1
|
Fukumoto T, Shimosawa T, Yakabe M, Yoshida S, Yoshida Y. Recent advances in biomarkers for senescence: Bridging basic research to clinic. Geriatr Gerontol Int 2025; 25:139-147. [PMID: 39754295 DOI: 10.1111/ggi.15054] [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: 03/11/2024] [Revised: 10/31/2024] [Accepted: 12/14/2024] [Indexed: 01/06/2025]
Abstract
In this review, we review the current status of biomarkers for aging and possible perspectives on anti-aging or rejuvenation from the standpoint of biomarkers. Aging is observed in all cells and organs, and we focused on research into senescence in the skin, musculoskeletal system, immune system, and cardiovascular system. Commonly used biomarkers include SA-βgal, cell-cycle markers, senescence-associated secretory phenotype (SASP) factors, damage-associated molecular patterns (DAMPs), and DNA-damage-related markers. In addition, each organ or cell has its specific markers. Generally speaking, a combination of biomarkers is required to define age-related changes. When considering the translation of basic research, biomarkers that are highly sensitive, highly specific, with validation and reliability as well as being non-invasive are optimal; however, currently reported markers do not fulfill the prerequisite for biomarkers. In addition, rodent models of aging do not necessarily represent human aging, and markers in rodent or cell models are not applicable in clinical settings. The prerequisite of clinically applicable biomarkers is that they provide useful information for clinical decision-making, such as predicting disease risk, diagnosing disease, monitoring disease progression, or guiding treatment decisions. Therefore, the development of non-invasive robust, reliable, and useful biomarkers in humans is necessary to develop anti-aging therapy for humans. Geriatr Gerontol Int 2025; 25: 139-147.
Collapse
Affiliation(s)
- Takeshi Fukumoto
- Division of Dermatology, Department of Internal Related, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Tatsuo Shimosawa
- Department of Clinical Laboratory, Graduate School of Medicine, International University of Health and Welfare, Hyogo, Japan
| | - Mitsutaka Yakabe
- Department of Geriatric Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Shota Yoshida
- Department of Geriatric and General Medicine, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Yohko Yoshida
- Department of Advanced Senotherapeutics and Department of Cardiovascular Biology and Medicine, Juntendo University Graduate School of Medicine, Tokyo, Japan
| |
Collapse
|
2
|
Ávila BM, Zanini BM, Luduvico KP, Oliveira TL, Hense JD, Garcia DN, Prosczek J, Stefanello FM, da Cruz PH, Giongo JL, Vaucher RA, Mason JB, Masternak MM, Schneider A. Effect of senolytic drugs in young female mice chemically induced to estropause. Life Sci 2024; 357:123073. [PMID: 39307182 DOI: 10.1016/j.lfs.2024.123073] [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: 05/27/2024] [Revised: 09/10/2024] [Accepted: 09/18/2024] [Indexed: 09/28/2024]
Abstract
AIMS This study aimed to assess metabolic responses and senescent cell burden in young female mice induced to estropause and treated with senolytic drugs. MAIN METHODS Estropause was induced by 4-vinylcyclohexene diepoxide (VCD) injection in two-month-old mice. The senolytics dasatinib and quercetin (D + Q) or fisetin were given by oral gavage once a month from five to 11 months of age. KEY FINDINGS VCD-induced estropause led to increased body mass and reduced albumin concentrations compared to untreated cyclic mice, without affecting insulin sensitivity, lipid profile, liver enzymes, or total proteins. Estropause decreased catalase activity in adipose tissue but had no significant effect on other redox parameters in adipose and hepatic tissues. Fisetin treatment reduced ROS levels in the hepatic tissue of estropause mice. Estropause did not influence senescence-associated beta-galactosidase activity in adipose and hepatic tissues but increased senescent cell markers and fibrosis in ovaries. Senolytic treatment did not decrease ovarian cellular senescence induced by estropause. SIGNIFICANCE Overall, the findings suggest that estropause leads to minor metabolic changes in young females, and the senolytics D + Q and fisetin had no protective effects despite increased ovarian senescence.
Collapse
Affiliation(s)
- Bianca M Ávila
- Nutrition College, Universidade Federal de Pelotas, Pelotas, RS, Brazil
| | - Bianka M Zanini
- Nutrition College, Universidade Federal de Pelotas, Pelotas, RS, Brazil
| | - Karina P Luduvico
- Center of Chemical, Pharmaceutical, and Food Sciences, Universidade Federal de Pelotas, Pelotas, RS, Brazil
| | - Thais L Oliveira
- Biotechnology Center, Universidade Federal de Pelotas, Pelotas, RS, Brazil
| | - Jéssica D Hense
- Nutrition College, Universidade Federal de Pelotas, Pelotas, RS, Brazil
| | - Driele N Garcia
- Nutrition College, Universidade Federal de Pelotas, Pelotas, RS, Brazil
| | - Juliane Prosczek
- Nutrition College, Universidade Federal de Pelotas, Pelotas, RS, Brazil
| | - Francieli M Stefanello
- Center of Chemical, Pharmaceutical, and Food Sciences, Universidade Federal de Pelotas, Pelotas, RS, Brazil
| | - Pedro H da Cruz
- Center of Chemical, Pharmaceutical, and Food Sciences, Microorganism Biochemistry and Molecular Biology Research Laboratory, Universidade Federal de Pelotas, Pelotas, RS, Brazil
| | - Janice L Giongo
- Center of Chemical, Pharmaceutical, and Food Sciences, Microorganism Biochemistry and Molecular Biology Research Laboratory, Universidade Federal de Pelotas, Pelotas, RS, Brazil
| | - Rodrigo A Vaucher
- Center of Chemical, Pharmaceutical, and Food Sciences, Microorganism Biochemistry and Molecular Biology Research Laboratory, Universidade Federal de Pelotas, Pelotas, RS, Brazil
| | - Jeffrey B Mason
- Center of Chemical, Pharmaceutical, and Food Sciences, Microorganism Biochemistry and Molecular Biology Research Laboratory, Universidade Federal de Pelotas, Pelotas, RS, Brazil
| | - Michal M Masternak
- College of Medicine, Burnett School of Biomedical Sciences, University of Central Florida, Orlando, FL, USA; Department of Head and Neck Surgery, Poznan University of Medical Sciences, Poznan, Poland
| | - Augusto Schneider
- Nutrition College, Universidade Federal de Pelotas, Pelotas, RS, Brazil.
| |
Collapse
|
3
|
Zhang M, Lin Y, Han Z, Huang X, Zhou S, Wang S, Zhou Y, Han X, Chen H. Exploring mechanisms of skin aging: insights for clinical treatment. Front Immunol 2024; 15:1421858. [PMID: 39582871 PMCID: PMC11581952 DOI: 10.3389/fimmu.2024.1421858] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Accepted: 10/23/2024] [Indexed: 11/26/2024] Open
Abstract
The skin is the largest organ in the human body and is made up of various cells and structures. Over time, the skin will age, which is not only influenced by internal factors, but also by external environmental factors, especially ultraviolet radiation. Aging causes immune system weakening in the elderly, which makes them more susceptible to dermatosis, such as type 2 inflammatory mediated pruritus. The immune response in this condition is marked by senescent cells consistently releasing low amounts of pro-inflammatory cytokines through a senescence-associated secretory phenotype (SASP). This continuous inflammation may accelerate immune system aging and establish a connection between immune aging and type 2 inflammatory skin diseases. In addition, two chronic pigmentation disorders, vitiligo and chloasma, are also associated with skin aging. Aged cells escape the immune system and accumulate in tissues, forming a microenvironment that promotes cancer. At the same time, "photoaging" caused by excessive exposure to ultraviolet radiation is also an important cause of skin cancer. This manuscript describes the possible links between skin aging and type 2 inflammation, chronic pigmentation disorders, and skin cancer and suggests some treatment options.
Collapse
Affiliation(s)
- Meiqi Zhang
- School of Medical and Life Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yumeng Lin
- Health Management Center, Nanjing Tongren Hospital, School of Medicine, Southeast University, Nanjing, China
| | - Zhongyu Han
- School of Medical and Life Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, China
- Zhongda Hospital, School of Medicine, Southeast University, Nanjing, China
| | - Xuewen Huang
- School of Medical and Life Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, China
- Science and Education Department, Chengdu Xinhua Hospital Affiliated to North Sichuan Medical College, Chengdu, China
| | - Shuwei Zhou
- Jiangsu Key Laboratory of Molecular and Functional Imaging, Department of Radiology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, China
| | - Siyu Wang
- Science and Education Department, Chengdu Xinhua Hospital Affiliated to North Sichuan Medical College, Chengdu, China
- Department of Gastroenterology, The First Hospital of Hunan University of Chinese Medicine, Changsha, China
| | - Yan Zhou
- Science and Education Department, Chengdu Xinhua Hospital Affiliated to North Sichuan Medical College, Chengdu, China
- Department of Dermatology, Guangzhou Dermatology Hospital, Guangzhou, China
| | - Xuan Han
- Science and Education Department, Chengdu Xinhua Hospital Affiliated to North Sichuan Medical College, Chengdu, China
- First Clinical College of Changzhi Medical College, Changzhi, China
| | - Haoran Chen
- Science and Education Department, Chengdu Xinhua Hospital Affiliated to North Sichuan Medical College, Chengdu, China
| |
Collapse
|
4
|
Bernhardt A, Jamil A, Morshed MT, Ponnath P, Gille V, Stephan N, Sauer H, Wartenberg M. Oxidative stress and regulation of adipogenic differentiation capacity by sirtuins in adipose stem cells derived from female patients of advancing age. Sci Rep 2024; 14:19885. [PMID: 39191852 DOI: 10.1038/s41598-024-70382-x] [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: 03/12/2024] [Accepted: 08/16/2024] [Indexed: 08/29/2024] Open
Abstract
Patient age is critical for mesenchymal stem cell quality and differentiation capacity. We demonstrate that proliferation and adipogenic capacity of subcutaneous adipose stem cells (ASCs) from female patients declined with advanced age, associated with reduction in cell nucleus size, increase in nuclear lamina protein lamin B1/B2, and lamin A, upregulation of senescence marker p16INK4a and senescence-associated β-galactosidase activity. Adipogenic induction resulted in differentiation of adipocytes and upregulation of adipogenic genes CCAAT enhancer binding protein alpha, fatty acid binding protein 4, lipoprotein lipase, and peroxisome proliferator-activated receptor-γ, which was not affected by the Sirt-1 activator YK-3-237 or the Sirt-1 inhibitor EX-527. Protein expression of the stem cell markers Oct4 and Sox2 was not significantly downregulated with advanced patient age. Mitochondrial reactive oxygen species were increased in ASCs from old-aged patients, whereas protein expression of NADPH oxidases NOX1 and NOX4 was downregulated, and dual oxidase isoforms remained unchanged. Generation of nitric oxide and iNOS expression was downregulated. Protein expression of Sirt-1 and Sirt-3 decreased with patient age, whereas Sirt-2 and Sirt-5 remained unchanged. Induction of adipogenesis stimulated protein expression of Sirt-1 and Sirt-3, which was not affected upon pre-incubation with the Sirt-1-activator YK-3-237 or the Sirt-1-inhibitor EX-527. The Sirt-1 inhibitor Sirtinol downregulated adiponectin protein expression and the number of adipocytes, whereas YK-3-237 exerted stimulatory effects. In summary, our data demonstrate increased oxidative stress in ASCs of aging patients, and decline of adipogenic capacity due to Sirt-1- mediated adiponectin downregulation in elderly patients.
Collapse
Affiliation(s)
- Anne Bernhardt
- Department of Physiology, Justus Liebig University Giessen, Giessen, Germany
- Department of Internal Medicine I, Division of Cardiology, University Hospital Jena, Friedrich Schiller University, Am Klinikum 1, 07747, Jena, Germany
| | - Alan Jamil
- Department of Physiology, Justus Liebig University Giessen, Giessen, Germany
| | - Md Tanvir Morshed
- Department of Physiology, Justus Liebig University Giessen, Giessen, Germany
| | - Pia Ponnath
- Department of Internal Medicine I, Division of Cardiology, University Hospital Jena, Friedrich Schiller University, Am Klinikum 1, 07747, Jena, Germany
| | - Veronika Gille
- Department of Internal Medicine I, Division of Cardiology, University Hospital Jena, Friedrich Schiller University, Am Klinikum 1, 07747, Jena, Germany
| | - Nadine Stephan
- Department of Internal Medicine I, Division of Cardiology, University Hospital Jena, Friedrich Schiller University, Am Klinikum 1, 07747, Jena, Germany
| | - Heinrich Sauer
- Department of Physiology, Justus Liebig University Giessen, Giessen, Germany
| | - Maria Wartenberg
- Department of Internal Medicine I, Division of Cardiology, University Hospital Jena, Friedrich Schiller University, Am Klinikum 1, 07747, Jena, Germany.
| |
Collapse
|
5
|
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 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
|
6
|
Llewellyn J, Hubbard SJ, Swift J. Translation is an emerging constraint on protein homeostasis in ageing. Trends Cell Biol 2024; 34:646-656. [PMID: 38423854 DOI: 10.1016/j.tcb.2024.02.001] [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: 08/25/2023] [Revised: 01/23/2024] [Accepted: 02/01/2024] [Indexed: 03/02/2024]
Abstract
Proteins are molecular machines that provide structure and perform vital transport, signalling and enzymatic roles. Proteins expressed by cells require tight regulation of their concentration, folding, localisation, and modifications; however, this state of protein homeostasis is continuously perturbed by tissue-level stresses. While cells in healthy tissues are able to buffer against these perturbations, for example, by expression of chaperone proteins, protein homeostasis is lost in ageing, and can lead to protein aggregation characteristic of protein folding diseases. Here, we review reports of a progressive disconnect between transcriptomic and proteomic regulation during cellular ageing. We discuss how age-associated changes to cellular responses to specific stressors in the tissue microenvironment are exacerbated by loss of ribosomal proteins, ribosomal pausing, and mistranslation.
Collapse
Affiliation(s)
- Jack Llewellyn
- Wellcome Centre for Cell-Matrix Research, Oxford Road, Manchester, M13 9PT, UK; Division of Cell Matrix Biology and Regenerative Medicine, School of Biological Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester, M13 9PT, UK
| | - Simon J Hubbard
- Division of Evolution and Genomic Sciences, School of Biological Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester, M13 9PT, UK.
| | - Joe Swift
- Wellcome Centre for Cell-Matrix Research, Oxford Road, Manchester, M13 9PT, UK; Division of Cell Matrix Biology and Regenerative Medicine, School of Biological Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester, M13 9PT, UK.
| |
Collapse
|
7
|
Tan CYR, Morenc M, Setiawan M, Lim ZZY, Soon AL, Bierman JC, Vires L, Laughlin T, DeAngelis YM, Rovito H, Jarrold BB, Nguyen TQN, Lim JSY, Kent O, Määttä A, Benham AM, Hawkins TJ, Lee XE, Ehrman MC, Oblong JE, Dreesen O, Bellanger S. Para-Hydroxycinnamic Acid Mitigates Senescence and Inflammaging in Human Skin Models. Int J Mol Sci 2024; 25:8153. [PMID: 39125721 PMCID: PMC11312399 DOI: 10.3390/ijms25158153] [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: 06/21/2024] [Revised: 07/21/2024] [Accepted: 07/22/2024] [Indexed: 08/12/2024] Open
Abstract
Para-hydroxycinnamic acid (pHCA) is one of the most abundant naturally occurring hydroxycinnamic acids, a class of chemistries known for their antioxidant properties. In this study, we evaluated the impact of pHCA on different parameters of skin aging in in vitro skin models after H2O2 and UV exposure. These parameters include keratinocyte senescence and differentiation, inflammation, and energy metabolism, as well as the underlying molecular mechanisms. Here we demonstrate that pHCA prevents oxidative stress-induced premature senescence of human primary keratinocytes in both 2D and 3D skin models, while improving clonogenicity in 2D. As aging is linked to inflammation, referred to as inflammaging, we analyzed the release of IL-6, IL-8, and PGE2, known to be associated with senescence. All of them were downregulated by pHCA in both normal and oxidative stress conditions. Mechanistically, DNA damage induced by oxidative stress is prevented by pHCA, while pHCA also exerts a positive effect on the mitochondrial and glycolytic functions under stress. Altogether, these results highlight the protective effects of pHCA against inflammaging, and importantly, help to elucidate its potential mechanisms of action.
Collapse
Affiliation(s)
- Christina Yan Ru Tan
- A*STAR Skin Research Labs (A*SRL), Skin Research Institute of Singapore (SRIS), Agency for Science, Technology and Research (A*STAR), 8A Biomedical Grove, #06-06 Immunos, Singapore 138648, Singapore; (C.Y.R.T.); (M.S.); (Z.Z.Y.L.); (A.L.S.); (T.Q.N.N.); (J.S.Y.L.); (X.E.L.); (O.D.)
| | - Malgorzata Morenc
- A*STAR Skin Research Labs (A*SRL), Skin Research Institute of Singapore (SRIS), Agency for Science, Technology and Research (A*STAR), 8A Biomedical Grove, #06-06 Immunos, Singapore 138648, Singapore; (C.Y.R.T.); (M.S.); (Z.Z.Y.L.); (A.L.S.); (T.Q.N.N.); (J.S.Y.L.); (X.E.L.); (O.D.)
| | - Melina Setiawan
- A*STAR Skin Research Labs (A*SRL), Skin Research Institute of Singapore (SRIS), Agency for Science, Technology and Research (A*STAR), 8A Biomedical Grove, #06-06 Immunos, Singapore 138648, Singapore; (C.Y.R.T.); (M.S.); (Z.Z.Y.L.); (A.L.S.); (T.Q.N.N.); (J.S.Y.L.); (X.E.L.); (O.D.)
| | - Zen Zhi Yan Lim
- A*STAR Skin Research Labs (A*SRL), Skin Research Institute of Singapore (SRIS), Agency for Science, Technology and Research (A*STAR), 8A Biomedical Grove, #06-06 Immunos, Singapore 138648, Singapore; (C.Y.R.T.); (M.S.); (Z.Z.Y.L.); (A.L.S.); (T.Q.N.N.); (J.S.Y.L.); (X.E.L.); (O.D.)
| | - Ai Ling Soon
- A*STAR Skin Research Labs (A*SRL), Skin Research Institute of Singapore (SRIS), Agency for Science, Technology and Research (A*STAR), 8A Biomedical Grove, #06-06 Immunos, Singapore 138648, Singapore; (C.Y.R.T.); (M.S.); (Z.Z.Y.L.); (A.L.S.); (T.Q.N.N.); (J.S.Y.L.); (X.E.L.); (O.D.)
| | - John C. Bierman
- The Procter & Gamble Company, Mason, OH 45040, USA; (J.C.B.); (L.V.); (T.L.); (Y.M.D.); (H.R.); (B.B.J.); (J.E.O.)
| | - Laura Vires
- The Procter & Gamble Company, Mason, OH 45040, USA; (J.C.B.); (L.V.); (T.L.); (Y.M.D.); (H.R.); (B.B.J.); (J.E.O.)
| | - Timothy Laughlin
- The Procter & Gamble Company, Mason, OH 45040, USA; (J.C.B.); (L.V.); (T.L.); (Y.M.D.); (H.R.); (B.B.J.); (J.E.O.)
| | - Yvonne M. DeAngelis
- The Procter & Gamble Company, Mason, OH 45040, USA; (J.C.B.); (L.V.); (T.L.); (Y.M.D.); (H.R.); (B.B.J.); (J.E.O.)
| | - Holly Rovito
- The Procter & Gamble Company, Mason, OH 45040, USA; (J.C.B.); (L.V.); (T.L.); (Y.M.D.); (H.R.); (B.B.J.); (J.E.O.)
| | - Bradley B. Jarrold
- The Procter & Gamble Company, Mason, OH 45040, USA; (J.C.B.); (L.V.); (T.L.); (Y.M.D.); (H.R.); (B.B.J.); (J.E.O.)
| | - Thi Quynh Ngoc Nguyen
- A*STAR Skin Research Labs (A*SRL), Skin Research Institute of Singapore (SRIS), Agency for Science, Technology and Research (A*STAR), 8A Biomedical Grove, #06-06 Immunos, Singapore 138648, Singapore; (C.Y.R.T.); (M.S.); (Z.Z.Y.L.); (A.L.S.); (T.Q.N.N.); (J.S.Y.L.); (X.E.L.); (O.D.)
| | - John Soon Yew Lim
- A*STAR Skin Research Labs (A*SRL), Skin Research Institute of Singapore (SRIS), Agency for Science, Technology and Research (A*STAR), 8A Biomedical Grove, #06-06 Immunos, Singapore 138648, Singapore; (C.Y.R.T.); (M.S.); (Z.Z.Y.L.); (A.L.S.); (T.Q.N.N.); (J.S.Y.L.); (X.E.L.); (O.D.)
| | - Olivia Kent
- Department of Biosciences, Durham University, South Road, Durham DH1 3LE, UK; (O.K.); (A.M.); (A.M.B.); (T.J.H.)
| | - Arto Määttä
- Department of Biosciences, Durham University, South Road, Durham DH1 3LE, UK; (O.K.); (A.M.); (A.M.B.); (T.J.H.)
| | - Adam M. Benham
- Department of Biosciences, Durham University, South Road, Durham DH1 3LE, UK; (O.K.); (A.M.); (A.M.B.); (T.J.H.)
| | - Timothy J. Hawkins
- Department of Biosciences, Durham University, South Road, Durham DH1 3LE, UK; (O.K.); (A.M.); (A.M.B.); (T.J.H.)
| | - Xin Er Lee
- A*STAR Skin Research Labs (A*SRL), Skin Research Institute of Singapore (SRIS), Agency for Science, Technology and Research (A*STAR), 8A Biomedical Grove, #06-06 Immunos, Singapore 138648, Singapore; (C.Y.R.T.); (M.S.); (Z.Z.Y.L.); (A.L.S.); (T.Q.N.N.); (J.S.Y.L.); (X.E.L.); (O.D.)
| | - Matthew C. Ehrman
- Procter & Gamble International Operations SA SG Branch, 70 Biopolis Street, Singapore 138547, Singapore;
| | - John E. Oblong
- The Procter & Gamble Company, Mason, OH 45040, USA; (J.C.B.); (L.V.); (T.L.); (Y.M.D.); (H.R.); (B.B.J.); (J.E.O.)
| | - Oliver Dreesen
- A*STAR Skin Research Labs (A*SRL), Skin Research Institute of Singapore (SRIS), Agency for Science, Technology and Research (A*STAR), 8A Biomedical Grove, #06-06 Immunos, Singapore 138648, Singapore; (C.Y.R.T.); (M.S.); (Z.Z.Y.L.); (A.L.S.); (T.Q.N.N.); (J.S.Y.L.); (X.E.L.); (O.D.)
| | - Sophie Bellanger
- A*STAR Skin Research Labs (A*SRL), Skin Research Institute of Singapore (SRIS), Agency for Science, Technology and Research (A*STAR), 8A Biomedical Grove, #06-06 Immunos, Singapore 138648, Singapore; (C.Y.R.T.); (M.S.); (Z.Z.Y.L.); (A.L.S.); (T.Q.N.N.); (J.S.Y.L.); (X.E.L.); (O.D.)
| |
Collapse
|
8
|
Song Y, Shao L, Yu X. Transcriptome Analysis of Transiently Reversible Cell Vacuolization Caused by Excessive Serum Concentration in Scophthalmus maximus. BIOLOGY 2024; 13:545. [PMID: 39056737 PMCID: PMC11274238 DOI: 10.3390/biology13070545] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/21/2024] [Revised: 07/16/2024] [Accepted: 07/17/2024] [Indexed: 07/28/2024]
Abstract
As an important research tool, cell lines play a vital role in life science research, medical research, and drug development. During the culture of the Scophthalmus maximus head kidney (TK) cell line, we found a phenomenon of cell vacuolization caused by excessive serum concentration. Moreover, the vacuolization of the cells gradually disappeared after passage by trypsin digestion. In clarifying the formation mechanism of this reversible cellular vacuolation, transcriptomics was utilized to explore the mechanism of cell vacuolization caused by excessive serum concentration. Transcriptome analysis indicated that excessive serum concentration could cause the up-regulated expression of PORCN and other genes to promote cell proliferation. Compared with cells whose vacuolization disappeared after trypsin digestion and passage, the expression of mitosis-related genes (BUB1, ttk, Mad2, Cdc20, CDK1, CCNB1), nuclear stability-related genes LMNB1 and tissue stress and repair-related genes HMMR in vacuolated cells caused by excessive serum concentration was significantly up-regulated. There is a regulatory system related to adaptation and stress repair in the cells, which can maintain cell stability to a certain extent. This study provides a theoretical basis for the stable culture of fish cell lines and the solution to the problem of cell vacuolation.
Collapse
Affiliation(s)
- Yuting Song
- College of Marine Life Sciences, Ocean University of China, Qingdao 266003, China;
- Institute of Evolution & Marine Biodiversity, Ocean University of China, Qingdao 266003, China
| | - Lijun Shao
- School of Public Health, Shandong Second Medical University, Weifang 261053, China;
| | - Xiaoli Yu
- School of Public Health, Shandong Second Medical University, Weifang 261053, China;
| |
Collapse
|
9
|
Chauhan R, Gupta A, Dagar G, Sharma S, Sadida HQ, Hashem S, Verghese AM, Tanwar M, Macha MA, Uddin S, Al-Shabeeb Akil AS, Pandita TK, Bhat AA, Singh M. Role of lamins in cellular physiology and cancer. ADVANCES IN PROTEIN CHEMISTRY AND STRUCTURAL BIOLOGY 2024; 143:119-153. [PMID: 39843134 DOI: 10.1016/bs.apcsb.2024.06.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/24/2025]
Abstract
Lamins, which are crucial type V intermediate filament proteins found in the nuclear lamina, are essential for maintaining the stability and function of the nucleus in higher vertebrates. They are classified into A- and B-types, and their distinct expression patterns contribute to cellular survival, development, and functionality. Lamins emerged during the transition from open to closed mitosis, with their complexity increasing alongside organism evolution. Derived from the LMNA, LMNB1, and LMNB2 genes, lamins undergo alternative splicing to produce seven variants, influencing cellular processes such as stiffness, chromatin condensation, and cell cycle regulation. The lamin network interacts with the cytoskeleton via Linkers of the nucleoskeleton to the cytoskeleton (LINC) complexes, playing a critical role in cellular stability and mechanotransduction. Lamins also regulate active transport into and out of the nucleus, affecting nuclear integrity, positioning, DNA maintenance, and gene expression. Genetic mutations in lamin genes lead to laminopathies, highlighting their functional significance and organizational roles. Changes in lamin subtype composition within the nuclear lamina have significant implications for cancer development, impacting cellular stiffness, mobility, and the Epithelial-to-Mesenchymal Transition (EMT). Lamin A/C, in particular, plays multifaceted roles in cancer biology, influencing progression, metastasis, and therapy response through interactions with various proteins and pathways. Dysregulated lamin expression is commonly observed in cancers, suggesting their potential as diagnostic and prognostic markers. This chapter underscores the pivotal roles of lamins in nuclear architecture and cancer biology, emphasizing their impact on cellular functions and disease pathology. Understanding lamin behavior and regulation mechanisms holds promise for developing novel diagnostic tools and targeted therapies in cancer treatment.
Collapse
Affiliation(s)
- Ravi Chauhan
- Department of Medical Oncology (Lab), Dr. B.R. Ambedkar Institute Rotary Cancer Hospital, All India Institute of Medical Sciences, New Delhi, India
| | - Ashna Gupta
- Department of Medical Oncology (Lab), Dr. B.R. Ambedkar Institute Rotary Cancer Hospital, All India Institute of Medical Sciences, New Delhi, India
| | - Gunjan Dagar
- Department of Medical Oncology (Lab), Dr. B.R. Ambedkar Institute Rotary Cancer Hospital, All India Institute of Medical Sciences, New Delhi, India
| | - Shalini Sharma
- Department of Medical Oncology (Lab), Dr. B.R. Ambedkar Institute Rotary Cancer Hospital, All India Institute of Medical Sciences, New Delhi, India
| | - Hana Q Sadida
- Department of Human Genetics-Precision Medicine in Diabetes, Obesity and Cancer Program, Sidra Medicine, Doha, Qatar
| | - Sheema Hashem
- Department of Human Genetics, Sidra Medicine, Doha, Qatar
| | - Ann M Verghese
- Department of Human Genetics, Sidra Medicine, Doha, Qatar
| | - Mukesh Tanwar
- Department of Genetics, Maharishi Dayanand University Rohtak, Haryana, India
| | - Muzafar A Macha
- Watson-Crick Centre for Molecular Medicine, Islamic University of Science and Technology, Jammu and Kashmir, India
| | - Shahab Uddin
- Translational Research Institute, Academic Health System, Hamad Medical Corporation, Doha, Qatar
| | - Ammira S Al-Shabeeb Akil
- Department of Human Genetics-Precision Medicine in Diabetes, Obesity and Cancer Program, Sidra Medicine, Doha, Qatar
| | - Tej K Pandita
- Center for Genomics and Precision Medicine, Texas A&M College of Medicine, Houston, TX, United States
| | - Ajaz A Bhat
- Department of Human Genetics-Precision Medicine in Diabetes, Obesity and Cancer Program, Sidra Medicine, Doha, Qatar.
| | - Mayank Singh
- Department of Medical Oncology (Lab), Dr. B.R. Ambedkar Institute Rotary Cancer Hospital, All India Institute of Medical Sciences, New Delhi, India.
| |
Collapse
|
10
|
de Oliveira RF, Salazar M, Matos L, Almeida H, Rodrigues AR, Gouveia AM. High copper levels induce premature senescence in 3T3-L1 preadipocytes. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2024; 1871:119734. [PMID: 38642724 DOI: 10.1016/j.bbamcr.2024.119734] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Revised: 04/02/2024] [Accepted: 04/12/2024] [Indexed: 04/22/2024]
Abstract
Copper (Cu) dyshomeostasis has been linked to obesity and related morbidities and also to aging. Cu levels are higher in older or obese individuals, and adipose tissue (AT) Cu levels correlate with body mass index. Aging and obesity induce similar AT functional and structural changes, including an accumulation of senescent cells. To study the effect of Cu-mediated stress-induced premature senescent (Cu-SIPS) on preadipocytes, 3T3-L1 cell line was exposed to a subcytotoxic concentration of copper sulfate. After Cu treatment, preadipocytes acquired typical senescence characteristics including diminished cell proliferation, cell and nuclei enlargement and increased lysosomal mass (higher Lamp2 expression and a slight increased number of cells positive for β-galactosidase associated with senescence (SA-β-Gal)). Cell cycle arrest was due to upregulation of p16Ink4aInk4a and p21Waf1/Cip1. Accordingly, protein levels of the proliferation marker KI67 were reduced. Cu-SIPS relates with oxidative stress and, in this context, an increase of SOD1 and HO-1 expression was detected in Cu-treated cells. The mRNA expression of senescence-associated secretory phenotype factors, such as Mmp3, Il-6 and Tnf-α, increased in Cu-SIPS 3T3-L1 cells but no effect was observed on the expression of heterochromatin-associated protein 1(HP1). Although the downregulation of Lamin B1 expression is considered a hallmark of senescence, Cu-SIPS cells presented higher levels of Lamin B1. The dysregulation of nuclear lamina was accompanied by an increase of nuclear blebbing, but not of micronuclei number. To conclude, a Cu-SIPS model in 3T3-L1 preadipocytes is here described, which may be an asset to the study of AT dysregulation observed in obesity and aging.
Collapse
Affiliation(s)
- Ricardo F de Oliveira
- Departamento de Biomedicina, Unidade de Biologia Experimental, Faculdade de Medicina da Universidade do Porto, Alameda Prof Hernâni Monteiro, 4200-319 Porto, Portugal
| | - Maria Salazar
- Departamento de Biomedicina, Unidade de Biologia Experimental, Faculdade de Medicina da Universidade do Porto, Alameda Prof Hernâni Monteiro, 4200-319 Porto, Portugal; i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal; IBMC - Instituto de Biologia Molecular e Celular, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal
| | - Liliana Matos
- Departamento de Biomedicina, Unidade de Biologia Experimental, Faculdade de Medicina da Universidade do Porto, Alameda Prof Hernâni Monteiro, 4200-319 Porto, Portugal; i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal; IBMC - Instituto de Biologia Molecular e Celular, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal; Faculdade de Ciências da Nutrição e Alimentação, Universidade do Porto, Rua Dr. Roberto Frias, 4200-465 Porto, Portugal
| | - Henrique Almeida
- Departamento de Biomedicina, Unidade de Biologia Experimental, Faculdade de Medicina da Universidade do Porto, Alameda Prof Hernâni Monteiro, 4200-319 Porto, Portugal; i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal; IBMC - Instituto de Biologia Molecular e Celular, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal
| | - Adriana R Rodrigues
- Departamento de Biomedicina, Unidade de Biologia Experimental, Faculdade de Medicina da Universidade do Porto, Alameda Prof Hernâni Monteiro, 4200-319 Porto, Portugal; i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal; IBMC - Instituto de Biologia Molecular e Celular, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal; Faculdade de Ciências da Nutrição e Alimentação, Universidade do Porto, Rua Dr. Roberto Frias, 4200-465 Porto, Portugal
| | - Alexandra M Gouveia
- Departamento de Biomedicina, Unidade de Biologia Experimental, Faculdade de Medicina da Universidade do Porto, Alameda Prof Hernâni Monteiro, 4200-319 Porto, Portugal; i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal; IBMC - Instituto de Biologia Molecular e Celular, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal.
| |
Collapse
|
11
|
Kida M, Abe J, Hori H, Hirai Y. PRSS3/mesotrypsin as a putative regulator of the biophysical characteristics of epidermal keratinocytes in superficial layers. Sci Rep 2024; 14:12383. [PMID: 38811772 PMCID: PMC11137022 DOI: 10.1038/s41598-024-63271-w] [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: 03/08/2024] [Accepted: 05/27/2024] [Indexed: 05/31/2024] Open
Abstract
Mesotrypsin, encoded by the PRSS3 gene, is a distinctive trypsin isoform renowned for its exceptional resistance to traditional trypsin inhibitors and unique substrate specificity. Within the skin epidermis, this protein primarily expresses in the upper layers of the stratified epidermis and plays a crucial role in processing pro-filaggrin (Pro-FLG). Although prior studies have partially elucidated its functions using primary cultured keratinocytes, challenges persist due to these cells' differentiation-activated cell death program. In the present study, HaCaT keratinocytes, characterized by minimal endogenous mesotrypsin expression and sustained proliferation in differentiated states, were utilized to further scrutinize the function of mesotrypsin. Despite the ready degradation of the intact form of active mesotrypsin in these cells, fusion with Venus, flanked by a peptide linker, enables evasion from the protein elimination machinery, thus facilitating activation of the Pro-FLG processing system. Inducing Venus-mesotrypsin expression in the cells resulted in a flattened phenotype and reduced proliferative capacity. Moreover, these cells displayed altered F-actin assembly, enhanced E-cadherin adhesive activity, and facilitated tight junction formation without overtly influencing epidermal differentiation. These findings underscore mesotrypsin's potentially pivotal role in shaping the characteristic cellular morphology of upper epidermal layers.
Collapse
Affiliation(s)
- Moeko Kida
- Department of Biomedical Sciences, Graduate School of Science and Technology, Kwansei Gakuin University, 1 Gakuen-Uegahara, Sanda, 669-1330, Japan
| | - Junya Abe
- Department of Biomedical Sciences, Graduate School of Science and Technology, Kwansei Gakuin University, 1 Gakuen-Uegahara, Sanda, 669-1330, Japan
| | - Haruna Hori
- Department of Biomedical Sciences, Graduate School of Science and Technology, Kwansei Gakuin University, 1 Gakuen-Uegahara, Sanda, 669-1330, Japan
| | - Yohei Hirai
- Department of Biomedical Sciences, Graduate School of Science and Technology, Kwansei Gakuin University, 1 Gakuen-Uegahara, Sanda, 669-1330, Japan.
| |
Collapse
|
12
|
Foo MXR, Ong PF, Yap ZX, Maric M, Bong CJS, Dröge P, Burke B, Dreesen O. Genetic and pharmacological modulation of lamin A farnesylation determines its function and turnover. Aging Cell 2024; 23:e14105. [PMID: 38504487 PMCID: PMC11113360 DOI: 10.1111/acel.14105] [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: 10/05/2023] [Revised: 01/10/2024] [Accepted: 01/29/2024] [Indexed: 03/21/2024] Open
Abstract
Hutchinson-Gilford Progeria syndrome (HGPS) is a severe premature ageing disorder caused by a 50 amino acid truncated (Δ50AA) and permanently farnesylated lamin A (LA) mutant called progerin. On a cellular level, progerin expression leads to heterochromatin loss, impaired nucleocytoplasmic transport, telomeric DNA damage and a permanent growth arrest called cellular senescence. Although the genetic basis for HGPS has been elucidated 20 years ago, the question whether the Δ50AA or the permanent farnesylation causes cellular defects has not been addressed. Moreover, we currently lack mechanistic insight into how the only FDA-approved progeria drug Lonafarnib, a farnesyltransferase inhibitor (FTI), ameliorates HGPS phenotypes. By expressing a variety of LA mutants using a doxycycline-inducible system, and in conjunction with FTI, we demonstrate that the permanent farnesylation, and not the Δ50AA, is solely responsible for progerin-induced cellular defects, as well as its rapid accumulation and slow clearance. Importantly, FTI does not affect clearance of progerin post-farnesylation and we demonstrate that early, but not late FTI treatment prevents HGPS phenotypes. Collectively, our study unravels the precise contributions of progerin's permanent farnesylation to its turnover and HGPS cellular phenotypes, and how FTI treatment ameliorates these. These findings are applicable to other diseases associated with permanently farnesylated proteins, such as adult-onset autosomal dominant leukodystrophy.
Collapse
Affiliation(s)
- Mattheus Xing Rong Foo
- A*STAR Skin Research Labs, Cell Ageing Laboratory, Skin Research Institute of Singapore, Singapore, Singapore
| | - Peh Fern Ong
- A*STAR Skin Research Labs, Cell Ageing Laboratory, Skin Research Institute of Singapore, Singapore, Singapore
| | - Zi Xuan Yap
- A*STAR Skin Research Labs, Cell Ageing Laboratory, Skin Research Institute of Singapore, Singapore, Singapore
| | - Martina Maric
- A*STAR Skin Research Labs, Cell Ageing Laboratory, Skin Research Institute of Singapore, Singapore, Singapore
| | - Christopher Jue Shi Bong
- A*STAR Skin Research Labs, Cell Ageing Laboratory, Skin Research Institute of Singapore, Singapore, Singapore
| | - Peter Dröge
- LambdaGen Pte. Ltd., Singapore, Singapore
- School of Biological Sciences, Nanyang Technological University, Singapore, Singapore
| | - Brian Burke
- A*STAR Skin Research Labs, Cell Ageing Laboratory, Skin Research Institute of Singapore, Singapore, Singapore
| | - Oliver Dreesen
- A*STAR Skin Research Labs, Cell Ageing Laboratory, Skin Research Institute of Singapore, Singapore, Singapore
| |
Collapse
|
13
|
Gao S, Chen L, Lin Z, Xu Z, Wang Y, Ling H, Wu Z, Yin Y, Yao W, Wu K, Liu G. 8-Oxoguanine DNA glycosylase protects cells from senescence via the p53-p21 pathway. Acta Biochim Biophys Sin (Shanghai) 2024; 56:184-198. [PMID: 38282476 PMCID: PMC10984855 DOI: 10.3724/abbs.2023264] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Accepted: 09/08/2023] [Indexed: 01/30/2024] Open
Abstract
Cellular senescence is an important factor leading to pulmonary fibrosis. Deficiency of 8-oxoguanine DNA glycosylase (OGG1) in mice leads to alleviation of bleomycin (BLM)-induced mouse pulmonary fibrosis, and inhibition of the OGG1 enzyme reduces the epithelial mesenchymal transition (EMT) in lung cells. In the present study, we find decreased expression of OGG1 in aged mice and BLM-induced cell senescence. In addition, a decrease in OGG1 expression results in cell senescence, such as increases in the percentage of SA-β-gal-positive cells, and in the p21 and p-H2AX protein levels in response to BLM in lung cells. Furthermore, OGG1 promotes cell transformation in A549 cells in the presence of BLM. We also find that OGG1 siRNA impedes cell cycle progression and inhibits the levels of telomerase reverse transcriptase (TERT) and LaminB1 in BLM-treated lung cells. The increase in OGG1 expression results in the opposite phenomenon. The mRNA levels of senescence-associated secretory phenotype (SASP) components, including IL-1α, IL-1β, IL-6, IL-8, CXCL1/CXCL2, and MMP-3, in the absence of OGG1 are obviously increased in A549 cells treated with BLM. Interestingly, we demonstrate that OGG1 binds to p53 to inhibit the activation of p53 and that silencing of p53 reverses the inhibition of OGG1 on senescence in lung cells. Additionally, the augmented cell senescence is shown in vivo in OGG1-deficient mice. Overall, we provide direct evidence in vivo and in vitro that OGG1 plays an important role in protecting tissue cells against aging associated with the p53 pathway.
Collapse
Affiliation(s)
- Shenglan Gao
- Clinical Research CenterAffiliated Hospital of Guangdong Medical UniversityZhanjiang524001China
| | - Lujun Chen
- Department of Cardiovascularthe Affiliated Hospital of Guangdong Medical UniversityZhanjiang524001China
| | - Ziying Lin
- State Key Laboratory of Respiratory DiseasesGuangdong Key Laboratory of Vascular DiseasesNational Clinical Research Center for Respiratory DiseasesGuangzhou Institute of Respiratory Healththe First Affiliated Hospital of Guangzhou Medical UniversityGuangzhou510120China
| | - Zhiliang Xu
- Clinical Research CenterAffiliated Hospital of Guangdong Medical UniversityZhanjiang524001China
| | - Yahong Wang
- Clinical Research CenterAffiliated Hospital of Guangdong Medical UniversityZhanjiang524001China
| | - Huayu Ling
- Department of Respiratory and Critical Care MedicineAffiliated Hospital of Guangdong Medical UniversityZhanjiang524001China
| | - Zijun Wu
- Department of Cardiovascularthe Affiliated Hospital of Guangdong Medical UniversityZhanjiang524001China
| | - Yu Yin
- Department of Respiratory and Critical Care MedicineAffiliated Hospital of Guangdong Medical UniversityZhanjiang524001China
| | - Weimin Yao
- Department of Respiratory and Critical Care MedicineAffiliated Hospital of Guangdong Medical UniversityZhanjiang524001China
| | - Keng Wu
- Department of Cardiovascularthe Affiliated Hospital of Guangdong Medical UniversityZhanjiang524001China
| | - Gang Liu
- Clinical Research CenterAffiliated Hospital of Guangdong Medical UniversityZhanjiang524001China
- Department of Respiratory and Critical Care MedicineTangdu HospitalAir Force Military Medical UniversityXi’an710038China
| |
Collapse
|
14
|
Kodera K, Hishida R, Sakai A, Nyuzuki H, Matsui N, Yamanaka T, Saitoh A, Matsui H. GPATCH4 contributes to nucleolus morphology and its dysfunction impairs cell viability. Biochem Biophys Res Commun 2024; 693:149384. [PMID: 38113722 DOI: 10.1016/j.bbrc.2023.149384] [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: 11/14/2023] [Accepted: 12/11/2023] [Indexed: 12/21/2023]
Abstract
The nucleolus serves a multifaceted role encompassing not only rRNA transcription and ribosome synthesis, but also the intricate orchestration of cell cycle regulation and the modulation of cellular senescence. G-patch domain containing 4 (GPATCH4) stands as one among the nucleolar proteins; however, its functional significances remain still unclear. In order to elucidate the functions of GPATCH4, we examined the effects of its dysfunction on cellular proliferation, alterations in nucleolar architecture, apoptotic events, and cellular senescence. Through experimentation conducted on cultured neuroblastoma SH-SY5Y cells, the reduction of GPATCH4 caused inhibition of cellular proliferation, concurrently fostering escalated apoptotic susceptibilities upon exposure to high-dose etoposide. In the realm of nucleolar morphology comparisons, a discernible decline was noted in the count of nucleoli per nucleus, concomitant with a significant expansion in the area occupied by individual nucleoli. Upon induction of senescence prompted by low-dose etoposide, GPATCH4 knockdown resulted in decreased cell viability and increased expression of senescence-associated markers, namely senescence-associated β-galactosidase (SA-β-GAL) and p16. Furthermore, GPATCH4 dysfunction elicited alterations in the gene expression profile of the ribosomal system. In sum, our findings showed that GPATCH4 is a pivotal nucleolar protein that regulates nucleolar morphology and is correlated with cell viability.
Collapse
Affiliation(s)
- Kazuki Kodera
- Department of Neuroscience of Disease, Brain Research Institute, Niigata University, Niigata, 951-8585, Japan; Department of Paediatrics, Niigata University Graduate School of Medical and Dental Sciences, Niigata, 951-8510, Japan
| | - Ryuichi Hishida
- Department of Neuroscience of Disease, Brain Research Institute, Niigata University, Niigata, 951-8585, Japan
| | - Akiko Sakai
- Department of Neuroscience of Disease, Brain Research Institute, Niigata University, Niigata, 951-8585, Japan
| | - Hiromi Nyuzuki
- Department of Paediatrics, Niigata University Graduate School of Medical and Dental Sciences, Niigata, 951-8510, Japan
| | - Noriko Matsui
- Department of Neuroscience of Disease, Brain Research Institute, Niigata University, Niigata, 951-8585, Japan
| | - Tomoyuki Yamanaka
- Department of Neuroscience of Disease, Brain Research Institute, Niigata University, Niigata, 951-8585, Japan
| | - Akihiko Saitoh
- Department of Paediatrics, Niigata University Graduate School of Medical and Dental Sciences, Niigata, 951-8510, Japan
| | - Hideaki Matsui
- Department of Neuroscience of Disease, Brain Research Institute, Niigata University, Niigata, 951-8585, Japan.
| |
Collapse
|
15
|
Martins C, Magalhães S, Almeida I, Neto V, Rebelo S, Nunes A. Metabolomics to Study Human Aging: A Review. Curr Mol Med 2024; 24:457-477. [PMID: 37026499 DOI: 10.2174/1566524023666230407123727] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Revised: 02/17/2023] [Accepted: 02/17/2023] [Indexed: 04/08/2023]
Abstract
In the last years, with the increase in the average life expectancy, the world's population is progressively aging, which entails social, health and economic problems. In this sense, the need to better understand the physiology of the aging process becomes an urgent need. Since the study of aging in humans is challenging, cellular and animal models are widely used as alternatives. Omics, namely metabolomics, have emerged in the study of aging, with the aim of biomarker discovering, which may help to uncomplicate this complex process. This paper aims to summarize different models used for aging studies with their advantages and limitations. Also, this review gathers the published articles referring to biomarkers of aging already discovered using metabolomics approaches, comparing the results obtained in the different studies. Finally, the most frequently used senescence biomarkers are described, along with their importance in understanding aging.
Collapse
Affiliation(s)
- Claudia Martins
- Department of Medical Sciences, iBiMED: Institute of Biomedicine, University of Aveiro, Agra do Crasto, Aveiro 3810-193, Portugal
| | - Sandra Magalhães
- Department of Surgery and Physiology, Faculty of Medicine, UnIC@RISE, Cardiovascular Research & Development Centre, University of Porto, Alameda Prof. Hernâni Monteiro, Porto 4200-319, Portugal
| | - Idália Almeida
- Department of Medical Sciences, iBiMED: Institute of Biomedicine, University of Aveiro, Agra do Crasto, Aveiro 3810-193, Portugal
- CICECO: Aveiro Institute of Materials, Department of Chemistry, University of Aveiro, Campus Universitário de Santiago, Aveiro 3810-193, Portugal
| | - Vanessa Neto
- Department of Medical Sciences, iBiMED: Institute of Biomedicine, University of Aveiro, Agra do Crasto, Aveiro 3810-193, Portugal
| | - Sandra Rebelo
- Department of Medical Sciences, iBiMED: Institute of Biomedicine, University of Aveiro, Agra do Crasto, Aveiro 3810-193, Portugal
| | - Alexandra Nunes
- Department of Medical Sciences, iBiMED: Institute of Biomedicine, University of Aveiro, Agra do Crasto, Aveiro 3810-193, Portugal
| |
Collapse
|
16
|
Kim Y. The impact of altered lamin B1 levels on nuclear lamina structure and function in aging and human diseases. Curr Opin Cell Biol 2023; 85:102257. [PMID: 37806292 DOI: 10.1016/j.ceb.2023.102257] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Revised: 09/11/2023] [Accepted: 09/13/2023] [Indexed: 10/10/2023]
Abstract
The role of lamin B1 in human health and aging has attracted increasing attention as mounting evidence reveals its significance in diverse cellular processes. Both upregulation and downregulation of lamin B1 have been implicated in age-associated organ dysfunctions and various human diseases, including central nervous system disorders. Additionally, lamin B1 levels undergo alterations in cancer cells, and a tumor-specific association exists between lamin B1 abundance and cancer aggressiveness. Investigating the connectivity between lamin B1 abundance and human health is of utmost importance for further research. This review presents recent advancements in understanding lamin B1's role in nuclear lamina function and its implications for human health.
Collapse
Affiliation(s)
- Youngjo Kim
- Department of Integrated Biomedical Science and Soonchunhyang Institute of Medi-Bioscience, Soonchunhyang University, Cheon-an 31151, Republic of Korea.
| |
Collapse
|
17
|
Chin T, Lee XE, Ng PY, Lee Y, Dreesen O. The role of cellular senescence in skin aging and age-related skin pathologies. Front Physiol 2023; 14:1297637. [PMID: 38074322 PMCID: PMC10703490 DOI: 10.3389/fphys.2023.1297637] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Accepted: 11/07/2023] [Indexed: 12/21/2024] Open
Abstract
Aging is the result of a gradual functional decline at the cellular, and ultimately, organismal level, resulting in an increased risk of developing a variety of chronic illnesses, such as cardiovascular disease, stroke, cancer and diabetes. The skin is the largest organ of the human body, and the site where signs of aging are most visible. These signs include thin and dry skin, sagging, loss of elasticity, wrinkles, as well as aberrant pigmentation. The appearance of these features is accelerated by exposure to extrinsic factors such as ultraviolet (UV) radiation or pollution, as well as intrinsic factors including time, genetics, and hormonal changes. At the cellular level, aging is associated with impaired proteostasis and an accumulation of macromolecular damage, genomic instability, chromatin reorganization, telomere shortening, remodelling of the nuclear lamina, proliferation defects and premature senescence. Cellular senescence is a state of permanent growth arrest and a key hallmark of aging in many tissues. Due to their inability to proliferate, senescent cells no longer contribute to tissue repair or regeneration. Moreover, senescent cells impair tissue homeostasis, promote inflammation and extracellular matrix (ECM) degradation by secreting molecules collectively known as the "senescence-associated secretory phenotype" (SASP). Senescence can be triggered by a number of different stimuli such as telomere shortening, oncogene expression, or persistent activation of DNA damage checkpoints. As a result, these cells accumulate in aging tissues, including human skin. In this review, we focus on the role of cellular senescence during skin aging and the development of age-related skin pathologies, and discuss potential strategies to rejuvenate aged skin.
Collapse
Affiliation(s)
- Toby Chin
- Lee Kong Chiang School of Medicine, Nanyang Technological University, Singapore, Singapore
| | - Xin Er Lee
- A*STAR Skin Research Labs (A*SRL), Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | - Pei Yi Ng
- A*STAR Skin Research Labs (A*SRL), Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | - Yaelim Lee
- Mechanobiology Institute, National University of Singapore, T-Lab, Singapore, Singapore
| | - Oliver Dreesen
- A*STAR Skin Research Labs (A*SRL), Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
- Mechanobiology Institute, National University of Singapore, T-Lab, Singapore, Singapore
| |
Collapse
|
18
|
Shevelyov YY. Interactions of Chromatin with the Nuclear Lamina and Nuclear Pore Complexes. Int J Mol Sci 2023; 24:15771. [PMID: 37958755 PMCID: PMC10649103 DOI: 10.3390/ijms242115771] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2023] [Revised: 10/26/2023] [Accepted: 10/29/2023] [Indexed: 11/15/2023] Open
Abstract
Heterochromatin and euchromatin form different spatial compartments in the interphase nucleus, with heterochromatin being localized mainly at the nuclear periphery. The mechanisms responsible for peripheral localization of heterochromatin are still not fully understood. The nuclear lamina and nuclear pore complexes were obvious candidates for the role of heterochromatin binders. This review is focused on recent studies showing that heterochromatin interactions with the nuclear lamina and nuclear pore complexes maintain its peripheral localization. Differences in chromatin interactions with the nuclear envelope in cell populations and in individual cells are also discussed.
Collapse
Affiliation(s)
- Yuri Y Shevelyov
- Laboratory of Analysis of Gene Regulation, National Research Centre "Kurchatov Institute", Kurchatov Sq. 2, 123182 Moscow, Russia
| |
Collapse
|
19
|
Wyles SP, Carruthers JD, Dashti P, Yu G, Yap JQ, Gingery A, Tchkonia T, Kirkland JL. Cellular Senescence in Human Skin Aging: Leveraging Senotherapeutics. Gerontology 2023; 70:7-14. [PMID: 37879300 PMCID: PMC10873061 DOI: 10.1159/000534756] [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: 12/26/2022] [Accepted: 10/18/2023] [Indexed: 10/27/2023] Open
Abstract
BACKGROUND As the largest organ in the human body, the skin is continuously exposed to intrinsic and extrinsic stimuli that impact its functionality and morphology with aging. Skin aging entails dysregulation of skin cells and loss, fragmentation, or fragility of extracellular matrix fibers that are manifested macroscopically by wrinkling, laxity, and pigmentary abnormalities. Age-related skin changes are the focus of many surgical and nonsurgical treatments aimed at improving overall skin appearance and health. SUMMARY As a hallmark of aging, cellular senescence, an essentially irreversible cell cycle arrest with apoptosis resistance and a secretory phenotype, manifests across skin layers by affecting epidermal and dermal cells. Knowledge of skin-specific senescent cells, such as melanocytes (epidermal aging) and fibroblasts (dermal aging), will promote our understanding of age-related skin changes and how to optimize patient outcomes in esthetic procedures. KEY MESSAGES This review provides an overview of skin aging in the context of cellular senescence and discusses senolytic intervention strategies to selectively target skin senescent cells that contribute to premature skin aging.
Collapse
Affiliation(s)
- Saranya P. Wyles
- Department of Dermatology, Mayo Clinic, Rochester, MN, United States
| | - Jean D. Carruthers
- Department of Ophthalmology and Visual Sciences, University of British Columbia, Vancouver, BC, Canada
| | - Parisa Dashti
- Department of Dermatology, Mayo Clinic, Rochester, MN, United States
| | - Grace Yu
- Mayo Clinic Graduate School of Biomedical Sciences, Mayo Clinic Alix School of Medicine, and Mayo Clinic Medical Scientist Training Program, Rochester, MN
| | - Jane Q. Yap
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, United States
| | - Anne Gingery
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, MN, United States
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN United States
| | - Tamar Tchkonia
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, United States
| | - James L. Kirkland
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, United States
- Division of General Internal Medicine, Department of Medicine, Mayo Clinic, Rochester, MN, United States
| |
Collapse
|
20
|
Tanaka S, Sakaue H, Koiwai T, Okuda N, Okuyama K, Horioka Y, Hiramatsu Y, Kawashima M, Ishiguro N, Sato T. Near-infrared radiation causes sebaceous gland enlargement along with an ROS-dependent augmentation of epidermal growth factor receptor expression in hamsters. Exp Dermatol 2023; 32:1717-1724. [PMID: 37401827 DOI: 10.1111/exd.14878] [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: 01/27/2023] [Revised: 06/01/2023] [Accepted: 06/25/2023] [Indexed: 07/05/2023]
Abstract
As near-infrared radiation (NIR), which is a composition of sunlight with an 780-1400 nm wavelength, is associated with skin aging such as wrinkles and slacks, the biological actions of NIR with high dermal penetration remains unclear. In the present study, we found that NIR irradiation (40 J/cm2 ) at different levels of irradiance (95-190 mW/cm2 ) using a laboratory device with a xenon flash lamp (780-1700 nm) caused sebaceous gland enlargement concomitantly with skin thickening in the auricle skin of hamsters. The sebaceous gland enlargement resulted from the proliferation of sebocytes due to an increase in the number of proliferating cell nuclear antigen (PCNA)- and lamin B1-positive cells in vivo. In addition, NIR irradiation transcriptionally augmented the production of epidermal growth factor receptor (EGFR) accompanied with an increase in the reactive oxygen species (ROS) level in hamster sebocytes in vitro. Furthermore, the administration of hydrogen peroxide increased the level of EGFR mRNA in the sebocytes. Therefore, these results provide novel evidence that NIR irradiation causes the hyperplasia of sebaceous glands in hamsters by mechanisms in which EGFR production is transcriptionally augmented through ROS-dependent pathways in sebocytes.
Collapse
Affiliation(s)
- Shiho Tanaka
- Department of Biochemistry, Tokyo University of Pharmacy and Life Sciences, Tokyo, Japan
- Department of Dermatology, Tokyo Women's Medical University, Tokyo, Japan
| | - Hiroaki Sakaue
- Department of Biochemistry, Tokyo University of Pharmacy and Life Sciences, Tokyo, Japan
| | - Toshikazu Koiwai
- Department of Biochemistry, Tokyo University of Pharmacy and Life Sciences, Tokyo, Japan
| | - Natsuki Okuda
- Department of Biochemistry, Tokyo University of Pharmacy and Life Sciences, Tokyo, Japan
| | - Katsuki Okuyama
- Department of Biochemistry, Tokyo University of Pharmacy and Life Sciences, Tokyo, Japan
| | | | | | - Makoto Kawashima
- Department of Dermatology, Tokyo Women's Medical University, Tokyo, Japan
| | - Naoko Ishiguro
- Department of Dermatology, Tokyo Women's Medical University, Tokyo, Japan
| | - Takashi Sato
- Department of Biochemistry, Tokyo University of Pharmacy and Life Sciences, Tokyo, Japan
| |
Collapse
|
21
|
Wang D, Amen Y, Elsbaey M, Nagata M, Matsumoto M, Wang D, Shimizu K. Vanilla pompona Leaves and Stems as New Sources of Bioactive Compounds: The Therapeutic Potential for Skin Senescence. PLANTA MEDICA 2023; 89:1259-1268. [PMID: 37459861 DOI: 10.1055/a-2117-9233] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/15/2023]
Abstract
A large variety of natural plants are widely produced and utilised because of their remarkable pharmacological effects. In this study, two phenolic glycosides were isolated for the first time from Vanilla pompona Schiede (Orchidaceae) from Kyushu, Japan: bis [4-(β-D - O-glucopyranosyloxy)-benzyl] (S)-2-isopropylmalate (1: ) and bis 4-[β-D-O-glucopyranosyloxy)-benzyl]-(2R,3S)-2-isopropyl tartrate (2: ). We have discovered that the crude extract of V. pompona leaves and stems and its two phenolic glycosides (compounds 1: - 2: ) are highly effective in reversing skin senescence. V. pompona and compounds 1: - 2: were found to promote the synthesis of collagen, hyaluronic acid, and elastin in skin fibroblasts in a normal skin cell model; in a replicative senescence model, V. pompona and compounds 1: - 2: significantly reduced the ageing phenotype in skin fibroblasts. These compounds also demonstrated a significant protective effect in a UV-induced photo-senescence model; the possible mechanisms of this effect were investigated in this study. To the best of our knowledge, this study is the first to develop V. pompona leaves and stems as new sources of bioactive compounds and to examine their therapeutic potential for skin senescence. The development potential of V. pompona leaves and stems for use in the cosmetics, cosmeceutical, and pharmaceutical industries remains to be investigated.
Collapse
Affiliation(s)
- Duanyang Wang
- Department of Agro-Environmental Sciences, Graduate School of Bioresource and Bioenvironmental Sciences, Kyushu University, Fukuoka, Japan
| | - Yhiya Amen
- Department of Pharmacognosy, Faculty of Pharmacy, Mansoura University, Mansoura, Egypt
| | - Marwa Elsbaey
- Department of Pharmacognosy, Faculty of Pharmacy, Mansoura University, Mansoura, Egypt
| | - Maki Nagata
- Department of Agro-Environmental Sciences, Graduate School of Bioresource and Bioenvironmental Sciences, Kyushu University, Fukuoka, Japan
| | - Masako Matsumoto
- Department of Agro-Environmental Sciences, Graduate School of Bioresource and Bioenvironmental Sciences, Kyushu University, Fukuoka, Japan
| | - Dongmei Wang
- Department of Agro-Environmental Sciences, Graduate School of Bioresource and Bioenvironmental Sciences, Kyushu University, Fukuoka, Japan
| | - Kuniyoshi Shimizu
- Department of Agro-Environmental Sciences, Graduate School of Bioresource and Bioenvironmental Sciences, Kyushu University, Fukuoka, Japan
| |
Collapse
|
22
|
Kim BH, Chung YH, Woo TG, Kang SM, Park S, Park BJ. Progerin, an Aberrant Spliced Form of Lamin A, Is a Potential Therapeutic Target for HGPS. Cells 2023; 12:2299. [PMID: 37759521 PMCID: PMC10527460 DOI: 10.3390/cells12182299] [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: 08/08/2023] [Revised: 09/14/2023] [Accepted: 09/15/2023] [Indexed: 09/29/2023] Open
Abstract
Hutchinson-Gilford progeria syndrome (HGPS) is an extremely rare genetic disorder caused by the mutant protein progerin, which is expressed by the abnormal splicing of the LMNA gene. HGPS affects systemic levels, with the exception of cognition or brain development, in children, showing that cellular aging can occur in the short term. Studying progeria could be useful in unraveling the causes of human aging (as well as fatal age-related disorders). Elucidating the clear cause of HGPS or the development of a therapeutic medicine could improve the quality of life and extend the survival of patients. This review aimed to (i) briefly describe how progerin was discovered as the causative agent of HGPS, (ii) elucidate the puzzling observation of the absence of primary neurological disease in HGPS, (iii) present several studies showing the deleterious effects of progerin and the beneficial effects of its inhibition, and (iv) summarize research to develop a therapy for HGPS and introduce clinical trials for its treatment.
Collapse
Affiliation(s)
- Bae-Hoon Kim
- Rare Disease R&D Center, PRG S&T Co., Ltd., Busan 46274, Republic of Korea; (B.-H.K.); (Y.-H.C.); (T.-G.W.)
| | - Yeon-Ho Chung
- Rare Disease R&D Center, PRG S&T Co., Ltd., Busan 46274, Republic of Korea; (B.-H.K.); (Y.-H.C.); (T.-G.W.)
| | - Tae-Gyun Woo
- Rare Disease R&D Center, PRG S&T Co., Ltd., Busan 46274, Republic of Korea; (B.-H.K.); (Y.-H.C.); (T.-G.W.)
| | - So-Mi Kang
- Department of Molecular Biology, College of Natural Science, Pusan National University, Busan 46231, Republic of Korea; (S.-M.K.); (S.P.)
| | - Soyoung Park
- Department of Molecular Biology, College of Natural Science, Pusan National University, Busan 46231, Republic of Korea; (S.-M.K.); (S.P.)
| | - Bum-Joon Park
- Rare Disease R&D Center, PRG S&T Co., Ltd., Busan 46274, Republic of Korea; (B.-H.K.); (Y.-H.C.); (T.-G.W.)
- Department of Molecular Biology, College of Natural Science, Pusan National University, Busan 46231, Republic of Korea; (S.-M.K.); (S.P.)
| |
Collapse
|
23
|
Murray A, Gough G, Cindrić A, Vučković F, Koschut D, Borelli V, Petrović DJ, Bekavac A, Plećaš A, Hribljan V, Brunmeir R, Jurić J, Pučić-Baković M, Slana A, Deriš H, Frkatović A, Groet J, O'Brien NL, Chen HY, Yeap YJ, Delom F, Havlicek S, Gammon L, Hamburg S, Startin C, D'Souza H, Mitrečić D, Kero M, Odak L, Krušlin B, Krsnik Ž, Kostović I, Foo JN, Loh YH, Dunn NR, de la Luna S, Spector T, Barišić I, Thomas MSC, Strydom A, Franceschi C, Lauc G, Krištić J, Alić I, Nižetić D. Dose imbalance of DYRK1A kinase causes systemic progeroid status in Down syndrome by increasing the un-repaired DNA damage and reducing LaminB1 levels. EBioMedicine 2023; 94:104692. [PMID: 37451904 PMCID: PMC10435767 DOI: 10.1016/j.ebiom.2023.104692] [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: 02/02/2023] [Revised: 06/15/2023] [Accepted: 06/20/2023] [Indexed: 07/18/2023] Open
Abstract
BACKGROUND People with Down syndrome (DS) show clinical signs of accelerated ageing. Causative mechanisms remain unknown and hypotheses range from the (essentially untreatable) amplified-chromosomal-instability explanation, to potential actions of individual supernumerary chromosome-21 genes. The latter explanation could open a route to therapeutic amelioration if the specific over-acting genes could be identified and their action toned-down. METHODS Biological age was estimated through patterns of sugar molecules attached to plasma immunoglobulin-G (IgG-glycans, an established "biological-ageing-clock") in n = 246 individuals with DS from three European populations, clinically characterised for the presence of co-morbidities, and compared to n = 256 age-, sex- and demography-matched healthy controls. Isogenic human induced pluripotent stem cell (hiPSCs) models of full and partial trisomy-21 with CRISPR-Cas9 gene editing and two kinase inhibitors were studied prior and after differentiation to cerebral organoids. FINDINGS Biological age in adults with DS is (on average) 18.4-19.1 years older than in chronological-age-matched controls independent of co-morbidities, and this shift remains constant throughout lifespan. Changes are detectable from early childhood, and do not require a supernumerary chromosome, but are seen in segmental duplication of only 31 genes, along with increased DNA damage and decreased levels of LaminB1 in nucleated blood cells. We demonstrate that these cell-autonomous phenotypes can be gene-dose-modelled and pharmacologically corrected in hiPSCs and derived cerebral organoids. Using isogenic hiPSC models we show that chromosome-21 gene DYRK1A overdose is sufficient and necessary to cause excess unrepaired DNA damage. INTERPRETATION Explanation of hitherto observed accelerated ageing in DS as a developmental progeroid syndrome driven by DYRK1A overdose provides a target for early pharmacological preventative intervention strategies. FUNDING Main funding came from the "Research Cooperability" Program of the Croatian Science Foundation funded by the European Union from the European Social Fund under the Operational Programme Efficient Human Resources 2014-2020, Project PZS-2019-02-4277, and the Wellcome Trust Grants 098330/Z/12/Z and 217199/Z/19/Z (UK). All other funding is described in details in the "Acknowledgements".
Collapse
Affiliation(s)
- Aoife Murray
- Faculty of Medicine and Dentistry, Blizard Institute, Queen Mary University of London, London, UK; The London Down Syndrome Consortium (LonDownS), London, UK.
| | - Gillian Gough
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore
| | - Ana Cindrić
- Glycoscience Research Laboratory, Genos Ltd., Zagreb, Croatia
| | - Frano Vučković
- Glycoscience Research Laboratory, Genos Ltd., Zagreb, Croatia
| | - David Koschut
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore; Disease Intervention Technology Laboratory (DITL), Institute of Molecular and Cellular Biology (IMCB), Agency for Science, Technology and Research (A∗STAR), Singapore
| | - Vincenzo Borelli
- Department of Medical and Surgical Sciences, Alma Mater Studiorum University of Bologna, Italy
| | - Dražen J Petrović
- Glycoscience Research Laboratory, Genos Ltd., Zagreb, Croatia; Croatian Institute for Brain Research, School of Medicine, University of Zagreb, Zagreb, Croatia
| | - Ana Bekavac
- Croatian Institute for Brain Research, School of Medicine, University of Zagreb, Zagreb, Croatia
| | - Ante Plećaš
- Faculty of Veterinary Medicine, Department of Anatomy, Histology and Embryology, University of Zagreb, Zagreb, Croatia
| | - Valentina Hribljan
- Croatian Institute for Brain Research, School of Medicine, University of Zagreb, Zagreb, Croatia
| | - Reinhard Brunmeir
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore
| | - Julija Jurić
- Glycoscience Research Laboratory, Genos Ltd., Zagreb, Croatia
| | | | - Anita Slana
- Glycoscience Research Laboratory, Genos Ltd., Zagreb, Croatia
| | - Helena Deriš
- Glycoscience Research Laboratory, Genos Ltd., Zagreb, Croatia
| | - Azra Frkatović
- Glycoscience Research Laboratory, Genos Ltd., Zagreb, Croatia
| | - Jűrgen Groet
- Faculty of Medicine and Dentistry, Blizard Institute, Queen Mary University of London, London, UK; The London Down Syndrome Consortium (LonDownS), London, UK
| | - Niamh L O'Brien
- Faculty of Medicine and Dentistry, Blizard Institute, Queen Mary University of London, London, UK; The London Down Syndrome Consortium (LonDownS), London, UK
| | - Hong Yu Chen
- Institute of Molecular and Cell Biology (IMCB), A∗STAR, Singapore
| | - Yee Jie Yeap
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore
| | - Frederic Delom
- Faculty of Medicine and Dentistry, Blizard Institute, Queen Mary University of London, London, UK
| | - Steven Havlicek
- Laboratory of Neurogenetics, Genome Institute of Singapore, A∗STAR, Singapore
| | - Luke Gammon
- Faculty of Medicine and Dentistry, Blizard Institute, Queen Mary University of London, London, UK
| | - Sarah Hamburg
- The London Down Syndrome Consortium (LonDownS), London, UK
| | - Carla Startin
- The London Down Syndrome Consortium (LonDownS), London, UK; Department of Forensic and Neurodevelopmental Sciences, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK; Division of Psychiatry, University College London, London, UK; School of Psychology, University of Roehampton, London, UK
| | - Hana D'Souza
- The London Down Syndrome Consortium (LonDownS), London, UK; Centre for Brain and Cognitive Development, Birkbeck, University of London, London, UK
| | - Dinko Mitrečić
- Croatian Institute for Brain Research, School of Medicine, University of Zagreb, Zagreb, Croatia
| | - Mijana Kero
- Department of Medical Genetics, Children's Hospital Zagreb, Centre of Excellence for Reproductive and Regenerative Medicine, School of Medicine, University of Zagreb, Zagreb, Croatia
| | - Ljubica Odak
- Department of Medical Genetics, Children's Hospital Zagreb, Centre of Excellence for Reproductive and Regenerative Medicine, School of Medicine, University of Zagreb, Zagreb, Croatia
| | - Božo Krušlin
- Department of Pathology, School of Medicine, University of Zagreb, Zagreb, Croatia
| | - Željka Krsnik
- Croatian Institute for Brain Research, School of Medicine, University of Zagreb, Zagreb, Croatia
| | - Ivica Kostović
- Croatian Institute for Brain Research, School of Medicine, University of Zagreb, Zagreb, Croatia
| | - Jia Nee Foo
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore; Laboratory of Neurogenetics, Genome Institute of Singapore, A∗STAR, Singapore
| | - Yuin-Han Loh
- Institute of Molecular and Cell Biology (IMCB), A∗STAR, Singapore
| | - Norris Ray Dunn
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore; Institute of Molecular and Cell Biology (IMCB), A∗STAR, Singapore
| | - Susana de la Luna
- ICREA, Genome Biology Programme (CRG), Universitat Pompeu Fabra (UPF), CIBER of Rare Diseases, Barcelona, Spain
| | - Tim Spector
- Department of Twin Research and Genetic Epidemiology, King's College London, London, UK
| | - Ingeborg Barišić
- Department of Medical Genetics, Children's Hospital Zagreb, Centre of Excellence for Reproductive and Regenerative Medicine, School of Medicine, University of Zagreb, Zagreb, Croatia
| | - Michael S C Thomas
- The London Down Syndrome Consortium (LonDownS), London, UK; Centre for Brain and Cognitive Development, Birkbeck, University of London, London, UK
| | - Andre Strydom
- The London Down Syndrome Consortium (LonDownS), London, UK; Department of Forensic and Neurodevelopmental Sciences, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK; Division of Psychiatry, University College London, London, UK
| | - Claudio Franceschi
- Department of Medical and Surgical Sciences, Alma Mater Studiorum University of Bologna, Italy; Institute of Information Technologies, Mathematics and Mechanics, Lobachevsky State University, Nizhny Novgorod 603022, Russia
| | - Gordan Lauc
- Glycoscience Research Laboratory, Genos Ltd., Zagreb, Croatia; Faculty of Pharmacy and Biochemistry, University of Zagreb, Zagreb, Croatia
| | | | - Ivan Alić
- Faculty of Medicine and Dentistry, Blizard Institute, Queen Mary University of London, London, UK; Faculty of Veterinary Medicine, Department of Anatomy, Histology and Embryology, University of Zagreb, Zagreb, Croatia.
| | - Dean Nižetić
- Faculty of Medicine and Dentistry, Blizard Institute, Queen Mary University of London, London, UK; The London Down Syndrome Consortium (LonDownS), London, UK; Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore.
| |
Collapse
|
24
|
Song C, Hu Z, Xu D, Bian H, Lv J, Zhu X, Zhang Q, Su L, Yin H, Lu T, Li Y. STING signaling in inflammaging: a new target against musculoskeletal diseases. Front Immunol 2023; 14:1227364. [PMID: 37492580 PMCID: PMC10363987 DOI: 10.3389/fimmu.2023.1227364] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Accepted: 06/20/2023] [Indexed: 07/27/2023] Open
Abstract
Stimulator of Interferon Gene (STING) is a critical signaling linker protein that plays a crucial role in the intrinsic immune response, particularly in the cytoplasmic DNA-mediated immune response in both pathogens and hosts. It is also involved in various signaling processes in vivo. The musculoskeletal system provides humans with morphology, support, stability, and movement. However, its aging can result in various diseases and negatively impact people's lives. While many studies have reported that cellular aging is a leading cause of musculoskeletal disorders, it also offers insight into potential treatments. Under pathological conditions, senescent osteoblasts, chondrocytes, myeloid cells, and muscle fibers exhibit persistent senescence-associated secretory phenotype (SASP), metabolic disturbances, and cell cycle arrest, which are closely linked to abnormal STING activation. The accumulation of cytoplasmic DNA due to chromatin escape from the nucleus following DNA damage or telomere shortening activates the cGAS-STING signaling pathway. Moreover, STING activation is also linked to mitochondrial dysfunction, epigenetic modifications, and impaired cytoplasmic DNA degradation. STING activation upregulates SASP and autophagy directly and indirectly promotes cell cycle arrest. Thus, STING may be involved in the onset and development of various age-related musculoskeletal disorders and represents a potential therapeutic target. In recent years, many STING modulators have been developed and used in the study of musculoskeletal disorders. Therefore, this paper summarizes the effects of STING signaling on the musculoskeletal system at the molecular level and current understanding of the mechanisms of endogenous active ligand production and accumulation. We also discuss the relationship between some age-related musculoskeletal disorders and STING, as well as the current status of STING modulator development.
Collapse
Affiliation(s)
- Chenyu Song
- Jiangsu CM Clinical Innovation Center of Degenerative Bone & Joint Disease, Wuxi TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Wuxi, China
- Institute of Translational Medicine, Shanghai University, Shanghai, China
| | - Zhuoyi Hu
- Jiangsu CM Clinical Innovation Center of Degenerative Bone & Joint Disease, Wuxi TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Wuxi, China
| | - Dingjun Xu
- Department of Orthopaedics, Wenzhou Hospital of Integrated Traditional Chinese and Western Medicine, Zhejiang, China
| | - Huihui Bian
- Institute of Translational Medicine, Shanghai University, Shanghai, China
| | - Juan Lv
- Institute of Translational Medicine, Shanghai University, Shanghai, China
| | - Xuanxuan Zhu
- Jiangsu CM Clinical Innovation Center of Degenerative Bone & Joint Disease, Wuxi TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Wuxi, China
| | - Qiang Zhang
- Jiangsu CM Clinical Innovation Center of Degenerative Bone & Joint Disease, Wuxi TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Wuxi, China
| | - Li Su
- Institute of Translational Medicine, Shanghai University, Shanghai, China
| | - Heng Yin
- Jiangsu CM Clinical Innovation Center of Degenerative Bone & Joint Disease, Wuxi TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Wuxi, China
| | - Tong Lu
- Department of Critical Care Medicine, Changshu Hospital Affiliated to Nanjing University of Chinese Medicine, Changshu, China
| | - Yinghua Li
- Institute of Translational Medicine, Shanghai University, Shanghai, China
| |
Collapse
|
25
|
Sochorová M, Kremslehner C, Nagelreiter I, Ferrara F, Lisicin MM, Narzt M, Bauer C, Stiegler A, Golabi B, Vávrová K, Gruber F. Deletion of NRF2 disturbs composition, morphology, and differentiation of the murine tail epidermis in chronological aging. Biofactors 2023; 49:684-698. [PMID: 36772996 PMCID: PMC10946746 DOI: 10.1002/biof.1941] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Accepted: 01/09/2023] [Indexed: 02/12/2023]
Abstract
NRF2 is a master regulator of the cellular protection against oxidative damage in mammals and of multiple pathways relevant in the mammalian aging process. In the epidermis of the skin NRF2 contributes additionally to the formation of an antioxidant barrier to protect from environmental insults and is involved in the differentiation process of keratinocytes. In chronological aging of skin, the capacity for antioxidant responses and the ability to restore homeostasis after damage are impaired. Surprisingly, in absence of extrinsic stressors, NRF2 deficient mice do not show any obvious skin phenotype, not even at old age. We investigated the differences in chronological epidermal aging of wild type and NRF2-deficient mice to identify the changes in aged epidermis that may compensate for absence of this important transcriptional regulator. While both genotypes showed elevated epidermal senescence markers (increased Lysophospholipids, decreased LaminB1 expression), the aged NRF2 deficient mice displayed disturbed epidermal differentiation manifested in irregular keratin 10 and loricrin expression. The tail skin displayed less age-related epidermal thinning and a less pronounced decline in proliferating basal epidermal cells compared to the wildtype controls. The stratum corneum lipid composition also differed, as we observed elevated production of barrier protective linoleic acid (C18:2) and reduced abundance of longer chain saturated lignoceric acid (C24:0) among the stratum corneum fatty acids in the aged NRF2-deficient mice. Thus, despite epidermal differentiation being disturbed in aged NRF2-deficient animals in homeostasis, adaptations in keratinocyte proliferation and barrier lipid synthesis could explain the lack of a more severe phenotype.
Collapse
Affiliation(s)
- Michaela Sochorová
- Department of DermatologyMedical University of ViennaViennaAustria
- Faculty of Pharmacy in Hradec KrálovéCharles UniversityHradec KrálovéCzech Republic
| | | | | | - Francesca Ferrara
- Department of DermatologyMedical University of ViennaViennaAustria
- Department of Chemical, Pharmaceutical and Agricultural SciencesUniversity of FerraraFerraraItaly
| | | | | | - Christina Bauer
- Department of DermatologyMedical University of ViennaViennaAustria
| | | | - Bahar Golabi
- Department of DermatologyMedical University of ViennaViennaAustria
| | - Katerina Vávrová
- Faculty of Pharmacy in Hradec KrálovéCharles UniversityHradec KrálovéCzech Republic
| | - Florian Gruber
- Department of DermatologyMedical University of ViennaViennaAustria
| |
Collapse
|
26
|
Kristiani L, Kim Y. The Interplay between Oxidative Stress and the Nuclear Lamina Contributes to Laminopathies and Age-Related Diseases. Cells 2023; 12:cells12091234. [PMID: 37174634 PMCID: PMC10177617 DOI: 10.3390/cells12091234] [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: 04/04/2023] [Revised: 04/23/2023] [Accepted: 04/24/2023] [Indexed: 05/15/2023] Open
Abstract
Oxidative stress is a physiological condition that arises when there is an imbalance between the production of reactive oxygen species (ROS) and the ability of cells to neutralize them. ROS can damage cellular macromolecules, including lipids, proteins, and DNA, leading to cellular senescence and physiological aging. The nuclear lamina (NL) is a meshwork of intermediate filaments that provides structural support to the nucleus and plays crucial roles in various nuclear functions, such as DNA replication and transcription. Emerging evidence suggests that oxidative stress disrupts the integrity and function of the NL, leading to dysregulation of gene expression, DNA damage, and cellular senescence. This review highlights the current understanding of the interplay between oxidative stress and the NL, along with its implications for human health. Specifically, elucidation of the mechanisms underlying the interplay between oxidative stress and the NL is essential for the development of effective treatments for laminopathies and age-related diseases.
Collapse
Affiliation(s)
- Lidya Kristiani
- Department of Biomedicine, School of Life Science, Indonesia International Institute for Life Science, Jakarta 13210, Indonesia
| | - Youngjo Kim
- Department of Integrated Biomedical Science, Soonchunhyang Institute of Medi-Bioscience, Soonchunhyang University, Cheonan 31151, Republic of Korea
| |
Collapse
|
27
|
Pennarun G, Picotto J, Bertrand P. Close Ties between the Nuclear Envelope and Mammalian Telomeres: Give Me Shelter. Genes (Basel) 2023; 14:genes14040775. [PMID: 37107534 PMCID: PMC10137478 DOI: 10.3390/genes14040775] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Revised: 03/18/2023] [Accepted: 03/20/2023] [Indexed: 04/29/2023] Open
Abstract
The nuclear envelope (NE) in eukaryotic cells is essential to provide a protective compartment for the genome. Beside its role in connecting the nucleus with the cytoplasm, the NE has numerous important functions including chromatin organization, DNA replication and repair. NE alterations have been linked to different human diseases, such as laminopathies, and are a hallmark of cancer cells. Telomeres, the ends of eukaryotic chromosomes, are crucial for preserving genome stability. Their maintenance involves specific telomeric proteins, repair proteins and several additional factors, including NE proteins. Links between telomere maintenance and the NE have been well established in yeast, in which telomere tethering to the NE is critical for their preservation and beyond. For a long time, in mammalian cells, except during meiosis, telomeres were thought to be randomly localized throughout the nucleus, but recent advances have uncovered close ties between mammalian telomeres and the NE that play important roles for maintaining genome integrity. In this review, we will summarize these connections, with a special focus on telomere dynamics and the nuclear lamina, one of the main NE components, and discuss the evolutionary conservation of these mechanisms.
Collapse
Affiliation(s)
- Gaëlle Pennarun
- Université Paris Cité, INSERM, CEA, Stabilité Génétique Cellules Souches et Radiations, LREV/iRCM/IBFJ, F-92260 Fontenay-aux-Roses, France
- Université Paris-Saclay, INSERM, CEA, Stabilité Génétique Cellules Souches et Radiations, LREV/iRCM/IBFJ, F-92260 Fontenay-aux-Roses, France
| | - Julien Picotto
- Université Paris Cité, INSERM, CEA, Stabilité Génétique Cellules Souches et Radiations, LREV/iRCM/IBFJ, F-92260 Fontenay-aux-Roses, France
- Université Paris-Saclay, INSERM, CEA, Stabilité Génétique Cellules Souches et Radiations, LREV/iRCM/IBFJ, F-92260 Fontenay-aux-Roses, France
| | - Pascale Bertrand
- Université Paris Cité, INSERM, CEA, Stabilité Génétique Cellules Souches et Radiations, LREV/iRCM/IBFJ, F-92260 Fontenay-aux-Roses, France
- Université Paris-Saclay, INSERM, CEA, Stabilité Génétique Cellules Souches et Radiations, LREV/iRCM/IBFJ, F-92260 Fontenay-aux-Roses, France
| |
Collapse
|
28
|
Carollo PS, Barra V. Chromatin epigenetics and nuclear lamina keep the nucleus in shape: Examples from natural and accelerated aging. Biol Cell 2023; 115:e2200023. [PMID: 36117150 DOI: 10.1111/boc.202200023] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Revised: 09/11/2022] [Accepted: 09/12/2022] [Indexed: 01/07/2023]
Abstract
As the repository of genetic information, the cell nucleus must protect DNA integrity from mechanical stresses. The nuclear lamina, which resides within the nuclear envelope (NE), is made up of lamins, intermediate filaments bound to DNA. The nuclear lamina provides the nucleus with the ability to deal with inward as well as outward mechanical stimuli. Chromatin, in turn, through its degrees of compaction, shares this role with the nuclear lamina, thus, ensuring the plasticity of the nucleus. Perturbation of chromatin condensation or the nuclear lamina has been linked to a plethora of biological conditions, that range from cancer and genetic diseases (laminopathies) to aging, both natural and accelerated, such as the case of Hutchinson-Gilford Progeria Syndrome (HGPS). From the experimental results accumulated so far on the topic, a direct link between variations of the epigenetic pattern and nuclear lamina structure would be suggested, however, it has never been clarified thoroughly. This relationship, instead, has a downstream important implication on nucleus shape, genome preservation, force sensing, and, ultimately, aging-related disease onset. With this review, we aim to collect recent studies on the importance of both nuclear lamina components and chromatin status in nuclear mechanics. We also aim to bring to light evidence of the link between DNA methylation and nuclear lamina in natural and accelerated aging.
Collapse
Affiliation(s)
- Pietro Salvatore Carollo
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies, University of Palermo, Palermo, Italy
| | - Viviana Barra
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies, University of Palermo, Palermo, Italy
| |
Collapse
|
29
|
Kirkland NJ, Skalak SH, Whitehead AJ, Hocker JD, Beri P, Vogler G, Hum B, Wang M, Lakatta EG, Ren B, Bodmer R, Engler AJ. Age-dependent Lamin changes induce cardiac dysfunction via dysregulation of cardiac transcriptional programs. NATURE AGING 2023; 3:17-33. [PMID: 36845078 PMCID: PMC9956937 DOI: 10.1038/s43587-022-00323-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Accepted: 10/31/2022] [Indexed: 12/24/2022]
Abstract
As we age, structural changes contribute to progressive decline in organ function, which in the heart act through poorly characterized mechanisms. Taking advantage of the short lifespan and conserved cardiac proteome of the fruit fly, we found that cardiomyocytes exhibit progressive loss of Lamin C (mammalian Lamin A/C homologue) with age, coincident with decreasing nuclear size and increasing nuclear stiffness. Premature genetic reduction of Lamin C phenocopies aging's effects on the nucleus, and subsequently decreases heart contractility and sarcomere organization. Surprisingly, Lamin C reduction downregulates myogenic transcription factors and cytoskeletal regulators, possibly via reduced chromatin accessibility. Subsequently, we find a role for cardiac transcription factors in regulating adult heart contractility and show that maintenance of Lamin C, and cardiac transcription factor expression, prevents age-dependent cardiac decline. Our findings are conserved in aged non-human primates and mice, demonstrating that age-dependent nuclear remodeling is a major mechanism contributing to cardiac dysfunction.
Collapse
Affiliation(s)
- Natalie J. Kirkland
- Department of Bioengineering, University California San Diego; La Jolla, CA, USA 92093
- Sanford Consortium for Regenerative Medicine; La Jolla, CA, USA 92037
| | - Scott H. Skalak
- Department of Bioengineering, University California San Diego; La Jolla, CA, USA 92093
- Sanford Consortium for Regenerative Medicine; La Jolla, CA, USA 92037
| | - Alexander J. Whitehead
- Department of Bioengineering, University California San Diego; La Jolla, CA, USA 92093
- Sanford Consortium for Regenerative Medicine; La Jolla, CA, USA 92037
| | - James D. Hocker
- Cell and Molecular Medicine, University California San Diego; La Jolla, CA, USA 92093
- Biomedical Sciences Program, University California San Diego; La Jolla, CA, USA 92093
| | - Pranjali Beri
- Department of Bioengineering, University California San Diego; La Jolla, CA, USA 92093
- Sanford Consortium for Regenerative Medicine; La Jolla, CA, USA 92037
| | - Geo Vogler
- Development, Aging and Regeneration Program, Sanford-Burnham-Prebys Medical Discovery Institute; La Jolla, CA, USA 92037
| | - Bill Hum
- Development, Aging and Regeneration Program, Sanford-Burnham-Prebys Medical Discovery Institute; La Jolla, CA, USA 92037
| | - Mingyi Wang
- Laboratory of Cardiovascular Science, Intramural Research Program, National Institute on Aging, National Institutes of Health, Baltimore, MD, USA 21224
| | - Edward G. Lakatta
- Laboratory of Cardiovascular Science, Intramural Research Program, National Institute on Aging, National Institutes of Health, Baltimore, MD, USA 21224
| | - Bing Ren
- Cell and Molecular Medicine, University California San Diego; La Jolla, CA, USA 92093
- Biomedical Sciences Program, University California San Diego; La Jolla, CA, USA 92093
- Ludwig Institute for Cancer Research; La Jolla, CA, USA 92037
| | - Rolf Bodmer
- Development, Aging and Regeneration Program, Sanford-Burnham-Prebys Medical Discovery Institute; La Jolla, CA, USA 92037
| | - Adam J. Engler
- Department of Bioengineering, University California San Diego; La Jolla, CA, USA 92093
- Biomedical Sciences Program, University California San Diego; La Jolla, CA, USA 92093
- Sanford Consortium for Regenerative Medicine; La Jolla, CA, USA 92037
| |
Collapse
|
30
|
Morgunova VV, Sokolova OA, Sizova TV, Malaev LG, Babaev DS, Kwon DA, Kalmykova AI. Dysfunction of Lamin B and Physiological Aging Cause Telomere Instability in Drosophila Germline. BIOCHEMISTRY. BIOKHIMIIA 2022; 87:1600-1610. [PMID: 36717449 DOI: 10.1134/s000629792212015x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
Chromatin spatial organization in the nucleus is essential for the genome functioning and regulation of gene activity. The nuclear lamina and lamina-associated proteins, lamins, play a key role in this process. Lamin dysfunction leads to the decompaction and transcriptional activation of heterochromatin, which is associated with the premature aging syndrome. In many cell types, telomeres are located at the nuclear periphery, where their replication and stability are ensured by the nuclear lamina. Moreover, diseases associated with defects in lamins and telomeres have similar manifestations and resemble physiological aging. Understanding molecular changes associated with aging at the organismal level is especially important. In this study, we compared the effects caused by the mutation in lamin B and physiological aging in the germline of the model organism Drosophila melanogaster. We have shown that the impaired localization of lamin B leads to the heterochromatin decompaction and transcriptional activation of some transposable elements and telomeric repeats. Both DNA damage and activation of homologous recombination in the telomeres were observed in the germ cells of lamin B mutants. The instability of repeat-enriched heterochromatin can be directly related to the genome destabilization, germ cell death, and sterility observed in lamin B mutants. Similar processes were observed in Drosophila germline in the course of physiological aging, which indicates a close link between the maintenance of the heterochromatin stability at the nuclear periphery and mechanisms of aging.
Collapse
Affiliation(s)
- Valeriya V Morgunova
- Institute of Molecular Genetics of National Research Centre "Kurchatov Institute", Moscow, 123182, Russia
| | - Olesya A Sokolova
- Institute of Molecular Genetics of National Research Centre "Kurchatov Institute", Moscow, 123182, Russia
| | - Tatyana V Sizova
- Institute of Molecular Genetics of National Research Centre "Kurchatov Institute", Moscow, 123182, Russia
| | - Leonid G Malaev
- Institute of Molecular Genetics of National Research Centre "Kurchatov Institute", Moscow, 123182, Russia.,Faculty of Biotechnology, Lomonosov Moscow State University, Moscow, 119991, Russia
| | - Dmitry S Babaev
- Institute of Molecular Genetics of National Research Centre "Kurchatov Institute", Moscow, 123182, Russia.,Faculty of Biotechnology, Lomonosov Moscow State University, Moscow, 119991, Russia
| | - Dmitry A Kwon
- Kurchatov Center for Genome Research, National Research Centre "Kurchatov Institute", Moscow, 123182, Russia
| | - Alla I Kalmykova
- Institute of Molecular Genetics of National Research Centre "Kurchatov Institute", Moscow, 123182, Russia.
| |
Collapse
|
31
|
Abstract
Cellular senescence, once thought an artifact of in vitro culture or passive outcome of aging, has emerged as fundamental to tissue development and function. The senescence mechanism importantly halts cell cycle progression to protect against tumor formation, while transiently present senescent cells produce a complex secretome (or SASP) of inflammatory mediators, proteases, and growth factors that guide developmental remodeling and tissue regeneration. Transiently present senescence is important for skin repair, where it accelerates extracellular matrix formation, limits fibrosis, promotes reepithelialization, and modulates inflammation. Unfortunately, advanced age and diabetes drive pathological accumulation of senescent cells in chronic wounds, which is perpetuated by a proinflammatory SASP, advanced glycation end-products, and oxidative damage. Although the biology of wound senescence remains incompletely understood, drugs that selectively target senescent cells are showing promise in clinical trials for diverse pathological conditions. It may not be long before senescence-targeted therapies will be available for the management, or perhaps even prevention, of chronic wounds.
Collapse
Affiliation(s)
- Holly N Wilkinson
- Centre for Atherothrombosis and Metabolic Disease, Hull York Medical School, University of Hull, Hull HU6 7RX, United Kingdom
| | - Matthew J Hardman
- Centre for Atherothrombosis and Metabolic Disease, Hull York Medical School, University of Hull, Hull HU6 7RX, United Kingdom
| |
Collapse
|
32
|
Abstract
Cellular senescence is implicated in a wide range of physiological and pathological conditions throughout an organism's entire lifetime. In particular, it has become evident that senescence plays a causative role in aging and age-associated disorders. This is not due simply to the loss of function of senescent cells. Instead, the substantial alterations of the cellular activities of senescent cells, especially the array of secretory factors, impact the surrounding tissues or even entire organisms. Such non-cell-autonomous functionality is largely coordinated by tissue-specific genes, constituting a cell fate-determining state. Senescence can be viewed as a gain-of-function phenotype or a process of cell identity shift. Cellular functionality or lineage-specific gene expression is tightly linked to the cell type-specific epigenetic landscape, reinforcing the heterogeneity of senescence across cell types. Here, we aim to define the senescence cellular functionality and epigenetic features that may contribute to the gain-of-function phenotype.
Collapse
Affiliation(s)
- Ioana Olan
- Cancer Research UK Cambridge Institute, Li Ka Shing Centre, University of Cambridge, Cambridge, United Kingdom; ,
| | - Masashi Narita
- Cancer Research UK Cambridge Institute, Li Ka Shing Centre, University of Cambridge, Cambridge, United Kingdom; ,
| |
Collapse
|
33
|
Thompson EL, Pitcher LE, Niedernhofer LJ, Robbins PD. Targeting Cellular Senescence with Senotherapeutics: Development of New Approaches for Skin Care. Plast Reconstr Surg 2022; 150:12S-19S. [PMID: 36170431 PMCID: PMC9529240 DOI: 10.1097/prs.0000000000009668] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
SUMMARY Aging of the skin is evidenced by increased wrinkles, age spots, dryness, and thinning with decreased elasticity. Extrinsic and intrinsic factors including UV, pollution, and inflammation lead to an increase in senescent cells (SnCs) in skin with age that contribute to these observed pathological changes. Cellular senescence is induced by multiple types of damage and stress and is characterized by the irreversible exit from the cell cycle with upregulation of cell cycle-dependent kinase inhibitors p16INK4a and p21CIP1. Most SnCs also developed an inflammatory senescence-associated secretory phenotype (SASP) that drives further pathology through paracrine effects on neighboring cells and endocrine effects on cells at a distance. Recently, compounds able to kill senescent cells specifically, termed senolytics, or suppress the SASP, termed senomorphics, have been developed that have the potential to improve skin aging as well as systemic aging in general. Here, we provide a summary of the evidence for a key role in cellular senescence in driving skin aging. In addition, the evidence for the potential application of senotherapeutics for skin treatments is presented. Overall, topical, and possibly oral senotherapeutic treatments have tremendous potential to eventually become a standard of care for skin aging and related skin disorders.
Collapse
Affiliation(s)
- Elizabeth L Thompson
- From the Department of Biochemistry, Molecular Biology, and Biophysics, Institute on the Biology of Aging and Metabolism, University of Minnesota
| | - Louise E Pitcher
- From the Department of Biochemistry, Molecular Biology, and Biophysics, Institute on the Biology of Aging and Metabolism, University of Minnesota
| | - Laura J Niedernhofer
- From the Department of Biochemistry, Molecular Biology, and Biophysics, Institute on the Biology of Aging and Metabolism, University of Minnesota
| | - Paul D Robbins
- From the Department of Biochemistry, Molecular Biology, and Biophysics, Institute on the Biology of Aging and Metabolism, University of Minnesota
| |
Collapse
|
34
|
Cassidy LD, Narita M. Autophagy at the intersection of aging, senescence, and cancer. Mol Oncol 2022; 16:3259-3275. [PMID: 35689420 PMCID: PMC9490138 DOI: 10.1002/1878-0261.13269] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Revised: 05/24/2022] [Accepted: 06/09/2022] [Indexed: 01/18/2023] Open
Abstract
Autophagy is an evolutionarily conserved cellular process in which macromolecules undergo lysosomal degradation. It fulfills essential roles in quality controlling cellular constituents and in energy homeostasis. Basal autophagy is also widely accepted to provide a protective role in aging and aging-related disorders, and its decline with age might precipitate the onset of a variety of diseases. In this review, we discuss the role of basal autophagy in maintaining homeostasis, in part through the maintenance of stem cell populations and the prevention of cellular senescence. We also consider how stress-induced senescence, for example, during oncogene activation and in premalignant disease, might rely on autophagy, and the possibility that the age-associated decline in autophagy might promote tumour development through a variety of mechanisms. Ultimately, evidence suggests that autophagy is required for malignant cancer progression in a number of settings. Thus, autophagy appears to be tumour-suppressive during the early stages of tumorigenesis and tumour-promoting at later stages.
Collapse
Affiliation(s)
- Liam D. Cassidy
- Cancer Research UK Cambridge InstituteUniversity of CambridgeUK
| | - Masashi Narita
- Cancer Research UK Cambridge InstituteUniversity of CambridgeUK
- Tokyo Tech World Research Hub Initiative (WRHI), Institute of Innovative ResearchTokyo Institute of TechnologyYokohamaJapan
| |
Collapse
|
35
|
Jarrold BB, Tan CYR, Ho CY, Soon AL, Lam TT, Yang X, Nguyen C, Guo W, Chew YC, DeAngelis YM, Costello L, De Los Santos Gomez P, Przyborski S, Bellanger S, Dreesen O, Kimball AB, Oblong JE. Early onset of senescence and imbalanced epidermal homeostasis across the decades in photoexposed human skin: Fingerprints of inflammaging. Exp Dermatol 2022; 31:1748-1760. [DOI: 10.1111/exd.14654] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Revised: 06/11/2022] [Accepted: 07/29/2022] [Indexed: 11/29/2022]
Affiliation(s)
| | | | - Chin Yee Ho
- A*STAR Skin Research Labs Singapore City Singapore
| | - Ai Ling Soon
- A*STAR Skin Research Labs Singapore City Singapore
| | - TuKiet T. Lam
- Keck MS & Proteomics Resource Yale School of Medicine New Haven Connecticut USA
| | | | | | - Wei Guo
- Zymo Research Corporation Irvine California USA
| | | | | | | | | | | | | | | | - Alexa B. Kimball
- Beth Israel Deaconess Medical Center and Harvard Medical School Boston Massachusetts USA
| | | |
Collapse
|
36
|
Sladitschek-Martens HL, Guarnieri A, Brumana G, Zanconato F, Battilana G, Xiccato RL, Panciera T, Forcato M, Bicciato S, Guzzardo V, Fassan M, Ulliana L, Gandin A, Tripodo C, Foiani M, Brusatin G, Cordenonsi M, Piccolo S. YAP/TAZ activity in stromal cells prevents ageing by controlling cGAS-STING. Nature 2022; 607:790-798. [PMID: 35768505 PMCID: PMC7613988 DOI: 10.1038/s41586-022-04924-6] [Citation(s) in RCA: 132] [Impact Index Per Article: 44.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Accepted: 06/01/2022] [Indexed: 02/06/2023]
Abstract
Ageing is intimately connected to the induction of cell senescence1,2, but why this is so remains poorly understood. A key challenge is the identification of pathways that normally suppress senescence, are lost during ageing and are functionally relevant to oppose ageing3. Here we connected the structural and functional decline of ageing tissues to attenuated function of the master effectors of cellular mechanosignalling YAP and TAZ. YAP/TAZ activity declines during physiological ageing in stromal cells, and mimicking such decline through genetic inactivation of YAP/TAZ in these cells leads to accelerated ageing. Conversely, sustaining YAP function rejuvenates old cells and opposes the emergence of ageing-related traits associated with either physiological ageing or accelerated ageing triggered by a mechano-defective extracellular matrix. Ageing traits induced by inactivation of YAP/TAZ are preceded by induction of tissue senescence. This occurs because YAP/TAZ mechanotransduction suppresses cGAS-STING signalling, to the extent that inhibition of STING prevents tissue senescence and premature ageing-related tissue degeneration after YAP/TAZ inactivation. Mechanistically, YAP/TAZ-mediated control of cGAS-STING signalling relies on the unexpected role of YAP/TAZ in preserving nuclear envelope integrity, at least in part through direct transcriptional regulation of lamin B1 and ACTR2, the latter of which is involved in building the peri-nuclear actin cap. The findings demonstrate that declining YAP/TAZ mechanotransduction drives ageing by unleashing cGAS-STING signalling, a pillar of innate immunity. Thus, sustaining YAP/TAZ mechanosignalling or inhibiting STING may represent promising approaches for limiting senescence-associated inflammation and improving healthy ageing.
Collapse
Affiliation(s)
| | | | - Giulia Brumana
- Department of Molecular Medicine, University of Padua, Padua, Italy
| | | | - Giusy Battilana
- Department of Molecular Medicine, University of Padua, Padua, Italy
| | | | - Tito Panciera
- Department of Molecular Medicine, University of Padua, Padua, Italy
| | - Mattia Forcato
- Department of Life Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Silvio Bicciato
- Department of Life Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | | | - Matteo Fassan
- Department of Medicine, University of Padua, Padua, Italy
| | - Lorenzo Ulliana
- Department of Industrial Engineering, University of Padua, Padua, Italy
| | - Alessandro Gandin
- Department of Industrial Engineering, University of Padua, Padua, Italy
| | - Claudio Tripodo
- Department of Health Sciences Unit, Human Pathology Section, University of Palermo, Palermo, Italy
- IFOM ETS, the AIRC Institute of Molecular Oncology, Milan, Italy
| | - Marco Foiani
- IFOM ETS, the AIRC Institute of Molecular Oncology, Milan, Italy
- University of Milan, Milan, Italy
| | - Giovanna Brusatin
- Department of Industrial Engineering, University of Padua, Padua, Italy
| | | | - Stefano Piccolo
- Department of Molecular Medicine, University of Padua, Padua, Italy.
- IFOM ETS, the AIRC Institute of Molecular Oncology, Milan, Italy.
| |
Collapse
|
37
|
Molecular Mechanisms of Changes in Homeostasis of the Dermal Extracellular Matrix: Both Involutional and Mediated by Ultraviolet Radiation. Int J Mol Sci 2022; 23:ijms23126655. [PMID: 35743097 PMCID: PMC9223561 DOI: 10.3390/ijms23126655] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Revised: 06/12/2022] [Accepted: 06/12/2022] [Indexed: 02/01/2023] Open
Abstract
Skin aging is a multi-factorial process that affects nearly every aspect of skin biology and function. With age, an impairment of structures, quality characteristics, and functions of the dermal extracellular matrix (ECM) occurs in the skin, which leads to disrupted functioning of dermal fibroblasts (DFs), the main cells supporting morphofunctional organization of the skin. The DF functioning directly depends on the state of the surrounding collagen matrix (CM). The intact collagen matrix ensures proper adhesion and mechanical tension in DFs, which allows these cells to maintain collagen homeostasis while ECM correctly regulates cellular processes. When the integrity of CM is destroyed, mechanotransduction is disrupted, which is accompanied by impairment of DF functioning and destruction of collagen homeostasis, thereby contributing to the progression of aging processes in skin tissues. This article considers in detail the processes of skin aging and associated changes in the skin layers, as well as the mechanisms of these processes at the molecular level.
Collapse
|
38
|
Bellanger A, Madsen-Østerbye J, Galigniana NM, Collas P. Restructuring of Lamina-Associated Domains in Senescence and Cancer. Cells 2022; 11:1846. [PMID: 35681541 PMCID: PMC9180887 DOI: 10.3390/cells11111846] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Revised: 06/02/2022] [Accepted: 06/04/2022] [Indexed: 01/01/2023] Open
Abstract
Induction of cellular senescence or cancer is associated with a reshaping of the nuclear envelope and a broad reorganization of heterochromatin. At the periphery of mammalian nuclei, heterochromatin is stabilized at the nuclear lamina via lamina-associated domains (LADs). Alterations in the composition of the nuclear lamina during senescence lead to a loss of peripheral heterochromatin, repositioning of LADs, and changes in epigenetic states of LADs. Cancer initiation and progression are also accompanied by a massive reprogramming of the epigenome, particularly in domains coinciding with LADs. Here, we review recent knowledge on alterations in chromatin organization and in the epigenome that affect LADs and related genomic domains in senescence and cancer.
Collapse
Affiliation(s)
- Aurélie Bellanger
- Department of Molecular Medicine, Institute of Basic Medical Sciences, Faculty of Medicine, University of Oslo, 0372 Oslo, Norway; (A.B.); (J.M.-Ø.); (N.M.G.)
| | - Julia Madsen-Østerbye
- Department of Molecular Medicine, Institute of Basic Medical Sciences, Faculty of Medicine, University of Oslo, 0372 Oslo, Norway; (A.B.); (J.M.-Ø.); (N.M.G.)
| | - Natalia M. Galigniana
- Department of Molecular Medicine, Institute of Basic Medical Sciences, Faculty of Medicine, University of Oslo, 0372 Oslo, Norway; (A.B.); (J.M.-Ø.); (N.M.G.)
- Department of Immunology and Transfusion Medicine, Oslo University Hospital, 0372 Oslo, Norway
| | - Philippe Collas
- Department of Molecular Medicine, Institute of Basic Medical Sciences, Faculty of Medicine, University of Oslo, 0372 Oslo, Norway; (A.B.); (J.M.-Ø.); (N.M.G.)
- Department of Immunology and Transfusion Medicine, Oslo University Hospital, 0372 Oslo, Norway
| |
Collapse
|
39
|
Zorina A, Zorin V, Kudlay D, Kopnin P. Age-Related Changes in the Fibroblastic Differon of the Dermis: Role in Skin Aging. Int J Mol Sci 2022; 23:ijms23116135. [PMID: 35682813 PMCID: PMC9181700 DOI: 10.3390/ijms23116135] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Revised: 05/27/2022] [Accepted: 05/27/2022] [Indexed: 02/06/2023] Open
Abstract
Skin aging is a multi-factorial process that affects nearly every aspect of skin biology and function. The processes developing in the skin during aging are based on fundamental molecular mechanisms associated with fibroblasts, the main cellular population of the dermis. It has been revealed that the amount of fibroblasts decreases markedly with age and their functional activity is also reduced. This inevitably leads to a decrease in the regenerative abilities of the skin and the progression of its aging. In this review we consider the mechanisms underlying these processes, mainly the changes observed with age in the stem/progenitor cells that constitute the fibroblastic differon of the dermis and form their microenvironment (niches). These changes lead to the depletion of stem cells, which, in turn, leads to a decrease in the number of differentiated (mature) dermal fibroblasts responsible for the production of the dermal extracellular matrix and its remodeling. We also describe in detail DNA damages, their cellular and systemic consequences, molecular mechanisms of DNA damage response, and also the role of fibroblast senescence in skin aging.
Collapse
Affiliation(s)
- Alla Zorina
- Human Stem Cells Institute, 119333 Moscow, Russia; (A.Z.); (V.Z.)
| | - Vadim Zorin
- Human Stem Cells Institute, 119333 Moscow, Russia; (A.Z.); (V.Z.)
| | - Dmitry Kudlay
- Department of Pharmacology, Institute of Pharmacy, I. M. Sechenov First Moscow State Medical University (Sechenov University), 119991 Moscow, Russia;
| | - Pavel Kopnin
- N. N. Blokhin National Medical Research Oncology Center, Ministry of Health of Russia, 115478 Moscow, Russia
- Correspondence: ; Tel.: +7-49-9324-1739
| |
Collapse
|
40
|
Meqbel BRM, Gomes M, Omer A, Gallouzi IE, Horn HF. LINCing Senescence and Nuclear Envelope Changes. Cells 2022; 11:1787. [PMID: 35681483 PMCID: PMC9179861 DOI: 10.3390/cells11111787] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2022] [Revised: 05/23/2022] [Accepted: 05/24/2022] [Indexed: 01/27/2023] Open
Abstract
The nuclear envelope (NE) has emerged as a nexus for cellular organization, signaling, and survival. Beyond its role as a barrier to separate the nucleoplasm from the cytoplasm, the NE's role in supporting and maintaining a myriad of other functions has made it a target of study in many cellular processes, including senescence. The nucleus undergoes dramatic changes in senescence, many of which are driven by changes in the NE. Indeed, Lamin B1, a key NE protein that is consistently downregulated in senescence, has become a marker for senescence. Other NE proteins have also been shown to play a role in senescence, including LINC (linker of nucleoskeleton and cytoskeleton) complex proteins. LINC complexes span the NE, forming physical connections between the cytoplasm to the nucleoplasm. In this way, they integrate nuclear and cytoplasmic mechanical signals and are essential not only for a variety of cellular functions but are needed for cell survival. However, LINC complex proteins have been shown to have a myriad of functions in addition to forming a LINC complex, often existing as nucleoplasmic or cytoplasmic soluble proteins in a variety of isoforms. Some of these proteins have now been shown to play important roles in DNA repair, cell signaling, and nuclear shape regulation, all of which are important in senescence. This review will focus on some of these roles and highlight the importance of LINC complex proteins in senescence.
Collapse
Affiliation(s)
- Bakhita R. M. Meqbel
- College of Health and Life Sciences, Hamad Bin Khalifa University, Doha P.O. Box 34110, Qatar;
| | - Matilde Gomes
- KAUST Smart-Health Initiative and Biological and Environmental Science and Engineering (BESE) Division, King Abdullah University of Science and Technology (KAUST), Jeddah 21589, Saudi Arabia; (M.G.); (I.E.G.)
| | - Amr Omer
- Department of Biochemistry, McGill University, 3655 Promenade Sir William Osler, Montreal, QC H3G 1Y6, Canada;
| | - Imed E. Gallouzi
- KAUST Smart-Health Initiative and Biological and Environmental Science and Engineering (BESE) Division, King Abdullah University of Science and Technology (KAUST), Jeddah 21589, Saudi Arabia; (M.G.); (I.E.G.)
- Department of Biochemistry, McGill University, 3655 Promenade Sir William Osler, Montreal, QC H3G 1Y6, Canada;
| | - Henning F. Horn
- College of Health and Life Sciences, Hamad Bin Khalifa University, Doha P.O. Box 34110, Qatar;
| |
Collapse
|
41
|
Maurizi E, Merra A, Schiroli D, Ghezzi B, Macaluso C, Pellegrini G. Fluctuations in Corneal Endothelial LAP2 Expression Levels Correlate with Passage Dependent Declines in Their Cell Proliferative Activity. Int J Mol Sci 2022; 23:ijms23105859. [PMID: 35628669 PMCID: PMC9146651 DOI: 10.3390/ijms23105859] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2022] [Revised: 05/19/2022] [Accepted: 05/22/2022] [Indexed: 01/27/2023] Open
Abstract
The corneal endothelium is the inner corneal mono-layered epithelium, fundamental for preserving corneal hydration and transparency. However, molecular mechanisms that regulate corneal endothelial cells (CEnCs), in particular regarding their proliferative capacity, have been only partially elucidated. CEnCs are quiescent in vivo and they easily undergo endothelial to mesenchymal transition (EnMT) in vitro. This study aims to analyze CEnCs behavior and expression in vitro, either in sub-confluent growing (S) or confluent (C) CEnCs cultures. Primary rabbit and human CEnCs were cultured and used for RT-PCR, immunofluorescence or western blot analysis. These methods allowed identifying a novel molecular marker, LAP2, that is upregulated in S while downregulated in C human or rabbit CEnCs. Those results were observed for several subsequent passages in culture and this, together with the correlation between ki67 and LAP2 expression, suggested LAP2 as a novel possible indicator for culture ageing. Finally, treatment with FGF and TGFβ in rCEnCs highlighted how LAP2 can vary as the cells regulate their proliferative state. In conclusion, we have identified a novel marker for CEnCs, LAP2, that regulates its expression depending on the cells sub/confluent state and that correlates with CEnCs proliferation.
Collapse
Affiliation(s)
- Eleonora Maurizi
- Centre for Regenerative Medicine, University of Modena and Reggio Emilia, 41125 Modena, Italy;
- Correspondence:
| | - Alessia Merra
- Holostem Terapie Avanzate S.r.l., 41125 Modena, Italy;
| | - Davide Schiroli
- Transfusion Medicine Unit, Azienda USL-IRCCS, 42123 Reggio Emilia, Italy;
| | - Benedetta Ghezzi
- Dentistry Centre Lab, University of Parma, 43126 Parma, Italy; (B.G.); (C.M.)
| | - Claudio Macaluso
- Dentistry Centre Lab, University of Parma, 43126 Parma, Italy; (B.G.); (C.M.)
| | - Graziella Pellegrini
- Centre for Regenerative Medicine, University of Modena and Reggio Emilia, 41125 Modena, Italy;
- Holostem Terapie Avanzate S.r.l., 41125 Modena, Italy;
| |
Collapse
|
42
|
SENESCENCE-MEDIATED ANTI-CANCER EFFECTS OF QUERCETI. Nutr Res 2022; 104:82-90. [DOI: 10.1016/j.nutres.2022.04.007] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2021] [Revised: 04/22/2022] [Accepted: 04/23/2022] [Indexed: 02/07/2023]
|
43
|
Nacarelli T, Azar A, Potnis M, Johannes G, Mell J, Johnson FB, Brown-Borg H, Noguchi E, Sell C. The methyltransferase enzymes KMT2D, SETD1B, and ASH1L are key mediators of both metabolic and epigenetic changes during cellular senescence. Mol Biol Cell 2022; 33:ar36. [PMID: 35196069 PMCID: PMC9282020 DOI: 10.1091/mbc.e20-08-0523] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2020] [Revised: 02/08/2022] [Accepted: 02/17/2022] [Indexed: 11/11/2022] Open
Abstract
Cellular senescence is a terminal cell fate characterized by growth arrest and a metabolically active state characterized by high glycolytic activity. Human fibroblasts were placed in a unique metabolic state using a combination of methionine restriction (MetR) and rapamycin (Rapa). This combination induced a metabolic reprogramming that prevented the glycolytic shift associated with senescence. Surprisingly, cells treated in this manner did not undergo senescence but continued to divide at a slow rate even at high passage, in contrast with either Rapa treatment or MetR, both of which extended life span but eventually resulted in growth arrest. Transcriptome-wide analysis revealed a coordinated regulation of metabolic enzymes related to one-carbon metabolism including three methyltransferase enzymes (KMT2D, SETD1B, and ASH1L), key enzymes for both carnitine synthesis and histone modification. These enzymes appear to be involved in both the metabolic phenotype of senescent cells and the chromatin changes required for establishing the senescence arrest. Targeting one of these enzymes, ASH1L, produced both a glycolytic shift and senescence, providing proof of concept. These findings reveal a mechanistic link between a major metabolic hallmark of senescence and nuclear events required for senescence.
Collapse
Affiliation(s)
- Timothy Nacarelli
- Glaxosmithkline, Oncology Synthetic Lethal Research Unit, Collegeville, PA 19426
| | | | - Manali Potnis
- Department of Biochemistry and Molecular Biology, Drexel University College of Medicine, Philadelphia, PA 19102
| | | | - Joshua Mell
- Department of Microbiology and Immunology, Drexel University College of Medicine, Philadelphia, PA 19129
| | - F. Brad Johnson
- Department of Pathology, University of Pennsylvania, Philadelphia, PA 19104
| | - Holly Brown-Borg
- Department of Biomedical Sciences, University of North Dakota School of Medicine and Health Sciences, Grand Forks, ND 58203
| | - Eishi Noguchi
- Department of Biochemistry and Molecular Biology, Drexel University College of Medicine, Philadelphia, PA 19102
| | - Christian Sell
- Department of Biochemistry and Molecular Biology, Drexel University College of Medicine, Philadelphia, PA 19102
| |
Collapse
|
44
|
Kamikawa Y, Saito A, Imaizumi K. Impact of Nuclear Envelope Stress on Physiological and Pathological Processes in Central Nervous System. Neurochem Res 2022; 47:2478-2487. [PMID: 35486254 DOI: 10.1007/s11064-022-03608-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 04/07/2022] [Accepted: 04/12/2022] [Indexed: 01/10/2023]
Abstract
The nuclear envelope (NE) separates genomic DNA from the cytoplasm and provides the molecular platforms for nucleocytoplasmic transport, higher-order chromatin organization, and physical links between the nucleus and cytoskeleton. Recent studies have shown that the NE is often damaged by various stresses termed "NE stress", leading to critical cellular dysfunction. Accumulating evidence has revealed the crucial roles of NE stress in the pathology of a broad spectrum of diseases. In the central nervous system (CNS), NE dysfunction impairs neural development and is associated with several neurological disorders, such as Alzheimer's disease and autosomal dominant leukodystrophy. In this review, the structure and functions of the NE are summarized, and the concepts of NE stress and NE stress responses are introduced. Additionally, the significant roles of the NE in the development of CNS and the mechanistic connections between NE stress and neurological disorders are described.
Collapse
Affiliation(s)
- Yasunao Kamikawa
- Department of Biochemistry, Institute of Biomedical & Health Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima, 734-8553, Japan.
| | - Atsushi Saito
- Department of Biochemistry, Institute of Biomedical & Health Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima, 734-8553, Japan
| | - Kazunori Imaizumi
- Department of Biochemistry, Institute of Biomedical & Health Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima, 734-8553, Japan.
| |
Collapse
|
45
|
Mustafin RN, Khusnutdinova EK. The relationship of lamins with epigenetic factors during aging. Vavilovskii Zhurnal Genet Selektsii 2022; 26:40-49. [PMID: 35342861 PMCID: PMC8892175 DOI: 10.18699/vjgb-22-06] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2021] [Revised: 05/23/2021] [Accepted: 09/28/2021] [Indexed: 11/26/2022] Open
Abstract
The key factor of genome instability during aging is transposon dysregulation. This may be due to senile changes in the expression of lamins, which epigenetically modulate transposons. Lamins directly physically interact with transposons. Epigenetic regulators such as SIRT7, BAF, and microRNA can also serve as intermediaries for their interactions. There is also an inverse regulation, since transposons are sources of miRNAs that affect lamins. We suggest that lamins can be attributed to epigenetic factors, since they are part of the NURD, interact with histone deacetylases and regulate gene expression without changing the nucleotide sequences. The role of lamins in the etiopathogenesis of premature aging syndromes may be associated with interactions with transposons. In various human cells, LINE1 is present in the heterochromatin domains of the genome associated with lamins, while SIRT7 facilitates the interaction of this retroelement with lamins. Both retroelements and the nuclear lamina play an important role in the antiviral response of organisms. This may be due to the role of lamins in protection from both viruses and transposons, since viruses and transposons are evolutionarily related. Transposable elements and lamins are secondary messengers of environmental stressors that can serve as triggers for aging and carcinogenesis. Transposons play a role in the development of cancer, while the microRNAs derived from them, participating in the etiopathogenesis of tumors, are important in human aging. Lamins have similar properties, since lamins are dysregulated in cancer, and microRNAs affecting them are involved in carcinogenesis. Changes in the expression of specif ic microRNAs were also revealed
in laminopathies. Identif ication of the epigenetic mechanisms of interaction of lamins with transposons during
aging
can become the basis for the development of methods of life extension and targeted therapy of age-associated
cancer
Collapse
Affiliation(s)
| | - E. K. Khusnutdinova
- Institute of Biochemistry and Genetics – Subdivision of the Ufa Federal Research Centre of the Russian Academy of Sciences
| |
Collapse
|
46
|
Malvezzi H, Dobo C, Filippi RZ, Mendes do Nascimento H, Palmieri da Silva e Sousa L, Meola J, Piccinato CA, Podgaec S. Altered p16 Ink4a, IL-1β, and Lamin b1 Protein Expression Suggest Cellular Senescence in Deep Endometriotic Lesions. Int J Mol Sci 2022; 23:2476. [PMID: 35269619 PMCID: PMC8910415 DOI: 10.3390/ijms23052476] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2021] [Revised: 01/26/2022] [Accepted: 01/31/2022] [Indexed: 01/10/2023] Open
Abstract
Endometriosis causes immunological and cellular alterations. Endometriosis lesions have lower levels of lamin b1 than the endometrium. Moreover, high levels of pro-inflammatory markers are observed in the peritoneal fluid, follicular fluid, and serum in endometriosis lesions. Thus, we hypothesized that the accumulation of senescent cells in endometriosis tissues would facilitate endometriosis maintenance in an inflammatory microenvironment. To study senescent cell markers and the senescence-associated secretory phenotype (SASP) in endometriosis lesions, we conducted a cross-sectional study with 27 patients undergoing video laparoscopy for endometriosis resection and 19 patients without endometriosis. Endometriosis lesions were collected from patients with endometriosis, while eutopic endometrium was collected from patients both with and without endometriosis. Tissues were evaluated for senescence markers (p16Ink4a, lamin b1, and IL-1β) and interleukin concentrations. The expression of p16Ink4a increased in lesions compared to that in eutopic endometrium from endometriosis patients in the secretory phase. In the proliferative phase, lesions exhibited lower lamin b1 expression but higher IL-4 expression than the eutopic endometrium. Further, IL-1β levels were higher in the lesions than in the eutopic endometrium in both the secretory and proliferative phases. We believe that our findings may provide targets for better therapeutic interventions to alleviate the symptoms of endometriosis.
Collapse
Affiliation(s)
- Helena Malvezzi
- Hospital Israelita Albert Einstein, Av. Albert Einstein 627, Morumbi 05652-900, SP, Brazil; (C.D.); (R.Z.F.); (H.M.d.N.); (L.P.d.S.e.S.); (C.A.P.); (S.P.)
| | - Cristine Dobo
- Hospital Israelita Albert Einstein, Av. Albert Einstein 627, Morumbi 05652-900, SP, Brazil; (C.D.); (R.Z.F.); (H.M.d.N.); (L.P.d.S.e.S.); (C.A.P.); (S.P.)
| | - Renee Zon Filippi
- Hospital Israelita Albert Einstein, Av. Albert Einstein 627, Morumbi 05652-900, SP, Brazil; (C.D.); (R.Z.F.); (H.M.d.N.); (L.P.d.S.e.S.); (C.A.P.); (S.P.)
| | - Helen Mendes do Nascimento
- Hospital Israelita Albert Einstein, Av. Albert Einstein 627, Morumbi 05652-900, SP, Brazil; (C.D.); (R.Z.F.); (H.M.d.N.); (L.P.d.S.e.S.); (C.A.P.); (S.P.)
| | - Laura Palmieri da Silva e Sousa
- Hospital Israelita Albert Einstein, Av. Albert Einstein 627, Morumbi 05652-900, SP, Brazil; (C.D.); (R.Z.F.); (H.M.d.N.); (L.P.d.S.e.S.); (C.A.P.); (S.P.)
| | - Juliana Meola
- School of Medicine of Ribeirão Preto, University of São Paulo, Gynecology and Obstetrics, Av. Bandeirantes, 3900, Vila Monte Alegre 14049-900, SP, Brazil;
| | - Carla Azevedo Piccinato
- Hospital Israelita Albert Einstein, Av. Albert Einstein 627, Morumbi 05652-900, SP, Brazil; (C.D.); (R.Z.F.); (H.M.d.N.); (L.P.d.S.e.S.); (C.A.P.); (S.P.)
| | - Sérgio Podgaec
- Hospital Israelita Albert Einstein, Av. Albert Einstein 627, Morumbi 05652-900, SP, Brazil; (C.D.); (R.Z.F.); (H.M.d.N.); (L.P.d.S.e.S.); (C.A.P.); (S.P.)
| |
Collapse
|
47
|
Evangelisti C, Rusciano I, Mongiorgi S, Ramazzotti G, Lattanzi G, Manzoli L, Cocco L, Ratti S. The wide and growing range of lamin B-related diseases: from laminopathies to cancer. Cell Mol Life Sci 2022; 79:126. [PMID: 35132494 PMCID: PMC8821503 DOI: 10.1007/s00018-021-04084-2] [Citation(s) in RCA: 47] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2021] [Revised: 12/03/2021] [Accepted: 12/08/2021] [Indexed: 12/18/2022]
Abstract
B-type lamins are fundamental components of the nuclear lamina, a complex structure that acts as a scaffold for organization and function of the nucleus. Lamin B1 and B2, the most represented isoforms, are encoded by LMNB1 and LMNB2 gene, respectively. All B-type lamins are synthesized as precursors and undergo sequential post-translational modifications to generate the mature protein. B-type lamins are involved in a wide range of nuclear functions, including DNA replication and repair, regulation of chromatin and nuclear stiffness. Moreover, lamins B1 and B2 regulate several cellular processes, such as tissue development, cell cycle, cellular proliferation, senescence, and DNA damage response. During embryogenesis, B-type lamins are essential for organogenesis, in particular for brain development. As expected from the numerous and pivotal functions of B-type lamins, mutations in their genes or fluctuations in their expression levels are critical for the onset of several diseases. Indeed, a growing range of human disorders have been linked to lamin B1 or B2, increasing the complexity of the group of diseases collectively known as laminopathies. This review highlights the recent findings on the biological role of B-type lamins under physiological or pathological conditions, with a particular emphasis on brain disorders and cancer.
Collapse
Affiliation(s)
- Camilla Evangelisti
- Cellular Signalling Laboratory, Department of Biomedical and NeuroMotor Sciences (DIBINEM), University of Bologna, Bologna, Italy
| | - Isabella Rusciano
- Cellular Signalling Laboratory, Department of Biomedical and NeuroMotor Sciences (DIBINEM), University of Bologna, Bologna, Italy
| | - Sara Mongiorgi
- Cellular Signalling Laboratory, Department of Biomedical and NeuroMotor Sciences (DIBINEM), University of Bologna, Bologna, Italy
| | - Giulia Ramazzotti
- Cellular Signalling Laboratory, Department of Biomedical and NeuroMotor Sciences (DIBINEM), University of Bologna, Bologna, Italy
| | - Giovanna Lattanzi
- CNR Institute of Molecular Genetics "Luigi Luca Cavalli-Sforza", Unit of Bologna, Bologna, Italy
- IRCCS Istituto Ortopedico Rizzoli, Bologna, Italy
| | - Lucia Manzoli
- Cellular Signalling Laboratory, Department of Biomedical and NeuroMotor Sciences (DIBINEM), University of Bologna, Bologna, Italy.
| | - Lucio Cocco
- Cellular Signalling Laboratory, Department of Biomedical and NeuroMotor Sciences (DIBINEM), University of Bologna, Bologna, Italy.
| | - Stefano Ratti
- Cellular Signalling Laboratory, Department of Biomedical and NeuroMotor Sciences (DIBINEM), University of Bologna, Bologna, Italy
| |
Collapse
|
48
|
Abstract
Lamins interact with a host of nuclear membrane proteins, transcription factors, chromatin regulators, signaling molecules, splicing factors, and even chromatin itself to form a nuclear subcompartment, the nuclear lamina, that is involved in a variety of cellular processes such as the governance of nuclear integrity, nuclear positioning, mitosis, DNA repair, DNA replication, splicing, signaling, mechanotransduction and -sensation, transcriptional regulation, and genome organization. Lamins are the primary scaffold for this nuclear subcompartment, but interactions with lamin-associated peptides in the inner nuclear membrane are self-reinforcing and mutually required. Lamins also interact, directly and indirectly, with peripheral heterochromatin domains called lamina-associated domains (LADs) and help to regulate dynamic 3D genome organization and expression of developmentally regulated genes.
Collapse
Affiliation(s)
- Xianrong Wong
- Laboratory of Developmental and Regenerative Biology, Skin Research Institute of Singapore, Agency for Science, Technology and Research (A∗STAR), Singapore 138648
| | - Ashley J Melendez-Perez
- Department of Biological Chemistry and Center for Epigenetics, Johns Hopkins University of Medicine, Baltimore, Maryland 21205, USA
| | - Karen L Reddy
- Department of Biological Chemistry and Center for Epigenetics, Johns Hopkins University of Medicine, Baltimore, Maryland 21205, USA
- Sidney Kimmel Cancer Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland 21231, USA
| |
Collapse
|
49
|
Wang AS, Nakamizo S, Ishida Y, Klassen G, Chong P, Wada A, Lim JSY, Wright GD, Kabashima K, Dreesen O. Identification and quantification of senescent cell types by lamin B1 and HMGB1 in Actinic keratosis lesions. J Dermatol Sci 2022; 105:61-64. [PMID: 34930673 DOI: 10.1016/j.jdermsci.2021.12.001] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Revised: 11/16/2021] [Accepted: 12/06/2021] [Indexed: 10/19/2022]
Affiliation(s)
| | | | | | - Genevieve Klassen
- School of Biological Sciences, Nanyang Technological University, Singapore
| | - Priscilla Chong
- School of Biological Sciences, Nanyang Technological University, Singapore
| | - Aya Wada
- Cell Aging Laboratory, A⁎STAR Skin Research Labs (A⁎SRL), Singapore
| | | | | | - Kenji Kabashima
- Kyoto University Graduate School of Medicine, Japan; Skin Immunology Laboratory, A⁎SRL, Singapore
| | - Oliver Dreesen
- Cell Aging Laboratory, A⁎STAR Skin Research Labs (A⁎SRL), Singapore.
| |
Collapse
|
50
|
Liao CM, Luo T, von der Ohe J, de Juan Mora B, Schmitt R, Hass R. Human MSC-Derived Exosomes Reduce Cellular Senescence in Renal Epithelial Cells. Int J Mol Sci 2021; 22:13562. [PMID: 34948355 PMCID: PMC8709122 DOI: 10.3390/ijms222413562] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Revised: 12/10/2021] [Accepted: 12/14/2021] [Indexed: 12/23/2022] Open
Abstract
Cellular senescence of renal tubular cells is associated with chronic diseases and age-related kidney disorders. Therapies to antagonize senescence are, therefore, explored as novel approaches in nephropathy. Exosomes derived from human mesenchymal stroma-/stem-like cells (MSC) entail the transfer of multiple bioactive molecules, exhibiting profound regenerative potential in various tissues, including therapeutic effects in kidney diseases. Here, we first demonstrate that exosomes promote proliferation and reduce senescence in aged MSC cultures. For potential therapeutic perspectives in organ rejuvenation, we used MSC-derived exosomes to antagonize senescence in murine kidney primary tubular epithelial cells (PTEC). Exosome treatment efficiently reduced senescence while diminishing the transcription of senescence markers and senescence-associated secretory phenotype (SASP) factors. Concomitantly, we observed less DNA damage foci and more proliferating cells. These data provide new information regarding the therapeutic property of MSC exosomes in the development of renal senescence, suggesting a contribution to a new chapter of regenerative vehicles in senotherapy.
Collapse
Affiliation(s)
- Chieh Ming Liao
- Department of Nephrology and Hypertension, Hannover Medical School, 30625 Hannover, Germany; (C.M.L.); (B.d.J.M.); (R.S.)
| | - Tianjiao Luo
- Biochemistry and Tumor Biology Lab, Department of Gynecology and Obstetrics, Hannover Medical School, 30625 Hannover, Germany; (T.L.); (J.v.d.O.)
| | - Juliane von der Ohe
- Biochemistry and Tumor Biology Lab, Department of Gynecology and Obstetrics, Hannover Medical School, 30625 Hannover, Germany; (T.L.); (J.v.d.O.)
| | - Blanca de Juan Mora
- Department of Nephrology and Hypertension, Hannover Medical School, 30625 Hannover, Germany; (C.M.L.); (B.d.J.M.); (R.S.)
| | - Roland Schmitt
- Department of Nephrology and Hypertension, Hannover Medical School, 30625 Hannover, Germany; (C.M.L.); (B.d.J.M.); (R.S.)
| | - Ralf Hass
- Biochemistry and Tumor Biology Lab, Department of Gynecology and Obstetrics, Hannover Medical School, 30625 Hannover, Germany; (T.L.); (J.v.d.O.)
| |
Collapse
|