scispace - formally typeset
Search or ask a question

What are exhausted t cells? 


Best insight from top research papers

Exhausted T cells refer to a state of T-cell dysfunction characterized by reduced effector functions and increased expression of inhibitory receptors. This exhaustion can arise from prolonged antigen exposure in chronic viral infections, cancer, and immune-mediated inflammatory diseases, leading to T cells losing their effector functions and memory characteristics. Exhausted T cells, particularly CD8+ T cells, exhibit impaired proliferative and cytotoxic capacities, along with elevated expression of co-inhibitory receptors, impacting their ability to combat tumors effectively. Strategies to reinvigorate exhausted T cells in the tumor microenvironment include immune checkpoint blockade, transcription factor-based therapy, epigenetic therapy, metabolism-based therapy, and cytokine therapy, aiming to enhance anti-tumor immunity and improve clinical outcomes. Understanding and targeting exhausted T cells are crucial in advancing cancer immunotherapy and potentially treating immune-mediated inflammatory diseases.

Answers from top 5 papers

More filters
Papers (5)Insight
Open accessJournal ArticleDOI
Joji Nagasaki, Yosuke Togashi 
7 Citations
Exhausted T cells in the tumor microenvironment are dysfunctional effector T cells with reduced cytokine secretion and increased inhibitory receptor expression, such as PD-1, impacting antitumor immune responses.
Exhausted T-cells lose effector function and memory T-cell characteristics due to prolonged stimulation, observed in chronic infections, cancer, and autoimmune diseases like IMIDs, showing potential for immunotherapy.
Exhausted CD8+ T cells (CD8+ TEXs) are dysfunctional T cells in the tumor microenvironment with impaired function and increased expression of co-inhibitory receptors, impacting anti-tumor immunity.
Exhausted T cells are hypo-responsive cells resulting from chronic antigen exposure in viral infections or cancer, characterized by unclear molecular definition and inconsistent surface receptor reliance in research.
Exhausted T cells are a subset of T cells that undergo dysfunction due to prolonged antigen exposure, impacting cancer-immunity cycles and potentially transitioning from assets to liabilities in tumor surveillance.

Related Questions

How cd8 involved in exhausted T cell through Intestinal immune network for IgA production?5 answersCD8+ T cells play a crucial role in the context of exhausted T cells within the intestinal immune network for IgA production. Exhausted CD8+ T cells are characterized by a loss of effector functions and the expression of inhibitory receptors, impacting immune responses against chronic infections or cancer. In the setting of autoimmune diseases like Celiac disease, a subset of CD8 T cells with regulatory properties, known as CD8 Tregs, can be activated through the T cell receptor to eliminate pathogenic CD4 T cells, contributing to immune regulation. Additionally, in colorectal cancer, pre-exhausted T cells express specific biomarkers like CCT6A and TUBA1B, which are associated with T cell exhaustion and overall patient survival, highlighting the complexity of T cell exhaustion mechanisms in cancer immunotherapy.
Can ebv lead to t cell exhaustion?5 answersEBV (Epstein-Barr virus) infection can indeed lead to T cell exhaustion. Studies have shown that patients with activated PI3Kδ syndrome (APDS), caused by gain-of-function mutations in PI3Kδ, experience impaired immune responses, including T cell exhaustion. This exhaustion is characterized by increased expression of inhibitory receptors like PD-1 on CD8+ T cells, similar to what is observed in HIV patients. Furthermore, inhibition of PD-1 on EBV-specific CD8+ T cells from APDS patients has been shown to enhance their proliferation and effector cytokine production, indicating a potential therapeutic approach to alleviate T cell exhaustion. Therefore, EBV infection, especially in conditions like APDS, can contribute to T cell exhaustion, highlighting the importance of understanding and addressing this phenomenon in the context of viral infections like COVID-19.
What are the relationships between T cell exhaustion and immune regulation?4 answersT cell exhaustion is a state of T cell dysfunction that occurs during chronic infections and tumor progression. It is characterized by low effector function, high expression of inhibitory receptors, unique metabolic patterns, and altered transcriptional profiles. The epigenetic changes associated with exhaustion are distinct from those seen in effector and memory cell differentiation. T cell exhaustion is driven by environmental signals that promote epigenetic modifications, leading to a trajectory towards a dysfunctional state. Factors such as antigen stimulation signal strength, cytokines, and epigenetics play a role in T cell exhaustion. The interaction between immune T cells and other tumor infiltrating cells or inflammatory cytokines in the tumor microenvironment also affects T cell exhaustion. Understanding and interfering with the regulatory mechanisms associated with T cell exhaustion has implications for immunotherapy.
What mechanisms contribute to T cell exhaustion in human diseases?5 answersT cell exhaustion in human diseases is driven by various mechanisms. A transcriptome analysis revealed an overlap of upregulated and downregulated genes in exhausted CD8+ T cells, which were enriched in exhaustion response-related pathways. The emergence of inhibitory receptors, such as programmed cell death protein-1 (PD-1), on CD8+ T cells is a leading cause of exhaustion. Additionally, the expression of inhibitory receptors and transcription factors, including PD-1, TIM3, TOX, and BLIMP1, is upregulated in exhausted Tregs. Imbalance between inhibitory and stimulatory signals, reprogramming of metabolism, and high levels of inhibitory receptors like PD-1, CTLA-4, TIM-3, and Lag-3 contribute to T cell exhaustion in chronic infections and cancer. Furthermore, Thymocyte selection-associated high mobility group box protein (TOX) genes and TOX-associated pathways play a critical role in driving T cell exhaustion in chronic infection and cancer.
Are CMV-specific T cells exhausted in chronic infection?5 answersCMV-specific T cells are not specifically mentioned in the abstracts provided.
What are the mechanisms of CMV-specific T cell exhaustion?3 answersT cell exhaustion in chronic viral infections is driven by factors such as high viral titers, strong TCR stimulation, and high antigen concentrations associated with strong inflammatory signals. The presence or absence of a microbiome does not influence the progression of T cell exhaustion in chronic lymphocytic choriomeningitis virus (LCMV) infections. However, the absence of the microbiome delays the partial viral control typically seen in chronic LCMV infections, indicating that the microbiome is critical for effector T cell function. Non-coding RNAs, including microRNAs and long non-coding RNAs, have been shown to participate in the regulation of T cell exhaustion during chronic viral infections. In chronic hepatitis B and C virus infections, persistently elevated antigen levels drive CD8+ T cells towards a state of exhaustion, which is associated with metabolic and signaling deregulations, as well as epigenetic alterations.

See what other people are reading

How does 3-bromopyruvate work in low concentrations?
5 answers
In low concentrations, 3-bromopyruvate (3-BP) demonstrates significant effects in various biological systems. When administered in chronic low doses to aged rats, 3-BP shows a hormetic effect by enhancing membrane-bound ATPase activities and reducing redox biomarker levels, suggesting its potential as an anti-aging agent. In pancreatic cancer cells and mouse models, 3-BP inhibits hexokinase II (HK2) enzyme, leading to a dose-dependent decrease in cell growth and ATP production, while also damaging mitochondrial integrity. Additionally, 3-BP disrupts the interaction between HK2 and VDAC1, inhibits glycolysis, and induces apoptosis in tumor tissues, highlighting its efficacy as an anti-tumor drug against pancreatic cancer. These findings collectively emphasize the multifaceted mechanisms through which 3-BP operates at low concentrations to exert anti-aging and anti-cancer effects.
Is KLK6 assoacited with cancer?
5 answers
KLK6 is indeed associated with cancer. Research indicates that KLK6 is significantly upregulated in pancreatic tumors compared to normal pancreatic tissue, impacting patient survival in pancreatic adenocarcinoma. Moreover, KLK6 plays a crucial role in tumor growth and metastasis in melanoma and lung carcinoma, acting as a molecular link between cancer cells and macrophages. In bladder urothelial carcinoma, high KLK6 expression correlates with poor prognosis, suggesting it as an independent prognostic factor and a potential therapeutic target. Additionally, in metastatic breast cancer, KLK6 is silenced through hypermethylation, with its re-expression inhibiting tumor progression and promoting a less malignant phenotype. Therefore, KLK6's involvement in various cancers underscores its significance as a potential biomarker and therapeutic target in cancer research.
How does GDF15 promote angiogenesis in prostate cancer?
5 answers
Growth Differentiation Factor-15 (GDF15) plays a crucial role in prostate cancer progression by promoting angiogenesis. Research demonstrates that GDF15 interacts with connective tissue growth factor (CCN2) and inhibits CCN2-mediated tube formation in endothelial cells, thereby blocking angiogenesis. Moreover, GDF15 secreted by prostate cancer cells stimulates cancer cell growth and invasion, contributing to tumor progression. Additionally, GDF15 facilitates the growth of prostate cancer in bone by activating osteoclastogenesis through osteoblastic production of CCL2 and RANKL, ultimately promoting bone metastases. These findings highlight the multifaceted role of GDF15 in prostate cancer, where it not only influences cancer cell behavior but also modulates the tumor microenvironment to support angiogenesis and metastasis.
What's MIF's role in angiogenesis of prostate cancer?
5 answers
Macrophage migration inhibitory factor (MIF) plays a crucial role in angiogenesis in prostate cancer. MIF is known to enhance malignant growth and metastasis by stimulating tumor cell proliferation, suppressing apoptotic death, facilitating invasion of the extracellular matrix, and promoting angiogenesis. Studies suggest that inhibiting MIF activity reduces the phenotypic hallmarks of cancer, including angiogenesis, by downregulating signaling pathways like ERK and AKT. Additionally, MIF can promote tumorigenesis by acting directly on cancer cells and reversing immunosuppression. Targeting oxidized MIF (oxMIF), a disease-related isoform, has shown promising results in reducing cancer cell invasion, angiogenesis, and proinflammatory cytokine production. Furthermore, the Nodal/ALK4 pathway, regulated by miR-185, is crucial in prostate cancer angiogenesis, highlighting a potential therapeutic target to inhibit angiogenesis and tumor progression.
How does fatty diet affect immunity?
5 answers
A high-fat diet (HFD) impacts immunity by inducing inflammation and altering immune cell populations in adipose tissue. Studies show that HFD consumption leads to increased infiltration of pro-inflammatory Th17 cells, dendritic cells, and macrophages, while reducing regulatory T cells. This results in elevated levels of inflammatory cytokines and chemokines, along with changes in key signaling pathways like PPAR-γ. Additionally, dietary fatty acids, particularly saturated fatty acids like palmitic acid (PA), can act as inflammatory stimuli, inducing a hyper-inflammatory response and altering immune memory. PA exposure enhances inflammation in macrophages through ceramide synthesis, contributing to long-lived innate immune memory. Conversely, oleic acid reverses PA-induced inflammation. These findings highlight the significant impact of fatty diets on immunity, influencing immune cell function and inflammatory responses.
How have tumor-on-a-chip platforms evolved in recent years to facilitate the study of cancer-immune system crosstalk?
5 answers
Tumor-on-a-chip platforms have evolved significantly in recent years to enhance the study of cancer-immune system crosstalk. These platforms utilize microfluidic technologies to create 3D cell co-cultures that mimic the tumor microenvironment, enabling researchers to investigate immune cell recruitment, tumor progression, and response to immunotherapies in a patient-specific manner. By incorporating various cell types, such as cancer cells, immune cells, and stromal cells, these platforms can replicate the complex interactions within the tumor ecosystem. Additionally, advanced imaging techniques and computational algorithms allow for real-time monitoring of cellular behaviors and responses to treatments. Overall, tumor-on-a-chip platforms offer a promising approach to advancing our understanding of cancer-immune system interactions and improving the efficacy of immunotherapies through personalized medicine applications.
Inulin effects in mice?
5 answers
In mice, inulin supplementation has shown various beneficial effects. Studies have demonstrated that inulin can lead to weight loss, improved glucose tolerance, increased energy expenditure, and reduction in visceral adipose tissue. Inulin alters the gut microbiota composition, increasing the abundance of beneficial bacteria like Alistipes and promoting the production of short-chain fatty acids (SCFAs). Furthermore, inulin intake has been associated with anti-inflammatory properties, potentially through the inhibition of the NF-κB signaling pathway and the promotion of defense proteins like Hmox1. These findings suggest that inulin plays a significant role in modulating metabolic parameters, gut microbiota, and inflammatory responses in mice, highlighting its potential as a nutritional strategy for addressing metabolic diseases and obesity-related pathologies.
What are the effects of IL-21 on CD4 and CD8 T cells?
5 answers
IL-21 has distinct effects on CD4 and CD8 T cells. It activates STAT3 in CD8+ T cells, promoting anti-tumor immunity. IL-21 is crucial for the activation of CD8+ T cells, aiding in their function recovery from chronic infections and cancer. Additionally, IL-21 induces the expression of IL-1 receptor type I (IL-1RI) on CD8+ T cells, enhancing their effector function by increasing cytokine production and cytotoxic granule release. On the other hand, IL-21 can polarize Tc1 cells towards hyper cytotoxicity without hyper polarization, suggesting a potential role in driving extreme aberrancy in SLE CD8+ T cells. Overall, IL-21 plays a significant role in modulating the function and responses of both CD4 and CD8 T cells in various immune contexts.
What are the Clinical and preclinical states of CAR T cell therapy in vivo for solid tumors cancer?
5 answers
Clinical and preclinical states of CAR T-cell therapy for solid tumors cancer are evolving. Challenges like antigen heterogeneity and immunosuppressive tumor microenvironment hinder CAR T-cell efficacy in solid tumors. Strategies to enhance CAR T-cell function include incorporating co-stimulatory domains, developing armored CAR-T cells, and exploring other immune cells like CAR-NK and CAR-M cells. Novel CAR T-cell products like YTB323 show enhanced in vivo expansion and antitumor activity in preclinical models, with promising safety and efficacy profiles in clinical trials for r/r DLBCL. To overcome limitations such as restricted trafficking, hypoxic TME, antigen escape, and exhaustion, researchers are advancing CAR designs and combining therapies to optimize clinical efficacy in solid tumors.
What is the mechanism of reverse cholesterol transport pathway?
5 answers
The reverse cholesterol transport (RCT) pathway involves the transport of excess peripheral cholesterol to the liver for conversion into bile acids and subsequent excretion, reducing hyperlipidemia and cardiovascular disease risks. Key proteins involved in RCT include ATP-binding cassette sub-family A1 (ABCA1), scavenger receptor class B type I (SR-BI), apolipoprotein A-I (ApoA-I), and apolipoprotein A-II (ApoA-II). Additionally, liver X receptor (LXR) and its downstream regulators ABCA1 and ATP-binding cassette subfamily G member 1 (ABCG1) genes play a crucial role in regulating cholesterol metabolism and the RCT pathway. Furthermore, the ATP-binding cassette transporter A1 (ABCA1) is central in mediating the biosynthesis of high-density lipoprotein (HDL), facilitating cholesterol efflux from macrophages and preventing lipid deposition, thus contributing to RCT.
How did rosenzweig 1978 introduced enriched environmet into practice?
10 answers
The concept of an enriched environment (EE) as introduced by Rosenzweig in 1978 has significantly evolved and expanded in practice across various fields of research, particularly in understanding and enhancing brain function and recovery from neurological injuries. While Rosenzweig's foundational work laid the groundwork, subsequent studies have built upon and diversified the application of EE in experimental settings. EE involves modifications to the living conditions of laboratory animals to improve their biological conditions, promoting transcriptional and translational effects that enhance cellular plasticity and cognitive performance. This paradigm has been shown to ameliorate motor, sensory, and cognitive stimulation in animals, compared with those housed under standard conditions. The principle behind EE is to provide a combination of increased social interaction, physical activity, spatial complexity, and novelty, which collectively contribute to its beneficial effects on learning, memory, and recovery of function after brain injury. Research has demonstrated that EE can mitigate the effects of stress across generations by enhancing neurogenesis in the hippocampus, indicating its profound impact on cognitive development and resilience. In stroke recovery, EE, through multimodal stimulation including social interactions and sensorimotor stimulation, has shown promise in improving lost neurological function without affecting the extent of brain damage. This approach has also been explored in the context of behavioral epigenetics, where EE is used to correct aberrant epigenetic effects of stress, highlighting the intricate relationship between biological and biographical events in the understanding of stress. Moreover, EE's role extends beyond neurological and cognitive benefits. It has been found to have therapeutic potential in metabolic disorders, such as obesity, by restoring energy balance and improving metabolic alterations through enhanced central nervous system activity and reduced inflammation. These diverse applications underscore the multifaceted benefits of EE, rooted in the pioneering work of Rosenzweig, and its ongoing relevance in contemporary research across neurology, psychiatry, and beyond.