1
|
Hanna P, Hoover DB, Kirkland LG, Smith EH, Poston MD, Peirce SG, Garbe CG, Cha S, Mori S, Brennan JA, Armour JA, Rytkin E, Efimov IR, Ajijola OA, Ardell JL, Shivkumar K. Noradrenergic and cholinergic innervation of the normal human heart and changes associated with cardiomyopathy. Anat Rec (Hoboken) 2025. [PMID: 40365781 DOI: 10.1002/ar.25686] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2025] [Revised: 04/14/2025] [Accepted: 04/30/2025] [Indexed: 05/15/2025]
Abstract
Autonomic nerves are crucial in cardiac function and pathology. However, data on the distribution of cholinergic and noradrenergic nerves in normal and pathologic human hearts is lacking. Nonfailing donor hearts were pressure-perfusion fixed, imaged, and dissected. Left ventricular cardiomyopathy samples were also obtained. Fixed frozen sections were immunostained for nerves, and adjacent tissue underwent clearing for 3D visualization. Cholinergic and noradrenergic nerves were evenly abundant in both atria, except the sinoatrial node, where vesicular acetylcholine transporter (VAChT) nerves were dominant. Noradrenergic consistently outnumbered cholinergic nerves in right (RV) and left ventricular (LV) regions. Noradrenergic innervation of LV regions varied between donors. Cholinergic innervation was higher in RV compared to LV samples, which generally had reduced VAChT nerves. Marked neural remodeling occurred in three cardiomyopathy cases. Tyrosine hydroxylase (TH) nerve density was increased in the right atrial appendage, and all nerves showed a trend to decrease in the left atrial appendage. Cholinergic innervation was reduced in the LV, and TH innervation was heterogeneous. Noradrenergic nerves were present in granulation tissue but absent in regions of dense scar. Some border zone regions had reduced TH innervation but no hyperinnervation. Dual innervation of most atrial regions supports balanced regulation of atrial function. Higher cholinergic input to the sinoatrial node favors vagal dominance in heart rate regulation. Innervation patterns support a significant role of noradrenergic input to the ventricle, especially on the left. Both atrial and ventricular nerves remodel in cardiomyopathy, providing a foundation for asymmetric neural input and dysregulation of cardiac electromechanical function.
Collapse
Affiliation(s)
- Peter Hanna
- David Geffen School of Medicine, University of California Los Angeles (UCLA), Los Angeles, California, USA
- Molecular, Cellular, and Integrative Physiology Program, University of California Los Angeles (UCLA), Los Angeles, California, USA
| | - Donald B Hoover
- Department of Biomedical Sciences, Quillen College of Medicine, East Tennessee State University, Johnson City, Tennessee, USA
- Center of Excellence in Inflammation, Infectious Disease and Immunity, East Tennessee State University, Johnson City, Tennessee, USA
| | - Logan G Kirkland
- Department of Biomedical Sciences, Quillen College of Medicine, East Tennessee State University, Johnson City, Tennessee, USA
| | - Elizabeth H Smith
- Department of Biomedical Sciences, Quillen College of Medicine, East Tennessee State University, Johnson City, Tennessee, USA
| | - Megan D Poston
- Department of Biomedical Sciences, Quillen College of Medicine, East Tennessee State University, Johnson City, Tennessee, USA
| | - Stanley G Peirce
- Department of Biomedical Sciences, Quillen College of Medicine, East Tennessee State University, Johnson City, Tennessee, USA
| | - Chloe G Garbe
- Department of Biomedical Sciences, Quillen College of Medicine, East Tennessee State University, Johnson City, Tennessee, USA
| | - Steven Cha
- David Geffen School of Medicine, University of California Los Angeles (UCLA), Los Angeles, California, USA
| | - Shumpei Mori
- David Geffen School of Medicine, University of California Los Angeles (UCLA), Los Angeles, California, USA
| | - Jaclyn A Brennan
- Department of Biomedical Engineering, George Washington University, Washington, DC, USA
| | - John Andrew Armour
- David Geffen School of Medicine, University of California Los Angeles (UCLA), Los Angeles, California, USA
| | - Eric Rytkin
- Department of Biomedical Engineering, Northwestern University, Chicago, Illinois, USA
| | - Igor R Efimov
- Department of Biomedical Engineering, Northwestern University, Chicago, Illinois, USA
- Department of Medicine (Cardiology), Northwestern University, Chicago, Illinois, USA
| | - Olujimi A Ajijola
- David Geffen School of Medicine, University of California Los Angeles (UCLA), Los Angeles, California, USA
- Molecular, Cellular, and Integrative Physiology Program, University of California Los Angeles (UCLA), Los Angeles, California, USA
| | - Jeffrey L Ardell
- David Geffen School of Medicine, University of California Los Angeles (UCLA), Los Angeles, California, USA
| | - Kalyanam Shivkumar
- David Geffen School of Medicine, University of California Los Angeles (UCLA), Los Angeles, California, USA
- Molecular, Cellular, and Integrative Physiology Program, University of California Los Angeles (UCLA), Los Angeles, California, USA
| |
Collapse
|
2
|
Cao RHM, Diab J, Grimm MC, Berney CR. Brown bowel syndrome: a systematic review. Ann Gastroenterol 2025; 38:237-246. [PMID: 40371207 PMCID: PMC12070341 DOI: 10.20524/aog.2025.0965] [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: 12/22/2024] [Accepted: 04/10/2025] [Indexed: 05/16/2025] Open
Abstract
Brown bowel syndrome (BBS) is a rare disorder characterized by brown pigmentation of the intestinal wall, thought to be a consequence of lipofuscin accumulation. Celiac disease and vitamin E deficiency have been postulated to be risk factors. We systematically searched PubMed, Embase, Web of Science and Cochrane to identify all case reports and abstracts reporting clinical information on patients with a confirmed diagnosis of BBS. Forty-two studies met our inclusion criteria, including 63 patients with confirmed BBS. The most common symptoms of BBS were diarrhea (50.8%) and malnutrition (50.8%), followed by abdominal pain (39.7%) and vomiting (22.2%). BBS patients with celiac disease who presented with similar symptoms to non-celiac patients were significantly less likely to be hypoalbuminemic (15.4 vs. 45.5%) and showed a non-significant trend towards a higher mortality rate (36.4% vs. 15.4%). Nineteen (31.7%) BBS patients were also vitamin E deficient. The clinical presentation and outcomes in BBS patients with vitamin E deficiency and celiac disease were similar to those without vitamin E deficiency and celiac disease. Further studies are warranted to better define the diagnostic-therapeutic approach to patients with BBS.
Collapse
Affiliation(s)
- Rena HM Cao
- Royal Prince Alfred Hospital, Camperdown, NSW, Australia; University of Sydney, Faculty of Medicine (Rena HM Cao)
| | - Jason Diab
- Bankstown-Lidcombe Hospital, Bankstown, NSW, Australia (Jason Diab, Christophe R. Berney)
- University of New South Wales, Faculty of Medicine and Health, Sydney (Jason Diab, Michael C. Grimm, Christophe R. Berney)
- University of Notre Dame, School of Medicine, Sydney (Jason Diab)
| | - Michael C. Grimm
- University of New South Wales, Faculty of Medicine and Health, Sydney (Jason Diab, Michael C. Grimm, Christophe R. Berney)
- University of New South Wales, Faculty of Medicine and Health, Sydney; St George Hospital, Kogarah, NSW, Australia (Michael C. Grimm)
| | - Christophe R. Berney
- Bankstown-Lidcombe Hospital, Bankstown, NSW, Australia (Jason Diab, Christophe R. Berney)
- University of New South Wales, Faculty of Medicine and Health, Sydney (Jason Diab, Michael C. Grimm, Christophe R. Berney)
| |
Collapse
|
3
|
Walter S, Häseli SP, Baumgarten P, Deubel S, Jung T, Höhn A, Ott C, Grune T. Oxidized protein aggregate lipofuscin impairs cardiomyocyte contractility via late-stage autophagy inhibition. Redox Biol 2025; 81:103559. [PMID: 40068328 PMCID: PMC11938141 DOI: 10.1016/j.redox.2025.103559] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2024] [Revised: 02/10/2025] [Accepted: 02/18/2025] [Indexed: 03/22/2025] Open
Abstract
Aging of the heart is accompanied by impairment of cardiac structure and function. At molecular level, autophagy plays a crucial role in preserving cardiac health. Autophagy maintains cellular homeostasis by facilitating balanced degradation of cytoplasmic components including organelles and misfolded or aggregated proteins. The age-related decline in autophagy favors an accumulation of protein aggregates such as lipofuscin particularly in the heart, which is composed primarily of non-proliferating cells. Therefore, this study investigates whether lipofuscin accumulation contributes to age-related functional decline of primary adult cardiomyocytes isolated from C57BL/6J mice and examines the role of autophagic flux in mediating these effects. Results showed an age-associated reduction in cardiomyocyte contraction amplitude and an increase in autofluorescence, indicating the accumulation of lipofuscin with age. In vitro treatment of adult primary cardiomyocytes with artificial lipofuscin increased autofluorescence and decreased both contraction amplitude and cellular autophagic flux. Induction of autophagy with rapamycin mitigated contractile dysfunction in lipofuscin-treated cardiomyocytes, whereas inhibition of autophagic flux revealed stage-dependent effects. Late-stage autophagy inhibition using chloroquine or concanamycin A reduced cardiomyocyte contraction amplitude, whereas early-stage autophagy inhibition via 3-methyladenine did not affect contraction within 24 h. In conclusion, our results indicate that lipofuscin directly impairs cardiomyocyte function by diminishing late-stage autophagic flux. These findings highlight the essential role of the autophagy-lysosomal system in preserving age-related loss of cardiomyocyte function caused by accumulating protein aggregates.
Collapse
Affiliation(s)
- Sophia Walter
- Molecular Toxicology, German Institute of Human Nutrition Potsdam-Rehbruecke (DIfE), Nuthetal, Germany; TraceAge-DFG Research Unit on Interactions of Essential Trace Elements in Healthy and Diseased Elderly, Potsdam-Berlin-Jena-Wuppertal, Germany; DZHK (German Center for Cardiovascular Research), Partner Site Berlin, Berlin, Germany
| | - Steffen P Häseli
- Molecular Toxicology, German Institute of Human Nutrition Potsdam-Rehbruecke (DIfE), Nuthetal, Germany; DZHK (German Center for Cardiovascular Research), Partner Site Berlin, Berlin, Germany
| | - Patricia Baumgarten
- Molecular Toxicology, German Institute of Human Nutrition Potsdam-Rehbruecke (DIfE), Nuthetal, Germany; DZHK (German Center for Cardiovascular Research), Partner Site Berlin, Berlin, Germany
| | - Stefanie Deubel
- Molecular Toxicology, German Institute of Human Nutrition Potsdam-Rehbruecke (DIfE), Nuthetal, Germany
| | - Tobias Jung
- Molecular Toxicology, German Institute of Human Nutrition Potsdam-Rehbruecke (DIfE), Nuthetal, Germany
| | - Annika Höhn
- Molecular Toxicology, German Institute of Human Nutrition Potsdam-Rehbruecke (DIfE), Nuthetal, Germany; German Center for Diabetes Research (DZD), München-Neuherberg, Germany
| | - Christiane Ott
- Molecular Toxicology, German Institute of Human Nutrition Potsdam-Rehbruecke (DIfE), Nuthetal, Germany; TraceAge-DFG Research Unit on Interactions of Essential Trace Elements in Healthy and Diseased Elderly, Potsdam-Berlin-Jena-Wuppertal, Germany; DZHK (German Center for Cardiovascular Research), Partner Site Berlin, Berlin, Germany; Cardiac Aging and Nutrition, German Institute of Human Nutrition Potsdam-Rehbruecke (DIfE), Nuthetal, Germany.
| | - Tilman Grune
- Molecular Toxicology, German Institute of Human Nutrition Potsdam-Rehbruecke (DIfE), Nuthetal, Germany; TraceAge-DFG Research Unit on Interactions of Essential Trace Elements in Healthy and Diseased Elderly, Potsdam-Berlin-Jena-Wuppertal, Germany; DZHK (German Center for Cardiovascular Research), Partner Site Berlin, Berlin, Germany; Institute of Nutritional Science, Department of Food Chemistry, University of Potsdam, Potsdam, 14469, Germany; German Center for Diabetes Research (DZD), München-Neuherberg, Germany
| |
Collapse
|
4
|
Guo J, Bullock G, O’Brien DP, Johnson GS, Katz ML. An RB1CC1 Missense Variant in Nova Scotia Duck Tolling Retrievers with Degenerative Encephalopathy. Genes (Basel) 2025; 16:269. [PMID: 40149422 PMCID: PMC11941761 DOI: 10.3390/genes16030269] [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] [Subscribe] [Scholar Register] [Received: 02/01/2025] [Revised: 02/18/2025] [Accepted: 02/19/2025] [Indexed: 03/29/2025] Open
Abstract
BACKGROUND/OBJECTIVES A slowly progressive hereditary neurological disorder classified as degenerative encephalopathy (DE) occurs in Nova Scotia Duck Tolling Retrievers. The disease is characterized by frequent episodes of pronounced involuntary movements during sleep, cognitive impairment, anxiety, heightened sensitivity to sensory stimuli, and compulsive behaviors. The clinical signs are accompanied by the degeneration of several brain regions. A study was undertaken to identify the molecular genetic basis of this disorder. METHODS Whole genome sequences (WGSs) from the DNA of affected and unaffected Nova Scotia Duck Tolling Retrievers were aligned to the Dog10K_Boxer_Tasha reference genome assembly and to the WGSs of 334 additional control dogs generated by this laboratory. RESULTS A missense C>T variant was identified in RB1CC1 exon 22 chromosome 29:4891014 that was uniquely homozygous in the affected dog. This variant predicts a p.G1503R change in the amino acid sequence of RB1CC1. Genotyping of 2950 Nova Scotia Duck Tolling Retrievers at the variant locus found complete concordance between the disease phenotype and RB1CC1 genotype. CONCLUSIONS RBCC1 is an essential component of a protein complex that mediates the initiation of autophagosome formation. Therefore, it appears likely that the disease results, at least in part, from impaired autophagy. Consistent with this possibility, brain neurons of an affected dog were found to contain abnormal lysosomal storage body-like inclusions. This disorder could serve as a valuable model to elucidate the mechanisms underlying human diseases associated with impaired autophagy. Identification of the disease-causing DNA sequence variant will enable owners of Nova Scotia Duck Tolling Retrievers to screen their dogs for the RB1CC1 risk variant.
Collapse
Affiliation(s)
- Juyuan Guo
- Canine Genetics Laboratory, Department of Veterinary Pathobiology, College of Veterinary Medicine, University of Missouri, Columbia, MO 65211, USA; (J.G.); (G.B.)
| | - Garrett Bullock
- Canine Genetics Laboratory, Department of Veterinary Pathobiology, College of Veterinary Medicine, University of Missouri, Columbia, MO 65211, USA; (J.G.); (G.B.)
| | - Dennis P. O’Brien
- Department of Veterinary Medicine and Surgery, College of Veterinary Medicine, University of Missouri, Columbia, MO 65211, USA; (D.P.O.)
| | - Gary S. Johnson
- Department of Veterinary Medicine and Surgery, College of Veterinary Medicine, University of Missouri, Columbia, MO 65211, USA; (D.P.O.)
| | - Martin L. Katz
- Canine Genetics Laboratory, Department of Veterinary Pathobiology, College of Veterinary Medicine, University of Missouri, Columbia, MO 65211, USA; (J.G.); (G.B.)
- Neurodegenerative Diseases Research Laboratory, Department of Ophthalmology, University of Missouri, Columbia, MO 65212, USA
| |
Collapse
|
5
|
Tan Yi Shean L, Milne EM, Shaw DJ, Maxwell S, Del-Pozo J. Lipofuscin accumulates in ganglionic neurons in chronic equine dysautonomia. J Vet Diagn Invest 2024; 36:864-869. [PMID: 39113499 PMCID: PMC11529066 DOI: 10.1177/10406387241265715] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2024] Open
Abstract
Lipofuscin is a complex mixture of highly oxidized, cross-linked macromolecules that accumulates in neurons with age and some neurodegenerative diseases. Equine dysautonomia (ED) is a polyneuropathy that mainly affects autonomic and enteric nervous systems, resulting in alimentary tract dysfunction. Our main aim was to determine whether neuronal lipofuscin increased with increasing duration of ED. We investigated the prevalence of lipofuscin in cranial cervical ganglia of horses with acute (AED), subacute (SED), and chronic ED (CED), young controls (of similar age to ED cases), and aged controls (n = 8 per group). We used Schmorl stain for histologic detection of lipofuscin and assessed its accumulation in neurons using image analysis software. The percentage of neurons positive for lipofuscin increased with age in individual groups and all groups combined (p < 0.001). There were fewer positive neurons in AED and SED compared to aged controls (p < 0.001) and more in CED than AED cases (p = 0.042) and young controls (p = 0.012). We found a strong positive correlation between percentage positive neurons and percentage positive area of the neuron containing lipofuscin for combined groups (p < 0.001). Although neuronal lipofuscin increased in cranial cervical ganglion in CED cases, it remains to be determined whether this is a cause or consequence of neuronal degeneration.
Collapse
Affiliation(s)
- Lydia Tan Yi Shean
- Royal (Dick) School of Veterinary Studies, The University of Edinburgh, Easter Bush Campus, Roslin, Midlothian, UK
| | - Elspeth M. Milne
- Royal (Dick) School of Veterinary Studies, The University of Edinburgh, Easter Bush Campus, Roslin, Midlothian, UK
| | - Darren J. Shaw
- Royal (Dick) School of Veterinary Studies, The University of Edinburgh, Easter Bush Campus, Roslin, Midlothian, UK
| | - Scott Maxwell
- Royal (Dick) School of Veterinary Studies, The University of Edinburgh, Easter Bush Campus, Roslin, Midlothian, UK
| | - Jorge Del-Pozo
- Royal (Dick) School of Veterinary Studies, The University of Edinburgh, Easter Bush Campus, Roslin, Midlothian, UK
| |
Collapse
|
6
|
Tao P, Zhang HF, Zhou P, Wang YL, Tan YZ, Wang HJ. Growth differentiation factor 11 alleviates oxidative stress-induced senescence of endothelial progenitor cells via activating autophagy. Stem Cell Res Ther 2024; 15:370. [PMID: 39420391 PMCID: PMC11488219 DOI: 10.1186/s13287-024-03975-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Accepted: 10/05/2024] [Indexed: 10/19/2024] Open
Abstract
BACKGROUND Stem cell transplantation has been regarded as a promising therapeutic strategy for myocardial regeneration after myocardial infarction (MI). However, the survival and differentiation of the transplanted stem cells in the hostile ischaemic and inflammatory microenvironment are poor. Recent studies have focused on enhancing the survival and differentiation of the stem cells, while strategies to suppress the senescence of the transplanted stem cells is unknown. Therefore, we investigated the effect of growth differentiation factor 11 (GDF11) on attenuating oxidative stress-induced senescence in the engrafted endothelial progenitor cells (EPCs). METHODS Rat models of oxidative stress were established by hydrogen peroxide conditioning. Oxidative stress-induced senescence was assessed through senescence-associated β-galactosidase expression and lipofuscin accumulation. The effects of GDF11 treatment on senescence and autophagy of EPCs were evaluated 345, while improvement of myocardial regeneration, neovascularization and cardiac function were examined following transplantation of the self-assembling peptide (SAP) loaded EPCs and GDF11 in the rat MI models. RESULTS Following hydrogen peroxide conditioning, the level of ROS in EPCs decreased significantly upon treatment with GDF11. This resulted in reduction in the senescent cells and lipofuscin particles, as well as the damaged mitochondria and rough endoplasmic reticula. Concurrently, there was a significant increase in LC3-II expression, LC3-positive puncta and the presence of autophagic ultrastructures were increased significantly. The formulated SAP effectively adhered to EPCs and sustained the release of GDF11. Transplantation of SAP-loaded EPCs and GDF11 into the ischaemic abdominal pouch or myocardium resulted in a decreased number of the senescent EPCs. At four weeks after transplantation into the myocardium, neovascularization and myocardial regeneration were enhanced, reverse myocardial remodeling was attenuated, and cardiac function was improved effectively. CONCLUSIONS This study provides novel evidence suggesting that oxidative stress could induce senescence of the transplanted EPCs in the ischemic myocardium. GDF11 demonstrates the ability to mitigate oxidative stress-induced senescence in the transplanted EPCs within the myocardium by activating autophagy.
Collapse
Affiliation(s)
- Ping Tao
- Department of Anatomy, Histology and Embryology, Shanghai Medical School of Fudan University, 138 Yixueyuan Road, Shanghai, 200032, People's Republic of China
- Department of Laboratory Medicine, Shanghai Traditional Chinese Medicine-Integrated Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200086, People's Republic of China
| | - Hai-Feng Zhang
- Department of Anatomy, Histology and Embryology, Shanghai Medical School of Fudan University, 138 Yixueyuan Road, Shanghai, 200032, People's Republic of China
| | - Pei Zhou
- Department of Anatomy, Histology and Embryology, Shanghai Medical School of Fudan University, 138 Yixueyuan Road, Shanghai, 200032, People's Republic of China
| | - Yong-Li Wang
- Department of Anatomy, Histology and Embryology, Shanghai Medical School of Fudan University, 138 Yixueyuan Road, Shanghai, 200032, People's Republic of China
| | - Yu-Zhen Tan
- Department of Anatomy, Histology and Embryology, Shanghai Medical School of Fudan University, 138 Yixueyuan Road, Shanghai, 200032, People's Republic of China.
- Rehabilitation Therapy Department, School of Health Sciences, West Yunnan University of Applied Sciences, Dali, Yunnan Province, 671000, People's Republic of China.
| | - Hai-Jie Wang
- Department of Anatomy, Histology and Embryology, Shanghai Medical School of Fudan University, 138 Yixueyuan Road, Shanghai, 200032, People's Republic of China.
- Rehabilitation Therapy Department, School of Health Sciences, West Yunnan University of Applied Sciences, Dali, Yunnan Province, 671000, People's Republic of China.
| |
Collapse
|
7
|
Ogrodnik M, Carlos Acosta J, Adams PD, d'Adda di Fagagna F, Baker DJ, Bishop CL, Chandra T, Collado M, Gil J, Gorgoulis V, Gruber F, Hara E, Jansen-Dürr P, Jurk D, Khosla S, Kirkland JL, Krizhanovsky V, Minamino T, Niedernhofer LJ, Passos JF, Ring NAR, Redl H, Robbins PD, Rodier F, Scharffetter-Kochanek K, Sedivy JM, Sikora E, Witwer K, von Zglinicki T, Yun MH, Grillari J, Demaria M. Guidelines for minimal information on cellular senescence experimentation in vivo. Cell 2024; 187:4150-4175. [PMID: 39121846 PMCID: PMC11790242 DOI: 10.1016/j.cell.2024.05.059] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Revised: 05/27/2024] [Accepted: 05/31/2024] [Indexed: 08/12/2024]
Abstract
Cellular senescence is a cell fate triggered in response to stress and is characterized by stable cell-cycle arrest and a hypersecretory state. It has diverse biological roles, ranging from tissue repair to chronic disease. The development of new tools to study senescence in vivo has paved the way for uncovering its physiological and pathological roles and testing senescent cells as a therapeutic target. However, the lack of specific and broadly applicable markers makes it difficult to identify and characterize senescent cells in tissues and living organisms. To address this, we provide practical guidelines called "minimum information for cellular senescence experimentation in vivo" (MICSE). It presents an overview of senescence markers in rodent tissues, transgenic models, non-mammalian systems, human tissues, and tumors and their use in the identification and specification of senescent cells. These guidelines provide a uniform, state-of-the-art, and accessible toolset to improve our understanding of cellular senescence in vivo.
Collapse
Affiliation(s)
- Mikolaj Ogrodnik
- Ludwig Boltzmann Research Group Senescence and Healing of Wounds, 1200 Vienna, Austria; Ludwig Boltzmann Institute for Traumatology, The Research Centre in Cooperation with AUVA, 1200 Vienna, Austria; Austrian Cluster for Tissue Regeneration, Vienna, Austria.
| | - Juan Carlos Acosta
- Cancer Research UK Scotland Centre, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh EH4 2XR, UK; Instituto de Biomedicina y Biotecnología de Cantabria, IBBTEC (CSIC, Universidad de Cantabria), C/ Albert Einstein 22, 39011 Santander, Spain
| | - Peter D Adams
- Cancer Genome and Epigenetics Program, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA
| | - Fabrizio d'Adda di Fagagna
- IFOM ETS - The AIRC Institute of Molecular Oncology, Milan, Italy; Institute of Molecular Genetics IGM-CNR "Luigi Luca Cavalli-Sforza," Pavia, Italy
| | - Darren J Baker
- Department of Biochemistry and Molecular Biology, Department of Pediatric and Adolescent Medicine, Robert and Arlene Kogod Center on Aging, Mayo Clinic, 200 First Steet SW, Rochester, MN 55905, USA
| | - Cleo L Bishop
- Blizard Institute, Barts and The London Faculty of Medicine and Dentistry, Queen Mary University of London, 4 Newark Street, London E1 2AT, UK
| | - Tamir Chandra
- MRC Human Generics Unit, University of Edinburgh, Edinburgh, UK
| | - Manuel Collado
- Health Research Institute of Santiago de Compostela (IDIS), Santiago de Compostela, Spain; Department of Immunology and Oncology (DIO), Centro Nacional de Biotecnología (CNB-CSIC), Madrid, Spain
| | - Jesus Gil
- MRC Laboratory of Medical Sciences (LMS), Du Cane Road, London W12 0NN, UK; Institute of Clinical Sciences (ICS), Faculty of Medicine, Imperial College London, Du Cane Road, London W12 0NN, UK
| | - Vassilis Gorgoulis
- Molecular Carcinogenesis Group, Department of Histology and Embryology, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece; Biomedical Research Foundation, Academy of Athens, 11527 Athens, Greece; Ninewells Hospital and Medical School, University of Dundee, Dundee DD19SY, UK; Faculty Institute for Cancer Sciences, Manchester Academic Health Sciences Centre, University of Manchester, Manchester M20 4GJ, UK; Faculty of Health and Medical Sciences, University of Surrey, Surrey GU2 7YH, UK
| | - Florian Gruber
- Department of Dermatology, Medical University of Vienna, Vienna, Austria; Christian Doppler Laboratory for Skin Multimodal Imaging of Aging and Senescence - SKINMAGINE, Vienna, Austria
| | - Eiji Hara
- Research Institute for Microbial Diseases (RIMD), Osaka University, Suita 565-0871, Japan; Immunology Frontier Research Center (IFReC), Osaka University, Suita 565-0871, Japan
| | - Pidder Jansen-Dürr
- Institute for Biomedical Aging Research, University of Innsbruck, and Center for Molecular Biosciences Innsbruck (CMBI), Innsbruck, Austria
| | - Diana Jurk
- Mayo Clinic, Department of Physiology and Biomedical Engineering, Robert and Arlene Kogod Center on Aging, Rochester, MN, USA
| | - Sundeep Khosla
- Kogod Center on Aging and Division of Endocrinology, Mayo Clinic, Rochester, MN, USA
| | - James L Kirkland
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, USA; Division of General Internal Medicine, Department of Medicine, Mayo Clinic, Rochester, MN, USA
| | - Valery Krizhanovsky
- Department of Molecular Cell Biology, The Weizmann Institute of Science, 7610001 Rehovot, Israel
| | - Tohru Minamino
- Department of Cardiovascular Biology and Medicine, Juntendo University Graduate School of Medicine, Tokyo 113-8421, Japan; Japan Agency for Medical Research and Development-Core Research for Evolutionary Medical Science and Technology (AMED-CREST), Japan Agency for Medical Research and Development, Tokyo 100-0004, Japan
| | - Laura J Niedernhofer
- Institute on the Biology of Aging and Metabolism, Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, 6-155 Jackson Hall, 321 Church Street, SE, Minneapolis, MN 55455, USA
| | - João F Passos
- Mayo Clinic, Department of Physiology and Biomedical Engineering, Robert and Arlene Kogod Center on Aging, Rochester, MN, USA
| | - Nadja A R Ring
- Ludwig Boltzmann Research Group Senescence and Healing of Wounds, 1200 Vienna, Austria; Ludwig Boltzmann Institute for Traumatology, The Research Centre in Cooperation with AUVA, 1200 Vienna, Austria; Austrian Cluster for Tissue Regeneration, Vienna, Austria
| | - Heinz Redl
- Ludwig Boltzmann Research Group Senescence and Healing of Wounds, 1200 Vienna, Austria; Ludwig Boltzmann Institute for Traumatology, The Research Centre in Cooperation with AUVA, 1200 Vienna, Austria; Austrian Cluster for Tissue Regeneration, Vienna, Austria
| | - Paul D Robbins
- Institute on the Biology of Aging and Metabolism, Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, 6-155 Jackson Hall, 321 Church Street, SE, Minneapolis, MN 55455, USA
| | - Francis Rodier
- Centre de recherche du Centre hospitalier de l'Université de Montréal (CRCHUM), Montreal, QC, Canada; Institut du cancer de Montréal, Montreal, QC, Canada; Université de Montréal, Département de radiologie, radio-oncologie et médicine nucléaire, Montreal, QC, Canada
| | - Karin Scharffetter-Kochanek
- Department f Dermatology and Allergic Diseases, Ulm University Hospital, Albert-Einstein-Allee 23, 89081 Ulm, Germany
| | - John M Sedivy
- Department of Molecular, Cellular Biology and Biochemistry, Center on the Biology of Aging, Brown University, Providence, RI, USA
| | - Ewa Sikora
- Laboratory of Molecular Bases of Aging, Nencki Institute of Experimental Biology, Polish Academy of Sciences, 02-093 Warsaw, Poland
| | - Kenneth Witwer
- The Johns Hopkins University School of Medicine, Department of Molecular and Comparative Pathobiology, Baltimore, MD, USA; The Johns Hopkins University School of Medicine, Department of Neurology, Baltimore, MD, USA
| | - Thomas von Zglinicki
- Newcastle University Biosciences Institute, Ageing Biology Laboratories, Newcastle upon Tyne, UK
| | - Maximina H Yun
- Technische Universität Dresden, CRTD/Center for Regenerative Therapies Dresden, Dresden, Germany; Max Planck Institute of Molecular Cellular Biology and Genetics, Dresden, Germany; Physics of Life Excellence Cluster, Dresden, Germany
| | - Johannes Grillari
- Ludwig Boltzmann Institute for Traumatology, The Research Centre in Cooperation with AUVA, 1200 Vienna, Austria; Austrian Cluster for Tissue Regeneration, Vienna, Austria; Institute of Molecular Biotechnology, BOKU University, Vienna, Austria.
| | - Marco Demaria
- European Research Institute for the Biology of Ageing (ERIBA), University Medical Center Groningen (UMCG), University of Groningen (RUG), Groningen, the Netherlands.
| |
Collapse
|
8
|
Smyła-Gruca W, Szczurek-Wasilewicz W, Skrzypek M, Romuk E, Karmański A, Jurkiewicz M, Gąsior M, Osadnik T, Banach M, Jóźwiak JJ, Szyguła-Jurkiewicz B. Ceruloplasmin and Lipofuscin Serum Concentrations Are Associated with Presence of Hypertrophic Cardiomyopathy. Biomedicines 2024; 12:1767. [PMID: 39200231 PMCID: PMC11352126 DOI: 10.3390/biomedicines12081767] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Revised: 07/28/2024] [Accepted: 08/03/2024] [Indexed: 09/02/2024] Open
Abstract
Oxidative stress reflects an imbalance between the systemic manifestation of reactive oxygen species and cells' ability to neutralize them by antioxidant systems. The role of oxidative stress in hypertrophic cardiomyopathy (HCM) is not fully understood. The aim of the study was to examine selected parameters of oxidative stress in patients with HCM compared to the control group. We enrolled 85 consecutive HCM patients and 97 controls without HCM. The groups were matched for sex, the body mass index, and age. Oxidative stress markers included superoxide dismutase (SOD), ceruloplasmin (CER), and lipofuscin (LPS). The median age of the HCM patients was 53 (40-63) years, and 41.2% of them were male. HCM patients, compared to the control ones, had significantly increased levels of CER and LPS. The areas under the receiver operating characteristics curves (AUC) indicated a good discriminatory power of CER (AUC 0.924, sensitivity 84%, and specificity 88%), an acceptable discriminatory power of LPS (AUC 0.740, sensitivity 66%, and specificity 72%), and poor discriminatory power of SOD (AUC 0.556, sensitivity 34%, and specificity 94%) for HCM detection. CER with good predictive strength, as well as LPS with acceptable predictive power, allows for HCM detection. The utility of SOD for HCM detection is limited.
Collapse
Affiliation(s)
- Wiktoria Smyła-Gruca
- Student’s Scientific Society, 3rd Department of Cardiology, Faculty of Medical Sciences in Zabrze, Medical University of Silesia, 40-055 Katowice, Poland; (W.S.-G.); (M.J.)
| | | | - Michał Skrzypek
- Department of Biostatistics, Faculty of Public Health in Bytom, Medical University of Silesia in Katowice, 40-055 Katowice, Poland;
| | - Ewa Romuk
- Department of Biochemistry, Faculty of Medical Sciences in Zabrze, Medical University of Silesia, 40-055 Katowice, Poland;
| | - Andrzej Karmański
- Department of Descriptive and Topographic Anatomy, Faculty of Medical Sciences in Zabrze, Medical University of Silesia, 40-055 Katowice, Poland;
| | - Michał Jurkiewicz
- Student’s Scientific Society, 3rd Department of Cardiology, Faculty of Medical Sciences in Zabrze, Medical University of Silesia, 40-055 Katowice, Poland; (W.S.-G.); (M.J.)
| | - Mariusz Gąsior
- 3rd Department of Cardiology, Faculty of Medical Sciences in Zabrze, Medical University of Silesia, 40-055 Katowice, Poland; (M.G.); (B.S.-J.)
| | - Tadeusz Osadnik
- Department of Pharmacology, Faculty of Medical Sciences in Zabrze, Medical University of Silesia, 40-055 Katowice, Poland;
- Cardiology and Lipid Disorders Clinic, Independent Public Health Care Institution “REPTY” Upper Silesian Rehabilitation Centre, 42-600 Tarnowskie Góry, Poland
| | - Maciej Banach
- Polish Mothers Memorial Hospital Research Institute, 90-419 Łódź, Poland;
- Department of Hypertension, Medical University of Lodz, 90-419 Łódź, Poland
- Cardiovascular Research Centre, University of Zielona Gora, 65-046 Zielona Gora, Poland
| | - Jacek J. Jóźwiak
- Department of Family Medicine and Public Health, Faculty of Medicine, University of Opole, 45-040 Opole, Poland;
| | - Bożena Szyguła-Jurkiewicz
- 3rd Department of Cardiology, Faculty of Medical Sciences in Zabrze, Medical University of Silesia, 40-055 Katowice, Poland; (M.G.); (B.S.-J.)
| |
Collapse
|
9
|
Ford VJ, Applefeld WN, Wang J, Sun J, Solomon SB, Sidenko S, Feng J, Sheffield C, Klein HG, Yu Z, Torabi‐Parizi P, Danner RL, Sachdev V, Solomon MA, Chen MY, Natanson C. Cardiac Magnetic Resonance Studies in a Large Animal Model That Simulates the Cardiac Abnormalities of Human Septic Shock. J Am Heart Assoc 2024; 13:e034026. [PMID: 39101510 PMCID: PMC11964030 DOI: 10.1161/jaha.123.034026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Accepted: 04/30/2024] [Indexed: 08/06/2024]
Abstract
BACKGROUND Septic shock is associated with increases in end-diastolic volume (EDV) and decreases in ejection fraction that reverse within 10 days. Nonsurvivors do not develop EDV increases. The mechanism is unknown. METHODS AND RESULTS Purpose-bred beagles (n=33) were randomized to receive intrabronchial Staphylococcus aureus or saline. Over 96 hours, cardiac magnetic resonance imaging and echocardiograms were performed. Tissue was obtained at 66 hours. From 0 to 96 hours after bacterial challenge, septic animals versus controls had significantly increased left ventricular wall edema (6%) and wall thinning with loss of mass (15%). On histology, the major finding was nonocclusive microvascular injury with edema in myocytes, the interstitium, and endothelial cells. Edema was associated with significant worsening of biventricular ejection fractions, ventricular-arterial coupling, and circumferential strain. Early during sepsis, (0-24 hours), the EDV decreased; significantly more in nonsurvivors (ie, greater diastolic dysfunction). From 24 to 48 hours, septic animals' biventricular chamber sizes increased; in survivors significantly greater than baseline and nonsurvivors, whose EDVs were not different from baseline. Preload, afterload, or heart rate differences did not explain these differential changes. CONCLUSIONS The cardiac dysfunction of sepsis is associated with wall edema. In nonsurvivors, at 0 to 24 hours, sepsis induces a more severe diastolic dysfunction, further decreasing chamber size. The loss of left ventricular mass with wall thinning in septic survivors may, in part, explain the EDV increases from 24 to 48 hours because of a potentially reparative process removing damaged wall tissue. Septic cardiomyopathy is most consistent with a nonocclusive microvascular injury resulting in edema causing reversible systolic and diastolic dysfunction with more severe diastolic dysfunction being associated with a decreased EDV and death.
Collapse
MESH Headings
- Animals
- Dogs
- Disease Models, Animal
- Shock, Septic/physiopathology
- Shock, Septic/complications
- Stroke Volume
- Magnetic Resonance Imaging
- Edema, Cardiac/physiopathology
- Edema, Cardiac/pathology
- Edema, Cardiac/diagnostic imaging
- Ventricular Function, Left
- Time Factors
- Humans
- Staphylococcal Infections/complications
- Staphylococcal Infections/physiopathology
- Echocardiography
- Ventricular Dysfunction, Left/physiopathology
- Ventricular Dysfunction, Left/diagnostic imaging
- Ventricular Dysfunction, Left/etiology
- Male
Collapse
Affiliation(s)
- Verity J. Ford
- Critical Care Medicine Department, Clinical CenterNational Institutes of Health, (NIH, CC)BethesdaMDUSA
| | - Willard N. Applefeld
- Critical Care Medicine Department, Clinical CenterNational Institutes of Health, (NIH, CC)BethesdaMDUSA
- Division of CardiologyDuke University Medical CenterDurhamNCUSA
| | - Jeffrey Wang
- Critical Care Medicine Department, Clinical CenterNational Institutes of Health, (NIH, CC)BethesdaMDUSA
- Emory UniversityAtlantaGAUSA
| | - Junfeng Sun
- Critical Care Medicine Department, Clinical CenterNational Institutes of Health, (NIH, CC)BethesdaMDUSA
| | - Steven B. Solomon
- Critical Care Medicine Department, Clinical CenterNational Institutes of Health, (NIH, CC)BethesdaMDUSA
| | - Stanislav Sidenko
- National Heart Lung and Blood InstituteNational Institutes of HealthBethesdaMDUSA
| | - Jing Feng
- Critical Care Medicine Department, Clinical CenterNational Institutes of Health, (NIH, CC)BethesdaMDUSA
| | | | - Harvey G. Klein
- Department of Transfusion Medicine, Clinical CenterNational Institutes of Health, (NIH, CC)BethesdaMDUSA
| | - Zu‐Xi Yu
- National Heart Lung and Blood InstituteNational Institutes of HealthBethesdaMDUSA
| | | | - Robert L. Danner
- Critical Care Medicine Department, Clinical CenterNational Institutes of Health, (NIH, CC)BethesdaMDUSA
- National Heart Lung and Blood InstituteNational Institutes of HealthBethesdaMDUSA
| | - Vandana Sachdev
- National Heart Lung and Blood InstituteNational Institutes of HealthBethesdaMDUSA
| | - Michael A. Solomon
- Critical Care Medicine Department, Clinical CenterNational Institutes of Health, (NIH, CC)BethesdaMDUSA
- National Heart Lung and Blood InstituteNational Institutes of HealthBethesdaMDUSA
| | - Marcus Y. Chen
- National Heart Lung and Blood InstituteNational Institutes of HealthBethesdaMDUSA
| | - Charles Natanson
- Critical Care Medicine Department, Clinical CenterNational Institutes of Health, (NIH, CC)BethesdaMDUSA
- National Heart Lung and Blood InstituteNational Institutes of HealthBethesdaMDUSA
| |
Collapse
|
10
|
Yang L, Liu D, Jiang S, Li H, Chen L, Wu Y, Essien AE, Opoku M, Naranmandakh S, Liu S, Ru Q, Li Y. SIRT1 signaling pathways in sarcopenia: Novel mechanisms and potential therapeutic targets. Biomed Pharmacother 2024; 177:116917. [PMID: 38908209 DOI: 10.1016/j.biopha.2024.116917] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Revised: 05/29/2024] [Accepted: 06/09/2024] [Indexed: 06/24/2024] Open
Abstract
Sarcopenia is an aging-related skeletal disease characterized by decreased muscle mass, strength, and physical function, severely affecting the quality of life (QoL) of the elderly population. Sirtuin 1 (SIRT1), as a nicotinamide adenine dinucleotide (NAD+)-dependent histone deacetylases, has been reported to participate in various aging-related signaling pathways and exert protective effect on many human diseases. SIRT1 functioned as an important role in the occurrence and progression of sarcopenia through regulating key pathways related to protein homeostasis, apoptosis, mitochondrial dysfunction, insulin resistance and autophagy in skeletal muscle, including SIRT1/Forkhead Box O (FoxO), AMP-activated protein kinase (AMPK)/SIRT1/nuclear factor κB (NF-κB), SIRT1/p53, AMPK/SIRT1/peroxisome proliferator-activated receptor gamma coactivator-1α (PGC-1α), and SIRT1/live kinase B1 (LKB1)/AMPK pathways. However, the specific mechanisms of these processes have not been fully illuminated. Currently, several SIRT1-mediated interventions on sarcopenia have been preliminarily developed, such as SIRT1 activator polyphenolic compounds, exercising and calorie restriction. In this review, we summarized the predominant mechanisms of SIRT1 involved in sarcopenia and therapeutic modalities targeting the SIRT1 signaling pathways for the prevention and prognosis of sarcopenia.
Collapse
Affiliation(s)
- Luning Yang
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China; Xiangya School of Medicine, Central South University, Changsha, Hunan 410013, China
| | - Di Liu
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| | - Shide Jiang
- Department of Orthopedics, The Central Hospital of Yongzhou, Yongzhou 425000, China
| | - Hengzhen Li
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| | - Lin Chen
- Department of Health and Physical Education, Jianghan University, Wuhan 430056, China
| | - Yuxiang Wu
- Department of Health and Physical Education, Jianghan University, Wuhan 430056, China
| | - Anko Elijah Essien
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| | - Michael Opoku
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| | - Shinen Naranmandakh
- Department of chemistry, School of Arts and Sciences, National University of Mongolia, Ulaanbaatar 14201, Mongolia
| | - ShuGuang Liu
- Department of Joint Surgery, Honghui Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi, China.
| | - Qin Ru
- Department of Health and Physical Education, Jianghan University, Wuhan 430056, China.
| | - Yusheng Li
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China.
| |
Collapse
|
11
|
Alibhai FJ, Li RK. Rejuvenation of the Aging Heart: Molecular Determinants and Applications. Can J Cardiol 2024; 40:1394-1411. [PMID: 38460612 DOI: 10.1016/j.cjca.2024.03.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2023] [Revised: 02/20/2024] [Accepted: 03/04/2024] [Indexed: 03/11/2024] Open
Abstract
In Canada and worldwide, the elderly population (ie, individuals > 65 years of age) is increasing disproportionately relative to the total population. This is expected to have a substantial impact on the health care system, as increased aged is associated with a greater incidence of chronic noncommunicable diseases. Within the elderly population, cardiovascular disease is a leading cause of death, therefore developing therapies that can prevent or slow disease progression in this group is highly desirable. Historically, aging research has focused on the development of anti-aging therapies that are implemented early in life and slow the age-dependent decline in cell and organ function. However, accumulating evidence supports that late-in-life therapies can also benefit the aged cardiovascular system by limiting age-dependent functional decline. Moreover, recent studies have demonstrated that rejuvenation (ie, reverting cellular function to that of a younger phenotype) of the already aged cardiovascular system is possible, opening new avenues to develop therapies for older individuals. In this review, we first provide an overview of the functional changes that occur in the cardiomyocyte with aging and how this contributes to the age-dependent decline in heart function. We then discuss the various anti-aging and rejuvenation strategies that have been pursued to improve the function of the aged cardiomyocyte, with a focus on therapies implemented late in life. These strategies include 1) established systemic approaches (caloric restriction, exercise), 2) pharmacologic approaches (mTOR, AMPK, SIRT1, and autophagy-targeting molecules), and 3) emerging rejuvenation approaches (partial reprogramming, parabiosis/modulation of circulating factors, targeting endogenous stem cell populations, and senotherapeutics). Collectively, these studies demonstrate the exciting potential and limitations of current rejuvenation strategies and highlight future areas of investigation that will contribute to the development of rejuvenation therapies for the aged heart.
Collapse
Affiliation(s)
- Faisal J Alibhai
- Toronto General Research Hospital Institute, University Health Network, Toronto, Ontario, Canada
| | - Ren-Ke Li
- Toronto General Research Hospital Institute, University Health Network, Toronto, Ontario, Canada; Department of Surgery, Division of Cardiovascular Surgery, University of Toronto, Toronto, Ontario, Canada.
| |
Collapse
|
12
|
Ma C, Liu Y, Fu Z. Implications of endoplasmic reticulum stress and autophagy in aging and cardiovascular diseases. Front Pharmacol 2024; 15:1413853. [PMID: 39119608 PMCID: PMC11306071 DOI: 10.3389/fphar.2024.1413853] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2024] [Accepted: 06/24/2024] [Indexed: 08/10/2024] Open
Abstract
The average lifespan of humans has been increasing, resulting in a rapidly rising percentage of older individuals and high morbidity of aging-associated diseases, especially cardiovascular diseases (CVDs). Diverse intracellular and extracellular factors that interrupt homeostatic functions in the endoplasmic reticulum (ER) induce ER stress. Cells employ a dynamic signaling pathway of unfolded protein response (UPR) to buffer ER stress. Recent studies have demonstrated that ER stress triggers various cellular processes associated with aging and many aging-associated diseases, including CVDs. Autophagy is a conserved process involving lysosomal degradation and recycling of cytoplasmic components, proteins, organelles, and pathogens that invade the cytoplasm. Autophagy is vital for combating the adverse influence of aging on the heart. The present report summarizes recent studies on the mechanism of ER stress and autophagy and their overlap in aging and on CVD pathogenesis in the context of aging. It also discusses possible therapeutic interventions targeting ER stress and autophagy that might delay aging and prevent or treat CVDs.
Collapse
Affiliation(s)
- Chenguang Ma
- Department of Anesthesiology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Yang Liu
- 32295 Troops of P.L.A, Liaoyang, China
| | - Zhiling Fu
- Department of Anesthesiology, Shengjing Hospital of China Medical University, Shenyang, China
| |
Collapse
|
13
|
Davan-Wetton CSA, Montero-Melendez T. An optimised protocol for the detection of lipofuscin, a versatile and quantifiable marker of cellular senescence. PLoS One 2024; 19:e0306275. [PMID: 39008441 PMCID: PMC11249248 DOI: 10.1371/journal.pone.0306275] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2024] [Accepted: 06/13/2024] [Indexed: 07/17/2024] Open
Abstract
Lipofuscin is a yellow-brown pigment typically found in the lysosomes that contains a mixture of molecules including lipids, metals and misfolded proteins. The use of Sudan black B to detect lipofuscin accumulation, a well described marker of cellular senescence and ageing, was first described in 2013 by Georgakopoulou, et al. Here, we provide an optimisation of the original protocol. Firstly, we adjusted the staining methodology for increased ease of use on cultured cells. Secondly, we show that Sudan black B-stained lipofuscin emits strong fluorescence in the far-red channel making it suitable for fluorescence microscopy detection and quantification. Moreover, we also demonstrate that this optimised protocol can be utilised in conjunction with standard immunofluorescence staining techniques, making possible the simultaneous detection of lipofuscin and other cellular proteins of interest, like additional markers of senescence. This is a significant advantage over the most commonly used method for senescence detection, based on beta galactosidase enzymatic activity. We therefore believe that these findings and the provided optimised protocol will represent a useful tool for the scientific community in the field of cellular senescence.
Collapse
Affiliation(s)
- Camilla S. A. Davan-Wetton
- The William Harvey Research Institute, Faculty of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| | - Trinidad Montero-Melendez
- The William Harvey Research Institute, Faculty of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
- Centre for Inflammation and Therapeutic Innovation, Queen Mary University of London, London, United Kingdom
| |
Collapse
|
14
|
Zheng LY, Da YX, Luo X, Zhang X, Sun ZJ, Dong DL. Sorafenib extends the lifespan of C. elegans through mitochondrial uncoupling mechanism. Free Radic Biol Med 2024; 214:101-113. [PMID: 38360276 DOI: 10.1016/j.freeradbiomed.2024.02.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/29/2023] [Revised: 02/07/2024] [Accepted: 02/08/2024] [Indexed: 02/17/2024]
Abstract
Sorafenib is a targeted anticancer drug in clinic. Low-dose sorafenib has been reported to activate AMPK through inducing mitochondrial uncoupling without detectable toxicities. AMPK activation has been the approach for extending lifespan, therefore, we investigated the effect of sorafenib on lifespan and physical activity of C. elegans and the underlying mechanisms. In the present study, we found that the effect of sorafenib on C. elegans lifespan was typically hermetic. Sorafenib treatment at higher concentrations (100 μM) was toxic but at lower concentrations (1, 2.5, 5 μM) was beneficial to C. elegans. Sorafenib (1 μM) treatment for whole-life period extended C. elegans lifespan and improved C. elegans physical activity as manifested by increasing pharyngeal pumping and body movement, preserving intestinal barrier integrity, muscle fibers organization and mitochondrial morphology. In addition, sorafenib (1 μM) treatment enhanced C. elegans stress resistance. Sorafenib activated AMPK through inducing mitochondrial uncoupling in C. elegans. Sorafenib treatment activated DAF-16, SKN-1, and increased SOD-3, HSP-16.2, GST-4 expression in C. elegans. Sorafenib treatment induced AMPK-dependent autophagy in C. elegans. We conclude that low-dose sorafenib protects C. elegans against aging through activating AMPK/DAF-16 dependent anti-oxidant pathways and stimulating autophagy responses. Low-dose sorafenib could be a strategy for treating aging and aging-related diseases.
Collapse
Affiliation(s)
- Lu-Yao Zheng
- Department of Pharmacology, China Pharmaceutical University, Nanjing, People's Republic of China
| | - Yan-Xin Da
- Department of Pharmacology, China Pharmaceutical University, Nanjing, People's Republic of China
| | - Xiu Luo
- Department of Pharmacology, China Pharmaceutical University, Nanjing, People's Republic of China
| | - Xiao Zhang
- Department of Pharmacology, China Pharmaceutical University, Nanjing, People's Republic of China
| | - Zhi-Jie Sun
- Department of Pharmacology, China Pharmaceutical University, Nanjing, People's Republic of China.
| | - De-Li Dong
- Department of Pharmacology, China Pharmaceutical University, Nanjing, People's Republic of China.
| |
Collapse
|
15
|
Ford VJ, Applefeld WN, Wang J, Sun J, Solomon SB, Sidenko S, Feng J, Sheffield C, Klein HG, Yu ZX, Torabi-Parizi P, Danner RL, Sachdev V, Solomon MA, Chen MY, Natanson C. Cardiac Magnetic Resonance Studies in a Large Animal Model that Simulates the Cardiac Abnormalities of Human Septic Shock. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.02.05.578971. [PMID: 38903100 PMCID: PMC11188083 DOI: 10.1101/2024.02.05.578971] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/22/2024]
Abstract
Background Septic shock, in humans and in our well-established animal model, is associated with increases in biventricular end diastolic volume (EDV) and decreases in ejection fraction (EF). These abnormalities occur over 2 days and reverse within 10 days. Septic non-survivors do not develop an increase in EDV. The mechanism for this cardiac dysfunction and EDV differences is unknown. Methods Purpose-bred beagles randomized to receive intrabronchial Staphylococcus aureus (n=27) or saline (n=6) were provided standard ICU care including sedation, mechanical ventilation, and fluid resuscitation to a pulmonary arterial occlusion pressure of over 10mmHg. No catecholamines were administered. Over 96h, cardiac magnetic resonance imaging, echocardiograms, and invasive hemodynamics were serially performed, and laboratory data was collected. Tissue was obtained at 66h from six septic animals. Results From 0-96h after bacterial challenge, septic animals vs. controls had significantly increased left ventricular wall edema (6%) and wall thinning with loss of mass (15%) which was more pronounced at 48h in non-survivors than survivors. On histology, edema was located predominantly in myocytes, the interstitium, and endothelial cells. Edema was associated with significantly worse biventricular function (lower EFs), ventricular-arterial coupling, and circumferential strain. In septic animals, from 0-24h, the EDV decreased from baseline and, despite cardiac filling pressures being similar, decreased significantly more in non-survivors. From 24-48h, all septic animals had increases in biventricular chamber sizes. Survivors biventricular EDVs were significantly greater than baseline and in non-survivors, where biventricular EDVs were not different from baseline. Preload, afterload, or HR differences did not explain these differential serial changes in chamber size. Conclusion Systolic and diastolic cardiac dysfunction during sepsis is associated with ventricular wall edema. Rather than differences in preload, afterload, or heart rate, structural alterations to the ventricular wall best account for the volume changes associated with outcome during sepsis. In non-survivors, from 0-24h, sepsis induces a more severe diastolic dysfunction, further decreasing chamber size. The loss of left ventricular mass with wall thinning in septic survivors may, in part explain, the EDV increases from 24-48h. However, these changes continued and even accelerated into the recovery phase consistent with a reparative process rather than ongoing injury.
Collapse
Affiliation(s)
- Verity J. Ford
- Critical Care Medicine Department, Clinical Center, National Institutes of Health, (NIH, CC) Bethesda, Maryland 20892 USA
| | - Willard N. Applefeld
- Critical Care Medicine Department, Clinical Center, National Institutes of Health, (NIH, CC) Bethesda, Maryland 20892 USA
- Division of Cardiology, Duke University Medical Center, Durham, NC, USA
| | - Jeffrey Wang
- Critical Care Medicine Department, Clinical Center, National Institutes of Health, (NIH, CC) Bethesda, Maryland 20892 USA
- Emory, 100 Woodruff Circle, Atlanta, GA 30322
| | - Junfeng Sun
- Critical Care Medicine Department, Clinical Center, National Institutes of Health, (NIH, CC) Bethesda, Maryland 20892 USA
| | - Steven B. Solomon
- Critical Care Medicine Department, Clinical Center, National Institutes of Health, (NIH, CC) Bethesda, Maryland 20892 USA
| | - Stanislav Sidenko
- National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, Maryland 20892 USA
| | - Jing Feng
- Critical Care Medicine Department, Clinical Center, National Institutes of Health, (NIH, CC) Bethesda, Maryland 20892 USA
| | | | - Harvey G. Klein
- Department of Transfusion Medicine, Clinical Center, National Institutes of Health, (NIH, CC) Bethesda, Maryland 20892 USA
| | - Zu-Xi Yu
- National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, Maryland 20892 USA
| | - Parizad Torabi-Parizi
- National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, Maryland 20892 USA
| | - Robert L. Danner
- Critical Care Medicine Department, Clinical Center, National Institutes of Health, (NIH, CC) Bethesda, Maryland 20892 USA
| | - Vandana Sachdev
- National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, Maryland 20892 USA
| | - Michael A. Solomon
- Critical Care Medicine Department, Clinical Center, National Institutes of Health, (NIH, CC) Bethesda, Maryland 20892 USA
- National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, Maryland 20892 USA
| | - Marcus Y. Chen
- National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, Maryland 20892 USA
| | - Charles Natanson
- Critical Care Medicine Department, Clinical Center, National Institutes of Health, (NIH, CC) Bethesda, Maryland 20892 USA
- National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, Maryland 20892 USA
| |
Collapse
|
16
|
Favero G, Golic I, Arnaboldi F, Cappella A, Korac A, Monsalve M, Stacchiotti A, Rezzani R. Cardiometabolic Changes in Sirtuin1-Heterozygous Mice on High-Fat Diet and Melatonin Supplementation. Int J Mol Sci 2024; 25:860. [PMID: 38255934 PMCID: PMC10815439 DOI: 10.3390/ijms25020860] [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: 12/06/2023] [Revised: 01/04/2024] [Accepted: 01/08/2024] [Indexed: 01/24/2024] Open
Abstract
A hypercaloric fatty diet predisposes an individual to metabolic syndrome and cardiovascular complications. Sirtuin1 (SIRT1) belongs to the class III histone deacetylase family and sustains anabolism, mitochondrial biogenesis, and fat distribution. Epididymal white adipose tissue (eWAT) is involved in inflammation, whilst interscapular brown adipose tissue (iBAT) drives metabolism in obese rodents. Melatonin, a pineal indoleamine, acting as a SIRT1 modulator, may alleviate cardiometabolic damage. In the present study, we morphologically characterized the heart, eWAT, and iBAT in male heterozygous SIRT1+/- mice (HET mice) on a high-fat diet (60%E lard) versus a standard rodent diet (8.5% E fat) and drinking melatonin (10 mg/kg) for 16 weeks. Wild-type (WT) male C57Bl6/J mice were similarly fed for comparison. Cardiomyocyte fibrosis and endoplasmic reticulum (ER) stress response worsened in HET mice on a high-fat diet vs. other groups. Lipid peroxidation, ER, and mitochondrial stress were assessed by 4 hydroxy-2-nonenal (4HNE), glucose-regulated protein78 (GRP78), CCAA/enhancer-binding protein homologous protein (CHOP), heat shock protein 60 (HSP60), and mitofusin2 immunostainings. Ultrastructural analysis indicated the prevalence of atypical inter-myofibrillar mitochondria with short, misaligned cristae in HET mice on a lard diet despite melatonin supplementation. Abnormal eWAT adipocytes, crown-like inflammatory structures, tumor necrosis factor alpha (TNFα), and iBAT whitening characterized HET mice on a hypercaloric fatty diet and were maintained after melatonin supply. All these data suggest that melatonin's mechanism of action is strictly linked to full SIRT1 expression, which is required for the exhibition of effective antioxidant and anti-inflammatory properties.
Collapse
Affiliation(s)
- Gaia Favero
- Anatomy and Physiopathology Division, Department of Clinical and Experimental Sciences, University of Brescia, Viale Europa 11, 25123 Brescia, Italy; (G.F.); (R.R.)
- Interdipartimental University Center of Research “Adaption and Regeneration of Tissues and Organs (ARTO)”, University of Brescia, 25123 Brescia, Italy
| | - Igor Golic
- Center for Electron Microscopy, Faculty of Biology, University of Belgrade, Studentski trg 16, 11000 Belgrade, Serbia; (I.G.); (A.K.)
| | - Francesca Arnaboldi
- Department of Biomedical Sciences for Health, University of Milan, Via Mangiagalli 31, 20133 Milan, Italy; (F.A.); (A.C.)
| | - Annalisa Cappella
- Department of Biomedical Sciences for Health, University of Milan, Via Mangiagalli 31, 20133 Milan, Italy; (F.A.); (A.C.)
- U.O. Laboratorio di Morfologia Umana Applicata, IRCCS Policlinico San Donato, San Donato Milanese, 20097 Milan, Italy
| | - Aleksandra Korac
- Center for Electron Microscopy, Faculty of Biology, University of Belgrade, Studentski trg 16, 11000 Belgrade, Serbia; (I.G.); (A.K.)
| | - Maria Monsalve
- Instituto de Investigaciones Biomedicas “Alberto Sols” (CSIC-UAM), 28029 Madrid, Spain;
| | - Alessandra Stacchiotti
- Department of Biomedical Sciences for Health, University of Milan, Via Mangiagalli 31, 20133 Milan, Italy; (F.A.); (A.C.)
- U.O. Laboratorio di Morfologia Umana Applicata, IRCCS Policlinico San Donato, San Donato Milanese, 20097 Milan, Italy
| | - Rita Rezzani
- Anatomy and Physiopathology Division, Department of Clinical and Experimental Sciences, University of Brescia, Viale Europa 11, 25123 Brescia, Italy; (G.F.); (R.R.)
- Interdipartimental University Center of Research “Adaption and Regeneration of Tissues and Organs (ARTO)”, University of Brescia, 25123 Brescia, Italy
- Italian Society for the Study of Orofacial Pain (Società Italiana Studio Dolore Orofacciale—SISDO), 25123 Brescia, Italy
| |
Collapse
|
17
|
Różanowska MB. Lipofuscin, Its Origin, Properties, and Contribution to Retinal Fluorescence as a Potential Biomarker of Oxidative Damage to the Retina. Antioxidants (Basel) 2023; 12:2111. [PMID: 38136230 PMCID: PMC10740933 DOI: 10.3390/antiox12122111] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2023] [Revised: 12/05/2023] [Accepted: 12/09/2023] [Indexed: 12/24/2023] Open
Abstract
Lipofuscin accumulates with age as intracellular fluorescent granules originating from incomplete lysosomal digestion of phagocytosed and autophagocytosed material. The purpose of this review is to provide an update on the current understanding of the role of oxidative stress and/or lysosomal dysfunction in lipofuscin accumulation and its consequences, particularly for retinal pigment epithelium (RPE). Next, the fluorescence of lipofuscin, spectral changes induced by oxidation, and its contribution to retinal fluorescence are discussed. This is followed by reviewing recent developments in fluorescence imaging of the retina and the current evidence on the prognostic value of retinal fluorescence for the progression of age-related macular degeneration (AMD), the major blinding disease affecting elderly people in developed countries. The evidence of lipofuscin oxidation in vivo and the evidence of increased oxidative damage in AMD retina ex vivo lead to the conclusion that imaging of spectral characteristics of lipofuscin fluorescence may serve as a useful biomarker of oxidative damage, which can be helpful in assessing the efficacy of potential antioxidant therapies in retinal degenerations associated with accumulation of lipofuscin and increased oxidative stress. Finally, amendments to currently used fluorescence imaging instruments are suggested to be more sensitive and specific for imaging spectral characteristics of lipofuscin fluorescence.
Collapse
Affiliation(s)
- Małgorzata B. Różanowska
- School of Optometry and Vision Sciences, College of Biomedical and Life Sciences, Cardiff University, Maindy Road, Cardiff CF24 4HQ, Wales, UK;
- Cardiff Institute for Tissue Engineering and Repair (CITER), Redwood Building, King Edward VII Avenue, Cardiff CF10 3NB, Wales, UK
| |
Collapse
|
18
|
Walter S, Mertens C, Muckenthaler MU, Ott C. Cardiac iron metabolism during aging - Role of inflammation and proteolysis. Mech Ageing Dev 2023; 215:111869. [PMID: 37678569 DOI: 10.1016/j.mad.2023.111869] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Revised: 09/01/2023] [Accepted: 09/03/2023] [Indexed: 09/09/2023]
Abstract
Iron is the most abundant trace element in the human body. Since iron can switch between its 2-valent and 3-valent form it is essential in various physiological processes such as energy production, proliferation or DNA synthesis. Especially high metabolic organs such as the heart rely on iron-associated iron-sulfur and heme proteins. However, due to switches in iron oxidation state, iron overload exhibits high toxicity through formation of reactive oxygen species, underlining the importance of balanced iron levels. Growing evidence demonstrates disturbance of this balance during aging. While age-associated cardiovascular diseases are often related to iron deficiency, in physiological aging cardiac iron accumulates. To understand these changes, we focused on inflammation and proteolysis, two hallmarks of aging, and their role in iron metabolism. Via the IL-6-hepcidin axis, inflammation and iron status are strongly connected often resulting in anemia accompanied by infiltration of macrophages. This tight connection between anemia and inflammation highlights the importance of the macrophage iron metabolism during inflammation. Age-related decrease in proteolytic activity additionally affects iron balance due to impaired degradation of iron metabolism proteins. Therefore, this review accentuates alterations in iron metabolism during aging with regards to inflammation and proteolysis to draw attention to their implications and associations.
Collapse
Affiliation(s)
- Sophia Walter
- German Institute of Human Nutrition Potsdam-Rehbruecke, Department of Molecular Toxicology, Nuthetal, Germany; TraceAge-DFG Research Unit on Interactions of Essential Trace Elements in Healthy and Diseased Elderly, Potsdam-Berlin-Jena, Wuppertal, Germany; DZHK (German Center for Cardiovascular Research), partner site Berlin, Berlin, Germany
| | - Christina Mertens
- Center for Translational Biomedical Iron Research, Department of Pediatric Oncology, Immunology, and Hematology, University of Heidelberg, Heidelberg, Germany; DZHK (German Center for Cardiovascular Research), Heidelberg, Mannheim, Germany
| | - Martina U Muckenthaler
- Center for Translational Biomedical Iron Research, Department of Pediatric Oncology, Immunology, and Hematology, University of Heidelberg, Heidelberg, Germany; DZHK (German Center for Cardiovascular Research), Heidelberg, Mannheim, Germany; Molecular Medicine Partnership Unit, Heidelberg, Germany; Translational Lung Research Center Heidelberg (TLRC), German Center for Lung Research (DZL), Heidelberg, Germany
| | - Christiane Ott
- German Institute of Human Nutrition Potsdam-Rehbruecke, Department of Molecular Toxicology, Nuthetal, Germany; TraceAge-DFG Research Unit on Interactions of Essential Trace Elements in Healthy and Diseased Elderly, Potsdam-Berlin-Jena, Wuppertal, Germany; DZHK (German Center for Cardiovascular Research), partner site Berlin, Berlin, Germany.
| |
Collapse
|
19
|
Li A, Shami GJ, Griffiths L, Lal S, Irving H, Braet F. Giant mitochondria in cardiomyocytes: cellular architecture in health and disease. Basic Res Cardiol 2023; 118:39. [PMID: 37775647 PMCID: PMC10541842 DOI: 10.1007/s00395-023-01011-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Revised: 08/29/2023] [Accepted: 09/15/2023] [Indexed: 10/01/2023]
Abstract
Giant mitochondria are frequently observed in different disease models within the brain, kidney, and liver. In cardiac muscle, these enlarged organelles are present across diverse physiological and pathophysiological conditions including in ageing and exercise, and clinically in alcohol-induced heart disease and various cardiomyopathies. This mitochondrial aberration is widely considered an early structural hallmark of disease leading to adverse organ function. In this thematic paper, we discuss the current state-of-knowledge on the presence, structure and functional implications of giant mitochondria in heart muscle. Despite its demonstrated reoccurrence in different heart diseases, the literature on this pathophysiological phenomenon remains relatively sparse since its initial observations in the early 60s. We review historical and contemporary investigations from cultured cardiomyocytes to human tissue samples to address the role of giant mitochondria in cardiac health and disease. Finally, we discuss their significance for the future development of novel mitochondria-targeted therapies to improve cardiac metabolism and functionality.
Collapse
Affiliation(s)
- Amy Li
- Department of Rural Clinical Sciences, La Trobe Rural Health School, La Trobe University, Bendigo, VIC, Australia.
- Centre for Healthy Futures, Torrens University Australia, Surry Hills, NSW, Australia.
- School of Medical Sciences, The University of Sydney, Camperdown, NSW, Australia.
| | - Gerald J Shami
- School of Medical Sciences (Molecular and Cellular Biomedicine), The University of Sydney, Camperdown, NSW, Australia
- Australian Centre for Microscopy and Microanalysis, The University of Sydney, Camperdown, NSW, Australia
| | - Lisa Griffiths
- Anatomical Pathology, PathWest, QEII Medical Centre, Nedlands, WA, Australia
| | - Sean Lal
- School of Medical Sciences, The University of Sydney, Camperdown, NSW, Australia
| | - Helen Irving
- Department of Rural Clinical Sciences, La Trobe Rural Health School, La Trobe University, Bendigo, VIC, Australia
| | - Filip Braet
- School of Medical Sciences (Molecular and Cellular Biomedicine), The University of Sydney, Camperdown, NSW, Australia.
- Australian Centre for Microscopy and Microanalysis, The University of Sydney, Camperdown, NSW, Australia.
| |
Collapse
|
20
|
Lee JR, Jeong KW. N-retinylidene- N-retinylethanolamine degradation in human retinal pigment epithelial cells via memantine- and ifenprodil-mediated autophagy. THE KOREAN JOURNAL OF PHYSIOLOGY & PHARMACOLOGY : OFFICIAL JOURNAL OF THE KOREAN PHYSIOLOGICAL SOCIETY AND THE KOREAN SOCIETY OF PHARMACOLOGY 2023; 27:449-456. [PMID: 37641807 PMCID: PMC10466070 DOI: 10.4196/kjpp.2023.27.5.449] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Revised: 04/26/2023] [Accepted: 04/27/2023] [Indexed: 08/31/2023]
Abstract
N-methyl-D-aspartate (NMDA) receptors are ionic glutamine receptors involved in brain development and functions such as learning and memory formation. NMDA receptor inhibition is associated with autophagy activation. In this study, we investigated whether the NMDA receptor antagonists, memantine and ifenprodil, induce autophagy in human retinal pigment epithelial cells (ARPE-19) to remove Nretinylidene- N-retinylethanolamine (A2E), an intracellular lipofuscin component. Fluorometric analysis using labeled A2E (A2E-BDP) and confocal microscopic examination revealed that low concentrations of NMDA receptor antagonists, which did not induce cytotoxicity, significantly reduced A2E accumulation in ARPE-19 cells. In addition, memantine and ifenprodil activated autophagy in ARPE-19 cells as measured by microtubule-associated protein 1A/1B-light chain3-II formation and phosphorylated p62 protein levels. Further, to understand the correlation between memantine- and ifenprodil-mediated A2E degradation and autophagy, autophagy-related 5 (ATG5) was depleted using RNA interference. Memantine and ifenprodil failed to degrade A2E in ARPE-19 cells lacking ATG5. Taken together, our study indicates that the NMDA receptor antagonists, memantine and ifenprodil, can remove A2E accumulated in cells via autophagy activation in ARPE-19 cells.
Collapse
Affiliation(s)
- Jae Rim Lee
- College of Pharmacy, Gachon Research Institute of Pharmaceutical Sciences, Gachon University, Incheon 21936, Korea
| | - Kwang Won Jeong
- College of Pharmacy, Gachon Research Institute of Pharmaceutical Sciences, Gachon University, Incheon 21936, Korea
| |
Collapse
|
21
|
Song SB, Shim W, Hwang ES. Lipofuscin Granule Accumulation Requires Autophagy Activation. Mol Cells 2023; 46:486-495. [PMID: 37438887 PMCID: PMC10440269 DOI: 10.14348/molcells.2023.0019] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2023] [Revised: 04/13/2023] [Accepted: 05/13/2023] [Indexed: 07/14/2023] Open
Abstract
Lipofuscins are oxidized lipid and protein complexes that accumulate during cellular senescence and tissue aging, regarded as markers for cellular oxidative damage, tissue aging, and certain aging-associated diseases. Therefore, understanding their cellular biological properties is crucial for effective treatment development. Through traditional microscopy, lipofuscins are readily observed as fluorescent granules thought to accumulate in lysosomes. However, lipofuscin granule formation and accumulation in senescent cells are poorly understood. Thus, this study examined lipofuscin accumulation in human fibroblasts exposed to various stressors. Our results substantiate that in glucose-starved or replicative senescence cells, where elevated oxidative stress levels activate autophagy, lipofuscins predominately appear as granules that co-localize with autolysosomes due to lysosomal acidity or impairment. Meanwhile, autophagosome formation is attenuated in cells experiencing oxidative stress induced by a doxorubicin pulse and chase, and lipofuscin fluorescence granules seldom manifest in the cytoplasm. As Torin-1 treatment activates autophagy, granular lipofuscins intensify and dominate, indicating that autophagy activation triggers their accumulation. Our results suggest that high oxidative stress activates autophagy but fails in lipofuscin removal, leaving an abundance of lipofuscin-filled impaired autolysosomes, referred to as residual bodies. Therefore, future endeavors in treating lipofuscin pathology-associated diseases and dysfunctions through autophagy activation demand meticulous consideration.
Collapse
Affiliation(s)
- Seon Beom Song
- Department of Life Science, University of Seoul, Seoul 02504, Korea
| | - Woosung Shim
- Department of Life Science, University of Seoul, Seoul 02504, Korea
| | - Eun Seong Hwang
- Department of Life Science, University of Seoul, Seoul 02504, Korea
| |
Collapse
|
22
|
Hao Y, Li B, Huber SA, Liu W. Bibliometric analysis of trends in cardiac aging research over the past 20 years. Medicine (Baltimore) 2023; 102:e34870. [PMID: 37653740 PMCID: PMC10470686 DOI: 10.1097/md.0000000000034870] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/04/2023] [Revised: 07/31/2023] [Accepted: 08/01/2023] [Indexed: 09/02/2023] Open
Abstract
BACKGROUND In recent years, many studies have addressed cardiac aging and related diseases. This study aims to understand the research trend of cardiac aging and find new hot issues. METHODS We searched the web of science core collection database for articles published between 2003 and 2022 on the topic of "cardiac aging." Complete information including keywords, publication year, journal title, country, organization, and author were extracted for analysis. The VOS viewer software was used to generate network maps of keywords, countries, institutions, and author relationships for visual network analysis. RESULTS A total of 1002 papers were analyzed in the study. Overall, the number of annual publications on cardiac aging has increased since 2009, and new hot topics are emerging. The top 3 countries with the most publications were the United States (471 articles), China (209 articles) and Italy (101 articles). The University of Washington published the most papers (35 articles). The cluster analysis with author as the keyword found that the connections among different scholars are scattered and clustered in a small range. Network analysis based on keyword co-occurrence and year of publication identified relevant features and trends in cardiac aging research. According to the results of cluster analysis, all the articles are divided into 4 topics: "mechanisms of cardiac aging", "prevention and treatment of cardiac aging", "characteristics of cardiac aging", and "others." In recent years, the mechanism and treatment of cardiac aging have attracted the most attention. In both studies, animal models are used more often than in human populations. Mitochondrial dysfunction, autophagy and mitochondrial autophagy are hotspots in current research. CONCLUSION In this study, bibliometric analysis was used to analyze the research trend of cardiac aging in the past 20 years. The mechanism and treatment of cardiac aging are the most concerned contents. Mitochondrial dysfunction, autophagy and mitophagy are the focus of future research on cardiac aging.
Collapse
Affiliation(s)
- Yan Hao
- Department of Cardiology, The Fourth Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
- Harbin Medical University, Harbin, Heilongjiang, China
| | - Bohan Li
- Harbin Medical University, Harbin, Heilongjiang, China
| | - Sally A. Huber
- Department of Pathology and Laboratory Medicine, University of Vermont, Colchester, VT
| | - Wei Liu
- Harbin Medical University, Harbin, Heilongjiang, China
- Department of Geriatric Cardiovascular Division, Guangdong Provincial Geriatrics Institute, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| |
Collapse
|
23
|
Deng Y, Adam V, Nepovimova E, Heger Z, Valko M, Wu Q, Wei W, Kuca K. c-Jun N-terminal kinase signaling in cellular senescence. Arch Toxicol 2023; 97:2089-2109. [PMID: 37335314 DOI: 10.1007/s00204-023-03540-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Accepted: 06/06/2023] [Indexed: 06/21/2023]
Abstract
Cellular senescence leads to decreased tissue regeneration and inflammation and is associated with diabetes, neurodegenerative diseases, and tumorigenesis. However, the mechanisms of cellular senescence are not fully understood. Emerging evidence has indicated that c-Jun N-terminal kinase (JNK) signaling is involved in the regulation of cellular senescence. JNK can downregulate hypoxia inducible factor-1α to accelerate hypoxia-induced neuronal cell senescence. The activation of JNK inhibits mTOR activity and triggers autophagy, which promotes cellular senescence. JNK can upregulate the expression of p53 and Bcl-2 and accelerates cancer cell senescence; however, this signaling also mediates the expression of amphiregulin and PD-LI to achieve cancer cell immune evasion and prevents their senescence. The activation of JNK further triggers forkhead box O expression and its target gene Jafrac1 to extend the lifespan of Drosophila. JNK can also upregulate the expression of DNA repair protein poly ADP-ribose polymerase 1 and heat shock protein to delay cellular senescence. This review discusses recent advances in understanding the function of JNK signaling in cellular senescence and includes a comprehensive analysis of the molecular mechanisms underlying JNK-mediated senescence evasion and oncogene-induced cellular senescence. We also summarize the research progress in anti-aging agents that target JNK signaling. This study will contribute to a better understanding of the molecular targets of cellular senescence and provides insights into anti-aging, which may be used to develop drugs for the treatment of aging-related diseases.
Collapse
Affiliation(s)
- Ying Deng
- College of Life Science, Yangtze University, Jingzhou, 434025, China
| | - Vojtech Adam
- Department of Chemistry and Biochemistry, Mendel University in Brno, Brno, 613 00, Czech Republic
- Central European Institute of Technology, Brno University of Technology, Brno, 602 00, Czech Republic
| | - Eugenie Nepovimova
- Department of Chemistry, Faculty of Science, University of Hradec Králové, 500 03, Hradec Králové, Czech Republic
| | - Zbynek Heger
- Department of Chemistry and Biochemistry, Mendel University in Brno, Brno, 613 00, Czech Republic
- Central European Institute of Technology, Brno University of Technology, Brno, 602 00, Czech Republic
| | - Marian Valko
- Faculty of Chemical and Food Technology, Slovak University of Technology, 812 37, Bratislava, Slovakia
| | - Qinghua Wu
- College of Life Science, Yangtze University, Jingzhou, 434025, China.
- Department of Chemistry, Faculty of Science, University of Hradec Králové, 500 03, Hradec Králové, Czech Republic.
| | - Wei Wei
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-Products, Key Laboratory of Traceability for Agricultural Genetically Modified Organisms, Ministry of Agriculture and Rural Affairs, Zhejiang Academy of Agricultural Sciences, Hangzhou, 310021, China.
| | - Kamil Kuca
- Department of Chemistry, Faculty of Science, University of Hradec Králové, 500 03, Hradec Králové, Czech Republic.
- Andalusian Research Institute in Data Science and Computational Intelligence (DaSCI), University of Granada, Granada, Spain.
| |
Collapse
|
24
|
Baechle JJ, Chen N, Makhijani P, Winer S, Furman D, Winer DA. Chronic inflammation and the hallmarks of aging. Mol Metab 2023; 74:101755. [PMID: 37329949 PMCID: PMC10359950 DOI: 10.1016/j.molmet.2023.101755] [Citation(s) in RCA: 122] [Impact Index Per Article: 61.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Revised: 05/30/2023] [Accepted: 06/13/2023] [Indexed: 06/19/2023] Open
Abstract
BACKGROUND Recently, the hallmarks of aging were updated to include dysbiosis, disabled macroautophagy, and chronic inflammation. In particular, the low-grade chronic inflammation during aging, without overt infection, is defined as "inflammaging," which is associated with increased morbidity and mortality in the aging population. Emerging evidence suggests a bidirectional and cyclical relationship between chronic inflammation and the development of age-related conditions, such as cardiovascular diseases, neurodegeneration, cancer, and frailty. How the crosstalk between chronic inflammation and other hallmarks of aging underlies biological mechanisms of aging and age-related disease is thus of particular interest to the current geroscience research. SCOPE OF REVIEW This review integrates the cellular and molecular mechanisms of age-associated chronic inflammation with the other eleven hallmarks of aging. Extra discussion is dedicated to the hallmark of "altered nutrient sensing," given the scope of Molecular Metabolism. The deregulation of hallmark processes during aging disrupts the delicate balance between pro-inflammatory and anti-inflammatory signaling, leading to a persistent inflammatory state. The resultant chronic inflammation, in turn, further aggravates the dysfunction of each hallmark, thereby driving the progression of aging and age-related diseases. MAIN CONCLUSIONS The crosstalk between chronic inflammation and other hallmarks of aging results in a vicious cycle that exacerbates the decline in cellular functions and promotes aging. Understanding this complex interplay will provide new insights into the mechanisms of aging and the development of potential anti-aging interventions. Given their interconnectedness and ability to accentuate the primary elements of aging, drivers of chronic inflammation may be an ideal target with high translational potential to address the pathological conditions associated with aging.
Collapse
Affiliation(s)
- Jordan J Baechle
- Buck Artificial Intelligence Platform, the Buck Institute for Research on Aging, Novato, CA, USA
| | - Nan Chen
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada; Division of Cellular & Molecular Biology, Diabetes Research Group, Toronto General Hospital Research Institute (TGHRI), University Health Network, Toronto, ON, Canada
| | - Priya Makhijani
- Buck Artificial Intelligence Platform, the Buck Institute for Research on Aging, Novato, CA, USA; Department of Immunology, Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | - Shawn Winer
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
| | - David Furman
- Buck Artificial Intelligence Platform, the Buck Institute for Research on Aging, Novato, CA, USA; Stanford 1000 Immunomes Project, Stanford University School of Medicine, Stanford, CA, USA; Instituto de Investigaciones en Medicina Traslacional (IIMT), Universidad Austral, CONICET, Pilar, Argentina.
| | - Daniel A Winer
- Buck Artificial Intelligence Platform, the Buck Institute for Research on Aging, Novato, CA, USA; Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada; Department of Immunology, Faculty of Medicine, University of Toronto, Toronto, ON, Canada; Division of Cellular & Molecular Biology, Diabetes Research Group, Toronto General Hospital Research Institute (TGHRI), University Health Network, Toronto, ON, Canada; Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA, USA.
| |
Collapse
|
25
|
Huang Y, Zhou B. Mitochondrial Dysfunction in Cardiac Diseases and Therapeutic Strategies. Biomedicines 2023; 11:biomedicines11051500. [PMID: 37239170 DOI: 10.3390/biomedicines11051500] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Revised: 05/15/2023] [Accepted: 05/16/2023] [Indexed: 05/28/2023] Open
Abstract
Mitochondria are the main site of intracellular synthesis of ATP, which provides energy for various physiological activities of the cell. Cardiomyocytes have a high density of mitochondria and mitochondrial damage is present in a variety of cardiovascular diseases. In this paper, we describe mitochondrial damage in mitochondrial cardiomyopathy, congenital heart disease, coronary heart disease, myocardial ischemia-reperfusion injury, heart failure, and drug-induced cardiotoxicity, in the context of the key roles of mitochondria in cardiac development and homeostasis. Finally, we discuss the main current therapeutic strategies aimed at alleviating mitochondrial impairment-related cardiac dysfunction, including pharmacological strategies, gene therapy, mitochondrial replacement therapy, and mitochondrial transplantation. It is hoped that this will provide new ideas for the treatment of cardiovascular diseases.
Collapse
Affiliation(s)
- Yafei Huang
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Science and Peking Union Medical College, 167 North Lishi Road, Xicheng District, Beijing 100037, China
| | - Bingying Zhou
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Science and Peking Union Medical College, 167 North Lishi Road, Xicheng District, Beijing 100037, China
| |
Collapse
|
26
|
Villalón-García I, Povea-Cabello S, Álvarez-Córdoba M, Talaverón-Rey M, Suárez-Rivero JM, Suárez-Carrillo A, Munuera-Cabeza M, Reche-López D, Cilleros-Holgado P, Piñero-Pérez R, Sánchez-Alcázar JA. Vicious cycle of lipid peroxidation and iron accumulation in neurodegeneration. Neural Regen Res 2022; 18:1196-1202. [PMID: 36453394 PMCID: PMC9838166 DOI: 10.4103/1673-5374.358614] [Citation(s) in RCA: 40] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022] Open
Abstract
Lipid peroxidation and iron accumulation are closely associated with neurodegenerative diseases, such as Alzheimer's, Parkinson's, and Huntington's diseases, or neurodegeneration with brain iron accumulation disorders. Mitochondrial dysfunction, lipofuscin accumulation, autophagy disruption, and ferroptosis have been implicated as the critical pathomechanisms of lipid peroxidation and iron accumulation in these disorders. Currently, the connection between lipid peroxidation and iron accumulation and the initial cause or consequence in neurodegeneration processes is unclear. In this review, we have compiled the known mechanisms by which lipid peroxidation triggers iron accumulation and lipofuscin formation, and the effect of iron overload on lipid peroxidation and cellular function. The vicious cycle established between both pathological alterations may lead to the development of neurodegeneration. Therefore, the investigation of these mechanisms is essential for exploring therapeutic strategies to restrict neurodegeneration. In addition, we discuss the interplay between lipid peroxidation and iron accumulation in neurodegeneration, particularly in PLA2G6-associated neurodegeneration, a rare neurodegenerative disease with autosomal recessive inheritance, which belongs to the group of neurodegeneration with brain iron accumulation disorders.
Collapse
Affiliation(s)
- Irene Villalón-García
- Centro Andaluz de Biología del Desarrollo (CABD-CSIC-Universidad Pablo de Olavide de Sevilla), and Centro de Investigación Biomédica en Red: Enfermedades Raras, Instituto de Salud Carlos III, Sevilla, Spain
| | - Suleva Povea-Cabello
- Centro Andaluz de Biología del Desarrollo (CABD-CSIC-Universidad Pablo de Olavide de Sevilla), and Centro de Investigación Biomédica en Red: Enfermedades Raras, Instituto de Salud Carlos III, Sevilla, Spain
| | - Mónica Álvarez-Córdoba
- Centro Andaluz de Biología del Desarrollo (CABD-CSIC-Universidad Pablo de Olavide de Sevilla), and Centro de Investigación Biomédica en Red: Enfermedades Raras, Instituto de Salud Carlos III, Sevilla, Spain
| | - Marta Talaverón-Rey
- Centro Andaluz de Biología del Desarrollo (CABD-CSIC-Universidad Pablo de Olavide de Sevilla), and Centro de Investigación Biomédica en Red: Enfermedades Raras, Instituto de Salud Carlos III, Sevilla, Spain
| | - Juan M. Suárez-Rivero
- Centro Andaluz de Biología del Desarrollo (CABD-CSIC-Universidad Pablo de Olavide de Sevilla), and Centro de Investigación Biomédica en Red: Enfermedades Raras, Instituto de Salud Carlos III, Sevilla, Spain
| | - Alejandra Suárez-Carrillo
- Centro Andaluz de Biología del Desarrollo (CABD-CSIC-Universidad Pablo de Olavide de Sevilla), and Centro de Investigación Biomédica en Red: Enfermedades Raras, Instituto de Salud Carlos III, Sevilla, Spain
| | - Manuel Munuera-Cabeza
- Centro Andaluz de Biología del Desarrollo (CABD-CSIC-Universidad Pablo de Olavide de Sevilla), and Centro de Investigación Biomédica en Red: Enfermedades Raras, Instituto de Salud Carlos III, Sevilla, Spain
| | - Diana Reche-López
- Centro Andaluz de Biología del Desarrollo (CABD-CSIC-Universidad Pablo de Olavide de Sevilla), and Centro de Investigación Biomédica en Red: Enfermedades Raras, Instituto de Salud Carlos III, Sevilla, Spain
| | - Paula Cilleros-Holgado
- Centro Andaluz de Biología del Desarrollo (CABD-CSIC-Universidad Pablo de Olavide de Sevilla), and Centro de Investigación Biomédica en Red: Enfermedades Raras, Instituto de Salud Carlos III, Sevilla, Spain
| | - Rocío Piñero-Pérez
- Centro Andaluz de Biología del Desarrollo (CABD-CSIC-Universidad Pablo de Olavide de Sevilla), and Centro de Investigación Biomédica en Red: Enfermedades Raras, Instituto de Salud Carlos III, Sevilla, Spain
| | - José A. Sánchez-Alcázar
- Centro Andaluz de Biología del Desarrollo (CABD-CSIC-Universidad Pablo de Olavide de Sevilla), and Centro de Investigación Biomédica en Red: Enfermedades Raras, Instituto de Salud Carlos III, Sevilla, Spain,Correspondence to: José A. Sánchez-Alcázar, MD, PhD, .
| |
Collapse
|
27
|
Kaushik S, Tasset I, Arias E, Pampliega O, Wong E, Martinez-Vicente M, Cuervo AM. Autophagy and the hallmarks of aging. Ageing Res Rev 2021; 72:101468. [PMID: 34563704 DOI: 10.1016/j.arr.2021.101468] [Citation(s) in RCA: 169] [Impact Index Per Article: 42.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 09/12/2021] [Accepted: 09/20/2021] [Indexed: 12/13/2022]
Abstract
Autophagy, an essential cellular process that mediates degradation of proteins and organelles in lysosomes, has been tightly linked to cellular quality control for its role as part of the proteostasis network. The current interest in identifying the cellular and molecular determinants of aging, has highlighted the important contribution of malfunctioning of autophagy with age to the loss of proteostasis that characterizes all old organisms. However, the diversity of cellular functions of the different types of autophagy and the often reciprocal interactions of autophagy with other determinants of aging, is placing autophagy at the center of the aging process. In this work, we summarize evidence for the contribution of autophagy to health- and lifespan and provide examples of the bidirectional interplay between autophagic pathways and several of the so-called hallmarks of aging. This central role of autophagy in aging, and the dependence on autophagy of many geroprotective interventions, has motivated a search for direct modulators of autophagy that could be used to slow aging and extend healthspan. Here, we review some of those ongoing therapeutic efforts and comment on the potential of targeting autophagy in aging.
Collapse
|