101
|
Lorenzo EC, Torrance BL, Haynes L. Impact of senolytic treatment on immunity, aging, and disease. FRONTIERS IN AGING 2023; 4:1161799. [PMID: 37886012 PMCID: PMC10598643 DOI: 10.3389/fragi.2023.1161799] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/08/2023] [Accepted: 07/31/2023] [Indexed: 10/28/2023]
Abstract
Cellular senescence has been implicated in the pathophysiology of many age-related diseases. However, it also plays an important protective role in the context of tumor suppression and wound healing. Reducing senescence burden through treatment with senolytic drugs or the use of genetically targeted models of senescent cell elimination in animals has shown positive results in the context of mitigating disease and age-associated inflammation. Despite positive, albeit heterogenous, outcomes in clinical trials, very little is known about the short-term and long-term immunological consequences of using senolytics as a treatment for age-related conditions. Further, many studies examining cellular senescence and senolytic treatment have been demonstrated in non-infectious disease models. Several recent reports suggest that senescent cell elimination may have benefits in COVID-19 and influenza resolution and disease prognosis. In this review, we discuss the current clinical trials and pre-clinical studies that are exploring the impact of senolytics on cellular immunity. We propose that while eliminating senescent cells may have an acute beneficial impact on primary immune responses, immunological memory may be negatively impacted. Closer investigation of senolytics on immune function and memory generation would provide insight as to whether senolytics could be used to enhance the aging immune system and have potential to be used as therapeutics or prophylactics in populations that are severely and disproportionately affected by infections such as the elderly and immunocompromised.
Collapse
Affiliation(s)
- Erica C. Lorenzo
- UConn Health Center on Aging, University of Connecticut School of Medicine, Farmington, CT, United States
| | - Blake L. Torrance
- UConn Health Center on Aging, University of Connecticut School of Medicine, Farmington, CT, United States
- Department of Immunology, University of Connecticut School of Medicine, Farmington, CT, United States
| | - Laura Haynes
- UConn Health Center on Aging, University of Connecticut School of Medicine, Farmington, CT, United States
- Department of Immunology, University of Connecticut School of Medicine, Farmington, CT, United States
| |
Collapse
|
102
|
Zhang H, Zhou H, Shen X, Lin X, Zhang Y, Sun Y, Zhou Y, Zhang L, Zhang D. The role of cellular senescence in metabolic diseases and the potential for senotherapeutic interventions. Front Cell Dev Biol 2023; 11:1276707. [PMID: 37868908 PMCID: PMC10587568 DOI: 10.3389/fcell.2023.1276707] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2023] [Accepted: 09/18/2023] [Indexed: 10/24/2023] Open
Abstract
Cellular senescence represents an irreversible state of cell cycle arrest induced by various stimuli strongly associated with aging and several chronic ailments. In recent years, studies have increasingly suggested that the accumulation of senescent cells is an important contributor to the decline of organ metabolism, ultimately resulting in metabolic diseases. Conversely, the elimination of senescent cells can alleviate or postpone the onset and progression of metabolic diseases. Thus, a close relationship between senescent cells and metabolic diseases is found, and targeting senescent cells has emerged as an alternative therapy for the treatment of metabolic diseases. In this review, we summarize the role of cellular senescence in metabolic diseases, explore relevant therapeutic strategies for metabolic diseases by removing senescent cells, and provide new insights into the treatment of metabolic diseases.
Collapse
Affiliation(s)
- Huantong Zhang
- School of Medicine, Hangzhou City University, Hangzhou, China
| | - Han Zhou
- School of Medicine, Hangzhou City University, Hangzhou, China
| | - Xin Shen
- School of Medicine, Hangzhou City University, Hangzhou, China
| | - Xingchen Lin
- School of Medicine, Hangzhou City University, Hangzhou, China
| | - Yuke Zhang
- School of Medicine, Hangzhou City University, Hangzhou, China
| | - Yiyi Sun
- School of Medicine, Hangzhou City University, Hangzhou, China
| | - Yi Zhou
- School of Medicine, Hangzhou City University, Hangzhou, China
| | - Lei Zhang
- School of Economy and Management, Zhejiang Sci-Tech University, Hangzhou, China
- Taizhou Hospital of Zhejiang Province, Zhejiang University, Taizhou, China
| | - Dayong Zhang
- School of Medicine, Hangzhou City University, Hangzhou, China
| |
Collapse
|
103
|
Gulej R, Nyúl-Tóth Á, Ahire C, DelFavero J, Balasubramanian P, Kiss T, Tarantini S, Benyo Z, Pacher P, Csik B, Yabluchanskiy A, Mukli P, Kuan-Celarier A, Krizbai IA, Campisi J, Sonntag WE, Csiszar A, Ungvari Z. Elimination of senescent cells by treatment with Navitoclax/ABT263 reverses whole brain irradiation-induced blood-brain barrier disruption in the mouse brain. GeroScience 2023; 45:2983-3002. [PMID: 37642933 PMCID: PMC10643778 DOI: 10.1007/s11357-023-00870-x] [Citation(s) in RCA: 30] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2023] [Accepted: 07/06/2023] [Indexed: 08/31/2023] Open
Abstract
Whole brain irradiation (WBI), a commonly employed therapy for multiple brain metastases and as a prophylactic measure after cerebral metastasis resection, is associated with a progressive decline in neurocognitive function, significantly impacting the quality of life for approximately half of the surviving patients. Recent preclinical investigations have shed light on the multifaceted cerebrovascular injury mechanisms underlying this side effect of WBI. In this study, we aimed to test the hypothesis that WBI induces endothelial senescence, contributing to chronic disruption of the blood-brain barrier (BBB) and microvascular rarefaction. To accomplish this, we utilized transgenic p16-3MR mice, which enable the identification and selective elimination of senescent cells. These mice were subjected to a clinically relevant fractionated WBI protocol (5 Gy twice weekly for 4 weeks), and cranial windows were applied to both WBI-treated and control mice. Quantitative assessment of BBB permeability and capillary density was performed using two-photon microscopy at the 6-month post-irradiation time point. The presence of senescent microvascular endothelial cells was assessed by imaging flow cytometry, immunolabeling, and single-cell RNA-sequencing (scRNA-seq). WBI induced endothelial senescence, which associated with chronic BBB disruption and a trend for decreased microvascular density in the mouse cortex. In order to investigate the cause-and-effect relationship between WBI-induced senescence and microvascular injury, senescent cells were selectively removed from animals subjected to WBI treatment using Navitoclax/ABT263, a well-known senolytic drug. This intervention was carried out at the 3-month post-WBI time point. In WBI-treated mice, Navitoclax/ABT263 effectively eliminated senescent endothelial cells, which was associated with decreased BBB permeability and a trend for increased cortical capillarization. Our findings provide additional preclinical evidence that senolytic treatment approaches may be developed for prevention of the side effects of WBI.
Collapse
Affiliation(s)
- Rafal Gulej
- Vascular Cognitive Impairment, Neurodegeneration and Healthy Brain Aging Program, Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- International Training Program in Geroscience, Doctoral School of Basic and Translational Medicine/Department of Public Health, Semmelweis University, Budapest, Hungary
| | - Ádám Nyúl-Tóth
- Vascular Cognitive Impairment, Neurodegeneration and Healthy Brain Aging Program, Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- International Training Program in Geroscience, Doctoral School of Basic and Translational Medicine/Department of Public Health, Semmelweis University, Budapest, Hungary
- International Training Program in Geroscience, Institute of Biophysics, Biological Research Centre, Eötvös Loránd Research Network, Szeged, Hungary
| | - Chetan Ahire
- Vascular Cognitive Impairment, Neurodegeneration and Healthy Brain Aging Program, Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Jordan DelFavero
- Vascular Cognitive Impairment, Neurodegeneration and Healthy Brain Aging Program, Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Priya Balasubramanian
- Vascular Cognitive Impairment, Neurodegeneration and Healthy Brain Aging Program, Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Stephenson Cancer Center, University of Oklahoma, Oklahoma City, OK, USA
| | - Tamas Kiss
- Vascular Cognitive Impairment, Neurodegeneration and Healthy Brain Aging Program, Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- International Training Program in Geroscience, First Department of Pediatrics, Semmelweis University, Budapest, Hungary
- Eötvös Loránd Research Network and Semmelweis University (ELKH-SE) Cerebrovascular and Neurocognitive Disorders Research Group, Budapest, Hungary
- International Training Program in Geroscience, Doctoral School of Basic and Translational Medicine/Department of Translational Medicine, Semmelweis University, Budapest, Hungary
- Graduate School, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Stefano Tarantini
- Vascular Cognitive Impairment, Neurodegeneration and Healthy Brain Aging Program, Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- International Training Program in Geroscience, Doctoral School of Basic and Translational Medicine/Department of Public Health, Semmelweis University, Budapest, Hungary
- Stephenson Cancer Center, University of Oklahoma, Oklahoma City, OK, USA
| | - Zoltan Benyo
- Eötvös Loránd Research Network and Semmelweis University (ELKH-SE) Cerebrovascular and Neurocognitive Disorders Research Group, Budapest, Hungary
- International Training Program in Geroscience, Doctoral School of Basic and Translational Medicine/Department of Translational Medicine, Semmelweis University, Budapest, Hungary
- Graduate School, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Pal Pacher
- Laboratory of Cardiovascular Physiology and Tissue Injury, National Institute On Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD, USA
| | - Boglarka Csik
- Vascular Cognitive Impairment, Neurodegeneration and Healthy Brain Aging Program, Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- International Training Program in Geroscience, Doctoral School of Basic and Translational Medicine/Department of Public Health, Semmelweis University, Budapest, Hungary
| | - Andriy Yabluchanskiy
- Vascular Cognitive Impairment, Neurodegeneration and Healthy Brain Aging Program, Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- International Training Program in Geroscience, Doctoral School of Basic and Translational Medicine/Department of Public Health, Semmelweis University, Budapest, Hungary
| | - Peter Mukli
- Vascular Cognitive Impairment, Neurodegeneration and Healthy Brain Aging Program, Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- International Training Program in Geroscience, Doctoral School of Basic and Translational Medicine/Department of Public Health, Semmelweis University, Budapest, Hungary
| | - Anna Kuan-Celarier
- Vascular Cognitive Impairment, Neurodegeneration and Healthy Brain Aging Program, Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - István A Krizbai
- International Training Program in Geroscience, Institute of Biophysics, Biological Research Centre, Eötvös Loránd Research Network, Szeged, Hungary
- Institute of Life Sciences, Vasile Goldiş Western University of Arad, Arad, Romania
| | | | - William E Sonntag
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Anna Csiszar
- Vascular Cognitive Impairment, Neurodegeneration and Healthy Brain Aging Program, Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Stephenson Cancer Center, University of Oklahoma, Oklahoma City, OK, USA
- International Training Program in Geroscience, Doctoral School of Basic and Translational Medicine/Department of Translational Medicine, Semmelweis University, Budapest, Hungary
| | - Zoltan Ungvari
- Vascular Cognitive Impairment, Neurodegeneration and Healthy Brain Aging Program, Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA.
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA.
- International Training Program in Geroscience, Doctoral School of Basic and Translational Medicine/Department of Public Health, Semmelweis University, Budapest, Hungary.
- Stephenson Cancer Center, University of Oklahoma, Oklahoma City, OK, USA.
| |
Collapse
|
104
|
Evenepoel P, Stenvinkel P, Shanahan C, Pacifici R. Inflammation and gut dysbiosis as drivers of CKD-MBD. Nat Rev Nephrol 2023; 19:646-657. [PMID: 37488276 DOI: 10.1038/s41581-023-00736-7] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/19/2023] [Indexed: 07/26/2023]
Abstract
Two decades ago, Kidney Disease: Improving Global Outcomes coined the term chronic kidney disease-mineral and bone disorder (CKD-MBD) to describe the syndrome of biochemical, bone and extra-skeletal calcification abnormalities that occur in patients with CKD. CKD-MBD is a prevalent complication and contributes to the excessively high burden of fractures and cardiovascular disease, loss of quality of life and premature mortality in patients with CKD. Thus far, therapy has focused primarily on phosphate retention, abnormal vitamin D metabolism and parathyroid hormone disturbances, but these strategies have largely proved unsuccessful, thus calling for paradigm-shifting concepts and innovative therapeutic approaches. Interorgan crosstalk is increasingly acknowledged to have an important role in health and disease. Accordingly, mounting evidence suggests a role for both the immune system and the gut microbiome in bone and vascular biology. Gut dysbiosis, compromised gut epithelial barrier and immune cell dysfunction are prominent features of the uraemic milieu. These alterations might contribute to the inflammatory state observed in CKD and could have a central role in the pathogenesis of CKD-MBD. The emerging fields of osteoimmunology and osteomicrobiology add another level of complexity to the pathogenesis of CKD-MBD, but also create novel therapeutic opportunities.
Collapse
Affiliation(s)
- Pieter Evenepoel
- Laboratory of Nephrology, Department of Microbiology, Immunology, and Transplantation, KU Leuven, Herestraat, Leuven, Belgium.
| | - Peter Stenvinkel
- Department of Renal Medicine M99, Karolinska University Hospital, Stockholm, Sweden
| | - Catherine Shanahan
- British Heart Foundation Centre of Excellence, School of Cardiovascular and Metabolic Medicine and Sciences, King's College London, London, UK
| | - Roberto Pacifici
- Division of Endocrinology, Metabolism and Lipids, Department of Medicine, Emory Microbiome Research Center, and Immunology and Molecular Pathogenesis Program, Emory University, Atlanta, GA, USA
| |
Collapse
|
105
|
Fang Y, Medina D, Stockwell R, McFadden S, Quinn K, Peck MR, Bartke A, Hascup KN, Hascup ER. Sexual dimorphic metabolic and cognitive responses of C57BL/6 mice to Fisetin or Dasatinib and quercetin cocktail oral treatment. GeroScience 2023; 45:2835-2850. [PMID: 37296266 PMCID: PMC10643448 DOI: 10.1007/s11357-023-00843-0] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Accepted: 05/23/2023] [Indexed: 06/12/2023] Open
Abstract
Senolytic treatment in aged mice clears senescent cell burden leading to functional improvements. However, less is known regarding the effects of these compounds when administered prior to significant senescent cell accumulation. From 4-13 months of age, C57BL/6 male and female mice received monthly oral dosing of either 100 mg/kg Fisetin or a 5 mg/kg Dasatinib (D) plus 50 mg/kg Quercetin (Q) cocktail. During treatment, several aspects of healthy aging were assayed including glucose metabolism using an insulin and glucose tolerance test, cognitive performance using Morris water maze and novel object recognition, and energy metabolism using indirect calorimetry. Afterwards, mice were euthanized for plasma, tissue specific markers of senescence-associated secretory phenotype (SASP), and white adipose tissue accumulation (WAT). Sexually dimorphic treatment effects were observed. Fisetin treated male mice had reduced SASP, enhanced glucose and energy metabolism, improved cognitive performance, and increased mRNA expression of adiponectin receptor 1 and glucose transporter 4. D + Q treatment had minimal effects in male C57BL/6 mice, but was detrimental to females causing increased SASP expression along with accumulation of WAT depots. Reduced energy metabolism and cognitive performance were also noted. Fisetin treatment had no effect in female C57BL/6 mice potentially due to a slower rate of biological aging. In summary, the senolytic treatment in young adulthood, has beneficial, negligible, or detrimental effects in C57BL/6 mice dependent upon sex and treatment. These observations should serve as a note of caution in this rapidly evolving and expanding field of investigation. Male and female C57BL/6 mice were treated with once monthly oral doses of either Dasatinib (D) + Quercetin (Q) or Fisetin from 4-13 months of age. Males treated with Fisetin had reduced SASP markers (blue spheres) as well as improved metabolism (red flame) and cognition. Females treated with D + Q had increased adiposity and SASP markers (red spheres) along with decreased metabolism (blue flame) and cognitive performance. No effects were observed in females treated with Fisetin or males treated with D + Q.
Collapse
Affiliation(s)
- Yimin Fang
- Department of Neurology, Dale and Deborah Smith Center for Alzheimer's Research and Treatment, Neuroscience Institute, Southern Illinois University School of Medicine, Springfield, IL, 62702, USA
| | - David Medina
- Department of Internal Medicine, Southern Illinois University School of Medicine, Springfield, IL, 62702, USA
| | - Robert Stockwell
- Department of Internal Medicine, Southern Illinois University School of Medicine, Springfield, IL, 62702, USA
| | - Sam McFadden
- Department of Neurology, Dale and Deborah Smith Center for Alzheimer's Research and Treatment, Neuroscience Institute, Southern Illinois University School of Medicine, Springfield, IL, 62702, USA
| | - Kathleen Quinn
- Department of Neurology, Dale and Deborah Smith Center for Alzheimer's Research and Treatment, Neuroscience Institute, Southern Illinois University School of Medicine, Springfield, IL, 62702, USA
| | - Mackenzie R Peck
- Department of Neurology, Dale and Deborah Smith Center for Alzheimer's Research and Treatment, Neuroscience Institute, Southern Illinois University School of Medicine, Springfield, IL, 62702, USA
| | - Andrzej Bartke
- Department of Internal Medicine, Southern Illinois University School of Medicine, Springfield, IL, 62702, USA
- Department of Medical Microbiology, Immunology and Cell Biology, Southern Illinois University School of Medicine, Springfield, IL, 62702, USA
| | - Kevin N Hascup
- Department of Neurology, Dale and Deborah Smith Center for Alzheimer's Research and Treatment, Neuroscience Institute, Southern Illinois University School of Medicine, Springfield, IL, 62702, USA
- Department of Medical Microbiology, Immunology and Cell Biology, Southern Illinois University School of Medicine, Springfield, IL, 62702, USA
- Department of Pharmacology, Southern Illinois University School of Medicine, Springfield, IL, 62702, USA
| | - Erin R Hascup
- Department of Neurology, Dale and Deborah Smith Center for Alzheimer's Research and Treatment, Neuroscience Institute, Southern Illinois University School of Medicine, Springfield, IL, 62702, USA.
- Department of Pharmacology, Southern Illinois University School of Medicine, Springfield, IL, 62702, USA.
| |
Collapse
|
106
|
Clayton ZS, Rossman MJ, Mahoney SA, Venkatasubramanian R, Maurer GS, Hutton DA, VanDongen NS, Greenberg NT, Longtine AG, Ludwig KR, Brunt VE, LaRocca TJ, Campisi J, Melov S, Seals DR. Cellular Senescence Contributes to Large Elastic Artery Stiffening and Endothelial Dysfunction With Aging: Amelioration With Senolytic Treatment. Hypertension 2023; 80:2072-2087. [PMID: 37593877 PMCID: PMC10530538 DOI: 10.1161/hypertensionaha.123.21392] [Citation(s) in RCA: 34] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Accepted: 08/02/2023] [Indexed: 08/19/2023]
Abstract
BACKGROUND Here, we assessed the role of cellular senescence and the senescence associated secretory phenotype (SASP) in age-related aortic stiffening and endothelial dysfunction. METHODS We studied young (6-8 mo) and old (27-29 mo) p16-3MR mice, which allows for genetic-based clearance of senescent cells with ganciclovir (GCV). We also treated old C57BL/6N mice with the senolytic ABT-263. RESULTS In old mice, GCV reduced aortic stiffness assessed by aortic pulse wave velocity (PWV; 477±10 vs. 382±7 cm/s, P<0.05) to young levels (old-GCV vs. young-vehicle, P=0.35); ABT-263 also reduced aortic PWV in old mice (446±9 to 356±11 cm/s, P<0.05). Aortic adventitial collagen was reduced by GCV (P<0.05) and ABT-263 (P=0.12) in old mice. To show an effect of the circulating SASP, we demonstrated that plasma exposure from Old-vehicle p16-3MR mice, but not from Old-GCV mice, induced aortic stiffening assessed ex vivo (elastic modulus; P<0.05). Plasma proteomics implicated glycolysis in circulating SASP-mediated aortic stiffening. In old p16-3MR mice, GCV increased endothelial function assessed via peak carotid artery endothelium-dependent dilation (EDD; Old-GCV, 94±1% vs. Old-vehicle, 84±2%, P<0.05) to young levels (Old-GCV vs. young-vehicle, P=0.98), and EDD was higher in old C57BL/6N mice treated with ABT-263 vs. vehicle (96±1% vs. 82±3%, P<0.05). Improvements in endothelial function were mediated by increased nitric oxide (NO) bioavailability (P<0.05) and reduced oxidative stress (P<0.05). Circulating SASP factors related to NO signaling were associated with greater NO-mediated EDD following senescent cell clearance. CONCLUSIONS Cellular senescence and the SASP contribute to vascular aging and senolytics hold promise for improving age-related vascular function.
Collapse
Affiliation(s)
- Zachary S. Clayton
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO
| | - Matthew J. Rossman
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO
| | - Sophia A. Mahoney
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO
| | | | - Grace S. Maurer
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO
| | - David A. Hutton
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO
| | | | - Nathan T. Greenberg
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO
| | - Abigail G. Longtine
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO
| | - Katelyn R. Ludwig
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO
| | - Vienna E. Brunt
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO
| | - Thomas J. LaRocca
- Department of Health & Exercise Science, Colorado State University, Fort Collins, CO
- Center for Healthy Aging, Colorado State University, Fort Collins, CO
| | - Judith Campisi
- The Buck Institute for Research on Aging, Novato, CA
- Lawrence Berkeley National Laboratory, Berkeley, CA
| | - Simon Melov
- The Buck Institute for Research on Aging, Novato, CA
| | - Douglas R. Seals
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO
| |
Collapse
|
107
|
Suvakov S, Kattah AG, Gojkovic T, Enninga EAL, Pruett J, Jayachandran M, Sousa C, Santos J, Abou Hassan C, Gonzales-Suarez M, Garovic VD. Impact of Aging and Cellular Senescence in the Pathophysiology of Preeclampsia. Compr Physiol 2023; 13:5077-5114. [PMID: 37770190 DOI: 10.1002/cphy.c230003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/03/2023]
Abstract
The incidence of hypertensive disorders of pregnancy is increasing, which may be due to several factors, including an increased age at pregnancy and more comorbid health conditions during reproductive years. Preeclampsia, the most severe hypertensive disorder of pregnancy, has been associated with an increased risk of future disease, including cardiovascular and kidney diseases. Cellular senescence, the process of cell cycle arrest in response to many physiologic and maladaptive stimuli, may play an important role in the pathogenesis of preeclampsia and provide a mechanistic link to future disease. In this article, we will discuss the pathophysiology of preeclampsia, the many mechanisms of cellular senescence, evidence for the involvement of senescence in the development of preeclampsia, as well as evidence that cellular senescence may link preeclampsia to the risk of future disease. Lastly, we will explore how a better understanding of the role of cellular senescence in preeclampsia may lead to therapeutic trials. © 2023 American Physiological Society. Compr Physiol 13:5077-5114, 2023.
Collapse
Affiliation(s)
- Sonja Suvakov
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota, USA
| | - Andrea G Kattah
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota, USA
| | - Tamara Gojkovic
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota, USA
| | - Elizabeth A L Enninga
- Division of Research, Department of Obstetrics and Gynecology, Mayo Clinic College of Medicine, Rochester, Minnesota, USA
| | - Jacob Pruett
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota, USA
| | | | - Ciria Sousa
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota, USA
| | - Janelle Santos
- Division of Research, Department of Obstetrics and Gynecology, Mayo Clinic College of Medicine, Rochester, Minnesota, USA
| | - Coline Abou Hassan
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota, USA
| | | | - Vesna D Garovic
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota, USA
- Division of Research, Department of Obstetrics and Gynecology, Mayo Clinic College of Medicine, Rochester, Minnesota, USA
| |
Collapse
|
108
|
Jain A, Casanova D, Padilla AV, Paniagua Bojorges A, Kotla S, Ko KA, Samanthapudi VSK, Chau K, Nguyen MTH, Wen J, Hernandez Gonzalez SL, Rodgers SP, Olmsted-Davis EA, Hamilton DJ, Reyes-Gibby C, Yeung SCJ, Cooke JP, Herrmann J, Chini EN, Xu X, Yusuf SW, Yoshimoto M, Lorenzi PL, Hobbs B, Krishnan S, Koutroumpakis E, Palaskas NL, Wang G, Deswal A, Lin SH, Abe JI, Le NT. Premature senescence and cardiovascular disease following cancer treatments: mechanistic insights. Front Cardiovasc Med 2023; 10:1212174. [PMID: 37781317 PMCID: PMC10540075 DOI: 10.3389/fcvm.2023.1212174] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Accepted: 08/03/2023] [Indexed: 10/03/2023] Open
Abstract
Cardiovascular disease (CVD) is a leading cause of morbidity and mortality, especially among the aging population. The "response-to-injury" model proposed by Dr. Russell Ross in 1999 emphasizes inflammation as a critical factor in atherosclerosis development, with atherosclerotic plaques forming due to endothelial cell (EC) injury, followed by myeloid cell adhesion and invasion into the blood vessel walls. Recent evidence indicates that cancer and its treatments can lead to long-term complications, including CVD. Cellular senescence, a hallmark of aging, is implicated in CVD pathogenesis, particularly in cancer survivors. However, the precise mechanisms linking premature senescence to CVD in cancer survivors remain poorly understood. This article aims to provide mechanistic insights into this association and propose future directions to better comprehend this complex interplay.
Collapse
Affiliation(s)
- Ashita Jain
- Department of Cardiology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Diego Casanova
- Department of Cardiology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | | | | | - Sivareddy Kotla
- Department of Cardiology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Kyung Ae Ko
- Department of Cardiology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | | | - Khanh Chau
- Department of Cardiovascular Sciences, Houston Methodist Research Institute, Houston, TX, United States
| | - Minh T. H. Nguyen
- Department of Cardiovascular Sciences, Houston Methodist Research Institute, Houston, TX, United States
| | - Jake Wen
- Department of Cardiology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | | | - Shaefali P. Rodgers
- Department of Cardiovascular Sciences, Houston Methodist Research Institute, Houston, TX, United States
| | | | - Dale J. Hamilton
- Department of Medicine, Center for Bioenergetics, Houston Methodist Research Institute, Houston, TX, United States
| | - Cielito Reyes-Gibby
- Department of Emergency Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Sai-Ching J. Yeung
- Department of Emergency Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - John P. Cooke
- Department of Cardiovascular Sciences, Houston Methodist Research Institute, Houston, TX, United States
| | - Joerg Herrmann
- Cardio Oncology Clinic, Division of Preventive Cardiology, Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN, United States
| | - Eduardo N. Chini
- Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, Jacksonville, FL, United States
| | - Xiaolei Xu
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN, United States
| | - Syed Wamique Yusuf
- Department of Cardiology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Momoko Yoshimoto
- Center for Stem Cell & Regenerative Medicine, The University of Texas Health Science Center at Houston, Houston, TX, United States
| | - Philip L. Lorenzi
- Department of Bioinformatics and Computational Biology, Division of VP Research, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Brain Hobbs
- Department of Population Health, The University of Texas at Austin, Austin, TX, United States
| | - Sunil Krishnan
- Department of Neurosurgery, The University of Texas Health Science Center at Houston, Houston, TX, United States
| | - Efstratios Koutroumpakis
- Department of Cardiology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Nicolas L. Palaskas
- Department of Cardiology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Guangyu Wang
- Department of Cardiovascular Sciences, Houston Methodist Research Institute, Houston, TX, United States
| | - Anita Deswal
- Department of Cardiology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Steven H. Lin
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Jun-ichi Abe
- Department of Cardiology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Nhat-Tu Le
- Department of Cardiovascular Sciences, Houston Methodist Research Institute, Houston, TX, United States
| |
Collapse
|
109
|
Kumar M, Orkaby A, Tighe C, Villareal DT, Billingsley H, Nanna MG, Kwak MJ, Rohant N, Patel S, Goyal P, Hummel S, Al-Malouf C, Kolimas A, Krishnaswami A, Rich MW, Kirkpatrick J, Damluji AA, Kuchel GA, Forman DE, Alexander KP. Life's Essential 8: Optimizing Health in Older Adults. JACC. ADVANCES 2023; 2:100560. [PMID: 37664644 PMCID: PMC10470487 DOI: 10.1016/j.jacadv.2023.100560] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Revised: 06/26/2023] [Accepted: 07/07/2023] [Indexed: 09/05/2023]
Abstract
The population worldwide is getting older as a result of advances in public health, medicine, and technology. Older individuals are living longer with a higher prevalence of subclinical and clinical cardiovascular disease (CVD). In 2010, the American Heart Association introduced a list of key prevention targets, known as "Life's Simple 7" to increase CVD-free survival, longevity, and quality of life. In 2022, sleep health was added to expand the recommendations to "Life's Essential 8" (eat better, be more active, stop smoking, get adequate sleep, manage weight, manage cholesterol, manage blood pressure, and manage diabetes). These prevention targets are intended to apply regardless of chronologic age. During this same time, the understanding of aging biology and goals of care for older adults further enhanced the relevance of prevention across the range of functions. From a biological perspective, aging is a complex cellular process characterized by genomic instability, telomere attrition, loss of proteostasis, inflammation, deregulated nutrient-sensing, mitochondrial dysfunction, cellular senescence, stem cell exhaustion, and altered intercellular communication. These aging hallmarks are triggered by and enhanced by traditional CVD risk factors leading to geriatric syndromes (eg, frailty, sarcopenia, functional limitation, and cognitive impairment) which complicate efforts toward prevention. Therefore, we review Life's Essential 8 through the lens of aging biology, geroscience, and geriatric precepts to guide clinicians taking care of older adults.
Collapse
Affiliation(s)
- Manish Kumar
- Pat and Jim Calhoun Cardiology Center, University of Connecticut School of Medicine, Farmington, Connecticut, USA
- UConn Center on Aging, University of Connecticut School of Medicine, Farmington, Connecticut, USA
| | - Ariela Orkaby
- New England GRECC (Geriatric Research Education and Clinical Center), VA Boston HealthCare System, Boston, Massachusetts, USA
- Division of Aging, Brigham & Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Caitlan Tighe
- VISN 4 Mental Illness Research, Education and Clinical Center, Veterans Affairs Pittsburgh Healthcare System, Pittsburgh, Pennsylvania, USA
| | - Dennis T. Villareal
- Division of Endocrinology, Diabetes, and Metabolism, Baylor College of Medicine, Houston, Texas, USA
| | - Hayley Billingsley
- Division of Cardiovascular Medicine, University of Michigan Frankel Cardiovascular Center, Ann Arbor, Michigan, USA
| | - Michael G. Nanna
- Section of Cardiovascular Medicine, Yale School of Medicine, New Haven, Connecticut, USA
| | - Min Ji Kwak
- Division of Geriatric and Palliative Medicine, McGovern Medical School, The University of Texas Health Science Center, Houston, Texas, USA
| | - Namit Rohant
- Department of Cardiology, University of Arizona, Tucson, Arizona, USA
| | - Shreya Patel
- Department of Pharmacy Practice, School of Pharmacy and Health Sciences, Fairleigh Dickinson University, Florham Park, New Jersey, USA
| | - Parag Goyal
- Program for the Care and Study of Aging Heart, Department of Medicine, Weill Cornell of Medicine, New York, New York, USA
| | - Scott Hummel
- Division of Cardiovascular Medicine, University of Michigan Frankel Cardiovascular Center, Ann Arbor, Michigan, USA
| | - Christina Al-Malouf
- Department of Cardiology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Amie Kolimas
- Department of Internal Medicine, University of Arizona, Tucson, Arizona, USA
| | | | - Michael W. Rich
- Department of Medicine, Washington University, St Louise, Missouri, USA
| | - James Kirkpatrick
- Department of Cardiology, University of Washington, Seattle, Washington, USA
| | - Abdulla A. Damluji
- The Inova Center of Outcomes Research, Inova Heart and Vascular Institute, Falls Church, Virginia, USA
| | - George A. Kuchel
- UConn Center on Aging, University of Connecticut School of Medicine, Farmington, Connecticut, USA
| | - Daniel E. Forman
- Divisions of Cardiology and Geriatrics, Department of Medicine, University of Pittsburgh, Pittsburgh GRECC, VA Pittsburgh Healthcare System, Pittsburgh, Pennsylvania, USA
| | - Karen P. Alexander
- Division of Cardiology, Duke Medicine, Duke Clinical Research Institute, Durham, North Carolina, USA
| |
Collapse
|
110
|
Li A, Yan J, Zhao Y, Yu Z, Tian S, Khan AH, Zhu Y, Wu A, Zhang C, Tian XL. Vascular Aging: Assessment and Intervention. Clin Interv Aging 2023; 18:1373-1395. [PMID: 37609042 PMCID: PMC10441648 DOI: 10.2147/cia.s423373] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2023] [Accepted: 08/06/2023] [Indexed: 08/24/2023] Open
Abstract
Vascular aging represents a collection of structural and functional changes in a blood vessel with advancing age, including increased stiffness, vascular wall remodeling, loss of angiogenic ability, and endothelium-dependent vasodilation dysfunction. These age-related alterations may occur earlier in those who are at risk for or have cardiovascular diseases, therefore, are defined as early or premature vascular aging. Vascular aging contributes independently to cardio-cerebral vascular diseases (CCVDs). Thus, early diagnosis and interventions targeting vascular aging are of paramount importance in the delay or prevention of CCVDs. Here, we review the direct assessment of vascular aging by examining parameters that reflect changes in structure, function, or their compliance with age including arterial wall thickness and lumen diameter, endothelium-dependent vasodilation, arterial stiffness as well as indirect assessment through pathological studies of biomarkers including endothelial progenitor cell, lymphocytic telomeres, advanced glycation end-products, and C-reactive protein. Further, we evaluate how different types of interventions including lifestyle mediation, such as caloric restriction and salt intake, and treatments for hypertension, diabetes, and hyperlipidemia affect age-related vascular changes. As a single parameter or intervention targets only a certain vascular physiological change, it is recommended to use multiple parameters to evaluate and design intervention approaches accordingly to prevent systemic vascular aging in clinical practices or population-based studies.
Collapse
Affiliation(s)
- Ao Li
- Queen Mary School, Nanchang University, Nanchang, Jiangxi, 330031, People’s Republic of China
- Aging and Vascular Diseases, Human Aging Research Institute (HARI) and School of Life Science, Nanchang University, and Jiangxi Key Laboratory of Human Aging, Nanchang, Jiangxi, 330031, People’s Republic of China
| | - Jinhua Yan
- Department of Geriatrics, Institute of Gerontology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People’s Republic of China
| | - Ya Zhao
- Aging and Vascular Diseases, Human Aging Research Institute (HARI) and School of Life Science, Nanchang University, and Jiangxi Key Laboratory of Human Aging, Nanchang, Jiangxi, 330031, People’s Republic of China
| | - Zhenping Yu
- Institute of Translational Medicine, School of Life Science, Nanchang University, and Jiangxi Key Laboratory of Human Aging, Nanchang, Jiangxi, 330031, People’s Republic of China
| | - Shane Tian
- Department of Biochemistry/Chemistry, Ohio State University, Columbus, OH, USA
| | - Abdul Haseeb Khan
- Aging and Vascular Diseases, Human Aging Research Institute (HARI) and School of Life Science, Nanchang University, and Jiangxi Key Laboratory of Human Aging, Nanchang, Jiangxi, 330031, People’s Republic of China
| | - Yuanzheng Zhu
- Aging and Vascular Diseases, Human Aging Research Institute (HARI) and School of Life Science, Nanchang University, and Jiangxi Key Laboratory of Human Aging, Nanchang, Jiangxi, 330031, People’s Republic of China
| | - Andong Wu
- Aging and Vascular Diseases, Human Aging Research Institute (HARI) and School of Life Science, Nanchang University, and Jiangxi Key Laboratory of Human Aging, Nanchang, Jiangxi, 330031, People’s Republic of China
| | - Cuntai Zhang
- Department of Geriatrics, Institute of Gerontology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People’s Republic of China
| | - Xiao-Li Tian
- Aging and Vascular Diseases, Human Aging Research Institute (HARI) and School of Life Science, Nanchang University, and Jiangxi Key Laboratory of Human Aging, Nanchang, Jiangxi, 330031, People’s Republic of China
| |
Collapse
|
111
|
Forman DE, Kuchel GA, Newman JC, Kirkland JL, Volpi E, Taffet GE, Barzilai N, Pandey A, Kitzman DW, Libby P, Ferrucci L. Impact of Geroscience on Therapeutic Strategies for Older Adults With Cardiovascular Disease: JACC Scientific Statement. J Am Coll Cardiol 2023; 82:631-647. [PMID: 37389519 PMCID: PMC10414756 DOI: 10.1016/j.jacc.2023.05.038] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Revised: 05/09/2023] [Accepted: 05/31/2023] [Indexed: 07/01/2023]
Abstract
Geroscience posits that cardiovascular disease (CVD) and other chronic diseases result from progressive erosion of the effectiveness of homeostatic mechanisms that oppose age-related accumulation of molecular damage. This hypothetical common root to chronic diseases explains why patients with CVD are often affected by multimorbidity and frailty and why older age negatively affects CVD prognosis and treatment response. Gerotherapeutics enhance resilience mechanisms that counter age-related molecular damage to prevent chronic diseases, frailty, and disability, thereby extending healthspan. Here, we describe the main resilience mechanisms of mammalian aging, with a focus on how they can affect CVD pathophysiology. We next present novel gerotherapeutic approaches, some of which are already used in management of CVD, and explore their potential to transform care and management of CVD. The geroscience paradigm is gaining traction broadly in medical specialties, with potential to mitigate premature aging, reduce health care disparities, and improve population healthspan.
Collapse
Affiliation(s)
- Daniel E Forman
- Department of Medicine (Geriatrics and Cardiology) University of Pittsburgh, Pittsburgh, Pennsylvania, USA; GRECC, VA Pittsburgh Healthcare System, Pittsburgh, Pennsylvania, USA.
| | - George A Kuchel
- UConn Center on Aging, University of Connecticut School of Medicine, UConn Health, Farmington, Connecticut, USA
| | - John C Newman
- Buck Institute for Research on Aging, Novato California, USA; Division of Geriatrics, University of California San Francisco, San Francisco, California, USA
| | - James L Kirkland
- Division of General Internal Medicine, Department of Medicine and Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota, USA
| | - Elena Volpi
- Sealy Center on Aging, University of Texas Medical Branch, Galveston, Texas, USA
| | - George E Taffet
- Department of Medicine (Geriatrics and Cardiovascular Sciences), Baylor College of Medicine, Houston, Texas, USA
| | - Nir Barzilai
- Einstein Institute for Aging Research, Bronx, New York, USA; Einstein-NSC and Glenn Medical Center, Albert Einstein College of Medicine, Bronx, New York, USA
| | - Ambarish Pandey
- Division of Cardiology, Department of Internal Medicine, UT Southwestern Medical Center, Dallas, Texas, USA
| | - Dalane W Kitzman
- Cardiovascular Division, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Peter Libby
- Cardiovascular Medicine and Geriatrics, Department of Internal Medicine, Wake Forest University School of Medicine, Winston-Salem, North Carolina, USA
| | | |
Collapse
|
112
|
Holloway K, Neherin K, Dam KU, Zhang H. Cellular senescence and neurodegeneration. Hum Genet 2023; 142:1247-1262. [PMID: 37115318 DOI: 10.1007/s00439-023-02565-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2023] [Accepted: 04/20/2023] [Indexed: 04/29/2023]
Abstract
Advancing age is a major risk factor of Alzheimer's disease (AD). The worldwide prevalence of AD is approximately 50 million people, and this number is projected to increase substantially. The molecular mechanisms underlying the aging-associated susceptibility to cognitive impairment in AD are largely unknown. As a hallmark of aging, cellular senescence is a significant contributor to aging and age-related diseases including AD. Senescent neurons and glial cells have been detected to accumulate in the brains of AD patients and mouse models. Importantly, selective elimination of senescent cells ameliorates amyloid beta and tau pathologies and improves cognition in AD mouse models, indicating a critical role of cellular senescence in AD pathogenesis. Nonetheless, the mechanisms underlying when and how cellular senescence contributes to AD pathogenesis remain unclear. This review provides an overview of cellular senescence and discusses recent advances in the understanding of the impact of cellular senescence on AD pathogenesis, with brief discussions of the possible role of cellular senescence in other neurodegenerative diseases including Down syndrome, Parkinson's disease, multiple sclerosis, and amyotrophic lateral sclerosis.
Collapse
Affiliation(s)
- Kristopher Holloway
- Department of Pediatrics, University of Massachusetts Chan Medical School, Worcester, MA, 01655, USA
| | - Kashfia Neherin
- Department of Pediatrics, University of Massachusetts Chan Medical School, Worcester, MA, 01655, USA
| | - Kha Uyen Dam
- Department of Pediatrics, University of Massachusetts Chan Medical School, Worcester, MA, 01655, USA
| | - Hong Zhang
- Department of Pediatrics, University of Massachusetts Chan Medical School, Worcester, MA, 01655, USA.
| |
Collapse
|
113
|
Bloom SI, Liu Y, Tucker JR, Islam MT, Machin DR, Abdeahad H, Thomas TG, Bramwell RC, Lesniewski LA, Donato AJ. Endothelial cell telomere dysfunction induces senescence and results in vascular and metabolic impairments. Aging Cell 2023; 22:e13875. [PMID: 37259606 PMCID: PMC10410008 DOI: 10.1111/acel.13875] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Revised: 04/03/2023] [Accepted: 05/04/2023] [Indexed: 06/02/2023] Open
Abstract
In advanced age, increases in oxidative stress and inflammation impair endothelial function, which contributes to the development of cardiovascular disease (CVD). One plausible source of this oxidative stress and inflammation is an increase in the abundance of senescent endothelial cells. Cellular senescence is a cell cycle arrest that occurs in response to various damaging stimuli. In the present study, we tested the hypothesis that advanced age results in endothelial cell telomere dysfunction that induces senescence. In both human and mouse endothelial cells, advanced age resulted in an increased abundance of dysfunctional telomeres, characterized by activation of DNA damage signaling at telomeric DNA. To test whether this results in senescence, we selectively reduced the telomere shelterin protein telomere repeat binding factor 2 (Trf2) from endothelial cells of young mice. Trf2 reduction increased endothelial cell telomere dysfunction and resulted in cellular senescence. Furthermore, induction of endothelial cell telomere dysfunction increased inflammatory signaling and oxidative stress, resulting in impairments in endothelial function. Finally, we demonstrate that endothelial cell telomere dysfunction-induced senescence impairs glucose tolerance. This likely occurs through increases in inflammatory signaling in the liver and adipose tissue, as well as reductions in microvascular density and vasodilation to metabolic stimuli. Cumulatively, the findings of the present study identify age-related telomere dysfunction as a mechanism that leads to endothelial cell senescence. Furthermore, these data provide compelling evidence that senescent endothelial cells contribute to age-related increases in oxidative stress and inflammation that impair arterial and metabolic function.
Collapse
Affiliation(s)
- Samuel I. Bloom
- Department of Nutrition and Integrative PhysiologyThe University of UtahSalt Lake CityUtahUSA
| | - Yu Liu
- Department of GeriatricsTongji HospitalWuhanChina
- Division of Geriatrics, Department of Internal MedicineUniversity of Utah School of MedicineSalt Lake CityUtahUSA
| | - Jordan R. Tucker
- Division of Geriatrics, Department of Internal MedicineUniversity of Utah School of MedicineSalt Lake CityUtahUSA
| | - Md Torikul Islam
- Department of Nutrition and Integrative PhysiologyThe University of UtahSalt Lake CityUtahUSA
| | - Daniel R. Machin
- Division of Geriatrics, Department of Internal MedicineUniversity of Utah School of MedicineSalt Lake CityUtahUSA
- Department of Nutrition and Integrative PhysiologyFlorida State UniversityTallahasseeFloridaUSA
| | - Hossein Abdeahad
- Department of Nutrition and Integrative PhysiologyThe University of UtahSalt Lake CityUtahUSA
| | - Tyler G. Thomas
- Division of Geriatrics, Department of Internal MedicineUniversity of Utah School of MedicineSalt Lake CityUtahUSA
| | - R. Colton Bramwell
- Division of Geriatrics, Department of Internal MedicineUniversity of Utah School of MedicineSalt Lake CityUtahUSA
| | - Lisa A. Lesniewski
- Department of Nutrition and Integrative PhysiologyThe University of UtahSalt Lake CityUtahUSA
- Division of Geriatrics, Department of Internal MedicineUniversity of Utah School of MedicineSalt Lake CityUtahUSA
- Geriatric Research, Education and Clinical CenterVeteran's Affairs Medical Center‐Salt Lake CitySalt Lake CityUtahUSA
- Nora Eccles Harrison Cardiovascular Research and Training InstituteThe University of UtahSalt Lake CityUtahUSA
| | - Anthony J. Donato
- Department of Nutrition and Integrative PhysiologyThe University of UtahSalt Lake CityUtahUSA
- Division of Geriatrics, Department of Internal MedicineUniversity of Utah School of MedicineSalt Lake CityUtahUSA
- Geriatric Research, Education and Clinical CenterVeteran's Affairs Medical Center‐Salt Lake CitySalt Lake CityUtahUSA
- Nora Eccles Harrison Cardiovascular Research and Training InstituteThe University of UtahSalt Lake CityUtahUSA
- Department of BiochemistryThe University of UtahSalt Lake CityUtahUSA
| |
Collapse
|
114
|
Ya J, Bayraktutan U. Vascular Ageing: Mechanisms, Risk Factors, and Treatment Strategies. Int J Mol Sci 2023; 24:11538. [PMID: 37511296 PMCID: PMC10380571 DOI: 10.3390/ijms241411538] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Revised: 07/10/2023] [Accepted: 07/14/2023] [Indexed: 07/30/2023] Open
Abstract
Ageing constitutes the biggest risk factor for poor health and adversely affects the integrity and function of all the cells, tissues, and organs in the human body. Vascular ageing, characterised by vascular stiffness, endothelial dysfunction, increased oxidative stress, chronic low-grade inflammation, and early-stage atherosclerosis, may trigger or exacerbate the development of age-related vascular diseases, which each year contribute to more than 3.8 million deaths in Europe alone and necessitate a better understanding of the mechanisms involved. To this end, a large number of recent preclinical and clinical studies have focused on the exponential accumulation of senescent cells in the vascular system and paid particular attention to the specific roles of senescence-associated secretory phenotype, proteostasis dysfunction, age-mediated modulation of certain microRNA (miRNAs), and the contribution of other major vascular risk factors, notably diabetes, hypertension, or smoking, to vascular ageing in the elderly. The data generated paved the way for the development of various senotherapeutic interventions, ranging from the application of synthetic or natural senolytics and senomorphics to attempt to modify lifestyle, control diet, and restrict calorie intake. However, specific guidelines, considering the severity and characteristics of vascular ageing, need to be established before widespread use of these agents. This review briefly discusses the molecular and cellular mechanisms of vascular ageing and summarises the efficacy of widely studied senotherapeutics in the context of vascular ageing.
Collapse
Affiliation(s)
- Jingyuan Ya
- Academic Unit of Mental Health and Clinical Neuroscience, Nottingham University, Queen's Medical Centre, Nottingham NG7 2UH, UK
| | - Ulvi Bayraktutan
- Academic Unit of Mental Health and Clinical Neuroscience, Nottingham University, Queen's Medical Centre, Nottingham NG7 2UH, UK
| |
Collapse
|
115
|
Karnewar S, Karnewar V, Shankman LS, Owens GK. Treatment of advanced atherosclerotic mice with the senolytic agent ABT-263 is associated with reduced indices of plaque stability and increased mortality. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.07.12.548696. [PMID: 37502944 PMCID: PMC10369968 DOI: 10.1101/2023.07.12.548696] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/29/2023]
Abstract
The use of senolytic agents to remove senescent cells from atherosclerotic lesions is controversial. A common limitation of previous studies is the failure to rigorously define the effects of senolytic agent ABT-263 (Navitoclax) on smooth muscle cells (SMC) despite studies claiming that they are the major source of senescent cells. Moreover, there are no studies of the effect of ABT-263 on endothelial cells (EC), which along with SMC comprise 90% of α-SMA+ myofibroblast-like cells in the protective fibrous cap. Here we tested the hypothesis that treatment of advanced atherosclerotic mice with the ABT-263 will reduce lesion size and increase plaque stability. SMC (Myh11-CreERT2-eYFP) and EC (Cdh5-CreERT2-eYFP) lineage tracing Apoe-/- mice were fed a WD for 18 weeks, followed by ABT-263 100mg/kg/bw for six weeks or 50mg/kg/bw for nine weeks. ABT-263 treatment did not change lesion size or lumen area of the brachiocephalic artery (BCA). However, ABT-263 treatment reduced SMC by 90% and increased EC-contributions to lesions via EC-to-mesenchymal transition (EndoMT) by 60%. ABT-263 treatment also reduced α-SMA+ fibrous cap thickness by 60% and increased mortality by >50%. Contrary to expectations, treatment of WD-fed Apoe-/- mice with the senolytic agent ABT-263 resulted in multiple detrimental changes including reduced indices of stability, and increased mortality.
Collapse
Affiliation(s)
- Santosh Karnewar
- Robert M. Berne Cardiovascular Research Center, University of Virginia-School of Medicine, 415 Lane Road, Suite 1010, Charlottesville, VA, 22908, USA
| | - Vaishnavi Karnewar
- Robert M. Berne Cardiovascular Research Center, University of Virginia-School of Medicine, 415 Lane Road, Suite 1010, Charlottesville, VA, 22908, USA
| | - Laura S Shankman
- Robert M. Berne Cardiovascular Research Center, University of Virginia-School of Medicine, 415 Lane Road, Suite 1010, Charlottesville, VA, 22908, USA
| | - Gary K Owens
- Robert M. Berne Cardiovascular Research Center, University of Virginia-School of Medicine, 415 Lane Road, Suite 1010, Charlottesville, VA, 22908, USA
| |
Collapse
|
116
|
Wang Y, Che L, Chen X, He Z, Song D, Yuan Y, Liu C. Repurpose dasatinib and quercetin: Targeting senescent cells ameliorates postmenopausal osteoporosis and rejuvenates bone regeneration. Bioact Mater 2023; 25:13-28. [PMID: 37056256 PMCID: PMC10088057 DOI: 10.1016/j.bioactmat.2023.01.009] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Revised: 12/27/2022] [Accepted: 01/12/2023] [Indexed: 01/21/2023] Open
Abstract
Clinical therapies developed for estrogen-deficiency-driven postmenopausal osteoporosis (PMO) and related diseases, such as bone degeneration, show multiple adverse effects nowadays. Targeting senescent cells (SnCs) and the consequent senescence-associated secretory phenotype (SASP) with a combination of dasatinib and quercetin (DQ) is a recently developed novel therapy for multiple age-related diseases. Herein, we found that estrogen deficiency induced-bone loss was attributed to a pro-inflammatory microenvironment with SASP secretions and accelerated SnC accumulation, especially senescent mesenchymal stem cells (MSCs) characterized by exhaustion and dysfunction in middle aged rats. Systematically targeting SnCs with DQ strikingly ameliorated PMO and restored MSC function. Local administration of DQ and bone morphogenetic protein 2 (BMP2) in combination promoted osteogenic differentiation of MSCs and rejuvenated osteoporotic bone regeneration. Our results repurposed DQ as an attractive therapy for treating PMO and related diseases.
Collapse
Affiliation(s)
- Ying Wang
- Key Laboratory for Ultrafine Materials of Ministry of Education, Frontiers Science Center for Materiobiology and Dynamic Chemistry, Engineering Research Center for Biomaterials of Ministry of Education, School of Materials Science and Engineering, East China University of Science and Technology, Shanghai, 200237, PR China
| | - Lingbin Che
- Department of Orthopedics, Shanghai General Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200080, China
| | - Xi Chen
- Key Laboratory for Ultrafine Materials of Ministry of Education, Frontiers Science Center for Materiobiology and Dynamic Chemistry, Engineering Research Center for Biomaterials of Ministry of Education, School of Materials Science and Engineering, East China University of Science and Technology, Shanghai, 200237, PR China
| | - Zirui He
- Key Laboratory for Ultrafine Materials of Ministry of Education, Frontiers Science Center for Materiobiology and Dynamic Chemistry, Engineering Research Center for Biomaterials of Ministry of Education, School of Materials Science and Engineering, East China University of Science and Technology, Shanghai, 200237, PR China
| | - Dianwen Song
- Department of Orthopedics, Shanghai General Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200080, China
| | - Yuan Yuan
- Key Laboratory for Ultrafine Materials of Ministry of Education, Frontiers Science Center for Materiobiology and Dynamic Chemistry, Engineering Research Center for Biomaterials of Ministry of Education, School of Materials Science and Engineering, East China University of Science and Technology, Shanghai, 200237, PR China
| | - Changsheng Liu
- Key Laboratory for Ultrafine Materials of Ministry of Education, Frontiers Science Center for Materiobiology and Dynamic Chemistry, Engineering Research Center for Biomaterials of Ministry of Education, School of Materials Science and Engineering, East China University of Science and Technology, Shanghai, 200237, PR China
| |
Collapse
|
117
|
Witham MD, Granic A, Miwa S, Passos JF, Richardson GD, Sayer AA. New Horizons in cellular senescence for clinicians. Age Ageing 2023; 52:afad127. [PMID: 37466640 PMCID: PMC10355181 DOI: 10.1093/ageing/afad127] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Indexed: 07/20/2023] Open
Abstract
Cellular senescence has emerged as a fundamental biological mechanism underpinning the ageing process and has been implicated in the pathogenesis of an increasing number of age-related conditions. Cellular senescence is a cell fate originally defined as an irreversible loss of replicative potential although it is now clear that it can be induced by a variety of mechanisms independent of replication and telomere attrition. The drivers include a persistent DNA damage response causing multiple alterations in cellular function. Senescent cells secrete a range of mediators that drive chronic inflammation and can convert other cells to the senescent state-the senescence-associated secretory phenotype. Much research to date has been conducted in animal models, but it is now clear that senescent cells accompany ageing in humans and their presence is an important driver of disease across systems. Proof-of-concept work suggests that preventing or reversing senescence may be a viable strategy to counteract human ageing and age-related disease. Possible interventions include exercise, nutrition and senolytics/senostatic drugs although there are a number of potential limitations to the use of senotherapeutics. These interventions are generally tested for single-organ conditions, but the real power of this approach is the potential to tackle multiple age-related conditions. The litmus test for this exciting new class of therapies, however, will be whether they can improve healthy life expectancy rather than merely extending lifespan. The outcomes measured in clinical studies need to reflect these aims if senotherapeutics are to gain the trust of clinicians, patients and the public.
Collapse
Affiliation(s)
- Miles D Witham
- AGE Research Group, Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle, UK
- NIHR Newcastle Biomedical Research Centre, Newcastle upon Tyne Hospitals NHS Foundation Trust, Cumbria, Northumberland, Tyne and Wear NHS Foundation Trust and Newcastle University, Newcastle, UK
| | - Antoneta Granic
- AGE Research Group, Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle, UK
- NIHR Newcastle Biomedical Research Centre, Newcastle upon Tyne Hospitals NHS Foundation Trust, Cumbria, Northumberland, Tyne and Wear NHS Foundation Trust and Newcastle University, Newcastle, UK
| | - Satomi Miwa
- Biosciences Institute, Faculty of Medical Sciences, Newcastle University, Newcastle, UK
| | - Joao F Passos
- Department of Physiology and Biomedical Engineering and Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, MN, USA
| | - Gavin D Richardson
- Vascular Medicine and Biology Theme, Biosciences Institute, Faculty of Medical Sciences, Newcastle University, Newcastle, UK
| | - Avan A Sayer
- AGE Research Group, Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle, UK
- NIHR Newcastle Biomedical Research Centre, Newcastle upon Tyne Hospitals NHS Foundation Trust, Cumbria, Northumberland, Tyne and Wear NHS Foundation Trust and Newcastle University, Newcastle, UK
| |
Collapse
|
118
|
Bloom SI, Tuday E, Islam T, Gogulamudi VR, Lesniewski LA, Donato AJ. Senolytics Reduce Endothelial Cell DNA Damage and Telomere Dysfunction Despite Reductions in Telomere Length. AGING BIOLOGY 2023; 1:e20230007. [PMID: 40109908 PMCID: PMC11922086 DOI: 10.59368/agingbio.20230007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 03/22/2025]
Abstract
Aging results in cellular damage that can induce cell cycle arrest known as cellular senescence. Endothelial cells are one of the first cell types to become senescent in advancing age and contribute to age-related cardiovascular diseases. Drugs known as senolytics reduce endothelial cell senescence in cell culture. From a translational perspective, a key question is whether this occurs in vivo and if remaining cells appear healthier and display fewer hallmarks of cellular aging. In this study, we treated old mice with the senolytic cocktail dasatinib and quercetin (D+Q) or a vehicle control. In 24-month-old mice, D+Q treatment reduced p21 gene expression in carotid artery endothelial cells, indicative of reductions in senescence. In lung endothelial cells, we examined DNA damage, telomere dysfunction (DNA damage signaling at telomeres), and telomere length, which are hallmarks of aging associated with senescence and other deleterious effects on cellular function. D+Q treatment resulted in fewer endothelial cells with DNA damage and dysfunctional telomeres. Surprisingly, D+Q reduced endothelial cell telomere length, yet this did not result in critically short telomeres and thus telomere dysfunction. Mice have longer telomeres than humans; therefore, future studies on the effect of senolytics on telomere length are warranted. Collectively, this study provides important evidence on the effect of senolytics, including that they clear senescent endothelial cells in vivo, which reduces DNA damage and telomere dysfunction. These data indicate that the clearing of senescent endothelial cells in old age leaves behind a population of cells that exhibit fewer hallmarks of vascular aging.
Collapse
Affiliation(s)
- Samuel I Bloom
- Department of Nutrition and Integrative Physiology, The University of Utah, Salt Lake City, UT, USA
| | - Eric Tuday
- Department of Internal Medicine, Division of Cardiology, The University of Utah, Salt Lake City, UT, USA
- Veteran's Affairs Medical Center-Salt Lake City, Geriatric, Research, Education, and Clinical Center, Salt Lake City, UT, USA
| | - Torikul Islam
- Department of Nutrition and Integrative Physiology, The University of Utah, Salt Lake City, UT, USA
| | - Venkateswara R Gogulamudi
- Department of Internal Medicine, Division of Geriatrics, The University of Utah, Salt Lake City, UT, USA
| | - Lisa A Lesniewski
- Department of Nutrition and Integrative Physiology, The University of Utah, Salt Lake City, UT, USA
- Veteran's Affairs Medical Center-Salt Lake City, Geriatric, Research, Education, and Clinical Center, Salt Lake City, UT, USA
- Department of Internal Medicine, Division of Geriatrics, The University of Utah, Salt Lake City, UT, USA
- Nora Eccles Harrison Cardiovascular Research and Training Institute, The University of Utah, Salt Lake City, UT, USA
| | - Anthony J Donato
- Department of Nutrition and Integrative Physiology, The University of Utah, Salt Lake City, UT, USA
- Veteran's Affairs Medical Center-Salt Lake City, Geriatric, Research, Education, and Clinical Center, Salt Lake City, UT, USA
- Department of Internal Medicine, Division of Geriatrics, The University of Utah, Salt Lake City, UT, USA
- Nora Eccles Harrison Cardiovascular Research and Training Institute, The University of Utah, Salt Lake City, UT, USA
- Department of Biochemistry, The University of Utah, Salt Lake City, UT, USA
| |
Collapse
|
119
|
Torrance BL, Cadar AN, Martin DE, Panier HA, Lorenzo EC, Bartley JM, Xu M, Haynes L. Senolytic treatment with dasatinib and quercetin does not improve overall influenza responses in aged mice. FRONTIERS IN AGING 2023; 4:1212750. [PMID: 37396956 PMCID: PMC10313122 DOI: 10.3389/fragi.2023.1212750] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Accepted: 06/09/2023] [Indexed: 07/04/2023]
Abstract
Age is the greatest risk factor for adverse outcomes following influenza (flu) infection. The increased burden of senescent cells with age has been identified as a root cause in many diseases of aging and targeting these cells with drugs termed senolytics has shown promise in alleviating many age-related declines across organ systems. However, there is little known whether targeting these cells will improve age-related deficits in the immune system. Here, we utilized a well characterized senolytic treatment with a combination of dasatinib and quercetin (D + Q) to clear aged (18-20 months) mice of senescent cells prior to a flu infection. We comprehensively profiled immune responses during the primary infection as well as development of immune memory and protection following pathogen reencounter. Senolytic treatment did not improve any aspects of the immune response that were assayed for including: weight loss, viral load, CD8 T-cell infiltration, antibody production, memory T cell development, or recall ability. These results indicate that D + Q may not be an appropriate senolytic to improve aged immune responses to flu infection.
Collapse
Affiliation(s)
- Blake L. Torrance
- University of Connecticut Center on Aging, Farmington, CT, United States
- Department of Immunology, Farmington, CT, United States
| | - Andreia N. Cadar
- University of Connecticut Center on Aging, Farmington, CT, United States
- Department of Immunology, Farmington, CT, United States
| | - Dominique E. Martin
- University of Connecticut Center on Aging, Farmington, CT, United States
- Department of Immunology, Farmington, CT, United States
| | | | - Erica C. Lorenzo
- University of Connecticut Center on Aging, Farmington, CT, United States
| | - Jenna M. Bartley
- University of Connecticut Center on Aging, Farmington, CT, United States
- Department of Immunology, Farmington, CT, United States
| | - Ming Xu
- University of Connecticut Center on Aging, Farmington, CT, United States
- Department of Genetics and Genome Sciences, Farmington, CT, United States
| | - Laura Haynes
- University of Connecticut Center on Aging, Farmington, CT, United States
- Department of Immunology, Farmington, CT, United States
| |
Collapse
|
120
|
Li X, Li C, Zhang W, Wang Y, Qian P, Huang H. Inflammation and aging: signaling pathways and intervention therapies. Signal Transduct Target Ther 2023; 8:239. [PMID: 37291105 PMCID: PMC10248351 DOI: 10.1038/s41392-023-01502-8] [Citation(s) in RCA: 431] [Impact Index Per Article: 215.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2022] [Revised: 04/26/2023] [Accepted: 05/15/2023] [Indexed: 06/10/2023] Open
Abstract
Aging is characterized by systemic chronic inflammation, which is accompanied by cellular senescence, immunosenescence, organ dysfunction, and age-related diseases. Given the multidimensional complexity of aging, there is an urgent need for a systematic organization of inflammaging through dimensionality reduction. Factors secreted by senescent cells, known as the senescence-associated secretory phenotype (SASP), promote chronic inflammation and can induce senescence in normal cells. At the same time, chronic inflammation accelerates the senescence of immune cells, resulting in weakened immune function and an inability to clear senescent cells and inflammatory factors, which creates a vicious cycle of inflammation and senescence. Persistently elevated inflammation levels in organs such as the bone marrow, liver, and lungs cannot be eliminated in time, leading to organ damage and aging-related diseases. Therefore, inflammation has been recognized as an endogenous factor in aging, and the elimination of inflammation could be a potential strategy for anti-aging. Here we discuss inflammaging at the molecular, cellular, organ, and disease levels, and review current aging models, the implications of cutting-edge single cell technologies, as well as anti-aging strategies. Since preventing and alleviating aging-related diseases and improving the overall quality of life are the ultimate goals of aging research, our review highlights the critical features and potential mechanisms of inflammation and aging, along with the latest developments and future directions in aging research, providing a theoretical foundation for novel and practical anti-aging strategies.
Collapse
Affiliation(s)
- Xia Li
- Bone Marrow Transplantation Center, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, People's Republic of China
- Liangzhu Laboratory, Zhejiang University Medical Center, 1369 West Wenyi Road, Hangzhou, 311121, China
- Institute of Hematology, Zhejiang University & Zhejiang Engineering Laboratory for Stem Cell and Immunotherapy, Hangzhou, 310058, China
- Zhejiang Province Engineering Laboratory for Stem Cell and Immunity Therapy, Hangzhou, 310058, China
| | - Chentao Li
- Liangzhu Laboratory, Zhejiang University Medical Center, 1369 West Wenyi Road, Hangzhou, 311121, China
- Zhejiang University-University of Edinburgh Institute, Zhejiang University School of Medicine, Zhejiang University, Haining, China
| | - Wanying Zhang
- Zhejiang University-University of Edinburgh Institute, Zhejiang University School of Medicine, Zhejiang University, Haining, China
| | - Yanan Wang
- Zhejiang University-University of Edinburgh Institute, Zhejiang University School of Medicine, Zhejiang University, Haining, China
| | - Pengxu Qian
- Liangzhu Laboratory, Zhejiang University Medical Center, 1369 West Wenyi Road, Hangzhou, 311121, China.
- Institute of Hematology, Zhejiang University & Zhejiang Engineering Laboratory for Stem Cell and Immunotherapy, Hangzhou, 310058, China.
- Zhejiang Province Engineering Laboratory for Stem Cell and Immunity Therapy, Hangzhou, 310058, China.
- Center for Stem Cell and Regenerative Medicine and Bone Marrow Transplantation Center of the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310058, China.
| | - He Huang
- Bone Marrow Transplantation Center, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, People's Republic of China.
- Liangzhu Laboratory, Zhejiang University Medical Center, 1369 West Wenyi Road, Hangzhou, 311121, China.
- Institute of Hematology, Zhejiang University & Zhejiang Engineering Laboratory for Stem Cell and Immunotherapy, Hangzhou, 310058, China.
- Zhejiang Province Engineering Laboratory for Stem Cell and Immunity Therapy, Hangzhou, 310058, China.
| |
Collapse
|
121
|
Lee DY, Lee SJ, Chandrasekaran P, Lamichhane G, O'Connell JF, Egan JM, Kim Y. Dietary Curcumin Attenuates Hepatic Cellular Senescence by Suppressing the MAPK/NF-κB Signaling Pathway in Aged Mice. Antioxidants (Basel) 2023; 12:1165. [PMID: 37371895 DOI: 10.3390/antiox12061165] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Revised: 05/19/2023] [Accepted: 05/25/2023] [Indexed: 06/29/2023] Open
Abstract
Dietary interventions with bioactive compounds have been found to suppress the accumulation of senescent cells and senescence-associated secretory phenotypes (SASPs). One such compound, curcumin (CUR), has beneficial health and biological effects, including antioxidant and anti-inflammatory properties, but its ability to prevent hepatic cellular senescence is unclear. The objective of this study was to investigate the effects of dietary CUR as an antioxidant on hepatic cellular senescence and determine its benefits on aged mice. We screened the hepatic transcriptome and found that CUR supplementation led to the downregulation of senescence-associated hepatic gene expressions in both usually fed and nutritionally challenged aged mice. Our results showed that CUR supplementation enhanced antioxidant properties and suppressed mitogen-activated protein kinase (MAPK) signaling cascades in the liver, particularly c-Jun N-terminal kinase (JNK) in aged mice and p38 in diet-induced obese aged mice. Furthermore, dietary CUR decreased the phosphorylation of nuclear factor-κB (NF-κB), a downstream transcription factor of JNK and p38, and inhibited the mRNA expression of proinflammatory cytokines and SASPs. The potency of CUR administration was demonstrated in aged mice via enhanced insulin homeostasis along with declined body weight. Taken together, these results suggest that CUR supplementation may be a nutritional strategy to prevent hepatic cellular senescence.
Collapse
Affiliation(s)
- Da-Yeon Lee
- Department of Nutritional Sciences, Oklahoma State University, Stillwater, OK 74078, USA
| | - Su-Jeong Lee
- Department of Nutritional Sciences, Oklahoma State University, Stillwater, OK 74078, USA
| | - Prabha Chandrasekaran
- Laboratory of Clinical Investigation, National Institute on Aging, Baltimore, MD 21224, USA
| | - Gopal Lamichhane
- Department of Nutritional Sciences, Oklahoma State University, Stillwater, OK 74078, USA
| | - Jennifer F O'Connell
- Laboratory of Clinical Investigation, National Institute on Aging, Baltimore, MD 21224, USA
| | - Josephine M Egan
- Laboratory of Clinical Investigation, National Institute on Aging, Baltimore, MD 21224, USA
| | - Yoo Kim
- Department of Nutritional Sciences, Oklahoma State University, Stillwater, OK 74078, USA
| |
Collapse
|
122
|
Ramadhiani R, Ikeda K, Miyagawa K, Ryanto GRT, Tamada N, Suzuki Y, Kirita Y, Matoba S, Hirata KI, Emoto N. Endothelial cell senescence exacerbates pulmonary hypertension by inducing juxtacrine Notch signaling in smooth muscle cells. iScience 2023; 26:106662. [PMID: 37192975 PMCID: PMC10182325 DOI: 10.1016/j.isci.2023.106662] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2022] [Revised: 03/17/2023] [Accepted: 04/06/2023] [Indexed: 05/18/2023] Open
Abstract
Pulmonary arterial hypertension (PAH) is a fatal disease characterized by a progressive increase in pulmonary artery pressure caused by pathological pulmonary artery remodeling. Here, we demonstrate that endothelial cell (EC) senescence plays a negative role in pulmonary hypertension via juxtacrine interaction with smooth muscle cells (SMCs). By using EC-specific progeroid mice, we discovered that EC progeria deteriorated vascular remodeling in the lungs, and exacerbated pulmonary hypertension in mice. Mechanistically, senescent ECs overexpressed Notch ligands, which resulted in increased Notch signaling and activated proliferation and migration capacities in neighboring SMCs. Pharmacological inhibition of Notch signaling reduced the effects of senescent ECs on SMCs functions in vitro, and improved the worsened pulmonary hypertension in EC-specific progeroid mice in vivo. Our findings show that EC senescence is a critical disease-modifying factor in PAH and that EC-mediated Notch signaling is a pharmacotherapeutic target for the treatment of PAH, particularly in the elderly.
Collapse
Affiliation(s)
- Risa Ramadhiani
- Laboratory of Clinical Pharmaceutical Science, Kobe Pharmaceutical University, 4-19-1 Motoyamakitamachi, Higashinada, Kobe 658-8558, Japan
- Division of Cardiovascular Medicine, Department of Internal Medicine, Kobe University Graduate School of Medicine, 7-5-1 Kusunoki, Chuo, Kobe 6500017, Japan
| | - Koji Ikeda
- Laboratory of Clinical Pharmaceutical Science, Kobe Pharmaceutical University, 4-19-1 Motoyamakitamachi, Higashinada, Kobe 658-8558, Japan
- Department of Epidemiology for Longevity and Regional Health, Kyoto Prefectural University of Medicine, 465 Kajii, Kawaramachi-Hirokoji, Kamigyou, Kyoto 6028566, Japan
- Department of Cardiology and Nephrology, Kyoto Prefectural University of Medicine, 465 Kajii, Kawaramachi-Hirokoji, Kamigyou, Kyoto 6028566, Japan
| | - Kazuya Miyagawa
- Laboratory of Clinical Pharmaceutical Science, Kobe Pharmaceutical University, 4-19-1 Motoyamakitamachi, Higashinada, Kobe 658-8558, Japan
- Division of Cardiovascular Medicine, Department of Internal Medicine, Kobe University Graduate School of Medicine, 7-5-1 Kusunoki, Chuo, Kobe 6500017, Japan
| | - Gusty Rizky Tough Ryanto
- Laboratory of Clinical Pharmaceutical Science, Kobe Pharmaceutical University, 4-19-1 Motoyamakitamachi, Higashinada, Kobe 658-8558, Japan
- Division of Cardiovascular Medicine, Department of Internal Medicine, Kobe University Graduate School of Medicine, 7-5-1 Kusunoki, Chuo, Kobe 6500017, Japan
| | - Naoki Tamada
- Laboratory of Clinical Pharmaceutical Science, Kobe Pharmaceutical University, 4-19-1 Motoyamakitamachi, Higashinada, Kobe 658-8558, Japan
- Division of Cardiovascular Medicine, Department of Internal Medicine, Kobe University Graduate School of Medicine, 7-5-1 Kusunoki, Chuo, Kobe 6500017, Japan
| | - Yoko Suzuki
- Laboratory of Clinical Pharmaceutical Science, Kobe Pharmaceutical University, 4-19-1 Motoyamakitamachi, Higashinada, Kobe 658-8558, Japan
| | - Yuhei Kirita
- Department of Cardiology and Nephrology, Kyoto Prefectural University of Medicine, 465 Kajii, Kawaramachi-Hirokoji, Kamigyou, Kyoto 6028566, Japan
| | - Satoaki Matoba
- Department of Cardiology and Nephrology, Kyoto Prefectural University of Medicine, 465 Kajii, Kawaramachi-Hirokoji, Kamigyou, Kyoto 6028566, Japan
| | - Ken-ichi Hirata
- Division of Cardiovascular Medicine, Department of Internal Medicine, Kobe University Graduate School of Medicine, 7-5-1 Kusunoki, Chuo, Kobe 6500017, Japan
| | - Noriaki Emoto
- Laboratory of Clinical Pharmaceutical Science, Kobe Pharmaceutical University, 4-19-1 Motoyamakitamachi, Higashinada, Kobe 658-8558, Japan
- Division of Cardiovascular Medicine, Department of Internal Medicine, Kobe University Graduate School of Medicine, 7-5-1 Kusunoki, Chuo, Kobe 6500017, Japan
| |
Collapse
|
123
|
Samakkarnthai P, Saul D, Zhang L, Aversa Z, Doolittle ML, Sfeir JG, Kaur J, Atkinson EJ, Edwards JR, Russell GG, Pignolo RJ, Kirkland JL, Tchkonia T, Niedernhofer LJ, Monroe DG, Lebrasseur NK, Farr JN, Robbins PD, Khosla S. In vitro and in vivo effects of zoledronic acid on senescence and senescence-associated secretory phenotype markers. Aging (Albany NY) 2023; 15:3331-3355. [PMID: 37154858 PMCID: PMC10449299 DOI: 10.18632/aging.204701] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Accepted: 04/18/2023] [Indexed: 05/10/2023]
Abstract
In addition to reducing fracture risk, zoledronic acid has been found in some studies to decrease mortality in humans and extend lifespan and healthspan in animals. Because senescent cells accumulate with aging and contribute to multiple co-morbidities, the non-skeletal actions of zoledronic acid could be due to senolytic (killing of senescent cells) or senomorphic (inhibition of the secretion of the senescence-associated secretory phenotype [SASP]) actions. To test this, we first performed in vitro senescence assays using human lung fibroblasts and DNA repair-deficient mouse embryonic fibroblasts, which demonstrated that zoledronic acid killed senescent cells with minimal effects on non-senescent cells. Next, in aged mice treated with zoledronic acid or vehicle for 8 weeks, zoledronic acid significantly reduced circulating SASP factors, including CCL7, IL-1β, TNFRSF1A, and TGFβ1 and improved grip strength. Analysis of publicly available RNAseq data from CD115+ (CSF1R/c-fms+) pre-osteoclastic cells isolated from mice treated with zoledronic acid demonstrated a significant downregulation of senescence/SASP genes (SenMayo). To establish that these cells are potential senolytic/senomorphic targets of zoledronic acid, we used single cell proteomic analysis (cytometry by time of flight [CyTOF]) and demonstrated that zoledronic acid significantly reduced the number of pre-osteoclastic (CD115+/CD3e-/Ly6G-/CD45R-) cells and decreased protein levels of p16, p21, and SASP markers in these cells without affecting other immune cell populations. Collectively, our findings demonstrate that zoledronic acid has senolytic effects in vitro and modulates senescence/SASP biomarkers in vivo. These data point to the need for additional studies testing zoledronic acid and/or other bisphosphonate derivatives for senotherapeutic efficacy.
Collapse
Affiliation(s)
- Parinya Samakkarnthai
- Division of Endocrinology, Mayo Clinic, Rochester, MN 55905, USA
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, MN 55905, USA
- Division of Endocrinology, Phramongkutklao Hospital and College of Medicine, Bangkok 10400, Thailand
| | - Dominik Saul
- Division of Endocrinology, Mayo Clinic, Rochester, MN 55905, USA
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, MN 55905, USA
- Department of Trauma and Reconstructive Surgery, Eberhard Karls University Tübingen, BG Trauma Center Tübingen, Tübingen 72076, Germany
| | - Lei Zhang
- Institute on the Biology of Aging and Metabolism, Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Minneapolis, MN 55455, USA
| | - Zaira Aversa
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, MN 55905, USA
- Department of Physical Medicine and Rehabilitation, Mayo Clinic, Rochester, MN 55905, USA
| | - Madison L. Doolittle
- Division of Endocrinology, Mayo Clinic, Rochester, MN 55905, USA
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, MN 55905, USA
| | - Jad G. Sfeir
- Division of Endocrinology, Mayo Clinic, Rochester, MN 55905, USA
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, MN 55905, USA
| | - Japneet Kaur
- Division of Endocrinology, Mayo Clinic, Rochester, MN 55905, USA
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, MN 55905, USA
| | | | - James R. Edwards
- Botnar Research Centre, Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Oxford, OX3 7FY, UK
| | - Graham G. Russell
- Botnar Research Centre, Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Oxford, OX3 7FY, UK
- Mellanby Centre for Musculoskeletal Research, University of Sheffield, Sheffield, S10 2RX, UK
| | - Robert J. Pignolo
- Division of Endocrinology, Mayo Clinic, Rochester, MN 55905, USA
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, MN 55905, USA
- Division of Hospital Internal Medicine, Mayo Clinic, Rochester, MN 55905, USA
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN 55905, USA
| | - James L. Kirkland
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN 55905, USA
- Division of General Internal Medicine, Mayo Clinic, Rochester, MN 55905, USA
| | - Tamar Tchkonia
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, MN 55905, USA
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN 55905, USA
- Division of General Internal Medicine, Mayo Clinic, Rochester, MN 55905, USA
| | - Laura J. Niedernhofer
- Institute on the Biology of Aging and Metabolism, Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Minneapolis, MN 55455, USA
| | - David G. Monroe
- Division of Endocrinology, Mayo Clinic, Rochester, MN 55905, USA
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, MN 55905, USA
| | - Nathan K. Lebrasseur
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, MN 55905, USA
- Department of Physical Medicine and Rehabilitation, Mayo Clinic, Rochester, MN 55905, USA
| | - Joshua N. Farr
- Division of Endocrinology, Mayo Clinic, Rochester, MN 55905, USA
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, MN 55905, USA
| | - Paul D. Robbins
- Institute on the Biology of Aging and Metabolism, Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Minneapolis, MN 55455, USA
| | - Sundeep Khosla
- Division of Endocrinology, Mayo Clinic, Rochester, MN 55905, USA
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, MN 55905, USA
| |
Collapse
|
124
|
Suda M, Paul KH, Minamino T, Miller JD, Lerman A, Ellison-Hughes GM, Tchkonia T, Kirkland JL. Senescent Cells: A Therapeutic Target in Cardiovascular Diseases. Cells 2023; 12:1296. [PMID: 37174697 PMCID: PMC10177324 DOI: 10.3390/cells12091296] [Citation(s) in RCA: 46] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Revised: 04/27/2023] [Accepted: 04/29/2023] [Indexed: 05/15/2023] Open
Abstract
Senescent cell accumulation has been observed in age-associated diseases including cardiovascular diseases. Senescent cells lack proliferative capacity and secrete senescence-associated secretory phenotype (SASP) factors that may cause or worsen many cardiovascular diseases. Therapies targeting senescent cells, especially senolytic drugs that selectively induce senescent cell removal, have been shown to delay, prevent, alleviate, or treat multiple age-associated diseases in preclinical models. Some senolytic clinical trials have already been completed or are underway for a number of diseases and geriatric syndromes. Understanding how cellular senescence affects the various cell types in the cardiovascular system, such as endothelial cells, vascular smooth muscle cells, fibroblasts, immune cells, progenitor cells, and cardiomyocytes, is important to facilitate translation of senotherapeutics into clinical interventions. This review highlights: (1) the characteristics of senescent cells and their involvement in cardiovascular diseases, focusing on the aforementioned cardiovascular cell types, (2) evidence about senolytic drugs and other senotherapeutics, and (3) the future path and clinical potential of senotherapeutics for cardiovascular diseases.
Collapse
Affiliation(s)
- Masayoshi Suda
- Department of Physiology and Biomedical Engineering, Mayo Clinic, 200 First St., S.W., Rochester, MN 55905, USA
- Department of Cardiovascular Biology and Medicine, Juntendo University Graduate School of Medicine, 3-1-3 Hongo, Bunkyo-ku, Tokyo 113-8421, Japan
| | - Karl H. Paul
- Department of Physiology and Biomedical Engineering, Mayo Clinic, 200 First St., S.W., Rochester, MN 55905, USA
- Department of Physiology and Pharmacology, Karolinska Institutet, Solnavägen 9, 171 65 Solna, Sweden
| | - Tohru Minamino
- Department of Cardiovascular Biology and Medicine, Juntendo University Graduate School of Medicine, 3-1-3 Hongo, Bunkyo-ku, Tokyo 113-8421, Japan
- Japan Agency for Medical Research and Development-Core Research for Evolutionary Medical Science and Technology (AMED-CREST), Japan Agency for Medical Research and Development, Tokyo 100-0004, Japan
| | - Jordan D. Miller
- Division of Cardiovascular Surgery, Mayo Clinic College of Medicine, 200 First St., S.W., Rochester, MN 55905, USA
| | - Amir Lerman
- Department of Cardiovascular Medicine, Mayo Clinic, 200 First St., S.W., Rochester, MN 55905, USA
| | - Georgina M. Ellison-Hughes
- Centre for Human and Applied Physiological Sciences, School of Basic and Medical Biosciences, Faculty of Life Sciences & Medicine, Guy’s Campus, King’s College London, London SE1 1UL, UK
- Centre for Stem Cells and Regenerative Medicine, School of Basic and Medical Biosciences, Faculty of Life Sciences & Medicine, Guy’s Campus, King’s College London, London SE1 1UL, UK
| | - Tamar Tchkonia
- Department of Physiology and Biomedical Engineering, Mayo Clinic, 200 First St., S.W., Rochester, MN 55905, USA
| | - James L. Kirkland
- Department of Physiology and Biomedical Engineering, Mayo Clinic, 200 First St., S.W., Rochester, MN 55905, USA
- Division of General Internal Medicine, Department of Medicine, Mayo Clinic, 200 First St., S.W., Rochester, MN 55905, USA
| |
Collapse
|
125
|
Knopp RC, Erickson MA, Rhea EM, Reed MJ, Banks WA. Cellular senescence and the blood-brain barrier: Implications for aging and age-related diseases. Exp Biol Med (Maywood) 2023; 248:399-411. [PMID: 37012666 PMCID: PMC10281623 DOI: 10.1177/15353702231157917] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/05/2023] Open
Abstract
The blood-brain barrier (BBB) is a critical physiochemical interface that regulates communication between the brain and blood. It is comprised of brain endothelial cells which regulate the BBB's barrier and interface properties and is surrounded by supportive brain cell types including pericytes and astrocytes. Recent reports have suggested that the BBB undergoes dysfunction during normative aging and in disease. In this review, we consider the effect of cellular senescence, one of the nine hallmarks of aging, on the BBB. We first characterize known normative age-related changes at the BBB, and then evaluate changes in neurodegenerative diseases, with an emphasis on if/how cellular senescence is influencing these changes. We then discuss what insight has been gained from in vitro and in vivo studies of cellular senescence at the BBB. Finally, we evaluate mechanisms by which cellular senescence in peripheral pathologies can indirectly or directly affect BBB function.
Collapse
Affiliation(s)
- Rachel C Knopp
- Veterans Affairs Puget Sound Health Care
System, Geriatrics Research Education and Clinical Center (GRECC), Seattle, WA 98108,
USA
- Department of Medicine, Division of
Gerontology and Geriatric Medicine, University of Washington School of Medicine, Seattle, WA
98195, USA
| | - Michelle A Erickson
- Veterans Affairs Puget Sound Health Care
System, Geriatrics Research Education and Clinical Center (GRECC), Seattle, WA 98108,
USA
- Department of Medicine, Division of
Gerontology and Geriatric Medicine, University of Washington School of Medicine, Seattle, WA
98195, USA
| | - Elizabeth M Rhea
- Veterans Affairs Puget Sound Health Care
System, Geriatrics Research Education and Clinical Center (GRECC), Seattle, WA 98108,
USA
- Department of Medicine, Division of
Gerontology and Geriatric Medicine, University of Washington School of Medicine, Seattle, WA
98195, USA
| | - May J Reed
- Veterans Affairs Puget Sound Health Care
System, Geriatrics Research Education and Clinical Center (GRECC), Seattle, WA 98108,
USA
- Department of Medicine, Division of
Gerontology and Geriatric Medicine, University of Washington School of Medicine, Seattle, WA
98195, USA
| | - William A Banks
- Veterans Affairs Puget Sound Health Care
System, Geriatrics Research Education and Clinical Center (GRECC), Seattle, WA 98108,
USA
- Department of Medicine, Division of
Gerontology and Geriatric Medicine, University of Washington School of Medicine, Seattle, WA
98195, USA
| |
Collapse
|
126
|
Budamagunta V, Kumar A, Rani A, Bean L, Manohar‐Sindhu S, Yang Y, Zhou D, Foster TC. Effect of peripheral cellular senescence on brain aging and cognitive decline. Aging Cell 2023; 22:e13817. [PMID: 36959691 PMCID: PMC10186609 DOI: 10.1111/acel.13817] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Revised: 02/22/2023] [Accepted: 02/24/2023] [Indexed: 03/25/2023] Open
Abstract
We examine similar and differential effects of two senolytic treatments, ABT-263 and dasatinib + quercetin (D + Q), in preserving cognition, markers of peripheral senescence, and markers of brain aging thought to underlie cognitive decline. Male F344 rats were treated from 12 to 18 months of age with D + Q, ABT-263, or vehicle, and were compared to young (6 months). Both senolytic treatments rescued memory, preserved the blood-brain barrier (BBB) integrity, and prevented the age-related decline in hippocampal N-methyl-D-aspartate receptor (NMDAR) function associated with impaired cognition. Senolytic treatments decreased senescence-associated secretory phenotype (SASP) and inflammatory cytokines/chemokines in the plasma (IL-1β, IP-10, and RANTES), with some markers more responsive to D + Q (TNFα) or ABT-263 (IFNγ, leptin, EGF). ABT-263 was more effective in decreasing senescence genes in the spleen. Both senolytic treatments decreased the expression of immune response and oxidative stress genes and increased the expression of synaptic genes in the dentate gyrus (DG). However, D + Q influenced twice as many genes as ABT-263. Relative to D + Q, the ABT-263 group exhibited increased expression of DG genes linked to cell death and negative regulation of apoptosis and microglial cell activation. Furthermore, D + Q was more effective at decreasing morphological markers of microglial activation. The results indicate that preserved cognition was associated with the removal of peripheral senescent cells, decreasing systemic inflammation that normally drives neuroinflammation, BBB breakdown, and impaired synaptic function. Dissimilarities associated with brain transcription indicate divergence in central mechanisms, possibly due to differential access.
Collapse
Affiliation(s)
- Vivekananda Budamagunta
- Department of Neuroscience, McKnight Brain InstituteUniversity of FloridaGainesvilleFloridaUSA
- Genetics and Genomics Graduate Program, Genetics InstituteUniversity of FloridaGainesvilleFloridaUSA
- Department of Pharmacodynamics, College of PharmacyUniversity of FloridaGainesvilleFloridaUSA
| | - Ashok Kumar
- Department of Neuroscience, McKnight Brain InstituteUniversity of FloridaGainesvilleFloridaUSA
| | - Asha Rani
- Department of Neuroscience, McKnight Brain InstituteUniversity of FloridaGainesvilleFloridaUSA
| | - Linda Bean
- Department of Neuroscience, McKnight Brain InstituteUniversity of FloridaGainesvilleFloridaUSA
| | - Sahana Manohar‐Sindhu
- Genetics and Genomics Graduate Program, Genetics InstituteUniversity of FloridaGainesvilleFloridaUSA
| | - Yang Yang
- Department of Pharmacodynamics, College of PharmacyUniversity of FloridaGainesvilleFloridaUSA
- Department of Biochemistry and Structural BiologyUniversity of Texas Health Science Center at San AntonioSan AntonioTexasUSA
| | - Daohong Zhou
- Department of Biochemistry and Structural BiologyUniversity of Texas Health Science Center at San AntonioSan AntonioTexasUSA
| | - Thomas C. Foster
- Department of Neuroscience, McKnight Brain InstituteUniversity of FloridaGainesvilleFloridaUSA
- Genetics and Genomics Graduate Program, Genetics InstituteUniversity of FloridaGainesvilleFloridaUSA
| |
Collapse
|
127
|
Torrance BL, Panier HA, Cadar AN, Martin DE, Lorenzo EC, Jellison ER, Bartley JM, Haynes L. Cellular Senescence is a Double-Edged Sword in Regulating Aged Immune Responses to Influenza. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.04.10.536027. [PMID: 37090607 PMCID: PMC10120665 DOI: 10.1101/2023.04.10.536027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/25/2023]
Abstract
Clearance of senescent cells has demonstrated therapeutic potential in the context of chronic age-related diseases. Little is known, however, how clearing senescent cells affects the ability to respond to an acute infection and form quality immunological memory. We aimed to probe the effects of clearing senescent cells in aged mice on the immune response to influenza (flu) infection. We utilized a p16 trimodality reporter mouse model (p16-3MR) to allow for identification and selective deletion of p16-expressing senescent cells upon administration of ganciclovir (GCV). While p16-expressing senescent cells may exacerbate dysfunctional responses to a primary infection, our data suggest they may play a role in fostering memory cell generation. We demonstrate that although deletion of p16-expressing cells enhanced viral clearance, this also severely limited antibody production in the lungs of flu-infected aged mice. 30 days later, there were fewer flu-specific CD8 memory T cells and lower levels of flu-specific antibodies in the lungs of GCV treated mice. GCV treated mice were unable to mount an optimal memory response and demonstrated increased viral load following a heterosubtypic challenge. These results suggest that targeting senescent cells may potentiate primary responses while limiting the ability to form durable and protective immune memory with age.
Collapse
|
128
|
Zheng Y, Deng J, Wang G, Zhang X, Wang L, Ma X, Dai Y, E L, Liu X, Zhang R, Zhang Y, Liu H. P53 negatively regulates the osteogenic differentiation in jaw bone marrow MSCs derived from diabetic osteoporosis. Heliyon 2023; 9:e15188. [PMID: 37096002 PMCID: PMC10121411 DOI: 10.1016/j.heliyon.2023.e15188] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2022] [Revised: 03/27/2023] [Accepted: 03/29/2023] [Indexed: 04/05/2023] Open
Abstract
Patients with diabetic osteoporosis (DOP) often suffer from poor osseointegration of artificial implants, which is a challenge that affects implant outcomes. The osteogenic differentiation ability of human jaw bone marrow mesenchymal stem cells (JBMMSCs) is the key to implant osseointegration. Studies have shown that the microenvironment of hyperglycemia affects the osteogenic differentiation of mesenchymal stem cells (MSC), but the mechanism is still unclear. Therefore, the aim of this study was to isolate and culture JBMMSCs from surgically derived bone fragments from DOP patients and control patients to investigate the differences in their osteogenic differentiation ability and to elucidate its mechanisms. The results showed that the osteogenic ability of hJBMMSCs was significantly decreased in the DOP environment. Mechanism study showed that the expression of senescence marker gene P53 was significantly increased in DOP hJBMMSCs compared to control hJBMMSCs according to RNA-sequencing result. Further, DOP hJBMMSCs were found to display significant senescence using β-galactosidase staining, mitochondrial membrane potential and ROS assay, qRT-PCR and WB analysis. Overexpression of P53 in hJBMMSCs, knockdown of P53 in DOP hJBMMSCs, and knockdown followed by overexpression of P53 significantly affected the osteogenic differentiation ability of hJBMMSCs. These results suggest that MSC senescence is an important reason for decreasing osteogenic capacity in DOP patients. P53 is a key target in regulating hJBMMSCs aging, and knocking down P53 can effectively restore the osteogenic differentiation ability of DOP hJBMMSCs and promote osteosynthesis in DOP dental implants. It provided a new idea to elucidate the pathogenesis and treatment of diabetic bone metabolic diseases.
Collapse
Affiliation(s)
- Ying Zheng
- Medical School of Chinese PLA, Beijing 100853, China
- Institute of Stomatology & Oral Maxilla Facial Key Laboratory, Department of Stomatology, First Medical Center of Chinese PLA General Hospital, Beijing, 100853, China
| | - Junhao Deng
- Senior Department of Orthopedics, The Fourth Medical Center of Chinese PLA General Hospital, Beijing, 100048, China
| | - Gang Wang
- Medical School of Chinese PLA, Beijing 100853, China
- Institute of Stomatology & Oral Maxilla Facial Key Laboratory, Department of Stomatology, First Medical Center of Chinese PLA General Hospital, Beijing, 100853, China
| | - Xiaru Zhang
- Department of Experimental Hematology and Biochemistry, Beijing Institute of Radiation Medicine, Beijing 100085, China
| | - Lin Wang
- Medical School of Chinese PLA, Beijing 100853, China
- Institute of Stomatology & Oral Maxilla Facial Key Laboratory, Department of Stomatology, First Medical Center of Chinese PLA General Hospital, Beijing, 100853, China
| | - Xiaocao Ma
- Institute of Stomatology & Oral Maxilla Facial Key Laboratory, Department of Stomatology, First Medical Center of Chinese PLA General Hospital, Beijing, 100853, China
| | - Yawen Dai
- Medical School of Chinese PLA, Beijing 100853, China
- Institute of Stomatology & Oral Maxilla Facial Key Laboratory, Department of Stomatology, First Medical Center of Chinese PLA General Hospital, Beijing, 100853, China
| | - Lingling E
- Institute of Stomatology & Oral Maxilla Facial Key Laboratory, Department of Stomatology, First Medical Center of Chinese PLA General Hospital, Beijing, 100853, China
| | - Xiangwei Liu
- Medical School of Chinese PLA, Beijing 100853, China
- Institute of Stomatology & Oral Maxilla Facial Key Laboratory, Department of Stomatology, First Medical Center of Chinese PLA General Hospital, Beijing, 100853, China
| | - Rong Zhang
- Institute of Stomatology & Oral Maxilla Facial Key Laboratory, Department of Stomatology, First Medical Center of Chinese PLA General Hospital, Beijing, 100853, China
| | - Yi Zhang
- Department of Experimental Hematology and Biochemistry, Beijing Institute of Radiation Medicine, Beijing 100085, China
- Corresponding author.
| | - Hongchen Liu
- Medical School of Chinese PLA, Beijing 100853, China
- Institute of Stomatology & Oral Maxilla Facial Key Laboratory, Department of Stomatology, First Medical Center of Chinese PLA General Hospital, Beijing, 100853, China
- Corresponding author. Medical School of Chinese PLA, Beijing 100853, China.
| |
Collapse
|
129
|
Hao W, Shan W, Wan F, Luo J, Niu Y, Zhou J, Zhang Y, Xu N, Xie W. Canagliflozin Delays Aging of HUVECs Induced by Palmitic Acid via the ROS/p38/JNK Pathway. Antioxidants (Basel) 2023; 12:antiox12040838. [PMID: 37107212 PMCID: PMC10135379 DOI: 10.3390/antiox12040838] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2023] [Revised: 03/19/2023] [Accepted: 03/28/2023] [Indexed: 04/29/2023] Open
Abstract
Vascular aging is an important factor contributing to cardiovascular diseases, such as hypertension and atherosclerosis. Hyperlipidemia or fatty accumulation may play an important role in vascular aging and cardiovascular diseases. Canagliflozin (CAN), a sodium-glucose cotransporter inhibitor, can exert a cardiovascular protection effect that is likely independent of its hypoglycemic activities; however, the exact mechanisms remain undetermined. We hypothesized that CAN might have protective effects on blood vessels by regulating vascular aging induced by hyperlipidemia or fatty accumulation in blood vessel walls. In this study, which was undertaken on the basis of aging and inflammation, we investigated the protective effects and mechanisms of CAN in human umbilical vein endothelial cells induced by palmitic acid. We found that CAN could delay vascular aging, reduce the secretion of the senescence-associated secretory phenotype (SASP) and protect DNA from damage, as well as exerting an effect on the cell cycle of senescent cells. These actions likely occur through the attenuation of the excess reactive oxygen species (ROS) produced in vascular endothelial cells and/or down-regulation of the p38/JNK signaling pathway. In summary, our study revealed a new role for CAN as one of the sodium-dependent glucose transporter 2 inhibitors in delaying lipotoxicity-induced vascular aging by targeting the ROS/p38/JNK pathway, giving new medicinal value to CAN and providing novel therapeutic ideas for delaying vascular aging in patients with dyslipidemia.
Collapse
Affiliation(s)
- Wenhui Hao
- State Key Laboratory of Chemical Oncogenomics, Shenzhen International Graduate School, Tsinghua University, Shenzhen 518055, China
- Shenzhen Key Laboratory of Health Science and Technology, Institute of Biopharmaceutical and Health, Tsinghua University, Shenzhen 518055, China
| | - Wenjie Shan
- State Key Laboratory of Chemical Oncogenomics, Shenzhen International Graduate School, Tsinghua University, Shenzhen 518055, China
- Shenzhen Key Laboratory of Health Science and Technology, Institute of Biopharmaceutical and Health, Tsinghua University, Shenzhen 518055, China
- Open FIESTA Center, Shenzhen International Graduate School, Tsinghua University, Shenzhen 518055, China
| | - Fang Wan
- State Key Laboratory of Chemical Oncogenomics, Shenzhen International Graduate School, Tsinghua University, Shenzhen 518055, China
- Shenzhen Key Laboratory of Health Science and Technology, Institute of Biopharmaceutical and Health, Tsinghua University, Shenzhen 518055, China
| | - Jingyi Luo
- State Key Laboratory of Chemical Oncogenomics, Shenzhen International Graduate School, Tsinghua University, Shenzhen 518055, China
- Shenzhen Key Laboratory of Health Science and Technology, Institute of Biopharmaceutical and Health, Tsinghua University, Shenzhen 518055, China
| | - Yaoyun Niu
- State Key Laboratory of Chemical Oncogenomics, Shenzhen International Graduate School, Tsinghua University, Shenzhen 518055, China
- Shenzhen Key Laboratory of Health Science and Technology, Institute of Biopharmaceutical and Health, Tsinghua University, Shenzhen 518055, China
| | - Jin Zhou
- Institute for Ocean Engineering, Shenzhen International Graduate School, Tsinghua University, Shenzhen 518055, China
| | - Yaou Zhang
- State Key Laboratory of Chemical Oncogenomics, Shenzhen International Graduate School, Tsinghua University, Shenzhen 518055, China
- Shenzhen Key Laboratory of Health Science and Technology, Institute of Biopharmaceutical and Health, Tsinghua University, Shenzhen 518055, China
| | - Naihan Xu
- State Key Laboratory of Chemical Oncogenomics, Shenzhen International Graduate School, Tsinghua University, Shenzhen 518055, China
- Shenzhen Key Laboratory of Health Science and Technology, Institute of Biopharmaceutical and Health, Tsinghua University, Shenzhen 518055, China
| | - Weidong Xie
- State Key Laboratory of Chemical Oncogenomics, Shenzhen International Graduate School, Tsinghua University, Shenzhen 518055, China
- Shenzhen Key Laboratory of Health Science and Technology, Institute of Biopharmaceutical and Health, Tsinghua University, Shenzhen 518055, China
| |
Collapse
|
130
|
Li Z, Tian M, Wang G, Cui X, Ma J, Liu S, Shen B, Liu F, Wu K, Xiao X, Zhu C. Senotherapeutics: An emerging approach to the treatment of viral infectious diseases in the elderly. Front Cell Infect Microbiol 2023; 13:1098712. [PMID: 37065192 PMCID: PMC10094634 DOI: 10.3389/fcimb.2023.1098712] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Accepted: 03/06/2023] [Indexed: 03/31/2023] Open
Abstract
In the context of the global COVID-19 pandemic, the phenomenon that the elderly have higher morbidity and mortality is of great concern. Existing evidence suggests that senescence and viral infection interact with each other. Viral infection can lead to the aggravation of senescence through multiple pathways, while virus-induced senescence combined with existing senescence in the elderly aggravates the severity of viral infections and promotes excessive age-related inflammation and multiple organ damage or dysfunction, ultimately resulting in higher mortality. The underlying mechanisms may involve mitochondrial dysfunction, abnormal activation of the cGAS-STING pathway and NLRP3 inflammasome, the role of pre-activated macrophages and over-recruited immune cells, and accumulation of immune cells with trained immunity. Thus, senescence-targeted drugs were shown to have positive effects on the treatment of viral infectious diseases in the elderly, which has received great attention and extensive research. Therefore, this review focused on the relationship between senescence and viral infection, as well as the significance of senotherapeutics for the treatment of viral infectious diseases.
Collapse
Affiliation(s)
- Zhiqiang Li
- Department of Clinical Laboratory, Institute of Translational Medicine, Renmin Hospital of Wuhan University, Wuhan, China
| | - Mingfu Tian
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan, China
| | - Guolei Wang
- Department of Clinical Laboratory, Institute of Translational Medicine, Renmin Hospital of Wuhan University, Wuhan, China
| | - Xianghua Cui
- Department of Clinical Laboratory, Institute of Translational Medicine, Renmin Hospital of Wuhan University, Wuhan, China
| | - Jun’e Ma
- Department of Clinical Laboratory, Institute of Translational Medicine, Renmin Hospital of Wuhan University, Wuhan, China
| | - Siyu Liu
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan, China
| | - Bingzheng Shen
- Department of Pharmacy, Renmin Hospital of Wuhan University, Wuhan, China
| | - Fang Liu
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan, China
| | - Kailang Wu
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan, China
| | - Xuan Xiao
- Department of Clinical Laboratory, Institute of Translational Medicine, Renmin Hospital of Wuhan University, Wuhan, China
- *Correspondence: Chengliang Zhu, ; Xuan Xiao,
| | - Chengliang Zhu
- Department of Clinical Laboratory, Institute of Translational Medicine, Renmin Hospital of Wuhan University, Wuhan, China
- *Correspondence: Chengliang Zhu, ; Xuan Xiao,
| |
Collapse
|
131
|
Khalil R, Diab-Assaf M, Lemaitre JM. Emerging Therapeutic Approaches to Target the Dark Side of Senescent Cells: New Hopes to Treat Aging as a Disease and to Delay Age-Related Pathologies. Cells 2023; 12:915. [PMID: 36980256 PMCID: PMC10047596 DOI: 10.3390/cells12060915] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2023] [Revised: 03/05/2023] [Accepted: 03/07/2023] [Indexed: 03/19/2023] Open
Abstract
Life expectancy has drastically increased over the last few decades worldwide, with important social and medical burdens and costs. To stay healthy longer and to avoid chronic disease have become essential issues. Organismal aging is a complex process that involves progressive destruction of tissue functionality and loss of regenerative capacity. One of the most important aging hallmarks is cellular senescence, which is a stable state of cell cycle arrest that occurs in response to cumulated cell stresses and damages. Cellular senescence is a physiological mechanism that has both beneficial and detrimental consequences. Senescence limits tumorigenesis, lifelong tissue damage, and is involved in different biological processes, such as morphogenesis, regeneration, and wound healing. However, in the elderly, senescent cells increasingly accumulate in several organs and secrete a combination of senescence associated factors, contributing to the development of various age-related diseases, including cancer. Several studies have revealed major molecular pathways controlling the senescent phenotype, as well as the ones regulating its interactions with the immune system. Attenuating the senescence-associated secretory phenotype (SASP) or eliminating senescent cells have emerged as attractive strategies aiming to reverse or delay the onset of aging diseases. Here, we review current senotherapies designed to suppress the deleterious effect of SASP by senomorphics or to selectively kill senescent cells by "senolytics" or by immune system-based approaches. These recent investigations are promising as radical new controls of aging pathologies and associated multimorbidities.
Collapse
Affiliation(s)
- Roula Khalil
- IRMB, University Montpellier, INSERM, 34090 Montpellier, France;
| | - Mona Diab-Assaf
- Fanar Faculty of Sciences II, Lebanese University, Beirut P.O. Box 90656, Lebanon;
| | | |
Collapse
|
132
|
Rodríguez-Agustín A, Casanova V, Grau-Expósito J, Sánchez-Palomino S, Alcamí J, Climent N. Immunomodulatory Activity of the Tyrosine Kinase Inhibitor Dasatinib to Elicit NK Cytotoxicity against Cancer, HIV Infection and Aging. Pharmaceutics 2023; 15:pharmaceutics15030917. [PMID: 36986778 PMCID: PMC10055786 DOI: 10.3390/pharmaceutics15030917] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2023] [Revised: 03/07/2023] [Accepted: 03/09/2023] [Indexed: 03/14/2023] Open
Abstract
Tyrosine kinase inhibitors (TKIs) have been extensively used as a treatment for chronic myeloid leukemia (CML). Dasatinib is a broad-spectrum TKI with off-target effects that give it an immunomodulatory capacity resulting in increased innate immune responses against cancerous cells and viral infected cells. Several studies reported that dasatinib expanded memory-like natural killer (NK) cells and γδ T cells that have been related with increased control of CML after treatment withdrawal. In the HIV infection setting, these innate cells are associated with virus control and protection, suggesting that dasatinib could have a potential role in improving both the CML and HIV outcomes. Moreover, dasatinib could also directly induce apoptosis of senescence cells, being a new potential senolytic drug. Here, we review in depth the current knowledge of virological and immunogenetic factors associated with the development of powerful cytotoxic responses associated with this drug. Besides, we will discuss the potential therapeutic role against CML, HIV infection and aging.
Collapse
Affiliation(s)
| | - Víctor Casanova
- HIV Unit, Hospital Clínic-IDIBAPS, University of Barcelona, 08036 Barcelona, Spain
| | - Judith Grau-Expósito
- HIV Unit, Hospital Clínic-IDIBAPS, University of Barcelona, 08036 Barcelona, Spain
| | - Sonsoles Sánchez-Palomino
- HIV Unit, Hospital Clínic-IDIBAPS, University of Barcelona, 08036 Barcelona, Spain
- CIBER of Infectious Diseases (CIBERINFEC), 28029 Madrid, Spain
| | - José Alcamí
- CIBER of Infectious Diseases (CIBERINFEC), 28029 Madrid, Spain
- AIDS Immunopathogenesis Unit, Instituto de Salud Carlos III (ISCIII), 28029 Madrid, Spain
| | - Núria Climent
- HIV Unit, Hospital Clínic-IDIBAPS, University of Barcelona, 08036 Barcelona, Spain
- CIBER of Infectious Diseases (CIBERINFEC), 28029 Madrid, Spain
- Correspondence: ; Tel.: +34-93-2275400 (ext. 3144); Fax: +34-93-2271775
| |
Collapse
|
133
|
Goldstein DR, Abdel-Latif A. Immune mechanisms of cardiac aging. THE JOURNAL OF CARDIOVASCULAR AGING 2023; 3:17. [PMID: 37092016 PMCID: PMC10121185 DOI: 10.20517/jca.2023.02] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/12/2023]
Abstract
Advances in healthcare and improvements in living conditions have led to rising life expectancy worldwide. Aging is associated with excessive oxidative stress, a chronic inflammatory state, and limited tissue healing, all of which result in an increased risk of heart failure. In fact, the prevalence of heart failure approaches 40% in the ninth decade of life, with the majority of these cases suffering from heart failure with preserved ejection fraction (HFpEF). In cardiomyocytes (CMs), age-related mitochondrial dysfunction results in disrupted calcium signaling and covalent protein-linked aggregates, which cause cardiomyocyte functional disturbances, resulting in increased stiffness and diastolic dysfunction. Importantly, aging is also associated with chronic low-grade, sterile inflammation, which alters the function of interstitial cardiac cells and leads to cardiac fibrosis. Taken together, cardiac aging is associated with cellular, structural, and functional changes in the heart that contribute to the rising prevalence of heart failure in older people.
Collapse
Affiliation(s)
- Daniel R. Goldstein
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI 48109, USA
- Department of Microbiology and Immunology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Ahmed Abdel-Latif
- Division of Cardiovascular Medicine, Department of Internal Medicine CVC, University of Michigan, Ann Arbor, MI 48109, USA
- Ann Arbor VA Healthcare System, Ann Arbor, MI 48109, USA
| |
Collapse
|
134
|
Chao CT, Hung KY. Vascular frailty, a proposal for new frailty type: A narrative review. Kaohsiung J Med Sci 2023; 39:318-325. [PMID: 36866657 DOI: 10.1002/kjm2.12664] [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: 01/31/2023] [Revised: 02/08/2023] [Accepted: 02/09/2023] [Indexed: 03/04/2023] Open
Abstract
Frailty is the incremental accumulation of minute defects that progressively impair health and performance. Frailty is commonly observed in older adults; however, secondary frailty may also occur in patients with metabolic disorders or major organ failure. In addition to physical frailty, several distinct types of frailty have been identified, including oral, cognitive, and social frailty, each of which is of practical importance. This nomenclature suggests that detailed descriptions of frailty can potentially advance relevant researches. In this narrative review, we first summarize the clinical value and plausible biological origin of frailty, as well as how to appropriately assess it using physical frailty phenotypes and frailty indexes. In the second part, we discuss the issue of vascular tissue as a relatively underappreciated organ whose pathologies contribute to the development of physical frailty. Moreover, when vascular tissue undergoes degeneration, it exhibits vulnerability to subtle injuries and manifests a unique phenotype amenable to clinical assessment prior to or accompanying physical frailty development. Finally, we propose that vascular frailty, based on an extensive set of experimental and clinical evidence, can be considered a new frailty type that requires our attention. We also outline potential methods for the operationalization of vascular frailty. Further studies are required to validate our claim and sharpen the spectrum of this degenerative phenotype.
Collapse
Affiliation(s)
- Chia-Ter Chao
- Nephrology Division, Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan.,Nephrology Division, Department of Internal Medicine, National Taiwan University Hospital BeiHu Branch, Taipei, Taiwan.,Nephrology Division, Department of Internal Medicine, National Taiwan University College of Medicine, Taipei, Taiwan.,Graduate Institute of Toxicology, National Taiwan University College of Medicine, Taipei, Taiwan
| | - Kuan-Yu Hung
- Nephrology Division, Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan.,Nephrology Division, Department of Internal Medicine, National Taiwan University College of Medicine, Taipei, Taiwan
| |
Collapse
|
135
|
Lino CA, de Oliveira-Silva T, Lunardon G, Balbino-Silva C, Lima VM, Huang ZP, Donato J, Takano APC, Barreto-Chaves ML, Wang DZ, Diniz GP. Ablation of miRNA-22 protects against obesity-induced adipocyte senescence and ameliorates metabolic disorders in middle-aged mice. Mech Ageing Dev 2023; 210:111775. [PMID: 36641038 DOI: 10.1016/j.mad.2023.111775] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Revised: 12/28/2022] [Accepted: 01/09/2023] [Indexed: 01/13/2023]
Abstract
High-fat diet (HFD) promotes obesity-related metabolic complications by activating cellular senescence in white adipose tissue (WAT). Growing evidence supports the importance of microRNA-22 (miR-22) in metabolic disorders and cellular senescence. Recently, we showed that miR-22 deletion attenuates obesity-related metabolic abnormalities. However, whether miR-22 mediates HFD-induced cellular senescence of WAT remains unknown. Here, we uncovered that obese mice displayed increased pri-miR-22 levels and cellular senescence in WAT. However, miR-22 ablation protected mice against HFD-induced WAT senescence. In addition, in vitro studies showed that miR-22 deletion prevented preadipocyte senescence in response to Doxorubicin (Doxo). Loss-of-function studies in vitro and in vivo revealed that miR-22 increases H2ax mRNA and γH2ax levels in preadipocytes and WAT without inducing DNA damage. Intriguingly, miR-22 ablation prevented HFD-induced increase in γH2ax levels and DNA damage in WAT. Similarly, miR-22 deletion prevented Doxo-induced increase in γH2ax levels in preadipocytes. Adipose miR-22 levels were enhanced in middle-aged mice fed a HFD than those found in young mice. Furthermore, miR-22 deletion attenuated fat mass gain and glucose imbalance induced by HFD in middle-aged mice. Overall, our findings indicate that miR-22 is a key regulator of obesity-induced WAT senescence and metabolic disorders in middle-aged mice.
Collapse
Affiliation(s)
- Caroline A Lino
- Department of Anatomy, Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo, Brazil
| | | | - Guilherme Lunardon
- Department of Anatomy, Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo, Brazil
| | - Camila Balbino-Silva
- Department of Anatomy, Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo, Brazil
| | - Vanessa M Lima
- Department of Anatomy, Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo, Brazil
| | - Zhan-Peng Huang
- Center for Translational Medicine, The First Affiliated Hospital, NHC Key Laboratory of Assisted Circulation, Sun Yat-sen University, Guangzhou, China
| | - Jose Donato
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo, Brazil
| | - Ana Paula C Takano
- Department of Anatomy, Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo, Brazil
| | | | - Da-Zhi Wang
- Department of Cardiology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA; Center for Regenerative Medicine, USF Health Heart Institute, University of South Florida, Tampa, FL, USA
| | - Gabriela P Diniz
- Department of Anatomy, Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo, Brazil.
| |
Collapse
|
136
|
Wang Y, Wu J, Wang J, He L, Lai H, Zhang T, Wang X, Li W. Mitochondrial oxidative stress in brain microvascular endothelial cells: Triggering blood-brain barrier disruption. Mitochondrion 2023; 69:71-82. [PMID: 36709855 DOI: 10.1016/j.mito.2023.01.007] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Revised: 01/02/2023] [Accepted: 01/22/2023] [Indexed: 01/27/2023]
Abstract
Blood-brain barrier disruption plays an important role in central nervous system diseases. This review provides information on the role of mitochondrial oxidative stress in brain microvascular endothelial cells in cellular dysfunction, the disruption of intercellular junctions, transporter dysfunction, abnormal angiogenesis, neurovascular decoupling, and the involvement and aggravation of vascular inflammation and illustrates related molecular mechanisms. In addition, recent drug and nondrug therapies targeting cerebral vascular endothelial cell mitochondria to repair the blood-brain barrier are discussed. This review shows that mitochondrial oxidative stress disorder in brain microvascular endothelial cells plays a key role in the occurrence and development of blood-brain barrier damage and may be critical in various pathological mechanisms of blood-brain barrier damage. These new findings suggest a potential new strategy for the treatment of central nervous system diseases through mitochondrial modulation of cerebral vascular endothelial cells.
Collapse
Affiliation(s)
- Yi Wang
- Basic Medical College, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan Province 610000, PR China.
| | - Jing Wu
- Basic Medical College, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan Province 610000, PR China.
| | - Jiexin Wang
- Basic Medical College, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan Province 610000, PR China.
| | - Linxi He
- Basic Medical College, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan Province 610000, PR China.
| | - Han Lai
- School of Foreign Languages, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan Province 610000, PR China.
| | - Tian Zhang
- Basic Medical College, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan Province 610000, PR China.
| | - Xin Wang
- Basic Medical College, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan Province 610000, PR China.
| | - Weihong Li
- Basic Medical College, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan Province 610000, PR China.
| |
Collapse
|
137
|
Meenakshi-Siddharthan DV, Livia C, Peterson TE, Stalboerger P, Attia ZI, Clavell AL, Friedman PA, Kapa S, Noseworthy PA, Schafer MJ, Stulak JM, Behfar A, Boilson BA. Artificial Intelligence-Derived Electrocardiogram Assessment of Cardiac Age and Molecular Markers of Senescence in Heart Failure. Mayo Clin Proc 2023; 98:372-385. [PMID: 36868745 DOI: 10.1016/j.mayocp.2022.10.026] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/06/2022] [Revised: 05/27/2022] [Accepted: 10/12/2022] [Indexed: 03/05/2023]
Abstract
OBJECTIVE To ascertain whether heart failure (HF) itself is a senescent phenomenon independent of age, and how this is reflected at a molecular level in the circulating progenitor cell niche, and at a substrate level using a novel electrocardiogram (ECG)-based artificial intelligence platform. PATIENTS AND METHODS Between October 14, 2016, and October 29, 2020, CD34+ progenitor cells were analyzed by flow cytometry and isolated by magnetic-activated cell sorting from patients of similar age with New York Heart Association functional classes IV (n = 17) and I-II (n = 10) heart failure with reduced ejection fraction and healthy controls (n = 10). CD34+ cellular senescence was quantitated by human telomerase reverse transcriptase expression and telomerase expression by quantitative polymerase chain reaction, and senescence-associated secretory phenotype (SASP) protein expression assayed in plasma. An ECG-based artificial intelligence (AI) algorithm was used to determine cardiac age and difference from chronological age (AI ECG age gap). RESULTS CD34+ counts and telomerase expression were significantly reduced and AI ECG age gap and SASP expression increased in all HF groups compared with healthy controls. Expression of SASP protein was closely associated with telomerase activity and severity of HF phenotype and inflammation. Telomerase activity was more closely associated with CD34+ cell counts and AI ECG age gap. CONCLUSION We conclude from this pilot study that HF may promote a senescent phenotype independent of chronological age. We show for the first time that the AI ECG in HF shows a phenotype of cardiac aging beyond chronological age, and appears to be associated with cellular and molecular evidence of senescence.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | - Marissa J Schafer
- Department of Physiology and Biomedical Engineering; Robert and Arlene Kogod Center on Aging
| | | | - Atta Behfar
- Van Cleve Cardiac Regeneration Program; Department of Cardiovascular Diseases
| | | |
Collapse
|
138
|
Zhang L, Pitcher LE, Prahalad V, Niedernhofer LJ, Robbins PD. Targeting cellular senescence with senotherapeutics: senolytics and senomorphics. FEBS J 2023; 290:1362-1383. [PMID: 35015337 DOI: 10.1111/febs.16350] [Citation(s) in RCA: 266] [Impact Index Per Article: 133.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2021] [Revised: 11/17/2021] [Accepted: 01/10/2022] [Indexed: 12/15/2022]
Abstract
The concept of geroscience is that since ageing is the greatest risk factor for many diseases and conditions, targeting the ageing process itself will have the greatest impact on human health. Of the hallmarks of ageing, cellular senescence has emerged as a druggable therapeutic target for extending healthspan in model organisms. Cellular senescence is a cell state of irreversible proliferative arrest driven by different types of stress, including oncogene-induced stress. Many senescent cells (SnCs) develop a senescent-associated secretory phenotype (SASP) comprising pro-inflammatory cytokines, chemokines, proteases, bioactive lipids, inhibitory molecules, extracellular vesicles, metabolites, lipids and other factors, able to promote chronic inflammation and tissue dysfunction. SnCs up-regulate senescent cell anti-apoptotic pathways (SCAPs) that prevent them from dying despite the accumulation of damage to DNA and other organelles. These SCAPs and other pathways altered in SnCs represent therapeutic targets for the development of senotherapeutic drugs that induce selective cell death of SnCs, specifically termed senolytics or suppress markers of senescence, in particular the SASP, termed senomorphics. Here, we review the current state of the development of senolytics and senomorphics for the treatment of age-related diseases and disorders and extension of healthy longevity. In addition, the challenges of documenting senolytic and senomorphic activity in pre-clinical models and the current state of the clinical application of the different senotherapeutics will be discussed.
Collapse
Affiliation(s)
- Lei Zhang
- Department of Biochemistry, Molecular Biology and Biophysics, Institute on the Biology of Aging and Metabolism, University of Minnesota, Minneapolis, MN, USA
| | - Louise E Pitcher
- Department of Biochemistry, Molecular Biology and Biophysics, Institute on the Biology of Aging and Metabolism, University of Minnesota, Minneapolis, MN, USA
| | - Vaishali Prahalad
- Department of Biochemistry, Molecular Biology and Biophysics, Institute on the Biology of Aging and Metabolism, University of Minnesota, Minneapolis, MN, USA
| | - Laura J Niedernhofer
- Department of Biochemistry, Molecular Biology and Biophysics, Institute on the Biology of Aging and Metabolism, University of Minnesota, Minneapolis, MN, USA
| | - Paul D Robbins
- Department of Biochemistry, Molecular Biology and Biophysics, Institute on the Biology of Aging and Metabolism, University of Minnesota, Minneapolis, MN, USA
| |
Collapse
|
139
|
Sweeney M, Cook SA, Gil J. Therapeutic opportunities for senolysis in cardiovascular disease. FEBS J 2023; 290:1235-1255. [PMID: 35015342 PMCID: PMC10952275 DOI: 10.1111/febs.16351] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2021] [Revised: 12/20/2021] [Accepted: 01/10/2022] [Indexed: 12/19/2022]
Abstract
Cellular senescence within the cardiovascular system has, until recently, been understudied and unappreciated as a factor in the development of age-related cardiovascular diseases such as heart failure, myocardial infarction and atherosclerosis. This is in part due to challenges with defining senescence within post-mitotic cells such as cardiomyocytes. However, recent evidence has demonstrated senescent-like changes, including a senescence-associated secretory phenotype (SASP), in cardiomyocytes in response to ageing and cell stress. Other replicating cells, including fibroblasts and vascular smooth muscle cells, within the cardiovascular system have also been shown to undergo senescence and contribute to disease pathogenesis. These findings coupled with the emergence of senolytic therapies, to target and eliminate senescent cells, have provided fascinating new avenues for management of several age-related cardiovascular diseases with high prevalence. In this review, we discuss the role of senescent cells within the cardiovascular system and highlight the contribution of senescence cells to common cardiovascular diseases. We discuss the emerging role for senolytics in cardiovascular disease management while highlighting important aspects of senescence biology which must be clarified before the potential of senolytics can be fully realized.
Collapse
Affiliation(s)
- Mark Sweeney
- MRC London Institute of Medical Sciences (LMS)LondonUK
- Institute of Clinical Sciences (ICS)Faculty of MedicineImperial College LondonUK
- Wellcome Trust / National Institute of Health Research 4i Clinical Research FellowLondonUK
| | - Stuart A. Cook
- MRC London Institute of Medical Sciences (LMS)LondonUK
- Institute of Clinical Sciences (ICS)Faculty of MedicineImperial College LondonUK
| | - Jesús Gil
- MRC London Institute of Medical Sciences (LMS)LondonUK
- Institute of Clinical Sciences (ICS)Faculty of MedicineImperial College LondonUK
| |
Collapse
|
140
|
Murray KO, Mahoney SA, Venkatasubramanian R, Seals DR, Clayton ZS. Aging, aerobic exercise, and cardiovascular health: Barriers, alternative strategies and future directions. Exp Gerontol 2023; 173:112105. [PMID: 36731386 PMCID: PMC10068966 DOI: 10.1016/j.exger.2023.112105] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Revised: 01/19/2023] [Accepted: 01/23/2023] [Indexed: 02/04/2023]
Abstract
Age-associated cardiovascular (CV) dysfunction, namely arterial dysfunction, is a key antecedent to the development of CV disease (CVD). Arterial dysfunction with aging is characterized by impaired vascular endothelial function and stiffening of the large elastic arteries, each of which is an independent predictor of CVD. These processes are largely mediated by an excess production of reactive oxygen species (ROS) and an increase in chronic, low-grade inflammation that ultimately leads to a reduction in bioavailability of the vasodilatory molecule nitric oxide. Additionally, there are other fundamental aging mechanisms that may contribute to excessive ROS and inflammation termed the "hallmarks of aging"; these additional mechanisms of arterial dysfunction may represent therapeutic targets for improving CV health with aging. Aerobic exercise is the most well-known and effective intervention to prevent and treat the effects of aging on CV dysfunction. However, the majority of mid-life and older (ML/O) adults do not meet recommended exercise guidelines due to traditional barriers to aerobic exercise, such as reduced leisure time, motivation, or access to fitness facilities. Therefore, it is a biomedical research priority to develop and implement time- and resource-efficient alternative strategies to aerobic exercise to reduce the burden of CVD in ML/O adults. Alternative strategies that mimic or are inspired by aerobic exercise, that target pathways specific to the fundamental mechanisms of aging, represent a promising approach to accomplish this goal.
Collapse
Affiliation(s)
- Kevin O Murray
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO, United States of America
| | - Sophia A Mahoney
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO, United States of America
| | | | - Douglas R Seals
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO, United States of America
| | - Zachary S Clayton
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO, United States of America.
| |
Collapse
|
141
|
Samakkarnthai P, Saul D, Zhang L, Aversa Z, Doolittle ML, Sfeir JG, Kaur J, Atkinson EJ, Edwards JR, Russell RGG, Pignolo RJ, Kirkland JL, Tchkonia T, Niedernhofer LJ, Monroe DG, LeBrasseur NK, Farr JN, Robbins PD, Khosla S. In vitro and in vivo effects of zoledronate on senescence and senescence-associated secretory phenotype markers. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.02.23.529777. [PMID: 36865244 PMCID: PMC9980119 DOI: 10.1101/2023.02.23.529777] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/02/2023]
Abstract
In addition to reducing fracture risk, zoledronate has been found in some studies to decrease mortality in humans and extend lifespan and healthspan in animals. Because senescent cells accumulate with aging and contribute to multiple co-morbidities, the non-skeletal actions of zoledronate could be due to senolytic (killing of senescent cells) or senomorphic (inhibition of the secretion of the senescence-associated secretory phenotype [SASP]) actions. To test this, we first performed in vitro senescence assays using human lung fibroblasts and DNA repair-deficient mouse embryonic fibroblasts, which demonstrated that zoledronate killed senescent cells with minimal effects on non-senescent cells. Next, in aged mice treated with zoledronate or vehicle for 8 weeks, zoledronate significantly reduced circulating SASP factors, including CCL7, IL-1β, TNFRSF1A, and TGFβ1 and improved grip strength. Analysis of publicly available RNAseq data from CD115+ (CSF1R/c-fms+) pre-osteoclastic cells isolated from mice treated with zoledronate demonstrated a significant downregulation of senescence/SASP genes (SenMayo). To establish that these cells are potential senolytic/senomorphic targets of zoledronate, we used single cell proteomic analysis (cytometry by time of flight [CyTOF]) and demonstrated that zoledronate significantly reduced the number of pre-osteoclastic (CD115+/CD3e-/Ly6G-/CD45R-) cells and decreased protein levels of p16, p21, and SASP markers in these cells without affecting other immune cell populations. Collectively, our findings demonstrate that zoledronate has senolytic effects in vitro and modulates senescence/SASP biomarkers in vivo . These data point to the need for additional studies testing zoledronate and/or other bisphosphonate derivatives for senotherapeutic efficacy.
Collapse
|
142
|
Thong L, McElduff EJ, Henry MT. Trials and Treatments: An Update on Pharmacotherapy for Idiopathic Pulmonary Fibrosis. Life (Basel) 2023; 13:486. [PMID: 36836843 PMCID: PMC9963632 DOI: 10.3390/life13020486] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Revised: 02/07/2023] [Accepted: 02/08/2023] [Indexed: 02/12/2023] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) is a chronic and progressive fibrosing interstitial lung disease that occurs predominantly in the older population. There is increasing incidence and prevalence in IPF globally. The emergence of anti-fibrotic therapies in the last decade have improved patient survival though a cure is yet to be developed. In this review article, we aim to summarize the existing and novel pharmacotherapies for the treatment of IPF (excluding treatments for acute exacerbations), focusing on the current knowledge on the pathophysiology of the disease, mechanism of action of the drugs, and clinical trials.
Collapse
Affiliation(s)
- Lorraine Thong
- Department of Clinical Medicine, Trinity College Dublin, D08 W9RT Dublin, Ireland
| | - Enda James McElduff
- Department of Clinical Medicine, Royal College of Surgeons Ireland, D02 YN77 Dublin, Ireland
| | - Michael Thomas Henry
- Department of Respiratory Medicine, Cork University Hospital, T12 YE02 Cork, Ireland
| |
Collapse
|
143
|
Ruggiero AD, Davis MA, Davis AT, DeStephanis D, Williams AG, Vemuri R, Fanning KM, Sherrill C, Cline JM, Caudell DL, Kavanagh K. Delayed effects of radiation in adipose tissue reflect progenitor damage and not cellular senescence. GeroScience 2023; 45:507-521. [PMID: 36136223 PMCID: PMC9886706 DOI: 10.1007/s11357-022-00660-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2022] [Accepted: 09/08/2022] [Indexed: 02/03/2023] Open
Abstract
The pathogenesis of many age-related diseases is linked to cellular senescence, a state of inflammation-inducing, irreversible cell cycle arrest. The consequences and mechanisms of age-associated cellular senescence are often studied using in vivo models of radiation exposure. However, it is unknown whether radiation induces persistent senescence, like that observed in ageing. We performed analogous studies in mice and monkeys, where young mice and rhesus macaques received sub-lethal doses of ionizing radiation and were observed for ~ 15% of their expected lifespan. Assessments of 8-hydroxy-2' -deoxyguanosine (8-OHdG), senescence-associated beta-galactosidase (SAβ-gal), and p16Ink4a and p21 were performed on mitotic and post-mitotic tissues - liver and adipose tissue - 6 months and 3 years post-exposure for the mice and monkeys, respectively. No elevations in 8-OHdG, SA-βgal staining, or p16 Ink4a or p21 gene or protein expression were found in mouse and monkey liver or adipose tissue compared to control animals. Despite no evidence of senescence, progenitor cell dysfunction persisted after radiation exposure, as indicated by lower in situ CD34+ adipose cells (p = 0.03), and deficient adipose stromal vascular cell proliferation (p < 0.05) and differentiation (p = 0.04) ex vivo. Our investigation cautions that employing radiation to study senescence-related processes should be limited to the acute post-exposure period and that stem cell damage likely underpins the dysfunction associated with delayed effects of radiation.
Collapse
Affiliation(s)
- Alistaire D Ruggiero
- Department of Pathology, Wake Forest University School of Medicine, 575 N. Patterson Ave, Winston-Salem, NC, 27101, USA
| | - Matthew A Davis
- Department of Internal Medicine, Wake Forest University School of Medicine, Winston-Salem, NC, USA
| | - Ashley T Davis
- Department of Pathology, Wake Forest University School of Medicine, 575 N. Patterson Ave, Winston-Salem, NC, 27101, USA
| | - Darla DeStephanis
- Department of Pathology, Wake Forest University School of Medicine, 575 N. Patterson Ave, Winston-Salem, NC, 27101, USA
| | - Abigail G Williams
- Department of Pathology, Wake Forest University School of Medicine, 575 N. Patterson Ave, Winston-Salem, NC, 27101, USA
| | - Ravichandra Vemuri
- Department of Pathology, Wake Forest University School of Medicine, 575 N. Patterson Ave, Winston-Salem, NC, 27101, USA
| | - Katherine M Fanning
- Department of Pathology, Wake Forest University School of Medicine, 575 N. Patterson Ave, Winston-Salem, NC, 27101, USA
| | - Chrissy Sherrill
- Department of Pathology, Wake Forest University School of Medicine, 575 N. Patterson Ave, Winston-Salem, NC, 27101, USA
| | - J Mark Cline
- Department of Pathology, Wake Forest University School of Medicine, 575 N. Patterson Ave, Winston-Salem, NC, 27101, USA
| | - David L Caudell
- Department of Pathology, Wake Forest University School of Medicine, 575 N. Patterson Ave, Winston-Salem, NC, 27101, USA
| | - Kylie Kavanagh
- Department of Pathology, Wake Forest University School of Medicine, 575 N. Patterson Ave, Winston-Salem, NC, 27101, USA.
- College of Health and Medicine, University of Tasmania, Hobart, Australia.
| |
Collapse
|
144
|
Dugan B, Conway J, Duggal NA. Inflammaging as a target for healthy ageing. Age Ageing 2023; 52:7024516. [PMID: 36735849 DOI: 10.1093/ageing/afac328] [Citation(s) in RCA: 51] [Impact Index Per Article: 25.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2022] [Revised: 11/11/2022] [Indexed: 02/05/2023] Open
Abstract
Life expectancy has been on the rise for the past few decades, but healthy life expectancy has not kept pace, leading to a global burden of age-associated disorders. Advancing age is accompanied by a chronic increase in basal systemic inflammation, termed inflammaging, contributing towards an increased risk of developing chronic diseases in old age. This article reviews the recent literature to formulate hypotheses regarding how age-associated inflammaging plays a crucial role in driving chronic diseases and ill health in older adults. Here, we discuss how non-pharmacological intervention strategies (diet, nutraceutical supplements, phytochemicals, physical activity, microbiome-based therapies) targeting inflammaging restore health in older adults. We also consider alternative existing pharmacological interventions (Caloric restriction mimetics, p38 mitogen-activated protein kinase inhibitors) and explore novel targets (senolytics) aimed at combating inflammaging and optimising the ageing process to increase healthy lifespan.
Collapse
Affiliation(s)
- Ben Dugan
- Institute of Inflammation and Ageing, University of Birmingham, Birmingham, UK.,MRC Versus Arthritis Centre for Musculoskeletal Ageing Research, University of Birmingham, Birmingham, UK
| | - Jessica Conway
- Institute of Inflammation and Ageing, University of Birmingham, Birmingham, UK.,MRC Versus Arthritis Centre for Musculoskeletal Ageing Research, University of Birmingham, Birmingham, UK
| | - Niharika A Duggal
- Institute of Inflammation and Ageing, University of Birmingham, Birmingham, UK.,MRC Versus Arthritis Centre for Musculoskeletal Ageing Research, University of Birmingham, Birmingham, UK
| |
Collapse
|
145
|
Bone-targeted delivery of senolytics to eliminate senescent cells increases bone formation in senile osteoporosis. Acta Biomater 2023; 157:352-366. [PMID: 36470392 DOI: 10.1016/j.actbio.2022.11.056] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2022] [Revised: 10/31/2022] [Accepted: 11/28/2022] [Indexed: 12/12/2022]
Abstract
Systemic elimination of senescent cells using senolytic drugs presents therapeutic effects on age-related diseases, including senile osteoporosis. However, low bioavailability and potential side effects of senolytics restrict clinical application. Therefore, we developed a bone-targeted delivery system for senolytics to effective treatment of senile osteoporosis. In this study, quercetin was screened out as the ideal senolytics for eliminating senescent BMSCs. Treatment of quercetin efficiently decreased the senescence markers in senescent BMSCs models. After treatment with quercetin in vitro, cell mitosis and calcification staining assay confirmed that the proliferation and osteogenesis of the senescent BMSCs populations were enhanced. To enhance the effectiveness and minimize the side effect of treatment, liposomes decorated with bone affinity peptide (DSS)6 were constructed for bone-targeted delivery of quercetin. After administration of liposomes loading quercetin in two aged mice models, histological and cellular analysis confirmed that bone-targeted treatment with quercetin efficiently eliminated senescent cells in bone, restored the function of BMCSs, and promoted bone formation in aged mice models when compared to non-targeted treatment. Taken together, the bone-targeted delivery of senolytics efficiently eliminates senescent cells to recover bone mass and microarchitecture, showing an effective treatment for senile osteoporosis. STATEMENT OF SIGNIFICANCE: Senile osteoporosis, a common and hazardous chronic disease, has been still lacking effective therapy. How to effectively eliminate the hazards of senescent cells in skeleton to bone formation remains challenge. In this study, quercetin was screened out as the ideal senolytic drug for senescent BMSCs and could effectively eliminated senescent BMSCs to restore the cellular functions of senescent BMSCs models in vitro. Then, the bone-targeted liposomes were designed to encapsulate and deliver senolytics efficiently to senile bone tissue. Based on two aged mice models, we confirmed that bone-targeted delivery of quercetin efficiently eliminated senescent cells in skeleton and enhanced bone formation in vivo, suggesting the bone-targeted elimination of senescent cells is an effective treatment for senile osteoporosis.
Collapse
|
146
|
Song J, Farris D, Ariza P, Moorjani S, Varghese M, Blin M, Chen J, Tyrrell D, Zhang M, Singer K, Salmon M, Goldstein DR. Age-associated adipose tissue inflammation promotes monocyte chemotaxis and enhances atherosclerosis. Aging Cell 2023; 22:e13783. [PMID: 36683460 PMCID: PMC9924943 DOI: 10.1111/acel.13783] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2022] [Revised: 10/31/2022] [Accepted: 01/05/2023] [Indexed: 01/24/2023] Open
Abstract
Although aging enhances atherosclerosis, we do not know if this occurs via alterations in circulating immune cells, lipid metabolism, vasculature, or adipose tissue. Here, we examined whether aging exerts a direct pro-atherogenic effect on adipose tissue in mice. After demonstrating that aging augmented the inflammatory profile of visceral but not subcutaneous adipose tissue, we transplanted visceral fat from young or aged mice onto the right carotid artery of Ldlr-/- recipients. Aged fat transplants not only increased atherosclerotic plaque size with increased macrophage numbers in the adjacent carotid artery, but also in distal vascular territories, indicating that aging of the adipose tissue enhances atherosclerosis via secreted factors. By depleting macrophages from the visceral fat, we identified that adipose tissue macrophages are major contributors of the secreted factors. To identify these inflammatory factors, we found that aged fat transplants secreted increased levels of the inflammatory mediators TNFα, CXCL2, and CCL2, which synergized to promote monocyte chemotaxis. Importantly, the combined blockade of these inflammatory mediators impeded the ability of aged fat transplants to enhance atherosclerosis. In conclusion, our study reveals that aging enhances atherosclerosis via increased inflammation of visceral fat. Our study suggests that future therapies targeting the visceral fat may reduce atherosclerosis disease burden in the expanding older population.
Collapse
Affiliation(s)
- Jianrui Song
- Department of Internal Medicine, Division of Cardiovascular MedicineUniversity of MichiganAnn ArborMichiganUSA
| | - Diana Farris
- Department of Internal Medicine, Division of Cardiovascular MedicineUniversity of MichiganAnn ArborMichiganUSA
| | - Paola Ariza
- Department of Internal Medicine, Division of Cardiovascular MedicineUniversity of MichiganAnn ArborMichiganUSA
| | - Smriti Moorjani
- Department of Internal Medicine, Division of Cardiovascular MedicineUniversity of MichiganAnn ArborMichiganUSA
| | - Mita Varghese
- Department of Pediatrics, Division of EndocrinologyUniversity of MichiganAnn ArborMichiganUSA
| | - Muriel Blin
- Department of Internal Medicine, Division of Cardiovascular MedicineUniversity of MichiganAnn ArborMichiganUSA
| | - Judy Chen
- Department of Internal Medicine, Division of Cardiovascular MedicineUniversity of MichiganAnn ArborMichiganUSA
- Graduate Program in ImmunologyUniversity of MichiganAnn ArborMichiganUSA
| | - Daniel Tyrrell
- Department of Internal Medicine, Division of Cardiovascular MedicineUniversity of MichiganAnn ArborMichiganUSA
| | - Min Zhang
- Department of BiostatisticsUniversity of MichiganAnn ArborMichiganUSA
| | - Kanakadurga Singer
- Department of Pediatrics, Division of EndocrinologyUniversity of MichiganAnn ArborMichiganUSA
- Graduate Program in ImmunologyUniversity of MichiganAnn ArborMichiganUSA
| | - Morgan Salmon
- Department of Cardiac SurgeryUniversity of MichiganAnn ArborMichiganUSA
| | - Daniel R. Goldstein
- Department of Internal Medicine, Division of Cardiovascular MedicineUniversity of MichiganAnn ArborMichiganUSA
- Graduate Program in ImmunologyUniversity of MichiganAnn ArborMichiganUSA
- Department of Microbiology and ImmunologyUniversity of MichiganAnn ArborMichiganUSA
| |
Collapse
|
147
|
Yang N, Nakagawa M, Nishiura A, Yamada M, Morikuni H, Honda Y, Matsumoto N. Identification of Senescent Cells in Peri-Implantitis and Prevention of Mini-Implant Loss Using Senolytics. Int J Mol Sci 2023; 24:ijms24032507. [PMID: 36768829 PMCID: PMC9916936 DOI: 10.3390/ijms24032507] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Revised: 12/05/2022] [Accepted: 12/16/2022] [Indexed: 02/03/2023] Open
Abstract
Peri-implantitis is a disease that causes the detachment of orthodontic mini-implants. Recently, stress-induced senescent cells have been reported to be involved in various inflammatory diseases. Senescent cell-eliminating drugs, termed "senolytics", can improve the symptoms of such diseases. However, the relationship between peri-implantitis and senescent cells remains unclear. In this study, we evaluated the presence of senescent cells in a rat peri-implantitis model developed with a gum ring. The effect on bone resorption and implant loss was also investigated with and without senolytics (Dasatinib and Quercetin). The number of senescence markers (p19, p21, and p16) was found to increase, and implant detachment occurred in 24 days. After the administration of senolytics, the number of senescence markers decreased and implant detachment was inhibited. This study suggests that senescent cells aggravate peri-implantitis and senolytic administration latently reduces implant loss by inhibiting senescence-related mechanisms.
Collapse
Affiliation(s)
- Niuxin Yang
- Department of Orthodontics, Osaka Dental University, 8-1 Kuzuhahanazonocho, Hirakata 573-1121, Japan
| | - Masato Nakagawa
- Department of Oral Anatomy, Osaka Dental University, 8-1 Kuzuhahanazonocho, Hirakata 573-1121, Japan
- Correspondence: (M.N.); (Y.H.); Tel.: +81-90-1675-7895 (M.N.); +81-72-864-3013 (Y.H.)
| | - Aki Nishiura
- Department of Orthodontics, Osaka Dental University, 8-1 Kuzuhahanazonocho, Hirakata 573-1121, Japan
| | - Masahiro Yamada
- Division of Molecular and Regenerative Prosthodontics, Tohoku University Graduate School of Dentistry, 4-1 Seiryomachi, Sendai 980-8575, Japan
| | - Hidetoshi Morikuni
- Department of Orthodontics, Osaka Dental University, 8-1 Kuzuhahanazonocho, Hirakata 573-1121, Japan
| | - Yoshitomo Honda
- Department of Oral Anatomy, Osaka Dental University, 8-1 Kuzuhahanazonocho, Hirakata 573-1121, Japan
- Correspondence: (M.N.); (Y.H.); Tel.: +81-90-1675-7895 (M.N.); +81-72-864-3013 (Y.H.)
| | - Naoyuki Matsumoto
- Department of Orthodontics, Osaka Dental University, 8-1 Kuzuhahanazonocho, Hirakata 573-1121, Japan
| |
Collapse
|
148
|
McGovern KE, Sonar SA, Watanabe M, Coplen CP, Bradshaw CM, Nikolich JŽ. The aging of the immune system and its implications for transplantation. GeroScience 2023:10.1007/s11357-022-00720-2. [PMID: 36626019 PMCID: PMC9838392 DOI: 10.1007/s11357-022-00720-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2022] [Accepted: 12/21/2022] [Indexed: 01/11/2023] Open
Abstract
By the last third of life, most mammals, including humans, exhibit a decline in immune cell numbers, immune organ structure, and immune defense of the organism, commonly known as immunosenescence. This decline leads to clinical manifestations of increased susceptibility to infections, particularly those caused by emerging and reemerging microorganisms, which can reach staggering levels-infection with SARS-CoV-2 has been 270-fold more lethal to older adults over 80 years of age, compared to their 18-39-year-old counterparts. However, while this would be expected to be beneficial to situations where hyporeactivity of the immune system may be desirable, this is not always the case. Here, we discuss the cellular and molecular underpinnings of immunosenescence as they pertain to outcomes of solid organ and hematopoietic transplantation.
Collapse
Affiliation(s)
- Kathryn E McGovern
- Department of Immunobiology, University of Arizona, Tucson, AZ, 85724, USA
- Arizona Center On Aging, The University of Arizona, University of Arizona College of Medicine-Tucson, Tucson, AZ, 85724, USA
- BIO5 Institute, University of Arizona, Tucson, AZ, USA
| | - Sandip A Sonar
- Department of Immunobiology, University of Arizona, Tucson, AZ, 85724, USA
- Arizona Center On Aging, The University of Arizona, University of Arizona College of Medicine-Tucson, Tucson, AZ, 85724, USA
| | - Makiko Watanabe
- Department of Immunobiology, University of Arizona, Tucson, AZ, 85724, USA
- Arizona Center On Aging, The University of Arizona, University of Arizona College of Medicine-Tucson, Tucson, AZ, 85724, USA
| | - Christopher P Coplen
- Department of Immunobiology, University of Arizona, Tucson, AZ, 85724, USA
- Arizona Center On Aging, The University of Arizona, University of Arizona College of Medicine-Tucson, Tucson, AZ, 85724, USA
| | - Christine M Bradshaw
- Department of Immunobiology, University of Arizona, Tucson, AZ, 85724, USA
- Arizona Center On Aging, The University of Arizona, University of Arizona College of Medicine-Tucson, Tucson, AZ, 85724, USA
| | - Janko Ž Nikolich
- Department of Immunobiology, University of Arizona, Tucson, AZ, 85724, USA.
- Arizona Center On Aging, The University of Arizona, University of Arizona College of Medicine-Tucson, Tucson, AZ, 85724, USA.
- BIO5 Institute, University of Arizona, Tucson, AZ, USA.
- The Aegis Consortium for Pandemic-free Future, University of Arizona Health Sciences, University of Arizona, Tucson, 85719, USA.
| |
Collapse
|
149
|
Seara FAC, Maciel L, Kasai-Brunswick TH, Nascimento JHM, Campos-de-Carvalho AC. Extracellular Vesicles and Cardiac Aging. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2023; 1418:33-56. [PMID: 37603271 DOI: 10.1007/978-981-99-1443-2_3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/22/2023]
Abstract
Global population aging is a major challenge to health and socioeconomic policies. The prevalence of diseases progressively increases with aging, with cardiovascular disease being the major cause of mortality among elderly people. The allostatic overload imposed by the accumulation of cardiac senescent cells has been suggested to play a pivotal role in the aging-related deterioration of cardiovascular function. Senescent cells exhibit intrinsic disorders and release a senescence-associated secretory phenotype (SASP). Most of these SASP compounds and damaged molecules are released from senescent cells by extracellular vesicles (EVs). Once secreted, these EVs can be readily incorporated by recipient neighboring cells and elicit cellular damage or otherwise can promote extracellular matrix remodeling. This has been associated with the development of cardiac dysfunction, fibrosis, and vascular calcification, among others. The molecular signature of these EVs is highly variable and might provide important information for the development of aging-related biomarkers. Conversely, EVs released by the stem and progenitor cells can exert a rejuvenating effect, raising the possibility of future anti-aging therapies.
Collapse
Affiliation(s)
- Fernando A C Seara
- Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
- Department of Physiological Sciences, Institute of Health and Biological Sciences, Federal Rural University of Rio de Janeiro, Seropédica, Brazil
| | - Leonardo Maciel
- Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
- Federal University of Rio de Janeiro, Campus Professor Geraldo, Duque de Caxias, Brazil
| | - Tais Hanae Kasai-Brunswick
- Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
- National Center of Structural Biology and Bioimaging, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Jose H M Nascimento
- Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil.
- Laboratory of Cardiac Electrophysiology, Carlos Chagas Filho Institute of Biophysics, Health Sciences Centre, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil.
| | - Antonio C Campos-de-Carvalho
- Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
- National Center of Structural Biology and Bioimaging, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| |
Collapse
|
150
|
Abstract
Cellular senescence has become a subject of great interest within the ageing research field over the last 60 years, from the first observation in vitro by Leonard Hayflick and Paul Moorhead in 1961, to novel findings of phenotypic sub-types and senescence-like phenotype in post-mitotic cells. It has essential roles in wound healing, tumour suppression and the very first stages of human development, while causing widespread damage and dysfunction with age leading to a raft of age-related diseases. This chapter discusses these roles and their interlinking pathways, and how the observed accumulation of senescent cells with age has initiated a whole new field of ageing research, covering pathologies in the heart, liver, kidneys, muscles, brain and bone. This chapter will also examine how senescent cell accumulation presents in these different tissues, along with their roles in disease development. Finally, there is much focus on developing treatments for senescent cell accumulation in advanced age as a method of alleviating age-related disease. We will discuss here the various senolytic and senostatic treatment approaches and their successes and limitations, and the innovative new strategies being developed to address the differing effects of cellular senescence in ageing and disease.
Collapse
Affiliation(s)
- Rebecca Reed
- Biosciences Institute, Faculty of Medical Sciences, Campus for Ageing and Vitality, Newcastle University, Newcastle upon Tyne, UK
| | - Satomi Miwa
- Biosciences Institute, Faculty of Medical Sciences, Campus for Ageing and Vitality, Newcastle University, Newcastle upon Tyne, UK.
| |
Collapse
|