601
|
Hickson LJ, Langhi Prata LGP, Bobart SA, Evans TK, Giorgadze N, Hashmi SK, Herrmann SM, Jensen MD, Jia Q, Jordan KL, Kellogg TA, Khosla S, Koerber DM, Lagnado AB, Lawson DK, LeBrasseur NK, Lerman LO, McDonald KM, McKenzie TJ, Passos JF, Pignolo RJ, Pirtskhalava T, Saadiq IM, Schaefer KK, Textor SC, Victorelli SG, Volkman TL, Xue A, Wentworth MA, Wissler Gerdes EO, Zhu Y, Tchkonia T, Kirkland JL. Senolytics decrease senescent cells in humans: Preliminary report from a clinical trial of Dasatinib plus Quercetin in individuals with diabetic kidney disease. EBioMedicine 2019; 47:446-456. [PMID: 31542391 PMCID: PMC6796530 DOI: 10.1016/j.ebiom.2019.08.069] [Citation(s) in RCA: 811] [Impact Index Per Article: 135.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2019] [Revised: 08/16/2019] [Accepted: 08/29/2019] [Indexed: 01/01/2023] Open
Abstract
Background Senescent cells, which can release factors that cause inflammation and dysfunction, the senescence-associated secretory phenotype (SASP), accumulate with ageing and at etiological sites in multiple chronic diseases. Senolytics, including the combination of Dasatinib and Quercetin (D + Q), selectively eliminate senescent cells by transiently disabling pro-survival networks that defend them against their own apoptotic environment. In the first clinical trial of senolytics, D + Q improved physical function in patients with idiopathic pulmonary fibrosis (IPF), a fatal senescence-associated disease, but to date, no peer-reviewed study has directly demonstrated that senolytics decrease senescent cells in humans. Methods In an open label Phase 1 pilot study, we administered 3 days of oral D 100 mg and Q 1000 mg to subjects with diabetic kidney disease (N = 9; 68·7 ± 3·1 years old; 2 female; BMI:33·9 ± 2·3 kg/m2; eGFR:27·0 ± 2·1 mL/min/1·73m2). Adipose tissue, skin biopsies, and blood were collected before and 11 days after completing senolytic treatment. Senescent cell and macrophage/Langerhans cell markers and circulating SASP factors were assayed. Findings D + Q reduced adipose tissue senescent cell burden within 11 days, with decreases in p16INK4A-and p21CIP1-expressing cells, cells with senescence-associated β-galactosidase activity, and adipocyte progenitors with limited replicative potential. Adipose tissue macrophages, which are attracted, anchored, and activated by senescent cells, and crown-like structures were decreased. Skin epidermal p16INK4A+ and p21CIP1+ cells were reduced, as were circulating SASP factors, including IL-1α, IL-6, and MMPs-9 and −12. Interpretation “Hit-and-run” treatment with senolytics, which in the case of D + Q have elimination half-lives <11 h, significantly decreases senescent cell burden in humans. Fund NIH and Foundations. ClinicalTrials.gov Identifier: NCT02848131. Senescence, Frailty, and Mesenchymal Stem Cell Functionality in Chronic Kidney Disease: Effect of Senolytic Agents.
Collapse
Affiliation(s)
- LaTonya J Hickson
- Cellular Senescence and Translation and Pharmacology Programs, Robert and Arlene Kogod Center on Aging, Mayo Clinic, United States of America; Division of Geriatric Medicine and Gerontology, Department of Medicine, Mayo Clinic, United States of America; Division of Nephrology and Hypertension, Department of Medicine, Mayo Clinic, United States of America
| | - Larissa G P Langhi Prata
- Cellular Senescence and Translation and Pharmacology Programs, Robert and Arlene Kogod Center on Aging, Mayo Clinic, United States of America
| | - Shane A Bobart
- Division of Nephrology and Hypertension, Department of Medicine, Mayo Clinic, United States of America
| | - Tamara K Evans
- Cellular Senescence and Translation and Pharmacology Programs, Robert and Arlene Kogod Center on Aging, Mayo Clinic, United States of America; Department of Medicine Clinical Trials Unit, Department of Medicine, Mayo Clinic, United States of America
| | - Nino Giorgadze
- Cellular Senescence and Translation and Pharmacology Programs, Robert and Arlene Kogod Center on Aging, Mayo Clinic, United States of America
| | - Shahrukh K Hashmi
- Cellular Senescence and Translation and Pharmacology Programs, Robert and Arlene Kogod Center on Aging, Mayo Clinic, United States of America; Division of Hematology, Department of Medicine, Mayo Clinic, United States of America
| | - Sandra M Herrmann
- Division of Nephrology and Hypertension, Department of Medicine, Mayo Clinic, United States of America
| | - Michael D Jensen
- Division of Endocrinology, Department of Medicine, Mayo Clinic, United States of America
| | - Qingyi Jia
- Division of Endocrinology, Department of Medicine, Mayo Clinic, United States of America
| | - Kyra L Jordan
- Division of Nephrology and Hypertension, Department of Medicine, Mayo Clinic, United States of America
| | - Todd A Kellogg
- Department of Surgery, Mayo Clinic, United States of America
| | - Sundeep Khosla
- Cellular Senescence and Translation and Pharmacology Programs, Robert and Arlene Kogod Center on Aging, Mayo Clinic, United States of America; Division of Endocrinology, Department of Medicine, Mayo Clinic, United States of America
| | - Daniel M Koerber
- Cellular Senescence and Translation and Pharmacology Programs, Robert and Arlene Kogod Center on Aging, Mayo Clinic, United States of America
| | - Anthony B Lagnado
- Cellular Senescence and Translation and Pharmacology Programs, Robert and Arlene Kogod Center on Aging, Mayo Clinic, United States of America; Department of Physiology and Biomedical Engineering, Mayo Clinic, United States of America
| | - Donna K Lawson
- Division of Hospital Medicine, Department of Medicine, Mayo Clinic, United States of America
| | - Nathan K LeBrasseur
- Cellular Senescence and Translation and Pharmacology Programs, Robert and Arlene Kogod Center on Aging, Mayo Clinic, United States of America; Department of Physiology, Mayo Clinic, United States of America; Department of Physical Medicine and Rehabilitation, Mayo Clinic, United States of America
| | - Lilach O Lerman
- Division of Nephrology and Hypertension, Department of Medicine, Mayo Clinic, United States of America
| | - Kathleen M McDonald
- Cellular Senescence and Translation and Pharmacology Programs, Robert and Arlene Kogod Center on Aging, Mayo Clinic, United States of America; Office of Research Regulatory Support, Mayo Clinic, United States of America
| | | | - João F Passos
- Cellular Senescence and Translation and Pharmacology Programs, Robert and Arlene Kogod Center on Aging, Mayo Clinic, United States of America; Department of Physiology and Biomedical Engineering, Mayo Clinic, United States of America
| | - Robert J Pignolo
- Cellular Senescence and Translation and Pharmacology Programs, Robert and Arlene Kogod Center on Aging, Mayo Clinic, United States of America; Division of Geriatric Medicine and Gerontology, Department of Medicine, Mayo Clinic, United States of America; Division of Endocrinology, Department of Medicine, Mayo Clinic, United States of America; Division of Hospital Medicine, Department of Medicine, Mayo Clinic, United States of America; Department of Physiology, Mayo Clinic, United States of America
| | - Tamar Pirtskhalava
- Cellular Senescence and Translation and Pharmacology Programs, Robert and Arlene Kogod Center on Aging, Mayo Clinic, United States of America
| | - Ishran M Saadiq
- Division of Nephrology and Hypertension, Department of Medicine, Mayo Clinic, United States of America
| | - Kalli K Schaefer
- Cellular Senescence and Translation and Pharmacology Programs, Robert and Arlene Kogod Center on Aging, Mayo Clinic, United States of America
| | - Stephen C Textor
- Division of Nephrology and Hypertension, Department of Medicine, Mayo Clinic, United States of America
| | - Stella G Victorelli
- Cellular Senescence and Translation and Pharmacology Programs, Robert and Arlene Kogod Center on Aging, Mayo Clinic, United States of America; Department of Physiology and Biomedical Engineering, Mayo Clinic, United States of America
| | - Tammie L Volkman
- Cellular Senescence and Translation and Pharmacology Programs, Robert and Arlene Kogod Center on Aging, Mayo Clinic, United States of America; Department of Medicine Clinical Trials Unit, Department of Medicine, Mayo Clinic, United States of America
| | - Ailing Xue
- Cellular Senescence and Translation and Pharmacology Programs, Robert and Arlene Kogod Center on Aging, Mayo Clinic, United States of America
| | - Mark A Wentworth
- Cellular Senescence and Translation and Pharmacology Programs, Robert and Arlene Kogod Center on Aging, Mayo Clinic, United States of America; Office of Research Regulatory Support, Mayo Clinic, United States of America
| | - Erin O Wissler Gerdes
- Cellular Senescence and Translation and Pharmacology Programs, Robert and Arlene Kogod Center on Aging, Mayo Clinic, United States of America; Department of Medicine Clinical Trials Unit, Department of Medicine, Mayo Clinic, United States of America
| | - Yi Zhu
- Cellular Senescence and Translation and Pharmacology Programs, Robert and Arlene Kogod Center on Aging, Mayo Clinic, United States of America
| | - Tamara Tchkonia
- Cellular Senescence and Translation and Pharmacology Programs, Robert and Arlene Kogod Center on Aging, Mayo Clinic, United States of America.
| | - James L Kirkland
- Cellular Senescence and Translation and Pharmacology Programs, Robert and Arlene Kogod Center on Aging, Mayo Clinic, United States of America; Division of Geriatric Medicine and Gerontology, Department of Medicine, Mayo Clinic, United States of America; Division of Hospital Medicine, Department of Medicine, Mayo Clinic, United States of America; Division of General Internal Medicine, Department of Medicine, Mayo Clinic, United States of America.
| |
Collapse
|
602
|
Zarakowska E, Czerwinska J, Tupalska A, Yousefzadeh MJ, Gregg SQ, Croix CMS, Niedernhofer LJ, Foksinski M, Gackowski D, Szpila A, Starczak M, Tudek B, Olinski R. Oxidation Products of 5-Methylcytosine are Decreased in Senescent Cells and Tissues of Progeroid Mice. J Gerontol A Biol Sci Med Sci 2019; 73:1003-1009. [PMID: 29415265 DOI: 10.1093/gerona/gly012] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2017] [Accepted: 02/01/2018] [Indexed: 12/24/2022] Open
Abstract
5-Hydroxymethylcytosine and 5-formylcytosine are stable DNA base modifications generated from 5-methylcytosine by the ten-eleven translocation protein family that function as epigenetic markers. 5-Hydroxymethyluracil may also be generated from thymine by ten-eleven translocation enzymes. Here, we asked if these epigenetic changes accumulate in senescent cells, since they are thought to be inversely correlated with proliferation. Testing this in ERCC1-XPF-deficient cells and mice also enabled discovery if these DNA base changes are repaired by nucleotide excision repair. Epigenetic marks were measured in proliferating, quiescent and senescent wild-type (WT) and Ercc1-/- primary mouse embryonic fibroblasts. The pattern of epigenetic marks depended more on the proliferation status of the cells than their DNA repair capacity. The cytosine modifications were all decreased in senescent cells compared to quiescent or proliferating cells, whereas 5-(hydroxymethyl)-2'-deoxyuridine was increased. In vivo, both 5-(hydroxymethyl)-2'-deoxyuridine and 5-(hydroxymethyl)-2'-deoxycytidine were significantly increased in liver tissues of aged WT mice compared to young adult WT mice. Livers of Ercc1-deficient mice with premature senescence and aging had reduced level of 5-(hydroxymethyl)-2'-deoxycytidine and 5-formyl-2'-deoxycytidine compared to aged-matched WT controls. Taken together, we demonstrate for the first time, that 5-(hydroxymethyl)-2'-deoxycytidine is significantly reduced in senescent cells and tissue, potentially yielding a novel marker of senescence.
Collapse
Affiliation(s)
- Ewelina Zarakowska
- Department of Clinical Biochemistry, Faculty of Pharmacy, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Torun, Poland
| | - Jolanta Czerwinska
- Institute of Biochemistry and Biophysics, Polish Academy of Sciences, Warsaw, Poland
| | - Agnieszka Tupalska
- Institute of Genetics and Biotechnology, Faculty of Biology, University of Warsaw, Poland
| | - Matt J Yousefzadeh
- Department of Molecular Medicine, Center on Aging, The Scripps Research Institute, Jupiter, Florida
| | - Siobhán Q Gregg
- Department of Cell Biology, University of Pittsburgh, Pennsylvania
| | | | - Laura J Niedernhofer
- Department of Molecular Medicine, Center on Aging, The Scripps Research Institute, Jupiter, Florida
| | - Marek Foksinski
- Department of Clinical Biochemistry, Faculty of Pharmacy, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Torun, Poland
| | - Daniel Gackowski
- Department of Clinical Biochemistry, Faculty of Pharmacy, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Torun, Poland
| | - Anna Szpila
- Department of Clinical Biochemistry, Faculty of Pharmacy, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Torun, Poland
| | - Marta Starczak
- Department of Clinical Biochemistry, Faculty of Pharmacy, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Torun, Poland
| | - Barbara Tudek
- Institute of Biochemistry and Biophysics, Polish Academy of Sciences, Warsaw, Poland.,Institute of Genetics and Biotechnology, Faculty of Biology, University of Warsaw, Poland
| | - Ryszard Olinski
- Department of Clinical Biochemistry, Faculty of Pharmacy, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Torun, Poland
| |
Collapse
|
603
|
Feng M, Kim J, Field K, Reid C, Chatzistamou I, Shim M. Aspirin ameliorates the long-term adverse effects of doxorubicin through suppression of cellular senescence. FASEB Bioadv 2019; 1:579-590. [PMID: 32123852 PMCID: PMC6996307 DOI: 10.1096/fba.2019-00041] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2019] [Revised: 05/17/2019] [Accepted: 08/19/2019] [Indexed: 01/08/2023] Open
Abstract
A number of childhood cancer survivors develop adverse, late onset side effects of earlier cancer treatments, known as the late effects of cancer therapy. As the number of survivors continues to increase, this growing population is at increased risk for a number of health-related problems. In the present study, we have examined the effect of aspirin on the late effects of chemotherapy by treating juvenile mice with doxorubicin (DOX). This novel mouse model produced various long-term adverse effects, some of which resemble premature aging phenotypes. DOX also resulted in the tissue accumulation of senescent cells and up-regulation of cyclooxygenase-2 (COX2) expression. However, treatment with aspirin following juvenile exposure to DOX improved body weight gain, ameliorated the long-term adverse effects, and reduced the levels of senescence markers. Moreover, aspirin reduced p53 and p21 accumulation in DOX-treated human and mouse fibroblasts. However, the suppressive effect of aspirin on DOX-induced p53 accumulation was significantly decreased in COX2 knockout mouse embryonic fibroblasts. Additionally, treatment of senescent fibroblasts with aspirin or celecoxib, a COX2 specific inhibitor, reduced cell viability and decreased the levels of Bcl-xL protein. Taken together, these studies suggest that aspirin may be able to reduce the late effects of chemotherapy through the suppression of cellular senescence.
Collapse
Affiliation(s)
- Mingxiao Feng
- Department of Biological SciencesUniversity of South CarolinaColumbiaSCUSA
- Center for Colon Cancer ResearchUniversity of South CarolinaColumbiaSCUSA
| | - Joohwee Kim
- Department of Biological SciencesUniversity of South CarolinaColumbiaSCUSA
- Center for Colon Cancer ResearchUniversity of South CarolinaColumbiaSCUSA
| | - Kevin Field
- UNC School of MedicineUniversity of North Carolina at Chapel HillChapel HillNCUSA
| | - Christine Reid
- Department of Biological SciencesUniversity of South CarolinaColumbiaSCUSA
- Center for Colon Cancer ResearchUniversity of South CarolinaColumbiaSCUSA
| | - Ioulia Chatzistamou
- Department of Pathology, Microbiology & ImmunologySchool of MedicineUniversity of South CarolinaColumbiaSCUSA
| | - Minsub Shim
- Department of BiochemistryCollege of Graduate Studies and Arizona College of Osteopathic MedicineMidwestern UniversityGlendaleAZUSA
| |
Collapse
|
604
|
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
|
605
|
Sessions GA, Copp ME, Liu JY, Sinkler MA, D'Costa S, Diekman BO. Controlled induction and targeted elimination of p16 INK4a-expressing chondrocytes in cartilage explant culture. FASEB J 2019; 33:12364-12373. [PMID: 31408372 DOI: 10.1096/fj.201900815rr] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Cellular senescence is a phenotypic state that contributes to age-related diseases through the secretion of matrix-degrading and inflammatory molecules. An emerging therapeutic strategy for osteoarthritis (OA) is to selectively eliminate senescent cells by initiating apoptosis. This study establishes a cartilage explant model of senescence induction and senolytic clearance using p16Ink4a expression as a biomarker of senescence. Growth-factor stimulation of explants increased the expression of p16Ink4a at both the mRNA and protein levels. Applying this culture system to cartilage from p16tdTom reporter mice (a knockin allele with tdTomato fluorescent protein regulated by the endogenous p16Ink4a promoter) demonstrated the emergence of a p16-high population that was quantified using flow cytometry for tdTomato. Cell sorting was used to separate chondrocytes based on tdTomato fluorescence and p16-high cells showed higher senescence-associated β-galactosidase activity and increased gene expression of the senescence-associated secretory phenotype as compared with p16-low cells. The potential for effective senolysis within the cartilage extracellular matrix was assessed using navitoclax (ABT-263). Navitoclax treatment reduced the percentage of p16-high cells from 17.9 to 6.1% (mean of 13 matched pairs; P < 0.001) and increased cleaved caspase-3 confirmed apoptotic activity. Together, these findings establish a physiologically relevant cartilage explant model for testing the induction and elimination of senescent chondrocytes, which will support investigations of senolytic therapy for OA.-Sessions, G. A., Copp, M. E., Liu, J.-Y., Sinkler, M. A., D'Costa, S., Diekman, B. O. Controlled induction and targeted elimination of p16INK4a-expressing chondrocytes in cartilage explant culture.
Collapse
Affiliation(s)
- Garrett A Sessions
- Thurston Arthritis Research Center, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Michaela E Copp
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina-North Carolina State University, Raleigh, North Carolina, USA
| | - Jie-Yu Liu
- Lineberger Comprehensive Cancer Center, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Margaret A Sinkler
- Thurston Arthritis Research Center, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA.,Medical College of Georgia, Augusta University, Augusta, Georgia, USA
| | - Susan D'Costa
- Thurston Arthritis Research Center, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Brian O Diekman
- Thurston Arthritis Research Center, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA.,Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina-North Carolina State University, Raleigh, North Carolina, USA.,Lineberger Comprehensive Cancer Center, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| |
Collapse
|
606
|
Terlecki-Zaniewicz L, Lämmermann I, Latreille J, Bobbili MR, Pils V, Schosserer M, Weinmüllner R, Dellago H, Skalicky S, Pum D, Almaraz JCH, Scheideler M, Morizot F, Hackl M, Gruber F, Grillari J. Small extracellular vesicles and their miRNA cargo are anti-apoptotic members of the senescence-associated secretory phenotype. Aging (Albany NY) 2019; 10:1103-1132. [PMID: 29779019 PMCID: PMC5990398 DOI: 10.18632/aging.101452] [Citation(s) in RCA: 158] [Impact Index Per Article: 26.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2018] [Accepted: 05/10/2018] [Indexed: 12/15/2022]
Abstract
Loss of functionality during aging of cells and organisms is caused and accompanied by altered cell-to-cell communication and signalling. One factor thereby is the chronic accumulation of senescent cells and the concomitant senescence-associated secretory phenotype (SASP) that contributes to microenvironment remodelling and a pro-inflammatory status. While protein based SASP factors have been well characterized, little is known about small extracellular vesicles (sEVs) and their miRNA cargo. Therefore, we analysed secretion of sEVs from senescent human dermal fibroblasts and catalogued the therein contained miRNAs. We observed a four-fold increase of sEVs, with a concomitant increase of >80% of all cargo miRNAs. The most abundantly secreted miRNAs were predicted to collectively target mRNAs of pro-apoptotic proteins, and indeed, senescent cell derived sEVs exerted anti-apoptotic activity. In addition, we identified senescence-specific differences in miRNA composition of sEVs, with an increase of miR-23a-5p and miR-137 and a decrease of miR-625-3p, miR-766-3p, miR-199b-5p, miR-381-3p, miR-17-3p. By correlating intracellular and sEV-miRNAs, we identified miRNAs selectively retained in senescent cells (miR-21-3p and miR-17-3p) or packaged specifically into senescent cell derived sEVs (miR-15b-5p and miR-30a-3p). Therefore, we suggest sEVs and their miRNA cargo to be novel, members of the SASP that are selectively secreted or retained in cellular senescence.
Collapse
|
607
|
Faget DV, Ren Q, Stewart SA. Unmasking senescence: context-dependent effects of SASP in cancer. Nat Rev Cancer 2019; 19:439-453. [PMID: 31235879 DOI: 10.1038/s41568-019-0156-2] [Citation(s) in RCA: 540] [Impact Index Per Article: 90.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 05/15/2019] [Indexed: 01/10/2023]
Abstract
Cellular senescence plays a critical role in tumorigenesis. Once thought of as a tissue culture artefact by some researchers, senescence is now a major field of study. Although there are common molecular mechanisms that enforce the growth arrest that characterizes the phenotype, the impact of senescence is varied and can, in some instances, have opposite effects on tumorigenesis. It has become clearer that the cell of origin and the tissue in question dictate the impact of senescence on tumorigenesis. In this Review, we unravel this complexity by focusing on how senescence impacts tumorigenesis when it arises within incipient tumour cells versus stromal cells, and how these roles can change in different stages of disease progression. In addition, we highlight the diversity of the senescent phenotype and its functional output beyond growth arrest: the senescence-associated secretory phenotype (SASP). Fortunately, a number of new genetic and pharmacologic tools have been developed that are now allowing the senescence phenotype to be parsed further.
Collapse
Affiliation(s)
- Douglas V Faget
- Department of Cell Biology and Physiology, Washington University School of Medicine, St. Louis, MO, USA
| | - Qihao Ren
- Department of Cell Biology and Physiology, Washington University School of Medicine, St. Louis, MO, USA
| | - Sheila A Stewart
- Department of Cell Biology and Physiology, Washington University School of Medicine, St. Louis, MO, USA.
- Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA.
- ICCE Institute, Washington University School of Medicine, St. Louis, MO, USA.
- Siteman Cancer Center, Washington University School of Medicine, St. Louis, MO, USA.
| |
Collapse
|
608
|
Qian M, Liu B. Pharmaceutical Intervention of Aging. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1086:235-254. [PMID: 30232763 DOI: 10.1007/978-981-13-1117-8_15] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
The aging population represents a significant worldwide socioeconomic challenge. Aging is an inevitable and multifactorial biological process and primary risk factor for most age-related diseases, such as cardiovascular diseases, cancers, type 2 diabetes mellitus (T2DM), and neurodegenerative diseases. Pharmacological interventions targeting aging appear to be a more effective approach in preventing age-related disorders compared with the treatments targeted to specific disease. In this chapter, we focus on the latest findings on molecular compounds that mimic caloric restriction (CR), supplement nicotinamide adenine dinucleotide (NAD+) levels, and eliminate senescent cells, including metformin, resveratrol, spermidine, rapamycin, NAD+ boosters, as well as senolytics. All these interventions modulate the determinants and pathways responsible for aging/longevity, such as the kinase target of rapamycin (TOR), AMP-activated protein kinase (AMPK), sirtuins, and insulin-like growth factor (IGF-1) signaling (Fig. 15.1).
Collapse
Affiliation(s)
- Minxian Qian
- Guangdong Key Laboratory of Genome Stability and Human Disease Prevention, Medical Research Center, Department of Biochemistry and Molecular Biology, Shenzhen University Health Science Center, Shenzhen, China
| | - Baohua Liu
- Guangdong Key Laboratory of Genome Stability and Human Disease Prevention, Medical Research Center, Department of Biochemistry and Molecular Biology, Shenzhen University Health Science Center, Shenzhen, China.
| |
Collapse
|
609
|
Knoppert SN, Valentijn FA, Nguyen TQ, Goldschmeding R, Falke LL. Cellular Senescence and the Kidney: Potential Therapeutic Targets and Tools. Front Pharmacol 2019; 10:770. [PMID: 31354486 PMCID: PMC6639430 DOI: 10.3389/fphar.2019.00770] [Citation(s) in RCA: 63] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2019] [Accepted: 06/14/2019] [Indexed: 01/10/2023] Open
Abstract
Chronic kidney disease (CKD) is an increasing health burden (affecting approximately 13.4% of the population). Currently, no curative treatment options are available and treatment is focused on limiting the disease progression. The accumulation of senescent cells has been implicated in the development of kidney fibrosis by limiting tissue rejuvenation and through the secretion of pro-fibrotic and pro-inflammatory mediators termed as the senescence-associated secretory phenotype. The clearance of senescent cells in aging models results in improved kidney function, which shows promise for the options of targeting senescent cells in CKD. There are several approaches for the development of “senotherapies”, the most rigorous of which is the elimination of senescent cells by the so-called senolytic drugs either newly developed or repurposed for off-target effects in terms of selectively inducing apoptosis in senescent cells. Several chemotherapeutics and checkpoint inhibitors currently used in daily oncological practice show senolytic properties. However, the applicability of such senolytic compounds for the treatment of renal diseases has hardly been investigated. A serious concern is that systemic side effects will limit the use of senolytics for kidney fibrosis. Specifically targeting senescent cells and/or targeted drug delivery to the kidney might circumvent these side effects. In this review, we discuss the connection between CKD and senescence, the pharmacological options for targeting senescent cells, and the means to specifically target the kidney.
Collapse
Affiliation(s)
- Sebastian N Knoppert
- Department of Pathology, University Medical Center Utrecht, Utrecht, Netherlands
| | - Floris A Valentijn
- Department of Pathology, University Medical Center Utrecht, Utrecht, Netherlands
| | - Tri Q Nguyen
- Department of Pathology, University Medical Center Utrecht, Utrecht, Netherlands
| | - Roel Goldschmeding
- Department of Pathology, University Medical Center Utrecht, Utrecht, Netherlands
| | - Lucas L Falke
- Department of Pathology, University Medical Center Utrecht, Utrecht, Netherlands.,Department of Internal Medicine, Diakonessenhuis, University Medical Center Utrecht, Utrecht, Netherlands
| |
Collapse
|
610
|
Li W, Qin L, Feng R, Hu G, Sun H, He Y, Zhang R. Emerging senolytic agents derived from natural products. Mech Ageing Dev 2019; 181:1-6. [PMID: 31077707 DOI: 10.1016/j.mad.2019.05.001] [Citation(s) in RCA: 64] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2019] [Revised: 04/05/2019] [Accepted: 05/07/2019] [Indexed: 01/10/2023]
Abstract
Cellular senescence is a hallmark of aging, it is a permanent state of cell cycle arrest induced by cellular stresses. During the aging process, senescent cells (SCs) increasingly accumulate in tissues, causing a loss of tissue-repair capacity because of cell cycle arrest in progenitor cells and produce proinflammatory and matrix-degrading molecules which are known as the senescence-associated secretory phenotype (SASP), and thereby contribute to the development of various age-related diseases. Genetic evidence has demonstrated that clearance of SCs can delay aging and extend healthspan. Senolytics, small molecules that can selectively kill SCs, have been developed to treat various age-related diseases. In recent years, emerging natural compounds have been discovered to be effective senolytic agents, such as quercetin, fisetin, piperlongumine and the curcumin analog. Some of the compounds have been validated in animal models and have great potential to be pushed to clinical applications. In this review, we will discuss cellular senescence and its potential as a target for treating age-related diseases, and summarize the known natural compounds as senolytic agents and their applications.
Collapse
Affiliation(s)
- Wen Li
- School of Pharmaceutical Science and Yunnan Key Laboratory of Pharmacology for Natural Products, Kunming Medical University, Kunming, Yunnan 650500, China; Department of Endocrinology, The Third People's Hospital of Yunnan Province, Kunming, Yunnan 650011, China
| | - Lin Qin
- School of Pharmaceutical Science and Yunnan Key Laboratory of Pharmacology for Natural Products, Kunming Medical University, Kunming, Yunnan 650500, China; Department of Endocrinology, The First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan 650000, China
| | - Rennan Feng
- Department of Nutrition and Food Hygiene, School of Public Health, Harbin Medical University, Harbin, Heilongjiang 150081, China
| | - Guangrong Hu
- Department of Emergency, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150081, China
| | - Hui Sun
- Department of Emergency, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150081, China
| | - Yonghan He
- State Key Laboratory of Genetic Resources and Evolution, Kunming Institute of Zoology, The Chinese Academy of Sciences, Kunming 650223, China.
| | - Rongping Zhang
- School of Pharmaceutical Science and Yunnan Key Laboratory of Pharmacology for Natural Products, Kunming Medical University, Kunming, Yunnan 650500, China.
| |
Collapse
|
611
|
Soto-Gamez A, Quax WJ, Demaria M. Regulation of Survival Networks in Senescent Cells: From Mechanisms to Interventions. J Mol Biol 2019; 431:2629-2643. [DOI: 10.1016/j.jmb.2019.05.036] [Citation(s) in RCA: 71] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2019] [Revised: 05/20/2019] [Accepted: 05/23/2019] [Indexed: 01/10/2023]
|
612
|
Guo CJ, Xie JJ, Hong RH, Pan HS, Zhang FG, Liang YM. Puerarin alleviates streptozotocin (STZ)-induced osteoporosis in rats through suppressing inflammation and apoptosis via HDAC1/HDAC3 signaling. Biomed Pharmacother 2019; 115:108570. [DOI: 10.1016/j.biopha.2019.01.031] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2018] [Revised: 01/06/2019] [Accepted: 01/08/2019] [Indexed: 11/15/2022] Open
|
613
|
Chapman J, Fielder E, Passos JF. Mitochondrial dysfunction and cell senescence: deciphering a complex relationship. FEBS Lett 2019; 593:1566-1579. [PMID: 31211858 DOI: 10.1002/1873-3468.13498] [Citation(s) in RCA: 207] [Impact Index Per Article: 34.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2019] [Revised: 06/11/2019] [Accepted: 06/13/2019] [Indexed: 12/31/2022]
Abstract
Cellular senescence and mitochondrial dysfunction have both been defined as classical hallmarks of the ageing process. Here, we review the intricate relationship between the two. In the context of ageing, it is now well regarded that cellular senescence is a key driver in both ageing and the onset of a number of age-related pathologies. Emerging evidence has pinpointed mitochondria as one of the key modulators in the development of the senescence phenotype, particularly the pro-inflammatory senescence associated secretory phenotype (SASP). This review focuses on the contribution of homeostatic mechanisms, as well as of reactive oxygen species and mitochondrial metabolites in the senescence programme. Furthermore, we discuss emerging pathways and mitochondrial-mediated mechanisms that may be influencing the SASP and, subsequently, explore how these may be exploited to open up new therapeutic avenues.
Collapse
Affiliation(s)
- James Chapman
- Institute for Cell and Molecular Biosciences, Newcastle University Institute for Ageing, Newcastle University, Newcastle upon Tyne, UK
| | - Edward Fielder
- Institute for Cell and Molecular Biosciences, Newcastle University Institute for Ageing, Newcastle University, Newcastle upon Tyne, UK
| | - João F Passos
- Institute for Cell and Molecular Biosciences, Newcastle University Institute for Ageing, Newcastle University, Newcastle upon Tyne, UK.,Department of Physiology and Biochemical Engineering, Mayo Clinic, Rochester, NY, USA
| |
Collapse
|
614
|
Masaldan S, Belaidi AA, Ayton S, Bush AI. Cellular Senescence and Iron Dyshomeostasis in Alzheimer's Disease. Pharmaceuticals (Basel) 2019; 12:E93. [PMID: 31248150 PMCID: PMC6630536 DOI: 10.3390/ph12020093] [Citation(s) in RCA: 81] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2019] [Revised: 06/14/2019] [Accepted: 06/17/2019] [Indexed: 02/07/2023] Open
Abstract
Iron dyshomeostasis is a feature of Alzheimer's disease (AD). The impact of iron on AD is attributed to its interactions with the central proteins of AD pathology (amyloid precursor protein and tau) and/or through the iron-mediated generation of prooxidant molecules (e.g., hydroxyl radicals). However, the source of iron accumulation in pathologically relevant regions of the brain and its contribution to AD remains unclear. One likely contributor to iron accumulation is the age-associated increase in tissue-resident senescent cells that drive inflammation and contribute to various pathologies associated with advanced age. Iron accumulation predisposes ageing tissue to oxidative stress that can lead to cellular dysfunction and to iron-dependent cell death modalities (e.g., ferroptosis). Further, elevated brain iron is associated with the progression of AD and cognitive decline. Elevated brain iron presents a feature of AD that may be modified pharmacologically to mitigate the effects of age/senescence-associated iron dyshomeostasis and improve disease outcome.
Collapse
Affiliation(s)
- Shashank Masaldan
- Melbourne Dementia Research Centre, The Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, VIC 3052, Australia.
| | - Abdel Ali Belaidi
- Melbourne Dementia Research Centre, The Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, VIC 3052, Australia.
| | - Scott Ayton
- Melbourne Dementia Research Centre, The Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, VIC 3052, Australia.
| | - Ashley I Bush
- Melbourne Dementia Research Centre, The Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, VIC 3052, Australia.
| |
Collapse
|
615
|
Dai L, Qureshi AR, Witasp A, Lindholm B, Stenvinkel P. Early Vascular Ageing and Cellular Senescence in Chronic Kidney Disease. Comput Struct Biotechnol J 2019; 17:721-729. [PMID: 31303976 PMCID: PMC6603301 DOI: 10.1016/j.csbj.2019.06.015] [Citation(s) in RCA: 58] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2019] [Revised: 06/08/2019] [Accepted: 06/11/2019] [Indexed: 01/08/2023] Open
Abstract
Chronic kidney disease (CKD) is a clinical model of premature ageing characterized by progressive vascular disease, systemic inflammation, muscle wasting and frailty. The predominant early vascular ageing (EVA) process mediated by medial vascular calcification (VC) results in a marked discrepancy between chronological and biological vascular age in CKD. Though the exact underlying mechanisms of VC and EVA are not fully elucidated, accumulating evidence indicates that cellular senescence - and subsequent chronic inflammation through the senescence-associated secretary phenotype (SASP) - plays a fundamental role in its initiation and progression. In this review, we discuss the pathophysiological links between senescence and the EVA process in CKD, with focus on cellular senescence and media VC, and potential anti-ageing therapeutic strategies of senolytic drugs targeting cellular senescence and EVA in CKD.
Collapse
Affiliation(s)
| | | | | | | | - Peter Stenvinkel
- Division of Renal Medicine and Baxter Novum, Department of Clinical Science, Intervention and Technology, Karolinska Institutet, Campus Flemingsberg, Stockholm, Sweden
| |
Collapse
|
616
|
Smigiel JM, Taylor SE, Bryson BL, Tamagno I, Polak K, Jackson MW. Cellular plasticity and metastasis in breast cancer: a pre- and post-malignant problem. JOURNAL OF CANCER METASTASIS AND TREATMENT 2019; 5:47. [PMID: 32355893 PMCID: PMC7192216 DOI: 10.20517/2394-4722.2019.26] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
As a field we have made tremendous strides in treating breast cancer, with a decline in the past 30 years of overall breast cancer mortality. However, this progress is met with little affect once the disease spreads beyond the primary site. With a 5-year survival rate of 22%, 10-year of 13%, for those patients with metastatic breast cancer (mBC), our ability to effectively treat wide spread disease is minimal. A major contributing factor to this ineffectiveness is the complex make-up, or heterogeneity, of the primary site. Within a primary tumor, secreted factors, malignant and pre-malignant epithelial cells, immune cells, stromal fibroblasts and many others all reside alongside each other creating a dynamic environment contributing to metastasis. Furthermore, heterogeneity contributes to our lack of understanding regarding the cells' remarkable ability to undergo epithelial/non-cancer stem cell (CSC) to mesenchymal/CSC (E-M/CSC) plasticity. The enhanced invasion & motility, tumor-initiating potential, and acquired therapeutic resistance which accompanies E-M/CSC plasticity implicates a significant role in metastasis. While most work trying to understand E-M/CSC plasticity has been done on malignant cells, recent evidence is emerging concerning the ability for pre-malignant cells to undergo E-M/CSC plasticity and contribute to the metastatic process. Here we will discuss the importance of E-M/CSC plasticity within malignant and pre-malignant populations of the tumor. Moreover, we will discuss how one may potentially target these populations, ultimately disrupting the metastatic cascade and increasing patient survival for those with mBC.
Collapse
Affiliation(s)
- Jacob M. Smigiel
- Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA
| | - Sarah E. Taylor
- Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA
| | - Benjamin L. Bryson
- Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA
| | - Ilaria Tamagno
- Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA
| | - Kelsey Polak
- Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA
| | - Mark W. Jackson
- Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA
- Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, OH 44106, USA
| |
Collapse
|
617
|
Fleury H, Malaquin N, Tu V, Gilbert S, Martinez A, Olivier MA, Sauriol A, Communal L, Leclerc-Desaulniers K, Carmona E, Provencher D, Mes-Masson AM, Rodier F. Exploiting interconnected synthetic lethal interactions between PARP inhibition and cancer cell reversible senescence. Nat Commun 2019. [PMID: 31186408 DOI: 10.1038/s41467-019-10460-1] [] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
Senescence is a tumor suppression mechanism defined by stable proliferation arrest. Here we demonstrate that the known synthetic lethal interaction between poly(ADP-ribose) polymerase 1 inhibitors (PARPi) and DNA repair triggers p53-independent ovarian cancer cell senescence defined by senescence-associated phenotypic hallmarks including DNA-SCARS, inflammatory secretome, Bcl-XL-mediated apoptosis resistance, and proliferation restriction via Chk2 and p21 (CDKN1A). The concept of senescence as irreversible remains controversial and here we show that PARPi-senescent cells re-initiate proliferation upon drug withdrawal, potentially explaining the requirement for sustained PARPi therapy in the clinic. Importantly, PARPi-induced senescence renders ovarian and breast cancer cells transiently susceptible to second-phase synthetic lethal approaches targeting the senescence state using senolytic drugs. The combination of PARPi and a senolytic is effective in preclinical models of ovarian and breast cancer suggesting that coupling these synthetic lethalities provides a rational approach to their clinical use and may together be more effective in limiting resistance.
Collapse
Affiliation(s)
- Hubert Fleury
- Centre de recherche du Centre hospitalier de l'Université de Montréal (CRCHUM), Montreal, H2X 0A9, QC, Canada.,Institut du cancer de Montréal, Montreal, H2X 0A9, QC, Canada
| | - Nicolas Malaquin
- Centre de recherche du Centre hospitalier de l'Université de Montréal (CRCHUM), Montreal, H2X 0A9, QC, Canada.,Institut du cancer de Montréal, Montreal, H2X 0A9, QC, Canada
| | - Véronique Tu
- Centre de recherche du Centre hospitalier de l'Université de Montréal (CRCHUM), Montreal, H2X 0A9, QC, Canada.,Institut du cancer de Montréal, Montreal, H2X 0A9, QC, Canada
| | - Sophie Gilbert
- Centre de recherche du Centre hospitalier de l'Université de Montréal (CRCHUM), Montreal, H2X 0A9, QC, Canada.,Institut du cancer de Montréal, Montreal, H2X 0A9, QC, Canada
| | - Aurélie Martinez
- Centre de recherche du Centre hospitalier de l'Université de Montréal (CRCHUM), Montreal, H2X 0A9, QC, Canada.,Institut du cancer de Montréal, Montreal, H2X 0A9, QC, Canada
| | - Marc-Alexandre Olivier
- Centre de recherche du Centre hospitalier de l'Université de Montréal (CRCHUM), Montreal, H2X 0A9, QC, Canada.,Institut du cancer de Montréal, Montreal, H2X 0A9, QC, Canada
| | - Alexandre Sauriol
- Centre de recherche du Centre hospitalier de l'Université de Montréal (CRCHUM), Montreal, H2X 0A9, QC, Canada.,Institut du cancer de Montréal, Montreal, H2X 0A9, QC, Canada
| | - Laudine Communal
- Centre de recherche du Centre hospitalier de l'Université de Montréal (CRCHUM), Montreal, H2X 0A9, QC, Canada.,Institut du cancer de Montréal, Montreal, H2X 0A9, QC, Canada
| | - Kim Leclerc-Desaulniers
- Centre de recherche du Centre hospitalier de l'Université de Montréal (CRCHUM), Montreal, H2X 0A9, QC, Canada.,Institut du cancer de Montréal, Montreal, H2X 0A9, QC, Canada
| | - Euridice Carmona
- Centre de recherche du Centre hospitalier de l'Université de Montréal (CRCHUM), Montreal, H2X 0A9, QC, Canada.,Institut du cancer de Montréal, Montreal, H2X 0A9, QC, Canada
| | - Diane Provencher
- Centre de recherche du Centre hospitalier de l'Université de Montréal (CRCHUM), Montreal, H2X 0A9, QC, Canada.,Institut du cancer de Montréal, Montreal, H2X 0A9, QC, Canada.,Division of Gynecologic Oncology, Université de Montréal, Montreal, H3C 3J7, QC, Canada
| | - Anne-Marie Mes-Masson
- Centre de recherche du Centre hospitalier de l'Université de Montréal (CRCHUM), Montreal, H2X 0A9, QC, Canada. .,Institut du cancer de Montréal, Montreal, H2X 0A9, QC, Canada. .,Department of Medicine, Université de Montréal, Montreal, H3C 3J7, QC, Canada.
| | - Francis Rodier
- Centre de recherche du Centre hospitalier de l'Université de Montréal (CRCHUM), Montreal, H2X 0A9, QC, Canada. .,Institut du cancer de Montréal, Montreal, H2X 0A9, QC, Canada. .,Department of Radiology, Radio-Oncology and Nuclear Medicine, Université de Montréal, Montreal, H3C 3J7, QC, Canada.
| |
Collapse
|
618
|
Fleury H, Malaquin N, Tu V, Gilbert S, Martinez A, Olivier MA, Sauriol SA, Communal L, Leclerc-Desaulniers K, Carmona E, Provencher D, Mes-Masson AM, Rodier F. Exploiting interconnected synthetic lethal interactions between PARP inhibition and cancer cell reversible senescence. Nat Commun 2019; 10:2556. [PMID: 31186408 PMCID: PMC6560032 DOI: 10.1038/s41467-019-10460-1] [Citation(s) in RCA: 155] [Impact Index Per Article: 25.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2018] [Accepted: 05/09/2019] [Indexed: 12/19/2022] Open
Abstract
Senescence is a tumor suppression mechanism defined by stable proliferation arrest. Here we demonstrate that the known synthetic lethal interaction between poly(ADP-ribose) polymerase 1 inhibitors (PARPi) and DNA repair triggers p53-independent ovarian cancer cell senescence defined by senescence-associated phenotypic hallmarks including DNA-SCARS, inflammatory secretome, Bcl-XL-mediated apoptosis resistance, and proliferation restriction via Chk2 and p21 (CDKN1A). The concept of senescence as irreversible remains controversial and here we show that PARPi-senescent cells re-initiate proliferation upon drug withdrawal, potentially explaining the requirement for sustained PARPi therapy in the clinic. Importantly, PARPi-induced senescence renders ovarian and breast cancer cells transiently susceptible to second-phase synthetic lethal approaches targeting the senescence state using senolytic drugs. The combination of PARPi and a senolytic is effective in preclinical models of ovarian and breast cancer suggesting that coupling these synthetic lethalities provides a rational approach to their clinical use and may together be more effective in limiting resistance.
Collapse
Affiliation(s)
- Hubert Fleury
- Centre de recherche du Centre hospitalier de l'Université de Montréal (CRCHUM), Montreal, H2X 0A9, QC, Canada
- Institut du cancer de Montréal, Montreal, H2X 0A9, QC, Canada
| | - Nicolas Malaquin
- Centre de recherche du Centre hospitalier de l'Université de Montréal (CRCHUM), Montreal, H2X 0A9, QC, Canada
- Institut du cancer de Montréal, Montreal, H2X 0A9, QC, Canada
| | - Véronique Tu
- Centre de recherche du Centre hospitalier de l'Université de Montréal (CRCHUM), Montreal, H2X 0A9, QC, Canada
- Institut du cancer de Montréal, Montreal, H2X 0A9, QC, Canada
| | - Sophie Gilbert
- Centre de recherche du Centre hospitalier de l'Université de Montréal (CRCHUM), Montreal, H2X 0A9, QC, Canada
- Institut du cancer de Montréal, Montreal, H2X 0A9, QC, Canada
| | - Aurélie Martinez
- Centre de recherche du Centre hospitalier de l'Université de Montréal (CRCHUM), Montreal, H2X 0A9, QC, Canada
- Institut du cancer de Montréal, Montreal, H2X 0A9, QC, Canada
| | - Marc-Alexandre Olivier
- Centre de recherche du Centre hospitalier de l'Université de Montréal (CRCHUM), Montreal, H2X 0A9, QC, Canada
- Institut du cancer de Montréal, Montreal, H2X 0A9, QC, Canada
| | - Skye Alexandre Sauriol
- Centre de recherche du Centre hospitalier de l'Université de Montréal (CRCHUM), Montreal, H2X 0A9, QC, Canada
- Institut du cancer de Montréal, Montreal, H2X 0A9, QC, Canada
| | - Laudine Communal
- Centre de recherche du Centre hospitalier de l'Université de Montréal (CRCHUM), Montreal, H2X 0A9, QC, Canada
- Institut du cancer de Montréal, Montreal, H2X 0A9, QC, Canada
| | - Kim Leclerc-Desaulniers
- Centre de recherche du Centre hospitalier de l'Université de Montréal (CRCHUM), Montreal, H2X 0A9, QC, Canada
- Institut du cancer de Montréal, Montreal, H2X 0A9, QC, Canada
| | - Euridice Carmona
- Centre de recherche du Centre hospitalier de l'Université de Montréal (CRCHUM), Montreal, H2X 0A9, QC, Canada
- Institut du cancer de Montréal, Montreal, H2X 0A9, QC, Canada
| | - Diane Provencher
- Centre de recherche du Centre hospitalier de l'Université de Montréal (CRCHUM), Montreal, H2X 0A9, QC, Canada
- Institut du cancer de Montréal, Montreal, H2X 0A9, QC, Canada
- Division of Gynecologic Oncology, Université de Montréal, Montreal, H3C 3J7, QC, Canada
| | - Anne-Marie Mes-Masson
- Centre de recherche du Centre hospitalier de l'Université de Montréal (CRCHUM), Montreal, H2X 0A9, QC, Canada.
- Institut du cancer de Montréal, Montreal, H2X 0A9, QC, Canada.
- Department of Medicine, Université de Montréal, Montreal, H3C 3J7, QC, Canada.
| | - Francis Rodier
- Centre de recherche du Centre hospitalier de l'Université de Montréal (CRCHUM), Montreal, H2X 0A9, QC, Canada.
- Institut du cancer de Montréal, Montreal, H2X 0A9, QC, Canada.
- Department of Radiology, Radio-Oncology and Nuclear Medicine, Université de Montréal, Montreal, H3C 3J7, QC, Canada.
| |
Collapse
|
619
|
Exploiting interconnected synthetic lethal interactions between PARP inhibition and cancer cell reversible senescence. Nat Commun 2019. [PMID: 31186408 DOI: 10.1038/s41467-019-10460-1]+[] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
Abstract
Senescence is a tumor suppression mechanism defined by stable proliferation arrest. Here we demonstrate that the known synthetic lethal interaction between poly(ADP-ribose) polymerase 1 inhibitors (PARPi) and DNA repair triggers p53-independent ovarian cancer cell senescence defined by senescence-associated phenotypic hallmarks including DNA-SCARS, inflammatory secretome, Bcl-XL-mediated apoptosis resistance, and proliferation restriction via Chk2 and p21 (CDKN1A). The concept of senescence as irreversible remains controversial and here we show that PARPi-senescent cells re-initiate proliferation upon drug withdrawal, potentially explaining the requirement for sustained PARPi therapy in the clinic. Importantly, PARPi-induced senescence renders ovarian and breast cancer cells transiently susceptible to second-phase synthetic lethal approaches targeting the senescence state using senolytic drugs. The combination of PARPi and a senolytic is effective in preclinical models of ovarian and breast cancer suggesting that coupling these synthetic lethalities provides a rational approach to their clinical use and may together be more effective in limiting resistance.
Collapse
|
620
|
Lee YH, Chen YY, Yeh YL, Wang YJ, Chen RJ. Stilbene Compounds Inhibit Tumor Growth by the Induction of Cellular Senescence and the Inhibition of Telomerase Activity. Int J Mol Sci 2019; 20:ijms20112716. [PMID: 31159515 PMCID: PMC6600253 DOI: 10.3390/ijms20112716] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2019] [Revised: 05/30/2019] [Accepted: 05/31/2019] [Indexed: 12/17/2022] Open
Abstract
Cellular senescence is a state of cell cycle arrest characterized by a distinct morphology, gene expression pattern, and secretory phenotype. It can be triggered by multiple mechanisms, including those involved in telomere shortening, the accumulation of DNA damage, epigenetic pathways, and the senescence-associated secretory phenotype (SASP), and so on. In current cancer therapy, cellular senescence has emerged as a potent tumor suppression mechanism that restrains proliferation in cells at risk for malignant transformation. Therefore, compounds that stimulate the growth inhibition effects of senescence while limiting its detrimental effects are believed to have great clinical potential. In this review article, we first review the current knowledge of the pro- and antitumorigeneic functions of senescence and summarize the key roles of telomerase in the regulation of senescence in tumors. Second, we review the current literature regarding the anticancer effects of stilbene compounds that are mediated by the targeting of telomerase and cell senescence. Finally, we provide future perspectives on the clinical utilization of stilbene compounds, especially resveratrol and pterostilbene, as novel cancer therapeutic remedies. We conclude and propose that stilbene compounds may induce senescence and may potentially be used as the therapeutic or adjuvant agents for cancers with high telomerase activity.
Collapse
Affiliation(s)
- Yu-Hsuan Lee
- Department of Food Safety/Hygiene and Risk Management, College of Medicine, National Cheng Kung University, Tainan 70428, Taiwan.
| | - Yu-Ying Chen
- Department of Environmental and Occupational Health, College of Medicine, National Cheng Kung University, Tainan 70428, Taiwan.
| | - Ya-Ling Yeh
- Department of Environmental and Occupational Health, College of Medicine, National Cheng Kung University, Tainan 70428, Taiwan.
| | - Ying-Jan Wang
- Department of Environmental and Occupational Health, College of Medicine, National Cheng Kung University, Tainan 70428, Taiwan.
- Department of Medical Research, China Medical University Hospital, China Medical University, Taichung 40402, Taiwan.
| | - Rong-Jane Chen
- Department of Food Safety/Hygiene and Risk Management, College of Medicine, National Cheng Kung University, Tainan 70428, Taiwan.
| |
Collapse
|
621
|
Pagnotti GM, Styner M, Uzer G, Patel VS, Wright LE, Ness KK, Guise TA, Rubin J, Rubin CT. Combating osteoporosis and obesity with exercise: leveraging cell mechanosensitivity. Nat Rev Endocrinol 2019; 15:339-355. [PMID: 30814687 PMCID: PMC6520125 DOI: 10.1038/s41574-019-0170-1] [Citation(s) in RCA: 151] [Impact Index Per Article: 25.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Osteoporosis, a condition of skeletal decline that undermines quality of life, is treated with pharmacological interventions that are associated with poor adherence and adverse effects. Complicating efforts to improve clinical outcomes, the incidence of obesity is increasing, predisposing the population to a range of musculoskeletal complications and metabolic disorders. Pharmacological management of obesity has yet to deliver notable reductions in weight and debilitating complications are rarely avoided. By contrast, exercise shows promise as a non-invasive and non-pharmacological method of regulating both osteoporosis and obesity. The principal components of exercise - mechanical signals - promote bone and muscle anabolism while limiting formation and expansion of fat mass. Mechanical regulation of bone and marrow fat might be achieved by regulating functions of differentiated cells in the skeletal tissue while biasing lineage selection of their common progenitors - mesenchymal stem cells. An inverse relationship between adipocyte versus osteoblast fate selection from stem cells is implicated in clinical conditions such as childhood obesity and increased marrow adiposity in type 2 diabetes mellitus, as well as contributing to skeletal frailty. Understanding how exercise-induced mechanical signals can be used to improve bone quality while decreasing fat mass and metabolic dysfunction should lead to new strategies to treat chronic diseases such as osteoporosis and obesity.
Collapse
Affiliation(s)
- Gabriel M Pagnotti
- School of Medicine, Division of Endocrinology, Indiana University, Indianapolis, IN, USA
| | - Maya Styner
- Department of Medicine, Division of Endocrinology and Metabolism, University of North Carolina, Chapel Hill, NC, USA
| | - Gunes Uzer
- College of Mechanical and Biomedical Engineering, Boise State University, Boise, ID, USA
| | - Vihitaben S Patel
- Department of Biomedical Engineering, Stony Brook University, Stony Brook, NY, USA
| | - Laura E Wright
- School of Medicine, Division of Endocrinology, Indiana University, Indianapolis, IN, USA
| | - Kirsten K Ness
- Department of Epidemiology and Cancer Control, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Theresa A Guise
- School of Medicine, Division of Endocrinology, Indiana University, Indianapolis, IN, USA
| | - Janet Rubin
- Department of Medicine, Division of Endocrinology and Metabolism, University of North Carolina, Chapel Hill, NC, USA
| | - Clinton T Rubin
- Department of Biomedical Engineering, Stony Brook University, Stony Brook, NY, USA.
| |
Collapse
|
622
|
Palmer AK, Xu M, Zhu Y, Pirtskhalava T, Weivoda MM, Hachfeld CM, Prata LG, van Dijk TH, Verkade E, Casaclang‐Verzosa G, Johnson KO, Cubro H, Doornebal EJ, Ogrodnik M, Jurk D, Jensen MD, Chini EN, Miller JD, Matveyenko A, Stout MB, Schafer MJ, White TA, Hickson LJ, Demaria M, Garovic V, Grande J, Arriaga EA, Kuipers F, von Zglinicki T, LeBrasseur NK, Campisi J, Tchkonia T, Kirkland JL. Targeting senescent cells alleviates obesity-induced metabolic dysfunction. Aging Cell 2019; 18:e12950. [PMID: 30907060 PMCID: PMC6516193 DOI: 10.1111/acel.12950] [Citation(s) in RCA: 434] [Impact Index Per Article: 72.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2019] [Revised: 02/16/2019] [Accepted: 03/03/2019] [Indexed: 12/18/2022] Open
Abstract
Adipose tissue inflammation and dysfunction are associated with obesity-related insulin resistance and diabetes, but mechanisms underlying this relationship are unclear. Although senescent cells accumulate in adipose tissue of obese humans and rodents, a direct pathogenic role for these cells in the development of diabetes remains to be demonstrated. Here, we show that reducing senescent cell burden in obese mice, either by activating drug-inducible "suicide" genes driven by the p16Ink4a promoter or by treatment with senolytic agents, alleviates metabolic and adipose tissue dysfunction. These senolytic interventions improved glucose tolerance, enhanced insulin sensitivity, lowered circulating inflammatory mediators, and promoted adipogenesis in obese mice. Elimination of senescent cells also prevented the migration of transplanted monocytes into intra-abdominal adipose tissue and reduced the number of macrophages in this tissue. In addition, microalbuminuria, renal podocyte function, and cardiac diastolic function improved with senolytic therapy. Our results implicate cellular senescence as a causal factor in obesity-related inflammation and metabolic derangements and show that emerging senolytic agents hold promise for treating obesity-related metabolic dysfunction and its complications.
Collapse
|
623
|
Thompson PJ, Shah A, Ntranos V, Van Gool F, Atkinson M, Bhushan A. Targeted Elimination of Senescent Beta Cells Prevents Type 1 Diabetes. Cell Metab 2019; 29:1045-1060.e10. [PMID: 30799288 DOI: 10.1016/j.cmet.2019.01.021] [Citation(s) in RCA: 234] [Impact Index Per Article: 39.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/25/2018] [Revised: 07/08/2018] [Accepted: 01/23/2019] [Indexed: 12/25/2022]
Abstract
Type 1 diabetes (T1D) is an organ-specific autoimmune disease characterized by hyperglycemia due to progressive loss of pancreatic beta cells. Immune-mediated beta cell destruction drives the disease, but whether beta cells actively participate in the pathogenesis remains unclear. Here, we show that during the natural history of T1D in humans and the non-obese diabetic (NOD) mouse model, a subset of beta cells acquires a senescence-associated secretory phenotype (SASP). Senescent beta cells upregulated pro-survival mediator Bcl-2, and treatment of NOD mice with Bcl-2 inhibitors selectively eliminated these cells without altering the abundance of the immune cell types involved in the disease. Significantly, elimination of senescent beta cells halted immune-mediated beta cell destruction and was sufficient to prevent diabetes. Our findings demonstrate that beta cell senescence is a significant component of the pathogenesis of T1D and indicate that clearance of senescent beta cells could be a new therapeutic approach for T1D.
Collapse
Affiliation(s)
- Peter J Thompson
- Diabetes Center, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Ajit Shah
- Diabetes Center, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Vasilis Ntranos
- Department of Electrical Engineering and Computer Sciences, University of California, Berkeley, Berkeley, CA 94720, USA; Department of Electrical Engineering, Stanford University, Stanford, CA 94305, USA
| | - Frederic Van Gool
- Diabetes Center, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Mark Atkinson
- Diabetes Institute, University of Florida, Gainesville, FL 32610-0296, USA
| | - Anil Bhushan
- Diabetes Center, University of California, San Francisco, San Francisco, CA 94143, USA.
| |
Collapse
|
624
|
Abstract
Cellular senescence (CS) is one of hallmarks of aging and accumulation of senescent cells (SCs) with age contributes to tissue or organismal aging, as well as the pathophysiologies of diverse age-related diseases (ARDs). Genetic ablation of SCs in tissues lengthened health span and reduced the risk of age-related pathologies in a mouse model, suggesting a direct link between SCs, longevity, and ARDs. Therefore, senotherapeutics, medicines targeting SCs, might be an emerging strategy for the extension of health span, and prevention or treatment of ARDs. Senotherapeutics are classified as senolytics which kills SCs selectively; senomorphics which modulate functions and morphology of SCs to those of young cells, or delays the progression of young cells to SCs in tissues; and immune-system mediators of the clearance of SCs. Some senolytics and senomorphics have been proven to markedly prevent or treat ARDs in animal models. This review will present the current status of the development of senotherapeutics, in relation to aging itself and ARDs. Finally, future directions and opportunities for senotherapeutics use will discussed. This knowledge will provide information that can be used to develop novel senotherapeutics for health span and ARDs. [BMB Reports 2019; 52(1): 47-55].
Collapse
Affiliation(s)
- Eok-Cheon Kim
- Department of Biochemistry and Molecular Biology, Smart-aging Convergence Research Center, College of Medicine, Yeungnam University, Daegu 42415, Korea
| | - Jae-Ryong Kim
- Department of Biochemistry and Molecular Biology, Smart-aging Convergence Research Center, College of Medicine, Yeungnam University, Daegu 42415, Korea
| |
Collapse
|
625
|
Parikh P, Wicher S, Khandalavala K, Pabelick CM, Britt RD, Prakash YS. Cellular senescence in the lung across the age spectrum. Am J Physiol Lung Cell Mol Physiol 2019; 316:L826-L842. [PMID: 30785345 PMCID: PMC6589594 DOI: 10.1152/ajplung.00424.2018] [Citation(s) in RCA: 76] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2018] [Revised: 02/14/2019] [Accepted: 02/14/2019] [Indexed: 12/18/2022] Open
Abstract
Cellular senescence results in cell cycle arrest with secretion of cytokines, chemokines, growth factors, and remodeling proteins (senescence-associated secretory phenotype; SASP) that have autocrine and paracrine effects on the tissue microenvironment. SASP can promote remodeling, inflammation, infectious susceptibility, angiogenesis, and proliferation, while hindering tissue repair and regeneration. While the role of senescence and the contributions of senescent cells are increasingly recognized in the context of aging and a variety of disease states, relatively less is known regarding the portfolio and influences of senescent cells in normal lung growth and aging per se or in the induction or progression of lung diseases across the age spectrum such as bronchopulmonary dysplasia, asthma, chronic obstructive pulmonary disease, or pulmonary fibrosis. In this review, we introduce concepts of cellular senescence, the mechanisms involved in the induction of senescence, and the SASP portfolio that are relevant to lung cells, presenting the potential contribution of senescent cells and SASP to inflammation, hypercontractility, and remodeling/fibrosis: aspects critical to a range of lung diseases. The potential to blunt lung disease by targeting senescent cells using a novel class of drugs (senolytics) is discussed. Potential areas for future research on cellular senescence in the lung are identified.
Collapse
Affiliation(s)
- Pavan Parikh
- Department of Obstetrics and Gynecology, Division of Maternal Fetal Medicine, Mayo Clinic, Rochester, Minnesota
| | - Sarah Wicher
- Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, Rochester, Minnesota
| | - Karl Khandalavala
- Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, Rochester, Minnesota
| | - Christina M. Pabelick
- Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, Rochester, Minnesota
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota
| | - Rodney D. Britt
- Center for Perinatal Research, The Research Institute at Nationwide Children’s Hospital, Columbus, Ohio
- Department of Pediatrics, The Ohio State University, Columbus, Ohio
| | - Y. S. Prakash
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota
| |
Collapse
|
626
|
Abdelfatah N, Chen R, Duff HJ, Seifer CM, Buffo I, Huculak C, Clarke S, Clegg R, Jassal DS, Gordon PMK, Ober C, Frosk P, Gerull B. Characterization of a Unique Form of Arrhythmic Cardiomyopathy Caused by Recessive Mutation in LEMD2. JACC Basic Transl Sci 2019; 4:204-221. [PMID: 31061923 PMCID: PMC6488817 DOI: 10.1016/j.jacbts.2018.12.001] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/04/2018] [Revised: 11/02/2018] [Accepted: 12/03/2018] [Indexed: 02/08/2023]
Abstract
Nuclear envelope proteins have been shown to play an important role in the pathogenesis of inherited dilated cardiomyopathy. Here, we present a remarkable cardiac phenotype caused by a homozygous LEMD2 mutation in patients of the Hutterite population with juvenile cataract. Mutation carriers develop arrhythmic cardiomyopathy with mild impairment of left ventricular systolic function but severe ventricular arrhythmias leading to sudden cardiac death. Affected cardiac tissue from a deceased patient and fibroblasts exhibit elongated nuclei with abnormal condensed heterochromatin at the periphery. The patient fibroblasts demonstrate cellular senescence and reduced proliferation capacity, which may suggest an involvement of LEM domain containing protein 2 in chromatin remodeling processes and premature aging.
Collapse
Key Words
- ACM, arrhythmogenic cardiomyopathy
- BANF, barrier to autointegration factor
- CMR, cardiac magnetic resonance
- DAPI, 4′,6′-diamidino-2-phenylindole
- DCM, dilated cardiomyopathy
- DNA, deoxyribonucleic acid
- EMD, emerin
- ICD, implantable cardioverter-defibrillator
- LEMD2
- LEMD2, LEM domain containing protein 2
- LGE, late gadolinium enhancement
- LMNA, lamin A/C
- LV, left ventricular
- NE, nuclear envelope
- P, passage
- PBS, phosphate-buffered saline
- SAHF, senescence-associated heterochromatin foci
- SNV, single nucleotide variant
- chromatin remodeling
- dilated cardiomyopathy
- eGFP, enhanced green fluorescent protein
- inner nuclear membrane
- sudden death
Collapse
Affiliation(s)
- Nelly Abdelfatah
- Department of Cardiac Sciences, Libin Cardiovascular Institute of Alberta, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Ruping Chen
- Comprehensive Heart Failure Center, University Hospital Würzburg, Würzburg, Germany
| | - Henry J Duff
- Department of Cardiac Sciences, Libin Cardiovascular Institute of Alberta, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Colette M Seifer
- Section of Cardiology, Department of Internal Medicine, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Ilan Buffo
- Variety Children's Heart Centre, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Cathleen Huculak
- Department of Medical Genetics, Alberta Health Services, Calgary, Alberta, Canada
| | - Stephanie Clarke
- Department of Biochemistry and Medical Genetics, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Robin Clegg
- Department of Pediatrics, University of Calgary, Calgary, Alberta, Canada
| | - Davinder S Jassal
- Section of Cardiology, Department of Internal Medicine, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Paul M K Gordon
- Cumming School of Medicine Centre for Health Genomics and Informatics, University of Calgary, Calgary, Alberta, Canada
| | - Carole Ober
- Department of Human Genetics, The University of Chicago, Chicago, Illinois
| | | | - Patrick Frosk
- Department of Biochemistry and Medical Genetics, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, Manitoba, Canada.,Department of Pediatrics and Child Health, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Brenda Gerull
- Department of Cardiac Sciences, Libin Cardiovascular Institute of Alberta, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada.,Comprehensive Heart Failure Center, University Hospital Würzburg, Würzburg, Germany.,Department of Internal Medicine I, University Hospital Würzburg, Würzburg, Germany
| |
Collapse
|
627
|
|
628
|
Docherty MH, O'Sullivan ED, Bonventre JV, Ferenbach DA. Cellular Senescence in the Kidney. J Am Soc Nephrol 2019; 30:726-736. [PMID: 31000567 DOI: 10.1681/asn.2018121251] [Citation(s) in RCA: 179] [Impact Index Per Article: 29.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Senescent cells have undergone permanent growth arrest, adopt an altered secretory phenotype, and accumulate in the kidney and other organs with ageing and injury. Senescence has diverse physiologic roles and experimental studies support its importance in nephrogenesis, successful tissue repair, and in opposing malignant transformation. However, recent murine studies have shown that depletion of chronically senescent cells extends healthy lifespan and delays age-associated disease-implicating senescence and the senescence-associated secretory phenotype as drivers of organ dysfunction. Great interest is therefore focused on the manipulation of senescence as a novel therapeutic target in kidney disease. In this review, we examine current knowledge and areas of ongoing uncertainty regarding senescence in the human kidney and experimental models. We summarize evidence supporting the role of senescence in normal kidney development and homeostasis but also senescence-induced maladaptive repair, renal fibrosis, and transplant failure. Recent studies using senescent cell manipulation and depletion as novel therapies to treat renal disease are discussed, and we explore unanswered questions for future research.
Collapse
Affiliation(s)
| | - Eoin D O'Sullivan
- Department of Renal Medicine, Royal Infirmary of Edinburgh, Edinburgh, UK.,Centre for Inflammation Research, Queen's Medical Research Institute, University of Edinburgh, Edinburgh, UK; and
| | - Joseph V Bonventre
- Renal Division and Division of Engineering in Medicine, Brigham and Women's Hospital, Department of Medicine, Harvard Medical School, Boston, Massachusetts
| | - David A Ferenbach
- Department of Renal Medicine, Royal Infirmary of Edinburgh, Edinburgh, UK; .,Centre for Inflammation Research, Queen's Medical Research Institute, University of Edinburgh, Edinburgh, UK; and
| |
Collapse
|
629
|
Pignolo RJ, Samsonraj RM, Law SF, Wang H, Chandra A. Targeting Cell Senescence for the Treatment of Age-Related Bone Loss. Curr Osteoporos Rep 2019; 17:70-85. [PMID: 30806947 DOI: 10.1007/s11914-019-00504-2] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
PURPOSE OF REVIEW We review cell senescence in the context of age-related bone loss by broadly discussing aging mechanisms in bone, currently known inducers and markers of senescence, the senescence-associated secretory phenotype (SASP), and the emerging roles of senescence in bone homeostasis and pathology. RECENT FINDINGS Cellular senescence is a state of irreversible cell cycle arrest induced by insults or stressors including telomere attrition, oxidative stress, DNA damage, oncogene activation, and other intrinsic or extrinsic triggers and there is mounting evidence for the role of senescence in aging bone. Cellular aging also instigates a SASP that exerts detrimental paracrine and likely systemic effects. With aging, multiple cell types in the bone microenvironment become senescent, with osteocytes and myeloid cells as primary contributors to the SASP. Targeting undesired senescent cells may be a favorable strategy to promote bone anabolic and anti-resorptive functions in aging bone, with the possibility of improving bone quality and function with normal aging and/or disease.
Collapse
Affiliation(s)
- Robert J Pignolo
- Department of Medicine, Mayo Clinic, Rochester, MN, USA.
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, USA.
- Division of Geriatric Medicine & Gerontology, Mayo Clinic College of Medicine, 200 First Street SW, Rochester, MN, 55905, USA.
| | | | - Susan F Law
- Department of Medicine, Mayo Clinic, Rochester, MN, USA
| | - Haitao Wang
- Department of Medicine, Mayo Clinic, Rochester, MN, USA
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, USA
- Division of Geriatric Medicine & Gerontology, Mayo Clinic College of Medicine, 200 First Street SW, Rochester, MN, 55905, USA
| | - Abhishek Chandra
- Department of Medicine, Mayo Clinic, Rochester, MN, USA
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, USA
- Division of Geriatric Medicine & Gerontology, Mayo Clinic College of Medicine, 200 First Street SW, Rochester, MN, 55905, USA
| |
Collapse
|
630
|
Lujambio A, Banito A. Functional screening to identify senescence regulators in cancer. Curr Opin Genet Dev 2019; 54:17-24. [PMID: 30877988 DOI: 10.1016/j.gde.2019.02.001] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2018] [Accepted: 02/01/2019] [Indexed: 12/12/2022]
Abstract
Cellular senescence is implicated in numerous biological processes, and can play pleiotropic, sometimes opposing, roles in cancer. Several triggers, cell types, contexts, and senescence-associated phenotypes introduce a multitude of possibilities when studying this process and its biological consequences. Recent studies continue to characterize cellular senescence at different levels, using a combination of functional screens, in silico analysis, omics characterizations and more targeted studies. However, a comprehensive analysis of its context-dependent effects and multiple phenotypes is required. Application of state-of-the-art and emerging technologies will increase our understanding of this complex process and better guide future strategies to harness senescence to our advantage, or to target it when detrimental.
Collapse
Affiliation(s)
- Amaia Lujambio
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, USA; Liver Cancer Program, Division of Liver Diseases, Department of Medicine, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, USA; The Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, USA; Graduate School of Biomedical Sciences at Icahn School of Medicine at Mount Sinai, New York, USA
| | - Ana Banito
- Hopp Children's Cancer Center (KiTZ) and Division of Pediatric Neurooncology, German Cancer Research Center (DKFZ), Heidelberg, Germany.
| |
Collapse
|
631
|
Kritchevsky SB, Forman DE, Callahan KE, Ely EW, High KP, McFarland F, Pérez-Stable EJ, Schmader KE, Studenski SA, Williams J, Zieman S, Guralnik JM. Pathways, Contributors, and Correlates of Functional Limitation Across Specialties: Workshop Summary. J Gerontol A Biol Sci Med Sci 2019; 74:534-543. [PMID: 29697758 PMCID: PMC6417483 DOI: 10.1093/gerona/gly093] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2017] [Indexed: 12/25/2022] Open
Abstract
Traditional clinical care models focus on the measurement and normalization of individual organ systems and de-emphasize aspects of health related to the integration of physiologic systems. Measures of physical, cognitive and sensory, and psychosocial or emotional function predict important health outcomes like death and disability independently from the severity of a specific disease, cumulative co-morbidity, or disease severity measures. A growing number of clinical scientists in several subspecialties are exploring the utility of functional assessment to predict complication risk, indicate stress resistance, inform disease screening approaches and risk factor interpretation, and evaluate care. Because a substantial number of older adults in the community have some form of functional limitation, integrating functional assessment into clinical medicine could have a large impact. Although interest in functional implications for health and disease management is growing, the science underlying functional capacity, functional limitation, physical frailty, and functional metrics is often siloed among different clinicians and researchers, with fragmented concepts and methods. On August 25-26, 2016, participants at a trans-disciplinary workshop, supported by the National Institute on Aging and the John A. Hartford Foundation, explored what is known about the pathways, contributors, and correlates of physical, cognitive, and sensory functional measures across conditions and disease states; considered social determinants and health disparities; identified knowledge gaps, and suggested priorities for future research. This article summarizes those discussions.
Collapse
Affiliation(s)
- Stephen B Kritchevsky
- Department of Internal Medicine, Wake Forest School of Medicine, Winston-Salem, North Carolina
| | - Daniel E Forman
- Department of Medicine, University of Pittsburgh, Pennsylvania
| | - Kathryn E Callahan
- Department of Internal Medicine, Wake Forest School of Medicine, Winston-Salem, North Carolina
| | - E Wesley Ely
- VA Tennessee Valley Geriatric Research Education Clinical Center (GRECC) and Department of Medicine, Vanderbilt University, Nashville
| | - Kevin P High
- Department of Internal Medicine, Wake Forest School of Medicine, Winston-Salem, North Carolina
| | - Frances McFarland
- Department of Internal Medicine, Wake Forest School of Medicine, Winston-Salem, North Carolina
| | | | | | | | | | | | - Jack M Guralnik
- Department of Epidemiology and Public Health, University of Maryland School of Medicine, Baltimore
| |
Collapse
|
632
|
Tachikart Y, Malaise O, Mumme M, Jorgensen C, Brondello JM. Seno-suppressive molecules as new therapeutic perspectives in rheumatic diseases. Biochem Pharmacol 2019; 165:126-133. [PMID: 30878551 DOI: 10.1016/j.bcp.2019.03.017] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2019] [Accepted: 03/12/2019] [Indexed: 01/10/2023]
Abstract
Over the past years, through in vitro studies and unique animal models, biologists and clinicians have demonstrated that cellular senescence is at the root of numerous age-related chronic diseases including osteoarthritis and osteoporosis. This non-proliferative cellular syndrome can modify other surrounding tissue-resident cells through the establishment of a deleterious catabolic and inflammatory microenvironment. Targeting these deleterious cells through local or systemic seno-therapeutic agent delivery in pre-clinical models improves dramatically clinical signs and extends health span. In this review, we will summarize the current knowledge on cellular senescence, list the different strategies for identifying seno-suppressive therapeutic agents and their translations to rheumatic diseases.
Collapse
Affiliation(s)
- Yassin Tachikart
- IRMB (Institut of Regenerative Medicine and Biotherapies), Inserm U1183, Univ Montpellier, Montpellier, France
| | - Olivier Malaise
- IRMB (Institut of Regenerative Medicine and Biotherapies), Inserm U1183, Univ Montpellier, Montpellier, France; GIGA Research (Groupe Interdisciplinaire de Genoproteomique Appliquée), CHU de Liège & Université de Liège, Liège, Belgium
| | - Marcus Mumme
- IRMB (Institut of Regenerative Medicine and Biotherapies), Inserm U1183, Univ Montpellier, Montpellier, France; Clinic for Orthopedics and Traumatology, University Hospital of Basel, Basel, Switzerland
| | - Christian Jorgensen
- IRMB (Institut of Regenerative Medicine and Biotherapies), Inserm U1183, Univ Montpellier, Montpellier, France; Service de Rhumatologie, CHU La Peyronie, Montpellier, France
| | - Jean-Marc Brondello
- IRMB (Institut of Regenerative Medicine and Biotherapies), Inserm U1183, Univ Montpellier, Montpellier, France.
| |
Collapse
|
633
|
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
|
634
|
Habiballa L, Salmonowicz H, Passos JF. Mitochondria and cellular senescence: Implications for musculoskeletal ageing. Free Radic Biol Med 2019; 132:3-10. [PMID: 30336251 DOI: 10.1016/j.freeradbiomed.2018.10.417] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/20/2018] [Revised: 09/25/2018] [Accepted: 10/09/2018] [Indexed: 01/07/2023]
Abstract
Musculoskeletal ageing and its associated diseases are major contributors to the loss of independence and reduced quality of life in older people. Several recent studies indicate that cellular senescence is a contributor to age-related loss of function in various organs including muscle, bones and joints. Importantly, these studies indicate that therapies targeting specifically senescent cells have great therapeutic potential in improving musculoskeletal health during ageing. Senescent cells are characterised by dramatic changes in mitochondrial function, metabolism and homeostasis. Mitochondrial dysfunction has been shown to contribute to senescence and the SASP. Here we review the role of cellular senescence in musculoskeletal ageing as well as the potential mechanisms by which mitochondrial dysfunction may impact on the induction and development of the senescent phenotype.
Collapse
Affiliation(s)
- Leena Habiballa
- Institute for Cell and Molecular Biosciences & Newcastle University Institute for Ageing, Newcastle upon Tyne NE4 5PL, UK
| | - Hanna Salmonowicz
- Institute for Cell and Molecular Biosciences & Newcastle University Institute for Ageing, Newcastle upon Tyne NE4 5PL, UK
| | - João F Passos
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN 55905, USA.
| |
Collapse
|
635
|
Anderson R, Lagnado A, Maggiorani D, Walaszczyk A, Dookun E, Chapman J, Birch J, Salmonowicz H, Ogrodnik M, Jurk D, Proctor C, Correia-Melo C, Victorelli S, Fielder E, Berlinguer-Palmini R, Owens A, Greaves LC, Kolsky KL, Parini A, Douin-Echinard V, LeBrasseur NK, Arthur HM, Tual-Chalot S, Schafer MJ, Roos CM, Miller JD, Robertson N, Mann J, Adams PD, Tchkonia T, Kirkland JL, Mialet-Perez J, Richardson GD, Passos JF. Length-independent telomere damage drives post-mitotic cardiomyocyte senescence. EMBO J 2019; 38:embj.2018100492. [PMID: 30737259 PMCID: PMC6396144 DOI: 10.15252/embj.2018100492] [Citation(s) in RCA: 337] [Impact Index Per Article: 56.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2018] [Revised: 12/18/2018] [Accepted: 01/02/2019] [Indexed: 01/08/2023] Open
Abstract
Ageing is the biggest risk factor for cardiovascular disease. Cellular senescence, a process driven in part by telomere shortening, has been implicated in age‐related tissue dysfunction. Here, we address the question of how senescence is induced in rarely dividing/post‐mitotic cardiomyocytes and investigate whether clearance of senescent cells attenuates age‐related cardiac dysfunction. During ageing, human and murine cardiomyocytes acquire a senescent‐like phenotype characterised by persistent DNA damage at telomere regions that can be driven by mitochondrial dysfunction and crucially can occur independently of cell division and telomere length. Length‐independent telomere damage in cardiomyocytes activates the classical senescence‐inducing pathways, p21CIP and p16INK4a, and results in a non‐canonical senescence‐associated secretory phenotype, which is pro‐fibrotic and pro‐hypertrophic. Pharmacological or genetic clearance of senescent cells in mice alleviates detrimental features of cardiac ageing, including myocardial hypertrophy and fibrosis. Our data describe a mechanism by which senescence can occur and contribute to age‐related myocardial dysfunction and in the wider setting to ageing in post‐mitotic tissues.
Collapse
Affiliation(s)
- Rhys Anderson
- Ageing Research Laboratories, Institute for Ageing, Newcastle University, Newcastle upon Tyne, UK.,Institute for Cell and Molecular Biosciences, Newcastle University, Newcastle upon Tyne, UK
| | - Anthony Lagnado
- Ageing Research Laboratories, Institute for Ageing, Newcastle University, Newcastle upon Tyne, UK.,Institute for Cell and Molecular Biosciences, Newcastle University, Newcastle upon Tyne, UK
| | - Damien Maggiorani
- INSERM Institute of metabolic and cardiovascular diseases, University of Toulouse, Toulouse, France
| | - Anna Walaszczyk
- Cardiovascular Research Centre, Institute for Genetic Medicine, Newcastle University, Newcastle upon Tyne, UK
| | - Emily Dookun
- Institute for Cell and Molecular Biosciences, Newcastle University, Newcastle upon Tyne, UK.,Cardiovascular Research Centre, Institute for Genetic Medicine, Newcastle University, Newcastle upon Tyne, UK
| | - James Chapman
- Ageing Research Laboratories, Institute for Ageing, Newcastle University, Newcastle upon Tyne, UK.,Institute for Cell and Molecular Biosciences, Newcastle University, Newcastle upon Tyne, UK
| | - Jodie Birch
- Ageing Research Laboratories, Institute for Ageing, Newcastle University, Newcastle upon Tyne, UK.,Institute for Cell and Molecular Biosciences, Newcastle University, Newcastle upon Tyne, UK
| | - Hanna Salmonowicz
- Ageing Research Laboratories, Institute for Ageing, Newcastle University, Newcastle upon Tyne, UK.,Institute for Cell and Molecular Biosciences, Newcastle University, Newcastle upon Tyne, UK
| | - Mikolaj Ogrodnik
- Ageing Research Laboratories, Institute for Ageing, Newcastle University, Newcastle upon Tyne, UK.,Institute for Cell and Molecular Biosciences, Newcastle University, Newcastle upon Tyne, UK
| | - Diana Jurk
- Ageing Research Laboratories, Institute for Ageing, Newcastle University, Newcastle upon Tyne, UK.,Institute for Cell and Molecular Biosciences, Newcastle University, Newcastle upon Tyne, UK.,Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, USA
| | - Carole Proctor
- Ageing Research Laboratories, Institute for Ageing, Newcastle University, Newcastle upon Tyne, UK.,Institute for Cell and Molecular Biosciences, Newcastle University, Newcastle upon Tyne, UK
| | - Clara Correia-Melo
- Ageing Research Laboratories, Institute for Ageing, Newcastle University, Newcastle upon Tyne, UK.,Institute for Cell and Molecular Biosciences, Newcastle University, Newcastle upon Tyne, UK
| | - Stella Victorelli
- Ageing Research Laboratories, Institute for Ageing, Newcastle University, Newcastle upon Tyne, UK.,Institute for Cell and Molecular Biosciences, Newcastle University, Newcastle upon Tyne, UK
| | - Edward Fielder
- Ageing Research Laboratories, Institute for Ageing, Newcastle University, Newcastle upon Tyne, UK.,Institute for Cell and Molecular Biosciences, Newcastle University, Newcastle upon Tyne, UK
| | | | - Andrew Owens
- Cardiovascular Research Centre, Institute for Genetic Medicine, Newcastle University, Newcastle upon Tyne, UK
| | - Laura C Greaves
- Wellcome Trust Centre for Mitochondrial Research, Centre for Ageing and Vitality, Newcastle University, Newcastle upon Tyne, UK
| | - Kathy L Kolsky
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, MN, USA
| | - Angelo Parini
- INSERM Institute of metabolic and cardiovascular diseases, University of Toulouse, Toulouse, France
| | - Victorine Douin-Echinard
- INSERM Institute of metabolic and cardiovascular diseases, University of Toulouse, Toulouse, France
| | | | - Helen M Arthur
- Cardiovascular Research Centre, Institute for Genetic Medicine, Newcastle University, Newcastle upon Tyne, UK
| | - Simon Tual-Chalot
- Cardiovascular Research Centre, Institute for Genetic Medicine, Newcastle University, Newcastle upon Tyne, UK
| | - Marissa J Schafer
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, MN, USA
| | - Carolyn M Roos
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, MN, USA
| | - Jordan D Miller
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, MN, USA
| | - Neil Robertson
- Institute of Cancer Sciences, CR-UK Beatson Institute, University of Glasgow, Glasgow, UK
| | - Jelena Mann
- Institute of Cellular Medicine, Newcastle University, Newcastle upon Tyne, UK
| | - Peter D Adams
- Institute of Cancer Sciences, CR-UK Beatson Institute, University of Glasgow, Glasgow, UK.,Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA
| | - Tamara Tchkonia
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, MN, USA
| | - James L Kirkland
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, MN, USA
| | - Jeanne Mialet-Perez
- INSERM Institute of metabolic and cardiovascular diseases, University of Toulouse, Toulouse, France
| | - Gavin D Richardson
- Cardiovascular Research Centre, Institute for Genetic Medicine, Newcastle University, Newcastle upon Tyne, UK
| | - João F Passos
- Ageing Research Laboratories, Institute for Ageing, Newcastle University, Newcastle upon Tyne, UK .,Institute for Cell and Molecular Biosciences, Newcastle University, Newcastle upon Tyne, UK.,Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, USA
| |
Collapse
|
636
|
Zhang B, Lam EWF, Sun Y. Senescent cells: A new Achilles' heel to exploit for cancer medicine? Aging Cell 2019; 18:e12875. [PMID: 30450821 PMCID: PMC6351840 DOI: 10.1111/acel.12875] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2018] [Revised: 10/09/2018] [Accepted: 10/15/2018] [Indexed: 01/07/2023] Open
Abstract
Cellular senescence is a typical tumor‐suppressive mechanism that restricts the proliferation of premalignant cells. However, mounting evidence suggests that senescent cells, which also persist in vivo, can promote the incidence of aging‐related disorders principally via the senescence‐associated secretory phenotype (SASP), among which cancer is particularly devastating. Despite the beneficial effects of the SASP on certain physiological events such as wound healing and tissue repair, more studies have demonstrated that senescent cells can substantially contribute to pathological conditions and accelerate disease exacerbation, particularly cancer resistance, relapse and metastasis. To limit the detrimental properties while retaining the beneficial aspects of senescent cells, research advancements that support screening, design and optimization of anti‐aging therapeutic agents are in rapid progress in the setting of prospective development of clinical strategies, which together represent a new wave of efforts to control human malignancies or mitigate degenerative complications.
Collapse
Affiliation(s)
- Boyi Zhang
- Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institutes for Biological Sciences; University of Chinese Academy of Sciences, Chinese Academy of Sciences; Shanghai China
| | - Eric W.-F. Lam
- Department of Surgery and Cancer; Imperial College London; London UK
| | - Yu Sun
- Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institutes for Biological Sciences; University of Chinese Academy of Sciences, Chinese Academy of Sciences; Shanghai China
- Department of Medicine and VAPSHCS; University of Washington; Seattle Washington
| |
Collapse
|
637
|
Abstract
Originally thought of as a stress response end point, the view of cellular senescence has since evolved into one encompassing a wide range of physiological and pathological functions, including both protumorignic and antitumorigenic features. It has also become evident that senescence is a highly dynamic and heterogenous process. Efforts to reconcile the beneficial and detrimental features of senescence suggest that physiological functions require the transient presence of senescent cells in the tissue microenvironment. Here, we propose the concept of a physiological "senescence life cycle," which has pathological consequences if not executed in its entirety.
Collapse
Affiliation(s)
- Adelyne Sue Li Chan
- Cancer Research UK Cambridge Institute, Li Ka Shing Centre, University of Cambridge, Cambridge CB2 0RE, United Kingdom
| | - Masashi Narita
- Cancer Research UK Cambridge Institute, Li Ka Shing Centre, University of Cambridge, Cambridge CB2 0RE, United Kingdom
| |
Collapse
|
638
|
Li W, He Y, Zhang R, Zheng G, Zhou D. The curcumin analog EF24 is a novel senolytic agent. Aging (Albany NY) 2019; 11:771-782. [PMID: 30694217 PMCID: PMC6366974 DOI: 10.18632/aging.101787] [Citation(s) in RCA: 108] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2018] [Accepted: 01/15/2019] [Indexed: 05/07/2023]
Abstract
Cellular senescence is a hallmark of aging because senescent cells (SCs) accumulate with aging and play a causative role in age-related diseases. Selectively eliminating SCs has been emerging as a new strategy for treating age-related diseases and extending healthspan. Curcumin and its analogs have some anti-aging activities. However, the mechanisms of their action have not been fully elucidated. In the present study, we investigated whether various curcumin analogs can function as a senolytic agent. The results from our studies show that among these curcumin analogs EF24 is the most potent and broad-spectrum senolytic agent. Mechanistically, EF24 selectively kills SCs by inducing SC apoptosis in a reactive oxygen species (ROS) production independent manner but associated with an increase in the proteasome degradation of the Bcl-2 anti-apoptotic protein family proteins known to play an important role in protecting SCs from apoptosis. In addition, EF24 can synergistically kill SCs with ABT-263, a Bcl-2 and Bcl-xl inhibitor and a known senolytic agent. These findings provide new insights into the mechanisms by which curcumin analogs function as an anti-aging agent and suggest that the curcumin analog EF24 has the potential to be used as a novel senolytic agent for the treatment of age-related diseases.
Collapse
Affiliation(s)
- Wen Li
- School of Pharmaceutical Science and Yunnan Key Laboratory of Pharmacology for Natural Products, Kunming Medical University, Kunming, Yunnan 650500, China
- Department of Pharmacodynamics, College of Pharmacy, University of Florida, Gainesville, FL 32610, USA
- Department of Endocrinology, The Third People’s Hospital of Yunnan Province, Kunming, Yunnan 650011, China
| | - Yonghan He
- Department of Pharmacodynamics, College of Pharmacy, University of Florida, Gainesville, FL 32610, USA
| | - Rongping Zhang
- School of Pharmaceutical Science and Yunnan Key Laboratory of Pharmacology for Natural Products, Kunming Medical University, Kunming, Yunnan 650500, China
| | - Guangrong Zheng
- Department of Medicinal Chemistry, College of Pharmacy, University of Florida, Gainesville, FL 32610, USA
| | - Daohong Zhou
- Department of Pharmacodynamics, College of Pharmacy, University of Florida, Gainesville, FL 32610, USA
| |
Collapse
|
639
|
Calcinotto A, Kohli J, Zagato E, Pellegrini L, Demaria M, Alimonti A. Cellular Senescence: Aging, Cancer, and Injury. Physiol Rev 2019; 99:1047-1078. [PMID: 30648461 DOI: 10.1152/physrev.00020.2018] [Citation(s) in RCA: 785] [Impact Index Per Article: 130.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
Cellular senescence is a permanent state of cell cycle arrest that occurs in proliferating cells subjected to different stresses. Senescence is, therefore, a cellular defense mechanism that prevents the cells to acquire an unnecessary damage. The senescent state is accompanied by a failure to re-enter the cell cycle in response to mitogenic stimuli, an enhanced secretory phenotype and resistance to cell death. Senescence takes place in several tissues during different physiological and pathological processes such as tissue remodeling, injury, cancer, and aging. Although senescence is one of the causative processes of aging and it is responsible of aging-related disorders, senescent cells can also play a positive role. In embryogenesis and tissue remodeling, senescent cells are required for the proper development of the embryo and tissue repair. In cancer, senescence works as a potent barrier to prevent tumorigenesis. Therefore, the identification and characterization of key features of senescence, the induction of senescence in cancer cells, or the elimination of senescent cells by pharmacological interventions in aging tissues is gaining consideration in several fields of research. Here, we describe the known key features of senescence, the cell-autonomous, and noncell-autonomous regulators of senescence, and we attempt to discuss the functional role of this fundamental process in different contexts in light of the development of novel therapeutic targets.
Collapse
Affiliation(s)
- Arianna Calcinotto
- Institute of Oncology Research (IOR), Oncology Institute of Southern Switzerland , Bellinzona , Switzerland ; University of Groningen, European Research Institute for the Biology of Ageing, University Medical Center Groningen , Groningen , The Netherlands ; IOR, Oncology Institute of Southern Switzerland , Bellinzona , Switzerland ; Università della Svizzera Italiana, Faculty of Biomedical Sciences , Lugano , Italy ; Faculty of Biology and Medicine, University of Lausanne UNIL , Lausanne , Switzerland ; and Department of Medicine, Venetian Institute of Molecular Medicine, University of Padova , Padova , Italy
| | - Jaskaren Kohli
- Institute of Oncology Research (IOR), Oncology Institute of Southern Switzerland , Bellinzona , Switzerland ; University of Groningen, European Research Institute for the Biology of Ageing, University Medical Center Groningen , Groningen , The Netherlands ; IOR, Oncology Institute of Southern Switzerland , Bellinzona , Switzerland ; Università della Svizzera Italiana, Faculty of Biomedical Sciences , Lugano , Italy ; Faculty of Biology and Medicine, University of Lausanne UNIL , Lausanne , Switzerland ; and Department of Medicine, Venetian Institute of Molecular Medicine, University of Padova , Padova , Italy
| | - Elena Zagato
- Institute of Oncology Research (IOR), Oncology Institute of Southern Switzerland , Bellinzona , Switzerland ; University of Groningen, European Research Institute for the Biology of Ageing, University Medical Center Groningen , Groningen , The Netherlands ; IOR, Oncology Institute of Southern Switzerland , Bellinzona , Switzerland ; Università della Svizzera Italiana, Faculty of Biomedical Sciences , Lugano , Italy ; Faculty of Biology and Medicine, University of Lausanne UNIL , Lausanne , Switzerland ; and Department of Medicine, Venetian Institute of Molecular Medicine, University of Padova , Padova , Italy
| | - Laura Pellegrini
- Institute of Oncology Research (IOR), Oncology Institute of Southern Switzerland , Bellinzona , Switzerland ; University of Groningen, European Research Institute for the Biology of Ageing, University Medical Center Groningen , Groningen , The Netherlands ; IOR, Oncology Institute of Southern Switzerland , Bellinzona , Switzerland ; Università della Svizzera Italiana, Faculty of Biomedical Sciences , Lugano , Italy ; Faculty of Biology and Medicine, University of Lausanne UNIL , Lausanne , Switzerland ; and Department of Medicine, Venetian Institute of Molecular Medicine, University of Padova , Padova , Italy
| | - Marco Demaria
- Institute of Oncology Research (IOR), Oncology Institute of Southern Switzerland , Bellinzona , Switzerland ; University of Groningen, European Research Institute for the Biology of Ageing, University Medical Center Groningen , Groningen , The Netherlands ; IOR, Oncology Institute of Southern Switzerland , Bellinzona , Switzerland ; Università della Svizzera Italiana, Faculty of Biomedical Sciences , Lugano , Italy ; Faculty of Biology and Medicine, University of Lausanne UNIL , Lausanne , Switzerland ; and Department of Medicine, Venetian Institute of Molecular Medicine, University of Padova , Padova , Italy
| | - Andrea Alimonti
- Institute of Oncology Research (IOR), Oncology Institute of Southern Switzerland , Bellinzona , Switzerland ; University of Groningen, European Research Institute for the Biology of Ageing, University Medical Center Groningen , Groningen , The Netherlands ; IOR, Oncology Institute of Southern Switzerland , Bellinzona , Switzerland ; Università della Svizzera Italiana, Faculty of Biomedical Sciences , Lugano , Italy ; Faculty of Biology and Medicine, University of Lausanne UNIL , Lausanne , Switzerland ; and Department of Medicine, Venetian Institute of Molecular Medicine, University of Padova , Padova , Italy
| |
Collapse
|
640
|
Lau L, David G. Pro- and anti-tumorigenic functions of the senescence-associated secretory phenotype. Expert Opin Ther Targets 2019; 23:1041-1051. [PMID: 30616404 DOI: 10.1080/14728222.2019.1565658] [Citation(s) in RCA: 50] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Introduction: Cellular senescence is a stable form of cell cycle exit. Though they no longer divide, senescent cells remain metabolically active and secrete a plethora of proteins collectively termed the senescence-associated secretory phenotype (SASP). Although senescence-associated cell cycle exit likely evolved as an anti-tumor mechanism, the SASP contains both anti- and pro-tumorigenic potential.Areas covered: In this review, we briefly discuss the discovery of senescent cells and its relationship to cancer and aging. We also describe the initiation and regulation of the SASP upon senescence stimulus onset. We focus on both the pro- and anti-tumorigenic properties of the SASP. Finally, we speculate on the potential benefits of therapy-induced senescence combined with selective SASP inhibition for the treatment of cancer.Expert opinion: Further identification and characterization of the SASP factors that are pro-tumorigenic and those that are anti-tumorigenic in specific contexts will be crucial in order to develop personalized therapeutics for the successful treatment of cancer.
Collapse
Affiliation(s)
- Lena Lau
- Department of Biochemistry and Molecular Pharmacology, New York University School of Medicine, New York, NY, USA
| | - Gregory David
- Department of Biochemistry and Molecular Pharmacology, New York University School of Medicine, New York, NY, USA.,Department of Urology, New York University School of Medicine, New York, NY, USA.,NYU Cancer Institute, New York University School of Medicine, New York, NY, USA
| |
Collapse
|
641
|
Justice JN, Nambiar AM, Tchkonia T, LeBrasseur NK, Pascual R, Hashmi SK, Prata L, Masternak MM, Kritchevsky SB, Musi N, Kirkland JL. Senolytics in idiopathic pulmonary fibrosis: Results from a first-in-human, open-label, pilot study. EBioMedicine 2019; 40:554-563. [PMID: 30616998 PMCID: PMC6412088 DOI: 10.1016/j.ebiom.2018.12.052] [Citation(s) in RCA: 796] [Impact Index Per Article: 132.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2018] [Revised: 12/14/2018] [Accepted: 12/21/2018] [Indexed: 01/19/2023] Open
Abstract
Background Cellular senescence is a key mechanism that drives age-related diseases, but has yet to be targeted therapeutically in humans. Idiopathic pulmonary fibrosis (IPF) is a progressive, fatal cellular senescence-associated disease. Selectively ablating senescent cells using dasatinib plus quercetin (DQ) alleviates IPF-related dysfunction in bleomycin-administered mice. Methods A two-center, open-label study of intermittent DQ (D:100 mg/day, Q:1250 mg/day, three-days/week over three-weeks) was conducted in participants with IPF (n = 14) to evaluate feasibility of implementing a senolytic intervention. The primary endpoints were retention rates and completion rates for planned clinical assessments. Secondary endpoints were safety and change in functional and reported health measures. Associations with the senescence-associated secretory phenotype (SASP) were explored. Findings Fourteen patients with stable IPF were recruited. The retention rate was 100% with no DQ discontinuation; planned clinical assessments were complete in 13/14 participants. One serious adverse event was reported. Non-serious events were primarily mild-moderate, with respiratory symptoms (n = 16 total events), skin irritation/bruising (n = 14), and gastrointestinal discomfort (n = 12) being most frequent. Physical function evaluated as 6-min walk distance, 4-m gait speed, and chair-stands time was significantly and clinically-meaningfully improved (p < .05). Pulmonary function, clinical chemistries, frailty index (FI-LAB), and reported health were unchanged. DQ effects on circulat.ing SASP factors were inconclusive, but correlations were observed between change in function and change in SASP-related matrix-remodeling proteins, microRNAs, and pro-inflammatory cytokines (23/48 markers r ≥ 0.50). Interpretation Our first-in-humans open-label pilot supports study feasibility and provides initial evidence that senolytics may alleviate physical dysfunction in IPF, warranting evaluation of DQ in larger randomized controlled trials for senescence-related diseases. ClinicalTrials.gov identifier: NCT02874989 (posted 2016–2018).
Collapse
Affiliation(s)
- Jamie N Justice
- Sticht Center for Healthy Aging and Alzheimer's Prevention, Internal Medicine - Gerontology and Geriatric Medicine, Wake Forest School of Medicine (WFSM), 1 Medical Center Blvd, Winston-Salem, NC 27157, United States.
| | - Anoop M Nambiar
- Division of Pulmonary Diseases and Critical Care Medicine, Department of Internal Medicine, University of Texas Health Sciences Center at San Antonio (UTHSCSA) and South Texas Veterans Health Care System, San Antonio, TX 78229, United States.
| | - Tamar Tchkonia
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, MN 55905, United States.
| | - Nathan K LeBrasseur
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, MN 55905, United States.
| | - Rodolfo Pascual
- Internal Medicine - Pulmonary, Critical Care, Allergy, Immunologic Medicine, Wake Forest School of Medicine, 1 Medical Center Blvd, Winston-Salem, NC 27157, United States.
| | - Shahrukh K Hashmi
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, MN 55905, United States.
| | - Larissa Prata
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, MN 55905, United States.
| | - Michal M Masternak
- Burnett School of Biomedical Sciences, University of Central Florida, Orlando, FL 32827, United States.
| | - Stephen B Kritchevsky
- Sticht Center for Healthy Aging and Alzheimer's Prevention, Internal Medicine - Gerontology and Geriatric Medicine, Wake Forest School of Medicine (WFSM), 1 Medical Center Blvd, Winston-Salem, NC 27157, United States.
| | - Nicolas Musi
- Barshop Institute for Longevity and Aging Studies, Center for Healthy Aging, University of Texas Health Sciences Center at San Antonio and South Texas Veterans Health Care System, San Antonio, TX 78229, United States; San Antonio Geriatric Research, Education and Clinical Center, South Texas Veterans Health Care System, San Antonio, TX 78229, United States.
| | - James L Kirkland
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, MN 55905, United States.
| |
Collapse
|
642
|
Lau A, Kennedy BK, Kirkland JL, Tullius SG. Mixing old and young: enhancing rejuvenation and accelerating aging. J Clin Invest 2019; 129:4-11. [PMID: 30601138 DOI: 10.1172/jci123946] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Donor age and recipient age are factors that influence transplantation outcomes. Aside from age-associated differences in intrinsic graft function and alloimmune responses, the ability of young and old cells to exert either rejuvenating or aging effects extrinsically may also apply to the transplantation of hematopoietic stem cells or solid organ transplants. While the potential for rejuvenation mediated by the transfer of youthful cells is currently being explored for therapeutic applications, aspects that relate to accelerating aging are no less clinically significant. Those effects may be particularly relevant in transplantation with an age discrepancy between donor and recipient. Here, we review recent advances in understanding the mechanisms by which young and old cells modify their environments to promote rejuvenation- or aging-associated phenotypes. We discuss their relevance to clinical transplantation and highlight potential opportunities for therapeutic intervention.
Collapse
Affiliation(s)
- Ashley Lau
- Division of Transplant Surgery, Department of Surgery, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Brian K Kennedy
- Departments of Biochemistry and Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore.,Singapore Institute for Clinical Sciences, Singapore.,Agency for Science, Technology and Research (A*STAR), Singapore.,Buck Institute for Research on Aging, Novato, California, USA
| | - James L Kirkland
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, Minnesota, USA
| | - Stefan G Tullius
- Division of Transplant Surgery, Department of Surgery, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
643
|
Mahmoudi S, Xu L, Brunet A. Turning back time with emerging rejuvenation strategies. Nat Cell Biol 2019; 21:32-43. [PMID: 30602763 DOI: 10.1038/s41556-018-0206-0] [Citation(s) in RCA: 111] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2018] [Accepted: 08/24/2018] [Indexed: 01/10/2023]
Abstract
Ageing is associated with the functional decline of all tissues and a striking increase in many diseases. Although ageing has long been considered a one-way street, strategies to delay and potentially even reverse the ageing process have recently been developed. Here, we review four emerging rejuvenation strategies-systemic factors, metabolic manipulations, senescent cell ablation and cellular reprogramming-and discuss their mechanisms of action, cellular targets, potential trade-offs and application to human ageing.
Collapse
Affiliation(s)
- Salah Mahmoudi
- Department of Genetics, Stanford University, Stanford, CA, USA
| | - Lucy Xu
- Department of Genetics, Stanford University, Stanford, CA, USA.,Department of Biology, Stanford University, Stanford, CA, USA
| | - Anne Brunet
- Department of Genetics, Stanford University, Stanford, CA, USA. .,Glenn Laboratories for the Biology of Aging, Stanford University, Stanford, CA, USA.
| |
Collapse
|
644
|
Larsson L, Degens H, Li M, Salviati L, Lee YI, Thompson W, Kirkland JL, Sandri M. Sarcopenia: Aging-Related Loss of Muscle Mass and Function. Physiol Rev 2019; 99:427-511. [PMID: 30427277 PMCID: PMC6442923 DOI: 10.1152/physrev.00061.2017] [Citation(s) in RCA: 965] [Impact Index Per Article: 160.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2017] [Revised: 05/14/2018] [Accepted: 06/15/2018] [Indexed: 12/11/2022] Open
Abstract
Sarcopenia is a loss of muscle mass and function in the elderly that reduces mobility, diminishes quality of life, and can lead to fall-related injuries, which require costly hospitalization and extended rehabilitation. This review focuses on the aging-related structural changes and mechanisms at cellular and subcellular levels underlying changes in the individual motor unit: specifically, the perikaryon of the α-motoneuron, its neuromuscular junction(s), and the muscle fibers that it innervates. Loss of muscle mass with aging, which is largely due to the progressive loss of motoneurons, is associated with reduced muscle fiber number and size. Muscle function progressively declines because motoneuron loss is not adequately compensated by reinnervation of muscle fibers by the remaining motoneurons. At the intracellular level, key factors are qualitative changes in posttranslational modifications of muscle proteins and the loss of coordinated control between contractile, mitochondrial, and sarcoplasmic reticulum protein expression. Quantitative and qualitative changes in skeletal muscle during the process of aging also have been implicated in the pathogenesis of acquired and hereditary neuromuscular disorders. In experimental models, specific intervention strategies have shown encouraging results on limiting deterioration of motor unit structure and function under conditions of impaired innervation. Translated to the clinic, if these or similar interventions, by saving muscle and improving mobility, could help alleviate sarcopenia in the elderly, there would be both great humanitarian benefits and large cost savings for health care systems.
Collapse
Affiliation(s)
- Lars Larsson
- Department of Physiology and Pharmacology, Basic and Clinical Muscle Biology Group, Karolinska Institutet , Stockholm , Sweden ; Section of Clinical Neurophysiology, Department of Clinical Neuroscience, Karolinska Institutet , Stockholm , Sweden ; Department of Biobehavioral Health, The Pennsylvania State University , University Park, Pennsylvania ; School of Healthcare Science, Metropolitan University , Manchester , United Kingdom ; Institute of Sport Science and Innovations, Lithuanian Sports University , Kaunas , Lithuania ; Clinical Genetics Unit, Department of Woman and Child Health, University of Padova , Padova , Italy ; IRP Città della Speranza, Padova , Italy ; Department of Biology, Texas A&M University , College Station, Texas ; Robert and Arlene Kogod Center on Aging, Mayo Clinic , Rochester, Minnesota ; Department of Biomedical Science, Venetian Institute of Molecular Medicine, University of Padova , Padova , Italy
| | - Hans Degens
- Department of Physiology and Pharmacology, Basic and Clinical Muscle Biology Group, Karolinska Institutet , Stockholm , Sweden ; Section of Clinical Neurophysiology, Department of Clinical Neuroscience, Karolinska Institutet , Stockholm , Sweden ; Department of Biobehavioral Health, The Pennsylvania State University , University Park, Pennsylvania ; School of Healthcare Science, Metropolitan University , Manchester , United Kingdom ; Institute of Sport Science and Innovations, Lithuanian Sports University , Kaunas , Lithuania ; Clinical Genetics Unit, Department of Woman and Child Health, University of Padova , Padova , Italy ; IRP Città della Speranza, Padova , Italy ; Department of Biology, Texas A&M University , College Station, Texas ; Robert and Arlene Kogod Center on Aging, Mayo Clinic , Rochester, Minnesota ; Department of Biomedical Science, Venetian Institute of Molecular Medicine, University of Padova , Padova , Italy
| | - Meishan Li
- Department of Physiology and Pharmacology, Basic and Clinical Muscle Biology Group, Karolinska Institutet , Stockholm , Sweden ; Section of Clinical Neurophysiology, Department of Clinical Neuroscience, Karolinska Institutet , Stockholm , Sweden ; Department of Biobehavioral Health, The Pennsylvania State University , University Park, Pennsylvania ; School of Healthcare Science, Metropolitan University , Manchester , United Kingdom ; Institute of Sport Science and Innovations, Lithuanian Sports University , Kaunas , Lithuania ; Clinical Genetics Unit, Department of Woman and Child Health, University of Padova , Padova , Italy ; IRP Città della Speranza, Padova , Italy ; Department of Biology, Texas A&M University , College Station, Texas ; Robert and Arlene Kogod Center on Aging, Mayo Clinic , Rochester, Minnesota ; Department of Biomedical Science, Venetian Institute of Molecular Medicine, University of Padova , Padova , Italy
| | - Leonardo Salviati
- Department of Physiology and Pharmacology, Basic and Clinical Muscle Biology Group, Karolinska Institutet , Stockholm , Sweden ; Section of Clinical Neurophysiology, Department of Clinical Neuroscience, Karolinska Institutet , Stockholm , Sweden ; Department of Biobehavioral Health, The Pennsylvania State University , University Park, Pennsylvania ; School of Healthcare Science, Metropolitan University , Manchester , United Kingdom ; Institute of Sport Science and Innovations, Lithuanian Sports University , Kaunas , Lithuania ; Clinical Genetics Unit, Department of Woman and Child Health, University of Padova , Padova , Italy ; IRP Città della Speranza, Padova , Italy ; Department of Biology, Texas A&M University , College Station, Texas ; Robert and Arlene Kogod Center on Aging, Mayo Clinic , Rochester, Minnesota ; Department of Biomedical Science, Venetian Institute of Molecular Medicine, University of Padova , Padova , Italy
| | - Young Il Lee
- Department of Physiology and Pharmacology, Basic and Clinical Muscle Biology Group, Karolinska Institutet , Stockholm , Sweden ; Section of Clinical Neurophysiology, Department of Clinical Neuroscience, Karolinska Institutet , Stockholm , Sweden ; Department of Biobehavioral Health, The Pennsylvania State University , University Park, Pennsylvania ; School of Healthcare Science, Metropolitan University , Manchester , United Kingdom ; Institute of Sport Science and Innovations, Lithuanian Sports University , Kaunas , Lithuania ; Clinical Genetics Unit, Department of Woman and Child Health, University of Padova , Padova , Italy ; IRP Città della Speranza, Padova , Italy ; Department of Biology, Texas A&M University , College Station, Texas ; Robert and Arlene Kogod Center on Aging, Mayo Clinic , Rochester, Minnesota ; Department of Biomedical Science, Venetian Institute of Molecular Medicine, University of Padova , Padova , Italy
| | - Wesley Thompson
- Department of Physiology and Pharmacology, Basic and Clinical Muscle Biology Group, Karolinska Institutet , Stockholm , Sweden ; Section of Clinical Neurophysiology, Department of Clinical Neuroscience, Karolinska Institutet , Stockholm , Sweden ; Department of Biobehavioral Health, The Pennsylvania State University , University Park, Pennsylvania ; School of Healthcare Science, Metropolitan University , Manchester , United Kingdom ; Institute of Sport Science and Innovations, Lithuanian Sports University , Kaunas , Lithuania ; Clinical Genetics Unit, Department of Woman and Child Health, University of Padova , Padova , Italy ; IRP Città della Speranza, Padova , Italy ; Department of Biology, Texas A&M University , College Station, Texas ; Robert and Arlene Kogod Center on Aging, Mayo Clinic , Rochester, Minnesota ; Department of Biomedical Science, Venetian Institute of Molecular Medicine, University of Padova , Padova , Italy
| | - James L Kirkland
- Department of Physiology and Pharmacology, Basic and Clinical Muscle Biology Group, Karolinska Institutet , Stockholm , Sweden ; Section of Clinical Neurophysiology, Department of Clinical Neuroscience, Karolinska Institutet , Stockholm , Sweden ; Department of Biobehavioral Health, The Pennsylvania State University , University Park, Pennsylvania ; School of Healthcare Science, Metropolitan University , Manchester , United Kingdom ; Institute of Sport Science and Innovations, Lithuanian Sports University , Kaunas , Lithuania ; Clinical Genetics Unit, Department of Woman and Child Health, University of Padova , Padova , Italy ; IRP Città della Speranza, Padova , Italy ; Department of Biology, Texas A&M University , College Station, Texas ; Robert and Arlene Kogod Center on Aging, Mayo Clinic , Rochester, Minnesota ; Department of Biomedical Science, Venetian Institute of Molecular Medicine, University of Padova , Padova , Italy
| | - Marco Sandri
- Department of Physiology and Pharmacology, Basic and Clinical Muscle Biology Group, Karolinska Institutet , Stockholm , Sweden ; Section of Clinical Neurophysiology, Department of Clinical Neuroscience, Karolinska Institutet , Stockholm , Sweden ; Department of Biobehavioral Health, The Pennsylvania State University , University Park, Pennsylvania ; School of Healthcare Science, Metropolitan University , Manchester , United Kingdom ; Institute of Sport Science and Innovations, Lithuanian Sports University , Kaunas , Lithuania ; Clinical Genetics Unit, Department of Woman and Child Health, University of Padova , Padova , Italy ; IRP Città della Speranza, Padova , Italy ; Department of Biology, Texas A&M University , College Station, Texas ; Robert and Arlene Kogod Center on Aging, Mayo Clinic , Rochester, Minnesota ; Department of Biomedical Science, Venetian Institute of Molecular Medicine, University of Padova , Padova , Italy
| |
Collapse
|
645
|
Abstract
Chronic obstructive pulmonary disease (COPD) and idiopathic pulmonary fibrosis are regarded as a diseases of accelerated lung ageing and show all of the hallmarks of ageing, including telomere shortening, cellular senescence, activation of PI3 kinase-mTOR signaling, impaired autophagy, mitochondrial dysfunction, stem cell exhaustion, epigenetic changes, abnormal microRNA profiles, immunosenescence and a low grade chronic inflammation due to senescence-associated secretory phenotype (SASP). Many of these ageing mechanisms are driven by exogenous and endogenous oxidative stress. There is also a reduction in anti-ageing molecules, such as sirtuins and Klotho, which further accelerate the ageing process. Understanding these molecular mechanisms has identified several novel therapeutic targets and several drugs and dietary interventions are now in development to treat chronic lung disease.
Collapse
Affiliation(s)
- Peter J Barnes
- Airway Disease Section, National Heart and Lung Institute, Imperial College, London, UK.
| |
Collapse
|
646
|
Sedlackova L, Korolchuk VI. Mitochondrial quality control as a key determinant of cell survival. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2018; 1866:575-587. [PMID: 30594496 DOI: 10.1016/j.bbamcr.2018.12.012] [Citation(s) in RCA: 110] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/16/2018] [Revised: 12/03/2018] [Accepted: 12/21/2018] [Indexed: 01/09/2023]
Abstract
Mitochondria are the energy producing dynamic double-membraned organelles essential for cellular and organismal survival. A multitude of intra- and extra-cellular signals involved in the regulation of energy metabolism and cell fate determination converge on mitochondria to promote or prevent cell survival by modulating mitochondrial function and structure. Mitochondrial fitness is maintained by mitophagy, a pathway of selective degradation of dysfunctional organelles. Mitophagy impairment and altered clearance results in increased levels of dysfunctional and structurally aberrant mitochondria, changes in energy production, loss of responsiveness to intra- and extra-cellular signals and ultimately cell death. The decline of mitochondrial function and homeostasis with age is reported to be central to age-related pathologies. Here we discuss the molecular mechanisms controlling mitochondrial dynamics, mitophagy and cell death signalling and how their perturbation may contribute to ageing and age-related illness.
Collapse
Affiliation(s)
- Lucia Sedlackova
- Institute for Cell and Molecular Biosciences, Newcastle University, Newcastle upon Tyne NE4 5PL, UK
| | - Viktor I Korolchuk
- Institute for Cell and Molecular Biosciences, Newcastle University, Newcastle upon Tyne NE4 5PL, UK.
| |
Collapse
|
647
|
Ovadya Y, Landsberger T, Leins H, Vadai E, Gal H, Biran A, Yosef R, Sagiv A, Agrawal A, Shapira A, Windheim J, Tsoory M, Schirmbeck R, Amit I, Geiger H, Krizhanovsky V. Impaired immune surveillance accelerates accumulation of senescent cells and aging. Nat Commun 2018; 9:5435. [PMID: 30575733 PMCID: PMC6303397 DOI: 10.1038/s41467-018-07825-3] [Citation(s) in RCA: 363] [Impact Index Per Article: 51.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2018] [Accepted: 11/26/2018] [Indexed: 02/06/2023] Open
Abstract
Cellular senescence is a stress response that imposes stable cell-cycle arrest in damaged cells, preventing their propagation in tissues. However, senescent cells accumulate in tissues in advanced age, where they might promote tissue degeneration and malignant transformation. The extent of immune-system involvement in regulating age-related accumulation of senescent cells, and its consequences, are unknown. Here we show that Prf1-/- mice with impaired cell cytotoxicity exhibit both higher senescent-cell tissue burden and chronic inflammation. They suffer from multiple age-related disorders and lower survival. Strikingly, pharmacological elimination of senescent-cells by ABT-737 partially alleviates accelerated aging phenotype in these mice. In LMNA+/G609G progeroid mice, impaired cell cytotoxicity further promotes senescent-cell accumulation and shortens lifespan. ABT-737 administration during the second half of life of these progeroid mice abrogates senescence signature and increases median survival. Our findings shed new light on mechanisms governing senescent-cell presence in aging, and could motivate new strategies for regenerative medicine.
Collapse
Affiliation(s)
- Yossi Ovadya
- Department of Molecular Cell Biology, The Weizmann Institute of Science, 76100, Rehovot, Israel
| | - Tomer Landsberger
- Department of Immunology, The Weizmann Institute of Science, 76100, Rehovot, Israel
| | - Hanna Leins
- Institute of Molecular Medicine, Stem Cell and Aging, Ulm University, Ulm, 89081, Germany.,Department of Internal Medicine I, University Hospital of Ulm, Ulm, 89081, Germany
| | - Ezra Vadai
- Department of Molecular Cell Biology, The Weizmann Institute of Science, 76100, Rehovot, Israel
| | - Hilah Gal
- Department of Molecular Cell Biology, The Weizmann Institute of Science, 76100, Rehovot, Israel
| | - Anat Biran
- Department of Molecular Cell Biology, The Weizmann Institute of Science, 76100, Rehovot, Israel
| | - Reut Yosef
- Department of Molecular Cell Biology, The Weizmann Institute of Science, 76100, Rehovot, Israel
| | - Adi Sagiv
- Department of Molecular Cell Biology, The Weizmann Institute of Science, 76100, Rehovot, Israel
| | - Amit Agrawal
- Department of Molecular Cell Biology, The Weizmann Institute of Science, 76100, Rehovot, Israel
| | - Alon Shapira
- Department of Molecular Cell Biology, The Weizmann Institute of Science, 76100, Rehovot, Israel
| | - Joseph Windheim
- Department of Molecular Cell Biology, The Weizmann Institute of Science, 76100, Rehovot, Israel
| | - Michael Tsoory
- Department of Veterinary Resources, The Weizmann Institute of Science, 76100, Rehovot, Israel
| | - Reinhold Schirmbeck
- Department of Internal Medicine I, University Hospital of Ulm, Ulm, 89081, Germany
| | - Ido Amit
- Department of Immunology, The Weizmann Institute of Science, 76100, Rehovot, Israel
| | - Hartmut Geiger
- Institute of Molecular Medicine, Stem Cell and Aging, Ulm University, Ulm, 89081, Germany.,Experimental Hematology and Cancer Biology Cincinnati Children's Hospital Medical Center, 45229, Cincinnati, OH, USA
| | - Valery Krizhanovsky
- Department of Molecular Cell Biology, The Weizmann Institute of Science, 76100, Rehovot, Israel.
| |
Collapse
|
648
|
Prata LGPL, Ovsyannikova IG, Tchkonia T, Kirkland JL. Senescent cell clearance by the immune system: Emerging therapeutic opportunities. Semin Immunol 2018; 40:101275. [PMID: 31088710 PMCID: PMC7061456 DOI: 10.1016/j.smim.2019.04.003] [Citation(s) in RCA: 315] [Impact Index Per Article: 45.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/02/2018] [Revised: 11/01/2018] [Accepted: 04/15/2019] [Indexed: 12/19/2022]
Abstract
Senescent cells (SCs) arise from normal cells in multiple organs due to inflammatory, metabolic, DNA damage, or tissue damage signals. SCs are non-proliferating but metabolically active cells that can secrete a range of pro-inflammatory and proteolytic factors as part of the senescence-associated secretory phenotype (SASP). Senescent cell anti-apoptotic pathways (SCAPs) protect SCs from their own pro-apoptotic SASP. SCs can chemo-attract immune cells and are usually cleared by these immune cells. During aging and in multiple chronic diseases, SCs can accumulate in dysfunctional tissues. SCs can impede innate and adaptive immune responses. Whether immune system loss of capacity to clear SCs promotes immune system dysfunction, or conversely whether immune dysfunction permits SC accumulation, are important issues that are not yet fully resolved. SCs may be able to assume distinct states that interact differentially with immune cells, thereby promoting or inhibiting SC clearance, establishing a chronically pro-senescent and pro-inflammatory environment, leading to modulation of the SASP by the immune cells recruited and activated by the SASP. Therapies that enhance immune cell-mediated clearance of SCs could provide a lever for reducing SC burden. Such therapies could include vaccines, small molecule immunomodulators, or other approaches. Senolytics, drugs that selectively eliminate SCs by transiently disabling their SCAPs, may prove to alleviate immune dysfunction in older individuals and thereby accelerate immune-mediated clearance of SCs. The more that can be understood about the interplay between SCs and the immune system, the faster new interventions may be developed to delay, prevent, or treat age-related dysfunction and the multiple senescence-associated chronic diseases and disorders.
Collapse
Affiliation(s)
- Larissa G P Langhi Prata
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, 200 First St., S.W., Rochester, MN 55905, USA.
| | - Inna G Ovsyannikova
- Mayo Clinic Vaccine Research Group, Mayo Clinic, 200 First St., S.W., Rochester, MN 55905, USA.
| | - Tamara Tchkonia
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, 200 First St., S.W., Rochester, MN 55905, USA.
| | - James L Kirkland
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, 200 First St., S.W., Rochester, MN 55905, USA.
| |
Collapse
|
649
|
Shetty AK, Kodali M, Upadhya R, Madhu LN. Emerging Anti-Aging Strategies - Scientific Basis and Efficacy. Aging Dis 2018; 9:1165-1184. [PMID: 30574426 PMCID: PMC6284760 DOI: 10.14336/ad.2018.1026] [Citation(s) in RCA: 82] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2018] [Accepted: 11/30/2018] [Indexed: 12/11/2022] Open
Abstract
The prevalence of age-related diseases is in an upward trend due to increased life expectancy in humans. Age-related conditions are among the leading causes of morbidity and death worldwide currently. Therefore, there is an urgent need to find apt interventions that slow down aging and reduce or postpone the incidence of debilitating age-related diseases. This review discusses the efficacy of emerging anti-aging approaches for maintaining better health in old age. There are many anti-aging strategies in development, which include procedures such as augmentation of autophagy, elimination of senescent cells, transfusion of plasma from young blood, intermittent fasting, enhancement of adult neurogenesis, physical exercise, antioxidant intake, and stem cell therapy. Multiple pre-clinical studies suggest that administration of autophagy enhancers, senolytic drugs, plasma from young blood, drugs that enhance neurogenesis and BDNF are promising approaches to sustain normal health during aging and also to postpone age-related neurodegenerative diseases such as Alzheimer's disease. Stem cell therapy has also shown promise for improving regeneration and function of the aged or Alzheimer's disease brain. Several of these approaches are awaiting critical appraisal in clinical trials to determine their long-term efficacy and possible adverse effects. On the other hand, procedures such as intermittent fasting, physical exercise, intake of antioxidants such as resveratrol and curcumin have shown considerable promise for improving function in aging, some of which are ready for large-scale clinical trials, as they are non-invasive, and seem to have minimal side effects. In summary, several approaches are at the forefront of becoming mainstream therapies for combating aging and postponing age-related diseases in the coming years.
Collapse
Affiliation(s)
- Ashok K. Shetty
- Institute for Regenerative Medicine, Department of Molecular and Cellular Medicine, Texas A&M University Health Science Center College of Medicine, College Station, Texas 77843, USA
- Olin E. Teague Veterans’ Medical Center, Central Texas Veterans Health Care System, Temple, Texas 76504, USA
| | - Maheedhar Kodali
- Institute for Regenerative Medicine, Department of Molecular and Cellular Medicine, Texas A&M University Health Science Center College of Medicine, College Station, Texas 77843, USA
- Olin E. Teague Veterans’ Medical Center, Central Texas Veterans Health Care System, Temple, Texas 76504, USA
| | - Raghavendra Upadhya
- Institute for Regenerative Medicine, Department of Molecular and Cellular Medicine, Texas A&M University Health Science Center College of Medicine, College Station, Texas 77843, USA
- Olin E. Teague Veterans’ Medical Center, Central Texas Veterans Health Care System, Temple, Texas 76504, USA
| | - Leelavathi N. Madhu
- Institute for Regenerative Medicine, Department of Molecular and Cellular Medicine, Texas A&M University Health Science Center College of Medicine, College Station, Texas 77843, USA
| |
Collapse
|
650
|
Yabluchanskiy A, Ungvari Z, Csiszar A, Tarantini S. Advances and challenges in geroscience research: An update. Physiol Int 2018; 105:298-308. [PMID: 30587027 PMCID: PMC9341286 DOI: 10.1556/2060.105.2018.4.32] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/05/2023]
Abstract
Aging remains the most pervasive risk factor for a wide range of chronic diseases that afflict modern societies. In the United States alone, incidence of age-related diseases (e.g., cardiovascular disease, stroke, Alzheimer's disease, vascular cognitive impairment and dementia, cancer, hypertension, type-2 diabetes, chronic obstructive pulmonary disease, and osteoarthritis) is on the rise, posing an unsustainable socioeconomic burden even for the most developed countries. Tackling each and every age-related disease alone is proving to be costly and ineffective. The emerging field of geroscience has posed itself as an interdisciplinary approach that aims to understand the relationship between the biology of aging and the pathophysiology of chronic age-related diseases. According to the geroscience concept, aging is the single major risk factor that underlies several age-related chronic diseases, and manipulation of cellular and systemic aging processes can delay the manifestation and/or severity of these age-related chronic pathologies. The goal of this endeavor is to achieve health improvements by preventing/delaying the pathogenesis of several age-related diseases simultaneously in the elderly population by targeting key cellular and molecular processes of aging instead of managing diseases of aging as they arise individually. In this review, we discuss recent advances in the field of geroscience, highlighting their implications for potential future therapeutic targets and the associated scientific challenges and opportunities that lay ahead.
Collapse
Affiliation(s)
- A Yabluchanskiy
- 1 Vascular Cognitive Impairment and Neurodegeneration Program Reynolds Oklahoma Center on Aging, Department of Geriatric Medicine, University of Oklahoma Health Sciences Center , Oklahoma City, OK, USA
- 2 Translational Geroscience Laboratory, Department of Geriatric Medicine, University of Oklahoma Health Sciences Center , Oklahoma City, Oklahoma, USA
| | - Z Ungvari
- 1 Vascular Cognitive Impairment and Neurodegeneration Program Reynolds Oklahoma Center on Aging, Department of Geriatric Medicine, University of Oklahoma Health Sciences Center , Oklahoma City, OK, USA
- 2 Translational Geroscience Laboratory, Department of Geriatric Medicine, University of Oklahoma Health Sciences Center , Oklahoma City, Oklahoma, USA
- 3 Department of Medical Physics and Informatics, University of Szeged , Szeged, Hungary
- 4 Department of Pulmonology, Semmelweis University , Budapest, Hungary
| | - A Csiszar
- 1 Vascular Cognitive Impairment and Neurodegeneration Program Reynolds Oklahoma Center on Aging, Department of Geriatric Medicine, University of Oklahoma Health Sciences Center , Oklahoma City, OK, USA
- 2 Translational Geroscience Laboratory, Department of Geriatric Medicine, University of Oklahoma Health Sciences Center , Oklahoma City, Oklahoma, USA
- 3 Department of Medical Physics and Informatics, University of Szeged , Szeged, Hungary
| | - S Tarantini
- 1 Vascular Cognitive Impairment and Neurodegeneration Program Reynolds Oklahoma Center on Aging, Department of Geriatric Medicine, University of Oklahoma Health Sciences Center , Oklahoma City, OK, USA
- 2 Translational Geroscience Laboratory, Department of Geriatric Medicine, University of Oklahoma Health Sciences Center , Oklahoma City, Oklahoma, USA
| |
Collapse
|