Submitted Successfully!
To reward your contribution, here is a gift for you: A free trial for our video production service.
Thank you for your contribution! You can also upload a video entry or images related to this topic.
Version Summary Created by Modification Content Size Created at Operation
1 + 3056 word(s) 3056 2021-03-16 04:03:21 |
2 format correct Meta information modification 3056 2021-03-23 01:59:32 | |
3 format correct Meta information modification 3056 2021-03-23 10:21:55 |

Video Upload Options

Do you have a full video?

Confirm

Are you sure to Delete?
Cite
If you have any further questions, please contact Encyclopedia Editorial Office.
Wang, W. Thymic Aging Associated with COVID-19. Encyclopedia. Available online: https://encyclopedia.pub/entry/8173 (accessed on 01 July 2024).
Wang W. Thymic Aging Associated with COVID-19. Encyclopedia. Available at: https://encyclopedia.pub/entry/8173. Accessed July 01, 2024.
Wang, Weikan. "Thymic Aging Associated with COVID-19" Encyclopedia, https://encyclopedia.pub/entry/8173 (accessed July 01, 2024).
Wang, W. (2021, March 22). Thymic Aging Associated with COVID-19. In Encyclopedia. https://encyclopedia.pub/entry/8173
Wang, Weikan. "Thymic Aging Associated with COVID-19." Encyclopedia. Web. 22 March, 2021.
Thymic Aging Associated with COVID-19
Edit

Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) caused the global pandemic of coronavirus disease 2019 (COVID-19) and particularly exhibits severe symptoms and mortality in elderly individuals. Mounting evidence shows that the characteristics of the age-related clinical severity of COVID-19 are attributed to insufficient antiviral immune function and excessive self-damaging immune reaction, involving T cell immunity and associated with pre-existing basal inflammation in the elderly. Age-related changes to T cell immunosenescence is characterized by not only restricted T cell receptor (TCR) repertoire diversity, accumulation of exhausted and/or senescent memory T cells, but also by increased self-reactive T cell- and innate immune cell-induced chronic inflammation, and accumulated and functionally enhanced polyclonal regulatory T (Treg) cells. Many of these changes can be traced back to age-related thymic involution/degeneration. How these changes contribute to differences in COVID-19 disease severity between young and aged patients is an urgent area of investigation.

aged COVID-19 patients aged thymus thymic involution role of T cells immunopathology

1. Introduction

Currently, the global pandemic of coronavirus disease 2019 (COVID-19), caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), poses a greater threat to elderly people than to children and young adults, as shown by a higher frequency of severe symptoms and mortality in elderly patients, while children and young adults usually present with mild disease [1][2]. Differences in clinical severity are likely associated with immune system age [3]. Both the innate and adaptive immune systems are involved in antiviral responses. Although the innate immune system responds early, adaptive antiviral immunity is specific and robust, lasting longer in combating viral infection and generating immune memory. Adaptive antiviral immunity primarily includes neutralization antibodies (Ab) [4] associated with B cells, and cellular (mostly T cell)-mediated anti-SARS-CoV-2 immunity [5][6][7][8]. Although specific Abs are important for an immunoprotective barrier by blocking free viral particles from entering host cells, T cells and NK (nature killer, containing both innate and adaptive immune features) cells are more powerful because they destroy virally infected cells, thereby terminating viral replication. Generally, T cell priming is a key factor for effective immunity and vaccination, since T cells act not only as killer cells, but also as helper cells. For example, CD8+ T cells with cytotoxic T lymphocyte (CTL) function conduct killing of virally infected cells. Mild COVID-19 patients exhibit more CD8+ CTL cells [7][8], while patients with severe disease have predominantly increased SARS-CoV-2-specific CD4+ T cells in their recovery-stage of the disease [7][8]. These differences imply that different T cell subsets have different roles in disease severity and outcome. CD4+ T helper cells support the B cell-mediated antibody-producing humoral response. Additionally, some act as regulatory cells either via cytokine secretion, such as CD4+ Th1 (T-helper 1) cells, which primarily produce interferon-γ (IFN-γ), tumor necrosis factor-α (TNF-α), etc., and Th2 cells, which primarily produce interleukin-(IL)-4, IL-10, etc., and Th17 cells (producing IL-17), or facilitate immunosuppression (via multiple mechanisms, including inhibitory cytokines), such as CD4+FoxP3+ regulatory T (Treg) cells. Th1-biased cellular immune responses typically direct the killing of the virus, while Th2-biased responses are usually associated with lung allergy in respiratory infections [9]. The roles of Treg cells reported during COVID-19 are thus far contradictory, either reportedly decreased [10][11] or relatively increased in COVID-19 patients with severe disease or/and lymphopenia [6][12][13]. The roles of Treg cells in COVID-19 patients should perhaps be assessed based on their physiological localization and disease stage. If increased Treg cells are in the lung during an inflammatory cytokine storm, this will probably be beneficial for the alleviation of the excessive immune response [14][15], but if increased Treg cells are present early in the disease, it could be detrimental to the establishment of effective antiviral immunity.

Age-related changes to the T cell immune system include three main characteristics: (1) immunosenescence: low immune response, due to restriction of the TCR repertoire diversity, coupled with an increased oligoclonal expansion of peripheral memory/senescent T cells; (2) established chronic inflammation in the elderly, termed inflammaging, which is partially due to increased self-reactive T cell-induced chronic self-tissue damage, in addition to pro-inflammatory somatic cellular senescence-associated secretory phenotype (SASP); (3) enhanced polyclonal Treg cell generation in the aged, atrophied thymus and Treg accumulation in the aged peripheral secondary lymphoid organs. Evidence shows that all these changes are mainly attributed to age-related thymic involution [16].

Immunosenescence and inflammaging are high risk factors for severe COVID-19 in the elderly [1][2][17][18]. As age-related thymic involution contributes to immunosenescence and inflammaging (Figure 1A, Table 1 third column) [16], thymic function should also be considered as a potential player in aged populations versus young [19][20], and may also impact vaccination efficiency in the elderly. One indication that thymic function participates in COVID-19 disease severity has been reported, in which thymosin alpha-1 (T α 1, a synthetic thymic peptide) reduced the mortality of patients with severe COVID-19 [21], and a clinical trial with T α 1 to treat COVID-19 infection in elderly patients was approved (https://clinicaltrials.gov/ct2/show/NCT04428008 (12 January 2021)). Therefore, rejuvenation of aged thymic function in combination with an improvement in the pre-existing aged peripheral T cell microenvironment and inflammaging could improve protective immunity and efficient vaccination against viruses, including SARS-CoV-2, in the elderly.

Figure 1. How the aged thymus is involved in viral infection and a proposed comprehensive rejuvenation strategy for enhanced antiviral immunity and vaccination efficiency. (A) Left panels show the T cell pathway from the thymus to the lung during respiratory viral infection, such as SARS-CoV-2, using arrows. Middle panels show how immunosenescence and inflammaging are detrimental to antiviral immunity. (B) Right panels (red boxes) are proposed rejuvenation checkpoints where the dotted red lines are inhibition or blockade and the solid red lines with arrows are promotion or enhancement.

Table 1. Contributions of aged thymus to viral infection and potential rejuvenation therapeutics.

  Normal Thymus Maintains Homeostasis and Immunity Age-related Thymic Changes Contribute to Viral Infection Potential Rejuvenation Strategies
Thymus 1. Sufficient naïve T cell generation with highly diverse TCR repertoire
2. Minimal self-reactive T cell generation
3. tTreg generation balanced with tTcon generation
1. Reduced functional naïve T cells
2. Increased self-reactive T cells
3. Enhanced tTreg generation in proportion to tTcon output
Thymic rejuvenation via:
1. Injecting reprogrammed FoxN1 over-expressing fibroblasts
2. Providing exogenous factors such as growth hormone, IL-7, etc.
Peripheral lymphoid tissues and circulating blood 1. T cells with normal TCR repertoire → a broad recognition of foreign antigens
2. Potent T cell immune response to foreign antigens and homeostatic clearance of senescent somatic cells
3. pTreg cells balanced with pTcon cells → maintenance of immune tolerance and antiviral immunity.
1. Immunosenescence:
Restricted TCR repertoire diversity → compromised viral antigen recognition
Accumulated exhausted T cells → compromised anti-viral immune response and senescent somatic cell clearance → inflammaging
Accumulated pTreg → suppress normal antiviral immune responses
2. Inflammaging:
Self-reactive T cell induced tissue damage → chronic basal inflammation → inhibition of T and B cell activation for antiviral responses
1. Enhance peripheral T cell function via:
a. TGF-β blockade to inhibit iTreg cells
b. PD-1 blockade
2. Reduce chronic inflammatory conditions via low-dose mTOR inhibitors, aspirin, etc.
Lung 1. Sufficient cellular and humoral antiviral immunity
2. Timely clearance of virus by appropriate pro-inflammatory responses
1. Reduced antiviral function by T cells and plasma cells
2. Inflammatory cytokine storm facilitated by inflammaging
3. Lung tissue fibrosis after inflammation
TGF-β blockade to reduce fibrosis

References

  1. Chen, J.; Kelley, W.J.; Goldstein, D.R. Role of Aging and the Immune Response to Respiratory Viral Infections: Potential Implications for COVID-19. J. Immunol. 2020, 205, 313–320.
  2. O’Driscoll, M.; Dos Santos, G.R.; Wang, L.; Cummings, D.A.T.; Azman, A.S.; Paireau, J.; Fontanet, A.; Cauchemez, S.; Salje, H. Age-specific mortality and immunity patterns of SARS-CoV-2. Nature 2020, 590.
  3. Akbar, A.N.; Gilroy, D.W. Aging immunity may exacerbate COVID-19. Science 2020, 369, 256–257.
  4. Ni, L.; Ye, F.; Cheng, M.L.; Feng, Y.; Deng, Y.Q.; Zhao, H.; Wei, P.; Ge, J.; Gou, M.; Li, X.; et al. Detection of SARS-CoV-2-Specific Humoral and Cellular Immunity in COVID-19 Convalescent Individuals. Immunity 2020, 52, 971–977e3.
  5. Cox, R.J.; Brokstad, K.A. Not just antibodies: B cells and T cells mediate immunity to COVID-19. Nat. Rev. Immunol. 2020, 20, 581–582.
  6. Chen, Z.; Wherry, E.J. T cell responses in patients with COVID-19. Nat. Rev. Immunol. 2020, 20, 529–536.
  7. Swadling, L.; Maini, M.K. T cells in COVID-19—United in diversity. Nat. Immunol. 2020, 21, 1307–1308.
  8. Peng, Y.; Mentzer, A.J.; Liu, G.; Yao, X.; Yin, Z.; Dong, D.; Dejnirattisai, W.; Rostron, T.; Supasa, P.; Liu, C.; et al. Broad and strong memory CD4(+) and CD8(+) T cells induced by SARS-CoV-2 in UK convalescent individuals following COVID-19. Nat. Immunol. 2020, 21, 1336–1345.
  9. Yu, J.; Tostanoski, L.H.; Peter, L.; Mercado, N.B.; McMahan, K.; Mahrokhian, S.H.; Nkolola, J.P.; Liu, P.; Li, Z.; Chandrashekar, A.; et al. DNA vaccine protection against SARS-CoV-2 in rhesus macaques. Science 2020.
  10. Sadeghi, A.; Tahmasebi, S.; Mahmood, A.; Kuznetsova, M.; Valizadeh, H.; Taghizadieh, A.; Nazemiyeh, M.; Aghebati-Maleki, L.; Jadidi-Niaragh, F.; Abbaspour-Aghdam, S.; et al. Th17 and Treg cells function in SARS-CoV2 patients compared with healthy controls. J. Cell. Physiol. 2020.
  11. Meckiff, B.J.; Ramirez-Suastegui, C.; Fajardo, V.; Chee, S.J.; Kusnadi, A.; Simon, H.; Eschweiler, S.; Grifoni, A.; Pelosi, E.; Weiskopf, D.; et al. Imbalance of Regulatory and Cytotoxic SARS-CoV-2-Reactive CD4(+) T Cells in COVID-19. Cell 2020, 183, 1340–1353.e16.
  12. Gutiérrez-Bautista, J.F.; Rodriguez-Nicolas, A.; Rosales-Castillo, A.; Jiménez, P.; Garrido, F.; Anderson, P.; Ruiz-Cabello, F.; López-Ruz, M. Ángel Negative Clinical Evolution in COVID-19 Patients Is Frequently Accompanied with an Increased Proportion of Undifferentiated Th Cells and a Strong Underrepresentation of the Th1 Subset. Front. Immunol. 2020, 11, 596553.
  13. Chen, X.; Huang, J.; Huang, Y.; Chen, J.; Jiang, X.; Shi, Y. Characteristics of immune cells and cytokines in patients with coronavirus disease 2019 in Guangzhou, China. Hum. Immunol. 2020, 81, 702–708.
  14. Stephen-Victor, E.; Das, M.; Karnam, A.; Pitard, B.; Gautier, J.-F.; Bayry, J. Potential of regulatory T-cell-based therapies in the management of severe COVID-19. Eur. Respir. J. 2020, 56, 2002182.
  15. Ciabattini, A.; Garagnani, P.; Santoro, F.; Rappuoli, R.; Franceschi, C.; Medaglini, D. Shelter from the cytokine storm: Pitfalls and prospects in the development of SARS-CoV-2 vaccines for an elderly population. Semin. Immunopathol. 2020, 42, 619–634.
  16. Thomas, R.; Wang, W.; Su, D.-M. Contributions of Age-Related Thymic Involution to Immunosenescence and Inflammaging. Immun. Ageing 2020, 17, 1–17.
  17. Pietrobon, A.J.; Teixeira, F.M.E.; Sato, M.N. I mmunosenescence and Inflammaging: Risk Factors of Severe COVID-19 in Older People. Front. Immunol. 2020, 11, 579220.
  18. Hazeldine, J.; Lord, J.M. Immunesenescence: A Predisposing Risk Factor for the Development of COVID-19? Front. Immunol. 2020, 11, 573662.
  19. Rehman, S.; Majeed, T.; Ansari, M.A.; Ali, U.; Sabit, H.; Al-Suhaimi, E.A. Current scenario of COVID-19 in pediatric age group and physiology of immune and thymus response. Saudi J. Biol. Sci. 2020, 27, 2567–2573.
  20. Lins, M.P.; Smaniotto, S. Potential impact of SARS-CoV-2 infection on the thymus. Can. J. Microbiol. 2020, 67, 1–6.
  21. Liu, Y.; Pang, Y.; Hu, Z.; Wu, M.; Wang, C.; Feng, Z.; Mao, C.; Tan, Y.; Liu, Y.; Chen, L.; et al. Thymosin Alpha 1 Reduces the Mortality of Severe Coronavirus Disease 2019 by Restoration of Lymphocytopenia and Reversion of Exhausted T Cells. Clin. Infect. Dis. 2020, 71, 2150–2157.
  22. Wölfel, R.; Corman, V.M.; Guggemos, W.; Seilmaier, M.; Zange, S.; Müller, M.A.; Niemeyer, D.; Jones, T.C.; Vollmar, P.; Rothe, C.; et al. Virological assessment of hospitalized patients with COVID-2019. Nat. Cell Biol. 2020, 581, 465–469.
  23. Cao, Q.; Chen, Y.C.; Chen, C.L.; Chiu, C.H. SARS-CoV-2 infection in children: Transmission dynamics and clinical characteristics. J. Formos. Med. Assoc. 2020, 119, 670–673.
  24. Lai, C.C.; Shih, T.P.; Ko, W.C.; Tang, H.J.; Hsueh, P.R. Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and coronavirus disease-2019 (COVID-19): The epidemic and the challenges. Int. J. Antimicrob. Agents 2020, 55, 105924.
  25. Zamai, L. The Yin and Yang of ACE/ACE2 Pathways: The Rationale for the Use of Renin-Angiotensin System Inhibitors in COVID-19 Patients. Cells 2020, 9, 1704.
  26. Goronzy, J.J.; Weyand, C.M. Mechanisms underlying T cell ageing. Nat. Rev. Immunol. 2019, 19, 573–583.
  27. Nikolich-Žugich, J. The twilight of immunity: Emerging concepts in aging of the immune system. Nat. Immunol. 2018, 19, 10–19.
  28. Akbar, A.N.; Henson, S.M.; Lanna, A. Senescence of T Lymphocytes: Implications for Enhancing Human Immunity. Trends Immunol. 2016, 37, 866–876.
  29. Palmer, S.; Albergante, L.; Blackburn, C.C.; Newman, T.J. Thymic involution and rising disease incidence with age. Proc. Natl. Acad. Sci. USA 2018, 115, 1883–1888.
  30. Sun, L.; Brown, R.; Chen, S.; Zhuge, Q.; Su, D.-M. Aging induced decline in T-lymphopoiesis is primarily dependent on status of progenitor niches in the bone marrow and thymus. Aging 2012, 4, 606–619.
  31. Karlsson, A.C.; Humbert, M.; Buggert, M. The known unknowns of T cell immunity to COVID-19. Sci. Immunol. 2020, 5, eabe8063.
  32. Tan, L.; Wang, Q.; Zhang, D.; Ding, J.; Huang, Q.; Tang, Y.Q.; Wang, Q.; Miao, H. Lymphopenia predicts disease severity of COVID-19: A descriptive and predictive study. Signal Transduct. Target Ther. 2020, 5, 33.
  33. Le Bert, N.; Tan, A.T.; Kunasegaran, K.; Tham, C.Y.L.; Hafezi, M.; Chia, A.; Chng, M.H.Y.; Lin, M.; Tan, N.; Linster, M.; et al. SARS-CoV-2-specific T cell immunity in cases of COVID-19 and SARS, and uninfected controls. Nature 2020, 584, 457–462.
  34. Grifoni, A.; Weiskopf, D.; Ramirez, S.I.; Mateus, J.; Dan, J.M.; Moderbacher, C.R.; Rawlings, S.A.; Sutherland, A.; Premkumar, L.; Jadi, R.S.; et al. Targets of T Cell Responses to SARS-CoV-2 Coronavirus in Humans with COVID-19 Disease and Unexposed Individuals. Cell 2020, 181, 1489–1501.e15.
  35. Weiskopf, D.; Schmitz, K.S.; Raadsen, M.P.; Grifoni, A.; Okba, N.M.A.; Endeman, H.; van den Akker, J.P.C.; Molenkamp, R.; Koopmans, M.P.G.; van Gorp, E.C.M.; 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.
  36. Braun, J.; Loyal, L.; Frentsch, M.; Wendisch, D.; Georg, G.; Kurth, F.; Hippenstiel, S.; Dingeldey, M.; Kruse, B.; Fauchere, F.; et al. SARS-CoV-2-reactive T cells in healthy donors and patients with COVID-19. Nature 2020, 587, 270–274.
  37. Li, X.; Geng, M.; Peng, Y.; Meng, L.; Lu, S. Molecular immune pathogenesis and diagnosis of COVID-19. J. Pharm. Anal. 2020, 19, 1–7.
  38. Radzikowska, U.; Ding, M.; Tan, G.; Zhakparov, D.; Peng, Y.; Wawrzyniak, P.; Wang, M.; Li, S.; Morita, H.; Altunbulakli, C.; et al. Distribution of ACE2, CD147, CD26, and other SARS-CoV-2 associated molecules in tissues and immune cells in health and in asthma, COPD, obesity, hypertension, and COVID-19 risk factors. Allergy 2020, 75, 2829–2845.
  39. Alon, R.; Sportiello, M.; Kozlovski, S.; Kumar, A.; Reilly, E.C.; Zarbock, A.; Garbi, N.; Topham, D.J. Leukocyte trafficking to the lungs and beyond: Lessons from influenza for COVID-19. Nat. Rev. Immunol. 2021, 21, 49–64.
  40. Mackall, C.L.; Fleisher, T.A.; Wexler, L.H.; Gress, R.E.; Brown, M.R.; Andrich, M.P.; Chen, C.C.; Feuerstein, I.M.; Horowitz, M.E.; Magrath, I.T.; et al. Age, Thymopoiesis, and CD4+ T-Lymphocyte Regeneration after Intensive Chemotherapy. N. Engl. J. Med. 1995, 332, 143–149.
  41. Mitchell, W.A.; Lang, P.O.; Aspinall, R. Tracing thymic output in older individuals. Clin. Exp. Immunol. 2010, 161, 497–503.
  42. Gautier, D.; Beq, S.; Cortesão, C.S.; Sousa, A.E.; Cheynier, R. Efficient Thymopoiesis Contributes to the Maintenance of Peripheral CD4 T Cells during Chronic Human Immunodeficiency Virus Type 2 Infection. J. Virol. 2007, 81, 12685–12688.
  43. Nikolich-Zugich, J. T cell aging: Naive but not young. J. Exp. Med. 2005, 201, 837–840.
  44. Yi, J.S.; Cox, M.A.; Zajac, A.J. T-cell exhaustion: Characteristics, causes and conversion. Immunology 2010, 129, 474–481.
  45. Gui, J.; Mustachio, L.M.; Su, N.-M.; Craig, R.W. Thymus Size and Age-related Thymic Involution: Early Programming, Sexual Dimorphism, Progenitors and Stroma. Aging Dis. 2012, 3, 280–290.
  46. Gui, J.; Zhu, X.; Dohkan, J.; Cheng, L.; Barnes, P.F.; Su, D.-M. The aged thymus shows normal recruitment of lymphohematopoietic progenitors but has defects in thymic epithelial cells. Int. Immunol. 2007, 19, 1201–1211.
  47. Rezzani, R.; Nardo, L.; Favero, G.; Peroni, M.; Rodella, L.F. Thymus and aging: Morphological, radiological, and functional overview. Age 2014, 36, 313–351.
  48. Pawelec, G. Age and immunity: What is “immunosenescence”? Exp. Gerontol. 2018, 105, 4–9.
  49. Coder, B.D.; Wang, H.; Ruan, L.; Su, D.-M. Thymic Involution Perturbs Negative Selection Leading to Autoreactive T Cells That Induce Chronic Inflammation. J. Immunol. 2015, 194, 5825–5837.
  50. Freund, A.; Orjalo, A.V.; Desprez, P.-Y.; Campisi, J. Inflammatory networks during cellular senescence: Causes and consequences. Trends Mol. Med. 2010, 16, 238–246.
  51. De Martinis, M.; Franceschi, C.; Monti, D.; Ginaldi, L. Inflamm-ageing and lifelong antigenic load as major determinants of ageing rate and longevity. FEBS Lett. 2005, 579, 2035–2039.
  52. Franceschi, C.; Zaikin, A.; Gordleeva, S.; Ivanchenko, M.; Bonifazi, F.; Storci, G.; Bonafè, M. Inflammaging 2018: An update and a model. Semin. Immunol. 2018, 40, 1–5.
  53. Fulop, T.; Larbi, A.; Dupuis, G.; Le Page, A.; Frost, E.H.; Cohen, A.A.; Witkowski, J.M.; Franceschi, C. Immunosenescence and Inflamm-Aging as Two Sides of the Same Coin: Friends or Foes? Front. Immunol. 2018, 8, 1960.
  54. Oh, J.; Wang, W.; Thomas, R.; Su, D.-M. Capacity of tTreg generation is not impaired in the atrophied thymus. PLoS Biol. 2017, 15, e2003352.
  55. Raynor, J.; Lages, C.S.; Shehata, H.; Hildeman, D.A.; Chougnet, C.A. Homeostasis and function of regulatory T cells in aging. Curr. Opin. Immunol. 2012, 24, 482–487.
  56. Tsukamoto, H.; Clise-Dwyer, K.; Huston, G.E.; Duso, D.K.; Buck, A.L.; Johnson, L.L.; Haynes, L.; Swain, S.L. Age-associated increase in lifespan of naive CD4 T cells contributes to T-cell homeostasis but facilitates development of functional defects. Proc. Natl. Acad. Sci. USA 2009, 106, 18333–18338.
  57. Tsukamoto, H.; Huston, G.E.; Dibble, J.; Duso, D.K.; Swain, S.L. Bim Dictates Naive CD4 T Cell Lifespan and the Development of Age-Associated Functional Defects. J. Immunol. 2010, 185, 4535–4544.
  58. Chougnet, C.A.; Tripathi, P.; Lages, C.S.; Raynor, J.; Sholl, A.; Fink, P.; Plas, D.R.; Hildeman, D.A. A Major Role for Bim in Regulatory T Cell Homeostasis. J. Immunol. 2010, 186, 156–163.
  59. Linton, P.J.; Dorshkind, K. Age-related changes in lymphocyte development and function. Nat. Immunol. 2004, 5, 133–139.
  60. Vallejo, A.N. CD28 extinction in human T cells: Altered functions and the program of T-cell senescence. Immunol. Rev. 2005, 205, 158–169.
  61. Pangrazzi, L.; Weinberger, B. T cells, aging and senescence. Exp. Gerontol. 2020, 134, 110887.
  62. Sato, K.; Kato, A.; Sekai, M.; Hamazaki, Y.; Minato, N. Physiologic Thymic Involution Underlies Age-Dependent Accumulation of Senescence-Associated CD4+ T Cells. J. Immunol. 2017, 199, 138–148.
  63. Aiello, A.; Farzaneh, F.; Candore, G.; Caruso, C.; Davinelli, S.; Gambino, C.M.; Ligotti, M.E.; Zareian, N.; Accardi, G. Immunosenescence and Its Hallmarks: How to Oppose Aging Strategically? A Review of Potential Options for Therapeutic Intervention. Front. Immunol. 2019, 10, 2247.
  64. Liu, Y.; Sanoff, H.K.; Cho, H.; Burd, C.E.; Torrice, C.; Ibrahim, J.G.; Thomas, N.E.; Sharpless, N.E. Expression of p16(INK4a) in peripheral blood T-cells is a biomarker of human aging. Aging Cell 2009, 8, 439–448.
  65. Liu, Y.; Johnson, S.M.; Fedoriw, Y.; Rogers, A.B.; Yuan, H.; Krishnamurthy, J.; Sharpless, N.E. Expression of p16INK4a prevents cancer and promotes aging in lymphocytes. Blood 2011, 117, 3257–3267.
  66. Connoy, A.C.; Trader, M.; High, K.P. Age-related changes in cell surface and senescence markers in the spleen of DBA/2 mice: A flow cytometric analysis. Exp. Gerontol. 2006, 41, 225–229.
  67. Xia, J.; Wang, H.; Guo, J.; Zhang, Z.; Coder, B.; Su, N.-M. Age-Related Disruption of Steady-State Thymic Medulla Provokes Autoimmune Phenotype via Perturbing Negative Selection. Aging Dis. 2012, 3, 248–259.
  68. Coppé, J.-P.; Desprez, P.-Y.; Krtolica, A.; Campisi, J. The Senescence-Associated Secretory Phenotype: The Dark Side of Tumor Suppression. Annu. Rev. Pathol. Mech. Dis. 2010, 5, 99–118.
  69. Fulop, T.; Witkowski, J.M.; Olivieri, F.; Larbi, A. The integration of inflammaging in age-related diseases. Semin. Immunol. 2018, 40, 17–35.
  70. Wikby, A.; Nilsson, B.-O.; Forsey, R.; Thompson, J.; Strindhall, J.; Löfgren, S.; Ernerudh, J.; Pawelec, G.; Ferguson, F.; Johansson, B. The immune risk phenotype is associated with IL-6 in the terminal decline stage: Findings from the Swedish NONA immune longitudinal study of very late life functioning. Mech. Ageing Dev. 2006, 127, 695–704.
  71. Bryl, E.; Vallejo, A.N.; Weyand, C.M.; Goronzy, J.J. Down-Regulation of CD28 Expression by TNF-α. J. Immunol. 2001, 167, 3231–3238.
  72. Bryl, E.; Vallejo, A.N.; Matteson, E.L.; Witkowski, J.M.; Weyand, C.M.; Goronzy, J.J. Modulation of CD28 expression with anti-tumor necrosis factor α therapy in rheumatoid arthritis. Arthritis Rheum. 2005, 52, 2996–3003.
  73. Domingues, R.; Lippi, A.; Setz, C.; Outeiro, T.F.; Krisko, A. SARS-CoV-2, immunosenescence and inflammaging: Partners in the COVID-19 crime. Aging 2020, 12, 18778–18789.
  74. Cunha, L.L.; Perazzio, S.F.; Azzi, J.; Cravedi, P.; Riella, L.V. Remodeling of the Immune Response with Aging: Immunosenescence and Its Potential Impact on COVID-19 Immune Response. Front. Immunol. 2020, 11, 1748.
  75. Vellas, C.; Delobel, P.; Barreto, P.D.S.; Izopet, J. COVID-19, Virology and Geroscience: A Perspective. J. Nutr. Health Aging 2020, 24, 685–691.
  76. Kellogg, C.; Equils, O. The role of the thymus in COVID-19 disease severity: Implications for antibody treatment and immunization. Hum. Vaccines Immunother. 2020, 1–6.
  77. Schmidt, A.; Oberle, N.; Krammer, P.H. Molecular mechanisms of treg-mediated T cell suppression. Front. Immunol. 2012, 3, 51.
  78. Shevyrev, D.; Tereshchenko, V. Treg Heterogeneity, Function, and Homeostasis. Front. Immunol. 2020, 10, 3100.
  79. Garg, S.K.; Delaney, C.; Toubai, T.; Ghosh, A.; Reddy, P.; Banerjee, R.; Yung, R. Aging is associated with increased regulatory T-cell function. Aging Cell 2014, 13, 441–448.
  80. Lages, C.S.; Suffia, I.; Velilla, P.A.; Huang, B.; Warshaw, G.; Hildeman, D.A.; Belkaid, Y.; Chougnet, C. Functional Regulatory T Cells Accumulate in Aged Hosts and Promote Chronic Infectious Disease Reactivation. J. Immunol. 2008, 181, 1835–1848.
  81. Belkaid, Y. Regulatory T cells and infection: A dangerous necessity. Nat. Rev. Immunol. 2007, 7, 875–888.
  82. Overacre-Delgoffe, A.E.; Vignali, D.A. Treg Fragility: A Prerequisite for Effective Antitumor Immunity? Cancer Immunol. Res. 2018, 6, 882–887.
  83. Frimpong-Boateng, K.; Van Rooijen, N.; Geiben-Lynn, R. Regulatory T Cells Suppress Natural Killer Cells during Plasmid DNA Vaccination in Mice, Blunting the CD8+ T Cell Immune Response by the Cytokine TGFβ. PLoS ONE 2010, 5, e12281.
  84. Casares, N.; Rudilla, F.; Arribillaga, L.; Llopiz, D.; Riezu-Boj, J.-I.; Lozano, T.; López-Sagaseta, J.; Guembe, L.; Sarobe, P.; Prieto, J.; et al. A Peptide Inhibitor of FOXP3 Impairs Regulatory T Cell Activity and Improves Vaccine Efficacy in Mice. J. Immunol. 2010, 185, 5150–5159.
  85. Pierce, C.A.; Preston-Hurlburt, P.; Dai, Y.; Aschner, C.B.; Cheshenko, N.; Galen, B.; Garforth, S.J.; Herrera, N.G.; Jangra, R.K.; Morano, N.C.; et al. Immune responses to SARS-CoV-2 infection in hospitalized pediatric and adult patients. Sci. Transl. Med. 2020, 12.
  86. De Biasi, S.; Meschiari, M.; Gibellini, L.; Bellinazzi, C.; Borella, R.; Fidanza, L.; Gozzi, L.; Iannone, A.; Tartaro, D.L.; Mattioli, M.; et al. Marked T cell activation, senescence, exhaustion and skewing towards TH17 in patients with COVID-19 pneumonia. Nat. Commun. 2020, 11, 1–17.
More
Information
Subjects: Cell Biology
Contributor MDPI registered users' name will be linked to their SciProfiles pages. To register with us, please refer to https://encyclopedia.pub/register :
View Times: 460
Revisions: 3 times (View History)
Update Date: 28 Apr 2021
1000/1000
Video Production Service