scholarly journals Ancestral SARS-CoV-2-specific T cells cross-recognize Omicron (B.1.1.529)

Author(s):  
Yu Gao ◽  
Curtis Cai ◽  
Alba Grifoni ◽  
Thomas Müller ◽  
Julia Niessl ◽  
...  

Abstract The emergence of the SARS-CoV-2 variant-of-concern Omicron (B.1.1.529) has destabilized global efforts to control the impact of COVID-19. Recent data have suggested that B.1.1.529 can readily infect people with naturally acquired or vaccine-induced immunity, facilitated in some cases by viral escape from antibodies that neutralize ancestral SARS-CoV-2. However, severe disease appears to be relatively uncommon in such individuals, highlighting a potential role for other components of the adaptive immune system. We report here that SARS-CoV-2 spike-specific CD4+ and CD8+ T cells induced by prior infection and, more extensively, by mRNA vaccination provide comprehensive heterologous immune reactivity against B.1.1.529. Pairwise comparisons across groups further revealed that SARS-CoV-2 spike-reactive CD4+ and CD8+ T cells exhibited similar functional attributes, memory distributions, and phenotypic traits in response to the ancestral strain or B.1.1.529. Our data indicate that established SARS-CoV-2 spike-specific CD4+ and CD8+ T cell responses, especially after mRNA vaccination, remain largely intact against B.1.1.529.

2021 ◽  
pp. 1-19
Author(s):  
Sonia George ◽  
Trevor Tyson ◽  
Nolwen L. Rey ◽  
Rachael Sheridan ◽  
Wouter Peelaerts ◽  
...  

Background: α-Synuclein (α-syn) is the predominant protein in Lewy-body inclusions, which are pathological hallmarks of α- synucleinopathies, such as Parkinson’s disease (PD) and multiple system atrophy (MSA). Other hallmarks include activation of microglia, elevation of pro-inflammatory cytokines, as well as the activation of T and B cells. These immune changes point towards a dysregulation of both the innate and the adaptive immune system. T cells have been shown to recognize epitopes derived from α-syn and altered populations of T cells have been found in PD and MSA patients, providing evidence that these cells can be key to the pathogenesis of the disease. Objective To study the role of the adaptive immune system with respect to α-syn pathology. Methods: We injected human α-syn preformed fibrils (PFFs) into the striatum of immunocompromised mice (NSG) and assessed accumulation of phosphorylated α-syn pathology, proteinase K-resistant α-syn pathology and microgliosis in the striatum, substantia nigra and frontal cortex. We also assessed the impact of adoptive transfer of naïve T and B cells into PFF-injected immunocompromised mice. Results: Compared to wildtype mice, NSG mice had an 8-fold increase in phosphorylated α-syn pathology in the substantia nigra. Reconstituting the T cell population decreased the accumulation of phosphorylated α-syn pathology and resulted in persistent microgliosis in the striatum when compared to non-transplanted mice. Conclusion: Our work provides evidence that T cells play a role in the pathogenesis of experimental α-synucleinopathy.


2018 ◽  
Vol 92 (16) ◽  
Author(s):  
Pritesh Desai ◽  
Vikas Tahiliani ◽  
Georges Abboud ◽  
Jessica Stanfield ◽  
Shahram Salek-Ardakani

ABSTRACTRespiratory infection with vaccinia virus (VacV) elicits robust CD8+T cell responses that play an important role in host resistance. In the lung, VacV encounters multiple tissue-resident antigen-presenting cell (APC) populations, but which cell plays a dominant role in priming of virus-specific CD8+effector T cell responses remains poorly defined. We used Batf3−/−mice to investigate the impact of CD103+and CD8α+dendritic cell (DC) deficiency on anti-VacV CD8+T cell responses. We found that Batf3−/−mice were more susceptible to VacV infection, exhibiting profound weight loss, which correlated with impaired accumulation of gamma interferon (IFN-γ)-producing CD8+T cells in the lungs. This was largely due to defective priming since early in the response, antigen-specific CD8+T cells in the draining lymph nodes of Batf3−/−mice expressed significantly reduced levels of Ki67, CD25, and T-bet. These results underscore a specific role for Batf3-dependent DCs in regulating priming and expansion of effector CD8+T cells necessary for host resistance against acute respiratory VacV infection.IMPORTANCEDuring respiratory infection with vaccinia virus (VacV), a member ofPoxviridaefamily, CD8+T cells play important role in resolving the primary infection. Effector CD8+T cells clear the virus by accumulating in the infected lungs in large numbers and secreting molecules such as IFN-γ that kill virally infected cells. However, precise cell types that regulate the generation of effector CD8+T cells in the lungs are not well defined. Dendritic cells (DCs) are a heterogeneous population of immune cells that are recognized as key initiators and regulators of T-cell-mediated immunity. In this study, we reveal that a specific subset of DCs that are dependent on the transcription factor Batf3 for their development regulate the magnitude of CD8+T cell effector responses in the lungs, thereby providing protection during pulmonary VacV infection.


Cells ◽  
2021 ◽  
Vol 10 (9) ◽  
pp. 2365
Author(s):  
Christina E. Zielinski

T helper cell responses are tailored to their respective antigens and adapted to their specific tissue microenvironment. While a great proportion of T cells acquire a resident identity, a significant proportion of T cells continue circulating, thus encountering changing microenvironmental signals during immune surveillance. One signal, which has previously been largely overlooked, is sodium chloride. It has been proposed to have potent effects on T cell responses in the context of autoimmune, allergic and infectious tissue inflammation in mouse models and humans. Sodium chloride is stringently regulated in the blood by the kidneys but displays differential deposition patterns in peripheral tissues. Sodium chloride accumulation might furthermore be regulated by dietary intake and thus by intentional behavior. Together, these results make sodium chloride an interesting but still controversial signal for immune modulation. Its downstream cellular activities represent a potential therapeutic target given its effects on T cell cytokine production. In this review article, we provide an overview and critical evaluation of the impact of this ionic signal on T helper cell polarization and T helper cell effector functions. In addition, the impact of sodium chloride from the tissue microenvironment is assessed for human health and disease and for its therapeutic potential.


2018 ◽  
Vol 36 (5_suppl) ◽  
pp. 65-65 ◽  
Author(s):  
Robert J. Canter ◽  
Ethan Aguilar ◽  
Ziming Wang ◽  
Catherine Le ◽  
Lam Khuat ◽  
...  

65 Background: Obesity is increasingly prevalent and viewed as a critical co-factor in many pathologic conditions due to metabolic, inflammatory and immune perturbations. We performed a multi-species evaluation of the impact of obesity T cell effector functions and markers of immune exhaustion. Methods: We examined the impact of obesity on PD-1 and T cell-mediated responses across different pre-clinical models (tumor, infection, and autoimmune encephalomyelitis [EAE]) and species (mouse, dog, non-human primate, and human). Results: CD4 and CD8 T cells from obese mice, dogs, non-human primates and humans displayed increases in memory T cells and PD-1 expression, as well as impaired proliferative responses compared to lean controls, indicating a greater degree of T cell exhaustion at baseline. Following immunization with myelin oligodendrocyte glycoprotein, obese mice were resistant to induction of EAE, correlating with reduced antigen-specific CD4 T cells in the central nervous system. Administration of anti-PD-1 resulted in restoration of EAE and increased antigen-specific T cell numbers in obese mice. Tumors in obese mice exhibited accelerated growth compared to lean mice, and T cells displayed higher PD-1 expression correlating with RNAseq/molecular signatures of exhaustion compared to tumor-bearing lean mice. PD-1 blockade resulted in marked anti-tumor effects only in obese mice, and not lean. Impaired viral resistance to murine cytomegalovirus (MCMV) resulted was seen in obese mice, associated with increased PD-1/PD-L1 expression, which was reversible by PD-1/PD-L1 blockade. Conclusions: Obesity results in an increase in PD-1/PD-L1 expression and inhibition of T cell responses across species, and blockade not only reverses this inhibition but also leads to markedly augmented T cell effector responses compared to lean counterparts where no effects were observed. These results highlight how the immune system has evolved to control T cell responses using checkpoints contingent on dynamic host conditions and have translational relevance for predicting both efficacy and toxicity in clinical immuno-oncology.


2007 ◽  
Vol 204 (11) ◽  
pp. 2501-2504 ◽  
Author(s):  
Jonathan W. Yewdell ◽  
Heather D. Hickman

CD8+ T cells rapidly recognize virus-infected cells due to the generation of antigenic peptides from defective ribosomal products (DRiPs) that are encoded by standard open reading frames (ORFs). New data now show that alternative reading frame (ARF) DRiPs can also induce robust CD8+ T cell responses. ARF-specific T cells control retroviral replication and select for viral escape in monkeys, providing the most compelling evidence to date for the biological relevance of ARF immunosurveillance.


2021 ◽  
Vol 12 ◽  
Author(s):  
Gaëlle Tilly ◽  
Marion Cadoux ◽  
Alexandra Garcia ◽  
Jérémy Morille ◽  
Sandrine Wiertlewski ◽  
...  

Background and ObjectivesInhibition of de novo pyrimidine synthesis in proliferating T and B lymphocytes by teriflunomide, a pharmacological inhibitor of dihydroorotate dehydrogenase (DHODH), has been shown to be an effective therapy to treat patients with MS in placebo-controlled phase 3 trials. Nevertheless, the underlying mechanism contributing to the efficacy of DHODH inhibition has been only partially elucidated. Here, we aimed to determine the impact of teriflunomide on the immune compartment in a longitudinal high-dimensional follow-up of patients with relapse-remitting MS (RRMS) treated with teriflunomide.MethodsHigh-dimensional spectral flow cytometry was used to analyze the phenotype and the function of innate and adaptive immune system of patients with RRMS before and 12 months after teriflunomide treatment. In addition, we assessed the impact of teriflunomide on the migration of memory CD8 T cells in patients with RRMS, and we defined patient immune metabolic profiles.ResultsWe found that 12 months of treatment with teriflunomide in patients with RRMS does not affect the B cell or CD4 T cell compartments, including regulatory TREG follicular helper TFH cell and helper TH cell subsets. In contrast, we observed a specific impact of teriflunomide on the CD8 T cell compartment, which was characterized by decreased homeostatic proliferation and reduced production of TNFα and IFNγ. Furthermore, we showed that DHODH inhibition also had a negative impact on the migratory velocity of memory CD8 T cells in patients with RRMS. Finally, we showed that the susceptibility of memory CD8 T cells to DHODH inhibition was not related to impaired metabolism.DiscussionOverall, these findings demonstrate that the clinical efficacy of teriflunomide results partially in the specific susceptibility of memory CD8 T cells to DHODH inhibition in patients with RRMS and strengthens active roles for these T cells in the pathophysiological process of MS.


Blood ◽  
2013 ◽  
Vol 121 (3) ◽  
pp. 459-467 ◽  
Author(s):  
Jurjen Tel ◽  
Gerty Schreibelt ◽  
Simone P. Sittig ◽  
Till S. M. Mathan ◽  
Sonja I. Buschow ◽  
...  

Abstract In human peripheral blood, 4 populations of dendritic cells (DCs) can be distinguished, plasmacytoid dendritic cells (pDCs) and CD16+, CD1c+, and BDCA-3+ myeloid DCs (mDCs), each with distinct functional characteristics. DCs have the unique capacity to cross-present exogenously encountered antigens (Ags) to CD8+ T cells. Here we studied the ability of all 4 blood DC subsets to take up, process, and present tumor Ags to T cells. Although pDCs take up less Ags than CD1c+ and BDCA3+ mDCs, pDCs induce potent Ag-specific CD4+ and CD8+ T-cell responses. We show that pDCs can preserve Ags for prolonged periods of time and on stimulation show strong induction of both MHC class I and II, which explains their efficient activation of both CD4+ and CD8+ T cells. Furthermore, pDCs cross-present soluble and cell-associated tumor Ags to cytotoxic T lymphocytes equally well as BDCA3+ mDCs. These findings, and the fact that pDCs outnumber BDCA3+ mDCs, both in peripheral blood and lymph nodes, together with their potent IFN-I production, known to activate both components of the innate and adaptive immune system, put human pDCs forward as potent activators of CD8+ T cells in antitumor responses. Our findings may therefore have important consequences for the development of antitumor immunotherapy.


2020 ◽  
Vol 217 (7) ◽  
Author(s):  
Georgina Bowyer ◽  
Hannah Sharpe ◽  
Navin Venkatraman ◽  
Pierre Birahim Ndiaye ◽  
Djibril Wade ◽  
...  

CMV is associated with immunosenescence and reduced vaccine responses in the elderly (>70 yr). However, the impact of CMV in young adults is less clear. In this study, healthy UK and Senegalese adults aged 18–50 yr (average, 29 yr) were vaccinated with the Ebola vaccine candidate chimpanzee adenovirus type 3–vectored Ebola Zaire vaccine (ChAd3-EBO-Z) and boosted with modified vaccinia Ankara Ebola Zaire–vectored (MVA–EBO-Z) vaccine. CMV carriage was associated with an expansion of phenotypically senescent CD4+ and CD8+ T cells expressing CD57 and killer cell lectin-like receptor G1 (KLRG1), which was negatively associated with vaccine responses in both cohorts. Ebola-specific T cell responses induced by vaccination also contained significantly increased frequencies of terminally differentiated CD57+KLRG1+ cells in CMV seropositive (CMV+) individuals. This study suggests that CMV can also affect vaccine responses in younger adults and may have a particularly marked impact in many developing countries where CMV seroprevalence is almost universal.


2007 ◽  
Vol 81 (18) ◽  
pp. 10081-10091 ◽  
Author(s):  
Allison Imrie ◽  
Janet Meeks ◽  
Alexandra Gurary ◽  
Munkhzul Sukhbataar ◽  
Paul Kitsutani ◽  
...  

ABSTRACT Proinflammatory cytokines secreted by memory CD8+ and CD4+ T cells are thought to play a direct role in the pathogenesis of dengue virus infection by increasing vascular permeability and thereby inducing the pathophysiologic events associated with dengue hemorrhagic fever and dengue shock syndrome. Severe disease is frequently observed in the setting of secondary infection with heterologous dengue virus serotypes, suggesting a role for cross-reactive memory T cells in the immunopathogenesis of severe disease. We used a large panel of well-characterized dengue virus-specific CD8+ T-cell clones isolated from Pacific Islanders previously infected with dengue virus 1 to examine effector memory function, focusing on a novel dominant HLA-B*5502-restricted NS5329-337 epitope, and assessed T-cell responses to stimulation with variant peptides representing heterologous serotypes. Variant peptides were differentially recognized by dengue virus 1-specific effector CD8+ cytotoxic T lymphocytes (CTL) in a heterogeneous and clone-specific manner, in which cytolytic function and cytokine secretion could be enhanced, diminished, or abrogated compared with cognate peptide stimulation. Dengue virus-specific CTL stimulated with cognate and variant peptides demonstrated a cytokine response hierarchy of gamma IFN (IFN-γ) > tumor necrosis factor alpha (TNF-α) > interleukin-2 (IL-2), and a subset of clones also produced IL-4 and IL-6. Individual clones demonstrated greater avidity for variant peptides representing heterologous serotypes, including serotypes previously encountered by the subject, and IFN-γ and TNF-α secretion was enhanced by stimulation with these heterologous peptides. Altered antiviral T-cell responses in response to stimulation with heterologous dengue virus serotypes have implications for control of virus replication and for disease pathogenesis.


Blood ◽  
2017 ◽  
Vol 130 (Suppl_1) ◽  
pp. 77-77
Author(s):  
Jennifer A Ball ◽  
Andrew James Clear ◽  
Maria Calaminici ◽  
Andrew Stagg ◽  
James Lindsay ◽  
...  

Abstract Background: Gastro-intestinal (GI) graft-versus-host disease (GvHD) results in significant morbidity and mortality after allogeneic hematopoietic stem cell transplantation (AHSCT). Retinoic acid (RA), a metabolite of vitamin A, has diverse effects on immune cells. RA signaling promotes immune tolerance under steady state conditions. However in pro-inflammatory conditions RA increases Th1, Th17 CD4+ and Tc1 CD8+ T cell responses and is implicated in the pathogenesis of autoimmune inflammatory bowel disease. Furthermore, RA exposure and RA-receptor alpha (RARα) signaling in allogeneic T cells potentiates GI-GvHD in mice. Thus RA and cytokines that influence the effect of RA on T cells may represent potential therapeutic targets for GI-GvHD. However, the impact of RA at a tissue level in human GI-GvHD is unknown. We therefore investigated the role of RA-responsive T cells in human GvHD in vivo and human allogeneic T-cell responses in vitro . Methods: We scrutinized GI biopsies from 47 patients after AHSCT (36 with and 11 without GI-GvHD confirmed by conventional histological criteria). We also analysed skin biopsies from 11 AHSCT patients. We used conventional immunohistochemistry and a novel deep phenotyping method using sequential staining, stripping and re-probing on the same fixed embedded biopsy to determine cellular co-expression of multiple markers to identify RA-responsive T cells in human GvHD biopsies at sites of GvHD damage. Finally, we used an HLA-mismatched allogeneic mixed lymphocyte co-culture (MLRs) system, combining CFSE dye dilution and multi-parameter flow cytometry to enable the phenotyping of alloproliferative T cells after manipulation of RA signalling. Results: Firstly we observed thatthe number of cells expressing high levels of cytoplasmic RA binding proteins I/II (a surrogate marker of RA) was increased in gut biopsies in patients with GI-GvHD versus controls (median 155 vs 43 cells/mm2, p=0.056). Importantly the number of cells expressing RARα was also significantly increased (median 1315 vs 544 cells/mm2, p=0.001) consistent with RA-responsiveness. Crucially the number of RARα+ cells correlated with clinical stage of acute GI-GvHD with higher numbers in patients with severe (stage 3-4) versus less severe (stage 1-2) disease. Having demonstrated that RA-responsive cells were associated with both occurrence and severity of GI-GvHD we went on to identify potential RA-responsive T cell subsets. Additionally, as IL-23 and IL-33 direct RA-programming of T cells to either pro-inflammatory or tolerogenic phenotypes we also measured cellular expression of these cytokines and their receptors. CD8+ T cells co-localized with RARα+ cells and were present in significantly increased numbers in gut biopsies from patients with GI-GvHD, as were Tbet+ Th1/Tc1 cells. In contrast, numbers of RORγ+ Th17 cells were unchanged and CD4+ cells were decreased. IL-23p19+ and IL-23R+ cell numbers were significantly increased in GI-GvHD biopsies, whereas IL33R (ST2+)cells were unchanged. In order to determine if the RA-responsive, CD8+ and IL-23R+ cells represented a single effector subset, we used sequential stripping/re-probing to deep phenotype the T cells. We identified a unique single CD8+ T-cell population that co-expressed RARα+, T-bet and IL23R, present in significantly increased numbers in gut biopsies from patients with GI-GvHD versus controls. This is consistent with a RA-responsive, IL-23-dependent, inflammatory Tc1 effector cell mediating tissue damage in human gut GvHD. Importantly, there was no increase in this T cell subset in skin biopsies of skin GvHD patients demonstrating tissue-specificity. Finally we determined the impact of exogenous RA exposure on T cell alloresponses in MLRs. RA significantly increased the proportion of alloproliferative effector CD8+ T cells co-expressing Tbet and gut-homing molecules, confirming that RA can directly potentiate human alloreactive CD8 T cell responses with capacity to home to the GI tract. Conclusions: This is the first data to demonstrate a role for RA at a tissue level in human GI-GvHD. Furthermore we have identified a gut specific CD8+ T cell subset which co-expresses RARα, T-bet, and IL-23R localised in areas of GI-damage likely to represent the RA-responsive effector cell. Therapeutic blockade of IL-23R could target this cellular response to prevent or treat GI-GvHD. Disclosures Gribben: Acerta: Honoraria; Janssen: Honoraria; Kite: Honoraria; Celgene: Honoraria; TG Therapeutics: Honoraria; Karyopharm: Honoraria; Pharmacyclics: Honoraria; Genentech/Roche: Honoraria; Abbvie: Honoraria.


Sign in / Sign up

Export Citation Format

Share Document