1
|
Torres G, Salladay-Perez IA, Dhingra A, Covarrubias AJ. Genetic origins, regulators, and biomarkers of cellular senescence. Trends Genet 2024; 40:1018-1031. [PMID: 39341687 PMCID: PMC11717094 DOI: 10.1016/j.tig.2024.08.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2024] [Revised: 08/18/2024] [Accepted: 08/21/2024] [Indexed: 10/01/2024]
Abstract
This review comprehensively examines the molecular biology and genetic origins of cellular senescence. We focus on various cellular stressors and pathways leading to senescence, including recent advances in the understanding of the genetic influences driving senescence, such as telomere attrition, chemotherapy-induced DNA damage, pathogens, oncogene activation, and cellular and metabolic stress. This review also highlights the complex interplay of various signaling and metabolic pathways involved in cellular senescence and provides insights into potential therapeutic targets for aging-related diseases. Furthermore, this review outlines future research directions to deepen our understanding of senescence biology and develop effective interventions targeting senescent cells (SnCs).
Collapse
Affiliation(s)
- Grasiela Torres
- Department of Microbiology, Immunology, and Molecular Genetics, University of California, Los Angeles, Los Angeles, CA, USA; Molecular Biology Interdepartmental Doctoral Program, University of California, Los Angeles, Los Angeles, CA, USA; Molecular Biology Institute, University of California, Los Angeles, Los Angeles, CA, USA
| | - Ivan A Salladay-Perez
- Department of Microbiology, Immunology, and Molecular Genetics, University of California, Los Angeles, Los Angeles, CA, USA; Molecular Biology Interdepartmental Doctoral Program, University of California, Los Angeles, Los Angeles, CA, USA; Molecular Biology Institute, University of California, Los Angeles, Los Angeles, CA, USA
| | - Anika Dhingra
- Department of Microbiology, Immunology, and Molecular Genetics, University of California, Los Angeles, Los Angeles, CA, USA
| | - Anthony J Covarrubias
- Department of Microbiology, Immunology, and Molecular Genetics, University of California, Los Angeles, Los Angeles, CA, USA; Molecular Biology Institute, University of California, Los Angeles, Los Angeles, CA, USA.
| |
Collapse
|
2
|
Devillers R, Dos Santos A, Destombes Q, Laplante M, Elowe S. Recent insights into the causes and consequences of chromosome mis-segregation. Oncogene 2024; 43:3139-3150. [PMID: 39278989 DOI: 10.1038/s41388-024-03163-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2024] [Revised: 09/04/2024] [Accepted: 09/06/2024] [Indexed: 09/18/2024]
Abstract
Mitotic cells face the challenging task of ensuring accurate and equal segregation of their duplicated, condensed chromosomes between the nascent daughter cells. Errors in the process result in chromosome missegregation, a significant consequence of which is the emergence of aneuploidy-characterized by an imbalance in chromosome number-and the associated phenomenon of chromosome instability (CIN). Aneuploidy and CIN are common features of cancer, which leverages them to promote genome heterogeneity and plasticity, thereby facilitating rapid tumor evolution. Recent research has provided insights into how mitotic errors shape cancer genomes by inducing both numerical and structural chromosomal changes that drive tumor initiation and progression. In this review, we survey recent findings regarding the mitotic causes and consequences of aneuploidy. We discuss new findings into the types of chromosome segregation errors that lead to aneuploidy and novel pathways that protect genome integrity during mitosis. Finally, we describe new developments in our understanding of the immediate consequences of chromosome mis-segregation on the genome stability of daughter cells.
Collapse
Affiliation(s)
- Romain Devillers
- Centre de Recherche sur le Cancer, CHU de Québec-Université Laval, Québec City, QC, Canada
- Centre de recherche du Centre Hospitalier Universitaire (CHU) de Québec-Université Laval, Axe de reproduction, santé de la mère et de l'enfant, Québec, QC, Canada
- Centre de recherche de l'Institut universitaire de cardiologie et de pneumologie de Québec (CRIUCPQ), Faculté de Médecine, Université Laval, Québec, QC, Canada
- Regroupement Québécois de Recherche sur la Fonction, L'ingénierie et les Applications des Protéines, Québec, Canada
| | - Alexsandro Dos Santos
- Centre de Recherche sur le Cancer, CHU de Québec-Université Laval, Québec City, QC, Canada
- Centre de recherche du Centre Hospitalier Universitaire (CHU) de Québec-Université Laval, Axe de reproduction, santé de la mère et de l'enfant, Québec, QC, Canada
- Regroupement Québécois de Recherche sur la Fonction, L'ingénierie et les Applications des Protéines, Québec, Canada
| | - Quentin Destombes
- Centre de Recherche sur le Cancer, CHU de Québec-Université Laval, Québec City, QC, Canada
- Centre de recherche du Centre Hospitalier Universitaire (CHU) de Québec-Université Laval, Axe de reproduction, santé de la mère et de l'enfant, Québec, QC, Canada
- Regroupement Québécois de Recherche sur la Fonction, L'ingénierie et les Applications des Protéines, Québec, Canada
| | - Mathieu Laplante
- Centre de Recherche sur le Cancer, CHU de Québec-Université Laval, Québec City, QC, Canada
- Centre de recherche de l'Institut universitaire de cardiologie et de pneumologie de Québec (CRIUCPQ), Faculté de Médecine, Université Laval, Québec, QC, Canada
| | - Sabine Elowe
- Centre de Recherche sur le Cancer, CHU de Québec-Université Laval, Québec City, QC, Canada.
- Centre de recherche du Centre Hospitalier Universitaire (CHU) de Québec-Université Laval, Axe de reproduction, santé de la mère et de l'enfant, Québec, QC, Canada.
- Regroupement Québécois de Recherche sur la Fonction, L'ingénierie et les Applications des Protéines, Québec, Canada.
- Département de Pédiatrie, Faculté de Médecine, Université Laval, Québec City, QC, Canada.
| |
Collapse
|
3
|
Loaiza-Moss J, Braun U, Leitges M. Transcriptome Profiling of Mouse Embryonic Fibroblast Spontaneous Immortalization: A Comparative Analysis. Int J Mol Sci 2024; 25:8116. [PMID: 39125691 PMCID: PMC11311763 DOI: 10.3390/ijms25158116] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2024] [Revised: 07/17/2024] [Accepted: 07/23/2024] [Indexed: 08/12/2024] Open
Abstract
Cell immortalization, a hallmark of cancer development, is a process that cells can undergo on their path to carcinogenesis. Spontaneously immortalized mouse embryonic fibroblasts (MEFs) have been used for decades; however, changes in the global transcriptome during this process have been poorly described. In our research, we characterized the poly-A RNA transcriptome changes after spontaneous immortalization. To this end, differentially expressed genes (DEGs) were screened using DESeq2 and characterized by gene ontology enrichment analysis and protein-protein interaction (PPI) network analysis to identify the potential hub genes. In our study, we identified changes in the expression of genes involved in proliferation regulation, cell adhesion, immune response and transcriptional regulation in immortalized MEFs. In addition, we performed a comparative analysis with previously reported MEF immortalization data, where we propose a predicted gene regulatory network model in immortalized MEFs based on the altered expression of Mapk11, Cdh1, Chl1, Zic1, Hoxd10 and the novel hub genes Il6 and Itgb2.
Collapse
Affiliation(s)
| | | | - Michael Leitges
- Division of Biomedical Sciences, Faculty of Medicine, Memorial University of Newfoundland, 300 Prince Philip Drive, St. Johns, NL A1B 3V6, Canada; (J.L.-M.); (U.B.)
| |
Collapse
|
4
|
Samson N, Bosoi CR, Roy C, Turcotte L, Tribouillard L, Mouchiroud M, Berthiaume L, Trottier J, Silva HCG, Guerbette T, Plata-Gómez AB, Besse-Patin A, Montoni A, Ilacqua N, Lamothe J, Citron YR, Gélinas Y, Gobeil S, Zoncu R, Caron A, Morissette M, Pellegrini L, Rochette PJ, Estall JL, Efeyan A, Shum M, Audet-Walsh É, Barbier O, Marette A, Laplante M. HSDL2 links nutritional cues to bile acid and cholesterol homeostasis. SCIENCE ADVANCES 2024; 10:eadk9681. [PMID: 38820148 PMCID: PMC11141617 DOI: 10.1126/sciadv.adk9681] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Accepted: 04/29/2024] [Indexed: 06/02/2024]
Abstract
In response to energy and nutrient shortage, the liver triggers several catabolic processes to promote survival. Despite recent progress, the precise molecular mechanisms regulating the hepatic adaptation to fasting remain incompletely characterized. Here, we report the identification of hydroxysteroid dehydrogenase-like 2 (HSDL2) as a mitochondrial protein highly induced by fasting. We show that the activation of PGC1α-PPARα and the inhibition of the PI3K-mTORC1 axis stimulate HSDL2 expression in hepatocytes. We found that HSDL2 depletion decreases cholesterol conversion to bile acids (BAs) and impairs FXR activity. HSDL2 knockdown also reduces mitochondrial respiration, fatty acid oxidation, and TCA cycle activity. Bioinformatics analyses revealed that hepatic Hsdl2 expression positively associates with the postprandial excursion of various BA species in mice. We show that liver-specific HSDL2 depletion affects BA metabolism and decreases circulating cholesterol levels upon refeeding. Overall, our report identifies HSDL2 as a fasting-induced mitochondrial protein that links nutritional signals to BAs and cholesterol homeostasis.
Collapse
Affiliation(s)
- Nolwenn Samson
- Centre de Recherche de l'Institut universitaire de cardiologie et de pneumologie de Québec (CRIUCPQ), Université Laval, Québec, QC, Canada
- Centre de recherche sur le cancer de l’Université Laval, Université Laval, Québec, QC, Canada
| | - Cristina R. Bosoi
- Centre de Recherche de l'Institut universitaire de cardiologie et de pneumologie de Québec (CRIUCPQ), Université Laval, Québec, QC, Canada
| | - Christian Roy
- Centre de Recherche de l'Institut universitaire de cardiologie et de pneumologie de Québec (CRIUCPQ), Université Laval, Québec, QC, Canada
| | - Laurie Turcotte
- Centre de Recherche de l'Institut universitaire de cardiologie et de pneumologie de Québec (CRIUCPQ), Université Laval, Québec, QC, Canada
| | - Laura Tribouillard
- Centre de Recherche de l'Institut universitaire de cardiologie et de pneumologie de Québec (CRIUCPQ), Université Laval, Québec, QC, Canada
- Centre de recherche sur le cancer de l’Université Laval, Université Laval, Québec, QC, Canada
| | - Mathilde Mouchiroud
- Centre de Recherche de l'Institut universitaire de cardiologie et de pneumologie de Québec (CRIUCPQ), Université Laval, Québec, QC, Canada
- Centre de recherche sur le cancer de l’Université Laval, Université Laval, Québec, QC, Canada
| | - Line Berthiaume
- Centre de recherche sur le cancer de l’Université Laval, Université Laval, Québec, QC, Canada
- Centre de recherche du Centre Hospitalier Universitaire (CHU) de Québec-Université Laval, Axe Endocrinologie et néphrologie, Québec, QC, Canada
| | - Jocelyn Trottier
- Centre de recherche du Centre Hospitalier Universitaire (CHU) de Québec-Université Laval, Axe Endocrinologie et néphrologie, Québec, QC, Canada
| | - Heitor C. G. Silva
- Centre de recherche du Centre Hospitalier Universitaire (CHU) de Québec-Université Laval, Axe Endocrinologie et néphrologie, Québec, QC, Canada
- Faculté de médecine, Université Laval, Québec, QC, Canada
| | - Thomas Guerbette
- Centre de recherche du Centre Hospitalier Universitaire (CHU) de Québec-Université Laval, Axe Endocrinologie et néphrologie, Québec, QC, Canada
- Faculté de médecine, Université Laval, Québec, QC, Canada
| | - Ana Belén Plata-Gómez
- Metabolism and Cell Signaling Laboratory, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| | - Aurèle Besse-Patin
- Institut de recherches cliniques de Montréal (IRCM), Montréal, QC, Canada
| | - Alicia Montoni
- Axe Médecine régénératrice, Centre de Recherche du CHU de Québec-Université Laval, Hôpital du Saint-Sacrement, Québec, QC, Canada
| | - Nicolò Ilacqua
- Faculté de médecine, Université Laval, Québec, QC, Canada
- Centre de recherche CERVO, Québec, QC, Canada
| | - Jennifer Lamothe
- Centre de Recherche de l'Institut universitaire de cardiologie et de pneumologie de Québec (CRIUCPQ), Université Laval, Québec, QC, Canada
| | - Yemima R. Citron
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA, USA
- Innovative Genomics Initiative at the University of California, Berkeley, Berkeley, CA, USA
| | - Yves Gélinas
- Centre de Recherche de l'Institut universitaire de cardiologie et de pneumologie de Québec (CRIUCPQ), Université Laval, Québec, QC, Canada
| | | | - Roberto Zoncu
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA, USA
- Innovative Genomics Initiative at the University of California, Berkeley, Berkeley, CA, USA
| | - Alexandre Caron
- Centre de Recherche de l'Institut universitaire de cardiologie et de pneumologie de Québec (CRIUCPQ), Université Laval, Québec, QC, Canada
- Faculté de Pharmacie, Université Laval, Québec, QC, Canada
| | - Mathieu Morissette
- Centre de Recherche de l'Institut universitaire de cardiologie et de pneumologie de Québec (CRIUCPQ), Université Laval, Québec, QC, Canada
- Faculté de médecine, Université Laval, Québec, QC, Canada
| | - Luca Pellegrini
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, QC, Canada
| | - Patrick J. Rochette
- Faculté de médecine, Université Laval, Québec, QC, Canada
- Axe Médecine régénératrice, Centre de Recherche du CHU de Québec-Université Laval, Hôpital du Saint-Sacrement, Québec, QC, Canada
- Département d’Ophtalmologie et ORL – chirurgie cervico-faciale, Université Laval, Québec, QC, Canada
| | - Jennifer L. Estall
- Institut de recherches cliniques de Montréal (IRCM), Montréal, QC, Canada
- Faculté de médecine, Université de Montréal, Montréal, QC, Canada
| | - Alejo Efeyan
- Metabolism and Cell Signaling Laboratory, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| | - Michael Shum
- Centre de recherche du Centre Hospitalier Universitaire (CHU) de Québec-Université Laval, Axe Endocrinologie et néphrologie, Québec, QC, Canada
- Faculté de médecine, Université Laval, Québec, QC, Canada
| | - Étienne Audet-Walsh
- Centre de recherche sur le cancer de l’Université Laval, Université Laval, Québec, QC, Canada
- Centre de recherche du Centre Hospitalier Universitaire (CHU) de Québec-Université Laval, Axe Endocrinologie et néphrologie, Québec, QC, Canada
- Faculté de médecine, Université Laval, Québec, QC, Canada
| | - Olivier Barbier
- Centre de recherche du Centre Hospitalier Universitaire (CHU) de Québec-Université Laval, Axe Endocrinologie et néphrologie, Québec, QC, Canada
- Faculté de Pharmacie, Université Laval, Québec, QC, Canada
| | - André Marette
- Centre de Recherche de l'Institut universitaire de cardiologie et de pneumologie de Québec (CRIUCPQ), Université Laval, Québec, QC, Canada
- Faculté de médecine, Université Laval, Québec, QC, Canada
| | - Mathieu Laplante
- Centre de Recherche de l'Institut universitaire de cardiologie et de pneumologie de Québec (CRIUCPQ), Université Laval, Québec, QC, Canada
- Centre de recherche sur le cancer de l’Université Laval, Université Laval, Québec, QC, Canada
- Faculté de médecine, Université Laval, Québec, QC, Canada
| |
Collapse
|
5
|
Khashei Varnamkhasti K, Moghanibashi M, Naeimi S. Implications of ZNF334 gene in lymph node metastasis of lung SCC: potential bypassing of cellular senescence. J Transl Med 2024; 22:372. [PMID: 38637790 PMCID: PMC11025273 DOI: 10.1186/s12967-024-05115-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Accepted: 03/20/2024] [Indexed: 04/20/2024] Open
Abstract
BACKGROUND The primary goal of this work is to identify biomarkers associated with lung squamous cell carcinoma and assess their potential for early detection of lymph node metastasis. METHODS This study investigated gene expression in lymph node metastasis of lung squamous cell carcinoma using data from the Cancer Genome Atlas and R software. Protein-protein interaction networks, hub genes, and enriched pathways were analyzed. ZNF334 and TINAGL1, two less explored genes, were further examined through in vitro, ex vivo, and in vivo experiments to validate the findings from bioinformatics analyses. The role of ZNF334 and TINAGL1 in senescence induction was assessed after H2O2 and UV induced senescence phenotype determined using β-galactosidase activity and cell cycle status assay. RESULTS We identified a total of 611 up- and 339 down-regulated lung squamous cell carcinoma lymph node metastasis-associated genes (FDR < 0.05). Pathway enrichment analysis highlighted the central respiratory pathway within mitochondria for the subnet genes and the nuclear DNA-directed RNA polymerases for the hub genes. Significantly down regulation of ZNF334 gene was associated with malignancy lymph node progression and senescence induction has significantly altered ZNF334 expression (with consistency in bioinformatics, in vitro, ex vivo, and in vivo results). Deregulation of TINAGL1 expression with inconsistency in bioinformatics, in vitro (different types of lung squamous cancer cell lines), ex vivo, and in vivo results, was also associated with malignancy lymph node progression and altered in senescence phenotype. CONCLUSIONS ZNF334 is a highly generalizable gene to lymph node metastasis of lung squamous cell carcinoma and its expression alter certainly under senescence conditions.
Collapse
Affiliation(s)
| | - Mehdi Moghanibashi
- Department of Genetics, Faculty of Medicine, Kazerun Branch, Islamic Azad University, Kazerun, Iran.
| | - Sirous Naeimi
- Department of Genetics, Faculty of Basic Sciences, Kazerun Branch, Islamic Azad University, Kazerun, Iran
| |
Collapse
|
6
|
Lee E, Carreras-Gallo N, Lopez L, Turner L, Lin A, Mendez TL, Went H, Tomusiak A, Verdin E, Corley M, Ndhlovu L, Smith R, Dwaraka VB. Exploring the effects of Dasatinib, Quercetin, and Fisetin on DNA methylation clocks: a longitudinal study on senolytic interventions. Aging (Albany NY) 2024; 16:3088-3106. [PMID: 38393697 PMCID: PMC10929829 DOI: 10.18632/aging.205581] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2023] [Accepted: 01/19/2024] [Indexed: 02/25/2024]
Abstract
Senolytics, small molecules targeting cellular senescence, have emerged as potential therapeutics to enhance health span. However, their impact on epigenetic age remains unstudied. This study aimed to assess the effects of Dasatinib and Quercetin (DQ) senolytic treatment on DNA methylation (DNAm), epigenetic age, and immune cell subsets. In a Phase I pilot study, 19 participants received DQ for 6 months, with DNAm measured at baseline, 3 months, and 6 months. Significant increases in epigenetic age acceleration were observed in first-generation epigenetic clocks and mitotic clocks at 3 and 6 months, along with a notable decrease in telomere length. However, no significant differences were observed in second and third-generation clocks. Building upon these findings, a subsequent investigation evaluated the combination of DQ with Fisetin (DQF), a well-known antioxidant and antiaging senolytic molecule. After one year, 19 participants (including 10 from the initial study) received DQF for 6 months, with DNAm assessed at baseline and 6 months. Remarkably, the addition of Fisetin to the treatment resulted in non-significant increases in epigenetic age acceleration, suggesting a potential mitigating effect of Fisetin on the impact of DQ on epigenetic aging. Furthermore, our analyses unveiled notable differences in immune cell proportions between the DQ and DQF treatment groups, providing a biological basis for the divergent patterns observed in the evolution of epigenetic clocks. These findings warrant further research to validate and comprehensively understand the implications of these combined interventions.
Collapse
Affiliation(s)
- Edwin Lee
- Institute For Hormonal Balance, Orlando, FL 32819, USA
| | | | | | | | - Aaron Lin
- TruDiagnostic, Lexington, KY 40503, USA
| | | | | | - Alan Tomusiak
- Buck Institute for Research on Aging, Novato, CA 94945, USA
| | - Eric Verdin
- Buck Institute for Research on Aging, Novato, CA 94945, USA
| | | | | | | | | |
Collapse
|
7
|
Deng Y, Adam V, Nepovimova E, Heger Z, Valko M, Wu Q, Wei W, Kuca K. c-Jun N-terminal kinase signaling in cellular senescence. Arch Toxicol 2023; 97:2089-2109. [PMID: 37335314 DOI: 10.1007/s00204-023-03540-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Accepted: 06/06/2023] [Indexed: 06/21/2023]
Abstract
Cellular senescence leads to decreased tissue regeneration and inflammation and is associated with diabetes, neurodegenerative diseases, and tumorigenesis. However, the mechanisms of cellular senescence are not fully understood. Emerging evidence has indicated that c-Jun N-terminal kinase (JNK) signaling is involved in the regulation of cellular senescence. JNK can downregulate hypoxia inducible factor-1α to accelerate hypoxia-induced neuronal cell senescence. The activation of JNK inhibits mTOR activity and triggers autophagy, which promotes cellular senescence. JNK can upregulate the expression of p53 and Bcl-2 and accelerates cancer cell senescence; however, this signaling also mediates the expression of amphiregulin and PD-LI to achieve cancer cell immune evasion and prevents their senescence. The activation of JNK further triggers forkhead box O expression and its target gene Jafrac1 to extend the lifespan of Drosophila. JNK can also upregulate the expression of DNA repair protein poly ADP-ribose polymerase 1 and heat shock protein to delay cellular senescence. This review discusses recent advances in understanding the function of JNK signaling in cellular senescence and includes a comprehensive analysis of the molecular mechanisms underlying JNK-mediated senescence evasion and oncogene-induced cellular senescence. We also summarize the research progress in anti-aging agents that target JNK signaling. This study will contribute to a better understanding of the molecular targets of cellular senescence and provides insights into anti-aging, which may be used to develop drugs for the treatment of aging-related diseases.
Collapse
Affiliation(s)
- Ying Deng
- College of Life Science, Yangtze University, Jingzhou, 434025, China
| | - Vojtech Adam
- Department of Chemistry and Biochemistry, Mendel University in Brno, Brno, 613 00, Czech Republic
- Central European Institute of Technology, Brno University of Technology, Brno, 602 00, Czech Republic
| | - Eugenie Nepovimova
- Department of Chemistry, Faculty of Science, University of Hradec Králové, 500 03, Hradec Králové, Czech Republic
| | - Zbynek Heger
- Department of Chemistry and Biochemistry, Mendel University in Brno, Brno, 613 00, Czech Republic
- Central European Institute of Technology, Brno University of Technology, Brno, 602 00, Czech Republic
| | - Marian Valko
- Faculty of Chemical and Food Technology, Slovak University of Technology, 812 37, Bratislava, Slovakia
| | - Qinghua Wu
- College of Life Science, Yangtze University, Jingzhou, 434025, China.
- Department of Chemistry, Faculty of Science, University of Hradec Králové, 500 03, Hradec Králové, Czech Republic.
| | - Wei Wei
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-Products, Key Laboratory of Traceability for Agricultural Genetically Modified Organisms, Ministry of Agriculture and Rural Affairs, Zhejiang Academy of Agricultural Sciences, Hangzhou, 310021, China.
| | - Kamil Kuca
- Department of Chemistry, Faculty of Science, University of Hradec Králové, 500 03, Hradec Králové, Czech Republic.
- Andalusian Research Institute in Data Science and Computational Intelligence (DaSCI), University of Granada, Granada, Spain.
| |
Collapse
|
8
|
Zhao HG, Deininger M. Always stressed but never exhausted: how stem cells in myeloid neoplasms avoid extinction in inflammatory conditions. Blood 2023; 141:2797-2812. [PMID: 36947811 PMCID: PMC10315634 DOI: 10.1182/blood.2022017152] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Revised: 02/27/2023] [Accepted: 03/13/2023] [Indexed: 03/24/2023] Open
Abstract
Chronic or recurrent episodes of acute inflammation cause attrition of normal hematopoietic stem cells (HSCs) that can lead to hematopoietic failure but they drive progression in myeloid malignancies and their precursor clonal hematopoiesis. Mechanistic parallels exist between hematopoiesis in chronic inflammation and the continuously increased proliferation of myeloid malignancies, particularly myeloproliferative neoplasms (MPNs). The ability to enter dormancy, a state of deep quiescence characterized by low oxidative phosphorylation, low glycolysis, reduced protein synthesis, and increased autophagy is central to the preservation of long-term HSCs and likely MPN SCs. The metabolic features of dormancy resemble those of diapause, a state of arrested embryonic development triggered by adverse environmental conditions. To outcompete their normal counterparts in the inflammatory MPN environment, MPN SCs co-opt mechanisms used by HSCs to avoid exhaustion, including signal attenuation by negative regulators, insulation from activating cytokine signals, anti-inflammatory signaling, and epigenetic reprogramming. We propose that new therapeutic strategies may be derived from conceptualizing myeloid malignancies as an ecosystem out of balance, in which residual normal and malignant hematopoietic cells interact in multiple ways, only few of which have been characterized in detail. Disrupting MPN SC insulation to overcome dormancy, interfering with aberrant cytokine circuits that favor MPN cells, and directly boosting residual normal HSCs are potential strategies to tip the balance in favor of normal hematopoiesis. Although eradicating the malignant cell clones remains the goal of therapy, rebalancing the ecosystem may be a more attainable objective in the short term.
Collapse
Affiliation(s)
- Helong Gary Zhao
- Versiti Blood Research Institute and Medical College of Wisconsin, Milwaukee, WI
| | - Michael Deininger
- Versiti Blood Research Institute and Medical College of Wisconsin, Milwaukee, WI
| |
Collapse
|
9
|
An injectable and pH-responsive hyaluronic acid hydrogel as metformin carrier for prevention of breast cancer recurrence. Carbohydr Polym 2023; 304:120493. [PMID: 36641175 DOI: 10.1016/j.carbpol.2022.120493] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2022] [Revised: 12/13/2022] [Accepted: 12/20/2022] [Indexed: 12/25/2022]
Abstract
To achieve the pH-responsive release of metformin in tumor acidic microenvironment, we prepared OHA-Met by covalently grafting metformin (Met) onto oxidized hyaluronic acid (OHA) through imine bonds, and then prepared carboxymethyl chitosan (CMCS)/OHA-Met drug loaded hydrogels. The CMCS/OHA-Met hydrogels showed the in-situ injection performance. At pH = 7.4, the cumulative release rate of metformin from CMCS/OHA-Met20 hydrogel was 42.7 ± 2.6 % in 6 h, and the release tended to balance after 72 h. At pH = 5.5, the release kept constant and the cumulative release rate was 79.3 ± 4.7 % at 6 h, showing good pH-responsive behavior. Metformin induced apoptosis of MCF-7 cells through the caspase 3/PARP pathway. CMCS/OHA-Met20 hydrogel could effectively kill MCF-7 cells, while reducing the cytotoxicity of free metformin to L929 cells. In vivo breast cancer recurrence experiments showed CMCS/OHA-Met20 hydrogel could achieve local injection and pH-responsive smart drug delivery at the tumor resection site, inhibiting breast cancer recurrence. Compared with direct administration, CMCS/OHA-Met20 hydrogel reduced the metformin dosage, frequency of administration and systemic side effects.
Collapse
|
10
|
Shimoyama Y, Yamada K, Yoshida S, Kawamura A, Hannya Y, Imaizumi Y, Kumamoto T, Takeda Y, Shimoda M, Eto K, Yoshida K. Inhibition of protein kinase C delta leads to cellular senescence to induce anti-tumor effects in colorectal cancer. Cancer Sci 2023. [PMID: 36851883 DOI: 10.1111/cas.15768] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Revised: 02/10/2023] [Accepted: 02/21/2023] [Indexed: 03/01/2023] Open
Abstract
Protein kinase C delta (PKCδ) is a multifunctional serine-threonine kinase implicated in cell proliferation, differentiation, tumorigenesis, and therapeutic resistance. However, the molecular mechanism of PKCδ in colorectal cancer (CRC) remains unclear. In this study, we showed that PKCδ acts as a negative regulator of cellular senescence in p53 wild-type (wt-p53) CRC. Immunohistochemical analysis revealed that PKCδ levels in human CRC tissues were higher than those in the surrounding normal tissues. Deletion studies have shown that cell proliferation and tumorigenesis in wt-p53 CRC is sensitive to PKCδ expression. We found that PKCδ activates p21 via a p53-independent pathway and that PKCδ-kinase activity is essential for p21 activity. In addition, both repression of PKCδ expression and inhibition of PKCδ activity induced cellular senescence-like phenotypes, including increased senescence-associated β-galactosidase (SA-β-gal) staining, low LaminB1 expression, large nucleus size, and senescence-associated secretory phenotype (SASP) detection. Finally, a kinase inhibitor of PKCδ suppressed senescence-dependent tumorigenicity in a dose-dependent manner. These results offer a mechanistic insight into CRC survival and tumorigenesis. In addition, a novel therapeutic strategy for wt-p53 CRC is proposed.
Collapse
Affiliation(s)
- Yuya Shimoyama
- Department of Biochemistry, The Jikei University School of Medicine, Tokyo, Japan
| | - Kohji Yamada
- Department of Biochemistry, The Jikei University School of Medicine, Tokyo, Japan
| | - Saishu Yoshida
- Department of Biochemistry, The Jikei University School of Medicine, Tokyo, Japan
| | - Akira Kawamura
- Department of Biochemistry, The Jikei University School of Medicine, Tokyo, Japan
| | - Yoshito Hannya
- Department of Biochemistry, The Jikei University School of Medicine, Tokyo, Japan
| | - Yuta Imaizumi
- Department of Surgery, The Jikei University School of Medicine, Tokyo, Japan
| | - Tomotaka Kumamoto
- Department of Surgery, The Jikei University School of Medicine, Tokyo, Japan
| | - Yasuhiro Takeda
- Department of Surgery, The Jikei University School of Medicine, Tokyo, Japan
| | - Masayuki Shimoda
- Department of Pathology, The Jikei University School of Medicine, Tokyo, Japan
| | - Ken Eto
- Department of Surgery, The Jikei University School of Medicine, Tokyo, Japan
| | - Kiyotsugu Yoshida
- Department of Biochemistry, The Jikei University School of Medicine, Tokyo, Japan
| |
Collapse
|
11
|
Zhao J, Wu Y, Yue Y, Chen M, Xu Y, Liu X, Liu X, Gao X, Wang H, Si X, Zhong W, Zhang X, Zhang L, Wang M. The development of a tumor-associated autoantibodies panel to predict clinical outcomes for immune checkpoint inhibitor-based treatment in patients with advanced non-small-cell lung cancer. Thorac Cancer 2023; 14:497-505. [PMID: 36594104 PMCID: PMC9925345 DOI: 10.1111/1759-7714.14772] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Revised: 11/28/2022] [Accepted: 11/29/2022] [Indexed: 01/04/2023] Open
Abstract
BACKGROUND Immune checkpoint inhibitors (ICIs) have become one important therapeutic strategy for advanced non-small-cell lung cancer (NSCLC). It remains imperative to identify reliable and convenient biomarkers to predict both the efficacy and toxicity of immunotherapy, and tumor-associated autoantibodies (TAAbs) are recognized as one of the promising candidates for this. PATIENTS AND METHODS This study enrolled 97 advanced NSCLC patients with ICI-based immunotherapy treatment, who were divided into a training cohort (n = 48) and a validation cohort (n = 49), and measured for the serum level of 35 TAAbs. According to the statistical association between the serum positivity and clinical outcome of each TAAb in the training cohort, a TAAb panel was developed to predict the progression-free survival (PFS), and further examined in the validation cohort and in different subgroups. Similarly, another TAAb panel was derived to predict the occurrence of immune-related adverse events (irAEs). RESULTS In the training cohort, a 7-TAAb panel composed of p53, CAGE, MAGEA4, GAGE7, UTP14A, IMP2, and PSMC1 TAAbs was derived to predict PFS (median PFS [mPFS] 9.9 vs. 4.3 months, p = 0.043). The statistical association between the panel positivity and longer PFS was confirmed in the validation cohort (mPFS 11.1 vs. 4.8 months, p = 0.015) and in different subgroups of patients. Moreover, another 4-TAAb panel of BRCA2, MAGEA4, ZNF768, and PARP TAAbs was developed to predict the occurrence of irAEs, showing higher risk in panel-positive patients (71.43% vs. 28.91%, p = 0.0046). CONCLUSIONS Collectively, our study developed and validated two TAAb panels as valuable prognostic biomarkers for immunotherapy.
Collapse
Affiliation(s)
- Jing Zhao
- Department of Respiratory and Critical Care MedicinePeking Union Medical College HospitalBeijingChina
| | - Yang Wu
- School of MedicineTsinghua UniversityBeijingChina
| | - Yuan Yue
- Department of Respiratory and Critical Care MedicinePeking Union Medical College HospitalBeijingChina
| | - Minjiang Chen
- Department of Respiratory and Critical Care MedicinePeking Union Medical College HospitalBeijingChina
| | - Yan Xu
- Department of Respiratory and Critical Care MedicinePeking Union Medical College HospitalBeijingChina
| | - Xiangning Liu
- Department of Respiratory and Critical Care MedicinePeking Union Medical College HospitalBeijingChina
| | - Xiaoyan Liu
- Department of Respiratory and Critical Care MedicinePeking Union Medical College HospitalBeijingChina
| | - Xiaoxing Gao
- Department of Respiratory and Critical Care MedicinePeking Union Medical College HospitalBeijingChina
| | - Hanping Wang
- Department of Respiratory and Critical Care MedicinePeking Union Medical College HospitalBeijingChina
| | - Xiaoyan Si
- Department of Respiratory and Critical Care MedicinePeking Union Medical College HospitalBeijingChina
| | - Wei Zhong
- Department of Respiratory and Critical Care MedicinePeking Union Medical College HospitalBeijingChina
| | - Xiaotong Zhang
- Department of Respiratory and Critical Care MedicinePeking Union Medical College HospitalBeijingChina
| | - Li Zhang
- Department of Respiratory and Critical Care MedicinePeking Union Medical College HospitalBeijingChina
| | - Mengzhao Wang
- Department of Respiratory and Critical Care MedicinePeking Union Medical College HospitalBeijingChina
| |
Collapse
|
12
|
Kawamoto S, Matsumoto T, Takasugi M, Hara E. The 6th international cell senescence association conference. Genes Cells 2022; 27:517-525. [PMID: 35726163 DOI: 10.1111/gtc.12970] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Revised: 06/01/2022] [Accepted: 06/01/2022] [Indexed: 11/28/2022]
Abstract
The 6th conference of the international cell senescence association (ICSA) on the theme of "A New Era of Senescence Research: The Challenge of Controlling Aging and Cancer" was held on December 12-15, 2021 in Osaka, Japan as a Hybrid Meeting. The conference brought together basic and translational scientists to discuss recent developments in the field of cellular senescence research. In recent years, the study of cellular senescence has become a very hot field of research. It is clear that the ICSA, founded in 2015, has played an important role in this process. The 6th ICSA conference has provided another opportunity for exchanges and new connections between basic and translational scientists. The scientific program consisted of keynote lectures, invited talks, short talks selected from abstracts, a poster session, and luncheon seminars sponsored by the Japanese Society of Anti-Aging Medicine. In the Meet the Editor session, Dr Christoph Schmitt, Editor-in-Chief of Nature Metabolism, gave a short presentation about the journal and answered questions from the audience. Being a hybrid meeting, there was only so much that could be done, but we hope that the meeting was fruitful.
Collapse
Affiliation(s)
- Shimpei Kawamoto
- Research Institute for Microbial Diseases, Osaka University, Suita, Japan
| | - Tomonori Matsumoto
- Research Institute for Microbial Diseases, Osaka University, Suita, Japan
| | - Masaki Takasugi
- Graduate School of Medicine, Osaka Metropolitan University, Osaka, Japan
| | - Eiji Hara
- Research Institute for Microbial Diseases, Osaka University, Suita, Japan
| |
Collapse
|
13
|
Zha Y, Zhuang W, Yang Y, Zhou Y, Li H, Liang J. Senescence in Vascular Smooth Muscle Cells and Atherosclerosis. Front Cardiovasc Med 2022; 9:910580. [PMID: 35722104 PMCID: PMC9198250 DOI: 10.3389/fcvm.2022.910580] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Accepted: 05/04/2022] [Indexed: 12/12/2022] Open
Abstract
Vascular smooth muscle cells (VSMCs) are the primary cell type involved in the atherosclerosis process; senescent VSMCs are observed in both aged vessels and atherosclerotic plaques. Factors associated with the atherosclerotic process, including oxidative stress, inflammation, and calcium-regulating factors, are closely linked to senescence in VSMCs. A number of experimental studies using traditional cellular aging markers have suggested that anti-aging biochemical agents could be used to treat atherosclerosis. However, doubt has recently been cast on such potential due to the increasingly apparent complexity of VSMCs status and an incomplete understanding of the role that these cells play in the atherosclerosis process, as well as a lack of specific or spectrum-limited cellular aging markers. The utility of anti-aging drugs in atherosclerosis treatment should be reevaluated. Promotion of a healthy lifestyle, exploring in depth the characteristics of each cell type associated with atherosclerosis, including VSMCs, and development of targeted drug delivery systems will ensure efficacy whilst evaluation of the safety and tolerability of drug use should be key aims of future anti-atherosclerosis research. This review summarizes the characteristics of VSMC senescence during the atherosclerosis process, the factors regulating this process, as well as an overview of progress toward the development and application of anti-aging drugs.
Collapse
Affiliation(s)
- Yiwen Zha
- Medical College, Yangzhou University, Yangzhou, China
| | - Wenwen Zhuang
- Medical College, Yangzhou University, Yangzhou, China
| | - Yongqi Yang
- Medical College, Yangzhou University, Yangzhou, China
| | - Yue Zhou
- Medical College, Yangzhou University, Yangzhou, China
| | - Hongliang Li
- Medical College, Yangzhou University, Yangzhou, China
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, China
- *Correspondence: Hongliang Li,
| | - Jingyan Liang
- Medical College, Yangzhou University, Yangzhou, China
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, China
- Jiangsu Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Senile Diseases, Yangzhou University, Yangzhou, China
- Jingyan Liang,
| |
Collapse
|
14
|
Villot R, Poirier A, Devillers R, Kolnoguz A, Elowe S, Manem VSK, Joubert P, Mallette FA, Laplante M. ZNF768: controlling cellular senescence and proliferation with ten fingers. Mol Cell Oncol 2021; 8:1985930. [PMID: 35419475 PMCID: PMC8997246 DOI: 10.1080/23723556.2021.1985930] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
We recently identified Zinc-finger protein 768 (ZNF768) as a novel transcription factor controlling cell fate decision downstream of Rat sarcoma virus (RAS). We showed that ZNF768 depletion impairs cell cycle progression and triggers cellular senescence, while its overexpression allows cells to bypass oncogene-induced senescence. Elevated ZNF768 levels is common in tumors, suggesting that ZNF768 may help to escape cellular senescence, sustain proliferation and promote malignant transformation. Here, we discuss these recent findings and highlight key questions emerging from our work.
Collapse
Affiliation(s)
- Romain Villot
- Centre de Recherche de l’Institut Universitaire de Cardiologie et de Pneumologie de Québec (CRIUCPQ), Faculté de Médecine, Université Laval, Québec, QC, Canada
- Centre de Recherche sur le Cancer de l’Université Laval, Université Laval, Québec, QC, Canada
| | - Audrey Poirier
- Centre de Recherche de l’Institut Universitaire de Cardiologie et de Pneumologie de Québec (CRIUCPQ), Faculté de Médecine, Université Laval, Québec, QC, Canada
- Centre de Recherche sur le Cancer de l’Université Laval, Université Laval, Québec, QC, Canada
| | - Romain Devillers
- Centre de Recherche de l’Institut Universitaire de Cardiologie et de Pneumologie de Québec (CRIUCPQ), Faculté de Médecine, Université Laval, Québec, QC, Canada
- Centre de Recherche sur le Cancer de l’Université Laval, Université Laval, Québec, QC, Canada
- Centre de Recherche du CHU de Québec, Université Laval, Québec, QC, Canada
| | - Aliona Kolnoguz
- Centre de Recherche de l’Institut Universitaire de Cardiologie et de Pneumologie de Québec (CRIUCPQ), Faculté de Médecine, Université Laval, Québec, QC, Canada
- Centre de Recherche sur le Cancer de l’Université Laval, Université Laval, Québec, QC, Canada
| | - Sabine Elowe
- Centre de Recherche sur le Cancer de l’Université Laval, Université Laval, Québec, QC, Canada
- Centre de Recherche du CHU de Québec, Université Laval, Québec, QC, Canada
- Département de Pédiatrie, Faculté de Médecine, Université Laval, Québec, QC, Canada
| | - Venkata S. K. Manem
- Centre de Recherche de l’Institut Universitaire de Cardiologie et de Pneumologie de Québec (CRIUCPQ), Faculté de Médecine, Université Laval, Québec, QC, Canada
- Faculty of Pharmacy, Université Laval, Québec, QC, Canada
| | - Philippe Joubert
- Centre de Recherche de l’Institut Universitaire de Cardiologie et de Pneumologie de Québec (CRIUCPQ), Faculté de Médecine, Université Laval, Québec, QC, Canada
- Centre de Recherche sur le Cancer de l’Université Laval, Université Laval, Québec, QC, Canada
| | - Frédérick A. Mallette
- Département de Médecine, Université de Montréal, Montréal, QC, Canada
- Chromatin Structure and Cellular Senescence Research Unit, Maisonneuve-Rosemont Hospital Research Centre, Université de Montréal, Montréal, QC, Canada
- Département de Biochimie et Médecine Moléculaire, Université de Montréal, Montréal, QC, Canada
| | - Mathieu Laplante
- Centre de Recherche de l’Institut Universitaire de Cardiologie et de Pneumologie de Québec (CRIUCPQ), Faculté de Médecine, Université Laval, Québec, QC, Canada
- Centre de Recherche sur le Cancer de l’Université Laval, Université Laval, Québec, QC, Canada
- Département de Médecine, Faculté de Médecine, Université Laval, Québec, QC, Canada
| |
Collapse
|
15
|
ZNF768 Expression Associates with High Proliferative Clinicopathological Features in Lung Adenocarcinoma. Cancers (Basel) 2021; 13:cancers13164136. [PMID: 34439288 PMCID: PMC8391643 DOI: 10.3390/cancers13164136] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2021] [Revised: 08/03/2021] [Accepted: 08/10/2021] [Indexed: 12/24/2022] Open
Abstract
Simple Summary Zinc-finger protein 768 (ZNF768) is a transcription factor that was recently shown to promote proliferation and repress senescence downstream of growth factor signaling in vitro. This protein was found to be overexpressed in a small cohort of lung adenocarcinoma (LUAD) compared to normal tissue, but its clinical value in this cancer remains unknown. The aim of this study was to determine whether ZNF768 associates with clinicopathological features in LUAD. We found that ZNF768 levels are often elevated in LUAD and that ZNF768 protein levels positively correlate with Ki-67 and other proliferative clinicopathological features in this cancer. Supporting a role for ZNF768 in promoting proliferation, ZNF768 depletion severely impairs proliferation in several lung cancer cell lines. Our study, that extends previous in vitro data, provides the first clinical observations supporting a possible role for ZNF768 in promoting cancer cell proliferation and tumor development in humans. Abstract Lung adenocarcinoma (LUAD) is the most common type of lung cancer and a leading cause of cancer-related deaths worldwide. Despite important recent advances, the prognosis for LUAD patients is still unfavourable, with a 5 year-survival rate close to 15%. Improving the characterization of lung tumors is important to develop alternative options for the diagnosis and the treatment of this disease. Zinc-finger protein 768 (ZNF768) is a transcription factor that was recently shown to promote proliferation and repress senescence downstream of growth factor signaling. Although ZNF768 protein levels were found to be elevated in LUAD compared to normal lung tissue, it is currently unknown whether ZNF768 expression associates with clinicopathological features in LUAD. Here, using tissue microarrays of clinical LUAD surgical specimens collected from 364 patients, we observed that high levels of ZNF768 is a common characteristic of LUAD. We show that ZNF768 protein levels correlate with high proliferative features in LUAD, including the mitotic score and Ki-67 expression. Supporting a role for ZNF768 in promoting proliferation, we report that ZNF768 depletion severely impairs proliferation in several lung cancer cell lines in vitro. A marked decrease in the expression of key proliferative genes was observed in cancer cell lines depleted from ZNF768. Altogether, our findings support a role for ZNF768 in promoting proliferation of LUAD.
Collapse
|