Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
Cancer Sci ; 115(8): 2762-2773, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38802068

ABSTRACT

Senescent cells promote cancer development and progression through chronic inflammation caused by a senescence-associated secretory phenotype (SASP). Although various senotherapeutic strategies targeting senescent cells have been developed for the prevention and treatment of cancers, technology for the in vivo detection and evaluation of senescent cell accumulation has not yet been established. Here, we identified activatable fluorescent probes targeting dipeptidylpeptidase-4 (DPP4) as an effective probe for detecting senescent cells through an enzymatic activity-based screening of fluorescent probes. We also determined that these probes were highly, selectively, and rapidly activated in senescent cells during live cell imaging. Furthermore, we successfully visualized senescent cells in the organs of mice using DPP4-targeted probes. These results are expected to lead to the development of a diagnostic technology for noninvasively detecting senescent cells in vivo and could play a role in the application of DPP4 prodrugs for senotherapy.


Subject(s)
Cellular Senescence , Dipeptidyl Peptidase 4 , Fluorescent Dyes , Dipeptidyl Peptidase 4/metabolism , Dipeptidyl Peptidase 4/analysis , Animals , Mice , Humans
2.
Int J Mol Sci ; 24(3)2023 Jan 26.
Article in English | MEDLINE | ID: mdl-36768745

ABSTRACT

Senescent cells exhibit several typical features, including the senescence-associated secretory phenotype (SASP), promoting the secretion of various inflammatory proteins and small extracellular vesicles (EVs). SASP factors cause chronic inflammation, leading to age-related diseases. Recently, therapeutic strategies targeting senescent cells, known as senolytics, have gained attention; however, noninvasive methods to detect senescent cells in living organisms have not been established. Therefore, the goal of this study was to identify novel senescent markers using small EVs (sEVs). sEVs were isolated from young and senescent fibroblasts using three different methods, including size-exclusion chromatography, affinity column for phosphatidylserine, and immunoprecipitation using antibodies against tetraspanin proteins, followed by mass spectrometry. Principal component analysis revealed that the protein composition of sEVs released from senescent cells was significantly different from that of young cells. Importantly, we identified ATP6V0D1 and RTN4 as novel markers that are frequently upregulated in sEVs from senescent and progeria cells derived from patients with Werner syndrome. Furthermore, these two proteins were significantly enriched in sEVs from the serum of aged mice. This study supports the potential use of senescent markers from sEVs to detect the presence of senescent cells in vivo.


Subject(s)
Cellular Senescence , Extracellular Vesicles , Animals , Mice , Extracellular Vesicles/metabolism , Fibroblasts/metabolism
3.
Commun Biol ; 5(1): 1420, 2022 12 28.
Article in English | MEDLINE | ID: mdl-36577784

ABSTRACT

Cellular senescence caused by oncogenic stimuli is associated with the development of various age-related pathologies through the senescence-associated secretory phenotype (SASP). SASP is mediated by the activation of cytoplasmic nucleic acid sensors. However, the molecular mechanism underlying the accumulation of nucleotide ligands in senescent cells is unclear. In this study, we revealed that the expression of RNaseH2A, which removes ribonucleoside monophosphates (rNMPs) from the genome, is regulated by E2F transcription factors, and it decreases during cellular senescence. Residual rNMPs cause genomic DNA fragmentation and aberrant activation of cytoplasmic nucleic acid sensors, thereby provoking subsequent SASP factor gene expression in senescent cells. In addition, RNaseH2A expression was significantly decreased in aged mouse tissues and cells from individuals with Werner syndrome. Furthermore, RNaseH2A degradation using the auxin-inducible degron system induced the accumulation of nucleotide ligands and induction of certain tumourigenic SASP-like factors, promoting the metastatic properties of colorectal cancer cells. Our results indicate that RNaseH2A downregulation provokes SASP through nucleotide ligand accumulation, which likely contributes to the pathological features of senescent, progeroid, and cancer cells.


Subject(s)
DNA , Neoplasms , Animals , Mice , Cellular Senescence/genetics , DNA Fragmentation , Down-Regulation , Gene Expression , Genomics , Ligands , Neoplasms/genetics , Neoplasms/metabolism , Nucleotides , Phenotype , Humans , Cell Line
4.
Nat Commun ; 13(1): 4157, 2022 07 18.
Article in English | MEDLINE | ID: mdl-35851277

ABSTRACT

Cellular senescence and cell competition are important tumor suppression mechanisms that restrain cells with oncogenic mutations at the initial stage of cancer development. However, the link between cellular senescence and cell competition remains unclear. Senescent cells accumulated during the in vivo aging process contribute toward age-related cancers via the development of senescence-associated secretory phenotype (SASP). Here, we report that hepatocyte growth factor (HGF), a SASP factor, inhibits apical extrusion and promotes basal protrusion of Ras-mutated cells in the cell competition assay. Additionally, cellular senescence induced by a high-fat diet promotes the survival of cells with oncogenic mutations, whereas crizotinib, an inhibitor of HGF signaling, provokes the removal of mutated cells from mouse livers and intestines. Our study provides evidence that cellular senescence inhibits cell competition-mediated elimination of oncogenic cells through HGF signaling, suggesting that it may lead to cancer incidence during aging.


Subject(s)
Hepatocyte Growth Factor , Neoplasms , Animals , Carcinogenesis , Cell Competition , Cellular Senescence/genetics , Hepatocyte Growth Factor/genetics , Mice , Oncogenes/genetics
5.
Cells ; 11(12)2022 06 17.
Article in English | MEDLINE | ID: mdl-35741087

ABSTRACT

Cancer cells exhibit the unique characteristics of high proliferation and aberrant DNA damage response, which prevents cancer therapy from effectively eliminating them. The machinery required for telomere maintenance, such as telomerase and the alternative lengthening of telomeres (ALT), enables cancer cells to proliferate indefinitely. In addition, the molecules in this system are involved in noncanonical pro-tumorigenic functions. Of these, the function of the cyclic GMP-AMP synthase (cGAS)-stimulator of interferon genes (STING) pathway, which contains telomere-related molecules, is a well-known contributor to the tumor microenvironment (TME). This review summarizes the current knowledge of the role of telomerase and ALT in cancer regulation, with emphasis on their noncanonical roles beyond telomere maintenance. The components of the cGAS-STING pathway are summarized with respect to intercell communication in the TME. Elucidating the underlying functional connection between telomere-related molecules and TME regulation is important for the development of cancer therapeutics that target cancer-specific pathways in different contexts. Finally, strategies for designing new cancer therapies that target cancer cells and the TME are discussed.


Subject(s)
Neoplasms , Telomerase , Humans , Membrane Proteins/metabolism , Neoplasms/metabolism , Nucleotidyltransferases/metabolism , Telomerase/metabolism , Telomere/metabolism , Tumor Microenvironment
6.
Sci Immunol ; 7(72): eabl7209, 2022 06 24.
Article in English | MEDLINE | ID: mdl-35749514

ABSTRACT

Long-term senescent cells exhibit a secretome termed the senescence-associated secretory phenotype (SASP). Although the mechanisms of SASP factor induction have been intensively studied, the release mechanism and how SASP factors influence tumorigenesis in the biological context remain unclear. In this study, using a mouse model of obesity-induced hepatocellular carcinoma (HCC), we identified the release mechanism of SASP factors, which include interleukin-1ß (IL-1ß)- and IL-1ß-dependent IL-33, from senescent hepatic stellate cells (HSCs) via gasdermin D (GSDMD) amino-terminal-mediated pore. We found that IL-33 was highly induced in senescent HSCs in an IL-1ß-dependent manner in the tumor microenvironment. The release of both IL-33 and IL-1ß was triggered by lipoteichoic acid (LTA), a cell wall component of gut microbiota that was transferred and accumulated in the liver tissue of high-fat diet-fed mice, and the release of these factors was mediated through cell membrane pores formed by the GSDMD amino terminus, which was cleaved by LTA-induced caspase-11. We demonstrated that IL-33 release from HSCs promoted HCC development via the activation of ST2-positive Treg cells in the liver tumor microenvironment. The accumulation of GSDMD amino terminus was also detected in HSCs from human NASH-associated HCC patients, suggesting that similar mechanism could be involved in a certain type of human HCC. These results uncover a release mechanism for SASP factors from sensitized senescent HSCs in the tumor microenvironment, thereby facilitating obesity-associated HCC progression. Furthermore, our findings highlight the therapeutic potential of inhibitors of GSDMD-mediated pore formation for HCC treatment.


Subject(s)
Carcinoma, Hepatocellular , Liver Neoplasms , Phosphate-Binding Proteins/metabolism , Pore Forming Cytotoxic Proteins/metabolism , Animals , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/pathology , Cellular Senescence , Hepatic Stellate Cells/metabolism , Hepatic Stellate Cells/pathology , Humans , Interleukin-33/metabolism , Mice , Obesity/complications , Obesity/metabolism , Tumor Microenvironment
7.
Proc Natl Acad Sci U S A ; 118(35)2021 08 31.
Article in English | MEDLINE | ID: mdl-34426493

ABSTRACT

Cellular senescence causes a dramatic alteration of chromatin organization and changes the gene expression profile of proinflammatory factors, thereby contributing to various age-related pathologies through the senescence-associated secretory phenotype (SASP). Chromatin organization and global gene expression are maintained by the CCCTC-binding factor (CTCF); however, the molecular mechanism underlying CTCF regulation and its association with SASP gene expression remains unclear. We discovered that noncoding RNA (ncRNA) derived from normally silenced pericentromeric repetitive sequences directly impairs the DNA binding of CTCF. This CTCF disturbance increases the accessibility of chromatin and activates the transcription of SASP-like inflammatory genes, promoting malignant transformation. Notably, pericentromeric ncRNA was transferred into surrounding cells via small extracellular vesicles acting as a tumorigenic SASP factor. Because CTCF blocks the expression of pericentromeric ncRNA in young cells, the down-regulation of CTCF during cellular senescence triggers the up-regulation of this ncRNA and SASP-related inflammatory gene expression. In this study, we show that pericentromeric ncRNA provokes chromosomal alteration by inhibiting CTCF, leading to a SASP-like inflammatory response in a cell-autonomous and non-cell-autonomous manner and thus may contribute to the risk of tumorigenesis during aging.


Subject(s)
Aging/genetics , DNA-Binding Proteins/metabolism , DNA/metabolism , Inflammation/genetics , RNA, Untranslated/physiology , Senescence-Associated Secretory Phenotype/genetics , Animals , Cellular Senescence/genetics , Centromere , DNA, Neoplasm/metabolism , Female , Gene Expression Regulation , HEK293 Cells , Humans , Mice , Mice, Inbred BALB C , Neoplasms , Protein Binding/genetics
8.
Int J Mol Sci ; 21(10)2020 May 25.
Article in English | MEDLINE | ID: mdl-32466233

ABSTRACT

DNA damage, caused by various oncogenic stresses, can induce cell death or cellular senescence as an important tumor suppressor mechanism. Senescent cells display the features of a senescence-associated secretory phenotype (SASP), secreting inflammatory proteins into surrounding tissues, and contributing to various age-related pathologies. In addition to this inflammatory protein secretion, the release of extracellular vesicles (EVs) is also upregulated in senescent cells. However, the molecular mechanism underlying this phenomenon remains unclear. Here, we show that DNA damage activates the ceramide synthetic pathway, via the downregulation of sphingomyelin synthase 2 (SMS2) and the upregulation of neutral sphingomyelinase 2 (nSMase2), leading to an increase in senescence-associated EV (SA-EV) biogenesis. The EV biogenesis pathway, together with the autophagy-mediated degradation pathway, functions to block apoptosis by removing cytoplasmic DNA fragments derived from chromosomal DNA or bacterial infections. Our data suggest that this SA-EV pathway may play a prominent role in cellular homeostasis, particularly in senescent cells. In summary, DNA damage provokes SA-EV release by activating the ceramide pathway to protect cells from excessive inflammatory responses.


Subject(s)
Cellular Senescence , Ceramides/metabolism , DNA Damage , Extracellular Vesicles/metabolism , Animals , Autophagy , Cell Line , Cells, Cultured , Humans , Male , Mice , Mice, Inbred ICR , Sphingomyelin Phosphodiesterase/genetics , Sphingomyelin Phosphodiesterase/metabolism , Transferases (Other Substituted Phosphate Groups)/genetics , Transferases (Other Substituted Phosphate Groups)/metabolism
9.
Nat Commun ; 11(1): 1935, 2020 04 22.
Article in English | MEDLINE | ID: mdl-32321921

ABSTRACT

Although cellular senescence acts primarily as a tumour suppression mechanism, the accumulation of senescent cells in vivo eventually exerts deleterious side effects through inflammatory/tumour-promoting factor secretion. Thus, the development of new drugs that cause the specific elimination of senescent cells, termed senolysis, is anticipated. Here, by an unbiased high-throughput screening of chemical compounds and a bio-functional analysis, we identify BET family protein degrader (BETd) as a promising senolytic drug. BETd provokes senolysis through two independent but integrated pathways; the attenuation of non-homologous end joining (NHEJ), and the up-regulation of autophagic gene expression. BETd treatment eliminates senescent hepatic stellate cells in obese mouse livers, accompanied by the reduction of liver cancer development. Furthermore, the elimination of chemotherapy-induced senescent cells by BETd increases the efficacy of chemotherapy against xenograft tumours in immunocompromised mice. These results reveal the vulnerability of senescent cells and open up possibilities for its control.


Subject(s)
Antineoplastic Agents/administration & dosage , Autophagy/drug effects , Cellular Senescence/drug effects , DNA End-Joining Repair/drug effects , Neoplasms/physiopathology , Animals , Cell Cycle Proteins/antagonists & inhibitors , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Cell Line, Tumor , Humans , Mice , Mice, Inbred C57BL , Mice, Nude , Neoplasms/drug therapy , Neoplasms/genetics , Neoplasms/metabolism , Transcription Factors/antagonists & inhibitors , Transcription Factors/genetics , Transcription Factors/metabolism
10.
Cancer Sci ; 111(2): 304-311, 2020 Feb.
Article in English | MEDLINE | ID: mdl-31799772

ABSTRACT

Cellular senescence is historically regarded as a tumor suppression mechanism to prevent damaged cells from aberrant proliferation in benign and premalignant tumors. However, recent findings have suggested that senescent cells contribute to tumorigenesis and age-associated pathologies through the senescence-associated secretory phenotype (SASP). Therefore, to control age-associated cancer, it is important to understand the molecular mechanisms of the SASP in the cancer microenvironment. New findings have suggested that the cyclic GMP-AMP synthase (cGAS)-stimulator of interferon genes (STING) signaling pathway, a critical indicator of innate immune response, triggers the SASP in response to accumulation of cytoplasmic DNA (cytoplasmic chromatin fragments, mtDNA and cDNA) in senescent cells. Notably, the cGAS-STING signaling pathway promotes or inhibits tumorigenesis depending on the biological context in vivo, indicating that it may be a potential therapeutic target for cancer. Herein, we review the regulatory machinery and biological function of the SASP via the cGAS-STING signaling pathway in cancer.


Subject(s)
Membrane Proteins/metabolism , Neoplasms/metabolism , Nucleotidyltransferases/metabolism , Signal Transduction , Cellular Senescence , Cytoplasm/genetics , DNA Damage , Gene Expression Regulation, Neoplastic , Humans , Immunity, Innate , Neoplasms/genetics , Phenotype , Tumor Microenvironment
11.
Nat Commun ; 9(1): 1249, 2018 03 28.
Article in English | MEDLINE | ID: mdl-29593264

ABSTRACT

Accumulating evidence indicates that the senescence-associated secretory phenotype (SASP) contributes to many aspects of physiology and disease. Thus, controlling the SASP will have tremendous impacts on our health. However, our understanding of SASP regulation is far from complete. Here, we show that cytoplasmic accumulation of nuclear DNA plays key roles in the onset of SASP. Although both DNase2 and TREX1 rapidly remove the cytoplasmic DNA fragments emanating from the nucleus in pre-senescent cells, the expression of these DNases is downregulated in senescent cells, resulting in the cytoplasmic accumulation of nuclear DNA. This causes the aberrant activation of cGAS-STING cytoplasmic DNA sensors, provoking SASP through induction of interferon-ß. Notably, the blockage of this pathway prevents SASP in senescent hepatic stellate cells, accompanied by a decline of obesity-associated hepatocellular carcinoma development in mice. These findings provide valuable new insights into the roles and mechanisms of SASP and possibilities for their control.


Subject(s)
Cellular Senescence , Cytoplasm/enzymology , Deoxyribonucleases/metabolism , Down-Regulation , Animals , Carcinoma, Hepatocellular/enzymology , Cell Line , DNA Damage , DNA, Single-Stranded/metabolism , Exodeoxyribonucleases/metabolism , Hepatic Stellate Cells/enzymology , Humans , Interferon-beta/metabolism , Liver/enzymology , Liver Neoplasms/enzymology , Male , Mice , Mice, Inbred C57BL , Microscopy, Fluorescence , Phenotype , Phosphoproteins/metabolism , Phosphorylation , RNA Interference
12.
Cancer Discov ; 7(5): 522-538, 2017 05.
Article in English | MEDLINE | ID: mdl-28202625

ABSTRACT

Obesity increases the risk of cancers, including hepatocellular carcinomas (HCC). However, the precise molecular mechanisms through which obesity promotes HCC development are still unclear. Recent studies have shown that gut microbiota may influence liver diseases by transferring its metabolites and components. Here, we show that the hepatic translocation of obesity-induced lipoteichoic acid (LTA), a Gram-positive gut microbial component, promotes HCC development by creating a tumor-promoting microenvironment. LTA enhances the senescence-associated secretory phenotype (SASP) of hepatic stellate cells (HSC) collaboratively with an obesity-induced gut microbial metabolite, deoxycholic acid, to upregulate the expression of SASP factors and COX2 through Toll-like receptor 2. Interestingly, COX2-mediated prostaglandin E2 (PGE2) production suppresses the antitumor immunity through a PTGER4 receptor, thereby contributing to HCC progression. Moreover, COX2 overexpression and excess PGE2 production were detected in HSCs in human HCCs with noncirrhotic, nonalcoholic steatohepatitis (NASH), indicating that a similar mechanism could function in humans.Significance: We showed the importance of the gut-liver axis in obesity-associated HCC. The gut microbiota-driven COX2 pathway produced the lipid mediator PGE2 in senescent HSCs in the tumor microenvironment, which plays a pivotal role in suppressing antitumor immunity, suggesting that PGE2 and its receptor may be novel therapeutic targets for noncirrhotic NASH-associated HCC. Cancer Discov; 7(5); 522-38. ©2017 AACR.This article is highlighted in the In This Issue feature, p. 443.


Subject(s)
Carcinoma, Hepatocellular/metabolism , Dinoprostone/metabolism , Gastrointestinal Microbiome/physiology , Liver Neoplasms/metabolism , Obesity/complications , Animals , Carcinoma, Hepatocellular/immunology , Carcinoma, Hepatocellular/microbiology , Female , Humans , Lipopolysaccharides/metabolism , Liver Neoplasms/immunology , Liver Neoplasms/microbiology , Male , Mice, Inbred C57BL , Obesity/microbiology , Teichoic Acids/metabolism , Tumor Escape/immunology , Tumor Microenvironment/immunology
13.
Nature ; 499(7456): 97-101, 2013 Jul 04.
Article in English | MEDLINE | ID: mdl-23803760

ABSTRACT

Obesity has become more prevalent in most developed countries over the past few decades, and is increasingly recognized as a major risk factor for several common types of cancer. As the worldwide obesity epidemic has shown no signs of abating, better understanding of the mechanisms underlying obesity-associated cancer is urgently needed. Although several events were proposed to be involved in obesity-associated cancer, the exact molecular mechanisms that integrate these events have remained largely unclear. Here we show that senescence-associated secretory phenotype (SASP) has crucial roles in promoting obesity-associated hepatocellular carcinoma (HCC) development in mice. Dietary or genetic obesity induces alterations of gut microbiota, thereby increasing the levels of deoxycholic acid (DCA), a gut bacterial metabolite known to cause DNA damage. The enterohepatic circulation of DCA provokes SASP phenotype in hepatic stellate cells (HSCs), which in turn secretes various inflammatory and tumour-promoting factors in the liver, thus facilitating HCC development in mice after exposure to chemical carcinogen. Notably, blocking DCA production or reducing gut bacteria efficiently prevents HCC development in obese mice. Similar results were also observed in mice lacking an SASP inducer or depleted of senescent HSCs, indicating that the DCA-SASP axis in HSCs has key roles in obesity-associated HCC development. Moreover, signs of SASP were also observed in the HSCs in the area of HCC arising in patients with non-alcoholic steatohepatitis, indicating that a similar pathway may contribute to at least certain aspects of obesity-associated HCC development in humans as well. These findings provide valuable new insights into the development of obesity-associated cancer and open up new possibilities for its control.


Subject(s)
Cellular Senescence , Deoxycholic Acid/metabolism , Gastrointestinal Tract/metabolism , Gastrointestinal Tract/microbiology , Hepatic Stellate Cells/metabolism , Liver Neoplasms/metabolism , Obesity/metabolism , Animals , Anti-Bacterial Agents/pharmacology , Bacteria/metabolism , Bile Acids and Salts/metabolism , Carcinoma, Hepatocellular/complications , Carcinoma, Hepatocellular/etiology , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/prevention & control , Cells, Cultured , Cellular Senescence/drug effects , Cytokines/metabolism , DNA Damage/drug effects , Deoxycholic Acid/blood , Dietary Fats/adverse effects , Dietary Fats/pharmacology , Disease Models, Animal , Fatty Liver/complications , Fatty Liver/pathology , Gastrointestinal Tract/drug effects , Hepatic Stellate Cells/cytology , Hepatic Stellate Cells/drug effects , Humans , Interleukin-1beta/deficiency , Liver Neoplasms/complications , Liver Neoplasms/etiology , Liver Neoplasms/prevention & control , Male , Mice , Mice, Inbred C57BL , Non-alcoholic Fatty Liver Disease , Obesity/chemically induced , Phenotype , Risk Factors
SELECTION OF CITATIONS
SEARCH DETAIL