Tissue immunity to SARS-CoV-2: Role in protection and immunopathology*
Ksenia Rybkina
Department of Microbiology and Immunology, Columbia University Vagelos College of Physicians and Surgeons, New York, New York, USA
Search for more papers by this authorJulia Davis-Porada
Department of Microbiology and Immunology, Columbia University Vagelos College of Physicians and Surgeons, New York, New York, USA
Search for more papers by this authorCorresponding Author
Donna L. Farber
Department of Microbiology and Immunology, Columbia University Vagelos College of Physicians and Surgeons, New York, New York, USA
Department of Surgery, Columbia University Irving Medical Center, New York, New York, USA
Correspondence
Donna L. Farber, Department of Microbiology and Immunology, Columbia University Vagelos College of Physicians and Surgeons, New York, NY 10032, USA.
Email: [email protected]
Search for more papers by this authorKsenia Rybkina
Department of Microbiology and Immunology, Columbia University Vagelos College of Physicians and Surgeons, New York, New York, USA
Search for more papers by this authorJulia Davis-Porada
Department of Microbiology and Immunology, Columbia University Vagelos College of Physicians and Surgeons, New York, New York, USA
Search for more papers by this authorCorresponding Author
Donna L. Farber
Department of Microbiology and Immunology, Columbia University Vagelos College of Physicians and Surgeons, New York, New York, USA
Department of Surgery, Columbia University Irving Medical Center, New York, New York, USA
Correspondence
Donna L. Farber, Department of Microbiology and Immunology, Columbia University Vagelos College of Physicians and Surgeons, New York, NY 10032, USA.
Email: [email protected]
Search for more papers by this author*This article is part of a series of reviews covering SARS-CoV-2 Immunity appearing in Volume 309 of Immunological Reviews.
Ksenia Rybkina and Julia Davis-Porada contributed equally to this study.
Summary
The SARS-CoV-2 pandemic has demonstrated the importance of studying antiviral immunity within sites of infection to gain insights into mechanisms for immune protection and disease pathology. As SARS-CoV-2 is tropic to the respiratory tract, many studies of airway washes, lymph node aspirates, and postmortem lung tissue have revealed site-specific immune dynamics that are associated with the protection or immunopathology but are not readily observed in circulation. This review summarizes the growing body of work identifying immune processes in tissues and their interplay with immune responses in circulation during acute SARS-CoV-2 infection, severe disease, and memory persistence. Establishment of tissue resident immunity also may have implications for vaccination and the durability of immune memory and protection.
CONFLICT OF INTEREST
There are no conflicts of interest.
Open Research
DATA AVAILABILITY STATEMENT
Data sharing not applicable to this article as no datasets were generated or analysed during the current study
REFERENCES
- 1Kustin T, Harel N, Finkel U, et al. Evidence for increased breakthrough rates of SARS-CoV-2 variants of concern in BNT162b2-mRNA-vaccinated individuals. Nat Med. 2021; 27(8): 1379-1384.
- 2Abdool Karim SS, de Oliveira T. New SARS-CoV-2 variants – clinical, public health, and vaccine implications. N Engl J Med. 2021; 384(19): 1866-1868.
- 3Wang P, Casner RG, Nair MS, et al. Increased resistance of SARS-CoV-2 variant P.1 to antibody neutralization. Cell Host Microbe. 2021; 29(5): 747-751 e744.
- 4Ardain A, Marakalala MJ, Leslie A. Tissue-resident innate immunity in the lung. Immunology. 2020; 159(3): 245-256.
- 5Arimori Y, Nakamura R, Yamada H, et al. Type I interferon limits influenza virus-induced acute lung injury by regulation of excessive inflammation in mice. Antiviral Res. 2013; 99(3): 230-237.
- 6Teijaro JR, Turner D, Pham Q, Wherry EJ, Lefrancois L, Farber DL. Cutting edge: tissue-retentive lung memory CD4 T cells mediate optimal protection to respiratory virus infection. J Immunol. 2011; 187(11): 5510-5514.
- 7Son YM, Cheon IS, Wu Y, et al. Tissue-resident CD4(+) T helper cells assist the development of protective respiratory B and CD8(+) T cell memory responses. Sci Immunol. 2021; 6(55):eabb6852.
- 8Paik DH, Farber DL. Anti-viral protective capacity of tissue resident memory T cells. Curr Opin Virol. 2020; 46: 20-26.
- 9Paik DH, Farber DL. Influenza infection fortifies local lymph nodes to promote lung-resident heterosubtypic immunity. J Exp Med. 2021; 218(1):e20200218.
- 10Kinnear E, Lambert L, McDonald JU, Cheeseman HM, Caproni LJ, Tregoning JS. Airway T cells protect against RSV infection in the absence of antibody. Mucosal Immunol. 2018; 11(1): 249-256.
- 11Luangrath MA, Schmidt ME, Hartwig SM, Varga SM. Tissue-resident memory T cells in the lungs protect against acute respiratory syncytial virus infection. Immunohorizons. 2021; 5(2): 59-69.
- 12Zhao J, Zhao J, Mangalam AK, et al. Airway memory CD4(+) T cells mediate protective immunity against emerging respiratory coronaviruses. Immunity. 2016; 44(6): 1379-1391.
- 13Allie SR, Bradley JE, Mudunuru U, et al. The establishment of resident memory B cells in the lung requires local antigen encounter. Nat Immunol. 2019; 20(1): 97-108.
- 14Szabo PA, Miron M, Farber DL. Location, location, location: tissue resident memory T cells in mice and humans. Sci Immunol. 2019; 4(34):eaas9673.
- 15Kumar BV, Ma W, Miron M, et al. Human tissue-resident memory T cells are defined by core transcriptional and functional signatures in lymphoid and mucosal sites. Cell Rep. 2017; 20(12): 2921-2934.
- 16Weisel NM, Weisel FJ, Farber DL, et al. Comprehensive analyses of B-cell compartments across the human body reveal novel subsets and a gut-resident memory phenotype. Blood. 2020; 136(24): 2774-2785.
- 17Poon MML, Byington E, Meng W, et al. Heterogeneity of human anti-viral immunity shaped by virus, tissue, age, and sex. Cell Rep. 2021; 37(9):110071.
- 18Pizzolla A, Nguyen TH, Sant S, et al. Influenza-specific lung-resident memory T cells are proliferative and polyfunctional and maintain diverse TCR profiles. J Clin Invest. 2018; 128(2): 721-733.
- 19Wu Z, McGoogan JM. Characteristics of and important lessons from the coronavirus disease 2019 (COVID-19) outbreak in China: summary of a report of 72314 cases from the Chinese Center for Disease Control and Prevention. JAMA. 2020; 323(13): 1239-1242.
- 20Sungnak W, Huang N, Becavin C, et al. SARS-CoV-2 entry factors are highly expressed in nasal epithelial cells together with innate immune genes. Nat Med. 2020; 26(5): 681-687.
- 21Hoffmann M, Kleine-Weber H, Schroeder S, et al. SARS-CoV-2 cell entry depends on ACE2 and TMPRSS2 and is blocked by a clinically proven protease inhibitor. Cell. 2020; 181(2): 271-280 e278.
- 22Yan R, Zhang Y, Li Y, Xia L, Guo Y, Zhou Q. Structural basis for the recognition of SARS-CoV-2 by full-length human ACE2. Science. 2020; 367(6485): 1444-1448.
- 23Hou YJ, Okuda K, Edwards CE, et al. SARS-CoV-2 reverse genetics reveals a variable infection gradient in the respiratory tract. Cell. 2020; 182(2): 429-446 e414.
- 24Delorey TM, Ziegler CGK, Heimberg G, et al. COVID-19 tissue atlases reveal SARS-CoV-2 pathology and cellular targets. Nature. 2021; 595(7865): 107-113.
- 25Desai N, Neyaz A, Szabolcs A, et al. Temporal and spatial heterogeneity of host response to SARS-CoV-2 pulmonary infection. Nat Commun. 2020; 11(1): 6319.
- 26Puelles VG, Lutgehetmann M, Lindenmeyer MT, et al. Multiorgan and renal tropism of SARS-CoV-2. N Engl J Med. 2020; 383(6): 590-592.
- 27Wolfel R, Corman VM, Guggemos W, et al. Virological assessment of hospitalized patients with COVID-2019. Nature. 2020; 581(7809): 465-469.
- 28Bost P, De Sanctis F, Cane S, et al. Deciphering the state of immune silence in fatal COVID-19 patients. Nat Commun. 2021; 12(1): 1428.
- 29Grant RA, Morales-Nebreda L, Markov NS, et al. Circuits between infected macrophages and T cells in SARS-CoV-2 pneumonia. Nature. 2021; 590(7847): 635-641.
- 30Speranza E, Williamson BN, Feldmann F, et al. Single-cell RNA sequencing reveals SARS-CoV-2 infection dynamics in lungs of African green monkeys. Sci Transl Med. 2021; 13(578):eabe8146.
- 31Bruder D, Srikiatkhachorn A, Enelow RI. Cellular immunity and lung injury in respiratory virus infection. Viral Immunol. 2006; 19(2): 147-155.
- 32de Jong MD, Simmons CP, Thanh TT, et al. Fatal outcome of human influenza A (H5N1) is associated with high viral load and hypercytokinemia. Nat Med. 2006; 12(10): 1203-1207.
- 33Nienhold R, Ciani Y, Koelzer VH, et al. Two distinct immunopathological profiles in autopsy lungs of COVID-19. Nat Commun. 2020; 11(1): 5086.
- 34Melms JC, Biermann J, Huang H, et al. A molecular single-cell lung atlas of lethal COVID-19. Nature. 2021; 595(7865): 114-119.
- 35D'Agnillo F, Walters KA, Xiao Y, et al. Lung epithelial and endothelial damage, loss of tissue repair, inhibition of fibrinolysis, and cellular senescence in fatal COVID-19. Sci Transl Med. 2021; 13(620):eabj7790.
- 36Ziegler CGK, Allon SJ, Nyquist SK, et al. SARS-CoV-2 receptor ACE2 is an interferon-stimulated gene in human airway epithelial cells and is detected in specific cell subsets across tissues. Cell. 2020; 181(5): 1016-1035 e1019.
- 37Roach A, Chikwe J, Catarino P, et al. Lung transplantation for Covid-19-related respiratory failure in the United States. N Engl J Med. 2022; 386(12): 1187-1188.
- 38Gupta A, Madhavan MV, Sehgal K, et al. Extrapulmonary manifestations of COVID-19. Nat Med. 2020; 26(7): 1017-1032.
- 39Feldstein LR, Tenforde MW, Friedman KG, et al. Characteristics and outcomes of US children and adolescents with multisystem inflammatory syndrome in children (MIS-C) compared with severe acute COVID-19. JAMA. 2021; 325(11): 1074-1087.
- 40Premraj L, Kannapadi NV, Briggs J, et al. Mid and long-term neurological and neuropsychiatric manifestations of post-COVID-19 syndrome: a meta-analysis. J Neurol Sci. 2022; 434:120162.
- 41Freeman EE, McMahon DE, Lipoff JB, et al. The spectrum of COVID-19-associated dermatologic manifestations: an international registry of 716 patients from 31 countries. J Am Acad Dermatol. 2020; 83(4): 1118-1129.
- 42Dolhnikoff M, Ferreira Ferranti J, de Almeida Monteiro RA, et al. SARS-CoV-2 in cardiac tissue of a child with COVID-19-related multisystem inflammatory syndrome. Lancet Child Adolesc Health. 2020; 4(10): 790-794.
- 43Duarte-Neto AN, Caldini EG, Gomes-Gouvea MS, et al. An autopsy study of the spectrum of severe COVID-19 in children: from SARS to different phenotypes of MIS-C. EClinicalMedicine. 2021; 35:100850.
- 44Sperotto F, Friedman KG, Son MBF, VanderPluym CJ, Newburger JW, Dionne A. Cardiac manifestations in SARS-CoV-2-associated multisystem inflammatory syndrome in children: a comprehensive review and proposed clinical approach. Eur J Pediatr. 2021; 180(2): 307-322.
- 45Smith N, Goncalves P, Charbit B, et al. Distinct systemic and mucosal immune responses during acute SARS-CoV-2 infection. Nat Immunol. 2021; 22(11): 1428-1439.
- 46Szabo PA, Dogra P, Gray JI, et al. Longitudinal profiling of respiratory and systemic immune responses reveals myeloid cell-driven lung inflammation in severe COVID-19. Immunity. 2021; 54(4): 797-814 e796.
- 47Vu DL, Martinez-Murillo P, Pigny F, et al. Longitudinal analysis of inflammatory response to SARS-CoV-2 in the upper respiratory tract reveals an association with viral load, independent of symptoms. J Clin Immunol. 2021; 41(8): 1723-1732.
- 48Chua RL, Lukassen S, Trump S, et al. COVID-19 severity correlates with airway epithelium-immune cell interactions identified by single-cell analysis. Nat Biotechnol. 2020; 38(8): 970-979.
- 49Liao M, Liu Y, Yuan J, et al. Single-cell landscape of bronchoalveolar immune cells in patients with COVID-19. Nat Med. 2020; 26(6): 842-844.
- 50Reynolds D, Vazquez Guillamet C, Day A, et al. Comprehensive immunologic evaluation of bronchoalveolar lavage samples from human patients with moderate and severe seasonal influenza and severe COVID-19. J Immunol. 2021; 207(5): 1229-1238.
- 51Zhao Y, Kilian C, Turner JE, et al. Clonal expansion and activation of tissue-resident memory-like Th17 cells expressing GM-CSF in the lungs of severe COVID-19 patients. Sci Immunol. 2021; 6(56):eabf6692.
- 52Zhou Z, Ren L, Zhang L, et al. Heightened innate immune responses in the respiratory tract of COVID-19 patients. Cell Host Microbe. 2020; 27(6): 883-890 e882.
- 53Hadjadj J, Yatim N, Barnabei L, et al. Impaired type I interferon activity and inflammatory responses in severe COVID-19 patients. Science. 2020; 369(6504): 718-724.
- 54Mudd PA, Crawford JC, Turner JS, et al. Distinct inflammatory profiles distinguish COVID-19 from influenza with limited contributions from cytokine storm. Sci Adv. 2020; 6(50):eabe3024.
- 55Guo XJ, Thomas PG. New fronts emerge in the influenza cytokine storm. Semin Immunopathol. 2017; 39(5): 541-550.
- 56Lucas C, Wong P, Klein J, et al. Longitudinal analyses reveal immunological misfiring in severe COVID-19. Nature. 2020; 584(7821): 463-469.
- 57Wilk AJ, Rustagi A, Zhao NQ, et al. A single-cell atlas of the peripheral immune response in patients with severe COVID-19. Nat Med. 2020; 26(7): 1070-1076.
- 58Schulte-Schrepping J, Reusch N, Paclik D, et al. Severe COVID-19 is marked by a dysregulated myeloid cell compartment. Cell. 2020; 182(6): 1419-1440 e1423.
- 59Ouwendijk WJD, Raadsen MP, van Kampen JJA, et al. High levels of neutrophil extracellular traps persist in the lower respiratory tract of critically ill patients with coronavirus disease 2019. J Infect Dis. 2021; 223(9): 1512-1521.
- 60Veras FP, Pontelli MC, Silva CM, et al. SARS-CoV-2-triggered neutrophil extracellular traps mediate COVID-19 pathology. J Exp Med. 2020; 217(12):e20201129.
- 61Grant EJ, Josephs TM, Loh L, et al. Broad CD8(+) T cell cross-recognition of distinct influenza A strains in humans. Nat Commun. 2018; 9(1): 5427.
- 62Roukens AHE, Pothast CR, Konig M, et al. Prolonged activation of nasal immune cell populations and development of tissue-resident SARS-CoV-2-specific CD8(+) T cell responses following COVID-19. Nat Immunol. 2022; 23(1): 23-32.
- 63Zhang F, Mears JR, Shakib L, et al. IFN-gamma and TNF-alpha drive a CXCL10+ CCL2+ macrophage phenotype expanded in severe COVID-19 lungs and inflammatory diseases with tissue inflammation. Genome Med. 2021; 13(1): 64.
- 64Amanat F, Stadlbauer D, Strohmeier S, et al. A serological assay to detect SARS-CoV-2 seroconversion in humans. Nat Med. 2020; 26(7): 1033-1036.
- 65Long QX, Liu BZ, Deng HJ, et al. Antibody responses to SARS-CoV-2 in patients with COVID-19. Nat Med. 2020; 26(6): 845-848.
- 66Luchsinger LL, Ransegnola BP, Jin DK, et al. Serological assays estimate highly variable SARS-CoV-2 neutralizing antibody activity in recovered COVID-19 patients. J Clin Microbiol. 2020; 58(12):e02005-20.
- 67Pickering S, Betancor G, Galao RP, et al. Comparative assessment of multiple COVID-19 serological technologies supports continued evaluation of point-of-care lateral flow assays in hospital and community healthcare settings. PLoS Pathog. 2020; 16(9):e1008817.
- 68Feng S, Phillips DJ, White T, et al. Correlates of protection against symptomatic and asymptomatic SARS-CoV-2 infection. Nat Med. 2021; 27(11): 2032-2040.
- 69Seow J, Graham C, Merrick B, et al. Longitudinal observation and decline of neutralizing antibody responses in the three months following SARS-CoV-2 infection in humans. Nat Microbiol. 2020; 5(12): 1598-1607.
- 70Roltgen K, Powell AE, Wirz OF, et al. Defining the features and duration of antibody responses to SARS-CoV-2 infection associated with disease severity and outcome. Sci Immunol. 2020; 5(54):eabe0240.
- 71Weisberg SP, Connors TJ, Zhu Y, et al. Distinct antibody responses to SARS-CoV-2 in children and adults across the COVID-19 clinical spectrum. Nat Immunol. 2021; 22(1): 25-31.
- 72Liu L, To KK, Chan KH, et al. High neutralizing antibody titer in intensive care unit patients with COVID-19. Emerg Microbes Infect. 2020; 9(1): 1664-1670.
- 73Lucas C, Klein J, Sundaram ME, et al. Delayed production of neutralizing antibodies correlates with fatal COVID-19. Nat Med. 2021; 27(7): 1178-1186.
- 74Sterlin D, Mathian A, Miyara M, et al. IgA dominates the early neutralizing antibody response to SARS-CoV-2. Sci Transl Med. 2021; 13(577):eabd2223.
- 75Cagigi A, Yu M, Osterberg B, et al. Airway antibodies emerge according to COVID-19 severity and wane rapidly but reappear after SARS-CoV-2 vaccination. JCI Insight. 2021; 6(22):e151463.
- 76Froberg J, Gillard J, Philipsen R, et al. SARS-CoV-2 mucosal antibody development and persistence and their relation to viral load and COVID-19 symptoms. Nat Commun. 2021; 12(1): 5621.
- 77Sokal A, Chappert P, Barba-Spaeth G, et al. Maturation and persistence of the anti-SARS-CoV-2 memory B cell response. Cell. 2021; 184(5): 1201-1213 e1214.
- 78Sakharkar M, Rappazzo CG, Wieland-Alter WF, et al. Prolonged evolution of the human B cell response to SARS-CoV-2 infection. Sci Immunol. 2021; 6(56):eabg6916.
- 79Dan JM, Mateus J, Kato Y, et al. Immunological memory to SARS-CoV-2 assessed for up to 8 months after infection. Science. 2021; 371(6529):eabf4063.
- 80Victora GD, Nussenzweig MC. Germinal centers. Annu Rev Immunol. 2012; 30: 429-457.
- 81Gaebler C, Wang Z, Lorenzi JCC, et al. Evolution of antibody immunity to SARS-CoV-2. Nature. 2021; 591(7851): 639-644.
- 82Haslbauer JD, Matter MS, Stalder AK, Tzankov A. Histomorphological patterns of regional lymph nodes in COVID-19 lungs. Pathologe. 2021; 42(Suppl 1): 89-97.
- 83Kaneko N, Kuo HH, Boucau J, et al. Loss of Bcl-6-expressing T follicular helper cells and germinal centers in COVID-19. Cell. 2020; 183(1): 143-157 e113.
- 84Roltgen K, Nielsen SCA, Silva O, et al. Immune imprinting, breadth of variant recognition, and germinal center response in human SARS-CoV-2 infection and vaccination. Cell. 2022; 185(6): 1025-1040 e1014.
- 85Hoehn KB, Turner JS, Miller FI, et al. Human B cell lineages associated with germinal centers following influenza vaccination are measurably evolving. Elife. 2021; 10:e70873.
- 86Dan JM, Lindestam Arlehamn CS, Weiskopf D, et al. A cytokine-independent approach to identify antigen-specific human germinal center T follicular helper cells and rare antigen-specific CD4+ T cells in blood. J Immunol. 2016; 197(3): 983-993.
- 87Grifoni A, Weiskopf D, Ramirez SI, et al. Targets of T cell responses to SARS-CoV-2 coronavirus in humans with COVID-19 disease and unexposed individuals. Cell. 2020; 181(7): 1489-1501 e1415.
- 88Rydyznski Moderbacher C, Ramirez SI, Dan JM, et al. Antigen-specific adaptive immunity to SARS-CoV-2 in acute COVID-19 and associations with age and disease severity. Cell. 2020; 183(4): 996-1012 e1019.
- 89Weiskopf D, Schmitz KS, Raadsen MP, et al. Phenotype and kinetics of SARS-CoV-2-specific T cells in COVID-19 patients with acute respiratory distress syndrome. Sci Immunol. 2020; 5(48):eabd2071.
- 90Hogan RJ, Usherwood EJ, Zhong W, et al. Activated antigen-specific CD8+ T cells persist in the lungs following recovery from respiratory virus infections. J Immunol. 2001; 166(3): 1813-1822.
- 91Heide J, Schulte S, Kohsar M, et al. Broadly directed SARS-CoV-2-specific CD4+ T cell response includes frequently detected peptide specificities within the membrane and nucleoprotein in patients with acute and resolved COVID-19. PLoS Pathog. 2021; 17(9):e1009842.
- 92Gangaev A, Ketelaars SLC, Isaeva OI, et al. Identification and characterization of a SARS-CoV-2 specific CD8(+) T cell response with immunodominant features. Nat Commun. 2021; 12(1): 2593.
- 93Dai Y, Wang J, Jeong HH, Chen W, Jia P, Zhao Z. Association of CXCR6 with COVID-19 severity: delineating the host genetic factors in transcriptomic regulation. Hum Genet. 2021; 140(9): 1313-1328.
- 94Rodda LB, Netland J, Shehata L, et al. Functional SARS-CoV-2-specific immune memory persists after mild COVID-19. Cell. 2021; 184(1): 169-183 e117.
- 95Balachandran H, Phetsouphanh C, Agapiou D, et al. Maintenance of broad neutralizing antibodies and memory B cells 1 year post-infection is predicted by SARS-CoV-2-specific CD4+ T cell responses. Cell Rep. 2022; 38(6):110345.
- 96Milne G, Hames T, Scotton C, et al. Does infection with or vaccination against SARS-CoV-2 lead to lasting immunity? Lancet Respir Med. 2021; 9(12): 1450-1466.
- 97Li C, Yu D, Wu X, et al. Twelve-month specific IgG response to SARS-CoV-2 receptor-binding domain among COVID-19 convalescent plasma donors in Wuhan. Nat Commun. 2021; 12(1): 4144.
- 98Abayasingam A, Balachandran H, Agapiou D, et al. Long-term persistence of RBD(+) memory B cells encoding neutralizing antibodies in SARS-CoV-2 infection. Cell Rep Med. 2021; 2(4):100228.
- 99Cohen KW, Linderman SL, Moodie Z, et al. Longitudinal analysis shows durable and broad immune memory after SARS-CoV-2 infection with persisting antibody responses and memory B and T cells. Cell Rep Med. 2021; 2(7):100354.
- 100Iyer AS, Jones FK, Nodoushani A, et al. Persistence and decay of human antibody responses to the receptor binding domain of SARS-CoV-2 spike protein in COVID-19 patients. Sci Immunol. 2020; 5(52):eabe0367.
- 101Wang Z, Muecksch F, Schaefer-Babajew D, et al. Naturally enhanced neutralizing breadth against SARS-CoV-2 one year after infection. Nature. 2021; 595(7867): 426-431.
- 102Hartley GE, Edwards ESJ, Aui PM, et al. Rapid generation of durable B cell memory to SARS-CoV-2 spike and nucleocapsid proteins in COVID-19 and convalescence. Sci Immunol. 2020; 5(54):eabf8891.
- 103Adamo S, Michler J, Zurbuchen Y, et al. Signature of long-lived memory CD8(+) T cells in acute SARS-CoV-2 infection. Nature. 2022; 602(7895): 148-155.
- 104Tan AT, Lim JM, Le Bert N, et al. Rapid measurement of SARS-CoV-2 spike T cells in whole blood from vaccinated and naturally infected individuals. J Clin Invest. 2021; 131(17):e152379.
- 105Farber DL. Tissues, not blood, are where immune cells function. Nature. 2021; 593(7860): 506-509.
- 106Poon MML, Farber DL. The whole body as the system in systems immunology. iScience. 2020; 23(9):101509.
- 107Weisberg SP, Ural BB, Farber DL. Tissue-specific immunity for a changing world. Cell. 2021; 184(6): 1517-1529.
- 108Carpenter DJ, Granot T, Matsuoka N, et al. Human immunology studies using organ donors: impact of clinical variations on immune parameters in tissues and circulation. Am J Transplant. 2018; 18(1): 74-88.
- 109Dogra P, Rancan C, Ma W, et al. Tissue determinants of human NK cell development, function, and residence. Cell. 2020; 180(4): 749-763 e713.
- 110Granot T, Senda T, Carpenter DJ, et al. Dendritic cells display subset and tissue-specific maturation dynamics over human life. Immunity. 2017; 46(3): 504-515.
- 111Kumar BV, Connors TJ, Farber DL. Human T cell development, localization, and function throughout life. Immunity. 2018; 48(2): 202-213.
- 112Thome JJ, Yudanin N, Ohmura Y, et al. Spatial map of human T cell compartmentalization and maintenance over decades of life. Cell. 2014; 159(4): 814-828.
- 113Gordon CL, Miron M, Thome JJ, et al. Tissue reservoirs of antiviral T cell immunity in persistent human CMV infection. J Exp Med. 2017; 214(3): 651-667.
- 114Poon MML, Rybkina K, Kato Y, et al. SARS-CoV-2 infection generates tissue-localized immunological memory in humans. Sci Immunol. 2021; 6(65):eabl9105.
- 115Grau-Exposito J, Sanchez-Gaona N, Massana N, et al. Peripheral and lung resident memory T cell responses against SARS-CoV-2. Nat Commun. 2021; 12(1): 3010.
- 116Slifka MK, Antia R, Whitmire JK, Ahmed R. Humoral immunity due to long-lived plasma cells. Immunity. 1998; 8: 363-372.
- 117Turner JS, Kim W, Kalaidina E, et al. SARS-CoV-2 infection induces long-lived bone marrow plasma cells in humans. Nature. 2021; 595(7867): 421-425.
- 118Shaan Lakshmanappa Y, Elizaldi SR, Roh JW, et al. SARS-CoV-2 induces robust germinal center CD4 T follicular helper cell responses in rhesus macaques. Nat Commun. 2021; 12(1): 541.
- 119Israelow B, Mao T, Klein J, et al. Adaptive immune determinants of viral clearance and protection in mouse models of SARS-CoV-2. Sci Immunol. 2021; 6(64):eabl4509.
- 120Khoury DS, Cromer D, Reynaldi A, et al. Neutralizing antibody levels are highly predictive of immune protection from symptomatic SARS-CoV-2 infection. Nat Med. 2021; 27(7): 1205-1211.
- 121Earle KA, Ambrosino DM, Fiore-Gartland A, et al. Evidence for antibody as a protective correlate for COVID-19 vaccines. Vaccine. 2021; 39(32): 4423-4428.
- 122Deng W, Bao L, Liu J, et al. Primary exposure to SARS-CoV-2 protects against reinfection in rhesus macaques. Science. 2020; 369(6505): 818-823.
- 123Schmidt F, Muecksch F, Weisblum Y, et al. Plasma neutralization of the SARS-CoV-2 Omicron variant. N Engl J Med. 2022; 386(6): 599-601.
- 124Liu C, Ginn HM, Dejnirattisai W, et al. Reduced neutralization of SARS-CoV-2 B.1.617 by vaccine and convalescent serum. Cell. 2021; 184(16): 4220-4236 e4213.
- 125Zhou D, Dejnirattisai W, Supasa P, et al. Evidence of escape of SARS-CoV-2 variant B.1.351 from natural and vaccine-induced sera. Cell. 2021; 184(9): 2348-2361 e2346.
- 126Tarke A, Coelho CH, Zhang Z, et al. SARS-CoV-2 vaccination induces immunological T cell memory able to cross-recognize variants from Alpha to Omicron. Cell. 2022; 185(5): 847-859 e811.
- 127McMahan K, Yu J, Mercado NB, et al. Correlates of protection against SARS-CoV-2 in rhesus macaques. Nature. 2021; 590(7847): 630-634.
- 128Baden LR, El Sahly HM, Essink B, et al. Efficacy and safety of the mRNA-1273 SARS-CoV-2 vaccine. N Engl J Med. 2020; 384(5): 403-416.
- 129Polack FP, Thomas SJ, Kitchin N, et al. Safety and efficacy of the BNT162b2 mRNA Covid-19 vaccine. N Engl J Med. 2020; 383(27): 2603-2615.
- 130Evans JP, Zeng C, Carlin C, et al. Neutralizing antibody responses elicited by SARS-CoV-2 mRNA vaccination wane over time and are boosted by breakthrough infection. Sci Transl Med. 2022; 14(637):eabn8057.
- 131Goel RR, Painter MM, Apostolidis SA, et al. mRNA vaccines induce durable immune memory to SARS-CoV-2 and variants of concern. Science. 2021; 374(6572):eabm0829.
- 132Lederer K, Bettini E, Parvathaneni K, et al. Germinal center responses to SARS-CoV-2 mRNA vaccines in healthy and immunocompromised individuals. Cell. 2022; 185(6): 1008-1024 e1015.
- 133Turner JS, O'Halloran JA, Kalaidina E, et al. SARS-CoV-2 mRNA vaccines induce persistent human germinal centre responses. Nature. 2021; 596(7870): 109-113.
- 134Herati RS, Muselman A, Vella L, et al. Successive annual influenza vaccination induces a recurrent oligoclonotypic memory response in circulating T follicular helper cells. Sci Immunol. 2017; 2(8):eaag2152.
- 135Cromer D, Juno JA, Khoury D, et al. Prospects for durable immune control of SARS-CoV-2 and prevention of reinfection. Nat Rev Immunol. 2021; 21(6): 395-404.
- 136Tregoning JS, Flight KE, Higham SL, Wang Z, Pierce BF. Progress of the COVID-19 vaccine effort: viruses, vaccines and variants versus efficacy, effectiveness and escape. Nat Rev Immunol. 2021; 21(10): 626-636.
- 137Zens KD, Chen J-K, Farber DL. Vaccine-generated lung tissue-resident memory T cells provide heterosubtypic protection to influenza infection. J Clin Invest Insight. 2016; 1(10):e85832.
- 138Afkhami S, D'Agostino MR, Zhang A, et al. Respiratory mucosal delivery of next-generation COVID-19 vaccine provides robust protection against both ancestral and variant strains of SARS-CoV-2. Cell. 2022; 185(5): 896-915 e819.
- 139Darrah PA, Zeppa JJ, Maiello P, et al. Prevention of tuberculosis in macaques after intravenous BCG immunization. Nature. 2020; 577(7788): 95-102.
- 140van Doremalen N, Purushotham JN, Schulz JE, et al. Intranasal ChAdOx1 nCoV-19/AZD1222 vaccination reduces viral shedding after SARS-CoV-2 D614G challenge in preclinical models. Sci Transl Med. 2021; 13(607):eabh0755.
- 141Goel RR, Apostolidis SA, Painter MM, et al. Distinct antibody and memory B cell responses in SARS-CoV-2 naive and recovered individuals following mRNA vaccination. Sci Immunol. 2021; 6(58):eabi6950.
- 142Mazzoni A, Di Lauria N, Maggi L, et al. First-dose mRNA vaccination is sufficient to reactivate immunological memory to SARS-CoV-2 in subjects who have recovered from COVID-19. J Clin Invest. 2021; 131(12):e149150.
- 143Reynolds CJ, Pade C, Gibbons JM, et al. Prior SARS-CoV-2 infection rescues B and T cell responses to variants after first vaccine dose. Science. 2021; 372(6549): 1418-1423.
- 144Stamatatos L, Czartoski J, Wan YH, et al. mRNA vaccination boosts cross-variant neutralizing antibodies elicited by SARS-CoV-2 infection. Science. 2021; 372(6549): 1413-148.
- 145Servellita V, Syed AM, Morris MK, et al. Neutralizing immunity in vaccine breakthrough infections from the SARS-CoV-2 Omicron and Delta variants. Cell. 2022; 185(9): 1539-1548 e1535.
- 146Goldberg Y, Mandel M, Bar-On YM, et al. Protection and Waning of Natural and Hybrid Immunity to SARS-CoV-2. N Engl J Med. 2022; 386: 2201-2212.
- 147Medigeshi GR, Batra G, Murugesan DR, et al. Sub-optimal neutralisation of Omicron (B.1.1.529) variant by antibodies induced by vaccine alone or SARS-CoV-2 infection plus vaccine (hybrid immunity) post 6-months. EBioMedicine. 2022; 78:103938.
- 148Nordstrom P, Ballin M, Nordstrom A. Risk of SARS-CoV-2 reinfection and COVID-19 hospitalisation in individuals with natural and hybrid immunity: a retrospective, total population cohort study in Sweden. Lancet Infect Dis. 2022; 22(6): 781-790.
- 149Gao Y, Cai C, Grifoni A, et al. Ancestral SARS-CoV-2-specific T cells cross-recognize the Omicron variant. Nat Med. 2022; 28(3): 472-476.
- 150GeurtsvanKessel CH, Geers D, Schmitz KS, et al. Divergent SARS-CoV-2 Omicron-reactive T and B cell responses in COVID-19 vaccine recipients. Sci Immunol. 2022; 7(69):eabo2202.
- 151Keeton R, Tincho MB, Ngomti A, et al. T cell responses to SARS-CoV-2 spike cross-recognize Omicron. Nature. 2022; 603(7901): 488-492.
- 152Mudd PA, Minervina AA, Pogorelyy MV, et al. SARS-CoV-2 mRNA vaccination elicits a robust and persistent T follicular helper cell response in humans. Cell. 2022; 185(4): 603-613 e615.