Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 683
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.
Front Immunol ; 15: 1358725, 2024.
Article in English | MEDLINE | ID: mdl-39148728

ABSTRACT

Introduction: The immunological characteristics that could protect children with coronavirus disease 2019 (COVID-19) from severe or fatal illnesses have not been fully understood yet. Methods: Here, we performed single-cell RNA sequencing (scRNA-seq) analysis on peripheral blood samples of 15 children (8 with COVID-19) and compared them to 18 adults (13 with COVID-19). Results: The child-adult integrated single cell data indicated that children with the disease presented a restrained response to type I interferon in most of the major immune cell types, along with suppression of upstream interferon regulatory factor and toll-like receptor expression in monocytes, which was confirmed by in vitro interferon stimulation assays. Unlike adult patients, children with COVID-19 showed lower frequencies of activated proinflammatory CD14+ monocytes, possibly explaining the rareness of cytokine storm in them. Notably, natural killer (NK) cells in pediatric patients displayed potent cytotoxicity with a rich expression of cytotoxic molecules and upregulated cytotoxic pathways, whereas the cellular senescence, along with the Notch signaling pathway, was significantly downregulated in NK cells, all suggesting more robust cytotoxicity in NK cells of children than adult patients that was further confirmed by CD107a degranulation assays. Lastly, a modest adaptive immune response was evident with more naïve T cells but less activated and proliferated T cells while less naïve B cells but more activated B cells in children over adult patients. Conclusion: Conclusively, this preliminary study revealed distinct cell frequency and activation status of major immune cell types, particularly more robust NK cell cytotoxicity in PBMC that might help protect children from severe COVID-19.


Subject(s)
COVID-19 , Killer Cells, Natural , SARS-CoV-2 , Single-Cell Analysis , Humans , COVID-19/immunology , Child , Adult , SARS-CoV-2/immunology , Male , Female , Killer Cells, Natural/immunology , Child, Preschool , Adolescent , Monocytes/immunology , Monocytes/metabolism , Middle Aged , Adaptive Immunity , Cytotoxicity, Immunologic , Young Adult , Interferon Type I/immunology , Cellular Senescence/immunology
3.
PLoS Biol ; 22(8): e3002380, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39137219

ABSTRACT

Quantifying the kinetics with which memory T cell populations are generated and maintained is essential for identifying the determinants of the duration of immunity. The quality and persistence of circulating CD4 effector memory (TEM) and central memory (TCM) T cells in mice appear to shift with age, but it is unclear whether these changes are driven by the aging host environment, by cell age effects, or both. Here, we address these issues by combining DNA labelling methods, established fate-mapping systems, a novel reporter mouse strain, and mathematical models. Together, these allow us to quantify the dynamics of both young and established circulating memory CD4 T cell subsets, within both young and old mice. We show that that these cells and their descendents become more persistent the longer they reside within the TCM and TEM pools. This behaviour may limit memory CD4 T cell diversity by skewing TCR repertoires towards clones generated early in life, but may also compensate for functional defects in new memory cells generated in old age.


Subject(s)
Aging , CD4-Positive T-Lymphocytes , Cellular Senescence , Memory T Cells , Animals , Memory T Cells/immunology , CD4-Positive T-Lymphocytes/immunology , Mice , Cellular Senescence/immunology , Aging/immunology , Aging/physiology , Mice, Inbred C57BL , Immunologic Memory
4.
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
5.
Nat Cell Biol ; 26(8): 1336-1345, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39103548

ABSTRACT

The accumulation of senescent cells promotes ageing and age-related diseases, but molecular mechanisms that senescent cells use to evade immune clearance and accumulate in tissues remain to be elucidated. Here we report that p16-positive senescent cells upregulate the immune checkpoint protein programmed death-ligand 1 (PD-L1) to accumulate in ageing and chronic inflammation. We show that p16-mediated inhibition of cell cycle kinases CDK4/6 induces PD-L1 stability in senescent cells via downregulation of its ubiquitin-dependent degradation. p16-expressing senescent alveolar macrophages elevate PD-L1 to promote an immunosuppressive environment that can contribute to an increased burden of senescent cells. Treatment with activating anti-PD-L1 antibodies engaging Fcγ receptors on effector cells leads to the elimination of PD-L1 and p16-positive cells. Our study uncovers a molecular mechanism of p16-dependent regulation of PD-L1 protein stability in senescent cells and reveals the potential of targeting PD-L1 to improve immunosurveillance of senescent cells and ameliorate senescence-associated inflammation.


Subject(s)
B7-H1 Antigen , Cellular Senescence , Cyclin-Dependent Kinase Inhibitor p16 , Protein Stability , Cellular Senescence/immunology , B7-H1 Antigen/metabolism , B7-H1 Antigen/genetics , Cyclin-Dependent Kinase Inhibitor p16/metabolism , Cyclin-Dependent Kinase Inhibitor p16/genetics , Animals , Humans , Cyclin-Dependent Kinase 4/metabolism , Cyclin-Dependent Kinase 4/genetics , Immunologic Surveillance , Mice, Inbred C57BL , Cyclin-Dependent Kinase 6/metabolism , Cyclin-Dependent Kinase 6/genetics , Mice , Proteolysis , Receptors, IgG/metabolism , Inflammation/immunology , Inflammation/metabolism , Inflammation/pathology , Inflammation/genetics
6.
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
7.
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
8.
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
9.
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
10.
Biogerontology ; 25(5): 749-773, 2024 Oct.
Article in English | MEDLINE | ID: mdl-38954358

ABSTRACT

The accumulation of pro-inflammatory senescent cells within tissues is a common hallmark of the aging process and many age-related diseases. This modification has been called the senescence-associated secretory phenotype (SASP) and observed in cultured cells and in cells isolated from aged tissues. Currently, there is a debate whether the accumulation of senescent cells within tissues should be attributed to increased generation of senescent cells or to a defect in their elimination from aging tissues. Emerging studies have revealed that senescent cells display an increased expression of several inhibitory immune checkpoint ligands, especially those of the programmed cell death protein-1 (PD-1) ligand-1 (PD-L1) proteins. It is known that the PD-L1 ligands, especially those of cancer cells, target the PD-1 receptor of cytotoxic CD8+ T and natural killer (NK) cells disturbing their functions, e.g., evoking a decline in their cytotoxic activity and promoting their exhaustion and even apoptosis. An increase in the level of the PD-L1 protein in senescent cells was able to suppress their immune surveillance and inhibit their elimination by cytotoxic CD8+ T and NK cells. Senescent cells are known to express ligands for several inhibitory immune checkpoint receptors, i.e., PD-1, LILRB4, NKG2A, TIM-3, and SIRPα receptors. Here, I will briefly describe those pathways and examine whether these inhibitory checkpoints could be involved in the immune evasion of senescent cells with aging and age-related diseases. It seems plausible that an enhanced inhibitory checkpoint signaling can prevent the elimination of senescent cells from tissues and thus promote the aging process.


Subject(s)
Aging , Cellular Senescence , Humans , Aging/immunology , Aging/metabolism , Cellular Senescence/immunology , Cellular Senescence/physiology , Animals , Immune Checkpoint Proteins/metabolism , Senescence-Associated Secretory Phenotype , B7-H1 Antigen/metabolism , Immunologic Surveillance
11.
Semin Cancer Biol ; 104-105: 1-15, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39032717

ABSTRACT

Ovarian cancer is a common gynecological malignancy, and its treatment remains challenging. Although ovarian cancer may respond to immunotherapy because of endogenous immunity at the molecular or T cell level, immunotherapy has so far not had the desired effect. The functional status of preexisting T cells is an indispensable determinant of powerful antitumor immunity and immunotherapy. T cell exhaustion and senescence are two crucial states of T cell dysfunction, which share some overlapping phenotypic and functional features, but each status possesses unique molecular and developmental signatures. It has been widely accepted that exhaustion and senescence of T cells are important strategies for cancer cells to evade immunosurveillance and maintain the immunosuppressive microenvironment. Herein, this review summarizes the phenotypic and functional features of exhaust and senescent T cells, and describes the key drivers of the two T cell dysfunctional states in the tumor microenvironment and their functional roles in ovarian cancer. Furthermore, we present a summary of the molecular machinery and signaling pathways governing T cell exhaustion and senescence. Possible strategies that can prevent and/or reverse T cell dysfunction are also explored. An in-depth understanding of exhausted and senescent T cells will provide novel strategies to enhance immunotherapy of ovarian cancer through redirecting tumor-specific T cells away from a dysfunctional developmental trajectory.


Subject(s)
Cellular Senescence , Immunotherapy , Ovarian Neoplasms , T-Lymphocytes , Tumor Microenvironment , Humans , Female , Ovarian Neoplasms/immunology , Ovarian Neoplasms/therapy , Ovarian Neoplasms/pathology , Immunotherapy/methods , Cellular Senescence/immunology , Tumor Microenvironment/immunology , T-Lymphocytes/immunology , Animals , T-Cell Exhaustion
12.
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
13.
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
14.
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
15.
Nat Aging ; 4(8): 1053-1063, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38867059

ABSTRACT

Chronological aging correlates with epigenetic modifications at specific loci, calibrated to species lifespan. Such 'epigenetic clocks' appear conserved among mammals, but whether they are cell autonomous and restricted by maximal organismal lifespan remains unknown. We used a multilifetime murine model of repeat vaccination and memory T cell transplantation to test whether epigenetic aging tracks with cellular replication and if such clocks continue 'counting' beyond species lifespan. Here we found that memory T cell epigenetic clocks tick independently of host age and continue through four lifetimes. Instead of recording chronological time, T cells recorded proliferative experience through modification of cell cycle regulatory genes. Applying this epigenetic profile across a range of human T cell contexts, we found that naive T cells appeared 'young' regardless of organism age, while in pediatric patients, T cell acute lymphoblastic leukemia appeared to have epigenetically aged for up to 200 years. Thus, T cell epigenetic clocks measure replicative history and can continue to accumulate well-beyond organismal lifespan.


Subject(s)
Cellular Senescence , Epigenesis, Genetic , Animals , Humans , Mice , Cellular Senescence/genetics , Cellular Senescence/immunology , Aging/immunology , Aging/genetics , T-Lymphocytes/immunology , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/genetics , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/immunology , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/pathology , Memory T Cells/immunology , Memory T Cells/metabolism , Mice, Inbred C57BL , Male , T-Cell Senescence
16.
Adv Sci (Weinh) ; 11(30): e2400630, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38867377

ABSTRACT

Senescent cancer cells are endowed with high immunogenic potential that has been leveraged to elicit antitumor immunity and potentially complement anticancer therapies. However, the efficacy of live senescent cancer cell-based vaccination is limited by interference from immunosuppressive senescence-associated secretory phenotype and pro-tumorigenic capacity of senescent cells. Here, a senescent cancer cell-based nanovaccine with strong immunogenicity and favorable potential for immunotherapy is reported. The biomimetic nanovaccine integrating a senescent cancer cell membrane-coated nanoadjuvant outperforms living senescent cancer cells in enhancing dendritic cells (DCs) internalization, improving lymph node targeting, and enhancing immune responses. In contrast to nanovaccines generated from immunogenic cell death-induced tumor cells, senescent nanovaccines facilitate DC maturation, eliciting superior antitumor protection and improving therapeutic outcomes in melanoma-challenged mice with fewer side effects when combined with αPD-1. The study suggests a versatile biomanufacturing approach to maximize immunogenic potential and minimize adverse effects of senescent cancer cell-based vaccination and advances the design of biomimetic nanovaccines for cancer immunotherapy.


Subject(s)
Cancer Vaccines , Cellular Senescence , Immunotherapy , Animals , Mice , Cancer Vaccines/immunology , Immunotherapy/methods , Cellular Senescence/immunology , Disease Models, Animal , Biomimetics/methods , Mice, Inbred C57BL , Cell Membrane/immunology , Nanoparticles , Humans , Cell Line, Tumor , Female , Biomimetic Materials , Dendritic Cells/immunology , Nanovaccines
17.
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
18.
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
19.
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
20.
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
SELECTION OF CITATIONS
SEARCH DETAIL