401
|
Yang J, Peng S, Zhang B, Houten S, Schadt E, Zhu J, Suh Y, Tu Z. Human geroprotector discovery by targeting the converging subnetworks of aging and age-related diseases. GeroScience 2020; 42:353-372. [PMID: 31637571 PMCID: PMC7031474 DOI: 10.1007/s11357-019-00106-x] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2019] [Accepted: 09/13/2019] [Indexed: 12/15/2022] Open
Abstract
A key goal of geroscience research is to identify effective interventions to extend human healthspan, the years of healthy life. Currently, majority of the geroprotectors are found by screening compounds in model organisms; whether they will be effective in humans is largely unknown. Here we present a new strategy called ANDRU (aging network based drug discovery) to help the discovery of human geroprotectors. It first identifies human aging subnetworks that putatively function at the interface between aging and age-related diseases; it then screens for pharmacological interventions that may "reverse" the age-associated transcriptional changes occurred in these subnetworks. We applied ANDRU to human adipose gene expression data from the Genotype Tissue Expression (GTEx) project. For the top 31 identified compounds, 19 of them showed at least some evidence supporting their function in improving metabolic traits or lifespan, which include type 2 diabetes drugs such as pioglitazone. As the query aging genes were refined to the ones with more intimate links to diseases, ANDRU identified more meaningful drug hits than the general approach without considering the underlying network structures. In summary, ANDRU represents a promising human data-driven strategy that may speed up the discovery of interventions to extend human healthspan.
Collapse
Affiliation(s)
- Jialiang Yang
- Institute of Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, New York City, USA
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, 1425 Madison Ave, IMI 3-70F, New York City, NY, 10029, USA
| | - Shouneng Peng
- Institute of Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, New York City, USA
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, 1425 Madison Ave, IMI 3-70F, New York City, NY, 10029, USA
| | - Bin Zhang
- Institute of Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, New York City, USA
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, 1425 Madison Ave, IMI 3-70F, New York City, NY, 10029, USA
| | - Sander Houten
- Institute of Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, New York City, USA
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, 1425 Madison Ave, IMI 3-70F, New York City, NY, 10029, USA
| | - Eric Schadt
- Institute of Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, New York City, USA
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, 1425 Madison Ave, IMI 3-70F, New York City, NY, 10029, USA
| | - Jun Zhu
- Institute of Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, New York City, USA
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, 1425 Madison Ave, IMI 3-70F, New York City, NY, 10029, USA
| | - Yousin Suh
- Department of Genetics, Albert Einstein College of Medicine, New York, New York City, USA
- Department of Medicine Endocrinology, Albert Einstein College of Medicine, New York, New York City, USA
| | - Zhidong Tu
- Institute of Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, New York City, USA.
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, 1425 Madison Ave, IMI 3-70F, New York City, NY, 10029, USA.
| |
Collapse
|
402
|
Barinda AJ, Ikeda K, Nugroho DB, Wardhana DA, Sasaki N, Honda S, Urata R, Matoba S, Hirata KI, Emoto N. Endothelial progeria induces adipose tissue senescence and impairs insulin sensitivity through senescence associated secretory phenotype. Nat Commun 2020; 11:481. [PMID: 31980643 PMCID: PMC6981212 DOI: 10.1038/s41467-020-14387-w] [Citation(s) in RCA: 65] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2019] [Accepted: 01/06/2020] [Indexed: 12/17/2022] Open
Abstract
Vascular senescence is thought to play a crucial role in an ageing-associated decline of organ functions; however, whether vascular senescence is causally implicated in age-related disease remains unclear. Here we show that endothelial cell (EC) senescence induces metabolic disorders through the senescence-associated secretory phenotype. Senescence-messaging secretomes from senescent ECs induced a senescence-like state and reduced insulin receptor substrate-1 in adipocytes, which thereby impaired insulin signaling. We generated EC-specific progeroid mice that overexpressed the dominant negative form of telomeric repeat-binding factor 2 under the control of the Tie2 promoter. EC-specific progeria impaired systemic metabolic health in mice in association with adipose tissue dysfunction even while consuming normal chow. Notably, shared circulation with EC-specific progeroid mice by parabiosis sufficiently transmitted the metabolic disorders into wild-type recipient mice. Our data provides direct evidence that EC senescence impairs systemic metabolic health, and thus establishes EC senescence as a bona fide risk for age-related metabolic disease.
Collapse
Affiliation(s)
- Agian Jeffilano Barinda
- Laboratory of Clinical Pharmaceutical Science, Kobe Pharmaceutical University, 4-19-1 Motoyamakitamachi, Higashinada, Kobe, 658-8558, Japan.,Department of Pharmacology and Therapeutic, Faculty of Medicine, Universitas Indonesia, Salemba Raya 6, Jakarta, 10430, Indonesia
| | - Koji Ikeda
- Laboratory of Clinical Pharmaceutical Science, Kobe Pharmaceutical University, 4-19-1 Motoyamakitamachi, Higashinada, Kobe, 658-8558, Japan.
| | - Dhite Bayu Nugroho
- Laboratory of Clinical Pharmaceutical Science, Kobe Pharmaceutical University, 4-19-1 Motoyamakitamachi, Higashinada, Kobe, 658-8558, Japan
| | - Donytra Arby Wardhana
- Laboratory of Clinical Pharmaceutical Science, Kobe Pharmaceutical University, 4-19-1 Motoyamakitamachi, Higashinada, Kobe, 658-8558, Japan
| | - Naoto Sasaki
- Laboratory of Medical Pharmaceutics, Kobe Pharmaceutical University, 4-19-1 Motoyamakitamachi, Higashinada, Kobe, 658-8558, Japan
| | - Sakiko Honda
- Department of Cardiology, Kyoto Prefectural University Graduate School of Medical Science, 465 Kajii, Kawaramachi-Hirokoji, Kyoto, 602-8566, Japan
| | - Ryota Urata
- Department of Cardiology, Kyoto Prefectural University Graduate School of Medical Science, 465 Kajii, Kawaramachi-Hirokoji, Kyoto, 602-8566, Japan
| | - Satoaki Matoba
- Department of Cardiology, Kyoto Prefectural University Graduate School of Medical Science, 465 Kajii, Kawaramachi-Hirokoji, Kyoto, 602-8566, Japan
| | - Ken-Ichi Hirata
- Division of Cardiovascular Medicine, Department of Internal Medicine, Kobe University Graduate School of Medicine, 7-5-1 Kusunoki, Chuo, Kobe, 6500017, Japan
| | - Noriaki Emoto
- Laboratory of Clinical Pharmaceutical Science, Kobe Pharmaceutical University, 4-19-1 Motoyamakitamachi, Higashinada, Kobe, 658-8558, Japan.,Division of Cardiovascular Medicine, Department of Internal Medicine, Kobe University Graduate School of Medicine, 7-5-1 Kusunoki, Chuo, Kobe, 6500017, Japan
| |
Collapse
|
403
|
Zhang X, Ehrlich KC, Yu F, Hu X, Meng XH, Deng HW, Shen H, Ehrlich M. Osteoporosis- and obesity-risk interrelationships: an epigenetic analysis of GWAS-derived SNPs at the developmental gene TBX15. Epigenetics 2020; 15:728-749. [PMID: 31975641 PMCID: PMC7574382 DOI: 10.1080/15592294.2020.1716491] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
A major challenge in translating findings from genome-wide association studies (GWAS) to biological mechanisms is pinpointing functional variants because only a very small percentage of variants associated with a given trait actually impact the trait. We used an extensive epigenetics, transcriptomics, and genetics analysis of the TBX15/WARS2 neighbourhood to prioritize this region's best-candidate causal variants for the genetic risk of osteoporosis (estimated bone density, eBMD) and obesity (waist-hip ratio or waist circumference adjusted for body mass index). TBX15 encodes a transcription factor that is important in bone development and adipose biology. Manual curation of 692 GWAS-derived variants gave eight strong candidates for causal SNPs that modulate TBX15 transcription in subcutaneous adipose tissue (SAT) or osteoblasts, which highly and specifically express this gene. None of these SNPs were prioritized by Bayesian fine-mapping. The eight regulatory causal SNPs were in enhancer or promoter chromatin seen preferentially in SAT or osteoblasts at TBX15 intron-1 or upstream. They overlap strongly predicted, allele-specific transcription factor binding sites. Our analysis suggests that these SNPs act independently of two missense SNPs in TBX15. Remarkably, five of the regulatory SNPs were associated with eBMD and obesity and had the same trait-increasing allele for both. We found that WARS2 obesity-related SNPs can be ascribed to high linkage disequilibrium with TBX15 intron-1 SNPs. Our findings from GWAS index, proxy, and imputed SNPs suggest that a few SNPs, including three in a 0.7-kb cluster, act as causal regulatory variants to fine-tune TBX15 expression and, thereby, affect both obesity and osteoporosis risk.
Collapse
Affiliation(s)
- Xiao Zhang
- Tulane Center for Bioinformatics and Genomics, Department of Biostatistics and Data Science, School of Public Health and Tropical Medicine, Tulane University , New Orleans, LA, USA
| | - Kenneth C Ehrlich
- Tulane Center for Bioinformatics and Genomics, Department of Biostatistics and Data Science, School of Public Health and Tropical Medicine, Tulane University , New Orleans, LA, USA
| | - Fangtang Yu
- Tulane Center for Bioinformatics and Genomics, Department of Biostatistics and Data Science, School of Public Health and Tropical Medicine, Tulane University , New Orleans, LA, USA
| | - Xiaojun Hu
- Tulane Center for Bioinformatics and Genomics, Department of Biostatistics and Data Science, School of Public Health and Tropical Medicine, Tulane University , New Orleans, LA, USA.,Department of Orthopedics, People's Hospital of Rongchang District , Chongqing, China
| | - Xiang-He Meng
- Laboratory of Molecular and Statistical Genetics, College of Life Sciences, Hunan Normal University , Changsha, Hunan, China
| | - Hong-Wen Deng
- Tulane Center for Bioinformatics and Genomics, Department of Biostatistics and Data Science, School of Public Health and Tropical Medicine, Tulane University , New Orleans, LA, USA
| | - Hui Shen
- Tulane Center for Bioinformatics and Genomics, Department of Biostatistics and Data Science, School of Public Health and Tropical Medicine, Tulane University , New Orleans, LA, USA
| | - Melanie Ehrlich
- Tulane Center for Bioinformatics and Genomics, Department of Biostatistics and Data Science, School of Public Health and Tropical Medicine, Tulane University , New Orleans, LA, USA.,Tulane Cancer Center, Hayward Human Genetics Program, Tulane University Health Sciences , New Orleans, LA, USA
| |
Collapse
|
404
|
Buemann B, Uvnäs-Moberg K. Oxytocin may have a therapeutical potential against cardiovascular disease. Possible pharmaceutical and behavioral approaches. Med Hypotheses 2020; 138:109597. [PMID: 32032912 DOI: 10.1016/j.mehy.2020.109597] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2019] [Revised: 01/11/2020] [Accepted: 01/22/2020] [Indexed: 12/22/2022]
Abstract
Based on the ancient role of oxytocin and its homologues as amplifiers of reproduction we argue for an evolutionary coupling of oxytocin to signaling pathway which support restorative mechanisms of cells and tissue. In particular, the survival and function of different categories of stem cells and primordial cells are enhanced by mitogen-activated protein kinase (MAPK) pathways. Furthermore, oxytocin stimulates the AMP-activated protein kinase pathway (AMPK) in numerous of cell types which promotes the maintenance of different cell structures. This involves autophagic processes and, in particular, may support the renewal of mitochondria. Mitochondrial fitness may protect against oxidative and inflammatory stress - a well-documented effect of oxytocin. The combined specific trophic and protective effects oxytocin may delay several degenerative phenomena including sarcopenia, type-2 diabetes and atherosclerosis. These effects may be exerted both on a central level supporting the function and integrity of the hypothalamus and peripherally acting directly on blood vessels, pancreas, heart, skeletal muscles and adipose tissue etc. Furthermore, in the capacity of being both a hormone and neuromodulator, oxytocin interacts with numerous of regulatory mechanisms particularly the autonomic nervous system and HPA-axis which may reduce blood pressure and affect the immune function. The potential of the oxytocin system as a behavioral and molecular target for the prevention and treatment of cardiovascular disease is discussed. Focus is put on the affiliative and sexual significance and the different options and limitations associated with a pharmaceutical approach. MeSH: Aging, Atherosclerosis, Heart, Hypothalamus, Inflammation, Love, Orgasm, Oxytocin.
Collapse
Affiliation(s)
| | - Kerstin Uvnäs-Moberg
- Department of Animal Environment and Health, Swedish University of Agricultural Sciences, Skara, Sweden
| |
Collapse
|
405
|
Caseiro A, Rocha C, Silva AM, Ferreira C, Silva I, Clemente M, Cipriano I, Saraiva M, Barreira R, Azenha J, Loureiro MH, Martins A, Pereira T. Effects of A Personalized Intervention Program on the Biochemical and Hematological Profile in Community Dwelling Old Adults-The AGA@4life Intervention Model. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2020; 17:ijerph17030718. [PMID: 31979121 PMCID: PMC7038043 DOI: 10.3390/ijerph17030718] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/29/2019] [Revised: 01/13/2020] [Accepted: 01/15/2020] [Indexed: 12/20/2022]
Abstract
Aging is a social and economic challenge of the highest importance and a multidisciplinary intervention seems to be a promising approach for improving the quality of life of elderly individuals. This project was designed aimed at promoting an active and healthy aging through the implementation of an intervention program based on the comprehensive geriatric assessment model (AGA@4life), focused on promoting health and wellbeing, independence and autonomy, mobility, and social inclusion. A non-randomized interventional study was designed to evaluate the effect of only a dietetic and nutritional approach (control group (CG)) and the combination of a tailored exercise program and a dietetic and nutritional approach (intervention group (IG)) in the biochemical and hematological profile of older adults in the framework of AGA@4life. The 34 participants enrolled, aged 65 years or over, were subject to a thorough baseline (T0) multidisciplinary diagnostic evaluation, including the gathering of clinical information and a battery of biochemical and hematological determinations, and reevaluated after eight weeks of intervention (T1). Between T0 and T1, an increase in albumin and total proteins serum levels were observed in both groups (p < 0.01); the hematological profile in CG and IG showed an increase in red cell count and hemoglobin (p < 0.05). In IG, an increase of HDL cholesterol (p < 0.001) and a decrease of triglycerides (p = 0.001) were still observed. The AGA@4life multidisciplinary intervention improved the hematological and biochemical profile of old adults, potentially contributing to delay the development of several aging comorbidities and increase the quality of life of participants.
Collapse
Affiliation(s)
- Armando Caseiro
- Politécnico de Coimbra, ESTESC, Ciências Biomédicas Laboratoriais, Rua 5 de Outubro, 3046-854 Coimbra, Portugal; (A.M.S.); (C.F.); (I.S.); (M.C.); (R.B.)
- LABINSAÚDE—Laboratório de Investigação em Ciências Aplicadas à Saúde, Instituto Politécnico de Coimbra, ESTeSC, Rua 5 de Outubro, 3046-854 Coimbra, Portugal; (M.H.L.); (A.M.); (T.P.)
- Unidade I&D Química-Física Molecular, Faculdade de Ciências e Tecnologia, Universidade de Coimbra, 3004-535 Coimbra, Portugal
- Correspondence: ; Tel.: +351-2398-02430
| | - Clara Rocha
- Politécnico de Coimbra, ESTESC, Ciências Complementares, Rua 5 de Outubro, 3046-854 Coimbra, Portugal;
- INESC Coimbra, Department of Electrical and Computer Engineering, University of Coimbra, Polo 2, 3030-290 Coimbra, Portugal
| | - Ana Margarida Silva
- Politécnico de Coimbra, ESTESC, Ciências Biomédicas Laboratoriais, Rua 5 de Outubro, 3046-854 Coimbra, Portugal; (A.M.S.); (C.F.); (I.S.); (M.C.); (R.B.)
| | - Carla Ferreira
- Politécnico de Coimbra, ESTESC, Ciências Biomédicas Laboratoriais, Rua 5 de Outubro, 3046-854 Coimbra, Portugal; (A.M.S.); (C.F.); (I.S.); (M.C.); (R.B.)
| | - Isabel Silva
- Politécnico de Coimbra, ESTESC, Ciências Biomédicas Laboratoriais, Rua 5 de Outubro, 3046-854 Coimbra, Portugal; (A.M.S.); (C.F.); (I.S.); (M.C.); (R.B.)
| | - Mariana Clemente
- Politécnico de Coimbra, ESTESC, Ciências Biomédicas Laboratoriais, Rua 5 de Outubro, 3046-854 Coimbra, Portugal; (A.M.S.); (C.F.); (I.S.); (M.C.); (R.B.)
| | - Inês Cipriano
- Politécnico de Coimbra, ESTESC, Fisiologia Clínica, Rua 5 de Outubro, 3046-854 Coimbra, Portugal;
| | - Marina Saraiva
- Politécnico de Coimbra, ESTESC, Fisioterapia, Rua 5 de Outubro, 3046-854 Coimbra, Portugal;
| | - Rogério Barreira
- Politécnico de Coimbra, ESTESC, Ciências Biomédicas Laboratoriais, Rua 5 de Outubro, 3046-854 Coimbra, Portugal; (A.M.S.); (C.F.); (I.S.); (M.C.); (R.B.)
- Serviço de Sangue e Medicina Transfusional, Centro Hospitalar e Universitário de Coimbra, 3004-561 Coimbra, Portugal
| | - Joana Azenha
- Politécnico de Coimbra, ESTESC, Dietética e Nutrição, Rua 5 de Outubro, 3046-854 Coimbra, Portugal;
| | - Maria Helena Loureiro
- LABINSAÚDE—Laboratório de Investigação em Ciências Aplicadas à Saúde, Instituto Politécnico de Coimbra, ESTeSC, Rua 5 de Outubro, 3046-854 Coimbra, Portugal; (M.H.L.); (A.M.); (T.P.)
- Politécnico de Coimbra, ESTESC, Dietética e Nutrição, Rua 5 de Outubro, 3046-854 Coimbra, Portugal;
| | - Anabela Martins
- LABINSAÚDE—Laboratório de Investigação em Ciências Aplicadas à Saúde, Instituto Politécnico de Coimbra, ESTeSC, Rua 5 de Outubro, 3046-854 Coimbra, Portugal; (M.H.L.); (A.M.); (T.P.)
- Politécnico de Coimbra, ESTESC, Fisioterapia, Rua 5 de Outubro, 3046-854 Coimbra, Portugal;
| | - Telmo Pereira
- LABINSAÚDE—Laboratório de Investigação em Ciências Aplicadas à Saúde, Instituto Politécnico de Coimbra, ESTeSC, Rua 5 de Outubro, 3046-854 Coimbra, Portugal; (M.H.L.); (A.M.); (T.P.)
- Politécnico de Coimbra, ESTESC, Fisiologia Clínica, Rua 5 de Outubro, 3046-854 Coimbra, Portugal;
| |
Collapse
|
406
|
Guaraldi G, Milic J. Why Am I Getting Fat? Exploring Immune-Metabolic Pathways to Central Fat Accumulation in Persons With HIV. J Infect Dis 2020; 221:343-345. [PMID: 31538199 DOI: 10.1093/infdis/jiz466] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2019] [Accepted: 09/09/2019] [Indexed: 11/12/2022] Open
Affiliation(s)
- Giovanni Guaraldi
- Department of Surgical, Medical, Dental and Morphological Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Jovana Milic
- Department of Surgical, Medical, Dental and Morphological Sciences, University of Modena and Reggio Emilia, Modena, Italy.,Clinical and Experimental Medicine PhD Program, University of Modena and Reggio Emilia, Modena, Italy
| |
Collapse
|
407
|
Feng J, Xu H, Pan F, Hu J, Wu Y, Lin N, Zhang X, Ji C, Hu Y, Zhong H, Yan L, Zhong T, Cui X. An Integrated Analysis of mRNA and lncRNA Expression Profiles Indicates Their Potential Contribution to Brown Fat Dysfunction With Aging. Front Endocrinol (Lausanne) 2020; 11:46. [PMID: 32127793 PMCID: PMC7039067 DOI: 10.3389/fendo.2020.00046] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/03/2019] [Accepted: 01/27/2020] [Indexed: 12/16/2022] Open
Abstract
Brown adipose tissue (BAT) can convert fatty acids and glucose into heat, exhibiting the potential to combat obesity and diabetes. The mass and activity of BAT gradually diminishes with aging. As a newly found regulator of gene expression, long non-coding RNAs (lncRNAs) exhibit a wide range of functions in life processes. However, whether long non-coding RNA (lncRNA) involves in BAT dysfunction with aging is still unclear. Here, using RNA-sequencing technology, we identified 3237 messenger RNAs (mRNAs) and 1312 lncRNAs as differentially expressed in BAT of 10-months-old mice compared with 6- to 8-week-old. The protein-protein interaction network and k-score analysis revealed that the core mRNAs were associated with two important aging-related pathways, including cell cycle and p53 signaling pathway. Gene set enrichment analysis indicated that these mRNAs might participate in lipid metabolism and brown fat dysfunction. Functional enrichment analyses demonstrated that dysregulated lncRNAs were associated with mitochondria, regulation of cellular senescence, cell cycle, metabolic and p53 signaling pathways. Moreover, we revealed that two lncRNAs (NONMMUT024512 and n281160) may involve in the regulation of their adjacent gene peroxisome proliferator-activated receptor alpha (Pparα), a thermogenesis regulator. Collectively, these results lay a foundation for extensive studies on the role of lncRNAs in age-related thermogenic degradation.
Collapse
Affiliation(s)
- Jie Feng
- Nanjing Maternity and Child Health Care Hospital, Women's Hospital of Nanjing Medical University, Nanjing, China
- Jiangsu Institute of Planned Parenthood Research, Nanjing, China
| | - Haoqin Xu
- Jiangsu Institute of Planned Parenthood Research, Nanjing, China
| | - Fenghui Pan
- Department of Geriatrics, Drum Tower Hospital Affiliated to Nanjing University Medical School, Nanjing, China
| | - Jiaojiao Hu
- Nanjing Maternity and Child Health Care Hospital, Women's Hospital of Nanjing Medical University, Nanjing, China
| | - Yulin Wu
- Jiangsu Institute of Planned Parenthood Research, Nanjing, China
| | - Ning Lin
- Jiangsu Institute of Planned Parenthood Research, Nanjing, China
| | - Xiaoxiao Zhang
- Nanjing Maternity and Child Health Care Hospital, Women's Hospital of Nanjing Medical University, Nanjing, China
| | - Chenbo Ji
- Nanjing Maternity and Child Health Care Hospital, Women's Hospital of Nanjing Medical University, Nanjing, China
| | - Yun Hu
- Department of Geriatrics, Drum Tower Hospital Affiliated to Nanjing University Medical School, Nanjing, China
| | - Hong Zhong
- Nanjing Maternity and Child Health Care Hospital, Women's Hospital of Nanjing Medical University, Nanjing, China
| | - Linping Yan
- Nanjing Maternity and Child Health Care Hospital, Women's Hospital of Nanjing Medical University, Nanjing, China
| | - Tianying Zhong
- Nanjing Maternity and Child Health Care Hospital, Women's Hospital of Nanjing Medical University, Nanjing, China
- *Correspondence: Tianying Zhong
| | - Xianwei Cui
- Nanjing Maternity and Child Health Care Hospital, Women's Hospital of Nanjing Medical University, Nanjing, China
- Xianwei Cui
| |
Collapse
|
408
|
Mau T, O’Brien M, Ghosh AK, Miller RA, Yung R. Life-span Extension Drug Interventions Affect Adipose Tissue Inflammation in Aging. J Gerontol A Biol Sci Med Sci 2020; 75:89-98. [PMID: 31353414 PMCID: PMC6909899 DOI: 10.1093/gerona/glz177] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2019] [Indexed: 02/02/2023] Open
Abstract
The National Institute on Aging (NIA)-sponsored Interventions Testing Program (ITP) has identified a number of dietary drug interventions that significantly extend life span, including rapamycin, acarbose, and 17-α estradiol. However, these drugs have diverse downstream targets, and their effects on age-associated organ-specific changes are unclear (Nadon NL, Strong R, Miller RA, Harrison DE. NIA Interventions Testing Program: investigating putative aging intervention agents in a genetically heterogeneous mouse model. EBioMedicine. 2017;21:3-4. doi:10.1016/j.ebiom.2016.11.038). Potential mechanisms by which these drugs extend life could be through their effect on inflammatory processes often noted in tissues of aging mice and humans. Our study focuses on the effects of three drugs in the ITP on inflammation in gonadal white adipose tissue (gWAT) of HET3 mice-including adiposity, adipose tissue macrophage (ATM) M1/M2 polarization, markers of cellular senescence, and endoplasmic reticulum stress. We found that rapamycin led to a 56% increase of CD45+ leukocytes in gWAT, where the majority of these are ATMs. Interestingly, rapamycin led to a 217% and 106% increase of M1 (CD45+CD64+CD206-) ATMs in females and males, respectively. Our data suggest rapamycin may achieve life-span extension in part through adipose tissue inflammation. Additionally, HET3 mice exhibit a spectrum of age-associated changes in the gWAT, but acarbose and 17-α estradiol do not strongly alter these phenotypes-suggesting that acarbose and 17- α estradiol may not influence life span through mechanisms involving adipose tissue inflammation.
Collapse
Affiliation(s)
- Theresa Mau
- Division of Geriatric and Palliative Medicine, Department of Internal Medicine, University of Michigan, Ann Arbor
- Graduate Program in Immunology, Program in Biomedical Sciences (PIBS), University of Michigan, Ann Arbor
| | - Martin O’Brien
- Division of Geriatric and Palliative Medicine, Department of Internal Medicine, University of Michigan, Ann Arbor
| | - Amiya K Ghosh
- Division of Geriatric and Palliative Medicine, Department of Internal Medicine, University of Michigan, Ann Arbor
| | - Richard A Miller
- Department of Pathology and Glenn Center for Biology of Aging Research, University of Michigan, Ann Arbor
| | - Raymond Yung
- Division of Geriatric and Palliative Medicine, Department of Internal Medicine, University of Michigan, Ann Arbor
- Graduate Program in Immunology, Program in Biomedical Sciences (PIBS), University of Michigan, Ann Arbor
- Department of Pathology and Glenn Center for Biology of Aging Research, University of Michigan, Ann Arbor
- Geriatric Research, Education, and Clinical Care Center (GRECC), VA Ann Arbor Health System, Michigan
| |
Collapse
|
409
|
Khan S, Chan YT, Revelo XS, Winer DA. The Immune Landscape of Visceral Adipose Tissue During Obesity and Aging. Front Endocrinol (Lausanne) 2020; 11:267. [PMID: 32499756 PMCID: PMC7243349 DOI: 10.3389/fendo.2020.00267] [Citation(s) in RCA: 69] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/16/2020] [Accepted: 04/14/2020] [Indexed: 12/16/2022] Open
Abstract
Obesity and aging represent major health burdens to the global adult population. Both conditions promote the development of associated metabolic diseases such as insulin resistance. The visceral adipose tissue (VAT) is a site that becomes dysfunctional during obesity and aging, and plays a significant role during their pathophysiology. The changes in obese and aging VAT are now recognized to be partly driven by a chronic local inflammatory state, characterized by immune cells that typically adopt an inflammatory phenotype during metabolic disease. Here, we summarize the current knowledge on the immune cell landscape of the VAT during lean, obese, and aged conditions, highlighting their similarities and differences. We also briefly discuss possible linked mechanisms that fuel obesity- and age-associated VAT dysfunction.
Collapse
Affiliation(s)
- Saad Khan
- Department of Immunology, University of Toronto, Toronto, ON, Canada
- Division of Cellular & Molecular Biology, Diabetes Research Group, Toronto General Hospital Research Institute (TGHRI), University Health Network, Toronto, ON, Canada
| | - Yi Tao Chan
- Department of Immunology, University of Toronto, Toronto, ON, Canada
- Division of Cellular & Molecular Biology, Diabetes Research Group, Toronto General Hospital Research Institute (TGHRI), University Health Network, Toronto, ON, Canada
| | - Xavier S. Revelo
- Center for Immunology, University of Minnesota, Minneapolis, MN, United States
- Department of Integrative Biology and Physiology, University of Minnesota, Minneapolis, MN, United States
- *Correspondence: Xavier S. Revelo
| | - Daniel A. Winer
- Department of Immunology, University of Toronto, Toronto, ON, Canada
- Division of Cellular & Molecular Biology, Diabetes Research Group, Toronto General Hospital Research Institute (TGHRI), University Health Network, Toronto, ON, Canada
- Department of Pathology, University Health Network, Toronto, ON, Canada
- Buck Institute for Research on Aging, Novato, CA, United States
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
- Daniel A. Winer
| |
Collapse
|
410
|
Papatheodoridi AM, Chrysavgis L, Koutsilieris M, Chatzigeorgiou A. The Role of Senescence in the Development of Nonalcoholic Fatty Liver Disease and Progression to Nonalcoholic Steatohepatitis. Hepatology 2020; 71:363-374. [PMID: 31230380 DOI: 10.1002/hep.30834] [Citation(s) in RCA: 144] [Impact Index Per Article: 28.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/26/2019] [Accepted: 06/17/2019] [Indexed: 12/17/2022]
Abstract
In recent years, cellular senescence has generated a lot of interest among researchers because of its involvement in both the normal aging process and common human diseases. During senescence, cells undergo alterations that include telomere shortening, nuclear area enlargement, and genomic and mitochondrial DNA damage, leading to irreversible cell cycle arrest, and secretion of proinflammatory cytokines. Evidence suggests that the complex process of senescence is involved in the development of a plethora of chronic diseases including metabolic and inflammatory disorders and tumorigenesis. Recently, several human and animal studies have emphasized the involvement of senescence in the pathogenesis and development of liver steatosis including the progression to nonalcoholic steatohepatitis (NASH) as characterized by the additional emergence of inflammation, hepatocyte ballooning, and liver fibrosis. The development of nonalcoholic fatty liver disease (NAFLD) and its progression to NASH are commonly accompanied by several pathophysiological events including metabolic dysregulation and inflammatory phenomena occurring within the liver that may contribute to or derive from cellular senescence, implying that the latter may be both a stimulus and a consequence of the disease. Conclusion: In this review, we summarize the current literature on the impact of cellular senescence in NAFLD/NASH and discuss the effectiveness and safety of novel senolytic drugs and therapeutic options available to delay or treat the disease. Finally, we identify the open questions and issues to be addressed in the near future.
Collapse
Affiliation(s)
| | - Lampros Chrysavgis
- Department of Physiology, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Michael Koutsilieris
- Department of Physiology, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Antonios Chatzigeorgiou
- Department of Physiology, Medical School, National and Kapodistrian University of Athens, Athens, Greece
- Institute for Clinical Chemistry and Laboratory Medicine, University Hospital and Faculty of Medicine Carl Gustav Carus of TU Dresden, Dresden, Germany
| |
Collapse
|
411
|
Merritt CE. Comparison of 14 body size variables on adult skeletal age methods: how body mass, BMI, fat and muscle influence age estimations. Ann Hum Biol 2019; 47:32-41. [PMID: 31868026 DOI: 10.1080/03014460.2019.1704872] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Background: Skeletal age estimations consistently under-age individuals with low body mass and over-age individuals with high body mass.Aim: To assess the directional biases of body size variables, including fat and muscle tissue, on skeletal age estimates.Subjects and methods: CT scans of 420 age- and size-selected cadavers were assessed using the fourth rib and pubic symphysis age estimation methods. MANCOVA was used to test the effects of stature, body mass, circumference, and fat and muscle tissue area on age estimation biases.Results: For both methods, there were biases based on body mass, BMI, circumference and fat tissue area. Individuals with the lowest measurements for these variables were under-aged while individuals with the highest measurements were over-aged. Age estimation biases were not affected by stature, muscle area, or skeletal measurements of bi-iliac breadth, femoral head diameter, or femoral length.Conclusion: Increased body mass, and specifically fat tissue, accelerates skeletal ageing. Increased muscle area generally did not show a protective effect on skeletal ageing. The accuracy of age estimations would be improved by factoring in body mass. However, femoral head diameter and bi-iliac breadth are not good proxies for body mass. Osteological markers of obesity offer a promising new approach.
Collapse
Affiliation(s)
- Catherine E Merritt
- Centre for Anatomy and Human Identification, University of Dundee, Dundee, UK
| |
Collapse
|
412
|
Bourgeois C, Gorwood J, Barrail-Tran A, Lagathu C, Capeau J, Desjardins D, Le Grand R, Damouche A, Béréziat V, Lambotte O. Specific Biological Features of Adipose Tissue, and Their Impact on HIV Persistence. Front Microbiol 2019; 10:2837. [PMID: 31921023 PMCID: PMC6927940 DOI: 10.3389/fmicb.2019.02837] [Citation(s) in RCA: 60] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2019] [Accepted: 11/22/2019] [Indexed: 12/19/2022] Open
Abstract
Although white AT can contribute to anti-infectious immune responses, it can also be targeted and perturbed by pathogens. The AT's immune involvement is primarily due to strong pro-inflammatory responses (with both local and paracrine effects), and the large number of fat-resident macrophages. Adipocytes also exert direct antimicrobial responses. In recent years, it has been found that memory T cells accumulate in AT, where they provide efficient secondary responses against viral pathogens. These observations have prompted researchers to re-evaluate the links between obesity and susceptibility to infections. In contrast, AT serves as a reservoir for several persistence pathogens, such as human adenovirus Ad-36, Trypanosoma gondii, Mycobacterium tuberculosis, influenza A virus, and cytomegalovirus (CMV). The presence and persistence of bacterial DNA in AT has led to the concept of a tissue-specific microbiota. The unexpected coexistence of immune cells and pathogens within the specific AT environment is intriguing, and its impact on anti-infectious immune responses requires further evaluation. AT has been recently identified as a site of HIV persistence. In the context of HIV infection, AT is targeted by both the virus and the antiretroviral drugs. AT's intrinsic metabolic features, large overall mass, and wide distribution make it a major tissue reservoir, and one that may contribute to the pathophysiology of chronic HIV infections. Here, we review the immune, metabolic, viral, and pharmacological aspects that contribute to HIV persistence in AT. We also evaluate the respective impacts of both intrinsic and HIV-induced factors on AT's involvement as a viral reservoir. Lastly, we examine the potential consequences of HIV persistence on the metabolic and immune activities of AT.
Collapse
Affiliation(s)
- Christine Bourgeois
- Center for Immunology of Viral Infections and Autoimmune Diseases, IDMIT Department, IBFJ, CEA, Université Paris Sud, INSERM U1184, Fontenay-aux-Roses, France
| | - Jennifer Gorwood
- INSERM UMR_S 938, Centre de Recherche Saint-Antoine, Institut Hospitalo-Universitaire de Cardio-Métabolisme et Nutrition (ICAN), Sorbonne Université, Paris, France
| | - Aurélie Barrail-Tran
- Center for Immunology of Viral Infections and Autoimmune Diseases, IDMIT Department, IBFJ, CEA, Université Paris Sud, INSERM U1184, Fontenay-aux-Roses, France
- AP-HP, Service de Médecine Interne et Immunologie Clinique, Hôpital Bicêtre, Groupe Hospitalier Universitaire Paris Sud, Le Kremlin-Bicêtre, France
| | - Claire Lagathu
- INSERM UMR_S 938, Centre de Recherche Saint-Antoine, Institut Hospitalo-Universitaire de Cardio-Métabolisme et Nutrition (ICAN), Sorbonne Université, Paris, France
| | - Jacqueline Capeau
- INSERM UMR_S 938, Centre de Recherche Saint-Antoine, Institut Hospitalo-Universitaire de Cardio-Métabolisme et Nutrition (ICAN), Sorbonne Université, Paris, France
| | - Delphine Desjardins
- Center for Immunology of Viral Infections and Autoimmune Diseases, IDMIT Department, IBFJ, CEA, Université Paris Sud, INSERM U1184, Fontenay-aux-Roses, France
| | - Roger Le Grand
- Center for Immunology of Viral Infections and Autoimmune Diseases, IDMIT Department, IBFJ, CEA, Université Paris Sud, INSERM U1184, Fontenay-aux-Roses, France
| | - Abderaouf Damouche
- Center for Immunology of Viral Infections and Autoimmune Diseases, IDMIT Department, IBFJ, CEA, Université Paris Sud, INSERM U1184, Fontenay-aux-Roses, France
| | - Véronique Béréziat
- INSERM UMR_S 938, Centre de Recherche Saint-Antoine, Institut Hospitalo-Universitaire de Cardio-Métabolisme et Nutrition (ICAN), Sorbonne Université, Paris, France
| | - Olivier Lambotte
- Center for Immunology of Viral Infections and Autoimmune Diseases, IDMIT Department, IBFJ, CEA, Université Paris Sud, INSERM U1184, Fontenay-aux-Roses, France
- AP-HP, Service de Médecine Interne et Immunologie Clinique, Hôpital Bicêtre, Groupe Hospitalier Universitaire Paris Sud, Le Kremlin-Bicêtre, France
| |
Collapse
|
413
|
O'Hara SP, Splinter PL, Trussoni CE, Guicciardi ME, Splinter NP, Al Suraih MS, Nasser-Ghodsi N, Stollenwerk D, Gores GJ, LaRusso NF. The transcription factor ETS1 promotes apoptosis resistance of senescent cholangiocytes by epigenetically up-regulating the apoptosis suppressor BCL2L1. J Biol Chem 2019; 294:18698-18713. [PMID: 31659122 PMCID: PMC6901313 DOI: 10.1074/jbc.ra119.010176] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2019] [Revised: 10/15/2019] [Indexed: 12/21/2022] Open
Abstract
Primary sclerosing cholangitis (PSC) is an idiopathic, progressive cholangiopathy. Cholangiocyte senescence is important in PSC pathogenesis, and we have previously reported that senescence is regulated by the transcription factor ETS proto-oncogene 1 (ETS1) and associated with overexpression of BCL2 like 1 (BCL2L1 or BCL-xL), an anti-apoptotic BCL2-family member. Here, we further explored the mechanisms regulating BCL-xL-mediated, apoptosis resistance in senescent cholangiocytes and uncovered that ETS1 and the histone acetyltransferase E1A-binding protein P300 (EP300 or p300) both promote BCL-xL transcription. Using immunofluorescence, we found that BCL-xL protein expression is increased both in cholangiocytes of livers from individuals with PSC and a mouse model of PSC. Using an in vitro model of lipopolysaccharide-induced senescence in normal human cholangiocytes (NHCs), we found increased BCL-xL mRNA and protein levels, and ChIP-PCRs indicated increased occupancy of ETS1, p300, and histone 3 Lys-27 acetylation (H3K27Ac) at the BCL-xL promoter. Using co-immunoprecipitation and proximity ligation assays, we further demonstrate that ETS1 and p300 physically interact in senescent but not control NHCs. Additionally, mutagenesis of predicted ETS1-binding sites within the BCL-xL promoter blocked luciferase reporter activity, and CRISPR/Cas9-mediated genetic deletion of ETS1 reduced senescence-associated BCL-xL expression. In senescent NHCs, TRAIL-mediated apoptosis was reduced ∼70%, and ETS1 deletion or RNAi-mediated BCL-xL suppression increased apoptosis. Overall, our results suggest that ETS1 and p300 promote senescent cholangiocyte resistance to apoptosis by modifying chromatin and inducing BCL-xL expression. These findings reveal ETS1 as a central regulator of both cholangiocyte senescence and the associated apoptosis-resistant phenotype.
Collapse
Affiliation(s)
- Steven P O'Hara
- Division of Gastroenterology and Hepatology and the Mayo Clinic Center for Cell Signaling in Gastroenterology, Mayo Clinic, Rochester, Minnesota 55905.
| | - Patrick L Splinter
- Division of Gastroenterology and Hepatology and the Mayo Clinic Center for Cell Signaling in Gastroenterology, Mayo Clinic, Rochester, Minnesota 55905
| | - Christy E Trussoni
- Division of Gastroenterology and Hepatology and the Mayo Clinic Center for Cell Signaling in Gastroenterology, Mayo Clinic, Rochester, Minnesota 55905
| | - Maria Eugenia Guicciardi
- Division of Gastroenterology and Hepatology and the Mayo Clinic Center for Cell Signaling in Gastroenterology, Mayo Clinic, Rochester, Minnesota 55905
| | - Noah P Splinter
- Division of Gastroenterology and Hepatology and the Mayo Clinic Center for Cell Signaling in Gastroenterology, Mayo Clinic, Rochester, Minnesota 55905
| | - Mohammed S Al Suraih
- Division of Gastroenterology and Hepatology and the Mayo Clinic Center for Cell Signaling in Gastroenterology, Mayo Clinic, Rochester, Minnesota 55905
| | - Navine Nasser-Ghodsi
- Division of Gastroenterology and Hepatology and the Mayo Clinic Center for Cell Signaling in Gastroenterology, Mayo Clinic, Rochester, Minnesota 55905
| | - Deborah Stollenwerk
- Division of Gastroenterology and Hepatology and the Mayo Clinic Center for Cell Signaling in Gastroenterology, Mayo Clinic, Rochester, Minnesota 55905
| | - Gregory J Gores
- Division of Gastroenterology and Hepatology and the Mayo Clinic Center for Cell Signaling in Gastroenterology, Mayo Clinic, Rochester, Minnesota 55905
| | - Nicholas F LaRusso
- Division of Gastroenterology and Hepatology and the Mayo Clinic Center for Cell Signaling in Gastroenterology, Mayo Clinic, Rochester, Minnesota 55905
| |
Collapse
|
414
|
de Toro-Martín J, Guénard F, Tchernof A, Hould FS, Lebel S, Julien F, Marceau S, Vohl MC. Body mass index is associated with epigenetic age acceleration in the visceral adipose tissue of subjects with severe obesity. Clin Epigenetics 2019; 11:172. [PMID: 31791395 PMCID: PMC6888904 DOI: 10.1186/s13148-019-0754-6] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2019] [Accepted: 09/23/2019] [Indexed: 12/19/2022] Open
Abstract
Background There is solid evidence that obesity induces the acceleration of liver epigenetic aging. However, unlike easily accessible blood or subcutaneous adipose tissue, little is known about the impact of obesity on epigenetic aging of metabolically active visceral adipose tissue (VAT). Herein, we aimed to test whether obesity accelerates VAT epigenetic aging in subjects with severe obesity. Results A significant and positive correlation between chronological age and epigenetic age, estimated with a reduced version of the Horvath’s epigenetic clock, was found in both blood (r = 0.78, p = 9.4 × 10−12) and VAT (r = 0.80, p = 1.1 × 10−12). Epigenetic age acceleration, defined as the residual resulting from regressing epigenetic age on chronological age, was significantly correlated with body mass index (BMI) in VAT (r = 0.29, p = 0.037). Multivariate linear regression analysis showed that, after adjusting for chronological age, sex and metabolic syndrome status, BMI remained significantly associated with epigenetic age acceleration in VAT (beta = 0.15, p = 0.035), equivalent to 2.3 years for each 10 BMI units. Binomial logistic regression showed that BMI-adjusted epigenetic age acceleration in VAT was significantly associated with a higher loss of excess body weight following biliopancreatic diversion with duodenal switch surgery (odds ratio = 1.21; 95% CI = 1.04–1.48; p = 0.03). Conclusions Epigenetic age acceleration increases with BMI in VAT, but not in blood, as previously reported in liver. These results suggest that obesity is associated with epigenetic age acceleration of metabolically active tissues. Further studies that deepen the physiological relevance of VAT epigenetic aging will help to better understand the onset of metabolic syndrome and weight loss dynamics following bariatric surgery.
Collapse
Affiliation(s)
- Juan de Toro-Martín
- Institute of Nutrition and Functional Foods (INAF), Université Laval, Pavillon des Services (2729 K), 2440, boul. Hochelaga, Quebec, QC, G1V 0A6, Canada.,School of Nutrition, Université Laval, Quebec, QC, Canada
| | - Frédéric Guénard
- Institute of Nutrition and Functional Foods (INAF), Université Laval, Pavillon des Services (2729 K), 2440, boul. Hochelaga, Quebec, QC, G1V 0A6, Canada.,School of Nutrition, Université Laval, Quebec, QC, Canada
| | - André Tchernof
- School of Nutrition, Université Laval, Quebec, QC, Canada.,Quebec Heart and Lung Institute Research Center, Quebec, QC, Canada
| | | | - Stéfane Lebel
- Department of Surgery, Université Laval, Quebec, QC, Canada
| | | | - Simon Marceau
- Department of Surgery, Université Laval, Quebec, QC, Canada
| | - Marie-Claude Vohl
- Institute of Nutrition and Functional Foods (INAF), Université Laval, Pavillon des Services (2729 K), 2440, boul. Hochelaga, Quebec, QC, G1V 0A6, Canada. .,School of Nutrition, Université Laval, Quebec, QC, Canada.
| |
Collapse
|
415
|
Abstract
Adipose tissue is highly dynamic and increases its size dependent on the status of nutrition. Generally, an increase of adipose tissue mass is attributed to two mechanisms, namely hypertrophy (increase in adipocyte size) and hyperplasia (increase in adipocyte number). Here, we analyzed the proliferation capacity of a pool of nutrition sensing preadipocytes after short-term high fat diet (HFD) feeding. We show that this process is age independent and that adipocyte hyperplasia seems not to be dependent on adipocyte hypertrophy. Further, we could show that the subsequent development into adipocytes is influenced by the duration of HFD feeding after proliferation. Our data also demonstrate that the studied pool of preadipocytes seems to be finite and cannot be reactivated by multiple bouts of HFD feeding. In conclusion, our results indicate an important link between stem cells, nutrition status and homeostasis in the epididymal adipose tissue.
Collapse
Affiliation(s)
- Elisabeth Kulenkampff
- Institute of Food Nutrition and Health, Eidgenössische Technische Hochschule Zürich (ETH), Schwerzenbach, Switzerland
| | - Christian Wolfrum
- Institute of Food Nutrition and Health, Eidgenössische Technische Hochschule Zürich (ETH), Schwerzenbach, Switzerland
| |
Collapse
|
416
|
Giovos G, Yavropoulou MP, Yovos JG. The role of cellular senescence in diabetes mellitus and osteoporosis: molecular pathways and potential interventions. Hormones (Athens) 2019; 18:339-351. [PMID: 31701490 DOI: 10.1007/s42000-019-00132-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/01/2019] [Accepted: 09/04/2019] [Indexed: 02/06/2023]
Abstract
The improving effectiveness of health care leads inevitably to a rapid increase in the elderly population worldwide. At advanced ages, however, people experience chronic disabilities, which significantly increase the social and economic burden while curtailing survival, independence, and quality of life of the aging population. As aging is a multifactorial process, apart from genetic predisposition, other environmental factors, such as chronic sterile inflammation and cellular senescence, contribute as crucial participants and have been targeted to reverse their deleterious effects on tissue homeostasis and functional integrity. Cellular senescence refers to the essentially irreversible inhibition of cellular proliferation when cells are subjected to extrinsic or endogenous stress. Although the process of cellular senescence has long been known, recent evidence demonstrated that it characterizes many aging phenotypes and that elimination of senescent cells at the tissue level can improve age-related tissue dysfunction. These observations have renewed scientific interest in possible therapeutic interventions. Two major chronic diseases associated with aging that impose an enormous burden on global health systems are type 2 diabetes and osteoporosis. This review presents current data on (i) the underlying molecular mechanisms of cellular senescence, (ii) its relationship to these two endocrine diseases that are today prevalent worldwide, and (iii) future prospects of targeted intervention with the aim of simultaneously improving the progression and prognosis of these serious problems of aging.
Collapse
Affiliation(s)
- Georgios Giovos
- Clinical Research Fellow in Endocrinology, Wisdem Centre, University Hospitals Coventry & Warwickshire, Coventry, UK
| | - Maria P Yavropoulou
- Endocrinology Unit, 1st Propaedeutic Department of Internal Medicine, National and Kapodistrian University of Athens, UOA, LAIKO General Hospital, 17 Agiou Thoma Str., 11527, Athens, Greece.
| | - John G Yovos
- Professor Emeritus in Internal Medicine and Endocrinology, Aristotle University of Thessaloniki, Thessaloniki, Greece
| |
Collapse
|
417
|
Lefranc C, Friederich-Persson M, Braud L, Palacios-Ramirez R, Karlsson S, Boujardine N, Motterlini R, Jaisser F, Nguyen Dinh Cat A. MR (Mineralocorticoid Receptor) Induces Adipose Tissue Senescence and Mitochondrial Dysfunction Leading to Vascular Dysfunction in Obesity. Hypertension 2019; 73:458-468. [PMID: 30624990 DOI: 10.1161/hypertensionaha.118.11873] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Adipose tissue (AT) senescence and mitochondrial dysfunction are associated with obesity. Studies in obese patients and animals demonstrate that the MR (mineralocorticoid receptor) contributes to obesity-associated cardiovascular complications through its specific role in AT. However, underlying mechanisms remain unclear. This study aims to elucidate whether MR regulates mitochondrial function in obesity, resulting in AT premature aging and vascular dysfunction. Obese (db/db) and lean (db/+) mice were treated with an MR antagonist or a specific mitochondria-targeted antioxidant. Mitochondrial and vascular functions were determined by respirometry and myography, respectively. Molecular mechanisms were probed by Western immunoblotting and real-time polymerase chain reaction in visceral AT and arteries and focused on senescence markers and redox-sensitive pathways. db/db mice displayed AT senescence with activation of the p53-p21 pathway and decreased SIRT (sirtuin) levels, as well as mitochondrial dysfunction. Furthermore, the beneficial anticontractile effects of perivascular AT were lost in db/db via ROCK (Rho kinase) activation. MR blockade prevented these effects. Thus, MR activation in obesity induces mitochondrial dysfunction and AT senescence and dysfunction, which consequently increases vascular contractility. In conclusion, our study identifies novel mechanistic insights involving MR, adipose mitochondria, and vascular function that may be of importance to develop new therapeutic strategies to limit obesity-associated cardiovascular complications.
Collapse
Affiliation(s)
- Clara Lefranc
- From the Department of Physiology, INSERM UMRS 1138 Team 1, Centre de Recherche des Cordeliers, Sorbonne University, Paris, France (C.L., R.P.-R., N.B., F.J., A.N.D.C.)
| | | | - Laura Braud
- Department of Pathophysiology of Cardiovascular and Respiratory Diseases, Development and Senescence, INSERM U955 Team 12, University Paris-Est Creteil, France (L.B., R.M.)
| | - Roberto Palacios-Ramirez
- From the Department of Physiology, INSERM UMRS 1138 Team 1, Centre de Recherche des Cordeliers, Sorbonne University, Paris, France (C.L., R.P.-R., N.B., F.J., A.N.D.C.)
| | - Susanne Karlsson
- Department of Medical Cell Biology, Uppsala University, Sweden (M.F.-P., S.K.)
| | - Nabiha Boujardine
- From the Department of Physiology, INSERM UMRS 1138 Team 1, Centre de Recherche des Cordeliers, Sorbonne University, Paris, France (C.L., R.P.-R., N.B., F.J., A.N.D.C.)
| | - Roberto Motterlini
- Department of Pathophysiology of Cardiovascular and Respiratory Diseases, Development and Senescence, INSERM U955 Team 12, University Paris-Est Creteil, France (L.B., R.M.)
| | - Frederic Jaisser
- From the Department of Physiology, INSERM UMRS 1138 Team 1, Centre de Recherche des Cordeliers, Sorbonne University, Paris, France (C.L., R.P.-R., N.B., F.J., A.N.D.C.)
| | - Aurelie Nguyen Dinh Cat
- From the Department of Physiology, INSERM UMRS 1138 Team 1, Centre de Recherche des Cordeliers, Sorbonne University, Paris, France (C.L., R.P.-R., N.B., F.J., A.N.D.C.)
| |
Collapse
|
418
|
Targeting normal and cancer senescent cells as a strategy of senotherapy. Ageing Res Rev 2019; 55:100941. [PMID: 31408714 DOI: 10.1016/j.arr.2019.100941] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2019] [Revised: 07/04/2019] [Accepted: 08/06/2019] [Indexed: 12/19/2022]
Abstract
Senotherapy is an antiageing strategy. It refers to selective killing of senescent cells by senolytic agents, strengthening the activity of immune cells that eliminate senescent cells or alleviating the secretory phenotype (SASP) of senescent cells. As senescent cells accumulate with age and are considered to be at the root of age-related disorders, senotherapy seems to be very promising in improving healthspan. Genetic approaches, which allowed to selectively induce death of senescent cells in transgenic mice, provided proof-of-concept evidence that elimination of senescent cells can be a therapeutic approach for treating many age-related diseases. Translating these results into humans is based on searching for synthetic and natural compounds, which are able to exert such beneficial effects. The major challenge in the field is to show efficacy, safety and tolerability of senotherapy in humans. The question is how these therapeutics can influence senescence of non-dividing post-mitotic cells. Another issue concerns senescence of cancer cells induced during therapy as there is a risk of resumption of senescent cell division that could terminate in cancer renewal. Thus, development of an effective senotherapeutic strategy is also an urgent issue in cancer treatment. Different aspects, both beneficial and potentially detrimental, will be discussed in this review.
Collapse
|
419
|
Kim SR, Jiang K, Ogrodnik M, Chen X, Zhu XY, Lohmeier H, Ahmed L, Tang H, Tchkonia T, Hickson LJ, Kirkland JL, Lerman LO. Increased renal cellular senescence in murine high-fat diet: effect of the senolytic drug quercetin. Transl Res 2019; 213:112-123. [PMID: 31356770 PMCID: PMC6783353 DOI: 10.1016/j.trsl.2019.07.005] [Citation(s) in RCA: 94] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/20/2018] [Revised: 07/05/2019] [Accepted: 07/11/2019] [Indexed: 12/25/2022]
Abstract
Obesity and dyslipidemia can be associated with cellular senescence, and may impair kidney function. However, whether senescence contributes to renal dysfunction in these conditions remains unclear. Quercetin is an abundant dietary flavonoid that selectively clears inhibiting PI3K/AKT and p53/p21/serpines and inducing apoptosis. We hypothesized that high-fat-diet-induced obesity causes renal senescence, which would be mitigated by quercetin. C57BL/6J mice fed either standard chow or high-fat diets (HFDs) were treated with quercetin (50 mg/kg) or vehicle 5-days biweekly via oral gavage for 10 weeks. Subsequently, renal function was studied in vivo using magnetic resonance imaging, and renal senescence and histology were evaluated ex vivo. Mice fed with a HFD developed obesity and hypercholesterolemia, whereas renal size remained unchanged. Murine obesity impaired renal function and cortical oxygenation, and induced glomerulomegaly. Renal markers of senescence (eg, expression of p16, p19, and p53) and its secretory phenotype were upregulated in the obese hypercholesterolemic compared to lean mice in renal tubular cells, but attenuated in quercetin-treated murine kidneys, as was renal fibrosis. Quercetin treatment also increased renal cortical oxygenation and decreased plasma creatinine levels in obese mice, whereas body weight and cholesterol levels were unaltered. Therefore, murine obesity and dyslipidemia induce renal tissue senescence and impairs kidney function, which is alleviated by chronic senolytic treatment. These findings implicate senescence in loss of kidney function in murine dyslipidemia and obesity, and support further studies of senolytic therapy in obesity.
Collapse
Affiliation(s)
- Seo Rin Kim
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota
| | - Kai Jiang
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota
| | - Mikolaj Ogrodnik
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, Minnesota
| | - Xiaojun Chen
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota
| | - Xiang-Yang Zhu
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota
| | - Hannah Lohmeier
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota
| | - Leena Ahmed
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota
| | - Hui Tang
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota
| | - Tamara Tchkonia
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, Minnesota
| | - LaTonya J Hickson
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota
| | - James L Kirkland
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, Minnesota
| | - Lilach O Lerman
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota.
| |
Collapse
|
420
|
Adipose Tissue Quality in Aging: How Structural and Functional Aspects of Adipose Tissue Impact Skeletal Muscle Quality. Nutrients 2019; 11:nu11112553. [PMID: 31652734 PMCID: PMC6893709 DOI: 10.3390/nu11112553] [Citation(s) in RCA: 60] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2019] [Revised: 10/19/2019] [Accepted: 10/21/2019] [Indexed: 12/20/2022] Open
Abstract
The interplay between adipose tissue and skeletal muscle and the impact on mobility and aging remain enigmatic. The progressive decline in mobility promoted by aging has been previously attributed to the loss of skeletal mass and function and more recently linked to changes in body fat composition and quantity. Regardless of body size, visceral and intermuscular adipose depots increase with aging and are associated with adverse health outcomes. However, the quality of adipose tissue, in particular abdominal subcutaneous as it is the largest depot, likely plays a significant role in aging outcomes, such as mobility decline, though its communication with other tissues such as skeletal muscle. In this review, we discuss the age-associated development of a pro-inflammatory profile, cellular senescence, and metabolic inflexibility in abdominal subcutaneous adipose tissue. Collectively, these facets of adipose tissue quality influence its secretory profile and crosstalk with skeletal muscle and likely contribute to the development of muscle atrophy and disability. Therefore, the identification of the key structural and functional components of adipose tissue quality—including necrosis, senescence, inflammation, self-renewal, metabolic flexibility—and adipose tissue-secreted proteins that influence mobility via direct effects on skeletal muscle are necessary to prevent morbidity/mortality in the aging population.
Collapse
|
421
|
Frasca D, Diaz A, Romero M, Blomberg BB. Leptin induces immunosenescence in human B cells. Cell Immunol 2019; 348:103994. [PMID: 31831137 DOI: 10.1016/j.cellimm.2019.103994] [Citation(s) in RCA: 55] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2019] [Revised: 10/14/2019] [Accepted: 10/15/2019] [Indexed: 11/16/2022]
Abstract
Leptin is an adipokine secreted primarily by the adipocytes. Leptin has endocrine and immune functions and increases the secretion of pro-inflammatory cytokines by immune cells. Here we show that incubation of B cells from young lean individuals with leptin increases the frequencies of pro-inflammatory B cells and induces intrinsic B cell inflammation, characterized by mRNA expression of pro-inflammatory cytokines (TNF-α and IL-6), chemokines (IL-8), micro-RNAs (miR-155 and miR-16), TLR4 and p16, a cell cycle regulator associated with immunosenescence. We have previously shown that the expression of these pro-inflammatory markers in unstimulated B cells is negatively associated with the response of the same B cells after in vivo or in vitro stimulation. B cells from young lean individuals, after in vitro incubation with leptin, show reduced class switch and influenza vaccine-specific IgG production. Our results altogether show that leptin makes B cells from youn lean individuals similar to those from young obese and elderly lean individuals, suggesting that leptin may be a mechanisms of immunosenescence in human B cells.
Collapse
Affiliation(s)
- Daniela Frasca
- Department of Microbiology and Immunology, University of Miami Miller School of Medicine, Miami, FL, USA.
| | - Alain Diaz
- Department of Microbiology and Immunology, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Maria Romero
- Department of Microbiology and Immunology, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Bonnie B Blomberg
- Department of Microbiology and Immunology, University of Miami Miller School of Medicine, Miami, FL, USA; Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL, USA
| |
Collapse
|
422
|
Hohensinner PJ, Kaun C, Ebenbauer B, Hackl M, Demyanets S, Richter D, Prager M, Wojta J, Rega-Kaun G. Reduction of Premature Aging Markers After Gastric Bypass Surgery in Morbidly Obese Patients. Obes Surg 2019; 28:2804-2810. [PMID: 29693219 PMCID: PMC6132736 DOI: 10.1007/s11695-018-3247-3] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Background Obesity is considered to be a major comorbidity. Obese patients suffer from an increased proinflammatory state associated with a premature aging phenotype including increased secretion of senescence-associated secretory proteins (SASP) and reduced telomere length. Micro-ribonucleic acids (miRNAs) are non-coding RNA molecules that could modify the post-transcriptional process. Several studies have reported associations between miRNAs and metabolic unhealthy conditions. Aim To determine if bariatric surgery and the resulting weight loss could reverse the premature aging phenotype. Methods We enrolled 58 morbidly obese patients undergoing bariatric surgery. Markers of premature aging including the SASP IL-6, CRP and PAI-1, 7 miRNAs, as well as telomere length and telomere oxidation in mononuclear cells were evaluated. Results Patients showed a significant drop of body mass index (BMI; 43.98 ± 3.5 versus 28.02 ± 4.1, p < 0.001). We observed a significant reduction in SASP including a reduction of 55% of plasma IL-6 levels (p = 0 < 0.001), 83% of CRP levels (p = 0.001) and 15% of plasma PAI-1 levels (p < 0.001). Telomere length doubled in the patient cohort (p < 0.001) and was accompanied by a reduction in the telomere oxidation index by 70% (p < 0.001). Telomere length was inversely correlated with telomere oxidation. The aging-associated miRNA miR10a_5p was upregulated significantly (p = 0.039), while the other tested miRNAs showed no difference. Conclusion Our data indicate a significant reduction of the proinflammatory SASP after bariatric surgery. We observed an increase in telomere length and reduced oxidative stress at telomeres. miR10a_5p which is downregulated during aging was upregulated after surgery. Overall, bariatric surgery ameliorated the premature aging phenotype.
Collapse
Affiliation(s)
- P J Hohensinner
- Department of Internal Medicine II, Division of Cardiology, Medical University of Vienna, Waehringer Guertel 18-20, 1090, Vienna, Austria.,Ludwig Boltzmann Cluster for Cardiovascular Research, Vienna, Austria
| | - C Kaun
- Department of Internal Medicine II, Division of Cardiology, Medical University of Vienna, Waehringer Guertel 18-20, 1090, Vienna, Austria
| | - B Ebenbauer
- Department of Internal Medicine II, Division of Cardiology, Medical University of Vienna, Waehringer Guertel 18-20, 1090, Vienna, Austria.,Ludwig Boltzmann Cluster for Cardiovascular Research, Vienna, Austria
| | - M Hackl
- TAmiRNA GmbH, Vienna, Austria
| | - S Demyanets
- Department of Internal Medicine II, Division of Cardiology, Medical University of Vienna, Waehringer Guertel 18-20, 1090, Vienna, Austria.,Department of Laboratory Medicine, Medical University of Vienna, Vienna, Austria
| | - D Richter
- Department of General Surgery, Territory Hospital Oberwart, Oberwart, Austria.,Department of Surgery, Hospital Hietzing, Vienna, Austria
| | - M Prager
- Department of General Surgery, Territory Hospital Oberwart, Oberwart, Austria.,Department of Surgery, Hospital Hietzing, Vienna, Austria
| | - J Wojta
- Department of Internal Medicine II, Division of Cardiology, Medical University of Vienna, Waehringer Guertel 18-20, 1090, Vienna, Austria.,Ludwig Boltzmann Cluster for Cardiovascular Research, Vienna, Austria.,Core Facilities, Medical University of Vienna, Vienna, Austria
| | - Gersina Rega-Kaun
- Department of Internal Medicine II, Division of Cardiology, Medical University of Vienna, Waehringer Guertel 18-20, 1090, Vienna, Austria. .,Ludwig Boltzmann Cluster for Cardiovascular Research, Vienna, Austria. .,5th Medical Department for Endocrinology and Rheumatology, Wilhelminenhospital, Vienna, Austria.
| |
Collapse
|
423
|
Ghosh AK, O’Brien M, Mau T, Qi N, Yung R. Adipose Tissue Senescence and Inflammation in Aging is Reversed by the Young Milieu. J Gerontol A Biol Sci Med Sci 2019; 74:1709-1715. [PMID: 30590424 PMCID: PMC6777079 DOI: 10.1093/gerona/gly290] [Citation(s) in RCA: 55] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2018] [Indexed: 11/14/2022] Open
Abstract
Visceral adipose tissue (VAT) inflammation plays a central role in longevity and multiple age-related disorders. Cellular senescence (SEN) is a fundamental aging mechanism that contributes to age-related chronic inflammation and organ dysfunction, including VAT. Recent studies using heterochronic parabiosis models strongly suggested that circulating factors in young plasma alter the aging phenotypes of old animals. Our study investigated if young plasma rescued SEN phenotypes in the VAT of aging mice. With heterochronic parabiosis model using young (3 months) and old (18 months) mice, we found significant reduction in the levels of pro-inflammatory cytokines and altered adipokine profile that are protective of SEN in the VAT of old mice. These data are indicative of protection from SEN of aging VAT by young blood circulation. Old parabionts also exhibited diminished expression of cyclin-dependent kinase inhibitors (CDKi) genes p16 (Cdkn2a) and p21 (Cdkn1a/Cip1) in the VAT. In addition, when exposed to young serum condition in an ex vivo culture system, aging adipose tissue-derived stromovascular fraction cells produced significantly lower amounts of pro-inflammatory cytokines (MCP-1 and IL-6) compared to old condition. Expressions of p16 and p21 genes were also diminished in the old stromovascular fraction cells under young serum condition. Finally, in 3T3-preadipocytes culture system, we found reduced pro-inflammatory cytokines (Mcp-1 and Il-6) and diminished expression of cyclin-dependent kinase inhibitor genes in the presence of young serum compared to old serum. In summary, this study demonstrates that young milieu is capable of protecting aging adipose tissue from SEN and thereby inflammation.
Collapse
Affiliation(s)
- Amiya Kumar Ghosh
- Division of Geriatric and Palliative Medicine, University of Michigan, Ann Arbor
| | - Martin O’Brien
- Division of Geriatric and Palliative Medicine, University of Michigan, Ann Arbor
| | - Theresa Mau
- Division of Geriatric and Palliative Medicine, University of Michigan, Ann Arbor
| | - Nathan Qi
- Animal Phenotyping Core, Division of Metabolism, Endocrinology, and Diabetes, Department of Internal Medicine, University of Michigan, Ann Arbor
| | - Raymond Yung
- Division of Geriatric and Palliative Medicine, University of Michigan, Ann Arbor
| |
Collapse
|
424
|
Palmer AK, Gustafson B, Kirkland JL, Smith U. Cellular senescence: at the nexus between ageing and diabetes. Diabetologia 2019; 62:1835-1841. [PMID: 31451866 PMCID: PMC6731336 DOI: 10.1007/s00125-019-4934-x] [Citation(s) in RCA: 209] [Impact Index Per Article: 34.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/15/2019] [Accepted: 05/21/2019] [Indexed: 12/11/2022]
Abstract
Ageing and diabetes lead to similar organ dysfunction that is driven by parallel molecular mechanisms, one of which is cellular senescence. The abundance of senescent cells in various tissues increases with age, obesity and diabetes. Senescent cells have been directly implicated in the generation of insulin resistance. Recently, drugs that preferentially target senescent cells, known as senolytics, have been described and recently entered clinical trials. In this review, we explore the biological links between ageing and diabetes, specifically focusing on cellular senescence. We summarise the current data on cellular senescence in key target tissues associated with the development and clinical phenotypes of type 2 diabetes and discuss the therapeutic potential of targeting cellular senescence in diabetes.
Collapse
Affiliation(s)
- Allyson K Palmer
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, 200 1st St SW, Rochester, MN, USA
| | - Birgit Gustafson
- Lundberg Laboratory for Diabetes Research, Department of Molecular and Clinical Medicine, Sahlgrenska University Hospital and University of Gothenburg, 413 45, Gothenburg, Sweden
| | - James L Kirkland
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, 200 1st St SW, Rochester, MN, USA.
| | - Ulf Smith
- Lundberg Laboratory for Diabetes Research, Department of Molecular and Clinical Medicine, Sahlgrenska University Hospital and University of Gothenburg, 413 45, Gothenburg, Sweden.
| |
Collapse
|
425
|
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
|
426
|
Sawaki D, Czibik G, Pini M, Ternacle J, Suffee N, Mercedes R, Marcelin G, Surenaud M, Marcos E, Gual P, Clément K, Hue S, Adnot S, Hatem SN, Tsuchimochi I, Yoshimitsu T, Hénégar C, Derumeaux G. Visceral Adipose Tissue Drives Cardiac Aging Through Modulation of Fibroblast Senescence by Osteopontin Production. Circulation 2019; 138:809-822. [PMID: 29500246 DOI: 10.1161/circulationaha.117.031358] [Citation(s) in RCA: 133] [Impact Index Per Article: 22.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND Aging induces cardiac structural and functional changes linked to the increased deposition of extracellular matrix proteins, including OPN (osteopontin), conducing to progressive interstitial fibrosis. Although OPN is involved in various pathological conditions, its role in myocardial aging remains unknown. METHODS OPN deficient mice (OPN-/-) with their wild-type (WT) littermates were evaluated at 2 and 14 months of age in terms of cardiac structure, function, histology and key molecular markers. OPN expression was determined by reverse-transcription polymerase chain reaction, immunoblot and immunofluorescence. Luminex assays were performed to screen plasma samples for various cytokines/adipokines in addition to OPN. Similar explorations were conducted in aged WT mice after surgical removal of visceral adipose tissue (VAT) or treatment with a small-molecule OPN inhibitor agelastatin A. Primary WT fibroblasts were incubated with plasma from aged WT and OPN-/- mice, and evaluated for senescence (senescence-associated β-galactosidase and p16), as well as fibroblast activation markers (Acta2 and Fn1). RESULTS Plasma OPN levels increased in WT mice during aging, with VAT showing the strongest OPN induction contrasting with myocardium that did not express OPN. VAT removal in aged WT mice restored cardiac function and decreased myocardial fibrosis in addition to a substantial reduction of circulating OPN and transforming growth factor β levels. OPN deficiency provided a comparable protection against age-related cardiac fibrosis and dysfunction. Intriguingly, a strong induction of senescence in cardiac fibroblasts was observed in both VAT removal and OPN-/- mice. The addition of plasma from aged OPN-/- mice to cultures of primary cardiac fibroblasts induced senescence and reduced their activation (compared to aged WT plasma). Finally, Agelastatin A treatment of aged WT mice fully reversed age-related myocardial fibrosis and dysfunction. CONCLUSIONS During aging, VAT represents the main source of OPN and alters heart structure and function via its profibrotic secretome. As a proof-of-concept, interventions targeting OPN, such as VAT removal and OPN deficiency, rescued the heart and induced a selective modulation of fibroblast senescence. Our work uncovers OPN's role in the context of myocardial aging and proposes OPN as a potential new therapeutic target for a healthy cardiac aging.
Collapse
Affiliation(s)
- Daigo Sawaki
- INSERM IMRB U955, Université Paris-Est Creteil (D.S., G.C., M.P., J.T., R.M., M.S., E.M., S.H., S.A., C.H., G.D.)
| | - Gabor Czibik
- INSERM IMRB U955, Université Paris-Est Creteil (D.S., G.C., M.P., J.T., R.M., M.S., E.M., S.H., S.A., C.H., G.D.)
| | - Maria Pini
- INSERM IMRB U955, Université Paris-Est Creteil (D.S., G.C., M.P., J.T., R.M., M.S., E.M., S.H., S.A., C.H., G.D.)
| | - Julien Ternacle
- INSERM IMRB U955, Université Paris-Est Creteil (D.S., G.C., M.P., J.T., R.M., M.S., E.M., S.H., S.A., C.H., G.D.)
- AP-HP, Department of Cardiology, Henri Mondor Hospital, DHU-ATVB (J.T., G.D.)
| | - Nadine Suffee
- Sorbonne Université, INSERM UMRS 1166, Institute of Cardiometabolism and Nutrition ICAN (N.S., S.H.)
| | - Raquel Mercedes
- INSERM IMRB U955, Université Paris-Est Creteil (D.S., G.C., M.P., J.T., R.M., M.S., E.M., S.H., S.A., C.H., G.D.)
| | - Geneviève Marcelin
- Institute of Cardiometabolism and Nutrition, ICAN, Pitié-Salpêtrière Hospital (G.M., K.C.)
- Sorbonne Universities, Université Pierre et Marie Curie, University of Paris 06, INSERM UMR_S 1166, Nutriomics Team 6 (G.M., K.C.)
| | - Mathieu Surenaud
- INSERM IMRB U955, Université Paris-Est Creteil (D.S., G.C., M.P., J.T., R.M., M.S., E.M., S.H., S.A., C.H., G.D.)
- AP-HP Vaccine Research Institute (VRI) (M.S., S.H.)
| | - Elisabeth Marcos
- INSERM IMRB U955, Université Paris-Est Creteil (D.S., G.C., M.P., J.T., R.M., M.S., E.M., S.H., S.A., C.H., G.D.)
| | - Philippe Gual
- INSERM, U1065, C3M, Team 8 "hepatic complications in obesity" (P.G.)
- Université Côte d'Azur (P.G.)
| | - Karine Clément
- Institute of Cardiometabolism and Nutrition, ICAN, Pitié-Salpêtrière Hospital (G.M., K.C.)
- Sorbonne Universities, Université Pierre et Marie Curie, University of Paris 06, INSERM UMR_S 1166, Nutriomics Team 6 (G.M., K.C.)
- Assistance Publique Hopitaux de Paris, AP-HP, Pitié-Salpêtrière Hospital, Nutrition and Endocrinology Department and Hepato-biliary and Digestive Surgery Department (K.C.)
| | - Sophie Hue
- INSERM IMRB U955, Université Paris-Est Creteil (D.S., G.C., M.P., J.T., R.M., M.S., E.M., S.H., S.A., C.H., G.D.)
- Sorbonne Université, INSERM UMRS 1166, Institute of Cardiometabolism and Nutrition ICAN (N.S., S.H.)
- AP-HP Vaccine Research Institute (VRI) (M.S., S.H.)
| | - Serge Adnot
- INSERM IMRB U955, Université Paris-Est Creteil (D.S., G.C., M.P., J.T., R.M., M.S., E.M., S.H., S.A., C.H., G.D.)
- AP-HP, Department of Physiology, Henri Mondor Hospital, DHU-ATVB (S.A.)
| | - Stéphane N Hatem
- Institut de Cardiologie, Hôpital Universitaire Pitié-Salpêtrière, Assistance Publique-Hôpitaux de Paris (S.H.)
| | - Izuru Tsuchimochi
- Laboratory of Synthetic Organic and Medicinal Chemistry, Division of Pharmaceutical Sciences, Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama University (I.T., T.Y.)
| | - Takehiko Yoshimitsu
- Laboratory of Synthetic Organic and Medicinal Chemistry, Division of Pharmaceutical Sciences, Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama University (I.T., T.Y.)
| | - Corneliu Hénégar
- INSERM IMRB U955, Université Paris-Est Creteil (D.S., G.C., M.P., J.T., R.M., M.S., E.M., S.H., S.A., C.H., G.D.)
| | - Geneviève Derumeaux
- INSERM IMRB U955, Université Paris-Est Creteil (D.S., G.C., M.P., J.T., R.M., M.S., E.M., S.H., S.A., C.H., G.D.)
- AP-HP, Department of Cardiology, Henri Mondor Hospital, DHU-ATVB (J.T., G.D.)
| |
Collapse
|
427
|
Slusher AL, Zúñiga TM, Acevedo EO. Inflamm-Aging Is Associated with Lower Plasma PTX3 Concentrations and an Impaired Capacity of PBMCs to Express hTERT following LPS Stimulation. Mediators Inflamm 2019; 2019:2324193. [PMID: 31611733 PMCID: PMC6757284 DOI: 10.1155/2019/2324193] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2019] [Revised: 05/22/2019] [Accepted: 06/20/2019] [Indexed: 12/31/2022] Open
Abstract
Age-related elevations in proinflammatory cytokines, known as inflamm-aging, are associated with shorter immune cell telomere lengths. Purpose. This study examined the relationship of plasma PTX3 concentrations, a biomarker of appropriate immune function, with telomere length in 15 middle-aged (40-64 years) and 15 young adults (20-31 years). In addition, PBMCs were isolated from middle-aged and young adults to examine their capacity to express a key mechanistic component of telomere length maintenance, human telomerase reverse transcriptase (hTERT), following ex vivo cellular stimulation. Methods. Plasma PTX3 and inflammatory cytokines (i.e., IL-6, IL-10, TGF-β, and TNF-α), PBMC telomere lengths, and PBMC hTERT gene expression and inflammatory protein secretion following exposure to LPS, PTX3, and PTX3+LPS were measured. Results. Aging was accompanied by the accumulation of centrally located visceral adipose tissue, without changes in body weight and BMI, and alterations in the systemic inflammatory milieu (decreased plasma PTX3 and TGF-β; increased TNF-α (p ≤ 0.050)). In addition, shorter telomere lengths in middle-aged compared to young adults (p = 0.011) were negatively associated with age, body fat percentages, and plasma TNF-α (r = -0.404, p = 0.027; r = -0.427, p = 0.019; and r = -0.323, p = 0.041, respectively). Finally, the capacity of PBMCs to increase hTERT gene expression following ex vivo stimulation was impaired in middle-aged compared to young adults (p = 0.033) and negatively associated with telomere lengths (r = 0.353, p = 0.028). Conclusions. Proinflammation and the impaired hTERT gene expression capacity of PBMCs may contribute to age-related telomere attrition and disease.
Collapse
Affiliation(s)
- Aaron L. Slusher
- Department of Kinesiology and Health Sciences, Virginia Commonwealth University, Richmond, VA, USA
- School of Kinesiology, University of Michigan, Ann Arbor, MI, USA
| | - Tiffany M. Zúñiga
- Department of Kinesiology and Health Sciences, Virginia Commonwealth University, Richmond, VA, USA
- Department of Nutritional Sciences, University of Arizona, Tucson, AZ, USA
| | - Edmund O. Acevedo
- Department of Kinesiology and Health Sciences, Virginia Commonwealth University, Richmond, VA, USA
| |
Collapse
|
428
|
Majerczak J, Grandys M, Frołow M, Szkutnik Z, Zakrzewska A, Niżankowski R, Duda K, Chlopicki S, Zoladz JA. Age-Dependent Impairment in Endothelial Function and Arterial Stiffness in Former High Class Male Athletes Is No Different to That in Men With No History of Physical Training. J Am Heart Assoc 2019; 8:e012670. [PMID: 31512551 PMCID: PMC6817998 DOI: 10.1161/jaha.119.012670] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Background Physical activity is generally considered to exert positive effects on the cardiovascular system in humans. However, surprisingly little is known about the delayed effect of professional physical training performed at a young age on endothelial function and arterial stiffness in aging athletes. The present study aimed to assess the impact of long‐lasting professional physical training (endurance and sprint) performed at a young age on the endothelial function and arterial stiffness reported in older age in relation to glycocalyx injury, prostacyclin and nitric oxide production, inflammation, basal blood lipid profile, and glucose homeostasis. Methods and Results This study involved 94 male subjects with varied training backgrounds, including young athletes (mean age ∼25 years), older former high class athletes (mean age ∼60 years), and aged‐matched untrained control groups. Aging increased arterial stiffness, as reflected by an enhancement in pulse wave velocity, augmentation index, and stiffness index (P<10−4), as well as decreased endothelial function, as judged by the attenuation of flow‐mediated vasodilation (FMD) in the brachial artery (P=0.03). Surprisingly, no effect of the training performed at a young age on endothelial function and arterial stiffness was observed in the former athletes. Moreover, no effect of training performed at a young age (P>0.05) on blood lipid profile, markers of inflammation, and glycocalyx shedding were observed in the former athletes. Conclusions Our study clearly shows that aging, but not physical training history, represents the main contributing factor responsible for decline in endothelial function and increase in arterial stiffness in former athletes.
Collapse
Affiliation(s)
- Joanna Majerczak
- Department of Neurobiology Poznan University of Physical Education Poznan Poland.,Department of Muscle Physiology Chair of Physiology and Biochemistry Faculty of Rehabilitation University School of Physical Education Krakow Poland
| | - Marcin Grandys
- Department of Muscle Physiology Chair of Physiology and Biochemistry Faculty of Rehabilitation University School of Physical Education Krakow Poland
| | - Marzena Frołow
- Jagiellonian Centre for Experimental Therapeutics (JCET) Jagiellonian University Krakow Poland
| | - Zbigniew Szkutnik
- Faculty of Applied Mathematics AGH-University of Science and Technology Krakow Poland
| | - Agnieszka Zakrzewska
- Jagiellonian Centre for Experimental Therapeutics (JCET) Jagiellonian University Krakow Poland
| | - Rafał Niżankowski
- Jagiellonian Centre for Experimental Therapeutics (JCET) Jagiellonian University Krakow Poland
| | - Krzysztof Duda
- Department of Muscle Physiology Chair of Physiology and Biochemistry Faculty of Rehabilitation University School of Physical Education Krakow Poland
| | - Stefan Chlopicki
- Jagiellonian Centre for Experimental Therapeutics (JCET) Jagiellonian University Krakow Poland.,Chair of Pharmacology Jagiellonian University Medical College Krakow Poland
| | - Jerzy A Zoladz
- Department of Muscle Physiology Chair of Physiology and Biochemistry Faculty of Rehabilitation University School of Physical Education Krakow Poland
| |
Collapse
|
429
|
Justice JN, Gregory H, Tchkonia T, LeBrasseur NK, Kirkland JL, Kritchevsky SB, Nicklas BJ. Cellular Senescence Biomarker p16INK4a+ Cell Burden in Thigh Adipose is Associated With Poor Physical Function in Older Women. J Gerontol A Biol Sci Med Sci 2019; 73:939-945. [PMID: 28658942 DOI: 10.1093/gerona/glx134] [Citation(s) in RCA: 97] [Impact Index Per Article: 16.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2017] [Indexed: 12/18/2022] Open
Abstract
Background Ample evidence implicates cellular senescence as a contributor to frailty and functional decline in rodents, but considerable effort remains to translate these findings to human aging. Methods We quantified senescence biomarker p16INK4a-expressing cells in thigh adipose tissue obtained from older women previously enrolled in a 5-month resistance training intervention, with or without caloric restriction (RT ± CR, n = 11 baseline, 8 pre-post-intervention pairs). Women in this subsample were older (72.9 ± 3.4 y) and overweight/obese (body mass index: 30.6 ± 2.4 kg/m2). p16INK4a+ cells were identified from 12 to 20 random visual fields/sample at 20× magnification (immunohistochemical, nuclear staining) and were present in all adipose samples. Results Cross-sectional associations were observed between p16INK4a+ cell burden and physical function, including grip strength (r = -0.74), 400-m walk time (r = 0.74), 4-m gait speed (r = -0.73), and self-perceived mobility (r = -0.78) (p ≤ .05). These relationships remained significant after independent adjustments for age and adiposity (p ≤ .05). p16INK4a+ cell abundance was lower following the intervention (pre: 5.47 ± 3.4%, post: 2.17 ± 1.1% count p16INK4a+ cells, p ≤ .05). Conclusions These results provide proof-of-concept that p16INK4a+ cells in thigh adipose are associated with physical function, and may be sensitive to change with RT ± CR in overweight/obese older women.
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), Winston-Salem, North Carolina
| | - Heather Gregory
- Sticht Center for Healthy Aging and Alzheimer's Prevention, Internal Medicine - Gerontology and Geriatric Medicine, Wake Forest School of Medicine (WFSM), Winston-Salem, North Carolina
| | - Tamar Tchkonia
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, Minnesota
| | | | - James L Kirkland
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, Minnesota
| | - Stephen B Kritchevsky
- Sticht Center for Healthy Aging and Alzheimer's Prevention, Internal Medicine - Gerontology and Geriatric Medicine, Wake Forest School of Medicine (WFSM), Winston-Salem, North Carolina
| | - Barbara J Nicklas
- Sticht Center for Healthy Aging and Alzheimer's Prevention, Internal Medicine - Gerontology and Geriatric Medicine, Wake Forest School of Medicine (WFSM), Winston-Salem, North Carolina
| |
Collapse
|
430
|
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
|
431
|
El-Kafoury BMA, Bahgat NM, Abdel-Hady EA, Samad AAAE, Shawky MK, Mohamed FA. Impaired metabolic and hepatic functions following subcutaneous lipectomy in adult obese rats. Exp Physiol 2019; 104:1661-1677. [PMID: 31443137 DOI: 10.1113/ep087670] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2019] [Accepted: 08/16/2019] [Indexed: 12/13/2022]
Abstract
NEW FINDINGS What is the central question of this study? What is the impact and drawbacks of subcutaneous lipectomy on body metabolism? What is the main finding and its importance? Subcutaneous lipectomy resulted in deterioration of hepatic functions, atherosclerotic lipid profile and disturbed redox state. While the results support lipectomy as an effective treatment for obesity, lipectomy induces unfavourable changes in health. ABSTRACT The number of obese older adults is on the rise, but data about proper treatment of obesity in the elderly is controversial. The present study was designed to investigate the effectiveness and consequences of partial subcutaneous lipectomy, as a rapid medical intervention against increased accumulation of body fat, in adult obese rats. The study was conducted on adult (9-12 months) female rats, in which obesity was induced by bilateral surgical ovariectomy. They were randomized into two main groups: short term (5 weeks) and long term (10 weeks). Both groups were subdivided into control, ovariectomized (OVX) and ovariectomized lipectomized groups. Body weight (BW) was measured and body mass index (BMI) calculated. Fasting blood glucose, lipid profile and plasma levels of total proteins, albumin, liver enzymes, malondialdehyde (MDA), leptin and adiponectin were determined. The content of both blood and hepatic tissue of reduced glutathione was estimated. In addition, histological study of the liver, aorta and peri-renal fat was performed. Compared to controls, OVX rats showed significant increase in BW, BMI and plasma levels of liver enzymes, MDA and leptin. Histological study revealed vacuolated ballooned hepatocytes and enlarged irregular visceral adipocytes with atherosclerotic changes in the wall of aorta. Following subcutaneous lipectomy, rats exhibited significant fasting hyperglycaemia, dyslipidaemia, lowered plasma albumin and disturbed redox state with aggravation of the histological changes. The findings indicate that although subcutaneous lipectomy appears to be effective in combating obesity in older females, it has unfavourable effects on both metabolic and hepatic functions.
Collapse
Affiliation(s)
| | - Nehal M Bahgat
- Physiology Department, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| | - Enas A Abdel-Hady
- Physiology Department, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| | | | - Mona K Shawky
- Physiology Department, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| | - Fatma A Mohamed
- Physiology Department, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| |
Collapse
|
432
|
Frasca D, Blomberg BB. Adipose Tissue: A Tertiary Lymphoid Organ: Does It Change with Age? Gerontology 2019; 66:114-121. [PMID: 31412335 DOI: 10.1159/000502036] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2019] [Accepted: 07/10/2019] [Indexed: 12/16/2022] Open
Abstract
In this manuscript, we summarize published results showing that obesity and aging are inflammatory conditions associated with serious health problems, increased risk for disease and death. We show that fat mass increases with age and represents a major contributor to insulin resistance and the metabolic syndrome. We summarize the effects of age on the adipose tissue (AT), related to the abundance, distribution, cellular composition, endocrine signaling and function of the tissue. The AT is an immunological tissue, with several hallmarks of innate and adaptive immune responses. We show that in both mice and humans, the AT is heavily infiltrated by immune cells that have receptors for pro-inflammatory cytokines and chemokines secreted by the adipocytes and also by the immune cells that have infiltrated the AT. We also show that the AT provides an environment for the secretion of IgG antibodies with anti-self (autoimmune) reactivity. As we have previously shown, this is due to the release of self antigens following cell death due to hypoxia, as well as to the expression of activation-induced cytidine deaminase, the enzyme of class switch recombination, and the transcription factor T-bet by the resident B cells, which also express the membrane marker CD11c, both involved in the production of autoimmune IgG antibodies. We show data in support of the AT as a tertiary lymphoid organ (TLO), showing the examples of TLOs that develop within the AT, such as fat-associated lymphoid clusters and milky spots, as well as artery TLOs that develop in the adventitia areas of the aorta.
Collapse
Affiliation(s)
- Daniela Frasca
- Department of Microbiology and Immunology, University of Miami Miller School of Medicine, Miami, Florida, USA,
| | - Bonnie B Blomberg
- Department of Microbiology and Immunology, University of Miami Miller School of Medicine, Miami, Florida, USA.,Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, Florida, USA
| |
Collapse
|
433
|
Ogrodnik M, Salmonowicz H, Jurk D, Passos JF. Expansion and Cell-Cycle Arrest: Common Denominators of Cellular Senescence. Trends Biochem Sci 2019; 44:996-1008. [PMID: 31345557 DOI: 10.1016/j.tibs.2019.06.011] [Citation(s) in RCA: 70] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2019] [Revised: 06/24/2019] [Accepted: 06/26/2019] [Indexed: 12/21/2022]
Abstract
Cellular senescence is a major driver of age-related diseases, and senotherapies are being tested in clinical trials. Despite its popularity, cellular senescence is weakly defined and is frequently referred to as irreversible cell-cycle arrest. In this article we hypothesize that cellular senescence is a phenotype that results from the coordination of two processes: cell expansion and cell-cycle arrest. We provide evidence for the compatibility of the proposed model with recent findings showing senescence in postmitotic tissues, wound healing, obesity, and development. We believe our model also explains why some characteristics of senescence can be found in non-senescent cells. Finally, we propose new avenues for research from our model.
Collapse
Affiliation(s)
- Mikolaj Ogrodnik
- Department of Physiology and Biochemical Engineering, Mayo Clinic, Rochester, MN, USA.
| | - Hanna Salmonowicz
- Institute for Cell and Molecular Biosciences, Newcastle University, Newcastle upon Tyne, UK
| | - Diana Jurk
- Department of Physiology and Biochemical Engineering, Mayo Clinic, Rochester, MN, USA; Institute for Cell and Molecular Biosciences, Newcastle University, Newcastle upon Tyne, UK
| | - João F Passos
- Department of Physiology and Biochemical Engineering, Mayo Clinic, Rochester, MN, USA; Institute for Cell and Molecular Biosciences, Newcastle University, Newcastle upon Tyne, UK.
| |
Collapse
|
434
|
Zaarur N, Desevin K, Mackenzie J, Lord A, Grishok A, Kandror KV. ATGL-1 mediates the effect of dietary restriction and the insulin/IGF-1 signaling pathway on longevity in C. elegans. Mol Metab 2019; 27:75-82. [PMID: 31311719 PMCID: PMC6717769 DOI: 10.1016/j.molmet.2019.07.001] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/20/2019] [Revised: 06/28/2019] [Accepted: 07/02/2019] [Indexed: 01/08/2023] Open
Abstract
Objective Animal lifespan is controlled through genetic pathways that are conserved from nematodes to humans. Lifespan-promoting conditions in nematodes include fasting and a reduction of insulin/IGF signaling. Here we aimed to investigate the input of the Caenorhabditis elegans homologue of the mammalian rate-limiting lipolytic enzyme Adipose Triglyceride Lipase, ATGL-1, in longevity control. Methods We used a combination of genetic and biochemical approaches to determine the role of ATGL-1 in accumulation of triglycerides and regulation of longevity. Results We found that expression of ATGL is increased in the insulin receptor homologue mutant daf-2 in a FoxO/DAF-16-dependent manner. ATGL-1 is also up-regulated by fasting and in the eat-2 loss-of-function mutant strain. Overexpression of ATGL-1 increases basal and maximal oxygen consumption rate and extends lifespan in C. elegans. Reduction of ATGL-1 function suppresses longevity of the long-lived mutants eat-2 and daf-2. Conclusion Our results demonstrate that ATGL is required for extended lifespan downstream of both dietary restriction and reduced insulin/IGF signaling. Expression of ATGL-1 in Caenorhabditis elegans is regulated by fasting and insulin/IGF1 signaling. Over-expression of ATGL-1 extends lifespan while loss-of-function mutant decreases lifespan of long-lived C. elegans models. The effect of ATGL-1 on longevity may be mediated by an increase in mitochondrial oxidation.
Collapse
Affiliation(s)
- Nava Zaarur
- Department of Biochemistry, Boston University School of Medicine, Boston, MA, 02118, USA
| | - Kathleen Desevin
- Department of Biochemistry, Boston University School of Medicine, Boston, MA, 02118, USA
| | - James Mackenzie
- Department of Biochemistry, Boston University School of Medicine, Boston, MA, 02118, USA
| | - Avery Lord
- Department of Biochemistry, Boston University School of Medicine, Boston, MA, 02118, USA
| | - Alla Grishok
- Department of Biochemistry, Boston University School of Medicine, Boston, MA, 02118, USA.
| | - Konstantin V Kandror
- Department of Biochemistry, Boston University School of Medicine, Boston, MA, 02118, USA.
| |
Collapse
|
435
|
Long-Chain Fatty Acids and Inflammatory Markers Coaccumulate in the Skeletal Muscle of Sarcopenic Old Rats. DISEASE MARKERS 2019; 2019:9140789. [PMID: 31354893 PMCID: PMC6636585 DOI: 10.1155/2019/9140789] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/15/2019] [Revised: 04/09/2019] [Accepted: 05/09/2019] [Indexed: 01/16/2023]
Abstract
Obesity and inflammation are reportedly associated with the pathogenesis of sarcopenia, which is characterized by age-related loss of skeletal muscle mass. Intramuscular fat deposits have been found to compromise muscle integrity; however, the relevant fat compounds and their roles as mediators of muscular inflammation are not known. The aim of this study was to identify potential correlations between inflammation markers and lipid compounds that accumulate in the quadriceps muscle of previously described Sprague-Dawley (SD) rat model for high-fat diet- (HFD-) induced muscle loss. Six-month-old SD rats were continuously fed a control (CD) or HFD until the age of 21 months. Magnetic resonance imaging (MRI) revealed a significant decline in muscle cross-sectional area in male SD rats as a result of HFD, but not in female rats. Here, we developed a new procedure to quantitatively identify and classify the fatty acid methyl esters (FAMEs) in rats' quadriceps muscles from our former study using gas chromatography-mass spectrometry (GC-MS). Fatty acid analysis revealed accumulation of octadecadienoic (linoleic acid), octadecanoic (stearic acid), and octadecenoic (vaccenic acid) acids exclusively in the quadriceps muscles of male rats. The designated fatty acids were mainly incorporated into triacylglycerols (TAGs) or free fatty acids (FFAs), and their proportions were significantly elevated by consumption of a HFD. Furthermore, the number of resident immune cells and the levels of the chemokines RANTES, MCP-1, and MIP-2 were significantly increased in quadriceps muscle tissue of HFD-fed male, but not female rats. Together, HFD-induced muscle loss in aged male SD rats is associated with greater deposits of long-chain fatty acid esters and increased levels of the inflammatory markers RANTES, MCP-1, and MIP-2 in skeletal muscle tissue. This trend is further reinforced by long-term consumption of a HFD, which may provoke synergistic crosstalk between long-chain fatty acids and inflammatory pathways in sarcopenic muscle.
Collapse
|
436
|
Zanfirescu A, Ungurianu A, Tsatsakis AM, Nițulescu GM, Kouretas D, Veskoukis A, Tsoukalas D, Engin AB, Aschner M, Margină D. A review of the alleged health hazards of monosodium glutamate. Compr Rev Food Sci Food Saf 2019; 18:1111-1134. [PMID: 31920467 PMCID: PMC6952072 DOI: 10.1111/1541-4337.12448] [Citation(s) in RCA: 92] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2019] [Accepted: 03/15/2019] [Indexed: 12/11/2022]
Abstract
Monosodium glutamate (MSG) is an umami substance widely used as flavor enhancer. Although it is generally recognized as being safe by food safety regulatory agencies, several studies have questioned its long-term safety. The purpose of this review was to survey the available literature on preclinical studies and clinical trials regarding the alleged adverse effects of MSG. Here, we aim to provide a comprehensive overview of the reported possible risks that may potentially arise following chronic exposure. Furthermore, we intend to critically evaluate the relevance of this data for dietary human intake. Preclinical studies have associated MSG administration with cardiotoxicity, hepatotoxicity, neurotoxicity, low-grade inflammation, metabolic disarray and premalignant alterations, along with behavioral changes. Moreover, links between MSG consumption and tumorigenesis, increased oxidative stress and apoptosis in thymocytes, as well as genotoxic effects in lymphocytes have been reported. However, in reviewing the available literature, we detected several methodological flaws, which led us to conclude that these studies have limited relevance for extrapolation to dietary human intakes of MSG risk exposure. Clinical trials have focused mainly on the effects of MSG on food intake and energy expenditure. Besides its well-known impact on food palatability, MSG enhances salivary secretion and interferes with carbohydrate metabolism, while the impact on satiety and post-meal recovery of hunger varied in relation to meal composition. Reports on MSG hypersensitivity, also known as 'Chinese restaurant syndrome', or links of its use to increased pain sensitivity and atopic dermatitis were found to have little supporting evidence. Based on the available literature, we conclude that further clinical and epidemiological studies are needed, with an appropriate design, accounting for both added and naturally occurring dietary MSG. Critical analysis of existing literature, establishes that many of the reported negative health effects of MSG have little relevance for chronic human exposure and are poorly informative as they are based on excessive dosing that does not meet with levels normally consumed in food products.
Collapse
Affiliation(s)
- Anca Zanfirescu
- Faculty of Pharmacy, “Carol Davila” University of Medicine and Pharmacy, Traian Vuia 6, Bucharest 020956, Romania
| | - Anca Ungurianu
- Faculty of Pharmacy, “Carol Davila” University of Medicine and Pharmacy, Traian Vuia 6, Bucharest 020956, Romania
| | - Aristides M. Tsatsakis
- Department of Forensic Sciences and Toxicology, Faculty of Medicine, University of Crete, Heraklion 71409, Crete, Greece
| | - George M. Nițulescu
- Faculty of Pharmacy, “Carol Davila” University of Medicine and Pharmacy, Traian Vuia 6, Bucharest 020956, Romania
| | - Demetrios Kouretas
- Department of Biochemistry and Biotechnology, University of Thessaly, Larissa 41500, Greece
| | - Aris Veskoukis
- Department of Biochemistry and Biotechnology, University of Thessaly, Larissa 41500, Greece
| | - Dimitrios Tsoukalas
- Department of Forensic Sciences and Toxicology, Faculty of Medicine, University of Crete, Heraklion 71409, Crete, Greece
- Metabolomic Medicine Clinic, Athens 10674, Greece
| | - Ayse B. Engin
- Gazi University, Faculty of Pharmacy, Department of Toxicology, Ankara 06330, Turkey
| | - Michael Aschner
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx NY 10463, USA
| | - Denisa Margină
- Faculty of Pharmacy, “Carol Davila” University of Medicine and Pharmacy, Traian Vuia 6, Bucharest 020956, Romania
| |
Collapse
|
437
|
In vitro model of chronological aging of adipocytes: Interrelationships with hypoxia and oxidation. Exp Gerontol 2019; 121:81-90. [DOI: 10.1016/j.exger.2019.03.011] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2018] [Revised: 03/25/2019] [Accepted: 03/26/2019] [Indexed: 01/07/2023]
|
438
|
Sun M, Tan Y, Rexiati M, Dong M, Guo W. Obesity is a common soil for premature cardiac aging and heart diseases - Role of autophagy. Biochim Biophys Acta Mol Basis Dis 2019; 1865:1898-1904. [DOI: 10.1016/j.bbadis.2018.09.004] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2018] [Revised: 08/22/2018] [Accepted: 09/04/2018] [Indexed: 12/31/2022]
|
439
|
Lipotoxicity, aging, and muscle contractility: does fiber type matter? GeroScience 2019; 41:297-308. [PMID: 31227962 DOI: 10.1007/s11357-019-00077-z] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2019] [Accepted: 06/05/2019] [Indexed: 12/18/2022] Open
Abstract
Sarcopenia is a universal characteristic of the aging process and is often accompanied by increases in whole-body adiposity. These changes in body composition have important clinical implications, given that loss of muscle and gain of fat mass are both significantly and independently associated with declining physical performance as well as an increased risk for disability, hospitalizations, and mortality in older individuals. This increased fat mass is not exclusively stored in adipose depots but may become deposited in non-adipose tissues, such as skeletal muscle, when the oxidative capacity of the adipose tissue itself is exceeded. The redistributed adipose tissue is thought to exert detrimental local effects on the muscle environment given the close proximity. Thus, sarcopenia observed with aging may be better defined in the context of loss of muscle quality rather than loss of muscle quantity per se. In this perspective, we briefly review the age-related physiological changes in cellularity, secretory profiles, and inflammatory status of adipose tissue which drive lipotoxicity (spillover) of skeletal muscle and then provide evidence of how this may affect specific fiber type contractility. We focus on biological contributors (cellular machinery) to contractility for which there is some evidence of vulnerability to lipid stress distinguishing between fiber types.
Collapse
|
440
|
Dipali SS, Ferreira CR, Zhou LT, Pritchard MT, Duncan FE. Histologic analysis and lipid profiling reveal reproductive age-associated changes in peri-ovarian adipose tissue. Reprod Biol Endocrinol 2019; 17:46. [PMID: 31189477 PMCID: PMC6563378 DOI: 10.1186/s12958-019-0487-6] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/04/2019] [Accepted: 05/07/2019] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Reproductive aging is a robust phenotype that occurs in all females and is characterized by a significant reduction in gamete quantity and quality, which can have negative consequences on both endocrine function and fertility. Age-associated differences in the oocyte, follicle, and ovary have been well-documented, but how the broader environment changes with age is less well understood. Fat is one of the largest organs in the body, and peri-gonadal adipose tissue surrounds the rodent ovary and comprises a local ovarian environment. The goal of this study was to characterize how peri-ovarian adipose tissue changes with advanced reproductive age. METHODS We isolated peri-gonadal adipose tissue from two cohorts of CB6F1 mice: reproductively young (6-12 weeks) and reproductively old (14-17 months). A comparative histological analysis was performed to evaluate adipocyte architecture. We then extracted lipids from the tissue and performed multiple reaction monitoring (MRM)-profiling, a mass spectrometry-based method of metabolite profiling, to compare the lipid profiles of peri-gonadal adipose tissue in these age cohorts. RESULTS We found that advanced reproductive age was associated with adipocyte hypertrophy and a corresponding decrease in the number of adipocytes per area. Of the 10 lipid classes examined, triacylglycerols (TAGs) had significantly different profiles between young and old cohorts, despite quantitative analysis revealing a decrease in the total amount of TAGs per weight of peri-gonadal adipose tissue with age. CONCLUSIONS These findings pinpoint age-associated physiological changes in peri-gonadal adipose tissue with respect to adipocyte morphology and lipid profiles and lay the foundation for future studies to examine how these alterations may influence both adipocyte and ovarian function.
Collapse
Affiliation(s)
- Shweta S Dipali
- Department of Obstetrics and Gynecology, Feinberg School of Medicine, Northwestern University, 303 E. Superior Street, Lurie 7-117, Chicago, IL, 60611, USA
| | - Christina R Ferreira
- Center for Analytical Instrumentation Development, Department of Chemistry, Purdue University, West Lafayette, IN, USA
- Bindley Bioscience Center, Purdue University, West Lafayette, IN, USA
| | - Luhan T Zhou
- Department of Obstetrics and Gynecology, Feinberg School of Medicine, Northwestern University, 303 E. Superior Street, Lurie 7-117, Chicago, IL, 60611, USA
| | - Michele T Pritchard
- Department of Pharmacology, Toxicology & Therapeutics, University of Kansas Medical Center, Kansas City, KS, USA
| | - Francesca E Duncan
- Department of Obstetrics and Gynecology, Feinberg School of Medicine, Northwestern University, 303 E. Superior Street, Lurie 7-117, Chicago, IL, 60611, USA.
| |
Collapse
|
441
|
mTOR and Aging: An Old Fashioned Dress. Int J Mol Sci 2019; 20:ijms20112774. [PMID: 31174250 PMCID: PMC6600378 DOI: 10.3390/ijms20112774] [Citation(s) in RCA: 65] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2019] [Revised: 06/03/2019] [Accepted: 06/04/2019] [Indexed: 12/12/2022] Open
Abstract
Aging is a physiologic/pathologic process characterized by a progressive impairment of cellular functions, supported by the alterations of several molecular pathways, leading to an increased cell susceptibility to injury. This deterioration is the primary risk factor for several major human pathologies. Numerous cellular processes, including genomic instability, telomere erosion, epigenetic alterations, loss of proteostasis, deregulated nutrient-sensing, mitochondrial dysfunction, stem cell exhaustion, and altered intercellular signal transduction represent common denominators of aging in different organisms. Mammalian target of rapamycin (mTOR) is an evolutionarily conserved nutrient sensing protein kinase that regulates growth and metabolism in all eukaryotic cells. Studies in flies, worms, yeast, and mice support the hypothesis that the mTOR signalling network plays a pivotal role in modulating aging. mTOR is emerging as the most robust mediator of the protective effects of various forms of dietary restriction, which has been shown to extend lifespan and slow the onset of age-related diseases across species. Herein we discuss the role of mTor signalling network in the development of classic age-related diseases, focused on cardiovascular system, immune response, and cancer.
Collapse
|
442
|
Sun W, von Meyenn F, Peleg‐Raibstein D, Wolfrum C. Environmental and Nutritional Effects Regulating Adipose Tissue Function and Metabolism Across Generations. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2019; 6:1900275. [PMID: 31179229 PMCID: PMC6548959 DOI: 10.1002/advs.201900275] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/04/2019] [Revised: 03/21/2019] [Indexed: 05/12/2023]
Abstract
The unabated rise in obesity prevalence during the last 40 years has spurred substantial interest in understanding the reasons for this epidemic. Studies in mice and humans have demonstrated that obesity is a highly heritable disease; however genetic variations within specific populations have so far not been able to explain this phenomenon to its full extent. Recent work has demonstrated that environmental cues can be sensed by an organism to elicit lasting changes, which in turn can affect systemic energy metabolism by different epigenetic mechanisms such as changes in small noncoding RNA expression, DNA methylation patterns, as well as histone modifications. These changes can directly modulate cellular function in response to environmental cues, however research during the last decade has demonstrated that some of these modifications might be transmitted to subsequent generations, thus modulating energy metabolism of the progeny in an inter- as well as transgenerational manner. In this context, adipose tissue has become a focus of research due to its plasticity, which allows the formation of energy storing (white) as well as energy wasting (brown/brite/beige) cells within the same depot. In this Review, the effects of environmental induced obesity with a particular focus on adipose tissue are discussed.
Collapse
Affiliation(s)
- Wenfei Sun
- Department of Health Science and TechnologiesETH ZürichSchorenstrasse 16SchwerzenbachCH‐8603Switzerland
| | - Ferdinand von Meyenn
- Department of Health Science and TechnologiesETH ZürichSchorenstrasse 16SchwerzenbachCH‐8603Switzerland
| | - Daria Peleg‐Raibstein
- Department of Health Science and TechnologiesETH ZürichSchorenstrasse 16SchwerzenbachCH‐8603Switzerland
| | - Christian Wolfrum
- Department of Health Science and TechnologiesETH ZürichSchorenstrasse 16SchwerzenbachCH‐8603Switzerland
| |
Collapse
|
443
|
Meng Z, Wang D, Liu W, Li R, Yan S, Jia M, Zhang L, Zhou Z, Zhu W. Perinatal exposure to Bisphenol S (BPS) promotes obesity development by interfering with lipid and glucose metabolism in male mouse offspring. ENVIRONMENTAL RESEARCH 2019; 173:189-198. [PMID: 30921577 DOI: 10.1016/j.envres.2019.03.038] [Citation(s) in RCA: 54] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/02/2018] [Revised: 03/14/2019] [Accepted: 03/15/2019] [Indexed: 05/27/2023]
Abstract
Bisphenol S (BPS), a substitute of bisphenol A (BPA), is widely used for manufacturing different polymers. Due to its wide range of applications, BPS has been frequently detected in the foodstuffs, environment and human blood and excreta. In this study, we examined the effects of the perinatal exposure to BPS on obesity development using 1H NMR based on metabolomics strategy combined with gene expression analysis in male mouse offspring at a dosage of 100 ng/g bw/day. We found that perinatal exposure to BPS significantly increased the body weight, the weights of liver and epididymal white adipose tissue (epiWAT), serum alanine aminotransferase (ALT) activity, and the contents of triglyceride (TG) and cholesterol (T-Cho) in the liver. Histopathological analysis showed that lipids were accumulated significantly in liver tissues and epiWAT with BPS exposure. Furthermore, expressions of genes involved in the inflammatory pathways were significantly increased in liver tissues and epiWAT. Meanwhile, serum metabolomics study showed significant changes in the contents of metabolites associated with lipid and glucose metabolism. Correspondingly, the relative expression levels of genes involved in lipid and glucose metabolism were significantly changed in the liver tissue and epiWAT of male mouse offspring. In conclusion, these results showed that perinatal exposure to BPS may increase the risk of obesity by interfering with lipid and glucose metabolism in male mouse offspring. The potential health risks of BPS in the human required further detailed studies evaluating.
Collapse
Affiliation(s)
- Zhiyuan Meng
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, Department of Applied Chemistry, China Agricultural University, Beijing, 100193, China
| | - Dezhen Wang
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, Department of Applied Chemistry, China Agricultural University, Beijing, 100193, China
| | - Wan Liu
- Department of Digestive, The Traditional Chinese Medicine Hospital of Xuzhou City Affiliated to Nanjing University of Chinese, Xuzhou, 221003, China
| | - Ruisheng Li
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, Department of Applied Chemistry, China Agricultural University, Beijing, 100193, China
| | - Sen Yan
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, Department of Applied Chemistry, China Agricultural University, Beijing, 100193, China
| | - Ming Jia
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, Department of Applied Chemistry, China Agricultural University, Beijing, 100193, China
| | - Luyao Zhang
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, Department of Applied Chemistry, China Agricultural University, Beijing, 100193, China
| | - Zhiqiang Zhou
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, Department of Applied Chemistry, China Agricultural University, Beijing, 100193, China
| | - Wentao Zhu
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, Department of Applied Chemistry, China Agricultural University, Beijing, 100193, China.
| |
Collapse
|
444
|
Gadecka A, Bielak-Zmijewska A. Slowing Down Ageing: The Role of Nutrients and Microbiota in Modulation of the Epigenome. Nutrients 2019; 11:nu11061251. [PMID: 31159371 PMCID: PMC6628342 DOI: 10.3390/nu11061251] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2019] [Revised: 05/27/2019] [Accepted: 05/28/2019] [Indexed: 12/13/2022] Open
Abstract
The human population is getting ageing. Both ageing and age-related diseases are correlated with an increased number of senescent cells in the organism. Senescent cells do not divide but are metabolically active and influence their environment by secreting many proteins due to a phenomenon known as senescence associated secretory phenotype (SASP). Senescent cells differ from young cells by several features. They possess more damaged DNA, more impaired mitochondria and an increased level of free radicals that cause the oxidation of macromolecules. However, not only biochemical and structural changes are related to senescence. Senescent cells have an altered chromatin structure, and in consequence, altered gene expression. With age, the level of heterochromatin decreases, and less condensed chromatin is more prone to DNA damage. On the one hand, some gene promoters are easily available for the transcriptional machinery; on the other hand, some genes are more protected (locally increased level of heterochromatin). The structure of chromatin is precisely regulated by the epigenetic modification of DNA and posttranslational modification of histones. The methylation of DNA inhibits transcription, histone methylation mostly leads to a more condensed chromatin structure (with some exceptions) and acetylation plays an opposing role. The modification of both DNA and histones is regulated by factors present in the diet. This means that compounds contained in daily food can alter gene expression and protect cells from senescence, and therefore protect the organism from ageing. An opinion prevailed for some time that compounds from the diet do not act through direct regulation of the processes in the organism but through modification of the physiology of the microbiome. In this review we try to explain the role of some food compounds, which by acting on the epigenetic level might protect the organism from age-related diseases and slow down ageing. We also try to shed some light on the role of microbiome in this process.
Collapse
Affiliation(s)
- Agnieszka Gadecka
- Nencki Institute of Experimental Biology, Polish Academy of Sciences, 3 Pasteur St., 02-093 Warsaw, Poland.
| | - Anna Bielak-Zmijewska
- Nencki Institute of Experimental Biology, Polish Academy of Sciences, 3 Pasteur St., 02-093 Warsaw, Poland.
| |
Collapse
|
445
|
Pratsinis H, Mavrogonatou E, Kletsas D. Scarless wound healing: From development to senescence. Adv Drug Deliv Rev 2019; 146:325-343. [PMID: 29654790 DOI: 10.1016/j.addr.2018.04.011] [Citation(s) in RCA: 58] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2017] [Revised: 03/29/2018] [Accepted: 04/09/2018] [Indexed: 12/21/2022]
Abstract
An essential element of tissue homeostasis is the response to injuries, cutaneous wound healing being the most studied example. In the adults, wound healing aims at quickly restoring the barrier function of the skin, leading however to scar, a dysfunctional fibrotic tissue. On the other hand, in fetuses a scarless tissue regeneration takes place. During ageing, the wound healing capacity declines; however, in the absence of comorbidities a higher quality in tissue repair is observed. Senescent cells have been found to accumulate in chronic unhealed wounds, but more recent reports indicate that their transient presence may be beneficial for tissue repair. In this review data on skin wound healing and scarring are presented, covering the whole spectrum from early embryonic development to adulthood, and furthermore until ageing of the organism.
Collapse
|
446
|
Postmus AC, Sturmlechner I, Jonker JW, van Deursen JM, van de Sluis B, Kruit JK. Senescent cells in the development of cardiometabolic disease. Curr Opin Lipidol 2019; 30:177-185. [PMID: 30913069 PMCID: PMC6530963 DOI: 10.1097/mol.0000000000000602] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
PURPOSE OF REVIEW Senescent cells have recently been identified as key players in the development of metabolic dysfunction. In this review, we will highlight recent developments in this field and discuss the concept of targeting these cells to prevent or treat cardiometabolic diseases. RECENT FINDINGS Evidence is accumulating that cellular senescence contributes to adipose tissue dysfunction, presumably through induction of low-grade inflammation and inhibition of adipogenic differentiation leading to insulin resistance and dyslipidaemia. Senescent cells modulate their surroundings through their bioactive secretome and only a relatively small number of senescent cells is sufficient to cause persistent physical dysfunction even in young mice. Proof-of-principle studies showed that selective elimination of senescent cells can prevent or delay the development of cardiometabolic diseases in mice. SUMMARY The metabolic consequences of senescent cell accumulation in various tissues are now unravelling and point to new therapeutic opportunities for the treatment of cardiometabolic diseases.
Collapse
Affiliation(s)
- Andrea C. Postmus
- Department of Pediatrics, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Ines Sturmlechner
- Departments of Pediatrics and Adolescent Medicine
- Department of Biochemistry and Molecular Biology, Mayo Clinic College of Medicine, Mayo Clinic, Rochester, Minnesota, USA
| | - Johan W. Jonker
- Department of Pediatrics, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Jan M. van Deursen
- Departments of Pediatrics and Adolescent Medicine
- Department of Biochemistry and Molecular Biology, Mayo Clinic College of Medicine, Mayo Clinic, Rochester, Minnesota, USA
| | - Bart van de Sluis
- Department of Pediatrics, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Janine K. Kruit
- Department of Pediatrics, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| |
Collapse
|
447
|
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
|
448
|
Tingstedt JL, Hove-Skovsgaard M, Gaardbo J, Ullum H, Nielsen SD, Gelpi M. The impact of concurrent HIV and type II diabetes on immune maturation, immune regulation and immune activation. APMIS 2019; 127:529-537. [PMID: 31017317 DOI: 10.1111/apm.12956] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2019] [Accepted: 04/12/2019] [Indexed: 12/15/2022]
Abstract
Chronic immune activation and inflammation are constant findings in people living with HIV (PLWH) and contribute to the risk of non-AIDS-related morbidities, including cardiovascular diseases (CVD). Type 2 diabetes (T2D) is also characterized by immune activation and inflammation. We aimed to investigate the impact of concurrent HIV infection and T2D on T-cell subsets. The study included PLWH with T2D (HIV+T2D+, N = 25) and without T2D (HIV+T2D-, N = 25) and HIV-negative controls with T2D (HIV-T2D+, N = 22) and without T2D (HIV-T2D-, N = 28). All PLWH in the study were receiving combination antiretroviral therapy. We examined T-cell homeostasis by determining T-cell subsets (immune maturation, immune regulation and immune activation) using flow cytometry. HIV+T2D- had lower proportion of Tc17 cells and higher proportion of apoptotic cells than HIV-T2D-. When comparing HIV+T2D+ and HIV+T2D- a lower proportion of CD4+ recent thymic emigrants (RTE) was found (p = 0.028). Furthermore, HIV+T2D+ had a higher proportion of non-suppressive CD4+ Tregs compared to HIV+T2D- (p = 0.010). In conclusion, even in the setting of treated HIV infection, distinct immunological alterations are found. In PLWH with concomitant T2D, most alterations in T-cell subsets were related to HIV and only few differences were found between PLWH with and without diabetes.
Collapse
Affiliation(s)
- Jeanette Linnea Tingstedt
- Viro-Immunology Research Unit, Department of Infectious Diseases, Rigshospitalet, University Hospital of Copenhagen, Copenhagen, Denmark.,Virus Research and Development Laboratory, Department of Microbiological Diagnostics and Virology, Statens Serum Institut, Copenhagen, Denmark
| | - Malene Hove-Skovsgaard
- Viro-Immunology Research Unit, Department of Infectious Diseases, Rigshospitalet, University Hospital of Copenhagen, Copenhagen, Denmark
| | - Julie Gaardbo
- Viro-Immunology Research Unit, Department of Infectious Diseases, Rigshospitalet, University Hospital of Copenhagen, Copenhagen, Denmark
| | - Henrik Ullum
- Department of Clinical Immunology, Rigshospitalet, University Hospital of Copenhagen, Copenhagen, Denmark
| | - Susanne Dam Nielsen
- Viro-Immunology Research Unit, Department of Infectious Diseases, Rigshospitalet, University Hospital of Copenhagen, Copenhagen, Denmark
| | - Marco Gelpi
- Viro-Immunology Research Unit, Department of Infectious Diseases, Rigshospitalet, University Hospital of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
449
|
Ogrodnik M, Zhu Y, Langhi LGP, Tchkonia T, Krüger P, Fielder E, Victorelli S, Ruswhandi RA, Giorgadze N, Pirtskhalava T, Podgorni O, Enikolopov G, Johnson KO, Xu M, Inman C, Palmer AK, Schafer M, Weigl M, Ikeno Y, Burns TC, Passos JF, von Zglinicki T, Kirkland JL, Jurk D. Obesity-Induced Cellular Senescence Drives Anxiety and Impairs Neurogenesis. Cell Metab 2019; 29:1061-1077.e8. [PMID: 30612898 PMCID: PMC6509403 DOI: 10.1016/j.cmet.2018.12.008] [Citation(s) in RCA: 315] [Impact Index Per Article: 52.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/19/2018] [Revised: 10/23/2018] [Accepted: 12/05/2018] [Indexed: 12/25/2022]
Abstract
Cellular senescence entails a stable cell-cycle arrest and a pro-inflammatory secretory phenotype, which contributes to aging and age-related diseases. Obesity is associated with increased senescent cell burden and neuropsychiatric disorders, including anxiety and depression. To investigate the role of senescence in obesity-related neuropsychiatric dysfunction, we used the INK-ATTAC mouse model, from which p16Ink4a-expressing senescent cells can be eliminated, and senolytic drugs dasatinib and quercetin. We found that obesity results in the accumulation of senescent glial cells in proximity to the lateral ventricle, a region in which adult neurogenesis occurs. Furthermore, senescent glial cells exhibit excessive fat deposits, a phenotype we termed "accumulation of lipids in senescence." Clearing senescent cells from high fat-fed or leptin receptor-deficient obese mice restored neurogenesis and alleviated anxiety-related behavior. Our study provides proof-of-concept evidence that senescent cells are major contributors to obesity-induced anxiety and that senolytics are a potential new therapeutic avenue for treating neuropsychiatric disorders.
Collapse
Affiliation(s)
- Mikolaj Ogrodnik
- Institute for Cell and Molecular Biosciences, Newcastle University Institute for Ageing, Campus for Ageing and Vitality, Newcastle upon Tyne NE4 5PL, UK; Robert and Arlene Kogod Center on Aging, Mayo Clinic, 200 First Street SW, Rochester, MN 55905, USA
| | - Yi Zhu
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, 200 First Street SW, Rochester, MN 55905, USA
| | - Larissa G P Langhi
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, 200 First Street SW, Rochester, MN 55905, USA
| | - Tamar Tchkonia
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, 200 First Street SW, Rochester, MN 55905, USA
| | - Patrick Krüger
- Institute for Cell and Molecular Biosciences, Newcastle University Institute for Ageing, Campus for Ageing and Vitality, Newcastle upon Tyne NE4 5PL, UK
| | - Edward Fielder
- Institute for Cell and Molecular Biosciences, Newcastle University Institute for Ageing, Campus for Ageing and Vitality, Newcastle upon Tyne NE4 5PL, UK
| | - Stella Victorelli
- Institute for Cell and Molecular Biosciences, Newcastle University Institute for Ageing, Campus for Ageing and Vitality, Newcastle upon Tyne NE4 5PL, UK
| | - Rifqha A Ruswhandi
- Institute for Cell and Molecular Biosciences, Newcastle University Institute for Ageing, Campus for Ageing and Vitality, Newcastle upon Tyne NE4 5PL, UK
| | - Nino Giorgadze
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, 200 First Street SW, Rochester, MN 55905, USA
| | - Tamar Pirtskhalava
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, 200 First Street SW, Rochester, MN 55905, USA
| | - Oleg Podgorni
- Department of Anesthesiology, Stony Brook School of Medicine, 101 Nicolls Road, Stony Brook, New York, NY 11794, USA; Center for Developmental Genetics, Stony Brook University, 100 Nicolls Road, Stony Brook, New York, NY 11794, USA
| | - Grigori Enikolopov
- Department of Anesthesiology, Stony Brook School of Medicine, 101 Nicolls Road, Stony Brook, New York, NY 11794, USA; Center for Developmental Genetics, Stony Brook University, 100 Nicolls Road, Stony Brook, New York, NY 11794, USA; Department of Nano-, Bio-, Information Technology and Cognitive Science, Moscow Institute of Physics and Technology, Moscow, Russia; Cold Spring Harbor Laboratory, Cold Spring Harbor, New York, NY, USA
| | - Kurt O Johnson
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, 200 First Street SW, Rochester, MN 55905, USA
| | - Ming Xu
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, 200 First Street SW, Rochester, MN 55905, USA
| | - Christine Inman
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, 200 First Street SW, Rochester, MN 55905, USA
| | - Allyson K Palmer
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, 200 First Street SW, Rochester, MN 55905, USA
| | - Marissa Schafer
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, 200 First Street SW, Rochester, MN 55905, USA
| | - Moritz Weigl
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, 200 First Street SW, Rochester, MN 55905, USA
| | - Yuji Ikeno
- The Barshop Institute for Longevity and Aging Studies, San Antonio, Department of Pathology, The University of Texas Health Science Center at San Antonio, Research Service, Audie L. Murphy VA Hospital (STVHCS), San Antonio, TX 78229, USA
| | - Terry C Burns
- Departments of Neurologic Surgery and Neuroscience, Mayo Clinic, 200 First Street SW, Rochester, MN 55905, USA
| | - João F Passos
- Institute for Cell and Molecular Biosciences, Newcastle University Institute for Ageing, Campus for Ageing and Vitality, Newcastle upon Tyne NE4 5PL, UK; Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN 55905, USA
| | - Thomas von Zglinicki
- Institute for Cell and Molecular Biosciences, Newcastle University Institute for Ageing, Campus for Ageing and Vitality, Newcastle upon Tyne NE4 5PL, UK; Near East University, Arts and Sciences Faculty, Molecular Biology and Genetics, Nicosia, North Cyprus POB 99138 Mersin 10, Turkey
| | - James L Kirkland
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, 200 First Street SW, Rochester, MN 55905, USA.
| | - Diana Jurk
- Institute for Cell and Molecular Biosciences, Newcastle University Institute for Ageing, Campus for Ageing and Vitality, Newcastle upon Tyne NE4 5PL, UK; Robert and Arlene Kogod Center on Aging, Mayo Clinic, 200 First Street SW, Rochester, MN 55905, USA; Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN 55905, USA.
| |
Collapse
|
450
|
Freedman BR, Mooney DJ. Biomaterials to Mimic and Heal Connective Tissues. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2019; 31:e1806695. [PMID: 30908806 PMCID: PMC6504615 DOI: 10.1002/adma.201806695] [Citation(s) in RCA: 136] [Impact Index Per Article: 22.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/15/2018] [Revised: 01/27/2019] [Indexed: 05/11/2023]
Abstract
Connective tissue is one of the four major types of animal tissue and plays essential roles throughout the human body. Genetic factors, aging, and trauma all contribute to connective tissue dysfunction and motivate the need for strategies to promote healing and regeneration. The goal here is to link a fundamental understanding of connective tissues and their multiscale properties to better inform the design and translation of novel biomaterials to promote their regeneration. Major clinical problems in adipose tissue, cartilage, dermis, and tendon are discussed that inspire the need to replace native connective tissue with biomaterials. Then, multiscale structure-function relationships in native soft connective tissues that may be used to guide material design are detailed. Several biomaterials strategies to improve healing of these tissues that incorporate biologics and are biologic-free are reviewed. Finally, important guidance documents and standards (ASTM, FDA, and EMA) that are important to consider for translating new biomaterials into clinical practice are highligted.
Collapse
Affiliation(s)
- Benjamin R Freedman
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, 02138, USA
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, 02115, USA
| | - David J Mooney
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, 02138, USA
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, 02115, USA
| |
Collapse
|