Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 77
Filtrar
1.
Eur J Immunol ; 54(4): e2350660, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38304946

RESUMO

Thawing of viably frozen human tissue T cells, ILCs, and NK cells and subsequent single-cell RNA sequencing reveals that recovery of cellular subclusters is variably impacted. While freeze-thawing does not alter the transcriptional profiles of cells, it upregulates genes and gene pathways associated with stress and activation.


Assuntos
Células Matadoras Naturais , Linfócitos T , Humanos , Congelamento , Análise de Sequência de RNA
2.
Nat Commun ; 15(1): 1752, 2024 Feb 26.
Artigo em Inglês | MEDLINE | ID: mdl-38409190

RESUMO

Stromal cells support epithelial cell and immune cell homeostasis and play an important role in inflammatory bowel disease (IBD) pathogenesis. Here, we quantify the stromal response to inflammation in pediatric IBD and reveal subset-specific inflammatory responses across colon segments and intestinal layers. Using data from a murine dynamic gut injury model and human ex vivo transcriptomic, protein and spatial analyses, we report that PDGFRA+CD142-/low fibroblasts and monocytes/macrophages co-localize in the intestine. In primary human fibroblast-monocyte co-cultures, intestinal PDGFRA+CD142-/low fibroblasts foster monocyte transition to CCR2+CD206+ macrophages through granulocyte-macrophage colony-stimulating factor (GM-CSF). Monocyte-derived CCR2+CD206+ cells from co-cultures have a phenotype similar to intestinal CCR2+CD206+ macrophages from newly diagnosed pediatric IBD patients, with high levels of PD-L1 and low levels of GM-CSF receptor. The study describes subset-specific changes in stromal responses to inflammation and suggests that the intestinal stroma guides intestinal macrophage differentiation.


Assuntos
Doenças Inflamatórias Intestinais , Monócitos , Humanos , Animais , Camundongos , Criança , Monócitos/metabolismo , Fator Estimulador de Colônias de Granulócitos e Macrófagos/metabolismo , Macrófagos/metabolismo , Inflamação/metabolismo , Doenças Inflamatórias Intestinais/genética , Doenças Inflamatórias Intestinais/metabolismo , Diferenciação Celular
4.
Nat Commun ; 15(1): 683, 2024 Jan 24.
Artigo em Inglês | MEDLINE | ID: mdl-38267402

RESUMO

Immune cell dysfunction within the tumor microenvironment (TME) undermines the control of cancer progression. Established tumors contain phenotypically distinct, tumor-specific natural killer (NK) cells; however, the temporal dynamics, mechanistic underpinning and functional significance of the NK cell compartment remains incompletely understood. Here, we use photo-labeling, combined with longitudinal transcriptomic and cellular analyses, to interrogate the fate of intratumoral NK cells. We reveal that NK cells rapidly lose effector functions and adopt a distinct phenotypic state with features associated with tissue residency. NK cell depletion from established tumors did not alter tumor growth, indicating that intratumoral NK cells cease to actively contribute to anti-tumor responses. IL-15 administration prevented loss of function and improved tumor control, generating intratumoral NK cells with both tissue-residency characteristics and enhanced effector function. Collectively, our data reveals the fate of NK cells after recruitment into tumors and provides insight into how their function may be revived.


Assuntos
Internato e Residência , Neoplasias , Humanos , Perfilação da Expressão Gênica , Células Matadoras Naturais , Transcriptoma , Microambiente Tumoral
5.
Cell Rep ; 42(11): 113425, 2023 11 28.
Artigo em Inglês | MEDLINE | ID: mdl-37950867

RESUMO

Innate lymphoid cells (ILCs) are tissue-resident effector cells with roles in tissue homeostasis, protective immunity, and inflammatory disease. Group 3 ILCs (ILC3s) are classically defined by the master transcription factor RORγt. However, ILC3 can be further subdivided into subsets that share type 3 effector modules that exhibit significant ontological, transcriptional, phenotypic, and functional heterogeneity. Notably lymphoid tissue inducer (LTi)-like ILC3s mediate effector functions not typically associated with other RORγt-expressing lymphocytes, suggesting that additional transcription factors contribute to dictate ILC3 subset phenotypes. Here, we identify Bcl6 as a subset-defining transcription factor of LTi-like ILC3s in mice and humans. Deletion of Bcl6 results in dysregulation of the LTi-like ILC3 transcriptional program and markedly enhances expression of interleukin-17A (IL-17A) and IL-17F in LTi-like ILC3s in a manner in part dependent upon the commensal microbiota-and associated with worsened inflammation in a model of colitis. Together, these findings redefine our understanding of ILC3 subset biology.


Assuntos
Linfócitos , Membro 3 do Grupo F da Subfamília 1 de Receptores Nucleares , Animais , Humanos , Camundongos , Imunidade Inata , Linfócitos/metabolismo , Tecido Linfoide/metabolismo , Membro 3 do Grupo F da Subfamília 1 de Receptores Nucleares/metabolismo , Proteínas Proto-Oncogênicas c-bcl-6/metabolismo , Fatores de Transcrição/metabolismo
6.
Immunity ; 56(6): 1285-1302.e7, 2023 06 13.
Artigo em Inglês | MEDLINE | ID: mdl-37269830

RESUMO

The integrin CD49a marks highly cytotoxic epidermal-tissue-resident memory (TRM) cells, but their differentiation from circulating populations remains poorly defined. We demonstrate enrichment of RUNT family transcription-factor-binding motifs in human epidermal CD8+CD103+CD49a+ TRM cells, paralleled by high RUNX2 and RUNX3 protein expression. Sequencing of paired skin and blood samples revealed clonal overlap between epidermal CD8+CD103+CD49a+ TRM cells and circulating memory CD8+CD45RA-CD62L+ T cells. In vitro stimulation of circulating CD8+CD45RA-CD62L+ T cells with IL-15 and TGF-ß induced CD49a expression and cytotoxic transcriptional profiles in a RUNX2- and RUNX3-dependent manner. We therefore identified a reservoir of circulating cells with cytotoxic TRM potential. In melanoma patients, high RUNX2, but not RUNX3, transcription correlated with a cytotoxic CD8+CD103+CD49a+ TRM cell signature and improved patient survival. Together, our results indicate that combined RUNX2 and RUNX3 activity promotes the differentiation of cytotoxic CD8+CD103+CD49a+ TRM cells, providing immunosurveillance of infected and malignant cells.


Assuntos
Linfócitos T CD8-Positivos , Melanoma , Humanos , Linfócitos T CD8-Positivos/metabolismo , Integrina alfa1/metabolismo , Integrinas/metabolismo , Subunidade alfa 1 de Fator de Ligação ao Core/genética , Subunidade alfa 1 de Fator de Ligação ao Core/metabolismo , Memória Imunológica , Antígenos Comuns de Leucócito/metabolismo , Melanoma/metabolismo
7.
Cell Rep Med ; 4(5): 101038, 2023 05 16.
Artigo em Inglês | MEDLINE | ID: mdl-37160121

RESUMO

Innate lymphoid cells (ILCs) are considered innate counterparts of adaptive T cells; however, their common and unique transcriptional signatures in pediatric inflammatory bowel disease (pIBD) are largely unknown. Here, we report a dysregulated colonic ILC composition in pIBD colitis that correlates with inflammatory activity, including accumulation of naive-like CD45RA+CD62L- ILCs. Weighted gene co-expression network analysis (WGCNA) reveals modules of genes that are shared or unique across innate and adaptive lymphocytes. Shared modules include genes associated with activation/tissue residency, naivety/quiescence, and antigen presentation. Lastly, nearest-neighbor-based analysis facilitates the identification of "most inflamed" and "least inflamed" lymphocytes in pIBD colon with unique transcriptional signatures. Our study reveals shared and unique transcriptional signatures of colonic ILCs and T cells in pIBD. We also provide insight into the transcriptional regulation of colonic inflammation, deepening our understanding of the potential mechanisms involved in pIBD.


Assuntos
Colite , Doenças Inflamatórias Intestinais , Humanos , Criança , Linfócitos , Imunidade Inata/genética , Colite/genética , Linfócitos T
8.
Front Immunol ; 14: 1151754, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37063885

RESUMO

Mast cells are tissue-resident cells playing major roles in homeostasis and disease conditions. Lung mast cells are particularly important in airway inflammatory diseases such as asthma. Human mast cells are classically divided into the subsets MCT and MCTC, where MCT express the mast cell protease tryptase and MCTC in addition express chymase, carboxypeptidase A3 (CPA3) and cathepsin G. Apart from the disctintion of the MCT and MCTC subsets, little is known about the heterogeniety of human lung mast cells and a deep analysis of their heterogeniety has previously not been performed. We therefore performed single cell RNA sequencing on sorted human lung mast cells using SmartSeq2. The mast cells showed high expression of classical mast cell markers. The expression of several individual genes varied considerably among the cells, however, no subpopulations were detected by unbiased clustering. Variable genes included the protease-encoding transcripts CMA1 (chymase) and CTSG (cathepsin G). Human lung mast cells are predominantly of the MCT subset and consistent with this, the expression of CMA1 was only detectable in a small proportion of the cells, and correlated moderately to CTSG. However, in contrast to established data for the protein, CPA3 mRNA was high in all cells and the correlation of CPA3 to CMA1 was weak.


Assuntos
Mastócitos , Peptídeo Hidrolases , Humanos , Quimases/genética , Quimases/metabolismo , Mastócitos/metabolismo , Catepsina G , Peptídeo Hidrolases/metabolismo , Triptases/genética , Triptases/metabolismo , Pulmão/metabolismo , Análise de Sequência de RNA
10.
Clin Transl Allergy ; 13(3): e12238, 2023 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-36973952

RESUMO

BACKGROUND: Obese asthma is a complex phenotype and further characterization of the pathophysiology is needed. This study aimed to explore inflammation-related plasma biomarkers in lean and overweight/obese asthmatics. METHODS: We elucidated levels of inflammation-related plasma proteins in obese asthma phenotypes in the population-based cohort BAMSE (Swedish: Children, Allergy, Milieu, Stockholm, Epidemiology) using data from 2069 24-26-year-olds. Subjects were divided into lean asthma (n = 166), lean controls (n = 1440), overweight/obese asthma (n = 73) and overweight/obese controls (n = 390). Protein levels (n = 92) were analysed using the Olink Proseek Multiplex Inflammation panel. RESULTS: Of the 92 included proteins, 41 were associated with lean and/or overweight/obese asthma. The majority of proteins associated with overweight/obese asthma also associated with overweight/obesity among non-asthmatics. Beta-nerve growth factor (BetaNGF), interleukin 10 (IL-10), and matrix metalloproteinase 10 (MMP10) were associated only with lean asthma while C-C motif chemokine 20 (CCL20), fibroblast growth factor 19 (FGF19), interleukin 5 (IL-5), leukemia inhibitory factor (LIF), tumor necrosis factor ligand superfamily member 9 (TNFRSF9), and urokinase-type plasminogen activator (uPA) were associated only with overweight/obese asthma. Overweight/obesity modified the association between asthma and 3 of the proteins: fibroblast growth factor 21 (FGF21), interleukin 4 (IL-4), and urokinase-type plasminogen activator (uPA). In the overweight/obese group, interleukin-6 (IL-6) was associated with non-allergic asthma but not allergic asthma. CONCLUSION: These data indicate distinct plasma protein phenotypes in lean and overweight/obese asthmatics which, in turn, can impact upon therapeutic approaches.

11.
Respir Res ; 24(1): 62, 2023 Feb 24.
Artigo em Inglês | MEDLINE | ID: mdl-36829233

RESUMO

BACKGROUND: COVID-19 remains a major public health challenge, requiring the development of tools to improve diagnosis and inform therapeutic decisions. As dysregulated inflammation and coagulation responses have been implicated in the pathophysiology of COVID-19 and sepsis, we studied their plasma proteome profiles to delineate similarities from specific features. METHODS: We measured 276 plasma proteins involved in Inflammation, organ damage, immune response and coagulation in healthy controls, COVID-19 patients during acute and convalescence phase, and sepsis patients; the latter included (i) community-acquired pneumonia (CAP) caused by Influenza, (ii) bacterial CAP, (iii) non-pneumonia sepsis, and (iv) septic shock patients. RESULTS: We identified a core response to infection consisting of 42 proteins altered in both COVID-19 and sepsis, although higher levels of cytokine storm-associated proteins were evident in sepsis. Furthermore, microbiologic etiology and clinical endotypes were linked to unique signatures. Finally, through machine learning, we identified biomarkers, such as TRIM21, PTN and CASP8, that accurately differentiated COVID-19 from CAP-sepsis with higher accuracy than standard clinical markers. CONCLUSIONS: This study extends the understanding of host responses underlying sepsis and COVID-19, indicating varying disease mechanisms with unique signatures. These diagnostic and severity signatures are candidates for the development of personalized management of COVID-19 and sepsis.


Assuntos
COVID-19 , Infecções Comunitárias Adquiridas , Pneumonia , Sepse , Humanos , COVID-19/complicações , Proteômica , Inflamação/complicações , Biomarcadores
12.
Scand J Immunol ; : e13195, 2022 Jun 02.
Artigo em Inglês | MEDLINE | ID: mdl-35652743

RESUMO

The Karolinska KI/K COVID-19 Immune Atlas project was conceptualized in March 2020 as a part of the academic research response to the developing SARS-CoV-2 pandemic. The aim was to rapidly provide a curated dataset covering the acute immune response towards SARS-CoV-2 infection in humans, as it occurred during the first wave. The Immune Atlas was built as an open resource for broad research and educational purposes. It contains a presentation of the response evoked by different immune and inflammatory cells in defined naïve patient-groups as they presented with moderate and severe COVID-19 disease. The present Resource Article describes how the Karolinska KI/K COVID-19 Immune Atlas allow scientists, students, and other interested parties to freely explore the nature of the immune response towards human SARS-CoV-2 infection in an online setting.

13.
Sci Immunol ; 7(70): eabj8301, 2022 04 15.
Artigo em Inglês | MEDLINE | ID: mdl-35427178

RESUMO

Innate lymphoid cells (ILCs) are highly plastic and predominantly mucosal tissue-resident cells that contribute to both homeostasis and inflammation depending on the microenvironment. The discovery of naïve-like ILCs suggests an ILC differentiation process that is akin to naïve T cell differentiation. Delineating the mechanisms that underlie ILC differentiation in tissues is crucial for understanding ILC biology in health and disease. Here, we showed that tonsillar ILCs expressing CD45RA lacked proliferative activity, indicative of cellular quiescence. CD62L distinguished two subsets of CD45RA+ ILCs. CD45RA+CD62L+ ILCs (CD62L+ ILCs) resembled circulating naïve ILCs because they lacked the transcriptional, metabolic, epigenetic, and cytokine production signatures of differentiated ILCs. CD45RA+CD62L- ILCs (CD62L- ILCs) were epigenetically similar to CD62L+ ILCs but showed a transcriptional, metabolic, and cytokine production signature that was more akin to differentiated ILCs. CD62L+ and CD62L- ILCs contained uni- and multipotent precursors of ILC1s/NK cells and ILC3s. Differentiation of CD62L+ and CD62L- ILCs led to metabolic reprogramming including up-regulation of genes associated with glycolysis, which was needed for their effector functions after differentiation. CD62L- ILCs with preferential differentiation capacity toward IL-22-producing ILC3s accumulated in the inflamed mucosa of patients with inflammatory bowel disease. These data suggested distinct differentiation potential of CD62L+ and CD62L- ILCs between tissue microenvironments and identified that manipulation of these cells is a possible approach to restore tissue-immune homeostasis.


Assuntos
Imunidade Inata , Células Matadoras Naturais , Diferenciação Celular , Humanos , Inflamação , Ativação Linfocitária
14.
Scand J Immunol ; 95(4): e13156, 2022 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-35274359

RESUMO

Innate lymphoid cells (ILC) can be viewed as the innate counterparts of T cells. In contrast to T cells, ILCs exert their functions in antigen-independent manners, relying on tissue-derived signals from other immune cells, stroma and neurons. Natural killer (NK) cells have been known for their antitumour effects for decades. However, the roles of other ILC subtypes in cancer immunity are just now starting to be unravelled. ILCs contribute to both homeostasis and inflammation in the intestinal mucosa. Intestinal inflammation predisposes the intestine for the development of colonic dysplasia and colorectal cancer (CRC). Recent data from mouse models and human studies indicate that ILCs play a role in CRC, exerting both protumoural and antitumoural functions. Studies also suggest that intratumoural ILC frequencies and expression of ILC signature genes can predict disease progression and response to PD-1 checkpoint therapy in CRC. In this mini-review, we focus on such recent insights and their implications for understanding the immunobiology of CRC. We also identify knowledge gaps and research areas that require further work.


Assuntos
Neoplasias Colorretais , Imunidade Inata , Animais , Inflamação , Intestinos , Células Matadoras Naturais , Linfócitos , Camundongos
15.
Nat Rev Immunol ; 22(11): 701-712, 2022 11.
Artigo em Inglês | MEDLINE | ID: mdl-35354980

RESUMO

More than a decade ago, type 2 innate lymphoid cells (ILC2s) were discovered to be members of a family of innate immune cells consisting of five subsets that form a first line of defence against infections before the recruitment of adaptive immune cells. Initially, ILC2s were implicated in the early immune response to parasitic infections, but it is now clear that ILC2s are highly diverse and have crucial roles in the regulation of tissue homeostasis and repair. ILC2s can also regulate the functions of other type 2 immune cells, including T helper 2 cells, type 2 macrophages and eosinophils. Dysregulation of ILC2s contributes to type 2-mediated pathology in a wide variety of diseases, potentially making ILC2s attractive targets for therapeutic interventions. In this Review, we focus on the spectrum of ILC2 phenotypes that have been described across different tissues and disease states with an emphasis on human ILC2s. We discuss recent insights in ILC2 biology and suggest how this knowledge might be used for novel disease treatments and improved human health.


Assuntos
Imunidade Inata , Linfócitos , Humanos , Células Th2
16.
Eur J Immunol ; 52(3): 503-510, 2022 03.
Artigo em Inglês | MEDLINE | ID: mdl-34837225

RESUMO

Corona disease 2019 (COVID-19) affects multiple organ systems. Recent studies have indicated perturbations in the circulating metabolome linked to COVID-19 severity. However, several questions pertain with respect to the metabolome in COVID-19. We performed an in-depth assessment of 1129 unique metabolites in 27 hospitalized COVID-19 patients and integrated results with large-scale proteomic and immunology data to capture multiorgan system perturbations. More than half of the detected metabolic alterations in COVID-19 were driven by patient-specific confounding factors ranging from comorbidities to xenobiotic substances. Systematically adjusting for this, a COVID-19-specific metabolic imprint was defined which, over time, underwent a switch in response to severe acute respiratory syndrome coronavirus-2 seroconversion. Integration of the COVID-19 metabolome with clinical, cellular, molecular, and immunological severity scales further revealed a network of metabolic trajectories aligned with multiple pathways for immune activation, and organ damage including neurological inflammation and damage. Altogether, this resource refines our understanding of the multiorgan system perturbations in severe COVID-19 patients.


Assuntos
COVID-19/imunologia , COVID-19/metabolismo , Metaboloma/imunologia , SARS-CoV-2 , Adolescente , Adulto , Idoso , COVID-19/complicações , Estudos de Casos e Controles , Doenças do Sistema Nervoso Central/etiologia , Doenças do Sistema Nervoso Central/imunologia , Doenças do Sistema Nervoso Central/metabolismo , Estudos de Coortes , Feminino , Humanos , Masculino , Metabolômica , Pessoa de Meia-Idade , Especificidade de Órgãos , Pandemias , Fenótipo , Proteômica , Índice de Gravidade de Doença , Adulto Jovem
17.
Front Immunol ; 12: 674080, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34745084

RESUMO

Tonsil hyperplasia is the most common cause of pediatric obstructive sleep apnea (OSA). Despite the growing knowledge in tissue immunology of tonsils, the immunopathology driving tonsil hyperplasia and OSA remains unknown. Here we used multi-parametric flow cytometry to analyze the composition and phenotype of tonsillar innate lymphoid cells (ILCs), T cells, and B cells from pediatric patients with OSA, who had previous polysomnography. Unbiased clustering analysis was used to delineate and compare lymphocyte heterogeneity between two patient groups: children with small tonsils and moderate OSA (n = 6) or large tonsils and very severe OSA (n = 13). We detected disturbed ILC and B cell proportions in patients with large tonsils, characterized by an increase in the frequency of naïve CD27-CD21hi B cells and a relative reduction of ILCs. The enrichment of naïve B cells was not commensurate with elevated Ki67 expression, suggesting defective differentiation and/or migration rather than cellular proliferation to be the causative mechanism. Finally, yet importantly, we provide the flow cytometry data to be used as a resource for additional translational studies aimed at investigating the immunological mechanisms of pediatric tonsil hyperplasia and OSA.


Assuntos
Linfócitos/imunologia , Tonsila Palatina/imunologia , Tonsila Palatina/patologia , Apneia Obstrutiva do Sono/imunologia , Pré-Escolar , Feminino , Citometria de Fluxo , Humanos , Hiperplasia , Imunidade Inata , Masculino , Células B de Memória/imunologia , Receptores CXCR5/análise , Membro 7 da Superfamília de Receptores de Fatores de Necrose Tumoral/análise
18.
Proc Natl Acad Sci U S A ; 118(40)2021 10 05.
Artigo em Inglês | MEDLINE | ID: mdl-34548411

RESUMO

Since the outset of the COVID-19 pandemic, increasing evidence suggests that the innate immune responses play an important role in the disease development. A dysregulated inflammatory state has been proposed as a key driver of clinical complications in COVID-19, with a potential detrimental role of granulocytes. However, a comprehensive phenotypic description of circulating granulocytes in severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2)-infected patients is lacking. In this study, we used high-dimensional flow cytometry for granulocyte immunophenotyping in peripheral blood collected from COVID-19 patients during acute and convalescent phases. Severe COVID-19 was associated with increased levels of both mature and immature neutrophils, and decreased counts of eosinophils and basophils. Distinct immunotypes were evident in COVID-19 patients, with altered expression of several receptors involved in activation, adhesion, and migration of granulocytes (e.g., CD62L, CD11a/b, CD69, CD63, CXCR4). Paired sampling revealed recovery and phenotypic restoration of the granulocytic signature in the convalescent phase. The identified granulocyte immunotypes correlated with distinct sets of soluble inflammatory markers, supporting pathophysiologic relevance. Furthermore, clinical features, including multiorgan dysfunction and respiratory function, could be predicted using combined laboratory measurements and immunophenotyping. This study provides a comprehensive granulocyte characterization in COVID-19 and reveals specific immunotypes with potential predictive value for key clinical features associated with COVID-19.


Assuntos
COVID-19/imunologia , Granulócitos/imunologia , COVID-19/sangue , COVID-19/diagnóstico , COVID-19/fisiopatologia , Granulócitos/citologia , Humanos , Imunidade Inata , Imunofenotipagem , Contagem de Leucócitos , Pulmão/fisiopatologia , Modelos Biológicos , Escores de Disfunção Orgânica , SARS-CoV-2 , Índice de Gravidade de Doença
19.
Sci Immunol ; 6(64): eabk0894, 2021 Oct 22.
Artigo em Inglês | MEDLINE | ID: mdl-34519539

RESUMO

Cross-reactive CD4+ T cells that recognize severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) are more commonly detected in the peripheral blood of unexposed individuals compared with SARS-CoV-2­reactive CD8+ T cells. However, large numbers of memory CD8+ T cells reside in tissues, feasibly harboring localized SARS-CoV-2­specific immune responses. To test this idea, we performed a comprehensive functional and phenotypic analysis of virus-specific T cells in tonsils, a major lymphoid tissue site in the upper respiratory tract, and matched peripheral blood samples obtained from children and adults before the emergence of COVID-19 (coronavirus disease 2019). We found that SARS-CoV-2­specific memory CD4+ T cells could be found at similar frequencies in the tonsils and peripheral blood in unexposed individuals, whereas functional SARS-CoV-2­specific memory CD8+ T cells were almost only detectable in the tonsils. Tonsillar SARS-CoV-2­specific memory CD8+ T cells displayed a follicular homing and tissue-resident memory phenotype, similar to tonsillar Epstein-Barr virus­specific memory CD8+ T cells, but were functionally less potent than other virus-specific memory CD8+ T cell responses. The presence of preexisting tissue-resident memory CD8+ T cells in unexposed individuals could potentially enable rapid sentinel immune responses against SARS-CoV-2.


Assuntos
Tonsila Faríngea/imunologia , Linfócitos T CD8-Positivos/imunologia , SARS-CoV-2/imunologia , Tonsila Faríngea/citologia , Adulto , Idoso , Pré-Escolar , Feminino , Citometria de Fluxo , Humanos , Masculino , Pessoa de Meia-Idade
20.
Allergy ; 76(6): 1925-1928, 2021 06.
Artigo em Inglês | MEDLINE | ID: mdl-33751599
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...