Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 2.398
Filter
1.
Sci Rep ; 14(1): 10337, 2024 05 06.
Article in English | MEDLINE | ID: mdl-38710802

ABSTRACT

Infectious diseases have long been a shaping force in human history, necessitating a comprehensive understanding of their dynamics. This study introduces a co-evolution model that integrates both epidemiological and evolutionary dynamics. Utilizing a system of differential equations, the model represents the interactions among susceptible, infected, and recovered populations for both ancestral and evolved viral strains. Methodologically rigorous, the model's existence and uniqueness have been verified, and it accommodates both deterministic and stochastic cases. A myriad of graphical techniques have been employed to elucidate the model's dynamics. Beyond its theoretical contributions, this model serves as a critical instrument for public health strategy, particularly predicting future outbreaks in scenarios where viral mutations compromise existing interventions.


Subject(s)
Stochastic Processes , Humans , Immune System/virology , Evolution, Molecular , Viruses/genetics , Viruses/immunology , Biological Evolution
2.
Chaos ; 34(5)2024 May 01.
Article in English | MEDLINE | ID: mdl-38809906

ABSTRACT

A time-delayed virus dynamic model is proposed with general monotonic incidence, different nonlinear CTL (cytotoxic T lymphocyte) responses [CTL elimination function pyg1(z) and CTL stimulation function cyg2(z)], and immune impairment. Indeed, the different CTL responses pose challenges in obtaining the dissipativeness of the model. By constructing appropriate Lyapunov functionals with some detailed analysis techniques, the global stability results of all equilibria of the model are obtained. By the way, we point out that the partial derivative fv(x,0) is increasing (but not necessarily strictly) in x>0 for the general monotonic incidence f(x,v). However, some papers defaulted that the partial derivative was strictly increasing. Our main results show that if the basic reproduction number R0≤1, the infection-free equilibrium E0 is globally asymptotically stable (GAS); if CTL stimulation function cyg2(z)=0 for z=0 and the CTL threshold parameter R1≤1

Subject(s)
T-Lymphocytes, Cytotoxic , T-Lymphocytes, Cytotoxic/immunology , Humans , Time Factors , Viruses/immunology , Virus Diseases/immunology , Models, Immunological , Models, Biological
3.
Pathog Dis ; 822024 Feb 07.
Article in English | MEDLINE | ID: mdl-38714349

ABSTRACT

Respiratory pathogens can cause severe disease and even death, especially in the very young and very old. Studies investigating their prevalence often focus on individuals presenting to healthcare providers with symptoms. However, the design of prevention strategies, e.g. which target groups to vaccinate, will benefit from knowledge on the prevalence of, risk factors for and host response to these pathogens in the general population. In this study, upper respiratory samples (n = 1311) were collected cross-sectionally during winter from 11- and 24-month old children, their parents, and adults ≥60 years of age that were recruited irrespective of seeking medical care. Almost all children, approximately two-thirds of parents and a quarter of older adults tested positive for at least one pathogen, often in the absence of symptoms. Viral interference was evident for the combination of rhinovirus and respiratory syncytial virus. Attending childcare facilities and having siblings associated with increased pathogen counts in children. On average, children showed increased levels of mucosal cytokines compared to parents and especially proinflammatory molecules associated with the presence of symptoms. These findings may guide further research into transmission patterns of respiratory pathogens and assist in determining the most appropriate strategies for the prediction and prevention of disease.


Subject(s)
Cytokines , Respiratory Tract Infections , Seasons , Humans , Cross-Sectional Studies , Netherlands/epidemiology , Infant , Male , Female , Respiratory Tract Infections/epidemiology , Respiratory Tract Infections/virology , Respiratory Tract Infections/immunology , Prevalence , Middle Aged , Adult , Cytokines/metabolism , Aged , Child, Preschool , Aged, 80 and over , Virus Diseases/epidemiology , Virus Diseases/virology , Virus Diseases/immunology , Viruses/isolation & purification , Viruses/classification , Viruses/immunology
4.
Viruses ; 16(5)2024 05 06.
Article in English | MEDLINE | ID: mdl-38793616

ABSTRACT

Interferons (IFNs) are antiviral cytokines that defend against viral infections by inducing the expression of interferon-stimulated genes (ISGs). Interferon-inducible transmembrane proteins (IFITMs) 1, 2, and 3 are crucial ISG products and members of the CD225 protein family. Compelling evidence shows that IFITMs restrict the infection of many unrelated viruses by inhibiting the virus-cell membrane fusion at the virus entry step via the modulation of lipid composition and membrane properties. Meanwhile, viruses can evade IFITMs' restrictions by either directly interacting with IFITMs via viral glycoproteins or by altering the native entry pathway. At the same time, cumulative evidence suggests context-dependent and multifaceted roles of IFITMs in modulating virus infections and cell signaling. Here, we review the diverse antiviral mechanisms of IFITMs, the viral antagonizing strategies, and the regulation of IFITM activity in host cells. The mechanisms behind the antiviral activity of IFITMs could aid the development of broad-spectrum antivirals and enhance preparedness for future pandemics.


Subject(s)
Interferons , Membrane Proteins , Virus Internalization , Humans , Membrane Proteins/metabolism , Membrane Proteins/immunology , Interferons/immunology , Interferons/metabolism , Virus Internalization/drug effects , Antiviral Agents/pharmacology , Immune Evasion , Animals , Virus Diseases/immunology , Virus Diseases/virology , Viruses/immunology , Viruses/drug effects , Host-Pathogen Interactions/immunology , Signal Transduction , Antigens, Differentiation/metabolism , Antigens, Differentiation/immunology
5.
Viruses ; 16(4)2024 04 11.
Article in English | MEDLINE | ID: mdl-38675930

ABSTRACT

Inflammation is a protective host response essential for controlling viral replication and promoting tissue repair [...].


Subject(s)
Inflammation , Virus Diseases , Inflammation/virology , Humans , Virus Diseases/immunology , Virus Diseases/virology , Animals , Viruses/immunology , Viruses/pathogenicity , Virus Replication , Host-Pathogen Interactions/immunology
6.
Viruses ; 16(4)2024 04 13.
Article in English | MEDLINE | ID: mdl-38675942

ABSTRACT

The epitranscriptomic modification m6A is a prevalent RNA modification that plays a crucial role in the regulation of various aspects of RNA metabolism. It has been found to be involved in a wide range of physiological processes and disease states. Of particular interest is the role of m6A machinery and modifications in viral infections, serving as an evolutionary marker for distinguishing between self and non-self entities. In this review article, we present a comprehensive overview of the epitranscriptomic modification m6A and its implications for the interplay between viruses and their host, focusing on immune responses and viral replication. We outline future research directions that highlight the role of m6A in viral nucleic acid recognition, initiation of antiviral immune responses, and modulation of antiviral signaling pathways. Additionally, we discuss the potential of m6A as a prognostic biomarker and a target for therapeutic interventions in viral infections.


Subject(s)
Immunity, Innate , Virus Diseases , Humans , Virus Diseases/immunology , Virus Diseases/virology , Methylation , Virus Replication , Viruses/immunology , Viruses/genetics , Animals , RNA, Viral/genetics , RNA, Viral/immunology , Signal Transduction , Host-Pathogen Interactions/immunology
7.
J Med Virol ; 96(5): e29622, 2024 May.
Article in English | MEDLINE | ID: mdl-38682614

ABSTRACT

RNA capping is an essential trigger for protein translation in eukaryotic cells. Many viruses have evolved various strategies for initiating the translation of viral genes and generating progeny virions in infected cells via synthesizing cap structure or stealing the RNA cap from nascent host messenger ribonucleotide acid (mRNA). In addition to protein translation, a new understanding of the role of the RNA cap in antiviral innate immunity has advanced the field of mRNA synthesis in vitro and therapeutic applications. Recent studies on these viral RNA capping systems have revealed startlingly diverse ways and molecular machinery. A comprehensive understanding of how viruses accomplish the RNA capping in infected cells is pivotal for designing effective broad-spectrum antiviral therapies. Here we systematically review the contemporary insights into the RNA-capping mechanisms employed by viruses causing human and animal infectious diseases, while also highlighting its impact on host antiviral innate immune response. The therapeutic applications of targeting RNA capping against viral infections and the development of RNA-capping inhibitors are also summarized.


Subject(s)
Antiviral Agents , RNA Caps , RNA, Viral , Virus Diseases , Animals , Humans , Antiviral Agents/therapeutic use , Antiviral Agents/pharmacology , Immunity, Innate , RNA Caps/metabolism , RNA, Viral/genetics , Virus Diseases/drug therapy , Virus Diseases/immunology , Virus Replication/drug effects , Viruses/genetics , Viruses/drug effects , Viruses/immunology
8.
Nature ; 628(8006): 162-170, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38538791

ABSTRACT

Ageing of the immune system is characterized by decreased lymphopoiesis and adaptive immunity, and increased inflammation and myeloid pathologies1,2. Age-related changes in populations of self-renewing haematopoietic stem cells (HSCs) are thought to underlie these phenomena3. During youth, HSCs with balanced output of lymphoid and myeloid cells (bal-HSCs) predominate over HSCs with myeloid-biased output (my-HSCs), thereby promoting the lymphopoiesis required for initiating adaptive immune responses, while limiting the production of myeloid cells, which can be pro-inflammatory4. Ageing is associated with increased proportions of my-HSCs, resulting in decreased lymphopoiesis and increased myelopoiesis3,5,6. Transfer of bal-HSCs results in abundant lymphoid and myeloid cells, a stable phenotype that is retained after secondary transfer; my-HSCs also retain their patterns of production after secondary transfer5. The origin and potential interconversion of these two subsets is still unclear. If they are separate subsets postnatally, it might be possible to reverse the ageing phenotype by eliminating my-HSCs in aged mice. Here we demonstrate that antibody-mediated depletion of my-HSCs in aged mice restores characteristic features of a more youthful immune system, including increasing common lymphocyte progenitors, naive T cells and B cells, while decreasing age-related markers of immune decline. Depletion of my-HSCs in aged mice improves primary and secondary adaptive immune responses to viral infection. These findings may have relevance to the understanding and intervention of diseases exacerbated or caused by dominance of the haematopoietic system by my-HSCs.


Subject(s)
Adaptive Immunity , Aging , Cell Lineage , Hematopoietic Stem Cells , Lymphocytes , Myeloid Cells , Rejuvenation , Animals , Female , Male , Mice , Adaptive Immunity/immunology , Aging/immunology , B-Lymphocytes/cytology , B-Lymphocytes/immunology , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/immunology , Inflammation/immunology , Inflammation/pathology , Lymphocytes/cytology , Lymphocytes/immunology , Lymphopoiesis , Myeloid Cells/cytology , Myeloid Cells/immunology , Myelopoiesis , Phenotype , T-Lymphocytes/cytology , T-Lymphocytes/immunology , Viruses/immunology
9.
Nature ; 624(7992): 645-652, 2023 Dec.
Article in English | MEDLINE | ID: mdl-38093014

ABSTRACT

People with diabetes feature a life-risking susceptibility to respiratory viral infection, including influenza and SARS-CoV-2 (ref. 1), whose mechanism remains unknown. In acquired and genetic mouse models of diabetes, induced with an acute pulmonary viral infection, we demonstrate that hyperglycaemia leads to impaired costimulatory molecule expression, antigen transport and T cell priming in distinct lung dendritic cell (DC) subsets, driving a defective antiviral adaptive immune response, delayed viral clearance and enhanced mortality. Mechanistically, hyperglycaemia induces an altered metabolic DC circuitry characterized by increased glucose-to-acetyl-CoA shunting and downstream histone acetylation, leading to global chromatin alterations. These, in turn, drive impaired expression of key DC effectors including central antigen presentation-related genes. Either glucose-lowering treatment or pharmacological modulation of histone acetylation rescues DC function and antiviral immunity. Collectively, we highlight a hyperglycaemia-driven metabolic-immune axis orchestrating DC dysfunction during pulmonary viral infection and identify metabolic checkpoints that may be therapeutically exploited in mitigating exacerbated disease in infected diabetics.


Subject(s)
Dendritic Cells , Diabetes Complications , Diabetes Mellitus , Disease Susceptibility , Hyperglycemia , Lung , Virus Diseases , Animals , Mice , Acetyl Coenzyme A/metabolism , Acetylation , Chromatin/genetics , Chromatin/metabolism , Dendritic Cells/immunology , Dendritic Cells/metabolism , Dendritic Cells/pathology , Diabetes Complications/immunology , Diabetes Complications/metabolism , Diabetes Mellitus/genetics , Diabetes Mellitus/immunology , Diabetes Mellitus/metabolism , Glucose/metabolism , Histones/metabolism , Hyperglycemia/complications , Hyperglycemia/immunology , Hyperglycemia/metabolism , Lung/immunology , Lung/metabolism , Lung/virology , T-Lymphocytes/immunology , Virus Diseases/complications , Virus Diseases/immunology , Virus Diseases/mortality , Viruses/immunology , Disease Models, Animal , Humans
11.
Nature ; 622(7984): 818-825, 2023 Oct.
Article in English | MEDLINE | ID: mdl-37821700

ABSTRACT

Effective pandemic preparedness relies on anticipating viral mutations that are able to evade host immune responses to facilitate vaccine and therapeutic design. However, current strategies for viral evolution prediction are not available early in a pandemic-experimental approaches require host polyclonal antibodies to test against1-16, and existing computational methods draw heavily from current strain prevalence to make reliable predictions of variants of concern17-19. To address this, we developed EVEscape, a generalizable modular framework that combines fitness predictions from a deep learning model of historical sequences with biophysical and structural information. EVEscape quantifies the viral escape potential of mutations at scale and has the advantage of being applicable before surveillance sequencing, experimental scans or three-dimensional structures of antibody complexes are available. We demonstrate that EVEscape, trained on sequences available before 2020, is as accurate as high-throughput experimental scans at anticipating pandemic variation for SARS-CoV-2 and is generalizable to other viruses including influenza, HIV and understudied viruses with pandemic potential such as Lassa and Nipah. We provide continually revised escape scores for all current strains of SARS-CoV-2 and predict probable further mutations to forecast emerging strains as a tool for continuing vaccine development ( evescape.org ).


Subject(s)
Evolution, Molecular , Forecasting , Immune Evasion , Mutation , Pandemics , Viruses , Humans , Drug Design , HIV Infections , Immune Evasion/genetics , Immune Evasion/immunology , Influenza, Human , Lassa virus , Nipah Virus , SARS-CoV-2/genetics , SARS-CoV-2/immunology , Viral Vaccines/immunology , Viruses/genetics , Viruses/immunology
12.
Nature ; 621(7977): 179-187, 2023 Sep.
Article in English | MEDLINE | ID: mdl-37648857

ABSTRACT

Tissue resident memory CD8+ T (TRM) cells offer rapid and long-term protection at sites of reinfection1. Tumour-infiltrating lymphocytes with characteristics of TRM cells maintain enhanced effector functions, predict responses to immunotherapy and accompany better prognoses2,3. Thus, an improved understanding of the metabolic strategies that enable tissue residency by T cells could inform new approaches to empower immune responses in tissues and solid tumours. Here, to systematically define the basis for the metabolic reprogramming supporting TRM cell differentiation, survival and function, we leveraged in vivo functional genomics, untargeted metabolomics and transcriptomics of virus-specific memory CD8+ T cell populations. We found that memory CD8+ T cells deployed a range of adaptations to tissue residency, including reliance on non-steroidal products of the mevalonate-cholesterol pathway, such as coenzyme Q, driven by increased activity of the transcription factor SREBP2. This metabolic adaptation was most pronounced in the small intestine, where TRM cells interface with dietary cholesterol and maintain a heightened state of activation4, and was shared by functional tumour-infiltrating lymphocytes in diverse tumour types in mice and humans. Enforcing synthesis of coenzyme Q through deletion of Fdft1 or overexpression of PDSS2 promoted mitochondrial respiration, memory T cell formation following viral infection and enhanced antitumour immunity. In sum, through a systematic exploration of TRM cell metabolism, we reveal how these programs can be leveraged to fuel memory CD8+ T cell formation in the context of acute infections and enhance antitumour immunity.


Subject(s)
CD8-Positive T-Lymphocytes , Lymphocytes, Tumor-Infiltrating , Neoplasms , Animals , Humans , Mice , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Cell Respiration , Cholesterol/metabolism , Cholesterol/pharmacology , Immunologic Memory , Intestine, Small/drug effects , Intestine, Small/metabolism , Lymphocytes, Tumor-Infiltrating/immunology , Lymphocytes, Tumor-Infiltrating/metabolism , Metabolomics , Mevalonic Acid/metabolism , Neoplasms/immunology , Ubiquinone/metabolism , Virus Diseases/immunology , Viruses/immunology , Mitochondria/metabolism
14.
Front Cell Infect Microbiol ; 13: 1173505, 2023.
Article in English | MEDLINE | ID: mdl-37465759

ABSTRACT

The inflammasome is a multiprotein complex that further regulates cell pyroptosis and inflammation by activating caspase-1. The assembly and activation of inflammasome are associated with a variety of diseases. Accumulative studies have shown that inflammasome is a key modulator of the host's defense response to viral infection. Indeed, it has been established that activation of inflammasome occurs during viral infection. At the same time, the host has evolved a variety of corresponding mechanisms to inhibit unnecessary inflammasome activation. Therefore, here, we review and summarize the latest research progress on the interaction between inflammosomes and viruses, highlight the assembly and activation of inflammosome in related cells after viral infection, as well as the corresponding molecular regulatory mechanisms, and elucidate the effects of this activation on virus immune escape and host innate and adaptive immune defenses. Finally, we also discuss the potential therapeutic strategies to prevent and/or ameliorate viral infection-related diseases via targeting inflammasomes and its products.


Subject(s)
Host Microbial Interactions , Inflammasomes , Virus Diseases , Viruses , Humans , Inflammasomes/immunology , Virus Diseases/immunology , Virus Diseases/therapy , Viruses/immunology , Host Microbial Interactions/immunology , Animals
15.
Science ; 380(6644): 478-484, 2023 05 05.
Article in English | MEDLINE | ID: mdl-37141353

ABSTRACT

Although all multicellular organisms have germ line-encoded innate receptors to sense pathogen-associated molecular patterns, vertebrates also evolved adaptive immunity based on somatically generated antigen receptors on B and T cells. Because randomly generated antigen receptors may also react with self-antigens, tolerance checkpoints operate to limit but not completely prevent autoimmunity. These two systems are intricately linked, with innate immunity playing an instrumental role in the induction of adaptive antiviral immunity. In this work, we review how inborn errors of innate immunity can instigate B cell autoimmunity. Increased nucleic acid sensing, often resulting from defects in metabolizing pathways or retroelement control, can break B cell tolerance and converge into TLR7-, cGAS-STING-, or MAVS-dominant signaling pathways. The resulting syndromes span a spectrum that ranges from chilblain and systemic lupus to severe interferonopathies.


Subject(s)
Autoimmunity , B-Lymphocytes , Host-Pathogen Interactions , Immunity, Innate , Virus Diseases , Viruses , Animals , Adaptive Immunity , Autoimmunity/genetics , B-Lymphocytes/immunology , Host-Pathogen Interactions/immunology , Immunity, Innate/genetics , Signal Transduction , Virus Diseases/immunology , Viruses/immunology , Humans
17.
J Mol Biol ; 435(16): 167976, 2023 08 15.
Article in English | MEDLINE | ID: mdl-36702393

ABSTRACT

The cellular defense against viruses involves the assembly of oligomers, granules and membraneless organelles (MLOs) that govern the activation of several arms of the innate immune response. Upon interaction with specific pathogen-derived ligands, a number of pattern recognition receptors (PRRs) undergo phase-separation thus triggering downstream signaling pathways. Among other relevant condensates, inflammasomes, apoptosis-associated speck-like protein containing a caspase-recruitment domain (ASC) specks, cyclic GMP-AMP synthase (cGAS) foci, protein kinase R (PKR) clusters, ribonuclease L-induced bodies (RLBs), stress granules (SGs), processing bodies (PBs) and promyelocytic leukemia protein nuclear bodies (PML NBs) play different roles in the immune response. In turn, viruses have evolved diverse strategies to evade the host defense. Viral DNA or RNA, as well as viral proteases or proteins carrying intrinsically disordered regions may interfere with condensate formation and function in multiple ways. In this review we discuss current and hypothetical mechanisms of viral escape that involve the disassembly, repurposing, or inactivation of membraneless condensates that govern innate immunity. We summarize emerging interconnections between these diverse condensates that ultimately determine the cellular outcome.


Subject(s)
Biomolecular Condensates , Immune Evasion , Immunity, Innate , Viruses , Biomolecular Condensates/immunology , Biomolecular Condensates/virology , Signal Transduction , Viruses/immunology
18.
Mol Cell ; 83(3): 481-495, 2023 02 02.
Article in English | MEDLINE | ID: mdl-36334591

ABSTRACT

Viral reproduction is contingent on viral protein synthesis that relies on the host ribosomes. As such, viruses have evolved remarkable strategies to hijack the host translational apparatus in order to favor viral protein production and to interfere with cellular innate defenses. Here, we describe the approaches viruses use to exploit the translation machinery, focusing on commonalities across diverse viral families, and discuss the functional relevance of this process. We illustrate the complementary strategies host cells utilize to block viral protein production and consider how cells ensure an efficient antiviral response that relies on translation during this tug of war over the ribosome. Finally, we highlight potential roles mRNA modifications and ribosome quality control play in translational regulation and innate immunity. We address these topics in the context of the COVID-19 pandemic and focus on the gaps in our current knowledge of these mechanisms, specifically in viruses with pandemic potential.


Subject(s)
COVID-19 , Protein Biosynthesis , Virus Diseases , Viruses , Humans , COVID-19/genetics , COVID-19/immunology , Pandemics , Protein Biosynthesis/genetics , Protein Biosynthesis/immunology , RNA, Viral/genetics , RNA, Viral/immunology , Viral Proteins/genetics , Viral Proteins/immunology , Virus Diseases/genetics , Virus Diseases/immunology , Viruses/genetics , Viruses/immunology , Ribosomes/genetics , Ribosomes/immunology , Ribosomes/virology
19.
Curr Opin Immunol ; 78: 102250, 2022 10.
Article in English | MEDLINE | ID: mdl-36209576

ABSTRACT

Recent advances in our understanding of nucleic acid pattern-recognition receptor (PRR) sensing of viruses have revealed a previously unappreciated level of complexity of the host antiviral response. As well as direct recognition of viral nucleic acid by PRRs, viruses also induce the release of host nucleic acid from the nucleus and mitochondria into the cytosol, which boosts nucleic acid activation of antiviral PRRs. Crosstalk and cooperation between DNA- and RNA-recognition signaling pathways has also been revealed, as has direct restriction of viral genomes in an interferon-independent manner by PRRs, and new roles for inflammasomes in sensing viral nucleic acid. Further, newly identified viral-evasion strategies targeting PRR pathways emphasize the importance of nucleic acid detection during viral infection at the host-pathogen innate immune interface.


Subject(s)
Immunity, Innate , Nucleic Acids , Virus Diseases , Humans , Antiviral Agents , Inflammasomes , Interferons , Nucleic Acids/immunology , Nucleic Acids/metabolism , Receptors, Pattern Recognition/metabolism , RNA , Virus Diseases/immunology , Virus Diseases/metabolism , Viruses/immunology
20.
Nature ; 609(7926): 354-360, 2022 09.
Article in English | MEDLINE | ID: mdl-35978192

ABSTRACT

CD8+ T cells that respond to chronic viral infections or cancer are characterized by the expression of inhibitory receptors such as programmed cell death protein 1 (PD-1) and by the impaired production of cytokines. This state of restrained functionality-which is referred to as T cell exhaustion1,2-is maintained by precursors of exhausted T (TPEX) cells that express the transcription factor T cell factor 1 (TCF1), self-renew and give rise to TCF1- exhausted effector T cells3-6. Here we show that the long-term proliferative potential, multipotency and repopulation capacity of exhausted T cells during chronic infection are selectively preserved in a small population of transcriptionally distinct CD62L+ TPEX cells. The transcription factor MYB is not only essential for the development of CD62L+ TPEX cells and maintenance of the antiviral CD8+ T cell response, but also induces functional exhaustion and thereby prevents lethal immunopathology. Furthermore, the proliferative burst in response to PD-1 checkpoint inhibition originates exclusively from CD62L+ TPEX cells and depends on MYB. Our findings identify CD62L+ TPEX cells as a stem-like population that is central to the maintenance of long-term antiviral immunity and responsiveness to immunotherapy. Moreover, they show that MYB is a transcriptional orchestrator of two fundamental aspects of exhausted T cell responses: the downregulation of effector function and the long-term preservation of self-renewal capacity.


Subject(s)
CD8-Positive T-Lymphocytes , Programmed Cell Death 1 Receptor , Proto-Oncogene Proteins c-myb , CD8-Positive T-Lymphocytes/cytology , CD8-Positive T-Lymphocytes/immunology , Cell Proliferation , Cell Self Renewal , Hepatocyte Nuclear Factor 1-alpha/metabolism , Immunotherapy , L-Selectin/metabolism , Precursor Cells, T-Lymphoid/cytology , Precursor Cells, T-Lymphoid/immunology , Programmed Cell Death 1 Receptor/immunology , Programmed Cell Death 1 Receptor/metabolism , Proto-Oncogene Proteins c-myb/metabolism , Viruses/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...