1
|
Lavarti R, Alvarez-Diaz T, Marti K, Kar P, Raju RP. The context-dependent effect of cellular senescence: From embryogenesis and wound healing to aging. Ageing Res Rev 2025; 109:102760. [PMID: 40318767 DOI: 10.1016/j.arr.2025.102760] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2025] [Revised: 04/20/2025] [Accepted: 04/26/2025] [Indexed: 05/07/2025]
Abstract
Aging is characterized by a steady loss of physiological integrity, leading to impaired function and increased vulnerability to death. Cell senescence is a biological process that progresses with aging and is believed to be a key driver of age-related diseases. Senescence, a hallmark of aging, also demonstrates its beneficial physiological aspects as an anti-cancer, pro-regenerative, homeostatic, and developmental mechanism. A transitory response in which the senescent cells are quickly formed and cleared may promote tissue regeneration and organismal fitness. At the same time, senescence-related secretory phenotypes associated with extended senescence can have devastating effects. The fact that the interaction between senescent cells and their surroundings is very context-dependent may also help to explain this seemingly opposing pleiotropic function. Further, mitochondrial dysfunction is an often-unappreciated hallmark of cellular senescence and figures prominently in multiple feedback loops that induce and maintain the senescent phenotype. This review summarizes the mechanism of cellular senescence and the significance of acute senescence. We concisely introduced the context-dependent role of senescent cells and SASP, aspects of mitochondrial biology altered in the senescent cells, and their impact on the senescent phenotype. Finally, we conclude with recent therapeutic advancements targeting cellular senescence, focusing on acute injuries and age-associated diseases. Collectively, these insights provide a future roadmap for the role of senescence in organismal fitness and life span extension.
Collapse
Affiliation(s)
- Rupa Lavarti
- Department of Pharmacology and Toxicology, Medical College of Georgia, Augusta University, Augusta, GA, United States
| | - Tatiana Alvarez-Diaz
- Department of Pharmacology and Toxicology, Medical College of Georgia, Augusta University, Augusta, GA, United States
| | - Kyarangelie Marti
- Department of Pharmacology and Toxicology, Medical College of Georgia, Augusta University, Augusta, GA, United States
| | - Parmita Kar
- Department of Pharmacology and Toxicology, Medical College of Georgia, Augusta University, Augusta, GA, United States
| | - Raghavan Pillai Raju
- Department of Pharmacology and Toxicology, Medical College of Georgia, Augusta University, Augusta, GA, United States; Charlie Norwood VA Medical Center, Augusta, GA, United States.
| |
Collapse
|
2
|
Rana KS, Marwah MK, Raja FNS, Dias I, Hindalekar YS, Al Tahan MA, Brown JE, Bellary S. The influence of senescent associated secretory phenotype on glucose homeostasis in C2C12 muscle cells: insights into potential p38 inhibitor interventions. J Recept Signal Transduct Res 2025; 45:118-127. [PMID: 40051308 DOI: 10.1080/10799893.2025.2475441] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2024] [Revised: 02/26/2025] [Accepted: 02/28/2025] [Indexed: 04/02/2025]
Abstract
Increased accumulation of senescent cells with aging is associated with reduced ability of insulin-target tissues to utilize glucose, resulting in increased insulin resistance and glucotoxicity. We investigated the role of the senescent-associated secretory phenotype (SASP) within C2C12, skeletal muscle cells on glucose homeostasis and if such effects could be reduced by blocking pro-inflammatory pathways. C2C12 myotubes were treated with 40% conditioned media from senescent fibroblasts. Indirect glucose uptake and glycogen content were measured. The effect of SASP on the generation of reactive oxygen species [1] and mitochondrial function was also measured. The experiments above were repeated with a p38 inhibitor. 40% SASP treatment significantly decreased glucose utilization and glycogen storage within myotubes (p < 0.0001). 40% SASP was successful in inducing oxidative stress and increased mitochondrial density, whilst reducing membrane potential following 48 h of incubation (p < 0.0001) and blocking NF-κβ, restored glucose utilization (p < 0.01) despite the presence of SASP. Co-incubation of 40% SASP with an NF-κβ inhibitor eliminates excessive ROS production and restores mitochondrial activity to levels comparable to control treatment (p < 0.0001). This study shows changes in glucose homeostasis in senescent cells is mediated through SASP, and interventions aimed at mitigating pro-inflammatory pathways can potentially improve insulin resistance.
Collapse
Affiliation(s)
- Karan S Rana
- School of Biosciences, College of Health and Life, Aston University, Birmingham, UK
| | - Mandeep K Marwah
- Aston Medical School, College of Health and Life Sciences, Aston University, Birmingham, UK
| | - Farah N S Raja
- School of Engineering and Physical Science, Aston University, Birmingham, UK
| | - Irundika Dias
- School of Engineering and Physical Science, Aston University, Birmingham, UK
| | | | - Mohamad Anas Al Tahan
- Aston Medical School, College of Health and Life Sciences, Aston University, Birmingham, UK
| | - James E Brown
- School of Biosciences, College of Health and Life, Aston University, Birmingham, UK
| | - Srikanth Bellary
- School of Biosciences, College of Health and Life, Aston University, Birmingham, UK
| |
Collapse
|
3
|
Calubag MF, Ademi I, Grunow I, Breuer L, Babygirija R, Lialios P, Le S, Minton D, Sonsalla MM, Illiano J, Knopf BA, Xiao F, Konopka AR, Harris DA, Lamming DW. Tissue-specific effects of dietary protein on cellular senescence are mediated by branched-chain amino acids. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.13.632607. [PMID: 39868338 PMCID: PMC11761368 DOI: 10.1101/2025.01.13.632607] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 01/28/2025]
Abstract
Dietary protein is a key regulator of healthy aging in both mice and humans. In mice, reducing dietary levels of the branched-chain amino acids (BCAAs) recapitulates many of the benefits of a low protein diet; BCAA-restricted diets extend lifespan, reduce frailty, and improve metabolic health, while BCAA supplementation shortens lifespan, promotes obesity, and impairs glycemic control. Recently, high protein diets have been shown to promote cellular senescence, a hallmark of aging implicated in many age-related diseases, in the liver of mice. Here, we test the hypothesis that the effects of high protein diets on metabolic health and on cell senescence are mediated by BCAAs. We find that reducing dietary levels of BCAAs protects male and female mice from the negative metabolic consequences of both normal and high protein diets. Further, we identify tissue-specific effects of BCAAs on cellular senescence, with restriction of all three BCAAs - but not individual BCAAs - protecting from hepatic cellular senescence while potentiating cell senescence in white adipose tissue. We find that the effects of BCAAs on hepatic cellular senescence are cell-autonomous, with lower levels of BCAAs protecting cultured cells from antimycin-A induced senescence. Our results demonstrate a direct effect of a specific dietary component on a hallmark of aging and suggest that cellular senescence may be highly susceptible to dietary interventions.
Collapse
Affiliation(s)
- Mariah F Calubag
- Department of Medicine, University of Wisconsin-Madison, Madison, WI 53705 USA
- William S. Middleton Memorial Veterans Hospital, Madison, WI 53705 USA
- Cell and Molecular Biology Graduate Program, University of Wisconsin-Madison, Madison, WI, USA
| | - Ismail Ademi
- Department of Medicine, University of Wisconsin-Madison, Madison, WI 53705 USA
- William S. Middleton Memorial Veterans Hospital, Madison, WI 53705 USA
| | - Isaac Grunow
- Department of Medicine, University of Wisconsin-Madison, Madison, WI 53705 USA
- William S. Middleton Memorial Veterans Hospital, Madison, WI 53705 USA
| | - Lucia Breuer
- Department of Medicine, University of Wisconsin-Madison, Madison, WI 53705 USA
- William S. Middleton Memorial Veterans Hospital, Madison, WI 53705 USA
| | - Reji Babygirija
- Department of Medicine, University of Wisconsin-Madison, Madison, WI 53705 USA
- William S. Middleton Memorial Veterans Hospital, Madison, WI 53705 USA
- Cell and Molecular Biology Graduate Program, University of Wisconsin-Madison, Madison, WI, USA
| | - Penelope Lialios
- Department of Medicine, University of Wisconsin-Madison, Madison, WI 53705 USA
- William S. Middleton Memorial Veterans Hospital, Madison, WI 53705 USA
| | - Sandra Le
- Department of Medicine, University of Wisconsin-Madison, Madison, WI 53705 USA
- William S. Middleton Memorial Veterans Hospital, Madison, WI 53705 USA
| | - Dennis Minton
- Department of Medicine, University of Wisconsin-Madison, Madison, WI 53705 USA
- William S. Middleton Memorial Veterans Hospital, Madison, WI 53705 USA
- Cell and Molecular Biology Graduate Program, University of Wisconsin-Madison, Madison, WI, USA
| | - Michelle M Sonsalla
- Department of Medicine, University of Wisconsin-Madison, Madison, WI 53705 USA
- William S. Middleton Memorial Veterans Hospital, Madison, WI 53705 USA
- Comparative Biomedical Sciences Graduate Program, University of Wisconsin-Madison, Madison, WI, USA
| | - Julia Illiano
- William S. Middleton Memorial Veterans Hospital, Madison, WI 53705 USA
- Wisconsin Laboratory for Surgical Metabolism, Department of Surgery, University of Wisconsin-Madison, Madison, WI, USA
| | - Bailey A Knopf
- Department of Medicine, University of Wisconsin-Madison, Madison, WI 53705 USA
- William S. Middleton Memorial Veterans Hospital, Madison, WI 53705 USA
- Cell and Molecular Biology Graduate Program, University of Wisconsin-Madison, Madison, WI, USA
| | - Fan Xiao
- Department of Medicine, University of Wisconsin-Madison, Madison, WI 53705 USA
- William S. Middleton Memorial Veterans Hospital, Madison, WI 53705 USA
- Nutrition and Metabolism Graduate Program, University of Wisconsin-Madison, Madison, WI, USA
| | - Adam R Konopka
- Department of Medicine, University of Wisconsin-Madison, Madison, WI 53705 USA
- William S. Middleton Memorial Veterans Hospital, Madison, WI 53705 USA
- Cell and Molecular Biology Graduate Program, University of Wisconsin-Madison, Madison, WI, USA
- University of Wisconsin Comprehensive Diabetes Center, University of Wisconsin-Madison, Madison, WI, 53705, USA
| | - David A Harris
- William S. Middleton Memorial Veterans Hospital, Madison, WI 53705 USA
- Wisconsin Laboratory for Surgical Metabolism, Department of Surgery, University of Wisconsin-Madison, Madison, WI, USA
- University of Wisconsin Comprehensive Diabetes Center, University of Wisconsin-Madison, Madison, WI, 53705, USA
| | - Dudley W Lamming
- Department of Medicine, University of Wisconsin-Madison, Madison, WI 53705 USA
- William S. Middleton Memorial Veterans Hospital, Madison, WI 53705 USA
- Cell and Molecular Biology Graduate Program, University of Wisconsin-Madison, Madison, WI, USA
- Comparative Biomedical Sciences Graduate Program, University of Wisconsin-Madison, Madison, WI, USA
- Nutrition and Metabolism Graduate Program, University of Wisconsin-Madison, Madison, WI, USA
- University of Wisconsin Comprehensive Diabetes Center, University of Wisconsin-Madison, Madison, WI, 53705, USA
- University of Wisconsin Carbone Comprehensive Cancer Center, University of Wisconsin-Madison, Madison, WI 53705, USA
| |
Collapse
|
4
|
Calubag MF, Robbins PD, Lamming DW. A nutrigeroscience approach: Dietary macronutrients and cellular senescence. Cell Metab 2024; 36:1914-1944. [PMID: 39178854 PMCID: PMC11386599 DOI: 10.1016/j.cmet.2024.07.025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Revised: 07/09/2024] [Accepted: 07/31/2024] [Indexed: 08/26/2024]
Abstract
Cellular senescence, a process in which a cell exits the cell cycle in response to stressors, is one of the hallmarks of aging. Senescence and the senescence-associated secretory phenotype (SASP)-a heterogeneous set of secreted factors that disrupt tissue homeostasis and promote the accumulation of senescent cells-reprogram metabolism and can lead to metabolic dysfunction. Dietary interventions have long been studied as methods to combat age-associated metabolic dysfunction, promote health, and increase lifespan. A growing body of literature suggests that senescence is responsive to diet, both to calories and specific dietary macronutrients, and that the metabolic benefits of dietary interventions may arise in part through reducing senescence. Here, we review what is currently known about dietary macronutrients' effect on senescence and the SASP, the nutrient-responsive molecular mechanisms that may mediate these effects, and the potential for these findings to inform the development of a nutrigeroscience approach to healthy aging.
Collapse
Affiliation(s)
- Mariah F Calubag
- Department of Medicine, University of Wisconsin-Madison, Madison, WI 53705, USA; William S. Middleton Memorial Veterans Hospital, Madison, WI 53705, USA; Graduate Program in Cellular and Molecular Biology, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Paul D Robbins
- Institute On the Biology of Aging and Metabolism, Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, 6-155 Jackson Hall, 321 Church Street, SE, Minneapolis, MN 55455, USA
| | - Dudley W Lamming
- Department of Medicine, University of Wisconsin-Madison, Madison, WI 53705, USA; William S. Middleton Memorial Veterans Hospital, Madison, WI 53705, USA; Graduate Program in Cellular and Molecular Biology, University of Wisconsin-Madison, Madison, WI 53705, USA.
| |
Collapse
|
5
|
Easter M, Hirsch MJ, Harris E, Howze PH, Matthews EL, Jones LI, Bollenbecker S, Vang S, Tyrrell DJ, Sanders YY, Birket SE, Barnes JW, Krick S. FGF receptors mediate cellular senescence in the cystic fibrosis airway epithelium. JCI Insight 2024; 9:e174888. [PMID: 38916962 PMCID: PMC11383597 DOI: 10.1172/jci.insight.174888] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2023] [Accepted: 06/20/2024] [Indexed: 06/27/2024] Open
Abstract
The number of adults living with cystic fibrosis (CF) has already increased significantly because of drastic improvements in life expectancy attributable to advances in treatment, including the development of highly effective modulator therapy. Chronic airway inflammation in CF contributes to morbidity and mortality, and aging processes like inflammaging and cell senescence influence CF pathology. Our results show that single-cell RNA sequencing data, human primary bronchial epithelial cells from non-CF and CF donors, a CF bronchial epithelial cell line, and Cftr-knockout (Cftr-/-) rats all demonstrated increased cell senescence markers in the CF bronchial epithelium. This was associated with upregulation of fibroblast growth factor receptors (FGFRs) and mitogen-activated protein kinase (MAPK) p38. Inhibition of FGFRs, specifically FGFR4 and to some extent FGFR1, attenuated cell senescence and improved mucociliary clearance, which was associated with MAPK p38 signaling. Mucociliary dysfunction could also be improved using a combination of senolytics in a CF ex vivo model. In summary, FGFR/MAPK p38 signaling contributes to cell senescence in CF airways, which is associated with impaired mucociliary clearance. Therefore, attenuation of cell senescence in the CF airways might be a future therapeutic strategy improving mucociliary dysfunction and lung disease in an aging population with CF.
Collapse
Affiliation(s)
- Molly Easter
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine
| | - Meghan June Hirsch
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine
| | - Elex Harris
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine
- Gregory Fleming James Cystic Fibrosis Research Center, and
| | - Patrick Henry Howze
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine
| | - Emma Lea Matthews
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine
| | - Luke I. Jones
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine
| | - Seth Bollenbecker
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine
| | - Shia Vang
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine
| | - Daniel J. Tyrrell
- Division of Molecular and Cellular Pathology, Department of Pathology, The University of Alabama at Birmingham (UAB), Birmingham, Alabama, USA
| | | | - Susan E. Birket
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine
- Gregory Fleming James Cystic Fibrosis Research Center, and
| | - Jarrod W. Barnes
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine
| | - Stefanie Krick
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine
- Gregory Fleming James Cystic Fibrosis Research Center, and
| |
Collapse
|
6
|
Wei B, Zhou Y, Li Q, Zhen S, Wu Q, Xiao Z, Liao J, Zhu B, Duan J, Yang X, Liang F. Outdoor fine particulate matter exposure and telomere length in humans: A systematic review and meta-analysis. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2024; 275:116206. [PMID: 38518608 DOI: 10.1016/j.ecoenv.2024.116206] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/15/2023] [Revised: 02/17/2024] [Accepted: 03/09/2024] [Indexed: 03/24/2024]
Abstract
Although the association between changes in human telomere length (TL) and ambient fine particulate matter (PM2.5) has been documented, there remains disagreement among the related literature. Our study conducted a systematic review and meta-analysis of epidemiological studies to investigate the health effects of outdoor PM2.5 exposure on human TL after a thorough database search. To quantify the overall effect estimates of TL changes associated with every 10 μg/m3 increase in PM2.5 exposure, we focused on two main topics, which were outdoor long-term exposure and prenatal exposure of PM2.5. Additionally, we included a summary of short-term PM2.5 exposure and its impact on TL due to limited data availability. Our qualitative analysis included 20 studies with 483,600 participants. The meta-analysis showed a statistically significant association between outdoor PM2.5 exposure and shorter human TL, with pooled impact estimates (β) of -0.12 (95% CI: -0.20, -0.03, I2= 95.4%) for general long-term exposure and -0.07 (95% CI: -0.15, 0.00, I2= 74.3%) for prenatal exposure. In conclusion, our findings suggest that outdoor PM2.5 exposure may contribute to TL shortening, and noteworthy associations were observed in specific subgroups, suggesting the impact of various research variables. Larger, high-quality studies using standardized methodologies are necessary to strengthen these conclusions further.
Collapse
Affiliation(s)
- Bincai Wei
- School of Public Health and Emergency Management, Southern University of Science and Technology, Shenzhen 518055, China
| | - Yawen Zhou
- School of Public Health and Emergency Management, Southern University of Science and Technology, Shenzhen 518055, China
| | - Qian Li
- School of Public Health and Emergency Management, Southern University of Science and Technology, Shenzhen 518055, China
| | - Shihan Zhen
- School of Public Health and Emergency Management, Southern University of Science and Technology, Shenzhen 518055, China
| | - Qingyao Wu
- School of Public Health and Emergency Management, Southern University of Science and Technology, Shenzhen 518055, China
| | - Zhiyi Xiao
- School of Public Health and Emergency Management, Southern University of Science and Technology, Shenzhen 518055, China
| | - Jian Liao
- School of Public Health and Emergency Management, Southern University of Science and Technology, Shenzhen 518055, China
| | - Bin Zhu
- School of Public Health and Emergency Management, Southern University of Science and Technology, Shenzhen 518055, China
| | - Jiahao Duan
- School of Public Health and Emergency Management, Southern University of Science and Technology, Shenzhen 518055, China
| | - Xueli Yang
- Department of Occupational and Environmental Health, School of Public Health, Tianjin Medical University, 22 Qixiangtai Road, Heping District, Tianjin 300070, China..
| | - Fengchao Liang
- School of Public Health and Emergency Management, Southern University of Science and Technology, Shenzhen 518055, China.
| |
Collapse
|
7
|
Bellone F, Sardella A, Muscianisi M, Basile G. Fatigue, sarcopenia, and frailty in older adults with inflammatory bowel disease. Minerva Gastroenterol (Torino) 2024; 70:79-88. [PMID: 33988010 DOI: 10.23736/s2724-5985.21.02886-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Inflammatory bowel diseases (IBDs) are characterized by a multifactorial clinical picture, in which age-related physical, functional and psychological symptoms may coexist. The accurate evaluation and identification of such symptomatology acquires considerable importance in the context of older adults, since those core factors typical of IBD may also expose older patients to an increased risk for age-related negative outcomes, such as frailty and disability. The purpose of the present review was to provide an updated overview on the evaluation and management of IBD in the elderly population, with regard to fatigue, sarcopenia, and frailty. The assessment of fatigue might contribute to the identification of early symptoms of IBD, such as pain and mood disorders, which should be treated timely to offer elderly patient a better quality of life. Similarly, an accurate evaluation of sarcopenia might represent a useful Prognostic Index to identify those patients at risk of developing physical frailty. Frailty in IBD should be evaluated not only in relation to the occurrence of negative outcomes, but also should be considered itself as an outcome itself in IBD. A recommendation for future research on this topic might be the implementation of randomized trials, which include older adults and evaluate fatigue, sarcopenia, and frailty. Similarly, the development of tailored intervention programs, based on both physical and psychological outcomes, with the purpose of improving patients' adaptation to the disease, and monitoring the evolution of symptoms and the response to therapies over time, should be encouraged.
Collapse
Affiliation(s)
- Federica Bellone
- Department of Clinical and Experimental Medicine, University of Messina, Messina, Italy
| | - Alberto Sardella
- Department of Clinical and Experimental Medicine, University of Messina, Messina, Italy
| | - Marco Muscianisi
- Department of Clinical and Experimental Medicine, University of Messina, Messina, Italy
| | - Giorgio Basile
- Department of Clinical and Experimental Medicine, University of Messina, Messina, Italy -
| |
Collapse
|
8
|
Schroeder HT, De Lemos Muller CH, Heck TG, Krause M, Homem de Bittencourt PI. Heat shock response during the resolution of inflammation and its progressive suppression in chronic-degenerative inflammatory diseases. Cell Stress Chaperones 2024; 29:116-142. [PMID: 38244765 PMCID: PMC10939074 DOI: 10.1016/j.cstres.2024.01.002] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Revised: 01/11/2024] [Accepted: 01/12/2024] [Indexed: 01/22/2024] Open
Abstract
The heat shock response (HSR) is a crucial biochemical pathway that orchestrates the resolution of inflammation, primarily under proteotoxic stress conditions. This process hinges on the upregulation of heat shock proteins (HSPs) and other chaperones, notably the 70 kDa family of heat shock proteins, under the command of the heat shock transcription factor-1. However, in the context of chronic degenerative disorders characterized by persistent low-grade inflammation (such as insulin resistance, obesity, type 2 diabetes, nonalcoholic fatty liver disease, and cardiovascular diseases) a gradual suppression of the HSR does occur. This work delves into the mechanisms behind this phenomenon. It explores how the Western diet and sedentary lifestyle, culminating in the endoplasmic reticulum stress within adipose tissue cells, trigger a cascade of events. This cascade includes the unfolded protein response and activation of the NOD-like receptor pyrin domain-containing protein-3 inflammasome, leading to the emergence of the senescence-associated secretory phenotype and the propagation of inflammation throughout the body. Notably, the activation of the NOD-like receptor pyrin domain-containing protein-3 inflammasome not only fuels inflammation but also sabotages the HSR by degrading human antigen R, a crucial mRNA-binding protein responsible for maintaining heat shock transcription factor-1 mRNA expression and stability on heat shock gene promoters. This paper underscores the imperative need to comprehend how chronic inflammation stifles the HSR and the clinical significance of evaluating the HSR using cost-effective and accessible tools. Such understanding is pivotal in the development of innovative strategies aimed at the prevention and treatment of these chronic inflammatory ailments, which continue to take a heavy toll on global health and well-being.
Collapse
Affiliation(s)
- Helena Trevisan Schroeder
- Laboratory of Cellular Physiology (FisCel), Department of Physiology, Institute of Basic Health Sciences (ICBS), Federal University of Rio Grande do Sul (UFRGS), Porto Alegre, Rio Grande do Sul, Brazil
| | - Carlos Henrique De Lemos Muller
- Laboratory of Inflammation, Metabolism and Exercise Research (LAPIMEX), Department of Physiology, ICBS, UFRGS, Porto Alegre, Rio Grande do Sul, Brazil
| | - Thiago Gomes Heck
- Post Graduate Program in Integral Health Care (PPGAIS-UNIJUÍ/UNICRUZ/URI), Regional University of Northwestern Rio Grande Do Sul State (UNIJUI) and Post Graduate Program in Mathematical and Computational Modeling (PPGMMC), UNIJUI, Ijuí, Rio Grande do Sul, Brazil
| | - Mauricio Krause
- Laboratory of Inflammation, Metabolism and Exercise Research (LAPIMEX), Department of Physiology, ICBS, UFRGS, Porto Alegre, Rio Grande do Sul, Brazil
| | - Paulo Ivo Homem de Bittencourt
- Laboratory of Cellular Physiology (FisCel), Department of Physiology, Institute of Basic Health Sciences (ICBS), Federal University of Rio Grande do Sul (UFRGS), Porto Alegre, Rio Grande do Sul, Brazil.
| |
Collapse
|
9
|
Karnik SJ, Margetts TJ, Wang HS, Movila A, Oblak AL, Fehrenbacher JC, Kacena MA, Plotkin LI. Mind the Gap: Unraveling the Intricate Dance Between Alzheimer's Disease and Related Dementias and Bone Health. Curr Osteoporos Rep 2024; 22:165-176. [PMID: 38285083 PMCID: PMC10912190 DOI: 10.1007/s11914-023-00847-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 12/19/2023] [Indexed: 01/30/2024]
Abstract
PURPOSE OF REVIEW This review examines the linked pathophysiology of Alzheimer's disease/related dementia (AD/ADRD) and bone disorders like osteoporosis. The emphasis is on "inflammaging"-a low-level inflammation common to both, and its implications in an aging population. RECENT FINDINGS Aging intensifies both ADRD and bone deterioration. Notably, ADRD patients have a heightened fracture risk, impacting morbidity and mortality, though it is uncertain if fractures worsen ADRD. Therapeutically, agents targeting inflammation pathways, especially Nuclear factor kappa-light-chain-enhancer of activated B cells (NF-kB) and TNF-α, appear beneficial for both conditions. Additionally, treatments like Sirtuin 1 (SIRT-1), known for anti-inflammatory and neuroprotective properties, are gaining attention. The interconnectedness of AD/ADRD and bone health necessitates a unified treatment approach. By addressing shared mechanisms, we can potentially transform therapeutic strategies, enriching our understanding and refining care in our aging society. This review article is part of a series of multiple manuscripts designed to determine the utility of using artificial intelligence for writing scientific reviews.
Collapse
Affiliation(s)
- Sonali J Karnik
- Department of Orthopaedic Surgery, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Tyler J Margetts
- Department of Orthopaedic Surgery, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Hannah S Wang
- Department of Orthopaedic Surgery, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Alexandru Movila
- Department of Biomedical Sciences and Comprehensive Care, Indiana University School of Dentistry, Indianapolis, IN, 46202, USA
- Indiana Center for Musculoskeletal Health, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Adrian L Oblak
- Department of Radiology & Imaging Sciences, Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
- Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Jill C Fehrenbacher
- Indiana Center for Musculoskeletal Health, Indiana University School of Medicine, Indianapolis, IN, USA
- Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
- Department of Pharmacology and Toxicology, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Melissa A Kacena
- Department of Orthopaedic Surgery, Indiana University School of Medicine, Indianapolis, IN, 46202, USA.
- Indiana Center for Musculoskeletal Health, Indiana University School of Medicine, Indianapolis, IN, USA.
- Richard L. Roudebush VA Medical Center, Indianapolis, IN, 46202, USA.
| | - Lilian I Plotkin
- Indiana Center for Musculoskeletal Health, Indiana University School of Medicine, Indianapolis, IN, USA.
- Richard L. Roudebush VA Medical Center, Indianapolis, IN, 46202, USA.
- Department of Anatomy, Cell Biology & Physiology, Indiana University School of Medicine, Indianapolis, IN, 46202, USA.
| |
Collapse
|
10
|
Kajdy A, Sys D, Modzelewski J, Bogusławska J, Cymbaluk-Płoska A, Kwiatkowska E, Bednarek-Jędrzejek M, Borowski D, Stefańska K, Rabijewski M, Baran A, Torbe A, Feduniw S, Kwiatkowski S. Evidence of Placental Aging in Late SGA, Fetal Growth Restriction and Stillbirth-A Systematic Review. Biomedicines 2023; 11:1785. [PMID: 37509425 PMCID: PMC10376283 DOI: 10.3390/biomedicines11071785] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2023] [Revised: 06/09/2023] [Accepted: 06/15/2023] [Indexed: 07/30/2023] Open
Abstract
During pregnancy, the placenta undergoes a natural aging process, which is considered normal. However, it has been hypothesized that an abnormally accelerated and premature aging of the placenta may contribute to placenta-related health issues. Placental senescence has been linked to several obstetric complications, including abnormal fetal growth, preeclampsia, preterm birth, and stillbirth, with stillbirth being the most challenging. A systematic search was conducted on Pubmed, Embase, and Scopus databases. Twenty-two full-text articles were identified for the final synthesis. Of these, 15 presented original research and 7 presented narrative reviews. There is a paucity of evidence in the literature on the role of placental aging in late small for gestational age (SGA), fetal growth restriction (FGR), and stillbirth. For future research, guidelines for both planning and reporting research must be implemented. The inclusion criteria should include clear differentiation between early and late SGA and FGR. As for stillbirths, only those with no other known cause of stillbirth should be included in the studies. This means excluding stillbirths due to congenital defects, infections, placental abruption, and maternal conditions affecting feto-maternal hemodynamics.
Collapse
Affiliation(s)
- Anna Kajdy
- First Department of Obstetrics and Gynecology, Centre of Postgraduate Medical Education, 01-004 Warsaw, Poland
| | - Dorota Sys
- Department of Medical Statistics, School of Public Health, Centre of Postgraduate Medical Education, 01-828 Warsaw, Poland
| | - Jan Modzelewski
- First Department of Obstetrics and Gynecology, Centre of Postgraduate Medical Education, 01-004 Warsaw, Poland
| | - Joanna Bogusławska
- Department of Biochemistry and Molecular Biology, Centre of Postgraduate Medical Education, 01-813 Warsaw, Poland
| | - Aneta Cymbaluk-Płoska
- Department of Gynecological Surgery and Gynecological Oncology of Adults and Adolescents, Pomeranian Medical University, 70-111 Szczecin, Poland
| | - Ewa Kwiatkowska
- Department of Nephrology, Transplantology and Internal Medicine, Pomeranian Medical University, 70-111 Szczecin, Poland
| | | | - Dariusz Borowski
- Clinic of Obstetrics and Gynaecology, Provincial Combined Hospital in Kielce, 25-736 Kielce, Poland
| | - Katarzyna Stefańska
- Department of Obstetrics, Medical University of Gdańsk, 80-210 Gdańsk, Poland
| | - Michał Rabijewski
- Department of Reproductive Health, Center of Postgraduate Medical Education, Centre of Postgraduate Medical Education, Żelazna 90 St., 01-004 Warsaw, Poland
| | - Arkadiusz Baran
- First Department of Obstetrics and Gynecology, Centre of Postgraduate Medical Education, 01-004 Warsaw, Poland
| | - Andrzej Torbe
- Department Obstetrics and Gynecology, Pomeranian Medical University, 70-111 Szczecin, Poland
| | - Stepan Feduniw
- Department of Gynecology, University Hospital Zürich, 8091 Zürich, Switzerland
| | - Sebastian Kwiatkowski
- Department Obstetrics and Gynecology, Pomeranian Medical University, 70-111 Szczecin, Poland
| |
Collapse
|
11
|
Choi S, Hill D, Young J, Cordeiro MF. Image processing and supervised machine learning for retinal microglia characterization in senescence. Methods Cell Biol 2023; 181:109-125. [PMID: 38302234 DOI: 10.1016/bs.mcb.2022.12.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
The process of senescence impairs the function of cells and can ultimately be a key factor in the development of disease. With an aging population, senescence-related diseases are increasing in prevalence. Therefore, understanding the mechanisms of cellular senescence within the central nervous system (CNS), including the retina, may yield new therapeutic pathways to slow or even prevent the development of neuro- and retinal degenerative diseases. One method of probing the changing functions of senescent retinal cells is to observe retinal microglial cells. Their morphological structure may change in response to their surrounding cellular environment. In this chapter, we show how microglial cells in the retina, which are implicated in aging and diseases of the CNS, can be identified, quantified, and classified into five distinct morphotypes using image processing and supervised machine learning algorithms. The process involves dissecting, staining, and mounting mouse retinas, before image capture via fluorescence microscopy. The resulting images can then be classified by morphotype using a support vector machine (SVM) we have recently described showing high accuracy. This SVM model uses shape metrics found to correspond with qualitative descriptions of the shape of each morphotype taken from existing literature. We encourage more objective and widespread use of methods of quantification such as this. We believe automatic delineation of the population of microglial cells in the retina, could potentially lead to their use as retinal imaging biomarkers for disease prediction in the future.
Collapse
Affiliation(s)
- Soyoung Choi
- UCL Institute of Ophthalmology, London, United Kingdom; Novai Ltd, Reading, United Kingdom
| | - Daniel Hill
- UCL Institute of Ophthalmology, London, United Kingdom
| | | | - Maria Francesca Cordeiro
- UCL Institute of Ophthalmology, London, United Kingdom; Novai Ltd, Reading, United Kingdom; Imperial College Ophthalmology Research Group, Imperial College London, London, United Kingdom.
| |
Collapse
|
12
|
Akram KM, Kulkarni NS, Brook A, Wyles MD, Anumba DOC. Transcriptomic analysis of the human placenta reveals trophoblast dysfunction and augmented Wnt signalling associated with spontaneous preterm birth. Front Cell Dev Biol 2022; 10:987740. [DOI: 10.3389/fcell.2022.987740] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Accepted: 10/13/2022] [Indexed: 11/13/2022] Open
Abstract
Preterm birth (PTB) is the leading cause of death in under-five children. Worldwide, annually, over 15 million babies are born preterm and 1 million of them die. The triggers and mechanisms of spontaneous PTB remain largely unknown. Most current therapies are ineffective and there is a paucity of reliable predictive biomarkers. Understanding the molecular mechanisms of spontaneous PTB is crucial for developing better diagnostics and therapeutics. To address this need, we conducted RNA-seq transcriptomic analysis, qRT-PCR and ELISA on fresh placental villous tissue from 20 spontaneous preterm and 20 spontaneous term deliveries, to identify genes and signalling pathways involved in the pathogenesis of PTB. Our differential gene expression, gene ontology and pathway analysis revealed several dysregulated genes (including OCLN, OPTN, KRT7, WNT7A, RSPO4, BAMBI, NFATC4, SLC6A13, SLC6A17, SLC26A8 and KLF8) associated with altered trophoblast functions. We identified dysregulated Wnt, oxytocin and cellular senescence signalling pathways in preterm placentas, where augmented Wnt signalling could play a pivotal role in the pathogenesis of PTB due to its diverse biological functions. We also reported two novel targets (ITPR2 and MYLK2) in the oxytocin signalling pathways for further study. Through bioinformatics analysis on DEGs, we identified four key miRNAs, - miR-524-5p, miR-520d-5p, miR-15a-5p and miR-424-5p - which were significantly downregulated in preterm placentas. These miRNAs may have regulatory roles in the aberrant gene expressions that we have observed in preterm placentas. We provide fresh molecular insight into the pathogenesis of spontaneous PTB which may drive further studies to develop new predictive biomarkers and therapeutics.
Collapse
|
13
|
Negre-Salvayre A, Swiader A, Salvayre R, Guerby P. Oxidative stress, lipid peroxidation and premature placental senescence in preeclampsia. Arch Biochem Biophys 2022; 730:109416. [PMID: 36179910 DOI: 10.1016/j.abb.2022.109416] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2022] [Revised: 09/19/2022] [Accepted: 09/23/2022] [Indexed: 11/16/2022]
Abstract
Accelerated placental senescence is associated with preeclampsia (PE) and other pregnancy complications. It is characterized by an accelerated decline in placental function due to the accumulation of senescence patterns such as telomere shortening, mitochondrial dysfunction, oxidative damages, increased expression of phosphorylated (serine-139) histone γ-H2AX, a sensitive marker of double-stranded DNA breaks, accumulation of cross-linked ubiquitinated proteins and sirtuin inhibition. Among the lipid oxidation products generated by the peroxidation of polyunsaturated fatty acids, aldehydes such as acrolein, 4-hydroxy-2-nonenal, 4-oxo-2-nonenal, are present in the blood and placenta from PE-affected women and could contribute to PE pathogenesis and accelerated placental aging. In this review we summarize the current knowledge on premature placental senescence and the role of oxidative stress and lipid oxidation-derived aldehydes in this process, as well as their links with PE pathogenesis. The interest of developing (or not) new therapeutic strategies targeting lipid peroxidation is discussed, the objective being a better understanding of accelerated placental aging in PE pathophysiology, and the prevention of PE bad outcomes.
Collapse
Affiliation(s)
| | | | | | - Paul Guerby
- lnfinity, CNRS, Inserm UMR 1291, University Toulouse III and Gynecology/Obstetrics Department, Paule-de-Viguier Hospital, Toulouse, France
| |
Collapse
|
14
|
Carroll A, Desforges M, Jones CJ, Heazell AE. Morphological and functional changes in placentas from prolonged pregnancies. Placenta 2022; 125:29-35. [DOI: 10.1016/j.placenta.2022.01.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/25/2021] [Revised: 12/23/2021] [Accepted: 01/10/2022] [Indexed: 11/27/2022]
|
15
|
Budhathoki S, Graham C, Sethu P, Kannappan R. Engineered Aging Cardiac Tissue Chip Model for Studying Cardiovascular Disease. Cells Tissues Organs 2022; 211:348-359. [PMID: 34365455 PMCID: PMC8818062 DOI: 10.1159/000516954] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2021] [Accepted: 04/29/2021] [Indexed: 01/03/2023] Open
Abstract
Due to the rapidly growing number of older people worldwide and the concomitant increase in cardiovascular complications, there is an urgent need for age-related cardiac disease modeling and drug screening platforms. In the present study, we developed a cardiac tissue chip model that incorporates hemodynamic loading and mimics essential aspects of the infarcted aging heart. We induced cellular senescence in H9c2 myoblasts using low-dose doxorubicin treatment. These senescent cells were then used to engineer cardiac tissue fibers, which were subjected to hemodynamic stresses associated with pressure-volume changes in the heart. Myocardial ischemia was modeled in the engineered cardiac tissue via hypoxic treatment. Our results clearly show that acute low-dose doxorubicin treatment-induced senescence, as evidenced by morphological and molecular markers, including enlarged and flattened nuclei, DNA damage response foci, and increased expression of cell cycle inhibitor p16INK4a, p53, and ROS. Under normal hemodynamic load, the engineered cardiac tissues demonstrated cell alignment and retained cardiac cell characteristics. Our senescent cardiac tissue model of hypoxia-induced myocardial infarction recapitulated the pathological disease hallmarks such as increased cell death and upregulated expression of ANP and BNP. In conclusion, the described methodology provides a novel approach to generate stress-induced aging cardiac cell phenotypes and engineer cardiac tissue chip models to study the cardiovascular disease pathologies associated with aging.
Collapse
Affiliation(s)
- Sachin Budhathoki
- Division of Cardiovascular Disease, Departments of Medicine, The University of Alabama at Birmingham, Birmingham, AL, USA
| | - Caleb Graham
- Division of Cardiovascular Disease, Departments of Medicine, The University of Alabama at Birmingham, Birmingham, AL, USA
| | - Palaniappan Sethu
- Division of Cardiovascular Disease, Departments of Medicine, The University of Alabama at Birmingham, Birmingham, AL, USA
| | - Ramaswamy Kannappan
- Division of Cardiovascular Disease, Departments of Medicine, The University of Alabama at Birmingham, Birmingham, AL, USA
| |
Collapse
|
16
|
Zambrano E, Nathanielsz PW, Rodríguez-González GL. Developmental programming and ageing of male reproductive function. Eur J Clin Invest 2021; 51:e13637. [PMID: 34107063 DOI: 10.1111/eci.13637] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Revised: 05/25/2021] [Accepted: 06/08/2021] [Indexed: 12/22/2022]
Abstract
Developmental programming predisposes offspring to metabolic, behavioural and reproductive dysfunction in adult life. Evidence is accumulating that ageing phenotype and longevity are in part developmentally programmed in each individual. Unfortunately, there are few studies addressing the effects of developmental programming by maternal nutrition on the rate of ageing of the male reproductive system. This review will discuss effects of foetal exposure to maternal environmental challenges on male offspring fertility and normal ageing of the male reproductive system. We focus on several key factors involved in reproductive ageing such as decreased hormone production, DNA fragmentation, oxidative stress, telomere shortening, epigenetics, maternal lifestyle and nutrition. There is compelling evidence that ageing of the male reproductive system is developmentally programmed. Both maternal over- or undernutrition accelerate ageing of male offspring reproductive function through similar mechanisms such as decreased serum testosterone levels, increase in oxidative stress biomarkers in both the testes and sperm and changes in sperm quality. Importantly, even in adult life, exercise in male offspring of obese mothers improves adverse effects of programming on reproductive function. Maternal consumption of a low-protein diet causes transgenerational effects in progeny via the paternal line. The seminal fluid has effects on the intrauterine environment. Programming by male factors may involve more than just the sperm. Improving knowledge on developmental programming ageing interactions will improve not only male health and life span but also the health of future generations by reducing programming via the paternal line.
Collapse
Affiliation(s)
- Elena Zambrano
- Reproductive Biology Department, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, México
| | | | - Guadalupe L Rodríguez-González
- Reproductive Biology Department, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, México
| |
Collapse
|
17
|
Wu Y, Xu J, Rong X, Wang F, Wang H, Zhao C. Gut microbiota alterations and health status in aging adults: From correlation to causation. Aging Med (Milton) 2021; 4:206-213. [PMID: 34553118 PMCID: PMC8444961 DOI: 10.1002/agm2.12167] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Revised: 05/31/2021] [Accepted: 06/01/2021] [Indexed: 12/17/2022] Open
Abstract
The deterioration of tissue structure and decline in physiological function during aging are accompanied by alterations to the gut microbiota. The elderly has higher risks of various diseases and chronic diseases. However, inter-individual differences are more apparent in elderly than younger, and a proportion of individuals have a delayed onset or even avoid developing chronic diseases. This difference in health status is influenced by both heredity and Lifestyle and environmental factors. During the process of aging, the gut microbiota is also affected by the external environment, and provides a buffer to external challenge, and thus the gut microbiota reflects an individual's personal experience. Moreover, the immune system undergoes a series of changes with age, which are related to chronic inflammation in the elderly. The formation, maturation and senescence of the intestinal immune system is closely related to the gut microbiota. Additionally, changes in the gut microbiota of elderly individuals may modulate the immune system, which may in turn affect health status. Herein, we summarize the correlations between the gut microbiota with individual health status in the elderly and explore the related mechanisms, which may provide a basis to maintain or enhance the health of the elderly though interventions targeting the gut microbiota.
Collapse
Affiliation(s)
- Yong‐Lin Wu
- Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS)School of Basic Medical SciencesShanghai Medical College, & National Clinical Research Center for Aging and MedicineHuashan HospitalFudan UniversityShanghaiChina
| | - Jun Xu
- Department of Orthopaedic SurgeryShanghai Sixth People’s HospitalShanghai Jiaotong UniversityShanghaiChina
| | - Xing‐Yu Rong
- Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS)School of Basic Medical SciencesShanghai Medical College, & National Clinical Research Center for Aging and MedicineHuashan HospitalFudan UniversityShanghaiChina
| | - Feifei Wang
- Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS)School of Basic Medical SciencesShanghai Medical College, & National Clinical Research Center for Aging and MedicineHuashan HospitalFudan UniversityShanghaiChina
| | - Hui‐Jing Wang
- Laboratory of NeuropsychopharmacologyCollege of Fundamental MedicineShanghai University of Medicine & Health ScienceShanghaiChina
| | - Chao Zhao
- Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS)School of Basic Medical SciencesShanghai Medical College, & National Clinical Research Center for Aging and MedicineHuashan HospitalFudan UniversityShanghaiChina
| |
Collapse
|
18
|
Parvizi M, Franchi F, Arendt BK, Ebtehaj S, Rodriguez-Porcel M, Lanza IR. Senolytic agents lessen the severity of abdominal aortic aneurysm in aged mice. Exp Gerontol 2021; 151:111416. [PMID: 34022272 PMCID: PMC11443445 DOI: 10.1016/j.exger.2021.111416] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Revised: 05/16/2021] [Accepted: 05/18/2021] [Indexed: 12/12/2022]
Abstract
Age is a major risk factor for abdominal aortic aneurysm (AAA), for which treatment options are limited to surgical intervention for large AAA and watchful waiting for small aneurysms. However, the factors that regulate the expansion of aneurysms are unclear. Development of new therapeutic strategies to prevent or treat small aneurysms awaits a more thorough understanding of the etiology of AAA formation and progression with aging. A variety of structural and functional changes have been reported in aging vasculature, but emerging evidence implicates senescent cells in the formation of AAA through their paracrine effects on vascular wall cell populations. Here we show that aging is associated with transcriptional changes in abdominal aortic tissue consistent with loss of smooth muscle cells, leukocyte adhesion, inflammation, and accumulation of senescent cells in the vascular wall and surrounding perivascular adipose tissue. Furthermore, aged mice demonstrated anatomical and histopathological features of AAA development in response to administration of angiotensin II over 28 days. Importantly, in our study we sought to determine if reducing senescent cells could lessen the severity of AAA in aged mice. We find that pretreatment of aged mice with oral senolytic agents (dasatinib + quercetin) reduced senescent cell abundance in the arterial walls and surrounding tissues and lessened the severity of AAA in response to angiotensin II administration. These data provide important preliminary evidence supporting a role of senescent cells in age-related AAA formation and progression and suggest that strategies to reduce senescent cell burden hold promise to lessen AAA severity.
Collapse
Affiliation(s)
- Mojtaba Parvizi
- Endocrine Research Unit, Division of Endocrinology, Department of Internal Medicine, Mayo Clinic, Rochester, MN, United States of America
| | - Federico Franchi
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN, United States of America
| | - Bonnie K Arendt
- Endocrine Research Unit, Division of Endocrinology, Department of Internal Medicine, Mayo Clinic, Rochester, MN, United States of America
| | - Sanam Ebtehaj
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN, United States of America
| | | | - Ian R Lanza
- Endocrine Research Unit, Division of Endocrinology, Department of Internal Medicine, Mayo Clinic, Rochester, MN, United States of America.
| |
Collapse
|
19
|
Martin PJ, Billet S, Landkocz Y, Fougère B. Inflammation at the Crossroads: the Combined Effects of COVID-19, Ageing, and Air Pollution. J Frailty Aging 2021; 10:281-285. [PMID: 34105713 PMCID: PMC7948651 DOI: 10.14283/jfa.2021.8] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The global COVID-19 pandemic has highlighted different vulnerability profiles among individuals. With the highest mortality rate, the elderly are a very sensitive group. With regard to the main symptoms, a failure of the respiratory system, associated with deregulation of the immune system, has been observed. These symptoms may also be encountered in chronic exposure of susceptible populations to air pollution, including exacerbation of the inflammatory response. Is there a relationship between age, pollution exposure and the severity of COVID-19? Although it is unclear how these parameters are related, the same pathways can be activated and appear to find a common mechanism of action in inflammation.
Collapse
Affiliation(s)
- P J Martin
- Dr. Sylvain Billet, Univ. Littoral Côte d'Opale, UR 4492, UCEIV, Maison de la Recherche en Environnement Industriel 2, 189A, Avenue Maurice Schumann, 59140 Dunkerque, France. Phone: +33-3 28 23 76 41, E-mail:
| | | | | | | |
Collapse
|
20
|
Kajdy A, Modzelewski J, Cymbaluk-Płoska A, Kwiatkowska E, Bednarek-Jędrzejek M, Borowski D, Stefańska K, Rabijewski M, Torbé A, Kwiatkowski S. Molecular Pathways of Cellular Senescence and Placental Aging in Late Fetal Growth Restriction and Stillbirth. Int J Mol Sci 2021; 22:4186. [PMID: 33919502 PMCID: PMC8072706 DOI: 10.3390/ijms22084186] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Revised: 04/13/2021] [Accepted: 04/14/2021] [Indexed: 12/20/2022] Open
Abstract
Abnormally accelerated, premature placental senescence plays a crucial role in the genesis of pregnancy pathologies. Abnormal growth in the third trimester can present as small for gestational age fetuses or fetal growth restriction. One differs from the other by the presence of signs of placental insufficiency and the risk of stillbirth. The majority of stillbirths occur in normally grown fetuses and are classified as "unexplained", which often leads to conclusions that they were unpreventable. The main characteristic of aging is a gradual decline in the function of cells, tissues, and organs. These changes result in the accumulation of senescent cells in mitotic tissues. These cells begin the aging process that disrupts tissues' normal functions by affecting neighboring cells, degrading the extracellular matrix, and reducing tissues' regeneration capacity. Different degrees of abnormal placentation result in the severity of fetal growth restriction and its sequelae, including fetal death. This review aims to present the current knowledge and identify future research directions to understand better placental aging in late fetal growth restriction and unexplained stillbirth. We hypothesized that the final diagnosis of placental insufficiency can be made only using markers of placental senescence.
Collapse
Affiliation(s)
- Anna Kajdy
- Department of Reproductive Health, Centre of Postgraduate Medical Education, Żelazna 90 St., 01-004 Warsaw, Poland; (J.M.); (M.R.)
| | - Jan Modzelewski
- Department of Reproductive Health, Centre of Postgraduate Medical Education, Żelazna 90 St., 01-004 Warsaw, Poland; (J.M.); (M.R.)
| | - Aneta Cymbaluk-Płoska
- Department of Gynecological Surgery and Gynecological Oncology of Adults and Adolescents, Pomeranian Medical University, Al. Powstańców Wlkp. 72, 70-111 Szczecin, Poland;
| | - Ewa Kwiatkowska
- Department of Nephrology, Transplantology and Internal Medicine, Pomeranian Medical University, Al. Powstańców Wlkp. 72, 70-111 Szczecin, Poland;
| | - Magdalena Bednarek-Jędrzejek
- Department Obstetrics and Gynecology, Pomeranian Medical University, Al. Powstańców Wlkp. 72, 70-111 Szczecin, Poland; (M.B.-J.); (A.T.)
| | - Dariusz Borowski
- Clinic of Fetal-Maternal, Gynecology and Neonatology, Collegium Medicum, Nicolaus Copernicus University in Bydgoszcz, Łukasiewicza 1 St., 85-821 Bydgoszcz, Poland;
| | - Katarzyna Stefańska
- Department of Obstetrics, Medical University of Gdańsk, Mariana Smoluchowskiego 17 St., 80-214 Gdańsk, Poland;
| | - Michał Rabijewski
- Department of Reproductive Health, Centre of Postgraduate Medical Education, Żelazna 90 St., 01-004 Warsaw, Poland; (J.M.); (M.R.)
| | - Andrzej Torbé
- Department Obstetrics and Gynecology, Pomeranian Medical University, Al. Powstańców Wlkp. 72, 70-111 Szczecin, Poland; (M.B.-J.); (A.T.)
| | - Sebastian Kwiatkowski
- Department Obstetrics and Gynecology, Pomeranian Medical University, Al. Powstańców Wlkp. 72, 70-111 Szczecin, Poland; (M.B.-J.); (A.T.)
| |
Collapse
|
21
|
The role of curcumin in aging and senescence: Molecular mechanisms. Biomed Pharmacother 2021; 134:111119. [DOI: 10.1016/j.biopha.2020.111119] [Citation(s) in RCA: 195] [Impact Index Per Article: 48.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2020] [Revised: 11/29/2020] [Accepted: 12/04/2020] [Indexed: 12/11/2022] Open
|
22
|
Molecular Mechanisms to Target Cellular Senescence in Hepatocellular Carcinoma. Cells 2020; 9:cells9122540. [PMID: 33255630 PMCID: PMC7761055 DOI: 10.3390/cells9122540] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2020] [Revised: 11/17/2020] [Accepted: 11/20/2020] [Indexed: 12/24/2022] Open
Abstract
Hepatocellular carcinoma (HCC) has emerged as a major cause of cancer-related death and is the most common type of liver cancer. Due to the current paucity of drugs for HCC therapy there is a pressing need to develop new therapeutic concepts. In recent years, the role of Serum Response Factor (SRF) and its coactivators, Myocardin-Related Transcription Factors A and B (MRTF-A and -B), in HCC formation and progression has received considerable attention. Targeting MRTFs results in HCC growth arrest provoked by oncogene-induced senescence. The induction of senescence acts as a tumor-suppressive mechanism and therefore gains consideration for pharmacological interventions in cancer therapy. In this article, we describe the key features and the functional role of senescence in light of the development of novel drug targets for HCC therapy with a focus on MRTFs.
Collapse
|
23
|
Kim GD, Park S. Effects of Cudrania tricuspidata on anti-senescence in high glucose-treated endothelial cells via the Akt/p53/p21 pathway. Food Sci Nutr 2020; 8:5999-6006. [PMID: 33282251 PMCID: PMC7684615 DOI: 10.1002/fsn3.1885] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2020] [Revised: 08/19/2020] [Accepted: 08/23/2020] [Indexed: 01/08/2023] Open
Abstract
The roles of Cudrania tricuspidata (CT) in the prevention of senescence and the underlying mechanisms have not been elucidated. In a high glucose (HG)-induced senescent endothelial cell (EC) culture, CT (20 µg/ml) reduced the number of senescence-associated β-galactosidase-positive cells by 8.3% compared with the control group and increased the expression of p-Sirt1 by more than twofold compared with the control group. Moreover, 20 μg/ml CT treatment doubled the activity of p-Akt, which was inhibited by HG, compared with the control group. In addition, CT treatment decreased the expression of p53, p21, and Rb, which was increased by HG. Overall, CT delays HG-induced senescence via the Akt/p53/p21 pathway, suggesting its potential as a functional agent for the protection of ECs.
Collapse
Affiliation(s)
- Gi Dae Kim
- Department of Food and NutritionKyungnam UniversityChangwon‐siRepublic of Korea
| | - Seonghee Park
- Department of Biological ScienceSookmyung Women's UniversitySeoulRepublic of Korea
| |
Collapse
|
24
|
Sun C, Groom KM, Oyston C, Chamley LW, Clark AR, James JL. The placenta in fetal growth restriction: What is going wrong? Placenta 2020; 96:10-18. [PMID: 32421528 DOI: 10.1016/j.placenta.2020.05.003] [Citation(s) in RCA: 126] [Impact Index Per Article: 25.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/30/2020] [Revised: 04/17/2020] [Accepted: 05/07/2020] [Indexed: 02/06/2023]
Abstract
The placenta is essential for the efficient delivery of nutrients and oxygen from mother to fetus to maintain normal fetal growth. Dysfunctional placental development underpins many pregnancy complications, including fetal growth restriction (FGR) a condition in which the fetus does not reach its growth potential. The FGR placenta is smaller than normal placentae throughout gestation and displays maldevelopment of both the placental villi and the fetal vasculature within these villi. Specialized epithelial cells called trophoblasts exhibit abnormal function and development in FGR placentae. This includes an altered balance between proliferation and apoptotic death, premature cellular senescence, and reduced colonisation of the maternal decidual tissue. Thus, the placenta undergoes aberrant changes at the macroscopic to cellular level in FGR, which can limit exchange capacity and downstream fetal growth. This review aims to compile stereological, in vitro, and imaging data to create a holistic overview of the FGR placenta and its pathophysiology, with a focus on the contribution of trophoblasts.
Collapse
Affiliation(s)
- Cherry Sun
- Department of Obstetrics and Gynaecology, Faculty of Medical and Health Sciences, The University of Auckland, 85 Park Road, Grafton, Auckland, 1023, New Zealand.
| | - Katie M Groom
- Liggins Institute, The University of Auckland, 85 Park Road, Grafton, Auckland, 1023, New Zealand
| | - Charlotte Oyston
- Department of Obstetrics and Gynaecology, Faculty of Medical and Health Sciences, The University of Auckland, 85 Park Road, Grafton, Auckland, 1023, New Zealand
| | - Lawrence W Chamley
- Department of Obstetrics and Gynaecology, Faculty of Medical and Health Sciences, The University of Auckland, 85 Park Road, Grafton, Auckland, 1023, New Zealand
| | - Alys R Clark
- Auckland Bioengineering Institute, The University of Auckland, Auckland Bioengineering, House, Level 6/70 Symonds Street, Grafton, Auckland, 1010, New Zealand
| | - Joanna L James
- Department of Obstetrics and Gynaecology, Faculty of Medical and Health Sciences, The University of Auckland, 85 Park Road, Grafton, Auckland, 1023, New Zealand
| |
Collapse
|
25
|
Park JH, Lee NK, Lim HJ, Ji ST, Kim YJ, Jang WB, Kim DY, Kang S, Yun J, Ha JS, Kim H, Lee D, Baek SH, Kwon SM. Pharmacological inhibition of mTOR attenuates replicative cell senescence and improves cellular function via regulating the STAT3-PIM1 axis in human cardiac progenitor cells. Exp Mol Med 2020; 52:615-628. [PMID: 32273566 PMCID: PMC7210934 DOI: 10.1038/s12276-020-0374-4] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2019] [Revised: 09/08/2019] [Accepted: 10/29/2019] [Indexed: 12/19/2022] Open
Abstract
The mammalian target of rapamycin (mTOR) signaling pathway efficiently regulates the energy state of cells and maintains tissue homeostasis. Dysregulation of the mTOR pathway has been implicated in several human diseases. Rapamycin is a specific inhibitor of mTOR and pharmacological inhibition of mTOR with rapamycin promote cardiac cell generation from the differentiation of mouse and human embryonic stem cells. These studies strongly implicate a role of sustained mTOR activity in the differentiating functions of embryonic stem cells; however, they do not directly address the required effect for sustained mTOR activity in human cardiac progenitor cells. In the present study, we evaluated the effect of mTOR inhibition by rapamycin on the cellular function of human cardiac progenitor cells and discovered that treatment with rapamycin markedly attenuated replicative cell senescence in human cardiac progenitor cells (hCPCs) and promoted their cellular functions. Furthermore, rapamycin not only inhibited mTOR signaling but also influenced signaling pathways, including STAT3 and PIM1, in hCPCs. Therefore, these data reveal a crucial function for rapamycin in senescent hCPCs and provide clinical strategies based on chronic mTOR activity.
Collapse
Affiliation(s)
- Ji Hye Park
- Laboratory of Regenerative Medicine and Stem Cell Biology, Department of Physiology, Medical Research Institute, School of Medicine, Pusan National University, Yangsan, 50612, Republic of Korea
- Research Institute of Convergence Biomedical Science and Technology, Pusan National University School of Medicine, Yangsan, 50612, Republic of Korea
- R&D Center for Advanced Pharmaceuticals & Evaluation, Korea Institute of Toxicology, Korea Research Institute of Chemical Technology, 141 Gajeong-ro, Yuseong-gu, Daejeon, 34114, South Korea
| | - Na Kyoung Lee
- Laboratory of Regenerative Medicine and Stem Cell Biology, Department of Physiology, Medical Research Institute, School of Medicine, Pusan National University, Yangsan, 50612, Republic of Korea
- Research Institute of Convergence Biomedical Science and Technology, Pusan National University School of Medicine, Yangsan, 50612, Republic of Korea
| | - Hye Ji Lim
- Laboratory of Regenerative Medicine and Stem Cell Biology, Department of Physiology, Medical Research Institute, School of Medicine, Pusan National University, Yangsan, 50612, Republic of Korea
- Research Institute of Convergence Biomedical Science and Technology, Pusan National University School of Medicine, Yangsan, 50612, Republic of Korea
| | - Seung Taek Ji
- Laboratory of Regenerative Medicine and Stem Cell Biology, Department of Physiology, Medical Research Institute, School of Medicine, Pusan National University, Yangsan, 50612, Republic of Korea
- Research Institute of Convergence Biomedical Science and Technology, Pusan National University School of Medicine, Yangsan, 50612, Republic of Korea
| | - Yeon-Ju Kim
- Laboratory of Regenerative Medicine and Stem Cell Biology, Department of Physiology, Medical Research Institute, School of Medicine, Pusan National University, Yangsan, 50612, Republic of Korea
- Research Institute of Convergence Biomedical Science and Technology, Pusan National University School of Medicine, Yangsan, 50612, Republic of Korea
| | - Woong Bi Jang
- Laboratory of Regenerative Medicine and Stem Cell Biology, Department of Physiology, Medical Research Institute, School of Medicine, Pusan National University, Yangsan, 50612, Republic of Korea
- Research Institute of Convergence Biomedical Science and Technology, Pusan National University School of Medicine, Yangsan, 50612, Republic of Korea
| | - Da Yeon Kim
- Laboratory of Regenerative Medicine and Stem Cell Biology, Department of Physiology, Medical Research Institute, School of Medicine, Pusan National University, Yangsan, 50612, Republic of Korea
- Research Institute of Convergence Biomedical Science and Technology, Pusan National University School of Medicine, Yangsan, 50612, Republic of Korea
| | - Songhwa Kang
- Laboratory of Regenerative Medicine and Stem Cell Biology, Department of Physiology, Medical Research Institute, School of Medicine, Pusan National University, Yangsan, 50612, Republic of Korea
- Research Institute of Convergence Biomedical Science and Technology, Pusan National University School of Medicine, Yangsan, 50612, Republic of Korea
| | - Jisoo Yun
- Laboratory of Regenerative Medicine and Stem Cell Biology, Department of Physiology, Medical Research Institute, School of Medicine, Pusan National University, Yangsan, 50612, Republic of Korea
- Research Institute of Convergence Biomedical Science and Technology, Pusan National University School of Medicine, Yangsan, 50612, Republic of Korea
| | - Jong Seong Ha
- Laboratory of Regenerative Medicine and Stem Cell Biology, Department of Physiology, Medical Research Institute, School of Medicine, Pusan National University, Yangsan, 50612, Republic of Korea
- Research Institute of Convergence Biomedical Science and Technology, Pusan National University School of Medicine, Yangsan, 50612, Republic of Korea
| | - Hyungtae Kim
- Department of Thoracic and Cardiovascular Surgery, Pusan National University Yangsan Hospital, School of Medicine, Pusan National University, Yangsan, 50612, Republic of Korea
| | - Dongjun Lee
- Department of Convergence Medical Science, Pusan National University School of Medicine, Yangsan, 50612, Republic of Korea.
| | - Sang Hong Baek
- Laboratory of Cardiovascular Disease, Division of Cardiology, School of Medicine, The Catholic University of Korea, Seoul, 137-040, Republic of Korea.
| | - Sang-Mo Kwon
- Laboratory of Regenerative Medicine and Stem Cell Biology, Department of Physiology, Medical Research Institute, School of Medicine, Pusan National University, Yangsan, 50612, Republic of Korea.
- Department of Thoracic and Cardiovascular Surgery, Pusan National University Yangsan Hospital, School of Medicine, Pusan National University, Yangsan, 50612, Republic of Korea.
| |
Collapse
|
26
|
Role of the Nuclear Lamina in Age-Associated Nuclear Reorganization and Inflammation. Cells 2020; 9:cells9030718. [PMID: 32183360 PMCID: PMC7140666 DOI: 10.3390/cells9030718] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2020] [Revised: 03/11/2020] [Accepted: 03/12/2020] [Indexed: 02/06/2023] Open
Abstract
Aging is characterized by the gradual loss of tissue function and integrity. Activation of inflammatory responses accelerates the deterioration of cells and tissues. Many studies have shown that alteration of the components of the nuclear lamina is associated with inflammation, both in vivo and in vitro. However, the mechanism by which the nuclear lamina regulates inflammation is largely unknown. Recent studies have suggested that the nuclear lamina regulates both organization of the three-dimensional chromatin structure at the nuclear periphery and global gene expression, such as the expression of inflammatory response genes. Here, we discuss the current updates in the research on nuclear lamina alteration, activation of inflammation, and nuclear reorganization in models of cellular senescence and organismal aging.
Collapse
|
27
|
Inflammation, Frailty and Cardiovascular Disease. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1216:55-64. [PMID: 31894547 DOI: 10.1007/978-3-030-33330-0_7] [Citation(s) in RCA: 155] [Impact Index Per Article: 31.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Chronic inflammation, which is called "inflamm-aging" , is characterized by an increased level of inflammatory cytokines in response to physiological and environmental stressors, and causes the immune system to function consistently at a low level, even though it is not effective. Possible causes of inflammaging include genetic susceptibility, visceral obesity, changes in gut microbiota and permeability, chronic infections and cellular senescence. Inflammation has a role in the development of many age-related diseases, such as frailty. Low grade chronic inflammation can also increase the risk of atherosclerosis and insulin resistance which are the leading mechanisms in the development of cardiovascular diseases (CVD). As it is well known that the risk of CVD is higher in older people with frailty and the risk of frailty is higher in patients with CVD, there may be relationship between inflammation and the development of CVD and frailty. Therefore, this important issue will be discussed in this chapter.
Collapse
|
28
|
O'Hara SP, Splinter PL, Trussoni CE, Guicciardi ME, Splinter NP, Al Suraih MS, Nasser-Ghodsi N, Stollenwerk D, Gores GJ, LaRusso NF. The transcription factor ETS1 promotes apoptosis resistance of senescent cholangiocytes by epigenetically up-regulating the apoptosis suppressor BCL2L1. J Biol Chem 2019; 294:18698-18713. [PMID: 31659122 PMCID: PMC6901313 DOI: 10.1074/jbc.ra119.010176] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2019] [Revised: 10/15/2019] [Indexed: 12/21/2022] Open
Abstract
Primary sclerosing cholangitis (PSC) is an idiopathic, progressive cholangiopathy. Cholangiocyte senescence is important in PSC pathogenesis, and we have previously reported that senescence is regulated by the transcription factor ETS proto-oncogene 1 (ETS1) and associated with overexpression of BCL2 like 1 (BCL2L1 or BCL-xL), an anti-apoptotic BCL2-family member. Here, we further explored the mechanisms regulating BCL-xL-mediated, apoptosis resistance in senescent cholangiocytes and uncovered that ETS1 and the histone acetyltransferase E1A-binding protein P300 (EP300 or p300) both promote BCL-xL transcription. Using immunofluorescence, we found that BCL-xL protein expression is increased both in cholangiocytes of livers from individuals with PSC and a mouse model of PSC. Using an in vitro model of lipopolysaccharide-induced senescence in normal human cholangiocytes (NHCs), we found increased BCL-xL mRNA and protein levels, and ChIP-PCRs indicated increased occupancy of ETS1, p300, and histone 3 Lys-27 acetylation (H3K27Ac) at the BCL-xL promoter. Using co-immunoprecipitation and proximity ligation assays, we further demonstrate that ETS1 and p300 physically interact in senescent but not control NHCs. Additionally, mutagenesis of predicted ETS1-binding sites within the BCL-xL promoter blocked luciferase reporter activity, and CRISPR/Cas9-mediated genetic deletion of ETS1 reduced senescence-associated BCL-xL expression. In senescent NHCs, TRAIL-mediated apoptosis was reduced ∼70%, and ETS1 deletion or RNAi-mediated BCL-xL suppression increased apoptosis. Overall, our results suggest that ETS1 and p300 promote senescent cholangiocyte resistance to apoptosis by modifying chromatin and inducing BCL-xL expression. These findings reveal ETS1 as a central regulator of both cholangiocyte senescence and the associated apoptosis-resistant phenotype.
Collapse
Affiliation(s)
- Steven P O'Hara
- Division of Gastroenterology and Hepatology and the Mayo Clinic Center for Cell Signaling in Gastroenterology, Mayo Clinic, Rochester, Minnesota 55905.
| | - Patrick L Splinter
- Division of Gastroenterology and Hepatology and the Mayo Clinic Center for Cell Signaling in Gastroenterology, Mayo Clinic, Rochester, Minnesota 55905
| | - Christy E Trussoni
- Division of Gastroenterology and Hepatology and the Mayo Clinic Center for Cell Signaling in Gastroenterology, Mayo Clinic, Rochester, Minnesota 55905
| | - Maria Eugenia Guicciardi
- Division of Gastroenterology and Hepatology and the Mayo Clinic Center for Cell Signaling in Gastroenterology, Mayo Clinic, Rochester, Minnesota 55905
| | - Noah P Splinter
- Division of Gastroenterology and Hepatology and the Mayo Clinic Center for Cell Signaling in Gastroenterology, Mayo Clinic, Rochester, Minnesota 55905
| | - Mohammed S Al Suraih
- Division of Gastroenterology and Hepatology and the Mayo Clinic Center for Cell Signaling in Gastroenterology, Mayo Clinic, Rochester, Minnesota 55905
| | - Navine Nasser-Ghodsi
- Division of Gastroenterology and Hepatology and the Mayo Clinic Center for Cell Signaling in Gastroenterology, Mayo Clinic, Rochester, Minnesota 55905
| | - Deborah Stollenwerk
- Division of Gastroenterology and Hepatology and the Mayo Clinic Center for Cell Signaling in Gastroenterology, Mayo Clinic, Rochester, Minnesota 55905
| | - Gregory J Gores
- Division of Gastroenterology and Hepatology and the Mayo Clinic Center for Cell Signaling in Gastroenterology, Mayo Clinic, Rochester, Minnesota 55905
| | - Nicholas F LaRusso
- Division of Gastroenterology and Hepatology and the Mayo Clinic Center for Cell Signaling in Gastroenterology, Mayo Clinic, Rochester, Minnesota 55905
| |
Collapse
|
29
|
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: 3.2] [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
|
30
|
Hartmann P, Chu H, Duan Y, Schnabl B. Gut microbiota in liver disease: too much is harmful, nothing at all is not helpful either. Am J Physiol Gastrointest Liver Physiol 2019; 316:G563-G573. [PMID: 30767680 PMCID: PMC6580239 DOI: 10.1152/ajpgi.00370.2018] [Citation(s) in RCA: 54] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/13/2018] [Revised: 02/15/2019] [Accepted: 02/15/2019] [Indexed: 01/31/2023]
Abstract
The intestinal microbiome plays a major role in the pathogenesis of liver disease, with a hallmark event being dysbiosis, or an imbalance of pathobionts and beneficial bacteria with the associated deleterious effects on their host. Reducing the number of intestinal bacteria with antibiotic treatment is generally advantageous in experimental liver diseases. Complete absence of intestinal microbiota as in germ-free rodents can be protective in autoimmune hepatitis and hepatic tumors induced by chemicals, or it can exacerbate disease as in acute toxic liver injury and liver fibrosis/cirrhosis. In alcoholic liver disease, nonalcoholic fatty liver disease, and autoimmune cholangiopathies, germ-free status can be associated with worsened or improved hepatic phenotype depending on the experimental model and type of rodent. Some of the unexpected outcomes can be explained by the limitations of rodents raised in a germ-free environment including a deficient immune system and an altered metabolism of lipids, cholesterol, xenobiotics/toxins, and bile acids. Given these limitations and to advance understanding of the interactions between host and intestinal microbiota, simplified model systems such as humanized gnotobiotic mice, or gnotobiotic mice monoassociated with a single bacterial strain or colonized with a defined set of microbes, are unique and useful models for investigation of liver disease in a complex ecosystem.
Collapse
Affiliation(s)
- Phillipp Hartmann
- Department of Pediatrics, University of California, San Diego, La Jolla, California
| | - Huikuan Chu
- Division of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology , Wuhan , China
- Department of Medicine, University of California, San Diego, La Jolla, California
| | - Yi Duan
- Department of Medicine, University of California, San Diego, La Jolla, California
- Department of Medicine, Veterans Affairs San Diego Healthcare System, San Diego, California
| | - Bernd Schnabl
- Department of Medicine, University of California, San Diego, La Jolla, California
- Department of Medicine, Veterans Affairs San Diego Healthcare System, San Diego, California
| |
Collapse
|
31
|
Major trauma and acceleration of the ageing process. Ageing Res Rev 2018; 48:32-39. [PMID: 30316759 DOI: 10.1016/j.arr.2018.10.001] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2018] [Revised: 09/07/2018] [Accepted: 10/08/2018] [Indexed: 12/30/2022]
Abstract
It is well established that numerous factors can affect the rate at which we age biologically. Diet, physical activity, lifestyle and our genes all play a major role in influencing the ageing trajectory and longevity. Major trauma affects millions globally, is the major cause of death in young adults and could influence ageing processes but has largely been ignored by biogenterologists. The long-term health consequences of physical trauma are well known in the medical community, how trauma effects the ageing process at a molecular level is not. It has long been difficult to assess ageing trajectories due to the absence of a biomarker of biological rather than chronological age. Recent advances in epigenetics have helped by identifying specific DNA methylation sites as good indicators of biological age. Recent investigations into the impact of psychological trauma and the associated physical stress on accelerating ageing as measured by epigenetic drift are promising. The physical and metabolic stress which is synonymous with physical trauma may also accelerate the ageing process. We suggest that long term epigenetic profiling is required to understand to what degree the ageing trajectory is altered by trauma, which will in turn add support for the development of novel therapies to improve health outcomes for survivors of traumatic injury.
Collapse
|
32
|
Castillo-Morales A, Monzón-Sandoval J, Urrutia AO, Gutiérrez H. Postmitotic cell longevity-associated genes: a transcriptional signature of postmitotic maintenance in neural tissues. Neurobiol Aging 2018; 74:147-160. [PMID: 30448614 DOI: 10.1016/j.neurobiolaging.2018.10.015] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2018] [Revised: 10/03/2018] [Accepted: 10/11/2018] [Indexed: 12/24/2022]
Abstract
Different cell types have different postmitotic maintenance requirements. Nerve cells, however, are unique in this respect as they need to survive and preserve their functional complexity for the entire lifetime of the organism, and failure at any level of their supporting mechanisms leads to a wide range of neurodegenerative conditions. Whether these differences across tissues arise from the activation of distinct cell type-specific maintenance mechanisms or the differential activation of a common molecular repertoire is not known. To identify the transcriptional signature of postmitotic cellular longevity (PMCL), we compared whole-genome transcriptome data from human tissues ranging in longevity from 120 days to over 70 years and found a set of 81 genes whose expression levels are closely associated with increased cell longevity. Using expression data from 10 independent sources, we found that these genes are more highly coexpressed in longer-living tissues and are enriched in specific biological processes and transcription factor targets compared with randomly selected gene samples. Crucially, we found that PMCL-associated genes are downregulated in the cerebral cortex and substantia nigra of patients with Alzheimer's and Parkinson's disease, respectively, as well as Hutchinson-Gilford progeria-derived fibroblasts, and that this downregulation is specifically linked to their underlying association with cellular longevity. Moreover, we found that sexually dimorphic brain expression of PMCL-associated genes reflects sexual differences in lifespan in humans and macaques. Taken together, our results suggest that PMCL-associated genes are part of a generalized machinery of postmitotic maintenance and functional stability in both neural and non-neural cells and support the notion of a common molecular repertoire differentially engaged in different cell types with different survival requirements.
Collapse
Affiliation(s)
- Atahualpa Castillo-Morales
- School of Life Sciences, University of Lincoln, Lincoln, UK; Milner Centre for Evolution, Department of Biology and Biochemistry, University of Bath, Bath, UK
| | - Jimena Monzón-Sandoval
- School of Life Sciences, University of Lincoln, Lincoln, UK; Milner Centre for Evolution, Department of Biology and Biochemistry, University of Bath, Bath, UK
| | - Araxi O Urrutia
- Milner Centre for Evolution, Department of Biology and Biochemistry, University of Bath, Bath, UK; Instituto de Ecología, Universidad Nacional Autónoma de México, Ciudad de México, Mexico.
| | | |
Collapse
|
33
|
Wu S, Zhu W, Thompson P, Hannun YA. Evaluating intrinsic and non-intrinsic cancer risk factors. Nat Commun 2018; 9:3490. [PMID: 30154431 PMCID: PMC6113228 DOI: 10.1038/s41467-018-05467-z] [Citation(s) in RCA: 228] [Impact Index Per Article: 32.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2017] [Accepted: 06/25/2018] [Indexed: 12/12/2022] Open
Abstract
Discriminating the contribution of unmodifiable random intrinsic DNA replication errors ('bad luck') to cancer development from those of other factors is critical for understanding cancer in humans and for directing public resources aimed at reducing the burden of cancer. Here, we review and highlight the evidence that demonstrates cancer causation is multifactorial, and provide several important examples where modification of risk factors has achieved cancer prevention. Furthermore, we stress the need and opportunities to advance understanding of cancer aetiology through integration of interaction effects between risk factors when estimating the contribution of individual and joint factors to cancer burden in a population. We posit that non-intrinsic factors drive most cancer risk, and stress the need for cancer prevention.
Collapse
Affiliation(s)
- Song Wu
- Department of Applied Mathematics and Statistics, Stony Brook University, Stony Brook, NY, 11794, USA
- Stony Brook Cancer Centre, Stony Brook University, Health Sciences Centre, Stony Brook, NY, 11794, USA
| | - Wei Zhu
- Department of Applied Mathematics and Statistics, Stony Brook University, Stony Brook, NY, 11794, USA
- Stony Brook Cancer Centre, Stony Brook University, Health Sciences Centre, Stony Brook, NY, 11794, USA
| | - Patricia Thompson
- Stony Brook Cancer Centre, Stony Brook University, Health Sciences Centre, Stony Brook, NY, 11794, USA
- Department of Pathology, Stony Brook University, Health Sciences Centre, Stony Brook, NY, 11794, USA
| | - Yusuf A Hannun
- Stony Brook Cancer Centre, Stony Brook University, Health Sciences Centre, Stony Brook, NY, 11794, USA.
- Department of Medicine, Stony Brook University, Health Sciences Centre, Stony Brook, NY, 11794, USA.
| |
Collapse
|
34
|
Alencar-Silva T, Braga MC, Santana GOS, Saldanha-Araujo F, Pogue R, Dias SC, Franco OL, Carvalho JL. Breaking the frontiers of cosmetology with antimicrobial peptides. Biotechnol Adv 2018; 36:2019-2031. [PMID: 30118811 DOI: 10.1016/j.biotechadv.2018.08.005] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2018] [Revised: 07/26/2018] [Accepted: 08/12/2018] [Indexed: 01/06/2023]
Abstract
Antimicrobial peptides (AMPs) are mostly endogenous, cationic, amphipathic polypeptides, produced by many natural sources. Recently, many biological functions beyond antimicrobial activity have been attributed to AMPs, and some of these have attracted the attention of the cosmetics industry. AMPs have revealed antioxidant, self-renewal and pro-collagen effects, which are desirable in anti-aging cosmetics. Additionally, AMPs may also be customized to act on specific cellular targets. Here, we review the recent literature that highlights the many possibilities presented by AMPs, focusing on the relevance and impact that this potentially novel class of active cosmetic ingredients might have in the near future, creating new market outlooks for the cosmetic industry with these molecules as a viable alternative to conventional cosmetics.
Collapse
Affiliation(s)
- Thuany Alencar-Silva
- Programa de Pós-graduação em Ciências Genômicas e Biotecnologia, Universidade Católica de Brasília, Brasília, DF, Brazil
| | - Mariana Carolina Braga
- Programa de Pós-graduação em Ciências Genômicas e Biotecnologia, Universidade Católica de Brasília, Brasília, DF, Brazil
| | - Gustavo Oliveira Silva Santana
- Programa de Pós-graduação em Ciências Genômicas e Biotecnologia, Universidade Católica de Brasília, Brasília, DF, Brazil
| | - Felipe Saldanha-Araujo
- Laboratório de Farmacologia Molecular, Departamento de Ciências da Saúde, Universidade de Brasília, Brasilia, DF, Brazil; Programa de Pós-graduação em Patologia Molecular, Universidade de Brasília, Brasília, DF, Brazil
| | - Robert Pogue
- Programa de Pós-graduação em Ciências Genômicas e Biotecnologia, Universidade Católica de Brasília, Brasília, DF, Brazil
| | - Simoni Campos Dias
- Programa de Pós-graduação em Ciências Genômicas e Biotecnologia, Universidade Católica de Brasília, Brasília, DF, Brazil; Universidade de Brasília, Pós-Graduação em Biologia Animal, Campus Darcy Ribeiro, Brasília/DF, 70910-900, Brazil
| | - Octavio Luiz Franco
- Programa de Pós-graduação em Ciências Genômicas e Biotecnologia, Universidade Católica de Brasília, Brasília, DF, Brazil; S-Inova Biotech, Pós-graduação em Biotecnologia, Universidade Católica Dom Bosco, Campo Grande, MS, Brazil; Programa de Pós-graduação em Patologia Molecular, Universidade de Brasília, Brasília, DF, Brazil; Centro de Análises Proteômicas e Bioquímicas, Programa de Pós-Graduação em Ciências Genômicas e Biotecnologia, Universidade Católica de Brasília, Brasília-DF, Brazil
| | - Juliana Lott Carvalho
- Programa de Pós-graduação em Ciências Genômicas e Biotecnologia, Universidade Católica de Brasília, Brasília, DF, Brazil.
| |
Collapse
|
35
|
Obesity and type-2 diabetes as inducers of premature cellular senescence and ageing. Biogerontology 2018; 19:447-459. [PMID: 30054761 PMCID: PMC6223730 DOI: 10.1007/s10522-018-9763-7] [Citation(s) in RCA: 126] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2018] [Accepted: 07/21/2018] [Indexed: 12/13/2022]
Abstract
Cellular senescence is now considered as a major mechanism in the development and progression of various diseases and this may include metabolic diseases such as obesity and type-2 diabetes. The presence of obesity and diabetes is a major risk factor in the development of additional health conditions, such as cardiovascular disease, kidney disease and cancer. Since senescent cells can drive disease development, obesity and diabetes can potentially create an environment that accelerates cell senescence within other tissues of the body. This can consequently manifest as age-related biological impairments and secondary diseases. Cell senescence in cell types linked with obesity and diabetes, namely adipocytes and pancreatic beta cells will be explored, followed by a discussion on the role of obesity and diabetes in accelerating ageing through induction of premature cell senescence mediated by high glucose levels and oxidised low-density lipoproteins. Particular emphasis will be placed on accelerated cell senescence in endothelial progenitor cells, endothelial cells and vascular smooth muscle cells with relation to cardiovascular disease and proximal tubular cells with relation to kidney disease. A summary of the potential strategies for therapeutically targeting senescent cells for improving health is also presented.
Collapse
|
36
|
Burton DG, Stolzing A. Cellular senescence: Immunosurveillance and future immunotherapy. Ageing Res Rev 2018; 43:17-25. [PMID: 29427795 DOI: 10.1016/j.arr.2018.02.001] [Citation(s) in RCA: 157] [Impact Index Per Article: 22.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2018] [Accepted: 02/02/2018] [Indexed: 12/12/2022]
Abstract
In response to persistent DNA damage, induction into cell senescence promotes an immunogenic program which facilitates immune clearance of these damaged cells. Under physiological conditions, senescent cells can activate both innate and adaptive immune responses, functioning to maintain tissue homeostasis. In addition, emerging findings suggest that programmed induction of cell senescence may be important for regulating reproductive processes, partly facilitated by immune clearance. However, likely owing to ageing of the immune system, a failure to eliminate senescent cells can contribute to their persistence in tissues, leading to the development and progression of age-related diseases. Such immune failure may in part be due to activation of the senescence program in immune cells, leading to their dysfunction. Furthermore, senescent cells under certain biological contexts have been shown to instead promote immune suppression, a response that may reflect differences between an acute verses chronic senescent phenotype. In this review, we provide an overview of the research to date concerning senescence immunosurviellance, including a focused discussion on the mechanisms by which macrophages may recognise senescent cells. Senescence immunotherapy strategies as an alternative to senolytics for the removal of senescent cells will also be discussed.
Collapse
|
37
|
|
38
|
Sultana Z, Maiti K, Dedman L, Smith R. Is there a role for placental senescence in the genesis of obstetric complications and fetal growth restriction? Am J Obstet Gynecol 2018; 218:S762-S773. [PMID: 29275823 DOI: 10.1016/j.ajog.2017.11.567] [Citation(s) in RCA: 106] [Impact Index Per Article: 15.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2017] [Revised: 10/23/2017] [Accepted: 11/08/2017] [Indexed: 12/11/2022]
Abstract
The placenta ages as pregnancy advances, yet its continued function is required for a successful pregnancy outcome. Placental aging is a physiological phenomenon; however, there are some placentas that show signs of aging earlier than others. Premature placental senescence and aging are implicated in a number of adverse pregnancy outcomes, including fetal growth restriction, preeclampsia, spontaneous preterm birth, and intrauterine fetal death. Here we discuss cellular senescence, a state of terminal proliferation arrest, and how senescence is regulated. We also explore the role of physiological placental senescence and how aberrant placental senescence alters placental function, contributing to the pathophysiology of fetal growth restriction, preeclampsia, spontaneous preterm labor/birth, and unexplained fetal death.
Collapse
|
39
|
Miragem AA, Homem de Bittencourt PI. Nitric oxide-heat shock protein axis in menopausal hot flushes: neglected metabolic issues of chronic inflammatory diseases associated with deranged heat shock response. Hum Reprod Update 2018; 23:600-628. [PMID: 28903474 DOI: 10.1093/humupd/dmx020] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2016] [Accepted: 06/28/2017] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Although some unequivocal underlying mechanisms of menopausal hot flushes have been demonstrated in animal models, the paucity of similar approaches in humans impedes further mechanistic outcomes. Human studies might show some as yet unexpected physiological mechanisms of metabolic adaptation that permeate the phase of decreased oestrogen levels in both symptomatic and asymptomatic women. This is particularly relevant because both the severity and time span of hot flushes are associated with increased risk of chronic inflammatory disease. On the other hand, oestrogen induces the expression of heat shock proteins of the 70 kDa family (HSP70), which are anti-inflammatory and cytoprotective protein chaperones, whose expression is modulated by different types of physiologically stressful situations, including heat stress and exercise. Therefore, lower HSP70 expression secondary to oestrogen deficiency increases cardiovascular risk and predisposes the patient to senescence-associated secretory phenotype (SASP) that culminates in chronic inflammatory diseases, such as obesities, type 2 diabetes, neuromuscular and neurodegenerative diseases. OBJECTIVE AND RATIONALE This review focuses on HSP70 and its accompanying heat shock response (HSR), which is an anti-inflammatory and antisenescent pathway whose intracellular triggering is also oestrogen-dependent via nitric oxide (NO) production. The main goal of the manuscript was to show that the vasomotor symptoms that accompany hot flushes may be a disguised clue for important neuroendocrine alterations linking oestrogen deficiency to the anti-inflammatory HSR. SEARCH METHODS Results from our own group and recent evidence on hypothalamic control of central temperature guided a search on PubMed and Google Scholar websites. OUTCOMES Oestrogen elicits rapid production of the vasodilatory gas NO, a powerful activator of HSP70 expression. Whence, part of the protective effects of oestrogen over cardiovascular and neuroendocrine systems is tied to its capacity of inducing the NO-elicited HSR. The hypothalamic areas involved in thermoregulation (infundibular nucleus in humans and arcuate nucleus in other mammals) and whose neurons are known to have their function altered after long-term oestrogen ablation, particularly kisspeptin-neurokinin B-dynorphin neurons, (KNDy) are the same that drive neuroprotective expression of HSP70 and, in many cases, this response is via NO even in the absence of oestrogen. From thence, it is not illogical that hot flushes might be related to an evolutionary adaptation to re-equip the NO-HSP70 axis during the downfall of circulating oestrogen. WIDER IMPLICATIONS Understanding of HSR could shed light on yet uncovered mechanisms of menopause-associated diseases as well as on possible manipulation of HSR in menopausal women through physiological, pharmacological, nutraceutical and prebiotic interventions. Moreover, decreased HSR indices (that can be clinically determined with ease) in perimenopause could be of prognostic value in predicting the moment and appropriateness of starting a HRT.
Collapse
Affiliation(s)
- Antônio Azambuja Miragem
- Laboratory of Cellular Physiology, Department of Physiology, Federal University of Rio Grande do Sul, Rua Sarmento Leite 500, ICBS, 2nd Floor, Suite 350, Porto Alegre, RS 90050-170, Brazil.,Federal Institute of Education, Science and Technology 'Farroupilha', Rua Uruguai 1675, Santa Rosa, RS 98900-000, Brazil
| | - Paulo Ivo Homem de Bittencourt
- Laboratory of Cellular Physiology, Department of Physiology, Federal University of Rio Grande do Sul, Rua Sarmento Leite 500, ICBS, 2nd Floor, Suite 350, Porto Alegre, RS 90050-170, Brazil
| |
Collapse
|
40
|
Eldawud R, Wagner A, Dong C, Stueckle TA, Rojanasakul Y, Dinu CZ. Carbon nanotubes physicochemical properties influence the overall cellular behavior and fate. NANOIMPACT 2018; 9:72-84. [PMID: 31544167 PMCID: PMC6753956 DOI: 10.1016/j.impact.2017.10.006] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/29/2023]
Abstract
The unique properties of single walled carbon nanotubes (SWCNTs) make them viable candidates for versatile implementation in the next generation of biomedical devices for targeted delivery of chemotherapeutic agents or cellular-sensing probes. Such implementation requires user-tailored changes in SWCNT's physicochemical characteristics to allow for efficient cellular integration while maintaining nanotubes' functionality. However, isolated reports showed that user-tailoring could induce deleterious effects in exposed cells, from decrease in cellular proliferation, to changes in cellular adhesion, generation of reactive oxygen species or phenotypical variations, just to name a few. Before full implementation of SWCNTs is achieved, their toxicological profiles need to be mechanistically correlated with their physicochemical properties to determine how the induced cellular fate is related to the exposure conditions or samples' characteristics. Our study provides a comprehensive analysis of the synergistic cyto- and genotoxic effects resulted from short-term exposure of human lung epithelial cells to pristine (as manufactured) and user-tailored SWCNTs, as a function of their physicochemical properties. Specifically, through a systematic approach we are correlating the nanotube uptake and nanotube-induced cellular changes to the sample's physicochemical characteristics (e.g., metal impurities, length, agglomerate size, surface area, dispersion, and surface functionalization). By identifying changes in active hallmarks involved in cell-cell connections and maintaining epithelial layer integrity, we also determine the role that short-term exposure to SWCNTs plays in the overall cellular fate and cellular transformation. Lastly, we assess cellular structure-function relationships to identify non-apoptotic pathways induced by SWCNTs exposure that could however lead to changes in cellular behavior and cellular transformation. Our results show that the degree of cell transformation is a function of the physicochemical properties of the SWCNT, with the nanotube with higher length, higher metal content and larger agglomerate size reducing cell viability to a larger extent. Such changes in cell viability are also complemented by changes in cell structure, cycle and cell-cell interactions, all responsible for maintaining cell fate.
Collapse
Affiliation(s)
- Reem Eldawud
- Department of Chemical and Biomedical Engineering, West Virginia University, Morgantown, WV 26506, USA
| | - Alixandra Wagner
- Department of Chemical and Biomedical Engineering, West Virginia University, Morgantown, WV 26506, USA
| | - Chenbo Dong
- Department of Chemical and Biomedical Engineering, West Virginia University, Morgantown, WV 26506, USA
| | - Todd A. Stueckle
- Health Effects Laboratory Division, National Institute for Occupational Safety and Health, Morgantown, WV 26505, USA
| | - Yon Rojanasakul
- Department of Pharmaceutical Sciences, West Virginia University, WV 26506, USA
| | - Cerasela Zoica Dinu
- Department of Chemical and Biomedical Engineering, West Virginia University, Morgantown, WV 26506, USA
- Department of Pharmaceutical Sciences, West Virginia University, WV 26506, USA
| |
Collapse
|
41
|
Fougère B, Boulanger E, Nourhashémi F, Guyonnet S, Cesari M. Chronic Inflammation: Accelerator of Biological Aging. J Gerontol A Biol Sci Med Sci 2017; 72:1218-1225. [PMID: 28003373 DOI: 10.1093/gerona/glw240] [Citation(s) in RCA: 181] [Impact Index Per Article: 22.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2016] [Accepted: 11/09/2016] [Indexed: 12/13/2022] Open
Abstract
Biological aging is characterized by a chronic low-grade inflammation level. This chronic phenomenon has been named "inflamm-aging" and is a highly significant risk factor for morbidity and mortality in the older persons. The most common theories of inflamm-aging include redox stress, mitochondrial dysfunction, glycation, deregulation of the immune system, hormonal changes, epigenetic modifications, and dysfunction telomere attrition. Inflamm-aging plays a role in the initiation and progression of age-related diseases such as type II diabetes, Alzheimer's disease, cardiovascular disease, frailty, sarcopenia, osteoporosis, and cancer. This review will cover the identification of pathways that control age-related inflammation across multiple systems and its potential causal role in contributing to adverse health outcomes.
Collapse
Affiliation(s)
- Bertrand Fougère
- Gérontopôle, Centre Hospitalier Universitaire de Toulouse, Toulouse, France.,Inserm UMR1027, Université de Toulouse III Paul Sabatier, Toulouse, France
| | | | - Fati Nourhashémi
- Gérontopôle, Centre Hospitalier Universitaire de Toulouse, Toulouse, France.,Inserm UMR1027, Université de Toulouse III Paul Sabatier, Toulouse, France
| | - Sophie Guyonnet
- Gérontopôle, Centre Hospitalier Universitaire de Toulouse, Toulouse, France.,Inserm UMR1027, Université de Toulouse III Paul Sabatier, Toulouse, France
| | - Matteo Cesari
- Gérontopôle, Centre Hospitalier Universitaire de Toulouse, Toulouse, France.,Inserm UMR1027, Université de Toulouse III Paul Sabatier, Toulouse, France
| |
Collapse
|
42
|
Senthil KKJ, Gokila VM, Wang SY. Activation of Nrf2-mediated anti-oxidant genes by antrodin C prevents hyperglycemia-induced senescence and apoptosis in human endothelial cells. Oncotarget 2017; 8:96568-96587. [PMID: 29228553 PMCID: PMC5722505 DOI: 10.18632/oncotarget.19951] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2017] [Accepted: 07/25/2017] [Indexed: 02/06/2023] Open
Abstract
In the present study, we investigated the effects of antrodin C (ADC), a maleimide derivative isolated from mycelia of Antrodia cinnamomea, on high glucose (HG, 30 mM)-accelerated endothelial dysfunction in vitro. HG-induced cytotoxicity in human umbilical vein endothelial cells (HUVECs) was significantly ameliorated by ADC. In addition, treatment with ADC significantly prevented HG-induced senescence, growth arrest at the G1-S transition phase and apoptosis in HUVECs. Moreover, the increased level of intracellular reactive oxygen species (ROS) under HG condition was significantly ameliorated by ADC. Further analysis revealed that ADC-mediated anti-oxidant effects were due to up-regulation of cellular anti-oxidant genes, such as HO-1 and NQO-1 via promotion of the transcriptional activity of Nrf2, which was further confirmed by the failure of ADC to protect HUVECs from HG-induced dysfunction under HO-1 inhibition or Nrf2 silencing. Furthermore, hyperosmotic glucose (HOG, 60 mM)-induced uncontrolled production of ROS, rapid apoptotic cell death and HUVEC injury were significantly prevented by ADC, whereas these preventive effects were barely observed in HO-1 inhibited or Nrf2 silenced cells. Taken together, these results suggest that ADC may represent a promising intervention in diabetic-associated cardiovascular diseases by activating the Nrf2-dependent cellular anti-oxidant defense system.
Collapse
Affiliation(s)
- Kumar K J Senthil
- Department of Forestry, National Chung Hsing University, Taichung, Taiwan
| | - Vani M Gokila
- Department of Forestry, National Chung Hsing University, Taichung, Taiwan.,National Chung Hsing University/University of California at Davis, Plant and Food Biotechnology Center, National Chung Hsing University, Taichung, Taiwan
| | - Sheng-Yang Wang
- Department of Forestry, National Chung Hsing University, Taichung, Taiwan.,National Chung Hsing University/University of California at Davis, Plant and Food Biotechnology Center, National Chung Hsing University, Taichung, Taiwan.,Agricultural Biotechnology Research Center, Academia Sinica, Taipei, Taiwan
| |
Collapse
|
43
|
Abstract
Individuals of the same age may not age at the same rate. Quantitative biomarkers of aging are valuable tools to measure physiological age, assess the extent of ‘healthy aging’, and potentially predict health span and life span for an individual. Given the complex nature of the aging process, the biomarkers of aging are multilayered and multifaceted. Here, we review the phenotypic and molecular biomarkers of aging. Identifying and using biomarkers of aging to improve human health, prevent age-associated diseases, and extend healthy life span are now facilitated by the fast-growing capacity of multilevel cross-sectional and longitudinal data acquisition, storage, and analysis, particularly for data related to general human populations. Combined with artificial intelligence and machine learning techniques, reliable panels of biomarkers of aging will have tremendous potential to improve human health in aging societies.
Collapse
Affiliation(s)
- Xian Xia
- Key Laboratory of Computational Biology, CAS Center for Excellence in Molecular Cell Science, Collaborative Innovation Center for Genetics and Developmental Biology, Chinese Academy of Sciences-Max Planck Partner Institute for Computational Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Weiyang Chen
- School of Information, Qilu University of Technology, Jinan, China
| | - Joseph McDermott
- Key Laboratory of Computational Biology, CAS Center for Excellence in Molecular Cell Science, Collaborative Innovation Center for Genetics and Developmental Biology, Chinese Academy of Sciences-Max Planck Partner Institute for Computational Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Jing-Dong Jackie Han
- Key Laboratory of Computational Biology, CAS Center for Excellence in Molecular Cell Science, Collaborative Innovation Center for Genetics and Developmental Biology, Chinese Academy of Sciences-Max Planck Partner Institute for Computational Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
| |
Collapse
|
44
|
O'Hara SP, Splinter PL, Trussoni CE, Pisarello MJL, Loarca L, Splinter NS, Schutte BF, LaRusso NF. ETS Proto-oncogene 1 Transcriptionally Up-regulates the Cholangiocyte Senescence-associated Protein Cyclin-dependent Kinase Inhibitor 2A. J Biol Chem 2017; 292:4833-4846. [PMID: 28184004 PMCID: PMC5377799 DOI: 10.1074/jbc.m117.777409] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2017] [Revised: 02/06/2017] [Indexed: 12/13/2022] Open
Abstract
Primary sclerosing cholangitis (PSC) is a chronic, fibroinflammatory cholangiopathy (disease of the bile ducts) of unknown pathogenesis. We reported that cholangiocyte senescence features prominently in PSC and that neuroblastoma RAS viral oncogene homolog (NRAS) is activated in PSC cholangiocytes. Additionally, persistent microbial insult (e.g. LPSs) induces cyclin-dependent kinase inhibitor 2A (CDKN2A/p16INK4a) expression and senescence in cultured cholangiocytes in an NRAS-dependent manner. However, the molecular mechanisms involved in LPS-induced cholangiocyte senescence and NRAS-dependent regulation of CDKN2A remain unclear. Using our in vitro senescence model, we found that LPS-induced CDKN2A expression coincided with a 4.5-fold increase in ETS1 (ETS proto-oncogene 1) mRNA, suggesting that ETS1 is involved in regulating CDKN2A This idea was confirmed by RNAi-mediated suppression or genetic deletion of ETS1, which blocked CDKN2A expression and reduced cholangiocyte senescence. Furthermore, site-directed mutagenesis of a predicted ETS-binding site within the CDKN2A promoter abolished luciferase reporter activity. Pharmacological inhibition of RAS/MAPK reduced ETS1 and CDKN2A protein expression and CDKN2A promoter-driven luciferase activity by ∼50%. In contrast, constitutively active NRAS expression induced ETS1 and CDKN2A protein expression, whereas ETS1 RNAi blocked this increase. Chromatin immunoprecipitation-PCR detected increased ETS1 and histone 3 lysine 4 trimethylation (H3K4Me3) at the CDKN2A promoter following LPS-induced senescence. Additionally, phospho-ETS1 expression was increased in cholangiocytes of human PSC livers and in the Abcb4 (Mdr2)-/- mouse model of PSC. These data pinpoint ETS1 and H3K4Me3 as key transcriptional regulators in NRAS-induced expression of CDKN2A, and this regulatory axis may therefore represent a potential therapeutic target for PSC treatment.
Collapse
Affiliation(s)
- Steven P O'Hara
- From the Division of Gastroenterology and Hepatology, and the Mayo Clinic Center for Cell Signaling in Gastroenterology, Mayo Clinic, Rochester, Minnesota 55905
| | - Patrick L Splinter
- From the Division of Gastroenterology and Hepatology, and the Mayo Clinic Center for Cell Signaling in Gastroenterology, Mayo Clinic, Rochester, Minnesota 55905
| | - Christy E Trussoni
- From the Division of Gastroenterology and Hepatology, and the Mayo Clinic Center for Cell Signaling in Gastroenterology, Mayo Clinic, Rochester, Minnesota 55905
| | - Maria J Lorenzo Pisarello
- From the Division of Gastroenterology and Hepatology, and the Mayo Clinic Center for Cell Signaling in Gastroenterology, Mayo Clinic, Rochester, Minnesota 55905
| | - Lorena Loarca
- From the Division of Gastroenterology and Hepatology, and the Mayo Clinic Center for Cell Signaling in Gastroenterology, Mayo Clinic, Rochester, Minnesota 55905
| | - Noah S Splinter
- From the Division of Gastroenterology and Hepatology, and the Mayo Clinic Center for Cell Signaling in Gastroenterology, Mayo Clinic, Rochester, Minnesota 55905
| | - Bryce F Schutte
- From the Division of Gastroenterology and Hepatology, and the Mayo Clinic Center for Cell Signaling in Gastroenterology, Mayo Clinic, Rochester, Minnesota 55905
| | - Nicholas F LaRusso
- From the Division of Gastroenterology and Hepatology, and the Mayo Clinic Center for Cell Signaling in Gastroenterology, Mayo Clinic, Rochester, Minnesota 55905
| |
Collapse
|
45
|
Sultana Z, Maiti K, Aitken J, Morris J, Dedman L, Smith R. Oxidative stress, placental ageing-related pathologies and adverse pregnancy outcomes. Am J Reprod Immunol 2017; 77. [PMID: 28240397 DOI: 10.1111/aji.12653] [Citation(s) in RCA: 182] [Impact Index Per Article: 22.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2016] [Accepted: 01/20/2017] [Indexed: 12/22/2022] Open
Abstract
Oxidative stress (OS), an imbalance between free radical generation and antioxidant defence, is recognized as a key factor in the pathogenesis of adverse pregnancy outcomes. Although OS is a common future of normal pregnancy, persistent, overwhelming OS leads to consumption and decline of antioxidants, affecting placental antioxidant capacity and reducing systems. The accumulation of OS causes damage to lipids, proteins and DNA in the placental tissue that induces a form of accelerated ageing. Premature ageing of the placenta is associated with placental insufficiency that prevents the organ meeting the needs of the foetus, and as a consequence, the viability of the foetus is compromised. This review summarizes the literature regarding the role of OS and premature placental ageing in the pathophysiology of pregnancy complications.
Collapse
Affiliation(s)
- Zakia Sultana
- Mothers and Babies Research Centre, Hunter Medical Research Institute, Newcastle, NSW, Australia.,Faculty of Health and Medicine, School of Medicine and Public Health, University of Newcastle, Newcastle, NSW, Australia.,Priority Research Centre for Reproductive Science, University of Newcastle, Newcastle, NSW, Australia
| | - Kaushik Maiti
- Mothers and Babies Research Centre, Hunter Medical Research Institute, Newcastle, NSW, Australia.,Faculty of Health and Medicine, School of Medicine and Public Health, University of Newcastle, Newcastle, NSW, Australia.,Priority Research Centre for Reproductive Science, University of Newcastle, Newcastle, NSW, Australia
| | - John Aitken
- Priority Research Centre for Reproductive Science, University of Newcastle, Newcastle, NSW, Australia
| | - Jonathan Morris
- Kolling Institute, Royal North Shore Hospital, University of Sydney, Sydney, NSW, Australia
| | - Lee Dedman
- Faculty of Science and Information Technology, School of Design, Communication and Information Technology, University of Newcastle, Newcastle, NSW, Australia
| | - Roger Smith
- Mothers and Babies Research Centre, Hunter Medical Research Institute, Newcastle, NSW, Australia.,Faculty of Health and Medicine, School of Medicine and Public Health, University of Newcastle, Newcastle, NSW, Australia.,Priority Research Centre for Reproductive Science, University of Newcastle, Newcastle, NSW, Australia
| |
Collapse
|
46
|
Serum from Varicose Patients Induces Senescence-Related Dysfunction of Vascular Endothelium Generating Local and Systemic Proinflammatory Conditions. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2016; 2016:2069290. [PMID: 27994710 PMCID: PMC5141312 DOI: 10.1155/2016/2069290] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/16/2016] [Revised: 10/25/2016] [Accepted: 11/01/2016] [Indexed: 11/17/2022]
Abstract
Although the role of endothelium in varicose vein development is indisputable, the effect of the pathology on biological properties of endothelial cells remains unclear. Here we examined if the presence of varicose veins affects senescence of endothelial cells (HUVECs) and, if so, what will be the local and systemic outcome of this effect. Experiments showed that HUVECs subjected to serum from varicose patients display improved proliferation, increased expression of senescence marker, SA-β-Gal, and increased generation of reactive oxygen species (ROS), as compared with serum from healthy donors. Both increased SA-β-Gal activity and ROS release were mediated by TGF-β1, the concentration of which in varicose serum was elevated and the activity of which in vitro was prevented using specific neutralizing antibody. Senescent HUVECs exposed to varicose serum generated increased amounts of ICAM-1, VCAM-1, P-selectin, uPA, PAI-1, and ET-1. Direct comparison of sera from varicose and healthy donors showed that pathological serum contained increased level of ICAM-1, VCAM-1, P-selectin, uPA, and ET-1. Calendar age of healthy subjects correlated positively with serum uPA and negatively with P-selectin. Age of varicose patients correlated positively with ICAM-1, VCAM-1, and ET-1. Collectively, our findings indicate that the presence of varicose veins causes a senescence-related dysfunction of vascular endothelium, which leads to the development of local and systemic proinflammatory environment.
Collapse
|
47
|
Tabibian JH, O’Hara SP, Trussoni CE, Tietz PS, Splinter PL, Mounajjed T, Hagey LR, LaRusso NF. Absence of the intestinal microbiota exacerbates hepatobiliary disease in a murine model of primary sclerosing cholangitis. Hepatology 2016; 63:185-196. [PMID: 26044703 PMCID: PMC4670294 DOI: 10.1002/hep.27927] [Citation(s) in RCA: 158] [Impact Index Per Article: 17.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/04/2014] [Accepted: 07/01/2015] [Indexed: 02/06/2023]
Abstract
UNLABELLED Primary sclerosing cholangitis (PSC) is a chronic, idiopathic, fibroinflammatory cholangiopathy. The role of the microbiota in PSC etiopathogenesis may be fundamentally important, yet remains obscure. We tested the hypothesis that germ-free (GF) mutltidrug resistance 2 knockout (mdr2(-/-) ) mice develop a distinct PSC phenotype, compared to conventionally housed (CV) mdr2(-/-) mice. Mdr2(-/-) mice (n = 12) were rederived as GF by embryo transfer, maintained in isolators, and sacrificed at 60 days in parallel with age-matched CV mdr2(-/-) mice. Serum biochemistries, gallbladder bile acids, and liver sections were examined. Histological findings were validated morphometrically, biochemically, and by immunofluorescence microscopy (IFM). Cholangiocyte senescence was assessed by p16(INK4a) in situ hybridization in liver tissue and by senescence-associated β-galactosidase staining in a culture-based model of insult-induced senescence. Serum biochemistries, including alkaline phosphatase, aspartate aminotransferase, and bilirubin, were significantly higher in GF mdr2(-/-) (P < 0.01). Primary bile acids were similar, whereas secondary bile acids were absent, in GF mdr2(-/-) mice. Fibrosis, ductular reaction, and ductopenia were significantly more severe histopathologically in GF mdr2(-/-) mice (P < 0.01) and were confirmed by hepatic morphometry, hydroxyproline assay, and IFM. Cholangiocyte senescence was significantly increased in GF mdr2(-/-) mice and abrogated in vitro by ursodeoxycholic acid (UDCA) treatment. CONCLUSIONS GF mdr2(-/-) mice exhibit exacerbated biochemical and histological features of PSC and increased cholangiocyte senescence, a characteristic and potential mediator of progressive biliary disease. UDCA, a commensal microbial metabolite, abrogates senescence in vitro. These findings demonstrate the importance of the commensal microbiota and its metabolites in protecting against biliary injury and suggest avenues for future studies of biomarkers and therapeutic interventions in PSC.
Collapse
Affiliation(s)
- James H. Tabibian
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester
- Center for Cell Signaling in Gastroenterology, Mayo Clinic, Rochester
| | - Steven P. O’Hara
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester
- Center for Cell Signaling in Gastroenterology, Mayo Clinic, Rochester
| | - Christy E. Trussoni
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester
- Center for Cell Signaling in Gastroenterology, Mayo Clinic, Rochester
| | - Pamela S. Tietz
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester
- Center for Cell Signaling in Gastroenterology, Mayo Clinic, Rochester
| | - Patrick L. Splinter
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester
- Center for Cell Signaling in Gastroenterology, Mayo Clinic, Rochester
| | | | - Lee R. Hagey
- Division of Gastroenterology, University of California, San Diego, La Jolla
| | - Nicholas F. LaRusso
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester
- Center for Cell Signaling in Gastroenterology, Mayo Clinic, Rochester
| |
Collapse
|
48
|
Mialet-Perez J, Douin-Echinard V, Cussac D, Bril A, Parini A. [Ageing: a matter of heart?]. Med Sci (Paris) 2015; 31:1006-13. [PMID: 26576608 DOI: 10.1051/medsci/20153111015] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Ageing is considered as a major risk factor for the development of chronic diseases. Among these, heart failure seems to be particularly important for both triggering and accelerating pathological ageing. In the present review, we give a general overview of the most relevant results concerning the mechanism of normal and premature senescence of cardiomyocytes and cardiac stromal cells. In particular, we will address the role of telomere dysfunction, DNA damage response, impairment of mitochondrial function, miRNAs and secretome of senescent cells in cardiac ageing and failure.
Collapse
Affiliation(s)
- Jeanne Mialet-Perez
- Inserm, UMR1048, institut des maladies métaboliques et cardiovasculaires, 1, avenue Jean Poulhès, BP 84225, 31432 Toulouse cedex 4, France - Université Paul Sabatier, Toulouse, France
| | - Victorine Douin-Echinard
- Inserm, UMR1048, institut des maladies métaboliques et cardiovasculaires, 1, avenue Jean Poulhès, BP 84225, 31432 Toulouse cedex 4, France - Université Paul Sabatier, Toulouse, France
| | - Daniel Cussac
- Inserm, UMR1048, institut des maladies métaboliques et cardiovasculaires, 1, avenue Jean Poulhès, BP 84225, 31432 Toulouse cedex 4, France - Université Paul Sabatier, Toulouse, France
| | - Antoine Bril
- Institut de recherches internationales Servier, Suresnes, France
| | - Angelo Parini
- Inserm, UMR1048, institut des maladies métaboliques et cardiovasculaires, 1, avenue Jean Poulhès, BP 84225, 31432 Toulouse cedex 4, France - Université Paul Sabatier, Toulouse, France
| |
Collapse
|
49
|
Manoli P, Antoniou A, Bashiardes E, Xenophontos S, Photiades M, Stribley V, Mylona M, Demetriou C, Cariolou MA. Sex-specific age association with primary DNA transfer. Int J Legal Med 2015; 130:103-12. [DOI: 10.1007/s00414-015-1291-2] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2015] [Accepted: 11/09/2015] [Indexed: 11/29/2022]
|
50
|
Hyperphosphatemia induces cellular senescence in human aorta smooth muscle cells through integrin linked kinase (ILK) up-regulation. Mech Ageing Dev 2015; 152:43-55. [PMID: 26467393 DOI: 10.1016/j.mad.2015.10.001] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2015] [Revised: 09/22/2015] [Accepted: 10/05/2015] [Indexed: 12/13/2022]
Abstract
Aging is conditioned by genetic and environmental factors. Hyperphosphatemia is related to some pathologies, affecting to vascular cells behavior. This work analyze whether high concentration of extracellular phosphate induces vascular smooth muscle cells senescence, exploring the intracellular mechanisms and highlighting the in vivo relevance of this phenomenon. Human aortic smooth muscle cells treated with β-Glycerophosphate (BGP, 10mM) suffered cellular senescence by increasing p53, p21 and p16 expression and the senescence associated β-galactosidase activity. In parallel, BGP induced ILK overexpression, dependent on the IGF-1 receptor activation, and oxidative stress. Down-regulating ILK expression prevented BGP-induced senescence and oxidative stress. Aortic rings from young rats treated with 10mM BGP for 48h, showed increased p53, p16 and ILK expression and SA-β-gal activity. Seven/eight nephrectomized rats feeding a hyperphosphatemic diet and fifteenth- month old mice showed hyperphosphatemia and aortic ILK, p53 and p16 expression. In conclusion, we demonstrated that high extracellular concentration of phosphate induced senescence in cultured smooth muscle through the activation of IGF-1 receptor and ILK overexpression and provided solid evidences for the in vivo relevance of these results since aged animals showed high levels of serum phosphate linked to increased expression of ILK and senescence genes.
Collapse
|