Thymic Aging Associated with COVID-19: History
Please note this is an old version of this entry, which may differ significantly from the current revision.
Subjects: Cell Biology
Contributor:

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

This entry is adapted from the peer-reviewed paper 10.3390/cells10030628

This entry is offline, you can click here to edit this entry!
Video Production Service