Our website provides four sections: (1) Introduction, (2) Methods, (3) Results, and (4) Conclusion. The Introduction section provides the research background of our study, including an overview of Alzheimer's disease and major depression disorder , as well as the brain-gut axis and our research ideas. The Methods section introduces virtual screening, molecular docking, metagenomic analysis, and other related techniques. The Results section presents our research achievements, including the identification and screening of key intestinal metabolite molecules, tetrahydrofolic acid and inosine. Additionally, potential genetic variations in the gut microbiota of AD and MDD patients were identified, particularly in genes encoding metabolic enzymes. The potential genetic variation of metabolic enzymes in AD and MDD patients and Gut Microbiota was identified. The Conclusion part summarizes the research content, and gives some reflections and prospects. Please see the details of this page below.
Alzheimer's disease (AD) is a progressive neurodegenerative disorder characterized primarily by cognitive impairment and behavioral disturbances, commonly occurring in the elderly or late-middle-aged individuals[1, 2].
Clinical symptoms of AD include memory loss, aphasia, apraxia, agnosia, and visuospatial deficits, often accompanied by changes in personality and behavior[3-5]. Previous studies suggest that the development of AD results from a combination of genetic, epigenetic, and environmental factors[6,7].
Given the unclear mechanisms underlying AD and the lack of effective treatments to cure or substantially reverse the disease, the search for effective therapies continues[8]. Pathologically, Alzheimer's disease patients exhibit the accumulation of large amyloid-beta (Aβ) plaques and neurofibrillary tangles (NFTs) in their brains, accompanied by neuroinflammation, synaptic dysfunction, mitochondrial and bioenergy disturbances, and vascular abnormalities[9].
Major depressive disorder (MDD) is a mental health condition characterized by significant physical changes, such as fatigue, weight loss, and decreased appetite, alongside widespread episodes, low treatment adherence, and high relapse rates. Approximately 300 million people worldwide are affected by MDD, making it one of the leading causes of disability [10].
The clinical symptoms of MDD include persistent low mood, loss of interest, changes in weight or appetite, and an increased risk of suicide [11]. However, due to the lack of in-depth understanding of pathogenesis and objective diagnostic evidence for MDD, it is difficult to identify and prevent early in clinic[12].
Due to the complexity of the pathophysiological mechanisms underlying MDD, accurate diagnostic methods and pharmacological treatments are relatively limited. Genetic factors, stress, comorbidities, and endocrine imbalances are considered to be related to the pathogenesis of MDD[11, 13].
In recent years, the gut-brain axis has gained significant attention for its role in maintaining homeostasis and contributing to disease development. The gut microbiota, often referred to as the "second brain," interacts with the central nervous system through various pathways, including the immune system, tryptophan metabolism, the vagus nerve, and the enteric nervous system. These interactions involve microbial metabolites such as short-chain fatty acids, branched-chain amino acids, and peptidoglycans[14]. It is worth noting that emerging studies have suggested that intestinal microbial metabolites play an important role in mediating the development of AD [15-17] and MDD [18-20] by influencing central nervous system receptors and cellular microenvironment. Based on literature research, nNOS and lilrB3 were selected as potential key targets for MDD and AD(Project. Background).
Therefore, in this study, we plan to screen and identify key gut microbiome metabolite molecules that interact with AD and MDD disease receptors nNOS and LilrB3, using computer-aided drug design (CADD) methods and computational chemistry based on the gut microbiome metabolic molecule library. In addition, we evaluated the upstream mechanisms by which key molecules of gut metabolites influence disease development through metagenomic sequencing of gut microbiota, combined with multi-omics databases for case-control studies of patients with AD and MDD.
A total of 1723 Human gut microbiota metabolite molecules were obtained from the Virtual Metabolic Human database (https://www.vmh.life/#microbes/metabolites)[21]. The metabolite molecular structure formula was obtained from PubChem, and manually drawn using Chemdraw (version 20.0). The format was converted through Openbable software (version 3.1.1).
PyRx software (version 0.8) was used for virtual screening[22]. The predicted 3D structure files of nNOS (AF-P29475-F1-model_v4) was obtained from AlphaFold Protein Structure Database[23, 24], and lilrB3 (8GRX) protein was obtained from PDB database[25, 26]. The ligands and receptors were pretreated with Pymol for dehydrating, hydrogenation and charge balance. Batch virtual filtering through the Vina Wizard in PyRx software, with getbox set to automatically maximize. Binding Affinity, rmsd/ub, rmsd/lb and other results were exported for subsequent analysis.
Autodock software (version 4.2.3) was used for molecular docking[27]. The specific operation is as follows: After preprocessing receptors and ligands, Autogrid is carried out according to the software instructions, with getbox set to automatically maximize. Autodock operation was performed based on the gpf file generated by Autogrid. Set the Number of GA Runs to 200 and the Maximum Number of evals to 2,500,000, the Maximum Number of generations to 27,000. The original file of the conformation of the docking result and Binding energy data are exported and visualized using PyMOL software (version 2.5.8).
Genome-wide Association Study (GWAS) sequencing data were obtained from the GWAS catalog . GWAS sequencing data of AD were obtained from GCST002245[28], GCST90079819[29], GCST90083805[29], GCST013196[30], GCST013197[30], GCST90276158[31], and data of MDD were obtained from GCST90078468[29], GCST90038650[32], GCST90078469[29], GCST90078637[29], GCST90043709[33] comprehensively. The R studio (version 4.3.1) was used for data quality control, and MAGMA (version 1.07), a multivariate regression model-based gene and pathway analysis tool, was used to analyze key single nucleotide polymorphism (SNPs) in the GWAS summary data[34]. SNPS are annotated based on snp151_hg38 (GRCh38).
Metagenomic sequencing data of Gut Microbiota in AD and MDD population cohorts were obtained from ENA Browser and CNGBdb[35] (China National GeneBank DataBase) database.
The metagenomic sequencing data of 75 amyloid-positive AD patients and 100 cognitively healthy controls participating in the AlzBiom study[36], and data of 156 MDD patients and 155 healthy controls were obtained[37] and quality controlled.
Metagenomic data was processed using inStrain (version 1.9.0) [38]. inStrain is an open-source tool designed for analyzing and comparing genomic data, particularly aimed at tracking genomic variation in bacteria and other microorganisms. The analysis was done by introducing a publicly available reference genome, the UHGG genome run inStrain (version 1.9.0), a collection of all microbial species known to exist in the human gut[39]. The profile and compare functions of inStrain can obtain the expected output results, mainly generated in the form of tsv tables.
1.1 Virtual screening of small-molecule metabolites of the gut microbiota potentially bound to the target receptor
A molecular database of human gut microbiota metabolites was constructed and virtual screening was performed as described above (Methods 1-2). Using an absolute value of predicted binding affinity > 7.6 as the criterion, filter out 34 small molecules with potential interactions with nNOS and 33 small molecules with potential interactions with lilrB3. Considering indicators such as toxicity, off-target effects, and the feasibility of engineered bacterial metabolic pathways, a comprehensive assessment was performed (Table 1-2).
1.2 Molecular docking of potential metabolite drug molecules to the key receptors of AD and MDD
Based on the virtual screening results, some promising small molecules were selected for molecular docking to further explore their ability to bind to disease receptors. Autodock was used for molecular docking as described above (Methods 3).
We found that inosine showed the largest clustering center of conformation binding energy among the molecular docking results of nNOS, and tetrahydrofolic acid showed the largest among the molecular docking results of lilrb3 (Figure 1-2). By further analysis of binding conformation, we found that the chemical bonds of inosine and nNOS were mainly distributed in amino acids such as PHE-709, ARG-419, ASN-702, MET-337, GLU-637, etc. (Figure 3). In the binding of tetrahydrofolic acid and lilrb3, the chemical bonds are mainly distributed in ALA-93, GLN-205, ARG-95, TYR-96, SER-65 and other amino acids (Figure 4).
Fig 1.Clustering binding energy center of candidate intestinal microbiota metabolite molecules docking with nNOS molecules.
Fig 2.Clustering binding energy center of candidate intestinal microbiota metabolite molecules docking with lilrB3 molecules.
Further, the Surface Plasmon Resonance (SPR) technique was used to evaluate the affinity, affinity constant and binding kinetics of ligand-receptor binding (Wet lab. Results. SPR Biacore molecular interaction). Ultimately, tetrahydrofolic acid and inosine were selected as potential drug molecules with specific binding to receptor targets associated with Alzheimer's disease (AD) and major depressive disorder (MDD), respectively (Figure 5).
Fig 5.Molecular structure diagram of inosine and tetrahydrofolic acid.
2.1 Evaluating the single nucleotide polymorphisms of key molecular metabolites in intestinal metabolites of AD and MDD patients based on GWAS analysis
Based on the method described previously (Methods 4), MAGMA was used to analyze AD and MDD patient-control GWAS sequencing data to identify mutations in genes encoding metabolic enzymes associated with key metabolic molecules inosine and tetrahydrofolate in the gut microbiota. In the analysis of AD patients, the genes related to tetrahydrofolate metabolism DHFR (rs80650414, P = 2.98*10-6), ALDH1L2 (rs374255885, P = 1.47*10-5), MTHFR (rs542060620, P = 3.96*10--5) were found to have significant SNPs (Figure 6). In addition, a strong effect mutation of APOE4 (rs1490374271, P = 1.95*107) was verified. For the analysis of MDD patients, the gene related to inosine metabolism, PPAT (rs1491299790, P = 1.82*10-6), NAMPT (rs1491400110, P = 9.3*10-6), ADSS1 (rs1490898184, P = 1.08*10-5) were found to have significant SNPs (Figure 7). These results suggest that the potential metabolic enzyme dysfunction caused by genetic variation of host metabolic enzymes is the cause of inosine and tetrahydrofolate deficiency.
Fig 6. Manhattan map for GWAS analysis of MDD.
Fig 7. Manhattan map for GWAS analysis of AD.
2.2 Evaluating the functional variation of Gut Microbiota metabolic enzymes in patients with AD and MDD based on metagenomic sequencing analysis
Based on the profile and compare functions in the inStrain analysis, the obtained table and image data can compare the differences between the sequences of the disease group and the control group. Specifically, it is whether there is a significant difference in the ANOVA analysis of variance of nucleotide diversity between the disease group and the control group and the proportion of variant sites in the disease group and the control group. The calculation formula for nucleotide diversity is: The calculation formula for nucleotide diversity is the sum of the square frequencies of each base: 1 - [(frequency of A)2 + (frequency of C)2 + (frequency of G)2 + (frequency of T)2] Among them, the presentation of nucleotide mutation ratio is mainly divided into four parameters: con-SNV, pop-SNV, SNS and SNV. Pop-SNV takes into account minor alleles compared to con-SNV. The variant sites can be further verified through the differences of these parameters between the disease group and the control group and the ANOVA p-value.
For the AD population, the significance comparison and variation site distribution analysis of the ANOVA p-value of the nucleotide diversity of clpX, folA, folC, folE, folK and folP genes in the disease group and the control group were mainly conducted (Figure 8). For the MDD population, add, deoD, gsk and yfiH were mainly analyzed (Figure 9). A significant difference in nucleotide diversity, along with corresponding differences in the composition of variant types were observed when the P value is less than 0.05, suggesting that there are potential genetic mutations in key metabolic genes of gut microbiota.
Fig 8. Comparison of the ANOVA p-value for nucleotide diversity between the AD and control groups, along with the proportion of variant types in both groups. a-b. gene folE. c-d. gene folk. e-f. gene folP. g-h. gene folC. i-j. gene folA. k-l. gene clpX.
Fig 9. Comparison of the ANOVA p-value for nucleotide diversity between the MDD and control groups, along with the proportion of variant types in both groups. a-b. gene gsk. c-d. gene deoD. e-f. gene add. g-h. gene yfiH.
In this study, we focused on disease targets lilrB3 and nNOS for cognitively related diseases AD and MDD, identified key gut microbiota metabolite molecules tetrahydrofolic acid and inosine, and evaluated their potential as potential therapeutic agents. In addtion, we explain the upstream mechanisms of potential genetic variation leading to tetrahydrofolate and inosine deficiencies at the host and gut microbiota genomics levels. Based on the modeling and analysis work, combined with the wet experiment, we constructed the engineering bacteria that stably expressed tetrahydrofolic acid and inosine.
1. Scheltens P, De Strooper B, Kivipelto M, Holstege H, Chételat G, Teunissen CE, Cummings J, van der Flier WM. Alzheimer's disease. Lancet (London, England) 2021: 397(10284): 1577-1590.
2. Khan S, Barve KH, Kumar MS. Recent Advancements in Pathogenesis, Diagnostics and Treatment of Alzheimer's Disease. Current neuropharmacology 2020: 18(11): 1106-1125.
3. McKhann GM, Knopman DS, Chertkow H, Hyman BT, Jack CR, Jr., Kawas CH, Klunk WE, Koroshetz WJ, Manly JJ, Mayeux R, Mohs RC, Morris JC, Rossor MN, Scheltens P, Carrillo MC, Thies B, Weintraub S, Phelps CH. The diagnosis of dementia due to Alzheimer's disease: recommendations from the National Institute on Aging-Alzheimer's Association workgroups on diagnostic guidelines for Alzheimer's disease. Alzheimer's & dementia : the journal of the Alzheimer's Association 2011: 7(3): 263-269.
4. Porsteinsson AP, Isaacson RS, Knox S, Sabbagh MN, Rubino I. Diagnosis of Early Alzheimer's Disease: Clinical Practice in 2021. The journal of prevention of Alzheimer's disease 2021: 8(3): 371-386.
5. Atri A. The Alzheimer's Disease Clinical Spectrum: Diagnosis and Management. The Medical clinics of North America 2019: 103(2): 263-293.
6. Nikolac Perkovic M, Videtic Paska A, Konjevod M, Kouter K, Svob Strac D, Nedic Erjavec G, Pivac N. Epigenetics of Alzheimer's Disease. Biomolecules 2021: 11(2).
7. Migliore L, Coppedè F. Gene-environment interactions in Alzheimer disease: the emerging role of epigenetics. Nature reviews Neurology 2022: 18(11): 643-660.
8. Breijyeh Z, Karaman R. Comprehensive Review on Alzheimer's Disease: Causes and Treatment. Molecules (Basel, Switzerland) 2020: 25(24).
9. Zhang J, Zhang Y, Wang J, Xia Y, Zhang J, Chen L. Recent advances in Alzheimer's disease: Mechanisms, clinical trials and new drug development strategies. Signal transduction and targeted therapy 2024: 9(1): 211.
10. Nagy C, Maitra M, Tanti A, Suderman M, Théroux JF, Davoli MA, Perlman K, Yerko V, Wang YC, Tripathy SJ, Pavlidis P, Mechawar N, Ragoussis J, Turecki G. Single-nucleus transcriptomics of the prefrontal cortex in major depressive disorder implicates oligodendrocyte precursor cells and excitatory neurons. Nature neuroscience 2020: 23(6): 771-781.
11. Cui L, Li S, Wang S, Wu X, Liu Y, Yu W, Wang Y, Tang Y, Xia M, Li B. Major depressive disorder: hypothesis, mechanism, prevention and treatment. Signal transduction and targeted therapy 2024: 9(1): 30.
12. Kovacs M, Lopez-Duran N. Prodromal symptoms and atypical affectivity as predictors of major depression in juveniles: implications for prevention. Journal of child psychology and psychiatry, and allied disciplines 2010: 51(4): 472-496.
13. Uher R, Pavlova B, Najafi S, Adepalli N, Ross B, Howes Vallis E, Freeman K, Parker R, Propper L, Palaniyappan L. Antecedents of major depressive, bipolar, and psychotic disorders: A systematic review and meta-analysis of prospective studies. Neuroscience and biobehavioral reviews 2024: 160: 105625.
14. Cryan JF, O'Riordan KJ, Cowan CSM, Sandhu KV, Bastiaanssen TFS, Boehme M, Codagnone MG, Cussotto S, Fulling C, Golubeva AV, Guzzetta KE, Jaggar M, Long-Smith CM, Lyte JM, Martin JA, Molinero-Perez A, Moloney G, Morelli E, Morillas E, O'Connor R, Cruz-Pereira JS, Peterson VL, Rea K, Ritz NL, Sherwin E, Spichak S, Teichman EM, van de Wouw M, Ventura-Silva AP, Wallace-Fitzsimons SE, Hyland N, Clarke G, Dinan TG. The Microbiota-Gut-Brain Axis. Physiological reviews 2019: 99(4): 1877-2013.
15. Ferreiro AL, Choi J, Ryou J, Newcomer EP, Thompson R, Bollinger RM, Hall-Moore C, Ndao IM, Sax L, Benzinger TLS, Stark SL, Holtzman DM, Fagan AM, Schindler SE, Cruchaga C, Butt OH, Morris JC, Tarr PI, Ances BM, Dantas G. Gut microbiome composition may be an indicator of preclinical Alzheimer's disease. Science translational medicine 2023: 15(700): eabo2984.
16. Chen C, Liao J, Xia Y, Liu X, Jones R, Haran J, McCormick B, Sampson TR, Alam A, Ye K. Gut microbiota regulate Alzheimer's disease pathologies and cognitive disorders via PUFA-associated neuroinflammation. Gut 2022: 71(11): 2233-2252.
17. Kesika P, Suganthy N, Sivamaruthi BS, Chaiyasut C. Role of gut-brain axis, gut microbial composition, and probiotic intervention in Alzheimer's disease. Life sciences 2021: 264: 118627.
18. Amin N, Liu J, Bonnechere B, MahmoudianDehkordi S, Arnold M, Batra R, Chiou YJ, Fernandes M, Ikram MA, Kraaij R, Krumsiek J, Newby D, Nho K, Radjabzadeh D, Saykin AJ, Shi L, Sproviero W, Winchester L, Yang Y, Nevado-Holgado AJ, Kastenmüller G, Kaddurah-Daouk R, van Duijn CM. Interplay of Metabolome and Gut Microbiome in Individuals With Major Depressive Disorder vs Control Individuals. JAMA psychiatry 2023: 80(6): 597-609.
19. Simpson CA, Diaz-Arteche C, Eliby D, Schwartz OS, Simmons JG, Cowan CSM. The gut microbiota in anxiety and depression - A systematic review. Clinical psychology review 2021: 83: 101943.
20. Sanada K, Nakajima S, Kurokawa S, Barceló-Soler A, Ikuse D, Hirata A, Yoshizawa A, Tomizawa Y, Salas-Valero M, Noda Y, Mimura M, Iwanami A, Kishimoto T. Gut microbiota and major depressive disorder: A systematic review and meta-analysis. Journal of affective disorders 2020: 266: 1-13.
21. Magnúsdóttir S, Heinken A, Kutt L, Ravcheev DA, Bauer E, Noronha A, Greenhalgh K, Jäger C, Baginska J, Wilmes P, Fleming RM, Thiele I. Generation of genome-scale metabolic reconstructions for 773 members of the human gut microbiota. Nature biotechnology 2017: 35(1): 81-89.
22. Dallakyan S, Olson AJ. Small-molecule library screening by docking with PyRx. Methods in molecular biology (Clifton, NJ) 2015: 1263: 243-250.
23. Varadi M, Bertoni D, Magana P, Paramval U, Pidruchna I, Radhakrishnan M, Tsenkov M, Nair S, Mirdita M, Yeo J, Kovalevskiy O, Tunyasuvunakool K, Laydon A, Žídek A, Tomlinson H, Hariharan D, Abrahamson J, Green T, Jumper J, Birney E, Steinegger M, Hassabis D, Velankar S. AlphaFold Protein Structure Database in 2024: providing structure coverage for over 214 million protein sequences. Nucleic acids research 2024: 52(D1): D368-d375.
24. Varadi M, Anyango S, Deshpande M, Nair S, Natassia C, Yordanova G, Yuan D, Stroe O, Wood G, Laydon A, Žídek A, Green T, Tunyasuvunakool K, Petersen S, Jumper J, Clancy E, Green R, Vora A, Lutfi M, Figurnov M, Cowie A, Hobbs N, Kohli P, Kleywegt G, Birney E, Hassabis D, Velankar S. AlphaFold Protein Structure Database: massively expanding the structural coverage of protein-sequence space with high-accuracy models. Nucleic acids research 2022: 50(D1): D439-d444.
25. Protein Data Bank: the single global archive for 3D macromolecular structure data. Nucleic acids research 2019: 47(D1): D520-d528.
26. Zhou J, Wang Y, Huang G, Yang M, Zhu Y, Jin C, Jing D, Ji K, Shi Y. LilrB3 is a putative cell surface receptor of APOE4. Cell research 2023: 33(2): 116-130.
27. Morris GM, Huey R, Lindstrom W, Sanner MF, Belew RK, Goodsell DS, Olson AJ. AutoDock4 and AutoDockTools4: Automated docking with selective receptor flexibility. Journal of computational chemistry 2009: 30(16): 2785-2791.
28. Lambert JC, Ibrahim-Verbaas CA, Harold D, Naj AC, Sims R, Bellenguez C, DeStafano AL, Bis JC, Beecham GW, Grenier-Boley B, Russo G, Thorton-Wells TA, Jones N, Smith AV, Chouraki V, Thomas C, Ikram MA, Zelenika D, Vardarajan BN, Kamatani Y, Lin CF, Gerrish A, Schmidt H, Kunkle B, Dunstan ML, Ruiz A, Bihoreau MT, Choi SH, Reitz C, Pasquier F, Cruchaga C, Craig D, Amin N, Berr C, Lopez OL, De Jager PL, Deramecourt V, Johnston JA, Evans D, Lovestone S, Letenneur L, Morón FJ, Rubinsztein DC, Eiriksdottir G, Sleegers K, Goate AM, Fiévet N, Huentelman MW, Gill M, Brown K, Kamboh MI, Keller L, Barberger-Gateau P, McGuiness B, Larson EB, Green R, Myers AJ, Dufouil C, Todd S, Wallon D, Love S, Rogaeva E, Gallacher J, St George-Hyslop P, Clarimon J, Lleo A, Bayer A, Tsuang DW, Yu L, Tsolaki M, Bossù P, Spalletta G, Proitsi P, Collinge J, Sorbi S, Sanchez-Garcia F, Fox NC, Hardy J, Deniz Naranjo MC, Bosco P, Clarke R, Brayne C, Galimberti D, Mancuso M, Matthews F, Moebus S, Mecocci P, Del Zompo M, Maier W, Hampel H, Pilotto A, Bullido M, Panza F, Caffarra P, Nacmias B, Gilbert JR, Mayhaus M, Lannefelt L, Hakonarson H, Pichler S, Carrasquillo MM, Ingelsson M, Beekly D, Alvarez V, Zou F, Valladares O, Younkin SG, Coto E, Hamilton-Nelson KL, Gu W, Razquin C, Pastor P, Mateo I, Owen MJ, Faber KM, Jonsson PV, Combarros O, O'Donovan MC, Cantwell LB, Soininen H, Blacker D, Mead S, Mosley TH, Jr., Bennett DA, Harris TB, Fratiglioni L, Holmes C, de Bruijn RF, Passmore P, Montine TJ, Bettens K, Rotter JI, Brice A, Morgan K, Foroud TM, Kukull WA, Hannequin D, Powell JF, Nalls MA, Ritchie K, Lunetta KL, Kauwe JS, Boerwinkle E, Riemenschneider M, Boada M, Hiltuenen M, Martin ER, Schmidt R, Rujescu D, Wang LS, Dartigues JF, Mayeux R, Tzourio C, Hofman A, Nöthen MM, Graff C, Psaty BM, Jones L, Haines JL, Holmans PA, Lathrop M, Pericak-Vance MA, Launer LJ, Farrer LA, van Duijn CM, Van Broeckhoven C, Moskvina V, Seshadri S, Williams J, Schellenberg GD, Amouyel P. Meta-analysis of 74,046 individuals identifies 11 new susceptibility loci for Alzheimer's disease. Nature genetics 2013: 45(12): 1452-1458.
29. Backman JD, Li AH, Marcketta A, Sun D, Mbatchou J, Kessler MD, Benner C, Liu D, Locke AE, Balasubramanian S, Yadav A, Banerjee N, Gillies CE, Damask A, Liu S, Bai X, Hawes A, Maxwell E, Gurski L, Watanabe K, Kosmicki JA, Rajagopal V, Mighty J, Jones M, Mitnaul L, Stahl E, Coppola G, Jorgenson E, Habegger L, Salerno WJ, Shuldiner AR, Lotta LA, Overton JD, Cantor MN, Reid JG, Yancopoulos G, Kang HM, Marchini J, Baras A, Abecasis GR, Ferreira MAR. Exome sequencing and analysis of 454,787 UK Biobank participants. Nature 2021: 599(7886): 628-634.
30. Wightman DP, Jansen IE, Savage JE, Shadrin AA, Bahrami S, Holland D, Rongve A, Børte S, Winsvold BS, Drange OK, Martinsen AE, Skogholt AH, Willer C, Bråthen G, Bosnes I, Nielsen JB, Fritsche LG, Thomas LF, Pedersen LM, Gabrielsen ME, Johnsen MB, Meisingset TW, Zhou W, Proitsi P, Hodges A, Dobson R, Velayudhan L, Heilbron K, Auton A, Sealock JM, Davis LK, Pedersen NL, Reynolds CA, Karlsson IK, Magnusson S, Stefansson H, Thordardottir S, Jonsson PV, Snaedal J, Zettergren A, Skoog I, Kern S, Waern M, Zetterberg H, Blennow K, Stordal E, Hveem K, Zwart JA, Athanasiu L, Selnes P, Saltvedt I, Sando SB, Ulstein I, Djurovic S, Fladby T, Aarsland D, Selbæk G, Ripke S, Stefansson K, Andreassen OA, Posthuma D. A genome-wide association study with 1,126,563 individuals identifies new risk loci for Alzheimer's disease. Nature genetics 2021: 53(9): 1276-1282.
31. Talyansky S, Le Guen Y, Kasireddy N, Belloy ME, Greicius MD. APOE-ε4 and BIN1 increase risk of Alzheimer's disease pathology but not specifically of Lewy body pathology. Acta neuropathologica communications 2023: 11(1): 149.
32. Dieppe PA, Lohmander LS. Pathogenesis and management of pain in osteoarthritis. Lancet (London, England) 2005: 365(9463): 965-973.
33. Jiang L, Zheng Z, Fang H, Yang J. A generalized linear mixed model association tool for biobank-scale data. Nature genetics 2021: 53(11): 1616-1621.
34. de Leeuw CA, Mooij JM, Heskes T, Posthuma D. MAGMA: generalized gene-set analysis of GWAS data. PLoS computational biology 2015: 11(4): e1004219.
35. Chen FZ, You LJ, Yang F, Wang LN, Guo XQ, Gao F, Hua C, Tan C, Fang L, Shan RQ, Zeng WJ, Wang B, Wang R, Xu X, Wei XF. CNGBdb: China National GeneBank DataBase. Yi chuan = Hereditas 2020: 42(8): 799-809.
36. Laske C, Müller S, Preische O, Ruschil V, Munk MHJ, Honold I, Peter S, Schoppmeier U, Willmann M. Signature of Alzheimer's Disease in Intestinal Microbiome: Results From the AlzBiom Study. Frontiers in neuroscience 2022: 16: 792996.
37. Hu X, Li Y, Wu J, Zhang H, Huang Y, Tan X, Wen L, Zhou X, Xie P, Olasunkanmi OI, Zhou J, Sun Z, Liu M, Zhang G, Yang J, Zheng P, Xie P. Changes of gut microbiota reflect the severity of major depressive disorder: a cross sectional study. Translational psychiatry 2023: 13(1): 137.
38. Olm MR, Crits-Christoph A, Bouma-Gregson K, Firek BA, Morowitz MJ, Banfield JF. inStrain profiles population microdiversity from metagenomic data and sensitively detects shared microbial strains. Nature biotechnology 2021: 39(6): 727-736.
39. Almeida A, Nayfach S, Boland M, Strozzi F, Beracochea M, Shi ZJ, Pollard KS, Sakharova E, Parks DH, Hugenholtz P, Segata N, Kyrpides NC, Finn RD. A unified catalog of 204,938 reference genomes from the human gut microbiome. Nature biotechnology 2021: 39(1): 105-114.