Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 677
Filter
1.
Front Immunol ; 15: 1346446, 2024.
Article in English | MEDLINE | ID: mdl-39114669

ABSTRACT

The cyclic GMP-AMP synthase (cGAS)-stimulator of interferon genes (STING) signaling pathway is one of the fundamental mechanisms of the body's defense, which responds to the abnormal presence of double-stranded DNA in the cytoplasm to establish an effective natural immune response. In addition to detecting microbial infections, the cGAS pathway may be triggered by any cytoplasmic DNA, which is absent from the normal cytoplasm, and only conditions such as senescence and mitochondrial stress can lead to its leakage and cause sterile inflammation. A growing body of research has shown that the cGAS-STING pathway is strongly associated with sterile inflammation. In this study, we reviewed the regulatory mechanisms and biological functions of the cGAS-STING pathway through its involvement in aseptic inflammation in liver disease, kidney disease, and cellular senescence.


Subject(s)
Cellular Senescence , Kidney Diseases , Liver Diseases , Membrane Proteins , Nucleotidyltransferases , Signal Transduction , Humans , Nucleotidyltransferases/metabolism , Cellular Senescence/immunology , Membrane Proteins/metabolism , Kidney Diseases/immunology , Kidney Diseases/metabolism , Kidney Diseases/pathology , Animals , Liver Diseases/immunology , Liver Diseases/metabolism
2.
Commun Biol ; 7(1): 934, 2024 Aug 02.
Article in English | MEDLINE | ID: mdl-39095531

ABSTRACT

Epigenetic clocks are age predictors that use machine-learning models trained on DNA CpG methylation values to predict chronological or biological age. Increases in predicted epigenetic age relative to chronological age (epigenetic age acceleration) are connected to aging-associated pathologies, and changes in epigenetic age are linked to canonical aging hallmarks. However, epigenetic clocks rely on training data from bulk tissues whose cellular composition changes with age. Here, we found that human naive CD8+ T cells, which decrease in frequency during aging, exhibit an epigenetic age 15-20 years younger than effector memory CD8+ T cells from the same individual. Importantly, homogenous naive T cells isolated from individuals of different ages show a progressive increase in epigenetic age, indicating that current epigenetic clocks measure two independent variables, aging and immune cell composition. To isolate the age-associated cell intrinsic changes, we created an epigenetic clock, the IntrinClock, that did not change among 10 immune cell types tested. IntrinClock shows a robust predicted epigenetic age increase in a model of replicative senescence in vitro and age reversal during OSKM-mediated reprogramming.


Subject(s)
Aging , CD8-Positive T-Lymphocytes , DNA Methylation , Epigenesis, Genetic , Humans , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Aging/genetics , Aging/immunology , Adult , Female , Male , Young Adult , Middle Aged , Aged , Cellular Senescence/genetics , Cellular Senescence/immunology , Adolescent
3.
Int J Mol Sci ; 25(14)2024 Jul 10.
Article in English | MEDLINE | ID: mdl-39062798

ABSTRACT

Fibrosing interstitial lung diseases (ILDs) are characterized by the gradual and irreversible accumulation of scar tissue in the lung parenchyma. The role of the immune response in the pathogenesis of pulmonary fibrosis remains unclear. In recent years, substantial advancements have been made in our comprehension of the pathobiology driving fibrosing ILDs, particularly concerning various age-related cellular disturbances and immune mechanisms believed to contribute to an inadequate response to stress and increased susceptibility to lung fibrosis. Emerging studies emphasize cellular senescence as a key mechanism implicated in the pathobiology of age-related diseases, including pulmonary fibrosis. Cellular senescence, marked by antagonistic pleiotropy, and the complex interplay with immunity, are pivotal in comprehending many aspects of lung fibrosis. Here, we review progress in novel concepts in cellular senescence, its association with the dysregulation of the immune response, and the evidence underlining its detrimental role in fibrosing ILDs.


Subject(s)
Cellular Senescence , Lung Diseases, Interstitial , Pulmonary Fibrosis , Humans , Cellular Senescence/immunology , Lung Diseases, Interstitial/immunology , Lung Diseases, Interstitial/pathology , Pulmonary Fibrosis/immunology , Pulmonary Fibrosis/pathology , Animals , Lung/immunology , Lung/pathology , Immunity
4.
Nat Commun ; 15(1): 6162, 2024 Jul 22.
Article in English | MEDLINE | ID: mdl-39039076

ABSTRACT

Senescent cells within tumors and their stroma exert complex pro- and anti-tumorigenic functions. However, the identities and traits of these cells, and the potential for improving cancer therapy through their targeting, remain poorly characterized. Here, we identify a senescent subset within previously-defined cancer-associated fibroblasts (CAFs) in pancreatic ductal adenocarcinomas (PDAC) and in premalignant lesions in mice and humans. Senescent CAFs isolated from mouse and humans expressed elevated levels of immune-regulatory genes. Depletion of senescent CAFs, either genetically or using the Bcl-2 inhibitor ABT-199 (venetoclax), increased the proportion of activated CD8+ T cells in mouse pancreatic carcinomas, whereas induction of CAF senescence had the opposite effect. Combining ABT-199 with an immune checkpoint therapy regimen significantly reduced mouse tumor burden. These results indicate that senescent CAFs in PDAC stroma limit the numbers of activated cytotoxic CD8+ T cells, and suggest that their targeted elimination through senolytic treatment may enhance immunotherapy.


Subject(s)
CD8-Positive T-Lymphocytes , Cancer-Associated Fibroblasts , Carcinoma, Pancreatic Ductal , Cellular Senescence , Immunotherapy , Pancreatic Neoplasms , Sulfonamides , Animals , Cancer-Associated Fibroblasts/immunology , Cancer-Associated Fibroblasts/metabolism , Cancer-Associated Fibroblasts/pathology , Pancreatic Neoplasms/immunology , Pancreatic Neoplasms/therapy , Pancreatic Neoplasms/pathology , Pancreatic Neoplasms/genetics , CD8-Positive T-Lymphocytes/immunology , Mice , Humans , Carcinoma, Pancreatic Ductal/immunology , Carcinoma, Pancreatic Ductal/therapy , Carcinoma, Pancreatic Ductal/pathology , Carcinoma, Pancreatic Ductal/genetics , Cellular Senescence/immunology , Immunotherapy/methods , Sulfonamides/pharmacology , Sulfonamides/therapeutic use , Mice, Inbred C57BL , Cell Line, Tumor , Lymphocyte Activation/immunology , Female , Immune Checkpoint Inhibitors/pharmacology , Immune Checkpoint Inhibitors/therapeutic use , Adenocarcinoma/immunology , Adenocarcinoma/therapy , Adenocarcinoma/genetics , Adenocarcinoma/pathology , Male , Bridged Bicyclo Compounds, Heterocyclic
5.
Int J Mol Sci ; 25(13)2024 Jun 28.
Article in English | MEDLINE | ID: mdl-39000211

ABSTRACT

Diabetes is associated with numerous comorbidities, one of which is increased vulnerability to infections. This review will focus on how diabetes mellitus (DM) affects the immune system and its various components, leading to the impaired proliferation of immune cells and the induction of senescence. We will explore how the pathology of diabetes-induced immune dysfunction may have similarities to the pathways of "inflammaging", a persistent low-grade inflammation common in the elderly. Inflammaging may increase the likelihood of conditions such as rheumatoid arthritis (RA) and periodontitis at a younger age. Diabetes affects bone marrow composition and cellular senescence, and in combination with advanced age also affects lymphopoiesis by increasing myeloid differentiation and reducing lymphoid differentiation. Consequently, this leads to a reduced immune system response in both the innate and adaptive phases, resulting in higher infection rates, reduced vaccine response, and increased immune cells' senescence in diabetics. We will also explore how some diabetes drugs induce immune senescence despite their benefits on glycemic control.


Subject(s)
Diabetes Mellitus , Humans , Diabetes Mellitus/immunology , Diabetes Mellitus/pathology , Animals , Cellular Senescence/immunology , Inflammation/immunology , Inflammation/pathology , Immune System/immunology
6.
Front Immunol ; 15: 1375730, 2024.
Article in English | MEDLINE | ID: mdl-39007138

ABSTRACT

Solid gastrointestinal tumors often respond poorly to immunotherapy for the complex tumor microenvironment (TME), which is exacerbated by immune system alterations. Immunosenescence is the process of increased diversification of immune genes due to aging and other factors, leading to a decrease in the recognition function of the immune system. This process involves immune organs, immune cells, and the senescence-associated secretory phenotype (SASP). The most fundamental change is DNA damage, resulting in TME remodeling. The main manifestations are worsening inflammation, increased immunosuppressive SASP production, decreased immune cell antitumor activity, and the accumulation of tumor-associated fibroblasts and myeloid-derived suppressor cells, making antitumor therapy less effective. Senotherapy strategies to remove senescent cells and block key senescence processes can have synergistic effects with other treatments. This review focuses on immunoenescence and its impact on the solid TME. We characterize the immunosenescent TME and discuss future directions for antitumor therapies targeting senescence.


Subject(s)
Gastrointestinal Neoplasms , Immunosenescence , Tumor Microenvironment , Humans , Gastrointestinal Neoplasms/immunology , Gastrointestinal Neoplasms/therapy , Tumor Microenvironment/immunology , Immunosenescence/immunology , Animals , Immunotherapy/methods , Senescence-Associated Secretory Phenotype/immunology , Cellular Senescence/immunology
7.
Braz J Med Biol Res ; 57: e13225, 2024.
Article in English | MEDLINE | ID: mdl-38896644

ABSTRACT

Innate immune system activation is crucial in the inflammatory response, but uncontrolled activation can lead to autoimmune diseases. Cellular exhaustion and senescence are two processes that contribute to innate immune tolerance breakdown. Exhausted immune cells are unable to respond adequately to specific antigens or stimuli, while senescent cells have impaired DNA replication and metabolic changes. These processes can impair immune system function and disrupt homeostasis, leading to the emergence of autoimmunity. However, the influence of innate immune exhaustion and senescence on autoimmune disorders is not well understood. This review aims to describe the current findings on the role of innate immune exhaustion and senescence in autoimmunity, focusing on the cellular and molecular changes involved in each process. Specifically, the article explores the markers and pathways associated with immune exhaustion, such as PD-1 and TIM-3, and senescence, including Β-galactosidase (ß-GAL), lamin B1, and p16ink4a, and their impact on autoimmune diseases, namely type 1 diabetes, rheumatoid arthritis, systemic lupus erythematosus, and immune-mediated myopathies. Understanding the mechanisms underlying innate immune exhaustion and senescence in autoimmunity may provide insights for the development of novel therapeutic strategies.


Subject(s)
Autoimmune Diseases , Cellular Senescence , Immunity, Innate , Humans , Immunity, Innate/immunology , Autoimmune Diseases/immunology , Cellular Senescence/immunology , Autoimmunity/immunology , Immune System Exhaustion
8.
Aging (Albany NY) ; 16(12): 10657-10665, 2024 06 26.
Article in English | MEDLINE | ID: mdl-38942604

ABSTRACT

Two recent seminal works have untangled the intricate role of tumor-associated senescent cells in cancer progression, or regression, by guiding our immune system against cancer cells. The characterization of these unique, yet diverse cell populations, should be considered, particularly when contemplating the use of senolytics, which are drugs that selectively eliminate senescent cells, in a cancer framework. Here, we will describe the current knowledge in this field. In particular, we will discuss how the presence of senescent cells in tumors could be used as a therapeutic target in immunogenic cancers and how we may hypothetically design an adaptive anti-aging vaccine.


Subject(s)
Aging , Cancer Vaccines , Cellular Senescence , Neoplasms , Humans , Neoplasms/immunology , Cellular Senescence/immunology , Aging/immunology , Cancer Vaccines/immunology , Animals
9.
Oncoimmunology ; 13(1): 2367777, 2024.
Article in English | MEDLINE | ID: mdl-38887372

ABSTRACT

T lymphocytes expressing CD57 and lacking costimulatory receptors CD27/CD28 have been reported to accumulate with aging, chronic infection, and cancer. These cells are described as senescent, with inability to proliferate but enhanced cytolytic and cytokine-producing capacity. However, robust functional studies on these cells taken directly from cancer patients are lacking. We isolated these T cells and their CD27/28+ counterparts from blood and tumor samples of 50 patients with previously untreated head and neck cancer. Functional studies confirmed that these cells have enhanced ability to degranulate and produce IFN-γ. They also retain the ability to proliferate, thus are not senescent. These data suggest that CD27/28-CD57+ CD8+ T cells are a subset of highly differentiated, CD45RA+ effector memory (TEMRA) cells with retained proliferative capacity. Patients with > 34% of these cells among CD8+ T cells in the blood had a higher rate of locoregional disease relapse, suggesting these cells may have prognostic significance.


Subject(s)
CD28 Antigens , CD57 Antigens , CD8-Positive T-Lymphocytes , Cellular Senescence , Head and Neck Neoplasms , Humans , CD28 Antigens/metabolism , CD57 Antigens/metabolism , Head and Neck Neoplasms/immunology , Head and Neck Neoplasms/pathology , Head and Neck Neoplasms/metabolism , Male , Middle Aged , Female , Aged , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Cellular Senescence/immunology , Interferon-gamma/metabolism , Adult , Cell Proliferation , Aged, 80 and over
10.
J Dermatol Sci ; 114(3): 94-103, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38806324

ABSTRACT

BACKGROUND: Elderly atopic dermatitis (AD) is a subtype of AD defined by age (≥ 60 years). The molecular characteristics of elderly AD remain to be clarified. OBJECTIVE: We sought to characterize the molecular features of skin lesions and peripheral blood mononuclear cells (PBMCs) in patients with AD across different age, focusing on elderly AD. METHODS: Skin and PBMCs samples were used for RNA sequencing. Analysis of differentially expressed genes and gene set variation analysis were performed. Immunofluorescence staining, quantitative real-time PCR (qRT-PCR), flow cytometry and transwell assay were used for validation. RESULTS: Compared with healthy controls, the skin transcriptome of AD patients showed common signatures of AD, like barrier dysfunction and enhanced Th1/Th2/Th17 immune pathways. In PBMCs, the expression of Th1/Th2 response genes was more remarkable in adult AD, while expression of Th17-related genes was significantly higher in childhood AD. The gene modules associated with natural killer (NK) cells were downregulated in elderly AD. In skin lesions, elderly AD exhibited enrichment of macrophages, fibroblasts and senescence-associated secretory phenotype (SASP) related genes. The correlation among fibroblasts, SASP and innate immune cells were revealed by the co-localization of fibroblasts, macrophages and NK cells in the lesions across different age groups. Fibroblasts under inflammation or senescence could induce stronger chemotaxis of macrophages and NK cells. CONCLUSION: We identified the molecular phenotypes of skin lesions and PBMCs in elderly AD individuals. Fibroblasts, innate immune cells, and SASP might play important roles in the pathogenesis of elderly AD.


Subject(s)
Cellular Senescence , Dermatitis, Atopic , Fibroblasts , Immunity, Innate , Killer Cells, Natural , Skin , Humans , Dermatitis, Atopic/immunology , Dermatitis, Atopic/pathology , Dermatitis, Atopic/genetics , Fibroblasts/immunology , Fibroblasts/metabolism , Aged , Middle Aged , Male , Female , Skin/immunology , Skin/pathology , Killer Cells, Natural/immunology , Cellular Senescence/immunology , Adult , Case-Control Studies , Transcriptome/immunology , Young Adult , Adolescent , Child , Leukocytes, Mononuclear/immunology , Leukocytes, Mononuclear/metabolism , Macrophages/immunology , Macrophages/metabolism , Age Factors , Gene Expression Profiling , Th17 Cells/immunology
11.
Aging Cell ; 23(7): e14162, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38689516

ABSTRACT

Clearance of senescent cells has demonstrated therapeutic potential in the context of chronic age-related diseases. Little is known, however, how clearing senescent cells affects the ability to respond to an acute infection and form quality immunological memory. We aimed to probe the effects of clearing senescent cells in aged mice on the immune response to influenza (flu) infection. We utilized a p16 trimodality reporter mouse model (p16-3MR) to allow for identification and selective clearance of p16-expressing cells upon administration of ganciclovir (GCV). While p16-expressing cells may exacerbate dysfunctional responses to a primary infection, our data suggest they may play a role in fostering memory cell generation. We demonstrate that although clearance of p16-expressing cells enhanced viral clearance, this also severely limited antibody production in the lungs of flu-infected aged mice. 30 days later, there were fewer flu-specific CD8 memory T cells and lower levels of flu-specific antibodies in the lungs of GCV-treated mice. Furthermore, GCV-treated mice were unable to mount an optimal memory response and demonstrated increased viral load following heterosubtypic challenge. These results suggest that targeting senescent cells may potentiate primary responses while limiting the ability to form durable and protective immune memory with age.


Subject(s)
Cellular Senescence , Orthomyxoviridae Infections , Animals , Cellular Senescence/immunology , Mice , Orthomyxoviridae Infections/immunology , Orthomyxoviridae Infections/virology , Aging/immunology , Mice, Inbred C57BL , Influenza, Human/immunology , Influenza, Human/virology
12.
Pharmacol Res ; 204: 107198, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38692466

ABSTRACT

In-depth studies of the tumor microenvironment (TME) have helped to elucidate its cancer-promoting mechanisms and inherent characteristics. Cellular senescence, which acts as a response to injury and can the release of senescence-associated secretory phenotypes (SASPs). These SASPs release various cytokines, chemokines, and growth factors, remodeling the TME. This continual development of a senescent environment could be associated with chronic inflammation and immunosuppressive TME. Additionally, SASPs could influence the phenotype and function of macrophages, leading to the recruitment of tumor-associated macrophages (TAMs). This contributes to tumor proliferation and metastasis in the senescent microenvironment, working in tandem with immune regulation, angiogenesis, and therapeutic resistance. This comprehensive review covers the evolving nature of the senescent microenvironment, macrophages, and TAMs in tumor development. We also explored the links between chronic inflammation, immunosuppressive TME, cellular senescence, and macrophages. Moreover, we compiled various tumor-specific treatment strategies centered on cellular senescence and the current challenges in cellular senescence research. This study aimed to clarify the mechanism of macrophages and the senescent microenvironment in tumor progression and advance the development of targeted tumor therapies.


Subject(s)
Cellular Senescence , Macrophages , Neoplasms , Tumor Microenvironment , Tumor-Associated Macrophages , Humans , Tumor Microenvironment/immunology , Animals , Neoplasms/immunology , Neoplasms/drug therapy , Neoplasms/pathology , Macrophages/immunology , Cellular Senescence/immunology , Tumor-Associated Macrophages/immunology , Tumor-Associated Macrophages/metabolism , Tumor-Associated Macrophages/drug effects , Senescence-Associated Secretory Phenotype
13.
Clin Exp Immunol ; 217(3): 279-290, 2024 Aug 09.
Article in English | MEDLINE | ID: mdl-38700066

ABSTRACT

Natural killer (NK) cells include different subsets with diverse effector capacities that are poorly understood in the context of parasitic diseases. Here, we investigated inhibitory and activating receptor expression on NK cells in patients with cutaneous leishmaniasis (CL) and explored their phenotypic and functional heterogeneity based on CD57 and NKG2C expression. The expression of CD57 identified NK cells that accumulated in CL patients and exhibited features of senescence. The CD57+ cells exhibited heightened levels of the activating receptor NKG2C and diminished expression of the inhibitory receptor NKG2A. RNA sequencing analyses based on NKG2C transcriptome have revealed two distinct profiles among CL patients associated with cytotoxic and functional genes. The CD57+NKG2C+ subset accumulated in the blood of patients and presented conspicuous features of senescence, including the expression of markers such as p16, yH2ax, and p38, as well as reduced proliferative capacity. In addition, they positively correlated with the number of days until lesion resolution. This study provides a broad understanding of the NK cell biology during Leishmania infection and reinforces the role of senescent cells in the adverse clinical outcomes of CL.


Subject(s)
CD57 Antigens , Cellular Senescence , Killer Cells, Natural , Leishmaniasis, Cutaneous , NK Cell Lectin-Like Receptor Subfamily C , Humans , Leishmaniasis, Cutaneous/immunology , Killer Cells, Natural/immunology , CD57 Antigens/metabolism , CD57 Antigens/immunology , NK Cell Lectin-Like Receptor Subfamily C/metabolism , NK Cell Lectin-Like Receptor Subfamily C/immunology , Cellular Senescence/immunology , Male , Female , Adult , Middle Aged , Young Adult
14.
Eur J Immunol ; 54(7): e2350603, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38752316

ABSTRACT

Systemic lupus erythematosus (SLE) is a complex autoimmune disease characterized by persistent activation of immune cells and overproduction of autoantibodies. The accumulation of senescent T and B cells has been observed in SLE and other immune-mediated diseases. However, the exact mechanistic pathways contributing to this process in SLE remain incompletely understood. In this study, we found that in SLE patients: (1) the frequency of CD4+CD57+ senescent T cells was significantly elevated and positively correlated with disease activity; (2) the expression levels of B-lymphoma-2 (BCL-2) family and interferon-induced genes (ISGs) were significantly upregulated; and (3) in vitro, the cytokine IL-15 stimulation increased the frequency of senescent CD4+ T cells and upregulated the expression of BCL-2 family and ISGs. Further, treatment with ABT-263 (a senolytic BCL-2 inhibitor) in MRL/lpr mice resulted in decreased: (1) frequency of CD4+CD44hiCD62L-PD-1+CD153+ senescent CD4+ T cells; (2) frequency of CD19+CD11c+T-bet+ age-related B cells; (3) level of serum antinuclear antibody; (4) proteinuria; (5) frequency of Tfh cells; and (6) renal histopathological abnormalities. Collectively, these results indicated a dominant role for CD4+CD57+ senescent CD4+ T cells in the pathogenesis of SLE and senolytic BCL-2 inhibitor ABT-263 may be the potential treatment in ameliorating lupus phenotypes.


Subject(s)
CD4-Positive T-Lymphocytes , Cellular Senescence , Lupus Erythematosus, Systemic , Proto-Oncogene Proteins c-bcl-2 , Sulfonamides , Lupus Erythematosus, Systemic/immunology , Lupus Erythematosus, Systemic/drug therapy , Animals , Humans , Mice , Proto-Oncogene Proteins c-bcl-2/antagonists & inhibitors , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins c-bcl-2/metabolism , Cellular Senescence/immunology , Cellular Senescence/drug effects , Sulfonamides/pharmacology , CD4-Positive T-Lymphocytes/immunology , Female , Adult , Aniline Compounds/pharmacology , Aniline Compounds/therapeutic use , Mice, Inbred MRL lpr , Middle Aged , Male , Senotherapeutics/pharmacology
16.
Cells ; 13(9)2024 Apr 25.
Article in English | MEDLINE | ID: mdl-38727285

ABSTRACT

With the increasing proportion of the aging population, neurodegenerative diseases have become one of the major health issues in society. Neurodegenerative diseases (NDs), including multiple sclerosis (MS), Alzheimer's disease (AD), Parkinson's disease (PD), and amyotrophic lateral sclerosis (ALS), are characterized by progressive neurodegeneration associated with aging, leading to a gradual decline in cognitive, emotional, and motor functions in patients. The process of aging is a normal physiological process in human life and is accompanied by the aging of the immune system, which is known as immunosenescence. T-cells are an important part of the immune system, and their senescence is the main feature of immunosenescence. The appearance of senescent T-cells has been shown to potentially lead to chronic inflammation and tissue damage, with some studies indicating a direct link between T-cell senescence, inflammation, and neuronal damage. The role of these subsets with different functions in NDs is still under debate. A growing body of evidence suggests that in people with a ND, there is a prevalence of CD4+ T-cell subsets exhibiting characteristics that are linked to senescence. This underscores the significance of CD4+ T-cells in NDs. In this review, we summarize the classification and function of CD4+ T-cell subpopulations, the characteristics of CD4+ T-cell senescence, the potential roles of these cells in animal models and human studies of NDs, and therapeutic strategies targeting CD4+ T-cell senescence.


Subject(s)
CD4-Positive T-Lymphocytes , Cellular Senescence , Neurodegenerative Diseases , Humans , Neurodegenerative Diseases/immunology , Neurodegenerative Diseases/pathology , Neurodegenerative Diseases/therapy , CD4-Positive T-Lymphocytes/immunology , Cellular Senescence/immunology , Animals , Aging/immunology , Aging/pathology , T-Cell Senescence
17.
Ageing Res Rev ; 97: 102296, 2024 06.
Article in English | MEDLINE | ID: mdl-38588867

ABSTRACT

Fibroblasts are abundant stromal cells which not only control the integrity of extracellular matrix (ECM) but also act as immune regulators. It is known that the structural cells within tissues can establish an organ-specific immunity expressing many immune-related genes and closely interact with immune cells. In fact, fibroblasts can modify their immune properties to display both pro-inflammatory and immunosuppressive activities in a context-dependent manner. After acute insults, fibroblasts promote tissue inflammation although they concurrently recruit immunosuppressive cells to enhance the resolution of inflammation. In chronic pathological states, tissue fibroblasts, especially senescent fibroblasts, can display many pro-inflammatory and immunosuppressive properties and stimulate the activities of different immunosuppressive cells. In return, immunosuppressive cells, such as M2 macrophages and myeloid-derived suppressor cells (MDSC), evoke an excessive conversion of fibroblasts into myofibroblasts, thus aggravating the severity of tissue fibrosis. Single-cell transcriptome studies on fibroblasts isolated from aged tissues have confirmed that tissue fibroblasts express many genes coding for cytokines, chemokines, and complement factors, whereas they lose some fibrogenic properties. The versatile immune properties of fibroblasts and their close cooperation with immune cells indicate that tissue fibroblasts have a crucial role in the aging process and age-related diseases.


Subject(s)
Aging , Fibroblasts , Fibroblasts/immunology , Humans , Aging/immunology , Aging/physiology , Animals , Cellular Senescence/immunology , Cellular Senescence/physiology
18.
Immunol Cell Biol ; 102(6): 425-428, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38650445

ABSTRACT

Unraveling the complexities of T cell aging is essential for developing targeted interventions to enhance immune function in the elderly. This article for the Highlights of 2023 Series integrates recent findings published in 2023, offering a panoramic view of the current understanding of T cell aging and its implications.


Subject(s)
Cellular Senescence , T-Lymphocytes , Humans , T-Lymphocytes/immunology , Cellular Senescence/immunology , Animals , Aging/immunology , T-Cell Senescence
19.
J Mol Med (Berl) ; 102(6): 733-750, 2024 06.
Article in English | MEDLINE | ID: mdl-38600305

ABSTRACT

The accumulation of senescent cells within tissues is a hallmark of the aging process. Senescent cells are also commonly present in many age-related diseases and in the cancer microenvironment. The escape of abnormal cells from immune surveillance indicates that there is some defect in the function of cytotoxic immune cells, e.g., CD8+ T cells and natural killer (NK) cells. Recent studies have revealed that the expression of programmed death-ligand 1 (PD-L1) protein is abundantly increased in senescent cells. An increase in the amount of PD-L1 protein protects senescent cells from clearance by the PD-1 checkpoint receptor in cytotoxic immune cells. In fact, the activation of the PD-1 receptor suppresses the cytotoxic properties of CD8+ T and NK cells, promoting a state of immunosenescence. The inhibitory PD-1/PD-L1 checkpoint pathway acts in cooperation with immunosuppressive cells; for example, activation of PD-1 receptor can enhance the differentiation of regulatory T cells (Treg), myeloid-derived suppressor cells (MDSC), and M2 macrophages, whereas the cytokines secreted by immunosuppressive cells stimulate the expression of the immunosuppressive PD-L1 protein. Interestingly, many signaling pathways known to promote cellular senescence and the aging process are crucial stimulators of the expression of PD-L1 protein, e.g., epigenetic regulation, inflammatory mediators, mTOR-related signaling, cGAS-STING pathway, and AhR signaling. It seems that the inhibitory PD-1/PD-L1 immune checkpoint axis has a crucial role in the accumulation of senescent cells and thus it promotes the aging process in tissues. Thus, the blockade of the PD-1/PD-L1 checkpoint signaling might be a potential anti-aging senolytic therapy. KEY MESSAGES: Senescent cells accumulate within tissues during aging and age-related diseases. Senescent cells are able to escape immune surveillance by cytotoxic immune cells. Expression of programmed death-ligand 1 (PD-L1) markedly increases in senescent cells. Age-related signaling stimulates the expression of PD-L1 protein in senescent cells. Inhibitory PD-1/PD-L1 checkpoint pathway suppresses clearance of senescent cells.


Subject(s)
Aging , B7-H1 Antigen , Cellular Senescence , Programmed Cell Death 1 Receptor , Signal Transduction , Humans , B7-H1 Antigen/metabolism , B7-H1 Antigen/genetics , Programmed Cell Death 1 Receptor/metabolism , Aging/immunology , Aging/metabolism , Animals , Cellular Senescence/immunology
20.
Cancer Discov ; 14(7): 1302-1323, 2024 Jul 01.
Article in English | MEDLINE | ID: mdl-38683161

ABSTRACT

The tumor microenvironment (TME) profoundly influences tumorigenesis, with gene expression in the breast TME capable of predicting clinical outcomes. The TME is complex and includes distinct cancer-associated fibroblast (CAF) subtypes whose contribution to tumorigenesis remains unclear. Here, we identify a subset of myofibroblast CAFs (myCAF) that are senescent (senCAF) in mouse and human breast tumors. Utilizing the MMTV-PyMT;INK-ATTAC (INK) mouse model, we found that senCAF-secreted extracellular matrix specifically limits natural killer (NK) cell cytotoxicity to promote tumor growth. Genetic or pharmacologic senCAF elimination unleashes NK cell killing, restricting tumor growth. Finally, we show that senCAFs are present in HER2+, ER+, and triple-negative breast cancer and in ductal carcinoma in situ (DCIS) where they predict tumor recurrence. Together, these findings demonstrate that senCAFs are potently tumor promoting and raise the possibility that targeting them by senolytic therapy could restrain breast cancer development. Significance: senCAFs limit NK cell-mediated killing, thereby contributing to breast cancer progression. Thus, targeting senCAFs could be a clinically viable approach to limit tumor progression. See related article by Belle et al., p. 1324.


Subject(s)
Breast Neoplasms , Cancer-Associated Fibroblasts , Disease Progression , Tumor Microenvironment , Animals , Female , Mice , Humans , Breast Neoplasms/immunology , Breast Neoplasms/pathology , Breast Neoplasms/genetics , Cancer-Associated Fibroblasts/metabolism , Cancer-Associated Fibroblasts/immunology , Tumor Microenvironment/immunology , Killer Cells, Natural/immunology , Cellular Senescence/immunology
SELECTION OF CITATIONS
SEARCH DETAIL