References for the main text and supplements of our journal article in Science Translational Medicine, "Progression to rheumatoid arthritis in at-risk individuals is defined by systemic inflammation and by T and B cell dysregulation" by He, Z. and Glass, M.C, et al., are provided below:
1. M. H. Buch, Defining refractory rheumatoid arthritis. Ann. Rheum. Dis. 77, 966–969 (2018).
doi:10.1136/annrheumdis-2017-212862
2. B. Combe, R. Landewe, C. I. Daien, C. Hua, D. Aletaha, J. M. Álvaro-Gracia, M. Bakkers, N. Brodin, G. R. Burmester, C. Codreanu, R. Conway, M. Dougados, P. Emery, G. Ferraccioli, J. Fonseca, K. Raza, L. Silva-Fernández, J. S. Smolen, D. Skingle, Z. Szekanecz, T. K. Kvien, A. van der Helm-van Mil, R. van Vollenhoven, 2016 update of the EULAR recommendations for the management of early arthritis. Ann. Rheum. Dis. 76, 948–959 (2017).
doi:10.1136/annrheumdis-2016-210602
3. K. C. Herold, B. N. Bundy, S. A. Long, J. A. Bluestone, L. A. DiMeglio, M. J. Dufort, S. E. Gitelman, P. A. Gottlieb, J. P. Krischer, P. S. Linsley, J. B. Marks, W. Moore, A. Moran, H. Rodriguez, W. E. Russell, D. Schatz, J. S. Skyler, E. Tsalikian, D. K. Wherrett, A.-G. Ziegler, C. J. Greenbaum, Type 1 Diabetes TrialNet Study Group, An Anti-CD3 Antibody, Teplizumab, in Relatives at Risk for Type 1 Diabetes. N. Engl. J. Med. 381, 603–613 (2019).
doi:10.1056/NEJMoa1902226
4. S. Rantapää-Dahlqvist, B. A. W. de Jong, E. Berglin, G. Hallmans, G. Wadell, H. Stenlund, U. Sundin, W. J. van Venrooij, Antibodies against cyclic citrullinated peptide and IgA rheumatoid factor predict the development of rheumatoid arthritis. Arthritis Rheumatol. 48, 2741–2749 (2003).
doi:10.1002/art.11223
5. M. M. J. Nielen, D. van Schaardenburg, H. W. Reesink, R. J. van de Stadt, I. E. van der Horst-Bruinsma, M. H. M. T. de Koning, M. R. Habibuw, J. P. Vandenbroucke, B. A. C. Dijkmans, Specific autoantibodies precede the symptoms of rheumatoid arthritis: a study of serial measurements in blood donors. Arthritis Rheumatol. 50, 380–386 (2004).
doi:10.1002/art.20018
6. L. B. Kelmenson, B. D. Wagner, B. K. McNair, A. Frazer-Abel, M. K. Demoruelle, D. T. Bergstedt, M. L. Feser, L. K. Moss, M. C. Parish, E. A. Mewshaw, T. R. Mikuls, J. D. Edison, V. M. Holers, K. D. Deane, Timing of Elevations of Autoantibody Isotypes Prior to Diagnosis of Rheumatoid Arthritis. Arthritis Rheumatol. 72, 251–261 (2020).
doi:10.1002/art.41091
7. L. Duquenne, E. M. Hensor, M. Wilson, L. Garcia-Montoya, J. L. Nam, J. Wu, K. Harnden, I. C. Anioke, A. Di Matteo, R. Chowdhury, N. Sidhu, F. Ponchel, K. Mankia, P. Emery, Predicting inflammatory arthritis in at-risk persons: Development of scores for risk stratification. Ann. Intern. Med. 176, 1027–1036 (2023).
doi:10.7326/M23-027
8. D. T. Bergstedt, W. J. Tarter, R. A. Peterson, M. L. Feser, M. C. Parish, C. C. Striebich, M. K. Demoruelle, L. Moss, E. A. Bemis, J. M. Norris, V. M. Holers, J. D. Edison, G. M. Thiele, T. R. Mikuls, K. D. Deane, Antibodies to Citrullinated Protein Antigens, Rheumatoid Factor Isotypes and the Shared Epitope and the Near-Term Development of Clinically-Apparent Rheumatoid Arthritis. Front. Immunol. 13, 916277 (2022).
doi:10.3389/fimmu.2022.916277
9. D. M. Gerlag, M. Safy, K. I. Maijer, M. W. Tang, S. W. Tas, M. J. F. Starmans-Kool, A. van Tubergen, M. Janssen, M. de Hair, M. Hansson, N. de Vries, A. H. Zwinderman, P. P. Tak, Effects of B-cell directed therapy on the preclinical stage of rheumatoid arthritis: the PRAIRI study. Ann. Rheum. Dis. 78, 179–185 (2019).
doi:10.1136/annrheumdis-2017-212763
10. A. P. Cope, M. Jasenecova, J. C. Vasconcelos, A. Filer, K. Raza, S. Qureshi, M. A. D’Agostino, I. B. McInnes, J. D. Isaacs, A. G. Pratt, B. A. Fisher, C. D. Buckley, P. Emery, P. Ho, M. H. Buch, C. Ciurtin, D. van Schaardenburg, T. Huizinga, R. Toes, E. Georgiou, J. Kelly, C. Murphy, A. T. Prevost, APIPPRA study investigators, Abatacept in individuals at high risk of rheumatoid arthritis (APIPPRA): a randomised, double-blind, multicentre, parallel, placebo-controlled, phase 2b clinical trial. Lancet 403, 838–849 (2024).
doi:10.1016/S0140-6736(23)02649-1
11. J. Rech, K. Tascilar, M. Hagen, A. Kleyer, B. Manger, V. Schoenau, A. J. Hueber, S. Kleinert, X. Baraliakos, J. Braun, U. Kiltz, M. Fleck, A. Rubbert-Roth, D. M. Kofler, F. Behrens, M. Feuchtenberger, M. Zaenker, R. Voll, N. Venhoff, J. Thiel, C. Glaser, E. Feist, G. R. Burmester, K. Karberg, J. Strunk, J. D. Cañete, L. Senolt, M. Filkova, E. Naredo, R. Largo, G. Krönke, M.-A. D’Agostino, M. Østergaard, G. Schett, Abatacept inhibits inflammation and onset of rheumatoid arthritis in individuals at high risk (ARIAA): a randomised, international, multicentre, double-blind, placebo-controlled trial. Lancet 403, 850–859 (2024).
doi:10.1016/S0140-6736(23)02650-8
12. K. D. Deane, C. I. O’Donnell, W. Hueber, D. S. Majka, A. A. Lazar, L. A. Derber, W. R. Gilliland, J. D. Edison, J. M. Norris, W. H. Robinson, V. M. Holers, The number of elevated cytokines and chemokines in preclinical seropositive rheumatoid arthritis predicts time to diagnosis in an age-dependent manner. Arthritis Rheumatol. 62, 3161–3172 (2010).
doi:10.1002/art.27638
13. H. Kokkonen, I. Söderström, J. Rocklöv, G. Hallmans, K. Lejon, S. Rantapää Dahlqvist, Up-regulation of cytokines and chemokines predates the onset of rheumatoid arthritis. Arthritis Rheumatol. 62, 383–391 (2010).
doi:10.1002/art.27186
14. J. Sokolove, R. Bromberg, K. D. Deane, L. J. Lahey, L. A. Derber, P. E. Chandra, J. D. Edison, W. R. Gilliland, R. J. Tibshirani, J. M. Norris, V. M. Holers, W. H. Robinson, Autoantibody epitope spreading in the pre-clinical phase predicts progression to rheumatoid arthritis. PLoS One 7, e35296 (2012).
doi:10.1371/journal.pone.0035296
15. K. T. Jørgensen, A. Wiik, M. Pedersen, C. J. Hedegaard, B. F. Vestergaard, R. E. Gislefoss, T. K. Kvien, J. Wohlfahrt, K. Bendtzen, M. Frisch, Cytokines, autoantibodies and viral antibodies in premorbid and postdiagnostic sera from patients with rheumatoid arthritis: case-control study nested in a cohort of Norwegian blood donors. Ann. Rheum. Dis. 67, 860–866 (2008).
doi:10.1136/ard.2007.073825
16. L. J. O’Neil, V. Spicer, I. Smolik, X. Meng, R. R. Goel, V. Anaparti, J. Wilkins, H. S. El-Gabalawy, Association of a Serum Protein Signature With Rheumatoid Arthritis Development. Arthritis Rheumatol. 73, 78–88 (2021).
doi:10.1002/art.41483
17. L. Hunt, E. M. Hensor, J. Nam, A. N. Burska, R. Parmar, P. Emery, F. Ponchel, T cell subsets: an immunological biomarker to predict progression to clinical arthritis in ACPA-positive individuals. Ann. Rheum. Dis. 75, 1884–1889 (2016).
doi:10.1136/annrheumdis-2015-207991
18. J. Inamo, J. Keegan, A. Griffith, T. Ghosh, A. Horisberger, K. Howard, J. F. Pulford, E. Murzin, B. Hancock, S. T. Dominguez, M. G. Gurra, S. Gurajala, A. H. Jonsson, J. A. Seifert, M. L. Feser, J. M. Norris, Y. Cao, W. Apruzzese, S. L. Bridges, V. P. Bykerk, S. Goodman, L. T. Donlin, G. S. Firestein, J. M. Bathon, L. B. Hughes, A. Filer, C. Pitzalis, J. H. Anolik, L. Moreland, N. Hacohen, J. M. Guthridge, J. A. James, C. M. Cuda, H. Perlman, M. B. Brenner, S. Raychaudhuri, J. A. Sparks, Accelerating Medicines Partnership RA/SLE Network, V. M. Holers, K. D. Deane, J. Lederer, D. A. Rao, F. Zhang, Deep immunophenotyping reveals circulating activated lymphocytes in individuals at risk for rheumatoid arthritis. J. Clin. Invest. 135 (2025).
doi:10.1172/JCI185217
19. H. Takada, M. K. Demoruelle, K. D. Deane, S. Nakamura, Y. Katsumata, K. Ikari, J. H. Buckner, W. H. Robinson, J. A. Seifert, M. L. Feser, L. Moss, J. M. Norris, M. Harigai, E. W. Y. Hsieh, V. M. Holers, Y. Okamoto, Expansion of HLA-DR Positive Peripheral Helper T and Naïve B cells in Anti-Citrullinated Protein Antibody-Positive Subjects At-Risk for Rheumatoid Arthritis. Arthritis Rheum. 76, 1023-1035 (2024).
doi:10.1002/art.42839
20. E. A. James, V. M. Holers, R. Iyer, E. B. Prideaux, N. L. Rao, C. Rims, V. S. Muir, S. E. Posso, M. S. Bloom, A. Zia, S. E. Elliott, J. Z. Adamska, R. Ai, R. C. Brewer, J. A. Seifert, L. Moss, S. Barzideh, M. K. Demoruelle, C. C. Striebich, Y. Okamoto, E. Sainbayar, A. A. Crook, R. A. Peterson, L. A. Vanderlinden, W. Wang, D. L. Boyle, W. H. Robinson, J. H. Buckner, G. S. Firestein, K. D. Deane, Multifaceted immune dysregulation characterizes individuals at-risk for rheumatoid arthritis. Nat. Commun. 14, 7637 (2023).
doi:10.1038/s41467-023-43091-8
21. H.-H. Chang, G.-Y. Liu, N. Dwivedi, B. Sun, Y. Okamoto, J. D. Kinslow, K. D. Deane, M. K. Demoruelle, J. M. Norris, P. R. Thompson, J. A. Sparks, D. A. Rao, E. W. Karlson, H.-C. Hung, V. M. Holers, I.-C. Ho, A molecular signature of preclinical rheumatoid arthritis triggered by dysregulated PTPN22. JCI Insight 1, e90045 (2016).
doi:10.1172/jci.insight.90045
22. J. D. Kinslow, L. K. Blum, K. D. Deane, M. K. Demoruelle, Y. Okamoto, M. C. Parish, S. Kongpachith, L. J. Lahey, J. M. Norris, W. H. Robinson, V. M. Holers, Elevated IgA Plasmablast Levels in Subjects at Risk of Developing Rheumatoid Arthritis. Arthritis Rheumatol. 68, 2372–2383 (2016).
doi:10.1002/art.39771
23. L. A. van de Stadt, M. H. M. T. de Koning, R. J. van de Stadt, G. Wolbink, B. A. C. Dijkmans, D. Hamann, D. van Schaardenburg, Development of the anti-citrullinated protein antibody repertoire prior to the onset of rheumatoid arthritis. Arthritis Rheumatol. 63, 3226–3233 (2011).
doi:10.1002/art.30537
24. Q. Gong, M. Sharma, E. L. Kuan, M. C. Glass, A. Chander, M. Singh, L. T. Graybuck, Z. J. Thomson, C. M. LaFrance, S. R. Zaim, T. Peng, L. Y. Okada, P. C. Genge, K. E. Henderson, E. M. Dornisch, E. D. Layton, P. J. Wittig, A. T. Heubeck, N. M. Mukuka, J. Reading, C. R. Roll, V. Hernandez, V. Parthasarathy, T. J. Stuckey, B. Musgrove, E. Swanson, C. Lord, M. D. A. Weiss, C. G. Phalen, R. R. Mettey, K. J. Lee, J. B. Johanneson, E. K. Kawelo, J. Garber, U. Krishnan, M. Smithmeyer, E. J. Wherry, L. Vella, S. E. Henrickson, M. S. Kopp, A. K. Savage, L. A. Becker, P. Meijer, E. M. Coffey, J. J. Goronzy, C. Speake, T. F. Bumol, A. W. Goldrath, T. R. Torgerson, X.-J. Li, P. J. Skene, J. H. Buckner, C. E. Gustafson, Longitudinal Multi-omic Immune Profiling Reveals Age-Related Immune Cell Dynamics in Healthy Adults. bioRxiv 2024.09.10.612119 (2024).
doi:10.1101/2024.09.10.612119
25. Data files S1 to S9.
doi:10.5061/dryad.1rn8pk13z.
26. Z. Ye, Y. Shen, K. Jin, J. Qiu, B. Hu, R. R. Jadhav, K. Sheth, C. M. Weyand, J. J. Goronzy, Arachidonic acid-regulated calcium signaling in T cells from patients with rheumatoid arthritis promotes synovial inflammation. Nat. Commun. 12, 907 (2021).
doi:10.1038/s41467-021-21242-z
27. D. E. Orange, V. Yao, K. Sawicka, J. Fak, M. O. Frank, S. Parveen, N. E. Blachere, C. Hale, F. Zhang, S. Raychaudhuri, O. G. Troyanskaya, R. B. Darnell, RNA Identification of PRIME Cells Predicting Rheumatoid Arthritis Flares. N. Engl. J. Med. 383, 218–228 (2020).
doi:10.1056/NEJMoa2004114
28. F. Zhang, K. Wei, K. Slowikowski, C. Y. Fonseka, D. A. Rao, S. Kelly, S. M. Goodman, D. Tabechian, L. B. Hughes, K. Salomon-Escoto, G. F. M. Watts, A. H. Jonsson, J. Rangel-Moreno, N. Meednu, C. Rozo, W. Apruzzese, T. M. Eisenhaure, D. J. Lieb, D. L. Boyle, A. M. Mandelin 2nd, Accelerating Medicines Partnership Rheumatoid Arthritis and Systemic Lupus Erythematosus (AMP RA/SLE) Consortium, B. F. Boyce, E. DiCarlo, E. M. Gravallese, P. K. Gregersen, L. Moreland, G. S. Firestein, N. Hacohen, C. Nusbaum, J. A. Lederer, H. Perlman, C. Pitzalis, A. Filer, V. M. Holers, V. P. Bykerk, L. T. Donlin, J. H. Anolik, M. B. Brenner, S. Raychaudhuri, Defining inflammatory cell states in rheumatoid arthritis joint synovial tissues by integrating single-cell transcriptomics and mass cytometry. Nat. Immunol. 20, 928–942 (2019).
doi:10.1038/s41590-019-0378-1
29. Z. Zhou, X. Zhou, X. Jiang, B. Yang, X. Lu, Y. Fei, L. Zhao, H. Chen, L. Zhang, X. Si, N. Liang, Y. Wang, D. Yang, Y. Peng, Y. Yang, Z. Yao, Y. He, X. Wu, W. Zhang, M. Wang, H. Yang, X. Zhang, Single-cell profiling identifies IL1Bhi macrophages associated with inflammation in PD-1 inhibitor-induced inflammatory arthritis. Nat. Commun. 15, 2107 (2024).
doi:10.1038/s41467-024-46195-x
30. S. Alivernini, L. MacDonald, A. Elmesmari, S. Finlay, B. Tolusso, M. R. Gigante, L. Petricca, C. Di Mario, L. Bui, S. Perniola, M. Attar, M. Gessi, A. L. Fedele, S. Chilaka, D. Somma, S. N. Sansom, A. Filer, C. McSharry, N. L. Millar, K. Kirschner, A. Nerviani, M. J. Lewis, C. Pitzalis, A. R. Clark, G. Ferraccioli, I. Udalova, C. D. Buckley, E. Gremese, I. B. McInnes, T. D. Otto, M. Kurowska-Stolarska, Distinct synovial tissue macrophage subsets regulate inflammation and remission in rheumatoid arthritis. Nat. Med. 26, 1295–1306 (2020).
doi:10.1038/s41591-020-0939-8
31. R. Yang, D. T. Avery, K. J. L. Jackson, M. Ogishi, I. Benhsaien, L. Du, X. Ye, J. Han, J. Rosain, J. N. Peel, M.-A. Alyanakian, B. Neven, S. Winter, A. Puel, B. Boisson, K. J. Payne, M. Wong, A. J. Russell, Y. Mizoguchi, S. Okada, G. Uzel, C. C. Goodnow, S. Latour, J. El Bakkouri, A. Bousfiha, K. Preece, P. E. Gray, B. Keller, K. Warnatz, S. Boisson-Dupuis, L. Abel, Q. Pan-Hammarström, J. Bustamante, C. S. Ma, J.-L. Casanova, S. G. Tangye, Human T-bet governs the generation of a distinct subset of CD11chighCD21low B cells. Sci. Immunol. 7, eabq3277 (2022).
doi:10.1126/sciimmunol.abq3277
32. S. Engelhart, R. J. Glynn, P. H. Schur, Disease associations with isolated elevations of each of the four IgG subclasses. Semin. Arthritis Rheum. 47, 276–280 (2017).
doi:10.1016/j.semarthrit.2017.03.021
33. Y. Yasumizu, D. Takeuchi, R. Morimoto, Y. Takeshima, T. Okuno, M. Kinoshita, T. Morita, Y. Kato, M. Wang, D. Motooka, D. Okuzaki, Y. Nakamura, N. Mikami, M. Arai, X. Zhang, A. Kumanogoh, H. Mochizuki, N. Ohkura, S. Sakaguchi, Single-cell transcriptome landscape of circulating CD4+ T cell populations in autoimmune diseases. Cell Genomics 0 (2024).
doi:10.1016/j.xgen.2023.100473
34. A. T. Bauquet, H. Jin, A. M. Paterson, M. Mitsdoerffer, I.-C. Ho, A. H. Sharpe, V. K. Kuchroo, The costimulatory molecule ICOS regulates the expression of c-Maf and IL-21 in the development of follicular T helper cells and TH-17 cells. Nat. Immunol. 10, 167–175 (2009).
doi:10.1038/ni.1690
35. L. Cosmi, R. De Palma, V. Santarlasci, L. Maggi, M. Capone, F. Frosali, G. Rodolico, V. Querci, G. Abbate, R. Angeli, L. Berrino, M. Fambrini, M. Caproni, F. Tonelli, E. Lazzeri, P. Parronchi, F. Liotta, E. Maggi, S. Romagnani, F. Annunziato, Human interleukin 17-producing cells originate from a CD161+CD4+ T cell precursor. J. Exp. Med. 205, 1903–1916 (2008).
doi:10.1084/jem.20080397
36. X. Shui, S. Chen, J. Lin, J. Kong, C. Zhou, J. Wu, Knockdown of lncRNA NEAT1 inhibits Th17/CD4+ T cell differentiation through reducing the STAT3 protein level. J. Cell. Physiol. 234, 22477–22484 (2019).
doi:10.1002/jcp.28811
37. G. P. Parzmair, M. Gereke, O. Haberkorn, M. Annemann, L. Podlasly, S. Kliche, A. Reinhold, B. Schraven, D. Bruder, ADAP plays a pivotal role in CD4+ T cell activation but is only marginally involved in CD8+ T cell activation, differentiation, and immunity to pathogens. J. Leukoc. Biol. 101, 407–419 (2017).
doi:10.1189/jlb.1A0216-090RR
38. Z. Thomson, Z. He, E. Swanson, K. Henderson, C. Phalen, S. R. Zaim, M.-P. Pebworth, L. Y. Okada, A. T. Heubeck, C. R. Roll, V. Hernandez, M. Weiss, P. C. Genge, J. Reading, J. R. Giles, S. Manne, J. Dougherty, C. J. Jasen, A. R. Greenplate, L. A. Becker, L. T. Graybuck, S. V. Vasaikar, G. L. Szeto, A. K. Savage, C. Speake, J. H. Buckner, X.-J. Li, T. F. Bumol, E. J. Wherry, T. R. Torgerson, L. A. Vella, S. E. Henrickson, P. J. Skene, C. E. Gustafson, Trimodal single-cell profiling reveals a novel pediatric CD8αα+ T cell subset and broad age-related molecular reprogramming across the T cell compartment. Nat. Immunol. 24, 1947–1959 (2023).
doi:10.1038/s41590-023-01641-8
39. C. S. Hopp, J. Skinner, S. L. Anzick, C. M. Tipton, M. E. Peterson, S. Li, S. Doumbo, K. Kayentao, A. Ongoiba, C. Martens, B. Traore, P. D. Crompton, Atypical B cells up-regulate costimulatory molecules during malaria and secrete antibodies with T follicular helper cell support. Sci. Immunol. 7, eabn1250 (2022).
doi:10.1126/sciimmunol.abn1250
40. B. Keller, V. Strohmeier, I. Harder, S. Unger, K. J. Payne, G. Andrieux, M. Boerries, P. T. Felixberger, J. J. M. Landry, A. Nieters, A. Rensing-Ehl, U. Salzer, N. Frede, S. Usadel, R. Elling, C. Speckmann, I. Hainmann, E. Ralph, K. Gilmour, M. W. J. Wentink, M. van der Burg, H. S. Kuehn, S. D. Rosenzweig, U. Kölsch, H. von Bernuth, P. Kaiser-Labusch, F. Gothe, S. Hambleton, A. D. Vlagea, A. Garcia Garcia, L. Alsina, G. Markelj, T. Avcin, J. Vasconcelos, M. Guedes, J.-Y. Ding, C.-L. Ku, B. Shadur, D. T. Avery, N. Venhoff, J. Thiel, H. Becker, L. Erazo-Borrás, C. M. Trujillo-Vargas, J. L. Franco, C. Fieschi, S. Okada, P. E. Gray, G. Uzel, J.-L. Casanova, M. Fliegauf, B. Grimbacher, H. Eibel, S. Ehl, R. E. Voll, M. Rizzi, P. Stepensky, V. Benes, C. S. Ma, C. Bossen, S. G. Tangye, K. Warnatz, The expansion of human T-bethighCD21low B cells is T cell dependent. Sci. Immunol. 6, eabh0891 (2021).
doi:10.1126/sciimmunol.abh0891
41. W. Zhang, H. Zhang, S. Liu, F. Xia, Z. Kang, Y. Zhang, Y. Liu, H. Xiao, L. Chen, C. Huang, N. Shen, H. Xu, F. Li, Excessive CD11c+Tbet+ B cells promote aberrant TFH differentiation and affinity-based germinal center selection in lupus. Proc. Natl. Acad. Sci. U. S. A. 116, 18550–18560 (2019).
doi:10.1073/pnas.1901340116
42. X. Gao, Q. Shen, J. A. Roco, B. Dalton, K. Frith, C. M. L. Munier, F. D. Ballard, K. Wang, H. G. Kelly, M. Nekrasov, J.-S. He, R. Jaeger, P. Carreira, J. I. Ellyard, L. Beattie, A. Enders, M. C. Cook, J. J. Zaunders, I. A. Cockburn, Zeb2 drives the formation of CD11c+ atypical B cells to sustain germinal centers that control persistent infection. Sci. Immunol. 9, eadj4748 (2024).
doi:10.1126/sciimmunol.adj4748
43. R. Argelaguet, B. Velten, D. Arnol, S. Dietrich, T. Zenz, J. C. Marioni, F. Buettner, W. Huber, O. Stegle, Multi-Omics Factor Analysis-a framework for unsupervised integration of multi-omics data sets. Mol. Syst. Biol. 14, e8124 (2018).
doi:10.15252/msb.20178124
44. E. Swanson, C. Lord, J. Reading, A. T. Heubeck, P. C. Genge, Z. Thomson, M. D. A. Weiss, X. J. Li, A. K. Savage, R. R. Green, T. R. Torgerson, T. F. Bumol, L. T. Graybuck, P. J. Skene, Simultaneous trimodal single-cell measurement of transcripts, epitopes, and chromatin accessibility using tea-seq. Elife 10 (2021).
doi:10.7554/ELIFE.63632
45. H. W. King, K. L. Wells, Z. Shipony, A. S. Kathiria, L. E. Wagar, C. Lareau, N. Orban, R. Capasso, M. M. Davis, L. M. Steinmetz, L. K. James, W. J. Greenleaf, Integrated single-cell transcriptomics and epigenomics reveals strong germinal center-associated etiology of autoimmune risk loci. Sci. Immunol. 6, eabh3768 (2021).
doi:10.1126/sciimmunol.abh3768
46. ENCODE Project Consortium, J. E. Moore, M. J. Purcaro, H. E. Pratt, C. B. Epstein, N. Shoresh, J. Adrian, T. Kawli, C. A. Davis, A. Dobin, R. Kaul, J. Halow, E. L. Van Nostrand, P. Freese, D. U. Gorkin, Y. Shen, Y. He, M. Mackiewicz, F. Pauli-Behn, B. A. Williams, A. Mortazavi, C. A. Keller, X.-O. Zhang, S. I. Elhajjajy, J. Huey, D. E. Dickel, V. Snetkova, X. Wei, X. Wang, J. C. Rivera-Mulia, J. Rozowsky, J. Zhang, S. B. Chhetri, J. Zhang, A. Victorsen, K. P. White, A. Visel, G. W. Yeo, C. B. Burge, E. Lécuyer, D. M. Gilbert, J. Dekker, J. Rinn, E. M. Mendenhall, J. R. Ecker, M. Kellis, R. J. Klein, W. S. Noble, A. Kundaje, R. Guigó, P. J. Farnham, J. M. Cherry, R. M. Myers, B. Ren, B. R. Graveley, M. B. Gerstein, L. A. Pennacchio, M. P. Snyder, B. E. Bernstein, B. Wold, R. C. Hardison, T. R. Gingeras, J. A. Stamatoyannopoulos, Z. Weng, Expanded encyclopaedias of DNA elements in the human and mouse genomes. Nature 583, 699–710 (2020).
doi:10.1038/s41586-020-2493-4
47. T. Iwasaki, R. Watanabe, H. Ito, T. Fujii, K. Ohmura, H. Yoshitomi, K. Murata, K. Murakami, A. Onishi, M. Tanaka, S. Matsuda, F. Matsuda, A. Morinobu, M. Hashimoto, Monocyte-derived transcriptomes explain the ineffectiveness of abatacept in rheumatoid arthritis. Arthritis Res. Ther. 26, 1 (2024).
doi:10.1186/s13075-023-03236-y
48. V. Farutin, T. Prod’homme, K. McConnell, N. Washburn, P. Halvey, C. J. Etzel, J. Guess, J. Duffner, K. Getchell, R. Meccariello, B. Gutierrez, C. Honan, G. Zhao, N. A. Cilfone, N. S. Gunay, J. L. Hillson, D. S. DeLuca, K. C. Saunders, D. A. Pappas, J. D. Greenberg, J. M. Kremer, A. M. Manning, L. E. Ling, I. Capila, Molecular profiling of rheumatoid arthritis patients reveals an association between innate and adaptive cell populations and response to anti-tumor necrosis factor. Arthritis Res. Ther. 21, 216 (2019).
doi:10.1186/s13075-019-1999-3
49. L. W. Moreland, R. Alten, F. Van den Bosch, T. Appelboom, M. Leon, P. Emery, S. Cohen, M. Luggen, W. Shergy, I. Nuamah, J.-C. Becker, Costimulatory blockade in patients with rheumatoid arthritis: a pilot, dose-finding, double-blind, placebo-controlled clinical trial evaluating CTLA-4Ig and LEA29Y eighty-five days after the first infusion. Arthritis Rheum. 46, 1470–1479 (2002).
doi:10.1002/art.10294
50. M. A. Kroenke, D. Eto, M. Locci, M. Cho, T. Davidson, E. K. Haddad, S. Crotty, Bcl6 and Maf cooperate to instruct human follicular helper CD4 T cell differentiation. J. Immunol. 188, 3734–3744 (2012).
doi:10.4049/jimmunol.1103246
51. Y. Chang, L. Bach, M. Hasiuk, L. Wen, T. Elmzzahi, C. Tsui, N. Gutiérrez-Melo, T. Steffen, D. T. Utzschneider, T. Raj, P. J. Jost, S. Heink, J. Cheng, O. T. Burton, J. Zeiträg, D. Alterauge, F. Dahlström, J.-C. Becker, M. Kastl, K. Symeonidis, M. van Uelft, M. Becker, S. Reschke, S. Krebs, H. Blum, Z. Abdullah, K. Paeschke, C. Ohnmacht, C. Neumann, A. Liston, F. Meissner, T. Korn, J. Hasenauer, V. Heissmeyer, M. Beyer, A. Kallies, L. T. Jeker, D. Baumjohann, TGF-β specifies TFH versus TH17 cell fates in murine CD4+ T cells through c-Maf. Sci. Immunol. 9, eadd4818 (2024).
doi:10.1126/sciimmunol.add4818
52. J. Zhao, Y. Chen, Q. Zhao, J. Shi, W. Yang, Z. Zhu, W. Yu, J. Guan, Y. Song, H. Wu, W. Jin, Y. Zhou, J. Liu, Increased circulating Tfh17 and PD-1+Tfh cells are associated with autoantibodies in Hashimoto’s thyroiditis. Autoimmunity 51, 352–359 (2018).
doi:10.1080/08916934.2018.1516761
53. R. Morita, N. Schmitt, S.-E. Bentebibel, R. Ranganathan, L. Bourdery, G. Zurawski, E. Foucat, M. Dullaers, S. Oh, N. Sabzghabaei, E. M. Lavecchio, M. Punaro, V. Pascual, J. Banchereau, H. Ueno, Human blood CXCR5+CD4+ T cells are counterparts of T follicular cells and contain specific subsets that differentially support antibody secretion. Immunity 34, 108–121 (2011).
doi:10.1016/j.immuni.2010.12.012
54. D. Rogers, A. Sood, H. Wang, J. J. P. van Beek, T. J. Rademaker, P. Artusa, C. Schneider, C. Shen, D. C. Wong, A. Bhagrath, M.-È. Lebel, S. A. Condotta, M. J. Richer, A. J. Martins, J. S. Tsang, L. B. Barreiro, P. François, D. Langlais, H. J. Melichar, J. Textor, J. N. Mandl, Pre-existing chromatin accessibility and gene expression differences among naive CD4+ T cells influence effector potential. Cell Rep. 37, 110064 (2021).
doi:10.1016/j.celrep.2021.110064
55. J. Choi, S. Crotty, Bcl6-mediated transcriptional regulation of follicular helper T cells (TFH). Trends Immunol. 42, 336–349 (2021).
doi:10.1016/j.it.2021.02.002
56. S. Fukuyo, S. Nakayamada, S. Iwata, S. Kubo, K. Saito, Y. Tanaka, Abatacept therapy reduces CD28+CXCR5+ follicular helper-like T cells in patients with rheumatoid arthritis. Clin. Exp. Rheumatol. 35, 562–570 (2017).
57. M. Nakou, G. Katsikas, P. Sidiropoulos, G. Bertsias, E. Papadimitraki, A. Raptopoulou, H. Koutala, H. A. Papadaki, H. Kritikos, D. T. Boumpas, Rituximab therapy reduces activated B cells in both the peripheral blood and bone marrow of patients with rheumatoid arthritis: depletion of memory B cells correlates with clinical response. Arthritis Res. Ther. 11, R131 (2009).
doi:10.1186/ar2798
58. H. Kristyanto, N. J. Blomberg, L. M. Slot, E. I. H. van der Voort, P. F. Kerkman, A. Bakker, L. E. Burgers, R. M. Ten Brinck, A. H. M. van der Helm-van Mil, H. Spits, D. L. Baeten, T. W. J. Huizinga, R. E. M. Toes, H. U. Scherer, Persistently activated, proliferative memory autoreactive B cells promote inflammation in rheumatoid arthritis. Sci. Transl. Med. 12 (2020).
doi:10.1126/scitranslmed.aaz5327
59. C. de Vries, W. Huang, R. K. Sharma, K. Wangriatisak, S. Turcinov, A. Cîrciumaru, L. Rönnblom, C. Grönwall, A. Hensvold, K. Lundberg, V. Malmström, Rheumatoid arthritis related B-cell changes are found already in the risk-RA phase. Eur. J. Immunol. 55, e202451391 (2025).
doi:10.1002/eji.202451391
60. A. Nellore, E. Zumaquero, C. D. Scharer, C. F. Fucile, C. M. Tipton, R. G. King, T. Mi, B. Mousseau, J. E. Bradley, F. Zhou, S. Mutneja, P. A. Goepfert, J. M. Boss, T. D. Randall, I. Sanz, A. F. Rosenberg, F. E. Lund, A transcriptionally distinct subset of influenza-specific effector memory B cells predicts long-lived antibody responses to vaccination in humans. Immunity 56, 847-863.e8 (2023).
doi:10.1016/j.immuni.2023.03.001
61. S. A. Jenks, K. S. Cashman, E. Zumaquero, U. M. Marigorta, A. V. Patel, X. Wang, D. Tomar, M. C. Woodruff, Z. Simon, R. Bugrovsky, E. L. Blalock, C. D. Scharer, C. M. Tipton, C. Wei, S. S. Lim, M. Petri, T. B. Niewold, J. H. Anolik, G. Gibson, F. E.-H. Lee, J. M. Boss, F. E. Lund, I. Sanz, Distinct Effector B Cells Induced by Unregulated Toll-like Receptor 7 Contribute to Pathogenic Responses in Systemic Lupus Erythematosus. Immunity 49, 725-739.e6 (2018).
doi:10.1016/j.immuni.2018.08.015
62. E. Wing, C. Sutherland, K. Miles, D. Gray, C. S. Goodyear, T. D. Otto, S. Breusch, G. Cowan, M. Gray, Double-negative-2 B cells are the major synovial plasma cell precursor in rheumatoid arthritis. Front. Immunol. 14, 1241474 (2023).
doi:10.3389/fimmu.2023.1241474
63. G. Dunlap, A. Wagner, N. Meednu, R. Wang, F. Zhang, J. C. Ekabe, A. H. Jonsson, K. Wei, S. Sakaue, A. Nathan, Accelerating Medicines Partnership Program: Rheumatoid Arthritis and Systemic Lupus Erythematosus (AMP RA/SLE) Network, V. P. Bykerk, L. T. Donlin, S. M. Goodman, G. S. Firestein, D. L. Boyle, V. M. Holers, L. W. Moreland, D. Tabechian, C. Pitzalis, A. Filer, S. Raychaudhuri, M. B. Brenner, J. Thakar, A. McDavid, D. A. Rao, J. H. Anolik, Clonal associations between lymphocyte subsets and functional states in rheumatoid arthritis synovium. Nat. Commun. 15, 4991 (2024).
doi:10.1038/s41467-024-49186-0
64. H. G. Evans, N. J. Gullick, S. Kelly, C. Pitzalis, G. M. Lord, B. W. Kirkham, L. S. Taams, In vivo activated monocytes from the site of inflammation in humans specifically promote Th17 responses. Proc. Natl. Acad. Sci. U. S. A. 106, 6232–6237 (2009).
doi:10.1073/pnas.0808144106
65. F. Zhang, A. H. Jonsson, A. Nathan, N. Millard, M. Curtis, Q. Xiao, M. Gutierrez-Arcelus, W. Apruzzese, G. F. M. Watts, D. Weisenfeld, S. Nayar, J. Rangel-Moreno, N. Meednu, K. E. Marks, I. Mantel, J. B. Kang, L. Rumker, J. Mears, K. Slowikowski, K. Weinand, D. E. Orange, L. Geraldino-Pardilla, K. D. Deane, D. Tabechian, A. Ceponis, G. S. Firestein, M. Maybury, I. Sahbudin, A. Ben-Artzi, A. M. Mandelin 2nd, A. Nerviani, M. J. Lewis, F. Rivellese, C. Pitzalis, L. B. Hughes, D. Horowitz, E. DiCarlo, E. M. Gravallese, B. F. Boyce, Accelerating Medicines Partnership: RA/SLE Network, L. W. Moreland, S. M. Goodman, H. Perlman, V. M. Holers, K. P. Liao, A. Filer, V. P. Bykerk, K. Wei, D. A. Rao, L. T. Donlin, J. H. Anolik, M. B. Brenner, S. Raychaudhuri, Deconstruction of rheumatoid arthritis synovium defines inflammatory subtypes. Nature 623, 616-624 (2023).
doi:10.1038/s41586-023-06708-y
66. D. Makrygiannakis, E. af Klint, I. E. Lundberg, R. Löfberg, A.-K. Ulfgren, L. Klareskog, A. I. Catrina, Citrullination is an inflammation-dependent process. Ann. Rheum. Dis. 65, 1219–1222 (2006).
doi:10.1136/ard.2005.049403
67. E. R. Vossenaar, T. J. M. Smeets, M. C. Kraan, J. M. Raats, W. J. van Venrooij, P. P. Tak, The presence of citrullinated proteins is not specific for rheumatoid synovial tissue. Arthritis Rheum. 50, 3485–3494 (2004).
doi:10.1002/art.20584
68. E. B. Prideaux, D. L. Boyle, E. Choi, J. H. Buckner, W. H. Robinson, V. M. Holers, K. D. Deane, G. S. Firestein, W. Wang, Epigenetic trajectory predicts development of clinical rheumatoid arthritis in ACPA+ individuals: Targeting Immune Responses for Prevention of Rheumatoid Arthritis (TIP-RA). bioRxiv 2024.10.15.618490 (2024).
doi:10.1101/2024.10.15.618490
69. C. A. Donado, E. Theisen, F. Zhang, A. Nathan, M. L. Fairfield, K. V. Rupani, D. Jones, K. P. Johannes, Accelerating Medicines Partnership RA/SLE Network, S. Raychaudhuri, D. F. Dwyer, A. H. Jonsson, M. B. Brenner, Granzyme K activates the entire complement cascade. Nature 641, 211-221 (2025).
doi:10.1038/s41586-025-08713-9
70. R. C. Brewer, T. V. Lanz, C. R. Hale, G. D. Sepich-Poore, C. Martino, A. D. Swafford, T. S. Carroll, S. Kongpachith, L. K. Blum, S. E. Elliott, N. E. Blachere, S. Parveen, J. Fak, V. Yao, O. Troyanskaya, M. O. Frank, M. S. Bloom, S. Jahanbani, A. M. Gomez, R. Iyer, N. S. Ramadoss, O. Sharpe, S. Chandrasekaran, L. B. Kelmenson, Q. Wang, H. Wong, H. L. Torres, M. Wiesen, D. T. Graves, K. D. Deane, V. M. Holers, R. Knight, R. B. Darnell, W. H. Robinson, D. E. Orange, Oral mucosal breaks trigger anti-citrullinated bacterial and human protein antibody responses in rheumatoid arthritis. Sci. Transl. Med. 15, eabq8476 (2023).
doi:10.1126/scitranslmed.abq8476
71. M. E. Chriswell, A. R. Lefferts, M. R. Clay, A. R. Hsu, J. Seifert, M. L. Feser, C. Rims, M. S. Bloom, E. A. Bemis, S. Liu, M. D. Maerz, D. N. Frank, M. K. Demoruelle, K. D. Deane, E. A. James, J. H. Buckner, W. H. Robinson, V. M. Holers, K. A. Kuhn, Clonal IgA and IgG autoantibodies from individuals at risk for rheumatoid arthritis identify an arthritogenic strain of Subdoligranulum. Sci. Transl. Med. 14, eabn5166 (2022).
doi:10.1126/scitranslmed.abq8476
72. N. Tajik, M. Frech, O. Schulz, F. Schälter, S. Lucas, V. Azizov, K. Dürholz, F. Steffen, Y. Omata, A. Rings, M. Bertog, A. Rizzo, A. Iljazovic, M. Basic, A. Kleyer, S. Culemann, G. Krönke, Y. Luo, K. Überla, U. S. Gaipl, B. Frey, T. Strowig, K. Sarter, S. C. Bischoff, S. Wirtz, J. D. Cañete, F. Ciccia, G. Schett, M. M. Zaiss, Targeting zonulin and intestinal epithelial barrier function to prevent onset of arthritis. Nat. Commun. 11, 1995 (2020).
doi:10.1038/s41467-020-15831-7
73. V. M. Holers, M. K. Demoruelle, K. A. Kuhn, J. H. Buckner, W. H. Robinson, Y. Okamoto, J. M. Norris, K. D. Deane, Rheumatoid arthritis and the mucosal origins hypothesis: protection turns to destruction. Nat. Rev. Rheumatol. 14, 542–557 (2018).
doi:10.1038/s41584-018-0070-0
74. D. I. Krijbolder, M. Verstappen, B. T. van Dijk, Y. J. Dakkak, L. E. Burgers, A. C. Boer, Y. J. Park, M. E. de Witt-Luth, K. Visser, M. R. Kok, E. T. H. Molenaar, P. H. P. de Jong, S. Böhringer, T. W. J. Huizinga, C. F. Allaart, E. Niemantsverdriet, A. H. M. van der Helm-van Mil, Intervention with methotrexate in patients with arthralgia at risk of rheumatoid arthritis to reduce the development of persistent arthritis and its disease burden (TREAT EARLIER): a randomised, double-blind, placebo-controlled, proof-of-concept trial. Lancet 400, 283–294 (2022).
doi:10.1016/S0140-6736(22)01193-X
75. L. Fraenkel, J. M. Bathon, B. R. England, E. W. St Clair, T. Arayssi, K. Carandang, K. D. Deane, M. Genovese, K. K. Huston, G. Kerr, J. Kremer, M. C. Nakamura, L. A. Russell, J. A. Singh, B. J. Smith, J. A. Sparks, S. Venkatachalam, M. E. Weinblatt, M. Al-Gibbawi, J. F. Baker, K. E. Barbour, J. L. Barton, L. Cappelli, F. Chamseddine, M. George, S. R. Johnson, L. Kahale, B. S. Karam, A. M. Khamis, I. Navarro-Millán, R. Mirza, P. Schwab, N. Singh, M. Turgunbaev, A. S. Turner, S. Yaacoub, E. A. Akl, 2021 American college of rheumatology guideline for the treatment of rheumatoid arthritis. Arthritis Rheumatol. 73, 1108–1123 (2021).
doi:10.1002/art.41752
76. D. Aletaha, T. Neogi, A. J. Silman, J. Funovits, D. T. Felson, C. O. Bingham 3rd, N. S. Birnbaum, G. R. Burmester, V. P. Bykerk, M. D. Cohen, B. Combe, K. H. Costenbader, M. Dougados, P. Emery, G. Ferraccioli, J. M. W. Hazes, K. Hobbs, T. W. J. Huizinga, A. Kavanaugh, J. Kay, T. K. Kvien, T. Laing, P. Mease, H. A. Ménard, L. W. Moreland, R. L. Naden, T. Pincus, J. S. Smolen, E. Stanislawska-Biernat, D. Symmons, P. P. Tak, K. S. Upchurch, J. Vencovsky, F. Wolfe, G. Hawker, 2010 rheumatoid arthritis classification criteria: an American College of Rheumatology/European League Against Rheumatism collaborative initiative. Ann. Rheum. Dis. 69, 1580–1588 (2010).
doi:10.1136/ard.2010.138461
77. V. Tsaltskan, K. Nguyen, C. Eaglin, V. M. Holers, K. D. Deane, G. S. Firestein, Comparison of web-based advertising and a social media platform as recruitment tools for underserved and hard-to-reach populations in rheumatology clinical research. ACR Open Rheumatol. 4, 623–630 (2022).
doi:10.1002/acr2.11448
78. P. C. Genge, C. R. Roll, A. T. Heubeck, E. Swanson, N. Kondza, C. Lord, M. Weiss, V. Hernandez, C. Phalen, Z. Thomson, T. R. Torgerson, P. J. Skene, T. F. Bumol, J. Reading, Optimized workflow for human PBMC multiomic immunosurveillance studies. STAR Protoc. 2, 100900 (2021).
doi:10.1016/j.xpro.2021.100900
79. A. Heubeck, A. Savage, K. Henderson, C. Roll, V. Hernandez, T. Torgerson, T. Bumol, J. Reading, Cross-platform immunophenotyping of human peripheral blood mononuclear cells with four high-dimensional flow cytometry panels. Cytometry A 103, 500-517 (2022).
doi:10.1002/cyto.a.24715
80. E. Swanson, J. Reading, L. T. Graybuck, P. J. Skene, BarWare: efficient software tools for barcoded single-cell genomics. BMC Bioinformatics 23, 106 (2022).
doi:10.1186/s12859-022-04620-2
81. Z. Gu, Complex heatmap visualization. Imeta 1, e43 (2022).
doi:10.1002/imt2.43
82. A. Kaushik, D. Dunham, Z. He, M. Manohar, M. Desai, K. C. Nadeau, S. Andorf, CyAnno: a semi-automated approach for cell type annotation of mass cytometry datasets. Bioinformatics 37, 4164–4171 (2021).
doi:10.1093/bioinformatics/btab409
83. F. A. Wolf, P. Angerer, F. J. Theis, SCANPY: large-scale single-cell gene expression data analysis. Genome Biol. 19, 15 (2018).
doi:10.1186/s13059-017-1382-0
84. D. R. Glass, A. G. Tsai, J. P. Oliveria, F. J. Hartmann, S. C. Kimmey, A. A. Calderon, L. Borges, M. C. Glass, L. E. Wagar, M. M. Davis, S. C. Bendall, An Integrated Multi-omic Single-Cell Atlas of Human B Cell Identity. Immunity 53, 217-232.e5 (2020).
doi:10.1016/j.immuni.2020.06.013
85. M. C. Glass, D. R. Glass, J.-P. Oliveria, B. Mbiribindi, C. O. Esquivel, S. M. Krams, S. C. Bendall, O. M. Martinez, Human IL-10-producing B cells have diverse states that are induced from multiple B cell subsets. Cell Rep. 39 (2022).
doi:10.1016/j.celrep.2022.110728
86. Q. Blampey, N. Bercovici, C.-A. Dutertre, I. Pic, J. M. Ribeiro, F. André, P.-H. Cournède, A biology-driven deep generative model for cell-type annotation in cytometry. Brief. Bioinform. 24, bbad260 (2023).
doi:10.1093/bib/bbad260
87. K. Quintelier, A. Couckuyt, A. Emmaneel, J. Aerts, Y. Saeys, S. Van Gassen, Analyzing high-dimensional cytometry data using FlowSOM. Nat. Protoc. 16, 3775–3801 (2021).
doi:10.1038/s41596-021-00550-0
88. P. Meijer, N. Howard, J. Liang, A. Kelsey, S. Subramanian, E. Johnson, P. Mariz, J. Harvey, M. Ambrose, V. Tereshchenko, A. Beaubien, N. Inala, Y. Aggoune, S. Pister, A. Vetto, M. Kinsey, T. Bumol, A. Goldrath, X. Li, T. Torgerson, P. Skene, L. Okada, C. La France, Z. Thomson, L. Graybuck, Provide proactive reproducible analysis transparency with every publication. arXiv (2024). https://arxiv.org/abs/2408.09103
89. S. L. Wolock, R. Lopez, A. M. Klein, Scrublet: Computational Identification of Cell Doublets in Single-Cell Transcriptomic Data. Cell Syst. 8, 281-291.e9 (2019).
doi:10.1016/j.cels.2018.11.005
90. C. Domínguez Conde, C. Xu, L. B. Jarvis, D. B. Rainbow, S. B. Wells, T. Gomes, S. K. Howlett, O. Suchanek, K. Polanski, H. W. King, L. Mamanova, N. Huang, P. A. Szabo, L. Richardson, L. Bolt, E. S. Fasouli, K. T. Mahbubani, M. Prete, L. Tuck, N. Richoz, Z. K. Tuong, L. Campos, H. S. Mousa, E. J. Needham, S. Pritchard, T. Li, R. Elmentaite, J. Park, E. Rahmani, D. Chen, D. K. Menon, O. A. Bayraktar, L. K. James, K. B. Meyer, N. Yosef, M. R. Clatworthy, P. A. Sims, D. L. Farber, K. Saeb-Parsy, J. L. Jones, S. A. Teichmann, Cross-tissue immune cell analysis reveals tissue-specific features in humans. Science 376, eabl5197 (2022).
doi:10.1126/science.abl5197
91. I. Korsunsky, N. Millard, J. Fan, K. Slowikowski, F. Zhang, K. Wei, Y. Baglaenko, M. Brenner, P.-R. Loh, S. Raychaudhuri, Fast, sensitive and accurate integration of single-cell data with Harmony. Nat. Methods 16, 1289–1296 (2019).
doi:10.1038/s41592-019-0619-0
92. G. V. Zuccarino-Catania, S. Sadanand, F. J. Weisel, M. M. Tomayko, H. Meng, S. H. Kleinstein, K. L. Good-Jacobson, M. J. Shlomchik, CD80 and PD-L2 define functionally distinct memory B cell subsets that are independent of antibody isotype. Nat. Immunol. 15, 631–637 (2014).
doi:10.1038/ni.2914
93. M. I. Love, W. Huber, S. Anders, Moderated estimation of fold change and dispersion for RNA-seq data with DESeq2. Genome Biol. 15, 550 (2014).
doi:10.1186/s13059-014-0550-8
94. P. Badia-I-Mompel, J. Vélez Santiago, J. Braunger, C. Geiss, D. Dimitrov, S. Müller-Dott, P. Taus, A. Dugourd, C. H. Holland, R. O. Ramirez Flores, J. Saez-Rodriguez, decoupleR: ensemble of computational methods to infer biological activities from omics data. Bioinform. Adv. 2, vbac016 (2022).
doi:10.1093/bioadv/vbac016
95. M. Büttner, J. Ostner, C. L. Müller, F. J. Theis, B. Schubert, scCODA is a Bayesian model for compositional single-cell data analysis. Nat. Commun. 12, 6876 (2021).
doi:10.1038/s41467-021-27150-6
96. S. V. Vasaikar, A. K. Savage, Q. Gong, E. Swanson, A. Talla, C. Lord, A. T. Heubeck, J. Reading, L. T. Graybuck, P. Meijer, T. R. Torgerson, P. J. Skene, T. F. Bumol, X.-J. Li, A comprehensive platform for analyzing longitudinal multi-omics data. Nat. Commun. 14, 1684 (2023).
doi:10.1038/s41467-023-37432-w
97. R. Z. Kunes, T. Walle, M. Land, T. Nawy, D. Pe’er, Supervised discovery of interpretable gene programs from single-cell data. Nat. Biotechnol. 42, 1084–1095 (2024).
doi:10.1038/s41587-023-01940-3
98. A. T. L. Lun, K. Bach, J. C. Marioni, Pooling across cells to normalize single-cell RNA sequencing data with many zero counts. Genome Biol. 17, 75 (2016).
doi:10.1186/s13059-016-0947-7
99. A. Federico, S. Monti, hypeR: an R package for geneset enrichment workflows. Bioinformatics 36, 1307–1308 (2020).
doi:10.1093/bioinformatics/btz700
100. A. J. Matthews, S. Zheng, L. J. DiMenna, J. Chaudhuri, Regulation of immunoglobulin class-switch recombination: choreography of noncoding transcription, targeted DNA deamination, and long-range DNA repair. Adv. Immunol. 122, 1–57 (2014).
doi:10.1016/B978-0-12-800267-4.00001-8
101. J. Stavnezer, Immunoglobulin class switching. Curr. Opin. Immunol. 8, 199–205 (1996).
doi:10.1016/S0952-7915(96)80058-6
102. M. Lorenz, S. Jung, A. Radbruch, Switch transcripts in immunoglobulin class switching. Science 267, 1825–1828 (1995).
doi:10.1126/science.7892607
103. D. Dimitrov, D. Türei, M. Garrido-Rodriguez, P. L. Burmedi, J. S. Nagai, C. Boys, R. O. Ramirez Flores, H. Kim, B. Szalai, I. G. Costa, A. Valdeolivas, A. Dugourd, J. Saez-Rodriguez, Comparison of methods and resources for cell-cell communication inference from single-cell RNA-Seq data. Nat. Commun. 13, 3224 (2022).
doi:10.1038/s41467-022-30755-0
104. M. Efremova, M. Vento-Tormo, S. A. Teichmann, R. Vento-Tormo, CellPhoneDB: inferring cell-cell communication from combined expression of multi-subunit ligand-receptor complexes. Nat. Protoc. 15, 1484–1506 (2020).
doi:10.1038/s41596-020-0292-x
105. M. S. B. Raredon, J. Yang, J. Garritano, M. Wang, D. Kushnir, J. C. Schupp, T. S. Adams, A. M. Greaney, K. L. Leiby, N. Kaminski, Y. Kluger, A. Levchenko, L. E. Niklason, Computation and visualization of cell-cell signaling topologies in single-cell systems data using Connectome. Sci. Rep. 12, 4187 (2022).
doi:10.1038/s41598-022-07959-x
106. S. Cabello-Aguilar, M. Alame, F. Kon-Sun-Tack, C. Fau, M. Lacroix, J. Colinge, SingleCellSignalR: inference of intercellular networks from single-cell transcriptomics. Nucleic Acids Res. 48, e55 (2020).
doi:10.1093/nar/gkaa183
107. R. Browaeys, W. Saelens, Y. Saeys, NicheNet: modeling intercellular communication by linking ligands to target genes. Nat. Methods 17, 159–162 (2020).
doi:10.1038/s41592-019-0667-5
108. D. Bredikhin, I. Kats, O. Stegle, MUON: multimodal omics analysis framework. Genome Biol. 23, 42 (2022).
doi:10.1186/s13059-021-02577-8
109. M. P. Mulè, A. J. Martins, J. S. Tsang, Normalizing and denoising protein expression data from droplet-based single cell profiling. bioRxiv 2020.02.24.963603 (2021).
doi:10.1101/2020.02.24.963603
110. J. M. Granja, M. R. Corces, S. E. Pierce, S. T. Bagdatli, H. Choudhry, H. Y. Chang, W. J. Greenleaf, ArchR is a scalable software package for integrative single-cell chromatin accessibility analysis. Nature Genetics 2021 53, 403–411 (2021).
doi:10.1038/s41588-021-00790-6
111. S. Rachid Zaim, M.-P. Pebworth, I. McGrath, L. Okada, M. Weiss, J. Reading, J. L. Czartoski, T. R. Torgerson, M. J. McElrath, T. F. Bumol, P. J. Skene, X.-J. Li, MOCHA’s advanced statistical modeling of scATAC-seq data enables functional genomic inference in large human cohorts. Nat. Commun. 15, 6828 (2024).
doi:10.1038/s41467-024-50612-6
112. A. N. Schep, B. Wu, J. D. Buenrostro, W. J. Greenleaf, chromVAR: inferring transcription-factor-associated accessibility from single-cell epigenomic data. Nat. Methods 14, 975–978 (2017).
doi:10.1038/nmeth.4401
113. R. Argelaguet, D. Arnol, D. Bredikhin, Y. Deloro, B. Velten, J. C. Marioni, O. Stegle, MOFA+: a statistical framework for comprehensive integration of multi-modal single-cell data. Genome Biol. 21, 111 (2020).
doi:10.1186/s13059-020-02015-1