201
|
Chen J, Zhang C, Wu Y. Does adjuvant chemotherapy improve outcomes in elderly patients with colorectal cancer? A systematic review and meta-analysis of real-world studies. Expert Rev Gastroenterol Hepatol 2022; 16:383-391. [PMID: 35303773 DOI: 10.1080/17474124.2022.2056014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/09/2022]
Abstract
BACKGROUND Elderly patients are frequently excluded from randomized trials. It is unclear if adjuvant chemotherapy improves outcomes of colorectal cancer in such patients. The current study aimed to review evidence on the impact of adjuvant chemotherapy on overall survival (OS) and disease-free survival (DFS) in elderly patients with stage II/III colorectal cancer by pooling data from real-world studies. METHODS PubMed, Embase, ScienceDirect, CENTRAL, and Google Scholar databases were searched for observational studies reporting adjusted data on OS and DFS in elderly (≥70 years) colorectal cancer patients based on receipt of adjuvant chemotherapy. RESULTS Thirteen studies included. The meta-analysis demonstrated statistically significant improved OS in elderly patients receiving adjuvant chemotherapy (p < 0.00001). Results were similar for sub-group analysis based on cancer stage and definition of elderly. Improvement in OS was noted in only Western population studies with no difference in Asian patients. The meta-analysis also revealed no statistically significant difference in DFS in elderly patients receiving adjuvant chemotherapy vs surgery alone (p = 0.14). CONCLUSION Real-world evidence indicates that adjuvant chemotherapy significantly improved OS but not DFS in elderly colorectal cancer patients. Scarce evidence suggests a limited role of adjuvant chemotherapy in Asian patients which needs confirmation by further studies.
Collapse
Affiliation(s)
- Jianbing Chen
- Department of Radiotherapy, Heping Hospital Affiliated to Changzhi Medical College, Changzhi, P.R China
| | - Chengda Zhang
- Department of Gastroenterology, The Third Hospital of Mianyang (Sichuan Mental Health Center), Mianyang, P.R China
| | - Yajuan Wu
- The Second Chest Radiotheropy Department, Shanxi Province Cancer Hospital/ Shanxi Hospital Affiliated to Cancer Hospital, Chinese Academy of Medical Sciences/Cancer Hospital Affiliated to Shanxi Medical University, Taiyuan, P.R China
| |
Collapse
|
202
|
Pretzsch E, Nieß H, Bösch F, Westphalen C, Jacob S, Neumann J, Werner J, Heinemann V, Angele M. Age and metastasis – How age influences metastatic spread in cancer. Colorectal cancer as a model. Cancer Epidemiol 2022; 77:102112. [PMID: 35104771 DOI: 10.1016/j.canep.2022.102112] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2021] [Revised: 01/15/2022] [Accepted: 01/17/2022] [Indexed: 12/12/2022]
|
203
|
Murakami T, Inagaki N, Kondoh H. Cellular Senescence in Diabetes Mellitus: Distinct Senotherapeutic Strategies for Adipose Tissue and Pancreatic β Cells. Front Endocrinol (Lausanne) 2022; 13:869414. [PMID: 35432205 PMCID: PMC9009089 DOI: 10.3389/fendo.2022.869414] [Citation(s) in RCA: 50] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/04/2022] [Accepted: 03/02/2022] [Indexed: 12/15/2022] Open
Abstract
Increased insulin resistance and impaired insulin secretion are significant characteristics manifested by patients with type 2 diabetes mellitus (T2DM). The degree and extent of these two features in T2DM vary among races and individuals. Insulin resistance is accelerated by obesity and is accompanied by accumulation of dysfunctional adipose tissues. In addition, dysfunction of pancreatic β-cells impairs insulin secretion. T2DM is significantly affected by aging, as the β-cell mass diminishes with age. Moreover, both obesity and hyperglycemia-related metabolic changes in developing diabetes are associated with accumulation of senescent cells in multiple organs, that is, organismal aging. Cellular senescence is defined as a state of irreversible cell cycle arrest with concomitant functional decline. It is caused by telomere shortening or senescence-inducing stress. Senescent cells secrete proinflammatory cytokines and chemokines, which is designated as the senescence-associated secretory phenotype (SASP), and this has a negative impact on adipose tissues and pancreatic β-cells. Recent advances in aging research have suggested that senolysis, the removal of senescent cells, can be a promising therapeutic approach to prevent or improve aging-related diseases, including diabetes. The attenuation of a SASP may be beneficial, although the pathophysiological involvement of cellular senescence in diabetes is not fully understood. In the clinical application of senotherapy, tissue-context-dependent senescent cells are increasingly being recognized as an issue to be solved. Recent studies have observed highly heterogenic and complex senescent cell populations that serve distinct roles among tissues, various stages of disease, and different ages. For example, in high-fat-diet induced diabetes with obesity, mouse adipose tissues display accumulation of p21Cip1-highly-expressing (p21high) cells in the early stage, followed by increases in both p21high and p16INK4a-highly-expressing (p16high) cells in the late stage. Interestingly, elimination of p21high cells in visceral adipose tissue can prevent or improve insulin resistance in mice with obesity, while p16high cell clearance is less effective in alleviating insulin resistance. Importantly, in immune-deficient mice transplanted with fat from obese patients, dasatinib plus quercetin, a senolytic cocktail that reduces the number of both p21high and p16high cells, improves both glucose tolerance and insulin resistance. On the other hand, in pancreatic β cells, p16high cells become increasingly predominant with age and development of diabetes. Consistently, elimination of p16high cells in mice improves both glucose tolerance and glucose-induced insulin secretion. Moreover, a senolytic compound, the anti-Bcl-2 inhibitor ABT263 reduces p16INK4a expression in islets and restores glucose tolerance in mice when combined with insulin receptor antagonist S961 treatment. In addition, efficacy of senotherapy in targeting mouse pancreatic β cells has been validated not only in T2DM, but also in type 1 diabetes mellitus. Indeed, in non-obese diabetic mice, treatment with anti-Bcl-2 inhibitors, such as ABT199, eliminates senescent pancreatic β cells, resulting in prevention of diabetes mellitus. These findings clearly indicate that features of diabetes are partly determined by which or where senescent cells reside in vivo, as adipose tissues and pancreatic β cells are responsible for insulin resistance and insulin secretion, respectively. In this review, we summarize recent advances in understanding cellular senescence in adipose tissues and pancreatic β cells in diabetes. We review the different potential molecular targets and distinctive senotherapeutic strategies in adipose tissues and pancreatic β cells. We propose the novel concept of a dual-target tailored approach in senotherapy against diabetes.
Collapse
Affiliation(s)
- Takaaki Murakami
- Department of Diabetes, Endocrinology and Nutrition, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Nobuya Inagaki
- Department of Diabetes, Endocrinology and Nutrition, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Hiroshi Kondoh
- Department of Diabetes, Endocrinology and Nutrition, Kyoto University Graduate School of Medicine, Kyoto, Japan
- Geriatric Unit, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| |
Collapse
|
204
|
Gautheron J, Lima L, Akinci B, Zammouri J, Auclair M, Ucar SK, Ozen S, Altay C, Bax BE, Nemazanyy I, Lenoir V, Prip-Buus C, Acquaviva-Bourdain C, Lascols O, Fève B, Vigouroux C, Noel E, Jéru I. Loss of thymidine phosphorylase activity disrupts adipocyte differentiation and induces insulin-resistant lipoatrophic diabetes. BMC Med 2022; 20:95. [PMID: 35341481 PMCID: PMC8958798 DOI: 10.1186/s12916-022-02296-2] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Accepted: 02/10/2022] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND Thymidine phosphorylase (TP), encoded by the TYMP gene, is a cytosolic enzyme essential for the nucleotide salvage pathway. TP catalyzes the phosphorylation of the deoxyribonucleosides, thymidine and 2'-deoxyuridine, to thymine and uracil. Biallelic TYMP variants are responsible for Mitochondrial NeuroGastroIntestinal Encephalomyopathy (MNGIE), an autosomal recessive disorder characterized in most patients by gastrointestinal and neurological symptoms, ultimately leading to death. Studies on the impact of TYMP variants in cellular systems with relevance to the organs affected in MNGIE are still scarce and the role of TP in adipose tissue remains unexplored. METHODS Deep phenotyping was performed in three patients from two families carrying homozygous TYMP variants and presenting with lipoatrophic diabetes. The impact of the loss of TP expression was evaluated using a CRISPR-Cas9-mediated TP knockout (KO) strategy in human adipose stem cells (ASC), which can be differentiated into adipocytes in vitro. Protein expression profiles and cellular characteristics were investigated in this KO model. RESULTS All patients had TYMP loss-of-function variants and first presented with generalized loss of adipose tissue and insulin-resistant diabetes. CRISPR-Cas9-mediated TP KO in ASC abolished adipocyte differentiation and decreased insulin response, consistent with the patients' phenotype. This KO also induced major oxidative stress, altered mitochondrial functions, and promoted cellular senescence. This translational study identifies a new role of TP by demonstrating its key regulatory functions in adipose tissue. CONCLUSIONS The implication of TP variants in atypical forms of monogenic diabetes shows that genetic diagnosis of lipodystrophic syndromes should include TYMP analysis. The fact that TP is crucial for adipocyte differentiation and function through the control of mitochondrial homeostasis highlights the importance of mitochondria in adipose tissue biology.
Collapse
Affiliation(s)
- Jérémie Gautheron
- Centre de Recherche Saint-Antoine (CRSA), Sorbonne Université-Inserm UMRS_938, 27 rue Chaligny 75571, 12, Paris Cedex, France.
- Institute of Cardiometabolism and Nutrition (ICAN), CHU Pitié-Salpêtrière - Saint-Antoine, Assistance Publique-Hôpitaux de Paris (AP-HP), 75012, Paris, France.
| | - Lara Lima
- Centre de Recherche Saint-Antoine (CRSA), Sorbonne Université-Inserm UMRS_938, 27 rue Chaligny 75571, 12, Paris Cedex, France
- Institute of Cardiometabolism and Nutrition (ICAN), CHU Pitié-Salpêtrière - Saint-Antoine, Assistance Publique-Hôpitaux de Paris (AP-HP), 75012, Paris, France
| | - Baris Akinci
- Department of Internal Medicine, Division of Endocrinology and Metabolism, Dokuz Eylul University, 35330, Izmir, Turkey
| | - Jamila Zammouri
- Centre de Recherche Saint-Antoine (CRSA), Sorbonne Université-Inserm UMRS_938, 27 rue Chaligny 75571, 12, Paris Cedex, France
- Institute of Cardiometabolism and Nutrition (ICAN), CHU Pitié-Salpêtrière - Saint-Antoine, Assistance Publique-Hôpitaux de Paris (AP-HP), 75012, Paris, France
| | - Martine Auclair
- Centre de Recherche Saint-Antoine (CRSA), Sorbonne Université-Inserm UMRS_938, 27 rue Chaligny 75571, 12, Paris Cedex, France
- Institute of Cardiometabolism and Nutrition (ICAN), CHU Pitié-Salpêtrière - Saint-Antoine, Assistance Publique-Hôpitaux de Paris (AP-HP), 75012, Paris, France
| | - Sema Kalkan Ucar
- Department of Pediatrics, Division of Metabolic Diseases, Ege University, 35100, Izmir, Turkey
| | - Samim Ozen
- Department of Pediatrics, Division of Pediatric Endocrinology, Ege University, 35100, Izmir, Turkey
| | - Canan Altay
- Department of Radiology, Dokuz Eylul University, 35100, Izmir, Turkey
| | - Bridget E Bax
- Institute of Molecular and Clinical Sciences, St George's University of London, London, SW17 0RE, UK
| | - Ivan Nemazanyy
- Platform for Metabolic Analyses, Structure Fédérative de Recherche Necker, Inserm, US24/CNRS UMS 3633, 75015, Paris, France
| | - Véronique Lenoir
- Institut Cochin, Université Paris Descartes-CNRS UMR8104, Paris, France
| | - Carina Prip-Buus
- Institut Cochin, Université Paris Descartes-CNRS UMR8104, Paris, France
| | - Cécile Acquaviva-Bourdain
- Service de Biochimie et Biologie Moléculaire Grand Est, Hospices Civils, UM Pathologies Héréditaires du Métabolisme et du Globule Rouge, CHU de Lyon, 69500, Bron, France
| | - Olivier Lascols
- Centre de Recherche Saint-Antoine (CRSA), Sorbonne Université-Inserm UMRS_938, 27 rue Chaligny 75571, 12, Paris Cedex, France
- Institute of Cardiometabolism and Nutrition (ICAN), CHU Pitié-Salpêtrière - Saint-Antoine, Assistance Publique-Hôpitaux de Paris (AP-HP), 75012, Paris, France
- Laboratoire commun de Biologie et Génétique Moléculaires, Hôpital Saint-Antoine, AP-HP, 75012, Paris, France
| | - Bruno Fève
- Centre de Recherche Saint-Antoine (CRSA), Sorbonne Université-Inserm UMRS_938, 27 rue Chaligny 75571, 12, Paris Cedex, France
- Institute of Cardiometabolism and Nutrition (ICAN), CHU Pitié-Salpêtrière - Saint-Antoine, Assistance Publique-Hôpitaux de Paris (AP-HP), 75012, Paris, France
- Centre National de Référence des Pathologies Rares de l'Insulino-Sécrétion et de l'Insulino-Sensibilité (PRISIS), Service de Diabétologie et Endocrinologie de la Reproduction, Hôpital Saint-Antoine, AP-HP, 75012, Paris, France
| | - Corinne Vigouroux
- Centre de Recherche Saint-Antoine (CRSA), Sorbonne Université-Inserm UMRS_938, 27 rue Chaligny 75571, 12, Paris Cedex, France
- Institute of Cardiometabolism and Nutrition (ICAN), CHU Pitié-Salpêtrière - Saint-Antoine, Assistance Publique-Hôpitaux de Paris (AP-HP), 75012, Paris, France
- Laboratoire commun de Biologie et Génétique Moléculaires, Hôpital Saint-Antoine, AP-HP, 75012, Paris, France
- Centre National de Référence des Pathologies Rares de l'Insulino-Sécrétion et de l'Insulino-Sensibilité (PRISIS), Service de Diabétologie et Endocrinologie de la Reproduction, Hôpital Saint-Antoine, AP-HP, 75012, Paris, France
| | - Esther Noel
- Département de Médecine Interne, Centre Hospitalier Universitaire, 67000, Strasbourg, France
| | - Isabelle Jéru
- Centre de Recherche Saint-Antoine (CRSA), Sorbonne Université-Inserm UMRS_938, 27 rue Chaligny 75571, 12, Paris Cedex, France.
- Institute of Cardiometabolism and Nutrition (ICAN), CHU Pitié-Salpêtrière - Saint-Antoine, Assistance Publique-Hôpitaux de Paris (AP-HP), 75012, Paris, France.
- Laboratoire commun de Biologie et Génétique Moléculaires, Hôpital Saint-Antoine, AP-HP, 75012, Paris, France.
| |
Collapse
|
205
|
Frasca D. Obesity Accelerates Age Defects in Human B Cells and Induces Autoimmunity. IMMUNOMETABOLISM 2022; 4:e220010. [PMID: 35433040 PMCID: PMC9012215 DOI: 10.20900/immunometab20220010] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Accepted: 03/17/2022] [Indexed: 11/24/2022]
Abstract
Aging is associated with systemic inflammation and decreased production of protective antibodies while the production of autoimmune antibodies is increased. Our results have shown that the human obese adipose tissue (AT), which increases in size with aging, contributes to systemic and B cell intrinsic inflammation, reduced protective and increased pathogenic B cell responses leading to increased secretion of autoimmune antibodies. With this R56 funding, we have been able to investigate the cellular and molecular mechanisms by which the human obese AT induces intrinsic B cell inflammation and dysfunctional B cell responses, stimulates the secretion of autoimmune antibodies, whose specificity has been characterized, and engages different AT cell types in antigen presentation pathways to allow secretion of these autoimmune antibodies. Briefly, immune cells are recruited to the AT by chemokines released by both non-immune (adipocytes) and by resident and infiltrating immune cells. We have identified several mechanisms responsible for the release of "self" antigens, and we have shown that reduced oxygen availability and hypoxia, cell cytotoxicity and DNA damage induce cell death and lead to further release of pro-inflammatory cytokines, "self" protein antigens, cell-free DNA and lipids. We have also identified different antigen presenting cells in the AT, responsible for the activation of pathogenic B cells, class switch and secretion of autoimmune IgG antibodies. The experiments performed have allowed the discovery of novel mechanisms for pathogenic responses and the identification of pathways to target in order to promote better humoral immunity during aging.
Collapse
Affiliation(s)
- Daniela Frasca
- Department of Microbiology and Immunology, University of Miami Miller School of Medicine, Miami, FL 33136, USA
- Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| |
Collapse
|
206
|
Park S. Biochemical, structural and physical changes in aging human skin, and their relationship. Biogerontology 2022; 23:275-288. [PMID: 35292918 DOI: 10.1007/s10522-022-09959-w] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Accepted: 02/25/2022] [Indexed: 11/02/2022]
Abstract
Skin is the largest organ of the human body, having the purpose of regulating temperature, protecting us from microbes or mechanical shocks, and allowing the sensations from touch. It is generally accepted that aging induces profound changes in the skin's biochemical, structural and physical properties, which can lead to impaired biological functions and/or diverse diseases. So far, the effects of aging on these skin properties have been well documented. However, very few studies have focused exclusively on the relationship among these critical properties in the aging process, which is this review's primary focus. Many in vivo, ex vivo, and in vitro techniques have been previously used to characterize these properties of the skin. This review aims to provide a comprehensive overview on the effects of aging on the changes in biochemical, structural, and physical properties, and explore the potential mechanisms of skin with the relation between these properties. First, we review different or contradictory results of aging-related changes in representative parameters of each property, including the interpretations of the findings. Next, we discuss the need for a standardized method to characterize aging-related changes in these properties, to improve the way of defining age-property relationship. Moreover, potential mechanisms based on the previous results are explored by linking the biochemical, structural, and physical properties. Finally, the need to study changes of various functional properties in the separate skin layers is addressed. This review can help understand the underlying mechanism of aging-related alterations, to improve the evaluation of the aging process and guide effective treatment strategies for aging-related diseases.
Collapse
Affiliation(s)
- Seungman Park
- Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD, 21218, USA. .,Department of Mechanical Engineering, Johns Hopkins University, Baltimore, MD, 21218, USA.
| |
Collapse
|
207
|
Zhu Y, Prata LGL, Gerdes EOW, Netto JME, Pirtskhalava T, Giorgadze N, Tripathi U, Inman CL, Johnson KO, Xue A, Palmer AK, Chen T, Schaefer K, Justice JN, Nambiar AM, Musi N, Kritchevsky SB, Chen J, Khosla S, Jurk D, Schafer MJ, Tchkonia T, Kirkland JL. Orally-active, clinically-translatable senolytics restore α-Klotho in mice and humans. EBioMedicine 2022; 77:103912. [PMID: 35292270 PMCID: PMC9034457 DOI: 10.1016/j.ebiom.2022.103912] [Citation(s) in RCA: 53] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2021] [Revised: 02/16/2022] [Accepted: 02/17/2022] [Indexed: 02/01/2023] Open
Abstract
BACKGROUND α-Klotho is a geroprotective protein that can attenuate or alleviate deleterious changes with ageing and disease. Declines in α-Klotho play a role in the pathophysiology of multiple diseases and age-related phenotypes. Pre-clinical evidence suggests that boosting α-Klotho holds therapeutic potential. However, readily clinically-translatable, practical strategies for increasing α-Klotho are not at hand. Here, we report that orally-active, clinically-translatable senolytics can increase α-Klotho in mice and humans. METHODS We examined α-Klotho expression in three different human primary cell types co-cultured with conditioned medium (CM) from senescent or non-senescent cells with or without neutralizing antibodies. We assessed α-Klotho expression in aged, obese, and senescent cell-transplanted mice treated with vehicle or senolytics. We assayed urinary α-Klotho in patients with idiopathic pulmonary fibrosis (IPF) who were treated with the senolytic drug combination, Dasatinib plus Quercetin (D+Q). FINDINGS We found exposure to the senescent cell secretome reduces α-Klotho in multiple nonsenescent human cell types. This was partially prevented by neutralizing antibodies against the senescence-associated secretory phenotype (SASP) factors, activin A and Interleukin 1α (IL-1α). Consistent with senescent cells' being a cause of decreased α-Klotho, transplanting senescent cells into younger mice reduced brain and urine α-Klotho. Selectively removing senescent cells genetically or pharmacologically increased α-Klotho in urine, kidney, and brain of mice with increased senescent cell burden, including naturally-aged, diet-induced obese (DIO), or senescent cell-transplanted mice. D+Q increased α-Klotho in urine of patients with IPF, a disease linked to cellular senescence. INTERPRETATION Senescent cells cause reduced α-Klotho, partially due to their production of activin A and IL-1α. Targeting senescent cells boosts α-Klotho in mice and humans. Thus, clearing senescent cells restores α-Klotho, potentially opening a novel, translationally-feasible avenue for developing orally-active small molecule, α-Klotho-enhancing clinical interventions. Furthermore, urinary α-Klotho may prove to be a useful test for following treatments in senolytic clinical trials. FUNDING This work was supported by National Institute of Health grants AG013925 (J.L.K.), AG062413 (J.L.K., S.K.), AG044271 (N.M.), AG013319 (N.M.), and the Translational Geroscience Network (AG061456: J.L.K., T.T., N.M., S.B.K., S.K.), Robert and Arlene Kogod (J.L.K.), the Connor Group (J.L.K.), Robert J. and Theresa W. Ryan (J.L.K.), and the Noaber Foundation (J.L.K.). The previous IPF clinical trial was supported by the Claude D. Pepper Older Americans Independence Centers at WFSM (AG021332: J.N.J., S.B.K.), UTHSCA (AG044271: A.M.N.), and the Translational Geroscience Network.
Collapse
Affiliation(s)
- Yi Zhu
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, 200 First St., S.W., Rochester, MN 55905, USA,Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN 55905, USA,Corresponding authors at: Robert and Arlene Kogod Center on Aging, Mayo Clinic, 200 First St., S.W., Rochester, MN 55905, USA.
| | | | - Erin O. Wissler Gerdes
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, 200 First St., S.W., Rochester, MN 55905, USA
| | | | - Tamar Pirtskhalava
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, 200 First St., S.W., Rochester, MN 55905, USA
| | - Nino Giorgadze
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, 200 First St., S.W., Rochester, MN 55905, USA
| | - Utkarsh Tripathi
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, 200 First St., S.W., Rochester, MN 55905, USA
| | - Christina L. Inman
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, 200 First St., S.W., Rochester, MN 55905, USA
| | - Kurt O. Johnson
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, 200 First St., S.W., Rochester, MN 55905, USA
| | - Ailing Xue
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, 200 First St., S.W., Rochester, MN 55905, USA
| | - Allyson K. Palmer
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, 200 First St., S.W., Rochester, MN 55905, USA
| | - Tingjun Chen
- Department of Neurology, Mayo Clinic, Rochester, MN 55905, USA
| | - Kalli Schaefer
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, 200 First St., S.W., Rochester, MN 55905, USA
| | - Jamie N. Justice
- Sticht Center for Healthy Aging and Alzheimer's Prevention, Internal Medicine, Gerontology and Geriatric Medicine, Wake Forest School of Medicine, Winston-Salem, NC 27157, USA
| | - Anoop M. Nambiar
- Division of Pulmonary Diseases and Critical Care Medicine, Department of Internal Medicine, University of Texas Health Sciences Center at San Antonio and South Texas Veterans Health Care System, San Antonio, TX 78229, USA
| | - Nicolas Musi
- Barshop Institute for Longevity and Aging Studies, Center for Healthy Aging, University of Texas Health Sciences Center at San Antonio and Geriatric Research, Education, and Clinical Center, South Texas Veterans Health Care System, San Antonio, TX 78229, USA
| | - Stephen B. Kritchevsky
- Sticht Center for Healthy Aging and Alzheimer's Prevention, Internal Medicine, Gerontology and Geriatric Medicine, Wake Forest School of Medicine, Winston-Salem, NC 27157, USA
| | - Jun Chen
- Division of Computational Biology, Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN 55905, USA
| | - Sundeep Khosla
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, 200 First St., S.W., Rochester, MN 55905, USA,Division of Endocrinology, Department of Medicine, Mayo Clinic, Rochester, MN 55905, USA
| | - Diana Jurk
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, 200 First St., S.W., Rochester, MN 55905, USA,Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN 55905, USA,Department of Neurology, Mayo Clinic, Rochester, MN 55905, USA
| | - Marissa J. Schafer
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, 200 First St., S.W., Rochester, MN 55905, USA,Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN 55905, USA
| | - Tamar Tchkonia
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, 200 First St., S.W., Rochester, MN 55905, USA,Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN 55905, USA
| | - James L. Kirkland
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, 200 First St., S.W., Rochester, MN 55905, USA,Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN 55905, USA,Division of General Internal Medicine, Department of Medicine, Mayo Clinic, Rochester, MN 55905, USA,Corresponding authors at: Robert and Arlene Kogod Center on Aging, Mayo Clinic, 200 First St., S.W., Rochester, MN 55905, USA.
| |
Collapse
|
208
|
Espinosa De Ycaza AE, Søndergaard E, Morgan-Bathke M, Lytle K, Delivanis DA, Ramos P, Carranza Leon BG, Jensen MD. Adipose Tissue Inflammation Is Not Related to Adipose Insulin Resistance in Humans. Diabetes 2022; 71:381-393. [PMID: 34857544 PMCID: PMC8893944 DOI: 10.2337/db21-0609] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/07/2021] [Accepted: 11/18/2021] [Indexed: 11/13/2022]
Abstract
The role of adipose tissue (AT) inflammation in AT function in humans is unclear. We tested whether AT macrophage (ATM) content, cytokine gene expression, and senescent cell burden (markers of AT inflammation) predict AT insulin resistance measured as the insulin concentration that suppresses lipolysis by 50% (IC50). We studied 86 volunteers with normal weight or obesity at baseline and a subgroup of 25 volunteers with obesity before and after weight loss. There was a strong positive relationship between IC50 and abdominal subcutaneous and femoral fat cell size (FCS). The positive, univariate relationships between IC50 and abdominal AT inflammatory markers CD68, CD14, CD206 ATM/100 adipocytes, senescent cells, IL-6, and TNF-α mRNA were not significant after adjustment for FCS. A 10% weight loss significantly reduced IC50; however, there was no reduction in adipose ATM content, senescent cells, or cytokine gene expression. Our study suggests that commonly used markers of AT inflammation are not causally linked to AT insulin resistance, whereas FCS is a strong predictor of AT insulin resistance with respect to lipolysis.
Collapse
Affiliation(s)
- Ana Elena Espinosa De Ycaza
- Endocrine Research Unit, Mayo Clinic, Rochester, MN
- Facultad de Medicina, Universidad de Panamá, Panama City, Republic of Panama
- Panamanian Institute of Biological Research, Panama City, Republic of Panama
| | - Esben Søndergaard
- Endocrine Research Unit, Mayo Clinic, Rochester, MN
- Steno Diabetes Center Aarhus, Aarhus, Denmark
- The Danish Diabetes Academy, Odense, Denmark
| | - Maria Morgan-Bathke
- Endocrine Research Unit, Mayo Clinic, Rochester, MN
- Nutrition and Dietetics, Viterbo University, La Crosse, WI
| | - Kelli Lytle
- Endocrine Research Unit, Mayo Clinic, Rochester, MN
| | | | - Paola Ramos
- Endocrine Research Unit, Mayo Clinic, Rochester, MN
| | - Barbara Gisella Carranza Leon
- Endocrine Research Unit, Mayo Clinic, Rochester, MN
- Division of Diabetes, Endocrinology and Metabolism, Vanderbilt University Medical Center, Nashville, TN
| | - Michael D. Jensen
- Endocrine Research Unit, Mayo Clinic, Rochester, MN
- Corresponding author: Michael D. Jensen,
| |
Collapse
|
209
|
The Shades of Grey in Adipose Tissue Reprogramming. Biosci Rep 2022; 42:230844. [PMID: 35211733 PMCID: PMC8905306 DOI: 10.1042/bsr20212358] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Revised: 02/22/2022] [Accepted: 02/24/2022] [Indexed: 11/22/2022] Open
Abstract
The adipose tissue (AT) has a major role in contributing to obesity-related pathologies through regulating systemic immunometabolism. The pathogenicity of the AT is underpinned by its remarkable plasticity to be reprogrammed during obesity, in the perspectives of tissue morphology, extracellular matrix (ECM) composition, angiogenesis, immunometabolic homoeostasis and circadian rhythmicity. Dysregulation in these features escalates the pathogenesis conferred by this endometabolic organ. Intriguingly, the potential to be reprogrammed appears to be an Achilles’ heel of the obese AT that can be targeted for the management of obesity and its associated comorbidities. Here, we provide an overview of the reprogramming processes of white AT (WAT), with a focus on their dynamics and pleiotropic actions over local and systemic homoeostases, followed by a discussion of potential strategies favouring therapeutic reprogramming. The potential involvement of AT remodelling in the pathogenesis of COVID-19 is also discussed.
Collapse
|
210
|
Pan Y, Li Y, Liu P, Zhang Y, Li B, Liu Z, Shui G, Ma L. Metabolomics-Based Frailty Biomarkers in Older Chinese Adults. Front Med (Lausanne) 2022; 8:830723. [PMID: 35155487 PMCID: PMC8825494 DOI: 10.3389/fmed.2021.830723] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Accepted: 12/20/2021] [Indexed: 11/14/2022] Open
Abstract
Background/Objectives Owing to accelerated population aging, health in older adults is becoming increasingly important. Frailty can reflect the health status and disease risks of older adults; however, appropriate biomarkers for early screening of frailty have not been identified. Here, we applied metabolomics to identify frailty biomarkers and potential pathogenic mechanisms of frailty. Methods Serum metabolic profiles from 25 frail and 49 non-frail (control) older adults were systematically investigated by liquid chromatography-mass spectrometry-based metabolomics. Results We identified 349 metabolites of 46 classes, with four increased and seven decreased metabolites in frail older adults. Pearson correlation analysis identified 11 and 21 metabolites that were positively and negatively correlated with grip strength, and 7 and 76 metabolites that were positively and negatively correlated with gait speed, respectively. Pathway analysis identified 10 metabolite sets and 13 pathways significantly associated with one or more frailty phenotype criteria. Conclusion These results revealed the metabolite characteristics of serum in frail older adults. Intermediates of carbohydrate metabolism (e.g., isocitrate, malate, fumarate, cis-aconitate, glucuronate, and pyruvate), saturated fatty acids (e.g., palmitic acid), unsaturated fatty acids (e.g., arachidonate and linoleic acid), and certain essential amino acids (e.g., tryptophan) may be candidate biomarkers for the early diagnosis of frailty. Mitochondrial function disorders, saturated fatty acid-mediated lipotoxicity, aberrant unsaturated fatty acid metabolism, and increased tryptophan degradation could be potential mechanisms of frailty.
Collapse
Affiliation(s)
- Yiming Pan
- Department of Geriatrics, National Research Center for Geriatric Medicine, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Yun Li
- Department of Geriatrics, National Research Center for Geriatric Medicine, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Pan Liu
- Department of Geriatrics, National Research Center for Geriatric Medicine, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Yaxin Zhang
- Department of Geriatrics, National Research Center for Geriatric Medicine, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Bowen Li
- LipidALL Technologies Company Limited, Changzhou, China
| | - Zuyun Liu
- Center for Clinical Big Data and Analytics, School of Public Health, Second Affiliated Hospital and Department of Big Data in Health Science, Zhejiang University School of Medicine, Hangzhou, China
| | - Guanghou Shui
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, China
| | - Lina Ma
- Department of Geriatrics, National Research Center for Geriatric Medicine, Xuanwu Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
211
|
Conte M, Petraglia L, Poggio P, Valerio V, Cabaro S, Campana P, Comentale G, Attena E, Russo V, Pilato E, Formisano P, Leosco D, Parisi V. Inflammation and Cardiovascular Diseases in the Elderly: The Role of Epicardial Adipose Tissue. Front Med (Lausanne) 2022; 9:844266. [PMID: 35242789 PMCID: PMC8887867 DOI: 10.3389/fmed.2022.844266] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2021] [Accepted: 01/13/2022] [Indexed: 01/08/2023] Open
Abstract
Human aging is a complex phenomenon characterized by a wide spectrum of biological changes which impact on behavioral and social aspects. Age-related changes are accompanied by a decline in biological function and increased vulnerability leading to frailty, thereby advanced age is identified among the major risk factors of the main chronic human diseases. Aging is characterized by a state of chronic low-grade inflammation, also referred as inflammaging. It recognizes a multifactorial pathogenesis with a prominent role of the innate immune system activation, resulting in tissue degeneration and contributing to adverse outcomes. It is widely recognized that inflammation plays a central role in the development and progression of numerous chronic and cardiovascular diseases. In particular, low-grade inflammation, through an increased risk of atherosclerosis and insulin resistance, promote cardiovascular diseases in the elderly. Low-grade inflammation is also promoted by visceral adiposity, whose accumulation is paralleled by an increased inflammatory status. Aging is associated to increase in epicardial adipose tissue (EAT), the visceral fat depot of the heart. Structural and functional changes in EAT have been shown to be associated with several heart diseases, including coronary artery disease, aortic stenosis, atrial fibrillation, and heart failure. EAT increase is associated with a greater production and secretion of pro-inflammatory mediators and neuro-hormones, so that thickened EAT can pathologically influence, in a paracrine and vasocrine manner, the structure and function of the heart and is associated to a worse cardiovascular outcome. In this review, we will discuss the evidence underlying the interplay between inflammaging, EAT accumulation and cardiovascular diseases. We will examine and discuss the importance of EAT quantification, its characteristics and changes with age and its clinical implication.
Collapse
Affiliation(s)
- Maddalena Conte
- Department of Translational Medical Sciences, University of Naples Federico II, Naples, Italy
- Casa di Cura San Michele, Maddaloni, Italy
| | - Laura Petraglia
- Department of Translational Medical Sciences, University of Naples Federico II, Naples, Italy
| | | | | | - Serena Cabaro
- Department of Translational Medical Sciences, University of Naples Federico II, Naples, Italy
| | - Pasquale Campana
- Department of Translational Medical Sciences, University of Naples Federico II, Naples, Italy
| | - Giuseppe Comentale
- Department of Advanced Biomedical Science, University of Naples Federico II, Naples, Italy
| | - Emilio Attena
- Department of Cardiology, Monaldi Hospital, Naples, Italy
| | - Vincenzo Russo
- Department of Medical Translational Sciences, Monaldi Hospital, University of Campania Luigi Vanvitelli, Campania, Italy
| | - Emanuele Pilato
- Department of Advanced Biomedical Science, University of Naples Federico II, Naples, Italy
| | - Pietro Formisano
- Department of Translational Medical Sciences, University of Naples Federico II, Naples, Italy
| | - Dario Leosco
- Department of Translational Medical Sciences, University of Naples Federico II, Naples, Italy
| | - Valentina Parisi
- Department of Translational Medical Sciences, University of Naples Federico II, Naples, Italy
| |
Collapse
|
212
|
Lee J, Jo SE, Lee J, Kim JH. An in vitro evaluation of luffa cylindrica stem sap in preadipocytes and dermal fibroblasts. Biochem Biophys Res Commun 2022; 599:100-105. [PMID: 35180468 DOI: 10.1016/j.bbrc.2022.02.024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2022] [Revised: 02/03/2022] [Accepted: 02/08/2022] [Indexed: 11/17/2022]
Abstract
BACKGROUND Luffa cylindrica stem sap (LuCS) has been ethnopharmacologically used as a cosmetic ingredients to improve the facial condition in Asians, but there is no scientific proof about the advantages of LuCS as a supplement for skin elasticity inducer. PURPOSE Presently, we have validated the beneficial effect of LuCS in human preadipocyte and fibroblast. METHODS In vitro activities of LuCS on expression of cellular elastin and collagen type I were validated using Western blot analysis in human fibroblasts. Effect of LuCS on preadipocyte development was performed using MDI medium containing isobutyl-methylxanthine, dexamethasone, and insulin and then evaluated using oil red O staining. RESULTS Treatment of LuCS stimulated the expression of cellular elastin and type I procollagen in human skin fibroblasts. Exposure to LuCS induced lipid accumulation of preadipocytes via activation of CEBP/α signaling pathway in preadipocytes. Expression of collagen I, elastin, or CEBP/α mRNA was decreased by age. 3-bromo-3-methylisoxazol-5-amine enhanced the synthesis of cellular lipid in preadipocytes. CONCLUSIONS Collectively, these results suggest the rationale of LuCS treatment in enhancing the skin condition.
Collapse
Affiliation(s)
- Jungwhoi Lee
- Department of Applied Life Science, Jeju National University, Jeju-do, 63243, Republic of Korea.
| | - Sung-Eun Jo
- [chanchanhee] Inc, Jeju, 63243, Republic of Korea.
| | - Jungsul Lee
- Department of Bio and Brain Engineering, KAIST, Daejeon, 34141, South Korea.
| | - Jae-Hoon Kim
- Department of Applied Life Science, Jeju National University, Jeju-do, 63243, Republic of Korea; Subtropical/tropical Organism Gene Bank, Jeju National University, Jeju, 63243, Republic of Korea.
| |
Collapse
|
213
|
Raffaele M, Kovacovicova K, Biagini T, Lo Re O, Frohlich J, Giallongo S, Nhan JD, Giannone AG, Cabibi D, Ivanov M, Tonchev AB, Mistrik M, Lacey M, Dzubak P, Gurska S, Hajduch M, Bartek J, Mazza T, Micale V, Curran SP, Vinciguerra M. Nociceptin/orphanin FQ opioid receptor (NOP) selective ligand MCOPPB links anxiolytic and senolytic effects. GeroScience 2022; 44:463-483. [PMID: 34820764 PMCID: PMC8612119 DOI: 10.1007/s11357-021-00487-y] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2021] [Accepted: 11/05/2021] [Indexed: 01/18/2023] Open
Abstract
Accumulation of senescent cells may drive age-associated alterations and pathologies. Senolytics are promising therapeutics that can preferentially eliminate senescent cells. Here, we performed a high-throughput automatized screening (HTS) of the commercial LOPAC®Pfizer library on aphidicolin-induced senescent human fibroblasts, to identify novel senolytics. We discovered the nociceptin receptor FQ opioid receptor (NOP) selective ligand 1-[1-(1-methylcyclooctyl)-4-piperidinyl]-2-[(3R)-3-piperidinyl]-1H-benzimidazole (MCOPPB, a compound previously studied as potential anxiolytic) as the best scoring hit. The ability of MCOPPB to eliminate senescent cells in in vitro models was further tested in mice and in C. elegans. MCOPPB reduced the senescence cell burden in peripheral tissues but not in the central nervous system. Mice and worms exposed to MCOPPB also exhibited locomotion and lipid storage changes. Mechanistically, MCOPPB treatment activated transcriptional networks involved in the immune responses to external stressors, implicating Toll-like receptors (TLRs). Our study uncovers MCOPPB as a NOP ligand that, apart from anxiolytic effects, also shows tissue-specific senolytic effects.
Collapse
Affiliation(s)
- Marco Raffaele
- International Clinical Research Center, St. Anne's University Hospital, Brno, Czech Republic
| | - Kristina Kovacovicova
- International Clinical Research Center, St. Anne's University Hospital, Brno, Czech Republic
- Psychogenics Inc, Tarrytown, NY, USA
| | - Tommaso Biagini
- Laboratory of Bioinformatics, Fondazione IRCCS Casa Sollievo Della Sofferenza, San Giovanni Rotondo, Italy
| | - Oriana Lo Re
- International Clinical Research Center, St. Anne's University Hospital, Brno, Czech Republic
- Department of Translational Stem Cell Biology, Research Institute of the Medical University of Varna, Varna, Bulgaria
| | - Jan Frohlich
- International Clinical Research Center, St. Anne's University Hospital, Brno, Czech Republic
| | - Sebastiano Giallongo
- International Clinical Research Center, St. Anne's University Hospital, Brno, Czech Republic
- Department of Biology, Faculty of Medicine, Masaryk University, Brno, Czech Republic
| | - James D Nhan
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA, USA
- Department of Molecular and Computational Biology, Arts, and Sciences, Dornsife College of Letters, University of Southern California, Los Angeles, CA, USA
| | - Antonino Giulio Giannone
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties, Pathologic Anatomy Unit-University of Palermo, Palermo, Italy
| | - Daniela Cabibi
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties, Pathologic Anatomy Unit-University of Palermo, Palermo, Italy
| | - Martin Ivanov
- Department of Anatomy and Cell Biology, Research Institute of the Medical University of Varna, Varna, Bulgaria
| | - Anton B Tonchev
- Department of Translational Stem Cell Biology, Research Institute of the Medical University of Varna, Varna, Bulgaria
- Department of Anatomy and Cell Biology, Research Institute of the Medical University of Varna, Varna, Bulgaria
| | - Martin Mistrik
- Institute of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacky University, Olomouc, Czech Republic
| | - Matthew Lacey
- Institute of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacky University, Olomouc, Czech Republic
| | - Petr Dzubak
- Institute of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacky University, Olomouc, Czech Republic
| | - Sona Gurska
- Institute of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacky University, Olomouc, Czech Republic
| | - Marian Hajduch
- Institute of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacky University, Olomouc, Czech Republic
| | - Jiri Bartek
- Institute of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacky University, Olomouc, Czech Republic
- Genome Integrity Unit, Danish Cancer Society Research Center, Copenhagen, Denmark
- Division of Genome Biology, Department of Medical Biochemistry and Biophysics, Science for Life Laboratory, Karolinska Institute, Stockholm, Sweden
| | - Tommaso Mazza
- Laboratory of Bioinformatics, Fondazione IRCCS Casa Sollievo Della Sofferenza, San Giovanni Rotondo, Italy
| | - Vincenzo Micale
- Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy
| | - Sean P Curran
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA, USA
- Department of Molecular and Computational Biology, Arts, and Sciences, Dornsife College of Letters, University of Southern California, Los Angeles, CA, USA
- Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Manlio Vinciguerra
- International Clinical Research Center, St. Anne's University Hospital, Brno, Czech Republic.
- Department of Translational Stem Cell Biology, Research Institute of the Medical University of Varna, Varna, Bulgaria.
| |
Collapse
|
214
|
Liebert A, Bicknell B, Laakso EL, Jalilitabaei P, Tilley S, Kiat H, Mitrofanis J. Remote Photobiomodulation Treatment for the Clinical Signs of Parkinson's Disease: A Case Series Conducted During COVID-19. Photobiomodul Photomed Laser Surg 2022; 40:112-122. [PMID: 34919459 DOI: 10.1089/photob.2021.0056] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Objective: To assess whether remote application of photobiomodulation (PBM) is effective in reducing clinical signs of Parkinson's disease (PD). Background: PD is a progressive neurodegenerative disease for which there is no cure and few treatment options. There is a strong link between the microbiome-gut-brain axis and PD. PBM in animal models can reduce the signs of PD and protect the neurons from damage when applied directly to the head or to remote parts of the body. In a clinical study, PBM has been shown to improve clinical signs of PD for up to 1 year. Methods: Seven participants were treated with PBM to the abdomen and neck three times per week for 12 weeks. Participants were assessed for mobility, balance, cognition, fine motor skill, and sense of smell on enrolment, after 12 weeks of treatment in a clinic and after 33 weeks of home treatment. Results: A number of clinical signs of PD were shown to be improved by remote PBM treatment, including mobility, cognition, dynamic balance, spiral test, and sense of smell. Improvements were individual to the participant. Some improvements were lost for certain participants during at-home treatment, which coincided with a number of enforced coronavirus disease 2019 (COVID-19) pandemic lockdown periods. Conclusions: Remote application of PBM was shown to be an effective treatment for a number of clinical signs of PD, with some being maintained for 45 weeks, despite lockdown restrictions. Improvements in clinical signs were similar to those seen with the application of remote plus transcranial PBM treatment in a previous study. Clinical Trial Registration number: U1111-1205-2035.
Collapse
Affiliation(s)
- Ann Liebert
- Faculty of Medicine and Health Sciences, Sydney University, Camperdown, Australia.,Office of Research and Governance, Adventist Hospital, Wahroonga, Australia
| | - Brian Bicknell
- Faculty of Health Sciences, Australian Catholic University, North Sydney, Australia
| | - E-Liisa Laakso
- Mater Research Institute, University of Queensland, South Brisbane, Australia.,Menzies Health Institute, Griffith University, Gold Coast, Australia
| | | | | | - Hosen Kiat
- Cardiac Health Institute, Epping, Australia.,Department of Clinical Medicine, Macquarie University, Macquarie Park, Australia
| | - John Mitrofanis
- Faculty of Medicine and Health Sciences, Sydney University, Camperdown, Australia
| |
Collapse
|
215
|
Dookun E, Passos JF, Arthur HM, Richardson GD. Therapeutic Potential of Senolytics in Cardiovascular Disease. Cardiovasc Drugs Ther 2022; 36:187-196. [PMID: 32979174 PMCID: PMC8770386 DOI: 10.1007/s10557-020-07075-w] [Citation(s) in RCA: 43] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 09/03/2020] [Indexed: 02/07/2023]
Abstract
Ageing is the biggest risk factor for impaired cardiovascular health, with cardiovascular disease being the leading cause of death in 40% of individuals over 65 years old. Ageing is associated with both an increased prevalence of cardiovascular disease including heart failure, coronary artery disease, and myocardial infarction. Furthermore, ageing is associated with a poorer prognosis to these diseases. Genetic models allowing the elimination of senescent cells revealed that an accumulation of senescence contributes to the pathophysiology of cardiovascular ageing and promotes the progression of cardiovascular disease through the expression of a proinflammatory and profibrotic senescence-associated secretory phenotype. These studies have resulted in an effort to identify pharmacological therapeutics that enable the specific elimination of senescent cells through apoptosis induction. These senescent cell apoptosis-inducing compounds are termed senolytics and their potential to ameliorate age-associated cardiovascular disease is the focus of this review.
Collapse
Affiliation(s)
- Emily Dookun
- Biosciences Institute, Newcastle University, Newcastle upon Tyne, NE1 3BZ, UK
| | - João F Passos
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, USA
| | - Helen M Arthur
- Biosciences Institute, Newcastle University, Newcastle upon Tyne, NE1 3BZ, UK
| | - Gavin D Richardson
- Biosciences Institute, Newcastle University, Newcastle upon Tyne, NE1 3BZ, UK.
| |
Collapse
|
216
|
Abstract
ABSTRACT Metabolic changes represent the most common sign of aging and lead to increased risk of developing diseases typical of old age. Age-associated metabolic changes, such as decreased insulin sensitivity, decreased mitochondrial function, and dysregulated nutrient uptake, fuel the low-grade chronic systemic inflammation, known as inflammaging, a leading cause of morbidity and mortality, linked to the development of several diseases of old age. How aging affects the metabolic phenotype of immune cells, and B cells in particular, is not well known and is under intensive investigation by several groups. In this study, we summarized the few published results linking intrinsic B-cell metabolism and B-cell function in different groups of young and elderly individuals: healthy, with type-2 diabetes mellitus, or with HIV infection. Although preliminary, these results suggest the intriguing possibility that metabolic pathways can represent potential novel therapeutic targets to reduce inflammaging and improve humoral immunity.
Collapse
Affiliation(s)
- Daniela Frasca
- Department of Microbiology and Immunology, University of Miami Miller School of Medicine, Miami, FL; and
- Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL
| | - Suresh Pallikkuth
- Department of Microbiology and Immunology, University of Miami Miller School of Medicine, Miami, FL; and
| | - Savita Pahwa
- Department of Microbiology and Immunology, University of Miami Miller School of Medicine, Miami, FL; and
| |
Collapse
|
217
|
Lake JE, Overton T, Naggie S, Sulkowski M, Loomba R, Kleiner DE, Price JC, Chew KW, Chung RT, Corey KE. Expert Panel Review on Nonalcoholic Fatty Liver Disease in Persons With Human Immunodeficiency Virus. Clin Gastroenterol Hepatol 2022; 20:256-268. [PMID: 33069882 PMCID: PMC9069630 DOI: 10.1016/j.cgh.2020.10.018] [Citation(s) in RCA: 46] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/07/2020] [Revised: 10/07/2020] [Accepted: 10/12/2020] [Indexed: 02/07/2023]
Abstract
Nonalcoholic fatty liver disease (NAFLD) affects 25% of adults in the general population and is a disease spectrum ranging from steatosis to nonalcoholic steatohepatitis (NASH) to end-stage liver disease. NAFLD is an independent risk factor for cardiovascular disease, diabetes mellitus, and all-cause mortality, and NASH cirrhosis is a frequent indication for liver transplantation. In persons with human immunodeficiency virus (PWH), chronic liver disease is the second leading cause of non-human immunodeficiency virus-related mortality. Between 20% and 63% of PWH have NASH, and 14% to 63% have NASH with fibrosis. However, little is known about the optimal diagnostic strategies, risk factors for, and treatment of NAFLD in PWH. Here, we review current data on and identify knowledge gaps in the epidemiology, pathophysiology, diagnosis, and management of NAFLD in PWH and highlight priorities for research.
Collapse
Affiliation(s)
- Jordan E Lake
- Division of Infectious Disease, University of Texas Health Sciences Center at Houston, Houston, Texas.
| | - Turner Overton
- Division of Infectious Disease, University of Alabama School of Medicine, Birmingham, Alabama
| | - Susanna Naggie
- Division of Infections Disease, Duke University School of Medicine, Durham, North Carolina
| | - Mark Sulkowski
- Division of Infectious Disease, Johns Hopkins School of Medicine, Baltimore, Maryland
| | - Rohit Loomba
- Division of Gastroenterology, University of California San Diego School of Medicine, San Diego, California
| | - David E Kleiner
- Center for Cancer Research, National Cancer Institute, Bethesda, Maryland
| | - Jennifer C Price
- Division of Gastroenterology, University of California San Francisco School of Medicine, San Francisco, California
| | - Kara W Chew
- Division of Infectious Diseases, University of California Los Angeles School of Medicine, Los Angeles, California
| | - Raymond T Chung
- Division of Gastroenterology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Kathleen E Corey
- Division of Gastroenterology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
218
|
Conventional type 1 dendritic cells protect against age-related adipose tissue dysfunction and obesity. Cell Mol Immunol 2022; 19:260-275. [PMID: 34983945 PMCID: PMC8803960 DOI: 10.1038/s41423-021-00812-7] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Revised: 11/19/2021] [Accepted: 11/22/2021] [Indexed: 02/08/2023] Open
Abstract
Conventional dendritic cells (cDCs) scan and integrate environmental cues in almost every tissue, including exogenous metabolic signals. While cDCs are critical in maintaining immune balance, their role in preserving energy homeostasis is unclear. Here, we showed that Batf3-deficient mice lacking conventional type 1 DCs (cDC1s) had increased body weight and adiposity during aging. This led to impaired energy expenditure and glucose tolerance, insulin resistance, dyslipidemia, and liver steatosis. cDC1 deficiency caused adipose tissue inflammation that was preceded by a paucity of NK1.1+ invariant NKT (iNKT) cells. Accordingly, among antigen-presenting cells, cDC1s exhibited notable induction of IFN-γ production by iNKT cells, which plays a metabolically protective role in lean adipose tissue. Flt3L treatment, which expands the dendritic cell (DC) compartment, mitigated diet-induced obesity and hyperlipidemia in a Batf3-dependent manner. This effect was partially mediated by NK1.1+ cells. These results reveal a new critical role for the cDC1-iNKT cell axis in the regulation of adipose tissue homeostasis.
Collapse
|
219
|
Minteer C, Morselli M, Meer M, Cao J, Higgins‐Chen A, Lang SM, Pellegrini M, Yan Q, Levine M. Tick tock, tick tock: Mouse culture and tissue aging captured by an epigenetic clock. Aging Cell 2022; 21:e13553. [PMID: 35104377 PMCID: PMC8844113 DOI: 10.1111/acel.13553] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2021] [Revised: 12/11/2021] [Accepted: 01/05/2022] [Indexed: 12/11/2022] Open
Abstract
Aging is associated with dramatic changes to DNA methylation (DNAm), although the causes and consequences of such alterations are unknown. Our ability to experimentally uncover mechanisms of epigenetic aging will be greatly enhanced by our ability to study and manipulate these changes using in vitro models. However, it remains unclear whether the changes elicited by cells in culture can serve as a model of what is observed in aging tissues in vivo. To test this, we serially passaged mouse embryonic fibroblasts (MEFs) and assessed changes in DNAm at each time point via reduced representation bisulfite sequencing. By developing a measure that tracked cellular aging in vitro, we tested whether it tracked physiological aging in various mouse tissues and whether anti-aging interventions modulate this measure. Our measure, termed CultureAGE, was shown to strongly increase with age when examined in multiple tissues (liver, lung, kidney, blood, and adipose). As a control, we confirmed that the measure was not a marker of cellular senescence, suggesting that it reflects a distinct yet progressive cellular aging phenomena that can be induced in vitro. Furthermore, we demonstrated slower epigenetic aging in animals undergoing caloric restriction and a resetting of our measure in lung and kidney fibroblasts when re-programmed to iPSCs. Enrichment and clustering analysis implicated EED and Polycomb group (PcG) factors as potentially important chromatin regulators in translational culture aging phenotypes. Overall, this study supports the concept that physiologically relevant aging changes can be induced in vitro and used to uncover mechanistic insights into epigenetic aging.
Collapse
Affiliation(s)
| | - Marco Morselli
- Department of Molecular, Cell, and Developmental BiologyUniversity of California Los AngelesLos AngelesCaliforniaUSA
| | - Margarita Meer
- Department of PathologyYale School of MedicineNew HavenConnecticutUSA
| | - Jian Cao
- Department of PathologyYale School of MedicineNew HavenConnecticutUSA
- Rutgers Cancer Institute of New JerseyNew BrunswickNew JerseyUSA
| | | | - Sabine M. Lang
- Department of PathologyYale School of MedicineNew HavenConnecticutUSA
| | - Matteo Pellegrini
- Department of Molecular, Cell, and Developmental BiologyUniversity of California Los AngelesLos AngelesCaliforniaUSA
| | - Qin Yan
- Department of PathologyYale School of MedicineNew HavenConnecticutUSA
| | - Morgan E. Levine
- Department of PathologyYale School of MedicineNew HavenConnecticutUSA
| |
Collapse
|
220
|
Bale LK, Schafer MJ, Atkinson EJ, Le Brasseur NK, Haak AJ, Oxvig C, Conover CA. Pregnancy‐associated plasma protein‐A (PAPP‐A) is a key component of an interactive cellular mechanism promoting pulmonary fibrosis. J Cell Physiol 2022; 237:2220-2229. [PMID: 35098542 PMCID: PMC9050837 DOI: 10.1002/jcp.30687] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2021] [Revised: 12/21/2021] [Accepted: 01/10/2022] [Indexed: 12/25/2022]
Abstract
Idiopathic pulmonary fibrosis (IPF) is a progressive lung disease with few effective treatment options. We found a highly significant correlation between pregnancy-associated plasma protein (PAPP)-A expression in IPF lung tissue and disease severity as measured by various pulmonary and physical function tests. PAPP-A is a metalloproteinase that enhances local insulin-like growth factor (IGF) activity. We used primary cultures of normal adult human lung fibroblasts (NHLF) to test the hypothesis that PAPP-A plays an important role in the development of pulmonary fibrosis. Treatment of NHLF with pro-fibrotic transforming growth factor (TGF)-β stimulated marked increases in IGF-I mRNA expression (>20-fold) and measurable IGF-I levels in 72-h conditioned medium (CM). TGF-β treatment also increased PAPP-A levels in CM fourfold (p = 0.004) and proteolytic activity ~2-fold. There was an indirect effect of TGF-β to stimulate signaling through the PI3K/Akt pathway, which was significantly inhibited by both IGF-I-inactivating and PAPP-A inhibitory antibodies. Induction of senescence in NHLF increased PAPP-A levels in CM 10-fold (p = 0.006) with attendant increased proteolytic activity. Thus, PAPP-A is a novel component of the senescent lung fibroblast secretome. In addition, NHLF secreted extracellular vehicles (EVs) with surface-bound active PAPP-A that were increased fivefold with senescence. Regulation of PAPP-A and IGF signaling by TGF-β and cell senescence suggests an interactive cellular mechanism underlying the resistance to apoptosis and the progression of fibrosis in IPF. Furthermore, PAPP-A-associated EVs may be a means of pro-fibrotic, pro-senescent communication with other cells in the lung and, thus, a potential therapeutic target for IPF.
Collapse
Affiliation(s)
- Laurie K. Bale
- Division of Endocrinology Metabolism and Nutrition, Endocrine Research Unit Rochester Minnesota USA
| | - Marissa J. Schafer
- Department of Physiology and Biomedical Engineering Rochester Minnesota USA
| | | | | | - Andrew J. Haak
- Department of Physiology and Biomedical Engineering Rochester Minnesota USA
| | - Claus Oxvig
- Department of Molecular Biology and Genetics University of Aarhus Aarhus Denmark
| | - Cheryl A. Conover
- Division of Endocrinology Metabolism and Nutrition, Endocrine Research Unit Rochester Minnesota USA
| |
Collapse
|
221
|
Cai Z, Liu D, Yang Y, Xie W, He M, Yu D, Wu Y, Wang X, Xiao W, Li Y. The role and therapeutic potential of stem cells in skeletal muscle in sarcopenia. Stem Cell Res Ther 2022; 13:28. [PMID: 35073997 PMCID: PMC8785537 DOI: 10.1186/s13287-022-02706-5] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2021] [Accepted: 01/05/2022] [Indexed: 01/23/2023] Open
Abstract
Sarcopenia is a common age-related skeletal muscle disorder featuring the loss of muscle mass and function. In regard to tissue repair in the human body, scientists always consider the use of stem cells. In skeletal muscle, satellite cells (SCs) are adult stem cells that maintain tissue homeostasis and repair damaged regions after injury to preserve skeletal muscle integrity. Muscle-derived stem cells (MDSCs) and SCs are the two most commonly studied stem cell populations from skeletal muscle. To date, considerable progress has been achieved in understanding the complex associations between stem cells in muscle and the occurrence and treatment of sarcopenia. In this review, we first give brief introductions to sarcopenia, SCs and MDSCs. Then, we attempt to untangle the differences and connections between these two types of stem cells and further elaborate on the interactions between sarcopenia and stem cells. Finally, our perspectives on the possible application of stem cells for the treatment of sarcopenia in future are presented. Several studies emerging in recent years have shown that changes in the number and function of stem cells can trigger sarcopenia, which in turn leads to adverse influences on stem cells because of the altered internal environment in muscle. A better understanding of the role of stem cells in muscle, especially SCs and MDSCs, in sarcopenia will facilitate the realization of novel therapy approaches based on stem cells to combat sarcopenia.
Collapse
Affiliation(s)
- Zijun Cai
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China.,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
| | - Di Liu
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China.,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
| | - Yuntao Yang
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China.,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
| | - Wenqing Xie
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China.,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
| | - Miao He
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China.,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
| | - Dengjie Yu
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China.,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
| | - Yuxiang Wu
- School of Kinesiology, Jianghan University, Wuhan, 430056, China
| | - Xiuhua Wang
- Xiang Ya Nursing School, Central South University, Changsha, 410008, Hunan, China
| | - Wenfeng Xiao
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China. .,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China.
| | - Yusheng Li
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China. .,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China.
| |
Collapse
|
222
|
Analysis of Clinical Characteristics, Radiological Predictors, Pathological Features, and Perioperative Outcomes Associated with Perinephric Fat Adhesion Degree. JOURNAL OF ONCOLOGY 2022; 2021:9095469. [PMID: 34987581 PMCID: PMC8723850 DOI: 10.1155/2021/9095469] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Accepted: 12/01/2021] [Indexed: 11/25/2022]
Abstract
Background To assess the clinical characteristics, radiological predictors, and pathological features of perinephric fat adhesion degree (PFAD) graded based on fixed criteria and to determine the impact of adherent perinephric fat (APF) on retroperitoneal laparoscopic partial nephrectomy (RLPN) outcomes. Methods 84 patients undergoing RLPN were included and graded into 4 groups based on PFAD. Univariate and multivariate analyses were performed for clinical characteristics and radiological predictors of PFAD. Perioperative data were compared between APF groups and non-APF groups. Masson staining determined collagen fibers. Immunohistochemistry detected CD45 immune cells and CD34 vessels. Results 20, 28, 18, and 18 patients were graded as normal perinephric fat (NPF), mild adherent perinephric fat (MiPF), moderate adherent perinephric fat (MoPF), and severe adherent perinephric fat (SPF), respectively. Multivariate analysis revealed that gender (p < 0.001), age (p = 0.003), and hypertension (p = 0.006) were significant clinical risk factors of PFAD, while radiological predictors included perinephric stranding (p = 0.001), posterior perinephric fat thickness (p = 0.009), and perinephric fat density (p = 0.02). APF was associated with drain output (p = 0.012) and accompanied by immune cells gathering in renal cortex near thickened renal capsule with many vessels. Conclusions Clinical characteristics and radiological predictors can evaluate PFAD and may assist to guide preoperative surgical option. Pathological features of APF reflect decapsulation and bleeding during kidney mobilization at RLPN.
Collapse
|
223
|
Wang L, Wang B, Gasek NS, Zhou Y, Cohn RL, Martin DE, Zuo W, Flynn WF, Guo C, Jellison ER, Kim T, Prata LGPL, Palmer AK, Li M, Inman CL, Barber LS, Al-Naggar IMA, Zhou Y, Du W, Kshitiz, Kuchel GA, Meves A, Tchkonia T, Kirkland JL, Robson P, Xu M. Targeting p21 Cip1 highly expressing cells in adipose tissue alleviates insulin resistance in obesity. Cell Metab 2022; 34:75-89.e8. [PMID: 34813734 PMCID: PMC8732323 DOI: 10.1016/j.cmet.2021.11.002] [Citation(s) in RCA: 94] [Impact Index Per Article: 31.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Revised: 09/30/2021] [Accepted: 11/03/2021] [Indexed: 01/07/2023]
Abstract
Insulin resistance is a pathological state often associated with obesity, representing a major risk factor for type 2 diabetes. Limited mechanism-based strategies exist to alleviate insulin resistance. Here, using single-cell transcriptomics, we identify a small, critically important, but previously unexamined cell population, p21Cip1 highly expressing (p21high) cells, which accumulate in adipose tissue with obesity. By leveraging a p21-Cre mouse model, we demonstrate that intermittent clearance of p21high cells can both prevent and alleviate insulin resistance in obese mice. Exclusive inactivation of the NF-κB pathway within p21high cells, without killing them, attenuates insulin resistance. Moreover, fat transplantation experiments establish that p21high cells within fat are sufficient to cause insulin resistance in vivo. Importantly, a senolytic cocktail, dasatinib plus quercetin, eliminates p21high cells in human fat ex vivo and mitigates insulin resistance following xenotransplantation into immuno-deficient mice. Our findings lay the foundation for pursuing the targeting of p21high cells as a new therapy to alleviate insulin resistance.
Collapse
Affiliation(s)
- Lichao Wang
- UConn Center on Aging, UConn Health, Farmington, CT 06030, USA; Department of Genetics and Genome Sciences, UConn Health, Farmington, CT 06030, USA
| | - Binsheng Wang
- UConn Center on Aging, UConn Health, Farmington, CT 06030, USA; Department of Genetics and Genome Sciences, UConn Health, Farmington, CT 06030, USA
| | - Nathan S Gasek
- UConn Center on Aging, UConn Health, Farmington, CT 06030, USA; Department of Genetics and Genome Sciences, UConn Health, Farmington, CT 06030, USA
| | - Yueying Zhou
- Xiangya Stomatological Hospital, Central South University, Changsha, 86-410000, China; Center for Regenerative Medicine and Skeletal Development, UConn Health, Farmington, CT 06030, USA
| | - Rachel L Cohn
- UConn Center on Aging, UConn Health, Farmington, CT 06030, USA; Department of Genetics and Genome Sciences, UConn Health, Farmington, CT 06030, USA
| | - Dominique E Martin
- UConn Center on Aging, UConn Health, Farmington, CT 06030, USA; Biomedical Science Graduate Program, UConn Health, Farmington, CT 06030, USA
| | - Wulin Zuo
- The Jackson Laboratory for Genomic Medicine, Farmington, CT 06030, USA
| | - William F Flynn
- The Jackson Laboratory for Genomic Medicine, Farmington, CT 06030, USA
| | - Chun Guo
- UConn Center on Aging, UConn Health, Farmington, CT 06030, USA
| | - Evan R Jellison
- Department of Immunology, UConn Health, Farmington, CT 06030, USA
| | - Taewan Kim
- UConn Center on Aging, UConn Health, Farmington, CT 06030, USA; Biomedical Science Graduate Program, UConn Health, Farmington, CT 06030, USA
| | | | - Allyson K Palmer
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, MN 55905, USA
| | - Ming Li
- Department of Dermatology, Mayo Clinic, Rochester, MN 55905, USA
| | - Christina L Inman
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, MN 55905, USA
| | - Lauren S Barber
- UConn Center on Aging, UConn Health, Farmington, CT 06030, USA
| | | | - Yanjiao Zhou
- Department of Medicine, UConn Health, Farmington, CT 06030, USA
| | - Wenqiang Du
- Department of Biomedical Engineering, UConn Health, Farmington, CT 06030, USA
| | - Kshitiz
- Department of Biomedical Engineering, UConn Health, Farmington, CT 06030, USA
| | - George A Kuchel
- UConn Center on Aging, UConn Health, Farmington, CT 06030, USA
| | - Alexander Meves
- Department of Dermatology, Mayo Clinic, Rochester, MN 55905, USA
| | - Tamar Tchkonia
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, MN 55905, USA
| | - James L Kirkland
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, MN 55905, USA
| | - Paul Robson
- Department of Genetics and Genome Sciences, UConn Health, Farmington, CT 06030, USA; The Jackson Laboratory for Genomic Medicine, Farmington, CT 06030, USA
| | - Ming Xu
- UConn Center on Aging, UConn Health, Farmington, CT 06030, USA; Department of Genetics and Genome Sciences, UConn Health, Farmington, CT 06030, USA.
| |
Collapse
|
224
|
Naphatthalung J, Chairuk P, Yorsin S, Kanokwiroon K, Radenahmad N, Jansakul C. Decreased body-fat accumulation and increased vasorelaxation to glyceryl trinitrate in middle-aged male rats following six-weeks consumption of coconut milk protein. BRAZ J PHARM SCI 2022. [DOI: 10.1590/s2175-97902022e20510] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
|
225
|
Yamada T. Intramuscular adipogenesis in cattle: Effects of body fat distribution and macrophage infiltration. Anim Sci J 2022; 93:e13785. [PMID: 36443236 DOI: 10.1111/asj.13785] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Revised: 10/18/2022] [Accepted: 11/01/2022] [Indexed: 11/30/2022]
Abstract
Ectopic fat is defined by the deposition of adipose tissue within non-adipose tissue such as skeletal muscle. Japanese Black cattle (Wagyu) are characterized by the ability to accumulate high amounts of intramuscular adipose tissue. Obese conditions enhance the accumulation of ectopic fat. This review shows the effects of subcutaneous and visceral fat distribution on Wagyu intramuscular adipogenesis. Obese conditions also stimulate the macrophage infiltration into adipose tissues. Adipose tissue macrophages have reported to regulate adipose tissue growth and ectopic fat accumulation in humans and rodents. Wagyu is characterized by the higher capacity for intramuscular adipogenesis than Holsteins. This review discusses the depot-specific effects of macrophage infiltration among subcutaneous, visceral, and intramuscular adipose tissue on intramuscular adipogenesis in Wagyu and Holstein cattle. Recently, metabolome analysis has been used to identify obesity-related biomarkers by comparing the biological samples between lean and obese patients. This review introduces the metabolomic profiles of plasma and intramuscular adipose tissue between Wagyu and Holsteins.
Collapse
Affiliation(s)
- Tomoya Yamada
- National Agriculture and Food Research Organization Oda Shimane Japan
| |
Collapse
|
226
|
Dattilo A, Ceccarini G, Scabia G, Magno S, Quintino L, Pelosini C, Salvetti G, Cusano R, Massidda M, Montanelli L, Gilio D, Gatti G, Giacomina A, Costa M, Santini F, Maffei M. Circulating Levels of MiRNAs From 320 Family in Subjects With Lipodystrophy: Disclosing Novel Signatures of the Disease. Front Endocrinol (Lausanne) 2022; 13:866679. [PMID: 35733784 PMCID: PMC9207177 DOI: 10.3389/fendo.2022.866679] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Accepted: 05/05/2022] [Indexed: 11/13/2022] Open
Abstract
Lipodystrophy (LD) indicates a group of rare disorders, with generalized or partial loss of white adipose tissue (WAT) often associated with metabolic derangements. Heterogeneity/wide spectrum of the disease and lack of biomarkers make diagnosis often difficult. MicroRNAs are important to maintain a correct WAT function and WAT is a source of circulating miRNAs (cmiRs). miRNAs from 320 family were previously detected in the WAT and variably associated to the metabolic syndrome. Our aim was then to investigate if LD can result in altered abundance of cmiRs-320. We collected samples from a cohort of LD subjects of various subtypes and from age matched controls. Use of quantitative PCR determined that cmiRs- 320a-3p, 320b, 320c, 320e are upregulated, while 320d is downregulated in LD. CmiRs-320 power as classifiers was more powerful in the most extreme and defined forms of LD, including the generalized and the Dunnigan subtypes. cmiR-320a-3p showed significant inverse relationships with plasma leptin (P < 0.0001), typically low in LD. The hepatic enzymes gamma-glutamyl transferase (GGT), aspartate aminotransferase (AST), alanine aminotransferase (ALT) and the marker of inflammation C-reactive protein (CRP) were inversely related to cmiR 320d (P < 0.05, for CRP and GGT; P < 0.01, for AST and ALT). Gene ontology analysis revealed cell-cell adhesion as a process regulated by 320 miRNAs targets, thus disclosing a novel route to investigate origin of WAT loss/dysfunction. In conclusion, cmiRs-320 constitute novel biomarkers of LD, abundance of miR320a-3p is inversely associated to indicators related to WAT function, while downregulation of cmiR-320d predicts an altered hepatic profile and higher inflammation.
Collapse
Affiliation(s)
- Alessia Dattilo
- Institute of Life Sciences, Scuola Superiore Sant’Anna, Pisa, Italy
- Obesity and Lipodystrophy Center, Endocrinology Unit, Pisa University Hospital, Pisa, Italy
| | - Giovanni Ceccarini
- Obesity and Lipodystrophy Center, Endocrinology Unit, Pisa University Hospital, Pisa, Italy
| | - Gaia Scabia
- Obesity and Lipodystrophy Center, Endocrinology Unit, Pisa University Hospital, Pisa, Italy
- National Research Council, Institute of Clinical Physiology, Pisa, Italy
| | - Silvia Magno
- Obesity and Lipodystrophy Center, Endocrinology Unit, Pisa University Hospital, Pisa, Italy
| | - Lara Quintino
- Obesity and Lipodystrophy Center, Endocrinology Unit, Pisa University Hospital, Pisa, Italy
| | - Caterina Pelosini
- Obesity and Lipodystrophy Center, Endocrinology Unit, Pisa University Hospital, Pisa, Italy
| | - Guido Salvetti
- Obesity and Lipodystrophy Center, Endocrinology Unit, Pisa University Hospital, Pisa, Italy
| | - Roberto Cusano
- Center for Advanced Studies, Research and Development in Sardinia, Pula (CA), Italy
| | - Matteo Massidda
- Center for Advanced Studies, Research and Development in Sardinia, Pula (CA), Italy
| | - Lucia Montanelli
- Department of Clinical and Experimental Medicine, Endocrinology Unit, Pisa University Hospital, Pisa, Italy
| | - Donatella Gilio
- Obesity and Lipodystrophy Center, Endocrinology Unit, Pisa University Hospital, Pisa, Italy
| | - Gianluca Gatti
- Plastic and Reconstructive Surgery Unit, Hospital of Pisa, Pisa, Italy
| | | | - Mario Costa
- National Research Council, Institute of Neuroscience, Pisa, Italy
- Centro Pisano Flash Radiotherapy, Center for Instrument Sharing of the University of Pisa (CPFR@CISUP), Pisa University Hospital, Pisa, Italy
| | - Ferruccio Santini
- Obesity and Lipodystrophy Center, Endocrinology Unit, Pisa University Hospital, Pisa, Italy
| | - Margherita Maffei
- Obesity and Lipodystrophy Center, Endocrinology Unit, Pisa University Hospital, Pisa, Italy
- National Research Council, Institute of Clinical Physiology, Pisa, Italy
- *Correspondence: Margherita Maffei,
| |
Collapse
|
227
|
Kemoun P, Ader I, Planat-Benard V, Dray C, Fazilleau N, Monsarrat P, Cousin B, Paupert J, Ousset M, Lorsignol A, Raymond-Letron I, Vellas B, Valet P, Kirkwood T, Beard J, Pénicaud L, Casteilla L. A gerophysiology perspective on healthy ageing. Ageing Res Rev 2022; 73:101537. [PMID: 34883201 DOI: 10.1016/j.arr.2021.101537] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2021] [Revised: 11/23/2021] [Accepted: 12/03/2021] [Indexed: 12/16/2022]
Abstract
Improvements in public health and health care have resulted in significant increases in lifespan globally, but also in a significant increase in chronic disease prevalence. This has led to a focus on healthy ageing bringing a shift from a pathology-centered to an intrinsic capacity and function-centered view. In parallel, the emerging field of geroscience has promoted the exploration of the biomolecular drivers of ageing towards a transverse vision by proposing an integrated set of molecular hallmarks. In this review, we propose to take a step further in this direction, highlighting a gerophysiological perspective that considers the notion of homeostasis/allostasis relating to robustness/fragility respectively. While robustness is associated with homeostasis achieved by an optimal structure/function relationship in all organs, successive repair processes occurring after daily injuries and infections result in accumulation of scar healing leading to progressive tissue degeneration, allostasis and frailty. Considering biological ageing as the accumulation of scarring at the level of the whole organism emphasizes three transverse and shared elements in the body - mesenchymal stroma cells/immunity/metabolism (SIM). This SIM tryptich drives tissue and organ fate to regulate the age-related evolution of body functions. It provides the basis of a gerophysiology perspective, possibly representing a better way to decipher healthy ageing, not only by defining a composite biomarker(s) but also by developing new preventive/curative strategies.
Collapse
|
228
|
Campos JTADM, Oliveira MSD, Soares LP, Medeiros KAD, Campos LRDS, Lima JG. DNA repair-related genes and adipogenesis: Lessons from congenital lipodystrophies. Genet Mol Biol 2022; 45:e20220086. [DOI: 10.1590/1678-4685-gmb-2022-0086] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Accepted: 09/20/2022] [Indexed: 11/09/2022] Open
|
229
|
Rubio-Tomás T, Rueda-Robles A, Plaza-Díaz J, Álvarez-Mercado AI. Nutrition and cellular senescence in obesity-related disorders. J Nutr Biochem 2022; 99:108861. [PMID: 34517097 DOI: 10.1016/j.jnutbio.2021.108861] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2020] [Revised: 05/29/2021] [Accepted: 08/10/2021] [Indexed: 02/06/2023]
Abstract
Adequate nutrition is vital for immune homeostasis. However, the incidence of obesity is increasing worldwide due to the adoption of the Western diet and a sedentary lifestyle. Obesity is associated with chronic inflammation which alters the function of adipose tissue, liver, pancreas, and the nervous system. Inflammation is related to cellular senescence, distinguished by irreversible cell cycle arrest. Senescent cells secrete the senescence-associated secretory phenotype (SASP) which contains pro-inflammatory factors. Targeting processes in senescence might have a salutary approach to obesity. The present review highlights the impact of an unhealthy diet on tissues affected by obesity, and the mechanisms that promote the consequent inflammation and senescence.
Collapse
Affiliation(s)
- Teresa Rubio-Tomás
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain; School of Medicine, University of Crete, Herakleion, Crete, Greece
| | - Ascensión Rueda-Robles
- Institute of Nutrition and Food Technology "José Mataix", Center of Biomedical Research, University of Granada, Armilla, Granada, Spain
| | - Julio Plaza-Díaz
- Children's Hospital of Eastern Ontario Research Institute, Ottawa, ON Canada; Department of Biochemistry and Molecular Biology II, School of Pharmacy, University of Granada, Granada, Spain; Instituto de Investigación Biosanitaria ibs.GRANADA, Complejo Hospitalario Universitario de Granada, Granada Spain.
| | - Ana I Álvarez-Mercado
- Institute of Nutrition and Food Technology "José Mataix", Center of Biomedical Research, University of Granada, Armilla, Granada, Spain; Department of Biochemistry and Molecular Biology II, School of Pharmacy, University of Granada, Granada, Spain; Instituto de Investigación Biosanitaria ibs.GRANADA, Complejo Hospitalario Universitario de Granada, Granada Spain.
| |
Collapse
|
230
|
Bicknell B, Laakso EL, Liebert A, Kiat H. Modifying the Microbiome as a Potential Mechanism of Photobiomodulation: A Case Report. Photobiomodul Photomed Laser Surg 2021; 40:88-97. [PMID: 34962422 DOI: 10.1089/photob.2021.0057] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Objective: The objective of this case study was to elucidate the effect of photobiomodulation (PBM) on the microbiome. Background: The gut microbiome has been identified as a key component of health, with gut dysbiosis, characterized by decreased microbial diversity and an altered microbial composition, being recognized as instrumental in many diseases and disorders. Previous research has suggested that the gut microbiome can be favorably altered in animal models using PBM. Materials and methods: The participant had their microbiome tested on nine occasions, three times before any treatment, three times after radiotherapy and commencement of immunotherapy for breast cancer, and three times after PBM treatment. The PBM treatment consisted of infrared laser treatment (904 nm; 700 Hz pulse frequency, 861.3 total joules) to the abdomen three times per week for 11 weeks. Results: The microbiome of the participant showed significant changes in diversity after PBM treatment, but not after cancer therapy, with an increase in the number of known beneficial bacteria (Akkermansia, Faecalibacterium, and Roseburia) and decrease in the number of potentially pathogenic genera. Conclusions: The results suggested the possibility that PBM may alter the microbiome and thus it represents a therapeutic avenue for chronic diseases with otherwise limited treatment options.
Collapse
Affiliation(s)
- Brian Bicknell
- Faculty of Health Sciences, Australian Catholic University, North Sydney, Australia
| | - E-Liisa Laakso
- Mater Research Institute, University of Queensland, South Brisbane, Australia.,Menzies Health Institute, Queensland, Griffith University, Gold Coast, Australia
| | - Ann Liebert
- School of Medical Sciences, Sydney University, Camperdown, Australia.,Office of Research and Governance, Adventist Hospital, Wahroonga, Australia
| | - Hosen Kiat
- Cardiac Health Institute, Epping, Australia.,Department of Clinical Medicine, Macquarie University, Macquarie Park, Australia
| |
Collapse
|
231
|
Bridge-Comer PE, Vickers MH, Morton-Jones J, Spada A, Rong J, Reynolds CM. Impact of Maternal Intake of Artificial Sweetener, Acesulfame-K, on Metabolic and Reproductive Health Outcomes in Male and Female Mouse Offspring. Front Nutr 2021; 8:745203. [PMID: 34938757 PMCID: PMC8687087 DOI: 10.3389/fnut.2021.745203] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Accepted: 10/14/2021] [Indexed: 11/13/2022] Open
Abstract
Guidelines advising pregnant women to avoid food and beverages with high fat and sugar have led to an increase in the consumption of "diet" options sweetened by artificial sweeteners (AS). Yet, there is limited information regarding the impact of AS intake during pregnancy on the long-term risk of cardiometabolic and reproductive complications in adult offspring. This study examined the influence of maternal acesulfame-K (Ace-K) and fructose consumption on metabolic and reproductive outcomes in offspring. Pregnant C57BL/6 mice received standard chow ad-libitum with either water (CD), fructose (Fr; 20% kcal intake), or AS (AS; 12.5 mM Ace-K) throughout pregnancy and lactation (n = 8/group). Postweaning offspring were maintained on a CD diet for the remainder of the experiment. Body weight, food intake, and water intake were measured weekly. Oral glucose tolerance tests (OGTT) were undertaken at 12 weeks, and the offspring were culled at week 14. Female, but not male, AS groups exhibited decreased glucose tolerance compared to Fr. There was an increase in gonadal fat adipocyte size in male offspring from AS and Fr groups compared to CD groups. In female offspring, adipocyte size was increased in the Fr group compared to the CD group. In female, but not male offspring, there was a trend toward increase in Fasn gene expression in AS group compared to the CD group. Maternal AS and Fr also negatively impacted upon female offspring estrus cycles and induced alterations to markers associated with ovulation. In summary, exposure to Ace-k via the maternal diet leads to impaired glucose tolerance and impacts adipocyte size in a sex-specific manner as well as significantly affecting estrus cycles and related gene markers in female offspring. This has implications in terms of providing tailored dietary advice for pregnant women and highlights the potential negative influence of artificial sweetener intake in the context of intergenerational impacts.
Collapse
Affiliation(s)
| | - Mark H Vickers
- Liggins Institute, University of Auckland, Auckland, New Zealand
| | | | - Ana Spada
- Liggins Institute, University of Auckland, Auckland, New Zealand
| | - Jing Rong
- Liggins Institute, University of Auckland, Auckland, New Zealand
| | - Clare M Reynolds
- Liggins Institute, University of Auckland, Auckland, New Zealand.,School of Public Health, Physiotherapy and Sports Science/Conway Institute/Institute of Food and Health/Diabetes Complications Research Centre, University College Dublin, Belfield, Ireland
| |
Collapse
|
232
|
Gudiksen A, Qoqaj A, Ringholm S, Wojtaszewski J, Plomgaard P, Pilegaard H. Ameliorating effects of lifelong physical activity on healthy aging and mitochondrial function in human white adipose tissue. J Gerontol A Biol Sci Med Sci 2021; 77:1101-1111. [PMID: 34875059 DOI: 10.1093/gerona/glab356] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Indexed: 11/12/2022] Open
Abstract
Growing old is patently among the most prominent risk factors for lifestyle related diseases and deterioration in physical performance. Aging in particular affects mitochondrial homeostasis and maintaining a well-functioning mitochondrial pool is imperative in order to avoid age-associated metabolic decline. White adipose tissue (WAT) is a key organ in energy balance and impaired mitochondrial function in adipocytes has been associated with increased low-grade inflammation, altered metabolism, excessive ROS production and an accelerated aging phenotype. Exercise training improves mitochondrial health but whether lifelong exercise training can sufficiently maintain WAT mitochondrial function is currently unknown. Therefore, to dissect the role and dose-dependence of lifelong exercise training on aging WAT metabolic parameters and mitochondrial function, young and older untrained, as well as moderately and highly exercise trained older male subjects were recruited and abdominal subcutaneous (s)WAT biopsies and venous blood samples were obtained to measure mitochondrial function and key metabolic factors in WAT and plasma. Mitochondrial intrinsic respiratory capacity was lower in sWAT from older than in young subjects. In spite of this, maximal mitochondrial respiration per wet weight, markers of oxidative capacity, and mitophagic capacity were increased in sWAT from lifelong highly exercise trained than all other groups. Furthermore, ROS emission was generally lower in sWAT from lifelong highly exercise trained than older untrained subjects. Taken together, aging reduces intrinsic mitochondrial respiration in human sWAT, but lifelong high volume exercise training increases oxidative capacity by increasing mitochondrial volume likely contributing to healthy aging.
Collapse
Affiliation(s)
- Anders Gudiksen
- Section for Cell Biology and Physiology, Department of Biology, University of Copenhagen, Copenhagen, Denmark
| | - Albina Qoqaj
- Section for Cell Biology and Physiology, Department of Biology, University of Copenhagen, Copenhagen, Denmark
| | - Stine Ringholm
- Section for Cell Biology and Physiology, Department of Biology, University of Copenhagen, Copenhagen, Denmark
| | - Jørgen Wojtaszewski
- Section of Integrative Physiology, Department of Nutrition, Exercise and Sports, University of Copenhagen, Copenhagen, Denmark
| | - Peter Plomgaard
- Department of Clinical Biochemistry, Centre of Inflammation and Metabolism, and Centre for Physical Activity Research, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark. Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Henriette Pilegaard
- Section for Cell Biology and Physiology, Department of Biology, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
233
|
Lu B, Huang L, Cao J, Li L, Wu W, Chen X, Ding C. Adipose tissue macrophages in aging-associated adipose tissue function. J Physiol Sci 2021; 71:38. [PMID: 34863096 PMCID: PMC10717320 DOI: 10.1186/s12576-021-00820-2] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2021] [Accepted: 11/04/2021] [Indexed: 12/29/2022]
Abstract
"Inflammaging" refers to the chronic, low-grade inflammation that characterizes aging. Aging, like obesity, is associated with visceral adiposity and insulin resistance. Adipose tissue macrophages (ATMs) have played a major role in obesity-associated inflammation and insulin resistance. Macrophages are elevated in adipose tissue in aging. However, the changes and also possibly functions of ATMs in aging and aging-related diseases are unclear. In this review, we will summarize recent advances in research on the role of adipose tissue macrophages with aging-associated insulin resistance and discuss their potential therapeutic targets for preventing and treating aging and aging-related diseases.
Collapse
Affiliation(s)
- Bangchao Lu
- Department of Geriatrics, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, Jiangshu, China
| | - Liang Huang
- Department of Geriatrics, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, Jiangshu, China
| | - Juan Cao
- Department of Geriatrics, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, Jiangshu, China
| | - Lingling Li
- Department of Geriatrics, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, Jiangshu, China
| | - Wenhui Wu
- Department of Geriatrics, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, Jiangshu, China
| | - Xiaolin Chen
- Department of Geriatrics, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, Jiangshu, China
| | - Congzhu Ding
- Department of Geriatrics, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, Jiangshu, China.
| |
Collapse
|
234
|
Englund DA, Zhang X, Aversa Z, LeBrasseur NK. Skeletal muscle aging, cellular senescence, and senotherapeutics: Current knowledge and future directions. Mech Ageing Dev 2021; 200:111595. [PMID: 34742751 PMCID: PMC8627455 DOI: 10.1016/j.mad.2021.111595] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Revised: 10/29/2021] [Accepted: 11/01/2021] [Indexed: 12/16/2022]
Abstract
Cellular senescence is a state of cell cycle arrest induced by several forms of metabolic stress. Senescent cells accumulate with advancing age and have a distinctive phenotype, characterized by profound chromatin alterations and a robust senescence-associated secretory phenotype (SASP) that exerts negative effects on tissue health, both locally and systemically. In preclinical models, pharmacological agents that eliminate senescent cells (senotherapeutics) restore health and youthful properties in multiple tissues. To date, however, very little is understood about the vulnerability of terminally-differentiated skeletal muscle fibers and the resident mononuclear cells that populate the interstitial microenvironment of skeletal muscle to senescence, and their contribution to the onset and progression of skeletal muscle loss and dysfunction with aging. Scientific advances in these areas have the potential to highlight new therapeutic approaches to optimize late-life muscle health. To this end, this review highlights the current evidence and the key questions that need to be addressed to advance the field's understanding of cellular senescence as a mediator of skeletal muscle aging and the potential for emerging senescent cell-targeting therapies to counter age-related deficits in muscle mass, strength, and function. This article is part of the Special Issue - Senolytics - Edited by Joao Passos and Diana Jurk.
Collapse
Affiliation(s)
- Davis A Englund
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, MN, USA; Department of Physical Medicine and Rehabilitation, Mayo Clinic, Rochester, MN, USA
| | - Xu Zhang
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, MN, USA; Department of Physical Medicine and Rehabilitation, Mayo Clinic, Rochester, MN, USA
| | - Zaira Aversa
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, MN, USA; Department of Physical Medicine and Rehabilitation, Mayo Clinic, Rochester, MN, USA
| | - Nathan K LeBrasseur
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, MN, USA; Department of Physical Medicine and Rehabilitation, Mayo Clinic, Rochester, MN, USA; Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, USA.
| |
Collapse
|
235
|
Dixit M, Duran‐Ortiz S, Yildirim G, Poudel SB, Louis LD, Bartke A, Schaffler MB, Kopchick JJ, Yakar S. Induction of somatopause in adult mice compromises bone morphology and exacerbates bone loss during aging. Aging Cell 2021; 20:e13505. [PMID: 34811875 PMCID: PMC8672783 DOI: 10.1111/acel.13505] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2021] [Revised: 08/31/2021] [Accepted: 09/29/2021] [Indexed: 12/23/2022] Open
Abstract
Somatopause refers to the gradual declines in growth hormone (GH) and insulin‐like growth factor‐1 throughout aging. To define how induced somatopause affects skeletal integrity, we used an inducible GH receptor knockout (iGHRKO) mouse model. Somatopause, induced globally at 6 months of age, resulted in significantly more slender bones in both male and female iGHRKO mice. In males, induced somatopause was associated with progressive expansion of the marrow cavity leading to significant thinning of the cortices, which compromised bone strength. We report progressive declines in osteocyte lacunar number, and increases in lacunar volume, in iGHRKO males, and reductions in lacunar number accompanied by ~20% loss of overall canalicular connectivity in iGHRKO females by 30 months of age. Induced somatopause did not affect mineral/matrix ratio assessed by Raman microspectroscopy. We found significant increases in bone marrow adiposity and high levels of sclerostin, a negative regulator of bone formation in iGHRKO mice. Surprisingly, however, despite compromised bone morphology, osteocyte senescence was reduced in the iGHRKO mice. In this study, we avoided the confounded effects of constitutive deficiency in the GH/IGF‐1 axis on the skeleton during growth, and specifically dissected its effects on the aging skeleton. We show here, for the first time, that induced somatopause compromises bone morphology and the bone marrow environment.
Collapse
Affiliation(s)
- Manisha Dixit
- David B. Kriser Dental Center Department of Molecular Pathobiology New York University College of Dentistry New York New York NY USA
| | - Silvana Duran‐Ortiz
- Edison Biotechnology Institute and Dept. of Biomedical Sciences Ohio University Athens OH USA
| | - Godze Yildirim
- David B. Kriser Dental Center Department of Molecular Pathobiology New York University College of Dentistry New York New York NY USA
| | - Sher Bahadur Poudel
- David B. Kriser Dental Center Department of Molecular Pathobiology New York University College of Dentistry New York New York NY USA
| | - Leeann D. Louis
- Department of Biomedical Engineering City College of New York New York NY USA
| | - Andrzej Bartke
- Southern Illinois University School of Medicine Springfield IL USA
| | | | - John J. Kopchick
- Edison Biotechnology Institute and Dept. of Biomedical Sciences Ohio University Athens OH USA
| | - Shoshana Yakar
- David B. Kriser Dental Center Department of Molecular Pathobiology New York University College of Dentistry New York New York NY USA
| |
Collapse
|
236
|
Jayawardena TU, Nagahawatta D, Lu YA, Yang HW, Je JG, Kim SY, Jeon YJ. Ishige okamurae and diphloroethohydoxycarmalol inhibit palmitic acid-impaired skeletal myogenesis and improve muscle regenerative potential. J Funct Foods 2021. [DOI: 10.1016/j.jff.2021.104832] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
|
237
|
Matsunaga T, Iske J, Schroeter A, Azuma H, Zhou H, Tullius SG. The potential of Senolytics in transplantation. Mech Ageing Dev 2021; 200:111582. [PMID: 34606875 PMCID: PMC10655132 DOI: 10.1016/j.mad.2021.111582] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Revised: 09/29/2021] [Accepted: 10/01/2021] [Indexed: 12/19/2022]
Abstract
Older organs provide a substantial unrealized potential with the capacity to close the gap between demand and supply in organ transplantation. The potential of senolytics in improving age-related conditions has been shown in various experimental studies and early clinical trials. Those encouraging data may also be of relevance for transplantation. As age-differences between donor and recipients are not uncommon, aging may be accelerated in recipients when transplanting older organs; young organs may, at least in theory, have the potential to 'rejuvenate' old recipients. Here, we review the relevance of senescent cells and the effects of senolytics on organ quality, alloimmune responses and outcomes in solid organ transplantation. This article is part of the Special Issue - Senolytics - Edited by Joao Passos and Diana Jurk.
Collapse
Affiliation(s)
- Tomohisa Matsunaga
- Division of Transplant Surgery, Department of Surgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA; Department of Urology, Osaka Medical and Pharmaceutical University, Takatsuki, Osaka, Japan
| | - Jasper Iske
- Division of Transplant Surgery, Department of Surgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA; Institute of Transplant Immunology, Hannover Medical School, Hannover, Lower Saxony, Germany
| | - Andreas Schroeter
- Division of Transplant Surgery, Department of Surgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA; Regenerative Medicine and Experimental Surgery, Department of General, Visceral and Transplant Surgery, Hannover Medical School, Hannover, Lower Saxony, Germany
| | - Haruhito Azuma
- Department of Urology, Osaka Medical and Pharmaceutical University, Takatsuki, Osaka, Japan
| | - Hao Zhou
- Division of Transplant Surgery, Department of Surgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Stefan G Tullius
- Division of Transplant Surgery, Department of Surgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
238
|
Zhuang H, Karvinen S, Törmäkangas T, Zhang X, Ojanen X, Velagapudi V, Alen M, Britton SL, Koch LG, Kainulainen H, Cheng S, Wiklund P. Interactive effects of aging and aerobic capacity on energy metabolism-related metabolites of serum, skeletal muscle, and white adipose tissue. GeroScience 2021; 43:2679-2691. [PMID: 34089174 PMCID: PMC8602622 DOI: 10.1007/s11357-021-00387-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Accepted: 05/17/2021] [Indexed: 12/25/2022] Open
Abstract
Aerobic capacity is a strong predictor of longevity. With aging, aerobic capacity decreases concomitantly with changes in whole body metabolism leading to increased disease risk. To address the role of aerobic capacity, aging, and their interaction on metabolism, we utilized rat models selectively bred for low and high intrinsic aerobic capacity (LCRs/HCRs) and compared the metabolomics of serum, muscle, and white adipose tissue (WAT) at two time points: Young rats were sacrificed at 9 months of age, and old rats were sacrificed at 21 months of age. Targeted and semi-quantitative metabolomics analysis was performed on the ultra-pressure liquid chromatography tandem mass spectrometry (UPLC-MS) platform. The effects of aerobic capacity, aging, and their interaction were studied via regression analysis. Our results showed that high aerobic capacity is associated with an accumulation of isovalerylcarnitine in muscle and serum at rest, which is likely due to more efficient leucine catabolism in muscle. With aging, several amino acids were downregulated in muscle, indicating more efficient amino acid metabolism, whereas in WAT less efficient amino acid metabolism and decreased mitochondrial β-oxidation were observed. Our results further revealed that high aerobic capacity and aging interactively affect lipid metabolism in muscle and WAT, possibly combating unfavorable aging-related changes in whole body metabolism. Our results highlight the significant role of WAT metabolism for healthy aging.
Collapse
Affiliation(s)
- Haihui Zhuang
- School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, China
- Key Laboratory of Systems Biomedicine (Ministry of Education), and Exercise Translational Medicine Center, Shanghai Center for Systems Biomedicine, Shanghai Jiao Tong University, Shanghai, China
- Faculty of Sport and Health Sciences, University of Jyväskylä, Jyväskylä, Finland
| | - Sira Karvinen
- Faculty of Sport and Health Sciences, University of Jyväskylä, Jyväskylä, Finland.
| | - Timo Törmäkangas
- Faculty of Sport and Health Sciences, University of Jyväskylä, Jyväskylä, Finland
| | - Xiaobo Zhang
- School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, China
- Key Laboratory of Systems Biomedicine (Ministry of Education), and Exercise Translational Medicine Center, Shanghai Center for Systems Biomedicine, Shanghai Jiao Tong University, Shanghai, China
| | - Xiaowei Ojanen
- School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, China
| | - Vidya Velagapudi
- Metabolomics Unit, Institute for Molecular Medicine Finland (FIMM), HiLIFE, University of Helsinki, Helsinki, Finland
| | - Markku Alen
- Department of Medical Rehabilitation, Oulu University Hospital, Oulu, Finland
| | - Steven L Britton
- Department of Anesthesiology, University of Michigan, Ann Arbor, MI, USA
- Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI, USA
| | - Lauren G Koch
- Department of Physiology and Pharmacology, The University of Toledo College of Medicine and Life Sciences, Toledo, OH, USA
| | - Heikki Kainulainen
- Faculty of Sport and Health Sciences, University of Jyväskylä, Jyväskylä, Finland
| | - Sulin Cheng
- School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, China
- Key Laboratory of Systems Biomedicine (Ministry of Education), and Exercise Translational Medicine Center, Shanghai Center for Systems Biomedicine, Shanghai Jiao Tong University, Shanghai, China
- Faculty of Sport and Health Sciences, University of Jyväskylä, Jyväskylä, Finland
| | - Petri Wiklund
- Key Laboratory of Systems Biomedicine (Ministry of Education), and Exercise Translational Medicine Center, Shanghai Center for Systems Biomedicine, Shanghai Jiao Tong University, Shanghai, China
- Huawei Helsinki Research Center, Huawei Technologies Oy (Finland) Co. Ltd, Helsinki, Finland
| |
Collapse
|
239
|
Abstract
Cell membrane fusion and multinucleation in macrophages are associated with physiologic homeostasis as well as disease. Osteoclasts are multinucleated macrophages that resorb bone through increased metabolic activity resulting from cell fusion. Fusion of macrophages also generates multinucleated giant cells (MGCs) in white adipose tissue (WAT) of obese individuals. For years, our knowledge of MGCs in WAT has been limited to their description as part of crown-like structures (CLS) surrounding damaged adipocytes. However, recent evidence indicates that these cells can phagocytose oversized lipid remnants, suggesting that, as in osteoclasts, cell fusion and multinucleation are required for specialized catabolic functions. We thus reason that WAT MGCs can be viewed as functionally analogous to osteoclasts and refer to them in this article as adipoclasts. We first review current knowledge on adipoclasts and their described functions. In view of recent advances in single cell genomics, we describe WAT macrophages from a ‘fusion perspective’ and speculate on the ontogeny of adipoclasts. Specifically, we highlight the role of CD9 and TREM2, two plasma membrane markers of lipid-associated macrophages in WAT, which have been previously described as regulators of fusion and multinucleation in osteoclasts and MGCs. Finally, we consider whether strategies aiming to target WAT macrophages can be more selectively directed against adipoclasts.
Collapse
|
240
|
Zhu Y, Wang T, He S, Pu S, Zhao H, Zhou Z, Wu Q. Comparison of Antiobesity Effects of Adipose-Derived Stromal/Stem Cells from Different Sources in a Natural Aging Model. Diabetes Metab Syndr Obes 2021; 14:4535-4546. [PMID: 34815680 PMCID: PMC8604647 DOI: 10.2147/dmso.s334044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/19/2021] [Accepted: 10/26/2021] [Indexed: 11/23/2022] Open
Abstract
PURPOSE Our previous study found that white adipose stem cells (W-ASCs) derived from abdominal and femoral sulcus white adipose stem cells (ASCs) have antiaging and age-related obesity effects. Whether interscapular brown adipose stem cells (B-ASCs) have the same effect has not been reported. The study objective was to compare the effects of ASCs from different tissues on aging and aging-related obesity. PATIENTS AND METHODS C57BL/6J mice at 22 months of age were transplanted with either B-ASCs or W-ASCs from young mice at 2 months of age. Changes in body weight, biochemistry, cytokines, hormone secretion, cell senescence, lipid metabolism, and ASC function were assessed after transplanted 1 month. RESULTS W-ASCs were superior to B-ASCs as aging and age-related obesity indicators, based on change in body weight, organ weight, antioxidant and anti-inflammatory activity, lipid metabolism, and liver and kidney function. CONCLUSION Difference in the tissue source was reflected by the heterogeneity of antiaging and age-related obesity effects of transplanted ASCs. Based on the study results, we recommend W-ASCs over B-ASCs in aging and age-related obesity applications.
Collapse
Affiliation(s)
- Yu Zhu
- School of Life Sciences, Guangxi Normal University, Guilin, Guangxi Zhuang Autonomous Region, People’s Republic of China
- Guangxi Universities Key Laboratory of Stem Cell and Biopharmaceutical Technology, Guangxi Normal University, Guilin, Guangxi Zhuang Autonomous Region, People’s Republic of China
- Research Center for Biomedical Sciences, Guangxi Normal University, Guilin, Guangxi Zhuang Autonomous Region, People’s Republic of China
| | - Tao Wang
- School of Life Sciences, Guangxi Normal University, Guilin, Guangxi Zhuang Autonomous Region, People’s Republic of China
- Guangxi Universities Key Laboratory of Stem Cell and Biopharmaceutical Technology, Guangxi Normal University, Guilin, Guangxi Zhuang Autonomous Region, People’s Republic of China
- Research Center for Biomedical Sciences, Guangxi Normal University, Guilin, Guangxi Zhuang Autonomous Region, People’s Republic of China
| | - Shuangli He
- School of Life Sciences, Guangxi Normal University, Guilin, Guangxi Zhuang Autonomous Region, People’s Republic of China
- Guangxi Universities Key Laboratory of Stem Cell and Biopharmaceutical Technology, Guangxi Normal University, Guilin, Guangxi Zhuang Autonomous Region, People’s Republic of China
- Research Center for Biomedical Sciences, Guangxi Normal University, Guilin, Guangxi Zhuang Autonomous Region, People’s Republic of China
| | - Shiming Pu
- School of Life Sciences, Guangxi Normal University, Guilin, Guangxi Zhuang Autonomous Region, People’s Republic of China
- Guangxi Universities Key Laboratory of Stem Cell and Biopharmaceutical Technology, Guangxi Normal University, Guilin, Guangxi Zhuang Autonomous Region, People’s Republic of China
- Research Center for Biomedical Sciences, Guangxi Normal University, Guilin, Guangxi Zhuang Autonomous Region, People’s Republic of China
| | - Hongxia Zhao
- School of Life Sciences, Guangxi Normal University, Guilin, Guangxi Zhuang Autonomous Region, People’s Republic of China
- Guangxi Universities Key Laboratory of Stem Cell and Biopharmaceutical Technology, Guangxi Normal University, Guilin, Guangxi Zhuang Autonomous Region, People’s Republic of China
- Research Center for Biomedical Sciences, Guangxi Normal University, Guilin, Guangxi Zhuang Autonomous Region, People’s Republic of China
| | - Zuping Zhou
- School of Life Sciences, Guangxi Normal University, Guilin, Guangxi Zhuang Autonomous Region, People’s Republic of China
- Guangxi Universities Key Laboratory of Stem Cell and Biopharmaceutical Technology, Guangxi Normal University, Guilin, Guangxi Zhuang Autonomous Region, People’s Republic of China
- Research Center for Biomedical Sciences, Guangxi Normal University, Guilin, Guangxi Zhuang Autonomous Region, People’s Republic of China
| | - Qiong Wu
- School of Life Sciences, Guangxi Normal University, Guilin, Guangxi Zhuang Autonomous Region, People’s Republic of China
- Guangxi Universities Key Laboratory of Stem Cell and Biopharmaceutical Technology, Guangxi Normal University, Guilin, Guangxi Zhuang Autonomous Region, People’s Republic of China
- Research Center for Biomedical Sciences, Guangxi Normal University, Guilin, Guangxi Zhuang Autonomous Region, People’s Republic of China
| |
Collapse
|
241
|
Abstract
Autophagy is an evolutionarily conserved, lysosome-dependent catabolic process whereby cytoplasmic components, including damaged organelles, protein aggregates and lipid droplets, are degraded and their components recycled. Autophagy has an essential role in maintaining cellular homeostasis in response to intracellular stress; however, the efficiency of autophagy declines with age and overnutrition can interfere with the autophagic process. Therefore, conditions such as sarcopenic obesity, insulin resistance and type 2 diabetes mellitus (T2DM) that are characterized by metabolic derangement and intracellular stresses (including oxidative stress, inflammation and endoplasmic reticulum stress) also involve the accumulation of damaged cellular components. These conditions are prevalent in ageing populations. For example, sarcopenia is an age-related loss of skeletal muscle mass and strength that is involved in the pathogenesis of both insulin resistance and T2DM, particularly in elderly people. Impairment of autophagy results in further aggravation of diabetes-related metabolic derangements in insulin target tissues, including the liver, skeletal muscle and adipose tissue, as well as in pancreatic β-cells. This Review summarizes the role of autophagy in the pathogenesis of metabolic diseases associated with or occurring in the context of ageing, including insulin resistance, T2DM and sarcopenic obesity, and describes its potential as a therapeutic target.
Collapse
Affiliation(s)
- Munehiro Kitada
- Department of Diabetology and Endocrinology, Kanazawa Medical University, Uchinada, Ishikawa, Japan
- Division of Anticipatory Molecular Food Science and Technology, Medical Research Institute, Kanazawa Medical University, Uchinada, Ishikawa, Japan
| | - Daisuke Koya
- Department of Diabetology and Endocrinology, Kanazawa Medical University, Uchinada, Ishikawa, Japan.
- Division of Anticipatory Molecular Food Science and Technology, Medical Research Institute, Kanazawa Medical University, Uchinada, Ishikawa, Japan.
- Department of General Internal Medicine, Kusatsu General Hospital, Kusatsu, Shiga, Japan.
| |
Collapse
|
242
|
Low E, Alimohammadiha G, Smith LA, Costello LF, Przyborski SA, von Zglinicki T, Miwa S. How good is the evidence that cellular senescence causes skin ageing? Ageing Res Rev 2021; 71:101456. [PMID: 34487917 PMCID: PMC8524668 DOI: 10.1016/j.arr.2021.101456] [Citation(s) in RCA: 55] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2021] [Revised: 08/25/2021] [Accepted: 08/31/2021] [Indexed: 12/11/2022]
Abstract
Skin is the largest organ of the body with important protective functions, which become compromised with time due to both intrinsic and extrinsic ageing processes. Cellular senescence is the primary ageing process at cell level, associated with loss of proliferative capacity, mitochondrial dysfunction and significantly altered patterns of expression and secretion of bioactive molecules. Intervention experiments have proven cell senescence as a relevant cause of ageing in many organs. In case of skin, accumulation of senescence in all major compartments with ageing is well documented and might be responsible for most, if not all, the molecular changes observed during ageing. Incorporation of senescent cells into in-vitro skin models (specifically 3D full thickness models) recapitulates changes typically associated with skin ageing. However, crucial evidence is still missing. A beneficial effect of senescent cell ablation on skin ageing has so far only been shown following rather unspecific interventions or in transgenic mouse models. We conclude that evidence for cellular senescence as a relevant cause of intrinsic skin ageing is highly suggestive but not yet completely conclusive.
Collapse
Affiliation(s)
- Evon Low
- Ageing Biology Laboratories, Newcastle University Biosciences Institute, Newcastle University, Newcastle upon Tyne NE4 5PL, UK
| | - Ghazaleh Alimohammadiha
- Ageing Biology Laboratories, Newcastle University Biosciences Institute, Newcastle University, Newcastle upon Tyne NE4 5PL, UK
| | - Lucy A Smith
- Department of Biosciences, Durham University, South Road, Durham DH1 3LE, UK
| | - Lydia F Costello
- Department of Biosciences, Durham University, South Road, Durham DH1 3LE, UK
| | - Stefan A Przyborski
- Department of Biosciences, Durham University, South Road, Durham DH1 3LE, UK
| | - Thomas von Zglinicki
- Ageing Biology Laboratories, Newcastle University Biosciences Institute, Newcastle University, Newcastle upon Tyne NE4 5PL, UK.
| | - Satomi Miwa
- Ageing Biology Laboratories, Newcastle University Biosciences Institute, Newcastle University, Newcastle upon Tyne NE4 5PL, UK
| |
Collapse
|
243
|
Khosla S, Samakkarnthai P, Monroe DG, Farr JN. Update on the pathogenesis and treatment of skeletal fragility in type 2 diabetes mellitus. Nat Rev Endocrinol 2021; 17:685-697. [PMID: 34518671 PMCID: PMC8605611 DOI: 10.1038/s41574-021-00555-5] [Citation(s) in RCA: 131] [Impact Index Per Article: 32.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 08/06/2021] [Indexed: 02/08/2023]
Abstract
Fracture risk is increased in patients with type 2 diabetes mellitus (T2DM). In addition, these patients sustain fractures despite having higher levels of areal bone mineral density, as measured by dual-energy X-ray absorptiometry, than individuals without T2DM. Thus, additional factors such as alterations in bone quality could have important roles in mediating skeletal fragility in patients with T2DM. Although the pathogenesis of increased fracture risk in T2DM is multifactorial, impairments in bone material properties and increases in cortical porosity have emerged as two key skeletal abnormalities that contribute to skeletal fragility in patients with T2DM. In addition, indices of bone formation are uniformly reduced in patients with T2DM, with evidence from mouse studies published over the past few years linking this abnormality to accelerated skeletal ageing, specifically cellular senescence. In this Review, we highlight the latest advances in our understanding of the mechanisms of skeletal fragility in patients with T2DM and suggest potential novel therapeutic approaches to address this problem.
Collapse
Affiliation(s)
- Sundeep Khosla
- Division of Endocrinology and Kogod Center on Aging, Mayo Clinic, Rochester, MN, USA.
| | - Parinya Samakkarnthai
- Division of Endocrinology and Kogod Center on Aging, Mayo Clinic, Rochester, MN, USA
- Division of Endocrinology, Phramongkutklao Hospital and College of Medicine, Bangkok, Thailand
| | - David G Monroe
- Division of Endocrinology and Kogod Center on Aging, Mayo Clinic, Rochester, MN, USA
| | - Joshua N Farr
- Division of Endocrinology and Kogod Center on Aging, Mayo Clinic, Rochester, MN, USA
| |
Collapse
|
244
|
Gries KJ, Zysik VS, Jobe TK, Griffin N, Leeds BP, Lowery JW. Muscle-derived factors influencing bone metabolism. Semin Cell Dev Biol 2021; 123:57-63. [PMID: 34756782 DOI: 10.1016/j.semcdb.2021.10.009] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2021] [Revised: 09/28/2021] [Accepted: 10/20/2021] [Indexed: 12/11/2022]
Abstract
A significant amount of attention has been brought to the endocrine-like function of skeletal muscle on various tissues, particularly with bone. Several lines of investigation indicate that the physiology of both bone and muscle systems may be regulated by a given stimulus, such as exercise, aging, and inactivity. Moreover, emerging evidence indicates that bone is heavily influenced by soluble factors derived from skeletal muscle (i.e., muscle-to-bone communication). The purpose of this review is to discuss the regulation of bone remodeling (formation and/or resorption) through skeletal muscle-derived cytokines (hereafter myokines) including the anti-inflammatory cytokine METRNL and pro-inflammatory cytokines (e.g., TNF-α, IL-6, FGF-2 and others). Our goal is to highlight possible therapeutic opportunities to improve muscle and bone health in aging.
Collapse
Affiliation(s)
- Kevin J Gries
- Program in Exercise & Sports Science, Marian University, 3200 Cold Spring Road, Indianapolis, IN 46222, USA; Bone & Muscle Research Group, Marian University, 3200 Cold Spring Road, Indianapolis, IN 46222, USA; Division of Biomedical Science, Marian University College of Osteopathic Medicine, 3200 Cold Spring Road, Indianapolis, IN 46222, USA.
| | - Victoria S Zysik
- Bone & Muscle Research Group, Marian University, 3200 Cold Spring Road, Indianapolis, IN 46222, USA; Marian University College of Osteopathic Medicine, 3200 Cold Spring Road, Indianapolis, IN 46222, USA
| | - Tyler K Jobe
- Program in Exercise & Sports Science, Marian University, 3200 Cold Spring Road, Indianapolis, IN 46222, USA
| | - Nicole Griffin
- Bone & Muscle Research Group, Marian University, 3200 Cold Spring Road, Indianapolis, IN 46222, USA; Marian University College of Osteopathic Medicine, 3200 Cold Spring Road, Indianapolis, IN 46222, USA
| | - Benjamin P Leeds
- Bone & Muscle Research Group, Marian University, 3200 Cold Spring Road, Indianapolis, IN 46222, USA; Division of Clinical Affairs, Marian University College of Osteopathic Medicine, 3200 Cold Spring Road, Indianapolis, IN 46222, USA
| | - Jonathan W Lowery
- Bone & Muscle Research Group, Marian University, 3200 Cold Spring Road, Indianapolis, IN 46222, USA; Division of Biomedical Science, Marian University College of Osteopathic Medicine, 3200 Cold Spring Road, Indianapolis, IN 46222, USA
| |
Collapse
|
245
|
Henson SM, Aksentijevic D. Senescence and Type 2 Diabetic Cardiomyopathy: How Young Can You Die of Old Age? Front Pharmacol 2021; 12:716517. [PMID: 34690759 PMCID: PMC8529062 DOI: 10.3389/fphar.2021.716517] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Accepted: 09/16/2021] [Indexed: 01/10/2023] Open
Abstract
Inflammation is well understood to be a physiological process of ageing however it also underlies many chronic diseases, including conditions without an obvious pathogenic inflammatory element. Recent findings have unequivocally identified type 2 diabetes (T2D) as a chronic inflammatory disease characterized by inflammation and immune senescence. Immunosenescence is a hallmark of the prolonged low-grade systemic inflammation, in particular associated with metabolic syndrome and can be a cause as well as a consequence of T2D. Diabetes is a risk factor for cardiovascular mortality and remodelling and with particular changes to myocardial structure, function, metabolism and energetics collectively resulting in diabetic cardiomyopathy. Both cardiomyocytes and immune cells undergo metabolic remodelling in T2D and as a result become trapped in a vicious cycle of lost metabolic flexibility, thus losing their key adaptive mechanisms to dynamic changes in O2 and nutrient availability. Immunosenescence driven by metabolic stress may be both the cause and key contributing factor to cardiac dysfunction in diabetic cardiomyopathy by inducing metabolic perturbations that can lead to impaired energetics, a strong predictor of cardiac mortality. Here we review our current understanding of the cross-talk between inflammaging and cardiomyocytes in T2D cardiomyopathy. We discuss potential mechanisms of metabolic convergence between cell types which, we hypothesize, might tip the balance between resolution of the inflammation versus adverse cardiac metabolic remodelling in T2D cardiomyopathy. A better understanding of the multiple biological paradigms leading to T2D cardiomyopathy including the immunosenescence associated with inflammaging will provide a powerful target for successful therapeutic interventions.
Collapse
Affiliation(s)
- Sian M Henson
- Centre for Translational Medicine and Therapeutics, London, United Kingdom
| | - Dunja Aksentijevic
- Centre for Biochemical Pharmacology, London, United Kingdom.,Centre for Inflammation and Therapeutic Innovation William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| |
Collapse
|
246
|
Abstract
Obesity is a major risk factor for the development of comorbidities such as type 2 diabetes, neurodegenerative disorders, osteoarthritis, cancer, cardiovascular and renal diseases. The onset of obesity is linked to an increase of senescent cells within adipose tissue and other organs. Cellular senescence is a stress response that has been shown to be causally linked to aging and development of various age-related diseases such as obesity. The senescence-associated-secretory phenotype of senescent cells creates a chronic inflammatory milieu that leads to local and systemic dysfunction. The elimination of senescent cells using pharmacological approaches (i.e., senolytics) has been shown to delay, prevent, or alleviate obesity-related organ dysfunction.
Collapse
Affiliation(s)
- Selim Chaib
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, MN, USA
| | - Tamara Tchkonia
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, MN, USA
| | - James L Kirkland
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, MN, USA.
| |
Collapse
|
247
|
Wilfred SA, Becker CB, Kanzler KE, Musi N, Espinoza SE, Kilpela LS. Binge eating among older women: prevalence rates and health correlates across three independent samples. J Eat Disord 2021; 9:132. [PMID: 34666821 PMCID: PMC8524882 DOI: 10.1186/s40337-021-00484-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/19/2021] [Accepted: 09/22/2021] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Emerging research indicates that binge eating (BE; consuming unusually large amounts of food in one siting while feeling a loss of control) is prevalent among older women. Yet, health correlates of BE in older adult populations are poorly understood. The original study aimed to investigate BE prevalence, frequency, and health correlates in a sample of older adult women. Based on results from this first study, we then sought to replicate findings in two additional samples of older adult women from separate studies. METHOD Using self-reported frequencies of BE from three separate samples of older women with very different demographics, we compared BE prevalence, frequency, and health correlates among older women. Study 1 (N = 185) includes data collected online (86% White; 59% overweight/obese status). Study 2 (N = 64) was conducted in person at a local food pantry (65% Hispanic; 47% household income < $10,000/year). Study 3 (N = 100) comprises data collected online (72% White; 50% Masters/Doctoral Degree). RESULTS Per DSM-5 frequency criterion of BE at least weekly, we found prevalence rates ranging from 19 to 26% across the three samples. Correlates of BE frequency included elevated negative mood, worry, BMI, and less nutritious food consumption. CONCLUSIONS Across three very different samples in terms of race/ethnicity, education, food security status, measurements, and sampling methodology, we found fairly consistent rates of self-reported BE at least weekly (19-26%). Results suggest that BE is related to negative health indices among older women and support the need for more research in this population.
Collapse
Affiliation(s)
| | | | | | - Nicolas Musi
- Barshop Institute, UT Health San Antonio, San Antonio, TX, USA
- South Texas VA Health System, Audie Murphy Veterans Hospital, San Antonio, TX, USA
| | - Sara E Espinoza
- Barshop Institute, UT Health San Antonio, San Antonio, TX, USA
- South Texas VA Health System, Audie Murphy Veterans Hospital, San Antonio, TX, USA
| | - Lisa Smith Kilpela
- ReACH Center, UT Health San Antonio, San Antonio, TX, USA.
- Barshop Institute, UT Health San Antonio, San Antonio, TX, USA.
| |
Collapse
|
248
|
Morales-Valencia J, David G. The Contribution of Physiological and Accelerated Aging to Cancer Progression Through Senescence-Induced Inflammation. Front Oncol 2021; 11:747822. [PMID: 34621683 PMCID: PMC8490756 DOI: 10.3389/fonc.2021.747822] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2021] [Accepted: 09/06/2021] [Indexed: 01/10/2023] Open
Abstract
Senescent cells are found to accumulate in aged individuals, as well as in cancer patients that receive chemotherapeutic treatment. Although originally believed to halt cancer progression due to their characteristic growth arrest, senescent cells remain metabolically active and secrete a combination of inflammatory agents, growth factors and proteases, collectively known as the senescence-associated secretory phenotype (SASP). In this review, we discuss the contribution of senescent cells to cancer progression through their ability to alter cancer cells’ properties and to generate a microenvironment that promotes tumor growth. Furthermore, recent evidence suggests that senescent cells are able resume proliferation and drive cancer relapse, pointing to the use of senolytics and SASP modulators as a potential approach to prevent tumor resurgence following treatment cessation. Thus, a better understanding of the hallmarks of senescence and the impact of the SASP will allow the development of improved targeted therapeutic strategies to leverage vulnerabilities associated with this cellular state.
Collapse
Affiliation(s)
- Jorge Morales-Valencia
- Department of Biochemistry and Molecular Pharmacology, New York University School of Medicine, New York, NY, United States.,NYU Cancer Institute, New York University School of Medicine, New York, NY, United States
| | - Gregory David
- Department of Biochemistry and Molecular Pharmacology, New York University School of Medicine, New York, NY, United States.,NYU Cancer Institute, New York University School of Medicine, New York, NY, United States.,Department of Urology, New York University School of Medicine, New York, NY, United States
| |
Collapse
|
249
|
Li Q, Hagberg CE, Silva Cascales H, Lang S, Hyvönen MT, Salehzadeh F, Chen P, Alexandersson I, Terezaki E, Harms MJ, Kutschke M, Arifen N, Krämer N, Aouadi M, Knibbe C, Boucher J, Thorell A, Spalding KL. Obesity and hyperinsulinemia drive adipocytes to activate a cell cycle program and senesce. Nat Med 2021; 27:1941-1953. [PMID: 34608330 DOI: 10.1038/s41591-021-01501-8] [Citation(s) in RCA: 99] [Impact Index Per Article: 24.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Accepted: 08/12/2021] [Indexed: 01/10/2023]
Abstract
Obesity is considered an important factor for many chronic diseases, including diabetes, cardiovascular disease and cancer. The expansion of adipose tissue in obesity is due to an increase in both adipocyte progenitor differentiation and mature adipocyte cell size. Adipocytes, however, are thought to be unable to divide or enter the cell cycle. We demonstrate that mature human adipocytes unexpectedly display a gene and protein signature indicative of an active cell cycle program. Adipocyte cell cycle progression associates with obesity and hyperinsulinemia, with a concomitant increase in cell size, nuclear size and nuclear DNA content. Chronic hyperinsulinemia in vitro or in humans, however, is associated with subsequent cell cycle exit, leading to a premature senescent transcriptomic and secretory profile in adipocytes. Premature senescence is rapidly becoming recognized as an important mediator of stress-induced tissue dysfunction. By demonstrating that adipocytes can activate a cell cycle program, we define a mechanism whereby mature human adipocytes senesce. We further show that by targeting the adipocyte cell cycle program using metformin, it is possible to influence adipocyte senescence and obesity-associated adipose tissue inflammation.
Collapse
Affiliation(s)
- Qian Li
- Department of Cell and Molecular Biology, Karolinska Institutet, Stockholm, Sweden
| | - Carolina E Hagberg
- Karolinska Institutet/AstraZeneca Integrated Cardio Metabolic Centre (KI/AZ ICMC), Department of Medicine Huddinge, Karolinska Institutet, Stockholm, Sweden.,Cardiovascular Medicine Division, Department of Medicine, Solna, Karolinska Institutet, Stockholm, Sweden
| | - Helena Silva Cascales
- Karolinska Institutet/AstraZeneca Integrated Cardio Metabolic Centre (KI/AZ ICMC), Department of Medicine Huddinge, Karolinska Institutet, Stockholm, Sweden
| | - Shuai Lang
- Department of Cell and Molecular Biology, Karolinska Institutet, Stockholm, Sweden
| | - Mervi T Hyvönen
- Department of Cell and Molecular Biology, Karolinska Institutet, Stockholm, Sweden.,School of Pharmacy, University of Eastern Finland, Kuopio, Finland
| | - Firoozeh Salehzadeh
- Karolinska Institutet/AstraZeneca Integrated Cardio Metabolic Centre (KI/AZ ICMC), Department of Medicine Huddinge, Karolinska Institutet, Stockholm, Sweden
| | - Ping Chen
- Karolinska Institutet/AstraZeneca Integrated Cardio Metabolic Centre (KI/AZ ICMC), Department of Medicine Huddinge, Karolinska Institutet, Stockholm, Sweden.,Center for Infectious Medicine, Department of Medicine, Karolinska Institute, Stockolm, Sweden
| | - Ida Alexandersson
- Bioscience Metabolism, Research and Early Development, Cardiovascular, Renal and Metabolism (CVRM), BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Eleni Terezaki
- Karolinska Institutet/AstraZeneca Integrated Cardio Metabolic Centre (KI/AZ ICMC), Department of Medicine Huddinge, Karolinska Institutet, Stockholm, Sweden
| | - Matthew J Harms
- Bioscience Metabolism, Research and Early Development, Cardiovascular, Renal and Metabolism (CVRM), BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Maria Kutschke
- Karolinska Institutet/AstraZeneca Integrated Cardio Metabolic Centre (KI/AZ ICMC), Department of Medicine Huddinge, Karolinska Institutet, Stockholm, Sweden.,Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University, Stockholm, Sweden
| | - Nahida Arifen
- Karolinska Institutet/AstraZeneca Integrated Cardio Metabolic Centre (KI/AZ ICMC), Department of Medicine Huddinge, Karolinska Institutet, Stockholm, Sweden
| | - Niels Krämer
- Department of Cell and Molecular Biology, Karolinska Institutet, Stockholm, Sweden
| | - Myriam Aouadi
- Karolinska Institutet/AstraZeneca Integrated Cardio Metabolic Centre (KI/AZ ICMC), Department of Medicine Huddinge, Karolinska Institutet, Stockholm, Sweden.,Center for Infectious Medicine, Department of Medicine, Karolinska Institute, Stockolm, Sweden
| | - Carole Knibbe
- CarMeN Laboratory, Lyon University, INRIA, INSA Lyon, Lyon, France
| | - Jeremie Boucher
- Bioscience Metabolism, Research and Early Development, Cardiovascular, Renal and Metabolism (CVRM), BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden.,The Lundberg Laboratory for Diabetes Research, University of Gothenburg, Gothenburg, Sweden.,Wallenberg Centre for Molecular and Translational Medicine, University of Gothenburg, Gothenburg, Sweden
| | - Anders Thorell
- Department of Clinical Science, Danderyds Hospital, Karolinska Institutet and Department of Surgery, Ersta Hospital, Karolinska Institutet, Stockholm, Sweden
| | - Kirsty L Spalding
- Department of Cell and Molecular Biology, Karolinska Institutet, Stockholm, Sweden. .,Karolinska Institutet/AstraZeneca Integrated Cardio Metabolic Centre (KI/AZ ICMC), Department of Medicine Huddinge, Karolinska Institutet, Stockholm, Sweden.
| |
Collapse
|
250
|
Tarantini S, Balasubramanian P, Delfavero J, Csipo T, Yabluchanskiy A, Kiss T, Nyúl-Tóth Á, Mukli P, Toth P, Ahire C, Ungvari A, Benyo Z, Csiszar A, Ungvari Z. Treatment with the BCL-2/BCL-xL inhibitor senolytic drug ABT263/Navitoclax improves functional hyperemia in aged mice. GeroScience 2021; 43:2427-2440. [PMID: 34427858 PMCID: PMC8599595 DOI: 10.1007/s11357-021-00440-z] [Citation(s) in RCA: 68] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2021] [Accepted: 08/11/2021] [Indexed: 12/13/2022] Open
Abstract
Moment-to-moment adjustment of regional cerebral blood flow to neuronal activity via neurovascular coupling (NVC or "functional hyperemia") has a critical role in maintenance of healthy cognitive function. Aging-induced impairment of NVC responses importantly contributes to age-related cognitive decline. Advanced aging is associated with increased prevalence of senescent cells in the cerebral microcirculation, but their role in impaired NVC responses remains unexplored. The present study was designed to test the hypothesis that a validated senolytic treatment can improve NVC responses and cognitive performance in aged mice. To achieve this goal, aged (24-month-old) C57BL/6 mice were treated with ABT263/Navitoclax, a potent senolytic agent known to eliminate senescent cells in the aged mouse brain. Mice were behaviorally evaluated (radial arms water maze) and NVC was assessed by measuring CBF responses (laser speckle contrast imaging) in the somatosensory whisker barrel cortex evoked by contralateral whisker stimulation. We found that NVC responses were significantly impaired in aged mice. ABT263/Navitoclax treatment improved NVC response, which was associated with significantly improved hippocampal-encoded functions of learning and memory. ABT263/Navitoclax treatment did not significantly affect endothelium-dependent acetylcholine-induced relaxation of aorta rings. Thus, increased presence of senescent cells in the aged brain likely contributes to age-related neurovascular uncoupling, exacerbating cognitive decline. The neurovascular protective effects of ABT263/Navitoclax treatment highlight the preventive and therapeutic potential of senolytic treatments (as monotherapy or as part of combination treatment regimens) as effective interventions in patients at risk for vascular cognitive impairment (VCI).
Collapse
Affiliation(s)
- Stefano Tarantini
- Vascular Cognitive Impairment and Neurodegeneration Program, Oklahoma Center for Geroscience and Healthy Brain Aging, Department of Biochemistry & Molecular Biology, University of Oklahoma Health Sciences Center, 975 NE 10th Street, BRC 1311, Oklahoma City, OK, 731042, USA.
- Peggy and Charles Stephenson Cancer Center, Oklahoma City, OK, 73104, USA.
- Department of Health Promotion Sciences, College of Public Health, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA.
- International Training Program in Geroscience, Doctoral School of Basic and Translational Medicine/Department of Public Health, Semmelweis University, Budapest, Hungary.
| | - Priya Balasubramanian
- Vascular Cognitive Impairment and Neurodegeneration Program, Oklahoma Center for Geroscience and Healthy Brain Aging, Department of Biochemistry & Molecular Biology, University of Oklahoma Health Sciences Center, 975 NE 10th Street, BRC 1311, Oklahoma City, OK, 731042, USA
| | - Jordan Delfavero
- Vascular Cognitive Impairment and Neurodegeneration Program, Oklahoma Center for Geroscience and Healthy Brain Aging, Department of Biochemistry & Molecular Biology, University of Oklahoma Health Sciences Center, 975 NE 10th Street, BRC 1311, Oklahoma City, OK, 731042, USA
| | - Tamas Csipo
- Vascular Cognitive Impairment and Neurodegeneration Program, Oklahoma Center for Geroscience and Healthy Brain Aging, Department of Biochemistry & Molecular Biology, University of Oklahoma Health Sciences Center, 975 NE 10th Street, BRC 1311, Oklahoma City, OK, 731042, USA
- International Training Program in Geroscience, Doctoral School of Basic and Translational Medicine/Department of Public Health, Semmelweis University, Budapest, Hungary
| | - Andriy Yabluchanskiy
- Vascular Cognitive Impairment and Neurodegeneration Program, Oklahoma Center for Geroscience and Healthy Brain Aging, Department of Biochemistry & Molecular Biology, University of Oklahoma Health Sciences Center, 975 NE 10th Street, BRC 1311, Oklahoma City, OK, 731042, USA
- Peggy and Charles Stephenson Cancer Center, Oklahoma City, OK, 73104, USA
- Department of Health Promotion Sciences, College of Public Health, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Tamas Kiss
- Vascular Cognitive Impairment and Neurodegeneration Program, Oklahoma Center for Geroscience and Healthy Brain Aging, Department of Biochemistry & Molecular Biology, University of Oklahoma Health Sciences Center, 975 NE 10th Street, BRC 1311, Oklahoma City, OK, 731042, USA
- Department of Pediatrics, University of Szeged, Szeged, Hungary
- First Department of Pediatrics, Semmelweis University, Budapest, Hungary
| | - Ádám Nyúl-Tóth
- Vascular Cognitive Impairment and Neurodegeneration Program, Oklahoma Center for Geroscience and Healthy Brain Aging, Department of Biochemistry & Molecular Biology, University of Oklahoma Health Sciences Center, 975 NE 10th Street, BRC 1311, Oklahoma City, OK, 731042, USA
- International Training Program in Geroscience, Doctoral School of Basic and Translational Medicine/Department of Public Health, Semmelweis University, Budapest, Hungary
- International Training Program in Geroscience, Institute of Biophysics, Biological Research Centre, Eötvös Loránd Research Network (ELKH), Szeged, Hungary
| | - Peter Mukli
- Vascular Cognitive Impairment and Neurodegeneration Program, Oklahoma Center for Geroscience and Healthy Brain Aging, Department of Biochemistry & Molecular Biology, University of Oklahoma Health Sciences Center, 975 NE 10th Street, BRC 1311, Oklahoma City, OK, 731042, USA
- International Training Program in Geroscience, Doctoral School of Basic and Translational Medicine/Department of Physiology, Semmelweis University, Budapest, Hungary
| | - Peter Toth
- Vascular Cognitive Impairment and Neurodegeneration Program, Oklahoma Center for Geroscience and Healthy Brain Aging, Department of Biochemistry & Molecular Biology, University of Oklahoma Health Sciences Center, 975 NE 10th Street, BRC 1311, Oklahoma City, OK, 731042, USA
- International Training Program in Geroscience, Doctoral School of Basic and Translational Medicine/Department of Public Health, Semmelweis University, Budapest, Hungary
- Department of Neurosurgery, University of Pécs Clinical Center, Pecs, Hungary
- MTA-PTE Clinical Neuroscience MR Research Group, Pecs, Hungary
| | - Chetan Ahire
- Vascular Cognitive Impairment and Neurodegeneration Program, Oklahoma Center for Geroscience and Healthy Brain Aging, Department of Biochemistry & Molecular Biology, University of Oklahoma Health Sciences Center, 975 NE 10th Street, BRC 1311, Oklahoma City, OK, 731042, USA
| | - Anna Ungvari
- Vascular Cognitive Impairment and Neurodegeneration Program, Oklahoma Center for Geroscience and Healthy Brain Aging, Department of Biochemistry & Molecular Biology, University of Oklahoma Health Sciences Center, 975 NE 10th Street, BRC 1311, Oklahoma City, OK, 731042, USA
| | - Zoltan Benyo
- International Training Program in Geroscience/Vascular Cognitive Impairment and Neurodegeneration Program, Department of Translational Medicine, Semmelweis University, Budapest, Hungary
| | - Anna Csiszar
- Vascular Cognitive Impairment and Neurodegeneration Program, Oklahoma Center for Geroscience and Healthy Brain Aging, Department of Biochemistry & Molecular Biology, University of Oklahoma Health Sciences Center, 975 NE 10th Street, BRC 1311, Oklahoma City, OK, 731042, USA
- Peggy and Charles Stephenson Cancer Center, Oklahoma City, OK, 73104, USA
- International Training Program in Geroscience/Vascular Cognitive Impairment and Neurodegeneration Program, Department of Translational Medicine, Semmelweis University, Budapest, Hungary
| | - Zoltan Ungvari
- Vascular Cognitive Impairment and Neurodegeneration Program, Oklahoma Center for Geroscience and Healthy Brain Aging, Department of Biochemistry & Molecular Biology, University of Oklahoma Health Sciences Center, 975 NE 10th Street, BRC 1311, Oklahoma City, OK, 731042, USA
- Peggy and Charles Stephenson Cancer Center, Oklahoma City, OK, 73104, USA
- Department of Health Promotion Sciences, College of Public Health, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- International Training Program in Geroscience, Doctoral School of Basic and Translational Medicine/Department of Public Health, Semmelweis University, Budapest, Hungary
| |
Collapse
|