1
|
Xie J, Shu X, Xie Z, Tang J, Wang G. Pharmacological modulation of cellular senescence: Implications for breast cancer progression and therapeutic strategies. Eur J Pharmacol 2025; 997:177475. [PMID: 40049574 DOI: 10.1016/j.ejphar.2025.177475] [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: 12/09/2024] [Revised: 02/26/2025] [Accepted: 03/04/2025] [Indexed: 05/02/2025]
Abstract
Senescence, defined by the cessation of cell proliferation, plays a critical and multifaceted role in breast cancer progression and treatment. Senescent cells produce senescence-associated secretory phenotypes (SASP) comprising inflammatory cytokines, chemokines, and small molecules, which actively shape the tumor microenvironment, influencing cancer development, progression, and metastasis. This review provides a comprehensive analysis of the types and origins of senescent cells in breast cancer, alongside their markers and detection methods. Special focus is placed on pharmacological strategies targeting senescence, including drugs that induce or inhibit senescence, their molecular mechanisms, and their roles in therapeutic outcomes when combined with chemotherapy and radiotherapy. By exploring these pharmacological interventions and their impact on breast cancer treatment, this review underscores the potential of senescence-targeting therapies to revolutionize breast cancer management.
Collapse
Affiliation(s)
- Jialing Xie
- Department of Clinical Pharmacology, Xiangya Hospital, Institute of Clinical Pharmacology, Hunan Key Laboratory of Pharmacogenetics, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, People's Republic of China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, 87 Xiangya Road, Changsha, 410008, People's Republic of China
| | - Xianlong Shu
- Department of Clinical Pharmacology, Xiangya Hospital, Institute of Clinical Pharmacology, Hunan Key Laboratory of Pharmacogenetics, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, People's Republic of China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, 87 Xiangya Road, Changsha, 410008, People's Republic of China
| | - Zilan Xie
- Department of Clinical Pharmacology, Xiangya Hospital, Institute of Clinical Pharmacology, Hunan Key Laboratory of Pharmacogenetics, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, People's Republic of China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, 87 Xiangya Road, Changsha, 410008, People's Republic of China
| | - Jie Tang
- Department of Clinical Pharmacology, Xiangya Hospital, Institute of Clinical Pharmacology, Hunan Key Laboratory of Pharmacogenetics, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, People's Republic of China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, 87 Xiangya Road, Changsha, 410008, People's Republic of China.
| | - Guo Wang
- Department of Clinical Pharmacology, Xiangya Hospital, Institute of Clinical Pharmacology, Hunan Key Laboratory of Pharmacogenetics, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, People's Republic of China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, 87 Xiangya Road, Changsha, 410008, People's Republic of China.
| |
Collapse
|
2
|
Guerrero-Navarro L, Monfort-Lanzas P, Krichbaumer V, De Araújo MEG, Monfregola J, Huber LA, Ballabio A, Jansen-Dürr P, Cavinato M. TFEB Orchestrates Stress Recovery and Paves the Way for Senescence Induction in Human Dermal Fibroblasts. Aging Cell 2025:e70083. [PMID: 40312996 DOI: 10.1111/acel.70083] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2025] [Accepted: 04/07/2025] [Indexed: 05/03/2025] Open
Abstract
Cells experience oxidative stress and widespread cellular damage during stress-induced premature senescence (SIPS). Senescent cells show an increase in lysosomal content, which may contribute to mitigating cellular damage by promoting autophagy. This study investigates the dynamics of lysosomal quality control in human dermal fibroblasts (HDF), specifically examining lysosomal signaling pathways during oxidative stress-induced SIPS. Our results reveal distinct signaling responses between the initial stress phase and the ensuing senescent phenotype. During the stress phase, treatment with tBHP, which undermines the antioxidant response, leads to elevated reactive oxygen species (ROS) and lysosomal damage. ROS accumulation activates AMP-activated protein kinase (AMPK) and inhibits Akt, which correlates with the suppression of mammalian target of rapamycin (mTOR). Inactivation of mTOR during this phase aligns with the activation of transcription factor EB (TFEB), a key regulator of autophagy and lysosomal biogenesis. TFEB knockdown under stress increased apoptosis, highlighting the protective role of TFEB in the stress response. As cells transition to senescence, TFEB activity, required for the autophagic damage repair, becomes less critical. The decrease in ROS levels leads to the normalization of AMPK and Akt signaling, accompanied by the reactivation of mTOR. This reactivation of mTOR, which is critical for establishing the senescent state, is observed alongside the inactivation of TFEB. Consequently, as damage decreases, TFEB activity decreases. Our results suggest a dynamic interplay between TFEB and mTOR, highlighting a critical role of TFEB in ensuring cellular survival during SIPS induction but becoming dispensable once senescence is established.
Collapse
Affiliation(s)
- Lena Guerrero-Navarro
- Institute for Biomedical Aging Research, Universität Innsbruck, Innsbruck, Austria
- Center for Molecular Biosciences Innsbruck (CMBI), Innsbruck, Austria
| | - Pablo Monfort-Lanzas
- Institute of Medical Biochemistry, Biocenter, Innsbruck Medical University, Innsbruck, Austria
- Institute of Bioinformatics, Biocenter, Innsbruck Medical University, Innsbruck, Austria
| | - Vinzenz Krichbaumer
- Institute for Biomedical Aging Research, Universität Innsbruck, Innsbruck, Austria
- Center for Molecular Biosciences Innsbruck (CMBI), Innsbruck, Austria
| | - Mariana E G De Araújo
- Biocenter, Division of Cell Biology, Innsbruck Medical University, Innsbruck, Austria
| | | | - Lukas A Huber
- Biocenter, Division of Cell Biology, Innsbruck Medical University, Innsbruck, Austria
| | - Andrea Ballabio
- Telethon Institute of Genetics and Medicine (TIGEM), Naples, Italy
| | - Pidder Jansen-Dürr
- Institute for Biomedical Aging Research, Universität Innsbruck, Innsbruck, Austria
- Center for Molecular Biosciences Innsbruck (CMBI), Innsbruck, Austria
| | - Maria Cavinato
- Institute for Biomedical Aging Research, Universität Innsbruck, Innsbruck, Austria
- Center for Molecular Biosciences Innsbruck (CMBI), Innsbruck, Austria
| |
Collapse
|
3
|
Czajkowski K, Herbet M, Murias M, Piątkowska-Chmiel I. Senolytics: charting a new course or enhancing existing anti-tumor therapies? Cell Oncol (Dordr) 2025; 48:351-371. [PMID: 39633108 PMCID: PMC11996976 DOI: 10.1007/s13402-024-01018-5] [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] [Accepted: 11/21/2024] [Indexed: 12/07/2024] Open
Abstract
Cell senescence is a natural response within our organisms. Initially, it was considered an effective anti-tumor mechanism. However, it is now believed that while cell senescence initially acts as a robust barrier against tumor initiation, the subsequent accumulation of senescent cells can paradoxically promote cancer recurrence and cause damage to neighboring tissues. This intricate balance between cell proliferation and senescence plays a pivotal role in maintaining tissue homeostasis. Moreover, senescence cells secrete many bioactive molecules collectively termed the senescence-associated secretory phenotype (SASP), which can induce chronic inflammation, alter tissue architecture, and promote tumorigenesis through paracrine signaling. Among the myriads of compounds, senotherapeutic drugs have emerged as exceptionally promising candidates in anticancer treatment. Their ability to selectively target senescent cells while sparing healthy tissues represents a paradigm shift in therapeutic intervention, offering new avenues for personalized oncology medicine. Senolytics have introduced new therapeutic possibilities by enabling the targeted removal of senescent cells. As standalone agents, they can clear tumor cells in a senescent state and, when combined with chemo- or radiotherapy, eliminate residual senescent cancer cells after treatment. This dual approach allows for the intentional use of lower-dose therapies or the removal of unintended senescent cells post-treatment. Additionally, by targeting non-cancerous senescent cells, senolytics may help reduce tumor formation risk, limit recurrence, and slow disease progression. This article examines the mechanisms of cellular senescence, its role in cancer treatment, and the importance of senotherapy, with particular attention to the therapeutic potential of senolytic drugs.
Collapse
Affiliation(s)
- Konrad Czajkowski
- Department of Toxicology, Faculty of Pharmacy, Medical University of Lublin, Lublin, Poland
| | - Mariola Herbet
- Department of Toxicology, Faculty of Pharmacy, Medical University of Lublin, Lublin, Poland
| | - Marek Murias
- Department of Toxicology, Poznan University of Medical Sciences, Poznań, Poland
| | - Iwona Piątkowska-Chmiel
- Department of Toxicology, Faculty of Pharmacy, Medical University of Lublin, Lublin, Poland.
| |
Collapse
|
4
|
Jolles B, Stierlé V. Doxorubicin-induced senescence is modulated by the eukaryotic release factor 3a and its polyglycine expansion in HCT116 cells. Biochimie 2025; 233:81-87. [PMID: 40015471 DOI: 10.1016/j.biochi.2025.02.009] [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: 10/10/2024] [Revised: 02/17/2025] [Accepted: 02/24/2025] [Indexed: 03/01/2025]
Abstract
In humans, the release factor eRF3a exists in several forms that differ in the length of the polyglycine tract (7, 10, 11 or 12 glycines) in its N-terminal domain. For the 12-Gly eRF3a, an association with cancer risk and a decreased affinity for the cytoplasmic poly (A) binding protein have already been established. In this work, HCT116 colon cancer cells were treated with low doses of doxorubicin, which is known to induce senescence in these cells with high efficiency. The expression of p21 and p53 (senescence marker proteins) as well as lysosomal β-galactosidase activity were reduced when 12-Gly-eRF3a was overexpressed or eRF3a was depleted in cells. If low activity of mTORC1 pathway might be responsible for reduced senescence onset after eRF3a depletion, its activity is maintained in cells overexpressing 12-Gly-eRF3a. In both cases, a defect in termination efficiency could be involved.
Collapse
Affiliation(s)
- Béatrice Jolles
- Sorbonne Université, Institute of Biology Paris-Seine, IBPS, CNRS, UMR 8256, Biological Adaptation and Ageing, B2A, F 75005, Paris, France
| | - Vérène Stierlé
- Sorbonne Université, Institute of Biology Paris-Seine, IBPS, CNRS, UMR 8256, Biological Adaptation and Ageing, B2A, F 75005, Paris, France; Sorbonne Université, Institute of Biology Paris-Seine, IBPS, CNRS, UMR 8263, INSERM U1345 Development Adaptation and Ageing, Dev2A, F 75005, Paris, France.
| |
Collapse
|
5
|
Li L, Yang L, Shen L, Zhao Y, Wang L, Zhang H. Fat Mass and Obesity-Associated Protein Regulates Granulosa Cell Aging by Targeting Matrix Metalloproteinase-2 Gene Via an N6-Methyladenosine-YT521-B Homology Domain Family Member 2-Dependent Pathway in Aged Mice. Reprod Sci 2024; 31:3498-3511. [PMID: 38995602 PMCID: PMC11527923 DOI: 10.1007/s43032-024-01632-6] [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: 04/18/2024] [Accepted: 06/22/2024] [Indexed: 07/13/2024]
Abstract
In this study, we aimed to investigate the molecular mechanisms of RNA N6-methyladenosine (m6A) modification and how its associated proteins affect granulosa cell aging. A granulosa cell senescence model was constructed to detect the differences in total RNA m6A modification levels and the expression of related enzymes. Changes in downstream molecular expression and the effects on the cellular senescence phenotype were explored by repeatedly knocking down and overexpressing the key genes fat mass and obesity-associated protein (FTO), YT521-B homology domain family member 2 (YTHDF2), and matrix metalloproteinase-2 (MMP2). There was an increased total RNA m6A modification and decreased expression of the demethylase FTO and target gene MMP2 in senescent granulosa cells. FTO and MMP2 knockdown promoted granulosa cell senescence, whereas FTO and MMP2 overexpression retarded it. YTHDF2 and FTO can bind to the messenger RNA of MMP2. The extracellular signal-regulated kinase (ERK) pathway, which is downstream of MMP2, retarded the process of granulosa cell senescence through ERK activators. In granulosa cells, FTO can regulate the expression of MMP2 in an m6A-YTHDF2-dependent manner, influencing the activation status of the ERK pathway and contributing to the aging process of granulosa cells.
Collapse
Affiliation(s)
- Linshuang Li
- Reproductive Medicine Center, Tongji Hospital, Tongji Medicine College, Huazhong University of Science and Technology, Jiefang Avenue 1095#, Wuhan, 430030, People's Republic of China
| | - Le Yang
- Reproductive Medicine Center, Tongji Hospital, Tongji Medicine College, Huazhong University of Science and Technology, Jiefang Avenue 1095#, Wuhan, 430030, People's Republic of China
| | - Lin Shen
- Reproductive Medicine Center, Tongji Hospital, Tongji Medicine College, Huazhong University of Science and Technology, Jiefang Avenue 1095#, Wuhan, 430030, People's Republic of China
| | - Yiqing Zhao
- Reproductive Medicine Center, Tongji Hospital, Tongji Medicine College, Huazhong University of Science and Technology, Jiefang Avenue 1095#, Wuhan, 430030, People's Republic of China
| | - Lan Wang
- Reproductive Medicine Center, Tongji Hospital, Tongji Medicine College, Huazhong University of Science and Technology, Jiefang Avenue 1095#, Wuhan, 430030, People's Republic of China.
| | - Hanwang Zhang
- Reproductive Medicine Center, Tongji Hospital, Tongji Medicine College, Huazhong University of Science and Technology, Jiefang Avenue 1095#, Wuhan, 430030, People's Republic of China.
| |
Collapse
|
6
|
Pretto L, Nabinger E, Filippi-Chiela EC, Fraga LR. Cellular senescence in reproduction: a two-edged sword†. Biol Reprod 2024; 110:660-671. [PMID: 38480995 DOI: 10.1093/biolre/ioae025] [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: 08/08/2023] [Revised: 01/31/2024] [Accepted: 02/02/2024] [Indexed: 04/16/2024] Open
Abstract
Cellular senescence (CS) is the state when cells are no longer capable to divide even after stimulation with grown factors. Cells that begin to undergo CS stop in the cell cycle and enter a suspended state without committing to programmed cell death. These cells assume a specific phenotype and influence their microenvironment by secreting molecules and extracellular vesicles that are part of the so-called senescent cell-associated secretory phenotype (SASP). Cellular senescence is intertwined with physiological and pathological conditions in the human organism. In terms of reproduction, senescent cells are present from reproductive tissues and germ cells to gestational tissues, and participate from fertilization to delivery, going through adverse reproductive outcomes such as pregnancy losses. Furthermore, various SASP molecules are enriched in gestational tissues throughout pregnancy. Thus, the aim of this review is to provide a basis about the features and potential roles played by CS throughout the reproductive process, encompassing its implication in each step of it and proposing a way to manage it in adverse reproductive contexts.
Collapse
Affiliation(s)
- Luiza Pretto
- Post-Graduate Program in Medicine: Medical Sciences, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
- Laboratory of Genomic Medicine, Experimental Research Center, Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil
| | - Eduarda Nabinger
- Laboratory of Genomic Medicine, Experimental Research Center, Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil
| | - Eduardo Cremonesi Filippi-Chiela
- Department of Morphological Science, Institute of Health Sciences, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
- Post-Graduate Program in Cellular and Molecular Biology, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
- Post-Graduate Program in Biological Sciences: Physiology, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Lucas Rosa Fraga
- Post-Graduate Program in Medicine: Medical Sciences, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
- Laboratory of Genomic Medicine, Experimental Research Center, Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil
- Department of Morphological Science, Institute of Health Sciences, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
- Teratology Information System (SIAT), Medical Genetics Service, Hospital de Clínicas de Porto Alegre (HCPA), Porto Alegre, Brazil
| |
Collapse
|
7
|
Wang P, Konja D, Singh S, Zhang B, Wang Y. Endothelial Senescence: From Macro- to Micro-Vasculature and Its Implications on Cardiovascular Health. Int J Mol Sci 2024; 25:1978. [PMID: 38396653 PMCID: PMC10889199 DOI: 10.3390/ijms25041978] [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: 12/27/2023] [Revised: 02/02/2024] [Accepted: 02/03/2024] [Indexed: 02/25/2024] Open
Abstract
Endothelial cells line at the most inner layer of blood vessels. They act to control hemostasis, arterial tone/reactivity, wound healing, tissue oxygen, and nutrient supply. With age, endothelial cells become senescent, characterized by reduced regeneration capacity, inflammation, and abnormal secretory profile. Endothelial senescence represents one of the earliest features of arterial ageing and contributes to many age-related diseases. Compared to those in arteries and veins, endothelial cells of the microcirculation exhibit a greater extent of heterogeneity. Microcirculatory endothelial senescence leads to a declined capillary density, reduced angiogenic potentials, decreased blood flow, impaired barrier properties, and hypoperfusion in a tissue or organ-dependent manner. The heterogeneous phenotypes of microvascular endothelial cells in a particular vascular bed and across different tissues remain largely unknown. Accordingly, the mechanisms underlying macro- and micro-vascular endothelial senescence vary in different pathophysiological conditions, thus offering specific target(s) for therapeutic development of senolytic drugs.
Collapse
Affiliation(s)
- Peichun Wang
- State Key Laboratory of Pharmaceutical Biotechnology, The University of Hong Kong, Hong Kong SAR, China; (P.W.); (D.K.); (S.S.); (B.Z.)
- Department of Pharmacology and Pharmacy, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Daniels Konja
- State Key Laboratory of Pharmaceutical Biotechnology, The University of Hong Kong, Hong Kong SAR, China; (P.W.); (D.K.); (S.S.); (B.Z.)
- Department of Pharmacology and Pharmacy, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Sandeep Singh
- State Key Laboratory of Pharmaceutical Biotechnology, The University of Hong Kong, Hong Kong SAR, China; (P.W.); (D.K.); (S.S.); (B.Z.)
- Department of Pharmacology and Pharmacy, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Beijia Zhang
- State Key Laboratory of Pharmaceutical Biotechnology, The University of Hong Kong, Hong Kong SAR, China; (P.W.); (D.K.); (S.S.); (B.Z.)
- Department of Pharmacology and Pharmacy, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Yu Wang
- State Key Laboratory of Pharmaceutical Biotechnology, The University of Hong Kong, Hong Kong SAR, China; (P.W.); (D.K.); (S.S.); (B.Z.)
- Department of Pharmacology and Pharmacy, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| |
Collapse
|
8
|
Knopp RC, Erickson MA, Rhea EM, Reed MJ, Banks WA. Cellular senescence and the blood-brain barrier: Implications for aging and age-related diseases. Exp Biol Med (Maywood) 2023; 248:399-411. [PMID: 37012666 PMCID: PMC10281623 DOI: 10.1177/15353702231157917] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/05/2023] Open
Abstract
The blood-brain barrier (BBB) is a critical physiochemical interface that regulates communication between the brain and blood. It is comprised of brain endothelial cells which regulate the BBB's barrier and interface properties and is surrounded by supportive brain cell types including pericytes and astrocytes. Recent reports have suggested that the BBB undergoes dysfunction during normative aging and in disease. In this review, we consider the effect of cellular senescence, one of the nine hallmarks of aging, on the BBB. We first characterize known normative age-related changes at the BBB, and then evaluate changes in neurodegenerative diseases, with an emphasis on if/how cellular senescence is influencing these changes. We then discuss what insight has been gained from in vitro and in vivo studies of cellular senescence at the BBB. Finally, we evaluate mechanisms by which cellular senescence in peripheral pathologies can indirectly or directly affect BBB function.
Collapse
Affiliation(s)
- Rachel C Knopp
- Veterans Affairs Puget Sound Health Care
System, Geriatrics Research Education and Clinical Center (GRECC), Seattle, WA 98108,
USA
- Department of Medicine, Division of
Gerontology and Geriatric Medicine, University of Washington School of Medicine, Seattle, WA
98195, USA
| | - Michelle A Erickson
- Veterans Affairs Puget Sound Health Care
System, Geriatrics Research Education and Clinical Center (GRECC), Seattle, WA 98108,
USA
- Department of Medicine, Division of
Gerontology and Geriatric Medicine, University of Washington School of Medicine, Seattle, WA
98195, USA
| | - Elizabeth M Rhea
- Veterans Affairs Puget Sound Health Care
System, Geriatrics Research Education and Clinical Center (GRECC), Seattle, WA 98108,
USA
- Department of Medicine, Division of
Gerontology and Geriatric Medicine, University of Washington School of Medicine, Seattle, WA
98195, USA
| | - May J Reed
- Veterans Affairs Puget Sound Health Care
System, Geriatrics Research Education and Clinical Center (GRECC), Seattle, WA 98108,
USA
- Department of Medicine, Division of
Gerontology and Geriatric Medicine, University of Washington School of Medicine, Seattle, WA
98195, USA
| | - William A Banks
- Veterans Affairs Puget Sound Health Care
System, Geriatrics Research Education and Clinical Center (GRECC), Seattle, WA 98108,
USA
- Department of Medicine, Division of
Gerontology and Geriatric Medicine, University of Washington School of Medicine, Seattle, WA
98195, USA
| |
Collapse
|
9
|
Li Z, Tian M, Wang G, Cui X, Ma J, Liu S, Shen B, Liu F, Wu K, Xiao X, Zhu C. Senotherapeutics: An emerging approach to the treatment of viral infectious diseases in the elderly. Front Cell Infect Microbiol 2023; 13:1098712. [PMID: 37065192 PMCID: PMC10094634 DOI: 10.3389/fcimb.2023.1098712] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Accepted: 03/06/2023] [Indexed: 03/31/2023] Open
Abstract
In the context of the global COVID-19 pandemic, the phenomenon that the elderly have higher morbidity and mortality is of great concern. Existing evidence suggests that senescence and viral infection interact with each other. Viral infection can lead to the aggravation of senescence through multiple pathways, while virus-induced senescence combined with existing senescence in the elderly aggravates the severity of viral infections and promotes excessive age-related inflammation and multiple organ damage or dysfunction, ultimately resulting in higher mortality. The underlying mechanisms may involve mitochondrial dysfunction, abnormal activation of the cGAS-STING pathway and NLRP3 inflammasome, the role of pre-activated macrophages and over-recruited immune cells, and accumulation of immune cells with trained immunity. Thus, senescence-targeted drugs were shown to have positive effects on the treatment of viral infectious diseases in the elderly, which has received great attention and extensive research. Therefore, this review focused on the relationship between senescence and viral infection, as well as the significance of senotherapeutics for the treatment of viral infectious diseases.
Collapse
Affiliation(s)
- Zhiqiang Li
- Department of Clinical Laboratory, Institute of Translational Medicine, Renmin Hospital of Wuhan University, Wuhan, China
| | - Mingfu Tian
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan, China
| | - Guolei Wang
- Department of Clinical Laboratory, Institute of Translational Medicine, Renmin Hospital of Wuhan University, Wuhan, China
| | - Xianghua Cui
- Department of Clinical Laboratory, Institute of Translational Medicine, Renmin Hospital of Wuhan University, Wuhan, China
| | - Jun’e Ma
- Department of Clinical Laboratory, Institute of Translational Medicine, Renmin Hospital of Wuhan University, Wuhan, China
| | - Siyu Liu
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan, China
| | - Bingzheng Shen
- Department of Pharmacy, Renmin Hospital of Wuhan University, Wuhan, China
| | - Fang Liu
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan, China
| | - Kailang Wu
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan, China
| | - Xuan Xiao
- Department of Clinical Laboratory, Institute of Translational Medicine, Renmin Hospital of Wuhan University, Wuhan, China
- *Correspondence: Chengliang Zhu, ; Xuan Xiao,
| | - Chengliang Zhu
- Department of Clinical Laboratory, Institute of Translational Medicine, Renmin Hospital of Wuhan University, Wuhan, China
- *Correspondence: Chengliang Zhu, ; Xuan Xiao,
| |
Collapse
|
10
|
Geben LC, Brockman AA, Chalkley MBL, Sweet SR, Gallagher JE, Scheuing AL, Simerly RB, Ess KC, Irish JM, Ihrie RA. Dephosphorylation of 4EBP1/2 Induces Prenatal Neural Stem Cell Quiescence. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.02.14.528513. [PMID: 36824760 PMCID: PMC9948964 DOI: 10.1101/2023.02.14.528513] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/17/2023]
Abstract
A limiting factor in the regenerative capacity of the adult brain is the abundance and proliferative ability of neural stem cells (NSCs). Adult NSCs are derived from a subpopulation of embryonic NSCs that temporarily enter quiescence during mid-gestation and remain quiescent until postnatal reactivation. Here we present evidence that the mechanistic/mammalian target of rapamycin (mTOR) pathway regulates quiescence entry in embryonic NSCs of the developing forebrain. Throughout embryogenesis, two downstream effectors of mTOR, p-4EBP1/2 T37/46 and p-S6 S240/244, were mutually exclusive in NSCs, rarely occurring in the same cell. While 4EBP1/2 was phosphorylated in stem cells undergoing mitosis at the ventricular surface, S6 was phosphorylated in more differentiated cells migrating away from the ventricle. Phosphorylation of 4EBP1/2, but not S6, was responsive to quiescence induction in cultured embryonic NSCs. Further, inhibition of p-4EBP1/2, but not p-S6, was sufficient to induce quiescence. Collectively, this work offers new insight into the regulation of quiescence entry in embryonic NSCs and, thereby, correct patterning of the adult brain. These data suggest unique biological functions of specific posttranslational modifications and indicate that the preferential inhibition of such modifications may be a useful therapeutic approach in neurodevelopmental diseases where NSC numbers, proliferation, and differentiation are altered.
Collapse
Affiliation(s)
- Laura C. Geben
- Program in Pharmacology, Vanderbilt University, Nashville, TN, 37235, USA
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, TN, 37235, USA
| | - Asa A. Brockman
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, TN, 37235, USA
| | | | - Serena R. Sweet
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN 37235, USA
| | - Julia E. Gallagher
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, TN, 37235, USA
| | - Alexandra L. Scheuing
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, TN, 37235, USA
| | - Richard B. Simerly
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN 37235, USA
- Vanderbilt Brain Institute, Vanderbilt University, Nashville TN 37235, USA
| | - Kevin C. Ess
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, TN, 37235, USA
- Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN 37235, USA
- Vanderbilt Brain Institute, Vanderbilt University, Nashville TN 37235, USA
| | - Jonathan M. Irish
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, TN, 37235, USA
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN 37235, USA
- Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, TN 37235, USA
| | - Rebecca A. Ihrie
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, TN, 37235, USA
- Department of Neurological Surgery, Vanderbilt University Medical Center, Nashville, TN 37235, USA
- Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, TN 37235, USA
- Vanderbilt Brain Institute, Vanderbilt University, Nashville TN 37235, USA
| |
Collapse
|
11
|
Alhaddad L, Chuprov-Netochin R, Pustovalova M, Osipov AN, Leonov S. Polyploid/Multinucleated Giant and Slow-Cycling Cancer Cell Enrichment in Response to X-ray Irradiation of Human Glioblastoma Multiforme Cells Differing in Radioresistance and TP53/PTEN Status. Int J Mol Sci 2023; 24:ijms24021228. [PMID: 36674747 PMCID: PMC9865596 DOI: 10.3390/ijms24021228] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2022] [Revised: 12/27/2022] [Accepted: 01/04/2023] [Indexed: 01/11/2023] Open
Abstract
Radioresistance compromises the efficacy of radiotherapy for glioblastoma multiforme (GBM), the most devastating and common brain tumor. The present study investigated the relationship between radiation tolerance and formation of polyploid/multinucleated giant (PGCC/MGCC) and quiescent/senescent slow-cycling cancer cells in human U-87, LN-229, and U-251 cell lines differing in TP53/PTEN status and radioresistance. We found significant enrichment in MGCC populations of U-87 and LN-229 cell lines, and generation of numerous small mononuclear (called Raju cells, or RJ cells) U-87-derived cells that eventually form cell colonies, in a process termed neosis, in response to X-ray irradiation (IR) at single acute therapeutic doses of 2-6 Gy. For the first time, single-cell high-content imaging and analysis of Ki-67- and EdU-coupled fluorescence demonstrated that the IR exposure dose-dependently augments two distinct GBM cell populations. Bifurcation of Ki-67 staining suggests fast-cycling and slow-cycling populations with a normal-sized nuclear area, and with an enlarged nuclear area, including one resembling the size of PGCC/MGCCs, that likely underlie the highest radioresistance and propensity for repopulation of U-87 cells. Proliferative activity and anchorage-independent survival of GBM cell lines seem to be related to neosis, low level of apoptosis, fraction of prematurely stress-induced senescent MGCCs, and the expression of p63 and p73, members of p53 family transcription factors, but not to the mutant p53. Collectively, our data support the importance of the TP53wt/PTENmut genotype for the maintenance of cycling radioresistant U-87 cells to produce a significant amount of senescent MGCCs as an IR stress-induced adaptation response to therapeutic irradiation doses.
Collapse
Affiliation(s)
- Lina Alhaddad
- School of Biological and Medical Physics, Moscow Institute of Physics and Technology, 141700 Dolgoprudny, Russia
| | - Roman Chuprov-Netochin
- School of Biological and Medical Physics, Moscow Institute of Physics and Technology, 141700 Dolgoprudny, Russia
| | - Margarita Pustovalova
- School of Biological and Medical Physics, Moscow Institute of Physics and Technology, 141700 Dolgoprudny, Russia
- State Research Center-Burnasyan Federal Medical Biophysical Center of Federal Medical Biological Agency (SRC-FMBC), 123098 Moscow, Russia
| | - Andreyan N. Osipov
- School of Biological and Medical Physics, Moscow Institute of Physics and Technology, 141700 Dolgoprudny, Russia
- State Research Center-Burnasyan Federal Medical Biophysical Center of Federal Medical Biological Agency (SRC-FMBC), 123098 Moscow, Russia
- N.N. Semenov Federal Research Center for Chemical Physics, Russian Academy of Sciences, 119991 Moscow, Russia
- Correspondence:
| | - Sergey Leonov
- School of Biological and Medical Physics, Moscow Institute of Physics and Technology, 141700 Dolgoprudny, Russia
- Institute of Cell Biophysics, Russian Academy of Sciences, 142290 Pushchino, Russia
| |
Collapse
|
12
|
Lozano-Torres B, García-Fernández A, Domínguez M, Sancenón F, Blandez JF, Martínez-Máñez R. β-Galactosidase-Activatable Nile Blue-Based NIR Senoprobe for the Real-Time Detection of Cellular Senescence. Anal Chem 2022; 95:1643-1651. [PMID: 36580602 PMCID: PMC9850349 DOI: 10.1021/acs.analchem.2c04766] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Cellular senescence is a stable cell cycle arrest in response to stress or other damage stimuli to maintain tissue homeostasis. However, the accumulation of senescent cells can lead to the progression of various senescence-related disorders. In this paper, we describe the development of a β-galactosidase-activatable near-infrared (NIR) senoprobe, NBGal, for the detection of senescent cells based on the use of the FDA-approved Nile blue (NB) fluorophore. NBGal was validated in chemotherapeutic-induced senescence cancer models in vitro using SK-Mel 103 and 4T1 cell lines. In vivo monitoring of cellular senescence was evaluated in orthotopic triple-negative breast cancer-bearing mice treated with palbociclib to induce senescence. In all cases, NBGal exhibited a selective tracking of senescent cells mainly ascribed to the overexpressed β-galactosidase enzyme responsible for hydrolyzing the NBGal probe generating the highly emissive NB fluorophore. In this way, NBGal has proven to be a qualitative, rapid, and minimally invasive probe that allows the direct detection of senescent cells in vivo.
Collapse
Affiliation(s)
- Beatriz Lozano-Torres
- Instituto
Interuniversitario de Investigación de Reconocimiento Molecular
y Desarrollo Tecnológico (IDM), Universitat
Politècnica de València-Universitat de València, Camí de Vera S/N, Valencia 46022, Spain,Unidad
Mixta UPV-CIPF de Investigación en Mecanismos de Enfermedades
y Nanomedicina, Universitat Politècnica
de València, Centro de Investigación Príncipe
Felipe, C/ Eduardo Primo
Yúfera 3, Valencia 46012, Spain,CIBER
de Bioingeniería, Biomateriales y Nanomedicina, Av. Monforte de Lemos, 3-5, Pabellón
11, Planta 0, Madrid 28029, Spain
| | - Alba García-Fernández
- Instituto
Interuniversitario de Investigación de Reconocimiento Molecular
y Desarrollo Tecnológico (IDM), Universitat
Politècnica de València-Universitat de València, Camí de Vera S/N, Valencia 46022, Spain,Unidad
Mixta UPV-CIPF de Investigación en Mecanismos de Enfermedades
y Nanomedicina, Universitat Politècnica
de València, Centro de Investigación Príncipe
Felipe, C/ Eduardo Primo
Yúfera 3, Valencia 46012, Spain,CIBER
de Bioingeniería, Biomateriales y Nanomedicina, Av. Monforte de Lemos, 3-5, Pabellón
11, Planta 0, Madrid 28029, Spain,Unidad
Mixta de Investigación en Nanomedicina y Sensores, Universitat Politècnica de València, IIS La Fe, Av. Fernando Abril Martorell,
10, Torre A 7a̲ planta, Valencia 46026, Spain
| | - Marcia Domínguez
- Instituto
Interuniversitario de Investigación de Reconocimiento Molecular
y Desarrollo Tecnológico (IDM), Universitat
Politècnica de València-Universitat de València, Camí de Vera S/N, Valencia 46022, Spain,CIBER
de Bioingeniería, Biomateriales y Nanomedicina, Av. Monforte de Lemos, 3-5, Pabellón
11, Planta 0, Madrid 28029, Spain
| | - Félix Sancenón
- Instituto
Interuniversitario de Investigación de Reconocimiento Molecular
y Desarrollo Tecnológico (IDM), Universitat
Politècnica de València-Universitat de València, Camí de Vera S/N, Valencia 46022, Spain,Unidad
Mixta UPV-CIPF de Investigación en Mecanismos de Enfermedades
y Nanomedicina, Universitat Politècnica
de València, Centro de Investigación Príncipe
Felipe, C/ Eduardo Primo
Yúfera 3, Valencia 46012, Spain,CIBER
de Bioingeniería, Biomateriales y Nanomedicina, Av. Monforte de Lemos, 3-5, Pabellón
11, Planta 0, Madrid 28029, Spain,Unidad
Mixta de Investigación en Nanomedicina y Sensores, Universitat Politècnica de València, IIS La Fe, Av. Fernando Abril Martorell,
10, Torre A 7a̲ planta, Valencia 46026, Spain,
| | - Juan F. Blandez
- Instituto
Interuniversitario de Investigación de Reconocimiento Molecular
y Desarrollo Tecnológico (IDM), Universitat
Politècnica de València-Universitat de València, Camí de Vera S/N, Valencia 46022, Spain,CIBER
de Bioingeniería, Biomateriales y Nanomedicina, Av. Monforte de Lemos, 3-5, Pabellón
11, Planta 0, Madrid 28029, Spain,Unidad
Mixta de Investigación en Nanomedicina y Sensores, Universitat Politècnica de València, IIS La Fe, Av. Fernando Abril Martorell,
10, Torre A 7a̲ planta, Valencia 46026, Spain,
| | - Ramón Martínez-Máñez
- Instituto
Interuniversitario de Investigación de Reconocimiento Molecular
y Desarrollo Tecnológico (IDM), Universitat
Politècnica de València-Universitat de València, Camí de Vera S/N, Valencia 46022, Spain,Unidad
Mixta UPV-CIPF de Investigación en Mecanismos de Enfermedades
y Nanomedicina, Universitat Politècnica
de València, Centro de Investigación Príncipe
Felipe, C/ Eduardo Primo
Yúfera 3, Valencia 46012, Spain,CIBER
de Bioingeniería, Biomateriales y Nanomedicina, Av. Monforte de Lemos, 3-5, Pabellón
11, Planta 0, Madrid 28029, Spain,Unidad
Mixta de Investigación en Nanomedicina y Sensores, Universitat Politècnica de València, IIS La Fe, Av. Fernando Abril Martorell,
10, Torre A 7a̲ planta, Valencia 46026, Spain,
| |
Collapse
|
13
|
Yamashita G, Takano N, Kazama H, Tsukahara K, Miyazawa K. p53 regulates lysosomal membrane permeabilization as well as cytoprotective autophagy in response to DNA-damaging drugs. Cell Death Dis 2022; 8:502. [PMID: 36581628 PMCID: PMC9800408 DOI: 10.1038/s41420-022-01293-x] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2022] [Revised: 12/21/2022] [Accepted: 12/22/2022] [Indexed: 12/31/2022]
Abstract
Lysosomes are single-membraned organelles that mediate the intracellular degradation of macromolecules. Various stress can induce lysosomal membrane permeabilization (LMP), translocating intralysosomal components, such as cathepsins, to the cytoplasm, which induces lysosomal-dependent cell death (LDCD). This study reports that p53 regulates LMP in response to DNA-damaging drugs. Treating wild-type TP53 A549 cells with DNA-damaging drugs (namely, doxorubicin, carboplatin, and etoposide) induced LMP and accelerated cell death more rapidly than treating TP53-knockout (KO) A549 cells. This suggested p53-dependent LMP and LDCD induction in response to DNA damage. LMP was induced by p53-dependent BID upregulation and activation, followed by translocation of truncated BID to lysosomes. Simultaneously, autophagy for damaged lysosome elimination (lysophagy) was activated via the p53-mTOR-TEFB/TFE3 pathways in response to DNA damage. These data suggested the dichotomous nature of p53 for LMP regulation; LMP induction and repression via the p53-BID axis and p53-mTOR-TFEB/TFE3 pathway, respectively. Blocking autophagy with hydroxychloroquine or azithromycin as well as ATG5 KO enhanced LMP and LDCD induction after exposure to DNA-damaging drugs. Furthermore, lysosomal membrane stabilization using U18666A, a cholesterol transporter Niemann-Pick disease C1 (NPC1) inhibitor, suppressed LMP as well as LDCD in wild-type TP53, but not in TP53-KO, A549 cells. Thus, LMP is finely regulated by TP53 after exposure to DNA-damaging drugs.
Collapse
Affiliation(s)
- Gai Yamashita
- grid.412781.90000 0004 1775 2495Department of Otorhinolaryngology, Head and Neck Surgery, Tokyo Medical University Hospital, Shinjuku-ku, Tokyo, 160-0023 Japan
| | - Naoharu Takano
- grid.410793.80000 0001 0663 3325Department of Biochemistry, Tokyo Medical University, Shinjuku-ku, Tokyo, 160-8402 Japan
| | - Hiromi Kazama
- grid.410793.80000 0001 0663 3325Department of Biochemistry, Tokyo Medical University, Shinjuku-ku, Tokyo, 160-8402 Japan
| | - Kiyoaki Tsukahara
- grid.412781.90000 0004 1775 2495Department of Otorhinolaryngology, Head and Neck Surgery, Tokyo Medical University Hospital, Shinjuku-ku, Tokyo, 160-0023 Japan
| | - Keisuke Miyazawa
- grid.410793.80000 0001 0663 3325Department of Biochemistry, Tokyo Medical University, Shinjuku-ku, Tokyo, 160-8402 Japan
| |
Collapse
|
14
|
Liu X, Oh S, Kirschner MW. The uniformity and stability of cellular mass density in mammalian cell culture. Front Cell Dev Biol 2022; 10:1017499. [PMID: 36313562 PMCID: PMC9597509 DOI: 10.3389/fcell.2022.1017499] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2022] [Accepted: 09/23/2022] [Indexed: 12/04/2022] Open
Abstract
Cell dry mass is principally determined by the sum of biosynthesis and degradation. Measurable change in dry mass occurs on a time scale of hours. By contrast, cell volume can change in minutes by altering the osmotic conditions. How changes in dry mass and volume are coupled is a fundamental question in cell size control. If cell volume were proportional to cell dry mass during growth, the cell would always maintain the same cellular mass density, defined as cell dry mass dividing by cell volume. The accuracy and stability against perturbation of this proportionality has never been stringently tested. Normalized Raman Imaging (NoRI), can measure both protein and lipid dry mass density directly. Using this new technique, we have been able to investigate the stability of mass density in response to pharmaceutical and physiological perturbations in three cultured mammalian cell lines. We find a remarkably narrow mass density distribution within cells, that is, significantly tighter than the variability of mass or volume distribution. The measured mass density is independent of the cell cycle. We find that mass density can be modulated directly by extracellular osmolytes or by disruptions of the cytoskeleton. Yet, mass density is surprisingly resistant to pharmacological perturbations of protein synthesis or protein degradation, suggesting there must be some form of feedback control to maintain the homeostasis of mass density when mass is altered. By contrast, physiological perturbations such as starvation or senescence induce significant shifts in mass density. We have begun to shed light on how and why cell mass density remains fixed against some perturbations and yet is sensitive during transitions in physiological state.
Collapse
Affiliation(s)
| | | | - Marc W. Kirschner
- Department of Systems Biology, Harvard Medical School, Boston, MA, United States
| |
Collapse
|
15
|
Targeting Phospholipase D Pharmacologically Prevents Phagocytic Function Loss of Retinal Pigment Epithelium Cells Exposed to High Glucose Levels. Int J Mol Sci 2022; 23:ijms231911823. [PMID: 36233124 PMCID: PMC9570224 DOI: 10.3390/ijms231911823] [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] [Received: 09/09/2022] [Revised: 09/23/2022] [Accepted: 10/01/2022] [Indexed: 11/17/2022] Open
Abstract
We previously described the participation of canonical phospholipase D isoforms (PLD1 and PLD2) in the inflammatory response of retinal pigment epithelium (RPE) cells exposed to high glucose concentrations (HG). Here, we studied the role of the PLD pathway in RPE phagocytic function. For this purpose, ARPE-19 cells were exposed to HG (33 mM) or to normal glucose concentration (NG, 5.5 mM) and phagocytosis was measured using pHrodo™ green bioparticles® or photoreceptor outer segments (POS). HG exposure for 48 and 72 h reduced phagocytic function of ARPE-19 cells, and this loss of function was prevented when cells were treated with 5 μM of PLD1 (VU0359595 or PLD1i) or PLD2 (VU0285655-1 or PLD2i) selective inhibitors. Furthermore, PLD1i and PLD2i did not affect RPE phagocytosis under physiological conditions and prevented oxidative stress induced by HG. In addition, we demonstrated PLD1 and PLD2 expression in ABC cells, a novel human RPE cell line. Under physiological conditions, PLD1i and PLD2i did not affect ABC cell viability, and partial silencing of both PLDs did not affect ABC cell POS phagocytosis. In conclusion, PLD1i and PLD2i prevent the loss of phagocytic function of RPE cells exposed to HG without affecting RPE function or viability under non-inflammatory conditions.
Collapse
|
16
|
Wahlmueller M, Narzt MS, Missfeldt K, Arminger V, Krasensky A, Lämmermann I, Schaedl B, Mairhofer M, Suessner S, Wolbank S, Priglinger E. Establishment of In Vitro Models by Stress-Induced Premature Senescence for Characterizing the Stromal Vascular Niche in Human Adipose Tissue. Life (Basel) 2022; 12:life12101459. [PMID: 36294893 PMCID: PMC9605485 DOI: 10.3390/life12101459] [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] [Received: 09/02/2022] [Accepted: 09/14/2022] [Indexed: 12/02/2022] Open
Abstract
Acting as the largest energy reservoir in the body, adipose tissue is involved in longevity and progression of age-related metabolic dysfunction. Here, cellular senescence plays a central role in the generation of a pro-inflammatory environment and in the evolution of chronic diseases. Within the complexity of a tissue, identification and targeting of senescent cells is hampered by their heterogeneity. In this study, we generated stress-induced premature senescence 2D and 3D in vitro models for the stromal vascular niche of human adipose tissue. We established treatment conditions for senescence induction using Doxorubicin (Dox), starting from adipose-derived stromal/stem cells (ASCs), which we adapted to freshly isolated microtissue-stromal vascular fraction (MT-SVF), where cells are embedded within their native extracellular matrix. Senescence hallmarks for the established in vitro models were verified on different cellular levels, including morphology, cell cycle arrest, senescence-associated β-galactosidase activity (SA-βgal) and gene expression. Two subsequent exposures with 200 nM Dox for six days were suitable to induce senescence in our in vitro models. We demonstrated induction of senescence in the 2D in vitro models through SA-βgal activity, at the mRNA level (LMNB1, CDK1, p21) and additionally by G2/M phase cell cycle arrest in ASCs. Significant differences in Lamin B1 and p21 protein expression confirmed senescence in our MT-SVF 3D model. MT-SVF 3D cultures were composed of multiple cell types, including CD31, CD34 and CD68 positive cells, while cell death remained unaltered upon senescence induction. As heterogeneity and complexity of adipose tissue senescence is given by multiple cell types, our established senescence models that represent the perivascular niche embedded within its native extracellular matrix are highly relevant for future clinical studies.
Collapse
Affiliation(s)
- Marlene Wahlmueller
- Ludwig Boltzmann Institute for Traumatology in Cooperation with the AUVA, 1200 Vienna, Austria
- Austrian Cluster for Tissue Regeneration, 1200 Vienna, Austria
- MorphoMed GmbH, 1030 Vienna, Austria
| | - Marie-Sophie Narzt
- Ludwig Boltzmann Institute for Traumatology in Cooperation with the AUVA, 1200 Vienna, Austria
- Austrian Cluster for Tissue Regeneration, 1200 Vienna, Austria
- MorphoMed GmbH, 1030 Vienna, Austria
| | - Karin Missfeldt
- Ludwig Boltzmann Institute for Traumatology in Cooperation with the AUVA, 1200 Vienna, Austria
- Austrian Cluster for Tissue Regeneration, 1200 Vienna, Austria
| | - Verena Arminger
- Ludwig Boltzmann Institute for Traumatology in Cooperation with the AUVA, 1200 Vienna, Austria
- Austrian Cluster for Tissue Regeneration, 1200 Vienna, Austria
| | - Anna Krasensky
- Ludwig Boltzmann Institute for Traumatology in Cooperation with the AUVA, 1200 Vienna, Austria
- Austrian Cluster for Tissue Regeneration, 1200 Vienna, Austria
| | - Ingo Lämmermann
- Christian Doppler Laboratory for the Biotechnology of Skin Aging, Department of Biotechnology, Institute of Molecular Biotechnology, University of Natural Resources and Life Sciences, 1190 Vienna, Austria
- Rockfish Bio AG, 1010 Vienna, Austria
| | - Barbara Schaedl
- Ludwig Boltzmann Institute for Traumatology in Cooperation with the AUVA, 1200 Vienna, Austria
- Austrian Cluster for Tissue Regeneration, 1200 Vienna, Austria
- University Clinic of Dentistry, Medical University of Vienna, 1090 Vienna, Austria
| | - Mario Mairhofer
- Department of Hematology and Internal Oncology, Johannes Kepler University, 4020 Linz, Austria
| | - Susanne Suessner
- Austrian Red Cross Blood Transfusion Service for Upper Austria, 4020 Linz, Austria
| | - Susanne Wolbank
- Ludwig Boltzmann Institute for Traumatology in Cooperation with the AUVA, 1200 Vienna, Austria
- Austrian Cluster for Tissue Regeneration, 1200 Vienna, Austria
| | - Eleni Priglinger
- Ludwig Boltzmann Institute for Traumatology in Cooperation with the AUVA, 1200 Vienna, Austria
- Austrian Cluster for Tissue Regeneration, 1200 Vienna, Austria
- MorphoMed GmbH, 1030 Vienna, Austria
- Correspondence:
| |
Collapse
|
17
|
Skin-Aging Pigmentation: Who Is the Real Enemy? Cells 2022; 11:cells11162541. [PMID: 36010618 PMCID: PMC9406699 DOI: 10.3390/cells11162541] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2022] [Revised: 08/10/2022] [Accepted: 08/11/2022] [Indexed: 12/21/2022] Open
Abstract
Skin aging is induced and sustained by chronological aging and photoaging. Aging skin pigmentation such as mottled pigmentation (senile lentigo) and melasma are typical signs of photoaging. The skin, like other human organs, undergoes cellular senescence, and senescent cells in the skin increase with age. The crosstalk between melanocytes as pigmentary cells and other adjacent types of aged skin cells such as senescent fibroblasts play a role in skin-aging pigmentation. In this review, we provide an overview of cellular senescence during the skin-aging process. The discussion also includes cellular senescence related to skin-aging pigmentation and the therapeutic potential of regulating the senescence process.
Collapse
|
18
|
Pustovalova M, Blokhina T, Alhaddad L, Chigasova A, Chuprov-Netochin R, Veviorskiy A, Filkov G, Osipov AN, Leonov S. CD44+ and CD133+ Non-Small Cell Lung Cancer Cells Exhibit DNA Damage Response Pathways and Dormant Polyploid Giant Cancer Cell Enrichment Relating to Their p53 Status. Int J Mol Sci 2022; 23:ijms23094922. [PMID: 35563313 PMCID: PMC9101266 DOI: 10.3390/ijms23094922] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2022] [Revised: 04/25/2022] [Accepted: 04/26/2022] [Indexed: 01/10/2023] Open
Abstract
Cancer stem cells (CSCs) play a critical role in the initiation, progression and therapy relapse of many cancers including non-small cell lung cancer (NSCLC). Here, we aimed to address the question of whether the FACS-sorted CSC-like (CD44 + &CD133 +) vs. non-CSC (CD44-/CD133- isogenic subpopulations of p53wt A549 and p53null H1299 cells differ in terms of DNA-damage signaling and the appearance of "dormant" features, including polyploidy, which are early markers (predictors) of their sensitivity to genotoxic stress. X-ray irradiation (IR) at 5 Gy provoked significantly higher levels of the ATR-Chk1/Chk2-pathway activity in CD44-/CD133- and CD133+ subpopulations of H1299 cells compared to the respective subpopulations of A549 cells, which only excited ATR-Chk2 activation as demonstrated by the Multiplex DNA-Damage/Genotoxicity profiling. The CD44+ subpopulations did not demonstrate IR-induced activation of ATR, while significantly augmenting only Chk2 and Chk1/2 in the A549- and H1299-derived cells, respectively. Compared to the A549 cells, all the subpopulations of H1299 cells established an increased IR-induced expression of the γH2AX DNA-repair protein. The CD44-/CD133- and CD133+ subpopulations of the A549 cells revealed IR-induced activation of ATR-p53-p21 cell dormancy signaling-mediated pathway, while none of the CD44+ subpopulations of either cell line possessed any signs of such activity. Our data indicated, for the first time, the transcription factor MITF-FAM3C axis operative in p53-deficient H1299 cells, specifically their CD44+ and CD133+ populations, in response to IR, which warrants further investigation. The p21-mediated quiescence is likely the predominant surviving pathway in CD44-/CD133- and CD133+ populations of A549 cells as indicated by single-cell high-content imaging and analysis of Ki67- and EdU-coupled fluorescence after IR stress. SA-beta-galhistology revealed that cellular-stress-induced premature senescence (SIPS) likely has a significant influence on the temporary dormant state of H1299 cells. For the first time, we demonstrated polyploid giant and/or multinucleated cancer-cell (PGCC/MGCC) fractions mainly featuring the progressively augmenting Ki67low phenotype in CD44+ and CD133+ A549 cells at 24-48 h after IR. In contrast, the Ki67high phenotype enrichment in the same fractions of all the sorted H1299 cells suggested an increase in their cycling/heterochromatin reorganization activity after IR stress. Our results proposed that entering the "quiescence" state rather than p21-mediated SIPS may play a significant role in the survival of p53wt CSC-like NSCLC cells after IR. The results obtained are important for the selection of therapeutic schemes for the treatment of patients with NSCLC, depending on the functioning of the p53 system in tumor cells.
Collapse
Affiliation(s)
- Margarita Pustovalova
- School of Biological and Medical Physics, Moscow Institute of Physics and Technology, 141700 Dolgoprudny, Russia; (T.B.); (L.A.); (A.C.); (R.C.-N.); (G.F.); (A.N.O.)
- State Research Center—Burnasyan Federal Medical Biophysical Center of Federal Medical Biological Agency (SRC-FMBC), 123098 Moscow, Russia
- Correspondence: (M.P.); (S.L.)
| | - Taisia Blokhina
- School of Biological and Medical Physics, Moscow Institute of Physics and Technology, 141700 Dolgoprudny, Russia; (T.B.); (L.A.); (A.C.); (R.C.-N.); (G.F.); (A.N.O.)
- State Research Center—Burnasyan Federal Medical Biophysical Center of Federal Medical Biological Agency (SRC-FMBC), 123098 Moscow, Russia
- N.N. Semenov Federal Research Center for Chemical Physics, Russian Academy of Sciences, 119991 Moscow, Russia;
| | - Lina Alhaddad
- School of Biological and Medical Physics, Moscow Institute of Physics and Technology, 141700 Dolgoprudny, Russia; (T.B.); (L.A.); (A.C.); (R.C.-N.); (G.F.); (A.N.O.)
| | - Anna Chigasova
- School of Biological and Medical Physics, Moscow Institute of Physics and Technology, 141700 Dolgoprudny, Russia; (T.B.); (L.A.); (A.C.); (R.C.-N.); (G.F.); (A.N.O.)
- N.N. Semenov Federal Research Center for Chemical Physics, Russian Academy of Sciences, 119991 Moscow, Russia;
| | - Roman Chuprov-Netochin
- School of Biological and Medical Physics, Moscow Institute of Physics and Technology, 141700 Dolgoprudny, Russia; (T.B.); (L.A.); (A.C.); (R.C.-N.); (G.F.); (A.N.O.)
| | - Alexander Veviorskiy
- N.N. Semenov Federal Research Center for Chemical Physics, Russian Academy of Sciences, 119991 Moscow, Russia;
| | - Gleb Filkov
- School of Biological and Medical Physics, Moscow Institute of Physics and Technology, 141700 Dolgoprudny, Russia; (T.B.); (L.A.); (A.C.); (R.C.-N.); (G.F.); (A.N.O.)
- Laboratory of Medical Informatics, Novgorod Technical School, Yaroslav-the-Wise Novgorod State University, 173003 Veliky Novgorod, Russia
| | - Andreyan N. Osipov
- School of Biological and Medical Physics, Moscow Institute of Physics and Technology, 141700 Dolgoprudny, Russia; (T.B.); (L.A.); (A.C.); (R.C.-N.); (G.F.); (A.N.O.)
- State Research Center—Burnasyan Federal Medical Biophysical Center of Federal Medical Biological Agency (SRC-FMBC), 123098 Moscow, Russia
- N.N. Semenov Federal Research Center for Chemical Physics, Russian Academy of Sciences, 119991 Moscow, Russia;
| | - Sergey Leonov
- School of Biological and Medical Physics, Moscow Institute of Physics and Technology, 141700 Dolgoprudny, Russia; (T.B.); (L.A.); (A.C.); (R.C.-N.); (G.F.); (A.N.O.)
- Institute of Cell Biophysics, Russian Academy of Sciences, 142290 Pushchino, Russia
- Correspondence: (M.P.); (S.L.)
| |
Collapse
|
19
|
Lysosomal ATP Transporter SLC17A9 Controls Cell Viability via Regulating Cathepsin D. Cells 2022; 11:cells11050887. [PMID: 35269509 PMCID: PMC8909234 DOI: 10.3390/cells11050887] [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] [Received: 08/28/2021] [Revised: 02/26/2022] [Accepted: 02/28/2022] [Indexed: 12/04/2022] Open
Abstract
SLC17A9 (solute carrier family 17 member 9) functions as an ATP transporter in lysosomes as well as other secretory vesicles. SLC17A9 inhibition or silence leads to cell death. However, the molecular mechanisms causing cell death are unclear. In this study, we report that cell death induced by SLC17A9 deficiency is rescued by the transcription factor EB (TFEB), a master gene for lysosomal protein expression, suggesting that SLC17A9 deficiency may be the main cause of lysosome dysfunction, subsequently leading to cell death. Interestingly, Cathepsin D, a lysosomal aspartic protease, is inhibited by SLC17A9 deficiency. Heterologous expression of Cathepsin D successfully rescues lysosomal dysfunction and cell death induced by SLC17A9 deficiency. On the other hand, the activity of Cathepsin B, a lysosomal cysteine protease, is not altered by SLC17A9 deficiency, and Cathepsin B overexpression does not rescue lysosomal dysfunction and cell death induced by SLC17A9 deficiency. Our data suggest that lysosomal ATP and SLC17A9 play critical roles in lysosomal function and cell viability by regulating Cathepsin D activity.
Collapse
|
20
|
Asatryan A, Calandria JM, Kautzmann MAI, Jun B, Gordon WC, Do KV, Bhattacharjee S, Pham TL, Bermúdez V, Mateos MV, Heap J, Bazan NG. New Retinal Pigment Epithelial Cell Model to Unravel Neuroprotection Sensors of Neurodegeneration in Retinal Disease. Front Neurosci 2022; 16:926629. [PMID: 35873810 PMCID: PMC9301569 DOI: 10.3389/fnins.2022.926629] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Accepted: 05/30/2022] [Indexed: 01/02/2023] Open
Abstract
Retinal pigment epithelial (RPE) cells sustain photoreceptor integrity, and when this function is disrupted, retinal degenerations ensue. Herein, we characterize a new cell line from human RPE that we termed ABC. These cells remarkably recapitulate human eye native cells. Distinctive from other epithelia, RPE cells originate from the neural crest and follow a neural development but are terminally differentiated into "epithelial" type, thus sharing characteristics with their neuronal lineages counterparts. Additionally, they form microvilli, tight junctions, and honeycomb packing and express distinctive markers. In these cells, outer segment phagocytosis, phagolysosome fate, phospholipid metabolism, and lipid mediator release can be studied. ABC cells display higher resistance to oxidative stress and are protected from senescence through mTOR inhibition, making them more stable in culture. The cells are responsive to Neuroprotectin D1 (NPD1), which downregulates inflammasomes and upregulates antioxidant and anti-inflammatory genes. ABC gene expression profile displays close proximity to native RPE lineage, making them a reliable cell system to unravel signaling in uncompensated oxidative stress (UOS) and retinal degenerative disease to define neuroprotection sites.
Collapse
Affiliation(s)
- Aram Asatryan
- Neuroscience Center of Excellence, School of Medicine, Louisiana State University Health New Orleans, New Orleans, LA, United States
| | - Jorgelina M Calandria
- Neuroscience Center of Excellence, School of Medicine, Louisiana State University Health New Orleans, New Orleans, LA, United States
| | - Marie-Audrey I Kautzmann
- Neuroscience Center of Excellence, School of Medicine, Louisiana State University Health New Orleans, New Orleans, LA, United States
| | - Bokkyoo Jun
- Neuroscience Center of Excellence, School of Medicine, Louisiana State University Health New Orleans, New Orleans, LA, United States
| | - William C Gordon
- Neuroscience Center of Excellence, School of Medicine, Louisiana State University Health New Orleans, New Orleans, LA, United States
| | - Khanh V Do
- Neuroscience Center of Excellence, School of Medicine, Louisiana State University Health New Orleans, New Orleans, LA, United States
| | - Surjyadipta Bhattacharjee
- Neuroscience Center of Excellence, School of Medicine, Louisiana State University Health New Orleans, New Orleans, LA, United States
| | - Thang L Pham
- Neuroscience Center of Excellence, School of Medicine, Louisiana State University Health New Orleans, New Orleans, LA, United States
| | - Vicente Bermúdez
- Neuroscience Center of Excellence, School of Medicine, Louisiana State University Health New Orleans, New Orleans, LA, United States
| | - Melina Valeria Mateos
- Neuroscience Center of Excellence, School of Medicine, Louisiana State University Health New Orleans, New Orleans, LA, United States
| | - Jessica Heap
- Neuroscience Center of Excellence, School of Medicine, Louisiana State University Health New Orleans, New Orleans, LA, United States
| | - Nicolas G Bazan
- Neuroscience Center of Excellence, School of Medicine, Louisiana State University Health New Orleans, New Orleans, LA, United States
| |
Collapse
|
21
|
Csekes E, Račková L. Skin Aging, Cellular Senescence and Natural Polyphenols. Int J Mol Sci 2021; 22:12641. [PMID: 34884444 PMCID: PMC8657738 DOI: 10.3390/ijms222312641] [Citation(s) in RCA: 143] [Impact Index Per Article: 35.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Revised: 11/13/2021] [Accepted: 11/18/2021] [Indexed: 01/10/2023] Open
Abstract
The skin, being the barrier organ of the body, is constitutively exposed to various stimuli impacting its morphology and function. Senescent cells have been found to accumulate with age and may contribute to age-related skin changes and pathologies. Natural polyphenols exert many health benefits, including ameliorative effects on skin aging. By affecting molecular pathways of senescence, polyphenols are able to prevent or delay the senescence formation and, consequently, avoid or ameliorate aging and age-associated pathologies of the skin. This review aims to provide an overview of the current state of knowledge in skin aging and cellular senescence, and to summarize the recent in vitro studies related to the anti-senescent mechanisms of natural polyphenols carried out on keratinocytes, melanocytes and fibroblasts. Aged skin in the context of the COVID-19 pandemic will be also discussed.
Collapse
Affiliation(s)
- Erika Csekes
- Centre of Experimental Medicine, Institute of Experimental Pharmacology and Toxicology, Slovak Academy of Sciences, Dúbravská Cesta 9, 841 04 Bratislava, Slovakia
| | - Lucia Račková
- Centre of Experimental Medicine, Institute of Experimental Pharmacology and Toxicology, Slovak Academy of Sciences, Dúbravská Cesta 9, 841 04 Bratislava, Slovakia
| |
Collapse
|
22
|
Takenaka Y, Inoue I, Nakano T, Ikeda M, Kakinuma Y. Prolonged disturbance of proteostasis induces cellular senescence via temporal mitochondrial dysfunction and subsequent mitochondrial accumulation in human fibroblasts. FEBS J 2021; 289:1650-1667. [PMID: 34689411 DOI: 10.1111/febs.16249] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Revised: 09/15/2021] [Accepted: 10/22/2021] [Indexed: 01/18/2023]
Abstract
Proteolytic activity declines with age, resulting in the accumulation of aggregated proteins in aged organisms. To investigate how disturbance in proteostasis causes cellular senescence, we developed a stress-induced premature senescence (SIPS) model, in which normal human fibroblast MRC-5 cells were treated with the proteasome inhibitor MG132 or the vacuolar-type ATPase inhibitor bafilomycin A1 (BAFA1) for 5 days. Time-course studies revealed a significant increase in intracellular reactive oxygen species (ROS) and mitochondrial superoxide during and after drug treatment. Mitochondrial membrane potential initially decreased, suggesting temporal mitochondrial dysfunction during drug treatment, but was restored along with mitochondrial accumulation after drug treatment. AMP-activated protein kinase alpha was notably activated during treatment; thereafter, intracellular ATP levels significantly increased. SIPS induction by MG132 or BAFA1 was partially attenuated by co-treatment with vitamin E or rapamycin, in which the levels of ROS, mitochondrial accumulation, and protein aggregates were suppressed, implying the critical involvement of oxidative stress and mitochondrial function in SIPS progression. Rapamycin co-treatment also augmented the expression of HSP70 and activation of AKT, which could recover proteostasis and promote cell survival, respectively. Our study proposes a possible pathway from the disturbed proteostasis to cellular senescence via excess ROS production as well as functional and quantitative changes in mitochondria.
Collapse
Affiliation(s)
- Yasuhiro Takenaka
- Department of Physiology, Graduate School of Medicine, Nippon Medical School, Tokyo, Japan.,Department of Diabetes and Endocrinology, Saitama Medical University, Japan
| | - Ikuo Inoue
- Department of Diabetes and Endocrinology, Saitama Medical University, Japan
| | - Takanari Nakano
- Department of Biochemistry, Saitama Medical University, Japan
| | - Masaaki Ikeda
- Department of Physiology, Saitama Medical University, Japan
| | - Yoshihiko Kakinuma
- Department of Physiology, Graduate School of Medicine, Nippon Medical School, Tokyo, Japan
| |
Collapse
|
23
|
Liu H, Han X, Yang H, Cao Y, Zhang C, Du J, Diao S, Fan Z. GREM1 inhibits osteogenic differentiation, senescence and BMP transcription of adipose-derived stem cells. Connect Tissue Res 2021; 62:325-336. [PMID: 32151168 DOI: 10.1080/03008207.2020.1736054] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Purpose: Adipose-derived stem cells (ADSCs) are ideal for cell-based therapies to support bone regeneration. It is vital to understand the critical genes and molecular mechanisms involved in the functional regulation of ADSCs for enhancing bone regeneration. In the present study, we investigated the Gremlin 1 (GREM1) effect on ADSCs osteogenic differentiation and senescence.Materials and methods: The in vitro ADSCs osteogenic differentiation potential was evaluated by determining alkaline phosphatase (ALP) activity, mineralization ability, and the expression of osteogenic markers. Cell senescence is determined by SA-β-gal staining, telomerase assay, and the expression of aging markers.Results: GREM1 overexpression in ADSCs reduced ALP activity and mineralization, inhibited the expression of osteogenic related genes OCN, OPN, DSPP, DMP1, and BSP, and key transcription factors, RUNX2 and OSX. GREM1 knockdown in ADSCs enhanced ALP activity and mineralization, promoted the expression of OCN, OPN, DSPP, DMP1, BSP, RUNX2, and OSX. GREM1 overexpression in ADSCs reduced the percent SA-β-Gal positive cells, P16 and P53 expressions, and increased telomerase activity. GREM1 knockdown in ADSCs increased the percentage of SA-β-Gal positive cells, P16 and P53 expressions, and reduced telomerase activity. Furthermore, GREM1 reduced the mRNA expression levels of BMP2, BMP6, and BMP7.Conclusions: In summary, our findings suggested that GREM1 inhibited ADSCs senescence and osteogenic differentiation and antagonized BMP transcription.
Collapse
Affiliation(s)
- Huina Liu
- Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, Beijing Stomatological Hospital, School of Stomatology, Capital Medical University, Beijing, China
| | - Xiao Han
- Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, Beijing Stomatological Hospital, School of Stomatology, Capital Medical University, Beijing, China
| | - Haoqing Yang
- Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, Beijing Stomatological Hospital, School of Stomatology, Capital Medical University, Beijing, China
| | - Yangyang Cao
- Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, Beijing Stomatological Hospital, School of Stomatology, Capital Medical University, Beijing, China
| | - Chen Zhang
- Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, Beijing Stomatological Hospital, School of Stomatology, Capital Medical University, Beijing, China
| | - Juan Du
- Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, Beijing Stomatological Hospital, School of Stomatology, Capital Medical University, Beijing, China
| | - Shu Diao
- Department of Pediatric Dentistry, Beijing Stomatological Hospital, School of Stomatology, Capital Medical University, Beijing, China
| | - Zhipeng Fan
- Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, Beijing Stomatological Hospital, School of Stomatology, Capital Medical University, Beijing, China
| |
Collapse
|
24
|
Zhou D, Borsa M, Simon AK. Hallmarks and detection techniques of cellular senescence and cellular ageing in immune cells. Aging Cell 2021; 20:e13316. [PMID: 33524238 PMCID: PMC7884036 DOI: 10.1111/acel.13316] [Citation(s) in RCA: 72] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2020] [Revised: 01/03/2021] [Accepted: 01/09/2021] [Indexed: 12/15/2022] Open
Abstract
The ageing of the global population brings about unprecedented challenges. Chronic age-related diseases in an increasing number of people represent an enormous burden for health and social care. The immune system deteriorates during ageing and contributes to many of these age-associated diseases due to its pivotal role in pathogen clearance, tissue homeostasis and maintenance. Moreover, in order to develop treatments for COVID-19, we urgently need to acquire more knowledge about the aged immune system, as older adults are disproportionally and more severely affected. Changes with age lead to impaired responses to infections, malignancies and vaccination, and are accompanied by chronic, low-degree inflammation, which together is termed immunosenescence. However, the molecular and cellular mechanisms that underlie immunosenescence, termed immune cell senescence, are mostly unknown. Cellular senescence, characterised by an irreversible cell cycle arrest, is thought to be the cause of tissue and organismal ageing. Thus, better understanding of cellular senescence in immune populations at single-cell level may provide us with insight into how immune cell senescence develops over the life time of an individual. In this review, we will briefly introduce the phenotypic characterisation of aged innate and adaptive immune cells, which also contributes to overall immunosenescence, including subsets and function. Next, we will focus on the different hallmarks of cellular senescence and cellular ageing, and the detection techniques most suitable for immune cells. Applying these techniques will deepen our understanding of immune cell senescence and to discover potential druggable pathways, which can be modulated to reverse immune ageing.
Collapse
Affiliation(s)
- Dingxi Zhou
- The Kennedy Institute of RheumatologyUniversity of OxfordOxfordUK
| | - Mariana Borsa
- The Kennedy Institute of RheumatologyUniversity of OxfordOxfordUK
| | | |
Collapse
|
25
|
Abstract
Senescence is a state of long-term cell cycle arrest that arises in cells that have incurred sublethal damage. While senescent cells no longer replicate, they remain metabolically active and further develop unique and stable phenotypes that are not present in proliferating cells. On one hand, senescent cells increase in size, maintain an active mTORC1 complex, and produce and secrete a substantial amount of inflammatory proteins as part of the senescence-associated secretory phenotype (SASP). On the other hand, these progrowth phenotypes contrast with the p53-mediated growth arrest typical of senescent cells that is associated with nucleolar stress and an inhibition of rRNA processing and ribosome biogenesis. In sum, translation in senescent cells paradoxically comprises both a global repression of translation triggered by DNA damage and a select increase in the translation of specific proteins, including SASP factors.
Collapse
|
26
|
Singh BK, Tripathi M, Sandireddy R, Tikno K, Zhou J, Yen PM. Decreased autophagy and fuel switching occur in a senescent hepatic cell model system. Aging (Albany NY) 2020; 12:13958-13978. [PMID: 32712601 PMCID: PMC7425478 DOI: 10.18632/aging.103740] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2020] [Accepted: 06/18/2020] [Indexed: 02/05/2023]
Abstract
Although aging in the liver contributes to the development of chronic liver diseases such as NAFLD and insulin resistance, little is known about the molecular and metabolic details of aging in hepatic cells. To examine these issues, we used sequential oxidative stress with hydrogen peroxide to induce premature senescence in AML12 hepatic cells. The senescent cells exhibited molecular and metabolic signatures, increased SA-βGal and γH2A.X staining, and elevated senescence and pro-inflammatory gene expression that resembled livers from aged mice. Metabolic phenotyping showed fuel switching towards glycolysis and mitochondrial glutamine oxidation as well as impaired energy production. The senescent AML12 cells also had increased mTOR signaling and decreased autophagy which likely contributed to the fuel switching from β-oxidation that occurred in normal AML12 cells. Additionally, senescence-associated secretory phenotype (SASP) proteins from conditioned media of senescent cells sensitized normal AML12 cells to palmitate-induced toxicity, a known pathological effect of hepatic aging. In summary, we have generated senescent AML12 cells which displayed the molecular hallmarks of aging and also exhibited the aberrant metabolic phenotype, mitochondrial function, and cell signaling that occur in the aged liver.
Collapse
Affiliation(s)
- Brijesh Kumar Singh
- Cardiovascular and Metabolic Disorders Program, Duke-NUS Medical School, Singapore 169857, Singapore
| | - Madhulika Tripathi
- Cardiovascular and Metabolic Disorders Program, Duke-NUS Medical School, Singapore 169857, Singapore
| | - Reddemma Sandireddy
- Cardiovascular and Metabolic Disorders Program, Duke-NUS Medical School, Singapore 169857, Singapore
| | - Keziah Tikno
- Cardiovascular and Metabolic Disorders Program, Duke-NUS Medical School, Singapore 169857, Singapore
| | - Jin Zhou
- Cardiovascular and Metabolic Disorders Program, Duke-NUS Medical School, Singapore 169857, Singapore
| | - Paul Michael Yen
- Cardiovascular and Metabolic Disorders Program, Duke-NUS Medical School, Singapore 169857, Singapore.,Duke University School of Medicine, Durham, NC 27710, USA
| |
Collapse
|
27
|
Song P, Zhao Q, Zou MH. Targeting senescent cells to attenuate cardiovascular disease progression. Ageing Res Rev 2020; 60:101072. [PMID: 32298812 DOI: 10.1016/j.arr.2020.101072] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2019] [Revised: 04/07/2020] [Accepted: 04/09/2020] [Indexed: 12/19/2022]
Abstract
Cardiovascular disease (CVD) is the most common disease to increase as life expectancy increases. Most high-profile pharmacological treatments for age-related CVD have led to inefficacious results, implying that novel approaches to treating these pathologies are needed. Emerging data have demonstrated that senescent cardiovascular cells, which are characterized by irreversible cell cycle arrest and a distinct senescence-associated secretory phenotype, accumulate in aged or diseased cardiovascular systems, suggesting that they may impair cardiovascular function. This review discusses the evidence implicating senescent cells in cardiovascular ageing, the onset and progression of CVD, and the molecular mechanisms underlying cardiovascular cell senescence. We also review eradication of senescent cardiovascular cells by small-molecule-drug-mediated apoptosis and immune cell-mediated efferocytosis and toxicity as promising and precisely targeted therapeutics for CVD prevention and treatment.
Collapse
|
28
|
Blagosklonny MV. From causes of aging to death from COVID-19. Aging (Albany NY) 2020; 12:10004-10021. [PMID: 32534452 PMCID: PMC7346074 DOI: 10.18632/aging.103493] [Citation(s) in RCA: 78] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Accepted: 06/08/2020] [Indexed: 12/19/2022]
Abstract
COVID-19 is not deadly early in life, but mortality increases exponentially with age, which is the strongest predictor of mortality. Mortality is higher in men than in women, because men age faster, and it is especially high in patients with age-related diseases, such as diabetes and hypertension, because these diseases are manifestations of aging and a measure of biological age. At its deepest level, aging (a program-like continuation of developmental growth) is driven by inappropriately high cellular functioning. The hyperfunction theory of quasi-programmed aging explains why COVID-19 vulnerability (lethality) is an age-dependent syndrome, linking it to other age-related diseases. It also explains inflammaging and immunosenescence, hyperinflammation, hyperthrombosis, and cytokine storms, all of which are associated with COVID-19 vulnerability. Anti-aging interventions, such as rapamycin, may slow aging and age-related diseases, potentially decreasing COVID-19 vulnerability.
Collapse
|
29
|
Lin C, Li H, Liu J, Hu Q, Zhang S, Zhang N, Liu L, Dai Y, Cao D, Li X, Huang B, Lu J, Zhang Y. Arginine hypomethylation-mediated proteasomal degradation of histone H4-an early biomarker of cellular senescence. Cell Death Differ 2020; 27:2697-2709. [PMID: 32447347 DOI: 10.1038/s41418-020-0562-8] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2019] [Revised: 05/06/2020] [Accepted: 05/11/2020] [Indexed: 01/10/2023] Open
Abstract
Senescence is accompanied with histones level alteration; however, the roles and the mechanisms of histone reduction in cellular senescence are largely unknown. Protein arginine methyltransferase 1 (PRMT1) is the major enzyme that generates monomethyl and asymmetrical dimethyl arginine. Here we showed that abrogation of PRMT1-mediated senescence was accompanied with decreasing histone H4 level. Consistently, under multiple classic senescence models, H4 decreasing was also been found prior to the other 3 core histones. Noticeably, asymmetric demethylation of histone H4 at arginine 3 (H4R3me2as), catalyzed by PRMT1, was decreased prior to histone H4. In addition, we showed that the PRMT1-mediated H4R3me2as maintained H4 stability. Reduction of H4R3me2as level increased the interaction between proteasome activator PA200 and histone H4, which catalyzes the poly-ubiquitin-independent degradation of H4. Moreover, H4 degradation promoted nucleosome decomposition, resulting in increased senescence-associated genes transcription. Significantly, H4 was restored by 3 well-informed anti-aging drugs (metformin, rapamycin, and resveratrol) much earlier than other senescence markers detected under H2O2-induced senescence. Thus, we uncovered a novel function of H4R3me2as in modulation of cellular senescence via regulating H4 stability. This finding also points to the value of histone H4 as a senescence indicator and a potential anti-aging drug screening marker.
Collapse
Affiliation(s)
- Cong Lin
- The Key Laboratory of Molecular Epigenetics of Ministry of Education (MOE), Northeast Normal University, Changchun, China
| | - Hongxin Li
- The Key Laboratory of Molecular Epigenetics of Ministry of Education (MOE), Northeast Normal University, Changchun, China
| | - Jiwei Liu
- The Institute of Genetics and Cytology, Northeast Normal University, Changchun, China
| | - Qianying Hu
- The Key Laboratory of Molecular Epigenetics of Ministry of Education (MOE), Northeast Normal University, Changchun, China
| | - Shuai Zhang
- The Institute of Genetics and Cytology, Northeast Normal University, Changchun, China
| | - Na Zhang
- The Institute of Genetics and Cytology, Northeast Normal University, Changchun, China
| | - Lingxia Liu
- The Key Laboratory of Molecular Epigenetics of Ministry of Education (MOE), Northeast Normal University, Changchun, China
| | - Yingjie Dai
- The Key Laboratory of Molecular Epigenetics of Ministry of Education (MOE), Northeast Normal University, Changchun, China
| | - Donghui Cao
- Pathological Diagnostic Center, First Hospital of Jilin University, Changchun, China
| | - Xiaoxue Li
- The Key Laboratory of Molecular Epigenetics of Ministry of Education (MOE), Northeast Normal University, Changchun, China
| | - Baiqu Huang
- The Institute of Genetics and Cytology, Northeast Normal University, Changchun, China
| | - Jun Lu
- The Institute of Genetics and Cytology, Northeast Normal University, Changchun, China.
| | - Yu Zhang
- The Key Laboratory of Molecular Epigenetics of Ministry of Education (MOE), Northeast Normal University, Changchun, China.
| |
Collapse
|
30
|
Chen Z, Jiang K, Zou Z, Luo X, Lim CT, Wen C. High-throughput and label-free isolation of senescent murine mesenchymal stem cells. BIOMICROFLUIDICS 2020; 14:034106. [PMID: 32477445 PMCID: PMC7244328 DOI: 10.1063/5.0011925] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/03/2020] [Accepted: 04/30/2020] [Indexed: 06/11/2023]
Abstract
Under internal or external insults such as aging and oxidative stresses, cells are induced into a senescent state and stop cellular division permanently. As senescent cells (SnCs) accumulate, the regeneration capacity of biological tissue would be compromised, which has been found to be associated with a plethora of age-related disorders. Therefore, isolating SnCs becomes necessary. To address the lack of effective surface markers for SnCs isolation, a label-free microfluidic device was proposed in this paper, in which a spiral microchannel was deployed to isolate SnCs based on their size differences. We adopted a well-received cellular senescence model by exerting excessive oxidative stress to murine mesenchymal stem cells. This model was then validated through a series of SnCs characterizations including size measurement, p16INK4a expression level, senescence-associated beta-galactosidase, and doubling time. The senescence chip demonstrated an efficiency of 75% and viability over 85% at a flow rate of 5 ml/min. The average cell size from the inner outlet was 5 μm larger than that from the outer outlet. The isolated cells had a sixfold higher p16INK4a expression level. Overall, the chip had an area under curve of 0.719 in the receiver operating characteristic analysis, showing decent performance in sorting SnCs. By having the ability to perform size-based sorting at a high flow rate, such a microfluidic device can provide high-throughput and label-free isolation of SnCs. To further improve the isolation performance, the device can be modified to introduce additional physical biomarkers of SnCs such as stiffness. This device poses a good potential in purification for cytotherapy or estimation of biological age.
Collapse
Affiliation(s)
- Zhengkun Chen
- Department of Biomedical Engineering, Faculty of Engineering, The Hong Kong Polytechnic University, Hong Kong, China
| | - Kuan Jiang
- Mechanobiology Institute, National University of Singapore, Singapore 117411
| | - Zhou Zou
- Department of Biomedical Engineering, Faculty of Engineering, The Hong Kong Polytechnic University, Hong Kong, China
| | - Xiaohe Luo
- Department of Biomedical Engineering, Faculty of Engineering, The Hong Kong Polytechnic University, Hong Kong, China
| | | | - Chunyi Wen
- Department of Biomedical Engineering, Faculty of Engineering, The Hong Kong Polytechnic University, Hong Kong, China
| |
Collapse
|
31
|
Martínez-Carreres L, Puyal J, Leal-Esteban LC, Orpinell M, Castillo-Armengol J, Giralt A, Dergai O, Moret C, Barquissau V, Nasrallah A, Pabois A, Zhang L, Romero P, Lopez-Mejia IC, Fajas L. CDK4 Regulates Lysosomal Function and mTORC1 Activation to Promote Cancer Cell Survival. Cancer Res 2019; 79:5245-5259. [PMID: 31395606 DOI: 10.1158/0008-5472.can-19-0708] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2019] [Revised: 06/28/2019] [Accepted: 08/01/2019] [Indexed: 02/06/2023]
Abstract
Cyclin-dependent kinase 4 (CDK4) is well-known for its role in regulating the cell cycle, however, its role in cancer metabolism, especially mTOR signaling, is undefined. In this study, we established a connection between CDK4 and lysosomes, an emerging metabolic organelle crucial for mTORC1 activation. On the one hand, CDK4 phosphorylated the tumor suppressor folliculin (FLCN), regulating mTORC1 recruitment to the lysosomal surface in response to amino acids. On the other hand, CDK4 directly regulated lysosomal function and was essential for lysosomal degradation, ultimately regulating mTORC1 activity. Pharmacologic inhibition or genetic inactivation of CDK4, other than retaining FLCN at the lysosomal surface, led to the accumulation of undigested material inside lysosomes, which impaired the autophagic flux and induced cancer cell senescence in vitro and in xenograft models. Importantly, the use of CDK4 inhibitors in therapy is known to cause senescence but not cell death. To overcome this phenomenon and based on our findings, we increased the autophagic flux in cancer cells by using an AMPK activator in combination with a CDK4 inhibitor. The cotreatment induced autophagy (AMPK activation) and impaired lysosomal function (CDK4 inhibition), resulting in cell death and tumor regression. Altogether, we uncovered a previously unknown role for CDK4 in lysosomal biology and propose a novel therapeutic strategy to target cancer cells. SIGNIFICANCE: These findings uncover a novel function of CDK4 in lysosomal biology, which promotes cancer progression by activating mTORC1; targeting this function offers a new therapeutic strategy for cancer treatment.
Collapse
Affiliation(s)
| | - Julien Puyal
- Department of Fundamental Neurosciences, University of Lausanne, Lausanne, Switzerland
| | | | | | | | - Albert Giralt
- Center for Integrative Genomics, University of Lausanne, Lausanne, Switzerland
| | - Oleksandr Dergai
- Center for Integrative Genomics, University of Lausanne, Lausanne, Switzerland
| | - Catherine Moret
- Center for Integrative Genomics, University of Lausanne, Lausanne, Switzerland
| | - Valentin Barquissau
- Center for Integrative Genomics, University of Lausanne, Lausanne, Switzerland
| | - Anita Nasrallah
- Center for Integrative Genomics, University of Lausanne, Lausanne, Switzerland
| | - Angélique Pabois
- Department of Fundamental Oncology, University of Lausanne, Epalinges, Switzerland.,Ludwig Institute for Cancer Research, University of Lausanne, Epalinges, Switzerland
| | - Lianjun Zhang
- Department of Fundamental Oncology, University of Lausanne, Epalinges, Switzerland
| | - Pedro Romero
- Department of Fundamental Oncology, University of Lausanne, Epalinges, Switzerland
| | | | - Lluis Fajas
- Center for Integrative Genomics, University of Lausanne, Lausanne, Switzerland.
| |
Collapse
|
32
|
Bezzerri V, Piacenza F, Caporelli N, Malavolta M, Provinciali M, Cipolli M. Is cellular senescence involved in cystic fibrosis? Respir Res 2019; 20:32. [PMID: 30764828 PMCID: PMC6376730 DOI: 10.1186/s12931-019-0993-2] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2018] [Accepted: 01/31/2019] [Indexed: 02/06/2023] Open
Abstract
Pulmonary disease is the main cause of the morbidity and mortality of patients affected by cystic fibrosis (CF). The lung pathology is dominated by excessive recruitment of neutrophils followed by an exaggerated inflammatory process that has also been reported to occur in the absence of apparent pathogenic infections. Airway surface dehydration and mucus accumulation are the driving forces of this process. The continuous release of reactive oxygen species and proteases by neutrophils contributes to tissue damage, which eventually leads to respiratory insufficiency. CF has been considered a paediatric problem for several decades. Nevertheless, during the last 40 years, therapeutic options for CF have been greatly improved, turning CF into a chronic disease and extending the life expectancy of patients. Unfortunately, chronic inflammatory processes, which are characterized by a substantial release of cytokines and chemokines, along with ROS and proteases, can accelerate cellular senescence, leading to further complications in adulthood. The alterations and mechanisms downstream of CFTR functional defects that can stimulate cellular senescence remain unclear. However, while there are correlative data suggesting that cellular senescence may be implicated in CF, a causal or consequential relationship between cellular senescence and CF is still far from being established. Senescence can be both beneficial and detrimental. Senescence may suppress bacterial infections and cooperate with tissue repair. Additionally, it may act as an effective anticancer mechanism. However, it may also promote a pro-inflammatory environment, thereby damaging tissues and leading to chronic age-related diseases. In this review, we present the most current knowledge on cellular senescence and contextualize its possible involvement in CF.
Collapse
Affiliation(s)
- Valentino Bezzerri
- Cystic Fibrosis Center, Azienda Ospedaliera Universitaria Ospedali Riuniti, 60121, Ancona, Italy
| | - Francesco Piacenza
- Advanced Technology Center for Aging Research, Scientific Technological Area, IRCCS INRCA, 60121, Ancona, Italy
| | - Nicole Caporelli
- Cystic Fibrosis Center, Azienda Ospedaliera Universitaria Ospedali Riuniti, 60121, Ancona, Italy
| | - Marco Malavolta
- Advanced Technology Center for Aging Research, Scientific Technological Area, IRCCS INRCA, 60121, Ancona, Italy
| | - Mauro Provinciali
- Advanced Technology Center for Aging Research, Scientific Technological Area, IRCCS INRCA, 60121, Ancona, Italy
| | - Marco Cipolli
- Cystic Fibrosis Center, Azienda Ospedaliera Universitaria Ospedali Riuniti, 60121, Ancona, Italy.
| |
Collapse
|
33
|
Abstract
Rapamycin inhibits cell proliferation, yet preserves (re)-proliferative potential (RPP). RPP is a potential of quiescent cells that is lost in senescent cells. mTOR drives conversion from quiescence to senescence (geroconversion). By suppressing geroconversion, rapamycin preserves RPP. Geroconversion is characterized by proliferation-like levels of phospho-S6K/S6/4E-BP1 in nonproliferating cells arrested by p16 and/or p21. mTOR-driven geroconversion is associated with cellular hyperfunction, which in turn leads to organismal aging manifested by age-related diseases.
Collapse
|
34
|
Eberhardt K, Matthäus C, Marthandan S, Diekmann S, Popp J. Raman and infrared spectroscopy reveal that proliferating and quiescent human fibroblast cells age by biochemically similar but not identical processes. PLoS One 2018; 13:e0207380. [PMID: 30507927 PMCID: PMC6277109 DOI: 10.1371/journal.pone.0207380] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2018] [Accepted: 10/30/2018] [Indexed: 12/22/2022] Open
Abstract
Dermal fibroblast cells can adopt different cell states such as proliferation, quiescence, apoptosis or senescence, in order to ensure tissue homeostasis. Proliferating (dividing) cells pass through the phases of the cell cycle, while quiescent and senescent cells exist in a non-proliferating cell cycle-arrested state. However, the reversible quiescence state is in contrast to the irreversible senescence state. Long-term quiescent cells transit into senescence indicating that cells age also when not passing through the cell cycle. Here, by label-free in vitro vibrational spectroscopy, we studied the biomolecular composition of quiescent dermal fibroblast cells and compared them with those of proliferating and senescent cells. Spectra were examined by multivariate statistical analysis using a PLS-LDA classification model, revealing differences in the biomolecular composition between the cell states mainly associated with protein alterations (variations in the side chain residues of amino acids and protein secondary structure), but also within nucleic acids and lipids. We observed spectral changes in quiescent compared to proliferating cells, which increased with quiescence cultivation time. Raman and infrared spectroscopy, which yield complementary biochemical information, clearly distinguished contact-inhibited from serum-starved quiescent cells. Furthermore, the spectra displayed spectral differences between quiescent cells and proliferating cells, which had recovered from quiescence. This became more distinct with increasing quiescence cultivation time. When comparing proliferating, (in particular long-term) quiescent and senescent cells, we found that Raman as well as infrared spectroscopy can separate these three cellular states from each other due to differences in their biomolecular composition. Our spectroscopic analysis shows that proliferating and quiescent fibroblast cells age by similar but biochemically not identical processes. Despite their aging induced changes, over long time periods quiescent cells can return into the cell cycle. Finally however, the cell cycle arrest becomes irreversible indicating senescence.
Collapse
Affiliation(s)
- Katharina Eberhardt
- Spectroscopy and Imaging, Leibniz Institute of Photonic Technology, Jena, Germany
| | - Christian Matthäus
- Spectroscopy and Imaging, Leibniz Institute of Photonic Technology, Jena, Germany
- Institute for Physical Chemistry and Abbe Center of Photonics, Friedrich Schiller University, Jena, Germany
| | - Shiva Marthandan
- Department of Molecular Biology, Leibniz Institute on Aging – Fritz Lipmann Institute, Jena, Germany
| | - Stephan Diekmann
- Department of Molecular Biology, Leibniz Institute on Aging – Fritz Lipmann Institute, Jena, Germany
| | - Jürgen Popp
- Spectroscopy and Imaging, Leibniz Institute of Photonic Technology, Jena, Germany
- Institute for Physical Chemistry and Abbe Center of Photonics, Friedrich Schiller University, Jena, Germany
- * E-mail:
| |
Collapse
|
35
|
Hinze C, Boucrot E. Endocytosis in proliferating, quiescent and terminally differentiated cells. J Cell Sci 2018; 131:131/23/jcs216804. [PMID: 30504135 DOI: 10.1242/jcs.216804] [Citation(s) in RCA: 54] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Endocytosis mediates nutrient uptake, receptor internalization and the regulation of cell signaling. It is also hijacked by many bacteria, viruses and toxins to mediate their cellular entry. Several endocytic routes exist in parallel, fulfilling different functions. Most studies on endocytosis have used transformed cells in culture. However, as the majority of cells in an adult body have exited the cell cycle, our understanding is biased towards proliferating cells. Here, we review the evidence for the different pathways of endocytosis not only in dividing, but also in quiescent, senescent and terminally differentiated cells. During mitosis, residual endocytosis is dedicated to the internalization of caveolae and specific receptors. In non-dividing cells, clathrin-mediated endocytosis (CME) functions, but the activity of alternative processes, such as caveolae, macropinocytosis and clathrin-independent routes, vary widely depending on cell types and functions. Endocytosis supports the quiescent state by either upregulating cell cycle arrest pathways or downregulating mitogen-induced signaling, thereby inhibiting cell proliferation. Endocytosis in terminally differentiated cells, such as skeletal muscles, adipocytes, kidney podocytes and neurons, supports tissue-specific functions. Finally, uptake is downregulated in senescent cells, making them insensitive to proliferative stimuli by growth factors. Future studies should reveal the molecular basis for the differences in activities between the different cell states.
Collapse
Affiliation(s)
- Claudia Hinze
- Institute of Structural and Molecular Biology, Division of Biosciences, University College London, London WC1E 6BT, UK
| | - Emmanuel Boucrot
- Institute of Structural and Molecular Biology, Division of Biosciences, University College London, London WC1E 6BT, UK .,Institute of Structural and Molecular Biology, Department of Biological Sciences, Birkbeck College, London WC1E 7HX, UK
| |
Collapse
|
36
|
Detecting senescent fate in mesenchymal stem cells: a combined cytofluorimetric and ultrastructural approach. Biogerontology 2018; 19:401-414. [PMID: 30101381 DOI: 10.1007/s10522-018-9766-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2018] [Accepted: 08/06/2018] [Indexed: 02/08/2023]
Abstract
Senescence can impair the therapeutic potential of stem cells. In this study, senescence-associated morphofunctional changes in periosteum-derived progenitor cells (PDPCs) from old and young individuals were investigated by combining cytofluorimetry, immunohistochemistry, and transmission electron microscopy. Cell cycle analysis demonstrated a large number of G0/G1 phase cells in PDPCs from old subjects and a progressive accumulation of G0/G1 cells during passaging in cultures from young subjects. Cytofluorimetry documented significant changes in light scattering parameters and closely correlated with the ultrastructural features, especially changes in mitochondrial shape and autophagy, which are consistent with the mitochondrial-lysosomal axis theory of ageing. The combined morphological, biofunctional, and ultrastructural approach enhanced the flow cytometric study of PDPC ageing. We speculate that impaired autophagy, documented in replicative senescent and old PDPCs, reflect a switch from quiescence to senescence. Its demonstration in a tissue with limited turnover-like the cambium layer of the periosteum, where reversible quiescence is the normal stem cell state throughout life-adds a new piece to the regenerative medicine jigsaw in an ageing society.
Collapse
|
37
|
Schottler J, Randoll N, Lucius R, Caliebe A, Roider J, Klettner A. Long-term treatment with anti-VEGF does not induce cell aging in primary retinal pigment epithelium. Exp Eye Res 2018. [DOI: 10.1016/j.exer.2018.03.002] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
38
|
Abstract
Inhibitors of mTOR, including clinically available rapalogs such as rapamycin (Sirolimus) and Everolimus, are gerosuppressants, which suppress cellular senescence. Rapamycin slows aging and extends life span in a variety of species from worm to mammals. Rapalogs can prevent age-related diseases, including cancer, atherosclerosis, obesity, neurodegeneration and retinopathy and potentially rejuvenate stem cells, immunity and metabolism. Here, I further suggest how rapamycin can be combined with metformin, inhibitors of angiotensin II signaling (Losartan, Lisinopril), statins (simvastatin, atorvastatin), propranolol, aspirin and a PDE5 inhibitor. Rational combinations of these drugs with physical exercise and an anti-aging diet (Koschei formula) can maximize their anti-aging effects and decrease side effects.
Collapse
|
39
|
Hernandez-Segura A, Nehme J, Demaria M. Hallmarks of Cellular Senescence. Trends Cell Biol 2018; 28:436-453. [PMID: 29477613 DOI: 10.1016/j.tcb.2018.02.001] [Citation(s) in RCA: 1622] [Impact Index Per Article: 231.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2017] [Revised: 02/01/2018] [Accepted: 02/02/2018] [Indexed: 01/10/2023]
Abstract
Cellular senescence is a permanent state of cell cycle arrest that promotes tissue remodeling during development and after injury, but can also contribute to the decline of the regenerative potential and function of tissues, to inflammation, and to tumorigenesis in aged organisms. Therefore, the identification, characterization, and pharmacological elimination of senescent cells have gained attention in the field of aging research. However, the nonspecificity of current senescence markers and the existence of different senescence programs strongly limit these tasks. Here, we describe the molecular regulators of senescence phenotypes and how they are used for identifying senescent cells in vitro and in vivo. We also highlight the importance that these levels of regulations have in the development of therapeutic targets.
Collapse
Affiliation(s)
- Alejandra Hernandez-Segura
- European Research Institute for the Biology of Ageing (ERIBA), University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Jamil Nehme
- European Research Institute for the Biology of Ageing (ERIBA), University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Marco Demaria
- European Research Institute for the Biology of Ageing (ERIBA), University Medical Center Groningen, University of Groningen, Groningen, The Netherlands.
| |
Collapse
|
40
|
Induction of accelerated senescence by the microtubule-stabilizing agent peloruside A. Invest New Drugs 2017; 35:706-717. [PMID: 28733703 DOI: 10.1007/s10637-017-0493-5] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2017] [Accepted: 07/12/2017] [Indexed: 10/19/2022]
Abstract
Chemotherapeutic agents can induce accelerated senescence in tumor cells, an irreversible state of cell cycle arrest. Paclitaxel, a microtubule-stabilizing agent used to treat solid tumors of the breast, ovary, and lung and discodermolide, another stabilizing agent from a marine sponge, induce senescence in cultured cancer cells. The aim of this study was to determine if the microtubule-stabilizing agent peloruside A, a polyketide natural product from a marine sponge, can induce accelerated senescence in a breast cancer cell line MCF7. Doxorubicin, a DNA-damaging agent, paclitaxel, and discodermolide were used as positive controls. Senescence-associated-β-galactosidase activity was increased by peloruside A, similar to paclitaxel, discodermolde, and doxorubicin, with a potency heirarchy of doxorubicin > paclitaxel > discodermolide > peloruside, based on IC25 concentrations that inhibit proliferation. Clonogenic survival was significantly decreased by peloruside A, similar to doxorubicin and the two other microtubule-stabilizing agents. The tumor suppressor protein p53 increased after treatment, whereas pRb decreased in response to all four compounds. It was concluded that in addition to apoptosis, peloruside A causes accelerated senescence in a subpopulation of MCF7 cells that contributes to its potential anticancer activity in a breast cancer cell line.
Collapse
|
41
|
Leontieva OV, Blagosklonny MV. While reinforcing cell cycle arrest, rapamycin and Torins suppress senescence in UVA-irradiated fibroblasts. Oncotarget 2017; 8:109848-109856. [PMID: 29312653 PMCID: PMC5752566 DOI: 10.18632/oncotarget.17827] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2017] [Accepted: 05/02/2017] [Indexed: 12/20/2022] Open
Abstract
Sunlight predisposes to skin cancer and melanomas. Ultraviolet A (UVA), a long wave component of sunlight, can reach dermal fibroblasts. Here we studied UVA-induced senescence in human fibroblasts in vitro. It is known that senescence occurs, when cell cycle is arrested, but mTOR is still active, thus converting arrest to senescence (geroconversion). We showed that, while arresting cell cycle, UVA did not inhibit mTOR, enabling geroconversion. In UVA-treated cells, mTOR remained fully active. Rapamycin and Torins 1/ 2 prevented UVA-induced senescent phenotype, although they further re-enforced cell cycle arrest. Given that senescent stromal fibroblasts support tumorigenesis, we envision that mTOR inhibitors may potentially be used to prevent sunlight-caused tumors as well as skin photo-aging.
Collapse
Affiliation(s)
- Olga V Leontieva
- Cell Stress Biology, Roswell Park Cancer Institute, Buffalo, NY, USA
| | | |
Collapse
|
42
|
Mathiassen SG, De Zio D, Cecconi F. Autophagy and the Cell Cycle: A Complex Landscape. Front Oncol 2017; 7:51. [PMID: 28409123 PMCID: PMC5374984 DOI: 10.3389/fonc.2017.00051] [Citation(s) in RCA: 142] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2017] [Accepted: 03/10/2017] [Indexed: 12/11/2022] Open
Abstract
Autophagy is a self-degradation pathway, in which cytoplasmic material is sequestered in double-membrane vesicles and delivered to the lysosome for degradation. Under basal conditions, autophagy plays a homeostatic function. However, in response to various stresses, the pathway can be further induced to mediate cytoprotection. Defective autophagy has been linked to a number of human pathologies, including neoplastic transformation, even though autophagy can also sustain the growth of tumor cells in certain contexts. In recent years, a considerable correlation has emerged between autophagy induction and stress-related cell-cycle responses, as well as unexpected roles for autophagy factors and selective autophagic degradation in the process of cell division. These advances have obvious implications for our understanding of the intricate relationship between autophagy and cancer. In this review, we will discuss our current knowledge of the reciprocal regulation connecting the autophagy pathway and cell-cycle progression. Furthermore, key findings involving nonautophagic functions for autophagy-related factors in cell-cycle regulation will be addressed.
Collapse
Affiliation(s)
- Søs Grønbæk Mathiassen
- Cell Stress and Survival Unit, Danish Cancer Society Research Center, Copenhagen, Denmark
| | - Daniela De Zio
- Cell Stress and Survival Unit, Danish Cancer Society Research Center, Copenhagen, Denmark
| | - Francesco Cecconi
- Cell Stress and Survival Unit, Danish Cancer Society Research Center, Copenhagen, Denmark.,Department of Biology, University of Rome Tor Vergata, Rome, Italy.,Department of Pediatric Hematology and Oncology, Istituto di Ricovero e Cura a Carattere Scientifico Bambino Gesù Children's Hospital, Rome, Italy
| |
Collapse
|
43
|
Walenbergh SMA, Houben T, Rensen SS, Bieghs V, Hendrikx T, van Gorp PJ, Oligschlaeger Y, Jeurissen MLJ, Gijbels MJJ, Buurman WA, Vreugdenhil ACE, Greve JWM, Plat J, Hofker MH, Kalhan S, Pihlajamäki J, Lindsey P, Koek GH, Shiri-Sverdlov R. Plasma cathepsin D correlates with histological classifications of fatty liver disease in adults and responds to intervention. Sci Rep 2016; 6:38278. [PMID: 27922112 PMCID: PMC5138820 DOI: 10.1038/srep38278] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2016] [Accepted: 10/25/2016] [Indexed: 02/06/2023] Open
Abstract
Non-alcoholic steatohepatitis (NASH) is characterized by liver lipid accumulation and inflammation. The mechanisms that trigger hepatic inflammation are poorly understood and subsequently, no specific non-invasive markers exist. We previously demonstrated a reduction in the plasma lysosomal enzyme, cathepsin D (CatD), in children with NASH compared to children without NASH. Recent studies have raised the concept that non-alcoholic fatty liver disease (NAFLD) in adults is distinct from children due to a different histological pattern in the liver. Yet, the link between plasma CatD to adult NASH was not examined. In the current manuscript, we investigated whether plasma CatD in adults correlates with NASH development and regression. Biopsies were histologically evaluated for inflammation and NAFLD in three complementary cohorts of adults (total n = 248). CatD and alanine aminotransferase (ALT) were measured in plasma. Opposite to our previous observations with childhood NASH, we observed increased levels of plasma CatD in patients with NASH compared to adults without hepatic inflammation. Furthermore, after surgical intervention, we found a reduction of plasma CatD compared to baseline. Our observations highlight a distinct pathophysiology between NASH in children and adults. The observation that plasma CatD correlated with NASH development and regression is promising for NASH diagnosis.
Collapse
Affiliation(s)
- Sofie M A Walenbergh
- Molecular Genetics, General Surgery, Paediatrics, Pathology, Population Genetics, Human Biology, Maastricht University Medical Centre, 6200MD, Maastricht, The Netherlands
| | - Tom Houben
- Molecular Genetics, General Surgery, Paediatrics, Pathology, Population Genetics, Human Biology, Maastricht University Medical Centre, 6200MD, Maastricht, The Netherlands
| | - Sander S Rensen
- Molecular Genetics, General Surgery, Paediatrics, Pathology, Population Genetics, Human Biology, Maastricht University Medical Centre, 6200MD, Maastricht, The Netherlands
| | - Veerle Bieghs
- Molecular Genetics, General Surgery, Paediatrics, Pathology, Population Genetics, Human Biology, Maastricht University Medical Centre, 6200MD, Maastricht, The Netherlands
| | - Tim Hendrikx
- Molecular Genetics, General Surgery, Paediatrics, Pathology, Population Genetics, Human Biology, Maastricht University Medical Centre, 6200MD, Maastricht, The Netherlands
| | - Patrick J van Gorp
- Molecular Genetics, General Surgery, Paediatrics, Pathology, Population Genetics, Human Biology, Maastricht University Medical Centre, 6200MD, Maastricht, The Netherlands
| | - Yvonne Oligschlaeger
- Molecular Genetics, General Surgery, Paediatrics, Pathology, Population Genetics, Human Biology, Maastricht University Medical Centre, 6200MD, Maastricht, The Netherlands
| | - Mike L J Jeurissen
- Molecular Genetics, General Surgery, Paediatrics, Pathology, Population Genetics, Human Biology, Maastricht University Medical Centre, 6200MD, Maastricht, The Netherlands
| | - Marion J J Gijbels
- Molecular Genetics, General Surgery, Paediatrics, Pathology, Population Genetics, Human Biology, Maastricht University Medical Centre, 6200MD, Maastricht, The Netherlands
| | - Wim A Buurman
- Molecular Genetics, General Surgery, Paediatrics, Pathology, Population Genetics, Human Biology, Maastricht University Medical Centre, 6200MD, Maastricht, The Netherlands
| | - Anita C E Vreugdenhil
- Molecular Genetics, General Surgery, Paediatrics, Pathology, Population Genetics, Human Biology, Maastricht University Medical Centre, 6200MD, Maastricht, The Netherlands
| | - Jan Willem M Greve
- Surgery, Atrium Medical Center Parkstad, 6419PC, Heerlen, The Netherlands
| | - Jogchum Plat
- Molecular Genetics, General Surgery, Paediatrics, Pathology, Population Genetics, Human Biology, Maastricht University Medical Centre, 6200MD, Maastricht, The Netherlands
| | - Marten H Hofker
- Pathology and Medical Biology, Molecular Genetics, Medical Biology Section, University Medical Center Groningen, 9713GZ, Groningen, The Netherlands
| | - Satish Kalhan
- Pathobiology, Lerner Research Institute, The Cleveland Clinic Foundation, 9500 Euclid Avenue, Cleveland, USA
| | - Jussi Pihlajamäki
- Clinical Nutrition, University of Eastern Finland, FI-70211 Kuopio, Finland.,Clinical Nutrition and Obesity Center, Kuopio University Hospital, FI-70211 Kuopio, Finland
| | - Patrick Lindsey
- Molecular Genetics, General Surgery, Paediatrics, Pathology, Population Genetics, Human Biology, Maastricht University Medical Centre, 6200MD, Maastricht, The Netherlands
| | - Ger H Koek
- Internal Medicine, Division of Gastroenterology and Hepatology, Maastricht University Medical Centre, 6200MD, Maastricht, The Netherlands
| | - Ronit Shiri-Sverdlov
- Molecular Genetics, General Surgery, Paediatrics, Pathology, Population Genetics, Human Biology, Maastricht University Medical Centre, 6200MD, Maastricht, The Netherlands
| |
Collapse
|
44
|
Terzi MY, Izmirli M, Gogebakan B. The cell fate: senescence or quiescence. Mol Biol Rep 2016; 43:1213-1220. [DOI: 10.1007/s11033-016-4065-0] [Citation(s) in RCA: 97] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2016] [Accepted: 08/16/2016] [Indexed: 12/31/2022]
|
45
|
Leontieva OV, Demidenko ZN, Blagosklonny MV. Dual mTORC1/C2 inhibitors suppress cellular geroconversion (a senescence program). Oncotarget 2016; 6:23238-48. [PMID: 26177051 PMCID: PMC4695114 DOI: 10.18632/oncotarget.4836] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2015] [Accepted: 07/24/2015] [Indexed: 11/30/2022] Open
Abstract
In proliferating cells, mTOR is active and promotes cell growth. When the cell cycle is arrested, then mTOR converts reversible arrest to senescence (geroconversion). Rapamycin and other rapalogs suppress geroconversion, maintaining quiescence instead. Here we showed that ATP-competitive kinase inhibitors (Torin1 and PP242), which inhibit both mTORC1 and TORC2, also suppressed geroconversion. Despite inhibition of proliferation (in proliferating cells), mTOR inhibitors preserved re-proliferative potential (RP) in arrested cells. In p21-arrested cells, Torin 1 and PP242 detectably suppressed geroconversion at concentrations as low as 1-3 nM and 10-30 nM, reaching maximal gerosuppression at 30 nM and 300 nM, respectively. Near-maximal gerosuppression coincided with inhibition of p-S6K(T389) and p-S6(S235/236). Dual mTOR inhibitors prevented senescent morphology and hypertrophy. Our study warrants investigation into whether low doses of dual mTOR inhibitors will prolong animal life span and delay age-related diseases. A new class of potential anti-aging drugs can be envisioned.
Collapse
Affiliation(s)
- Olga V Leontieva
- Department of Cell Stress Biology, Roswell Park Cancer Institute, Buffalo, NY, USA
| | - Zoya N Demidenko
- Department of Cell Stress Biology, Roswell Park Cancer Institute, Buffalo, NY, USA
| | | |
Collapse
|
46
|
Rejuvenating immunity: "anti-aging drug today" eight years later. Oncotarget 2016; 6:19405-12. [PMID: 25844603 PMCID: PMC4637294 DOI: 10.18632/oncotarget.3740] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2015] [Accepted: 03/28/2015] [Indexed: 01/02/2023] Open
Abstract
The 2014 year ended with celebration: Everolimus, a rapamycin analog, was shown to improve immunity in old humans, heralding ‘a turning point’ in research and new era in human quest for immortality. Yet, this turning point was predicted a decade ago. But what will cause human death, when aging will be abolished?
Collapse
|
47
|
Sardiello M. Transcription factor EB: from master coordinator of lysosomal pathways to candidate therapeutic target in degenerative storage diseases. Ann N Y Acad Sci 2016; 1371:3-14. [PMID: 27299292 PMCID: PMC5032832 DOI: 10.1111/nyas.13131] [Citation(s) in RCA: 96] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2016] [Revised: 05/15/2016] [Accepted: 05/16/2016] [Indexed: 12/14/2022]
Abstract
The lysosome is the main catabolic hub of the cell. Owing to its role in fundamental processes such as autophagy, plasma membrane repair, mTOR signaling, and maintenance of cellular homeostasis, the lysosome has a profound influence on cellular metabolism and human health. Indeed, inefficient or impaired lysosomal function has been implicated in the pathogenesis of a number of degenerative diseases affecting various organs and tissues, most notably the brain, liver, and muscle. The discovery of the coordinated lysosomal expression and regulation (CLEAR) genetic program and its master controller, transcription factor EB (TFEB), has provided an unprecedented tool to study and manipulate lysosomal function. Most lysosome-based processes-including macromolecule degradation, autophagy, lysosomal exocytosis, and proteostasis-are under the transcriptional control of TFEB. Interestingly, impaired TFEB signaling has been suggested to be a contributing factor in the pathogenesis of several degenerative storage diseases. Preclinical studies based on TFEB exogenous expression to reinstate TFEB activity or promote CLEAR network-based lysosomal enhancement have highlighted TFEB as a candidate therapeutic target for the treatment of various degenerative storage diseases.
Collapse
Affiliation(s)
- Marco Sardiello
- Department of Molecular and Human Genetics, Baylor College of Medicine, and Jan and Dan Duncan Neurological Research Institute at Texas Children's Hospital, Houston, Texas
| |
Collapse
|
48
|
Leontieva OV, Blagosklonny MV. Tumor promoter-induced cellular senescence: cell cycle arrest followed by geroconversion. Oncotarget 2015; 5:12715-27. [PMID: 25587030 PMCID: PMC4350340 DOI: 10.18632/oncotarget.3011] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2014] [Accepted: 12/26/2014] [Indexed: 02/07/2023] Open
Abstract
Phorbol ester (PMA or TPA), a tumor promoter, can cause either proliferation or cell cycle arrest, depending on cellular context. For example, in SKBr3 breast cancer cells, PMA hyper-activates the MEK/MAPK pathway, thus inducing p21 and cell cycle arrest. Here we showed that PMA-induced arrest was followed by conversion to cellular senescence (geroconversion). Geroconversion was associated with active mTOR and S6 kinase (S6K). Rapamycin suppressed geroconversion, maintaining quiescence instead. In this model, PMA induced arrest (step one of a senescence program), whereas constitutively active mTOR drove geroconversion (step two). Without affecting Akt phosphorylation, PMA increased phosphorylation of S6K (T389) and S6 (S240/244), and that was completely prevented by rapamycin. Yet, T421/S424 and S235/236 (p-S6K and p-S6, respectively) phosphorylation became rapamycin-insensitive in the presence of PMA. Either MEK or mTOR was sufficient to phosphorylate these PMA-induced rapamycin-resistant sites because co-treatment with U0126 and rapamycin was required to abrogate them. We next tested whether activation of rapamycin-insensitive pathways would shift quiescence towards senescence. In HT-p21 cells, cell cycle arrest was caused by IPTG-inducible p21 and was spontaneously followed by mTOR-dependent geroconversion. Rapamycin suppressed geroconversion, whereas PMA partially counteracted the effect of rapamycin, revealing the involvement of rapamycin-insensitive gerogenic pathways. In normal RPE cells arrested by serum withdrawal, the mTOR/pS6 pathway was inhibited and cells remained quiescent. PMA transiently activated mTOR, enabling partial geroconversion. We conclude that PMA can initiate a senescent program by either inducing arrest or fostering geroconversion or both. Rapamycin can decrease gero-conversion by PMA, without preventing PMA-induced arrest. The tumor promoter PMA is a gero-promoter, which may be useful to study aging in mammals.
Collapse
Affiliation(s)
- Olga V Leontieva
- Department of Cell Stress Biology, Roswell Park Cancer Institute, Buffalo, NY, USA
| | | |
Collapse
|
49
|
Senescence as a general cellular response to stress: A mini-review. Exp Gerontol 2015; 72:124-8. [PMID: 26435346 DOI: 10.1016/j.exger.2015.09.021] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2015] [Revised: 09/24/2015] [Accepted: 09/30/2015] [Indexed: 12/22/2022]
Abstract
Cellular senescence was initially described as the phenomenon of limited cell divisions that normal cells in culture can undergo during long-term-cultivation. Later it was found that senescence may be induced by various stress factors. The intriguing resemblance between stress-induced and replicative senescence makes questionable the distinction between both types and suggests that the cellular senescence is a common outcome of stress response. Growing evidences support the idea that stress-induced senescence is the cell-type specific.
Collapse
|
50
|
Affiliation(s)
- Peiqing Sun
- Departments of Cell and Molecular Biology, The Scripps Research Institute, La Jolla, CA
| |
Collapse
|