Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2024 Feb 1.
Published in final edited form as: Curr Opin Immunol. 2022 Dec 22;80:102278. doi: 10.1016/j.coi.2022.102278

Lung Tissue-Resident Memory T cells: The Gatekeeper To Respiratory Viral (Re)-Infection

Jinyi Tang 1,2, Jie Sun 1,2,#
PMCID: PMC9911367  NIHMSID: NIHMS1860553  PMID: 36565508

Abstract

The discovery of lung tissue-resident memory T (TRM) cells and the elucidation of their function in antiviral immunity have inspired considerable efforts to leverage the power of TRM cells, in defense to the infections and reinfections by respiratory viruses. Here, we have reviewed lung TRM cell identification, molecular regulation and function after influenza and SARS-CoV-2 infections. Furthermore, we have discussed emerging data on TRM responses induced by systemic and mucosal vaccination strategies. We hope that our current outstanding of TRM cells in this review could provide insights towards the development of vaccines capable of inducing highly efficacious mucosal TRM responses for protection against respiratory viral infections.

Keywords: Tissue resident memory, Influenza, SARS-CoV-2, Nasal vaccine

Introduction

Respiratory viral infections are a leading cause of mortality, causing an estimated 2 million deaths per year[1]. Respiratory viruses can cause catastrophic viral pandemics evidenced by the 1918 H1N1 pandemic and the current COVID-19 pandemic, which have claimed more than 40 and 6 million lives respectively[2,3]. Long term protection against reinfection by the same or similar viruses is mediated by memory T and B cells, as well as antibodies produced by long-lived plasma cells. Pathogens like influenza virus and SARS-CoV-2 have the ability to change their surface antigens frequently, escaping antibody-mediated viral neutralization. In this case, memory T cells are believed to play a crucial role in providing effective protection against viral dissemination and the development of severe diseases[4].

The respiratory mucosal interface is the front line encountering pathogens that spread via airborne transmission. Following infection, a subset of memory T cells termed tissue-resident memory T (TRM) cells, are retained in the respiratory tract. TRM cells are phenotypically and functionally different from the circulating central memory T (TCM) and effector memory T (TEM) cells[5]. Numerous reports have already demonstrated that compared to the circulating TCM and TEM cells, TRM (both CD4+ and CD8+) play a pivotal, if not dominant, role in the protection against secondary viral infection in the respiratory tract[5]. As the local sword, TRM cells rapidly respond upon pathogen invasion, killing virus-infected cells directly and activating a series of downstream immune events to impede viral replication and dissemination. Despite the important roles of TRM cells in protection against respiratory infection and re-infection, induction of strong antigen-specific TRM cells is not usually exploited by most available or candidate vaccines. It is thus needed to make more efforts on the development of TRM–based mucosal vaccines against the infection and transmission of respiratory viruses, in particular influenza and SARS-CoV-2[6]. In the following sections, we will mainly focus on the regulation and protective functions of CD8+ and CD4+ TRM in the contexts of influenza and SARS-CoV-2 natural infections or vaccinations.

Lung TRM identification and regulation

More than a decade ago, pioneering work on vesicular stomatitis virus (VSV) and listeria monocytogenes revealed the presence and persistence of non-circulating resident memory T cells in non-lymphoid organs after the resolution of the primary infection[7]. Almost at the same time, the presence of antigen-specific memory T cells was identified in the lungs following Sendai virus, influenza and respiratory syncytial virus (RSV) infections in mice[8,9]. Since then, the development of lung TRM cells has been observed in the context of almost all major respiratory viral, bacterial, fungal and parasitic infections. Aside from flow cytometric staining of TRM surface markers, techniques such as intravascular labeling, parabiosis, and tissue transplantation have greatly facilitated TRM research in animal models[10]. The identification and study of human respiratory TRM cells are emerging but remain challenging due to technical difficulties and the availability of human tissues.

Upon respiratory infection, naïve T cells are generally primed in the lung draining mediastinal lymph node (mLN), where they encounter antigen carried by antigen presenting cells (APCs) migrated from the lungs. Primed effector T cells then egress from the mLN and enter the lungs to combat the infection. After viral clearance, TRM cells emerge and persist in the lungs following contraction of the majority of effector T cells. Compared to the circulating counterparts (TCM and TEM cells), TRM cells highly express the transcriptional factors Blimp1, Hobit, Runx3, Bhlhe40, Notch, while downregulate the expression of KLF2, T-bet, Eomes and TCF1[11]. As a result, surface markers that maintain tissue localization and retention are upregulated. Most notably, the expression of CD69, which antagonizes the function of tissue-exiting molecule S1PR1, is uniform between mouse and human lung TRM cells, rendering its expression as the most distinctive feature of TRM cells compared to circulating memory cells. A group of TRM cells also express integrins CD103 and/or CD49a, further supporting their retention by interaction with epithelial E-cadherin and collagen in extracellular matrix respectively. Respiratory TRM cells also express the chemokine receptor CXCR3, allowing them to enter the lungs and be trapped by the local chemotactic ligand CXCL9/10/11 gradient[11]. Immunohistochemistry studies revealed the location of CD8+ TRM cells, mostly outside but also neighboring the area of inducible bronchus-associated lymphoid tissue (iBALT) structures in the lung parenchyma[12]. In addition, CXCR6 expression by TRM cells further promotes their localization into the airway[13,14] (Figure 1).

Figure 1. Lung TRM cell generation, maintenance and function.

Figure 1.

After respiratory viral infection (such as influenza and SARS-CoV-2) or intranasal vaccination, dendritic cells carrying viral antigen migrate from lung to draining mediastinal lymph nodes (mLNs), where naive T cells are activated. Primed effector T cells then egress from the mLNs and migrate to the lung for viral clearance. During this process, TRM precursors highly express transcriptional factors Blimp1, Hobit, Runx3, Bhlhe40 and Notch, while downregulate KLF2, T-bet, Eomes and TCF1. As a result, CD69, CD103 (selectively) and CXCR3 are upregulated to assist TRM cells localization and retention in the lung. In addition, CXCR6 expression further promotes TRM cell localization into the airway. In this context, CD4+ TRM cells can differentiate into IFN-γ- producing CD4+ TRM1 and IL-21-producing CD4+ TRH cells, with the latter help IFN-γ-producing CD8+ TRM cell maintenance and local B cell responses. CD8+ TRM cells can also kill the infected cells.

Besides chemotactic and retention signals, lung TRM cell programming and/or maintenance are also modulated by a variety of local tissue signals. For instance, cytokines such as IL-33, TGF-β, type I interferons and TNF are important to program lung TRM cell development, while IL-7 and IL-15 have been shown to be important for the maintenance of lung TRM cells[11]. Of note, it is also required for TRM cell development to re-encounter antigen locally after effector T cell migration into the lungs[15,16]. Additionally, the maintenance of a subset of lung PD-1+ TRM cells has been shown to be dependent on the persistent antigen stimulation in the memory phase long after the clearance of infectious virus[17]. Interestingly, the oxygen-rich and nutrient-deprived local environment in the respiratory tract imposes high levels of cellular stress in TRM cells[18]. Lung TRM cells are relatively short-lived compared to TRM cells in other organs such as the skin[18,19]. The limited interstitial space, enhanced TRM cell death, removal by mucociliary elevator and/or transportation via retrograde migration are likely the reasons for the steadily diminishment of TRM cells in the respiratory tract compared to those in other organs [20]. As a result, CD8+ TRM in the lung can last for approximately 200 days in the mice after viral infection, while variable data shows over a year in human following transplantation[21,22]. Such a short lifespan of lung TRM cells poses unique challenges for the development of TRM-based vaccines for combating respiratory viral infection.

Lung CD8+ TRM function following influenza and SARS-CoV-2 infection

The roles of lung CD8+ TRM cells in protection against respiratory viral infections have most extensively been studied in the murine influenza infection model. It is now clear that CD8+ TRM cells are vital for protection against influenza re-infection, particularly in the context of heterotypic influenza viruses that escape pre-existing antibodies[23]. CD8+ TRM cells are able to produce large quantities of cytokines such as IFN-γ conferring this protective function. IFN-γ blockade significantly decreased the survival rate in mice[24]. Similarly, these virus-specific TRM cells were also observed in human lungs[25], exhibiting high proliferative capacity and poly-functionality after ex vivo stimulation. Further T cell receptor (TCR) repertoire analysis indicated that the diversity of TCRαβ was maintained within the pool of lung CD8+ TRM cells, compared to other memory T cell populations in the lung [26]. Importantly, these virus-specific TRM cells also showed cross-reactivity against different influenza strains as observed in mice[24,26,27].

Regarding SARS-CoV-2 infection, the unprecedented efforts during the COVID-19 pandemic have shed considerable insight into human CD8+ TRM responses following infection. Early single-cell RNA sequencing of cells in bronchoalveolar lavage (BAL) fluid from patients with acute COVID-19 have identified the presence of effector CD8+ T cells and potential CD8+ TRM precursors after infection[28]. Later, our group found that increased antigen-specific CD8+ TRM cells can be detected for at least 2–3 months in the respiratory tract after the recovery from severe COVID-19[29]. The presence of CD8+ TRM cells in the lung and potentially other organs after acute SARS-CoV-2 infection were subsequently observed in a number of studies[3034]. Strikingly, these CD8+ TRM cells in the lung produce IFN-γ after antigenic stimulation and can persist up to 10 months after initial infection[30], suggesting a potential role for TRM cell-mediated long-term protection against SARS-CoV-2 reinfection in COVID-19 convalescents. Emerging animal studies have shown that antigen-specific CD8+ T cells capable of producing effector cytokines are maintained in the lungs after SARS-CoV-2 viral clearance[35,36]. Notably, lung CD8+ T cells can be detected up to 120 days post infection with a mouse-adapted SARS-CoV-2 strain (MA10) in mice[37]. The protective function of CD8+ TRM cells in the context of COVID-19 remains to be firmly established. One study using the K18-hACE2 mouse model concluded that CD8+ TRM cells are dispensable for SARS-CoV-2 protection[38]. However, SARS-CoV-2 mortality in K18-hACE2 mice may not be dependent on the pulmonary pathology[39], which puts the utility of the model for studying TRM-mediated protection into the question. To this end, CD8+ TRM cells were shown to provide substantial protection from lethal infection by an attenuated mouse-adapted SARS-CoV strain (MA15) in mice[40].

In certain circumstances, CD8+ TRM cells are not always “the more, the better”[41,42]. In aged COVID-19 convalescents, the levels of CD8+ TRM cell number in the respiratory tract appear positively correlated with the post COVID-19 lung sequelae [29,34]. Such a phenomena is consistent with the case of influenza-infected aged mice[43]. Late depletion of CD8+ TRM cells after viral clearance decreased chronic lung pathology and improved lung function in influenza-infected aged mice[29]. Hence, it is important to promote CD8+ TRM cell-mediated protective function, while minimizing their pathogenic potential to avoid chronic lung pathology.

Lung CD4+ TRM function following infection

Unlike CD8+ TRM cells, CD4+ TRM cells are generally more diverse, serving divergent functions against a variety of infections. Similar to effector CD4+ T cells, CD4+ TRM cells can be divided into different subsets based on their cytokine production, including IFN-γ-producing TRM1, IL-4/5/13-producing TRM2, IL-17-producing TRM17 cells and IL-21-producing resident helper (TRH) cells[44,45]. Additionally, Foxp3-expressing regulatory T (TREG) cells can also gain lung residency and become resident tissue TREG cells[44,45]. In the context of respiratory viral infection, TRM1 and TRH cells are usually the dominant CD4+ TRM cell responses in the lung[16,46].

CD4+ TRM cells have been shown to provide protection via a variety of potential mechanisms in response to heterosubtypic influenza infection[4648]. For instance, CD4+ TRM1 derived IFN-γ promotes an antiviral state in the lung and directly contribute to influenza clearance[49,50]. TRH cells localizing within the iBALT can assist in CD8+ TRM cell maintenance, providing the optimal local T cell mediated protection[46]. Moreover, TRH cells also aid local humoral responses in the lung by augmenting the development of tissue resident memory B (BRM) cells and local antibody production[16,46].

Our current understanding of CD4+ TRM cell response following SARS-CoV-2 infection is relatively limited. Data from both human and mouse studies have demonstrated CD4+ T cells can persist in the respiratory tract for a period of time after infection, even up to 120 days in the lung post infection with MA10 in mice[2833,37,51]. Occasionally, pre-existing CD4+ TRM cells could be also detected from respiratory tract of unexposed individuals[52], although the function of these cells has not been determined. In this regard, previous studies on SARS-CoV infection showed that CD4+ TRM cells can confer protective immunity against re-infection via IFN-γ production[53]. It is thus reasonable to assume that CD4+ TRM cells may mediate similar function after SARS-CoV-2 infection. Whether SARS-CoV-2 infection promotes the development of TRH cells is still unknown. Notably, SARS-CoV-2 infection could induce IL-17A- and GM-CSF-producing CD4+ TRM17 cells in the BAL, which may potentially contribute to pulmonary hyperinflammation observed in severe COVID-19 individuals[51].

Lung CD4+ and CD8+ TRM interaction

Pioneering work two decades ago have established an essential role of CD4+ T cell help in establishing CD8+ memory T cells[54,55]. Similarly, it was demonstrated that early CD4+ T cell help is important for the formation and localization of CD8+ TRM cells in the lung following influenza infection[48]. In the absence of CD4+ T cell-derived IFN-γ, “unhelped” CD8+ TRM cells downregulate CD103 expression and mis-positioned away from the airway epithelium, failing to provide optimal protection against heterosubtypic viral challenge[48]. Recently, we found that late local CD4+ T cell help, mediated mainly by TRH cells, is critical for long-term maintenance of a subset of PD-1hi CD8+ TRM cells in the lung[46]. This help is mainly dependent on TRH cell-derived IL-21, and IL-21R blockade after viral clearance resulted in decreased PD-1hi CD8+ TRM response[46]. In addition, TRH ablation also impaired BRM responses in the lung and decreased antibody secreting cells (ASCs) following viral rechallenge[16,46]. Interestingly, TRH development also requires B cell presence[16]. Altogether, these data suggest an interactive resident lymphocyte network co-operates to promote optimal local immune memory development and/or maintenance after viral reinfection.

Induction of lung TRM response after vaccination

Given the power of lung TRM cells in conferring protection, vaccine strategies that can promote robust TRM responses are expected to provide superb protection against respiratory pathogens[6]. Unfortunately, the current intramuscular inactivated influenza vaccines fail to generate notable resident immunity in the lung[56]. Conceptually, intranasal live-attenuated influenza virus vaccines (LAIV, FluMist) are thought to be better in generating TRM and mucosal antibody responses, which would result in higher protective efficacy compared to inactivated vaccines in children[57]. In spite of this observation in children, the efficacy of LAIV in adults decreases, evidenced by the inefficient induction of immune responses. This is likely due to diminished LAIV infection in adults as a result of pre-existing anti-influenza immunity[58]. The drawback of LAIV might be potentially overcame by future mucosal protein- or DNA/mRNA-based nanocarriers vaccines, which can be adjuvanted specifically to enhance the immunogenicity in the respiratory tract. The small size of nanocarriers with positive charge may potentially enhance the adhesion of antigens to the mucosa, avoid the physical and biological elimination[59,60], and facilitate the transfection into or taking up by targeted cells including APCs. These strategies are expected to provide better mucosal protection via the induction of strong TRM responses.

Current approved COVID-19 mRNA vaccines were demonstrated to provide high protection against symptomatic infections, mediated by strong circulating neutralizing antibody (nAb), as well as memory T and B cell responses[61]. However, within the respiratory tract, these immune responses were found to be suboptimal compared to COVID-19 convalescents[62,63]. Consistently, recent studies demonstrated resident T cells and nAb in nasal mucosa could be only detected after SARS-CoV-2 breakthrough infection (infection in vaccinated individuals), but was extremely rare in vaccinees alone[6466], suggesting that intramuscular immunization alone might not prevent infection and transmission efficiently. Remarkably, low dose of intramuscular mRNA immunization in mice is also inefficient in generating resident T cell response in the respiratory tract[62,67,68]. However, high doses of intramuscular mRNA immunization may generate antigen-specific resident T cell in the lung[69,70], indicating that the dose of peripheral immunization may dictate TRM generation in the lung, but potential side effects with high dose mRNA vaccination should be considered.

Recently, two COVID-19 intranasal vaccines have been approved in China and India. Both vaccines use the adenoviral vector-based vaccine platform, and appear to be well-tolerated[71]. Intranasal adenoviral immunization promotes strong mucosal humoral and TRM responses in animal models[62,72]. Therefore, it is tempting to speculate that these two COVID-19 intranasal vaccines may provide more effective protection against SARS-CoV-2 variants than the intramuscular ones, although the real-world efficacy remain to be determined. More recently, results from one small-scale clinical trial of intranasal delivery of approved adenoviral vector-based vaccine ChAdOx1 nCoV-19 (AZD1222) have also shown acceptable tolerability, but exhibited relatively weak immunogenicity even as an intranasal booster [73]. Thus, different formulations or subtypes of adenoviral vectors may have differential immunogenicity in the respiratory mucosa. In addition to adenoviral vectors, many other types of mucosal vaccine formulations are ongoing in clinical development including protein subunits, attenuated viruses and other viral vector-based vaccines [6]. Nevertheless, a “hybrid” systemic prime and mucosal boost strategy is expected to induce both long-lived circulating memory T cells and robust mucosal immunity (which are likely shorter-lived than circulating immunity), ultimately providing highly efficacious and long-lasting immunity against future viral infections.

Conclusion

Since the discovery of lung TRM cells, great advances have been made in the last decade regarding their characteristics, molecular regulation and protective function. Emerging studies have also begun to elucidate mechanisms modulating TRM heterogeneity and longevity in the lung. It is the time for the field to take advantage of what we have learned and devote even greater efforts for rational design of future vaccination strategies capable of harnessing the power of TRM cells for viral protection. Meanwhile, we also should not ignore the other side of the coin that excessive TRM cell activation may cause lung immunopathology and tissue destruction. Therefore, it is critical to harness TRM protective function while minimizing their pathogenic potential during the development of mucosal vaccines aiming to promote robust respiratory TRM responses.

Highlights.

  • Tissue-resident memory T (TRM) cell development following influenza or SARS-CoV-2 infection.

  • Lung TRM cell characteristics, mechanisms of regulation and function.

  • Robust TRM cell response confers optimal protection against heterosubtypic viral infection.

  • Intranasal booster immunization is a promising strategy to augment mucosal immunity.

Acknowledgements

We thank Yue Wu and Harish Narasimhan for critical reading of this manuscript. This work was supported by the National Institutes of Health (AI147394, AI154598, AI112844 and AG069264 to J.S.).

Footnotes

Conflict of interest statement

J.S. is a consultant for the Teneofour company, which does not directly involve with this work.

Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.

References and recommended reading

Papers of particular interest, published within the period of review, have been highlighted as:

* of special interest

** of outstanding interest

  • 1.Collaborators GL: Estimates of the global, regional, and national morbidity, mortality, and aetiologies of lower respiratory tract infections in 195 countries: a systematic analysis for the Global Burden of Disease Study 2015. Lancet Infect Dis 2017, 17:1133–1161. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Taubenberger JK, Morens DM: 1918 Influenza: the mother of all pandemics. Emerg Infect Dis 2006, 12:15–22. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.WHO: WHO Coronavius (COVID-19) Dashboard. World Health Organization (WHO) 2022. (update to 11/15/2022). [Google Scholar]
  • 4.Grifoni A, Sette A: From Alpha to omicron: The response of T cells. Curr Res Immunol 2022, 3:146–150. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Rosato PC, Beura LK, Masopust D: Tissue resident memory T cells and viral immunity. Curr Opin Virol 2017, 22:44–50. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Topol EJ, Iwasaki A: Operation Nasal Vaccine-Lightning speed to counter COVID-19. Sci Immunol 2022, 7:eadd9947. [DOI] [PubMed] [Google Scholar]
  • 7.Masopust D, Vezys V, Marzo AL, Lefrancois L: Preferential localization of effector memory cells in nonlymphoid tissue. Science 2001, 291:2413–2417. [DOI] [PubMed] [Google Scholar]
  • 8.Hogan RJ, Usherwood EJ, Zhong W, Roberts AA, Dutton RW, Harmsen AG, Woodland DL: Activated antigen-specific CD8+ T cells persist in the lungs following recovery from respiratory virus infections. J Immunol 2001, 166:1813–1822. [DOI] [PubMed] [Google Scholar]
  • 9.Ostler T, Hussell T, Surh CD, Openshaw P, Ehl S: Long-term persistence and reactivation of T cell memory in the lung of mice infected with respiratory syncytial virus. Eur J Immunol 2001, 31:2574–2582. [DOI] [PubMed] [Google Scholar]
  • 10.Szabo PA, Miron M, Farber DL: Location, location, location: Tissue resident memory T cells in mice and humans. Sci Immunol 2019, 4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Zheng MZM, Wakim LM: Tissue resident memory T cells in the respiratory tract. Mucosal Immunol 2022, 15:379–388. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Yoshizawa A, Bi K, Keskin DB, Zhang G, Reinhold B, Reinherz EL: TCR-pMHC encounter differentially regulates transcriptomes of tissue-resident CD8 T cells. Eur J Immunol 2018, 48:128–150. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Takamura S, Kato S, Motozono C, Shimaoka T, Ueha S, Matsuo K, Miyauchi K, Masumoto T, Katsushima A, Nakayama T, et al. : Interstitial-resident memory CD8(+) T cells sustain frontline epithelial memory in the lung. J Exp Med 2019, 216:2736–2747. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Wein AN, McMaster SR, Takamura S, Dunbar PR, Cartwright EK, Hayward SL, McManus DT, Shimaoka T, Ueha S, Tsukui T, et al. : CXCR6 regulates localization of tissue-resident memory CD8 T cells to the airways. J Exp Med 2019, 216:2748–2762. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Uddback I, Cartwright EK, Scholler AS, Wein AN, Hayward SL, Lobby J, Takamura S, Thomsen AR, Kohlmeier JE, Christensen JP: Long-term maintenance of lung resident memory T cells is mediated by persistent antigen. Mucosal Immunol 2021, 14:92–99. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Swarnalekha N, Schreiner D, Litzler LC, Iftikhar S, Kirchmeier D, Kunzli M, Son YM, Sun J, Moreira EA, King CG: T resident helper cells promote humoral responses in the lung. Sci Immunol 2021, 6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Wang Z, Wang S, Goplen NP, Li C, Cheon IS, Dai Q, Huang S, Shan J, Ma C, Ye Z, et al. : PD-1(hi) CD8(+) resident memory T cells balance immunity and fibrotic sequelae. Sci Immunol 2019, 4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Hayward SL, Scharer CD, Cartwright EK, Takamura S, Li ZT, Boss JM, Kohlmeier JE: Environmental cues regulate epigenetic reprogramming of airway-resident memory CD8(+) T cells. Nat Immunol 2020, 21:309–320. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Morris SE, Farber DL, Yates AJ: Tissue-Resident Memory T Cells in Mice and Humans: Towards a Quantitative Ecology. J Immunol 2019, 203:2561–2569. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Stolley JM, Johnston TS, Soerens AG, Beura LK, Rosato PC, Joag V, Wijeyesinghe SP, Langlois RA, Osum KC, Mitchell JS, et al. : Retrograde migration supplies resident memory T cells to lung-draining LN after influenza infection. J Exp Med 2020, 217. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Slutter B, Van Braeckel-Budimir N, Abboud G, Varga SM, Salek-Ardakani S, Harty JT: Dynamics of influenza-induced lung-resident memory T cells underlie waning heterosubtypic immunity. Sci Immunol 2017, 2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Snyder ME, Finlayson MO, Connors TJ, Dogra P, Senda T, Bush E, Carpenter D, Marboe C, Benvenuto L, Shah L, et al. : Generation and persistence of human tissue-resident memory T cells in lung transplantation. Sci Immunol 2019, 4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Wu T, Hu Y, Lee YT, Bouchard KR, Benechet A, Khanna K, Cauley LS: Lung-resident memory CD8 T cells (TRM) are indispensable for optimal cross-protection against pulmonary virus infection. J Leukoc Biol 2014, 95:215–224. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.McMaster SR, Wilson JJ, Wang H, Kohlmeier JE: Airway-Resident Memory CD8 T Cells Provide Antigen-Specific Protection against Respiratory Virus Challenge through Rapid IFN-gamma Production. J Immunol 2015, 195:203–209. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Turner DL, Bickham KL, Thome JJ, Kim CY, D’Ovidio F, Wherry EJ, Farber DL: Lung niches for the generation and maintenance of tissue-resident memory T cells. Mucosal Immunol 2014, 7:501–510. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Pizzolla A, Nguyen TH, Sant S, Jaffar J, Loudovaris T, Mannering SI, Thomas PG, Westall GP, Kedzierska K, Wakim LM: Influenza-specific lung-resident memory T cells are proliferative and polyfunctional and maintain diverse TCR profiles. J Clin Invest 2018, 128:721–733. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Koutsakos M, Illing PT, Nguyen THO, Mifsud NA, Crawford JC, Rizzetto S, Eltahla AA, Clemens EB, Sant S, Chua BY, et al. : Human CD8(+) T cell cross-reactivity across influenza A, B and C viruses. Nat Immunol 2019, 20:613–625. [DOI] [PubMed] [Google Scholar]
  • 28.Liao M, Liu Y, Yuan J, Wen Y, Xu G, Zhao J, Cheng L, Li J, Wang X, Wang F, et al. : Single-cell landscape of bronchoalveolar immune cells in patients with COVID-19. Nat Med 2020, 26:842–844. [DOI] [PubMed] [Google Scholar]
  • 29.Cheon IS, Li C, Son YM, Goplen NP, Wu Y, Cassmann T, Wang Z, Wei X, Tang J, Li Y, et al. : Immune signatures underlying post-acute COVID-19 lung sequelae. Sci Immunol 2021, 6:eabk1741. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Grau-Exposito J, Sanchez-Gaona N, Massana N, Suppi M, Astorga-Gamaza A, Perea D, Rosado J, Falco A, Kirkegaard C, Torrella A, et al. : Peripheral and lung resident memory T cell responses against SARS-CoV-2. Nat Commun 2021, 12:3010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Roukens AHE, Pothast CR, Konig M, Huisman W, Dalebout T, Tak T, Azimi S, Kruize Y, Hagedoorn RS, Zlei 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, 23:23–32. [DOI] [PubMed] [Google Scholar]
  • 32.Poon MML, Rybkina K, Kato Y, Kubota M, Matsumoto R, Bloom NI, Zhang Z, Hastie KM, Grifoni A, Weiskopf D, et al. : SARS-CoV-2 infection generates tissue-localized immunological memory in humans. Sci Immunol 2021, 6:eabl9105. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Szabo PA, Dogra P, Gray JI, Wells SB, Connors TJ, Weisberg SP, Krupska I, Matsumoto R, Poon MML, Idzikowski E, et al. : Longitudinal profiling of respiratory and systemic immune responses reveals myeloid cell-driven lung inflammation in severe COVID-19. Immunity 2021, 54:797–814 e796. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Vijayakumar B, Boustani K, Ogger PP, Papadaki A, Tonkin J, Orton CM, Ghai P, Suveizdyte K, Hewitt RJ, Desai SR, et al. : Immuno-proteomic profiling reveals aberrant immune cell regulation in the airways of individuals with ongoing post-COVID-19 respiratory disease. Immunity 2022, 55:542–556 e545. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Wong LR, Zheng J, Wilhelmsen K, Li K, Ortiz ME, Schnicker NJ, Thurman A, Pezzulo AA, Szachowicz PJ, Li P, et al. : Eicosanoid signalling blockade protects middle-aged mice from severe COVID-19. Nature 2022, 605:146–151. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Mao T, Israelow B, Pena-Hernandez MA, Suberi A, Zhou L, Luyten S, Reschke M, Dong H, Homer RJ, Saltzman WM, et al. : Unadjuvanted intranasal spike vaccine elicits protective mucosal immunity against sarbecoviruses. Science 2022:eabo2523. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Dinnon KH 3rd, Leist SR, Okuda K, Dang H, Fritch EJ, Gully KL, De la Cruz G, Evangelista MD, Asakura T, Gilmore RC, et al. : SARS-CoV-2 infection produces chronic pulmonary epithelial and immune cell dysfunction with fibrosis in mice. Sci Transl Med 2022, 14:eabo5070. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Roberts LM, Jessop F, Wehrly TD, Bosio CM: Cutting Edge: Lung-Resident T Cells Elicited by SARS-CoV-2 Do Not Mediate Protection against Secondary Infection. J Immunol 2021, 207:2399–2404. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Seehusen F, Clark JJ, Sharma P, Bentley EG, Kirby A, Subramaniam K, Wunderlin-Giuliani S, Hughes GL, Patterson EI, Michael BD, et al. : Neuroinvasion and Neurotropism by SARS-CoV-2 Variants in the K18-hACE2 Mouse. Viruses 2022, 14. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Channappanavar R, Fett C, Zhao J, Meyerholz DK, Perlman S: Virus-specific memory CD8 T cells provide substantial protection from lethal severe acute respiratory syndrome coronavirus infection. J Virol 2014, 88:11034–11044. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Goplen NP, Cheon IS, Sun J: Age-Related Dynamics of Lung-Resident Memory CD8(+) T Cells in the Age of COVID-19. Front Immunol 2021, 12:636118. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Narasimhan H, Wu Y, Goplen NP, Sun J: Immune determinants of chronic sequelae after respiratory viral infection. Sci Immunol 2022, 7:eabm7996. [DOI] [PubMed] [Google Scholar]
  • 43.Goplen NP, Wu Y, Son YM, Li C, Wang Z, Cheon IS, Jiang L, Zhu B, Ayasoufi K, Chini EN, et al. : Tissue-resident CD8(+) T cells drive age-associated chronic lung sequelae after viral pneumonia. Sci Immunol 2020, 5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Pruner KB, Pepper M: Local memory CD4 T cell niches in respiratory viral infection. J Exp Med 2021, 218. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Son YM, Sun J: Co-Ordination of Mucosal B Cell and CD8 T Cell Memory by Tissue-Resident CD4 Helper T Cells. Cells 2021, 10. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Son YM, Cheon IS, Wu Y, Li C, Wang Z, Gao X, Chen Y, Takahashi Y, Fu YX, Dent AL, 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. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Teijaro 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, 187:5510–5514. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Laidlaw BJ, Zhang N, Marshall HD, Staron MM, Guan T, Hu Y, Cauley LS, Craft J, Kaech SM: CD4+ T cell help guides formation of CD103+ lung-resident memory CD8+ T cells during influenza viral infection. Immunity 2014, 41:633–645. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Brown DM, Lee S, Garcia-Hernandez Mde L, Swain SL: Multifunctional CD4 cells expressing gamma interferon and perforin mediate protection against lethal influenza virus infection. J Virol 2012, 86:6792–6803. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.McKinstry KK, Strutt TM, Kuang Y, Brown DM, Sell S, Dutton RW, Swain SL: Memory CD4+ T cells protect against influenza through multiple synergizing mechanisms. J Clin Invest 2012, 122:2847–2856. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Zhao Y, Kilian C, Turner JE, Bosurgi L, Roedl K, Bartsch P, Gnirck AC, Cortesi F, Schultheiss C, Hellmig M, 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. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Diniz MO, Mitsi E, Swadling L, Rylance J, Johnson M, Goldblatt D, Ferreira D, Maini MK: Airway-resident T cells from unexposed individuals cross-recognize SARS-CoV-2. Nat Immunol 2022, 23:1324–1329. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Zhao J, Zhao J, Mangalam AK, Channappanavar R, Fett C, Meyerholz DK, Agnihothram S, Baric RS, David CS, Perlman S: Airway Memory CD4(+) T Cells Mediate Protective Immunity against Emerging Respiratory Coronaviruses. Immunity 2016, 44:1379–1391. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Shedlock DJ, Shen H: Requirement for CD4 T cell help in generating functional CD8 T cell memory. Science 2003, 300:337–339. [DOI] [PubMed] [Google Scholar]
  • 55.Sun JC, Bevan MJ: Defective CD8 T cell memory following acute infection without CD4 T cell help. Science 2003, 300:339–342. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Cox RJ, Brokstad KA, Ogra P: Influenza virus: immunity and vaccination strategies. Comparison of the immune response to inactivated and live, attenuated influenza vaccines. Scand J Immunol 2004, 59:1–15. [DOI] [PubMed] [Google Scholar]
  • 57.Ambrose CS, Wu X, Knuf M, Wutzler P: The efficacy of intranasal live attenuated influenza vaccine in children 2 through 17 years of age: a meta-analysis of 8 randomized controlled studies. Vaccine 2012, 30:886–892. [DOI] [PubMed] [Google Scholar]
  • 58.DiazGranados CA, Denis M, Plotkin S: Seasonal influenza vaccine efficacy and its determinants in children and non-elderly adults: a systematic review with meta-analyses of controlled trials. Vaccine 2012, 31:49–57. [DOI] [PubMed] [Google Scholar]
  • 59.Gupta NK, Tomar P, Sharma V, Dixit VK: Development and characterization of chitosan coated poly-(varepsilon-caprolactone) nanoparticulate system for effective immunization against influenza. Vaccine 2011, 29:9026–9037. [DOI] [PubMed] [Google Scholar]
  • 60.Tang J, Cai L, Xu C, Sun S, Liu Y, Rosenecker J, Guan S: Nanotechnologies in Delivery of DNA and mRNA Vaccines to the Nasal and Pulmonary Mucosa. Nanomaterials (Basel) 2022, 12. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Vacchio MS, Ciucci T, Gao Y, Watanabe M, Balmaceno-Criss M, McGinty MT, Huang A, Xiao Q, McConkey C, Zhao Y, et al. : A Thpok-Directed Transcriptional Circuitry Promotes Bcl6 and Maf Expression to Orchestrate T Follicular Helper Differentiation. Immunity 2019. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Tang J, Zeng C, Cox TM, Li C, Son YM, Cheon IS, Wu Y, Behl S, Taylor JJ, Chakraborty R, et al. : Respiratory mucosal immunity against SARS-CoV-2 following mRNA vaccination. Sci Immunol 2022:eadd4853. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Pieren DKJ, Kuguel SG, Rosado J, Robles AG, Rey-Cano J, Mancebo C, Esperalba J, Falcó V, Buzón MJ, M G: Limited induction of lung-resident memory T cell responses against SARS-CoV-2 by mRNA vaccination. medRxiv 10.1101/2022.05.25.22275300. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.Lim JME, Tan AT, Le Bert N, Hang SK, Low JGH, Bertoletti A: SARS-CoV-2 breakthrough infection in vaccinees induces virus-specific nasal-resident CD8+ and CD4+ T cells of broad specificity. J Exp Med 2022, 219. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Delphine Planas IS, Françoise Porot, Florence Guivel-Benhassine, Lynda Handala, Mathieu Prot, William-Henry Bolland, Julien Puech, Hélène Péré, David Veyer, Aymeric Sève, Etienne-Simon-Lorière, Timothée Bruel, Thierry Prazuck, Karl Stefic, Laurent Hocqueloux, Olivier Schwartz: Duration of BA.5 neutralization in sera and nasal swabs from SARS-CoV-2 vaccinated individuals, with or without Omicron breakthrough infection. Med (N Y) 2022. doi: 10.1016/j.medj.2022.09.010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Ssemaganda A, Nguyen HM, Nuhu F, Jahan N, Card CM, Kiazyk S, Severini G, Keynan Y, Su RC, Ji H, et al. : Expansion of cytotoxic tissue-resident CD8(+) T cells and CCR6(+)CD161(+) CD4(+) T cells in the nasal mucosa following mRNA COVID-19 vaccination. Nat Commun 2022, 13:3357. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Israelow B, Mao T, Klein J, Song E, Menasche B, Omer SB, Iwasaki A: Adaptive immune determinants of viral clearance and protection in mouse models of SARS-CoV-2. Sci Immunol 2021, 6:eabl4509. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Lapuente D, Fuchs J, Willar J, Vieira Antao A, Eberlein V, Uhlig N, Issmail L, Schmidt A, Oltmanns F, Peter AS, et al. : Protective mucosal immunity against SARS-CoV-2 after heterologous systemic prime-mucosal boost immunization. Nat Commun 2021, 12:6871. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Laczko D, Hogan MJ, Toulmin SA, Hicks P, Lederer K, Gaudette BT, Castano D, Amanat F, Muramatsu H, Oguin TH, 3rd, et al. : A Single Immunization with Nucleoside-Modified mRNA Vaccines Elicits Strong Cellular and Humoral Immune Responses against SARS-CoV-2 in Mice. Immunity 2020, 53:724–732 e727. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Künzli M, O’Flanagan SD, LaRue M, Talukder P, Dileepan T, Soerens AG, Quarnstrom CF, Wijeyesinghe S, Ye Y, McPartlan J, et al. : Route of self-amplifying mRNA vaccination modulates the establishment of pulmonary resident memory CD8 and CD4 T cells. bioRxiv 10.1101/2022.06.02.494574. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Li JX, Wu SP, Guo XL, Tang R, Huang BY, Chen XQ, Chen Y, Hou LH, Liu JX, Zhong J, et al. : Safety and immunogenicity of heterologous boost immunisation with an orally administered aerosolised Ad5-nCoV after two-dose priming with an inactivated SARS-CoV-2 vaccine in Chinese adults: a randomised, open-label, single-centre trial. Lancet Respir Med 2022, 10:739–748. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Hassan AO, Kafai NM, Dmitriev IP, Fox JM, Smith BK, Harvey IB, Chen RE, Winkler ES, Wessel AW, Case JB, et al. : A Single-Dose Intranasal ChAd Vaccine Protects Upper and Lower Respiratory Tracts against SARS-CoV-2. Cell 2020, 183:169–184 e113. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73.Madhavan M, Ritchie AJ, Aboagye J, Jenkin D, Provstgaad-Morys S, Tarbet I, Woods D, Davies S, Baker M, Platt A, et al. : Tolerability and immunogenicity of an intranasally-administered adenovirus-vectored COVID-19 vaccine: An open-label partially-randomised ascending dose phase I trial. EBioMedicine 2022, 85:104298. [DOI] [PMC free article] [PubMed] [Google Scholar]

RESOURCES