Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 39.561
Filter
Add more filters

Uruguay Oncology Collection
Publication year range
1.
Annu Rev Immunol ; 40: 271-294, 2022 04 26.
Article in English | MEDLINE | ID: mdl-35080919

ABSTRACT

Vertebrate immune systems suppress viral infection using both innate restriction factors and adaptive immunity. Viruses mutate to escape these defenses, driving hosts to counterevolve to regain fitness. This cycle recurs repeatedly, resulting in an evolutionary arms race whose outcome depends on the pace and likelihood of adaptation by host and viral genes. Although viruses evolve faster than their vertebrate hosts, their proteins are subject to numerous functional constraints that impact the probability of adaptation. These constraints are globally defined by evolutionary landscapes, which describe the fitness and adaptive potential of all possible mutations. We review deep mutational scanning experiments mapping the evolutionary landscapes of both host and viral proteins engaged in arms races. For restriction factors and some broadly neutralizing antibodies, landscapes favor the host, which may help to level the evolutionary playing field against rapidly evolving viruses. We discuss the biophysical underpinnings of these landscapes and their therapeutic implications.


Subject(s)
Virus Diseases , Viruses , Animals , Biological Evolution , Humans , Mutation , Viral Proteins , Virus Diseases/genetics , Viruses/genetics
2.
Annu Rev Immunol ; 38: 511-539, 2020 04 26.
Article in English | MEDLINE | ID: mdl-32340578

ABSTRACT

The continuous interactions between host and pathogens during their coevolution have shaped both the immune system and the countermeasures used by pathogens. Natural killer (NK) cells are innate lymphocytes that are considered central players in the antiviral response. Not only do they express a variety of inhibitory and activating receptors to discriminate and eliminate target cells but they can also produce immunoregulatory cytokines to alert the immune system. Reciprocally, several unrelated viruses including cytomegalovirus, human immunodeficiency virus, influenza virus, and dengue virus have evolved a multitude of mechanisms to evade NK cell function, such as the targeting of pathways for NK cell receptors and their ligands, apoptosis, and cytokine-mediated signaling. The studies discussed in this article provide further insights into the antiviral function of NK cells and the pathways involved, their constituent proteins, and ways in which they could be manipulated for host benefit.


Subject(s)
Host-Pathogen Interactions/immunology , Immune Evasion , Killer Cells, Natural/immunology , Killer Cells, Natural/metabolism , Viruses/immunology , Animals , Biomarkers , Cytokines/metabolism , Humans , Receptors, Natural Killer Cell/metabolism , Signal Transduction , Virus Diseases/immunology , Virus Diseases/metabolism , Virus Diseases/virology
3.
Annu Rev Immunol ; 37: 247-267, 2019 04 26.
Article in English | MEDLINE | ID: mdl-30633609

ABSTRACT

Recognition of foreign nucleic acids is the primary mechanism by which a type I interferon-mediated antiviral response is triggered. Given that human cells are replete with DNA and RNA, this evolutionary strategy poses an inherent biological challenge, i.e., the fundamental requirement to reliably differentiate self-nucleic acids from nonself nucleic acids. We suggest that the group of Mendelian inborn errors of immunity referred to as the type I interferonopathies relate to a breakdown of self/nonself discrimination, with the associated mutant genotypes involving molecules playing direct or indirect roles in nucleic acid signaling. This perspective begs the question as to the sources of self-derived nucleic acids that drive an inappropriate immune response. Resolving this question will provide fundamental insights into immune tolerance, antiviral signaling, and complex autoinflammatory disease states. Here we develop these ideas, discussing type I interferonopathies within the broader framework of nucleic acid-driven inflammation.


Subject(s)
Antigens, Viral/immunology , Autoantigens/immunology , Immune System Diseases/immunology , Nucleic Acids/immunology , Virus Diseases/immunology , Animals , Humans , Immune System Diseases/genetics , Immune Tolerance , Immunity, Innate , Interferon Type I/metabolism , Virus Diseases/genetics
4.
Annu Rev Immunol ; 37: 457-495, 2019 04 26.
Article in English | MEDLINE | ID: mdl-30676822

ABSTRACT

Exhausted CD8 T (Tex) cells are a distinct cell lineage that arise during chronic infections and cancers in animal models and humans. Tex cells are characterized by progressive loss of effector functions, high and sustained inhibitory receptor expression, metabolic dysregulation, poor memory recall and homeostatic self-renewal, and distinct transcriptional and epigenetic programs. The ability to reinvigorate Tex cells through inhibitory receptor blockade, such as αPD-1, highlights the therapeutic potential of targeting this population. Emerging insights into the mechanisms of exhaustion are informing immunotherapies for cancer and chronic infections. However, like other immune cells, Tex cells are heterogeneous and include progenitor and terminal subsets with unique characteristics and responses to checkpoint blockade. Here, we review our current understanding of Tex cell biology, including the developmental paths, transcriptional and epigenetic features, and cell intrinsic and extrinsic factors contributing to exhaustion and how this knowledge may inform therapeutic targeting of Tex cells in chronic infections, autoimmunity, and cancer.


Subject(s)
Costimulatory and Inhibitory T-Cell Receptors/metabolism , Immunotherapy/methods , Neoplasms/immunology , Programmed Cell Death 1 Receptor/metabolism , T-Lymphocytes/physiology , Virus Diseases/immunology , Animals , Cellular Senescence , Chronic Disease , Clonal Anergy , Epigenesis, Genetic , Humans , Neoplasms/therapy , Virus Diseases/therapy
5.
Annu Rev Immunol ; 37: 125-144, 2019 04 26.
Article in English | MEDLINE | ID: mdl-30485751

ABSTRACT

Platelets have dual physiologic roles as both cellular mediators of thrombosis and immune modulatory cells. Historically, the thrombotic function of platelets has received significant research and clinical attention, but emerging research indicates that the immune regulatory roles of platelets may be just as important. We now know that in addition to their role in the acute thrombotic event at the time of myocardial infarction, platelets initiate and accelerate inflammatory processes that are part of the pathogenesis of atherosclerosis and myocardial infarction expansion. Furthermore, it is increasingly apparent from recent studies that platelets impact the pathogenesis of many vascular inflammatory processes such as autoimmune diseases, sepsis, viral infections, and growth and metastasis of many types of tumors. Therefore, we must consider platelets as immune cells that affect all phases of immune responses.


Subject(s)
Atherosclerosis/immunology , Autoimmune Diseases/immunology , Blood Platelets/immunology , Inflammation , Myocardial Infarction/immunology , Thrombosis/immunology , Virus Diseases/immunology , Animals , Carcinogenesis/immunology , Humans , Immunomodulation
6.
Annu Rev Immunol ; 37: 1-17, 2019 04 26.
Article in English | MEDLINE | ID: mdl-30379594

ABSTRACT

Each of us is a story. Mine is a story of doing science for 60 years, and I am honored to be asked to tell it. Even though this autobiography was written for the Annual Review of Immunology, I have chosen to describe my whole career in science because the segment that was immunology is so intertwined with all else I was doing. This article is an elongation and modification of a talk I gave at my 80th birthday celebration at Caltech on March 23, 2018.


Subject(s)
Allergy and Immunology/history , NF-kappa B/metabolism , RNA Viruses/physiology , Virus Diseases/immunology , Animals , Disease Models, Animal , Gene Rearrangement , History, 20th Century , History, 21st Century , Humans , Mice , Protein-Tyrosine Kinases/metabolism , Reverse Transcription , United States
7.
Annu Rev Immunol ; 37: 349-375, 2019 04 26.
Article in English | MEDLINE | ID: mdl-30673536

ABSTRACT

Detection of double-stranded RNAs (dsRNAs) is a central mechanism of innate immune defense in many organisms. We here discuss several families of dsRNA-binding proteins involved in mammalian antiviral innate immunity. These include RIG-I-like receptors, protein kinase R, oligoadenylate synthases, adenosine deaminases acting on RNA, RNA interference systems, and other proteins containing dsRNA-binding domains and helicase domains. Studies suggest that their functions are highly interdependent and that their interdependence could offer keys to understanding the complex regulatory mechanisms for cellular dsRNA homeostasis and antiviral immunity. This review aims to highlight their interconnectivity, as well as their commonalities and differences in their dsRNA recognition mechanisms.


Subject(s)
Immunity, Innate/genetics , RNA, Double-Stranded/genetics , Virus Diseases/immunology , 2',5'-Oligoadenylate Synthetase/metabolism , Animals , DEAD Box Protein 58/metabolism , Humans , Immunomodulation , Mammals , Nucleotide Deaminases/metabolism , RNA Interference , eIF-2 Kinase/metabolism
8.
Annu Rev Immunol ; 36: 667-694, 2018 04 26.
Article in English | MEDLINE | ID: mdl-29677479

ABSTRACT

Pattern recognition receptors (PRRs) survey intra- and extracellular spaces for pathogen-associated molecular patterns (PAMPs) within microbial products of infection. Recognition and binding to cognate PAMP ligand by specific PRRs initiates signaling cascades that culminate in a coordinated intracellular innate immune response designed to control infection. In particular, our immune system has evolved specialized PRRs to discriminate viral nucleic acid from host. These are critical sensors of viral RNA to trigger innate immunity in the vertebrate host. Different families of PRRs of virus infection have been defined and reveal a diversity of PAMP specificity for wide viral pathogen coverage to recognize and extinguish virus infection. In this review, we discuss recent insights in pathogen recognition by the RIG-I-like receptors, related RNA helicases, Toll-like receptors, and other RNA sensor PRRs, to present emerging themes in innate immune signaling during virus infection.


Subject(s)
DEAD Box Protein 58/metabolism , Host-Pathogen Interactions/genetics , Host-Pathogen Interactions/immunology , Virus Diseases/etiology , Virus Diseases/metabolism , Viruses/immunology , Animals , DEAD-box RNA Helicases/metabolism , Humans , Protein Processing, Post-Translational , RNA Helicases/metabolism , RNA, Viral/genetics , RNA, Viral/metabolism , Receptors, Immunologic , Signal Transduction , Toll-Like Receptors/metabolism
9.
Annu Rev Immunol ; 35: 313-336, 2017 04 26.
Article in English | MEDLINE | ID: mdl-28142323

ABSTRACT

Protective immune responses to viral infection are initiated by innate immune sensors that survey extracellular and intracellular space for foreign nucleic acids. The existence of these sensors raises fundamental questions about self/nonself discrimination because of the abundance of self-DNA and self-RNA that occupy these same compartments. Recent advances have revealed that enzymes that metabolize or modify endogenous nucleic acids are essential for preventing inappropriate activation of the innate antiviral response. In this review, we discuss rare human diseases caused by dysregulated nucleic acid sensing, focusing primarily on intracellular sensors of nucleic acids. We summarize lessons learned from these disorders, we rationalize the existence of these diseases in the context of evolution, and we propose that this framework may also apply to a number of more common autoimmune diseases for which the underlying genetics and mechanisms are not yet fully understood.


Subject(s)
Autoimmune Diseases of the Nervous System/immunology , Autoimmunity , Lupus Erythematosus, Systemic/immunology , Nervous System Malformations/immunology , Nucleic Acids/immunology , Virus Diseases/immunology , Animals , Humans , Immunity, Innate , Interferon Type I/metabolism , Toll-Like Receptors/metabolism
10.
Annu Rev Immunol ; 34: 575-608, 2016 05 20.
Article in English | MEDLINE | ID: mdl-27168245

ABSTRACT

Mucosal surfaces provide a remarkably effective barrier against potentially dangerous pathogens. Therefore, enhancing mucosal immunity through vaccines-strengthening that first line of defense-holds significant promise for reducing the burden of viral diseases. The large and varied class of viral pathogens, however, continues to present thorny challenges to vaccine development. Two primary difficulties exist: Viruses exhibit a stunning diversity of strategies for evading the host immune response, and even when we understand the nature of effective immune protection against a given virus, eliciting that protection is technically challenging. Only a few mucosal vaccines have surmounted these obstacles thus far. Recent developments, however, could greatly improve vaccine design. In this review, we first sketch out our understanding of mucosal immunity and then compare the herpes simplex virus, human immunodeficiency virus, and influenza virus to illustrate the distinct challenges of developing successful vaccines and to outline potential solutions.


Subject(s)
HIV/immunology , Immune Evasion , Immunity, Mucosal , Orthomyxoviridae/immunology , Simplexvirus/immunology , Viral Vaccines/immunology , Virus Diseases/immunology , Animals , Antibodies, Neutralizing/metabolism , Antibodies, Viral/metabolism , Humans , Immunologic Memory , Virus Diseases/prevention & control
11.
Cell ; 186(17): 3619-3631.e13, 2023 08 17.
Article in English | MEDLINE | ID: mdl-37595565

ABSTRACT

During viral infection, cells can deploy immune strategies that deprive viruses of molecules essential for their replication. Here, we report a family of immune effectors in bacteria that, upon phage infection, degrade cellular adenosine triphosphate (ATP) and deoxyadenosine triphosphate (dATP) by cleaving the N-glycosidic bond between the adenine and sugar moieties. These ATP nucleosidase effectors are widely distributed within multiple bacterial defense systems, including cyclic oligonucleotide-based antiviral signaling systems (CBASS), prokaryotic argonautes, and nucleotide-binding leucine-rich repeat (NLR)-like proteins, and we show that ATP and dATP degradation during infection halts phage propagation. By analyzing homologs of the immune ATP nucleosidase domain, we discover and characterize Detocs, a family of bacterial defense systems with a two-component phosphotransfer-signaling architecture. The immune ATP nucleosidase domain is also encoded within diverse eukaryotic proteins with immune-like architectures, and we show biochemically that eukaryotic homologs preserve the ATP nucleosidase activity. Our findings suggest that ATP and dATP degradation is a cell-autonomous innate immune strategy conserved across the tree of life.


Subject(s)
Virus Diseases , Humans , Eukaryotic Cells , Prokaryotic Cells , Adenosine Triphosphate , N-Glycosyl Hydrolases
12.
Cell ; 186(22): 4851-4867.e20, 2023 10 26.
Article in English | MEDLINE | ID: mdl-37848036

ABSTRACT

Post-acute sequelae of COVID-19 (PASC, "Long COVID") pose a significant global health challenge. The pathophysiology is unknown, and no effective treatments have been found to date. Several hypotheses have been formulated to explain the etiology of PASC, including viral persistence, chronic inflammation, hypercoagulability, and autonomic dysfunction. Here, we propose a mechanism that links all four hypotheses in a single pathway and provides actionable insights for therapeutic interventions. We find that PASC are associated with serotonin reduction. Viral infection and type I interferon-driven inflammation reduce serotonin through three mechanisms: diminished intestinal absorption of the serotonin precursor tryptophan; platelet hyperactivation and thrombocytopenia, which impacts serotonin storage; and enhanced MAO-mediated serotonin turnover. Peripheral serotonin reduction, in turn, impedes the activity of the vagus nerve and thereby impairs hippocampal responses and memory. These findings provide a possible explanation for neurocognitive symptoms associated with viral persistence in Long COVID, which may extend to other post-viral syndromes.


Subject(s)
Post-Acute COVID-19 Syndrome , Serotonin , Humans , COVID-19/complications , Disease Progression , Inflammation , Post-Acute COVID-19 Syndrome/blood , Post-Acute COVID-19 Syndrome/pathology , Serotonin/blood , Virus Diseases
13.
Cell ; 186(5): 957-974.e28, 2023 03 02.
Article in English | MEDLINE | ID: mdl-36812912

ABSTRACT

Bats are distinctive among mammals due to their ability to fly, use laryngeal echolocation, and tolerate viruses. However, there are currently no reliable cellular models for studying bat biology or their response to viral infections. Here, we created induced pluripotent stem cells (iPSCs) from two species of bats: the wild greater horseshoe bat (Rhinolophus ferrumequinum) and the greater mouse-eared bat (Myotis myotis). The iPSCs from both bat species showed similar characteristics and had a gene expression profile resembling that of cells attacked by viruses. They also had a high number of endogenous viral sequences, particularly retroviruses. These results suggest that bats have evolved mechanisms to tolerate a large load of viral sequences and may have a more intertwined relationship with viruses than previously thought. Further study of bat iPSCs and their differentiated progeny will provide insights into bat biology, virus host relationships, and the molecular basis of bats' special traits.


Subject(s)
Chiroptera , Pluripotent Stem Cells , Virus Diseases , Viruses , Animals , Viruses/genetics , Transcriptome , Phylogeny
14.
Cell ; 186(10): 2144-2159.e22, 2023 05 11.
Article in English | MEDLINE | ID: mdl-37172565

ABSTRACT

Bats are special in their ability to live long and host many emerging viruses. Our previous studies showed that bats have altered inflammasomes, which are central players in aging and infection. However, the role of inflammasome signaling in combating inflammatory diseases remains poorly understood. Here, we report bat ASC2 as a potent negative regulator of inflammasomes. Bat ASC2 is highly expressed at both the mRNA and protein levels and is highly potent in inhibiting human and mouse inflammasomes. Transgenic expression of bat ASC2 in mice reduced the severity of peritonitis induced by gout crystals and ASC particles. Bat ASC2 also dampened inflammation induced by multiple viruses and reduced mortality of influenza A virus infection. Importantly, it also suppressed SARS-CoV-2-immune-complex-induced inflammasome activation. Four key residues were identified for the gain of function of bat ASC2. Our results demonstrate that bat ASC2 is an important negative regulator of inflammasomes with therapeutic potential in inflammatory diseases.


Subject(s)
Apoptosis Regulatory Proteins , Chiroptera , Inflammasomes , Ribonucleoproteins , Virus Diseases , Animals , Humans , Mice , Apoptosis Regulatory Proteins/metabolism , Chiroptera/immunology , COVID-19 , Inflammasomes/immunology , Ribonucleoproteins/metabolism , SARS-CoV-2 , Virus Diseases/immunology , Virus Physiological Phenomena
15.
Annu Rev Immunol ; 33: 79-106, 2015.
Article in English | MEDLINE | ID: mdl-25493335

ABSTRACT

Cell proliferation and cell death are integral elements in maintaining homeostatic balance in metazoans. Disease pathologies ensue when these processes are disturbed. A plethora of evidence indicates that malfunction of cell death can lead to inflammation, autoimmunity, or immunodeficiency. Programmed necrosis or necroptosis is a form of nonapoptotic cell death driven by the receptor interacting protein kinase 3 (RIPK3) and its substrate, mixed lineage kinase domain-like (MLKL). RIPK3 partners with its upstream adaptors RIPK1, TRIF, or DAI to signal for necroptosis in response to death receptor or Toll-like receptor stimulation, pathogen infection, or sterile cell injury. Necroptosis promotes inflammation through leakage of cellular contents from damaged plasma membranes. Intriguingly, many of the signal adaptors of necroptosis have dual functions in innate immune signaling. This unique signature illustrates the cooperative nature of necroptosis and innate inflammatory signaling pathways in managing cell and organismal stresses from pathogen infection and sterile tissue injury.


Subject(s)
Inflammation/metabolism , Inflammation/pathology , Necrosis/metabolism , Signal Transduction , Animals , Bacterial Infections/genetics , Bacterial Infections/metabolism , Bacterial Infections/pathology , Biological Evolution , Cell Death , Humans , Inflammasomes/metabolism , Inflammation/genetics , Interleukin-1beta/metabolism , NF-kappa B/metabolism , Parasitic Diseases/genetics , Parasitic Diseases/metabolism , Parasitic Diseases/pathology , Phosphorylation , Protein Binding , Receptor-Interacting Protein Serine-Threonine Kinases/genetics , Receptor-Interacting Protein Serine-Threonine Kinases/metabolism , Ubiquitination , Virus Diseases/genetics , Virus Diseases/metabolism , Virus Diseases/pathology
16.
Cell ; 185(13): 2210-2212, 2022 06 23.
Article in English | MEDLINE | ID: mdl-35750032

ABSTRACT

Many approved drugs, including antivirals, are small-molecule inhibitors of disease-causing proteins. Such inhibitors often elicit resistance during treatment. Chaturvedi et al. propose new, feedback-disruptor (FD) antivirals that efficiently cure infected cells from viruses and minimize the chance of resistance, providing a new paradigm to treat viral infections and possibly other diseases.


Subject(s)
Antiviral Agents , Virus Diseases , Antiviral Agents/pharmacology , Antiviral Agents/therapeutic use , Feedback , Humans , Virus Diseases/drug therapy
17.
Annu Rev Immunol ; 32: 189-225, 2014.
Article in English | MEDLINE | ID: mdl-24423116

ABSTRACT

Adoptive immunotherapy, or the infusion of lymphocytes, is a promising approach for the treatment of cancer and certain chronic viral infections. The application of the principles of synthetic biology to enhance T cell function has resulted in substantial increases in clinical efficacy. The primary challenge to the field is to identify tumor-specific targets to avoid off-tumor, on-target toxicity. Given recent advances in efficacy in numerous pilot trials, the next steps in clinical development will require multicenter trials to establish adoptive immunotherapy as a mainstream technology.


Subject(s)
Immunotherapy, Adoptive , Neoplasms/immunology , Neoplasms/therapy , Virus Diseases/immunology , Virus Diseases/therapy , Adoptive Transfer , Animals , Antigens/genetics , Antigens/immunology , Biomarkers , Cell- and Tissue-Based Therapy , Gene Transfer Techniques , Genetic Therapy , Humans , Neoplasms/genetics , Receptors, Antigen, T-Cell/immunology , Receptors, Antigen, T-Cell/metabolism , T-Cell Antigen Receptor Specificity/immunology , T-Lymphocyte Subsets/cytology , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , Transduction, Genetic , Virus Diseases/genetics
18.
Cell ; 184(6): 1604-1620, 2021 03 18.
Article in English | MEDLINE | ID: mdl-33740455

ABSTRACT

Historically, emerging viruses appear constantly and have cost millions of human lives. Currently, climate change and intense globalization have created favorable conditions for viral transmission. Therefore, effective antivirals, especially those targeting the conserved protein in multiple unrelated viruses, such as the compounds targeting RNA-dependent RNA polymerase, are urgently needed to combat more emerging and re-emerging viruses in the future. Here we reviewed the development of antivirals with common targets, including those against the same protein across viruses, or the same viral function, to provide clues for development of antivirals for future epidemics.


Subject(s)
Antiviral Agents/therapeutic use , Communicable Diseases, Emerging/drug therapy , Communicable Diseases, Emerging/epidemiology , Molecular Targeted Therapy/methods , Pandemics , Virus Diseases/drug therapy , Virus Diseases/epidemiology , Viruses/enzymology , Animals , Antiviral Agents/pharmacology , Communicable Diseases, Emerging/virology , Humans , RNA-Dependent RNA Polymerase/antagonists & inhibitors , Viral Envelope Proteins/antagonists & inhibitors , Virus Diseases/virology , Virus Internalization/drug effects
19.
Nat Immunol ; 24(10): 1685-1697, 2023 Oct.
Article in English | MEDLINE | ID: mdl-37697097

ABSTRACT

Natural killer (NK) cells are innate cytotoxic lymphocytes with adaptive immune features, including antigen specificity, clonal expansion and memory. As such, NK cells share many transcriptional and epigenetic programs with their adaptive CD8+ T cell siblings. Various signals ranging from antigen, co-stimulation and proinflammatory cytokines are required for optimal NK cell responses in mice and humans during virus infection; however, the integration of these signals remains unclear. In this study, we identified that the transcription factor IRF4 integrates signals to coordinate the NK cell response during mouse cytomegalovirus infection. Loss of IRF4 was detrimental to the expansion and differentiation of virus-specific NK cells. This defect was partially attributed to the inability of IRF4-deficient NK cells to uptake nutrients required for survival and memory generation. Altogether, these data suggest that IRF4 is a signal integrator that acts as a secondary metabolic checkpoint to orchestrate the adaptive response of NK cells during viral infection.


Subject(s)
Cytomegalovirus Infections , Virus Diseases , Humans , Mice , Animals , Trained Immunity , Killer Cells, Natural , CD8-Positive T-Lymphocytes , Immunologic Memory
20.
Nat Immunol ; 24(2): 280-294, 2023 02.
Article in English | MEDLINE | ID: mdl-36543960

ABSTRACT

T cell dysfunctionality prevents the clearance of chronic infections and cancer. Furthermore, epigenetic programming in dysfunctional CD8+ T cells limits their response to immunotherapies, including immune checkpoint blockade (ICB). However, it is unclear which upstream signals drive acquisition of dysfunctional epigenetic programs, and whether therapeutically targeting these signals can remodel terminally dysfunctional T cells to an ICB-responsive state. Here we innovate an in vitro model system of stable human T cell dysfunction and show that chronic TGFß1 signaling in posteffector CD8+ T cells accelerates their terminal dysfunction through stable epigenetic changes. Conversely, boosting bone morphogenetic protein (BMP) signaling while blocking TGFß1 preserved effector and memory programs in chronically stimulated human CD8+ T cells, inducing superior responses to tumors and synergizing the ICB responses during chronic viral infection. Thus, rebalancing TGFß1/BMP signals provides an exciting new approach to unleash dysfunctional CD8+ T cells and enhance T cell immunotherapies.


Subject(s)
CD8-Positive T-Lymphocytes , Virus Diseases , Humans , Immune Checkpoint Inhibitors/pharmacology , Immune Checkpoint Inhibitors/therapeutic use , Immunotherapy , Signal Transduction , Transforming Growth Factor beta1/metabolism , Bone Morphogenetic Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL