1
|
Elmitwalli O, Darwish R, Al-Jabery L, Algahiny A, Roy S, Butler AE, Hasan AS. The Emerging Role of p21 in Diabetes and Related Metabolic Disorders. Int J Mol Sci 2024; 25:13209. [PMID: 39684919 DOI: 10.3390/ijms252313209] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2024] [Revised: 12/02/2024] [Accepted: 12/05/2024] [Indexed: 12/18/2024] Open
Abstract
In the context of cell cycle inhibition, anti-proliferation, and the dysregulation observed in certain cancer pathologies, the protein p21 assumes a pivotal role. p21 links DNA damage responses to cellular processes such as apoptosis, senescence, and cell cycle arrest, primarily functioning as a regulator of the cell cycle. However, accumulating empirical evidence suggests that p21 is both directly and indirectly linked to a number of different metabolic processes. Intriguingly, recent investigations indicate that p21 significantly contributes to the pathogenesis of diabetes. In this review, we present a comprehensive evaluation of the scientific literature regarding the involvement of p21 in metabolic processes, diabetes etiology, pancreatic function, glucose homeostasis, and insulin resistance. Furthermore, we provide an encapsulated overview of therapies that target p21 to alleviate metabolic disorders. A deeper understanding of the complex interrelationship between p21 and diabetes holds promise for informing current and future therapeutic strategies to address this rapidly escalating health crisis.
Collapse
Affiliation(s)
- Omar Elmitwalli
- Department of Medicine, Royal College of Surgeons in Ireland-Medical University of Bahrain Busaiteen, Adliya P.O. Box 15503, Bahrain
| | - Radwan Darwish
- Department of Medicine, Royal College of Surgeons in Ireland-Medical University of Bahrain Busaiteen, Adliya P.O. Box 15503, Bahrain
| | - Lana Al-Jabery
- Department of Medicine, Royal College of Surgeons in Ireland-Medical University of Bahrain Busaiteen, Adliya P.O. Box 15503, Bahrain
| | - Ahmed Algahiny
- Department of Medicine, Royal College of Surgeons in Ireland-Medical University of Bahrain Busaiteen, Adliya P.O. Box 15503, Bahrain
| | - Sornali Roy
- Department of Medicine, Royal College of Surgeons in Ireland-Medical University of Bahrain Busaiteen, Adliya P.O. Box 15503, Bahrain
| | - Alexandra E Butler
- Department of Postgraduate Studies and Research, Royal College of Surgeons in Ireland-Medical University of Bahrain Busaiteen, Adliya P.O. Box 15503, Bahrain
| | - Ammar S Hasan
- Department of Postgraduate Studies and Research, Royal College of Surgeons in Ireland-Medical University of Bahrain Busaiteen, Adliya P.O. Box 15503, Bahrain
| |
Collapse
|
2
|
Mansfield L, Ramponi V, Gupta K, Stevenson T, Mathew AB, Barinda AJ, Herbstein F, Morsli S. Emerging insights in senescence: pathways from preclinical models to therapeutic innovations. NPJ AGING 2024; 10:53. [PMID: 39578455 PMCID: PMC11584693 DOI: 10.1038/s41514-024-00181-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Accepted: 10/25/2024] [Indexed: 11/24/2024]
Abstract
Senescence is a crucial hallmark of ageing and a significant contributor to the pathology of age-related disorders. As committee members of the young International Cell Senescence Association (yICSA), we aim to synthesise recent advancements in the identification, characterisation, and therapeutic targeting of senescence for clinical translation. We explore novel molecular techniques that have enhanced our understanding of senescent cell heterogeneity and their roles in tissue regeneration and pathology. Additionally, we delve into in vivo models of senescence, both non-mammalian and mammalian, to highlight tools available for advancing the contextual understanding of in vivo senescence. Furthermore, we discuss innovative diagnostic tools and senotherapeutic approaches, emphasising their potential for clinical application. Future directions of senescence research are explored, underscoring the need for precise, context-specific senescence classification and the integration of advanced technologies such as machine learning, long-read sequencing, and multifunctional senoprobes and senolytics. The dual role of senescence in promoting tissue homoeostasis and contributing to chronic diseases highlights the complexity of targeting these cells for improved clinical outcomes.
Collapse
Affiliation(s)
- Luke Mansfield
- The Bateson Centre, School of Medicine and Population Health, The University of Sheffield, Western Bank, Sheffield, UK
| | - Valentina Ramponi
- Cellular Plasticity and Disease Group, Institute for Research in Biomedicine (IRB Barcelona), Barcelona Institute of Science and Technology (BIST), Barcelona, Spain
| | - Kavya Gupta
- Department of Cellular and Molecular Biology and Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA, USA
| | | | - Abraham Binoy Mathew
- Department of Developmental Biology and Genetics, Biological Sciences, Indian Institute of Science, Bangalore, India
| | - Agian Jeffilano Barinda
- Department of Pharmacology and Therapeutics, Faculty of Medicine, Universitas Indonesia, Jakarta, Indonesia
- Metabolic, Cardiovascular, and Aging Cluster, Indonesia Medical Education and Research Institute (IMERI), Faculty of Medicine, Universitas Indonesia, Jakarta, Indonesia
| | - Florencia Herbstein
- Instituto de Investigación en Biomedicina de Buenos Aires (IBioBA) - CONICET - Partner Institute of the Max Planck Society, Buenos Aires, Argentina.
| | - Samir Morsli
- Karolinska Institutet, Department of Cell and Molecular Biology, Biomedicum Q6A, Stockholm, Sweden.
| |
Collapse
|
3
|
Papismadov N, Levi N, Roitman L, Agrawal A, Ovadya Y, Cherqui U, Yosef R, Akiva H, Gal H, Krizhanovsky V. p21 regulates expression of ECM components and promotes pulmonary fibrosis via CDK4 and Rb. EMBO J 2024; 43:5360-5380. [PMID: 39349844 PMCID: PMC11574164 DOI: 10.1038/s44318-024-00246-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2024] [Revised: 08/05/2024] [Accepted: 09/05/2024] [Indexed: 11/20/2024] Open
Abstract
Fibrosis and accumulation of senescent cells are common tissue changes associated with aging. Here, we show that the CDK inhibitor p21 (CDKN1A), known to regulate the cell cycle and the viability of senescent cells, also controls the expression of extracellular matrix (ECM) components in senescent and proliferating cells of the fibrotic lung, in a manner dependent on CDK4 and Rb phosphorylation. p21 knockout protects mice from the induction of lung fibrosis. Moreover, inducible p21 silencing during fibrosis development alleviates disease pathology, decreasing the inflammatory response and ECM accumulation in the lung, and reducing the amount of senescent cells. Furthermore, p21 silencing limits fibrosis progression even when introduced during disease development. These findings show that one common mechanism regulates both cell cycle progression and expression of ECM components, and suggest that targeting p21 might be a new approach for treating age-related fibrotic pathologies.
Collapse
Affiliation(s)
- Nurit Papismadov
- Department of Molecular Cell Biology, The Weizmann Institute of Science, 7610001, Rehovot, Israel
| | - Naama Levi
- Department of Molecular Cell Biology, The Weizmann Institute of Science, 7610001, Rehovot, Israel
| | - Lior Roitman
- Department of Molecular Cell Biology, The Weizmann Institute of Science, 7610001, Rehovot, Israel
| | - Amit Agrawal
- Department of Molecular Cell Biology, The Weizmann Institute of Science, 7610001, Rehovot, Israel
| | - Yossi Ovadya
- Department of Molecular Cell Biology, The Weizmann Institute of Science, 7610001, Rehovot, Israel
| | - Ulysse Cherqui
- Department of Molecular Cell Biology, The Weizmann Institute of Science, 7610001, Rehovot, Israel
| | - Reut Yosef
- Department of Molecular Cell Biology, The Weizmann Institute of Science, 7610001, Rehovot, Israel
| | - Hagay Akiva
- Department of Molecular Cell Biology, The Weizmann Institute of Science, 7610001, Rehovot, Israel
| | - Hilah Gal
- Department of Molecular Cell Biology, The Weizmann Institute of Science, 7610001, Rehovot, Israel
| | - Valery Krizhanovsky
- Department of Molecular Cell Biology, The Weizmann Institute of Science, 7610001, Rehovot, Israel.
| |
Collapse
|
4
|
Pun R, Kumari N, Monieb RH, Wagh S, North BJ. BubR1 and SIRT2: Insights into aneuploidy, aging, and cancer. Semin Cancer Biol 2024; 106-107:201-216. [PMID: 39490401 PMCID: PMC11625622 DOI: 10.1016/j.semcancer.2024.10.005] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2024] [Revised: 10/22/2024] [Accepted: 10/23/2024] [Indexed: 11/05/2024]
Abstract
Aging is a significant risk factor for cancer which is due, in part, to heightened genomic instability. Mitotic surveillance proteins such as BubR1 play a pivotal role in ensuring accurate chromosomal segregation and preventing aneuploidy. BubR1 levels have been shown to naturally decline with age and its loss is associated with various age-related pathologies. Sirtuins, a class of NAD+-dependent deacylases, are implicated in cancer and genomic instability. Among them, SIRT2 acts as an upstream regulator of BubR1, offering a critical pathway that can potentially mitigate age-related diseases, including cancer. In this review, we explore BubR1 as a key regulator of cellular processes crucial for aging-related phenotypes. We delve into the intricate mechanisms through which BubR1 influences genomic stability and cellular senescence. Moreover, we highlight the role of NAD+ and SIRT2 in modulating BubR1 expression and function, emphasizing its potential as a therapeutic target. The interaction between BubR1 and SIRT2 not only serves as a fundamental regulatory pathway in cellular homeostasis but also represents a promising avenue for developing targeted therapies against age-related diseases, particularly cancer.
Collapse
Affiliation(s)
- Renju Pun
- Biomedical Sciences Department, Creighton University School of Medicine, Omaha, NE, USA
| | - Niti Kumari
- Biomedical Sciences Department, Creighton University School of Medicine, Omaha, NE, USA
| | - Rodaina Hazem Monieb
- Biomedical Sciences Department, Creighton University School of Medicine, Omaha, NE, USA
| | - Sachin Wagh
- Biomedical Sciences Department, Creighton University School of Medicine, Omaha, NE, USA
| | - Brian J North
- Biomedical Sciences Department, Creighton University School of Medicine, Omaha, NE, USA.
| |
Collapse
|
5
|
Chen S, Zhao W, Chen R, Sheng F, Gu Y, Hao S, Wu D, Lu B, Chen L, Wu Y, Xu Y, Han Y, Zhou L, Riazuddin SA, Fu Q, Yao K. FYCO1 regulates autophagy and senescence via PAK1/p21 in cataract. Arch Biochem Biophys 2024; 761:110180. [PMID: 39395618 DOI: 10.1016/j.abb.2024.110180] [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: 07/28/2024] [Revised: 09/21/2024] [Accepted: 10/10/2024] [Indexed: 10/14/2024]
Abstract
BACKGROUND ARC (Age-related cataract) is one of the leading causes of vision impairment and blindness; however, its pathogenesis remains unclear. FYCO1 (FYVE and coiled-coil domain containing 1) serves as an autophagy adaptor. The present study investigated the role of FYCO1 in cataract. METHODS Ultraviolet-B (UVB) irradiation was used to establish a cataract mice model. Hematoxylin and eosin (H&E) assay were used to observe lens morphology. Cell models were constructed by cultivating SRA 01/04 cells with H2O2 and UVB. Cell counting kit-8 (CCK8) and Senescence-associated β-galactosidase (SA-β-Gal) assay were performed to explore proliferation and senescence. The gene and protein expression were assessed by quantitative real-time PCR (qRT-PCR), Western blot and immunofluorescence staining. RESULTS We demonstrated lens structural damage and downregulation of FYCO1 in mice with UVB-induced cataracts. In vitro results revealed a deletion in autophagy levels along with the decrease of FYCO1 expression in human lens epithelial cells (HLECs) after H2O2 treatment, which was confirmed in vivo. The knockout of FYCO1 in the HLECs did not change basal autophagy and senescence but suppressed HLECs response in the induction of both. Further investigation indicated that FYCO1 knockout inhibited senescence and p21 levels by suppressing the expression of p21 activated kinase 1 (PAK1) in cataract cell models. CONCLUSIONS This study has newly characterized the role of FYCO1 in UVB-induced cataracts and in oxidative stress, both of which are associated with ARCs. A novel association between FYCO1 and PAK1/p21 in lens epithelial cell autophagy, senescence, and cataractogenesis also appears to have been established.
Collapse
Affiliation(s)
- Shuying Chen
- Eye Center, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Zhejiang Provincial Key Laboratory of Ophthalmology, Zhejiang Provincial Clinical Research Center for Eye Diseases, Zhejiang Provincial Engineering Institute on Eye Diseases, Hangzhou, Zhejiang, China
| | - Wei Zhao
- Eye Center, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Zhejiang Provincial Key Laboratory of Ophthalmology, Zhejiang Provincial Clinical Research Center for Eye Diseases, Zhejiang Provincial Engineering Institute on Eye Diseases, Hangzhou, Zhejiang, China; Eye Department, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, China
| | - Rongrong Chen
- Eye Center, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Zhejiang Provincial Key Laboratory of Ophthalmology, Zhejiang Provincial Clinical Research Center for Eye Diseases, Zhejiang Provincial Engineering Institute on Eye Diseases, Hangzhou, Zhejiang, China
| | - Feiyin Sheng
- Eye Center, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Zhejiang Provincial Key Laboratory of Ophthalmology, Zhejiang Provincial Clinical Research Center for Eye Diseases, Zhejiang Provincial Engineering Institute on Eye Diseases, Hangzhou, Zhejiang, China
| | - Yuzhou Gu
- Eye Center, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Zhejiang Provincial Key Laboratory of Ophthalmology, Zhejiang Provincial Clinical Research Center for Eye Diseases, Zhejiang Provincial Engineering Institute on Eye Diseases, Hangzhou, Zhejiang, China
| | - Shengjie Hao
- Eye Center, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Zhejiang Provincial Key Laboratory of Ophthalmology, Zhejiang Provincial Clinical Research Center for Eye Diseases, Zhejiang Provincial Engineering Institute on Eye Diseases, Hangzhou, Zhejiang, China
| | - Di Wu
- Eye Center, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Zhejiang Provincial Key Laboratory of Ophthalmology, Zhejiang Provincial Clinical Research Center for Eye Diseases, Zhejiang Provincial Engineering Institute on Eye Diseases, Hangzhou, Zhejiang, China
| | - Bing Lu
- Eye Center, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Zhejiang Provincial Key Laboratory of Ophthalmology, Zhejiang Provincial Clinical Research Center for Eye Diseases, Zhejiang Provincial Engineering Institute on Eye Diseases, Hangzhou, Zhejiang, China
| | - Lu Chen
- Eye Center, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Zhejiang Provincial Key Laboratory of Ophthalmology, Zhejiang Provincial Clinical Research Center for Eye Diseases, Zhejiang Provincial Engineering Institute on Eye Diseases, Hangzhou, Zhejiang, China
| | - Yuhao Wu
- Eye Center, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Zhejiang Provincial Key Laboratory of Ophthalmology, Zhejiang Provincial Clinical Research Center for Eye Diseases, Zhejiang Provincial Engineering Institute on Eye Diseases, Hangzhou, Zhejiang, China
| | - Yili Xu
- Eye Center, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Zhejiang Provincial Key Laboratory of Ophthalmology, Zhejiang Provincial Clinical Research Center for Eye Diseases, Zhejiang Provincial Engineering Institute on Eye Diseases, Hangzhou, Zhejiang, China
| | - Yu Han
- Eye Center, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Zhejiang Provincial Key Laboratory of Ophthalmology, Zhejiang Provincial Clinical Research Center for Eye Diseases, Zhejiang Provincial Engineering Institute on Eye Diseases, Hangzhou, Zhejiang, China
| | - Lei Zhou
- School of Optometry, Department of Applied Biology and Chemical Technology, Research Centre for SHARP Vision (RCSV), The Hong Kong Polytechnic University, Hong Kong; Centre for Eye and Vision Research (CEVR), 17W Hong Kong Science Park, Hong Kong
| | - S Amer Riazuddin
- The Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Qiuli Fu
- Eye Center, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Zhejiang Provincial Key Laboratory of Ophthalmology, Zhejiang Provincial Clinical Research Center for Eye Diseases, Zhejiang Provincial Engineering Institute on Eye Diseases, Hangzhou, Zhejiang, China.
| | - Ke Yao
- Eye Center, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Zhejiang Provincial Key Laboratory of Ophthalmology, Zhejiang Provincial Clinical Research Center for Eye Diseases, Zhejiang Provincial Engineering Institute on Eye Diseases, Hangzhou, Zhejiang, China.
| |
Collapse
|
6
|
Vanalderwiert L, Henry A, Wahart A, Carvajal Berrio DA, Brauchle EM, El Kaakour L, Schenke-Layland K, Brinckmann J, Steenbock H, Debelle L, Six I, Faury G, Jaisson S, Gillery P, Durlach V, Sartelet H, Maurice P, Bennasroune A, Martiny L, Duca L, Romier B, Blaise S. Metabolic syndrome-associated murine aortic wall stiffening is associated with premature elastic fibers aging. Am J Physiol Cell Physiol 2024; 327:C698-C715. [PMID: 38946422 DOI: 10.1152/ajpcell.00615.2023] [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: 11/12/2023] [Revised: 05/13/2024] [Accepted: 05/14/2024] [Indexed: 07/02/2024]
Abstract
Type 2 diabetes (T2D) constitutes a major public health problem, and despite prevention efforts, this pandemic disease is one of the deadliest diseases in the world. In 2022, 6.7 million patients with T2D died prematurely from vascular complications. Indeed, diabetes increases the risk of myocardial infarction or stroke eightfold. The identification of the molecular factors involved in the occurrence of cardiovascular complications and their prevention are therefore major axes. Our hypothesis is that factors brought into play during physiological aging appear prematurely with diabetes progression. Our study focused on the aging of the extracellular matrix (ECM), a major element in the maintenance of vascular homeostasis. We characterized the morphological and functional aspects of aorta, with a focus on the collagen and elastic fibers of diabetic mice aged from 6 mo to nondiabetic mice aged 6 mo and 20 mo. The comparison with the two nondiabetic models (young and old) highlighted an exacerbated activity of proteases, which could explain a disturbance in the collagen accumulation and an excessive degradation of elastic fibers. Moreover, the generation of circulating elastin-derived peptides reflects premature aging of the ECM. These extracellular elements contribute to the appearance of vascular rigidity, often the origin of pathologies such as hypertension and atherosclerosis. In conclusion, we show that diabetic mice aged 6 mo present the same characteristics of ECM wear as those observed in mice aged 20 mo. This accelerated aortic wall remodeling could then explain the early onset of cardiovascular diseases and, therefore, the premature death of patients with T2D.NEW & NOTEWORTHY Aortic elastic fibers of young (6-mo old) individuals with diabetes degrade prematurely and exhibit an appearance like that found in aged (20-mo old) nondiabetic mice. Exacerbated elastolysis and elastin-derived peptide production are characteristic elements, contributing to early aortic wall rigidity and hypertension development. Therefore, limiting this early aging could be a judicious therapeutic approach to reduce cardiovascular complications and premature death in patients with diabetes.
Collapse
MESH Headings
- Animals
- Elastic Tissue/metabolism
- Elastic Tissue/pathology
- Vascular Stiffness/physiology
- Mice
- Aorta/metabolism
- Aorta/pathology
- Aorta/physiopathology
- Mice, Inbred C57BL
- Extracellular Matrix/metabolism
- Extracellular Matrix/pathology
- Male
- Metabolic Syndrome/metabolism
- Metabolic Syndrome/pathology
- Metabolic Syndrome/physiopathology
- Elastin/metabolism
- Collagen/metabolism
- Diabetes Mellitus, Experimental/metabolism
- Diabetes Mellitus, Experimental/pathology
- Diabetes Mellitus, Experimental/physiopathology
- Aging/pathology
- Aging/metabolism
- Diabetes Mellitus, Type 2/metabolism
- Diabetes Mellitus, Type 2/pathology
- Diabetes Mellitus, Type 2/physiopathology
- Aging, Premature/metabolism
- Aging, Premature/pathology
- Aging, Premature/physiopathology
Collapse
Affiliation(s)
| | - Auberi Henry
- UMR CNRS 7369 MEDyc, University of Reims Champagne-Ardenne, Reims, France
| | - Amandine Wahart
- UMR CNRS 7369 MEDyc, University of Reims Champagne-Ardenne, Reims, France
| | - Daniel A Carvajal Berrio
- Department for Medical Technologies and Regenerative Medicine, Institute of Biomedical Engineering, Eberhard Karls University Tübingen, Tübingen, Germany
| | - Eva M Brauchle
- Department for Medical Technologies and Regenerative Medicine, Institute of Biomedical Engineering, Eberhard Karls University Tübingen, Tübingen, Germany
- NMI Natural and Medical Sciences Institute, Reutlingen, Germany
| | - Lara El Kaakour
- UMR CNRS 7369 MEDyc, University of Reims Champagne-Ardenne, Reims, France
| | - Katja Schenke-Layland
- Department for Medical Technologies and Regenerative Medicine, Institute of Biomedical Engineering, Eberhard Karls University Tübingen, Tübingen, Germany
- NMI Natural and Medical Sciences Institute, Reutlingen, Germany
- Division of Cardiology, Department of Medicine/Cardiology, Cardiovascular Research Laboratories, David Geffen School of Medicine at UCLA, Los Angeles, California, United States
| | - Juergen Brinckmann
- Institute of Virology and Cell Biology, University of Lübeck, Lübeck, Germany
| | - Heiko Steenbock
- Institute of Virology and Cell Biology, University of Lübeck, Lübeck, Germany
| | - Laurent Debelle
- UMR CNRS 7369 MEDyc, University of Reims Champagne-Ardenne, Reims, France
| | - Isabelle Six
- Research Unit 7517, Pathophysiological Mechanisms and Consequences of Cardiovascular Calcifications (MP3CV), University of Picardie Jules Verne, Amiens, France
| | - Gilles Faury
- University Grenoble Alpes, INSERM, CHU Grenoble Alpes, Grenoble, France
| | - Stéphane Jaisson
- UMR CNRS 7369 MEDyc, University of Reims Champagne-Ardenne, Reims, France
- Biochemistry Department, University Hospital of Reims, Reims, France
| | - Philippe Gillery
- UMR CNRS 7369 MEDyc, University of Reims Champagne-Ardenne, Reims, France
- Biochemistry Department, University Hospital of Reims, Reims, France
| | - Vincent Durlach
- UMR CNRS 7369 MEDyc, University of Reims Champagne-Ardenne, Reims, France
- Cardiovascular and Thoracic Division, Hôpital Robert Debré, Reims, France
| | - Hervé Sartelet
- UMR CNRS 7369 MEDyc, University of Reims Champagne-Ardenne, Reims, France
| | - Pascal Maurice
- UMR CNRS 7369 MEDyc, University of Reims Champagne-Ardenne, Reims, France
| | - Amar Bennasroune
- UMR CNRS 7369 MEDyc, University of Reims Champagne-Ardenne, Reims, France
| | - Laurent Martiny
- UMR CNRS 7369 MEDyc, University of Reims Champagne-Ardenne, Reims, France
| | - Laurent Duca
- UMR CNRS 7369 MEDyc, University of Reims Champagne-Ardenne, Reims, France
| | - Béatrice Romier
- UMR CNRS 7369 MEDyc, University of Reims Champagne-Ardenne, Reims, France
| | - Sébastien Blaise
- UMR CNRS 7369 MEDyc, University of Reims Champagne-Ardenne, Reims, France
| |
Collapse
|
7
|
Matin M, Joshi T, Wang D, Tzvetkov NT, Matin FB, Wierzbicka A, Jóźwik A, Horbańczuk JO, Atanasov AG. Effects of Ginger ( Zingiber officinale) on the Hallmarks of Aging. Biomolecules 2024; 14:940. [PMID: 39199328 PMCID: PMC11352747 DOI: 10.3390/biom14080940] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Revised: 07/22/2024] [Accepted: 07/29/2024] [Indexed: 09/01/2024] Open
Abstract
Ginger (Zingiber officinale Roscoe) is broadly used as a traditional remedy and food ingredient, and numerous preclinical and clinical studies have demonstrated health benefits in a range of age-related disorders. Moreover, longevity-promoting effects have been demonstrated in several (preclinical) research models. With this work, we aimed to comprehensively review the reported effects of ginger and its bioactive constituents on the twelve established hallmarks of aging, with the ultimate goal of gaining a deeper understanding of the potential for future interventions in the area of longevity-extension and counteracting of aging-related diseases. The reviewed literature supports the favorable effects of ginger and some of its constituents on all twelve hallmarks of aging, with a particularly high number of animal research studies indicating counteraction of nutrient-sensing dysregulations, mitochondrial dysfunction, chronic inflammation, and dysbiosis. On this background, validation in human clinical trials is still insufficient or is entirely missing, with the exception of some studies indicating positive effects on deregulated nutrient-sensing, chronic inflammation, and dysbiosis. Thus, the existing body of literature clearly supports the potential of ginger to be further studied in clinical trials as a supplement for the promotion of both lifespan and health span.
Collapse
Affiliation(s)
- Maima Matin
- Institute of Genetics and Animal Biotechnology of the Polish Academy of Sciences, Jastrzebiec, 05-552 Magdalenka, Poland; (M.M.); (A.W.); (A.J.); (J.O.H.)
| | - Tanuj Joshi
- Department of Pharmaceutical Sciences, Bhimtal, Kumaun University, Nainital 263002, India;
| | - Dongdong Wang
- Centre for Metabolism, Obesity and Diabetes Research, Department of Medicine, McMaster University, 1280 Main Street West, Hamilton, ON L8S 4K1, Canada;
| | - Nikolay T. Tzvetkov
- Department of Biochemical Pharmacology and Drug Design, Institute of Molecular Biology “Roumen Tsanev”, Bulgarian Academy of Sciences, 1113 Sofia, Bulgaria;
| | - Farhan Bin Matin
- Department of Pharmacy, East West University, Aftabnagar, Dhaka 1212, Bangladesh;
| | - Agnieszka Wierzbicka
- Institute of Genetics and Animal Biotechnology of the Polish Academy of Sciences, Jastrzebiec, 05-552 Magdalenka, Poland; (M.M.); (A.W.); (A.J.); (J.O.H.)
| | - Artur Jóźwik
- Institute of Genetics and Animal Biotechnology of the Polish Academy of Sciences, Jastrzebiec, 05-552 Magdalenka, Poland; (M.M.); (A.W.); (A.J.); (J.O.H.)
| | - Jarosław Olav Horbańczuk
- Institute of Genetics and Animal Biotechnology of the Polish Academy of Sciences, Jastrzebiec, 05-552 Magdalenka, Poland; (M.M.); (A.W.); (A.J.); (J.O.H.)
| | - Atanas G. Atanasov
- Institute of Genetics and Animal Biotechnology of the Polish Academy of Sciences, Jastrzebiec, 05-552 Magdalenka, Poland; (M.M.); (A.W.); (A.J.); (J.O.H.)
- Laboratory of Natural Products and Medicinal Chemistry (LNPMC), Center for Global Health Research, Saveetha Medical College and Hospital, Saveetha Institute of Medical and Technical Sciences (SIMATS), Thandalam, Chennai 602105, India
- Ludwig Boltzmann Institute Digital Health and Patient Safety, Medical University of Vienna, Spitalgasse 23, 1090 Vienna, Austria
| |
Collapse
|
8
|
Cvekl A, Vijg J. Aging of the eye: Lessons from cataracts and age-related macular degeneration. Ageing Res Rev 2024; 99:102407. [PMID: 38977082 DOI: 10.1016/j.arr.2024.102407] [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: 03/11/2024] [Revised: 06/18/2024] [Accepted: 07/01/2024] [Indexed: 07/10/2024]
Abstract
Aging is the greatest risk factor for chronic human diseases, including many eye diseases. Geroscience aims to understand the effects of the aging process on these diseases, including the genetic, molecular, and cellular mechanisms that underlie the increased risk of disease over the lifetime. Understanding of the aging eye increases general knowledge of the cellular physiology impacted by aging processes at various biological extremes. Two major diseases, age-related cataract and age-related macular degeneration (AMD) are caused by dysfunction of the lens and retina, respectively. Lens transparency and light refraction are mediated by lens fiber cells lacking nuclei and other organelles, which provides a unique opportunity to study a single aging hallmark, i.e., loss of proteostasis, within an environment of limited metabolism. In AMD, local dysfunction of the photoreceptors/retinal pigmented epithelium/Bruch's membrane/choriocapillaris complex in the macula leads to the loss of photoreceptors and eventually loss of central vision, and is driven by nearly all the hallmarks of aging and shares features with Alzheimer's disease, Parkinson's disease, cardiovascular disease, and diabetes. The aging eye can function as a model for studying basic mechanisms of aging and, vice versa, well-defined hallmarks of aging can be used as tools to understand age-related eye disease.
Collapse
Affiliation(s)
- Ales Cvekl
- Departments of Genetics and Ophthalmology and Visual Sciences, Albert Einstein College of Medicine, Bronx, NY 10461, USA.
| | - Jan Vijg
- Departments of Genetics and Ophthalmology and Visual Sciences, Albert Einstein College of Medicine, Bronx, NY 10461, USA.
| |
Collapse
|
9
|
van der Linden J, Trap L, Scherer CV, Roks AJM, Danser AHJ, van der Pluijm I, Cheng C. Model Systems to Study the Mechanism of Vascular Aging. Int J Mol Sci 2023; 24:15379. [PMID: 37895059 PMCID: PMC10607365 DOI: 10.3390/ijms242015379] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Revised: 10/16/2023] [Accepted: 10/17/2023] [Indexed: 10/29/2023] Open
Abstract
Cardiovascular diseases are the leading cause of death globally. Within cardiovascular aging, arterial aging holds significant importance, as it involves structural and functional alterations in arteries that contribute substantially to the overall decline in cardiovascular health during the aging process. As arteries age, their ability to respond to stress and injury diminishes, while their luminal diameter increases. Moreover, they experience intimal and medial thickening, endothelial dysfunction, loss of vascular smooth muscle cells, cellular senescence, extracellular matrix remodeling, and deposition of collagen and calcium. This aging process also leads to overall arterial stiffening and cellular remodeling. The process of genomic instability plays a vital role in accelerating vascular aging. Progeria syndromes, rare genetic disorders causing premature aging, exemplify the impact of genomic instability. Throughout life, our DNA faces constant challenges from environmental radiation, chemicals, and endogenous metabolic products, leading to DNA damage and genome instability as we age. The accumulation of unrepaired damages over time manifests as an aging phenotype. To study vascular aging, various models are available, ranging from in vivo mouse studies to cell culture options, and there are also microfluidic in vitro model systems known as vessels-on-a-chip. Together, these models offer valuable insights into the aging process of blood vessels.
Collapse
Affiliation(s)
- Janette van der Linden
- Division of Vascular Medicine and Pharmacology, Department of Internal Medicine, Erasmus MC, 3015 GD Rotterdam, The Netherlands
- Department of Molecular Genetics, Cancer Genomics Center Netherlands, Erasmus MC, 3015 GD Rotterdam, The Netherlands
| | - Lianne Trap
- Department of Pulmonary Medicine, Erasmus MC, 3015 GD Rotterdam, The Netherlands
- Department of Internal Medicine, Erasmus MC, 3015 GD Rotterdam, The Netherlands
| | - Caroline V. Scherer
- Department of Molecular Genetics, Cancer Genomics Center Netherlands, Erasmus MC, 3015 GD Rotterdam, The Netherlands
| | - Anton J. M. Roks
- Division of Vascular Medicine and Pharmacology, Department of Internal Medicine, Erasmus MC, 3015 GD Rotterdam, The Netherlands
| | - A. H. Jan Danser
- Division of Vascular Medicine and Pharmacology, Department of Internal Medicine, Erasmus MC, 3015 GD Rotterdam, The Netherlands
| | - Ingrid van der Pluijm
- Department of Molecular Genetics, Cancer Genomics Center Netherlands, Erasmus MC, 3015 GD Rotterdam, The Netherlands
- Department of Vascular Surgery, Cardiovascular Institute, Erasmus MC, 3015 GD Rotterdam, The Netherlands
| | - Caroline Cheng
- Division of Experimental Cardiology, Department of Cardiology, Erasmus MC, 3015 GD Rotterdam, The Netherlands
- Department of Nephrology and Hypertension, Division of Internal Medicine and Dermatology, University Medical Center Utrecht, 3584 CX Utrecht, The Netherlands
| |
Collapse
|
10
|
Ma Y, Farny NG. Connecting the dots: Neuronal senescence, stress granules, and neurodegeneration. Gene 2023; 871:147437. [PMID: 37084987 PMCID: PMC10205695 DOI: 10.1016/j.gene.2023.147437] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Revised: 04/09/2023] [Accepted: 04/14/2023] [Indexed: 04/23/2023]
Abstract
Cellular senescence increases with aging. While senescence is associated with an exit of the cell cycle, there is ample evidence that post-mitotic cells including neurons can undergo senescence as the brain ages, and that senescence likely contributes significantly to the progression of neurodegenerative diseases (ND) such as Alzheimer's Disease (AD) and Amyotrophic Lateral Sclerosis (ALS). Stress granules (SGs) are stress-induced cytoplasmic biomolecular condensates of RNA and proteins, which have been linked to the development of AD and ALS. The SG seeding hypothesis of NDs proposes that chronic stress in aging neurons results in static SGs that progress into pathological aggregates Alterations in SG dynamics have also been linked to senescence, though studies that link SGs and senescence in the context of NDs and the aging brain have not yet been performed. In this Review, we summarize the literature on senescence, and explore the contribution of senescence to the aging brain. We describe senescence phenotypes in aging neurons and glia, and their links to neuroinflammation and the development of AD and ALS. We further examine the relationships of SGs to senescence and to ND. We propose a new hypothesis that neuronal senescence may contribute to the mechanism of SG seeding in ND by altering SG dynamics in aged cells, thereby providing additional aggregation opportunities within aged neurons.
Collapse
Affiliation(s)
- Yizhe Ma
- Department of Biology and Biotechnology, Worcester Polytechnic Institute, Worcester, MA, USA
| | - Natalie G Farny
- Department of Biology and Biotechnology, Worcester Polytechnic Institute, Worcester, MA, USA.
| |
Collapse
|
11
|
Wu S, Ohba S, Matsushita Y. Single-Cell RNA-Sequencing Reveals the Skeletal Cellular Dynamics in Bone Repair and Osteoporosis. Int J Mol Sci 2023; 24:9814. [PMID: 37372962 DOI: 10.3390/ijms24129814] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Revised: 05/29/2023] [Accepted: 06/03/2023] [Indexed: 06/29/2023] Open
Abstract
The bone is an important organ that performs various functions, and the bone marrow inside the skeleton is composed of a complex intermix of hematopoietic, vascular, and skeletal cells. Current single-cell RNA sequencing (scRNA-seq) technology has revealed heterogeneity and sketchy differential hierarchy of skeletal cells. Skeletal stem and progenitor cells (SSPCs) are located upstream of the hierarchy and differentiate into chondrocytes, osteoblasts, osteocytes, and bone marrow adipocytes. In the bone marrow, multiple types of bone marrow stromal cells (BMSCs), which have the potential of SSPCs, are spatiotemporally located in distinct areas, and SSPCs' potential shift of BMSCs may occur with the advancement of age. These BMSCs contribute to bone regeneration and bone diseases, such as osteoporosis. In vivo lineage-tracing technologies show that various types of skeletal lineage cells concomitantly gather and contribute to bone regeneration. In contrast, these cells differentiate into adipocytes with aging, leading to senile osteoporosis. scRNA-seq analysis has revealed that alteration in the cell-type composition is a major cause of tissue aging. In this review, we discuss the cellular dynamics of skeletal cell populations in bone homeostasis, regeneration, and osteoporosis.
Collapse
Affiliation(s)
- Sixun Wu
- Department of Cell Biology, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki 852-8588, Japan
| | - Shinsuke Ohba
- Department of Cell Biology, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki 852-8588, Japan
- Department of Tissue and Developmental Biology, Osaka University Graduate School of Dentistry, Osaka 565-0871, Japan
| | - Yuki Matsushita
- Department of Cell Biology, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki 852-8588, Japan
| |
Collapse
|
12
|
Xu X, Li G, Zhang D, Zhu H, Liu G, Zhang Z. Gut Microbiota is Associated with Aging-Related Processes of a Small Mammal Species under High-Density Crowding Stress. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2205346. [PMID: 36965140 PMCID: PMC10190659 DOI: 10.1002/advs.202205346] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/15/2022] [Revised: 02/15/2023] [Indexed: 05/18/2023]
Abstract
Humans and animals frequently encounter high-density crowding stress, which may accelerate their aging processes; however, the roles of gut microbiota in the regulation of aging-related processes under high-density crowding stress remain unclear. In the present study, it is found that high housing density remarkably increases the stress hormone (corticosterone), accelerates aging-related processes as indicated by telomere length (in brain and liver cells) and DNA damage or inflammation (as revealed by tumor necrosis factor-α and interleukin-10 levels), and reduces the lifespan of Brandt's vole (Lasiopodomys brandtii). Fecal microbiota transplantation from donor voles of habitats with different housing densities induces similar changes in aging-related processes in recipient voles. The elimination of high housing density or butyric acid administration delays the appearance of aging-related markers in the brain and liver cells of voles housed at high-density. This study suggests that gut microorganisms may play a significant role in regulating the density-dependent aging-related processes and subsequent population dynamics of animals, and can be used as potential targets for alleviating stress-related aging in humans exposed to high-density crowding stress.
Collapse
Affiliation(s)
- Xiaoming Xu
- State Key Laboratory of Integrated Management of Pest Insects and RodentsInstitute of ZoologyChinese Academy of SciencesBeijing100101China
- University of Chinese Academy of SciencesBeijing100049China
| | - Guoliang Li
- State Key Laboratory of Integrated Management of Pest Insects and RodentsInstitute of ZoologyChinese Academy of SciencesBeijing100101China
- CAS Center for Excellence in Biotic InteractionsUniversity of Chinese Academy of SciencesBeijing100049China
| | - Da Zhang
- State Key Laboratory of Integrated Management of Pest Insects and RodentsInstitute of ZoologyChinese Academy of SciencesBeijing100101China
- University of Chinese Academy of SciencesBeijing100049China
| | - Hanyi Zhu
- State Key Laboratory of Integrated Management of Pest Insects and RodentsInstitute of ZoologyChinese Academy of SciencesBeijing100101China
- University of Chinese Academy of SciencesBeijing100049China
| | - Guang‐hui Liu
- Institute for Stem cell and RegenerationCASBeijing100049China
- State Key Laboratory of Membrane BiologyInstitute of ZoologyChinese Academy of SciencesBeijing100101China
- Beijing Institute for Stem Cell and Regenerative MedicineBeijing100101China
| | - Zhibin Zhang
- State Key Laboratory of Integrated Management of Pest Insects and RodentsInstitute of ZoologyChinese Academy of SciencesBeijing100101China
- CAS Center for Excellence in Biotic InteractionsUniversity of Chinese Academy of SciencesBeijing100049China
| |
Collapse
|
13
|
Levi N, Papismadov N, Majewska J, Roitman L, Wigoda N, Eilam R, Tsoory M, Rotkopf R, Ovadya Y, Akiva H, Regev O, Krizhanovsky V. p21 facilitates chronic lung inflammation via epithelial and endothelial cells. Aging (Albany NY) 2023; 15:2395-2417. [PMID: 36996500 PMCID: PMC10120903 DOI: 10.18632/aging.204622] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Accepted: 03/17/2023] [Indexed: 04/01/2023]
Abstract
Cellular senescence is a stable state of cell cycle arrest that regulates tissue integrity and protects the organism from tumorigenesis. However, the accumulation of senescent cells during aging contributes to age-related pathologies. One such pathology is chronic lung inflammation. p21 (CDKN1A) regulates cellular senescence via inhibition of cyclin-dependent kinases (CDKs). However, its role in chronic lung inflammation and functional impact on chronic lung disease, where senescent cells accumulate, is less understood. To elucidate the role of p21 in chronic lung inflammation, we subjected p21 knockout (p21-/-) mice to repetitive inhalations of lipopolysaccharide (LPS), an exposure that leads to chronic bronchitis and accumulation of senescent cells. p21 knockout led to a reduced presence of senescent cells, alleviated the pathological manifestations of chronic lung inflammation, and improved the fitness of the mice. The expression profiling of the lung cells revealed that resident epithelial and endothelial cells, but not immune cells, play a significant role in mediating the p21-dependent inflammatory response following chronic LPS exposure. Our results implicate p21 as a critical regulator of chronic bronchitis and a driver of chronic airway inflammation and lung destruction.
Collapse
Affiliation(s)
- Naama Levi
- Department of Molecular Cell Biology, The Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Nurit Papismadov
- Department of Molecular Cell Biology, The Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Julia Majewska
- Department of Molecular Cell Biology, The Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Lior Roitman
- Department of Molecular Cell Biology, The Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Noa Wigoda
- Department of Life Sciences Core Facilities, The Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Raya Eilam
- Department of Veterinary Resources, The Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Michael Tsoory
- Department of Veterinary Resources, The Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Ron Rotkopf
- Department of Life Sciences Core Facilities, The Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Yossi Ovadya
- Department of Molecular Cell Biology, The Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Hagay Akiva
- Department of Molecular Cell Biology, The Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Ofer Regev
- Department of Immunology, The Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Valery Krizhanovsky
- Department of Molecular Cell Biology, The Weizmann Institute of Science, Rehovot 7610001, Israel
| |
Collapse
|
14
|
Moiseeva V, Cisneros A, Cobos AC, Tarrega AB, Oñate CS, Perdiguero E, Serrano AL, Muñoz-Cánoves P. Context-dependent roles of cellular senescence in normal, aged, and disease states. FEBS J 2023; 290:1161-1185. [PMID: 35811491 DOI: 10.1111/febs.16573] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2022] [Revised: 04/20/2022] [Accepted: 07/07/2022] [Indexed: 01/10/2023]
Abstract
Cellular senescence is a state of irreversible cell cycle arrest that often emerges after tissue damage and in age-related diseases. Through the production of a multicomponent secretory phenotype (SASP), senescent cells can impact the regeneration and function of tissues. However, the effects of senescent cells and their SASP are very heterogeneous and depend on the tissue environment and type as well as the duration of injury, the degree of persistence of senescent cells and the organism's age. While the transient presence of senescent cells is widely believed to be beneficial, recent data suggest that it is detrimental for tissue regeneration after acute damage. Furthermore, although senescent cell persistence is typically associated with the progression of age-related chronic degenerative diseases, it now appears to be also necessary for correct tissue function in the elderly. Here, we discuss what is currently known about the roles of senescent cells and their SASP in tissue regeneration in ageing and age-related diseases, highlighting their (negative and/or positive) contributions. We provide insight for future research, including the possibility of senolytic-based therapies and cellular reprogramming, with aims ranging from enhancing tissue repair to extending a healthy lifespan.
Collapse
Affiliation(s)
- Victoria Moiseeva
- Department of Experimental and Health Sciences, Pompeu Fabra University (UPF), CIBER on Neurodegenerative Diseases (CIBERNED), Barcelona, Spain
| | - Andrés Cisneros
- Department of Experimental and Health Sciences, Pompeu Fabra University (UPF), CIBER on Neurodegenerative Diseases (CIBERNED), Barcelona, Spain
| | - Aina Calls Cobos
- Department of Experimental and Health Sciences, Pompeu Fabra University (UPF), CIBER on Neurodegenerative Diseases (CIBERNED), Barcelona, Spain
| | - Aida Beà Tarrega
- Department of Experimental and Health Sciences, Pompeu Fabra University (UPF), CIBER on Neurodegenerative Diseases (CIBERNED), Barcelona, Spain
| | - Claudia Santos Oñate
- Department of Experimental and Health Sciences, Pompeu Fabra University (UPF), CIBER on Neurodegenerative Diseases (CIBERNED), Barcelona, Spain
| | - Eusebio Perdiguero
- Department of Experimental and Health Sciences, Pompeu Fabra University (UPF), CIBER on Neurodegenerative Diseases (CIBERNED), Barcelona, Spain
| | - Antonio L Serrano
- Department of Experimental and Health Sciences, Pompeu Fabra University (UPF), CIBER on Neurodegenerative Diseases (CIBERNED), Barcelona, Spain
| | - Pura Muñoz-Cánoves
- Department of Experimental and Health Sciences, Pompeu Fabra University (UPF), CIBER on Neurodegenerative Diseases (CIBERNED), Barcelona, Spain.,ICREA, Barcelona, Spain.,Spanish National Center on Cardiovascular Research (CNIC), Madrid, Spain
| |
Collapse
|
15
|
Oranger A, Storlino G, Dicarlo M, Zerlotin R, Pignataro P, Sanesi L, Narici M, Pišot R, Simunič B, Colaianni G, Grano M, Colucci S. Impact of 10-day bed rest on serum levels of irisin and markers of musculoskeletal metabolism. FASEB J 2023; 37:e22668. [PMID: 36475382 DOI: 10.1096/fj.202201005rr] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2022] [Revised: 10/17/2022] [Accepted: 11/11/2022] [Indexed: 12/12/2022]
Abstract
The bed rest (BR) is a ground-based model to simulate microgravity mimicking skeletal-muscle alterations as in spaceflight. Molecular coupling between bone and muscle might be involved in physiological and pathological conditions. Thus, the new myokine irisin and bone-muscle turnover markers have been studied during and after 10 days of BR. Ten young male individuals were subjected to 10 days of horizontal BR. Serum concentrations of irisin, myostatin, sclerostin, and haptoglobin were assessed, and muscle tissue gene expression on vastus lateralis biopsies was determined. During 10-days BR, we observed no significant fluctuation levels of irisin, myostatin, and sclerostin. Two days after BR (R+2), irisin serum levels significantly decreased while myostatin, sclerostin, and haptoglobin were significantly increased compared with BR0. Gene expression of myokines, inflammatory molecules, transcription factors, and markers of muscle atrophy and senescence on muscle biopsies were not altered, suggesting that muscle metabolism of young, healthy subjects is able to adapt to the hypomobility condition during 10-day BR. However, when subjects were divided according to irisin serum levels at BR9, muscle ring finger-1 mRNA expression was significantly lower in subjects with higher irisin serum levels, suggesting that this myokine may prevent the triggering of muscle atrophy. Moreover, the negative correlation between p21 mRNA and irisin at BR9 indicated a possible inhibitory effect of the myokine on the senescence marker. In conclusion, irisin could be a prognostic marker of hypomobility-induced muscle atrophy, and its serum levels could protect against muscle deterioration by preventing and/or delaying the expression of atrophy and senescence cellular markers.
Collapse
Affiliation(s)
- Angela Oranger
- Department of Precision and Regenerative Medicine and Ionian Area, University of Bari, Bari, Italy
| | - Giuseppina Storlino
- Department of Translational Biomedicine and Neuroscience, University of Bari, Bari, Italy
| | - Manuela Dicarlo
- Department of Translational Biomedicine and Neuroscience, University of Bari, Bari, Italy
| | - Roberta Zerlotin
- Department of Precision and Regenerative Medicine and Ionian Area, University of Bari, Bari, Italy
| | - Patrizia Pignataro
- Department of Translational Biomedicine and Neuroscience, University of Bari, Bari, Italy
| | - Lorenzo Sanesi
- Department of Translational Biomedicine and Neuroscience, University of Bari, Bari, Italy
| | - Marco Narici
- Department of Biomedical Sciences, University of Padova, Padova, Italy
| | - Rado Pišot
- Institute of Kinesiology Research, Science and Research Centre, Koper, Slovenia
| | - Bostjan Simunič
- Institute of Kinesiology Research, Science and Research Centre, Koper, Slovenia
| | - Graziana Colaianni
- Department of Precision and Regenerative Medicine and Ionian Area, University of Bari, Bari, Italy
| | - Maria Grano
- Department of Precision and Regenerative Medicine and Ionian Area, University of Bari, Bari, Italy
| | - Silvia Colucci
- Department of Translational Biomedicine and Neuroscience, University of Bari, Bari, Italy
| |
Collapse
|
16
|
Wu S, Lin S, Zhang X, Alizada M, Wang L, Zheng Y, Ke Q, Xu J. Recent advances in cell-based and cell-free therapeutic approaches for sarcopenia. FASEB J 2022; 36:e22614. [PMID: 36250337 DOI: 10.1096/fj.202200675r] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2022] [Revised: 07/02/2022] [Accepted: 10/04/2022] [Indexed: 11/11/2022]
Abstract
Sarcopenia is a progressive loss of muscle mass and function that is connected with increased hospital expenditures, falls, fractures, and mortality. Although muscle loss has been related to aging, injury, hormonal imbalances, and diseases such as malignancies, chronic obstructive pulmonary disease, heart failure, and kidney failure, the underlying pathogenic mechanisms of sarcopenia are unclear. Exercise-based interventions and multimodal strategies are currently being considered as potential therapeutic approaches to prevent or treat these diseases. Although drug therapy research is ongoing, no drug has yet been proven to have a substantial safety and clinical value to be the first drug therapy to be licensed for sarcopenia. To better understand the molecular alterations underlying sarcopenia and effective treatments, we review leading research and available findings from the systemic change to the muscle-specific microenvironment. Furthermore, we explore possible mechanisms of sarcopenia and provide new knowledge for the development of novel cell-free and cell-based therapeutics. This review will assist researchers in developing better therapies to improve muscle health in the elderly.
Collapse
Affiliation(s)
- Shiqiang Wu
- Department of Orthopedic, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, China
| | - Shu Lin
- Centre of Neurological and Metabolic Research, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, China.,Group of Neuroendocrinology, Garvan Institute of Medical Research, Sydney, New South Wales, Australia
| | - Xiaolu Zhang
- Department of Orthopedic, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, China
| | - Mujahid Alizada
- Department of Neurosurgery, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, China
| | - Liangmin Wang
- Department of Orthopedic, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, China
| | - Yiqiang Zheng
- Department of Orthopedic, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, China
| | - Qingfeng Ke
- Department of Orthopedic, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, China
| | - Jie Xu
- Shengli Clinical Medical College of Fujian Medical University, Fuzhou, China.,Department of Orthopedic, Fujian Provincial Hospital, Fuzhou, China
| |
Collapse
|
17
|
Zhang Y, Pignolo RJ, Bram RJ. Accelerated aging in cyclophilin B deficient mice downstream of
p21‐Cip1
/Waf1. JBMR Plus 2022; 6:e10674. [PMID: 36248275 PMCID: PMC9549704 DOI: 10.1002/jbm4.10674] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Revised: 08/08/2022] [Accepted: 08/17/2022] [Indexed: 11/05/2022] Open
Abstract
Loss of bone mass and strength is a common problem of advanced age in humans. Defective bone is also a primary finding in osteogenesis imperfecta (OI), a genetic condition most commonly caused by autosomal dominant mutations in the type I collagen genes. Although altered collagen has been proposed to correlate with cellular processes that underlie aging, the causal relationships between them in vivo have not yet been completely explored. Whether aging plays a promoting role in OI development or whether OI contributes to aging, also remains unknown. The PpiB gene encodes cyclophilin B (CypB), a prolyl isomerase residing in the endoplasmic reticulum required for normal assembly of collagen. Germline deletion or mutations of CypB in mice or humans cause autosomal recessive OI (type IX). Here, we show that mice lacking CypB develop early onset of aging‐associated phenotypes, including kyphosis, fat reduction and weight loss, as well as abnormal teeth, skin, and muscle. Elevated senescence‐associated beta‐galactosidase (SA‐β‐Gal) activity was observed in fat tissues and in bone marrow–derived multipotent stromal cells. Protein levels of the cyclin‐dependent kinase (cdk)‐inhibitor p21‐Cip1/Waf1, a well known senescence marker, were significantly elevated in CypB‐deficient primary cells and mouse tissues. Importantly, loss of p21 in CypB knockout mice attenuated SA‐β‐Gal activity and delayed the development of kyphosis. In addition, less adipose tissue depot and higher SA‐β‐Gal activity were observed in a second OI model, Cola2oim mutant mice. A potential upregulation of p21 was also revealed in a limited number of these mice. These findings suggest that some of the features in OI patients may be mediated in part through activation of the p21‐dependent pathway, one of which is closely associated with senescence and aging. This study provides new mechanistic insight into relationships between OI and aging and raises the possibility of using senolytics drugs to treat OI in the future. © 2022 The Authors. JBMR Plus published by Wiley Periodicals LLC on behalf of American Society for Bone and Mineral Research.
Collapse
Affiliation(s)
- Ying Zhang
- Department of Pediatric and Adolescent Medicine Mayo Clinic College of Medicine Rochester MN USA
| | - Robert J Pignolo
- Department of Medicine, Division of Geriatric Medicine and Gerontology Mayo Clinic College of Medicine Rochester MN USA
- Robert and Arlene Kogod Center on Aging Mayo Clinic College of Medicine Rochester MN USA
| | - Richard J Bram
- Department of Pediatric and Adolescent Medicine Mayo Clinic College of Medicine Rochester MN USA
- Department of Immunology Mayo Clinic College of Medicine Rochester MN USA
| |
Collapse
|
18
|
Chandra A, Lagnado AB, Farr JN, Doolittle M, Tchkonia T, Kirkland JL, LeBrasseur NK, Robbins PD, Niedernhofer LJ, Ikeno Y, Passos JF, Monroe DG, Pignolo RJ, Khosla S. Targeted clearance of p21- but not p16-positive senescent cells prevents radiation-induced osteoporosis and increased marrow adiposity. Aging Cell 2022; 21:e13602. [PMID: 35363946 PMCID: PMC9124310 DOI: 10.1111/acel.13602] [Citation(s) in RCA: 69] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2021] [Revised: 03/09/2022] [Accepted: 03/20/2022] [Indexed: 12/14/2022] Open
Abstract
Cellular senescence, which is a major cause of tissue dysfunction with aging and multiple other conditions, is known to be triggered by p16Ink4a or p21Cip1 , but the relative contributions of each pathway toward inducing senescence are unclear. Here, we directly addressed this issue by first developing and validating a p21-ATTAC mouse with the p21Cip1 promoter driving a "suicide" transgene encoding an inducible caspase-8 which, upon induction, selectively kills p21Cip1 -expressing senescent cells. Next, we used the p21-ATTAC mouse and the established p16-INK-ATTAC mouse to directly compare the contributions of p21Cip1 versus p16Ink4a in driving cellular senescence in a condition where a tissue phenotype (bone loss and increased marrow adiposity) is clearly driven by cellular senescence-specifically, radiation-induced osteoporosis. Using RNA in situ hybridization, we confirmed the reduction in radiation-induced p21Cip1 - or p16Ink4a -driven transcripts following senescent cell clearance in both models. However, only clearance of p21Cip1 +, but not p16Ink4a +, senescent cells prevented both radiation-induced osteoporosis and increased marrow adiposity. Reduction in senescent cells with dysfunctional telomeres following clearance of p21Cip1 +, but not p16Ink4a +, senescent cells also reduced several of the radiation-induced pro-inflammatory senescence-associated secretory phenotype factors. Thus, by directly comparing senescent cell clearance using two parallel genetic models, we demonstrate that radiation-induced osteoporosis is driven predominantly by p21Cip1 - rather than p16Ink4a -mediated cellular senescence. Further, this approach can be used to dissect the contributions of these pathways in other senescence-associated conditions, including aging across tissues.
Collapse
Affiliation(s)
- Abhishek Chandra
- Department of Physiology and Biomedical EngineeringMayo ClinicRochesterMinnesotaUSA
- Robert and Arlene Kogod Center on AgingMayo ClinicRochesterMinnesotaUSA
| | - Anthony B. Lagnado
- Department of Physiology and Biomedical EngineeringMayo ClinicRochesterMinnesotaUSA
- Robert and Arlene Kogod Center on AgingMayo ClinicRochesterMinnesotaUSA
| | - Joshua N. Farr
- Department of Physiology and Biomedical EngineeringMayo ClinicRochesterMinnesotaUSA
- Robert and Arlene Kogod Center on AgingMayo ClinicRochesterMinnesotaUSA
- Division of EndocrinologyMayo ClinicRochesterMinnesotaUSA
| | - Madison Doolittle
- Robert and Arlene Kogod Center on AgingMayo ClinicRochesterMinnesotaUSA
- Division of EndocrinologyMayo ClinicRochesterMinnesotaUSA
| | - Tamara Tchkonia
- Department of Physiology and Biomedical EngineeringMayo ClinicRochesterMinnesotaUSA
- Robert and Arlene Kogod Center on AgingMayo ClinicRochesterMinnesotaUSA
| | - James L. Kirkland
- Department of Physiology and Biomedical EngineeringMayo ClinicRochesterMinnesotaUSA
- Robert and Arlene Kogod Center on AgingMayo ClinicRochesterMinnesotaUSA
| | - Nathan K. LeBrasseur
- Robert and Arlene Kogod Center on AgingMayo ClinicRochesterMinnesotaUSA
- Department of Physical Medicine and RehabilitationMayo ClinicRochesterMinnesotaUSA
| | - Paul D. Robbins
- Institute on the Biology of Aging and MetabolismDepartment of Biochemistry, Molecular Biology and BiophysicsUniversity of MinnesotaMinneapolisMinnesotaUSA
| | - Laura J. Niedernhofer
- Institute on the Biology of Aging and MetabolismDepartment of Biochemistry, Molecular Biology and BiophysicsUniversity of MinnesotaMinneapolisMinnesotaUSA
| | - Yuji Ikeno
- Department of Pathology and Laboratory MedicineUniversity of Texas Health Science CenterSan AntonioTexasUSA
| | - João F. Passos
- Department of Physiology and Biomedical EngineeringMayo ClinicRochesterMinnesotaUSA
- Robert and Arlene Kogod Center on AgingMayo ClinicRochesterMinnesotaUSA
| | - David G. Monroe
- Robert and Arlene Kogod Center on AgingMayo ClinicRochesterMinnesotaUSA
- Division of EndocrinologyMayo ClinicRochesterMinnesotaUSA
| | - Robert J. Pignolo
- Department of Physiology and Biomedical EngineeringMayo ClinicRochesterMinnesotaUSA
- Robert and Arlene Kogod Center on AgingMayo ClinicRochesterMinnesotaUSA
- Division of EndocrinologyMayo ClinicRochesterMinnesotaUSA
| | - Sundeep Khosla
- Department of Physiology and Biomedical EngineeringMayo ClinicRochesterMinnesotaUSA
- Robert and Arlene Kogod Center on AgingMayo ClinicRochesterMinnesotaUSA
- Division of EndocrinologyMayo ClinicRochesterMinnesotaUSA
| |
Collapse
|
19
|
Lu J, An J, Wang J, Cao X, Cao Y, Huang C, Jiao S, Yan D, Lin X, Zhou X. Znhit1 Regulates p21Cip1 to Control Mouse Lens Differentiation. Invest Ophthalmol Vis Sci 2022; 63:18. [PMID: 35472217 PMCID: PMC9055562 DOI: 10.1167/iovs.63.4.18] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2021] [Accepted: 04/06/2022] [Indexed: 01/22/2023] Open
Abstract
Purpose The transparency of the ocular lens is essential for refracting and focusing light onto the retina, and transparency is controlled by many factors and signaling pathways. Here we showed a critical role of chromatin remodeler zinc finger HIT-type containing 1 (Znhit1) in maintaining lens transparency. Methods To explore the roles of Znhit1 in lens development, the cre-loxp system was used to generate lens-specific Znhit1 knockout mice (Znhit1Mlr10-Cre; Znhit1 cKO). Morphological changes in mice lenses were examined using hematoxylin and eosin staining. RNA sequencing (RNA-seq) and assay for transposase accessible chromatin using sequencing (ATAC-seq) were applied to screen transcriptome changes. Immunofluorescence staining were performed to assess proteins distribution and terminal deoxynucleotidyl transferase dUTP nick-end labeling staining were used for determining apoptosis. The mRNAs expression was examined by quantitative RT-PCR and proteins expression by Western blot. Results Lens-specific conditional knockout mice had a severe cataract, microphthalmia phenotype, and seriously abnormal lens fiber cells differentiation. Deletion of Znhit1 in the lens resulted in decreased cell proliferation and increased cell apoptosis of the lens epithelia. ATAC-seq showed that Znhit1 deficiency increased chromatin accessibility of cyclin-dependent kinase inhibitors, including p57Kip2 and p21Cip1, and upregulated the expression of these genes in mRNA and protein levels. And we also showed that loss of Znhit1 lead to lens fibrosis by upregulating the expression of p21Cip1. Conclusions Our findings suggested that Znhit1 is required for the survival of lens epithelial cells. The loss of Znhit1 leads to the overexpression of p21Cip1, further resulting in lens fibrosis, and impacted the establishment of lens transparency.
Collapse
Affiliation(s)
- Juan Lu
- School of Optometry and Ophthalmology and Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China
- State Key Laboratory of Optometry, Ophthalmology and Vision Science, Wenzhou, Zhejiang, China
| | - Jianhong An
- School of Optometry and Ophthalmology and Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China
- State Key Laboratory of Optometry, Ophthalmology and Vision Science, Wenzhou, Zhejiang, China
| | - Jiawei Wang
- School of Optometry and Ophthalmology and Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China
- State Key Laboratory of Optometry, Ophthalmology and Vision Science, Wenzhou, Zhejiang, China
| | - Xiaowen Cao
- School of Optometry and Ophthalmology and Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China
- State Key Laboratory of Optometry, Ophthalmology and Vision Science, Wenzhou, Zhejiang, China
| | - Yuqing Cao
- School of Optometry and Ophthalmology and Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China
- State Key Laboratory of Optometry, Ophthalmology and Vision Science, Wenzhou, Zhejiang, China
| | - Chengjie Huang
- School of Optometry and Ophthalmology and Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China
- State Key Laboratory of Optometry, Ophthalmology and Vision Science, Wenzhou, Zhejiang, China
| | - Shiming Jiao
- School of Optometry and Ophthalmology and Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China
- State Key Laboratory of Optometry, Ophthalmology and Vision Science, Wenzhou, Zhejiang, China
| | - Dongsheng Yan
- School of Optometry and Ophthalmology and Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China
- State Key Laboratory of Optometry, Ophthalmology and Vision Science, Wenzhou, Zhejiang, China
| | - Xinhua Lin
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Xiangtian Zhou
- School of Optometry and Ophthalmology and Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China
- State Key Laboratory of Optometry, Ophthalmology and Vision Science, Wenzhou, Zhejiang, China
- National Clinical Research Center for Ocular Diseases, Wenzhou, China
- Oujiang Laboratory, Zhejiang Lab for Regenerative Medicine, Vision and Brain Health, Wenzhou, Zhejiang, China
- Research Unit of Myopia Basic Research and Clinical Prevention and Control, Chinese Academy of Medical Sciences, Wenzhou, China
| |
Collapse
|
20
|
Murakami T, Inagaki N, Kondoh H. Cellular Senescence in Diabetes Mellitus: Distinct Senotherapeutic Strategies for Adipose Tissue and Pancreatic β Cells. Front Endocrinol (Lausanne) 2022; 13:869414. [PMID: 35432205 PMCID: PMC9009089 DOI: 10.3389/fendo.2022.869414] [Citation(s) in RCA: 50] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/04/2022] [Accepted: 03/02/2022] [Indexed: 12/15/2022] Open
Abstract
Increased insulin resistance and impaired insulin secretion are significant characteristics manifested by patients with type 2 diabetes mellitus (T2DM). The degree and extent of these two features in T2DM vary among races and individuals. Insulin resistance is accelerated by obesity and is accompanied by accumulation of dysfunctional adipose tissues. In addition, dysfunction of pancreatic β-cells impairs insulin secretion. T2DM is significantly affected by aging, as the β-cell mass diminishes with age. Moreover, both obesity and hyperglycemia-related metabolic changes in developing diabetes are associated with accumulation of senescent cells in multiple organs, that is, organismal aging. Cellular senescence is defined as a state of irreversible cell cycle arrest with concomitant functional decline. It is caused by telomere shortening or senescence-inducing stress. Senescent cells secrete proinflammatory cytokines and chemokines, which is designated as the senescence-associated secretory phenotype (SASP), and this has a negative impact on adipose tissues and pancreatic β-cells. Recent advances in aging research have suggested that senolysis, the removal of senescent cells, can be a promising therapeutic approach to prevent or improve aging-related diseases, including diabetes. The attenuation of a SASP may be beneficial, although the pathophysiological involvement of cellular senescence in diabetes is not fully understood. In the clinical application of senotherapy, tissue-context-dependent senescent cells are increasingly being recognized as an issue to be solved. Recent studies have observed highly heterogenic and complex senescent cell populations that serve distinct roles among tissues, various stages of disease, and different ages. For example, in high-fat-diet induced diabetes with obesity, mouse adipose tissues display accumulation of p21Cip1-highly-expressing (p21high) cells in the early stage, followed by increases in both p21high and p16INK4a-highly-expressing (p16high) cells in the late stage. Interestingly, elimination of p21high cells in visceral adipose tissue can prevent or improve insulin resistance in mice with obesity, while p16high cell clearance is less effective in alleviating insulin resistance. Importantly, in immune-deficient mice transplanted with fat from obese patients, dasatinib plus quercetin, a senolytic cocktail that reduces the number of both p21high and p16high cells, improves both glucose tolerance and insulin resistance. On the other hand, in pancreatic β cells, p16high cells become increasingly predominant with age and development of diabetes. Consistently, elimination of p16high cells in mice improves both glucose tolerance and glucose-induced insulin secretion. Moreover, a senolytic compound, the anti-Bcl-2 inhibitor ABT263 reduces p16INK4a expression in islets and restores glucose tolerance in mice when combined with insulin receptor antagonist S961 treatment. In addition, efficacy of senotherapy in targeting mouse pancreatic β cells has been validated not only in T2DM, but also in type 1 diabetes mellitus. Indeed, in non-obese diabetic mice, treatment with anti-Bcl-2 inhibitors, such as ABT199, eliminates senescent pancreatic β cells, resulting in prevention of diabetes mellitus. These findings clearly indicate that features of diabetes are partly determined by which or where senescent cells reside in vivo, as adipose tissues and pancreatic β cells are responsible for insulin resistance and insulin secretion, respectively. In this review, we summarize recent advances in understanding cellular senescence in adipose tissues and pancreatic β cells in diabetes. We review the different potential molecular targets and distinctive senotherapeutic strategies in adipose tissues and pancreatic β cells. We propose the novel concept of a dual-target tailored approach in senotherapy against diabetes.
Collapse
Affiliation(s)
- Takaaki Murakami
- Department of Diabetes, Endocrinology and Nutrition, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Nobuya Inagaki
- Department of Diabetes, Endocrinology and Nutrition, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Hiroshi Kondoh
- Department of Diabetes, Endocrinology and Nutrition, Kyoto University Graduate School of Medicine, Kyoto, Japan
- Geriatric Unit, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| |
Collapse
|
21
|
Ye D, Xu Y, Shi Y, Ji J, Lu X, Chen H, Huang R, Lu P, Li Y, Cheng L, Li Y, Cui K, Tang X, Luo L, Huang J. Occurrence of Oxidative Stress and Premature Senescence in the Anterior Segment of Acute Primary Angle-Closure Eyes. Invest Ophthalmol Vis Sci 2022; 63:34. [PMID: 35077549 PMCID: PMC8802011 DOI: 10.1167/iovs.63.1.34] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Purpose To explore whether oxidative stress and premature senescence occur in the anterior segment of acute primary angle-closure (APAC) eyes after increased intraocular pressure. Methods The eye samples of 21 APAC patients, 22 age-related cataract patients, and 10 healthy donors were included. Aqueous humor (AqH), iris, and anterior lens capsule samples were collected. The levels of oxidative stress markers and senescence-associated secretory phenotype (SASP)–related cytokines in AqH were estimated using relevant reagent kits and multiplex bead immunoassay technique. The intensity of relevant markers in anterior segment tissues was examined by immunofluorescence- and senescence-associated β-galactosidase (SA-β-gal) staining. Results Oxidative stress marker levels elevated significantly in the AqH of APAC eyes. Reactive oxygen species (ROS) and 8-hydroxydeoxyguanosine levels were positively correlated with preoperative peak intraocular pressure and age, whereas reduced glutathione/oxidized glutathione (GSH/GSSH) ratio was negatively correlated with both parameters. The levels of several SASP-related cytokines were markedly increased. ROS and malondialdehyde levels were positively correlated with the levels of some SASP-related cytokines, whereas superoxide dismutase level and GSH/GSSH ratio showed an opposite trend. The number of cells positive for oxidative mitochondrial DNA damage and apoptosis-related markers increased in the iris and anterior lens capsule of the APAC group. Senescence-associated markers (p16, p21, and p53) and SA-β-gal activity were increased in the iris of the APAC group. Conclusions Oxidative stress and premature senescence occurred in the anterior segment of APAC patients, suggesting that they may be involved in the development of pathological changes in the anterior segment of APAC eyes.
Collapse
Affiliation(s)
- Dan Ye
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangdong Provincial Clinical Research Center for Ocular Diseases, Guangzhou, China
| | - Yue Xu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangdong Provincial Clinical Research Center for Ocular Diseases, Guangzhou, China.,Interdepartmental Program in Vascular Biology and Therapeutics, Department of Pathology, Yale University School of Medicine, New Haven, Connecticut, United States
| | - Yuxun Shi
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangdong Provincial Clinical Research Center for Ocular Diseases, Guangzhou, China
| | - Jianping Ji
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangdong Provincial Clinical Research Center for Ocular Diseases, Guangzhou, China
| | - Xi Lu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangdong Provincial Clinical Research Center for Ocular Diseases, Guangzhou, China
| | - Hailiu Chen
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangdong Provincial Clinical Research Center for Ocular Diseases, Guangzhou, China
| | - Rong Huang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangdong Provincial Clinical Research Center for Ocular Diseases, Guangzhou, China
| | - Peng Lu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangdong Provincial Clinical Research Center for Ocular Diseases, Guangzhou, China
| | - Yunxuan Li
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangdong Provincial Clinical Research Center for Ocular Diseases, Guangzhou, China
| | - Lu Cheng
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangdong Provincial Clinical Research Center for Ocular Diseases, Guangzhou, China
| | - Yangyunhui Li
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangdong Provincial Clinical Research Center for Ocular Diseases, Guangzhou, China
| | - Kaixuan Cui
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangdong Provincial Clinical Research Center for Ocular Diseases, Guangzhou, China
| | - Xiaoyu Tang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangdong Provincial Clinical Research Center for Ocular Diseases, Guangzhou, China
| | - Lixia Luo
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangdong Provincial Clinical Research Center for Ocular Diseases, Guangzhou, China
| | - Jingjing Huang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangdong Provincial Clinical Research Center for Ocular Diseases, Guangzhou, China
| |
Collapse
|
22
|
Saito Y, Chikenji TS. Diverse Roles of Cellular Senescence in Skeletal Muscle Inflammation, Regeneration, and Therapeutics. Front Pharmacol 2021; 12:739510. [PMID: 34552495 PMCID: PMC8450382 DOI: 10.3389/fphar.2021.739510] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2021] [Accepted: 08/23/2021] [Indexed: 12/19/2022] Open
Abstract
Skeletal muscle undergoes vigorous tissue remodeling after injury. However, aging, chronic inflammatory diseases, sarcopenia, and neuromuscular disorders cause muscle loss and degeneration, resulting in muscular dysfunction. Cellular senescence, a state of irreversible cell cycle arrest, acts during normal embryonic development and remodeling after tissue damage; when these processes are complete, the senescent cells are eliminated. However, the accumulation of senescent cells is a hallmark of aging tissues or pathological contexts and may lead to progressive tissue degeneration. The mechanisms responsible for the effects of senescent cells have not been fully elucidated. Here, we review current knowledge about the beneficial and detrimental effects of senescent cells in tissue repair, regeneration, aging, and age-related disease, especially in skeletal muscle. We also discuss how senescence of muscle stem cells and muscle-resident fibro-adipogenic progenitors affects muscle pathologies or regeneration, and consider the possibility that immunosenescence leads to muscle pathogenesis. Finally, we explore senotherapy, the therapeutic targeting of senescence to treat age-related disease, from the standpoint of improving muscle regeneration.
Collapse
Affiliation(s)
- Yuki Saito
- Department of Anatomy, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Takako S Chikenji
- Department of Anatomy, Sapporo Medical University School of Medicine, Sapporo, Japan.,Department of Health Sciences, School of Medicine, Hokkaido University, Sapporo, Japan
| |
Collapse
|
23
|
Cui X, Du C, Wan S, Wu D, Yan L, Zhang J, Li J, Li H, Yang Z, Zhang H, Zhang J, Mu H, Zhang F, Peng X, Liu M, Hu Y. Deficiency of heat shock factor 4 promotes lens epithelial cell senescence through upregulating p21 cip1 expression. Biochim Biophys Acta Mol Basis Dis 2021; 1867:166233. [PMID: 34339841 DOI: 10.1016/j.bbadis.2021.166233] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Revised: 07/19/2021] [Accepted: 07/27/2021] [Indexed: 01/29/2023]
Abstract
Genetic mutations in heat shock factor 4 (Hsf4) is associated with both congenital and age-related cataracts. Hsf4 regulates lens development through its ability to both activate and inhibit transcription. Previous studies suggested Hsf4 is involved in modulating cellular senescence depending on p21cip1 and p27 kip1 expression in MEF cells. Here, we found that Hsf4 acts as a suppressor of p21cip1 expression and plays an anti-senescence role during lens development. Knocking out Hsf4 facilitated UVB-induced cellular senescence in mouse lens epithelial cells (mLECs). p21cip1 was upregulated at both the mRNA and protein levels in HSF4-/- mLECs under control and UVB-treated conditions, and knockdown of p21cip1 by siRNA alleviated UVB-induced cellular senescence. HSF4 directly bound to the p21cip1 promoter and increased H3K27m3 levels at the p21cip1 proximal promoter region by recruiting the methyltransferase EZH2. In animal models, p21cip1 was gradually upregulated in wild-type mouse lenses with increasing age, while Hsf4 levels decreased. We generated a Hsf4 mutant mice line (Hsf4del-42) which displayed obvious congenital cataract phenotype. The expression of p21cip1 and senescence-associated cytokines were induced in the cataractous lenses of Hsf4del-42 mice. H3K27m3 and EZH2 levels decreased in p21cip1 promoters in the lenses of Hsf4del-42 mice. The SA-β-Gal activities were positive in lens epithelia of aged Hsf4null zebrafish compared to wild-type lenses. p21cip1 and senescence-associated cytokines levels were also upregulated in lenses of Hsf4null zebrafish. Accordingly, we propose that HSF4 plays a protective role in lens epithelial cells against cellular senescence during lens development and aging, partly by fine-tuning p21cip1 expression.
Collapse
Affiliation(s)
- Xiukun Cui
- Jointed National Lab for antibody drug engineering, The First Affiliated Hospital, Henan University School of Medicine, Kaifeng, China.
| | - Chunxiao Du
- Jointed National Lab for antibody drug engineering, The First Affiliated Hospital, Henan University School of Medicine, Kaifeng, China
| | - Simin Wan
- Jointed National Lab for antibody drug engineering, The First Affiliated Hospital, Henan University School of Medicine, Kaifeng, China
| | - Dandan Wu
- Jointed National Lab for antibody drug engineering, The First Affiliated Hospital, Henan University School of Medicine, Kaifeng, China
| | - Longjun Yan
- Jointed National Lab for antibody drug engineering, The First Affiliated Hospital, Henan University School of Medicine, Kaifeng, China
| | - Jing Zhang
- Jointed National Lab for antibody drug engineering, The First Affiliated Hospital, Henan University School of Medicine, Kaifeng, China
| | - Jing Li
- Jointed National Lab for antibody drug engineering, The First Affiliated Hospital, Henan University School of Medicine, Kaifeng, China
| | - Hui Li
- Jointed National Lab for antibody drug engineering, The First Affiliated Hospital, Henan University School of Medicine, Kaifeng, China
| | - Zhengyan Yang
- Jointed National Lab for antibody drug engineering, The First Affiliated Hospital, Henan University School of Medicine, Kaifeng, China
| | - Hailong Zhang
- Jointed National Lab for antibody drug engineering, The First Affiliated Hospital, Henan University School of Medicine, Kaifeng, China
| | - Jun Zhang
- Jointed National Lab for antibody drug engineering, The First Affiliated Hospital, Henan University School of Medicine, Kaifeng, China
| | - Hongmei Mu
- Kaifeng Key Laboratory of Myopia and cataract, Kaifeng central Hospital Kaifeng, China
| | - Fengyan Zhang
- The Laboratory for ophthalmology and Vision Science, Department of Ophthalmology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Xuyan Peng
- The Laboratory for ophthalmology and Vision Science, Department of Ophthalmology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Mugen Liu
- College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Yanzhong Hu
- Jointed National Lab for antibody drug engineering, The First Affiliated Hospital, Henan University School of Medicine, Kaifeng, China; Kaifeng Key Laboratory of Myopia and cataract, Kaifeng central Hospital Kaifeng, China; The Laboratory for ophthalmology and Vision Science, Department of Ophthalmology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.
| |
Collapse
|
24
|
Takeshita H, Yamamoto K, Mogi M, Wang Y, Nozato Y, Fujimoto T, Yokoyama S, Hongyo K, Nakagami F, Akasaka H, Takami Y, Takeya Y, Sugimoto K, Horiuchi M, Rakugi H. Double Deletion of Angiotensin II Type 2 and Mas Receptors Accelerates Aging-Related Muscle Weakness in Male Mice. J Am Heart Assoc 2021; 10:e021030. [PMID: 34212761 PMCID: PMC8403326 DOI: 10.1161/jaha.120.021030] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Accepted: 04/16/2021] [Indexed: 12/28/2022]
Abstract
Background The activation of AT2 (angiotensin II type 2 receptor ) and Mas receptor by angiotensin II and angiotensin-(1-7), respectively, is the primary process that counteracts activation of the canonical renin-angiotensin system (RAS). Although inhibition of canonical RAS could delay the progression of physiological aging, we recently reported that deletion of Mas had no impact on the aging process in mice. Here, we used male mice with a deletion of only AT2 or a double deletion of AT2 and Mas to clarify whether these receptors contribute to the aging process in a complementary manner, primarily by focusing on aging-related muscle weakness. Methods and Results Serial changes in grip strength of these mice up to 24 months of age showed that AT2/Mas knockout mice, but not AT2 knockout mice, had significantly weaker grip strength than wild-type mice from the age of 18 months. AT2/Mas knockout mice exhibited larger sizes, but smaller numbers and increased frequency of central nucleation (a marker of aged muscle) of single skeletal muscle fibers than AT2 knockout mice. Canonical RAS-associated genes, inflammation-associated genes, and senescence-associated genes were highly expressed in skeletal muscles of AT2/Mas knockout mice. Muscle angiotensin II content increased in AT2/Mas knockout mice. Conclusions Double deletion of AT2 and Mas in mice exaggerated aging-associated muscle weakness, accompanied by signatures of activated RAS, inflammation, and aging in skeletal muscles. Because aging-associated phenotypes were absent in single deletions of the receptors, AT2 and Mas could complement each other in preventing local activation of RAS during aging.
Collapse
MESH Headings
- Age Factors
- Animals
- Fibrosis
- Gene Expression Regulation
- Genetic Predisposition to Disease
- Hand Strength
- Inflammation Mediators/metabolism
- Male
- Mice, Inbred C57BL
- Mice, Knockout
- Muscle Strength/genetics
- Muscle Weakness/genetics
- Muscle Weakness/metabolism
- Muscle Weakness/pathology
- Muscle Weakness/physiopathology
- Muscle, Skeletal/metabolism
- Muscle, Skeletal/pathology
- Muscle, Skeletal/physiopathology
- Phenotype
- Proto-Oncogene Mas
- Proto-Oncogene Proteins/deficiency
- Proto-Oncogene Proteins/genetics
- Receptor, Angiotensin, Type 2/deficiency
- Receptor, Angiotensin, Type 2/genetics
- Receptors, G-Protein-Coupled/deficiency
- Receptors, G-Protein-Coupled/genetics
- Renin-Angiotensin System/genetics
- Mice
Collapse
Affiliation(s)
- Hikari Takeshita
- Department of Geriatric and General MedicineOsaka University Graduate School of MedicineSuitaJapan
| | - Koichi Yamamoto
- Department of Geriatric and General MedicineOsaka University Graduate School of MedicineSuitaJapan
| | - Masaki Mogi
- Department of PharmacologyEhime University Graduate School of MedicineEhimeJapan
| | - Yu Wang
- Department of Geriatric and General MedicineOsaka University Graduate School of MedicineSuitaJapan
| | - Yoichi Nozato
- Department of Geriatric and General MedicineOsaka University Graduate School of MedicineSuitaJapan
| | - Taku Fujimoto
- Department of Geriatric and General MedicineOsaka University Graduate School of MedicineSuitaJapan
| | - Serina Yokoyama
- Department of Geriatric and General MedicineOsaka University Graduate School of MedicineSuitaJapan
| | - Kazuhiro Hongyo
- Department of Geriatric and General MedicineOsaka University Graduate School of MedicineSuitaJapan
| | - Futoshi Nakagami
- Department of Geriatric and General MedicineOsaka University Graduate School of MedicineSuitaJapan
| | - Hiroshi Akasaka
- Department of Geriatric and General MedicineOsaka University Graduate School of MedicineSuitaJapan
| | - Yoichi Takami
- Department of Geriatric and General MedicineOsaka University Graduate School of MedicineSuitaJapan
| | - Yasushi Takeya
- Department of Geriatric and General MedicineOsaka University Graduate School of MedicineSuitaJapan
| | - Ken Sugimoto
- Department of Geriatric and General MedicineOsaka University Graduate School of MedicineSuitaJapan
| | - Masatsugu Horiuchi
- Department of Molecular Cardiovascular Biology and PharmacologyEhime University Graduate School of MedicineEhimeJapan
| | - Hiromi Rakugi
- Department of Geriatric and General MedicineOsaka University Graduate School of MedicineSuitaJapan
| |
Collapse
|
25
|
Faranda AP, Shihan MH, Wang Y, Duncan MK. The aging mouse lens transcriptome. Exp Eye Res 2021; 209:108663. [PMID: 34119483 DOI: 10.1016/j.exer.2021.108663] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Revised: 05/04/2021] [Accepted: 06/01/2021] [Indexed: 02/06/2023]
Abstract
Age is a major risk factor for cataract (ARC). However, the influence of aging on the lens transcriptome is under studied. Lens epithelial (LEC) and fiber cells (LFC) were isolated from young (3 month old) and aged (24 month old) C57BL/6J mice, and the transcriptome elucidated via RNAseq. EdgeR estimated differential gene expression in pairwise contrasts, and Advaita's Ipathway guide and custom R scripts were used to evaluate the potential biological significance of differentially expressed genes (DEGs). This analysis revealed age-dependent decreases in lens differentiation marker expression in both LECs and LFCs, with gamma crystallin transcripts downregulating nearly 50 fold in aged LFCs. The expression of the transcription factors Hsf4 and Maf, which are known to activate lens fiber cell preferred genes, are downregulated, while FoxE3, which represses gamma crystallin expression, is upregulated in aged fibers. Aged LECs upregulate genes controlling the immune response, complement pathways, and cellular stress responses, including glutathione peroxidase 3 (Gpx3). Aged LFCs exhibit broad changes in the expression of genes regulating cell communication, and upregulate genes involved in antigen processing/presentation and cholesterol metabolism, while changes in the expression of mitochondrial respiratory chain genes are consistent with mitochondrial stress, including upregulation of NDufa4l2, which encodes an alternate electron transport chain protein. However, age did not profoundly affect the response of LECs to injury as both young and aged LECs upregulate inflammatory gene signatures at 24 h post injury to similar extents. These RNAseq profiles provide a rich data set that can be mined to understand the genetic regulation of lens aging and how this impinges on the pathophysiology of age related cataract.
Collapse
Affiliation(s)
- Adam P Faranda
- Department of Biological Sciences University of Delaware Newark, DE, 19716, USA
| | - Mahbubul H Shihan
- Department of Biological Sciences University of Delaware Newark, DE, 19716, USA
| | - Yan Wang
- Department of Biological Sciences University of Delaware Newark, DE, 19716, USA
| | - Melinda K Duncan
- Department of Biological Sciences University of Delaware Newark, DE, 19716, USA.
| |
Collapse
|
26
|
Xue Y, Chen H, Zhang S, Bao L, Chen B, Gong H, Zhao Y, Qi R. Resveratrol Confers Vascular Protection by Suppressing TLR4/Syk/NLRP3 Signaling in Oxidized Low-Density Lipoprotein-Activated Platelets. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:8819231. [PMID: 33728029 PMCID: PMC7935581 DOI: 10.1155/2021/8819231] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/04/2020] [Revised: 02/04/2021] [Accepted: 02/11/2021] [Indexed: 12/24/2022]
Abstract
This study investigated the effect of resveratrol on Toll-like receptor 4- (TLR4-) mediated matrix metalloproteinase 3 (MMP3) and MMP9 expression in oxidized low-density lipoprotein- (ox-LDL-) activated platelets and the potential molecule mechanism. Human platelets were used in the present study. The results showed that resveratrol suppressed TLR4, MMP3, and MMP9 expression in ox-LDL-activated platelets. The TLR4 inhibitor CLI-095 also inhibited MMP3 and MMP9 expression and secretion in ox-LDL- and lipopolysaccharide- (LPS-) activated platelets. The combination of resveratrol and CLI-095 synergistically suppressed MMP3 and MMP9 expression in ox-LDL- and LPS-activated platelets. These findings suggest that the resveratrol-induced inhibition of MMP3 and MMP9 expression is linked to the suppression of TLR4 activation. Resveratrol also suppressed spleen tyrosine kinase (Syk) phosphorylation and nucleotide-binding domain leucine-rich repeat containing protein 3 (NLRP3) expression and IL-1β secretion in ox-LDL- and LPS-treated platelets. The coimmunoprecipitation results showed that resveratrol inhibited the binding of Syk and NLRP3. Finally, resveratrol reduced vascular senescence cells and the expression of TLR4, MMP3, and MMP9 and prevented alterations of vascular structure in 52-week-old mice. Our findings demonstrated that resveratrol decreased inflammatory protein expression and improved vascular structure in aged mice. Resveratrol inhibited the expression of TLR4 and secretion of MMP3, MMP9, and IL-1β. The mechanism of action of resveratrol appears to be associated with the inhibition of TLR4/Syk/NLRP3 activation in ox-LDL-activated platelets.
Collapse
Affiliation(s)
- Yun Xue
- MOH Key Laboratory of Geriatrics, Beijing Hospital, National Center of Gerontology, Beijing, China
- Graduate School of Peking Union Medical College, Beijing, China
| | - Huilian Chen
- MOH Key Laboratory of Geriatrics, Beijing Hospital, National Center of Gerontology, Beijing, China
| | - Shenghao Zhang
- MOH Key Laboratory of Geriatrics, Beijing Hospital, National Center of Gerontology, Beijing, China
| | - Li Bao
- MOH Key Laboratory of Geriatrics, Beijing Hospital, National Center of Gerontology, Beijing, China
| | - Beidong Chen
- MOH Key Laboratory of Geriatrics, Beijing Hospital, National Center of Gerontology, Beijing, China
| | - Huan Gong
- MOH Key Laboratory of Geriatrics, Beijing Hospital, National Center of Gerontology, Beijing, China
| | - Yanyang Zhao
- MOH Key Laboratory of Geriatrics, Beijing Hospital, National Center of Gerontology, Beijing, China
| | - Ruomei Qi
- MOH Key Laboratory of Geriatrics, Beijing Hospital, National Center of Gerontology, Beijing, China
- Graduate School of Peking Union Medical College, Beijing, China
| |
Collapse
|
27
|
Vasudevan A, Schukken KM, Sausville EL, Girish V, Adebambo OA, Sheltzer JM. Aneuploidy as a promoter and suppressor of malignant growth. Nat Rev Cancer 2021; 21:89-103. [PMID: 33432169 DOI: 10.1038/s41568-020-00321-1] [Citation(s) in RCA: 108] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 11/12/2020] [Indexed: 12/13/2022]
Abstract
Aneuploidy has been recognized as a hallmark of tumorigenesis for more than 100 years, but the connection between chromosomal errors and malignant growth has remained obscure. New evidence emerging from both basic and clinical research has illuminated a complicated relationship: despite its frequency in human tumours, aneuploidy is not a universal driver of cancer development and instead can exert substantial tumour-suppressive effects. The specific consequences of aneuploidy are highly context dependent and are influenced by a cell's genetic and environmental milieu. In this Review, we discuss the diverse facets of cancer biology that are shaped by aneuploidy, including metastasis, drug resistance and immune recognition, and we highlight aneuploidy's distinct roles as both a tumour promoter and an anticancer vulnerability.
Collapse
|
28
|
Colaianni G, Errede M, Sanesi L, Notarnicola A, Celi M, Zerlotin R, Storlino G, Pignataro P, Oranger A, Pesce V, Tarantino U, Moretti B, Grano M. Irisin Correlates Positively With BMD in a Cohort of Older Adult Patients and Downregulates the Senescent Marker p21 in Osteoblasts. J Bone Miner Res 2021; 36:305-314. [PMID: 33053231 DOI: 10.1002/jbmr.4192] [Citation(s) in RCA: 48] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/09/2020] [Revised: 09/23/2020] [Accepted: 10/03/2020] [Indexed: 12/25/2022]
Abstract
Irisin is a myokine produced by skeletal muscle during exercise in both mice and humans. We previously showed that irisin treatment ameliorates immobility-induced osteoporosis and muscular atrophy in mice. Data in humans showed a positive association between irisin and bone mineral density (BMD) in athletes and a population of healthy children. However, the role of this myokine regarding the state of muscle and bone in the same population remained to be determined. For this purpose, 62 patients (age 68.71 ± 12.31 years) undergoing total hip or knee replacement were recruited. Our results showed that irisin serum levels negatively correlated with age (R = -0.515; p = .000018) and positively correlated with femoral BMD (R = 0.619; p = .001) and vertebral BMD (R = 0.201; p = .0001). Irisin was also positively associated with Fndc5 mRNA in muscle biopsies (R = 0.248; p = .016), as well as with Osteocalcin (Ocn) mRNA in bone biopsies (R = 0.708; p = .006). In skeletal muscle, FNDC5 positive fibers positively correlate with BMD of total femur (R = 0.765; p = .0014) and BMD of femoral neck (R = 0.575; p = .031), Interestingly, by analyzing patients divided by their T-score, we found lower irisin levels (p = .0011) in patients with osteopenia/osteoporosis (OP) compared to healthy controls matched for age and sex. By analyzing the senescence marker p21, we found a significant increase of its mRNA expression in the bone biopsies of OP patients compared to control ones. Therefore, we investigated in vitro whether rec-irisin had a direct effect on this senescence marker, showing that p21 mRNA expression was significantly downregulated in osteoblasts by the treatment with irisin. Overall, these results indicate that higher irisin levels are associated with a lower rate of age-related osteoporosis and that irisin could be effective in delaying the osteoblast aging process, suggesting a potential senolytic action of this myokine. © 2020 American Society for Bone and Mineral Research (ASBMR).
Collapse
Affiliation(s)
- Graziana Colaianni
- Department of Emergency and Organ Transplantation, University of Bari, Bari, Italy
| | - Mariella Errede
- Department of Basic Medical Sciences, Neuroscience and Sense Organs, University of Bari, Bari, Italy
| | - Lorenzo Sanesi
- Department of Emergency and Organ Transplantation, University of Bari, Bari, Italy
| | - Angela Notarnicola
- Department of Basic Medical Sciences, Neuroscience and Sense Organs, University of Bari, Bari, Italy
| | - Monica Celi
- Department of Orthopedics and Traumatology, Tor Vergata University of Rome, Rome, Italy
| | - Roberta Zerlotin
- Department of Emergency and Organ Transplantation, University of Bari, Bari, Italy
| | - Giuseppina Storlino
- Department of Emergency and Organ Transplantation, University of Bari, Bari, Italy
| | - Patrizia Pignataro
- Department of Basic Medical Sciences, Neuroscience and Sense Organs, University of Bari, Bari, Italy
| | - Angela Oranger
- Department of Emergency and Organ Transplantation, University of Bari, Bari, Italy
| | - Vito Pesce
- Department of Basic Medical Sciences, Neuroscience and Sense Organs, University of Bari, Bari, Italy
| | - Umberto Tarantino
- Department of Orthopedics and Traumatology, Tor Vergata University of Rome, Rome, Italy
| | - Biagio Moretti
- Department of Basic Medical Sciences, Neuroscience and Sense Organs, University of Bari, Bari, Italy
| | - Maria Grano
- Department of Emergency and Organ Transplantation, University of Bari, Bari, Italy
| |
Collapse
|
29
|
Mu X, Lin CY, Hambright WS, Tang Y, Ravuri S, Lu A, Matre P, Chen W, Gao X, Cui Y, Zhong L, Wang B, Huard J. Aberrant RhoA activation in macrophages increases senescence-associated secretory phenotypes and ectopic calcification in muscular dystrophic mice. Aging (Albany NY) 2020; 12:24853-24871. [PMID: 33361519 PMCID: PMC7803538 DOI: 10.18632/aging.202413] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2020] [Accepted: 11/21/2020] [Indexed: 12/17/2022]
Abstract
Duchenne Muscular Dystrophy (DMD) patients often suffer from both muscle wasting and osteoporosis. Our previous studies have revealed reduced regeneration potential in skeletal muscle and bone, concomitant with ectopic calcification of soft tissues in double knockout (dKO, dystrophin-/-; utrophin-/-) mice, a severe murine model for DMD. We found significant involvement of RhoA/ROCK (Rho-Associated Protein Kinase) signaling in mediating ectopic calcification of muscles in dKO mice. However, the cellular identity of these RhoA+ cells, and the role that RhoA plays in the chronic inflammation-associated pathologies has not been elucidated. Here, we report that CD68+ macrophages are highly prevalent at the sites of ectopic calcification of dKO mice, and that these macrophages highly express RhoA. Macrophages from dKO mice feature a shift towards a more pro-inflammatory M1 polarization and an increased expression of various senescence-associated secretory phenotype (SASP) factors that was reduced with the RhoA/ROCK inhibitor Y-27632. Further, systemic inhibition of RhoA activity in dKO mice led to reduced number of RhoA+/CD68+ cells, as well as a reduction in fibrosis and ectopic calcification. Together, these data revealed that RhoA signaling may be a key regulator of imbalanced mineralization in the dystrophic musculoskeletal system and consequently a therapeutic target for the treatment of DMD or other related muscle dystrophies.
Collapse
Affiliation(s)
- Xiaodong Mu
- Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China.,Steadman Philippon Research Institute, Center for Regenerative Sports Medicine, Vail, CO 81657, USA
| | - Chi-Yi Lin
- Department of Orthopedic Surgery, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - William S Hambright
- Steadman Philippon Research Institute, Center for Regenerative Sports Medicine, Vail, CO 81657, USA
| | - Ying Tang
- Department of Orthopaedic Surgery, University of Pittsburgh, Pittsburgh, PA 15260, USA
| | - Sudheer Ravuri
- Steadman Philippon Research Institute, Center for Regenerative Sports Medicine, Vail, CO 81657, USA
| | - Aiping Lu
- Steadman Philippon Research Institute, Center for Regenerative Sports Medicine, Vail, CO 81657, USA
| | - Polina Matre
- Department of Orthopedic Surgery, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Wanqun Chen
- Department of Orthopedic Surgery, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX 77030, USA.,Department of Biochemistry and Molecular Biology, Jinan University, Guangzhou, China
| | - Xueqin Gao
- Steadman Philippon Research Institute, Center for Regenerative Sports Medicine, Vail, CO 81657, USA
| | - Yan Cui
- Department of Orthopedic Surgery, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Ling Zhong
- Department of Orthopedic Surgery, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Bing Wang
- Department of Orthopaedic Surgery, University of Pittsburgh, Pittsburgh, PA 15260, USA
| | - Johnny Huard
- Steadman Philippon Research Institute, Center for Regenerative Sports Medicine, Vail, CO 81657, USA
| |
Collapse
|
30
|
Engler M, Fidan M, Nandi S, Cirstea IC. Senescence in RASopathies, a possible novel contributor to a complex pathophenoype. Mech Ageing Dev 2020; 194:111411. [PMID: 33309600 DOI: 10.1016/j.mad.2020.111411] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Revised: 11/17/2020] [Accepted: 11/23/2020] [Indexed: 01/07/2023]
Abstract
Senescence is a biological process that induces a permanent cell cycle arrest and a specific gene expression program in response to various stressors. Following studies over the last few decades, the concept of senescence has evolved from an antiproliferative mechanism in cancer (oncogene-induced senescence) to a critical component of physiological processes associated with embryonic development, tissue regeneration, ageing and its associated diseases. In somatic cells, oncogenic mutations in RAS-MAPK pathway genes are associated with oncogene-induced senescence and cancer, while germline mutations in the same pathway are linked to a group of monogenic developmental disorders generally termed RASopathies. Here, we consider that in these disorders, senescence induction may result in opposing outcomes, a tumour protective effect and a possible contributor to a premature ageing phenotype identified in Costello syndrome, which belongs to the RASopathy group. In this review, we will highlight the role of senescence in organismal homeostasis and we will describe the current knowledge about senescence in RASopathies. Additionally, we provide a perspective on examples of experimentally characterised RASopathy mutations that, alone or in combination with various stressors, may also trigger an age-dependent chronic senescence, possibly contributing to the age-dependent worsening of RASopathy pathophenotype and the reduction of lifespan.
Collapse
Affiliation(s)
- Melanie Engler
- Institute of Comparative Molecular Endocrinology, Ulm University, Helmholtzstr. 8/1, 89081, Ulm, Germany
| | - Miray Fidan
- Institute of Comparative Molecular Endocrinology, Ulm University, Helmholtzstr. 8/1, 89081, Ulm, Germany
| | - Sayantan Nandi
- Institute of Comparative Molecular Endocrinology, Ulm University, Helmholtzstr. 8/1, 89081, Ulm, Germany
| | - Ion Cristian Cirstea
- Institute of Comparative Molecular Endocrinology, Ulm University, Helmholtzstr. 8/1, 89081, Ulm, Germany.
| |
Collapse
|
31
|
Spinelli R, Parrillo L, Longo M, Florese P, Desiderio A, Zatterale F, Miele C, Raciti GA, Beguinot F. Molecular basis of ageing in chronic metabolic diseases. J Endocrinol Invest 2020; 43:1373-1389. [PMID: 32358737 PMCID: PMC7481162 DOI: 10.1007/s40618-020-01255-z] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/21/2020] [Accepted: 04/10/2020] [Indexed: 02/06/2023]
Abstract
AIM Over the last decades, the shift in age distribution towards older ages and the progressive ageing which has occurred in most populations have been paralleled by a global epidemic of obesity and its related metabolic disorders, primarily, type 2 diabetes (T2D). Dysfunction of the adipose tissue (AT) is widely recognized as a significant hallmark of the ageing process that, in turn, results in systemic metabolic alterations. These include insulin resistance, accumulation of ectopic lipids and chronic inflammation, which are responsible for an elevated risk of obesity and T2D onset associated to ageing. On the other hand, obesity and T2D, the paradigms of AT dysfunction, share many physiological characteristics with the ageing process, such as an increased burden of senescent cells and epigenetic alterations. Thus, these chronic metabolic disorders may represent a state of accelerated ageing. MATERIALS AND METHODS A more precise explanation of the fundamental ageing mechanisms that occur in AT and a deeper understanding of their role in the interplay between accelerated ageing and AT dysfunction can be a fundamental leap towards novel therapies that address the causes, not just the symptoms, of obesity and T2D, utilizing strategies that target either senescent cells or DNA methylation. RESULTS In this review, we summarize the current knowledge of the pathways that lead to AT dysfunction in the chronological ageing process as well as the pathophysiology of obesity and T2D, emphasizing the critical role of cellular senescence and DNA methylation. CONCLUSION Finally, we highlight the need for further research focused on targeting these mechanisms.
Collapse
Affiliation(s)
- R Spinelli
- Department of Translation Medicine, Federico II University of Naples, 80131, Naples, Italy
- URT Genomic of Diabetes, Institute of Experimental Endocrinology and Oncology, National Research Council, 80131, Naples, Italy
| | - L Parrillo
- Department of Translation Medicine, Federico II University of Naples, 80131, Naples, Italy
- URT Genomic of Diabetes, Institute of Experimental Endocrinology and Oncology, National Research Council, 80131, Naples, Italy
| | - M Longo
- Department of Translation Medicine, Federico II University of Naples, 80131, Naples, Italy
- URT Genomic of Diabetes, Institute of Experimental Endocrinology and Oncology, National Research Council, 80131, Naples, Italy
| | - P Florese
- Department of Translation Medicine, Federico II University of Naples, 80131, Naples, Italy
- URT Genomic of Diabetes, Institute of Experimental Endocrinology and Oncology, National Research Council, 80131, Naples, Italy
| | - A Desiderio
- Department of Translation Medicine, Federico II University of Naples, 80131, Naples, Italy
- URT Genomic of Diabetes, Institute of Experimental Endocrinology and Oncology, National Research Council, 80131, Naples, Italy
| | - F Zatterale
- Department of Translation Medicine, Federico II University of Naples, 80131, Naples, Italy
- URT Genomic of Diabetes, Institute of Experimental Endocrinology and Oncology, National Research Council, 80131, Naples, Italy
| | - C Miele
- Department of Translation Medicine, Federico II University of Naples, 80131, Naples, Italy
- URT Genomic of Diabetes, Institute of Experimental Endocrinology and Oncology, National Research Council, 80131, Naples, Italy
| | - G Alexander Raciti
- Department of Translation Medicine, Federico II University of Naples, 80131, Naples, Italy
- URT Genomic of Diabetes, Institute of Experimental Endocrinology and Oncology, National Research Council, 80131, Naples, Italy
| | - F Beguinot
- Department of Translation Medicine, Federico II University of Naples, 80131, Naples, Italy.
- URT Genomic of Diabetes, Institute of Experimental Endocrinology and Oncology, National Research Council, 80131, Naples, Italy.
| |
Collapse
|
32
|
Venkatesh D, Stockwell BR, Prives C. p21 can be a barrier to ferroptosis independent of p53. Aging (Albany NY) 2020; 12:17800-17814. [PMID: 32979260 PMCID: PMC7585094 DOI: 10.18632/aging.103961] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Accepted: 08/03/2020] [Indexed: 01/24/2023]
Abstract
Traditionally, the p21 protein has been viewed as limiting cancer progression and promoting aging. In contrast, there are reports that p21 can enhance cancer survival and limit tissue damage, depending on the tissue of origin and type of stressor involved. Here, we provide evidence to support these latter two roles of p21 by exploring its ability to regulate ferroptosis. Ferroptosis is a form of cell death that is associated with certain degenerative diseases, some of which are aging-related. Our results reveal a correlation between p21 protein levels in cell lines that are resistant to ferroptosis (p21 high) versus cell lines that are sensitive and easily undergo ferroptosis (p21 low). We also show that p21 levels themselves are differentially regulated in response to ferroptosis in a p53-independent manner. Further, experimentally altering the abundance of p21 protein inverts the ferroptosis-sensitivity of both resistant and sensitive human cancer cell lines. Our data also indicate that the interaction of p21 with CDKs is crucial for its ability to restrict the progression of ferroptosis. While this study was performed in cancer cell lines, our results support the potential of p21 to aid in maintenance of healthy tissues by blocking the damage incurred due to ferroptosis.
Collapse
Affiliation(s)
- Divya Venkatesh
- Department of Biological Sciences, Columbia University, New York, NY 10027, USA
| | - Brent R. Stockwell
- Department of Biological Sciences, Columbia University, New York, NY 10027, USA,Department of Chemistry, Columbia University, New York, NY 10027, USA
| | - Carol Prives
- Department of Biological Sciences, Columbia University, New York, NY 10027, USA
| |
Collapse
|
33
|
Oberlohr V, Lengel H, Hambright WS, Whitney KE, Evans TA, Huard J. Biologics for Skeletal Muscle Healing: The Role of Senescence and Platelet-Based Treatment Modalities. OPER TECHN SPORT MED 2020. [DOI: 10.1016/j.otsm.2020.150754] [Citation(s) in RCA: 59] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
|
34
|
Wilkinson HN, Hardman MJ. Senescence in Wound Repair: Emerging Strategies to Target Chronic Healing Wounds. Front Cell Dev Biol 2020; 8:773. [PMID: 32850866 PMCID: PMC7431694 DOI: 10.3389/fcell.2020.00773] [Citation(s) in RCA: 105] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2020] [Accepted: 07/22/2020] [Indexed: 01/10/2023] Open
Abstract
Cellular senescence is a fundamental stress response that restrains tumour formation. Yet, senescence cells are also present in non-cancerous states, accumulating exponentially with chronological age and contributing to age- and diabetes-related cellular dysfunction. The identification of hypersecretory and phagocytic behaviours in cells that were once believed to be non-functional has led to a recent explosion of senescence research. Here we discuss the profound, and often opposing, roles identified for short-lived vs. chronic tissue senescence. Transiently induced senescence is required for development, regeneration and acute wound repair, while chronic senescence is widely implicated in tissue pathology. We recently demonstrated that sustained senescence contributes to impaired diabetic healing via the CXCR2 receptor, which when blocked promotes repair. Further studies have highlighted the beneficial effects of targeting a range of senescence-linked processes to fight disease. Collectively, these findings hold promise for developing clinically viable strategies to tackle senescence in chronic wounds and other cutaneous pathologies.
Collapse
Affiliation(s)
- Holly N Wilkinson
- Centre for Atherothrombosis and Metabolic Disease, Hull York Medical School, University of Hull, Hull, United Kingdom
| | - Matthew J Hardman
- Centre for Atherothrombosis and Metabolic Disease, Hull York Medical School, University of Hull, Hull, United Kingdom
| |
Collapse
|
35
|
Sieben CJ, Jeganathan KB, Nelson GG, Sturmlechner I, Zhang C, van Deursen WH, Bakker B, Foijer F, Li H, Baker DJ, van Deursen JM. BubR1 allelic effects drive phenotypic heterogeneity in mosaic-variegated aneuploidy progeria syndrome. J Clin Invest 2020; 130:171-188. [PMID: 31738183 DOI: 10.1172/jci126863] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2018] [Accepted: 09/18/2019] [Indexed: 12/21/2022] Open
Abstract
Mosaic-variegated aneuploidy (MVA) syndrome is a rare childhood disorder characterized by biallelic BUBR1, CEP57, or TRIP13 aberrations; increased chromosome missegregation; and a broad spectrum of clinical features, including various cancers, congenital defects, and progeroid pathologies. To investigate the mechanisms underlying this disorder and its phenotypic heterogeneity, we mimicked the BUBR1L1012P mutation in mice (BubR1L1002P) and combined it with 2 other MVA variants, BUBR1X753 and BUBR1H, generating a truncated protein and low amounts of wild-type protein, respectively. Whereas BubR1X753/L1002P and BubR1H/X753 mice died prematurely, BubR1H/L1002P mice were viable and exhibited many MVA features, including cancer predisposition and various progeroid phenotypes, such as short lifespan, dwarfism, lipodystrophy, sarcopenia, and low cardiac stress tolerance. Strikingly, although these mice had a reduction in total BUBR1 and spectrum of MVA phenotypes similar to that of BubR1H/H mice, several progeroid pathologies were attenuated in severity, which in skeletal muscle coincided with reduced senescence-associated secretory phenotype complexity. Additionally, mice carrying monoallelic BubR1 mutations were prone to select MVA-related pathologies later in life, with predisposition to sarcopenia correlating with mTORC1 hyperactivity. Together, these data demonstrate that BUBR1 allelic effects beyond protein level and aneuploidy contribute to disease heterogeneity in both MVA patients and heterozygous carriers of MVA mutations.
Collapse
Affiliation(s)
| | | | | | | | - Cheng Zhang
- Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, Minnesota, USA
| | | | - Bjorn Bakker
- European Research Institute for the Biology of Aging, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Floris Foijer
- European Research Institute for the Biology of Aging, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Hu Li
- Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, Minnesota, USA
| | - Darren J Baker
- Departments of Biochemistry and Molecular Biology.,Pediatric and Adolescent Medicine, and
| | - Jan M van Deursen
- Departments of Biochemistry and Molecular Biology.,Pediatric and Adolescent Medicine, and
| |
Collapse
|
36
|
Mankhong S, Kim S, Moon S, Kwak HB, Park DH, Kang JH. Experimental Models of Sarcopenia: Bridging Molecular Mechanism and Therapeutic Strategy. Cells 2020; 9:E1385. [PMID: 32498474 PMCID: PMC7348939 DOI: 10.3390/cells9061385] [Citation(s) in RCA: 82] [Impact Index Per Article: 16.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Revised: 05/27/2020] [Accepted: 05/27/2020] [Indexed: 12/15/2022] Open
Abstract
Sarcopenia has been defined as a progressive decline of skeletal muscle mass, strength, and functions in elderly people. It is accompanied by physical frailty, functional disability, falls, hospitalization, and mortality, and is becoming a major geriatric disorder owing to the increasing life expectancy and growing older population worldwide. Experimental models are critical to understand the pathophysiology of sarcopenia and develop therapeutic strategies. Although its etiologies remain to be further elucidated, several mechanisms of sarcopenia have been identified, including cellular senescence, proteostasis imbalance, oxidative stress, and "inflammaging." In this article, we address three main aspects. First, we describe the fundamental aging mechanisms. Next, we discuss both in vitro and in vivo experimental models based on molecular mechanisms that have the potential to elucidate the biochemical processes integral to sarcopenia. The use of appropriate models to reflect sarcopenia and/or its underlying pathways will enable researchers to understand sarcopenia and develop novel therapeutic strategies for sarcopenia. Lastly, we discuss the possible molecular targets and the current status of drug candidates for sarcopenia treatment. In conclusion, the development of experimental models for sarcopenia is essential to discover molecular targets that are valuable as biochemical biomarkers and/or therapeutic targets for sarcopenia.
Collapse
Affiliation(s)
- Sakulrat Mankhong
- Department of Pharmacology, Hypoxia-related Disease Research Center, College of Medicine, Inha University, Incheon 22212, Korea; (S.M.); (S.K.); (S.M.)
| | - Sujin Kim
- Department of Pharmacology, Hypoxia-related Disease Research Center, College of Medicine, Inha University, Incheon 22212, Korea; (S.M.); (S.K.); (S.M.)
- Department of Kinesiology, Inha University, Incheon 22212, Korea; (H.-B.K.); (D.-H.P.)
- Institute of Sports & Arts Convergence (ISAC), Inha University, Incheon 22212, Korea
| | - Sohee Moon
- Department of Pharmacology, Hypoxia-related Disease Research Center, College of Medicine, Inha University, Incheon 22212, Korea; (S.M.); (S.K.); (S.M.)
| | - Hyo-Bum Kwak
- Department of Kinesiology, Inha University, Incheon 22212, Korea; (H.-B.K.); (D.-H.P.)
- Institute of Sports & Arts Convergence (ISAC), Inha University, Incheon 22212, Korea
| | - Dong-Ho Park
- Department of Kinesiology, Inha University, Incheon 22212, Korea; (H.-B.K.); (D.-H.P.)
- Institute of Sports & Arts Convergence (ISAC), Inha University, Incheon 22212, Korea
| | - Ju-Hee Kang
- Department of Pharmacology, Hypoxia-related Disease Research Center, College of Medicine, Inha University, Incheon 22212, Korea; (S.M.); (S.K.); (S.M.)
- Institute of Sports & Arts Convergence (ISAC), Inha University, Incheon 22212, Korea
| |
Collapse
|
37
|
Insights from In Vivo Studies of Cellular Senescence. Cells 2020; 9:cells9040954. [PMID: 32295081 PMCID: PMC7226957 DOI: 10.3390/cells9040954] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2020] [Revised: 04/09/2020] [Accepted: 04/10/2020] [Indexed: 01/07/2023] Open
Abstract
Cellular senescence is the dynamic process of durable cell-cycle arrest. Senescent cells remain metabolically active and often acquire a distinctive bioactive secretory phenotype. Much of our molecular understanding in senescent cell biology comes from studies using mammalian cell lines exposed to stress or extended culture periods. While less well understood mechanistically, senescence in vivo is becoming appreciated for its numerous biological implications, both in the context of beneficial processes, such as development, tumor suppression, and wound healing, and in detrimental conditions, where senescent cell accumulation has been shown to contribute to aging and age-related diseases. Importantly, clearance of senescent cells, through either genetic or pharmacological means, has been shown to not only extend the healthspan of prematurely and naturally aged mice but also attenuate pathology in mouse models of chronic disease. These observations have prompted an investigation of how and why senescent cells accumulate with aging and have renewed exploration into the characteristics of cellular senescence in vivo. Here, we highlight our molecular understanding of the dynamics that lead to a cellular arrest and how various effectors may explain the consequences of senescence in tissues. Lastly, we discuss how exploitation of strategies to eliminate senescent cells or their effects may have clinical utility.
Collapse
|
38
|
Montano M, Bhasin S, D'Aquila RT, Erlandson KM, Evans WJ, Funderburg NT, Justice A, Ndhlovu LC, Ojikutu B, Pahor M, Pahwa S, Ryan AS, Schrack J, Schultz MB, Sebastiani P, Sinclair DA, Tripp J, Walker B, Womack JA, Yung R, Reeves RK. Harvard HIV and Aging Workshop: Perspectives and Priorities from Claude D. Pepper Centers and Centers for AIDS Research. AIDS Res Hum Retroviruses 2019; 35:999-1012. [PMID: 31456412 DOI: 10.1089/aid.2019.0130] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
People aging with HIV (PAWH) infection experience greater impairments in physical and cognitive function, in addition to higher rates of peripheral comorbid conditions (e.g., renal failure, diabetes, bone fracture, hypertension, cardiovascular disease, polypharmacy, and multimorbidity). While multifactorial drivers, including HIV infection itself, antiretroviral therapy-related toxicities, disparities in care, and biobehavioral factors, likely contribute, there remains an overarching question as to what are the relevant age-related mechanisms and models that could inform interventions that promote health span and life span in PAWH? This workshop was convened to hear from experts on the biology of aging and HIV researchers studying PAWH to focus on advancing investigations at the interface of HIV and Aging. In this study, we summarize the discussions from the Harvard Center for AIDS Research and Boston Claude D. Pepper cosponsored workshop on HIV and Aging, which took place in October 2018.
Collapse
Affiliation(s)
- Monty Montano
- Boston Pepper OAIC, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
- Men's Health: Aging and Metabolism, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Shalender Bhasin
- Boston Pepper OAIC, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
- Men's Health: Aging and Metabolism, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | | | | | - William J Evans
- Department of Nutritional Sciences and Toxicology, University of California, Berkeley, California
| | - Nicholas T Funderburg
- Division of Medical Laboratory Science, School of Health and Rehabilitation Sciences, Ohio State University, Columbus, Ohio
| | - Amy Justice
- Department of Medicine, Yale University School of Medicine, New Haven, Connecticut
- VA Connecticut Healthcare System, West Haven, Connecticut
| | - Lishomwa C Ndhlovu
- Department of Tropical Medicine, Medical Microbiology and Pharmacology, John A. Burns School of Medicine, University of Hawaii at Manoa, Honolulu, Hawaii
| | - Bisola Ojikutu
- Division of Global Health Equity, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Marco Pahor
- Institute on Aging, Department of Aging and Geriatric Research, College of Medicine, University of Florida, Gainesville, Florida
| | - Savita Pahwa
- Department of Microbiology and Immunology, Miller School of Medicine, University of Miami, Miami, Florida
| | - Alice S Ryan
- Division of Gerontology and Geriatric Medicine, Department of Medicine, University of Maryland School of Medicine, Baltimore, Maryland
- Baltimore Veterans Affairs Geriatric Research Education and Clinical Center and Research and Development Service, Baltimore, Maryland
| | - Jennifer Schrack
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland
| | - Michael B Schultz
- Department of Genetics, Paul F. Glenn Labs for the Biology of Aging, Blavatnik Institute, Harvard Medical School, Boston, Massachusetts
| | - Paola Sebastiani
- Department of Biostatistics, Boston University, Boston, Massachusetts
| | - David A Sinclair
- Department of Genetics, Paul F. Glenn Labs for the Biology of Aging, Blavatnik Institute, Harvard Medical School, Boston, Massachusetts
| | - Julia Tripp
- Harvard University Center for AIDS Research, Cambridge, Massachusetts
| | - Bruce Walker
- Ragon Institute of MGH, MIT and Harvard, Cambridge, Massachusetts
| | - Julie A Womack
- VA Connecticut Healthcare System, West Haven, Connecticut
- Yale School of Nursing, West Haven, Connecticut
| | - Raymond Yung
- Division of Geriatric and Palliative Medicine, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan
| | - R Keith Reeves
- Ragon Institute of MGH, MIT and Harvard, Cambridge, Massachusetts
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
39
|
Folgueras AR, Freitas-Rodríguez S, Velasco G, López-Otín C. Mouse Models to Disentangle the Hallmarks of Human Aging. Circ Res 2019; 123:905-924. [PMID: 30355076 DOI: 10.1161/circresaha.118.312204] [Citation(s) in RCA: 59] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Model organisms have provided fundamental evidence that aging can be delayed and longevity extended. These findings gave rise to a new era in aging research aimed at elucidating the pathways and networks controlling this complex biological process. The identification of 9 hallmarks of aging has established a framework to evaluate the relative contribution of each hallmark and the interconnections among them. In this review, we revisit these hallmarks with the information obtained exclusively through the generation of genetically modified mouse models that have a significant impact on the aging process. We discuss within each hallmark those interventions that accelerate aging or that have been successful at increasing lifespan, with the final goal of identifying the most promising antiaging avenues based on the current knowledge provided by in vivo models.
Collapse
Affiliation(s)
- Alicia R Folgueras
- From the Departamento de Bioquímica y Biología Molecular, Facultad de Medicina, Instituto Universitario de Oncología del Principado de Asturias, Universidad de Oviedo, Spain
| | - Sandra Freitas-Rodríguez
- From the Departamento de Bioquímica y Biología Molecular, Facultad de Medicina, Instituto Universitario de Oncología del Principado de Asturias, Universidad de Oviedo, Spain
| | - Gloria Velasco
- From the Departamento de Bioquímica y Biología Molecular, Facultad de Medicina, Instituto Universitario de Oncología del Principado de Asturias, Universidad de Oviedo, Spain
| | - Carlos López-Otín
- From the Departamento de Bioquímica y Biología Molecular, Facultad de Medicina, Instituto Universitario de Oncología del Principado de Asturias, Universidad de Oviedo, Spain
| |
Collapse
|
40
|
Hart CR, Ryan ZC, Pfaffenbach KT, Dasari S, Parvizi M, Lalia AZ, Lanza IR. Attenuated activation of the unfolded protein response following exercise in skeletal muscle of older adults. Aging (Albany NY) 2019; 11:7587-7604. [PMID: 31525732 PMCID: PMC6781982 DOI: 10.18632/aging.102273] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2019] [Accepted: 09/05/2019] [Indexed: 12/11/2022]
Abstract
Sarcopenia is linked with impaired adaptive responses to exercise in aging skeletal muscle. The unfolded protein response (UPR) is an important intramyocellular molecular response pathway that is activated by exercise. The influence of age on skeletal muscle adaptive UPR in response to exercise, and the relationship to other key exercise-responsive regulatory pathways is not well-understood. We evaluated age-related changes in transcriptional markers of UPR activation following a single bout of resistance exercise in 12 young (27 ± 5yrs) and 12 older (75 ± 5yrs) healthy men and women. At baseline, there were modest differences in expression of UPR-related genes in young and older adults. Following exercise, transcriptional markers of UPR pathway activation were attenuated in older adults compared to young based on specific salient UPR-related genes and gene set enrichment analysis. The coordination of post-exercise transcriptional patterns between the UPR pathway, p53/p21 axis of autophagy, and satellite cell differentiation were less evident in older compared to young adults. In conclusion, transcriptomic analysis revealed an age-related decline in the adaptive UPR transcriptional response following a single bout of exercise that could contribute to impaired exercise responsiveness with age.
Collapse
Affiliation(s)
- Corey R. Hart
- Division of Endocrinology and Metabolism, Mayo Clinic College of Medicine, Rochester, MN 55905, USA
| | - Zachary C. Ryan
- Division of Endocrinology and Metabolism, Mayo Clinic College of Medicine, Rochester, MN 55905, USA
| | - Kyle T. Pfaffenbach
- Department of Physical Activity and Health, Eastern Oregon University, La Grande, OR 97850, USA
| | - Surendra Dasari
- Division of Biostatistics and Informatics, Mayo Clinic College of Medicine, Rochester, MN 55905, USA
| | - Mojtaba Parvizi
- Division of Endocrinology and Metabolism, Mayo Clinic College of Medicine, Rochester, MN 55905, USA
| | - Antigoni Z. Lalia
- Division of Endocrinology and Metabolism, Mayo Clinic College of Medicine, Rochester, MN 55905, USA
| | - Ian R. Lanza
- Division of Endocrinology and Metabolism, Mayo Clinic College of Medicine, Rochester, MN 55905, USA
| |
Collapse
|
41
|
Yousefzadeh MJ, Melos KI, Angelini L, Burd CE, Robbins PD, Niedernhofer LJ. Mouse Models of Accelerated Cellular Senescence. Methods Mol Biol 2019; 1896:203-230. [PMID: 30474850 DOI: 10.1007/978-1-4939-8931-7_17] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Senescent cells accumulate in multiple tissues as virtually all vertebrate organisms age. Senescence is a highly conserved response to many forms of cellular stress intended to block the propagation of damaged cells. Senescent cells have been demonstrated to play a causal role in aging via their senescence-associated secretory phenotype and by impeding tissue regeneration. Depletion of senescent cells either through genetic or pharmacologic methods has been demonstrated to extend murine lifespan and delay the onset of age-related diseases. Measuring the burden and location of senescent cells in vivo remains challenging, as there is no marker unique to senescent cells. Here, we describe multiple methods to detect the presence and extent of cellular senescence in preclinical models, with a special emphasis on murine models of accelerated aging that exhibit a more rapid onset of cellular senescence.
Collapse
Affiliation(s)
- Matthew J Yousefzadeh
- Institute on the Biology of Aging and Metabolism, University of Minnesota, Minneapolis, MN, USA
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Minneapolis, MN, USA
- Department of Molecular Medicine, The Scripps Research Institute, Jupiter, FL, USA
| | - Kendra I Melos
- Department of Molecular Medicine, The Scripps Research Institute, Jupiter, FL, USA
| | - Luise Angelini
- Institute on the Biology of Aging and Metabolism, University of Minnesota, Minneapolis, MN, USA
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Minneapolis, MN, USA
- Department of Molecular Medicine, The Scripps Research Institute, Jupiter, FL, USA
| | - Christin E Burd
- Department of Molecular Genetics, The Ohio State University, Columbus, OH, USA
- Department of Cancer Biology and Genetics, The Ohio State University, Columbus, OH, USA
| | - Paul D Robbins
- Institute on the Biology of Aging and Metabolism, University of Minnesota, Minneapolis, MN, USA
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Minneapolis, MN, USA
- Department of Molecular Medicine, The Scripps Research Institute, Jupiter, FL, USA
| | - Laura J Niedernhofer
- Institute on the Biology of Aging and Metabolism, University of Minnesota, Minneapolis, MN, USA.
| |
Collapse
|
42
|
Shakeri H, Gevaert AB, Schrijvers DM, De Meyer GRY, De Keulenaer GW, Guns PJDF, Lemmens K, Segers VF. Neuregulin-1 attenuates stress-induced vascular senescence. Cardiovasc Res 2019. [PMID: 29528383 DOI: 10.1093/cvr/cvy059] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Aims Cardiovascular ageing is a key determinant of life expectancy. Cellular senescence, a state of irreversible cell cycle arrest, is an important contributor to ageing due to the accumulation of damaged cells. Targeting cellular senescence could prevent age-related cardiovascular diseases. In this study, we investigated the effects of neuregulin-1 (NRG-1), an epidermal growth factor with cardioprotective and anti-atherosclerotic effects, on cellular senescence. Methods and results Senescence was induced in cultured rat aortic endothelial cells (ECs) and aortic smooth muscle cells (SMCs) by 2 h exposure to 30 µM hydrogen peroxide (H2O2). Cellular senescence was confirmed after 72 h using senescence-associated-β-galactosidase staining (SA-β-gal), cell surface area, and western blot analyses of SA pathways (acetyl-p53, p21). Recombinant human NRG-1 (rhNRG-1, 20 ng/mL) significantly reduced H2O2-induced senescence, as shown by a lower number of SA-β-gal positive cells, smaller surface area and lower expression of acetyl-p53. In C57BL/6 male mice rendered diabetic with streptozotocin (STZ), rhNRG-1 attenuated cellular senescence in aortic ECs and SMCs. Next, we created mice with SMC-specific knockdown of the NRG-1 receptor ErbB4. Aortic SMCs isolated from SMC-specific ErbB4 deficient mice (ErbB4f/+ SM22α-Cre+) showed earlier cellular senescence in vitro compared with wild-type (ErbB4+/+ SM22α-Cre+) SMCs. Furthermore, when rendered diabetic with STZ, ErbB4f/+ SM22α-Cre+ male mice showed significantly more vascular senescence than their diabetic wild-type littermates and had increased mortality. Conclusions This study is the first to explore the role of NRG-1 in vascular senescence. Our data demonstrate that NRG-1 markedly inhibits stress-induced premature senescence in vascular cells in vitro and in the aorta of diabetic mice in vivo. Consistently, deficiency in the NRG-1 receptor ErbB4 provokes cellular senescence in vitro as well as in vivo.
Collapse
Affiliation(s)
- Hadis Shakeri
- Laboratory of Physiopharmacology, University of Antwerp, Universiteitsplein 1, 2610 Antwerp, Belgium
| | - Andreas B Gevaert
- Laboratory of Physiopharmacology, University of Antwerp, Universiteitsplein 1, 2610 Antwerp, Belgium.,Department of Cardiology.,Laboratory for Cellular and Molecular Cardiology, Department of Cardiology, Antwerp University Hospital (UZA), Antwerp, Belgium
| | - Dorien M Schrijvers
- Laboratory of Physiopharmacology, University of Antwerp, Universiteitsplein 1, 2610 Antwerp, Belgium
| | - Guido R Y De Meyer
- Laboratory of Physiopharmacology, University of Antwerp, Universiteitsplein 1, 2610 Antwerp, Belgium
| | - Gilles W De Keulenaer
- Laboratory of Physiopharmacology, University of Antwerp, Universiteitsplein 1, 2610 Antwerp, Belgium
| | - Pieter-Jan D F Guns
- Laboratory of Physiopharmacology, University of Antwerp, Universiteitsplein 1, 2610 Antwerp, Belgium
| | - Katrien Lemmens
- Laboratory of Physiopharmacology, University of Antwerp, Universiteitsplein 1, 2610 Antwerp, Belgium
| | - Vincent F Segers
- Laboratory of Physiopharmacology, University of Antwerp, Universiteitsplein 1, 2610 Antwerp, Belgium.,Department of Cardiology
| |
Collapse
|
43
|
Cohen J, Torres C. Astrocyte senescence: Evidence and significance. Aging Cell 2019; 18:e12937. [PMID: 30815970 PMCID: PMC6516680 DOI: 10.1111/acel.12937] [Citation(s) in RCA: 187] [Impact Index Per Article: 31.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2018] [Revised: 01/14/2019] [Accepted: 02/04/2019] [Indexed: 12/12/2022] Open
Abstract
Astrocytes participate in numerous aspects of central nervous system (CNS) physiology ranging from ion balance to metabolism, and disruption of their physiological roles can therefore be a contributor to CNS dysfunction and pathology. Cellular senescence, one of the mechanisms of aging, has been proposed as a central component of the age dependency of neurodegenerative disorders. Cumulative evidence supports an integral role of astrocytes in the initiation and progression of neurodegenerative disease and cognitive decline with aging. The loss of astrocyte function or the gain of neuroinflammatory function as a result of cellular senescence could have profound implications for the aging brain and neurodegenerative disorders, and we propose the term “astrosenescence” to describe this phenotype. This review summarizes the current evidence pertaining to astrocyte senescence from early evidence, in vitro characterization and relationship to age‐related neurodegenerative disease. We discuss the significance of targeting senescent astrocytes as a novel approach toward therapies for age‐associated neurodegenerative disease.
Collapse
Affiliation(s)
- Justin Cohen
- Department of Pathology and Laboratory Medicine Drexel University College of Medicine Philadelphia Pennsylvania
| | - Claudio Torres
- Department of Pathology and Laboratory Medicine Drexel University College of Medicine Philadelphia Pennsylvania
| |
Collapse
|
44
|
Postmus AC, Sturmlechner I, Jonker JW, van Deursen JM, van de Sluis B, Kruit JK. Senescent cells in the development of cardiometabolic disease. Curr Opin Lipidol 2019; 30:177-185. [PMID: 30913069 PMCID: PMC6530963 DOI: 10.1097/mol.0000000000000602] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
PURPOSE OF REVIEW Senescent cells have recently been identified as key players in the development of metabolic dysfunction. In this review, we will highlight recent developments in this field and discuss the concept of targeting these cells to prevent or treat cardiometabolic diseases. RECENT FINDINGS Evidence is accumulating that cellular senescence contributes to adipose tissue dysfunction, presumably through induction of low-grade inflammation and inhibition of adipogenic differentiation leading to insulin resistance and dyslipidaemia. Senescent cells modulate their surroundings through their bioactive secretome and only a relatively small number of senescent cells is sufficient to cause persistent physical dysfunction even in young mice. Proof-of-principle studies showed that selective elimination of senescent cells can prevent or delay the development of cardiometabolic diseases in mice. SUMMARY The metabolic consequences of senescent cell accumulation in various tissues are now unravelling and point to new therapeutic opportunities for the treatment of cardiometabolic diseases.
Collapse
Affiliation(s)
- Andrea C. Postmus
- Department of Pediatrics, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Ines Sturmlechner
- Departments of Pediatrics and Adolescent Medicine
- Department of Biochemistry and Molecular Biology, Mayo Clinic College of Medicine, Mayo Clinic, Rochester, Minnesota, USA
| | - Johan W. Jonker
- Department of Pediatrics, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Jan M. van Deursen
- Departments of Pediatrics and Adolescent Medicine
- Department of Biochemistry and Molecular Biology, Mayo Clinic College of Medicine, Mayo Clinic, Rochester, Minnesota, USA
| | - Bart van de Sluis
- Department of Pediatrics, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Janine K. Kruit
- Department of Pediatrics, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| |
Collapse
|
45
|
Liu X, Wan M. A tale of the good and bad: Cell senescence in bone homeostasis and disease. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2019; 346:97-128. [PMID: 31122396 DOI: 10.1016/bs.ircmb.2019.03.005] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Historically, cellular senescence has been viewed as an irreversible cell-cycle arrest process with distinctive phenotypic alterations that were implicated primarily in aging and tumor suppression. Recent discoveries suggest that cellular senescence represents a series of diverse, dynamic, and heterogeneous cellular states with the senescence-associated secretory phenotype (SASP). Although senescent cells typically contribute to aging and age-related diseases, accumulating evidence has shown that they also have important physiological functions during embryonic development, late pubertal bone growth cessation, and adulthood tissue remodeling. Here, we review the recent research on cellular senescence and SASP, highlighting the key pathways that mediate senescence cell-cycle arrest and initiate SASP. We also summarize recent literature on the role of cellular senescence in maintaining bone homeostasis and mediating age-associated osteoporosis, discussing both the beneficial and adverse roles of cellular senescence in bone during different physiological stages, including bone development, childhood bone growth, adulthood bone remodeling, and bone aging.
Collapse
Affiliation(s)
- Xiaonan Liu
- Department of Orthopaedic Surgery, The Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Mei Wan
- Department of Orthopaedic Surgery, The Johns Hopkins University School of Medicine, Baltimore, MD, United States.
| |
Collapse
|
46
|
Sannicandro AJ, Soriano-Arroquia A, Goljanek-Whysall K. Micro(RNA)-managing muscle wasting. J Appl Physiol (1985) 2019; 127:619-632. [PMID: 30991011 DOI: 10.1152/japplphysiol.00961.2018] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Progressive skeletal muscle wasting is a natural consequence of aging and is common in chronic and acute diseases. Loss of skeletal muscle mass and function (strength) often leads to frailty, decreased independence, and increased risk of hospitalization. Despite progress made in our understanding of the mechanisms underlying muscle wasting, there is still no treatment available, with exercise training and dietary supplementation improving, but not restoring, muscle mass and/or function. There has been slow progress in developing novel therapies for muscle wasting, either during aging or disease, partially due to the complex nature of processes underlying muscle loss. The mechanisms of muscle wasting are multifactorial, with a combination of factors underlying age- and disease-related functional muscle decline. These factors include well-characterized changes in muscle such as changes in protein turnover and more recently described mechanisms such as autophagy or satellite cell senescence. Advances in transcriptomics and other high-throughput approaches have highlighted significant deregulation of skeletal muscle gene and protein levels during aging and disease. These changes are regulated at different levels, including posttranscriptional gene expression regulation by microRNAs. microRNAs, potent regulators of gene expression, modulate many processes in muscle, and microRNA-based interventions have been recently suggested as a promising new therapeutic strategy against alterations in muscle homeostasis. Here, we review recent developments in understanding the aging-associated mechanisms of muscle wasting and explore potential microRNA-based therapeutic avenues.
Collapse
Affiliation(s)
- Anthony J Sannicandro
- Department of Physiology, School of Medicine, National University of Ireland, Galway, Ireland
| | - Ana Soriano-Arroquia
- Institute of Ageing and Chronic Disease, University of Liverpool, United Kingdom
| | - Katarzyna Goljanek-Whysall
- Department of Physiology, School of Medicine, National University of Ireland, Galway, Ireland.,Institute of Ageing and Chronic Disease, University of Liverpool, United Kingdom
| |
Collapse
|
47
|
He Q, Gao Y, Wang T, Zhou L, Zhou W, Yuan Z. Deficiency of Yes-Associated Protein Induces Cataract in Mice. Aging Dis 2019; 10:293-306. [PMID: 31011480 PMCID: PMC6457047 DOI: 10.14336/ad.2018.0910] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2018] [Accepted: 09/10/2018] [Indexed: 12/25/2022] Open
Abstract
Cataract is a major cause of blindness worldwide, its complicated and unclear etiopathogenesis limit effective therapy. Here, we found that Yap, a downstream effector of the Hippo pathway, is specifically expressed in lens epithelial cells and Yap conditional knockout (cKO) in the lens leads to cataract. Histologically, Yap deficient lens show fewer epithelial cells, retention of nuclei and accumulation of morgagnian globules in the transitional zone and the posterior area. Mechanistically, GFAP-mediated Yap cKO leads to the reduced proliferation of epithelial cells, delayed fiber cell denucleation and increased cellular senescence in lens. Further RNA profiling analysis reveals Yap cKO results in a significant alteration in gene transcription that is involved in eye development, lens structure, inflammation, cellular proliferation and polarity. Collectively, our data reveal a novel function of Yap in the lens and links Yap deficiency with the development of cataract, making Yap a promising target for cataract therapy.
Collapse
Affiliation(s)
- Qing He
- 1State Key Laboratory of Brain and Cognitive Sciences, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China.,2The Brain Science Center, Beijing Institute of Basic Medical Sciences, Beijing 100850, China.,3College of Life Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yuhao Gao
- 1State Key Laboratory of Brain and Cognitive Sciences, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China.,2The Brain Science Center, Beijing Institute of Basic Medical Sciences, Beijing 100850, China.,3College of Life Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Tongxing Wang
- 4Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China.,5State Key Laboratory of Toxicology and Medical Countermeasures, Beijing 100850, China
| | - Lujun Zhou
- 2The Brain Science Center, Beijing Institute of Basic Medical Sciences, Beijing 100850, China.,3College of Life Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Wenxia Zhou
- 4Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China.,5State Key Laboratory of Toxicology and Medical Countermeasures, Beijing 100850, China
| | - Zengqiang Yuan
- 2The Brain Science Center, Beijing Institute of Basic Medical Sciences, Beijing 100850, China
| |
Collapse
|
48
|
Age-related inflammation triggers skeletal stem/progenitor cell dysfunction. Proc Natl Acad Sci U S A 2019; 116:6995-7004. [PMID: 30894483 DOI: 10.1073/pnas.1810692116] [Citation(s) in RCA: 144] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Aging is associated with impaired tissue regeneration. Stem cell number and function have been identified as potential culprits. We first demonstrate a direct correlation between stem cell number and time to bone fracture union in a human patient cohort. We then devised an animal model recapitulating this age-associated decline in bone healing and identified increased cellular senescence caused by a systemic and local proinflammatory environment as the major contributor to the decline in skeletal stem/progenitor cell (SSPC) number and function. Decoupling age-associated systemic inflammation from chronological aging by using transgenic Nfkb1KO mice, we determined that the elevated inflammatory environment, and not chronological age, was responsible for the decrease in SSPC number and function. By using a pharmacological approach inhibiting NF-κB activation, we demonstrate a functional rejuvenation of aged SSPCs with decreased senescence, increased SSPC number, and increased osteogenic function. Unbiased, whole-genome RNA sequencing confirmed the reversal of the aging phenotype. Finally, in an ectopic model of bone healing, we demonstrate a functional restoration of regenerative potential in aged SSPCs. These data identify aging-associated inflammation as the cause of SSPC dysfunction and provide mechanistic insights into its reversal.
Collapse
|
49
|
Melo Pereira S, Ribeiro R, Logarinho E. Approaches towards Longevity: Reprogramming, Senolysis, and Improved Mitotic Competence as Anti-Aging Therapies. Int J Mol Sci 2019; 20:E938. [PMID: 30795536 PMCID: PMC6413205 DOI: 10.3390/ijms20040938] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2019] [Revised: 02/09/2019] [Accepted: 02/18/2019] [Indexed: 02/06/2023] Open
Abstract
Mainstream approaches that are currently used as anti-aging therapies primarily explore the senescence and epigenetic drift aging hallmarks and they are at two ends of the spectrum. While senolytic therapies include either the selective elimination of senescent cells or the disruption of their secretome with the use of drugs or natural compounds, cellular reprogramming uses genetic manipulation to revert cells all the way back to pluripotency. Here, we describe the progress that has been made on these therapies, while highlighting the major challenges involved. Moreover, based on recent findings elucidating the impact of mitotic shutdown and aneuploidy in cellular senescence, we discuss the modulation of mitotic competence as an alternative strategy to delay the hallmarks of aging. We propose that a regulated rise in mitotic competence of cells could circumvent certain limitations that are present in the senolytic and reprogramming approaches, by acting to decelerate senescence and possibly restore the epigenetic landscape.
Collapse
Affiliation(s)
- Sofia Melo Pereira
- Ageing and Aneuploidy Laboratory, IBMC, Instituto de Biologia Molecular e Celular, Universidade do Porto, 4200-135 Porto, Portugal.
- i3S, Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal.
| | - Rui Ribeiro
- Ageing and Aneuploidy Laboratory, IBMC, Instituto de Biologia Molecular e Celular, Universidade do Porto, 4200-135 Porto, Portugal.
- i3S, Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal.
| | - Elsa Logarinho
- Ageing and Aneuploidy Laboratory, IBMC, Instituto de Biologia Molecular e Celular, Universidade do Porto, 4200-135 Porto, Portugal.
- i3S, Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal.
- Cell Division Unit, Faculty of Medicine, Department of Experimental Biology, Universidade do Porto, 4200-319 Porto, Portugal.
| |
Collapse
|
50
|
Goulart-Filho JAV, Montalli VAM, Passador-Santos F, de Araújo NS, de Araújo VC. Role of apoptotic, autophagic and senescence pathways in minor salivary gland adenoid cystic carcinoma. Diagn Pathol 2019; 14:14. [PMID: 30736793 PMCID: PMC6368765 DOI: 10.1186/s13000-019-0796-2] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2018] [Accepted: 02/01/2019] [Indexed: 12/16/2022] Open
Abstract
Background Adenoid cystic carcinoma (ACC) is a salivary gland malignancy with poor long-term survival, which warrants studies aimed at clarifying the pathogenesis of this disease in order to widen the scope of therapeutic options currently available. Alterations in regulatory mechanisms relating to vascular support, cell death and autophagy are important pathways for tumor growth in cancer. Thus, the present study aimed to access vascular supply, apoptosis, autophagy and cell senescence in ACC of minor salivary glands. Methods We analyzed 25 cases of minor salivary gland ACC by immunohistochemistry using anti-CD34, anti-CD105, anti-D2–40, anti-Bax, anti-Bcl-2, anti-Beclin-1, anti-LC3B, anti-p21 and anti-p16. Results Microvessel density was low and based on anti-CD34, anti-CD105 and anti-D2–40 immunostaining. There was positivity for anti-CD34, anti-Bcl-2, anti-Beclin, anti-LC3B and anti-p21 and a positive correlation between Bcl-2 and Beclin (p = 0.014). Conclusions Our results showed that ACC does not depend on neo-angiogenesis and is probably associated to anti-apoptotic, autophagic and anti-senescence events.
Collapse
Affiliation(s)
- João Augusto Vianna Goulart-Filho
- Department of Oral Pathology, São Leopoldo Mandic Research Institute, Rua José Rocha Junqueira, 13, CEP, Campinas, SP, 13045-610, Brazil
| | - Victor Angelo Martins Montalli
- Department of Oral Pathology, São Leopoldo Mandic Research Institute, Rua José Rocha Junqueira, 13, CEP, Campinas, SP, 13045-610, Brazil
| | - Fabrício Passador-Santos
- Department of Oral Pathology, São Leopoldo Mandic Research Institute, Rua José Rocha Junqueira, 13, CEP, Campinas, SP, 13045-610, Brazil
| | - Ney Soares de Araújo
- Department of Oral Pathology, São Leopoldo Mandic Research Institute, Rua José Rocha Junqueira, 13, CEP, Campinas, SP, 13045-610, Brazil
| | - Vera Cavalcanti de Araújo
- Department of Oral Pathology, São Leopoldo Mandic Research Institute, Rua José Rocha Junqueira, 13, CEP, Campinas, SP, 13045-610, Brazil.
| |
Collapse
|