351
|
Dysregulated Autophagy Mediates Sarcopenic Obesity and Its Complications via AMPK and PGC1α Signaling Pathways: Potential Involvement of Gut Dysbiosis as a Pathological Link. Int J Mol Sci 2020; 21:ijms21186887. [PMID: 32961822 PMCID: PMC7555990 DOI: 10.3390/ijms21186887] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2020] [Revised: 09/17/2020] [Accepted: 09/18/2020] [Indexed: 12/13/2022] Open
Abstract
Sarcopenic obesity (SOB), which is closely related to being elderly as a feature of aging, is recently gaining attention because it is associated with many other age-related diseases that present as altered intercellular communication, dysregulated nutrient sensing, and mitochondrial dysfunction. Along with insulin resistance and inflammation as the core pathogenesis of SOB, autophagy has recently gained attention as a significant mechanism of muscle aging in SOB. Known as important cellular metabolic regulators, the AMP-activated protein kinase (AMPK) and the peroxisome proliferator-activated receptor-gamma coactivator-1 alpha (PGC-1α) signaling pathways play an important role in autophagy, inflammation, and insulin resistance, as well as mutual communication between skeletal muscle, adipose tissue, and the liver. Furthermore, AMPK and PGC-1α signaling pathways are implicated in the gut microbiome-muscle axis. In this review, we describe the pathological link between SOB and its associated complications such as metabolic, cardiovascular, and liver disease, falls and fractures, osteoarthritis, pulmonary disease, and mental health via dysregulated autophagy controlled by AMPK and/or PGC-1α signaling pathways. Here, we propose potential treatments for SOB by modulating autophagy activity and gut dysbiosis based on plausible pathological links.
Collapse
|
352
|
Chen FX, Du N, Hu J, Ning F, Mei X, Li Q, Peng L. Intramuscular accumulation of pentadecanoic acid activates AKT1 to phosphorylate NCOR1 and triggers FOXM1-mediated apoptosis in the pathogenesis of sarcopenia. Am J Transl Res 2020; 12:5064-5079. [PMID: 33042406 PMCID: PMC7540105] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Accepted: 08/23/2020] [Indexed: 06/11/2023]
Abstract
Sarcopenia is an age-associated disorder that results in skeletal muscle loss. Apoptosis and inflammation are the two major contributors to sarcopenia. Emerging evidence has shown that long-chain fatty acids (LCFAs) are implicated in the muscles of sarcopenic animal models. However, it is unknown whether LCFAs are correlated with apoptosis or inflammation in the pathogenesis of sarcopenia. Herein, we found that pentadecanoic acid (PDA), a C15 LCFA, was significantly accumulated in human sarcopenic muscles. In vitro PDA treatment could dose-dependently induce the expression of the transcription factor FOXM1 (forkhead box M1) and several proapoptotic genes, such as PUMA (p53-upregulated modulator of apoptosis), BAX (B-cell/lymphoma 2-associated X) and APAF1 (apoptotic peptidase activating factor 1), thereby causing apoptosis. Mechanically, PDA activated AKT1 (AKT serine/threonine kinase 1) to phosphorylate NCOR1 (nuclear receptor corepressor 1). The phosphorylated NCOR1 disassociated from the NCOR1-FOXM1 transcriptional complex and could not repress FOXM1-mediated transcription, leading to the induction of PUMA. The activated PUMA further triggered downstream apoptotic signaling, including activation of the BAX, APAF1 and caspase cascades, leading to the occurrence of apoptosis. Alkaline phosphatase or knockdown of AKT1 in vitro reversed the FOXM1-mediated apoptotic signaling. Collectively, our results provide new evidence that LCFAs are involved in the pathogenesis of sarcopenia by activating apoptotic signaling. Attempts to decrease the intake of PDA-containing foods or blocking AKT1 may improve the symptoms of sarcopenia.
Collapse
Affiliation(s)
- Fa-Xiu Chen
- Department of Geriatrics, Jiangxi Provincial People's Hospital Affiliated to Nanchang University Nanchang 330006, Jiangxi, China
| | - Ning Du
- Department of Geriatrics, Jiangxi Provincial People's Hospital Affiliated to Nanchang University Nanchang 330006, Jiangxi, China
| | - Jian Hu
- Department of Geriatrics, Jiangxi Provincial People's Hospital Affiliated to Nanchang University Nanchang 330006, Jiangxi, China
| | - Fang Ning
- Department of Geriatrics, Jiangxi Provincial People's Hospital Affiliated to Nanchang University Nanchang 330006, Jiangxi, China
| | - Xun Mei
- Department of Geriatrics, Jiangxi Provincial People's Hospital Affiliated to Nanchang University Nanchang 330006, Jiangxi, China
| | - Qiang Li
- Department of Geriatrics, Jiangxi Provincial People's Hospital Affiliated to Nanchang University Nanchang 330006, Jiangxi, China
| | - Le Peng
- Department of Geriatrics, Jiangxi Provincial People's Hospital Affiliated to Nanchang University Nanchang 330006, Jiangxi, China
| |
Collapse
|
353
|
Frasca D, Blomberg BB. Adipose tissue, immune aging, and cellular senescence. Semin Immunopathol 2020; 42:573-587. [PMID: 32785750 DOI: 10.1007/s00281-020-00812-1] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Accepted: 07/28/2020] [Indexed: 12/14/2022]
Abstract
Obesity represents a serious health problem as it is rapidly increasing worldwide. Obesity is associated with reduced healthspan and lifespan, decreased responses to infections and vaccination, and increased frequency of inflammatory conditions typical of old age. Obesity is characterized by increased fat mass and remodeling of the adipose tissue (AT). In this review, we summarize published data on the different types of AT present in mice and humans, and their roles as fat storage as well as endocrine and immune tissues. We review the age-induced changes, including those in the distribution of fat in the body, in abundance and function of adipocytes and their precursors, and in the infiltration of immune cells from the peripheral blood. We also show that cells with a senescent-associated secretory phenotype accumulate in the AT of mice and humans with age, where they secrete several factors involved in the establishment and maintenance of local inflammation, oxidative stress, cell death, tissue remodeling, and infiltration of pro-inflammatory immune cells. Not only adipocytes and pre-adipocytes but also immune cells show a senescent phenotype in the AT. With the increase in human lifespan, it is crucial to identify strategies of intervention and target senescent cells in the AT to reduce local and systemic inflammation and the development of age-associated diseases. Several studies have indeed shown that senescent cells can be effectively targeted in the AT by selectively removing them or by inhibiting the pathways that lead to the secretion of pro-inflammatory factors.
Collapse
Affiliation(s)
- Daniela Frasca
- 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.
| | - 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
|
354
|
Yamada T, Kamiya M, Higuchi M. Breed differences in macrophage infiltration and senescence state in adipose tissues of Wagyu and Holsteins. Anim Sci J 2020; 91:e13443. [PMID: 32779259 DOI: 10.1111/asj.13443] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2020] [Revised: 06/07/2020] [Accepted: 07/15/2020] [Indexed: 11/30/2022]
Abstract
Obesity stimulates the macrophage infiltration and senescence state in adipose tissues of humans and rodents. The adipogenesis capacity of Japanese Black cattle (Wagyu) is higher than that of Holsteins. We hypothesized that breed differences between Wagyu and Holsteins may affect the level of macrophage infiltration and senescence state in adipose tissues. The macrophage infiltration, senescence marker gene expression and activity of senescence-associated β-galactosidase (SA-βgal) in visceral and intramuscular adipose tissue of Wagyu were higher than those of Holsteins. In contrast, there were no differences in macrophage infiltration, senescence marker gene expression and activity of SA-βgal in subcutaneous adipose tissue between the breeds. Expression of p53 gene, the master regulator of macrophage infiltration and senescence state, in visceral and intramuscular adipose tissue of Wagyu was higher than that of Holsteins. In contrast, there was no difference in the expression of p53 gene in subcutaneous adipose tissue between the breeds. These results suggest that breed differences in macrophage infiltration and senescence state in adipose tissues of Wagyu and Holsteins are affected by p53 expression.
Collapse
Affiliation(s)
- Tomoya Yamada
- Division of Livestock Feeding and Management, National Agriculture and Food Research Organization, Nasushiobara-shi, Japan
| | - Mituru Kamiya
- Division of Livestock Feeding and Management, National Agriculture and Food Research Organization, Nasushiobara-shi, Japan
| | - Mikito Higuchi
- Division of Livestock Feeding and Management, National Agriculture and Food Research Organization, Nasushiobara-shi, Japan
| |
Collapse
|
355
|
Wissler Gerdes EO, Zhu Y, Weigand BM, Tripathi U, Burns TC, Tchkonia T, Kirkland JL. Cellular senescence in aging and age-related diseases: Implications for neurodegenerative diseases. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2020; 155:203-234. [PMID: 32854855 PMCID: PMC7656525 DOI: 10.1016/bs.irn.2020.03.019] [Citation(s) in RCA: 64] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Aging is the major predictor for developing multiple neurodegenerative diseases, including Alzheimer's disease (AD) other dementias, and Parkinson's disease (PD). Senescent cells, which can drive aging phenotypes, accumulate at etiological sites of many age-related chronic diseases. These cells are resistant to apoptosis and can cause local and systemic dysfunction. Decreasing senescent cell abundance using senolytic drugs, agents that selectively target these cells, alleviates neurodegenerative diseases in preclinical models. In this review, we consider roles of senescent cells in neurodegenerative diseases and potential implications of senolytic agents as an innovative treatment.
Collapse
Affiliation(s)
| | - Yi Zhu
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, MN, United States
| | - B Melanie Weigand
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, MN, United States
| | - Utkarsh Tripathi
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, MN, United States
| | - Terence C Burns
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, MN, United States
| | - Tamar Tchkonia
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, MN, United States
| | - James L Kirkland
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, MN, United States.
| |
Collapse
|
356
|
Wyld L, Bellantuono I, Tchkonia T, Morgan J, Turner O, Foss F, George J, Danson S, Kirkland JL. Senescence and Cancer: A Review of Clinical Implications of Senescence and Senotherapies. Cancers (Basel) 2020; 12:cancers12082134. [PMID: 32752135 PMCID: PMC7464619 DOI: 10.3390/cancers12082134] [Citation(s) in RCA: 156] [Impact Index Per Article: 31.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Revised: 07/27/2020] [Accepted: 07/28/2020] [Indexed: 12/19/2022] Open
Abstract
Cellular senescence is a key component of human aging that can be induced by a range of stimuli, including DNA damage, cellular stress, telomere shortening, and the activation of oncogenes. Senescence is generally regarded as a tumour suppressive process, both by preventing cancer cell proliferation and suppressing malignant progression from pre-malignant to malignant disease. It may also be a key effector mechanism of many types of anticancer therapies, such as chemotherapy, radiotherapy, and endocrine therapies, both directly and via bioactive molecules released by senescent cells that may stimulate an immune response. However, senescence may contribute to reduced patient resilience to cancer therapies and may provide a pathway for disease recurrence after cancer therapy. A new group of drugs, senotherapies, (drugs which interact with senescent cells to interfere with their pro-aging impacts by either selectively destroying senescent cells (senolytic drugs) or inhibiting their function (senostatic drugs)) are under active investigation to determine whether they can enhance the efficacy of cancer therapies and improve resilience to cancer treatments. Senolytic drugs include quercetin, navitoclax, and fisetin and preclinical and early phase clinical data are emerging of their potential role in cancer treatments, although none are yet in routine use clinically. This article provides a review of these issues.
Collapse
Affiliation(s)
- Lynda Wyld
- The Healthy Lifespan Institute, Department of Oncology and Metabolism, The Medical School, University of Sheffield, Beech Hill Road, Sheffield S10 2RX, UK; (I.B.); (J.M.); (O.T.); (J.G.); (S.D.)
- Correspondence:
| | - Ilaria Bellantuono
- The Healthy Lifespan Institute, Department of Oncology and Metabolism, The Medical School, University of Sheffield, Beech Hill Road, Sheffield S10 2RX, UK; (I.B.); (J.M.); (O.T.); (J.G.); (S.D.)
| | - Tamara Tchkonia
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, MN 55905, USA;
| | - Jenna Morgan
- The Healthy Lifespan Institute, Department of Oncology and Metabolism, The Medical School, University of Sheffield, Beech Hill Road, Sheffield S10 2RX, UK; (I.B.); (J.M.); (O.T.); (J.G.); (S.D.)
| | - Olivia Turner
- The Healthy Lifespan Institute, Department of Oncology and Metabolism, The Medical School, University of Sheffield, Beech Hill Road, Sheffield S10 2RX, UK; (I.B.); (J.M.); (O.T.); (J.G.); (S.D.)
| | - Fiona Foss
- Department of Pathology, Sheffield Teaching Hospitals NHS Foundation Trust, Sheffield S10 2JF, UK;
| | - Jayan George
- The Healthy Lifespan Institute, Department of Oncology and Metabolism, The Medical School, University of Sheffield, Beech Hill Road, Sheffield S10 2RX, UK; (I.B.); (J.M.); (O.T.); (J.G.); (S.D.)
| | - Sarah Danson
- The Healthy Lifespan Institute, Department of Oncology and Metabolism, The Medical School, University of Sheffield, Beech Hill Road, Sheffield S10 2RX, UK; (I.B.); (J.M.); (O.T.); (J.G.); (S.D.)
| | - James L. Kirkland
- Departments of Internal Medicine, Geriatric Medicine and Gerontology, The Mayo Clinic, Rochester, MN 55905, USA;
| |
Collapse
|
357
|
Gourronc FA, Markan KR, Kulhankova K, Zhu Z, Sheehy R, Quelle DE, Zingman LV, Kurago ZB, Ankrum JA, Klingelhutz AJ. Pdgfrα-Cre mediated knockout of the aryl hydrocarbon receptor protects mice from high-fat diet induced obesity and hepatic steatosis. PLoS One 2020; 15:e0236741. [PMID: 32730300 PMCID: PMC7392206 DOI: 10.1371/journal.pone.0236741] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2020] [Accepted: 07/13/2020] [Indexed: 01/04/2023] Open
Abstract
Aryl hydrocarbon receptor (AHR) agonists such as dioxin have been associated with obesity and the development of diabetes. Whole-body Ahr knockout mice on high-fat diet (HFD) have been shown to resist obesity and hepatic steatosis. Tissue-specific knockout of Ahr in mature adipocytes via adiponectin-Cre exacerbates obesity while knockout in liver increases steatosis without having significant effects on obesity. Our previous studies demonstrated that treatment of subcutaneous preadipocytes with exogenous or endogenous AHR agonists disrupts maturation into functional adipocytes in vitro. Here, we used platelet-derived growth factor receptor alpha (Pdgfrα)-Cre mice, a Cre model previously established to knock out genes in preadipocyte lineages and other cell types, but not liver cells, to further define AHR's role in obesity. We demonstrate that Pdgfrα-Cre Ahr-floxed (Ahrfl/fl) knockout mice are protected from HFD-induced obesity compared to non-knockout Ahrfl/fl mice (control mice). The Pdgfrα-Cre Ahrfl/fl knockout mice were also protected from increased adiposity, enlargement of adipocyte size, and liver steatosis while on the HFD compared to control mice. On a regular control diet, knockout and non-knockout mice showed no differences in weight gain, indicating the protective phenotype arises only when animals are challenged by a HFD. At the cellular level, cultured cells from brown adipose tissue (BAT) of Pdgfrα-Cre Ahrfl/fl mice were more responsive than cells from controls to transcriptional activation of the thermogenic uncoupling protein 1 (Ucp1) gene by norepinephrine, suggesting an ability to burn more energy under certain conditions. Collectively, our results show that knockout of Ahr mediated by Pdgfrα-Cre is protective against diet-induced obesity and suggest a mechanism by which enhanced UCP1 activity within BAT might confer these effects.
Collapse
Affiliation(s)
- Francoise A. Gourronc
- Department of Microbiology and Immunology, University of Iowa, Iowa City, IA, United States of America
| | - Kathleen R. Markan
- Department of Neuroscience and Pharmacology, Fraternal Order of Eagles Diabetes Research Center, University of Iowa, Iowa City, IA, United States of America
| | - Katarina Kulhankova
- Department of Pediatrics, University of Iowa, Iowa City, IA, United States of America
| | - Zhiyong Zhu
- Department of Internal Medicine, University of Iowa, Iowa City, IA, United States of America
| | - Ryan Sheehy
- Department of Pharmacology, Kansas City University, Kansas City, KS, United States of America
| | - Dawn E. Quelle
- Department of Neuroscience and Pharmacology, Fraternal Order of Eagles Diabetes Research Center, University of Iowa, Iowa City, IA, United States of America
| | - Leonid V. Zingman
- Department of Internal Medicine, Fraternal Order of Eagles Diabetes Research Center, University of Iowa, Iowa City, IA, United States of America
| | - Zoya B. Kurago
- Department of Oral Biology and Diagnostic Sciences, Department of Pathology, Augusta University, Augusta, GA, United States of America
| | - James A. Ankrum
- Roy J. Carver Department of Biomedical Engineering, Fraternal Order of Eagles Diabetes Research Center, University of Iowa, Iowa City, IA, United States of America
| | - Aloysius J. Klingelhutz
- Department of Microbiology and Immunology, Fraternal Order of Eagles Diabetes Research Center, University of Iowa, Iowa City, IA, United States of America
- * E-mail:
| |
Collapse
|
358
|
Álvaro-Afonso FJ, Sanz-Corbalán I, Lázaro-Martínez JL, Kakagia D, Papanas N. Adipose-Derived Mesenchymal Stem Cells in the Treatment of Diabetic Foot Ulcers: A Review of Preclinical and Clinical Studies. Angiology 2020; 71:853-863. [PMID: 32723090 DOI: 10.1177/0003319720939467] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
This review provides an outline of the use of adipose-derived mesenchymal stem cells (AMSCs) in the treatment of diabetic foot ulcers (DFUs). A systematic search of PubMed and the Cochrane database was performed on October 2, 2019. Eighteen studies were identified (14 preclinical and 4 clinical). Studies in animal models have demonstrated that AMSCs enhance diabetic wound healing, accelerate granulation tissue formation, and increase reepithelialization and neovascularization. Only 1 randomized control trial has been published so far. Patients (n = 25) with DFUs were treated using an allogeneic AMSC directly on the wound bed as a primary dressing, and improvements were found in complete wound closure in the treatment group (n = 16). Three clinical studies showed that autologous AMSC might be a safe alternative to achieve therapeutic angiogenesis in patients with diabetes and peripheral arterial disease. Based on the available evidence, AMSCs hold promise in the treatment of DFUs. However, this evidence requires confirmation by well-designed trials. Additional studies are also required to understand some issues regarding this treatment for DFUs. For example, the potential application of autologous or allogeneic AMSCs in different types of DFUs, optimal dose/infusion schedules, safety evaluations, and cost-effectiveness.
Collapse
Affiliation(s)
- Francisco Javier Álvaro-Afonso
- Diabetic Foot Unit, University Podiatric Clinic, Edificio Facultad de Medicina, 16734Complutense University of Madrid, Instituto de Investigación Sanitaria del Hospital Clínico San Carlos (IdISSC), Madrid, Spain
| | - Irene Sanz-Corbalán
- Diabetic Foot Unit, University Podiatric Clinic, Edificio Facultad de Medicina, 16734Complutense University of Madrid, Instituto de Investigación Sanitaria del Hospital Clínico San Carlos (IdISSC), Madrid, Spain
| | - José Luis Lázaro-Martínez
- Diabetic Foot Unit, University Podiatric Clinic, Edificio Facultad de Medicina, 16734Complutense University of Madrid, Instituto de Investigación Sanitaria del Hospital Clínico San Carlos (IdISSC), Madrid, Spain
| | - Despoina Kakagia
- Department of Plastic Surgery, Democritus University of Thrace, University Hospital of Alexandroupolis, Greece
| | - Nikolaos Papanas
- Diabetes Centre-Diabetic Foot Clinic, Second Department of Internal Medicine, Democritus University of Thrace, University Hospital of Alexandroupolis, Greece
| |
Collapse
|
359
|
Agosti E, De Feudis M, Angelino E, Belli R, Alves Teixeira M, Zaggia I, Tamiso E, Raiteri T, Scircoli A, Ronzoni FL, Muscaritoli M, Graziani A, Prodam F, Sampaolesi M, Costelli P, Ferraro E, Reano S, Filigheddu N. Both ghrelin deletion and unacylated ghrelin overexpression preserve muscles in aging mice. Aging (Albany NY) 2020; 12:13939-13957. [PMID: 32712599 PMCID: PMC7425472 DOI: 10.18632/aging.103802] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2020] [Accepted: 07/13/2020] [Indexed: 02/06/2023]
Abstract
Sarcopenia, the decline in muscle mass and functionality during aging, might arise from age-associated endocrine dysfunction. Ghrelin is a hormone circulating in both acylated (AG) and unacylated (UnAG) forms with anti-atrophic activity on skeletal muscle. Here, we show that not only lifelong overexpression of UnAG (Tg) in mice, but also the deletion of ghrelin gene (Ghrl KO) attenuated the age-associated muscle atrophy and functionality decline, as well as systemic inflammation. Yet, the aging of Tg and Ghrl KO mice occurs with different dynamics: while old Tg mice seem to preserve the characteristics of young animals, Ghrl KO mice features deteriorate with aging. However, young Ghrl KO mice show more favorable traits compared to WT animals that result, on the whole, in better performances in aged Ghrl KO animals. Treatment with pharmacological doses of UnAG improved muscle performance in old mice without modifying the feeding behavior, body weight, and adipose tissue mass. The antiatrophic effect on muscle mass did not correlate with modifications of protein catabolism. However, UnAG treatment induced a strong shift towards oxidative metabolism in muscle. Altogether, these data confirmed and expanded some of the previously reported findings and advocate for the design of UnAG analogs to treat sarcopenia.
Collapse
Affiliation(s)
- Emanuela Agosti
- Department of Translational Medicine, University of Piemonte Orientale, Novara, Italy.,Department of Health Sciences, University of Piemonte Orientale, Novara, Italy
| | - Marilisa De Feudis
- Department of Translational Medicine, University of Piemonte Orientale, Novara, Italy
| | - Elia Angelino
- Division of Oncology, San Raffaele Scientific Institute and Vita-Salute San Raffaele University, Milano, Italy.,Molecular Biotechnology Center, Department of Molecular Biotechnology and Health Sciences, University of Torino, Turin, Italy
| | - Roberta Belli
- Department of Translational and Precision Medicine, Sapienza University, Rome, Italy
| | | | - Ivan Zaggia
- Department of Translational Medicine, University of Piemonte Orientale, Novara, Italy
| | - Edoardo Tamiso
- Department of Translational Medicine, University of Piemonte Orientale, Novara, Italy
| | - Tommaso Raiteri
- Department of Translational Medicine, University of Piemonte Orientale, Novara, Italy
| | - Andrea Scircoli
- Department of Translational Medicine, University of Piemonte Orientale, Novara, Italy
| | - Flavio L Ronzoni
- Department of Public Health, Experimental and Forensic Medicine, Institute of Human Anatomy, University of Pavia, Pavia, Italy.,Department of Biomedical Sciences, Humanitas University, Rozzano, Italy
| | - Maurizio Muscaritoli
- Department of Translational and Precision Medicine, Sapienza University, Rome, Italy
| | - Andrea Graziani
- Division of Oncology, San Raffaele Scientific Institute and Vita-Salute San Raffaele University, Milano, Italy.,Molecular Biotechnology Center, Department of Molecular Biotechnology and Health Sciences, University of Torino, Turin, Italy
| | - Flavia Prodam
- Department of Health Sciences, University of Piemonte Orientale, Novara, Italy
| | - Maurilio Sampaolesi
- Department of Public Health, Experimental and Forensic Medicine, Institute of Human Anatomy, University of Pavia, Pavia, Italy.,Center for Health Technologies (CHT), University of Pavia, Pavia, Italy.,Stem Cell Institute, KU Leuven, Leuven, Belgium.,Istituto Interuniversitario di Miologia (IIM)
| | - Paola Costelli
- Department of Clinical and Biological Sciences, University of Torino, Turin, Italy.,Istituto Interuniversitario di Miologia (IIM)
| | - Elisabetta Ferraro
- Division of Orthopaedics and Traumatology, Hospital "Maggiore della Carità", Novara, Italy
| | - Simone Reano
- Department of Translational Medicine, University of Piemonte Orientale, Novara, Italy
| | - Nicoletta Filigheddu
- Department of Translational Medicine, University of Piemonte Orientale, Novara, Italy.,Istituto Interuniversitario di Miologia (IIM)
| |
Collapse
|
360
|
Biomarkers of senescence in non-human primate adipose depots relate to aging. GeroScience 2020; 43:343-352. [PMID: 32705409 DOI: 10.1007/s11357-020-00230-z] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Accepted: 07/07/2020] [Indexed: 10/23/2022] Open
Abstract
Forty-three female African green monkeys (Chlorocebus aethiops sabaeus) were selected to represent young adult to advanced geriatric ages (7-24 years) to exhibit a wide range of obesity status (8-53% body fat) and diverse metabolic syndrome criteria such as diabetes, dyslipidemia, and hypertension. Subcutaneous and visceral adipose tissues were collected and evaluated for the presence of senescence cells in both whole tissue and single-cell isolates from subcutaneous sources, utilizing senescence-associated β-galactosidase (SAβ-gal) staining. Plasma samples were analyzed for selected metabolic and inflammatory biomarkers related to the senescence-associated secretory profile. Our results indicated that tissue staining scores did not differ between subcutaneous and intra-abdominal visceral depots and were highly related within individuals. Tissue staining was significantly associated with chronological age; however, no associations with fatness or metabolic syndrome criteria were observed. Associations with age were unchanged when obesity status was included in regression models. Isolated cell staining did positively relate to age but not tissue staining, suggesting some of the SAβ-gal-positive cells were stromal vascular cells or small adipocytes, but that mature large adipocytes, filtered out in the cell isolation process, are also likely to exhibit positive SAβ-gal staining. Plasminogen activator inhibitor-1 (PAI-1) concentration in circulation was the sole inflammation-related biomarker that positively associated with age and is considered to be a marker of senescent cell burden. Our study is the largest, most comprehensive assessment of adipose SAβ-gal staining in a relevant animal model of human aging, and confirms that this senescence-associated biomarker specifically indicates an age-related process.
Collapse
|
361
|
Föger-Samwald U, Dovjak P, Azizi-Semrad U, Kerschan-Schindl K, Pietschmann P. Osteoporosis: Pathophysiology and therapeutic options. EXCLI JOURNAL 2020; 19:1017-1037. [PMID: 32788914 PMCID: PMC7415937 DOI: 10.17179/excli2020-2591] [Citation(s) in RCA: 90] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Accepted: 07/07/2020] [Indexed: 12/12/2022]
Abstract
Osteoporosis is a metabolic bone disease that, on a cellular level, results from osteoclastic bone resorption not compensated by osteoblastic bone formation. This causes bones to become weak and fragile, thus increasing the risk of fractures. Traditional pathophysiological concepts of osteoporosis focused on endocrine mechanisms such as estrogen or vitamin D deficiency as well as secondary hyperparathyroidism. However, research over the last decades provided exiting new insights into mechanisms contributing to the onset of osteoporosis, which go far beyond this. Selected mechanisms such as interactions between bone and the immune system, the gut microbiome, and cellular senescence are reviewed in this article. Furthermore, an overview on currently available osteoporosis medications including antiresorptive and bone forming drugs is provided and an outlook on potential future treatment options is given.
Collapse
Affiliation(s)
- Ursula Föger-Samwald
- Institute of Pathophysiology and Allergy Research, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| | - Peter Dovjak
- Department of Acute Geriatrics, Salzkammergut Klinikum Gmunden, Gmunden, Austria
| | - Ursula Azizi-Semrad
- Department of Emergency Medicine, Medical University of Vienna, Vienna, Austria
| | - Katharina Kerschan-Schindl
- Department of Physical Medicine, Rehabilitation and Occupational Medicine, Medical University of Vienna, Vienna, Austria
| | - Peter Pietschmann
- Institute of Pathophysiology and Allergy Research, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
362
|
Bone marrow fat: friend or foe in people with diabetes mellitus? Clin Sci (Lond) 2020; 134:1031-1048. [PMID: 32337536 DOI: 10.1042/cs20200220] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2020] [Revised: 04/15/2020] [Accepted: 04/20/2020] [Indexed: 12/22/2022]
Abstract
Global trends in the prevalence of overweight and obesity put the adipocyte in the focus of huge medical interest. This review highlights a new topic in adipose tissue biology, namely the emerging pathogenic role of fat accumulation in bone marrow (BM). Specifically, we summarize current knowledge about the origin and function of BM adipose tissue (BMAT), provide evidence for the association of excess BMAT with diabetes and related cardiovascular complications, and discuss potential therapeutic approaches to correct BMAT dysfunction. There is still a significant uncertainty about the origins and function of BMAT, although several subpopulations of stromal cells have been suggested to have an adipogenic propensity. BM adipocytes are higly plastic and have a distinctive capacity to secrete adipokines that exert local and endocrine functions. BM adiposity is abundant in elderly people and has therefore been interpreted as a component of the whole-body ageing process. BM senescence and BMAT accumulation has been also reported in patients and animal models with Type 2 diabetes, being more pronounced in those with ischaemic complications. Understanding the mechanisms responsible for excess and altered function of BMAT could lead to new treatments able to preserve whole-body homeostasis.
Collapse
|
363
|
The role of adipose tissue senescence in obesity- and ageing-related metabolic disorders. Clin Sci (Lond) 2020; 134:315-330. [PMID: 31998947 DOI: 10.1042/cs20190966] [Citation(s) in RCA: 84] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2019] [Revised: 01/07/2020] [Accepted: 01/14/2020] [Indexed: 12/19/2022]
Abstract
Adipose tissue as the largest energy reservoir and endocrine organ is essential for maintenance of systemic glucose, lipid and energy homeostasis, but these metabolic functions decline with ageing and obesity. Adipose tissue senescence is one of the common features in obesity and ageing. Although cellular senescence is a defensive mechanism preventing tumorigenesis, its occurrence in adipose tissue causatively induces defective adipogenesis, inflammation, aberrant adipocytokines production and insulin resistance, leading to adipose tissue dysfunction. In addition to these paracrine effects, adipose tissue senescence also triggers systemic inflammation and senescence as well as insulin resistance in the distal metabolic organs, resulting in Type 2 diabetes and other premature physiological declines. Multiple cell types including mature adipocytes, immune cells, endothelial cells and progenitor cells gradually senesce at different levels in different fat depots with ageing and obesity, highlighting the heterogeneity and complexity of adipose tissue senescence. In this review, we discuss the causes and consequences of adipose tissue senescence, and the major cell types responsible for adipose tissue senescence in ageing and obesity. In addition, we summarize the pharmacological approaches and lifestyle intervention targeting adipose tissue senescence for the treatment of obesity- and ageing-related metabolic diseases.
Collapse
|
364
|
Kim YJ, Tu TH, Yang S, Kim JK, Kim JG. Characterization of Fatty Acid Composition Underlying Hypothalamic Inflammation in Aged Mice. Molecules 2020; 25:molecules25143170. [PMID: 32664475 PMCID: PMC7397167 DOI: 10.3390/molecules25143170] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2020] [Revised: 07/07/2020] [Accepted: 07/09/2020] [Indexed: 02/07/2023] Open
Abstract
Degenerative diseases, which can develop during aging, are underlined by inflammatory processes. Hypothalamic inflammation triggered by elevation in circulating fatty acid levels is directly coupled to metabolic disorders. The present study aimed to investigate and characterize the hypothalamic inflammation and composition of fatty acids in the hypothalami of aged mice. We verified that inflammation and microglial activation occur in the hypothalami of aged mice by performing quantitative real-time PCR and using immunohistochemistry methods. In addition, we observed increased levels of various saturated fatty acids in the hypothalami of aged mice, whereas no major changes in the levels of circulating fatty acids were detected using gas chromatography with a flame ionization detector. Collectively, our current findings suggest that increases in saturated fatty acid levels are coupled to hypothalamic inflammation and thereby cause perturbations in energy metabolism during the aging process.
Collapse
Affiliation(s)
- Ye Jin Kim
- Division of Life Sciences, College of Life Sciences and Bioengineering, Incheon National University, Incheon 406–772, Korea; (Y.J.K.); (T.H.T.)
| | - Thai Hien Tu
- Division of Life Sciences, College of Life Sciences and Bioengineering, Incheon National University, Incheon 406–772, Korea; (Y.J.K.); (T.H.T.)
| | - Sunggu Yang
- Department of Nano-Bioengineering, Incheon National University, Incheon 406–772, Korea;
| | - Jae Kwang Kim
- Division of Life Sciences, College of Life Sciences and Bioengineering, Incheon National University, Incheon 406–772, Korea; (Y.J.K.); (T.H.T.)
- Division of Life Sciences and Bio-Resource and Environmental Center, Incheon National University, Incheon 406–772, Korea
- Correspondence: (J.K.K.); (J.G.K.); Tel.: +82-32-835-8241 (J.K.K.); +82-32-835-8256 (J.G.K.)
| | - Jae Geun Kim
- Division of Life Sciences, College of Life Sciences and Bioengineering, Incheon National University, Incheon 406–772, Korea; (Y.J.K.); (T.H.T.)
- Correspondence: (J.K.K.); (J.G.K.); Tel.: +82-32-835-8241 (J.K.K.); +82-32-835-8256 (J.G.K.)
| |
Collapse
|
365
|
D-galactose: a model of accelerated ageing sufficiently sensitive to reflect preventative efficacy of an antioxidant treatment. Biogerontology 2020; 21:745-761. [PMID: 32638260 DOI: 10.1007/s10522-020-09891-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2020] [Accepted: 07/02/2020] [Indexed: 02/07/2023]
Abstract
Considering that the phenomenon of accelerated ageing contributes to early onset of various chronic diseases, modelling of the relevant dysregulated systems or responses is vital for research aimed at identification of potential therapeutic targets. Here, we aimed to establish a model capable of simulating the redox and inflammatory changes of accelerated ageing-specifically, the aim was early phase accelerated ageing, which would allow therapeutic intervention in a preventative approach prior to clinical disease manifestation. A secondary aim was to evaluate the sensitivity of the model to reflect preventative treatment efficacy. Daily D-galactose injections (250 mg/kg body mass/day) for 8 weeks in 9-week-old male Wistar rats induced a model of early accelerated ageing (decreased plasma FRAP; P < 0.05 and altered inflammatory signalling) and an aged profile in lymph node ultrastructure, but did not yet result in telomere shortening. Preventative daily oral antioxidant administration (grape seed-derived polyphenol, 100 mg/kg body mass) prevented tissue ageing, beneficially modulated the inflammatory response, including neutrophil chemokinetic capacity, and tended to increase absolute telomere length. Data suggests that using a mild model of D-galactose administration than those employed to induce neurodegeneration, simulated the point where oxidative stress starts to overwhelm the endogenous antioxidant response and where a pro-inflammatory phenotype switch manifests. Furthermore, despite the expected small effect size, the model was sufficiently sensitive to reflect benefits of preventative antioxidant treatment in the context of ageing. This model presents a practical model for use in drug discovery, particularly in the context of preventative medicine aimed at limiting oxidative stress-associated ageing. Since this starting point of accelerated ageing as illustrated by current data, is not expected to reflect major ageing-associated changes yet, we recommend that future preventative drug discovery studies employ a longitudinal study design in order to clearly demonstrate the delay of this starting point by preventative strategies.
Collapse
|
366
|
Kumar A, Bano D, Ehninger D. Cellular senescence in vivo: From cells to tissues to pathologies. Mech Ageing Dev 2020; 190:111308. [PMID: 32622996 DOI: 10.1016/j.mad.2020.111308] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2020] [Revised: 06/24/2020] [Accepted: 06/26/2020] [Indexed: 01/22/2023]
Abstract
Senescent cells accumulate during aging in a variety of tissues. Although scarce, they could influence tissue function non-cell-autonomously via secretion of a range of factors in their neighborhood. Recent studies support a role of senescent cells in age-related morbidity, including neurodegenerative diseases, cardiovascular pathologies, cancers, aging-associated nephrological alterations, chronic pulmonary disease and osteoarthritis, indicating that senescent cells could represent an interesting target for therapeutic exploitation across a range of pathophysiological contexts. In this article, we review data available to indicate which cell types can undergo senescence within various mammalian tissue environments and how these processes may contribute to tissue-specific pathologies associated with old age. We also consider markers used to identify senescent cells in vitro and in vivo. The data discussed may serve as an important starting point for an extended definition of molecular and functional characteristics of senescent cells in different organs and may hence promote the development and refinement of targeting strategies aimed at removing senescent cells from aging tissues.
Collapse
Affiliation(s)
- Avadh Kumar
- Translational Biogerontology Lab, German Center for Neurodegenerative Diseases (DZNE), Venusberg-Campus 1, 53127 Bonn, Germany
| | - Daniele Bano
- Aging and Neurodegeneration Lab, German Center for Neurodegenerative Diseases (DZNE), Venusberg-Campus 1, 53127 Bonn, Germany
| | - Dan Ehninger
- Translational Biogerontology Lab, German Center for Neurodegenerative Diseases (DZNE), Venusberg-Campus 1, 53127 Bonn, Germany.
| |
Collapse
|
367
|
Pires GN, Benedetto L, Cortese R, Gozal D, Gulia KK, Kumar VM, Tufik S, Andersen ML. Effects of sleep modulation during pregnancy in the mother and offspring: Evidences from preclinical research. J Sleep Res 2020; 30:e13135. [PMID: 32618040 DOI: 10.1111/jsr.13135] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2020] [Revised: 06/04/2020] [Accepted: 06/09/2020] [Indexed: 12/18/2022]
Abstract
Disturbed sleep during gestation may lead to adverse outcomes for both mother and child. Animal research plays an important role in providing insights into this research field by enabling ethical and methodological requirements that are not possible in humans. Here, we present an overview and discuss the main research findings related to the effects of prenatal sleep deprivation in animal models. Using systematic review approaches, we retrieved 42 articles dealing with some type of sleep alteration. The most frequent research topics in this context were maternal sleep deprivation, maternal behaviour, offspring behaviour, development of sleep-wake cycles in the offspring, hippocampal neurodevelopment, pregnancy viability, renal physiology, hypertension and metabolism. This overview indicates that the number of basic studies in this field is growing, and provides biological plausibility to suggest that sleep disturbances might be detrimental to both mother and offspring by promoting increased risk at the behavioural, hormonal, electrophysiological, metabolic and epigenetic levels. More studies on the effects of maternal sleep deprivation are needed, in light of their major translational perspective.
Collapse
Affiliation(s)
- Gabriel Natan Pires
- Departamento de Psicobiologia, Universidade Federal de São Paulo, São Paulo, Brazil.,Department of Physiological Sciences, Santa Casa de São Paulo School of Medical Sciences, São Paulo, Brazil
| | - Luciana Benedetto
- Departamento de Fisiología, Facultad de Medicina, Universidad de la República, Montevideo, Uruguay
| | - Rene Cortese
- Department of Child Health and Child Health Research Institute, University of Missouri School of Medicine, Columbia, MO, USA
| | - David Gozal
- Department of Child Health and Child Health Research Institute, University of Missouri School of Medicine, Columbia, MO, USA
| | - Kamalesh K Gulia
- Division of Sleep Research, Biomedical Technology Wing - Sree Chitra Tirunal Institute for Medical Sciences and Technology, Trivandrum, Kerala, India
| | | | - Sergio Tufik
- Departamento de Psicobiologia, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Monica Levy Andersen
- Departamento de Psicobiologia, Universidade Federal de São Paulo, São Paulo, Brazil
| |
Collapse
|
368
|
Chen J, Jiang Q, Xia X, Liu K, Yu Z, Tao W, Gong W, Han JJ. Individual variation of the SARS-CoV-2 receptor ACE2 gene expression and regulation. Aging Cell 2020; 19:e13168. [PMID: 32558150 PMCID: PMC7323071 DOI: 10.1111/acel.13168] [Citation(s) in RCA: 289] [Impact Index Per Article: 57.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2020] [Revised: 04/28/2020] [Accepted: 05/11/2020] [Indexed: 01/08/2023] Open
Abstract
The COVID-19 coronavirus is now spreading worldwide. Its pathogen, SARS-CoV-2, has been shown to use angiotensin-converting enzyme 2 (ACE2) as its host cell receptor, same as the severe acute respiratory syndrome coronavirus (SARS-CoV) in 2003. Epidemiology studies found males although only slightly more likely to be infected than females account for the majority of the severely ill and fatality, which also bias for people older than 60 years or with metabolic and cardiovascular diseases. Here by analyzing GTEx and other public data in 30 tissues across thousands of individuals, we found a significantly higher level in Asian females, an age-dependent decrease in all ethnic groups, and a highly significant decrease in type II diabetic patients of ACE2 expression. Consistently, the most significant expression quantitative loci (eQTLs) contributing to high ACE2 expression are close to 100% in East Asians, >30% higher than other ethnic groups. A shockingly common enrichment of viral infection pathways was found among ACE2 anti-expressed genes, and multiple binding sites of virus infection related transcription factors and sex hormone receptors locate at ACE2 regulatory regions. Human and mice data analysis further revealed ACE2 expression is reduced in T2D patients and with inflammatory cytokine treatment and upregulated by estrogen and androgen (both decrease with age). Our findings revealed a negative correlation between ACE2 expression and COVID-19 fatality at both population and molecular levels. These results will be instrumental when designing potential prevention and treatment strategies for ACE2 binding coronaviruses in general.
Collapse
Affiliation(s)
- Jiawei Chen
- Peking‐Tsinghua Center for Life SciencesAcademy for Advanced Interdisciplinary StudiesCenter for Quantitative Biology (CQB)Peking UniversityBeijingChina
| | - Quanlong Jiang
- CAS Key Laboratory of Computational BiologyCAS‐MPG Partner Institute for Computational BiologyShanghai Institute of Nutrition and HealthChinese Academy of Sciences Center for Excellence in Molecular Cell ScienceCollaborative Innovation Center for Genetics and Developmental BiologyShanghai Institutes for Biological SciencesChinese Academy of SciencesShanghaiChina
| | - Xian Xia
- CAS Key Laboratory of Computational BiologyCAS‐MPG Partner Institute for Computational BiologyShanghai Institute of Nutrition and HealthChinese Academy of Sciences Center for Excellence in Molecular Cell ScienceCollaborative Innovation Center for Genetics and Developmental BiologyShanghai Institutes for Biological SciencesChinese Academy of SciencesShanghaiChina
| | - Kangping Liu
- Peking‐Tsinghua Center for Life SciencesAcademy for Advanced Interdisciplinary StudiesCenter for Quantitative Biology (CQB)Peking UniversityBeijingChina
| | - Zhengqing Yu
- Peking‐Tsinghua Center for Life SciencesAcademy for Advanced Interdisciplinary StudiesCenter for Quantitative Biology (CQB)Peking UniversityBeijingChina
| | - Wanyu Tao
- Peking‐Tsinghua Center for Life SciencesAcademy for Advanced Interdisciplinary StudiesCenter for Quantitative Biology (CQB)Peking UniversityBeijingChina
| | - Wenxuan Gong
- CAS Key Laboratory of Computational BiologyCAS‐MPG Partner Institute for Computational BiologyShanghai Institute of Nutrition and HealthChinese Academy of Sciences Center for Excellence in Molecular Cell ScienceCollaborative Innovation Center for Genetics and Developmental BiologyShanghai Institutes for Biological SciencesChinese Academy of SciencesShanghaiChina
| | - Jing‐Dong J. Han
- Peking‐Tsinghua Center for Life SciencesAcademy for Advanced Interdisciplinary StudiesCenter for Quantitative Biology (CQB)Peking UniversityBeijingChina
- CAS Key Laboratory of Computational BiologyCAS‐MPG Partner Institute for Computational BiologyShanghai Institute of Nutrition and HealthChinese Academy of Sciences Center for Excellence in Molecular Cell ScienceCollaborative Innovation Center for Genetics and Developmental BiologyShanghai Institutes for Biological SciencesChinese Academy of SciencesShanghaiChina
| |
Collapse
|
369
|
Tencerova M, Frost M, Figeac F, Nielsen TK, Ali D, Lauterlein JJL, Andersen TL, Haakonsson AK, Rauch A, Madsen JS, Ejersted C, Højlund K, Kassem M. Obesity-Associated Hypermetabolism and Accelerated Senescence of Bone Marrow Stromal Stem Cells Suggest a Potential Mechanism for Bone Fragility. Cell Rep 2020; 27:2050-2062.e6. [PMID: 31091445 DOI: 10.1016/j.celrep.2019.04.066] [Citation(s) in RCA: 88] [Impact Index Per Article: 17.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2018] [Revised: 03/06/2019] [Accepted: 04/12/2019] [Indexed: 02/07/2023] Open
Abstract
Obesity is associated with increased risk for fragility fractures. However, the cellular mechanisms are unknown. Using a translational approach combining RNA sequencing and cellular analyses, we investigated bone marrow stromal stem cells (BM-MSCs) of 54 men divided into lean, overweight, and obese groups on the basis of BMI. Compared with BM-MSCs obtained from lean, obese BM-MSCs exhibited a shift of molecular phenotype toward committed adipocytic progenitors and increased expression of metabolic genes involved in glycolytic and oxidoreductase activity. Interestingly, compared with paired samples of peripheral adipose tissue-derived stromal cells (AT-MSCs), insulin signaling of obese BM-MSCs was enhanced and accompanied by increased abundance of insulin receptor positive (IR+) and leptin receptor positive (LEPR+) cells in BM-MSC cultures. Their hyper-activated metabolic state was accompanied by an accelerated senescence phenotype. Our data provide a plausible explanation for the bone fragility in obesity caused by enhanced insulin signaling leading to accelerated metabolic senescence of BM-MSCs.
Collapse
Affiliation(s)
- Michaela Tencerova
- Department of Molecular Endocrinology, KMEB, University of Southern Denmark and Odense University Hospital, 5000 Odense C, Denmark; OPEN, Odense Patient Data Explorative Network, Odense University Hospital, Odense, Denmark.
| | - Morten Frost
- Department of Molecular Endocrinology, KMEB, University of Southern Denmark and Odense University Hospital, 5000 Odense C, Denmark; Steno Diabetes Center Odense, Odense University Hospital, 5000 Odense C, Denmark
| | - Florence Figeac
- Department of Molecular Endocrinology, KMEB, University of Southern Denmark and Odense University Hospital, 5000 Odense C, Denmark
| | - Tina Kamilla Nielsen
- Department of Molecular Endocrinology, KMEB, University of Southern Denmark and Odense University Hospital, 5000 Odense C, Denmark
| | - Dalia Ali
- Department of Molecular Endocrinology, KMEB, University of Southern Denmark and Odense University Hospital, 5000 Odense C, Denmark
| | - Jens-Jacob Lindegaard Lauterlein
- Department of Molecular Endocrinology, KMEB, University of Southern Denmark and Odense University Hospital, 5000 Odense C, Denmark
| | - Thomas Levin Andersen
- Clinical Cell Biology, Department of Pathology, Odense University Hospital, 5000 Odense C, Denmark; Department of Clinical Research, University of Southern Denmark, 5000 Odense C, Denmark; Department of Molecular Medicine, University of Southern Denmark, 5000 Odense C, Denmark
| | - Anders Kristian Haakonsson
- Department of Molecular Endocrinology, KMEB, University of Southern Denmark and Odense University Hospital, 5000 Odense C, Denmark; OPEN, Odense Patient Data Explorative Network, Odense University Hospital, Odense, Denmark
| | - Alexander Rauch
- Department of Molecular Endocrinology, KMEB, University of Southern Denmark and Odense University Hospital, 5000 Odense C, Denmark
| | - Jonna Skov Madsen
- Institute of Regional Health Science, University of Southern Denmark, 5000 Odense C, Denmark; Department of Biochemistry and Immunology, Lillebaelt Hospital, 7100 Vejle, Denmark
| | - Charlotte Ejersted
- Department of Endocrinology, Odense University Hospital, 5000 Odense C, Denmark
| | - Kurt Højlund
- Steno Diabetes Center Odense, Odense University Hospital, 5000 Odense C, Denmark; Department of Clinical Research, University of Southern Denmark, 5000 Odense C, Denmark
| | - Moustapha Kassem
- Department of Molecular Endocrinology, KMEB, University of Southern Denmark and Odense University Hospital, 5000 Odense C, Denmark; Department of Cellular and Molecular Medicine, DanStem (Danish Stem Cell Center), Panum Institute, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
370
|
Sierra-Ramirez A, López-Aceituno JL, Costa-Machado LF, Plaza A, Barradas M, Fernandez-Marcos PJ. Transient metabolic improvement in obese mice treated with navitoclax or dasatinib/quercetin. Aging (Albany NY) 2020; 12:11337-11348. [PMID: 32584785 PMCID: PMC7343475 DOI: 10.18632/aging.103607] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Accepted: 06/09/2020] [Indexed: 01/10/2023]
Abstract
Senescent cells accumulate with obesity in the white adipose tissue of mice and humans. These senescent cells enhance the pro-inflammatory environment that, with time, contributes to the onset of glucose intolerance and type 2 diabetes. Glucose intolerance in mouse models of obesity has been successfully reversed by the elimination of senescent cells with the senolytic compounds navitoclax or the combination of dasatinib and quercetin (D/Q). In this work, we generated obese mice by high-fat diet feeding, and treated them with five consecutive cycles of navitoclax or D/Q during 16 weeks. We observed an efficient reduction in the white adipose tissue of the senescence markers senescence-associated β-galactosidase activity, Cdkn2a-p16 and Cdkn2a-p19 at the end of the 5 cycles. Mice treated with both navitoclax and D/Q showed an improvement of their insulin sensitivity and glucose tolerance during a short period of time (cycles 3 and 4), that disappeared at the fifth cycle. Also, these mice tended to increase the expression at their adipose tissue of the adipogenic genes Pparg and, Cebpa, as well as their plasma adiponectin levels. Together, our work shows that two different senolytic treatments, acting through independent pathways, are transiently effective in the treatment of obesity-induced metabolic disorders.
Collapse
Affiliation(s)
- Arantzazu Sierra-Ramirez
- Metabolic Syndrome Group - BIOPROMET, Madrid Institute for Advanced Studies - IMDEA Food, CEI UAM+CSIC, Madrid, Spain
| | - José Luis López-Aceituno
- Metabolic Syndrome Group - BIOPROMET, Madrid Institute for Advanced Studies - IMDEA Food, CEI UAM+CSIC, Madrid, Spain
| | - Luis Filipe Costa-Machado
- Metabolic Syndrome Group - BIOPROMET, Madrid Institute for Advanced Studies - IMDEA Food, CEI UAM+CSIC, Madrid, Spain
| | - Adrián Plaza
- Metabolic Syndrome Group - BIOPROMET, Madrid Institute for Advanced Studies - IMDEA Food, CEI UAM+CSIC, Madrid, Spain
| | - Marta Barradas
- Metabolic Syndrome Group - BIOPROMET, Madrid Institute for Advanced Studies - IMDEA Food, CEI UAM+CSIC, Madrid, Spain
| | - Pablo Jose Fernandez-Marcos
- Metabolic Syndrome Group - BIOPROMET, Madrid Institute for Advanced Studies - IMDEA Food, CEI UAM+CSIC, Madrid, Spain
| |
Collapse
|
371
|
Luo X, Li Y, Yang P, Chen Y, Wei L, Yu T, Xia J, Ruan XZ, Zhao L, Chen Y. Obesity induces preadipocyte CD36 expression promoting inflammation via the disruption of lysosomal calcium homeostasis and lysosome function. EBioMedicine 2020; 56:102797. [PMID: 32516742 PMCID: PMC7281849 DOI: 10.1016/j.ebiom.2020.102797] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2020] [Revised: 04/13/2020] [Accepted: 04/28/2020] [Indexed: 12/31/2022] Open
Abstract
Background Preadipocyte is closely related to obesity-induced inflammation. The impairment of autophagic flux by defective lysosomal function has been observed in adipose tissue from obese mice. While the fatty acid translocase CD36 is an important immuno-metabolic receptor, it remains unclear whether preadipocyte CD36 is involved in adipose tissue inflammation and whether CD36 regulates lysosomal function. Methods Using visceral adipose tissue from obese patients, a high-fat diet (HFD)-induced obese mice model, primary mouse preadipocytes and 3T3L1 cells we analyzed whether and how preadipocyte CD36 modulates lysosomal function and adipose tissue inflammation. Findings CD36 expression in preadipocytes is induced in obese patients and HFD-fed mice, accompanied with the disruption of lysosome function. CD36 knockout protects primary preadipocytes of HFD-fed mice from lysosomal impairment. In vitro, CD36 interacts with Fyn to phosphorylate and activate Inositol (1,4,5)-trisphosphate receptor 1 (IP3R1), causing excess calcium transport from endoplasmic reticulum (ER) to lysosome, which results in lysosomal impairment and inflammation. Moreover, IP3R inhibitor 2-aminoethoxydiphenyl borate (2APB) attenuates lysosomal impairment, inflammation and lipid accumulation in CD36-overexpressing preadipocytes. Interpretation Our data support that the abnormal upregulation of CD36 in preadipocytes may contribute to the development of adipose tissue inflammation. CD36/Fyn/IP3R1-mediated lysosomal calcium overload leads to lysosomal impairment and inflammation in preadipocyte. Thus targeting improving lysosomal calcium homeostasis may represent a novel strategy for treating obesity-induced inflammation.
Collapse
Affiliation(s)
- Xiaoxiao Luo
- Centre for Lipid Research, Key Laboratory of Molecular Biology for Infectious Diseases, Ministry of Education, Department of Infectious Diseases, Institute for Viral Hepatitis, The Second Affiliated Hospital, Chongqing Medical University, Chongqing 400016, China
| | - Yanping Li
- Centre for Lipid Research, Key Laboratory of Molecular Biology for Infectious Diseases, Ministry of Education, Department of Infectious Diseases, Institute for Viral Hepatitis, The Second Affiliated Hospital, Chongqing Medical University, Chongqing 400016, China
| | - Ping Yang
- Centre for Lipid Research, Key Laboratory of Molecular Biology for Infectious Diseases, Ministry of Education, Department of Infectious Diseases, Institute for Viral Hepatitis, The Second Affiliated Hospital, Chongqing Medical University, Chongqing 400016, China
| | - Yao Chen
- Medical Examination Center, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
| | - Li Wei
- Centre for Lipid Research, Key Laboratory of Molecular Biology for Infectious Diseases, Ministry of Education, Department of Infectious Diseases, Institute for Viral Hepatitis, The Second Affiliated Hospital, Chongqing Medical University, Chongqing 400016, China
| | - Ting Yu
- Centre for Lipid Research, Key Laboratory of Molecular Biology for Infectious Diseases, Ministry of Education, Department of Infectious Diseases, Institute for Viral Hepatitis, The Second Affiliated Hospital, Chongqing Medical University, Chongqing 400016, China
| | - Jun Xia
- Centre for Lipid Research, Key Laboratory of Molecular Biology for Infectious Diseases, Ministry of Education, Department of Infectious Diseases, Institute for Viral Hepatitis, The Second Affiliated Hospital, Chongqing Medical University, Chongqing 400016, China
| | - Xiong Z Ruan
- Centre for Lipid Research, Key Laboratory of Molecular Biology for Infectious Diseases, Ministry of Education, Department of Infectious Diseases, Institute for Viral Hepatitis, The Second Affiliated Hospital, Chongqing Medical University, Chongqing 400016, China; National Clinical Research Center for Aging and Medicine, Huashan Hospital, Fudan University, Hanghai, China; John Moorhead Research Laboratory, Centre for Nephrology, University College London Medical School, Royal Free Campus, University College London, London, United Kingdom
| | - Lei Zhao
- Centre for Lipid Research, Key Laboratory of Molecular Biology for Infectious Diseases, Ministry of Education, Department of Infectious Diseases, Institute for Viral Hepatitis, The Second Affiliated Hospital, Chongqing Medical University, Chongqing 400016, China.
| | - Yaxi Chen
- Centre for Lipid Research, Key Laboratory of Molecular Biology for Infectious Diseases, Ministry of Education, Department of Infectious Diseases, Institute for Viral Hepatitis, The Second Affiliated Hospital, Chongqing Medical University, Chongqing 400016, China.
| |
Collapse
|
372
|
Shook BA, Wasko RR, Mano O, Rutenberg-Schoenberg M, Rudolph MC, Zirak B, Rivera-Gonzalez GC, López-Giráldez F, Zarini S, Rezza A, Clark DA, Rendl M, Rosenblum MD, Gerstein MB, Horsley V. Dermal Adipocyte Lipolysis and Myofibroblast Conversion Are Required for Efficient Skin Repair. Cell Stem Cell 2020; 26:880-895.e6. [PMID: 32302523 PMCID: PMC7853423 DOI: 10.1016/j.stem.2020.03.013] [Citation(s) in RCA: 183] [Impact Index Per Article: 36.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2019] [Revised: 11/20/2019] [Accepted: 03/18/2020] [Indexed: 12/26/2022]
Abstract
Mature adipocytes store fatty acids and are a common component of tissue stroma. Adipocyte function in regulating bone marrow, skin, muscle, and mammary gland biology is emerging, but the role of adipocyte-derived lipids in tissue homeostasis and repair is poorly understood. Here, we identify an essential role for adipocyte lipolysis in regulating inflammation and repair after injury in skin. Genetic mouse studies revealed that dermal adipocytes are necessary to initiate inflammation after injury and promote subsequent repair. We find through histological, ultrastructural, lipidomic, and genetic experiments in mice that adipocytes adjacent to skin injury initiate lipid release necessary for macrophage inflammation. Tamoxifen-inducible genetic lineage tracing of mature adipocytes and single-cell RNA sequencing revealed that dermal adipocytes alter their fate and generate ECM-producing myofibroblasts within wounds. Thus, adipocytes regulate multiple aspects of repair and may be therapeutic for inflammatory diseases and defective wound healing associated with aging and diabetes.
Collapse
Affiliation(s)
- Brett A Shook
- Molecular, Cellular and Developmental Biology, Yale University, New Haven, CT 06511, USA
| | - Renee R Wasko
- Molecular, Cellular and Developmental Biology, Yale University, New Haven, CT 06511, USA
| | - Omer Mano
- Molecular, Cellular and Developmental Biology, Yale University, New Haven, CT 06511, USA; Interdepartmental Neuroscience Program, Yale University, New Haven, CT 06511, USA
| | - Michael Rutenberg-Schoenberg
- Program in Computational Biology and Bioinformatics, Yale University, New Haven, CT 06511, USA; Department of Molecular Biophysics and Biochemistry, Yale University, New Haven, CT 06511, USA
| | - Michael C Rudolph
- Division of Endocrinology, Metabolism and Diabetes, University of Colorado, Denver Anschutz Medical Campus, CO 80045, USA
| | - Bahar Zirak
- Department of Dermatology, University of California, San Francisco, San Francisco, CA 94143, USA
| | | | | | - Simona Zarini
- Interdepartmental Neuroscience Program, Yale University, New Haven, CT 06511, USA
| | - Amélie Rezza
- Black Family Stem Cell Institute, Icahn School of Medicine at Mount Sinai, New York, NY 11766, USA; Department of Developmental and Regenerative Biology, Icahn School of Medicine at Mount Sinai, New York, NY 11766, USA
| | - Damon A Clark
- Molecular, Cellular and Developmental Biology, Yale University, New Haven, CT 06511, USA; Interdepartmental Neuroscience Program, Yale University, New Haven, CT 06511, USA
| | - Michael Rendl
- Black Family Stem Cell Institute, Icahn School of Medicine at Mount Sinai, New York, NY 11766, USA; Department of Developmental and Regenerative Biology, Icahn School of Medicine at Mount Sinai, New York, NY 11766, USA
| | - Michael D Rosenblum
- Department of Dermatology, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Mark B Gerstein
- Program in Computational Biology and Bioinformatics, Yale University, New Haven, CT 06511, USA; Department of Molecular Biophysics and Biochemistry, Yale University, New Haven, CT 06511, USA
| | - Valerie Horsley
- Molecular, Cellular and Developmental Biology, Yale University, New Haven, CT 06511, USA; Department of Dermatology, Yale University, New Haven, CT 06511, USA.
| |
Collapse
|
373
|
Chattopadhyay D, Thirumurugan K. Longevity-promoting efficacies of rutin in high fat diet fed Drosophila melanogaster. Biogerontology 2020; 21:653-668. [PMID: 32430858 DOI: 10.1007/s10522-020-09882-y] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2020] [Accepted: 05/12/2020] [Indexed: 12/24/2022]
Abstract
Composition of diet significantly impacts lifespan in Drosophila melanogaster. Diet-composition becomes even more crucial while assessing a phytocompound for probable pro-longevity effects in flies. Rutin is a flavonol glycoside present in apple, buckwheat, black tea and green tea. Our previous study had reported hormetic efficacy of rutin to improve longevity and other physiological parameters in Drosophila melanogaster fed with standard diet. This study aimed to understand whether rutin could exhibit similar longevity promoting effects in flies fed with a high fat diet (HFD). In this study, wild type Canton-S males and females were reared on high fat diet (HFD) treated with or without rutin at different doses (100-800 µM) and assessed for survival, food intake, fecundity, locomotion, development, resistance to various forms of stresses and relative mRNA expression of specific genes associated with ageing, namely dFoxO, MnSod, Cat, dTsc1, dTsc2, Thor, dAtg1, dAtg5, dAtg7 and dTor. Rutin at only 400 µM significantly improved survival in males fed with HFD; while at 200 µM and 400 µM it significantly improved survival in females. Doses beyond 400 µM proved detrimental for both sexes. Rutin at 200 µM and 400 µM significantly reduced average food intake in both males and females fed with HFD. A significant reduction in number of eggs laid per female per day was observed in females treated with rutin at 400 µM. Rutin at 200 µM and 400 µM significantly improved climbing efficiency in males and females. A significant reduction in eclosion time was observed in larvae fed with HFD and treated with rutin at 400 µM. Rutin at 400 µM significantly improved resistance of males and females to different stresses namely heat shock, cold shock and starvation stresses. Interestingly, rutin at 400 µM significantly reduced survival of males and females exposed to oxidative stress. In males fed with HFD, rutin at 200 µM showed significantly increased relative expression of dFoxo, MnSod, Cat, dAtg1, dAtg5 and dAtg7; at 400 µM it significantly increased the relative expression of dFoxO, MnSod, Cat, dTsc1, dTsc2, Thor, dAtg1, dAtg5, dAtg7 while decreasing relative expression of dTor. Thus, data from this study collectively showed that rutin at 400 µM and to an extent 200 µM positively impacted lifespan and modulated other physiological parameters in males and females fed with HFD.
Collapse
Affiliation(s)
- Debarati Chattopadhyay
- Department of Biotechnology, St Joseph's College (Autonomous), Bangalore, Karnataka, India
| | - Kavitha Thirumurugan
- 206, Structural Biology Lab, Centre for Biomedical Research, School of Bio Sciences & Technology, Vellore Institute of Technology, Vellore, Tamil Nadu, India.
| |
Collapse
|
374
|
Song HD, Kim SN, Saha A, Ahn SY, Akindehin S, Son Y, Cho YK, Kim M, Park JH, Jung YS, Lee YH. Aging-Induced Brain-Derived Neurotrophic Factor in Adipocyte Progenitors Contributes to Adipose Tissue Dysfunction. Aging Dis 2020; 11:575-587. [PMID: 32489703 PMCID: PMC7220283 DOI: 10.14336/ad.2019.0810] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2019] [Accepted: 08/10/2019] [Indexed: 12/22/2022] Open
Abstract
Aging-related adipose tissue dysfunction contributes to the progression of chronic metabolic diseases. We investigated the role of age-dependent expression of a neurotrophin, brain-derived neurotrophic factor (BDNF) in adipose tissue. Pro-BDNF expression was elevated in epididymal white adipose tissue (eWAT) with advanced age, which was associated with the reduction in sympathetic innervation. Interestingly, BDNF expression was enriched in PDGFRα+ adipocyte progenitors isolated from eWAT, with age-dependent increase in expression. In vitro pro-BDNF treatment caused apoptosis in adipocytes differentiated from C3H10T1/2 cells, and siRNA knockdown of sortilin mitigated these effects. Tamoxifen-inducible PDGFRα+ cell-specific deletion of BDNF (BDNFPdgfra KO) reduced pro-BDNF expression in eWAT, prevented age-associated declines in sympathetic innervation and mitochondrial content in eWAT, and improved insulin sensitivity. Moreover, BDNFPdgfra KO mice showed reduced expression of aging-induced inflammation and senescence markers in eWAT. Collectively, these results identified the upregulation of pro-BDNF expression in adipocyte progenitors as a feature of visceral white adipose tissue aging and suggested that inhibition of BDNF expression in adipocyte progenitors is potentially beneficial to prevent aging-related adipose tissue dysfunction.
Collapse
Affiliation(s)
- Hyun-Doo Song
- 1College of Pharmacy, Yonsei University, Incheon, Republic of Korea
| | - Sang Nam Kim
- 2College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, Republic of Korea
| | - Abhirup Saha
- 2College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, Republic of Korea
| | - Sang-Yeop Ahn
- 1College of Pharmacy, Yonsei University, Incheon, Republic of Korea
| | - Seun Akindehin
- 1College of Pharmacy, Yonsei University, Incheon, Republic of Korea
| | - Yeonho Son
- 2College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, Republic of Korea
| | - Yoon Keun Cho
- 2College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, Republic of Korea
| | - MinSu Kim
- 2College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, Republic of Korea
| | - Ji-Hyun Park
- 2College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, Republic of Korea
| | - Young-Suk Jung
- 3College of Pharmacy, Pusan National University, Busan, Republic of Korea
| | - Yun-Hee Lee
- 2College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, Republic of Korea
| |
Collapse
|
375
|
Goh KJ, Chen JH, Rocha N, Semple RK. Human pluripotent stem cell-based models suggest preadipocyte senescence as a possible cause of metabolic complications of Werner and Bloom Syndromes. Sci Rep 2020; 10:7490. [PMID: 32367056 PMCID: PMC7198505 DOI: 10.1038/s41598-020-64136-8] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2020] [Accepted: 04/08/2020] [Indexed: 11/09/2022] Open
Abstract
Werner Syndrome (WS) and Bloom Syndrome (BS) are disorders of DNA damage repair caused by biallelic disruption of the WRN or BLM DNA helicases respectively. Both are commonly associated with insulin resistant diabetes, usually accompanied by dyslipidemia and fatty liver, as seen in lipodystrophies. In keeping with this, progressive reduction of subcutaneous adipose tissue is commonly observed. To interrogate the underlying cause of adipose tissue dysfunction in these syndromes, CRISPR/Cas9 genome editing was used to generate human pluripotent stem cell (hPSC) lacking either functional WRN or BLM helicase. No deleterious effects were observed in WRN−/− or BLM−/− embryonic stem cells, however upon their differentiation into adipocyte precursors (AP), premature senescence emerged, impairing later stages of adipogenesis. The resulting adipocytes were also found to be senescent, with increased levels of senescent markers and senescence-associated secretory phenotype (SASP) components. SASP components initiate and reinforce senescence in adjacent cells, which is likely to create a positive feedback loop of cellular senescence within the adipocyte precursor compartment, as demonstrated in normal ageing. Such a scenario could progressively attenuate adipose mass and function, giving rise to “lipodystrophy-like” insulin resistance. Further assessment of pharmacological senolytic strategies are warranted to mitigate this component of Werner and Bloom syndromes.
Collapse
Affiliation(s)
- Kim Jee Goh
- The University of Cambridge Metabolic Research Laboratories, Wellcome Trust-MRC Institute of Metabolic Science, Cambridge, UK
| | - Jian-Hua Chen
- The University of Cambridge Metabolic Research Laboratories, Wellcome Trust-MRC Institute of Metabolic Science, Cambridge, UK.,The National Institute for Health Research Cambridge Biomedical Research Centre, Cambridge, UK
| | - Nuno Rocha
- The University of Cambridge Metabolic Research Laboratories, Wellcome Trust-MRC Institute of Metabolic Science, Cambridge, UK.,The National Institute for Health Research Cambridge Biomedical Research Centre, Cambridge, UK
| | - Robert K Semple
- The University of Cambridge Metabolic Research Laboratories, Wellcome Trust-MRC Institute of Metabolic Science, Cambridge, UK. .,The National Institute for Health Research Cambridge Biomedical Research Centre, Cambridge, UK. .,Centre for Cardiovascular Science, Queen's Medical Research Institute, University of Edinburgh, Edinburgh, UK.
| |
Collapse
|
376
|
|
377
|
Shang D, Sun D, Shi C, Xu J, Shen M, Hu X, Liu H, Tu Z. Activation of epidermal growth factor receptor signaling mediates cellular senescence induced by certain pro-inflammatory cytokines. Aging Cell 2020; 19:e13145. [PMID: 32323422 PMCID: PMC7253070 DOI: 10.1111/acel.13145] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2019] [Revised: 02/14/2020] [Accepted: 03/10/2020] [Indexed: 02/06/2023] Open
Abstract
It is well established that inflammation in the body promotes organism aging, and recent studies have attributed a similar effect to senescent cells. Considering that certain pro‐inflammatory cytokines can induce cellular senescence, systematically evaluating the effects of pro‐inflammatory cytokines in cellular senescence is an important and urgent scientific problem, especially given the ongoing surge in aging human populations. Treating IMR90 cells and HUVECs with pro‐inflammatory cytokines identified six factors able to efficiently induce cellular senescence. Of these senescence‐inducing cytokines, the activity of five (namely IL‐1β, IL‐13, MCP‐2, MIP‐3α, and SDF‐1α) was significantly inhibited by treatment with cetuximab (an antibody targeting epidermal growth factor receptor [EGFR]), gefitinib (a small molecule inhibitor of EGFR), and EGFR knockdown. In addition, treatment with one of the senescence‐inducing cytokines, SDF‐1α, significantly increased the phosphorylation levels of EGFR, as well as Erk1/2. These results suggested that pro‐inflammatory cytokines induce cellular senescence by activating EGFR signaling. Next, we found that EGF treatment could also induce cellular senescence of IMR90 cells and HUVECs. Mechanically, EGF induced cellular senescence via excessive activation of Ras and the Ras‐BRaf‐Erk1/2 signaling axis. Moreover, EGFR activation induced IMR90 cells to secrete certain senescence‐associated secretory phenotype factors (IL‐8 and MMP‐3). In summary, we report that certain pro‐inflammatory cytokines induce cellular senescence through activation of the EGFR‐Ras signaling pathway. Our study thus offers new insight into a long‐ignored mechanism by which EGFR could regulate cellular senescence and suggests that growth signals themselves may catalyze aging under certain conditions.
Collapse
Affiliation(s)
- Dongsheng Shang
- School of Pharmacy Jiangsu University Zhenjiang China
- Institute of Life Sciences Jiangsu University Zhenjiang China
| | - Danlin Sun
- Institute of Life Sciences Jiangsu University Zhenjiang China
| | - Chunyan Shi
- Obstetrics and Gynecology Department Affiliated Hospital of Jiangsu University Zhenjiang China
| | - Jun Xu
- Department of Cognitive Neurology China National Clinical Research Center for Neurological Diseases (NCRC‐ND) Beijing Tiantan Hospital Capital Medical University Beijing China
| | - Mingxiang Shen
- Institute of Life Sciences Jiangsu University Zhenjiang China
| | - Xing Hu
- Obstetrics and Gynecology Department Affiliated Hospital of Jiangsu University Zhenjiang China
| | - Hanqing Liu
- School of Pharmacy Jiangsu University Zhenjiang China
| | - Zhigang Tu
- Institute of Life Sciences Jiangsu University Zhenjiang China
| |
Collapse
|
378
|
Khosla S, Farr JN, Tchkonia T, Kirkland JL. The role of cellular senescence in ageing and endocrine disease. Nat Rev Endocrinol 2020; 16:263-275. [PMID: 32161396 PMCID: PMC7227781 DOI: 10.1038/s41574-020-0335-y] [Citation(s) in RCA: 335] [Impact Index Per Article: 67.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 02/14/2020] [Indexed: 12/19/2022]
Abstract
With the ageing of the global population, interest is growing in the 'geroscience hypothesis', which posits that manipulation of fundamental ageing mechanisms will delay (in parallel) the appearance or severity of multiple chronic, non-communicable diseases, as these diseases share the same underlying risk factor - namely, ageing. In this context, cellular senescence has received considerable attention as a potential target in preventing or treating multiple age-related diseases and increasing healthspan. Here we review mechanisms of cellular senescence and approaches to target this pathway therapeutically using 'senolytic' drugs that kill senescent cells or inhibitors of the senescence-associated secretory phenotype (SASP). Furthermore, we highlight the evidence that cellular senescence has a causative role in multiple diseases associated with ageing. Finally, we focus on the role of cellular senescence in a number of endocrine diseases, including osteoporosis, metabolic syndrome and type 2 diabetes mellitus, as well as other endocrine conditions. Although much remains to be done, considerable preclinical evidence is now leading to the initiation of proof-of-concept clinical trials using senolytics for several endocrine and non-endocrine diseases.
Collapse
Affiliation(s)
- Sundeep Khosla
- Division of Endocrinology, Mayo Clinic, Rochester, MN, USA.
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, MN, USA.
| | - Joshua N Farr
- Division of Endocrinology, Mayo Clinic, Rochester, MN, USA
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, MN, USA
| | - Tamara Tchkonia
- Division of Endocrinology, Mayo Clinic, Rochester, MN, USA
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, MN, USA
| | - James L Kirkland
- Division of Endocrinology, Mayo Clinic, Rochester, MN, USA.
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, MN, USA.
| |
Collapse
|
379
|
Wu Q, Li B, Sun S, Sun S. Unraveling Adipocytes and Cancer Links: Is There a Role for Senescence? Front Cell Dev Biol 2020; 8:282. [PMID: 32411704 PMCID: PMC7198697 DOI: 10.3389/fcell.2020.00282] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2020] [Accepted: 04/02/2020] [Indexed: 12/31/2022] Open
Abstract
Senescence is characterized by a permanent cell cycle arrest that is elicited in response to different stresses. In addition, senescent cells undergo multiple other phenotypic alterations, such as autophagy modulation, metabolic reprogramming, and the senescence-associated secretory phenotype (SASP). These senescence-related and inflammatory effects prevail within tumors and are strongly controlled by cancer properties, and inflammatory mediators further maintain and propagate the senescence process to adjacent cells. It is important to consider these detrimental effects that may drive tumorigenesis or cancer relapse. Importantly, cancer-associated adipocytes (CAAs) are one of the primary stromal cells in various tumor microenvironments and favor tumor progression by releasing various factors that can mediate local and systemic effects. However, it remains unclear whether CAAs possess senescent features. In this review, we discuss the complex relationship between senescence and CAAs and highlight important considerations for therapeutics.
Collapse
Affiliation(s)
- Qi Wu
- Department of Breast and Thyroid Surgery, Renmin Hospital of Wuhan University, Wuhan, China.,Faculty of Medicine, University of Paris Sud-Saclay, Le Kremlin-Bicêtre, France
| | - Bei Li
- Department of Breast and Thyroid Surgery, Renmin Hospital of Wuhan University, Wuhan, China
| | - Si Sun
- Department of Clinical Laboratory, Renmin Hospital of Wuhan University, Wuhan, China
| | - Shengrong Sun
- Department of Breast and Thyroid Surgery, Renmin Hospital of Wuhan University, Wuhan, China
| |
Collapse
|
380
|
Polonis K, Becari C, Chahal CAA, Zhang Y, Allen AM, Kellogg TA, Somers VK, Singh P. Chronic Intermittent Hypoxia Triggers a Senescence-like Phenotype in Human White Preadipocytes. Sci Rep 2020; 10:6846. [PMID: 32321999 PMCID: PMC7176724 DOI: 10.1038/s41598-020-63761-7] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2019] [Accepted: 04/03/2020] [Indexed: 12/13/2022] Open
Abstract
Obstructive sleep apnea (OSA) is a common sleep disorder associated with obesity. Emerging evidence suggest that OSA increases the risk of cardiovascular morbidity and mortality partly via accelerating the process of cellular aging. Thus, we sought to examine the effects of intermittent hypoxia (IH), a hallmark of OSA, on senescence in human white preadipocytes. We demonstrate that chronic IH is associated with an increased generation of mitochondrial reactive oxygen species along with increased prevalence of cells with nuclear localization of γH2AX & p16. A higher prevalence of cells positive for senescence-associated β-galactosidase activity was also evident with chronic IH exposure. Intervention with aspirin, atorvastatin or renin-angiotensin system (RAS) inhibitors effectively attenuated IH-mediated senescence-like phenotype. Importantly, the validity of in vitro findings was confirmed by examination of the subcutaneous abdominal adipose tissue which showed that OSA patients had a significantly higher percentage of cells with nuclear localization of γH2AX & p16 than non-OSA individuals (20.1 ± 10.8% vs. 10.3 ± 2.7%, Padjusted < 0.001). Furthermore, the frequency of dual positive γH2AX & p16 nuclei in adipose tissue of OSA patients receiving statin, aspirin, and/or RAS inhibitors was comparable to non-OSA individuals. This study identifies chronic IH as a trigger of senescence-like phenotype in preadipocytes. Together, our data suggest that OSA may be considered as a senescence-related disorder.
Collapse
Affiliation(s)
- Katarzyna Polonis
- Department of Cardiovascular Medicine, Mayo Clinic, MN, Rochester, USA
| | - Christiane Becari
- Department of Cardiovascular Medicine, Mayo Clinic, MN, Rochester, USA
- Department of Surgery and Anatomy, Ribeirao Preto Medical School, Ribeirão Preto, SP, Brazil
| | - C Anwar A Chahal
- Department of Cardiovascular Medicine, Mayo Clinic, MN, Rochester, USA
- Mayo Clinic Graduate School of Biomedical Sciences, MN, Rochester, USA
| | - Yuebo Zhang
- Department of Cardiovascular Medicine, Mayo Clinic, MN, Rochester, USA
| | - Alina M Allen
- Division of Gastroenterology and Hepatology, Mayo Clinic, MN, Rochester, USA
| | | | - Virend K Somers
- Department of Cardiovascular Medicine, Mayo Clinic, MN, Rochester, USA
| | - Prachi Singh
- Department of Cardiovascular Medicine, Mayo Clinic, MN, Rochester, USA.
- Pennington Biomedical Research Center, LA, Baton Rouge, USA.
| |
Collapse
|
381
|
Moderation of mitochondrial respiration mitigates metabolic syndrome of aging. Proc Natl Acad Sci U S A 2020; 117:9840-9850. [PMID: 32303655 DOI: 10.1073/pnas.1917948117] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Deregulation of mitochondrial dynamics leads to the accumulation of oxidative stress and unhealthy mitochondria; consequently, this accumulation contributes to premature aging and alterations in mitochondria linked to metabolic complications. We postulate that restrained mitochondrial ATP synthesis might alleviate age-associated disorders and extend healthspan in mammals. Herein, we prepared a previously discovered mitochondrial complex IV moderate inhibitor in drinking water and orally administered to standard-diet-fed, wild-type C57BL/6J mice every day for up to 16 mo. No manifestation of any apparent toxicity or deleterious effect on studied mouse models was observed. The impacts of an added inhibitor on a variety of mitochondrial functions were analyzed, such as respiratory activity, mitochondrial bioenergetics, and biogenesis, and a few age-associated comorbidities, including reactive oxygen species (ROS) production, glucose abnormalities, and obesity in mice. It was found that mitochondrial quality, dynamics, and oxidative metabolism were greatly improved, resulting in lean mice with a specific reduction in visceral fat plus superb energy and glucose homeostasis during their aging period compared to the control group. These results strongly suggest that a mild interference in ATP synthesis through moderation of mitochondrial activity could effectively up-regulate mitogenesis, reduce ROS production, and preserve mitochondrial integrity, thereby impeding the onset of metabolic syndrome. We conclude that this inhibitory intervention in mitochondrial respiration rectified the age-related physiological breakdown in mice by protecting mitochondrial function and markedly mitigated certain undesired primary outcomes of metabolic syndrome, such as obesity and type 2 diabetes. This intervention warrants further research on the treatment of metabolic syndrome of aging in humans.
Collapse
|
382
|
Gorwood J, Ejlalmanesh T, Bourgeois C, Mantecon M, Rose C, Atlan M, Desjardins D, Le Grand R, Fève B, Lambotte O, Capeau J, Béréziat V, Lagathu C. SIV Infection and the HIV Proteins Tat and Nef Induce Senescence in Adipose Tissue and Human Adipose Stem Cells, Resulting in Adipocyte Dysfunction. Cells 2020; 9:cells9040854. [PMID: 32244726 PMCID: PMC7226797 DOI: 10.3390/cells9040854] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2020] [Revised: 03/11/2020] [Accepted: 03/30/2020] [Indexed: 12/11/2022] Open
Abstract
Background: Aging is characterized by adipose tissue senescence, inflammation, and fibrosis, with trunk fat accumulation. Aging HIV-infected patients have a higher risk of trunk fat accumulation than uninfected individuals—suggesting that viral infection has a role in adipose tissue aging. We previously demonstrated that HIV/SIV infection and the Tat and Nef viral proteins were responsible for adipose tissue fibrosis and impaired adipogenesis. We hypothesized that SIV/HIV infection and viral proteins could induce adipose tissue senescence and thus lead to adipocyte dysfunctions. Methods: Features of tissue senescence were evaluated in subcutaneous and visceral adipose tissues of SIV-infected macaques and in human adipose stem cells (ASCs) exposed to Tat or Nef for up to 30 days. Results: p16 expression and p53 activation were higher in adipose tissue of SIV-infected macaques than in control macaques, indicating adipose tissue senescence. Tat and Nef induced higher senescence in ASCs, characterized by higher levels of senescence-associated beta-galactosidase activity, p16 expression, and p53 activation vs. control cells. Treatment with Tat and Nef also induced oxidative stress and mitochondrial dysfunction. Prevention of oxidative stress (using N-acetyl-cysteine) reduced senescence in ASCs. Adipocytes having differentiated from Nef-treated ASCs displayed alterations in adipogenesis with lower levels of triglyceride accumulation and adipocyte marker expression and secretion, and insulin resistance. Conclusion: HIV/SIV promotes adipose tissue senescence, which in turn may alter adipocyte function and contribute to insulin resistance.
Collapse
Affiliation(s)
- Jennifer Gorwood
- Lipodystrophies, Metabolic and Hormonal Adaptation, and Aging, UMR_S 938, Centre de Recherche Saint-Antoine-Institut Hospitalo-Universitaire de Cardiométabolisme et Nutrition (ICAN), INSERM, Sorbonne Université, F-75012 Paris, France; (J.G.); (T.E.); (M.M.); (C.R.); (M.A.); (B.F.); (J.C.)
| | - Tina Ejlalmanesh
- Lipodystrophies, Metabolic and Hormonal Adaptation, and Aging, UMR_S 938, Centre de Recherche Saint-Antoine-Institut Hospitalo-Universitaire de Cardiométabolisme et Nutrition (ICAN), INSERM, Sorbonne Université, F-75012 Paris, France; (J.G.); (T.E.); (M.M.); (C.R.); (M.A.); (B.F.); (J.C.)
| | - Christine Bourgeois
- Immunology of Viral infections and Autoimmune Diseases, IDMIT Department, IBFJ, U1184, INSERM-CEA-Université Paris Sud 11, F-92260 Fontenay-Aux-Roses and F-94270 Le Kremlin-Bicêtre, France; (C.B.); (O.L.)
| | - Matthieu Mantecon
- Lipodystrophies, Metabolic and Hormonal Adaptation, and Aging, UMR_S 938, Centre de Recherche Saint-Antoine-Institut Hospitalo-Universitaire de Cardiométabolisme et Nutrition (ICAN), INSERM, Sorbonne Université, F-75012 Paris, France; (J.G.); (T.E.); (M.M.); (C.R.); (M.A.); (B.F.); (J.C.)
| | - Cindy Rose
- Lipodystrophies, Metabolic and Hormonal Adaptation, and Aging, UMR_S 938, Centre de Recherche Saint-Antoine-Institut Hospitalo-Universitaire de Cardiométabolisme et Nutrition (ICAN), INSERM, Sorbonne Université, F-75012 Paris, France; (J.G.); (T.E.); (M.M.); (C.R.); (M.A.); (B.F.); (J.C.)
| | - Michael Atlan
- Lipodystrophies, Metabolic and Hormonal Adaptation, and Aging, UMR_S 938, Centre de Recherche Saint-Antoine-Institut Hospitalo-Universitaire de Cardiométabolisme et Nutrition (ICAN), INSERM, Sorbonne Université, F-75012 Paris, France; (J.G.); (T.E.); (M.M.); (C.R.); (M.A.); (B.F.); (J.C.)
- Plastic Surgery Department, Tenon Hospital, AP-HP, F-75020 Paris, France
| | - Delphine Desjardins
- IDMIT Department, Center for Immunology of Viral Infections and Autoimmune Diseases, Inserm, CEA, Université Paris Saclay, F-92260 Fontenay-aux-Roses, France; (D.D.); (R.L.G.)
| | - Roger Le Grand
- IDMIT Department, Center for Immunology of Viral Infections and Autoimmune Diseases, Inserm, CEA, Université Paris Saclay, F-92260 Fontenay-aux-Roses, France; (D.D.); (R.L.G.)
| | - Bruno Fève
- Lipodystrophies, Metabolic and Hormonal Adaptation, and Aging, UMR_S 938, Centre de Recherche Saint-Antoine-Institut Hospitalo-Universitaire de Cardiométabolisme et Nutrition (ICAN), INSERM, Sorbonne Université, F-75012 Paris, France; (J.G.); (T.E.); (M.M.); (C.R.); (M.A.); (B.F.); (J.C.)
- Diabétologie et Reproduction, PRISIS, Service d’Endocrinologie, Hôpital Saint-Antoine, AP-HP, F-75012 Paris, France
| | - Olivier Lambotte
- Immunology of Viral infections and Autoimmune Diseases, IDMIT Department, IBFJ, U1184, INSERM-CEA-Université Paris Sud 11, F-92260 Fontenay-Aux-Roses and F-94270 Le Kremlin-Bicêtre, France; (C.B.); (O.L.)
- Service de Médecine Interne et Immunologie Clinique, Groupe Hospitalier Universitaire Paris Sud, Hôpital Bicêtre, AP-HP, F-94270 Le Kremlin-Bicêtre, France
| | - Jacqueline Capeau
- Lipodystrophies, Metabolic and Hormonal Adaptation, and Aging, UMR_S 938, Centre de Recherche Saint-Antoine-Institut Hospitalo-Universitaire de Cardiométabolisme et Nutrition (ICAN), INSERM, Sorbonne Université, F-75012 Paris, France; (J.G.); (T.E.); (M.M.); (C.R.); (M.A.); (B.F.); (J.C.)
| | - Véronique Béréziat
- Lipodystrophies, Metabolic and Hormonal Adaptation, and Aging, UMR_S 938, Centre de Recherche Saint-Antoine-Institut Hospitalo-Universitaire de Cardiométabolisme et Nutrition (ICAN), INSERM, Sorbonne Université, F-75012 Paris, France; (J.G.); (T.E.); (M.M.); (C.R.); (M.A.); (B.F.); (J.C.)
- Correspondence: (V.B.); (C.L.); Tel.: +33140011321 (V.B.)
| | - Claire Lagathu
- Lipodystrophies, Metabolic and Hormonal Adaptation, and Aging, UMR_S 938, Centre de Recherche Saint-Antoine-Institut Hospitalo-Universitaire de Cardiométabolisme et Nutrition (ICAN), INSERM, Sorbonne Université, F-75012 Paris, France; (J.G.); (T.E.); (M.M.); (C.R.); (M.A.); (B.F.); (J.C.)
- Correspondence: (V.B.); (C.L.); Tel.: +33140011321 (V.B.)
| |
Collapse
|
383
|
Kiss T, Nyúl-Tóth Á, Balasubramanian P, Tarantini S, Ahire C, DelFavero J, Yabluchanskiy A, Csipo T, Farkas E, Wiley G, Garman L, Csiszar A, Ungvari Z. Single-cell RNA sequencing identifies senescent cerebromicrovascular endothelial cells in the aged mouse brain. GeroScience 2020; 42:429-444. [PMID: 32236824 PMCID: PMC7205992 DOI: 10.1007/s11357-020-00177-1] [Citation(s) in RCA: 133] [Impact Index Per Article: 26.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Accepted: 03/01/2020] [Indexed: 01/21/2023] Open
Abstract
Age-related phenotypic changes of cerebromicrovascular endothelial cells lead to dysregulation of cerebral blood flow and blood-brain barrier disruption, promoting the pathogenesis of vascular cognitive impairment (VCI). In recent years, endothelial cell senescence has emerged as a potential mechanism contributing to microvascular pathologies opening the avenue to the therapeutic exploitation of senolytic drugs in preclinical studies. However, difficulties with the detection of senescent endothelial cells in wild type mouse models of aging hinder the assessment of the efficiency of senolytic treatments. To detect senescent endothelial cells in the aging mouse brain, we analyzed 4233 cells in fractions enriched for cerebromicrovascular endothelial cells and other cells associated with the neurovascular unit obtained from young (3-month-old) and aged (28-month-old) C57BL/6 mice. We define 13 transcriptomic cell types by deep, single-cell RNA sequencing. We match transcriptomic signatures of cellular senescence to endothelial cells identified on the basis of their gene expression profile. Our study demonstrates that with advanced aging, there is an increased ratio of senescent endothelial cells (~ 10%) in the mouse cerebral microcirculation. We propose that our single-cell RNA sequencing-based method can be adapted to study the effect of aging on senescence in various brain cell types as well as to evaluate the efficiency of various senolytic regimens in multiple tissues.
Collapse
Affiliation(s)
- Tamas Kiss
- Department of Biochemistry and Molecular Biology, Reynolds Oklahoma Center on Aging/Center for Geroscience and Healthy Brain Aging, Vascular Cognitive Impairment and Neurodegeneration Program, University of Oklahoma Health Sciences Center, 975 NE 10th Street, Oklahoma City, OK, 73104, USA
- Department of Medical Physics and Informatics, International Training Program in Geroscience, Theoretical Medicine Doctoral School, University of Szeged, Szeged, Hungary
| | - Ádám Nyúl-Tóth
- Department of Biochemistry and Molecular Biology, Reynolds Oklahoma Center on Aging/Center for Geroscience and Healthy Brain Aging, Vascular Cognitive Impairment and Neurodegeneration Program, University of Oklahoma Health Sciences Center, 975 NE 10th Street, Oklahoma City, OK, 73104, USA
- Biological Research Centre, Institute of Biophysics, Szeged, Hungary
| | - Priya Balasubramanian
- Department of Biochemistry and Molecular Biology, Reynolds Oklahoma Center on Aging/Center for Geroscience and Healthy Brain Aging, Vascular Cognitive Impairment and Neurodegeneration Program, University of Oklahoma Health Sciences Center, 975 NE 10th Street, Oklahoma City, OK, 73104, USA
| | - Stefano Tarantini
- Department of Biochemistry and Molecular Biology, Reynolds Oklahoma Center on Aging/Center for Geroscience and Healthy Brain Aging, Vascular Cognitive Impairment and Neurodegeneration Program, University of Oklahoma Health Sciences Center, 975 NE 10th Street, Oklahoma City, OK, 73104, USA
- Department of Health Promotion Sciences, College of Public Health, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Department of Public Health, International Training Program in Geroscience, Doctoral School of Basic and Translational Medicine, Semmelweis University, Budapest, Hungary
| | - Chetan Ahire
- Department of Biochemistry and Molecular Biology, Reynolds Oklahoma Center on Aging/Center for Geroscience and Healthy Brain Aging, Vascular Cognitive Impairment and Neurodegeneration Program, University of Oklahoma Health Sciences Center, 975 NE 10th Street, Oklahoma City, OK, 73104, USA
| | - Jordan DelFavero
- Department of Biochemistry and Molecular Biology, Reynolds Oklahoma Center on Aging/Center for Geroscience and Healthy Brain Aging, Vascular Cognitive Impairment and Neurodegeneration Program, University of Oklahoma Health Sciences Center, 975 NE 10th Street, Oklahoma City, OK, 73104, USA
| | - Andriy Yabluchanskiy
- Department of Biochemistry and Molecular Biology, Reynolds Oklahoma Center on Aging/Center for Geroscience and Healthy Brain Aging, Vascular Cognitive Impairment and Neurodegeneration Program, University of Oklahoma Health Sciences Center, 975 NE 10th Street, Oklahoma City, OK, 73104, USA
- Department of Public Health, International Training Program in Geroscience, Doctoral School of Basic and Translational Medicine, Semmelweis University, Budapest, Hungary
| | - Tamas Csipo
- Department of Biochemistry and Molecular Biology, Reynolds Oklahoma Center on Aging/Center for Geroscience and Healthy Brain Aging, Vascular Cognitive Impairment and Neurodegeneration Program, University of Oklahoma Health Sciences Center, 975 NE 10th Street, Oklahoma City, OK, 73104, USA
- Department of Health Promotion Sciences, College of Public Health, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Department of Cardiology, International Training Program in Geroscience, Division of Clinical Physiology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Eszter Farkas
- Department of Medical Physics and Informatics, International Training Program in Geroscience, Theoretical Medicine Doctoral School, University of Szeged, Szeged, Hungary
| | - Graham Wiley
- Genes & Human Disease Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
| | - Lori Garman
- Genes & Human Disease Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
| | - Anna Csiszar
- Department of Biochemistry and Molecular Biology, Reynolds Oklahoma Center on Aging/Center for Geroscience and Healthy Brain Aging, Vascular Cognitive Impairment and Neurodegeneration Program, University of Oklahoma Health Sciences Center, 975 NE 10th Street, Oklahoma City, OK, 73104, USA.
- Department of Medical Physics and Informatics, International Training Program in Geroscience, Theoretical Medicine Doctoral School, University of Szeged, Szeged, Hungary.
- The Peggy and Charles Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, USA.
| | - Zoltan Ungvari
- Department of Biochemistry and Molecular Biology, Reynolds Oklahoma Center on Aging/Center for Geroscience and Healthy Brain Aging, Vascular Cognitive Impairment and Neurodegeneration Program, University of Oklahoma Health Sciences Center, 975 NE 10th Street, Oklahoma City, OK, 73104, USA
- Department of Medical Physics and Informatics, International Training Program in Geroscience, Theoretical Medicine Doctoral School, University of Szeged, Szeged, Hungary
- Department of Health Promotion Sciences, College of Public Health, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Department of Public Health, International Training Program in Geroscience, Doctoral School of Basic and Translational Medicine, Semmelweis University, Budapest, Hungary
- The Peggy and Charles Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, USA
| |
Collapse
|
384
|
Effects of low skeletal muscle mass and sarcopenic obesity on albuminuria: a 7-year longitudinal study. Sci Rep 2020; 10:5774. [PMID: 32238873 PMCID: PMC7113302 DOI: 10.1038/s41598-020-62841-y] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2019] [Accepted: 03/19/2020] [Indexed: 12/25/2022] Open
Abstract
We aimed to identify the association between low skeletal muscle, sarcopenic obesity, and the incidence of albuminuria in the general population using a longitudinal study. Data from 29,942 subjects who underwent two or more routine health examinations from 2006 to 2013 were retrospectively reviewed. Relative skeletal muscle mass was presented using the skeletal muscle mass index (SMI), a measure of body weight-adjusted appendicular skeletal muscle mass estimated by bioelectrical impedance analysis. The cumulative incidence of albuminuria was 981 (3.3%) during the 7-year follow-up period. The hazard ratio of incident albuminuria was 1.44 (95% CI: 1.22–1.71, p for trend <0.001) in the lowest SMI tertile relative to the highest SMI tertile after multivariable adjustment. After additionally adjusting for general and central obesity, the hazard ratio was 1.35 (95% CI: 1.13–1.61, p for trend = 0.001) and 1.30 (95% CI: 1.08–1.56, p for trend = 0.003), respectively. Furthermore, the risk of developing albuminuria was much higher in the sarcopenic obesity group (HR: 1.49, 95% CI: 1.21–1.81, p for trend <0.001) compared to the other groups. Sarcopenic obesity, as well as low skeletal muscle, may lead to albuminuria in general populations.
Collapse
|
385
|
Role of the Nuclear Lamina in Age-Associated Nuclear Reorganization and Inflammation. Cells 2020; 9:cells9030718. [PMID: 32183360 PMCID: PMC7140666 DOI: 10.3390/cells9030718] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2020] [Revised: 03/11/2020] [Accepted: 03/12/2020] [Indexed: 02/06/2023] Open
Abstract
Aging is characterized by the gradual loss of tissue function and integrity. Activation of inflammatory responses accelerates the deterioration of cells and tissues. Many studies have shown that alteration of the components of the nuclear lamina is associated with inflammation, both in vivo and in vitro. However, the mechanism by which the nuclear lamina regulates inflammation is largely unknown. Recent studies have suggested that the nuclear lamina regulates both organization of the three-dimensional chromatin structure at the nuclear periphery and global gene expression, such as the expression of inflammatory response genes. Here, we discuss the current updates in the research on nuclear lamina alteration, activation of inflammation, and nuclear reorganization in models of cellular senescence and organismal aging.
Collapse
|
386
|
Mikkola TM, Kautiainen H, von Bonsdorff MB, Salonen MK, Wasenius N, Kajantie E, Eriksson JG. Body composition and changes in health-related quality of life in older age: a 10-year follow-up of the Helsinki Birth Cohort Study. Qual Life Res 2020; 29:2039-2050. [PMID: 32124264 PMCID: PMC7363735 DOI: 10.1007/s11136-020-02453-1] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/17/2020] [Indexed: 12/19/2022]
Abstract
Purpose Most studies examining the associations between body composition and health-related quality of life (HRQoL) in older age have been cross-sectional and analyzed only fat or lean mass. Hence, it is poorly known whether fat and lean mass are independently associated with subsequent changes in HRQoL. We investigated whether baseline lean and fat mass are associated with changes in HRQoL over a 10-year period in older adults. Methods We studied 1044 men and women from the Helsinki Birth Cohort Study (age 57–70 years at baseline). Bioelectrical impedance analysis was used to derive baseline fat mass index (FMI, fat mass/height2) and lean mass index (lean mass/height2), dichotomized at sex-specific medians. HRQoL was assessed using RAND 36-item Health Survey at baseline and follow-up 10 years later. Results When controlled for lean mass and adjusted for potential confounders, high baseline FMI was associated with a greater decline in general health (standardized regression coefficient [β] = − 0.13, p = 0.001), physical functioning (β = − 0.11, p = 0.002), role physical (β = − 0.13, p = 0.003), vitality (β = − 0.08, p = 0.027), role emotional (β = − 0.12, p = 0.007), and physical component score (β = − 0.14, p < 0.001). High baseline FMI was also associated with low HRQoL in all physical domains at baseline (β: from − 0.38 to − 0.10). Lean mass was not strongly associated with HRQoL at baseline or change in HRQoL. Conclusion In older community-dwelling adults, higher fat mass is, independent of lean mass, associated with lower physical HRQoL and greater decline in HRQoL. Prevention of adiposity may contribute to preservation of a good quality of life in older age.
Collapse
Affiliation(s)
- Tuija M Mikkola
- Folkhälsan Research Center, Helsinki, Finland. .,Clinicum, Faculty of Medicine, University of Helsinki, Helsinki, Finland.
| | - Hannu Kautiainen
- Folkhälsan Research Center, Helsinki, Finland.,Primary Health Care Unit, Kuopio University Hospital, Kuopio, Finland
| | - Mikaela B von Bonsdorff
- Folkhälsan Research Center, Helsinki, Finland.,Gerontology Research Center, Faculty of Sport and Health Sciences, University of Jyväskylä, Jyvaskyla, Finland
| | - Minna K Salonen
- Folkhälsan Research Center, Helsinki, Finland.,Public Health Promotion Unit, National Institute for Health and Welfare, Helsinki, Finland
| | - Niko Wasenius
- Folkhälsan Research Center, Helsinki, Finland.,Department of General Practice and Primary Health Care, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Eero Kajantie
- Public Health Promotion Unit, National Institute for Health and Welfare, Helsinki, Finland.,PEDEGO Research Unit, MRC Oulu, Oulu University Hospital and University of Oulu, Oulu, Finland.,Department of Clinical and Molecular Medicine, Norwegian University for Science and Technology, Trondheim, Norway.,Children's Hospital, Helsinki University Hospital and University of Helsinki, Helsinki, Finland
| | - Johan G Eriksson
- Folkhälsan Research Center, Helsinki, Finland.,Department of General Practice and Primary Health Care, University of Helsinki and Helsinki University Hospital, Helsinki, Finland.,Singapore Institute for Clinical Sciences, Agency for Science, Technology, and Research, Singapore, Singapore.,Department of Obstetrics & Gynaecology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| |
Collapse
|
387
|
Blair MJ, Jones JD, Woessner AE, Quinn KP. Skin Structure-Function Relationships and the Wound Healing Response to Intrinsic Aging. Adv Wound Care (New Rochelle) 2020; 9:127-143. [PMID: 31993254 DOI: 10.1089/wound.2019.1021] [Citation(s) in RCA: 89] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2019] [Accepted: 08/09/2019] [Indexed: 02/06/2023] Open
Abstract
Significance: Chronic wounds, such as diabetic foot ulcers, venous stasis ulcers, and pressure ulcers affect millions of Americans each year, and disproportionately afflict our increasingly older population. Older individuals are predisposed to wound infection, repeated trauma, and the development of chronic wounds. However, a complete understanding of how the attributes of aging skin affect the wound healing process has remained elusive. Recent Advances: A variety of studies have demonstrated that the dermal matrix becomes thinner, increasingly crosslinked, and fragmented with advanced age. These structural changes, as well as an increase in cell senescence, result in altered collagen fiber remodeling and increased stiffness. Studies combining mechanical testing with advanced imaging techniques are providing new insights into the relationships between these age-related changes. Emerging research into the mechanobiology of aging and the wound healing process indicate that the altered mechanical environment of aged skin may have a significant effect on age-related delays in healing. Critical Issues: The interpretation and synthesis of clinical studies is confounded by the effects of common comorbidities that also contribute to the development of chronic wounds. A lack of quantitative biomarkers of wound healing and age-related changes makes understanding structure-function relationships during the wound healing process challenging. Future Directions: Additional work is needed to establish quantitative and mechanistic relationships among age-related changes in the skin microstructure, mechanical function, and the cellular responses to wound healing.
Collapse
Affiliation(s)
- Michael J. Blair
- Department of Biomedical Engineering, University of Arkansas, Fayetteville, Arkansas
| | - Jake D. Jones
- Department of Biomedical Engineering, University of Arkansas, Fayetteville, Arkansas
| | - Alan E. Woessner
- Department of Biomedical Engineering, University of Arkansas, Fayetteville, Arkansas
| | - Kyle P. Quinn
- Department of Biomedical Engineering, University of Arkansas, Fayetteville, Arkansas
| |
Collapse
|
388
|
Aquino-Martinez R, Rowsey JL, Fraser DG, Eckhardt BA, Khosla S, Farr JN, Monroe DG. LPS-induced premature osteocyte senescence: Implications in inflammatory alveolar bone loss and periodontal disease pathogenesis. Bone 2020; 132:115220. [PMID: 31904537 PMCID: PMC6990876 DOI: 10.1016/j.bone.2019.115220] [Citation(s) in RCA: 64] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/14/2019] [Revised: 12/13/2019] [Accepted: 12/31/2019] [Indexed: 12/21/2022]
Abstract
Cellular senescence is associated with inflammation and extracellular matrix tissue remodeling through the secretion of proteins termed the senescence-associated secretory phenotype (SASP). Although osteocyte senescence in older individuals in the skeleton is well recognized, whether young alveolar osteocytes can also become senescent is unknown. This is potentially important in the context of periodontal disease, which is an inflammatory condition caused by a gradual change from symbiotic to pathogenic oral microflora that can lead to tooth loss. Our aim was to identify whether senescent osteocytes accumulate in young alveolar bone and whether bacterial-derived lipopolysaccharide (LPS) can influence cellular senescence in alveolar bone. An osteocyte-enriched cell population isolated from alveolar bone expressed increased levels of the known senescence marker p16Ink4a, as well as select SASP markers known to be implicated alveolar bone resorption (Icam1, Il6, Il17, Mmp13 and Tnfα), compared to ramus control cells. Increased senescence of alveolar bone osteocytes was also observed in vivo using the senescence-associated distension of satellites (SADS) assay and increased γH2AX, a marker of DNA damage associated with senescent cells. To approximate a bacterial infection in vitro, alveolar osteocytes were treated with LPS. We found increased expression of various senescence and SASP markers, increased γH2AX staining, increased SA-β-Gal activity and the redistribution of F-actin leading to a larger and flattened cell morphology, all hallmarks of cellular senescence. In conclusion, our data suggests a model whereby bacterial-derived LPS stimulates premature alveolar osteocyte senescence, which in combination with the resultant SASP, could potentially contribute to the onset of alveolar bone loss.
Collapse
Affiliation(s)
- Ruben Aquino-Martinez
- Department of Medicine, Division of Endocrinology, Mayo Clinic College of Medicine, Rochester, MN, USA
| | - Jennifer L Rowsey
- Department of Medicine, Division of Endocrinology, Mayo Clinic College of Medicine, Rochester, MN, USA
| | - Daniel G Fraser
- Department of Medicine, Division of Endocrinology, Mayo Clinic College of Medicine, Rochester, MN, USA
| | - Brittany A Eckhardt
- Department of Medicine, Division of Endocrinology, Mayo Clinic College of Medicine, Rochester, MN, USA
| | - Sundeep Khosla
- Department of Medicine, Division of Endocrinology, Mayo Clinic College of Medicine, Rochester, MN, USA; Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, MN, USA
| | - Joshua N Farr
- Department of Medicine, Division of Endocrinology, Mayo Clinic College of Medicine, Rochester, MN, USA; Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, MN, USA.
| | - David G Monroe
- Department of Medicine, Division of Endocrinology, Mayo Clinic College of Medicine, Rochester, MN, USA; Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, MN, USA.
| |
Collapse
|
389
|
Gourronc FA, Perdew GH, Robertson LW, Klingelhutz AJ. PCB126 blocks the thermogenic beiging response of adipocytes. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2020; 27:8897-8904. [PMID: 31721030 PMCID: PMC7098842 DOI: 10.1007/s11356-019-06663-0] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/29/2019] [Accepted: 10/01/2019] [Indexed: 05/16/2023]
Abstract
Subcutaneous white adipose tissue is capable of becoming thermogenic in a process that is referred to as "beiging." Beiging is associated with activation of the uncoupling protein, UCP1, and is known to be important for preventing adipose hypertrophy and development of insulin resistance. Polychlorinated biphenyls (PCBs) accumulate in fat, and it is hypothesized that disruption of adipogenesis and adipocyte function by PCBs may be causative in the development of obesity and diabetes. We developed immortal human subcutaneous preadipocytes that, when differentiated, are capable of beiging. Preadipocytes that were treated with polychlorinated biphenyl congener 126 (PCB126), followed by differentiation, were suppressed for their ability to activate UCP1 upon β-adrenergic stimulation with norepinephrine (NE), demonstrating a block in the beiging response. Treatment of preadipocytes with another known endogenous AhR agonist, indoxyl sulfate (IS), followed by differentiation also blocked the NE-stimulated upregulation of UCP1. Knockdown of the aryl hydrocarbon receptor (AhR) caused the preadipocytes to be refractory to PCB126 and IS effects. The chemical AhR antagonist, CH223191, was effective at preventing the effects of PCB126 but not IS, indicating AhR ligand specificity of CH223191. Repression of NE-induced UCP1 upregulation was also observed when already-differentiated mature adipocytes were treated with PCB126 but not IS. These results indicate that exposure of preadipocytes to endogenous (IS) or exogenous (PCB126) AhR agonists is effective at blocking them from becoming functional adipocytes that are capable of the beiging response. Mature adipocytes may have differential responses. This finding suggests a mechanism by which dioxin-like PCBs such as PCB126 could lead to disruption in energy homeostasis, potentially leading to obesity and diabetes.
Collapse
Affiliation(s)
- Francoise A Gourronc
- Department of Microbiology and Immunology, University of Iowa, 3-612 BSB, 51 Newton Road, Iowa City, IA, 52242, USA
| | - Gary H Perdew
- Center for Molecular Toxicology and Carcinogenesis and the Department of Veterinary and Biomedical Sciences, Pennsylvania State University, University Park, 16802, PA, USA
| | - Larry W Robertson
- Department of Occupational & Environmental Health, College of Public Health, University of Iowa, Iowa City, IA, 52242, USA
| | - Aloysius J Klingelhutz
- Department of Microbiology and Immunology, University of Iowa, 3-612 BSB, 51 Newton Road, Iowa City, IA, 52242, USA.
| |
Collapse
|
390
|
Kopchick JJ, Berryman DE, Puri V, Lee KY, Jorgensen JOL. The effects of growth hormone on adipose tissue: old observations, new mechanisms. Nat Rev Endocrinol 2020; 16:135-146. [PMID: 31780780 PMCID: PMC7180987 DOI: 10.1038/s41574-019-0280-9] [Citation(s) in RCA: 99] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 10/16/2019] [Indexed: 12/18/2022]
Abstract
The ability of growth hormone (GH) to induce adipose tissue lipolysis has been known for over five decades; however, the molecular mechanisms that mediate this effect and the ability of GH to inhibit insulin-stimulated glucose uptake have scarcely been documented. In this same time frame, our understanding of adipose tissue has evolved to reveal a complex structure with distinct types of adipocyte, depot-specific differences, a biologically significant extracellular matrix and important endocrine properties mediated by adipokines. All these aforementioned features, in turn, can influence lipolysis. In this Review, we provide a historical and current overview of the lipolytic effect of GH in humans, mice and cultured cells. More globally, we explain lipolysis in terms of GH-induced intracellular signalling and its effect on obesity, insulin resistance and lipotoxicity. In this regard, findings that define molecular mechanisms by which GH induces lipolysis are described. Finally, data are presented for the differential effect of GH on specific adipose tissue depots and on distinct classes of metabolically active adipocytes. Together, these cellular, animal and human studies reveal novel cellular phenotypes and molecular pathways regulating the metabolic effects of GH on adipose tissue.
Collapse
Affiliation(s)
- John J Kopchick
- Edison Biotechnology Institute, Ohio University, Athens, OH, USA.
- The Diabetes Institute, Ohio University Heritage College of Osteopathic Medicine, Athens, OH, USA.
- Department of Biomedical Sciences, Ohio University College of Osteopathic Medicine, Athens, OH, USA.
| | - Darlene E Berryman
- Edison Biotechnology Institute, Ohio University, Athens, OH, USA
- The Diabetes Institute, Ohio University Heritage College of Osteopathic Medicine, Athens, OH, USA
- Department of Biomedical Sciences, Ohio University College of Osteopathic Medicine, Athens, OH, USA
| | - Vishwajeet Puri
- The Diabetes Institute, Ohio University Heritage College of Osteopathic Medicine, Athens, OH, USA
- Department of Biomedical Sciences, Ohio University College of Osteopathic Medicine, Athens, OH, USA
| | - Kevin Y Lee
- The Diabetes Institute, Ohio University Heritage College of Osteopathic Medicine, Athens, OH, USA
- Department of Biomedical Sciences, Ohio University College of Osteopathic Medicine, Athens, OH, USA
| | - Jens O L Jorgensen
- Department of Endocrinology and Diabetes, Aarhus University Hospital, Aarhus, Denmark
| |
Collapse
|
391
|
Sampathkumar NK, Bravo JI, Chen Y, Danthi PS, Donahue EK, Lai RW, Lu R, Randall LT, Vinson N, Benayoun BA. Widespread sex dimorphism in aging and age-related diseases. Hum Genet 2020; 139:333-356. [PMID: 31677133 PMCID: PMC7031050 DOI: 10.1007/s00439-019-02082-w] [Citation(s) in RCA: 77] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2019] [Accepted: 10/26/2019] [Indexed: 02/07/2023]
Abstract
Although aging is a conserved phenomenon across evolutionary distant species, aspects of the aging process have been found to differ between males and females of the same species. Indeed, observations across mammalian studies have revealed the existence of longevity and health disparities between sexes, including in humans (i.e. with a female or male advantage). However, the underlying mechanisms for these sex differences in health and lifespan remain poorly understood, and it is unclear which aspects of this dimorphism stem from hormonal differences (i.e. predominance of estrogens vs. androgens) or from karyotypic differences (i.e. XX vs. XY sex chromosome complement). In this review, we discuss the state of the knowledge in terms of sex dimorphism in various aspects of aging and in human age-related diseases. Where the interplay between sex differences and age-related differences has not been explored fully, we present the state of the field to highlight important future research directions. We also discuss various dietary, drug or genetic interventions that were shown to improve longevity in a sex-dimorphic fashion. Finally, emerging tools and models that can be leveraged to decipher the mechanisms underlying sex differences in aging are also briefly discussed.
Collapse
Affiliation(s)
- Nirmal K Sampathkumar
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA, 90089, USA
- Maurice Wohl Clinical Neuroscience Institute, King's College London, London, UK
| | - Juan I Bravo
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA, 90089, USA
- Graduate Program in the Biology of Aging, University of Southern California, Los Angeles, CA, 90089, USA
| | - Yilin Chen
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA, 90089, USA
- Masters Program in Nutrition, Healthspan, and Longevity, University of Southern California, Los Angeles, CA, 90089, USA
| | - Prakroothi S Danthi
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA, 90089, USA
| | - Erin K Donahue
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA, 90089, USA
- Neuroscience Graduate Program, University of Southern California, Los Angeles, CA, 90089, USA
| | - Rochelle W Lai
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA, 90089, USA
| | - Ryan Lu
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA, 90089, USA
- Graduate Program in the Biology of Aging, University of Southern California, Los Angeles, CA, 90089, USA
| | - Lewis T Randall
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA, 90089, USA
- Graduate Program in the Biology of Aging, University of Southern California, Los Angeles, CA, 90089, USA
| | - Nika Vinson
- Department of Urology, Pelvic Medicine and Reconstructive Surgery, UCLA David Geffen School of Medicine, Los Angeles, CA, 90024, USA
| | - Bérénice A Benayoun
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA, 90089, USA.
- USC Norris Comprehensive Cancer Center, Epigenetics and Gene Regulation, Los Angeles, CA, 90089, USA.
- USC Stem Cell Initiative, Los Angeles, CA, 90089, USA.
| |
Collapse
|
392
|
Frasca D, Diaz A, Romero M, Vazquez T, Strbo N, Romero L, McCormack RM, Podack ER, Blomberg BB. Impaired B Cell Function in Mice Lacking Perforin-2. Front Immunol 2020; 11:328. [PMID: 32180773 PMCID: PMC7057857 DOI: 10.3389/fimmu.2020.00328] [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] [Received: 10/30/2019] [Accepted: 02/10/2020] [Indexed: 01/12/2023] Open
Abstract
Perforin-2 (P2) is a pore-forming protein with cytotoxic activity against intracellular bacterial pathogens. P2 knockout (P2KO) mice are unable to control infections and die from normally non-lethal bacterial infections. Here we show that P2KO mice as compared to WT mice show significantly higher levels of systemic inflammation, measured by inflammatory markers in serum, due to continuous microbial translocation from the gut which cannot be controlled as these mice lack P2. Systemic inflammation in young and old P2KO mice induces intrinsic B cell inflammation. Systemic and B cell intrinsic inflammation are negatively associated with in vivo and in vitro antibody responses. Chronic inflammation leads to class switch recombination defects, which are at least in part responsible for the reduced in vivo and in vitro antibody responses in young and old P2KO vs. WT mice. These defects include the reduced expression of activation-induced cytidine deaminase (AID), the enzyme for class switch recombination, somatic hypermutation and IgG production and of its transcriptional activators E47 and Pax5. Of note, the response of young P2KO mice is not different from the one observed in old WT mice, suggesting that the chronic inflammatory status of mice lacking P2 may accelerate, or be equivalent, to that seen in old mice. The inflammatory status of the splenic B cells is associated with increased frequencies and numbers of the pro-inflammatory B cell subset called Age-associated B Cells (ABCs) in the spleen and the visceral adipose tissue (VAT) of P2KO old mice. We show that B cells differentiate into ABCs in the VAT following interaction with the adipocytes and their products, and this occurs more in the VAT of P2KO mice as compared to WT controls. This is to our knowledge the first study on B cell function and antibody responses in mice lacking P2.
Collapse
Affiliation(s)
- Daniela Frasca
- Department of Microbiology and Immunology, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Alain Diaz
- Department of Microbiology and Immunology, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Maria Romero
- Department of Microbiology and Immunology, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Thomas Vazquez
- Department of Microbiology and Immunology, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Natasa Strbo
- Department of Microbiology and Immunology, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Laura Romero
- Department of Microbiology and Immunology, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Ryan M McCormack
- Department of Microbiology and Immunology, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Eckhard R Podack
- Department of Microbiology and Immunology, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Bonnie B Blomberg
- Department of Microbiology and Immunology, University of Miami Miller School of Medicine, Miami, FL, United States
| |
Collapse
|
393
|
Abdalla BA, Li Z, Nie Q. A Novel DNA Methyltransferase Dnmt3a3 Splice Variant Represses Preadipocyte Proliferation and Differentiation. Front Genet 2020; 11:115. [PMID: 32158470 PMCID: PMC7052267 DOI: 10.3389/fgene.2020.00115] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2019] [Accepted: 01/30/2020] [Indexed: 01/02/2023] Open
Abstract
Proliferation and differentiation of preadipocyte are essential for the formation of fat tissues. However, the genes that regulate the early stage of preadipocyte differentiation in chicken have remained elusive. Here we identify a novel spliced variant of the DNA methyltransferase Dnmt3a gene, named Dnmt3a3, that controls early preadipocyte differentiation. Dnmt3a3 expression is increased at the onset of preadipocyte differentiation and remains elevated during differentiation. Overexpression of Dnmt3a3 in preadipocytes markedly inhibits proliferation and cell-cycle progression, and this is accompanied by inhibition of the mRNA and protein level of cell-cycle control genes, such as p21 and p27. In addition, forced expression of Dnmt3a3 in differentiating preadipocytes represses early preadipocyte differentiation, and this was found to be accompanied by inhibition of the mRNA expression levels of early preadipocyte differentiation markers, such as GATA2, GATA3, C/EBPα, C/EBPβ, AP2, and PPARγ, or the protein levels of GATA3, C/EBPβ, and PPARγ. Taken together, these data demonstrate the participation of Dnmt3a3 in the proliferation and differentiation process of chicken primary preadipocyte cells.
Collapse
Affiliation(s)
- Bahareldin Ali Abdalla
- Department of Animal Genetics, Breeding and Reproduction, College of Animal Science, South China Agricultural University, Guangzhou, China.,National-Local Joint Engineering Research Center for Livestock Breeding, Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding and the Key Lab of Chicken Genetics, Breeding and Reproduction, Ministry of Agriculture, Guangzhou, China
| | - Zhenhui Li
- Department of Animal Genetics, Breeding and Reproduction, College of Animal Science, South China Agricultural University, Guangzhou, China.,National-Local Joint Engineering Research Center for Livestock Breeding, Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding and the Key Lab of Chicken Genetics, Breeding and Reproduction, Ministry of Agriculture, Guangzhou, China
| | - Qinghua Nie
- Department of Animal Genetics, Breeding and Reproduction, College of Animal Science, South China Agricultural University, Guangzhou, China.,National-Local Joint Engineering Research Center for Livestock Breeding, Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding and the Key Lab of Chicken Genetics, Breeding and Reproduction, Ministry of Agriculture, Guangzhou, China
| |
Collapse
|
394
|
Bettedi L, Yan A, Schuster E, Alic N, Foukas LC. Increased mitochondrial and lipid metabolism is a conserved effect of Insulin/PI3K pathway downregulation in adipose tissue. Sci Rep 2020; 10:3418. [PMID: 32099025 PMCID: PMC7042323 DOI: 10.1038/s41598-020-60210-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2019] [Accepted: 01/31/2020] [Indexed: 11/28/2022] Open
Abstract
The Insulin/IGF-1 signalling (IIS) pathway plays an essential role in the regulation of glucose and lipid homeostasis. At the same time, a reduction in the IIS pathway activity can extend lifespan and healthspan in various model organisms. Amongst a number of body organs that sense and respond to insulin/IGF-1, the adipose tissue has a central role in both the metabolic and lifespan effects of IIS at the organismal level. Genetic inactivation of IIS components specifically in the adipose tissue has been shown before to improve metabolic profile and extend lifespan in various model organisms. We sought to identify conserved molecular mechanisms that may underlie the beneficial effects of IIS inhibition in the adipose tissue, specifically at the level of phosphoinositide 3-kinase (PI3K), a key IIS effector molecule. To this end, we inactivated PI3K by genetic means in the fly fat body and by pharmacological inhibition in mammalian adipocytes. Gene expression studies revealed changes to metabolism and upregulation of mitochondrial activity in mouse adipocytes and fly fat bodies with downregulated PI3K, which were confirmed by biochemical assays in mammalian adipocytes. These data suggest that PI3K inactivation has a conserved effect of upregulating mitochondrial metabolism in both fly and mammalian adipose tissue, which likely contributes to the health- and life-span extending effect of IIS pathway downregulation.
Collapse
Affiliation(s)
- Lucia Bettedi
- Institute of Healthy Ageing and Department of Genetics, Evolution and Environment, University College London, London, WC1E 6BT, UK.,National Institutes of Child Health and Human Development (NICHD), Bethesda, MD, 20814, USA
| | - Anqi Yan
- Institute of Healthy Ageing and Department of Genetics, Evolution and Environment, University College London, London, WC1E 6BT, UK
| | - Eugene Schuster
- Endocrinology Team, Breast Cancer Now, The Institute of Cancer Research, 237 Fulham Road, London, SW3 6JB, UK
| | - Nazif Alic
- Institute of Healthy Ageing and Department of Genetics, Evolution and Environment, University College London, London, WC1E 6BT, UK
| | - Lazaros C Foukas
- Institute of Healthy Ageing and Department of Genetics, Evolution and Environment, University College London, London, WC1E 6BT, UK.
| |
Collapse
|
395
|
Willard-Mack CL, Elmore SA, Hall WC, Harleman J, Kuper CF, Losco P, Rehg JE, Rühl-Fehlert C, Ward JM, Weinstock D, Bradley A, Hosokawa S, Pearse G, Mahler BW, Herbert RA, Keenan CM. Nonproliferative and Proliferative Lesions of the Rat and Mouse Hematolymphoid System. Toxicol Pathol 2020; 47:665-783. [PMID: 31526133 DOI: 10.1177/0192623319867053] [Citation(s) in RCA: 67] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The INHAND Project (International Harmonization of Nomenclature and Diagnostic Criteria for Lesions in Rats and Mice) is a joint initiative of the Societies of Toxicologic Pathology from Europe (ESTP), Great Britain (BSTP), Japan (JSTP), and North America (STP) to develop an internationally accepted nomenclature for proliferative and nonproliferative changes in rats and mice. The purpose of this publication is to provide a standardized nomenclature for classifying changes observed in the hematolymphoid organs, including the bone marrow, thymus, spleen, lymph nodes, mucosa-associated lymphoid tissues, and other lymphoid tissues (serosa-associated lymphoid clusters and tertiary lymphoid structures) with color photomicrographs illustrating examples of the lesions. Sources of material included histopathology databases from government, academia, and industrial laboratories throughout the world. Content includes spontaneous lesions as well as lesions induced by exposure to test materials. The nomenclature for these organs is divided into 3 terminologies: descriptive, conventional, and enhanced. Three terms are listed for each diagnosis. The rationale for this approach and guidance for its application to toxicologic pathology are described in detail below.
Collapse
Affiliation(s)
| | - Susan A Elmore
- Thymus subgroup lead.,National Toxicology Program, National Institute of Environmental Health Sciences, Research Triangle Park, NC, USA
| | | | - Johannes Harleman
- Lymph node subgroup lead.,Neoplasm subgroup leads.,Independent Consultant, Darmstadt, Germany
| | - C Frieke Kuper
- Associated lymphoid organs subgroup lead.,Independent Consultant, Utrecht, the Netherlands
| | - Patricia Losco
- General hematolymphoid subgroup lead.,Independent Consultant, West Chester, PA, USA
| | - Jerold E Rehg
- Spleen subgroup leads.,Neoplasm subgroup leads.,Saint Jude Children's Research Hospital, Memphis, TN, USA
| | | | - Jerrold M Ward
- Spleen subgroup leads.,Neoplasm subgroup leads.,Global VetPathology, Montgomery Village, MD, USA
| | | | - Alys Bradley
- Charles River Laboratories, Tranent, Scotland, United Kingdom
| | - Satoru Hosokawa
- Eisai Co, Ltd, Drug Safety Research Laboratories, Ibaraki, Japan
| | | | - Beth W Mahler
- Experimental Pathology Laboratories, Research Triangle Park, NC, USA
| | - Ronald A Herbert
- National Toxicology Program, National Institute of Environmental Health Sciences, Research Triangle Park, NC, USA
| | | |
Collapse
|
396
|
Pinto C, Ninfole E, Benedetti A, Maroni L, Marzioni M. Aging-Related Molecular Pathways in Chronic Cholestatic Conditions. Front Med (Lausanne) 2020; 6:332. [PMID: 32039217 PMCID: PMC6985088 DOI: 10.3389/fmed.2019.00332] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2019] [Accepted: 12/20/2019] [Indexed: 12/12/2022] Open
Abstract
Aging is commonly defined as the time-dependent functional decline of organs and tissues. Average life expectancy has increased considerably over the past century and is estimated to increase even further, consequently also the interest in understanding the aging processes. Although aging is not a disease, it is the major risk factor for the development of many chronic diseases. Pathologies, such as Primary Biliary Cholangitis (PBC) and Primary Sclerosing Cholangitis (PSC) are cholestatic liver diseases characterized by chronic inflammation, biliary damage and ultimately liver fibrosis, targeting specifically cholangiocytes. To date, the influence of aging in these biliary diseases is not fully understood. Currently, liver transplantation is the only solution because of lacking in efficiently therapies. Although liver cells have a high regenerative capacity, they undergo extensive molecular changes in response to aging. Following time-dependent damage induced by aging, the cells initially activate protective compensatory processes that, if hyperstimulated, can lead to the decline of regenerative ability and the development of pathologies. Recent studies have introduced novel therapeutic tools for cholangiopathies that have showed to have promising potential as novel therapies for PSC and PBC and for the development of new drugs. The recent advancements in understanding of molecular aging have undoubtedly the potential to unveil new pathways for selective drug treatments, but further studies are needed to deepen their knowledge.
Collapse
Affiliation(s)
- Claudio Pinto
- Department of Gastroenterology and Hepatology, Università Politecnica delle Marche, Ancona, Italy
| | - Elisabetta Ninfole
- Department of Gastroenterology and Hepatology, Università Politecnica delle Marche, Ancona, Italy
| | - Antonio Benedetti
- Department of Gastroenterology and Hepatology, Università Politecnica delle Marche, Ancona, Italy
| | - Luca Maroni
- Department of Gastroenterology and Hepatology, Università Politecnica delle Marche, Ancona, Italy
| | - Marco Marzioni
- Department of Gastroenterology and Hepatology, Università Politecnica delle Marche, Ancona, Italy
| |
Collapse
|
397
|
Borkowska J, Domaszewska-Szostek A, Kołodziej P, Wicik Z, Połosak J, Buyanovskaya O, Charzewski L, Stańczyk M, Noszczyk B, Puzianowska-Kuznicka M. Alterations in 5hmC level and genomic distribution in aging-related epigenetic drift in human adipose stem cells. Epigenomics 2020; 12:423-437. [PMID: 32031421 DOI: 10.2217/epi-2019-0131] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Aim: To clarify mechanisms affecting the level and distribution of 5-hydroxymethylcytosine (5hmC) during aging. Materials & methods: We examined levels and genomic distribution of 5hmC along with the expression of ten-eleven translocation methylcytosine dioxygenases (TETs) in adipose stem cells in young and age-advanced individuals. Results: 5hmC levels were higher in adipose stem cells of age-advanced than young individuals (p = 0.0003), but were not associated with age-related changes in expression of TETs. 5hmC levels correlated with population doubling time (r = 0.62; p = 0.01). We identified 58 differentially hydroxymethylated regions. Hypo-hydroxymethylated differentially hydroxymethylated regions were approximately twofold enriched in CCCTC-binding factor binding sites. Conclusion: Accumulation of 5hmC in aged cells can result from inefficient active demethylation due to altered TETs activity and reduced passive demethylation due to slower proliferation.
Collapse
Affiliation(s)
- Joanna Borkowska
- Department of Human Epigenetics, Mossakowski Medical Research Centre, PAS, 5 Pawinskiego Street, 02-106 Warsaw, Poland
| | - Anna Domaszewska-Szostek
- Department of Human Epigenetics, Mossakowski Medical Research Centre, PAS, 5 Pawinskiego Street, 02-106 Warsaw, Poland
| | - Paulina Kołodziej
- Department of Geriatrics & Gerontology, Medical Centre of Postgraduate Education, 61/63 Kleczewska Street, 01-826 Warsaw, Poland
| | - Zofia Wicik
- Department of Human Epigenetics, Mossakowski Medical Research Centre, PAS, 5 Pawinskiego Street, 02-106 Warsaw, Poland
| | - Jacek Połosak
- Department of Human Epigenetics, Mossakowski Medical Research Centre, PAS, 5 Pawinskiego Street, 02-106 Warsaw, Poland
| | - Olga Buyanovskaya
- Department of Human Epigenetics, Mossakowski Medical Research Centre, PAS, 5 Pawinskiego Street, 02-106 Warsaw, Poland
| | - Lukasz Charzewski
- Faculty of Physics, University of Warsaw, 5 Pasteur Street, 02-093 Warsaw, Poland
| | - Marek Stańczyk
- Department of General Surgery, Wolski Hospital, 17 Kasprzaka Street, 01-211 Warsaw, Poland
| | - Bartłomiej Noszczyk
- Department of Plastic Surgery, Medical Centre of Postgraduate Education, 99/103 Marymoncka Street, 01-813 Warsaw, Poland
| | - Monika Puzianowska-Kuznicka
- Department of Human Epigenetics, Mossakowski Medical Research Centre, PAS, 5 Pawinskiego Street, 02-106 Warsaw, Poland.,Department of Geriatrics & Gerontology, Medical Centre of Postgraduate Education, 61/63 Kleczewska Street, 01-826 Warsaw, Poland
| |
Collapse
|
398
|
Mohamed NS, Wilkie WA, Remily EA, Delanois RE. Can human placental extract help patients with osteoarthritis? ANNALS OF TRANSLATIONAL MEDICINE 2020; 8:62. [PMID: 32176220 PMCID: PMC7048971 DOI: 10.21037/atm.2019.12.135] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/10/2019] [Accepted: 12/16/2019] [Indexed: 11/06/2022]
Affiliation(s)
- Nequesha S Mohamed
- Rubin Institute for Advanced Orthopedics, Center for Joint Preservation and Replacement, Sinai Hospital of Baltimore, Baltimore, MD, USA
| | - Wayne A Wilkie
- Rubin Institute for Advanced Orthopedics, Center for Joint Preservation and Replacement, Sinai Hospital of Baltimore, Baltimore, MD, USA
| | - Ethan A Remily
- Rubin Institute for Advanced Orthopedics, Center for Joint Preservation and Replacement, Sinai Hospital of Baltimore, Baltimore, MD, USA
| | - Ronald E Delanois
- Rubin Institute for Advanced Orthopedics, Center for Joint Preservation and Replacement, Sinai Hospital of Baltimore, Baltimore, MD, USA
| |
Collapse
|
399
|
Abstract
The past two centuries have witnessed an unprecedented rise in human life expectancy. Sustaining longer lives with reduced periods of disability will require an understanding of the underlying mechanisms of ageing, and genetics is a powerful tool for identifying these mechanisms. Large-scale genome-wide association studies have recently identified many loci that influence key human ageing traits, including lifespan. Multi-trait loci have been linked with several age-related diseases, suggesting shared ageing influences. Mutations that drive accelerated ageing in prototypical progeria syndromes in humans point to an important role for genome maintenance and stability. Together, these different strands of genetic research are highlighting pathways for the discovery of anti-ageing interventions that may be applicable in humans.
Collapse
|
400
|
Stahl EC, Delgado ER, Alencastro F, LoPresti ST, Wilkinson PD, Roy N, Haschak MJ, Skillen CD, Monga SP, Duncan AW, Brown BN. Inflammation and Ectopic Fat Deposition in the Aging Murine Liver Is Influenced by CCR2. THE AMERICAN JOURNAL OF PATHOLOGY 2020; 190:372-387. [PMID: 31843499 PMCID: PMC7013280 DOI: 10.1016/j.ajpath.2019.10.016] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/30/2019] [Revised: 10/02/2019] [Accepted: 10/22/2019] [Indexed: 02/08/2023]
Abstract
Aging is associated with inflammation and metabolic syndrome, which manifests in the liver as nonalcoholic fatty liver disease (NAFLD). NAFLD can range in severity from steatosis to fibrotic steatohepatitis and is a major cause of hepatic morbidity. However, the pathogenesis of NAFLD in naturally aged animals is unclear. Herein, we performed a comprehensive study of lipid content and inflammatory signature of livers in 19-month-old aged female mice. These animals exhibited increased body and liver weight, hepatic triglycerides, and inflammatory gene expression compared with 3-month-old young controls. The aged mice also had a significant increase in F4/80+ hepatic macrophages, which coexpressed CD11b, suggesting a circulating monocyte origin. A global knockout of the receptor for monocyte chemoattractant protein (CCR2) prevented excess steatosis and inflammation in aging livers but did not reduce the number of CD11b+ macrophages, suggesting changes in macrophage accumulation precede or are independent from chemokine (C-C motif) ligand-CCR2 signaling in the development of age-related NAFLD. RNA sequencing further elucidated complex changes in inflammatory and metabolic gene expression in the aging liver. In conclusion, we report a previously unknown accumulation of CD11b+ macrophages in aged livers with robust inflammatory and metabolic transcriptomic changes. A better understanding of the hallmarks of aging in the liver will be crucial in the development of preventive measures and treatments for end-stage liver disease in elderly patients.
Collapse
Affiliation(s)
- Elizabeth C Stahl
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania; Department of Pathology, University of Pittsburgh School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Evan R Delgado
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania; Department of Pathology, University of Pittsburgh School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Frances Alencastro
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania; Department of Pathology, University of Pittsburgh School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Samuel T LoPresti
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania; Bioengineering Department, Swanson School of Engineering, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Patrick D Wilkinson
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania; Department of Pathology, University of Pittsburgh School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Nairita Roy
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania; Department of Pathology, University of Pittsburgh School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Martin J Haschak
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania; Bioengineering Department, Swanson School of Engineering, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Clint D Skillen
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Satdarshan P Monga
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania; Department of Pathology, University of Pittsburgh School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania; Pittsburgh Liver Research Center, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Andrew W Duncan
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania; Department of Pathology, University of Pittsburgh School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania; Pittsburgh Liver Research Center, University of Pittsburgh, Pittsburgh, Pennsylvania.
| | - Bryan N Brown
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania; Bioengineering Department, Swanson School of Engineering, University of Pittsburgh, Pittsburgh, Pennsylvania; Pittsburgh Liver Research Center, University of Pittsburgh, Pittsburgh, Pennsylvania.
| |
Collapse
|