedQuery": "Are there any known lifestyle interventions that can positively impact genes related to aging?"
            }
          },
          {
            "id": "38c89363-89a1-56d5-82f2-28c19fa0fbcb",
            "score": 0.6846686805685964,
            "metadata": {
              "text": "53. Mair W & Dillin A Aging and survival: the genetics of life span extension by dietary restriction. \nAnnu. Rev. Biochem. 77, 727754 (2008). [PubMed: 18373439] \n54. Masoro EJCaloric restriction-induced life extension of rats and mice: a critique of proposed \nmechanisms. Biochim. Biophys. Acta1790, 10401048 (2009). [PubMed: 19250959] \n55. Weindruch R, Walford RL, Fligiel S & Guthrie D The retardation of aging in mice by dietary",
              "title": "2021 - Gene-by-environment modulation of lifespan and weight gain in the murine BXD family.pdf",
              "version": "v0",
              "chunk_order": 149,
              "document_id": "4d082da4-fa48-5170-8147-c4fea47a5d4b",
              "extraction_id": "bca61863-81b3-5ef7-850d-10cc9577a9e1",
              "associatedQuery": "Are there any known lifestyle interventions that can positively impact genes related to aging?"
            }
          },
          {
            "id": "b9240ab4-370f-5bc1-8c33-9755ab788aac",
            "score": 0.682794431364705,
            "metadata": {
              "text": "In addition to genes associated with aging, research has\nfocused on identifying genes associated with the life-\nextending effects of CR. One method is to identify genesthat decrease or cancel out the life-extending effects of CRwhen mutated (Gems et al., 2002; Bishop and Guarente,2007). More than 100 such genes have been identified inmodel organisms (D. Wuttke, C. Vora, J. P. de Magalhes,unpublished observations). The growth hormone receptor(GHR) is the only gene so far identified in mammals that",
              "title": "2012 - Genome-Environment Interactions That Modulate.pdf",
              "version": "v0",
              "chunk_order": 68,
              "document_id": "b1a1997c-e9df-5dc0-9d12-a3977d0c64ec",
              "extraction_id": "13ca8905-ddbb-5437-b6a8-4012969daa43",
              "associatedQuery": "Are there any known lifestyle interventions that can positively impact genes related to aging?"
            }
          }
        ],
        "kg_search_results": null
      }
    }
  },
  "17": {
    "results": {
      "completion": {
        "id": "chatcmpl-AIFji2gbFHCW8aj8mLegsooXneEeb",
        "choices": [
          {
            "finish_reason": "stop",
            "index": 0,
            "message": {
              "content": "The 'epigenetic clock' is a molecular biomarker of aging that is based on the DNA methylation levels of specific CpG sites. These methylation patterns are highly correlated with an individual's chronological age, with a robust correlation coefficient of approximately 0.9 for individuals aged between 20 and 100 years [1]. The epigenetic clock serves as a reliable predictor of biological age, which refers to how well a person's body functions compared to their chronological age [2]. \n\nIn aging research, the epigenetic clock is used to estimate the biological age of cells, tissues, or organs by analyzing the methylation levels of select CpGs, often referred to as clock CpGs [8]. This estimated age, known as the epigenetic age, can indicate different aging rates between individuals with the same chronological age, providing insights into the biological basis of aging [9]. The epigenetic clock has been applied in various studies to understand the relationship between epigenetic aging and factors such as metabolism, and it is considered one of the most promising molecular estimators of biological age [6], [8].",
              "role": "assistant"
            }
          }
        ],
        "created": 1728913758,
        "model": "gpt-4o-2024-08-06",
        "object": "chat.completion",
        "system_fingerprint": "fp_6b68a8204b",
        "usage": {
          "completion_tokens": 225,
          "prompt_tokens": 1260,
          "total_tokens": 1485
        }
      },
      "search_results": {
        "vector_search_results": [
          {
            "id": "e2522b52-d927-5c1a-8569-8fcb706ecc1e",
            "score": 0.7772643102845974,
            "metadata": {
              "text": "vided one of the most reliable aging biomarkers. An epigenetic clock is a group of \nCpG sites with particular methylation patterns that are highly related to the chrono-\nlogical age of an individual. This correlation is very robust (r=0.9) for individuals \nbetween 20 and 100years. The epigenetic clock is a breakthrough discovery that \nwill allow novel experimental approaches to understand the biological basis of \naging [113]. For example, by using the epigenetic clock as a measure of cellular",
              "title": "2020 - Clinical Genetics and Genomics of Aging.pdf",
              "version": "v0",
              "chunk_order": 729,
              "document_id": "62b635c3-040e-512a-b016-6ef295308a1e",
              "extraction_id": "a81cc7a6-0cc6-5909-9192-ac0fab26fbc2",
              "associatedQuery": "What is the 'epigenetic clock,' and how is it used in aging research?"
            }
          },
          {
            "id": "c76f4517-c117-56e6-96b9-218f0fdae9f3",
            "score": 0.7516819982718244,
            "metadata": {
              "text": "Epigenetic Clock\nChronological age is the number of years a person has lived, and biological or phys-\niological age refers to a measure of how well your body functions compared to your \nchronological age. Biological age is influenced by multiple factors (genes, lifestyle, \nbehavior, environment, among others) and correlates with mortality and health sta-\ntus. The epigenetic clock is one potentially reliable predictor of biological age.",
              "title": "2020 - Clinical Genetics and Genomics of Aging.pdf",
              "version": "v0",
              "chunk_order": 1002,
              "document_id": "62b635c3-040e-512a-b016-6ef295308a1e",
              "extraction_id": "63c7bfe5-a409-5435-91ea-487534957b81",
              "associatedQuery": "What is the 'epigenetic clock,' and how is it used in aging research?"
            }
          },
          {
            "id": "4edf498a-20de-593a-b301-73c799b07691",
            "score": 0.7424924680599662,
            "metadata": {
              "text": "Background\nEpigenetic clocks are sets of CpG dinucleotides whose DNA methylation (DNAm) can\nbe used to accurately predict a person s chronological age [ 1]. In recent years, various\nepigenetic clocks have been developed [ 25]. Well-known examples are the clocks de-\nveloped by Hannum et al., trained on blood samples and containing 71 CpGs [ 2], and\nHorvath, a multi-tissue predictor consisting of 353 CpGs [ 3]. A popular application of",
              "title": "2022 - Functional genomics analysis identifies.pdf",
              "version": "v0",
              "chunk_order": 8,
              "document_id": "1fe1c748-9e73-51ba-8521-de924cc133d4",
              "extraction_id": "6d7c1694-2c53-554c-9070-2db848fc5a42",
              "associatedQuery": "What is the 'epigenetic clock,' and how is it used in aging research?"
            }
          },
          {
            "id": "99532996-c835-534a-b6e7-a2f95ec00e2c",
            "score": 0.7356219291687012,
            "metadata": {
              "text": "An EpigeneticClock\nThe aging transcriptome could be used to gauge the physiological \nage of worms, and in that way serve as an epigenetic clock revealing \nhow much of life span has been spent and how much remains (23). \nMiddle-aged worms show an aging transcriptome half-way between \nthe aging expression profiles of young and old worms. This provides \nan independent way to assess the age of an animal independent of \nits life span. This is important as there are at least 2 explanations to",
              "title": "2021 - Career Retrospective Tom Johnson?Genetics, Genomics.pdf",
              "version": "v0",
              "chunk_order": 30,
              "document_id": "f3a26f44-f5af-5b2b-aa1c-aec2fd99f17e",
              "extraction_id": "c6cc3d8b-3736-5fe8-a4ff-eb186679a37e",
              "associatedQuery": "What is the 'epigenetic clock,' and how is it used in aging research?"
            }
          },
          {
            "id": "cb09d819-b809-5844-a111-5c7c7b9f9a99",
            "score": 0.7303161025047302,
            "metadata": {
              "text": "The epigenetic aging clock measures the sum of all the age-related \npathways affecting cellular physiology in old age. The aging epigen-\netic clock is heavily enriched for germline- and intestinal-expressed \ngenes, but lack muscle- and neuronal-expressed genes (23, 25). \nExpression changes in the germline and intestine were expected as \nthere are massive changes in the morphology of gonad at the end of \nfertility and the intestine in old age. The aging transcriptome pro-",
              "title": "2021 - Career Retrospective Tom Johnson?Genetics, Genomics.pdf",
              "version": "v0",
              "chunk_order": 33,
              "document_id": "f3a26f44-f5af-5b2b-aa1c-aec2fd99f17e",
              "extraction_id": "c6cc3d8b-3736-5fe8-a4ff-eb186679a37e",
              "associatedQuery": "What is the 'epigenetic clock,' and how is it used in aging research?"
            }
          },
          {
            "id": "2d08a161-7a62-5d3f-b300-1ca93ee5b751",
            "score": 0.7297257619932761,
            "metadata": {
              "text": "etic mouse aging and may be used to inform future studies in other model organisms and humans \nfocused on studying the relationship between epigenetic aging and metabolism.\nIntroduction\nEpigenetic clocks are widely used molecular biomarkers of aging (Horvath and Raj, 2018). These \nDNA methylation (DNAm) age predictors are based on the methylation levels of select CpGs that are RESEARCH ARTICLE\n*For correspondence: \nkmozhui@uthsc.edu\nCompeting interest: See page \n22\nFunding: See page 22",
              "title": "2021 -Mozhui- Epigenetic aging.pdf",
              "version": "v0",
              "chunk_order": 6,
              "document_id": "d23daa43-4176-54e6-b3c3-b889843e92f1",
              "extraction_id": "6dfd0c51-91dd-5bb3-b7ae-a9c86ea22c35",
              "associatedQuery": "What is the 'epigenetic clock,' and how is it used in aging research?"
            }
          },
          {
            "id": "66c03d04-0af5-50e5-8d4a-9a645493db46",
            "score": 0.7297257619932761,
            "metadata": {
              "text": "etic mouse aging and may be used to inform future studies in other model organisms and humans \nfocused on studying the relationship between epigenetic aging and metabolism.\nIntroduction\nEpigenetic clocks are widely used molecular biomarkers of aging (Horvath and Raj, 2018). These \nDNA methylation (DNAm) age predictors are based on the methylation levels of select CpGs that are RESEARCH ARTICLE\n*For correspondence: \nkmozhui@uthsc.edu\nCompeting interest: See page \n22\nFunding: See page 22",
              "title": "2021 - Genetic loci and metabolic states associated with murine epigenetic aging.pdf",
              "version": "v0",
              "chunk_order": 6,
              "document_id": "b82bd9e1-2373-577b-a942-164565eaca6b",
              "extraction_id": "68ee1ea3-5caf-5df5-8efc-134943a456cb",
              "associatedQuery": "What is the 'epigenetic clock,' and how is it used in aging research?"
            }
          },
          {
            "id": "35c83256-6072-5e6a-b15e-0cae1991b034",
            "score": 0.7288637484474534,
            "metadata": {
              "text": "estimators epigenetic clocks; telomere length; transcriptomic-, proteomic-, and \nmetabolomic-based estimators; and composite biomarkers concluded that the epi-\ngenetic clock is the most promising molecular estimator of biological age [26]. \nEpigenetic age estimators are sets of CpGs (also known as clock CpGs) that are \ncoupled with a mathematical algorithm to estimate the age of a DNA source, such \nas cells, tissues, or organs. This estimated age, also referred to as epigenetic age or",
              "title": "2020 - Clinical Genetics and Genomics of Aging.pdf",
              "version": "v0",
              "chunk_order": 333,
              "document_id": "62b635c3-040e-512a-b016-6ef295308a1e",
              "extraction_id": "8f22afaf-a5fb-5f44-9fc2-18d4aeceede7",
              "associatedQuery": "What is the 'epigenetic clock,' and how is it used in aging research?"
            }
          },
          {
            "id": "39dfbf42-78ec-5b0a-8448-55f47c22830e",
            "score": 0.7156729271606859,
            "metadata": {
              "text": "proved epigenetic clock. It should be noted that building\na biological age predictor is difficult since there is no\nclear definition of biological age. Nevertheless, one of\nthe essential features of biological age is its ability to in-\ndicate the different ageing rates between individuals with\nthe same chronological age. A previous study has re-\nported a number of CpG sites that show variation in the\nlongitudinal changing rates between individuals [ 40].",
              "title": "2019 - Improved precision of epigenetic clock.pdf",
              "version": "v0",
              "chunk_order": 69,
              "document_id": "556d0179-023f-581f-9c2d-febe4e75722f",
              "extraction_id": "487cf1b1-1190-5d14-8b24-ba92f75aa6aa",
              "associatedQuery": "What is the 'epigenetic clock,' and how is it used in aging research?"
            }
          },
          {
            "id": "d5ae06ad-3d88-5c4f-972a-0510d2fc67f3",
            "score": 0.7122355103492737,
            "metadata": {
              "text": "ranging from 0.15 to 0.19 [ 8,9]. Individuals with epigenetic clock estimates greater than\ntheir chronological age display age acceleration and have been shown to be at a\ngreater risk of all-cause mortality and multiple adverse health outcomes [ 10]. Conse-\nquently, identification of genetic and environmental contributors to the variation in\nthese measures in populations has become a major goal in the field [ 11].\nThe first generation of epigenetic aging clocks used penalized regression models to",
              "title": "2021 - Genome-wide association studies identify.pdf",
              "version": "v0",
              "chunk_order": 16,
              "document_id": "60c2e869-1fee-53ea-b332-26d9c2abc747",
              "extraction_id": "53db6715-4f12-50ad-8fb9-acba4e2f4f37",
              "associatedQuery": "What is the 'epigenetic clock,' and how is it used in aging research?"
            }
          }
        ],
        "kg_search_results": null
      }
    }
  },
  "18": {
    "results": {
      "completion": {
        "id": "chatcmpl-AIFlJI3UdMQFpFb34IWrf77DCGGJY",
        "choices": [
          {
            "finish_reason": "stop",
            "index": 0,
            "message": {
              "content": "Researchers use model organisms like yeast and worms to study human aging due to their simpler genomes, short lifespans, and the ease with which they can be genetically and environmentally manipulated. These characteristics make them ideal for identifying and characterizing genes and signaling pathways involved in aging [3]. \n\nYeast, specifically Saccharomyces cerevisiae, is a highly informative model for aging studies because of its genetic tools and the ability to measure aging through replicative or chronological lifespan assays [2], [5]. Yeast has been extensively used to identify genes and interventions responsible for lifespan extension, providing insights into the aging processes of all eukaryotic organisms [10].\n\nSimilarly, the nematode Caenorhabditis elegans is another widely used model organism in biogerontology. Researchers study these organisms to understand whether the aging process is evolutionarily conserved and to what degree mechanisms in these simpler organisms can be indicative of aging mechanisms in humans [1], [6]. These model organisms help explore both genetic and environmental determinants of lifespan, contributing to hypotheses surrounding extended lifespan and healthspan [7].",
              "role": "assistant"
            }
          }
        ],
        "created": 1728913857,
        "model": "gpt-4o-2024-08-06",
        "object": "chat.completion",
        "system_fingerprint": "fp_6b68a8204b",
        "usage": {
          "completion_tokens": 220,
          "prompt_tokens": 1188,
          "total_tokens": 1408
        }
      },
      "search_results": {
        "vector_search_results": [
          {
            "id": "f125dd77-ecd5-59aa-9cf0-ae89333159d2",
            "score": 0.7750034928321838,
            "metadata": {
              "text": "the nematode Caenorhabditis elegans , and the budding yeast \nSaccharomyces cerevisiae , have emerged as the most widely \nused and, hence, best characterized, model organisms in bio-\ngerontology. \n When considering the use of simple eukaryotes to study \naging and age-related disease, it is pertinent to ask whether, and to what degree, the aging process is evolutionarily con-\nserved. Does a yeast cell age by the same mechanism(s) as a",
              "title": "2007 - Longevity Genomics Across Species.pdf",
              "version": "v0",
              "chunk_order": 3,
              "document_id": "1ab0b63f-d97c-5f5c-98ee-0bde785fa630",
              "extraction_id": "789baa84-292c-547a-8600-8568f34ea9de",
              "associatedQuery": "How do researchers use model organisms like yeast or worms to study human aging?"
            }
          },
          {
            "id": "35414229-a946-525c-b508-4b8f49a2702c",
            "score": 0.769577466534897,
            "metadata": {
              "text": "Studies on the aging of mammals are rather limited by the long life\nspan of the commonly used model organisms. Thus, both nonverte-brate and invertebrate organisms, with their shorter life span and ease\nof genetic and environmental manipulations, gained popularity amongresearchers in the aging field as experimental models for aging studies.\nAmong them, budding yeast or Saccharomyces cerevisiae is a highly in-\nformative organismal model for aging studies with its genetic tools,",
              "title": "2016 - Epigenetics and aging.pdf",
              "version": "v0",
              "chunk_order": 17,
              "document_id": "71b206ec-81bd-5194-8b21-ae522f8cbc2d",
              "extraction_id": "6b4e8da8-7d82-5531-b1a8-21935a6f4a51",
              "associatedQuery": "How do researchers use model organisms like yeast or worms to study human aging?"
            }
          },
          {
            "id": "1b3d84fb-c799-5d19-b3bd-a9032b7980fc",
            "score": 0.743890903708068,
            "metadata": {
              "text": "Abstract\nCellular models such as yeasts are a driving force in biogerontology studies. Their simpler genome, short lifespans and vast\ngenetic and genomics resources make them ideal to characterise pro-ageing and anti-ageing genes and signalling pathways.Over the last three decades, yeasts have contributed to the understanding of fundamental aspects of lifespan regulation\nincluding the roles of nutrient response, global protein translation rates and quality, DNA damage, oxidative stress,",
              "title": "2022 - Genome-wide screens in yeast models towards.pdf",
              "version": "v0",
              "chunk_order": 1,
              "document_id": "be6ba237-7f13-5aac-8cb1-6a43e82d326b",
              "extraction_id": "6977180a-bec7-5d39-ba97-b0fd664b6c96",
              "associatedQuery": "How do researchers use model organisms like yeast or worms to study human aging?"
            }
          },
          {
            "id": "5caecfbd-14ef-59e2-a281-2bc524ca0353",
            "score": 0.7376859188079834,
            "metadata": {
              "text": "usually chosen for convenience rather than for specific features \napplicable to human aging. Hence, choosing the suitable animal model to answer the specific question we aim to understand is \nof high importance in these types of studies. Among the most \nprevalent aging model organisms are Saccharomyces cerevisiae , \nCaenorhabditis elegans, Drosophila melanogaster, and Mus mus -\nculus . As a single-celled organism, S. cerevisiae is easily grown,",
              "title": "2018 - Genomic Instabilities, Cellular Senescence, and Aging In Vitro, In Vivo and Aging-Like Human Syndromes.pdf",
              "version": "v0",
              "chunk_order": 23,
              "document_id": "7de8d462-8a3c-5625-8cbb-374f3bb46425",
              "extraction_id": "fbb608d4-8b2b-502d-88cc-d1fcb0b89afb",
              "associatedQuery": "How do researchers use model organisms like yeast or worms to study human aging?"
            }
          },
          {
            "id": "c14402ec-2ad7-5857-9f09-39c71656bf0f",
            "score": 0.7347005605697632,
            "metadata": {
              "text": "mammalian genes that affect aging than any other model organism. Aging in yeast is assayed primarily by\nmeasurement of replicative or chronological life span. Here, we review the genes and mechanisms implicated\nin these two aging model systems and key remaining issues that need to be addressed for their optimization.",
              "title": "2012 - Replicative and Chronological Aging.pdf",
              "version": "v0",
              "chunk_order": 2,
              "document_id": "496e387e-4278-5f74-8ecc-4edc1cee7dfe",
              "extraction_id": "cb936ad1-1a4f-5b1c-89f1-f8991f059969",
              "associatedQuery": "How do researchers use model organisms like yeast or worms to study human aging?"
            }
          },
          {
            "id": "c103f3f8-b155-5787-bdd9-16f9d390369d",
            "score": 0.7263469912513197,
            "metadata": {
              "text": "be more exaggerated in more distantly related species (such \nas the worm and mouse models). There are, however, simi -\nlarities between aged humans and aged model organisms; \nthey all tend to have decreasing overall fitness, and there -\nfore, studies using model organisms continue as they may \nbe at least indicative of some aging mechanisms in humans.\nExtensions to life span in model organisms are mostly \nassociated with disruption to fundamental metabolic path -",
              "title": "2012 - Genomics and Successful Aging Grounds for Renewed.pdf",
              "version": "v0",
              "chunk_order": 49,
              "document_id": "6d2b82c3-4256-562a-9b23-ff7c71e9fd93",
              "extraction_id": "3d18e792-3d83-5cc3-b9ab-309322ecf55d",
              "associatedQuery": "How do researchers use model organisms like yeast or worms to study human aging?"
            }
          },
          {
            "id": "b19ebe3b-e87e-5cab-baef-24deddd303bb",
            "score": 0.7236888408660889,
            "metadata": {
              "text": "eukaryote model organisms, namely yeast, worms, ies,and sh, as well as mice and rats, to explore both genetic\nand environmental determinants of lifespan. While these\nshort-lived models have each yielded a number of fasci-\nnating ndings and insights into hypotheses surrounding\nextended lifespan and healthspan, they may also haveconstrained this complex, multifactorial eld to areas in\nwhich they are best suited, most notably short-term inter-",
              "title": "2016 - Unraveling the message insights into comparative genomics.pdf",
              "version": "v0",
              "chunk_order": 17,
              "document_id": "0deba7bb-c27a-5d9e-b1b2-e48a5574882c",
              "extraction_id": "0dc4e4f4-cd48-5ccd-991b-edd688533b5e",
              "associatedQuery": "How do researchers use model organisms like yeast or worms to study human aging?"
            }
          },
          {
            "id": "c32f3dbe-95d5-531a-9165-d4da7b2dc2a8",
            "score": 0.7108061525465895,
            "metadata": {
              "text": "et al., 2010 ). These effects require an intact germline, andTable 2. Repositories and Tools for Aging Research\nModels Description Link/Reference\nYeast Saccharomyces genome database http://www.yeastgenome.org/\npublished lifespan data http://lifespandb.sageweb.org/ (McCormick et al., 2015 )\nWilcoxon rank sum test to test signicance of lifespan\ndifferenceshttp://data.kaeberleinlab.org/scripts/ranksum.php\nyeast outgrowth data analyzer (YODA) for chronological\nlifespan assayshttp://yoda.sageweb.org/",
              "title": "2016 - Epigenetic Mechanisms of Longevity and Aging.pdf",
              "version": "v0",
              "chunk_order": 26,
              "document_id": "588185a0-e157-552f-a304-4beefb85d398",
              "extraction_id": "5ff88a27-4525-5e7f-99bd-e781cedd8005",
              "associatedQuery": "How do researchers use model organisms like yeast or worms to study human aging?"
            }
          },
          {
            "id": "91375d45-be1d-5c54-8d0f-a9b1dded69bb",
            "score": 0.7103649377822876,
            "metadata": {
              "text": "for molecular biological studies on aging. Although material \nfrom humans should be employed where possible, for prac- \ntical reasons animal model systems like rats and mice are \nindispensible. There is evidence that, provided their health sta- \ntus and husbandry is optimal, rodents age much in the same \nway as humans do (Burek 1978). For studying certain funda- \nmental processes, such as the occurrence of various types of \nDNA rearrangement, lower organisms and cell lines can also",
              "title": "1989 - Genetic instability and aging theories, facts, and future perspectives.pdf",
              "version": "v0",
              "chunk_order": 117,
              "document_id": "4d5b1800-b676-5865-a555-09ea740cc14a",
              "extraction_id": "b0531531-f629-512b-9835-24cc870b4ef3",
              "associatedQuery": "How do researchers use model organisms like yeast or worms to study human aging?"
            }
          },
          {
            "id": "ae5be149-52ad-5854-b40a-c24374545cf0",
            "score": 0.7083187103271484,
            "metadata": {
              "text": "short life span, and fully sequenced genome (20 ,21). Despite being uni-\ncellular, yeast has been an excellent model to identify and characterize\nconserved basic biological processes, including aging. Yeast has beenextensively used to identify genes and interventions responsible for lifespan extension and to gain insights into the aging processes of all eu-\nkaryotic organisms. In parallel, over the years, studies on invertebrate\norganisms, such as Drosophila melanogaster (flies) and Caenorhabditis",
              "title": "2016 - Epigenetics and aging.pdf",
              "version": "v0",
              "chunk_order": 18,
              "document_id": "71b206ec-81bd-5194-8b21-ae522f8cbc2d",
              "extraction_id": "6b4e8da8-7d82-5531-b1a8-21935a6f4a51",
              "associatedQuery": "How do researchers use model organisms like yeast or worms to study human aging?"
            }
          }
        ],
        "kg_search_results": null
      }
    }
  },
  "19": {
    "results": {
      "completion": {
        "id": "chatcmpl-AIFlT2nob40QrExWjGMMqZ4fSc8yC",
        "choices": [
          {
            "finish_reason": "stop",
            "index": 0,
            "message": {
              "content": "Yes, there are promising anti-aging therapies being developed based on genetic research. Several approaches are being explored:\n\n1. **Senolytic Drugs**: Research has shown that abolishing senescent cells through genetic manipulation or senolytic drugs can significantly halt biological aging in mice. Trials are underway to test the ability of senolytics to postpone age-associated pathologies in humans [3].\n\n2. **Genetic Discoveries in Aging**: A number of genes capable of altering the aging process have been identified in animal models and even in humans. This area of research is promising as it explores the association of multiple alleles with human exceptional longevity [6].\n\n3. **Manipulation of Aging-Related Genes**: There is potential in manipulating aging-related genes through diet, lifestyle, and pharmaceuticals to improve human health and develop drugs against age-related diseases such as cancer, heart disease, type 2 diabetes, obesity, and neurodegenerative diseases [8].\n\nThese developments indicate that genetic research is paving the way for potential anti-aging therapies.",
              "role": "assistant"
            }
          }
        ],
        "created": 1728913867,
        "model": "gpt-4o-2024-08-06",
        "object": "chat.completion",
        "system_fingerprint": "fp_6b68a8204b",
        "usage": {
          "completion_tokens": 208,
          "prompt_tokens": 1345,
          "total_tokens": 1553
        }
      },
      "search_results": {
        "vector_search_results": [
          {
            "id": "78733c6a-d870-5154-9128-eb66291fa967",
            "score": 0.7031545573792666,
            "metadata": {
              "text": "need to develop approaches and therapies targeting theaging process and age-related diseases (Butler et al.,2008). Delaying the process of aging, even slightly,would have profound social, medical and economic ben-efits (Olshansky et al., 2006; Butler et al., 2008). Forexample, slowing aging by a mere 7 years would cutmortality of age-related diseases by half at every age.Therefore, the potential benefits from research on thebasic biology and genetics of aging are unparalleled interms of improving quality",
              "title": "2012 - Genome-Environment Interactions That Modulate.pdf",
              "version": "v0",
              "chunk_order": 14,
              "document_id": "b1a1997c-e9df-5dc0-9d12-a3977d0c64ec",
              "extraction_id": "68b12e10-d643-570a-aa63-eda62de81928",
              "associatedQuery": "Are there any promising anti-aging therapies being developed based on genetic research?"
            }
          },
          {
            "id": "9da7c5dc-0deb-577c-bb22-83f987bd76dd",
            "score": 0.6974615815327716,
            "metadata": {
              "text": "raises the possibility of therapies to slow aging.  Therefore the discoveryof a gerontogene with even very rare mutations that increased longevitywould cause speculation about future trends in mortality.    However, thediscovery of such a gene would be relevant only to long-term (and, there-fore, very speculative) projections.\nProspective Epidemiologic Surveys that Include Genetic Information\nSome epidemiologic cohort studies of populations have collected",
              "title": "2001 - Demography in the age of genomics.pdf",
              "version": "v0",
              "chunk_order": 784,
              "document_id": "0f07fa43-feb6-5656-b7e7-b8faa86f5623",
              "extraction_id": "e3014138-3d5b-58bc-a1a5-5ac6f04cac1c",
              "associatedQuery": "Are there any promising anti-aging therapies being developed based on genetic research?"
            }
          },
          {
            "id": "3c636897-c47e-505d-9203-306124b73e0e",
            "score": 0.6919307892363904,
            "metadata": {
              "text": "Interestingly, when senescent cells are abolished either through genetic manipulation or via senolytic\ndrugs, biological aging is signicantly halted in mice [ 53,54]. Therefore, trials are now under way to\ntest the ability of senolytics to postpone age-associated pathologies in humans [ 55]. Notably, multi-\nple drugs are being pursued that either directly or indirectly impact DNA repair or the consequenceof DNA damage.\nFuture Prospects: Developing Interventions through DNA Repair",
              "title": "2020 - Protecting the Aging Genome.pdf",
              "version": "v0",
              "chunk_order": 57,
              "document_id": "bb774030-2570-5596-b2ab-b8f57ff81086",
              "extraction_id": "e5067ce2-69a6-5433-bed4-b95daeaa691e",
              "associatedQuery": "Are there any promising anti-aging therapies being developed based on genetic research?"
            }
          },
          {
            "id": "265126e3-2a4d-518f-93cf-21a201747eef",
            "score": 0.6850976911012499,
            "metadata": {
              "text": "5. Goldman DP, etal. Substantial health and economic returns from delayed aging may warrant \na new focus for medical research. Health Aff (Millwood). 2013;32(10):1698705.\n 6. Esplin ED, Oei L, Snyder MP.Personalized sequencing and the future of medicine: discov-\nery, diagnosis and defeat of disease. Pharmacogenomics. 2014;15(14):177190.\n 7. Marian AJ.Clinical applications of molecular genetic discoveries. Transl Res. 2016;168:614.",
              "title": "2020 - Clinical Genetics and Genomics of Aging.pdf",
              "version": "v0",
              "chunk_order": 1774,
              "document_id": "62b635c3-040e-512a-b016-6ef295308a1e",
              "extraction_id": "822571e2-b05d-5e17-9eaa-431151851111",
              "associatedQuery": "Are there any promising anti-aging therapies being developed based on genetic research?"
            }
          },
          {
            "id": "dcc13291-f18b-5094-83b6-4609322bc242",
            "score": 0.6829540322450489,
            "metadata": {
              "text": "J.L. Kirkland, Barriers to the Preclinical Development of Therapeutics that Target\nAging Mechanisms, J. Gerontol. A Biol. Sci. Med Sci. 71 (11) (2016) 1388 1394 .\n[2]D.J. Baker, B.G. Childs, M. Durik, M.E. Wijers, C.J. Sieben, J. Zhong, R.A. Saltness,\nK.B. Jeganathan, G.C. Verzosa, A. Pezeshki, K. Khazaie, J.D. Miller, J.M. van\nDeursen, Naturally occurringp16(Ink4a)-positive cells shorten healthy lifespan,\nNature 530 (7589) (2016) 184 189.",
              "title": "2018 - Spontaneous DNA damage to the nuclear genome promotes senescence.pdf",
              "version": "v0",
              "chunk_order": 136,
              "document_id": "08be7274-78a3-5e93-9e8c-3d4f6dbeacf9",
              "extraction_id": "005e73b5-7a93-53ff-946c-735fb4588de5",
              "associatedQuery": "Are there any promising anti-aging therapies being developed based on genetic research?"
            }
          },
          {
            "id": "1c4286b6-ede2-568b-9c18-b1e99ede17a6",
            "score": 0.6753207832192489,
            "metadata": {
              "text": "series of recent breakthroughs, a number of genes capable ofaltering the aging process as a whole  or at least to a largedegree  have been identified in animal models and even a fewin humans (Finch & Ruvkun, 2001; de Magalhes, 2005; Kenyon,2005). Furthermore, multiple alleles have been examined fortheir association with human exceptional longevity (Vijg & Suh,2005). This is a fascinating and important area of research, yetthere are now so many genes being associated with aging andlongevity that keeping",
              "title": "2009 - The Human Ageing Genomic Resources online.pdf",
              "version": "v0",
              "chunk_order": 10,
              "document_id": "e43cd3b6-ad8e-5422-ba7c-ceb6e66cc529",
              "extraction_id": "7ada6b55-99c2-5e20-bf96-d153f927256c",
              "associatedQuery": "Are there any promising anti-aging therapies being developed based on genetic research?"
            }
          },
          {
            "id": "2c5241f1-1655-5e36-a787-b966767b2534",
            "score": 0.6704605018543647,
            "metadata": {
              "text": "pharmaceutical and other interventions for human aging based on research that starts with the genomic information required to sustain adaptation, and thus health, in older fruit flies [36-39].   \n \nNaturally, any such genomic short-cut to reverse-engineering the evolution of slowed aging from fruit flies to humans is fraught with potential for error.  Such \nevolutionarily deep orthologies are sure to supply",
              "title": "2009 - Adaptation, aging, and genomic information.pdf",
              "version": "v0",
              "chunk_order": 70,
              "document_id": "54a993af-b86b-5cc3-a04b-bab03c244534",
              "extraction_id": "c2a8f947-44f2-5100-99e5-9c3a2f1284e9",
              "associatedQuery": "Are there any promising anti-aging therapies being developed based on genetic research?"
            }
          },
          {
            "id": "f20fd517-5f05-53ca-93a5-916bc891ad92",
            "score": 0.6675249336615335,
            "metadata": {
              "text": "century. Manipulation of aging-related genes by diet,lifestyle, and pharmaceuticals could dramatically im-prove human health and could be used to develop drugsagainst age-related diseases such as cancer, heart dis-ease, type 2 diabetes, obesity, and neurodegenerativediseases. The hundreds of aging-related genes and genesrelated to CR already identified offer enormous oppor-tunities for target discovery (Fig. 2). Although aging-related genes cannot be modified in humans, under-standing how these can be",
              "title": "2012 - Genome-Environment Interactions That Modulate.pdf",
              "version": "v0",
              "chunk_order": 170,
              "document_id": "b1a1997c-e9df-5dc0-9d12-a3977d0c64ec",
              "extraction_id": "8650652a-1765-563b-a98e-2e9336bcf29a",
              "associatedQuery": "Are there any promising anti-aging therapies being developed based on genetic research?"
            }
          },
          {
            "id": "69681eeb-6629-5091-b2b4-b4444e570913",
            "score": 0.6667904853599408,
            "metadata": {
              "text": "[7] Hughes, S.E., Evason, K., Xiong, C., Kornfeld, K. Genetic and \npharmacological factors that influence reproductive aging in nema-\ntodes. PLoS Genet.  2007 , 3: e25. \n[8] Vijg, J., Campisi, J. Puzzles, promises and a cure for ageing. Na-\nture 2008 , 454: 1065-1071. \n[9] Rolland, Y., Czerwinski, S., Abellan Van Kan, G., Morley, J.E., \nCesari, M., Onder, G., Woo, J., Baumgartner, R., Pillard, F., Boirie, \nY., Chumlea, W.M., Vellas, B. Sarcopenia: its assessment, etiol-",
              "title": "2009 - MicroRNAs in C. elegans Aging Molecular Insurance for Robustness.pdf",
              "version": "v0",
              "chunk_order": 98,
              "document_id": "dff49223-ac74-5419-a190-a0c7f43a5ee5",
              "extraction_id": "c8d6f90d-a25c-590a-a546-4500df09aa28",
              "associatedQuery": "Are there any promising anti-aging therapies being developed based on genetic research?"
            }
          },
          {
            "id": "5d8cc04f-7e13-5dbc-80c2-a35643954e9a",
            "score": 0.6647262576711921,
            "metadata": {
              "text": "for the aging process during the 20th Century. Thissituation poses a fundamental challenge to anti-aging\nmedicine: how to develop effective therapies for a\ngenomically complex pathology. We propose such astrategy. As a rst step, we recommend the use of modelsystems in which signicant genetic intervention is not\nproscribed or impractical. Second, we propose that work",
              "title": "2002 - Pharmacology, Genomics, and the Evolutionary Biology.pdf",
              "version": "v0",
              "chunk_order": 1,
              "document_id": "1bc636a3-6ce0-5fea-b549-0dae90a78f1b",
              "extraction_id": "6c9e1997-bfe6-5708-a476-07c833eed8fa",
              "associatedQuery": "Are there any promising anti-aging therapies being developed based on genetic research?"
            }
          }
        ],
        "kg_search_results": null
      }
    }
  },
  "20": {
    "results": {
      "completion": {
        "id": "chatcmpl-AIFlaWmRr9Bl0RbY7T3uxkIANSmkA",
        "choices": [
          {
            "finish_reason": "stop",
            "index": 0,
            "message": {
              "content": "Caloric restriction and diet have significant impacts on the genetics of aging through various mechanisms:\n\n1. **Gene Expression and Lifespan Extension**: Caloric restriction (CR) has been shown to delay age-related gene-expression changes in mice and, to some extent, in flies. This suggests that CR may influence the genetic pathways associated with aging, potentially contributing to lifespan extension [4].\n\n2. **Epigenetic and Post-Translational Mechanisms**: In calorie-restricted rats, transcriptome analysis indicates that CR involves epigenetic and post-translational mechanisms, which are implicated in neuroprotection and aging. These mechanisms may alter genome function to promote increased health and lifespan [3], [5].\n\n3. **mTOR Pathway**: Caloric restriction is associated with decelerating mTOR-driven aging, which is a significant pathway involved in cellular growth and metabolism. By modulating this pathway, CR may influence the genetic regulation of aging processes [5].\n\n4. **Genomic and Epigenetic Approaches**: Nutritional modulation, including caloric restriction, can impact aging through genomic and epigenetic approaches. This suggests that diet can influence the genetic and epigenetic landscape, potentially affecting the aging process [6].\n\nOverall, caloric restriction and diet can modulate genetic pathways and mechanisms that are crucial for aging, potentially leading to increased lifespan and improved health during aging.",
              "role": "assistant"
            }
          }
        ],
        "created": 1728913874,
        "model": "gpt-4o-2024-08-06",
        "object": "chat.completion",
        "system_fingerprint": "fp_6b68a8204b",
        "usage": {
          "completion_tokens": 283,
          "prompt_tokens": 1597,
          "total_tokens": 1880
        }
      },
      "search_results": {
        "vector_search_results": [
          {
            "id": "56e72d29-31c2-5096-b4f8-06c740bce06e",
            "score": 0.7473972511944728,
            "metadata": {
              "text": "caloric restriction. Physiol. Genom. 17, 307 315.Van Remmen, H., Ward, W.F., Sabia, R.V ., Richardson, A., 1995. Gene\nexpression and protein degradation. In: Masoro, E.J. (Ed.), Handbook ofPhysiology. Section 11: Aging. Oxford University Press, New York, pp.\n171234.\nWeindruch, R., Walford, R.L., 1982. Dietary restriction in mice beginning at\n1 year of age: effect on life-span and spontaneous cancer incidence.Science 215, 1415 1418.S.R. Spindler / Mechanisms of Ageing and Development 126 (2005) 960 966 966",
              "title": "2005 - Rapid and reversible induction of the longevity, anticancer.pdf",
              "version": "v0",
              "chunk_order": 70,
              "document_id": "0b1bf178-21e4-5382-97c9-c93cdc1a9e66",
              "extraction_id": "db18d213-92ed-5f3e-a242-60d3ed0ec8c8",
              "associatedQuery": "How do caloric restriction and diet impact the genetics of aging?"
            }
          },
          {
            "id": "2b081115-d36e-57ec-aedc-2fd9691bc5e9",
            "score": 0.7285994291305542,
            "metadata": {
              "text": "extension by dietary restriction.   Annu Rev Biochem  2008,\n77:727-54.\n8. Harper JM, Leathers CW, Austad SN: Does caloric restriction\nextend life iin wild mice?   Aging Cell  2006, 5:441-9.\n9. Forster MJ, Morris P, Sohal RS: Genotype and age influence the\neffect of caloric intake  on mortality in mice.   FASEB J  2003,\n17:690-2.\n10. Spindler SR, Mote PL: Screening candidate longevity therapeu-\ntics using gene-e xpression arrays.   Gerontology  2007, 53:306-21.",
              "title": "2009 - Genes and gene expression modules associated with caloric.pdf",
              "version": "v0",
              "chunk_order": 276,
              "document_id": "893ba204-2e69-563f-9046-7246ca61494f",
              "extraction_id": "0b45ae60-562c-5e48-a1c1-9eb29614a63c",
              "associatedQuery": "How do caloric restriction and diet impact the genetics of aging?"
            }
          },
          {
            "id": "bb028469-8295-5657-8061-a715cb314a4a",
            "score": 0.7226208613740898,
            "metadata": {
              "text": "analysis in calorie-restricted rats implicates epigenetic and post-translational\nmechanisms in neuroprotection and aging. Genome Biol. 2015;16:285.\n21. Gillespie ZE, Pickering J, Eskiw CH. Better living through chemistry: caloric\nrestriction (CR) and CR mimetics alter genome function to promote\nincreased health and lifespan. Front Genet. 2016;7:142.\n22. Jiang T, Liebman SE, Lucia MS, Phillips CL, Levi M. Calorie restriction modulates\nrenal expression of sterol regulatory element binding proteins, lipid",
              "title": "2017 - Dietary restriction protects from age-associated DNA methylation and induces epigenetic reprogramming of lipid metabolism.pdf",
              "version": "v0",
              "chunk_order": 146,
              "document_id": "fe573bb0-3d37-55e5-93fa-65b3fbc5f532",
              "extraction_id": "a29560f5-a9e0-56d0-95f2-138eef516ef5",
              "associatedQuery": "How do caloric restriction and diet impact the genetics of aging?"
            }
          },
          {
            "id": "e01c4c58-342d-5369-89e6-98344af55000",
            "score": 0.7175973653793335,
            "metadata": {
              "text": "Calorie restriction, a dietary regimen that extends \nthe lifespan of numerous organisms, also delays the \nmajority of age-related gene-expression changes in \nmice and, to a certain extent, in flies45,50. It is currently \nunclear whether the effect of calorie restriction on gene \nexpression underlies its beneficial effect on lifespan or is merely a consequence thereof. Findings in yeast suggest \nthat there may be a causal link: Sir2 not only facilitates \nheterochromatin and promotes DNA stability, but is",
              "title": "2007 - The role of nuclear architecture.pdf",
              "version": "v0",
              "chunk_order": 76,
              "document_id": "578e2f7d-ddd4-56c8-a5b0-670969f8ff1e",
              "extraction_id": "9655c555-838e-5cdf-85cf-13736c3cf028",
              "associatedQuery": "How do caloric restriction and diet impact the genetics of aging?"
            }
          },
          {
            "id": "1e116f55-36fd-525f-9950-9a1354c32f7b",
            "score": 0.7175322602708507,
            "metadata": {
              "text": "Transcriptome analysis in calorie-restricted rats implicates epigenetic and post-\ntranslational mechanisms in neuroprotection and aging. Genome Biol. 16,2 8\n(2015).\n204. M. V. Blagosklonny, Calorie restriction: Decelerating mTOR-driven aging from cells to or-\nganisms (including humans). Cell Cycle 9, 683 688 (2010).\n205. D. K. Ingram, G. S. Roth, Calorie restriction mimetics: Can you have your cake and eat it,\ntoo? Ageing Res. Rev. 20,4 662 (2015).",
              "title": "2016 - Epigenetics and aging.pdf",
              "version": "v0",
              "chunk_order": 322,
              "document_id": "71b206ec-81bd-5194-8b21-ae522f8cbc2d",
              "extraction_id": "91434549-bda3-5154-b089-28efed9c1089",
              "associatedQuery": "How do caloric restriction and diet impact the genetics of aging?"
            }
          },
          {
            "id": "b990eb0a-709a-500c-836e-83e202e0d6a6",
            "score": 0.7143113613128662,
            "metadata": {
              "text": "life-span extension by calorie restriction in Saccharomyces cerevisiae. Science\n289:21262128.\nMair W, Goymer P, Pletcher SD, and Partridge L (2003) Demography of dietary\nrestriction and death in Drosophila. Science 301:17311733.\nMasoro EJ (2005) Overview of caloric restriction and ageing. Mech Ageing Dev\n126:913922.\nMathers JC (2006) Nutritional modulation of ageing: genomic and epigenetic ap-\nproaches. Mech Ageing Dev 127:584589.\nMeric-Bernstam F and Gonzalez-Angulo AM (2009) Targeting the mTOR signaling",
              "title": "2012 - Genome-Environment Interactions That Modulate.pdf",
              "version": "v0",
              "chunk_order": 214,
              "document_id": "b1a1997c-e9df-5dc0-9d12-a3977d0c64ec",
              "extraction_id": "5745c701-a549-51c3-adcc-b19c47436740",
              "associatedQuery": "How do caloric restriction and diet impact the genetics of aging?"
            }
          },
          {
            "id": "833c9ddc-ec27-5301-9c3b-025eab95e28c",
            "score": 0.7139496803283691,
            "metadata": {
              "text": "Keywords: Caloric restriction; Short-term; Longevity; Cancer; Microarray; Affymetrix\nAging is widely assumed to result from the gradual age-\nrelated accumulation of essentially irreversible moleculardamage. In this context, CR is often viewed as preventing orslowing the accumulation of such damage, thereby slowingthe process of aging ( Bokov et al., 2004 ). This view is\nintuitively appealing, as it provides a straightforwardexplanation for the stochastic nature of aging and the onset",
              "title": "2005 - Rapid and reversible induction of the longevity, anticancer.pdf",
              "version": "v0",
              "chunk_order": 4,
              "document_id": "0b1bf178-21e4-5382-97c9-c93cdc1a9e66",
              "extraction_id": "f4edcf22-ca64-5fe6-9bfa-f97a017d2d08",
              "associatedQuery": "How do caloric restriction and diet impact the genetics of aging?"
            }
          },
          {
            "id": "8151fbcb-f498-56a7-84ce-8af647fd2245",
            "score": 0.7123688802279428,
            "metadata": {
              "text": "of short- and long-term caloric restriction effects in the liver of agingmice. Proc. Natl. Acad. Sci. U.S.A. 98, 10630 10635.Capstick, F., Brooks, B.A., Burns, C.M., Zilkens, R.R., Steinbeck, K.S.,\nYue, D.K., 1997. Very low calorie diet (VLCD): a useful alternative inthe treatment of the obese NIDDM patient. Diab. Res. Clin. Pract. 36,\n105111.\nChen, H., 2004. Gene expression by the anterior pituitary gland: effects of\nage and caloric restriction. Mol. Cell. Endocrinol. 222, 21 31.",
              "title": "2005 - Rapid and reversible induction of the longevity, anticancer.pdf",
              "version": "v0",
              "chunk_order": 60,
              "document_id": "0b1bf178-21e4-5382-97c9-c93cdc1a9e66",
              "extraction_id": "381d5314-5aa1-525f-a2a5-352b70ad86fa",
              "associatedQuery": "How do caloric restriction and diet impact the genetics of aging?"
            }
          },
          {
            "id": "537cff80-380d-5c6a-a25a-04b32e9b1bd7",
            "score": 0.7118311736533399,
            "metadata": {
              "text": "genomic effects of caloric restriction.    Mech. Ageing Dev.     126 : 960    966 .  \n   Sun ,  H. ,  R.J.   Bennett , and  N.   Maizels .  1999 .  The  Saccharomyces cerevisiae   \nSgs1 helicase effi  ciently unwinds G-G paired DNAs.    Nucleic Acids Res.    \n 27 : 1978    1984 .  \n   Thompson ,  L.H. , and  D.   Schild .  2002 .  Recombinational DNA repair and human \ndisease.    Mutat. Res.     509 : 49    78 .",
              "title": "2008 - Longevity mutation in SCH9 prevents recombination.pdf",
              "version": "v0",
              "chunk_order": 182,
              "document_id": "a6b022ba-653f-51d3-845a-dd6b3d61d4a4",
              "extraction_id": "bc856edb-7526-5424-a822-47075459a607",
              "associatedQuery": "How do caloric restriction and diet impact the genetics of aging?"
            }
          },
          {
            "id": "4f28b643-04a4-59e2-8226-ab050e698b64",
            "score": 0.7113211154937744,
            "metadata": {
              "text": "L. & Spindler, S.\n \nR. Genomic profiling of short- and \nlong-term caloric restriction effects in the liver of aging mice. Proc. Natl Acad. Sci. \nUSA 98, 1063010635 (2001).\n62.\n \nHarman, D. The aging process. Proc. Natl Acad. Sci. USA  78, 71247128  \n(1981).\n63.\n \nvan der Pluijm I, G.\n \nG.et.al. Impaired genome maintenance suppresses the growth \nhormoneinsulin-like growth factor 1 axis in mice with cockayne syndrome. PLoS Biol.  \n5, 2338 (2006).\n64.\n \nYang, H., Baur, J.",
              "title": "2008 - DNA damage and ageing.pdf",
              "version": "v0",
              "chunk_order": 105,
              "document_id": "f170e9cf-dfab-5758-ab23-08daff2af694",
              "extraction_id": "554b2b00-d006-5b97-aeb1-70ec31482641",
              "associatedQuery": "How do caloric restriction and diet impact the genetics of aging?"
            }
          }
        ],
        "kg_search_results": null
      }
    }
  }
}