51
|
Guo G, Watterson S, Zhang SD, Bjourson A, McGilligan V, Peace A, Rai TS. The role of senescence in the pathogenesis of atrial fibrillation: A target process for health improvement and drug development. Ageing Res Rev 2021; 69:101363. [PMID: 34023420 DOI: 10.1016/j.arr.2021.101363] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2020] [Revised: 01/24/2021] [Accepted: 05/12/2021] [Indexed: 12/11/2022]
Abstract
Cellular senescence is a state of growth arrest that occurs after cells encounter various stresses. Senescence contributes to tumour suppression, embryonic development, and wound healing. It impacts on the pathology of various diseases by secreting inflammatory chemokines, immune modulators and other bioactive factors. These secretory biosignatures ultimately cause inflammation, tissue fibrosis, immunosenescence and many ageing-related diseases such as atrial fibrillation (AF). Because the molecular mechanisms underpinning AF development remain unclear, current treatments are suboptimal and have serious side effects. In this review, we summarize recent results describing the role of senescence in AF. We propose that senescence factors induce AF and have a causative role. Hence, targeting senescence and its secretory phenotype may attenuate AF.
Collapse
|
52
|
Baran SW, Lim MA, Do JP, Stolyar P, Rabe MD, Schaevitz LR, Cadena SM. Digital Biomarkers Enable Automated, Longitudinal Monitoring in a Mouse Model of Aging. J Gerontol A Biol Sci Med Sci 2021; 76:1206-1213. [PMID: 33491048 DOI: 10.1093/gerona/glab024] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Indexed: 11/14/2022] Open
Abstract
To understand the growing needs of an aging human population, there is demand for scalable and reproducible approaches to study animal models of aging and to test novel therapeutic interventions. We investigated the sensitivity and utility of a continuous monitoring platform and its digital biomarkers (motion, breathing rate, and wheel running) to evaluate behavioral and physiological differences between "young" (12 weeks) and "old" (23 months) male C57BL/6J mice with or without running wheels in the home cage. Compared to young mice, old mice showed marked reductions in motion and breathing rate, as well as altered circadian rhythms. Mice without running wheels possessed lower breathing rates compared to their counterparts with running wheels. Digital biomarkers showed age-dependent changes in response to routine procedures (cage changes and blood sampling) and alterations in subjects that unexpectedly reached endpoint. Continuous collection of digital biomarkers in the home cage can enhance current approaches by providing unbiased longitudinal monitoring for large-scale aging studies.
Collapse
Affiliation(s)
- Szczepan W Baran
- Emerging Technologies, Laboratory Animal Services, Scientific Operations, Novartis Institutes for BioMedical Research (NIBR), Inc., Cambridge, Massachusetts, USA
| | | | | | - Polina Stolyar
- Chemical Biology and Therapeutics, Novartis Institutes for BioMedical Research (NIBR), Inc., Cambridge, Massachusetts, USA
| | | | | | - Samuel M Cadena
- Chemical Biology and Therapeutics, Novartis Institutes for BioMedical Research (NIBR), Inc., Cambridge, Massachusetts, USA
| |
Collapse
|
53
|
Balasubramanian P, Branen L, Sivasubramanian MK, Monteiro R, Subramanian M. Aging is associated with glial senescence in the brainstem - implications for age-related sympathetic overactivity. Aging (Albany NY) 2021; 13:13460-13473. [PMID: 34038388 PMCID: PMC8202881 DOI: 10.18632/aging.203111] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2020] [Accepted: 04/28/2021] [Indexed: 12/11/2022]
Abstract
Accumulating evidence suggests that the sympathetic nervous system (SNS) overactivity plays a crucial role in age-related increase in the risk for cardiovascular diseases such as hypertension, myocardial infarction, stroke and heart diseases. Previous studies indicate that neuroinflammation in key brainstem regions that regulate sympathetic outflow plays a pathogenic role in aging-mediated sympathoexcitation. However, the molecular mechanisms underlying this phenomenon are not clear. While senescent cells and their secretory phenotype (SASP) have been implicated in the pathogenesis of several age-related diseases, their role in age-related neuroinflammation in the brainstem and SNS overactivity has not been investigated. To test this, we isolated brainstems from young (2-4 months) and aged (24 months) male C57BL/6J mice and assessed senescence using a combination of RNA-in situ hybridization, PCR analysis, multiplex assay and SA-β gal staining. Our results show significant increases in p16Ink4a expression, increased activity of SA-β gal and increases in SASP levels in the aged brainstem, suggesting age-induced senescence in the brainstem. Further, analysis of senescence markers in glial cells enriched fraction from fresh brainstem samples demonstrated that glial cells are more susceptible to senesce with age in the brainstem. In conclusion, our study suggests that aging induces glial senescence in the brainstem which likely causes inflammation and SNS overactivity.
Collapse
Affiliation(s)
- Priya Balasubramanian
- Oklahoma Center for Geroscience and Healthy Brain Aging, The University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | - Lyndee Branen
- Department of Physiological Sciences, College of Veterinary Medicine, Oklahoma State University, Stillwater, OK 74078, USA
| | - Mahesh Kumar Sivasubramanian
- Department of Physiological Sciences, College of Veterinary Medicine, Oklahoma State University, Stillwater, OK 74078, USA
| | - Raisa Monteiro
- Department of Physiological Sciences, College of Veterinary Medicine, Oklahoma State University, Stillwater, OK 74078, USA
| | - Madhan Subramanian
- Department of Physiological Sciences, College of Veterinary Medicine, Oklahoma State University, Stillwater, OK 74078, USA
| |
Collapse
|
54
|
López-Domínguez JA, Rodríguez-López S, Ahumada-Castro U, Desprez PY, Konovalenko M, Laberge RM, Cárdenas C, Villalba JM, Campisi J. Cdkn1a transcript variant 2 is a marker of aging and cellular senescence. Aging (Albany NY) 2021; 13:13380-13392. [PMID: 34035185 PMCID: PMC8202863 DOI: 10.18632/aging.203110] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2021] [Accepted: 05/18/2021] [Indexed: 12/18/2022]
Abstract
Cellular senescence is a cell fate response characterized by a permanent cell cycle arrest driven primarily the by cell cycle inhibitor and tumor suppressor proteins p16Ink4a and p21Cip1/Waf1. In mice, the p21Cip1/Waf1 encoding locus, Cdkn1a, is known to generate two transcripts that produce identical proteins, but one of these transcript variants is poorly characterized. We show that the Cdkn1a transcript variant 2, but not the better-studied variant 1, is selectively elevated during natural aging across multiple mouse tissues. Importantly, mouse cells induced to senescence in culture by genotoxic stress (ionizing radiation or doxorubicin) upregulated both transcripts, but with different temporal dynamics: variant 1 responded nearly immediately to genotoxic stress, whereas variant 2 increased much more slowly as cells acquired senescent characteristics. Upon treating mice systemically with doxorubicin, which induces widespread cellular senescence in vivo, variant 2 increased to a larger extent than variant 1. Variant 2 levels were also more sensitive to the senolytic drug ABT-263 in naturally aged mice. Thus, variant 2 is a novel and more sensitive marker than variant 1 or total p21Cip1/Waf1 protein for assessing the senescent cell burden and clearance in mice.
Collapse
Affiliation(s)
| | - Sandra Rodríguez-López
- Departamento de Biología Celular, Fisiología e Inmunología, Universidad de Córdoba, Campus de Excelencia Internacional Agroalimentario, 14071, Córdoba, Spain
| | - Ulises Ahumada-Castro
- Center for Integrative Biology, Faculty of Sciences, Universidad Mayor, Santiago 2422, Chile
- Geroscience Center for Brain Health and Metabolism, Santiago, Chile
| | | | | | | | - César Cárdenas
- Buck Institute for Research on Aging, Novato, CA 94945, USA
- Center for Integrative Biology, Faculty of Sciences, Universidad Mayor, Santiago 2422, Chile
- Geroscience Center for Brain Health and Metabolism, Santiago, Chile
- Department of Chemistry and Biochemistry, University of California, Santa Barbara, CA 93106, USA
| | - José Manuel Villalba
- Departamento de Biología Celular, Fisiología e Inmunología, Universidad de Córdoba, Campus de Excelencia Internacional Agroalimentario, 14071, Córdoba, Spain
| | - Judith Campisi
- Buck Institute for Research on Aging, Novato, CA 94945, USA
- Lawrence Berkeley National Laboratory, University of California, Berkeley, CA 94720, USA
| |
Collapse
|
55
|
Stojanović SD, Fiedler J, Bauersachs J, Thum T, Sedding DG. Senescence-induced inflammation: an important player and key therapeutic target in atherosclerosis. Eur Heart J 2021; 41:2983-2996. [PMID: 31898722 PMCID: PMC7453834 DOI: 10.1093/eurheartj/ehz919] [Citation(s) in RCA: 96] [Impact Index Per Article: 32.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/22/2019] [Revised: 09/13/2019] [Accepted: 12/12/2019] [Indexed: 12/21/2022] Open
Abstract
Inflammation is a hallmark and potent driver of pathological vascular remodelling in atherosclerosis. However, current anti-inflammatory therapeutic strategies have shown mixed results. As an alternative perspective on the conundrum of chronic inflammation emerging evidence points towards a small subset of senescent cells as a critical player and central node driving atherosclerosis. Senescent cells belonging to various cell types are a dominant and chronic source of a large array of pro-inflammatory cytokines and various additional plaque destabilizing factors, being involved with various aspects of atherosclerosis pathogenesis. Antagonizing these key agitators of local chronic inflammation and plaque instability may provide a causative and multi-purpose therapeutic strategy to treat atherosclerosis. Anti-senescence treatment options with translational potential are currently in development. However, several questions and challenges remain to be addressed before these novel treatment approaches may enter the clinical setting.
Collapse
Affiliation(s)
- Stevan D Stojanović
- Institute of Molecular and Translational Therapeutic Strategies (IMTTS), Hannover Medical School, Carl-Neuberg-Strasse 1, 30625 Hannover, Germany.,Department of Cardiology and Angiology, Hannover Medical School, Carl-Neuberg-Strasse 1, 30625 Hannover, Germany
| | - Jan Fiedler
- Institute of Molecular and Translational Therapeutic Strategies (IMTTS), Hannover Medical School, Carl-Neuberg-Strasse 1, 30625 Hannover, Germany
| | - Johann Bauersachs
- Department of Cardiology and Angiology, Hannover Medical School, Carl-Neuberg-Strasse 1, 30625 Hannover, Germany
| | - Thomas Thum
- Institute of Molecular and Translational Therapeutic Strategies (IMTTS), Hannover Medical School, Carl-Neuberg-Strasse 1, 30625 Hannover, Germany
| | - Daniel G Sedding
- Department of Internal Medicine III, Cardiology, Angiology and Intensive Care Medicine, Martin-Luther-University Halle (Saale), Ernst-Grube-Strasse 40, 06120 Halle (Saale), Germany
| |
Collapse
|
56
|
Chromatin-Directed Proteomics Identifies ZNF84 as a p53-Independent Regulator of p21 in Genotoxic Stress Response. Cancers (Basel) 2021; 13:cancers13092115. [PMID: 33925586 PMCID: PMC8123910 DOI: 10.3390/cancers13092115] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Revised: 04/18/2021] [Accepted: 04/22/2021] [Indexed: 12/12/2022] Open
Abstract
Simple Summary Chemotherapy is a commonly applied anticancer treatment, however therapy-induced senescent growth arrest has been associated with aggressive disease recurrence. The p21 protein, encoded by CDKN1A, plays a vital role in the induction of senescence. Its transcriptional control by p53 is well-established. However, in many cancers where TP53 is mutated, p21 expression must be triggered by p53-independent mechanisms. We here used a chromatin-directed proteomic approach and identified ZNF84 as a regulator of CDKN1A gene expression in various p53-deficient cell lines. Knock-down of ZNF84, an as-yet un-characterized protein, inhibited p21 gene and protein expression in response to doxorubicin and facilitated senescence bypass. Intriguingly, ZNF84 depletion diminished genotoxic burden evoked by doxorubicin. Clinical data association studies indicated the relevance of ZNF84 expression for patient survival. Collectively, we identified ZNF84 as a critical regulator of senescence-proliferation outcome of chemotherapy, opening possibilities for its targeting in novel anti-cancer therapies of p53-mutated tumours. Abstract The p21WAF1/Cip1 protein, encoded by CDKN1A, plays a vital role in senescence, and its transcriptional control by the tumour suppressor p53 is well-established. However, p21 can also be regulated in a p53-independent manner, by mechanisms that still remain less understood. We aimed to expand the knowledge about p53-independent senescence by looking for novel players involved in CDKN1A regulation. We used a chromatin-directed proteomic approach and identified ZNF84 as a novel regulator of p21 in various p53-deficient cell lines treated with cytostatic dose of doxorubicin. Knock-down of ZNF84, an as-yet un-characterized protein, inhibited p21 gene and protein expression in response to doxorubicin, it attenuated senescence and was associated with enhanced proliferation, indicating that ZNF84-deficiency can favor senescence bypass. ZNF84 deficiency was also associated with transcriptomic changes in genes governing various cancer-relevant processes e.g., mitosis. In cells with ZNF84 knock-down we discovered significantly lower level of H2AX Ser139 phosphorylation (γH2AX), which is triggered by DNA double strand breaks. Intriguingly, we observed a reverse correlation between the level of ZNF84 expression and survival rate of colon cancer patients. In conclusion, ZNF84, whose function was previously not recognized, was identified here as a critical p53-independent regulator of senescence, opening possibilities for its targeting in novel therapies of p53-null cancers.
Collapse
|
57
|
Sándor S, Tátrai K, Czeibert K, Egyed B, Kubinyi E. CDKN2A Gene Expression as a Potential Aging Biomarker in Dogs. Front Vet Sci 2021; 8:660435. [PMID: 33981746 PMCID: PMC8107359 DOI: 10.3389/fvets.2021.660435] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Accepted: 03/18/2021] [Indexed: 12/04/2022] Open
Abstract
Describing evolutionary conserved physiological or molecular patterns, which can reliably mark the age of both model organisms and humans or predict the onset of age-related pathologies has become a priority in aging research. The age-related gene-expression changes of the Cyclin Dependent Kinase Inhibitor 2A (CDKN2A) gene have been well-documented in humans and rodents. However, data is lacking from other relevant species, including dogs. Therefore, we quantified the CDKN2A mRNA abundance in dogs of different ages, in four tissue types: the frontal cortex of the brain, temporal muscle, skin, and blood. We found a significant, positive correlation between CDKN2A relative expression values and age in the brain, muscle, and blood; however, no correlation was detected in the skin. The strongest correlation was detected in the brain tissue (CDKN2A/GAPDH: r = 0.757, p < 0.001), similarly to human findings, while the muscle and blood showed weaker, but significant correlation. Our results suggest that CDKN2A might be a potential blood-borne biomarker of aging in dogs, although the validation and optimization will require further, more focused research. Our current results also clearly demonstrate that the role of CDKN2A in aging is conserved in dogs, regarding both tissue specificity and a pivotal role of CDKN2A in brain aging.
Collapse
Affiliation(s)
- Sára Sándor
- Department of Ethology, Senior Family Dog Project, ELTE Eötvös Loránd University, Budapest, Hungary
| | - Kitti Tátrai
- Department of Ethology, Senior Family Dog Project, ELTE Eötvös Loránd University, Budapest, Hungary
- Department of Genetics, ELTE Eötvös Loránd University, Budapest, Hungary
| | - Kálmán Czeibert
- Department of Ethology, Senior Family Dog Project, ELTE Eötvös Loránd University, Budapest, Hungary
| | - Balázs Egyed
- Department of Genetics, ELTE Eötvös Loránd University, Budapest, Hungary
| | - Enikő Kubinyi
- Department of Ethology, Senior Family Dog Project, ELTE Eötvös Loránd University, Budapest, Hungary
| |
Collapse
|
58
|
Moustogiannis A, Philippou A, Taso O, Zevolis E, Pappa M, Chatzigeorgiou A, Koutsilieris M. The Effects of Muscle Cell Aging on Myogenesis. Int J Mol Sci 2021; 22:ijms22073721. [PMID: 33918414 PMCID: PMC8038215 DOI: 10.3390/ijms22073721] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2021] [Revised: 03/30/2021] [Accepted: 03/31/2021] [Indexed: 02/06/2023] Open
Abstract
The process of myogenesis gradually deteriorates as the skeletal muscle ages, contributing to muscle mass loss. The aim of this study is to investigate the effect of senescence/aging on skeletal myogenesis, in vitro. A model of multiple cell divisions of C2C12 myoblasts was used to replicate cell senescence. Control and aged myoblasts were investigated during myogenesis, i.e., at days 0, 2, and 6of differentiation. SA-β-gal activity and comet assay were used as markers of aging and DNA damage. Flow cytometry was performed to characterize potential differences in cell cycle between control and aged cells. Alterations in the mRNA and/or protein expression of myogenic regulatory factors (MRFs), IGF-1 isoforms, apoptotic, atrophy, inflammatory, metabolic and aging-related factors were evaluated. Compared with the control cells, aged myoblasts exhibited G0/G1 cell cycle arrest, DNA damage, increased SA-β-gal activity, and increased expression of aging-related factors p16 and p21 during differentiation. Moreover, aged myoblasts showed a reduction in the expression of MRFs and metabolic/anabolic factors, along with an increased expression of apoptotic, atrophy and inflammatory factors. A diminished differentiation capacity characterized the aged myoblasts which, in combination with the induction of apoptotic and atrophy factors, indicated a disrupted myogenic lineage in the senescent muscle cells.
Collapse
Affiliation(s)
- Athanasios Moustogiannis
- Department of Physiology, Medical School, National and Kapodistrian University of Athens, 75 MicrasAsias, 115 27 Goudi-Athens, Greece; (A.P.); (O.T.); (E.Z.); (A.C.); (M.K.)
- Correspondence: ; Tel.: +30-210-7462690; Fax: +30-210-7462571
| | - Anastassios Philippou
- Department of Physiology, Medical School, National and Kapodistrian University of Athens, 75 MicrasAsias, 115 27 Goudi-Athens, Greece; (A.P.); (O.T.); (E.Z.); (A.C.); (M.K.)
| | - Orjona Taso
- Department of Physiology, Medical School, National and Kapodistrian University of Athens, 75 MicrasAsias, 115 27 Goudi-Athens, Greece; (A.P.); (O.T.); (E.Z.); (A.C.); (M.K.)
| | - Evangelos Zevolis
- Department of Physiology, Medical School, National and Kapodistrian University of Athens, 75 MicrasAsias, 115 27 Goudi-Athens, Greece; (A.P.); (O.T.); (E.Z.); (A.C.); (M.K.)
| | - Maria Pappa
- First Department of Propaedeutic Internal Medicine, Joint Rheumatology Program, National and Kapodistrian University of Athens, 75 MicrasAsias, 115 27 Goudi-Athens, Greece;
| | - Antonios Chatzigeorgiou
- Department of Physiology, Medical School, National and Kapodistrian University of Athens, 75 MicrasAsias, 115 27 Goudi-Athens, Greece; (A.P.); (O.T.); (E.Z.); (A.C.); (M.K.)
| | - Michael Koutsilieris
- Department of Physiology, Medical School, National and Kapodistrian University of Athens, 75 MicrasAsias, 115 27 Goudi-Athens, Greece; (A.P.); (O.T.); (E.Z.); (A.C.); (M.K.)
| |
Collapse
|
59
|
Kumari R, Jat P. Mechanisms of Cellular Senescence: Cell Cycle Arrest and Senescence Associated Secretory Phenotype. Front Cell Dev Biol 2021; 9:645593. [PMID: 33855023 PMCID: PMC8039141 DOI: 10.3389/fcell.2021.645593] [Citation(s) in RCA: 685] [Impact Index Per Article: 228.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Accepted: 02/16/2021] [Indexed: 01/10/2023] Open
Abstract
Cellular senescence is a stable cell cycle arrest that can be triggered in normal cells in response to various intrinsic and extrinsic stimuli, as well as developmental signals. Senescence is considered to be a highly dynamic, multi-step process, during which the properties of senescent cells continuously evolve and diversify in a context dependent manner. It is associated with multiple cellular and molecular changes and distinct phenotypic alterations, including a stable proliferation arrest unresponsive to mitogenic stimuli. Senescent cells remain viable, have alterations in metabolic activity and undergo dramatic changes in gene expression and develop a complex senescence-associated secretory phenotype. Cellular senescence can compromise tissue repair and regeneration, thereby contributing toward aging. Removal of senescent cells can attenuate age-related tissue dysfunction and extend health span. Senescence can also act as a potent anti-tumor mechanism, by preventing proliferation of potentially cancerous cells. It is a cellular program which acts as a double-edged sword, with both beneficial and detrimental effects on the health of the organism, and considered to be an example of evolutionary antagonistic pleiotropy. Activation of the p53/p21WAF1/CIP1 and p16INK4A/pRB tumor suppressor pathways play a central role in regulating senescence. Several other pathways have recently been implicated in mediating senescence and the senescent phenotype. Herein we review the molecular mechanisms that underlie cellular senescence and the senescence associated growth arrest with a particular focus on why cells stop dividing, the stability of the growth arrest, the hypersecretory phenotype and how the different pathways are all integrated.
Collapse
Affiliation(s)
- Ruchi Kumari
- MRC Prion Unit at UCL, UCL Institute of Prion Diseases, London, United Kingdom
| | - Parmjit Jat
- MRC Prion Unit at UCL, UCL Institute of Prion Diseases, London, United Kingdom
| |
Collapse
|
60
|
Sorci G, Léchenault-Bergerot C, Faivre B. Age reduces resistance and tolerance in malaria-infected mice. INFECTION GENETICS AND EVOLUTION 2020; 88:104698. [PMID: 33370596 DOI: 10.1016/j.meegid.2020.104698] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/17/2020] [Revised: 12/16/2020] [Accepted: 12/23/2020] [Indexed: 12/30/2022]
Abstract
Once infected, hosts can rely on two strategies to cope with parasites: fight them (resist the infection) or minimize the damage they induce (tolerate the infection). While there is evidence that aging reduces resistance, how tolerance varies as hosts become old has been barely studied. Here, we used a rodent malaria parasite (Plasmodium yoelii) to investigate whether 2- and 12-month old house mice differ in their capacity to resist and tolerate the infection. We found that 12-month old mice harbored higher parasitemia, showing that age reduces resistance to malaria. Infection-induced deterioration of host health was assessed using red blood cell and body mass loss. Using both traits, the rate of decline in host health, as parasitemia increased, was more pronounced in 12- than in 2-month old mice, showing that age is also associated with impaired tolerance to malaria. Overall, resistance and tolerance positively covaried; however, this was only due to the age effect, since, within age classes, the two traits were not correlated. These results show that senescing individuals might be both more susceptible to infectious diseases and less able to cope with the damage that infection induces.
Collapse
Affiliation(s)
- Gabriele Sorci
- Biogéosciences, CNRS UMR 6282, Université de Bourgogne Franche-Comté, 6 Boulevard Gabriel, 21000 Dijon, France.
| | | | - Bruno Faivre
- Biogéosciences, CNRS UMR 6282, Université de Bourgogne Franche-Comté, 6 Boulevard Gabriel, 21000 Dijon, France
| |
Collapse
|
61
|
van der Rijt S, Molenaars M, McIntyre RL, Janssens GE, Houtkooper RH. Integrating the Hallmarks of Aging Throughout the Tree of Life: A Focus on Mitochondrial Dysfunction. Front Cell Dev Biol 2020; 8:594416. [PMID: 33324647 PMCID: PMC7726203 DOI: 10.3389/fcell.2020.594416] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2020] [Accepted: 11/04/2020] [Indexed: 12/14/2022] Open
Abstract
Since the identification and definition of the hallmarks of aging, these aspects of molecular and cellular decline have been most often described as isolated or distinct mechanisms. However, there is significant evidence demonstrating interplay between most of these hallmarks and that they have the capacity to influence and regulate one another. These interactions are demonstrable across the tree of life, yet not all aspects are conserved. Here, we describe an integrative view on the hallmarks of aging by using the hallmark "mitochondrial dysfunction" as a focus point, and illustrate its capacity to both influence and be influenced by the other hallmarks of aging. We discuss the effects of mitochondrial pathways involved in aging, such as oxidative phosphorylation, mitochondrial dynamics, mitochondrial protein synthesis, mitophagy, reactive oxygen species and mitochondrial DNA damage in relation to each of the primary, antagonistic and integrative hallmarks. We discuss the similarities and differences in these interactions throughout the tree of life, and speculate how speciation may play a role in the variation in these mechanisms. We propose that the hallmarks are critically intertwined, and that mapping the full extent of these interactions would be of significant benefit to the aging research community.
Collapse
Affiliation(s)
- Sanne van der Rijt
- Laboratory Genetic Metabolic Diseases, Amsterdam Gastroenterology Endocrinology Metabolism, Amsterdam Cardiovascular Sciences, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
| | - Marte Molenaars
- Laboratory Genetic Metabolic Diseases, Amsterdam Gastroenterology Endocrinology Metabolism, Amsterdam Cardiovascular Sciences, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
| | - Rebecca L McIntyre
- Laboratory Genetic Metabolic Diseases, Amsterdam Gastroenterology Endocrinology Metabolism, Amsterdam Cardiovascular Sciences, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
| | - Georges E Janssens
- Laboratory Genetic Metabolic Diseases, Amsterdam Gastroenterology Endocrinology Metabolism, Amsterdam Cardiovascular Sciences, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
| | - Riekelt H Houtkooper
- Laboratory Genetic Metabolic Diseases, Amsterdam Gastroenterology Endocrinology Metabolism, Amsterdam Cardiovascular Sciences, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
| |
Collapse
|
62
|
Hall BM, Gleiberman AS, Strom E, Krasnov PA, Frescas D, Vujcic S, Leontieva OV, Antoch MP, Kogan V, Koman IE, Zhu Y, Tchkonia T, Kirkland JL, Chernova OB, Gudkov AV. Immune checkpoint protein VSIG4 as a biomarker of aging in murine adipose tissue. Aging Cell 2020; 19:e13219. [PMID: 32856419 PMCID: PMC7576241 DOI: 10.1111/acel.13219] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2020] [Revised: 05/26/2020] [Accepted: 07/03/2020] [Indexed: 12/20/2022] Open
Abstract
Adipose tissue is recognized as a major source of systemic inflammation with age, driving age-related tissue dysfunction and pathogenesis. Macrophages (Mφ) are central to these changes yet adipose tissue Mφ (ATMs) from aged mice remain poorly characterized. To identify biomarkers underlying changes in aged adipose tissue, we performed an unbiased RNA-seq analysis of ATMs from young (8-week-old) and healthy aged (80-week-old) mice. One of the genes identified, V-set immunoglobulin-domain-containing 4 (VSIG4/CRIg), encodes a Mφ-associated complement receptor and B7 family-related immune checkpoint protein. Here, we demonstrate that Vsig4 expression is highly upregulated with age in perigonadal white adipose tissue (gWAT) in two mouse strains (inbred C57BL/6J and outbred NIH Swiss) independent of gender. The accumulation of VSIG4 was mainly attributed to a fourfold increase in the proportion of VSIG4+ ATMs (13%-52%). In a longitudinal study, VSIG4 expression in gWAT showed a strong correlation with age within a cohort of male and female mice and correlated strongly with physiological frailty index (PFI, a multi-parameter assessment of health) in male mice. Our results indicate that VSIG4 is a novel biomarker of aged murine ATMs. VSIG4 expression was also found to be elevated in other aging tissues (e.g., thymus) and was strongly induced in tumor-adjacent stroma in cases of spontaneous and xenograft lung cancer models. VSIG4 expression was recently associated with cancer and several inflammatory diseases with diagnostic and prognostic potential in both mice and humans. Further investigation is required to determine whether VSIG4-positive Mφ contribute to immunosenescence and/or systemic age-related deficits.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Olga V. Leontieva
- Department of Pharmacology and TherapeuticsRoswell Park Comprehensive Cancer CenterBuffaloNYUSA
| | - Marina P. Antoch
- Department of Pharmacology and TherapeuticsRoswell Park Comprehensive Cancer CenterBuffaloNYUSA
| | - Valeria Kogan
- Institute for Translational ResearchAriel UniversityArielIsrael
| | - Igor E. Koman
- Institute for Translational ResearchAriel UniversityArielIsrael
| | - Yi Zhu
- Robert and Arlene Kogod Center on AgingMayo ClinicRochesterMNUSA
| | - Tamara Tchkonia
- Robert and Arlene Kogod Center on AgingMayo ClinicRochesterMNUSA
| | | | | | - Andrei V. Gudkov
- Everon Biosciences IncBuffaloNYUSA
- Department of Cell Stress BiologyRoswell Park Comprehensive Cancer CenterBuffaloNYUSA
- Genome Protection IncBuffaloNYUSA
| |
Collapse
|
63
|
Wilkinson HN, Hardman MJ. Senescence in Wound Repair: Emerging Strategies to Target Chronic Healing Wounds. Front Cell Dev Biol 2020; 8:773. [PMID: 32850866 PMCID: PMC7431694 DOI: 10.3389/fcell.2020.00773] [Citation(s) in RCA: 74] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2020] [Accepted: 07/22/2020] [Indexed: 01/10/2023] Open
Abstract
Cellular senescence is a fundamental stress response that restrains tumour formation. Yet, senescence cells are also present in non-cancerous states, accumulating exponentially with chronological age and contributing to age- and diabetes-related cellular dysfunction. The identification of hypersecretory and phagocytic behaviours in cells that were once believed to be non-functional has led to a recent explosion of senescence research. Here we discuss the profound, and often opposing, roles identified for short-lived vs. chronic tissue senescence. Transiently induced senescence is required for development, regeneration and acute wound repair, while chronic senescence is widely implicated in tissue pathology. We recently demonstrated that sustained senescence contributes to impaired diabetic healing via the CXCR2 receptor, which when blocked promotes repair. Further studies have highlighted the beneficial effects of targeting a range of senescence-linked processes to fight disease. Collectively, these findings hold promise for developing clinically viable strategies to tackle senescence in chronic wounds and other cutaneous pathologies.
Collapse
Affiliation(s)
- Holly N Wilkinson
- Centre for Atherothrombosis and Metabolic Disease, Hull York Medical School, University of Hull, Hull, United Kingdom
| | - Matthew J Hardman
- Centre for Atherothrombosis and Metabolic Disease, Hull York Medical School, University of Hull, Hull, United Kingdom
| |
Collapse
|
64
|
Insulin-Like Growth Factor I Prevents Cellular Aging via Activation of Mitophagy. J Aging Res 2020; 2020:4939310. [PMID: 32802505 PMCID: PMC7416301 DOI: 10.1155/2020/4939310] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2019] [Revised: 05/22/2020] [Accepted: 07/06/2020] [Indexed: 12/18/2022] Open
Abstract
Mitochondrial dysfunction is a hallmark of cellular aging. Mitophagy is a critical mitochondrial quality control mechanism that removes dysfunctional mitochondria and contributes to cell survival. Insulin-like growth factor 1 (IGF-1) promotes survival of smooth muscle cells (SMCs), but its potential effect on cellular aging is unknown yet. We found that IGF-1 decreased cell senescence, prevented DNA telomere shortening, increased mitochondrial membrane potential, activated cytochrome C oxidase, and reduced mitochondrial DNA damage in long-term cultured (aged) aortic SMC, suggesting an antiaging effect. IGF-1 increased mitophagy in aged cells, and this was associated with decreased expression of cyclin-dependent kinase inhibitors p16 and p21 and elevated levels of Nrf2 and Sirt3, regulators of mitophagy and mitochondrial biogenesis. SiRNA-induced inhibition of either Nrf2 or Sirt3 blocked IGF-1-induced upregulation of mitophagy, suggesting that the Nrf2/Sirt3 pathway was required for IGF-1's effect on mitophagy. PINK1 is a master regulator of mitophagy. PINK1 silencing suppressed mitophagy and inhibited IGF-1-induced antiaging effects in aged SMC, consistent with an essential role of mitophagy in IGF-1's effect on cellular aging. Thus, IGF-1 inhibited cellular aging via Nrf2/Sirt3-dependent activation of mitophagy. Our data suggest that activation of IGF-1 signaling is a novel potential strategy to activate mitophagy and slow cellular aging.
Collapse
|
65
|
Guo J, Zheng HJ, Zhang W, Lou W, Xia C, Han XT, Huang WJ, Zhang F, Wang Y, Liu WJ. Accelerated Kidney Aging in Diabetes Mellitus. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2020; 2020:1234059. [PMID: 32774664 PMCID: PMC7407029 DOI: 10.1155/2020/1234059] [Citation(s) in RCA: 49] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Revised: 05/25/2020] [Accepted: 06/25/2020] [Indexed: 02/07/2023]
Abstract
With aging, the kidney undergoes inexorable and progressive changes in structural and functional performance. These aging-related alterations are more obvious and serious in diabetes mellitus (DM). Renal accelerated aging under DM conditions is associated with multiple stresses such as accumulation of advanced glycation end products (AGEs), hypertension, oxidative stress, and inflammation. The main hallmarks of cellular senescence in diabetic kidneys include cyclin-dependent kinase inhibitors, telomere shortening, and diabetic nephropathy-associated secretory phenotype. Lysosome-dependent autophagy and antiaging proteins Klotho and Sirt1 play a fundamental role in the accelerated aging of kidneys in DM, among which the autophagy-lysosome system is the convergent mechanism of the multiple antiaging pathways involved in renal aging under DM conditions. Metformin and the inhibitor of sodium-glucose cotransporter 2 are recommended due to their antiaging effects independent of antihyperglycemia, besides angiotensin-converting enzyme inhibitors/angiotensin receptor blockers. Additionally, diet intervention including low protein and low AGEs with antioxidants are suggested for patients with diabetic nephropathy (DN). However, their long-term benefits still need further study. Exploring the interactive relationships among antiaging protein Klotho, Sirt1, and autophagy-lysosome system may provide insight into better satisfying the urgent medical needs of elderly patients with aging-related DN.
Collapse
Affiliation(s)
- Jing Guo
- Renal Research Institution; Key Laboratory of Chinese Internal Medicine of Ministry of Education and Beijing, Dongzhimen Hospital Affiliated to Beijing University of Chinese Medicine, Beijing 100700, China
| | - Hui Juan Zheng
- Renal Research Institution; Key Laboratory of Chinese Internal Medicine of Ministry of Education and Beijing, Dongzhimen Hospital Affiliated to Beijing University of Chinese Medicine, Beijing 100700, China
| | - Wenting Zhang
- Renal Research Institution; Key Laboratory of Chinese Internal Medicine of Ministry of Education and Beijing, Dongzhimen Hospital Affiliated to Beijing University of Chinese Medicine, Beijing 100700, China
| | - Wenjiao Lou
- Renal Research Institution; Key Laboratory of Chinese Internal Medicine of Ministry of Education and Beijing, Dongzhimen Hospital Affiliated to Beijing University of Chinese Medicine, Beijing 100700, China
| | - Chenhui Xia
- Renal Research Institution; Key Laboratory of Chinese Internal Medicine of Ministry of Education and Beijing, Dongzhimen Hospital Affiliated to Beijing University of Chinese Medicine, Beijing 100700, China
| | - Xue Ting Han
- Renal Research Institution; Key Laboratory of Chinese Internal Medicine of Ministry of Education and Beijing, Dongzhimen Hospital Affiliated to Beijing University of Chinese Medicine, Beijing 100700, China
| | - Wei Jun Huang
- Renal Research Institution; Key Laboratory of Chinese Internal Medicine of Ministry of Education and Beijing, Dongzhimen Hospital Affiliated to Beijing University of Chinese Medicine, Beijing 100700, China
| | - Fan Zhang
- Renal Research Institution; Key Laboratory of Chinese Internal Medicine of Ministry of Education and Beijing, Dongzhimen Hospital Affiliated to Beijing University of Chinese Medicine, Beijing 100700, China
| | - Yaoxian Wang
- Renal Research Institution; Key Laboratory of Chinese Internal Medicine of Ministry of Education and Beijing, Dongzhimen Hospital Affiliated to Beijing University of Chinese Medicine, Beijing 100700, China
| | - Wei Jing Liu
- Renal Research Institution; Key Laboratory of Chinese Internal Medicine of Ministry of Education and Beijing, Dongzhimen Hospital Affiliated to Beijing University of Chinese Medicine, Beijing 100700, China
- Institute of Nephrology, and Zhanjiang Key Laboratory of Prevention and Management of Chronic Kidney Disease, Guangdong Medical University, No. 57th South Renmin Road, Zhanjiang, Guangdong 524001, China
| |
Collapse
|
66
|
Qin X, Du D, Chen Q, Wu M, Wu T, Wen J, Jin Y, Zhang J, Wang S. Metformin prevents murine ovarian aging. Aging (Albany NY) 2020; 11:3785-3794. [PMID: 31182682 PMCID: PMC6594816 DOI: 10.18632/aging.102016] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2019] [Accepted: 05/31/2019] [Indexed: 02/07/2023]
Abstract
A number of studies have shown that metformin can delay aging process and extend healthy lifespan in animals. However, its role in female reproductive lifespan is unclear. This study was aimed to explore the potential anti-aging effect of metformin on the ovary and its possible mechanisms. Female C57BL/6 mice of 27-week old were divided into two groups, the control group (CON) and metformin-treated group (MET). CON mice were fed ad libitum, while MET mice were fed on chows supplied with 100mg/kg metformin for half a year. Ovarian reserve and function were assessed by ovarian follicle counts, estrous cycle and sex hormones levels. The expressions of oxidized metabolites, such as 8-hydroxy-2´-deoxyguanosine (8-OHdG), 4-hydroxynonenal (4-HNE), nitrotyrosine (NTY), and ovarian aging associated proteins P16, SIRT1, p-rpS6 and Bcl2 were examined. The MET mice exhibited increased level of serum E2 hormone and higher percentage of regular estrous cycles after 6 months' feeding, compared to the CON mice. The amount of primordial and primary follicles and the expression of SIRT1 were significantly increased, but the levels of P16, 8-OHdG, 4-HNE and p-rpS6 were decreased in the MET mice. These results indicate that metformin can delay ovarian aging process, probably by inducing the expression of SIRT1 and reducing the oxidative damage.
Collapse
Affiliation(s)
- Xian Qin
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, People's Republic of China.,Department of Obstetrics and Gynecology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, People's Republic of China
| | - Dingfu Du
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, People's Republic of China
| | - Qian Chen
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, People's Republic of China
| | - Meng Wu
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, People's Republic of China
| | - Tong Wu
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, People's Republic of China
| | - Jingyi Wen
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, People's Republic of China
| | - Yan Jin
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, People's Republic of China
| | - Jinjin Zhang
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, People's Republic of China
| | - Shixuan Wang
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, People's Republic of China
| |
Collapse
|
67
|
Johnson AA, Shokhirev MN, Wyss-Coray T, Lehallier B. Systematic review and analysis of human proteomics aging studies unveils a novel proteomic aging clock and identifies key processes that change with age. Ageing Res Rev 2020; 60:101070. [PMID: 32311500 DOI: 10.1016/j.arr.2020.101070] [Citation(s) in RCA: 83] [Impact Index Per Article: 20.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Revised: 03/23/2020] [Accepted: 04/07/2020] [Indexed: 12/14/2022]
Abstract
The development of clinical interventions that significantly improve human healthspan requires robust markers of biological age as well as thoughtful therapeutic targets. To promote these goals, we performed a systematic review and analysis of human aging and proteomics studies. The systematic review includes 36 different proteomics analyses, each of which identified proteins that significantly changed with age. We discovered 1,128 proteins that had been reported by at least two or more analyses and 32 proteins that had been reported by five or more analyses. Each of these 32 proteins has known connections relevant to aging and age-related disease. GDF15, for example, extends both lifespan and healthspan when overexpressed in mice and is additionally required for the anti-diabetic drug metformin to exert beneficial effects on body weight and energy balance. Bioinformatic enrichment analyses of our 1,128 commonly identified proteins heavily implicated processes relevant to inflammation, the extracellular matrix, and gene regulation. We additionally propose a novel proteomic aging clock comprised of proteins that were reported to change with age in plasma in three or more different studies. Using a large patient cohort comprised of 3,301 subjects (aged 18-76 years), we demonstrate that this clock is able to accurately predict human age.
Collapse
|
68
|
Sex Differences in Progression of Diabetic Cardiomyopathy in OVE26 Type 1 Diabetic Mice. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2020; 2020:6961348. [PMID: 32509150 PMCID: PMC7244980 DOI: 10.1155/2020/6961348] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/13/2020] [Revised: 03/14/2020] [Accepted: 04/01/2020] [Indexed: 12/13/2022]
Abstract
OVE26 mice are a widely used transgenic model of early-onset type 1 diabetes. These mice overexpress calmodulin in their pancreatic β cells, develop severe diabetes within the first weeks of life, and progress to severe diabetic complications including diabetic nephropathy and diabetic cardiomyopathy (DCM). To date, diabetic nephropathy in OVE26 mice has been well explored, leaving the progression of DCM and the gender impact in this type 1 diabetes model still unrevealed. In our study, male and female OVE26 mice and age-matched nondiabetic FVB mice were examined at 4, 12, 24, and 36 weeks for their cardiac function, body weight, blood glucose, and heart weight/tibia length ratio. Further, histopathological examination and Western blot analysis for the key markers demonstrate that DCM appears at 24 weeks OVE26 mice, initiating with cardiac senescence, followed by fibrosis and then cardiac dysfunction. Mitochondrial respiration function analysis showed no indication of dysfunction in OVE26 mice at 24 weeks of age in both genders. In addition, no significant difference for the pathogenic progression was observed between OVE26 and FVB mice in both males and females. In conclusion, this study suggests cardiac senescence and fibrosis, which may be amended by sex differences, play key roles in the progression of DCM in OVE26 mice. The comprehensive characterization of diabetic cardiomyopathy progression and the sex difference impact in OVE26 mice provides a basis for future study on DCM using OVE26 mice.
Collapse
|
69
|
Yu Q, Xiao H, Jedrychowski MP, Schweppe DK, Navarrete-Perea J, Knott J, Rogers J, Chouchani ET, Gygi SP. Sample multiplexing for targeted pathway proteomics in aging mice. Proc Natl Acad Sci U S A 2020; 117:9723-9732. [PMID: 32332170 PMCID: PMC7211924 DOI: 10.1073/pnas.1919410117] [Citation(s) in RCA: 65] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Pathway proteomics strategies measure protein expression changes in specific cellular processes that carry out related functions. Using targeted tandem mass tags-based sample multiplexing, hundreds of proteins can be quantified across 10 or more samples simultaneously. To facilitate these highly complex experiments, we introduce a strategy that provides complete control over targeted sample multiplexing experiments, termed Tomahto, and present its implementation on the Orbitrap Tribrid mass spectrometer platform. Importantly, this software monitors via the external desktop computer to the data stream and inserts optimized MS2 and MS3 scans in real time based on an application programming interface with the mass spectrometer. Hundreds of proteins of interest from diverse biological samples can be targeted and accurately quantified in a sensitive and high-throughput fashion. It achieves sensitivity comparable to, if not better than, deep fractionation and requires minimal total sample input (∼10 µg). As a proof-of-principle experiment, we selected four pathways important in metabolism- and inflammation-related processes (260 proteins/520 peptides) and measured their abundance across 90 samples (nine tissues from five old and five young mice) to explore effects of aging. Tissue-specific aging is presented here and we highlight the role of inflammation- and metabolism-related processes in white adipose tissue. We validated our approach through comparison with a global proteome survey across the tissues, work that we also provide as a general resource for the community.
Collapse
Affiliation(s)
- Qing Yu
- Department of Cell Biology, Harvard Medical School, Boston, MA 02115
| | - Haopeng Xiao
- Department of Cell Biology, Harvard Medical School, Boston, MA 02115
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA 02115
| | - Mark P Jedrychowski
- Department of Cell Biology, Harvard Medical School, Boston, MA 02115
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA 02115
| | - Devin K Schweppe
- Department of Cell Biology, Harvard Medical School, Boston, MA 02115
| | | | | | - John Rogers
- Thermo Fisher Scientific, Rockford, IL 61101
| | - Edward T Chouchani
- Department of Cell Biology, Harvard Medical School, Boston, MA 02115
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA 02115
| | - Steven P Gygi
- Department of Cell Biology, Harvard Medical School, Boston, MA 02115;
| |
Collapse
|
70
|
Idda ML, McClusky WG, Lodde V, Munk R, Abdelmohsen K, Rossi M, Gorospe M. Survey of senescent cell markers with age in human tissues. Aging (Albany NY) 2020; 12:4052-4066. [PMID: 32160592 PMCID: PMC7093180 DOI: 10.18632/aging.102903] [Citation(s) in RCA: 98] [Impact Index Per Article: 24.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2020] [Accepted: 02/20/2020] [Indexed: 01/10/2023]
Abstract
Cellular senescence, triggered by sublethal damage, is characterized by indefinite growth arrest, altered gene expression patterns, and a senescence-associated secretory phenotype. While the accumulation of senescent cells during aging decreases tissue function and promotes many age-related diseases, at present there is no universal marker to detect senescent cells in tissues. Cyclin-dependent kinase inhibitors 2A (p16/CDKN2A) and 1A (p21/CDKN1A) can identify senescent cells, but few studies have examined the numbers of cells expressing these markers in different organs as a function of age. Here, we investigated systematically p16- and p21-positive cells in tissue arrays designed to include normal organs from persons across a broad spectrum of ages. Increased numbers of p21-positive and p16-positive cells with donor age were found in skin (epidermis), pancreas, and kidney, while p16-expressing cells increased in brain cortex, liver, spleen and intestine (colon), and p21-expressing cells increased in skin (dermis). The numbers of cells expressing p16 or p21 in lung did not change with age, and muscle did not appear to have p21- or p16-positive cells. In summary, different organs display different levels of the senescent proteins p16 and p21 as a function of age across the human life span.
Collapse
Affiliation(s)
- M Laura Idda
- Laboratory of Genetics and Genomics, National Institute on Aging Intramural Research Program, National Institutes of Health, Baltimore, MD 21224, USA.,Istituto di Ricerca Genetica e Biomedica, Consiglio Nazionale delle Ricerche, Sassari, Italy
| | - Waverly G McClusky
- Laboratory of Genetics and Genomics, National Institute on Aging Intramural Research Program, National Institutes of Health, Baltimore, MD 21224, USA
| | - Valeria Lodde
- Laboratory of Genetics and Genomics, National Institute on Aging Intramural Research Program, National Institutes of Health, Baltimore, MD 21224, USA.,Department of Biomedical Sciences, University of Sassari, Sassari, Italy
| | - Rachel Munk
- Laboratory of Genetics and Genomics, National Institute on Aging Intramural Research Program, National Institutes of Health, Baltimore, MD 21224, USA
| | - Kotb Abdelmohsen
- Laboratory of Genetics and Genomics, National Institute on Aging Intramural Research Program, National Institutes of Health, Baltimore, MD 21224, USA
| | - Martina Rossi
- Laboratory of Genetics and Genomics, National Institute on Aging Intramural Research Program, National Institutes of Health, Baltimore, MD 21224, USA
| | - Myriam Gorospe
- Laboratory of Genetics and Genomics, National Institute on Aging Intramural Research Program, National Institutes of Health, Baltimore, MD 21224, USA
| |
Collapse
|
71
|
Schmeer C, Kretz A, Wengerodt D, Stojiljkovic M, Witte OW. Dissecting Aging and Senescence-Current Concepts and Open Lessons. Cells 2019; 8:cells8111446. [PMID: 31731770 PMCID: PMC6912776 DOI: 10.3390/cells8111446] [Citation(s) in RCA: 82] [Impact Index Per Article: 16.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2019] [Revised: 11/12/2019] [Accepted: 11/13/2019] [Indexed: 01/10/2023] Open
Abstract
In contrast to the programmed nature of development, it is still a matter of debate whether aging is an adaptive and regulated process, or merely a consequence arising from a stochastic accumulation of harmful events that culminate in a global state of reduced fitness, risk for disease acquisition, and death. Similarly unanswered are the questions of whether aging is reversible and can be turned into rejuvenation as well as how aging is distinguishable from and influenced by cellular senescence. With the discovery of beneficial aspects of cellular senescence and evidence of senescence being not limited to replicative cellular states, a redefinition of our comprehension of aging and senescence appears scientifically overdue. Here, we provide a factor-based comparison of current knowledge on aging and senescence, which we converge on four suggested concepts, thereby implementing the newly emerging cellular and molecular aspects of geroconversion and amitosenescence, and the signatures of a genetic state termed genosenium. We also address the possibility of an aging-associated secretory phenotype in analogy to the well-characterized senescence-associated secretory phenotype and delineate the impact of epigenetic regulation in aging and senescence. Future advances will elucidate the biological and molecular fingerprints intrinsic to either process.
Collapse
Affiliation(s)
- Christian Schmeer
- Hans-Berger Department of Neurology, Jena University Hospital, 07747 Jena, Thuringia, Germany; (A.K.); (D.W.); (M.S.); (O.W.W.)
- Jena Center for Healthy Ageing, Jena University Hospital, 07747 Jena, Thuringia, Germany
- Correspondence:
| | - Alexandra Kretz
- Hans-Berger Department of Neurology, Jena University Hospital, 07747 Jena, Thuringia, Germany; (A.K.); (D.W.); (M.S.); (O.W.W.)
- Jena Center for Healthy Ageing, Jena University Hospital, 07747 Jena, Thuringia, Germany
| | - Diane Wengerodt
- Hans-Berger Department of Neurology, Jena University Hospital, 07747 Jena, Thuringia, Germany; (A.K.); (D.W.); (M.S.); (O.W.W.)
| | - Milan Stojiljkovic
- Hans-Berger Department of Neurology, Jena University Hospital, 07747 Jena, Thuringia, Germany; (A.K.); (D.W.); (M.S.); (O.W.W.)
- Jena Center for Healthy Ageing, Jena University Hospital, 07747 Jena, Thuringia, Germany
| | - Otto W. Witte
- Hans-Berger Department of Neurology, Jena University Hospital, 07747 Jena, Thuringia, Germany; (A.K.); (D.W.); (M.S.); (O.W.W.)
- Jena Center for Healthy Ageing, Jena University Hospital, 07747 Jena, Thuringia, Germany
| |
Collapse
|
72
|
Del Rey MJ, Valín Á, Usategui A, Ergueta S, Martín E, Municio C, Cañete JD, Blanco FJ, Criado G, Pablos JL. Senescent synovial fibroblasts accumulate prematurely in rheumatoid arthritis tissues and display an enhanced inflammatory phenotype. Immun Ageing 2019; 16:29. [PMID: 31708994 PMCID: PMC6833299 DOI: 10.1186/s12979-019-0169-4] [Citation(s) in RCA: 53] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2019] [Accepted: 10/17/2019] [Indexed: 01/05/2023]
Abstract
BACKGROUND Accumulation of senescent cells has been associated with pro-inflammatory effects with deleterious consequences in different human diseases. The purpose of this study was to analyze cell senescence in human synovial tissues (ST), and its impact on the pro-inflammatory function of synovial fibroblasts (SF). RESULTS The expression of the senescence marker p16INK4a (p16) was analyzed by immunohistochemistry in rheumatoid arthritis (RA), osteoarthritis (OA), and normal ST from variably aged donors. The proportion of p16(+) senescent cells in normal ST from older donors was higher than from younger ones. Although older RA and OA ST showed proportions of senescent cells similar to older normal ST, senescence was increased in younger RA ST compared to age-matched normal ST. The percentage of senescent SA-β-gal(+) SF after 14 days in culture positively correlated with donor's age. Initial exposure to H2O2 or TNFα enhanced SF senescence and increased mRNA expression of IL6, CXCL8, CCL2 and MMP3 and proteins secretion. Senescent SF show a heightened IL6, CXCL8 and MMP3 mRNA and IL-6 and IL-8 protein expression response upon further challenge with TNFα. Treatment of senescent SF with the senolytic drug fenofibrate normalized IL6, CXCL8 and CCL2 mRNA expression. CONCLUSIONS Accumulation of senescent cells in ST increases in normal aging and prematurely in RA patients. Senescence of cultured SF is accelerated upon exposure to TNFα or oxidative stress and may contribute to the pathogenesis of synovitis by increasing the production of pro-inflammatory mediators.
Collapse
Affiliation(s)
- Manuel J. Del Rey
- Grupo de Enfermedades Inflamatorias y Autoinmunes, Instituto de Investigación Hospital 12 de Octubre (i+12), Madrid, Spain
| | - Álvaro Valín
- Grupo de Enfermedades Inflamatorias y Autoinmunes, Instituto de Investigación Hospital 12 de Octubre (i+12), Madrid, Spain
| | - Alicia Usategui
- Grupo de Enfermedades Inflamatorias y Autoinmunes, Instituto de Investigación Hospital 12 de Octubre (i+12), Madrid, Spain
| | - Sandra Ergueta
- Grupo de Enfermedades Inflamatorias y Autoinmunes, Instituto de Investigación Hospital 12 de Octubre (i+12), Madrid, Spain
| | - Eduardo Martín
- Grupo de Enfermedades Inflamatorias y Autoinmunes, Instituto de Investigación Hospital 12 de Octubre (i+12), Madrid, Spain
| | - Cristina Municio
- Grupo de Enfermedades Inflamatorias y Autoinmunes, Instituto de Investigación Hospital 12 de Octubre (i+12), Madrid, Spain
| | - Juan D. Cañete
- Unitat d’Artritis, Servei de Reumatologia, Hospital Clínic de Barcelona and Institut d’Investigacions Biomèdiques August Pí i Sunyer, Barcelona, Spain
| | - Francisco J. Blanco
- Laboratorio de Investigación Osteoarticular y del Envejecimiento, Instituto de Investigación Biomédica de A Coruña, INIBIC, A Coruña, Spain
| | - Gabriel Criado
- Grupo de Enfermedades Inflamatorias y Autoinmunes, Instituto de Investigación Hospital 12 de Octubre (i+12), Madrid, Spain
- Centro de Investigación, Hospital 12 de Octubre, 28041 Madrid, Spain
| | - José L. Pablos
- Grupo de Enfermedades Inflamatorias y Autoinmunes, Instituto de Investigación Hospital 12 de Octubre (i+12), Madrid, Spain
- Servicio de Reumatología, Hospital 12 de Octubre, Universidad Complutense de Madrid, 28041 Madrid, Spain
| |
Collapse
|
73
|
Choi H, Joe S, Nam H. Development of Tissue-Specific Age Predictors Using DNA Methylation Data. Genes (Basel) 2019; 10:genes10110888. [PMID: 31690030 PMCID: PMC6896025 DOI: 10.3390/genes10110888] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2019] [Revised: 11/01/2019] [Accepted: 11/01/2019] [Indexed: 12/17/2022] Open
Abstract
DNA methylation patterns have been shown to change throughout the normal aging process. Several studies have found epigenetic aging markers using age predictors, but these studies only focused on blood-specific or tissue-common methylation patterns. Here, we constructed nine tissue-specific age prediction models using methylation array data from normal samples. The constructed models predict the chronological age with good performance (mean absolute error of 5.11 years on average) and show better performance in the independent test than previous multi-tissue age predictors. We also compared tissue-common and tissue-specific aging markers and found that they had different characteristics. Firstly, the tissue-common group tended to contain more positive aging markers with methylation values that increased during the aging process, whereas the tissue-specific group tended to contain more negative aging markers. Secondly, many of the tissue-common markers were located in Cytosine-phosphate-Guanine (CpG) island regions, whereas the tissue-specific markers were located in CpG shore regions. Lastly, the tissue-common CpG markers tended to be located in more evolutionarily conserved regions. In conclusion, our prediction models identified CpG markers that capture both tissue-common and tissue-specific characteristics during the aging process.
Collapse
Affiliation(s)
- Heeyeon Choi
- School of Electrical Engineering and Computer Science, Gwangju Institute of Science of Technology, Gwangju 61005, Korea.
| | - Soobok Joe
- School of Electrical Engineering and Computer Science, Gwangju Institute of Science of Technology, Gwangju 61005, Korea.
| | - Hojung Nam
- School of Electrical Engineering and Computer Science, Gwangju Institute of Science of Technology, Gwangju 61005, Korea.
| |
Collapse
|
74
|
Bang M, Kim DG, Gonzales EL, Kwon KJ, Shin CY. Etoposide Induces Mitochondrial Dysfunction and Cellular Senescence in Primary Cultured Rat Astrocytes. Biomol Ther (Seoul) 2019; 27:530-539. [PMID: 31646843 PMCID: PMC6824621 DOI: 10.4062/biomolther.2019.151] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2019] [Revised: 09/23/2019] [Accepted: 09/25/2019] [Indexed: 11/05/2022] Open
Abstract
Brain aging is an inevitable process characterized by structural and functional changes and is a major risk factor for neurodegenerative diseases. Most brain aging studies are focused on neurons and less on astrocytes which are the most abundant cells in the brain known to be in charge of various functions including the maintenance of brain physical formation, ion homeostasis, and secretion of various extracellular matrix proteins. Altered mitochondrial dynamics, defective mitophagy or mitochondrial damages are causative factors of mitochondrial dysfunction, which is linked to age-related disorders. Etoposide is an anti-cancer reagent which can induce DNA stress and cellular senescence of cancer cell lines. In this study, we investigated whether etoposide induces senescence and functional alterations in cultured rat astrocytes. Senescence-associated β-galactosidase (SA-β-gal) activity was used as a cellular senescence marker. The results indicated that etoposide-treated astrocytes showed cellular senescence phenotypes including increased SA-β-gal-positive cells number, increased nuclear size and increased senescence-associated secretory phenotypes (SASP) such as IL-6. We also observed a decreased expression of cell cycle markers, including Phospho- Histone H3/Histone H3 and CDK2, and dysregulation of cellular functions based on wound-healing, neuronal protection, and phagocytosis assays. Finally, mitochondrial dysfunction was noted through the determination of mitochondrial membrane potential using tetramethylrhodamine methyl ester (TMRM) and the measurement of mitochondrial oxygen consumption rate (OCR). These data suggest that etoposide can induce cellular senescence and mitochondrial dysfunction in astrocytes which may have implications in brain aging and neurodegenerative conditions.
Collapse
Affiliation(s)
- Minji Bang
- Department of Neuroscience, School of Medicine and Center for Neuroscience Research, Konkuk University, Seoul 05029, Republic of Korea
| | - Do Gyeong Kim
- Department of Neuroscience, School of Medicine and Center for Neuroscience Research, Konkuk University, Seoul 05029, Republic of Korea
| | - Edson Luck Gonzales
- Department of Neuroscience, School of Medicine and Center for Neuroscience Research, Konkuk University, Seoul 05029, Republic of Korea
| | - Kyoung Ja Kwon
- Department of Neuroscience, School of Medicine and Center for Neuroscience Research, Konkuk University, Seoul 05029, Republic of Korea
| | - Chan Young Shin
- Department of Neuroscience, School of Medicine and Center for Neuroscience Research, Konkuk University, Seoul 05029, Republic of Korea
| |
Collapse
|
75
|
Santos-Otte P, Leysen H, van Gastel J, Hendrickx JO, Martin B, Maudsley S. G Protein-Coupled Receptor Systems and Their Role in Cellular Senescence. Comput Struct Biotechnol J 2019; 17:1265-1277. [PMID: 31921393 PMCID: PMC6944711 DOI: 10.1016/j.csbj.2019.08.005] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2019] [Revised: 08/20/2019] [Accepted: 08/21/2019] [Indexed: 12/17/2022] Open
Abstract
Aging is a complex biological process that is inevitable for nearly all organisms. Aging is the strongest risk factor for development of multiple neurodegenerative disorders, cancer and cardiovascular disorders. Age-related disease conditions are mainly caused by the progressive degradation of the integrity of communication systems within and between organs. This is in part mediated by, i) decreased efficiency of receptor signaling systems and ii) an increasing inability to cope with stress leading to apoptosis and cellular senescence. Cellular senescence is a natural process during embryonic development, more recently it has been shown to be also involved in the development of aging disorders and is now considered one of the major hallmarks of aging. G-protein-coupled receptors (GPCRs) comprise a superfamily of integral membrane receptors that are responsible for cell signaling events involved in nearly every physiological process. Recent advances in the molecular understanding of GPCR signaling complexity have expanded their therapeutic capacity tremendously. Emerging data now suggests the involvement of GPCRs and their associated proteins in the development of cellular senescence. With the proven efficacy of therapeutic GPCR targeting, it is reasonable to now consider GPCRs as potential platforms to control cellular senescence and the consequently, age-related disorders.
Collapse
Key Words
- ADP-ribosylation factor GTPase-activating protein, (Arf-GAP)
- AT1R blockers, (ARB)
- Aging
- Angiotensin II, (Ang II)
- Ataxia telangiectasia mutated, (ATM)
- Cellular senescence
- G protein-coupled receptor kinase interacting protein 2 (GIT2)
- G protein-coupled receptor kinase interacting protein 2, (GIT2)
- G protein-coupled receptor kinase, (GRK)
- G protein-coupled receptors (GPCRs)
- G protein-coupled receptors, (GPCRs)
- Hutchinson–Gilford progeria syndrome, (HGPS)
- Lysophosphatidic acid, (LPA)
- Regulator of G-protein signaling, (RGS)
- Relaxin family receptor 3, (RXFP3)
- active state, (R*)
- angiotensin type 1 receptor, (AT1R)
- angiotensin type 2 receptor, (AT2R)
- beta2-adrenergic receptor, (β2AR)
- cyclin-dependent kinase 2, (CDK2)
- cyclin-dependent kinase inhibitor 1, (cdkn1A/p21)
- endothelial cell differentiation gene, (Edg)
- inactive state, (R)
- latent semantic indexing, (LSI)
- mitogen-activated protein kinase, (MAPK)
- nuclear factor kappa-light-chain-enhancer of activated B cells, (NF- κβ)
- protein kinases, (PK)
- purinergic receptors family, (P2Y)
- renin-angiotensin system, (RAS)
- retinoblastoma, (RB)
- senescence associated secretory phenotype, (SASP)
- stress-induced premature senescence, (SIPS)
- transcription factor E2F3, (E2F3)
- transmembrane, (TM)
- tumor suppressor gene PTEN, (PTEN)
- tumor suppressor protein 53, (p53)
- vascular smooth muscle cells, (VSMC)
- β-Arrestin
Collapse
Affiliation(s)
- Paula Santos-Otte
- Center for Molecular and Cellular Bioengineering (CMCB), Technische Universität Dresden, 01062 Dresden, Germany
| | - Hanne Leysen
- Receptor Biology Lab, University of Antwerp, 2610 Antwerp, Belgium
- Department of Biomedical Sciences, University of Antwerp, 2610 Antwerp, Belgium
| | - Jaana van Gastel
- Receptor Biology Lab, University of Antwerp, 2610 Antwerp, Belgium
- Department of Biomedical Sciences, University of Antwerp, 2610 Antwerp, Belgium
| | - Jhana O. Hendrickx
- Receptor Biology Lab, University of Antwerp, 2610 Antwerp, Belgium
- Department of Biomedical Sciences, University of Antwerp, 2610 Antwerp, Belgium
| | - Bronwen Martin
- Receptor Biology Lab, University of Antwerp, 2610 Antwerp, Belgium
| | - Stuart Maudsley
- Receptor Biology Lab, University of Antwerp, 2610 Antwerp, Belgium
- Department of Biomedical Sciences, University of Antwerp, 2610 Antwerp, Belgium
| |
Collapse
|
76
|
Hunt NJ, Kang SWS, Lockwood GP, Le Couteur DG, Cogger VC. Hallmarks of Aging in the Liver. Comput Struct Biotechnol J 2019; 17:1151-1161. [PMID: 31462971 PMCID: PMC6709368 DOI: 10.1016/j.csbj.2019.07.021] [Citation(s) in RCA: 161] [Impact Index Per Article: 32.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2019] [Revised: 07/30/2019] [Accepted: 07/31/2019] [Indexed: 02/07/2023] Open
Abstract
While the liver demonstrates remarkable resilience during aging, there is growing evidence that it undergoes all the cellular hallmarks of aging, which increases the risk of liver and systemic disease. The aging process in the liver is driven by alterations of the genome and epigenome that contribute to dysregulation of mitochondrial function and nutrient sensing pathways, leading to cellular senescence and low-grade inflammation. These changes promote multiple phenotypic changes in all liver cells (hepatocytes, liver sinusoidal endothelial, hepatic stellate and Küpffer cells) and impairment of hepatic function. In particular, age-related changes in the liver sinusoidal endothelial cells are a significant but under-recognized risk factor for the development of age-related cardiometabolic disease. Liver aging is driven by transcription and metabolic epigenome alterations. This leads to cellular senescence and low-grade inflammation. Hepatocyte, sinusoidal endothelial, stellate and Küpffer cells undergoes the hallmarks of aging. Each cell type demonstrates phenotypical cellular changes with age.
Collapse
Key Words
- AMPK, 5′ adenosine monophosphate-activated protein kinase
- CR, caloric restriction
- Endothelial
- FOXO, forkhead box O
- Genetic
- HSC, hepatic stellate cell
- Hepatocyte
- IGF-1, insulin like growth factor 1
- IL-6, interleukin 6
- IL-8, interleukin 8
- KC, Küpffer cell
- LSEC, liver sinusoidal endothelial cell
- Mitochondrial dysfunction
- NAD, nicotinamide adenine dinucleotide
- NAFLD, non-alcoholic fatty liver disease
- NO, nitric oxide
- Nutrient sensing pathways
- PDGF, platelet derived growth factor
- PGC-1α, peroxisome proliferator-activated receptor gamma coactivator 1-α
- ROS, reactive oxygen species
- SIRT1, sirtuin 1
- Senescence
- TNFα, tumor necrosis factor alpha
- VEGF, vascular endothelial growth factor
- mTOR, mammalian target of rapamycin
- miR, microRNA
- αSMA, alpha smooth muscle actin
Collapse
Affiliation(s)
- Nicholas J Hunt
- ANZAC Research Institute, Aging and Alzheimer's Institute, Centre for Education and Research on Ageing, Concord Repatriation General Hospital, Concord, NSW, Australia.,The University of Sydney, Concord Clinical School, Sydney Medical School, Sydney, NSW, Australia.,The University of Sydney, Nutrition Ecology, Charles Perkins Centre, Sydney, NSW, Australia
| | - Sun Woo Sophie Kang
- ANZAC Research Institute, Aging and Alzheimer's Institute, Centre for Education and Research on Ageing, Concord Repatriation General Hospital, Concord, NSW, Australia.,The University of Sydney, Nutrition Ecology, Charles Perkins Centre, Sydney, NSW, Australia
| | - Glen P Lockwood
- ANZAC Research Institute, Aging and Alzheimer's Institute, Centre for Education and Research on Ageing, Concord Repatriation General Hospital, Concord, NSW, Australia.,The University of Sydney, Nutrition Ecology, Charles Perkins Centre, Sydney, NSW, Australia
| | - David G Le Couteur
- ANZAC Research Institute, Aging and Alzheimer's Institute, Centre for Education and Research on Ageing, Concord Repatriation General Hospital, Concord, NSW, Australia.,The University of Sydney, Concord Clinical School, Sydney Medical School, Sydney, NSW, Australia.,The University of Sydney, Nutrition Ecology, Charles Perkins Centre, Sydney, NSW, Australia
| | - Victoria C Cogger
- ANZAC Research Institute, Aging and Alzheimer's Institute, Centre for Education and Research on Ageing, Concord Repatriation General Hospital, Concord, NSW, Australia.,The University of Sydney, Concord Clinical School, Sydney Medical School, Sydney, NSW, Australia.,The University of Sydney, Nutrition Ecology, Charles Perkins Centre, Sydney, NSW, Australia
| |
Collapse
|
77
|
Terlecki-Zaniewicz L, Lämmermann I, Latreille J, Bobbili MR, Pils V, Schosserer M, Weinmüllner R, Dellago H, Skalicky S, Pum D, Almaraz JCH, Scheideler M, Morizot F, Hackl M, Gruber F, Grillari J. Small extracellular vesicles and their miRNA cargo are anti-apoptotic members of the senescence-associated secretory phenotype. Aging (Albany NY) 2019; 10:1103-1132. [PMID: 29779019 PMCID: PMC5990398 DOI: 10.18632/aging.101452] [Citation(s) in RCA: 142] [Impact Index Per Article: 28.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2018] [Accepted: 05/10/2018] [Indexed: 12/15/2022]
Abstract
Loss of functionality during aging of cells and organisms is caused and accompanied by altered cell-to-cell communication and signalling. One factor thereby is the chronic accumulation of senescent cells and the concomitant senescence-associated secretory phenotype (SASP) that contributes to microenvironment remodelling and a pro-inflammatory status. While protein based SASP factors have been well characterized, little is known about small extracellular vesicles (sEVs) and their miRNA cargo. Therefore, we analysed secretion of sEVs from senescent human dermal fibroblasts and catalogued the therein contained miRNAs. We observed a four-fold increase of sEVs, with a concomitant increase of >80% of all cargo miRNAs. The most abundantly secreted miRNAs were predicted to collectively target mRNAs of pro-apoptotic proteins, and indeed, senescent cell derived sEVs exerted anti-apoptotic activity. In addition, we identified senescence-specific differences in miRNA composition of sEVs, with an increase of miR-23a-5p and miR-137 and a decrease of miR-625-3p, miR-766-3p, miR-199b-5p, miR-381-3p, miR-17-3p. By correlating intracellular and sEV-miRNAs, we identified miRNAs selectively retained in senescent cells (miR-21-3p and miR-17-3p) or packaged specifically into senescent cell derived sEVs (miR-15b-5p and miR-30a-3p). Therefore, we suggest sEVs and their miRNA cargo to be novel, members of the SASP that are selectively secreted or retained in cellular senescence.
Collapse
|
78
|
Ghanam AR, Cao J, Ouyang X, Song X. New Insights into Chronological Mobility of Retrotransposons In Vivo. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2019; 2019:2818415. [PMID: 31346359 PMCID: PMC6617872 DOI: 10.1155/2019/2818415] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/18/2019] [Accepted: 06/03/2019] [Indexed: 12/17/2022]
Abstract
Tissue aging is the gradual decline of physiological homeostasis accompanied with accumulation of senescent cells, decreased clearance of unwanted biological compounds, and depletion of stem cells. Senescent cells were cell cycle arrested in response to various stimuli and identified using distinct phenotypes and changes in gene expression. Senescent cells that accumulate with aging can compromise normal tissue function and inhibit or stop repair and regeneration. Selective removal of senescent cells can slow the aging process and inhibits age-associated diseases leading to extended lifespans in mice and thus provides a possibility for developing antiaging therapy. To monitor the appearance of senescent cells in vivo and target them, a clearer understanding of senescent cell expression markers is needed. We investigated the age-associated expression of three molecular hallmarks of aging: SA-β-gal, P16INK4a, and retrotransposable elements (RTEs), in different mouse tissues during chronological aging. Our data showed that the expression of these markers is variable with aging in the different tissues. P16INK4a showed consistent increases with age in most tissues, while expression of RTEs was variable among different tissues examined. These data suggest that biological changes occurring with physiological aging may be useful in choosing the appropriate timing of therapeutic interventions to slow the aging process or keep more susceptible organs healthier in the aging process.
Collapse
Affiliation(s)
- Amr. R. Ghanam
- Hefei National Laboratory for Physical Sciences at the Microscale, CAS Key Laboratory of Brain Function and Disease, Neurodegenerative Disorder Research Center, School of Life Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui 230026, China
| | - Jun Cao
- Hefei National Laboratory for Physical Sciences at the Microscale, CAS Key Laboratory of Brain Function and Disease, Neurodegenerative Disorder Research Center, School of Life Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui 230026, China
| | - Xuan Ouyang
- Hefei National Laboratory for Physical Sciences at the Microscale, CAS Key Laboratory of Brain Function and Disease, Neurodegenerative Disorder Research Center, School of Life Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui 230026, China
| | - Xiaoyuan Song
- Hefei National Laboratory for Physical Sciences at the Microscale, CAS Key Laboratory of Brain Function and Disease, Neurodegenerative Disorder Research Center, School of Life Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui 230026, China
| |
Collapse
|
79
|
Santelli A, Sun IO, Eirin A, Abumoawad AM, Woollard JR, Lerman A, Textor S, Puranik AS, Lerman LO. Senescent Kidney Cells in Hypertensive Patients Release Urinary Extracellular Vesicles. J Am Heart Assoc 2019; 8:e012584. [PMID: 31433703 PMCID: PMC6585370 DOI: 10.1161/jaha.119.012584] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/14/2019] [Accepted: 05/03/2019] [Indexed: 12/13/2022]
Abstract
Background Hypertension may be associated with renal cellular injury. Cells in distress release extracellular vesicles (EVs), and their numbers in urine may reflect renal injury. Cellular senescence, an irreversible growth arrest in response to a noxious milieu, is characterized by release of proinflammatory cytokines. We hypothesized that EVs released by senescent nephron cells can be identified in urine of patients with hypertension. Methods and Results We recruited patients with essential hypertension (EH) or renovascular hypertension and healthy volunteers (n=14 each). Renal oxygenation was assessed using magnetic resonance imaging and blood samples collected from both renal veins for cytokine-level measurements. EVs isolated from urine samples were characterized by imaging flow cytometry based on specific markers, including p16 (senescence marker), calyxin (podocytes), urate transporter 1 (proximal tubules), uromodulin (ascending limb of Henle's loop), and prominin-2 (distal tubules). Overall percentage of urinary p16+ EVs was elevated in EH and renovascular hypertension patients compared with healthy volunteers and correlated inversely with renal function and directly with renal vein cytokine levels. Urinary levels of p16+/urate transporter 1+ were elevated in all hypertensive subjects compared with healthy volunteers, whereas p16+/prominin-2+ levels were elevated only in EH versus healthy volunteers and p16+/uromodulin+ in renovascular hypertension versus EH. Conclusions Levels of p16+ EVs are elevated in urine of hypertensive patients and may reflect increased proximal tubular cellular senescence. In EH, EVs originate also from distal tubules and in renovascular hypertension from Henle's loop. Hence, urinary EVs levels may be useful to identify intrarenal sites of cellular senescence.
Collapse
Affiliation(s)
- Adrian Santelli
- Division of Nephrology and HypertensionMayo ClinicRochesterMN
- Department of PhysiopathologyHospital de ClinicasMontevideoUruguay
| | - In O. Sun
- Division of Nephrology and HypertensionMayo ClinicRochesterMN
| | - Alfonso Eirin
- Division of Nephrology and HypertensionMayo ClinicRochesterMN
| | | | | | - Amir Lerman
- Department of Cardiovascular DiseasesMayo ClinicRochesterMN
| | | | | | | |
Collapse
|
80
|
Bechshøft CJL, Jensen SM, Schjerling P, Andersen JL, Svensson RB, Eriksen CS, Mkumbuzi NS, Kjaer M, Mackey AL. Age and prior exercise in vivo determine the subsequent in vitro molecular profile of myoblasts and nonmyogenic cells derived from human skeletal muscle. Am J Physiol Cell Physiol 2019; 316:C898-C912. [PMID: 30917034 DOI: 10.1152/ajpcell.00049.2019] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
The decline in skeletal muscle regenerative capacity with age is partly attributed to muscle stem cell (satellite cell) dysfunction. Recent evidence has pointed to a strong interaction between myoblasts and fibroblasts, but the influence of age on this interaction is unknown. Additionally, while the native tissue environment is known to determine the properties of myogenic cells in vitro, how the aging process alters this cell memory has not been established at the molecular level. We recruited 12 young and 12 elderly women, who performed a single bout of heavy resistance exercise with the knee extensor muscles of one leg. Five days later, muscle biopsies were collected from both legs, and myogenic cells and nonmyogenic cells were isolated for in vitro experiments with mixed or separated cells and analyzed by immunostaining and RT-PCR. A lower myogenic fusion index was detected in the cells from the old versus young women, in association with differences in gene expression levels of key myogenic regulatory factors and senescence, which were further altered by performing exercise before tissue sampling. Coculture with nonmyogenic cells from the elderly led to a higher myogenic differentiation index compared with nonmyogenic cells from the young. These findings show that the in vitro phenotype and molecular profile of human skeletal muscle myoblasts and fibroblasts is determined by the age and exercise state of the original in vivo environment and help explain how exercise can enhance muscle stem cell function in old age.
Collapse
Affiliation(s)
- Cecilie J L Bechshøft
- Institute of Sports Medicine Copenhagen, Department of Orthopedic Surgery M, Bispebjerg Hospital and Center for Healthy Aging, Faculty of Health and Medical Sciences, University of Copenhagen , Copenhagen , Denmark
| | - Simon M Jensen
- Institute of Sports Medicine Copenhagen, Department of Orthopedic Surgery M, Bispebjerg Hospital and Center for Healthy Aging, Faculty of Health and Medical Sciences, University of Copenhagen , Copenhagen , Denmark
| | - Peter Schjerling
- Institute of Sports Medicine Copenhagen, Department of Orthopedic Surgery M, Bispebjerg Hospital and Center for Healthy Aging, Faculty of Health and Medical Sciences, University of Copenhagen , Copenhagen , Denmark
| | - Jesper L Andersen
- Institute of Sports Medicine Copenhagen, Department of Orthopedic Surgery M, Bispebjerg Hospital and Center for Healthy Aging, Faculty of Health and Medical Sciences, University of Copenhagen , Copenhagen , Denmark
| | - Rene B Svensson
- Institute of Sports Medicine Copenhagen, Department of Orthopedic Surgery M, Bispebjerg Hospital and Center for Healthy Aging, Faculty of Health and Medical Sciences, University of Copenhagen , Copenhagen , Denmark
| | - Christian S Eriksen
- Institute of Sports Medicine Copenhagen, Department of Orthopedic Surgery M, Bispebjerg Hospital and Center for Healthy Aging, Faculty of Health and Medical Sciences, University of Copenhagen , Copenhagen , Denmark
| | - Nonhlanhla S Mkumbuzi
- Division of Exercise Science and Sports Medicine, Department of Human Biology, University of Cape Town, Newlands, South Africa
| | - Michael Kjaer
- Institute of Sports Medicine Copenhagen, Department of Orthopedic Surgery M, Bispebjerg Hospital and Center for Healthy Aging, Faculty of Health and Medical Sciences, University of Copenhagen , Copenhagen , Denmark
| | - Abigail L Mackey
- Institute of Sports Medicine Copenhagen, Department of Orthopedic Surgery M, Bispebjerg Hospital and Center for Healthy Aging, Faculty of Health and Medical Sciences, University of Copenhagen , Copenhagen , Denmark.,Center for Healthy Aging, Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen , Copenhagen , Denmark
| |
Collapse
|
81
|
Strickland M, Yacoubi-Loueslati B, Bouhaouala-Zahar B, Pender SLF, Larbi A. Relationships Between Ion Channels, Mitochondrial Functions and Inflammation in Human Aging. Front Physiol 2019; 10:158. [PMID: 30881309 PMCID: PMC6405477 DOI: 10.3389/fphys.2019.00158] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2018] [Accepted: 02/08/2019] [Indexed: 12/19/2022] Open
Abstract
Aging is often associated with a loss of function. We believe aging to be more an adaptation to the various, and often continuous, stressors encountered during life in order to maintain overall functionality of the systems. The maladaptation of a system during aging may increase the susceptibility to diseases. There are basic cellular functions that may influence and/or are influenced by aging. Mitochondrial function is amongst these. Their presence in almost all cell types makes of these valuable targets for interventions to slow down or even reserve signs of aging. In this review, the role of mitochondria and essential physiological regulators of mitochondria and cellular functions, ion channels, will be discussed in the context of human aging. The origins of inflamm-aging, associated with poor clinical outcomes, will be linked to mitochondria and ion channel biology.
Collapse
Affiliation(s)
- Marie Strickland
- Singapore Immunology Network, Agency for Science Technology and Research, Singapore, Singapore
- Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, United Kingdom
| | - Besma Yacoubi-Loueslati
- Laboratory of Mycology, Pathologies and Biomarkers, Department of Biology, Faculty of Sciences, University Tunis El Manar, Tunis, Tunisia
| | - Balkiss Bouhaouala-Zahar
- Laboratory of Venoms and Therapeutic Molecules, Institut Pasteur de Tunis, University Tunis El Manar, Tunis, Tunisia
- Medical School of Tunis, University Tunis El Manar, Tunis, Tunisia
| | - Sylvia L. F. Pender
- Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, United Kingdom
- Chinese University of Hong Kong – University of Southampton Joint Lab for Stem Cell and Regenerative Medicine, Hong Kong, China
| | - Anis Larbi
- Singapore Immunology Network, Agency for Science Technology and Research, Singapore, Singapore
- Department of Biology, Faculty of Sciences, University Tunis El Manar, Tunis, Tunisia
- Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Geriatrics Division, Department of Medicine, Research Center on Aging, University of Sherbrooke, Sherbrooke, QC, Canada
| |
Collapse
|
82
|
Chen L, He PL, Yang J, Yang YF, Wang K, Amend B, Stenzl A, Zhang YM, Wang ZL, Xing SS, Luo X. NLRP3/IL1β inflammasome associated with the aging bladder triggers bladder dysfunction in female rats. Mol Med Rep 2019; 19:2960-2968. [PMID: 30720125 PMCID: PMC6423574 DOI: 10.3892/mmr.2019.9919] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2018] [Accepted: 01/10/2019] [Indexed: 12/15/2022] Open
Abstract
Bladder dysfunction is associated with fibrosis-mediated aging, but the corresponding mechanism remains to be elucidated. Activation of the NACHT, LRR and PYD domains-containing protein 3 (NLRP3) inflammasome is related to chronic diseases associated with aging, including organ fibrosis. The present study aimed to explore the role of NLRP3/interleukin 1β in aging-associated bladder dysfunction. Female Sprague-Dawley rats were divided into the following two groups (n=10 rats/group): 2-month-old group (young group) and 24-month-old group (old group). Urodynamics were performed to assess the bladder function of the rats. The histological alterations were identified using Masson's trichrome staining. The protein expression of the NLRP3 inflammasome and NAD-dependent protein deacetylase sirtuin-3, mitochondrial (SIRT3) were detected by western blot analysis, and immunohistochemistry was used to examine a senescence marker (p21) and the NLRP3 inflammasome in the bladder. The localization of the key molecule Caspase1 was determined using immunofluorescence. The voiding time was longer in the old group compared with the young group. The expression levels of SIRT3 were reduced in the bladders of the old group, while those of the NLRP3 inflammasome and the senescence marker were significantly higher in the bladders of the old group compared with the young group. Increased collagen deposition leads to chronic bladder fibrosis with increased NLRP3. In the histological examination, the bladders of the old group displayed increased collagen deposition, urothelial thinning and detrusor shrinkage compared with the young group. Tissue fibrosis and urothelial alterations are the principal causes of bladder dysfunction during aging. Downregulated SIRT3 and upregulated expression of the NLRP3 inflammasome are involved in the degradation of aging bladders. Inflamm-aging is a novel mechanism underlying bladder dysfunction.
Collapse
Affiliation(s)
- Lin Chen
- Department of Urology, Affiliated Hospital of Chengdu University, Chengdu, Sichuan 610000, P.R. China
| | - Ping-Lin He
- Department of Urology, Affiliated Hospital of Chengdu University, Chengdu, Sichuan 610000, P.R. China
| | - Jin Yang
- Department of Urology, Affiliated Hospital of Chengdu University, Chengdu, Sichuan 610000, P.R. China
| | - Ya-Fei Yang
- Department of Urology, Affiliated Hospital of Chengdu University, Chengdu, Sichuan 610000, P.R. China
| | - Kai Wang
- Department of Urology, Affiliated Hospital of Chengdu University, Chengdu, Sichuan 610000, P.R. China
| | - Bastian Amend
- Department of Urology, University of Tübingen, D-72074 Tübingen, Germany
| | - Arnulf Stenzl
- Department of Urology, University of Tübingen, D-72074 Tübingen, Germany
| | - Ya-Mei Zhang
- Central Laboratory, Affiliated Hospital of Chengdu University, Chengdu, Sichuan 610000, P.R. China
| | - Zi-Li Wang
- Department of Urology, Affiliated Hospital of Chengdu University, Chengdu, Sichuan 610000, P.R. China
| | - Sha-Sha Xing
- Central Laboratory, Affiliated Hospital of Chengdu University, Chengdu, Sichuan 610000, P.R. China
| | - Xu Luo
- Department of Urology, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563003, P.R. China
| |
Collapse
|
83
|
Burla R, La Torre M, Zanetti G, Bastianelli A, Merigliano C, Del Giudice S, Vercelli A, Di Cunto F, Boido M, Vernì F, Saggio I. p53-Sensitive Epileptic Behavior and Inflammation in Ft1 Hypomorphic Mice. Front Genet 2018; 9:581. [PMID: 30546381 PMCID: PMC6278696 DOI: 10.3389/fgene.2018.00581] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2018] [Accepted: 11/08/2018] [Indexed: 11/13/2022] Open
Abstract
Epilepsy is a complex clinical condition characterized by repeated spontaneous seizures. Seizures have been linked to multiple drivers including DNA damage accumulation. Investigation of epilepsy physiopathology in humans imposes ethical and practical limitations, for this reason model systems are mostly preferred. Among animal models, mouse mutants are particularly valuable since they allow conjoint behavioral, organismal, and genetic analyses. Along with this, since aging has been associated with higher frequency of seizures, prematurely aging mice, simulating human progeroid diseases, offer a further useful modeling element as they recapitulate aging over a short time-window. Here we report on a mouse mutant with progeroid traits that displays repeated spontaneous seizures. Mutant mice were produced by reducing the expression of the gene Ft1 (AKTIP in humans). In vitro, AKTIP/Ft1 depletion causes telomere aberrations, DNA damage, and cell senescence. AKTIP/Ft1 interacts with lamins, which control nuclear architecture and DNA function. Premature aging defects of Ft1 mutant mice include skeletal alterations and lipodystrophy. The epileptic behavior of Ft1 mutant animals was age and sex linked. Seizures were observed in 18 mutant mice (23.6% of aged ≥ 21 weeks), at an average frequency of 2.33 events/mouse. Time distribution of seizures indicated non-random enrichment of seizures over the follow-up period, with 75% of seizures happening in consecutive weeks. The analysis of epileptic brains did not reveal overt brain morphological alterations or severe neurodegeneration, however, Ft1 reduction induced expression of the inflammatory markers IL-6 and TGF-β. Importantly, Ft1 mutant mice with concomitant genetic reduction of the guardian of the genome, p53, showed no seizures or inflammatory marker activation, implicating the DNA damage response into these phenotypes. This work adds insights into the connection among DNA damage, brain function, and aging. In addition, it further underscores the importance of model organisms for studying specific phenotypes, along with permitting the analysis of genetic interactions at the organismal level.
Collapse
Affiliation(s)
- Romina Burla
- Department of Biology and Biotechnology, Sapienza University of Rome, Rome, Italy
| | - Mattia La Torre
- Department of Biology and Biotechnology, Sapienza University of Rome, Rome, Italy
| | - Giorgia Zanetti
- Department of Biology and Biotechnology, Sapienza University of Rome, Rome, Italy
| | - Alex Bastianelli
- Department of Biology and Biotechnology, Sapienza University of Rome, Rome, Italy
| | - Chiara Merigliano
- Department of Biology and Biotechnology, Sapienza University of Rome, Rome, Italy.,Nanyang Technological University, Singapore, Singapore
| | - Simona Del Giudice
- Department of Biology and Biotechnology, Sapienza University of Rome, Rome, Italy
| | - Alessandro Vercelli
- Neuroscience Institute Cavalieri Ottolenghi, Torino, Italy.,Department of Neuroscience, University of Torino, Piedmont, Italy
| | - Ferdinando Di Cunto
- Neuroscience Institute Cavalieri Ottolenghi, Torino, Italy.,Department of Neuroscience, University of Torino, Piedmont, Italy
| | - Marina Boido
- Neuroscience Institute Cavalieri Ottolenghi, Torino, Italy.,Department of Neuroscience, University of Torino, Piedmont, Italy
| | - Fiammetta Vernì
- Department of Biology and Biotechnology, Sapienza University of Rome, Rome, Italy
| | - Isabella Saggio
- Department of Biology and Biotechnology, Sapienza University of Rome, Rome, Italy.,Nanyang Technological University, Singapore, Singapore
| |
Collapse
|