351
|
Ren H, Guo Z, Liu Y, Song C. Stem Cell-derived Exosomal MicroRNA as Therapy for Vascular Age-related Diseases. Aging Dis 2022; 13:852-867. [PMID: 35656114 PMCID: PMC9116915 DOI: 10.14336/ad.2021.1110] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2021] [Accepted: 11/10/2021] [Indexed: 12/20/2022] Open
Abstract
Vascular age-related diseases describe a group of age-related chronic diseases that result in a considerable healthcare burden to society. Vascular aging includes structural changes and dysfunctions of endothelial cells (ECs) and smooth muscle cells (SMCs) in blood vessels. Compared with conventional treatment for vascular age-related diseases, stem cell (SC) therapy elicits better anti-aging effects viathe inhibition/delay ECs and SMCs from entering senescence. Exosomal noncoding RNA (ncRNAs) in vascular aging and stem cell-derived exosomal microRNAs (SCEV-miRNAs), especially in mesenchymal stem cells, have an important role in the development of age-related diseases. This review summarizes SCEV-miRNAs of diverse origins that may play a vital role in treating subclinical and clinical stages of vascular age-related disorders. We further explored possible age-related pathways and molecular targets of SCEV-miRNA, which are associated with dysfunctions of ECs and SMCs in the senescent stage. Moreover, the perspectives and difficulties of SCEV-miRNA clinical translation are discussed. This review aims to provide greater understanding of the biology of vascular aging and to identify critical therapeutic targets for SCEV-miRNAs. Though still in its infancy, the potential value of SCEV-miRNAs for vascular age-related diseases is clear.
Collapse
Affiliation(s)
- Hang Ren
- Department of Cardiovascular Internal Medicine, the Second Hospital of Jilin University, Changchun, China
| | - Ziyuan Guo
- Department of Cardiovascular Internal Medicine, the Second Hospital of Jilin University, Changchun, China
| | - Yang Liu
- Department of Cardiovascular Internal Medicine, the Second Hospital of Jilin University, Changchun, China
| | - Chunli Song
- Department of Cardiovascular Internal Medicine, the Second Hospital of Jilin University, Changchun, China
| |
Collapse
|
352
|
Sun Y, Wang X, Liu T, Zhu X, Pan X. The multifaceted role of the SASP in atherosclerosis: from mechanisms to therapeutic opportunities. Cell Biosci 2022; 12:74. [PMID: 35642067 PMCID: PMC9153125 DOI: 10.1186/s13578-022-00815-5] [Citation(s) in RCA: 40] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2022] [Accepted: 05/15/2022] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND The global population of older individuals is growing, and ageing is a key risk factor for atherosclerotic cardiovascular diseases. Abnormal accumulation of senescent cells can cause potentially deleterious effects on the organism with age. As a vital marker of cellular senescence, the senescence-associated secretory phenotype (SASP) is a novel mechanism to link cellular senescence with atherosclerosis. MAIN BODY In this review, we concretely describe the characteristics of the SASP and its regulation mechanisms. Importantly, we provide novel perspectives on how the SASP can promote atherosclerosis. The SASP from different types of senescent cells have vital roles in atherosclerosis progression. As a significant mediator of the harmful effects of senescent cells, it can play a pro-atherogenic role by producing inflammation and immune dysfunction. Furthermore, the SASP can deliver senescence signals to the surrounding vascular cells, gradually contributing to the development of atherosclerosis. Finally, we focus on a variety of novel therapeutic strategies aimed to reduce the burden of atherosclerosis in elderly individuals by targeting senescent cells and inhibiting the regulatory mechanisms of the SASP. CONCLUSION This review systematically summarizes the multiple roles of the SASP in atherosclerosis and can contribute to the exploration of new therapeutic opportunities.
Collapse
Affiliation(s)
- Yu Sun
- Department of Neurology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Xia Wang
- Department of Neurology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Tianwei Liu
- Institute of Cerebrovascular Diseases, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Xiaoyan Zhu
- Department of Critical Care Medicine, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China.
| | - Xudong Pan
- Department of Neurology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China.
| |
Collapse
|
353
|
Li T, Xu G, Yi J, Huang Y. Intraoperative Hypothermia Induces Vascular Dysfunction in the CA1 Region of Rat Hippocampus. Brain Sci 2022; 12:brainsci12060692. [PMID: 35741578 PMCID: PMC9221322 DOI: 10.3390/brainsci12060692] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2022] [Revised: 05/19/2022] [Accepted: 05/25/2022] [Indexed: 02/05/2023] Open
Abstract
Intraoperative hypothermia is very common and leads to memory decline. The hippocampus is responsible for memory formation. As a functional core area, the cornu ammonis 1 (CA1) region of the hippocampus contains abundant blood vessels and is susceptible to ischemia. The aim of the study was to explore vascular function and neuronal state in the CA1 region of rats undergoing intraoperative hypothermia. The neuronal morphological change and activity-regulated cytoskeleton-associated protein (Arc) expression were evaluated by haematoxylin-eosin staining and immunofluorescence respectively. Histology and immunohistochemistry were used to assess vascular function. Results showed that intraoperative hypothermia inhibited the expression of vascular endothelial growth factor and endothelial nitric oxide synthase, and caused reactive oxygen species accumulation. Additionally, the phenotype of vascular smooth muscle cells was transformed from contractile to synthetic, showing a decrease in smooth muscle myosin heavy chain and an increase in osteopontin. Ultimately, vascular dysfunction caused neuronal pyknosis in the CA1 region and reduced memory-related Arc expression. In conclusion, neuronal disorder in the CA1 region was caused by intraoperative hypothermia-related vascular dysfunction. This study could provide a novel understanding of the effect of intraoperative hypothermia in the hippocampus, which might identify a new research target and treatment strategy.
Collapse
|
354
|
Garcia-Garcia J, Usategui-Martin R, Sanabria MR, Fernandez-Perez E, Telleria JJ, Coco-Martin RM. Pathophysiology of Age-Related Macular Degeneration: Implications for Treatment. Ophthalmic Res 2022; 65:615-636. [PMID: 35613547 DOI: 10.1159/000524942] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2022] [Accepted: 04/29/2022] [Indexed: 12/16/2022]
Abstract
Age-related macular degeneration (AMD) is a complex, multifactorial, progressive retinal disease that affects millions of people worldwide and has become the leading cause of visual impairment in developed countries. The disease etiopathogenesis is not understood fully, although many triggers and processes that lead to dysfunction and degeneration of the retinal pigment epithelium (RPE) have already been identified. Thus, the lack of cellular control of oxidative stress, altered proteostasis, dysfunction of lipid homeostasis, and mitochondrial dysfunction form an internal feedback loop that causes the RPE to fail and allows accumulation of abnormal misfolded proteins and abnormal lipids that will form drusen. An inadequate antioxidant response, deficits in autophagy mechanisms, and dysregulation of the extracellular matrix (ECM) help to increase the deposition of abnormal drusen material over time. The drusen then act as inflammatory centers that trigger chronic inflammation of the subretinal space in which microglia and recruited macrophages are also involved, and where the complement system is a key component. Choriocapillaris degeneration and nutritional influences are also classic elements recognized in the AMD pathophysiology. The genetic component of the disease is embodied in the recognition of the described risk or protective polymorphisms of some complement and ECM related genes (mainly CFH and ARMS2/HTRA1). Thus, carriers of the risk haplotype at ARMS2/HTRA1 have a higher risk of developing late AMD at a younger age. Finally, gut microbiota and epigenetics may play a role in modulating the progression to advanced AMD with the presence of local inflammatory conditions. Because of multiple implicated processes, different complex combinations of treatments will probably be the best option to obtain the best visual results; they in turn will differ depending on the type and spectrum of disease affecting individual patients or the disease stage in each patient at a specific moment. This will undoubtedly lead to personalized medicine for control and hopefully find a future cure. This necessitates the continued unraveling of all the processes involved in the pathogenesis of AMD that must be understood to devise the combinations of treatments for different concurrent or subsequent problems.
Collapse
Affiliation(s)
- Julián Garcia-Garcia
- Instituto de Oftalmobiologia Aplicada (IOBA), University of Valladolid, Valladolid, Spain
| | - Ricardo Usategui-Martin
- Instituto de Oftalmobiologia Aplicada (IOBA), University of Valladolid, Valladolid, Spain
- RICORS of Inflammation and Immunopathology of Organs and Systems Network, ISCIII, Madrid, Spain
- Dpto. de Biología Celular, Histología y Farmacología, University of Valladolid, Valladolid, Spain
| | - Maria Rosa Sanabria
- Instituto de Oftalmobiologia Aplicada (IOBA), University of Valladolid, Valladolid, Spain
- RICORS of Inflammation and Immunopathology of Organs and Systems Network, ISCIII, Madrid, Spain
- Ophthalmology Department, Palencia University Hospital Complex, Palencia, Spain
| | - Esther Fernandez-Perez
- Instituto de Oftalmobiologia Aplicada (IOBA), University of Valladolid, Valladolid, Spain
| | - Juan Jose Telleria
- Institute of Biology and Molecular Genetics (IBGM) University of Valladolid, Valladolid, Spain
- Dpto. de Biología Celular, Histología y Farmacología, University of Valladolid, Valladolid, Spain
| | - Rosa M Coco-Martin
- Instituto de Oftalmobiologia Aplicada (IOBA), University of Valladolid, Valladolid, Spain
- RICORS of Inflammation and Immunopathology of Organs and Systems Network, ISCIII, Madrid, Spain
| |
Collapse
|
355
|
Mitochondrial health quality control: measurements and interpretation in the framework of predictive, preventive, and personalized medicine. EPMA J 2022; 13:177-193. [PMID: 35578648 PMCID: PMC9096339 DOI: 10.1007/s13167-022-00281-6] [Citation(s) in RCA: 41] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Accepted: 05/03/2022] [Indexed: 12/11/2022]
Abstract
Mitochondria are the “gatekeeper” in a wide range of cellular functions, signaling events, cell homeostasis, proliferation, and apoptosis. Consequently, mitochondrial injury is linked to systemic effects compromising multi-organ functionality. Although mitochondrial stress is common for many pathomechanisms, individual outcomes differ significantly comprising a spectrum of associated pathologies and their severity grade. Consequently, a highly ambitious task in the paradigm shift from reactive to predictive, preventive, and personalized medicine (PPPM/3PM) is to distinguish between individual disease predisposition and progression under circumstances, resulting in compromised mitochondrial health followed by mitigating measures tailored to the individualized patient profile. For the successful implementation of PPPM concepts, robust parameters are essential to quantify mitochondrial health sustainability. The current article analyses added value of Mitochondrial Health Index (MHI) and Bioenergetic Health Index (BHI) as potential systems to quantify mitochondrial health relevant for the disease development and its severity grade. Based on the pathomechanisms related to the compromised mitochondrial health and in the context of primary, secondary, and tertiary care, a broad spectrum of conditions can significantly benefit from robust quantification systems using MHI/BHI as a prototype to be further improved. Following health conditions can benefit from that: planned pregnancies (improved outcomes for mother and offspring health), suboptimal health conditions with reversible health damage, suboptimal life-style patterns and metabolic syndrome(s) predisposition, multi-factorial stress conditions, genotoxic environment, ischemic stroke of unclear aetiology, phenotypic predisposition to aggressive cancer subtypes, pathologies associated with premature aging and neuro/degeneration, acute infectious diseases such as COVID-19 pandemics, among others.
Collapse
|
356
|
Shamoon L, Espitia-Corredor JA, Dongil P, Menéndez-Ribes M, Romero A, Valencia I, Díaz-Araya G, Sánchez-Ferrer CF, Peiró C. RESOLVIN E1 ATTENUATES DOXORUBICIN-INDUCED ENDOTHELIAL SENESCENCE BY MODULATING NLRP3 INFLAMMASOME ACTIVATION. Biochem Pharmacol 2022; 201:115078. [PMID: 35551917 DOI: 10.1016/j.bcp.2022.115078] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2021] [Revised: 04/14/2022] [Accepted: 05/02/2022] [Indexed: 01/10/2023]
Abstract
Endothelial cell senescence contributes to chronic inflammation and endothelial dysfunction, while favoring cardiovascular disorders and frailty. Senescent cells acquire a pro-inflammatory secretory phenotype that further propagates inflammation and senescence to neighboring cells. Cell senescence can be provoked by plethora of stressors, including inflammatory molecules and chemotherapeutic drugs. Doxorubicin (Doxo) is a powerful anthracycline anticancer drug whose clinical application is constrained by a dose-limiting cardiovascular toxicity. We here investigated whether cell senescence can contribute to the vascular damage elicited by Doxo. In human umbilical vein endothelial cells (HUVEC) cultures, Doxo (10-100 nM) increased the number of SA-β-gal positive cells and the levels of γH2AX, p21 and p53, used as markers of senescence. Moreover, we identified Doxo-induced senescence to be mediated by the nucleotide-binding domain-like receptor protein 3 (NLRP3) inflammasome, a key player of the immune innate system capable of releasing interleukin (IL)-1β. In fact, IL-1β itself mimicked the stimulatory action of Doxo on both NLRP3 activation and cellular senescence, while the pharmacological blockade of IL-1 receptors markedly attenuated the pro-senescence effects of Doxo. In search of additional pharmacological strategies to attenuate Doxo-induced endothelial senescence, we identified resolvin E1 (RvE1), an endogenous pro-resolving mediator, as capable of reducing cell senescence induced by both Doxo and IL-1β by interfering with the increased expression of pP65, NLRP3, and pro-IL-1β proteins and with the formation of active NLRP3 inflammasome complexes. Overall, RvE1 and the blockade of the NLRP3 inflammasome-IL-1β axis may offer a novel therapeutic approach against Doxo-induced cardiovascular toxicity and subsequent sequelae.
Collapse
Affiliation(s)
- Licia Shamoon
- Department of Pharmacology, Faculty of Medicine, Universidad Autónoma de Madrid, Madrid, Spain; PhD Programme in Pharmacology and Physiology, Doctoral School, Universidad Autónoma de Madrid, Madrid, Spain; Instituto de Investigaciones Sanitarias (IdiPAZ), Madrid, Spain
| | - Jenaro A Espitia-Corredor
- Department of Pharmacology, Faculty of Medicine, Universidad Autónoma de Madrid, Madrid, Spain; PhD Programme in Pharmacology and Physiology, Doctoral School, Universidad Autónoma de Madrid, Madrid, Spain; Laboratorio de Farmacología Molecular (FARMOLAB), Department of Pharmaceutical and Toxicological Chemistry, Faculty of Chemical Sciences and Pharmacy, Universidad de Chile, Santiago, Chile
| | - Pilar Dongil
- Department of Pharmacology, Faculty of Medicine, Universidad Autónoma de Madrid, Madrid, Spain; Instituto de Investigaciones Sanitarias (IdiPAZ), Madrid, Spain
| | - Marta Menéndez-Ribes
- Department of Pharmacology, Faculty of Medicine, Universidad Autónoma de Madrid, Madrid, Spain; Instituto de Investigaciones Sanitarias (IdiPAZ), Madrid, Spain
| | - Alejandra Romero
- Department of Pharmacology, Faculty of Medicine, Universidad Autónoma de Madrid, Madrid, Spain; Instituto de Investigaciones Sanitarias (IdiPAZ), Madrid, Spain
| | - Inés Valencia
- Department of Pharmacology, Faculty of Medicine, Universidad Autónoma de Madrid, Madrid, Spain; PhD Programme in Pharmacology and Physiology, Doctoral School, Universidad Autónoma de Madrid, Madrid, Spain; Instituto de Investigaciones Sanitarias (IdiPAZ), Madrid, Spain
| | - Guillermo Díaz-Araya
- Laboratorio de Farmacología Molecular (FARMOLAB), Department of Pharmaceutical and Toxicological Chemistry, Faculty of Chemical Sciences and Pharmacy, Universidad de Chile, Santiago, Chile; Advanced Center for Chronic diseases ACCDiS, Universidad de Chile, Santiago, Chile.
| | - Carlos F Sánchez-Ferrer
- Department of Pharmacology, Faculty of Medicine, Universidad Autónoma de Madrid, Madrid, Spain; Instituto de Investigaciones Sanitarias (IdiPAZ), Madrid, Spain.
| | - Concepción Peiró
- Department of Pharmacology, Faculty of Medicine, Universidad Autónoma de Madrid, Madrid, Spain; Instituto de Investigaciones Sanitarias (IdiPAZ), Madrid, Spain.
| |
Collapse
|
357
|
Mukli P, Wu DH, Csipo T, Owens CD, Lipecz A, Racz FS, Zouein FA, Tabak A, Csiszar A, Ungvari Z, Tsitouras PD, Yabluchanskiy A. Urinary Biomarkers of Oxidative Stress in Aging: Implications for Prediction of Accelerated Biological Age in Prospective Cohort Studies. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:6110226. [PMID: 35571254 PMCID: PMC9106456 DOI: 10.1155/2022/6110226] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Revised: 03/05/2022] [Accepted: 03/28/2022] [Indexed: 11/17/2022]
Abstract
Background Aging is a major risk factor for a range of chronic diseases. Oxidative stress theory of aging has been previously proposed as one of the mechanisms responsible for the age-related decline in organ/tissue function and the development of age-related diseases. Urine contains rich biological information on the health status of every major organ system and can be an important noninvasive source for biomarkers of systemic oxidative stress in aging. Aims The objective of this cross-sectional study was to validate a novel panel of urinary oxidative stress biomarkers. Methods Nucleic acid oxidation adducts and oxidative damage markers of lipids and proteins were assessed in urine samples from nondiabetic and currently nonsmoking subjects (n = 198) across different ages (20 to 89 years old). Urinary parameters and chronological age were correlated then the biological age of enrolled individuals was determined from the urinary oxidative stress markers using the algorithm of Klemera and Doubal. Results Our findings showed that 8-oxo-7,8-deoxyguanosine (8-oxoG), 8-oxo-7,8-dihydroguanosine (8-OHdG), and dityrosine (DTyr) positively correlated with chronological age, while the level of an F2-isoprostane (iPF2 α-VI) correlated negatively with age. We found that 8-oxoG, DTyr, and iPF2 α-VI were significantly higher among accelerated agers compared to nonaccelerated agers and that a decision tree model could successfully identify accelerated agers with an accuracy of >92%. Discussion. Our results indicate that 8-oxoG and iPF2 α-VI levels in the urine reveal biological aging. Conclusion Assessing urinary biomarkers of oxidative stress may be an important approach for the evaluation of biological age by identifying individuals at accelerated risk for the development of age-related diseases.
Collapse
Affiliation(s)
- Peter Mukli
- Vascular Cognitive Impairment and Neurodegeneration Program, Oklahoma Center for Geroscience and Healthy Brain Aging, Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Department of Physiology, Semmelweis University, Budapest, Hungary
| | - Dee H. Wu
- Department of Radiological Sciences, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- The Peggy and Charles Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Tamas Csipo
- Vascular Cognitive Impairment and Neurodegeneration Program, Oklahoma Center for Geroscience and Healthy Brain Aging, Department of Biochemistry and Molecular Biology, 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
| | - Cameron D. Owens
- Vascular Cognitive Impairment and Neurodegeneration Program, Oklahoma Center for Geroscience and Healthy Brain Aging, Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Agnes Lipecz
- Vascular Cognitive Impairment and Neurodegeneration Program, Oklahoma Center for Geroscience and Healthy Brain Aging, Department of Biochemistry and Molecular Biology, 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
| | - Frigyes Samuel Racz
- Department of Physiology, Semmelweis University, Budapest, Hungary
- Department of Neurology, Dell Medical School, University of Texas at Austin, Austin, TX, USA
| | - Fouad A. Zouein
- The Cardiovascular, Renal, and Metabolic Diseases Research Center of Excellence, American University of Beirut Medical Center, Riad El-Solh, Beirut, Lebanon
- Department of Signaling and Cardiovascular Pathophysiology, UMR-S 1180, Inserm, Université Paris-Saclay, France
- Department of Pharmacology and Toxicology, School of Medicine, University of Mississippi Medical Center, Jackson, MS, USA
| | - Adam Tabak
- International Training Program in Geroscience, Doctoral School of Basic and Translational Medicine/Department of Public Health, Semmelweis University, Budapest, Hungary
- 1st Department of Medicine, Faculty of Medicine, Semmelweis University, Budapest, Hungary
| | - Anna Csiszar
- Vascular Cognitive Impairment and Neurodegeneration Program, Oklahoma Center for Geroscience and Healthy Brain Aging, Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- The Peggy and Charles Stephenson Cancer Center, University of Oklahoma Health Sciences Center, 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 and Neurodegeneration Program, Oklahoma Center for Geroscience and Healthy Brain Aging, Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Department of Radiological Sciences, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- The Peggy and Charles Stephenson Cancer Center, 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
- Department of Health Promotion Sciences, College of Public Health, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Panayiotis D. Tsitouras
- Vascular Cognitive Impairment and Neurodegeneration Program, Oklahoma Center for Geroscience and Healthy Brain Aging, Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Andriy Yabluchanskiy
- Vascular Cognitive Impairment and Neurodegeneration Program, Oklahoma Center for Geroscience and Healthy Brain Aging, Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- The Peggy and Charles Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Department of Health Promotion Sciences, College of Public Health, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| |
Collapse
|
358
|
Senthilkumar T, Kumarganesh S, Sivakumar P, Periyarselvam K. Primitive detection of Alzheimer’s disease using neuroimaging: A progression model for Alzheimer’s disease: Their applications, benefits, and drawbacks. JOURNAL OF INTELLIGENT & FUZZY SYSTEMS 2022. [DOI: 10.3233/jifs-220628] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Alzheimer’s disease (A.D.) is the most widespread type of Dementia, and it is not a curable neurodegenerative disease that affects millions of older people. Researchers were able to use their understanding of Alzheimer’s disease risk variables to develop enrichment processes for longitudinal imaging studies. Using this method, they reduced their sample size and study time. This paper describes the primitive detective of Alzheimer’s diseases using Neuroimaging techniques. Several preprocessing methods were used to ensure that the dataset was ready for subsequent feature extraction and categorization. The noise was reduced by converting and averaging many scan frames from real to DCT space. Both sides of the averaged image were filtered and combined into a single shot after being converted to real space. InceptionV3 and DenseNet201 are two pre-trained models used in the suggested model. The PCA approach was used to select the traits, and the resulting explained variance ratio was 0.99The Simons Foundation Autism Research Initiative (SFARI)—Simon’s Simplex Collection (SSC)—and UCI machine learning datasets showed that our method is faster and more successful at identifying complete long-risk patterns when compared to existing methods.
Collapse
Affiliation(s)
- T. Senthilkumar
- GRT Institute of Engineering and Technology, Tiruttani, Tamilnadu, India
| | | | - P. Sivakumar
- GRT Institute of Engineering and Technology, Tiruttani, Tamilnadu, India
| | - K. Periyarselvam
- GRT Institute of Engineering and Technology, Tiruttani, Tamilnadu, India
| |
Collapse
|
359
|
Mone P, Pansini A, Jankauskas SS, Varzideh F, Kansakar U, Lombardi A, Trimarco V, Frullone S, Santulli G. L-Arginine Improves Cognitive Impairment in Hypertensive Frail Older Adults. Front Cardiovasc Med 2022; 9:868521. [PMID: 35498050 PMCID: PMC9039514 DOI: 10.3389/fcvm.2022.868521] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2022] [Accepted: 02/28/2022] [Indexed: 12/11/2022] Open
Abstract
Cognitive impairment is a prevailing event in hypertensive patients and in frail older adults. Endothelial dysfunction has been shown to underlie both hypertension and cognitive dysfunction. Our hypothesis is that L-Arginine, which is known to ameliorate endothelial dysfunction, could counteract cognitive impairment in a high-risk population of hypertensive frail older adults. We designed a clinical trial to verify the effects of 4-weeks oral supplementation of L-Arginine on global cognitive function of hypertensive frail older patients. The study was successfully completed by 35 frail hypertensive elderly patients assigned to L-Arginine and 37 assigned to placebo. At follow-up, we found a significant difference in the Montreal Cognitive Assessment (MoCA) test score between the L-Arginine treated group and placebo (p: 0.0178). Moreover, we demonstrated that L-Arginine significantly attenuates Angiotensin II-induced mitochondrial oxidative stress in human endothelial cells. In conclusion, our findings indicate for the first time that oral L-Arginine supplementation significantly improves cognitive impairment in frail hypertensive older adults.
Collapse
Affiliation(s)
- Pasquale Mone
- Department of Medicine, Albert Einstein College of Medicine, New York, NY, United States.,Azienda Sanitaria Locale (ASL) Avellino, Avellino, Italy.,Campania University, Naples, Italy
| | | | | | - Fahimeh Varzideh
- Department of Medicine, Albert Einstein College of Medicine, New York, NY, United States
| | - Urna Kansakar
- Department of Medicine, Albert Einstein College of Medicine, New York, NY, United States
| | - Angela Lombardi
- Department of Medicine, Albert Einstein College of Medicine, New York, NY, United States
| | | | | | - Gaetano Santulli
- Department of Medicine, Albert Einstein College of Medicine, New York, NY, United States.,University of Naples "Federico II", Naples, Italy
| |
Collapse
|
360
|
Furukawa M, Matsuda K, Aoki Y, Yamada M, Wang J, Watanabe M, Kurosawa M, Shikama Y, Matsushita K. Analysis of senescence in gingival tissues and gingival fibroblast cultures. Clin Exp Dent Res 2022; 8:939-949. [PMID: 35491709 PMCID: PMC9382052 DOI: 10.1002/cre2.581] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2022] [Revised: 03/30/2022] [Accepted: 04/03/2022] [Indexed: 11/14/2022] Open
Abstract
Objective To determine senescence‐associated changes in the gingival tissues of aged mice and gingival fibroblast cultures. Materials and Methods The production of senescence‐associated β‐galactosidase (SA‐β‐gal) and mRNA expression of p16, p21, interleukin (IL)‐1β, and tumor necrosis factor α (TNF‐α) were evaluated in gingival tissues, gingival fibroblasts of 10‐ and 20‐month‐old C57BL/6NCrl mice, and multiple‐passaged and hydrogen peroxide‐stimulated human gingival fibroblasts (HGFs). Changes in molecular expression in HGF cultures due to senescent cell elimination by the senolytic drug ABT‐263 (Navitoclax) were analyzed. Results Compared to 10‐week‐old mice, the 20‐month‐old mice had higher numbers of M1 macrophages. The proportion of cells expressing SA‐β‐gal were also higher in 20‐ month‐old mice than in 10‐week‐old‐mice. Gingival fibroblasts in 20‐month‐old mice expressed less collagen 1a1, collagen 4a1, and collagen 4a2 mRNA than those in 10‐week‐old mice. Compared to control cells, H2O2 treated HGF cells expressed higher levels of SA‐β‐gal and p16, p21, IL‐1β, and TNF‐α. Furthermore, ABT‐263 suppressed HGF cell expression of cytokines after senescence induction. Conclusions Senescence‐associated changes were observed in the gingival tissues of aged mice and HGF cultures. In addition, the potential of senolytic drugs to modify aging‐related changes in the gingiva was shown.
Collapse
Affiliation(s)
- Masae Furukawa
- Department of Oral Disease Research National Center for Geriatrics and Gerontology Obu Japan
| | | | - Yu Aoki
- Daiichi Sankyo Healthcare Co., Ltd. Tokyo Japan
| | - Mitsuyoshi Yamada
- Department of Oral Disease Research National Center for Geriatrics and Gerontology Obu Japan
- Department of Operative Dentistry, School of Dentistry Aichi Gakuin University Nagoya Japan
| | - Jingshu Wang
- Department of Oral Disease Research National Center for Geriatrics and Gerontology Obu Japan
| | - Maki Watanabe
- Department of Oral Disease Research National Center for Geriatrics and Gerontology Obu Japan
| | - Mie Kurosawa
- Department of Oral Disease Research National Center for Geriatrics and Gerontology Obu Japan
| | - Yosuke Shikama
- Department of Oral Disease Research National Center for Geriatrics and Gerontology Obu Japan
| | - Kenji Matsushita
- Department of Oral Disease Research National Center for Geriatrics and Gerontology Obu Japan
| |
Collapse
|
361
|
Jusic A, Thomas PB, Wettinger SB, Dogan S, Farrugia R, Gaetano C, Tuna BG, Pinet F, Robinson EL, Tual-Chalot S, Stellos K, Devaux Y. Noncoding RNAs in age-related cardiovascular diseases. Ageing Res Rev 2022; 77:101610. [PMID: 35338919 DOI: 10.1016/j.arr.2022.101610] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Revised: 01/28/2022] [Accepted: 03/15/2022] [Indexed: 11/01/2022]
Abstract
Cardiovascular diseases (CVDs) are the leading cause of morbidity and mortality in the adult population worldwide and represent a severe economic burden and public health concern. The majority of human genes do not code for proteins. However, noncoding transcripts play important roles in ageing that significantly increases the risk for CVDs. Noncoding RNAs (ncRNAs) are critical regulators of multiple biological processes related to ageing such as oxidative stress, mitochondrial dysfunction and chronic inflammation. NcRNAs are also involved in pathophysiological developments within the cardiovascular system including arrhythmias, cardiac hypertrophy, fibrosis, myocardial infarction and heart failure. In this review article, we cover the roles of ncRNAs in cardiovascular ageing and disease as well as their potential therapeutic applications in CVDs.
Collapse
|
362
|
Kusumoto D, Yuasa S, Fukuda K. Induced Pluripotent Stem Cell-Based Drug Screening by Use of Artificial Intelligence. Pharmaceuticals (Basel) 2022; 15:562. [PMID: 35631387 PMCID: PMC9145330 DOI: 10.3390/ph15050562] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Revised: 04/25/2022] [Accepted: 04/27/2022] [Indexed: 12/10/2022] Open
Abstract
Induced pluripotent stem cells (iPSCs) are terminally differentiated somatic cells that differentiate into various cell types. iPSCs are expected to be used for disease modeling and for developing novel treatments because differentiated cells from iPSCs can recapitulate the cellular pathology of patients with genetic mutations. However, a barrier to using iPSCs for comprehensive drug screening is the difficulty of evaluating their pathophysiology. Recently, the accuracy of image analysis has dramatically improved with the development of artificial intelligence (AI) technology. In the field of cell biology, it has become possible to estimate cell types and states by examining cellular morphology obtained from simple microscopic images. AI can evaluate disease-specific phenotypes of iPS-derived cells from label-free microscopic images; thus, AI can be utilized for disease-specific drug screening using iPSCs. In addition to image analysis, various AI-based methods can be applied to drug development, including phenotype prediction by analyzing genomic data and virtual screening by analyzing structural formulas and protein-protein interactions of compounds. In the future, combining AI methods may rapidly accelerate drug discovery using iPSCs. In this review, we explain the details of AI technology and the application of AI for iPSC-based drug screening.
Collapse
Affiliation(s)
- Dai Kusumoto
- Department of Cardiology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo 160-8582, Japan;
- Center for Preventive Medicine, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo 160-8582, Japan
| | - Shinsuke Yuasa
- Department of Cardiology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo 160-8582, Japan;
| | - Keiichi Fukuda
- Department of Cardiology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo 160-8582, Japan;
| |
Collapse
|
363
|
Custodero C, Ciavarella A, Panza F, Gnocchi D, Lenato GM, Lee J, Mazzocca A, Sabbà C, Solfrizzi V. Role of inflammatory markers in the diagnosis of vascular contributions to cognitive impairment and dementia: a systematic review and meta-analysis. GeroScience 2022; 44:1373-1392. [PMID: 35486344 PMCID: PMC9213626 DOI: 10.1007/s11357-022-00556-w] [Citation(s) in RCA: 56] [Impact Index Per Article: 18.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2021] [Accepted: 03/23/2022] [Indexed: 12/29/2022] Open
Abstract
Vascular contribution to cognitive impairment and dementia (VCID) is a clinical label encompassing a wide range of cognitive disorders progressing from mild to major vascular cognitive impairment (VCI), which is also defined as vascular dementia (VaD). VaD diagnosis is mainly based on clinical and imaging findings. Earlier biomarkers are needed to identify subjects at risk to develop mild VCI and VaD. In the present meta-analysis, we comprehensively evaluated the role of inflammatory biomarkers in differential diagnosis between VaD and Alzheimer’s disease (AD), and assessed their prognostic value on predicting VaD incidence. We collected literature until January 31, 2021, assessing three inflammatory markers [interleukin(IL)-6, C-reactive protein (CRP), tumor necrosis factor (TNF)-α] from blood or cerebrospinal fluid (CSF) samples. Thirteen cross-sectional and seven prospective studies were included. Blood IL-6 levels were cross-sectionally significantly higher in people with VaD compared to AD patients (SMD: 0.40, 95% CI: 0.18 to 0.62) with low heterogeneity (I2: 41%, p = 0.13). Higher IL-6 levels were also associated to higher risk of incident VaD (relative risk: 1.28, 95% CI: 1.03 to 1.59, I2: 0%). IL-6 in CSF was significantly higher in people with VaD compared to healthy subjects (SMD: 0.77, 95% CI: 0.17 to 1.37, I2: 70%), and not compared to AD patients, but due to limited evidence and high inconsistency across studies, we could not draw definite conclusion. Higher blood IL-6 levels might represent a useful biomarker able to differentiate people with VaD from those with AD and might be correlated with higher risk of future VaD.
Collapse
Affiliation(s)
- Carlo Custodero
- Dipartimento Interdisciplinare di Medicina, Clinica Medica e Geriatria "Cesare Frugoni", University of Bari Aldo Moro, Bari, Italy
| | - Alessandro Ciavarella
- Dipartimento Interdisciplinare di Medicina, Clinica Medica e Geriatria "Cesare Frugoni", University of Bari Aldo Moro, Bari, Italy.,Fondazione IRCCS Ca' Granda-Ospedale Maggiore Policlinico, A. Bianchi Bonomi Hemophilia and Thrombosis Center, Milan, Italy
| | - Francesco Panza
- Population Health Unit-"Salus In Apulia Study", National Institute of Gastroenterology "Saverio de Bellis", Research Hospital, Castellana Grotte, Bari, Italy
| | - Davide Gnocchi
- Dipartimento Interdisciplinare di Medicina, Clinica Medica e Geriatria "Cesare Frugoni", University of Bari Aldo Moro, Bari, Italy
| | - Gennaro M Lenato
- Dipartimento Interdisciplinare di Medicina, Clinica Medica e Geriatria "Cesare Frugoni", University of Bari Aldo Moro, Bari, Italy
| | - Juhan Lee
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA
| | - Antonio Mazzocca
- Dipartimento Interdisciplinare di Medicina, Clinica Medica e Geriatria "Cesare Frugoni", University of Bari Aldo Moro, Bari, Italy
| | - Carlo Sabbà
- Dipartimento Interdisciplinare di Medicina, Clinica Medica e Geriatria "Cesare Frugoni", University of Bari Aldo Moro, Bari, Italy
| | - Vincenzo Solfrizzi
- Dipartimento Interdisciplinare di Medicina, Clinica Medica e Geriatria "Cesare Frugoni", University of Bari Aldo Moro, Bari, Italy.
| |
Collapse
|
364
|
Phoenix A, Chandran R, Ergul A. Cerebral Microvascular Senescence and Inflammation in Diabetes. Front Physiol 2022; 13:864758. [PMID: 35574460 PMCID: PMC9098835 DOI: 10.3389/fphys.2022.864758] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Accepted: 04/14/2022] [Indexed: 01/16/2023] Open
Abstract
Stress-induced premature senescence can contribute to the accelerated metabolic aging process in diabetes. Progressive accumulation of senescent cells in the brain, especially those displaying the harmful inflammatory senescence-associated secretory phenotype (SASP), may lead to cognitive impairment linked with metabolic disturbances. In this context, the senescence within the neurovascular unit (NVU) should be studied as much as in the neurons as emerging evidence shows that neurogliovascular communication is critical for brain health. It is also known that cerebrovascular dysfunction and decreased cerebral blood flow (CBF) precede the occurrence of neuronal pathologies and overt cognitive impairment. Various studies have shown that endothelial cells, the major component of the NVU, acquire a senescent phenotype via various molecular mediators and pathways upon exposure to high glucose and other conditions mimicking metabolic disturbances. In addition, senescence in the other cells that are part of the NVU, like pericytes and vascular smooth cells, was also triggered upon exposure to diabetic conditions. The senescence within the NVU may compromise functional and trophic coupling among glial, vascular, and neuronal cells and the resulting SASP may contribute to the chronic neurovascular inflammation observed in Alzheimer's Disease and Related Dementias (ADRD). The link between diabetes-mediated cerebral microvascular dysfunction, NVU senescence, inflammation, and cognitive impairment must be widely studied to design therapeutic strategies.
Collapse
Affiliation(s)
- Ashley Phoenix
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston, SC, United States
| | - Raghavendar Chandran
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston, SC, United States
| | - Adviye Ergul
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston, SC, United States,Ralph H. Johnson Veterans Affairs Medical Center, Charleston, SC, United States,*Correspondence: Adviye Ergul,
| |
Collapse
|
365
|
Valencia I, Vallejo S, Dongil P, Romero A, San Hipólito-Luengo Á, Shamoon L, Posada M, García-Olmo D, Carraro R, Erusalimsky JD, Romacho T, Peiró C, Sánchez-Ferrer CF. DPP4 Promotes Human Endothelial Cell Senescence and Dysfunction via the PAR2-COX-2-TP Axis and NLRP3 Inflammasome Activation. Hypertension 2022; 79:1361-1373. [PMID: 35477273 DOI: 10.1161/hypertensionaha.121.18477] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND Abnormal accumulation of senescent cells in the vessel wall leads to a compromised vascular function contributing to vascular aging. Soluble DPP4 (dipeptidyl peptidase 4; sDPP4) secretion from visceral adipose tissue is enhanced in obesity, now considered a progeric condition. sDPP4 triggers vascular deleterious effects, albeit its contribution to vascular aging is unknown. We aimed to explore sDPP4 involvement in vascular aging, unraveling the molecular pathway by which sDPP4 acts on the endothelium. METHODS Human endothelial cell senescence was assessed by senescence-associated β-galactosidase assay, visualization of DNA damage, and expression of prosenescent markers, whereas vascular function was evaluated by myography over human dissected microvessels. In visceral adipose tissue biopsies from a cohort of obese patients, we explored several age-related parameters in vitro and ex vivo. RESULTS By a common mechanism, sDPP4 triggers endothelial cell senescence and endothelial dysfunction in isolated human resistance arteries. sDPP4 activates the metabotropic receptor PAR2 (protease-activated receptor 2), COX-2 (cyclooxygenase 2) activity, and the production of TXA2 (thromboxane A2) acting over TP (thromboxane receptor) receptors (PAR2-COX-2-TP axis), leading to NLRP3 (nucleotide-binding oligomerization domain, leucine-rich repeat, and pyrin domain-containing 3) inflammasome activation. Obese patients exhibited impaired microarterial functionality in comparison to control nonobese counterparts. Importantly, endothelial dysfunction in obese patients positively correlated with greater expression of DPP4, prosenescent, and proinflammatory markers in visceral adipose tissue nearby the resistance arteries. Moreover, when DPP4 activity or sDPP4-induced prosenescent mechanism was blocked, endothelial dysfunction was restored back to levels of healthy subjects. CONCLUSIONS These results reveal sDPP4 as a relevant mediator in early vascular aging and highlight its capacity activating main proinflammatory mediators in the endothelium that might be pharmacologically tackled.
Collapse
Affiliation(s)
- Inés Valencia
- Department of Pharmacology and Therapeutics, School of Medicine, Universidad Autónoma de Madrid, Spain. (I.V., S.V., P.D., A.R., Á.S.H.-L., L.S., T.R., C.P., C.F.S.-F.).,PhD Programme in Pharmacology and Physiology, Doctoral School, Universidad Autónoma de Madrid, Spain. (I.V., L.S.).,Instituto de Investigación Sanitaria del Hospital Universitario La Paz, Madrid, Spain (I.V., S.V., P.D., A.R., Á.S.H.-L., L.S., T.R., C.P., C.F.S.-F.)
| | - Susana Vallejo
- Department of Pharmacology and Therapeutics, School of Medicine, Universidad Autónoma de Madrid, Spain. (I.V., S.V., P.D., A.R., Á.S.H.-L., L.S., T.R., C.P., C.F.S.-F.).,Instituto de Investigación Sanitaria del Hospital Universitario La Paz, Madrid, Spain (I.V., S.V., P.D., A.R., Á.S.H.-L., L.S., T.R., C.P., C.F.S.-F.)
| | - Pilar Dongil
- Department of Pharmacology and Therapeutics, School of Medicine, Universidad Autónoma de Madrid, Spain. (I.V., S.V., P.D., A.R., Á.S.H.-L., L.S., T.R., C.P., C.F.S.-F.).,Instituto de Investigación Sanitaria del Hospital Universitario La Paz, Madrid, Spain (I.V., S.V., P.D., A.R., Á.S.H.-L., L.S., T.R., C.P., C.F.S.-F.)
| | - Alejandra Romero
- Department of Pharmacology and Therapeutics, School of Medicine, Universidad Autónoma de Madrid, Spain. (I.V., S.V., P.D., A.R., Á.S.H.-L., L.S., T.R., C.P., C.F.S.-F.).,Instituto de Investigación Sanitaria del Hospital Universitario La Paz, Madrid, Spain (I.V., S.V., P.D., A.R., Á.S.H.-L., L.S., T.R., C.P., C.F.S.-F.)
| | - Álvaro San Hipólito-Luengo
- Department of Pharmacology and Therapeutics, School of Medicine, Universidad Autónoma de Madrid, Spain. (I.V., S.V., P.D., A.R., Á.S.H.-L., L.S., T.R., C.P., C.F.S.-F.).,Instituto de Investigación Sanitaria del Hospital Universitario La Paz, Madrid, Spain (I.V., S.V., P.D., A.R., Á.S.H.-L., L.S., T.R., C.P., C.F.S.-F.)
| | - Licia Shamoon
- Department of Pharmacology and Therapeutics, School of Medicine, Universidad Autónoma de Madrid, Spain. (I.V., S.V., P.D., A.R., Á.S.H.-L., L.S., T.R., C.P., C.F.S.-F.).,PhD Programme in Pharmacology and Physiology, Doctoral School, Universidad Autónoma de Madrid, Spain. (I.V., L.S.).,Instituto de Investigación Sanitaria del Hospital Universitario La Paz, Madrid, Spain (I.V., S.V., P.D., A.R., Á.S.H.-L., L.S., T.R., C.P., C.F.S.-F.)
| | - María Posada
- Service of Surgery and Instituto de Investigación Sanitaria del Hospital Fundación Jiménez Díaz, Madrid, Spain (M.P., D.G.-O.)
| | - Damián García-Olmo
- Service of Surgery and Instituto de Investigación Sanitaria del Hospital Fundación Jiménez Díaz, Madrid, Spain (M.P., D.G.-O.)
| | - Raffaelle Carraro
- Department of Medicine, School of Medicine, Universidad Autónoma de Madrid, Spain. (R.C.).,Service of Endocrinology and Instituto de Investigación Sanitaria del Hospital Universitario La Princesa, Madrid, Spain (R.C.)
| | - Jorge D Erusalimsky
- School of Sport and Health Sciences, Cardiff Metropolitan University, United Kingdom (J.D.E.)
| | - Tania Romacho
- Department of Pharmacology and Therapeutics, School of Medicine, Universidad Autónoma de Madrid, Spain. (I.V., S.V., P.D., A.R., Á.S.H.-L., L.S., T.R., C.P., C.F.S.-F.).,Instituto de Investigación Sanitaria del Hospital Universitario La Paz, Madrid, Spain (I.V., S.V., P.D., A.R., Á.S.H.-L., L.S., T.R., C.P., C.F.S.-F.)
| | - Concepción Peiró
- Department of Pharmacology and Therapeutics, School of Medicine, Universidad Autónoma de Madrid, Spain. (I.V., S.V., P.D., A.R., Á.S.H.-L., L.S., T.R., C.P., C.F.S.-F.).,Instituto de Investigación Sanitaria del Hospital Universitario La Paz, Madrid, Spain (I.V., S.V., P.D., A.R., Á.S.H.-L., L.S., T.R., C.P., C.F.S.-F.)
| | - Carlos F Sánchez-Ferrer
- Department of Pharmacology and Therapeutics, School of Medicine, Universidad Autónoma de Madrid, Spain. (I.V., S.V., P.D., A.R., Á.S.H.-L., L.S., T.R., C.P., C.F.S.-F.).,Instituto de Investigación Sanitaria del Hospital Universitario La Paz, Madrid, Spain (I.V., S.V., P.D., A.R., Á.S.H.-L., L.S., T.R., C.P., C.F.S.-F.)
| |
Collapse
|
366
|
Soma M, Lalam SK. The role of nicotinamide mononucleotide (NMN) in anti-aging, longevity, and its potential for treating chronic conditions. Mol Biol Rep 2022; 49:9737-9748. [PMID: 35441939 DOI: 10.1007/s11033-022-07459-1] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Accepted: 04/06/2022] [Indexed: 12/24/2022]
Abstract
Biosynthesis and regulation of nicotinamide adenine dinucleotide (NAD+) has recently gained a lot of attention. A systemic decline in NAD+ across many tissues is associated with all the hallmarks of aging. NAD+ can affect a variety of cellular processes, including metabolic pathways, DNA repair, and immune cell activity, both directly and indirectly. These cellular processes play a vital role in maintaining homeostasis, but as people get older, their tissue and cellular NAD+ levels decrease, and this drop in NAD+ levels has been connected to a number of age-related disorders. By restoring NAD+ levels, several of these age-related disorders can be delayed or even reversed. Some of the new studies conducted in mice and humans have targeted the NAD+ metabolism with NAD+ intermediates. Of these, nicotinamide mononucleotide (NMN) has been shown to offer great therapeutic potential with promising results in age-related chronic conditions such as diabetes, cardiovascular issues, cognitive impairment, and many others. Further, human interventions are required to study the long-term effects of supplementing NMN with varying doses. The paper focuses on reviewing the importance of NAD+ on human aging and survival, biosynthesis of NAD+ from its precursors, key clinical trial findings, and the role of NMN on various health conditions.
Collapse
|
367
|
Watanabe R, Hashimoto M. Aging-Related Vascular Inflammation: Giant Cell Arteritis and Neurological Disorders. Front Aging Neurosci 2022; 14:843305. [PMID: 35493934 PMCID: PMC9039280 DOI: 10.3389/fnagi.2022.843305] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2021] [Accepted: 03/22/2022] [Indexed: 12/16/2022] Open
Abstract
Aging is characterized by the functional decline of the immune system and constitutes the primary risk factor for infectious diseases, cardiovascular disorders, cancer, and neurodegenerative disorders. Blood vessels are immune-privileged sites and consist of endothelial cells, vascular smooth muscle cells, macrophages, dendritic cells, fibroblasts, and pericytes, among others. Aging also termed senescence inevitably affects blood vessels, making them vulnerable to inflammation. Atherosclerosis causes low-grade inflammation from the endothelial side; whereas giant cell arteritis (GCA) causes intense inflammation from the adventitial side. GCA is the most common autoimmune vasculitis in the elderly characterized by the formation of granulomas composed of T cells and macrophages in medium- and large-sized vessels. Recent studies explored the pathophysiology of GCA at unprecedented resolutions, and shed new light on cellular signaling pathways and metabolic fitness in wall-destructive T cells and macrophages. Moreover, recent reports have revealed that not only can cerebrovascular disorders, such as stroke and ischemic optic neuropathy, be initial or coexistent manifestations of GCA, but the same is true for dementia and neurodegenerative disorders. In this review, we first outline how aging affects vascular homeostasis. Subsequently, we review the updated pathophysiology of GCA and explain the similarities and differences between vascular aging and GCA. Then, we introduce the possible link between T cell aging, neurological aging, and GCA. Finally, we discuss therapeutic strategies targeting both senescence and vascular inflammation.
Collapse
|
368
|
Tai GJ, Yu QQ, Li JP, Wei W, Ji XM, Zheng RF, Li XX, Wei L, Xu M. NLRP3 inflammasome links vascular senescence to diabetic vascular lesions. Pharmacol Res 2022; 178:106143. [PMID: 35219871 DOI: 10.1016/j.phrs.2022.106143] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Revised: 01/31/2022] [Accepted: 02/22/2022] [Indexed: 01/10/2023]
Abstract
Vascular senescence is inextricably linked to the onset and progression of cardiovascular diseases (CVDs), which are the main cause of mortality in people with Type 2 diabetes (T2DM). Previous studies have emphasized the importance of chronic aseptic inflammation in diabetic vasculopathy. Here, we found the abnormal activation of NLRP3 inflammasome in the aorta of both old and T2DM mice by immunofluorescence and Western Blot analysis. Histopathological and isometry tension analysis showed that the presence of T2DM triggered or aggravated the increase of vascular aging markers, as well as age-associated vascular impairment and vasomotor dysfunction, which were improved by NLRP3 deletion or inhibition. Differential expression of aortic genes links to senescence activation and vascular remodeling supports the favorable benefits of NLRP3-/- during T2DM. In vitro results based on primary mice aortic endothelial cells (MAECs) and vascular smooth muscle cells (VSMCs) demonstrate that NLRP3 deficiency attenuated premature senescence and restored proliferation and migration capability under-stimulation, and partially ameliorated replicative senescence. These results provide an insight into the critical role of NLRP3 signaling in T2DM-induced vascular aging and loss of vascular homeostasis, and provide the possibility that targeting NLRP3 inflammasome might be a promising strategy to prevent diabetic vascular senescence and associated vascular lesions.
Collapse
Affiliation(s)
- Guang-Jie Tai
- Department of Clinical Pharmacy, School of Preclinical Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Qing-Qing Yu
- Department of Clinical Pharmacy, School of Preclinical Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Jia-Peng Li
- Department of Clinical Pharmacy, School of Preclinical Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Wei Wei
- Department of Clinical Pharmacy, School of Preclinical Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Xiao-Man Ji
- Department of Clinical Pharmacy, School of Preclinical Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Rui-Fang Zheng
- Xinjiang Key Laboratory of Uighur Medicines, Xinjiang Institute of Materia Medica, Urumchi, Xinjiang 830004, China
| | - Xiao-Xue Li
- Department of Cardiology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing 210009, China
| | - Li Wei
- Department of Practice and Policy, UCL School of Pharmacy, London WC1N 1AX, United Kingdom
| | - Ming Xu
- Department of Clinical Pharmacy, School of Preclinical Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing 210009, China.
| |
Collapse
|
369
|
Figuer A, Alique M, Valera G, Serroukh N, Ceprían N, de Sequera P, Morales E, Carracedo J, Ramírez R, Bodega G. Nuevos mecanismos implicados en el desarrollo de la enfermedad cardiovascular en la enfermedad renal crónica. Nefrologia 2022. [DOI: 10.1016/j.nefro.2022.03.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
|
370
|
Kong D, Yu Y. Prostaglandin D2 signaling and cardiovascular homeostasis. J Mol Cell Cardiol 2022; 167:97-105. [DOI: 10.1016/j.yjmcc.2022.03.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/28/2021] [Revised: 02/25/2022] [Accepted: 03/28/2022] [Indexed: 10/18/2022]
|
371
|
Kiss T, Nyúl-Tóth Á, Gulej R, Tarantini S, Csipo T, Mukli P, Ungvari A, Balasubramanian P, Yabluchanskiy A, Benyo Z, Conley SM, Wren JD, Garman L, Huffman DM, Csiszar A, Ungvari Z. Old blood from heterochronic parabionts accelerates vascular aging in young mice: transcriptomic signature of pathologic smooth muscle remodeling. GeroScience 2022; 44:953-981. [PMID: 35124764 PMCID: PMC9135944 DOI: 10.1007/s11357-022-00519-1] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Accepted: 01/16/2022] [Indexed: 02/07/2023] Open
Abstract
Vascular aging has a central role in the pathogenesis of cardiovascular diseases contributing to increased mortality of older adults. There is increasing evidence that, in addition to the documented role of cell-autonomous mechanisms of aging, cell-nonautonomous mechanisms also play a critical role in the regulation of vascular aging processes. Our recent transcriptomic studies (Kiss T. et al. Geroscience. 2020;42(2):727-748) demonstrated that circulating anti-geronic factors from young blood promote vascular rejuvenation in aged mice. The present study was designed to expand upon the results of this study by testing the hypothesis that circulating pro-geronic factors also contribute to the genesis of vascular aging phenotypes. To test this hypothesis, through heterochronic parabiosis, we determined the extent to which shifts in the vascular transcriptome (RNA-seq) are modulated by the old systemic environment. We reanalyzed existing RNA-seq data, comparing the transcriptome in the aorta arch samples isolated from isochronic parabiont aged (20-month-old) C57BL/6 mice [A-(A); parabiosis for 8 weeks] and young isochronic parabiont (6-month-old) mice [Y-(Y)] and also assessing transcriptomic changes in the aortic arch in young (6-month-old) parabiont mice [Y-(A); heterochronic parabiosis for 8 weeks] induced by the presence of old blood derived from aged (20-month-old) parabionts. We identified 528 concordant genes whose expression levels differed in the aged phenotype and were shifted towards the aged phenotype by the presence of old blood in young Y-(A) animals. Among them, the expression of 221 concordant genes was unaffected by the presence of young blood in A-(Y) mice. GO enrichment analysis suggests that old blood-regulated genes may contribute to pathologic vascular remodeling. IPA Upstream Regulator analysis (performed to identify upstream transcriptional regulators that may contribute to the observed transcriptomic changes) suggests that the mechanism of action of pro-geronic factors present in old blood may include inhibition of pathways mediated by SRF (serum response factor), insulin-like growth factor-1 (IGF-1) and VEGF-A. In conclusion, relatively short-term exposure to old blood can accelerate vascular aging processes. Our findings provide additional evidence supporting the significant plasticity of vascular aging and the existence of circulating pro-geronic factors mediating pathological remodeling of the vascular smooth muscle cells and the extracellular matrix.
Collapse
Affiliation(s)
- Tamas Kiss
- Vascular Cognitive Impairment and Neurodegeneration Program, Oklahoma Center for Geroscience and Healthy Brain Aging, Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK USA
- International Training Program in Geroscience, First Department of Pediatrics, Semmelweis University, Budapest, Hungary
- International Training Program in Geroscience, Doctoral School of Basic and Translational Medicine/Department of Translational Medicine, Semmelweis University, Budapest, Hungary
| | - Ádám Nyúl-Tóth
- Vascular Cognitive Impairment and Neurodegeneration Program, Oklahoma Center for Geroscience and Healthy Brain Aging, Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK USA
- International Training Program in Geroscience, Institute of Biophysics, Biological Research Centre, Eötvös Loránd Research Network (ELKH), Szeged, Hungary
- International Training Program in Geroscience, Doctoral School of Basic and Translational Medicine/Department of Public Health, Semmelweis University, Budapest, Hungary
| | - Rafal Gulej
- Vascular Cognitive Impairment and Neurodegeneration Program, Oklahoma Center for Geroscience and Healthy Brain Aging, Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK USA
| | - Stefano Tarantini
- Vascular Cognitive Impairment and Neurodegeneration Program, Oklahoma Center for Geroscience and Healthy Brain Aging, Department of Biochemistry and Molecular Biology, 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
- Department of Health Promotion Sciences, College of Public Health, University of Oklahoma Health Sciences Center, Oklahoma City, OK USA
- The Peggy and Charles Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104 USA
| | - Tamas Csipo
- Vascular Cognitive Impairment and Neurodegeneration Program, Oklahoma Center for Geroscience and Healthy Brain Aging, Department of Biochemistry and Molecular Biology, 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 and Neurodegeneration Program, Oklahoma Center for Geroscience and Healthy Brain Aging, Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK USA
- International Training Program in Geroscience, Doctoral School of Basic and Translational Medicine/Department of Physiology, Semmelweis University, Budapest, Hungary
| | - Anna Ungvari
- Vascular Cognitive Impairment and Neurodegeneration Program, Oklahoma Center for Geroscience and Healthy Brain Aging, Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK USA
| | - Priya Balasubramanian
- Vascular Cognitive Impairment and Neurodegeneration Program, Oklahoma Center for Geroscience and Healthy Brain Aging, Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK USA
| | - Andriy Yabluchanskiy
- Vascular Cognitive Impairment and Neurodegeneration Program, Oklahoma Center for Geroscience and Healthy Brain Aging, Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK USA
| | - Zoltan Benyo
- International Training Program in Geroscience, Doctoral School of Basic and Translational Medicine/Department of Translational Medicine, Semmelweis University, Budapest, Hungary
| | - Shannon M. Conley
- Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK USA
| | - Jonathan D. Wren
- Oklahoma Medical Research Foundation, Genes & Human Disease Research Program, Oklahoma City, OK USA
| | - Lori Garman
- Oklahoma Medical Research Foundation, Genes & Human Disease Research Program, Oklahoma City, OK USA
| | - Derek M. Huffman
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY 10461 USA
- Institute for Aging Research, Albert Einstein College of Medicine, Bronx, NY USA
- Department of Medicine, Albert Einstein College of Medicine, Bronx, NY USA
| | - Anna Csiszar
- Vascular Cognitive Impairment and Neurodegeneration Program, Oklahoma Center for Geroscience and Healthy Brain Aging, Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK USA
- International Training Program in Geroscience, Doctoral School of Basic and Translational Medicine/Department of Translational Medicine, Semmelweis University, Budapest, Hungary
- The Peggy and Charles Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104 USA
- International Training Program in Geroscience, Theoretical Medicine Doctoral School, University of Szeged, Szeged, Hungary
| | - Zoltan Ungvari
- Vascular Cognitive Impairment and Neurodegeneration Program, Oklahoma Center for Geroscience and Healthy Brain Aging, Department of Biochemistry and Molecular Biology, 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
- Department of Health Promotion Sciences, College of Public Health, University of Oklahoma Health Sciences Center, Oklahoma City, OK USA
- The Peggy and Charles Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104 USA
| |
Collapse
|
372
|
ABSTRACTS (BY NUMBER). Tissue Eng Part A 2022. [DOI: 10.1089/ten.tea.2022.29025.abstracts] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
|
373
|
Ouyang M, Zhang Q, Shu J, Wang Z, Fan J, Yu K, Lei L, Li Y, Wang Q. Capsaicin Ameliorates the Loosening of Mitochondria-Associated Endoplasmic Reticulum Membranes and Improves Cognitive Function in Rats With Chronic Cerebral Hypoperfusion. Front Cell Neurosci 2022; 16:822702. [PMID: 35370565 PMCID: PMC8968035 DOI: 10.3389/fncel.2022.822702] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2021] [Accepted: 02/10/2022] [Indexed: 12/17/2022] Open
Abstract
Based on accumulating evidence, vascular factors contribute to cognitive decline and dementia. Mitochondrial dysfunction is the core pathophysiological mechanism. Mitochondria-associated endoplasmic reticulum membranes (MAMs) are subcellular structures that physically and biologically connect mitochondria with the endoplasmic reticulum (ER) and regulate multiple functions ranging from calcium transfer to mitochondrial dynamics and bioenergetics. MAMs dysfunction has been speculated to be a key factor contributing to the pathogenesis of cognitive disorders and a new therapeutic target. However, the alteration of MAMs in vascular cognitive impairment remains to be revealed. Capsaicin, a specific agonist known to activated the transient receptor potential vanilloid type 1 (TRPV1), is involved in hippocampal synaptic plasticity and memory, but the detailed mechanism is still unclear. In this study, chronic cerebral hypoperfusion (CCH) model rats were created by bilateral common carotid artery occlusion (BCCAO), which is a widely used model to study vascular dementia. We observed that CCH rats showed obvious cognitive deficits, and ER-mitochondria contacts were loosener with lower expression of mitofusin2 (MFN2), a key protein connecting MAMs, in the hippocampal CA1 region, compared to the sham group. After capsaicin treatment for 12 weeks, we found that cognitive deficits induced by CCH were significantly alleviated and loosened ER-mitochondrial interactions were obviously improved. In conclusion, the findings of this study highlight that MAMs may contribute to the pathogenesis of cognitive impairment induced by CCH, and our new evidence that capsaicin improves cognitive function highlights a novel opportunity for drug discovery.
Collapse
Affiliation(s)
- Mengqi Ouyang
- Department of Neurology, The General Hospital of Western Theater Command, Chengdu, China
| | - Qi Zhang
- Department of Pharmacology, Gaoping District People’s Hospital of Nanchong, Nanchong, China
| | - Jiahui Shu
- Department of Pharmacology, Yichang Yiling Hospital, Yichang, China
| | - Zhiqiang Wang
- Department of Neurology, Chengdu BOE Hospital, Chengdu, China
| | - Jin Fan
- Department of Neurology, The General Hospital of Western Theater Command, Chengdu, China
| | - Ke Yu
- Department of Neurology, The General Hospital of Western Theater Command, Chengdu, China
| | - Lei Lei
- Department of Neurology, The General Hospital of Western Theater Command, Chengdu, China
| | - Yuxia Li
- Department of Neurology, Chengdu BOE Hospital, Chengdu, China
| | - Qingsong Wang
- Department of Neurology, The General Hospital of Western Theater Command, Chengdu, China
- *Correspondence: Qingsong Wang,
| |
Collapse
|
374
|
Fang X, Crumpler RF, Thomas KN, Mazique JN, Roman RJ, Fan F. Contribution of cerebral microvascular mechanisms to age-related cognitive impairment and dementia. Physiol Int 2022; 109:10.1556/2060.2022.00020. [PMID: 35238800 PMCID: PMC10710737 DOI: 10.1556/2060.2022.00020] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Accepted: 02/03/2022] [Indexed: 11/19/2022]
Abstract
Cognitive impairment and dementia are significant health burdens worldwide. Aging, hypertension, and diabetes are the primary risk factors for Alzheimer's disease and Alzheimer's disease and related dementias (AD/ADRD). There are no effective treatments for AD/ADRD to date. An emerging body of evidence indicates that cerebral vascular dysfunction and hypoperfusion precedes the development of other AD pathological phenotypes and cognitive impairment. However, vascular contribution to dementia is not currently well understood. This commentary highlights the emerging concepts and mechanisms underlying the microvascular contribution to AD/ADRD, including hypotheses targeting the anterograde and retrograde cerebral vascular pathways, as well as the cerebral capillaries and the venous system. We also briefly discuss vascular endothelial dysfunction, oxidative stress, inflammation, and cellular senescence that may contribute to impaired cerebral blood flow autoregulation, neurovascular uncoupling, and dysfunction of cerebral capillaries and the venous system.
Collapse
Affiliation(s)
- Xing Fang
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, MS 39216, USA
| | - Reece F. Crumpler
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, MS 39216, USA
| | - Kirby N. Thomas
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, MS 39216, USA
| | - Jena’ N. Mazique
- Program in Neuroscience, University of Mississippi Medical Center, Jackson, MS 39216, USA
| | - Richard J. Roman
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, MS 39216, USA
| | - Fan Fan
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, MS 39216, USA
- Program in Neuroscience, University of Mississippi Medical Center, Jackson, MS 39216, USA
| |
Collapse
|
375
|
Ji H, Kwan AC, Chen M, Ouyang D, Ebinger JE, Bell SP, Niiranen T, Bello NA, Cheng S. Sex Differences in Myocardial and Vascular Aging. Circ Res 2022; 130:566-577. [PMID: 35175845 PMCID: PMC8863105 DOI: 10.1161/circresaha.121.319902] [Citation(s) in RCA: 87] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
It is well known that cardiovascular disease manifests differently in women and men. The underlying causes of these differences during the aging lifespan are less well understood. Sex differences in cardiac and vascular phenotypes are seen in childhood and tend to track along distinct trajectories related to dimorphism in genetic factors as well as response to risk exposures and hormonal changes during the life course. These differences underlie sex-specific variation in cardiovascular events later in life, including myocardial infarction, heart failure, ischemic stroke, and peripheral vascular disease. With respect to cardiac phenotypes, females have intrinsically smaller body size-adjusted cardiac volumes and they tend to experience greater age-related wall thickening and myocardial stiffening with aging. With respect to vascular phenotypes, sexual dimorphism in both physiology and pathophysiology are also seen, including overt differences in blood pressure trajectories. The majority of sex differences in myocardial and vascular alterations that manifest with aging seem to follow relatively consistent trajectories from the very early to the very later stages of life. This review aims to synthesize recent cardiovascular aging-related research to highlight clinically relevant studies in diverse female and male populations that can inform approaches to improving the diagnosis, management, and prognosis of cardiovascular disease risks in the aging population at large.
Collapse
Affiliation(s)
- Hongwei Ji
- Department of Cardiology, the Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
- Department of Cardiology, Shanghai Tenth People’s Hospital, Tongji University, Shanghai, China
| | - Alan C. Kwan
- Department of Cardiology, Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Melanie Chen
- Department of Cardiology, Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - David Ouyang
- Department of Cardiology, Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Joseph E. Ebinger
- Department of Cardiology, Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Susan P. Bell
- Division of Cardiovascular Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Teemu Niiranen
- Department of Internal Medicine, University of Turku, Turku, Finland
- Department of Public Health Solutions, Finnish Institute for Health and Welfare, Turku, Finland
| | - Natalie A. Bello
- Department of Cardiology, Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Susan Cheng
- Department of Cardiology, Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| |
Collapse
|
376
|
Effect of Subclinical Hypothyroidism on the Association between Hemoglobin A1c and Reduced Renal Function: A Prospective Study. Diagnostics (Basel) 2022; 12:diagnostics12020462. [PMID: 35204553 PMCID: PMC8871099 DOI: 10.3390/diagnostics12020462] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2022] [Revised: 02/07/2022] [Accepted: 02/08/2022] [Indexed: 02/06/2023] Open
Abstract
Subclinical hypothyroidism (SCH) was reported to be associated with accelerating endothelial dysfunction, which is recognized as one of the upstream mechanisms that leads to glomerular injury (lower glomerular filtration rate (GFR)). SCH was also reported to be associated with hyperglycemia, which is associated with higher hemoglobin A1c (HbA1c) levels and induces endothelial dysfunction. Therefore, SCH status could influence the association between HbA1c and reduced eGFR. To clarify those associations, we conducted a prospective study of 1580 Japanese individuals who participated in an annual health check-up in 2014 with 2.8 years of follow-up. All participants had free triiodothyronine (T3) and free thyroxine (T4) levels in the normal range. Among study participants, 88 were diagnosed as having SCH. Even though no significant correlation was observed between HbA1c and annual change in estimated GFR among participants without SCH (multi-adjusted standardized parameter estimate (β) = 0.03, p = 0.250), a significant inverse association was observed among participants with SCH (β = −0.26, p = 0.014). When those analyses were performed among participants who were not taking glucose lowering medication, the observed associations were essentially the same: β = 0.03, p = 0.266 for participants without SCH and β = −0.32, p = 0.006 for participants with SCH, respectively. Therefore, SCH status could influence the association between HbA1c and renal function.
Collapse
|
377
|
Montano D. Frequency and Associations of Adverse Reactions of COVID-19 Vaccines Reported to Pharmacovigilance Systems in the European Union and the United States. Front Public Health 2022; 9:756633. [PMID: 35186864 PMCID: PMC8850379 DOI: 10.3389/fpubh.2021.756633] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2021] [Accepted: 12/10/2021] [Indexed: 12/12/2022] Open
Abstract
IntroductionThis study aims to provide a risk assessment of the adverse reactions related to the COVID-19 vaccines manufactured by AstraZeneca, Janssen, Moderna, and Pfizer-BioNTech which have been in use in the European Union and the United States between December 2020 and October 2021.MethodsData from the European Database of Suspected Adverse Drug Reaction (EudraVigilance) and the Vaccine Adverse Events Reporting System (VAERS) from 2020 to October 2021 are analysed. More than 7.8 million adverse reactions of about 1.6 million persons are included. The adverse reactions are classified with the Common Toxicity Criteria (CTC) categories. COVID-19 vaccine exposures and adverse reactions reported between December 2020 and October 2021 are compared to influenza vaccine exposures and adverse reactions reported between 2020 and 2021. The population-level vaccine exposures to COVID-19 and influenza vaccines comprised about 451 million and 437 million exposures, respectively. Absolute and relative risk estimates are calculated by CTC categories and COVID-19 vaccines for the EU and US populations aged 18 years and older.ResultsA higher risk of reporting serious adverse reactions was observed for the COVID-19 vaccines in comparison to the influenza vaccines. Individuals age 65 and older were associated with a higher frequency of death, hospitalisations, and life-threatening reactions than younger individuals (relative risk estimates between 1.49 99% CI [1.44–1.55] and 8.61 99% CI [8.02–9.23]). Outcome onset of serious adverse reactions occurred within the first 7 days after vaccination in about 77.6–89.1% of cases. The largest absolute risks were observed for allergic, constitutional reactions, dermatological, gastrointestinal, neurological reactions, and localised and non-localised pain. The largest relative risks between COVID-19 vs. influenza vaccines were observed for allergic reactions, arrhythmia, general cardiovascular events, coagulation, haemorrhages, gastrointestinal, ocular, sexual organs reactions, and thrombosis.ConclusionThe present study provides an overview of adverse reactions frequently reported to the pharmacovigilance systems following COVID-19 vaccination in the EU and US populations. Despite the limitations of passive reporting systems, these results may inform further clinical research investigating in more detail the pathophysiological mechanisms potentially associated with the COVID-19 vaccines.
Collapse
|
378
|
Chen Y, Liu Z, Chen H, Huang X, Huang X, Lei Y, Liang Q, Wei J, Zhang Q, Guo X, Huang Q. p53 SUMOylation Mediates AOPP-Induced Endothelial Senescence and Apoptosis Evasion. Front Cardiovasc Med 2022; 8:795747. [PMID: 35187108 PMCID: PMC8850781 DOI: 10.3389/fcvm.2021.795747] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2021] [Accepted: 12/30/2021] [Indexed: 12/14/2022] Open
Abstract
The aging of endothelial cells plays a critical role in the development of age-related vascular disease. We established a model of endothelial premature senescence by application of Advanced oxidation protein products (AOPPs) modified bovine serum albumin (AOPP-BSA) in human umbilical vein endothelial cells (HUVECs). This cellular senescence was accompanied with endothelial barrier dysfunction and angiogenesis impairment. It was further revealed that these senescent HUVECs underwent apoptosis evasion and the receptor for advanced glycation endproducts (RAGE) played a role in these processes. The AOPP-induced senescence was regulated by the state of autophagy in HUVECs. We further proved that AOPP-BSA attenuated the autophagy of HUVECs, led to p53 SUMOylation at K386, resulting in endothelial senescence. We also established the animal model of vascular senescence by using ApoE−/− mice fed with high-fat diet plus daily injection of AOPP-BSA to verify the role of p53 SUMOylation in vascular senescence. Combined with intraperitoneal injection of rapamycin, the effect of autophagy on AOPP-induced p53 SUMOylation was also confirmed in vivo. Our data indicates that p53 SUMOylation at K386 plays an important role in AOPP-induced endothelial senescence and apoptosis evasion, suggesting that p53 K386 SUMOylation may serve as a potential therapeutic target in protecting against vascular senescence.
Collapse
Affiliation(s)
- Yanjia Chen
- Department of Pathophysiology, Guangdong Provincial Key Laboratory of Shock and Microcirculation, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
- Department of Anesthesiology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Zhuanhua Liu
- Department of Pathophysiology, Guangdong Provincial Key Laboratory of Shock and Microcirculation, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Hongyu Chen
- Department of Pathophysiology, Guangdong Provincial Key Laboratory of Shock and Microcirculation, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Xingfu Huang
- Department of Cardiology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Xiaoxia Huang
- Department of Pathophysiology, Guangdong Provincial Key Laboratory of Shock and Microcirculation, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Yang Lei
- Department of Pathophysiology, Guangdong Provincial Key Laboratory of Shock and Microcirculation, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Qing Liang
- Department of Pathophysiology, Guangdong Provincial Key Laboratory of Shock and Microcirculation, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Jiayi Wei
- Department of Pathophysiology, Guangdong Provincial Key Laboratory of Shock and Microcirculation, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Qin Zhang
- Department of Pathophysiology, Guangdong Provincial Key Laboratory of Shock and Microcirculation, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Xiaohua Guo
- Department of Pathophysiology, Guangdong Provincial Key Laboratory of Shock and Microcirculation, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Qiaobing Huang
- Department of Pathophysiology, Guangdong Provincial Key Laboratory of Shock and Microcirculation, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
- *Correspondence: Qiaobing Huang
| |
Collapse
|
379
|
Tian Y, Fopiano KA, Buncha V, Lang L, Rudic RD, Filosa JA, Dou H, Bagi Z. Aging-induced impaired endothelial wall shear stress mechanosensing causes arterial remodeling via JAM-A/F11R shedding by ADAM17. GeroScience 2022; 44:349-369. [PMID: 34718985 PMCID: PMC8810930 DOI: 10.1007/s11357-021-00476-1] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2021] [Accepted: 10/08/2021] [Indexed: 11/25/2022] Open
Abstract
Physiological and pathological vascular remodeling is uniquely driven by mechanical forces from blood flow in which wall shear stress (WSS) mechanosensing by the vascular endothelium plays a pivotal role. This study aimed to determine the novel role for a disintegrin and metalloproteinase 17 (ADAM17) in impaired WSS mechanosensing, which was hypothesized to contribute to aging-associated abnormal vascular remodeling. Without changes in arterial blood pressure and blood flow rate, skeletal muscle resistance arteries of aged mice (30-month-old vs. 12-week-old) exhibited impaired WSS mechanosensing and displayed inward hypertrophic arterial remodeling. These vascular changes were recapitulated by in vivo confined, AAV9-mediated overexpression of ADAM17 in the resistance arteries of young mice. An aging-related increase in ADAM17 expression reduced the endothelial junction level of its cleavage substrate, junctional adhesion molecule-A/F11 receptor (JAM-A/F11R). In cultured endothelial cells subjected to steady WSS ADAM17 activation or JAM-A/F11R knockdown inhibited WSS mechanosensing. The ADAM17-activation induced, impaired WSS mechanosensing was normalized by overexpression of ADAM17 cleavage resistant, mutated JAM-AV232Y both in cultured endothelial cells and in resistance arteries of aged mice, in vivo. These data demonstrate a novel role for ADAM17 in JAM-A/F11R cleavage-mediated impaired endothelial WSS mechanosensing and subsequently developed abnormal arterial remodeling in aging. ADAM17 could prove to be a key regulator of WSS mechanosensing, whereby it can also play a role in pathological vascular remodeling in diseases.
Collapse
Affiliation(s)
- Yanna Tian
- Department of Physiology, Medical College of Georgia, Augusta University, Augusta, GA, 30912, USA
| | - Katie Anne Fopiano
- Department of Physiology, Medical College of Georgia, Augusta University, Augusta, GA, 30912, USA
| | - Vadym Buncha
- Department of Physiology, Medical College of Georgia, Augusta University, Augusta, GA, 30912, USA
| | - Liwei Lang
- Department of Physiology, Medical College of Georgia, Augusta University, Augusta, GA, 30912, USA
| | - R Daniel Rudic
- Department of Pharmacology and Toxicology, Medical College of Georgia, Augusta University, Augusta, GA, 30912, USA
| | - Jessica A Filosa
- Department of Physiology, Medical College of Georgia, Augusta University, Augusta, GA, 30912, USA
| | - Huijuan Dou
- Department of Physiology, Medical College of Georgia, Augusta University, Augusta, GA, 30912, USA
- Department of Medicine, Columbia University Medical Center, New York, NY, 10032, USA
| | - Zsolt Bagi
- Department of Physiology, Medical College of Georgia, Augusta University, Augusta, GA, 30912, USA.
| |
Collapse
|
380
|
Zhang H, Roman RJ, Fan F. Hippocampus is more susceptible to hypoxic injury: has the Rosetta Stone of regional variation in neurovascular coupling been deciphered? GeroScience 2022; 44:127-130. [PMID: 34453273 PMCID: PMC8810993 DOI: 10.1007/s11357-021-00449-4] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2021] [Accepted: 08/23/2021] [Indexed: 10/20/2022] Open
Abstract
Alzheimer's disease and Alzheimer's disease-related dementias (AD/ADRD) are associated with cerebral hypoperfusion or reductions in baseline cerebral blood flow (CBF). The neurovascular coupling (NVC) response or functional hyperemia regulates brain perfusion via a retrograde (capillary-to-arteriole) pathway by increasing regional CBF in response to local neuron activation. The hippocampus plays a significant role in spatial and non-spatial memory. Functional MRI (fMRI) has not established a solid positive correlation between hippocampal blood oxygen level-dependent (BOLD) signal and local neuronal activity. The inconsistency of NVC in the hippocampus compared to the neocortex is possibly due to anatomical and methodological difficulties to accurately detect hippocampal blood flow. A recent study reported that NVC and oxygenation are reduced in the hippocampus compared to the cortex using a novel invasive surgical approach by creating a cranial window with and without removing the neocortex. Results from these studies suggest that the hippocampus is more susceptible to hypoxic injury in pathological conditions when NVC is impaired, such as AD/ADRD, stroke, and traumatic brain injury (TBI). The Rosetta Stone of regional variation in the NVC and its significance in AD/ADRD has not been fully deciphered based on these results without addressing remaining concerns; however, we are one step closer, indeed.
Collapse
Affiliation(s)
- Huawei Zhang
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, 2500 North State Street, Jackson, MS, 39216, USA
| | - Richard J Roman
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, 2500 North State Street, Jackson, MS, 39216, USA
| | - Fan Fan
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, 2500 North State Street, Jackson, MS, 39216, USA.
| |
Collapse
|
381
|
Kaur M, Sharma S. Molecular mechanisms of cognitive impairment associated with stroke. Metab Brain Dis 2022; 37:279-287. [PMID: 35029798 DOI: 10.1007/s11011-022-00901-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/09/2021] [Accepted: 01/03/2022] [Indexed: 10/19/2022]
Abstract
Stroke is the second leading cause of death after coronary heart disease in developed countries and is the greatest cause of disability and cognitive impairment. Risk factors for cognitive impairment and dementia after stroke are multifactorial including older age, family history, hypertension, arterial fibrillation, diabetes, genetic variants, low educational status, vascular comorbidities, prior transient ischaemic attack or recurrent stroke, depressive illness duration of a stroke, location, volume, intensity, and degree of neuronal degeneration, location and size of infarction after stroke, time interval after stroke other cerebral dysfunctions. The pathophysiology of stroke associated cognitive impairment is complex and recent molecular, cellular, and animal models studies have revealed that multiple cellular changes have been implicated, including altered redox state, mitochondrial dysfunction, disruption of the blood-brain barrier, perivascular spacing, glymphatic system impairment, microglia activation and amyloid-β deposition in the parenchyma of the brain. These studies have also evidenced the involvement of various transcription factors, intracellular adhesion molecules, and endogenous growth factors in the pathogenesis of cognitive impairment associated with stroke and providing scope for developing therapeutic strategies for treatment. This review summarizes the latest research findings on molecular mechanisms involved in cognitive impairment associated with stroke.
Collapse
Affiliation(s)
- Mandeep Kaur
- Department of Pharmacology, School of Pharmaceutical Sciences, CT University, Ludhiana, Punjab, India
| | - Saurabh Sharma
- School of Pharmaceutical Sciences, CT University, Ludhiana, Punjab, India.
| |
Collapse
|
382
|
Zhang S, Laubrie JD, Mousavi SJ, Avril S. 3D finite-element modeling of vascular adaptation after endovascular aneurysm repair. INTERNATIONAL JOURNAL FOR NUMERICAL METHODS IN BIOMEDICAL ENGINEERING 2022; 38:e3547. [PMID: 34719114 DOI: 10.1002/cnm.3547] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Accepted: 10/27/2021] [Indexed: 06/13/2023]
Abstract
Aneurysm shrinkage is clinically observed after successful endovascular aortic aneurysm repair (EVAR). However, global understanding of post-operative aneurysm evolutions remains weak. In this work, we propose to study these effects using numerical simulation. We set up a 3D finite-element model of post-EVAR vascular adaptation within an open-source finite-element code, which was initially developed for growth and remodeling (G&R). We modeled the endograft with a set of uniaxial prestrained springs that apply radial forces on the inner surface of the artery. Constitutive equations, momentum balance equations, and equations related to the mechanobiology of the artery were formulated based on the homogenized constrained mixture theory. We performed a sensitivity analysis by varying different selected parameters, namely oversizing and compliance of the stent-graft, gain parameters related to collagen G&R, and the residual pressure in the aneurysm sac. This permitted us to evaluate how each factor influences post-EVAR vascular adaptation. It was found that oversizing, compliance or gain parameters have a limited influence compared to that of the residual pressure in the aneurysm sac, which was found to play a critical role in the stability of aneurysm after stent-graft implantation. An excessive residual pressure larger than 50 mmHg can induce a continuous expansion of the aneurysm while a moderate residual pressure below this critical threshold yields continuous shrinkage of the aneurysm. Moreover, it was found that elderly patients, with relatively lower amounts of remnant elastin in the arterial wall, are more sensitive to the effect of residual pressure. Therefore, these results show that elderly patients may present a higher potential risk of aortic sac expansion due to intra-aneurysm sac pressure after EVAR than younger patients.
Collapse
Affiliation(s)
- Shaojie Zhang
- Mines Saint-Étienne, Univ Lyon, Univ Jean Monnet, INSERM, U 1059 Sainbiose, Saint-Étienne, France
| | - Joan D Laubrie
- Mines Saint-Étienne, Univ Lyon, Univ Jean Monnet, INSERM, U 1059 Sainbiose, Saint-Étienne, France
| | - S Jamaleddin Mousavi
- Mines Saint-Étienne, Univ Lyon, Univ Jean Monnet, INSERM, U 1059 Sainbiose, Saint-Étienne, France
| | - Stéphane Avril
- Mines Saint-Étienne, Univ Lyon, Univ Jean Monnet, INSERM, U 1059 Sainbiose, Saint-Étienne, France
| |
Collapse
|
383
|
Banks NF, Rogers EM, Church DD, Ferrando AA, Jenkins NDM. The contributory role of vascular health in age-related anabolic resistance. J Cachexia Sarcopenia Muscle 2022; 13:114-127. [PMID: 34951146 PMCID: PMC8818606 DOI: 10.1002/jcsm.12898] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Revised: 10/18/2021] [Accepted: 11/22/2021] [Indexed: 12/25/2022] Open
Abstract
Sarcopenia, or the age-related loss of skeletal muscle mass and function, is an increasingly prevalent condition that contributes to reduced quality of life, morbidity, and mortality in older adults. Older adults display blunted anabolic responses to otherwise anabolic stimuli-a phenomenon that has been termed anabolic resistance (AR)-which is likely a casual factor in sarcopenia development. AR is multifaceted, but historically much of the mechanistic focus has been on signalling impairments, and less focus has been placed on the role of the vasculature in postprandial protein kinetics. The vascular endothelium plays an indispensable role in regulating vascular tone and blood flow, and age-related impairments in vascular health may impede nutrient-stimulated vasodilation and subsequently the ability to deliver nutrients (e.g. amino acids) to skeletal muscle. Although the majority of data has been obtained studying younger adults, the relatively limited data on the effect of blood flow on protein kinetics in older adults suggest that vasodilatory function, especially of the microvasculature, strongly influences the muscle protein synthetic response to amino acid feedings. In this narrative review, we examine evidence of AR in older adults following amino acid and mixed meal consumption, examine the evidence linking vascular dysfunction and insulin resistance to age-related AR, review the influence of nitric oxide and endothelin-1 on age-related vascular dysfunction as it relates to AR, briefly review the potential causal role of arterial stiffness in promoting skeletal muscle microvascular dysfunction and AR, and provide a brief overview and future considerations for research examining age-related AR.
Collapse
Affiliation(s)
- Nile F Banks
- Integrative Laboratory of Applied Physiology and Lifestyle Medicine, University of Iowa, Iowa City, IA, USA
| | - Emily M Rogers
- Integrative Laboratory of Applied Physiology and Lifestyle Medicine, University of Iowa, Iowa City, IA, USA
| | - David D Church
- Center for Translational Research in Aging and Longevity, College of Medicine, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Arny A Ferrando
- Center for Translational Research in Aging and Longevity, College of Medicine, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Nathaniel D M Jenkins
- Integrative Laboratory of Applied Physiology and Lifestyle Medicine, University of Iowa, Iowa City, IA, USA.,Abboud Cardiovascular Research Center, University of Iowa, Iowa City, IA, USA
| |
Collapse
|
384
|
You Q, Yu L, Li G, He H, Lv Y. Effects of Different Intensities and Durations of Aerobic Exercise on Vascular Endothelial Function in Middle-Aged and Elderly People: A Meta-analysis. Front Physiol 2022; 12:803102. [PMID: 35126182 PMCID: PMC8814456 DOI: 10.3389/fphys.2021.803102] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Accepted: 12/06/2021] [Indexed: 12/28/2022] Open
Abstract
Background Previous studies have found that aerobic exercise was more effective in improving vascular endothelial function than resistance training, high-intensity interval training (HIIT), and other types of exercise, while the effects between different intensities and durations of aerobic exercise were unclear. Therefore, we performed this meta-analysis to investigate the effects of different intensities and durations of aerobic exercise on the vascular endothelial function of middle-aged and elderly people. Methods: Databases were searched up to April 2021 for studies evaluating the influences of different intensities and durations of aerobic exercise on endothelial function assessed by flow-mediated dilation (FMD) among healthy middle-aged and elderly people. Data were pooled using random-effects models to obtain the weighted mean difference (WMD) and 95% confidence intervals (CIs). Results A total of 9 studies involving 221 participants fulfilled the inclusion criteria. Aerobic exercise improved the overall FMD of healthy middle-aged and elderly people [WMD, 1.33 (95% CI, 0.37–2.28), P < 0.05]. Specifically, vigorous-intensity exercise increased FMD significantly in healthy middle-aged and elderly people [WMD, 1.10 (95% CI, 0.27–1.93), P < 0.05], while moderate-intensity exercise had no significant association with FMD [WMD, 1.49 (95% CI, −0.62 to 3.60), P = 0.17]. In addition, long-term (8 weeks or above) aerobic exercise increased the FMD in healthy middle-aged and elderly people [WMD, 1.63 (95% CI, 0.61–2.66), P < 0.05], while one-time acute aerobic exercise had no significant association with FMD of healthy middle-aged and elderly people [WMD, 0.89 (95% CI, −1.47 to 3.24), P = 0.46]. Specifically, 8 weeks or above of vigorous-intensity exercise increased FMD significantly in healthy middle-aged and elderly people [WMD, 1.48 (95% CI, 1.06–1.90), P < 0.01], while 8 weeks or above of moderate aerobic exercise had no significant association with FMD [WMD, 1.49 (95% CI, −0.62 to 3.60), P = 0.17]. Conclusion Aerobic exercise, especially 8 weeks or above of vigorous-intensity aerobic exercise, improved the endothelial function in healthy middle-aged and elderly people.
Collapse
Affiliation(s)
- Qiuping You
- Sports Coaching College, Beijing Sport University, Beijing, China
| | - Laikang Yu
- Department of Strength and Conditioning Training, Beijing Sport University, Beijing, China
| | - Gen Li
- Department of Strength and Conditioning Training, Beijing Sport University, Beijing, China
| | - Hui He
- China Institute of Sport and Health Science, Beijing Sport University, Beijing, China
| | - Yuanyuan Lv
- China Institute of Sport and Health Science, Beijing Sport University, Beijing, China
- *Correspondence: Yuanyuan Lv
| |
Collapse
|
385
|
Age-related Activation of Cyclic GMP-AMP synthase-Stimulator of Interferon Genes Signaling in the Auditory System is Associated with Presbycusis in C57BL/6J Male Mice. Neuroscience 2022; 481:73-84. [PMID: 34848262 DOI: 10.1016/j.neuroscience.2021.11.031] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2021] [Revised: 10/29/2021] [Accepted: 11/20/2021] [Indexed: 11/21/2022]
Abstract
Presbycusis, or age-related hearing loss (ARHL), is primarily associated with sensory or transduction nerve cell degeneration in the peripheral and/or central auditory systems. During aging, the auditory system shows mitochondrial dysfunction and increased inflammatory responses. Mitochondrial dysfunction promotes leakage of mitochondrial DNA (mtDNA) into the cytosol, which activates the cyclic GMP-AMP synthase (cGAS)-stimulator of interferon genes (STING) pathway to induce type I interferon and inflammatory responses. However, whether this pathway is involved in the occurrence and development of ARHL is unknown. This study aimed to determine whether there are age-related changes in the levels of cytosolic mtDNA and cGAS-STING pathway activation in the auditory pathway and to explore their relationship with ARHL. The results showed that cGAS-positive immunoreactive cells were observed in the cochlea, inferior colliculus, and auditory cortex. Levels of cytosolic mtDNA, cGAS, STING, phosphorylated interferon regulatory factor 3, and cytokines were significantly increased in the cochlea, inferior colliculus, and auditory cortex of 6-, 9-, and 12-month-old mice compared with 3-month-old mice. These findings suggested that cytosolic mtDNA may play an important role in the pathogenesis of ARHL by activating cGAS-STING-mediated type I interferon and inflammatory responses.
Collapse
|
386
|
Ge F, Pan Q, Qin Y, Jia M, Ruan C, Wei X, Jing Q, Zhi X, Wang X, Jiang L, Osto E, Guo J, Meng D. Single-Cell Analysis Identify Transcription Factor BACH1 as a Master Regulator Gene in Vascular Cells During Aging. Front Cell Dev Biol 2022; 9:786496. [PMID: 35004685 PMCID: PMC8740196 DOI: 10.3389/fcell.2021.786496] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Accepted: 11/23/2021] [Indexed: 12/13/2022] Open
Abstract
Vascular aging is a potent driver of cardiovascular and cerebrovascular diseases. Vascular aging features cellular and functional changes, while its molecular mechanisms and the cell heterogeneity are poorly understood. This study aims to 1) explore the cellular and molecular properties of aged cardiac vasculature in monkey and mouse and 2) demonstrate the role of transcription factor BACH1 in the regulation of endothelial cell (EC) senescence and its mechanisms. Here we analyzed published single-cell RNA sequencing (scRNA-seq) data from monkey coronary arteries and aortic arches and mouse hearts. We revealed that the gene expression of YAP1, insulin receptor, and VEGF receptor 2 was downregulated in both aged ECs of coronary arteries’ of monkey and aged cardiac capillary ECs of mouse, and proliferation-related cardiac capillary ECs were significantly decreased in aged mouse. Increased interaction of ECs and immunocytes was observed in aged vasculature of both monkey and mouse. Gene regulatory network analysis identified BACH1 as a master regulator of aging-related genes in both coronary and aorta ECs of monkey and cardiac ECs of mouse. The expression of BACH1 was upregulated in aged cardiac ECs and aortas of mouse. BACH1 aggravated endothelial cell senescence under oxidative stress. Mechanistically, BACH1 occupied at regions of open chromatin and bound to CDKN1A (encoding for P21) gene enhancers, activating its transcription in senescent human umbilical vein endothelial cells (HUVECs). Thus, these findings demonstrate that BACH1 plays an important role in endothelial cell senescence and vascular aging.
Collapse
Affiliation(s)
- Fei Ge
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Department of Rheumatology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Qi Pan
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Department of Rheumatology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Yue Qin
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Department of Rheumatology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Mengping Jia
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Department of Rheumatology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Chengchao Ruan
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Department of Rheumatology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Xiangxiang Wei
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Department of Rheumatology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Qing Jing
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Xiuling Zhi
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Department of Rheumatology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Xinhong Wang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Department of Rheumatology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Lindi Jiang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Department of Rheumatology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Elena Osto
- Institute of Clinical Chemistry and Department of Cardiology, University Heart Center, University and University Hospital Zurich, Zurich, Switzerland
| | - Jieyu Guo
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Department of Rheumatology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Dan Meng
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Department of Rheumatology, Zhongshan Hospital, Fudan University, Shanghai, China
| |
Collapse
|
387
|
Wang X, Schepler H, Neufurth M, Wang S, Schröder HC, Müller WEG. Polyphosphate in Chronic Wound Healing: Restoration of Impaired Metabolic Energy State. PROGRESS IN MOLECULAR AND SUBCELLULAR BIOLOGY 2022; 61:51-82. [PMID: 35697937 DOI: 10.1007/978-3-031-01237-2_4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Many pathological conditions are characterized by a deficiency of metabolic energy. A prominent example is nonhealing or difficult-to-heal chronic wounds. Because of their unique ability to serve as a source of metabolic energy, inorganic polyphosphates (polyP) offer the opportunity to develop novel strategies to treat such wounds. The basis is the generation of ATP from the polymer through the joint action of two extracellular or plasma membrane-bound enzymes alkaline phosphatase and adenylate kinase, which enable the transfer of energy-rich phosphate from polyP to AMP with the formation of ADP and finally ATP. Building on these findings, it was possible to develop novel regeneratively active materials for wound therapy, which have already been successfully evaluated in first studies on patients.
Collapse
Affiliation(s)
- Xiaohong Wang
- ERC Advanced Investigator Group, Institute for Physiological Chemistry, University Medical Center of the Johannes Gutenberg University, Mainz, Germany
| | - Hadrian Schepler
- Department of Dermatology, University Clinic Mainz, Mainz, Germany
| | - Meik Neufurth
- ERC Advanced Investigator Group, Institute for Physiological Chemistry, University Medical Center of the Johannes Gutenberg University, Mainz, Germany
| | - Shunfeng Wang
- ERC Advanced Investigator Group, Institute for Physiological Chemistry, University Medical Center of the Johannes Gutenberg University, Mainz, Germany
| | - Heinz C Schröder
- ERC Advanced Investigator Group, Institute for Physiological Chemistry, University Medical Center of the Johannes Gutenberg University, Mainz, Germany
| | - Werner E G Müller
- ERC Advanced Investigator Group, Institute for Physiological Chemistry, University Medical Center of the Johannes Gutenberg University, Mainz, Germany.
| |
Collapse
|
388
|
Sun X, Meng X, Zhang P, Wang L, Ren Y, Xu G, Yang T, Liu M. Quantification of pulmonary vessel volumes on low-dose computed tomography in a healthy male Chinese population: the effects of aging and smoking. Quant Imaging Med Surg 2022; 12:406-416. [PMID: 34993089 DOI: 10.21037/qims-21-160] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2021] [Accepted: 06/24/2021] [Indexed: 12/20/2022]
Abstract
BACKGROUND This study sought to determine pulmonary vascular volumes (PVVs) on low-dose computed tomography (LDCT) in a healthy male Chinese population and analyze the effects of aging and smoking on PVVs. METHODS A total of 1,320 healthy male participants (comprising 720 non-smokers, 445 smokers, and 155 ex-smokers) who underwent LDCT were retrospectively included in this study. Their demographic data and smoking status data were collected. An automatic integration segmentation approach for LDCT was used to segment pulmonary vessels semi-automatically. The PVVs of the whole lung, left lung, and right lung on LDCT were calculated, and correlations between PVVs and age and smoking status were then compared. RESULTS The inter-rater correlation coefficient of the whole lung, left lung, and right lung PVVs was 0.98 [95% confidence interval (CI): 0.95-0.99], 0.97 (95% CI: 0.93-0.98), and 0.97 (95% CI: 0.94-0.99), respectively. The intra-class correlation coefficient of the whole lung left lung, and right lung PVVs was 0.98 (95% CI: 0.95-0.99), 0.96 (95% CI: 0.95-0.99), and 0.96 (95% CI: 0.92-0.98), respectively. In non-smokers, PVVs decreased with age. The PVVs of heavy smokers were higher than those of light smokers, ex-smokers, and non-smokers. The PVVs of ex-smokers were comparable to those of light smokers. CONCLUSIONS The PVVs measured on LDCT tended to decrease with age in healthy male non-smokers gradually. Compared to non-smokers, the PVVs of smokers increased, even with the normal lung function.
Collapse
Affiliation(s)
- Xuebiao Sun
- Department of Radiology, China-Japan Friendship Hospital, Beijing, China
| | - Xiapei Meng
- Department of Radiology, China-Japan Friendship Hospital, Beijing, China
| | - Peiyao Zhang
- Department of Radiology, China-Japan Friendship Hospital, Beijing, China
| | - Lei Wang
- Department of Radiology, China-Japan Friendship Hospital, Beijing, China
| | - Yanhong Ren
- Department of Pulmonary and Critical Care Medicine, China-Japan Friendship Hospital, Beijing, China
| | - Guodong Xu
- Institute of Clinical Medicine, China-Japan Friendship Hospital, Beijing, China
| | - Ting Yang
- Department of Pulmonary and Critical Care Medicine, China-Japan Friendship Hospital, Beijing, China
| | - Min Liu
- Department of Radiology, China-Japan Friendship Hospital, Beijing, China
| |
Collapse
|
389
|
Franco ACC, Carneiro LDS, Franco RSM, Góes Junior AMDO. Influence of sex and age on inferior vena cava diameter and implications for the implantation of vena cava filters. J Vasc Bras 2022. [DOI: 10.1590/1677-5449.202101472] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Abstract Background Measuring the venous diameter and choosing a compatible vena cava filter are essential to reduce the risk of complications resulting from implantation of these devices. However, there is little information on how the diameter of the inferior vena cava varies with sex and age. Objectives To determine the influence of patients’ gender and age on their inferior vena cava diameter and the suitability of the different models of available filters. Methods Retrospective analytical study based on computed tomography images. The diameter of the inferior vena cava was measured at 3 points: above the confluence of the common iliac veins, below the renal veins, and midway between these two points (cranial point, caudal point, and midpoint) using Arya® and Carestream PACS® software. The results were classified by sex and age groups. Results CT scans of 417 patients were analyzed: 245 women and 172 men. The diameters at the midpoint and caudal point were, respectively, 19.1 mm and 20.6 mm in women from 81 to 92 years old and were statistically smaller (p< 0.05) when compared to women aged 19 to 40 years (midpoint: 22.7 mm; caudal point: 23 mm). Similar results were seen in men. Venous diameters at the cranial and caudal points in patients aged from 51 to 70 years were statistically larger in men (cranial point: 24.4 mm; caudal point:22.3 mm) than in women (cranial point: 22.6 mm; caudal point:20.8 mm) (p< 0.05). Conclusions A smaller diameter was found for the inferior vena cava in older patients of both sexes and the rate of diameter change was similar among men and women.
Collapse
|
390
|
Labissiere X, Zigmond ZM, Challa A, Montoya C, Manzur-Pineda K, Abraham A, Tabbara M, Salama A, Pan Y, Salman LH, Yang X, Vazquez-Padron RI, Martinez L. Vein morphometry in end-stage kidney disease: Teasing out the contribution of age, comorbidities, and vintage to chronic wall remodeling. Front Cardiovasc Med 2022; 9:1005030. [PMID: 36419492 PMCID: PMC9676677 DOI: 10.3389/fcvm.2022.1005030] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Accepted: 10/10/2022] [Indexed: 11/09/2022] Open
Abstract
Background Chronic kidney disease (CKD) is a highly comorbid condition with significant effects on vascular health and remodeling. Upper extremity veins are important in end-stage kidney disease (ESKD) due to their potential use to create vascular accesses. However, unlike arteries, the contribution of CKD-associated factors to the chronic remodeling of veins has been barely studied. Methods We measured morphometric parameters in 315 upper extremity veins, 131 (85% basilic) from stage 5 CKD/ESKD patients and 184 (89% basilic) from non-CKD organ donors. Associations of demographic and clinical characteristics with intimal hyperplasia (IH) and medial fibrosis were evaluated using multivariate regression models. Results The study cohort included 33% females, 30% blacks, 32% Hispanics, and 37% whites. Over 60% had hypertension, and 25% had diabetes independent of CKD status. Among kidney disease participants, 26% had stage 5 CKD, while 22 and 52% had ESKD with and without history of a previous arteriovenous fistula/graft (AVF/AVG), respectively. Intimal hyperplasia was associated with older age (β = 0.13 per year, confidence interval [CI] = 0.002-0.26), dialysis vintage > 12 months (β = 0.22, CI = 0.09-0.35), and previous AVF/AVG creation (β = 0.19, CI = 0.06-0.32). Upper quartile values of IH were significantly associated with diabetes (odds ratio [OR] = 2.02, CI = 1.08-3.80), which demonstrated an additive effect with previous AVF/AVG history and longer vintage in exacerbating IH. Medial fibrosis also increased as a function of age (β = 0.17, CI = 0.04-0.30) and among patients with diabetes (β = 0.15, CI = 0.03-0.28). Age was the predominant factor predicting upper quartile values of fibrosis (OR = 1.03 per year, CI = 1.01-1.05) independent of other comorbidities. Conclusion Age and diabetes are the most important risk factors for chronic development of venous IH and fibrosis independent of CKD status. Among kidney disease patients, longer dialysis vintage, and history of a previous AVF/AVG are strong predictors of IH.
Collapse
Affiliation(s)
- Xochilt Labissiere
- DeWitt Daughtry Family Department of Surgery, Leonard M. Miller School of Medicine, University of Miami, Miami, FL, United States
| | - Zachary M Zigmond
- Bruce W. Carter Veterans Affairs Medical Center, Miami, FL, United States
| | - Akshara Challa
- DeWitt Daughtry Family Department of Surgery, Leonard M. Miller School of Medicine, University of Miami, Miami, FL, United States
| | - Christopher Montoya
- DeWitt Daughtry Family Department of Surgery, Leonard M. Miller School of Medicine, University of Miami, Miami, FL, United States
| | - Karen Manzur-Pineda
- DeWitt Daughtry Family Department of Surgery, Leonard M. Miller School of Medicine, University of Miami, Miami, FL, United States
| | - Amalia Abraham
- DeWitt Daughtry Family Department of Surgery, Leonard M. Miller School of Medicine, University of Miami, Miami, FL, United States
| | - Marwan Tabbara
- DeWitt Daughtry Family Department of Surgery, Leonard M. Miller School of Medicine, University of Miami, Miami, FL, United States
| | - Alghidak Salama
- DeWitt Daughtry Family Department of Surgery, Leonard M. Miller School of Medicine, University of Miami, Miami, FL, United States
| | - Yue Pan
- Department of Public Health Sciences, Leonard M. Miller School of Medicine, University of Miami, Miami, FL, United States
| | - Loay H Salman
- Division of Nephrology, Albany Medical College, Albany, NY, United States
| | - Xiaofeng Yang
- Lewis Katz School of Medicine, Temple University, Philadelphia, PA, United States
| | - Roberto I Vazquez-Padron
- DeWitt Daughtry Family Department of Surgery, Leonard M. Miller School of Medicine, University of Miami, Miami, FL, United States.,Bruce W. Carter Veterans Affairs Medical Center, Miami, FL, United States
| | - Laisel Martinez
- DeWitt Daughtry Family Department of Surgery, Leonard M. Miller School of Medicine, University of Miami, Miami, FL, United States
| |
Collapse
|
391
|
Du Y, Mao L, Wang Z, Yan K, Zhang L, Zou J. Osteopontin - The stirring multifunctional regulatory factor in multisystem aging. Front Endocrinol (Lausanne) 2022; 13:1014853. [PMID: 36619570 PMCID: PMC9813443 DOI: 10.3389/fendo.2022.1014853] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Accepted: 12/09/2022] [Indexed: 12/24/2022] Open
Abstract
Osteopontin (OPN) is a multifunctional noncollagenous matrix phosphoprotein that is expressed both intracellularly and extracellularly in various tissues. As a growth regulatory protein and proinflammatory immunochemokine, OPN is involved in the pathological processes of many diseases. Recent studies have found that OPN is widely involved in the aging processes of multiple organs and tissues, such as T-cell senescence, atherosclerosis, skeletal muscle regeneration, osteoporosis, neurodegenerative changes, hematopoietic stem cell reconstruction, and retinal aging. However, the regulatory roles and mechanisms of OPN in the aging process of different tissues are not uniform, and OPN even has diverse roles in different developmental stages of the same tissue, generating uncertainty for the future study and utilization of OPN. In this review, we will summarize the regulatory role and molecular mechanism of OPN in different tissues and cells, such as the musculoskeletal system, central nervous system, cardiovascular system, liver, and eye, during senescence. We believe that a better understanding of the mechanism of OPN in the aging process will help us develop targeted and comprehensive therapeutic strategies to fight the spread of age-related diseases.
Collapse
|
392
|
Dieffenbach PB, Aravamudhan A, Fredenburgh LE, Tschumperlin DJ. The Mechanobiology of Vascular Remodeling in the Aging Lung. Physiology (Bethesda) 2022; 37:28-38. [PMID: 34514871 PMCID: PMC8742727 DOI: 10.1152/physiol.00019.2021] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
Aging is accompanied by declining lung function and increasing susceptibility to lung diseases. The role of endothelial dysfunction and vascular remodeling in these changes is supported by growing evidence, but underlying mechanisms remain elusive. In this review we summarize functional, structural, and molecular changes in the aging pulmonary vasculature and explore how interacting aging and mechanobiological cues may drive progressive vascular remodeling in the lungs.
Collapse
Affiliation(s)
- Paul B. Dieffenbach
- 1Division of Pulmonary and Critical Care Medicine, Department of Medicine, Brigham and Women’s Hospital, Boston, Massachusetts
| | - Aja Aravamudhan
- 2Department of Physiology and Biomedical Engineering, Mayo Clinic College of Medicine and Science, Rochester, Minnesota
| | - Laura E. Fredenburgh
- 1Division of Pulmonary and Critical Care Medicine, Department of Medicine, Brigham and Women’s Hospital, Boston, Massachusetts
| | - Daniel J. Tschumperlin
- 2Department of Physiology and Biomedical Engineering, Mayo Clinic College of Medicine and Science, Rochester, Minnesota
| |
Collapse
|
393
|
Zhang H, Che W, Shi K, Huang Y, Xu C, Fei M, Fan X, Zhang J, Hu X, Hu F, Qin S, Zhang X, Huang Q, Yu F. FT4/FT3 ratio: A novel biomarker predicts coronary microvascular dysfunction (CMD) in euthyroid INOCA patients. Front Endocrinol (Lausanne) 2022; 13:1021326. [PMID: 36187090 PMCID: PMC9520241 DOI: 10.3389/fendo.2022.1021326] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Accepted: 08/29/2022] [Indexed: 11/17/2022] Open
Abstract
BACKGROUND Ischemia and no obstructive coronary artery disease (INOCA) patients who presented coronary microvascular dysfunction (CMD) demonstrate a poor prognosis, yet the risk factors for CMD remain unclear. Subtle changes in thyroid hormone levels within the normal range, especially the free thyroxine (FT4)/free triiodothyronine (FT3) ratio, have been shown to regulate the cardiovascular system. This prospective study investigated the correlation between FT4/FT3 ratio and CMD in euthyroid patients with INOCA. METHODS This prospective study (www.chictr.org.cn/, ChiCTR2000037112) recruited patients with myocardial ischemia symptoms who underwent both coronary angiography (CAG) and myocardial perfusion imaging (MPI) with dynamic single-photon emission computed tomography (D-SPECT). INOCA was defined as coronary stenosis< 50% and CMD was defined as coronary flow reserve (CFR)<2.5. All patients were excluded from abnormal thyroid function and thyroid disease history. RESULTS Among 71 INOCA patients (15 [21.1%] CMD), FT4 and FT4/FT3 ratio in CMD group were significantly higher and both showed significantly moderate correlation with CFR (r=-0.25, p=0.03; r=-0.34, p=0.003, respectively). The ROC curve revealed that FT4/FT3 ratio had the highest efficacy for predicting CMD with an optimized cutoff value>3.39 (AUC 0.78, p<0.001, sensitivity, 80.0%; specificity, 71.4%). Multivariate logistic regression showed that FT4/FT3 ratio was an independent predictor of CMD (OR 7.62, 95% CI 1.12-51.89, p=0.038, P for trend=0.006). CONCLUSION In euthyroid INOCA patients, increased FT4/FT3 ratio levels are associated with the occurrence of CMD, presenting a novel biomarker for improving the risk stratification.
Collapse
Affiliation(s)
- Han Zhang
- Department of Nuclear Medicine, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Shanghai, China
- Institute of Nuclear Medicine, Tongji University School of Medicine, Shanghai, China
| | - Wenliang Che
- Department of Cardiology, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Shanghai, China
| | - Kuangyu Shi
- Department of Nuclear Medicine, University of Bern, Bern, Switzerland
- Department of Informatics, Technical University of Munich, Munich, Germany
| | - Yan Huang
- Department of Nuclear Medicine, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Shanghai, China
- Institute of Nuclear Medicine, Tongji University School of Medicine, Shanghai, China
| | - Chong Xu
- Department of Cardiology, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Shanghai, China
| | - Mengyu Fei
- Department of Radiology, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Shanghai, China
| | - Xin Fan
- Department of Nuclear Medicine, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Shanghai, China
- Institute of Nuclear Medicine, Tongji University School of Medicine, Shanghai, China
| | - Jiajia Zhang
- Department of Nuclear Medicine, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Shanghai, China
- Institute of Nuclear Medicine, Tongji University School of Medicine, Shanghai, China
| | - Xueping Hu
- Department of Nuclear Medicine, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Shanghai, China
- Institute of Nuclear Medicine, Tongji University School of Medicine, Shanghai, China
| | - Fan Hu
- Department of Nuclear Medicine, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Shanghai, China
- Institute of Nuclear Medicine, Tongji University School of Medicine, Shanghai, China
| | - Shanshan Qin
- Department of Nuclear Medicine, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Shanghai, China
- Institute of Nuclear Medicine, Tongji University School of Medicine, Shanghai, China
| | - Xiaoying Zhang
- Department of Nuclear Medicine, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Shanghai, China
- Institute of Nuclear Medicine, Tongji University School of Medicine, Shanghai, China
| | - Qingqing Huang
- Shanghai Key Laboratory of Molecular Imaging, Shanghai University of Medicine and Health Sciences, Shanghai, China
- *Correspondence: Qingqing Huang, ; Fei Yu,
| | - Fei Yu
- Department of Nuclear Medicine, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Shanghai, China
- Shanghai Key Laboratory of Molecular Imaging, Shanghai University of Medicine and Health Sciences, Shanghai, China
- *Correspondence: Qingqing Huang, ; Fei Yu,
| |
Collapse
|
394
|
Franco ACC, Carneiro LDS, Franco RSM, Góes Junior AMDO. Influência do sexo e da idade sobre o diâmetro da cava inferior e implicações para o implante de filtros de veia cava. J Vasc Bras 2022; 21:e20210147. [DOI: 10.1590/1677-5449.202101471] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Accepted: 07/15/2022] [Indexed: 11/06/2022] Open
Abstract
Resumo Contexto A aferição do diâmetro venoso e a escolha de um filtro de veia cava compatível são fundamentais para diminuir o risco de complicações decorrentes do implante desses dispositivos. Entretanto, são escassas as informações sobre como o diâmetro da cava inferior varia de acordo com o sexo e a idade. Objetivos Determinar a influência do sexo e da idade dos pacientes sobre o diâmetro da cava inferior e a adequação dos diferentes modelos de filtro disponíveis. Métodos Estudo analítico retrospectivo, realizado a partir de imagens de tomografia computadorizada. O diâmetro no segmento infrarrenal da veia cava inferior foi aferido em três pontos (cranial, médio e caudal). Os resultados foram classificados de acordo com o sexo e as faixas etárias. Resultados Foram analisadas tomografias de 417 pacientes: 245 mulheres e 172 homens. Os diâmetros nos pontos médio e caudal foram, respectivamente, 19,1 mm e 20,6 mm em mulheres de 81 a 92 anos, sendo estatisticamente menores (p < 0,05) quando comparados aos de mulheres com idade entre 19 e 40 anos (diâmetro no ponto médio: 22,7 mm; diâmetro no ponto caudal: 23 mm). Resultados semelhantes foram observados em homens. Os diâmetros venosos nos pontos cranial e caudal foram estatisticamente maiores em homens (ponto cranial: 24,4 mm; ponto caudal: 22,3 mm) do que em mulheres (ponto cranial: 22,6 mm; ponto caudal: 20,8 mm) em pacientes com idade entre 51 e 70 anos (p < 0,05). Conclusões O diâmetro da veia cava inferior foi menor em pacientes com idade mais avançada em ambos os sexos, e a taxa de variação do diâmetro foi semelhante entre homens e mulheres.
Collapse
|
395
|
Hu Z, Guan Y, Hu W, Xu Z, Ishfaq M. An overview of pharmacological activities of baicalin and its aglycone baicalein: New insights into molecular mechanisms and signaling pathways. IRANIAN JOURNAL OF BASIC MEDICAL SCIENCES 2022; 25:14-26. [PMID: 35656442 PMCID: PMC9118284 DOI: 10.22038/ijbms.2022.60380.13381] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Accepted: 12/27/2021] [Indexed: 11/28/2022]
Abstract
The flavonoids, baicalin, and its aglycone baicalein possess multi-fold therapeutic properties and are mainly found in the roots of Oroxylum indicum (L.) Kurz and Scutellaria baicalensis Georgi. These flavonoids have been reported to possess various pharmacological properties, including antibacterial, antiviral, anticancer, anticonvulsant, anti-oxidant, hepatoprotective, and neuroprotective effects. The pharmacological properties of baicalin and baicalein are due to their abilities to scavenge reactive oxygen species (ROS) and interaction with various signaling molecules associated with apoptosis, inflammation, autophagy, cell cycle, mitochondrial dynamics, and cytoprotection. In this review, we summarized the molecular mechanisms underlying the chemopreventive and chemotherapeutic applications of baicalin and baicalein in the treatment of cancer and inflammatory diseases. In addition, the preventive effects of baicalin and baicalein on mitochondrial dynamics and functions were highlighted with a particular emphasis on their anti-oxidative and cytoprotective properties. The current review highlights could be useful for future prospective studies to further improve the pharmacological applications of baicalein and baicalin. These studies should define the threshold for optimal drug exposure, dose optimization and focus on therapeutic drug monitoring, objective disease markers, and baicalin/baicalein drug levels.
Collapse
Affiliation(s)
- Zhihua Hu
- College of Computer Science, Huanggang Normal University, Huanggang 438000, China.,These authors contributed equally to this work
| | - Yurong Guan
- College of Computer Science, Huanggang Normal University, Huanggang 438000, China.,These authors contributed equally to this work
| | - Wanying Hu
- College of Veterinary Medicine, Northeast Agricultural University, 600 Changjiang Road, Xiangfang District, Harbin 150030, P. R. China
| | - Zhiyong Xu
- Hubei Zhiying Medical Imaging Center, Radiology Department of Huanggang Hospital of Traditional Chinese Medicine, China
| | - Muhammad Ishfaq
- College of Computer Science, Huanggang Normal University, Huanggang 438000, China
| |
Collapse
|
396
|
Seki A, Fishbein MC. Age-related cardiovascular changes and diseases. Cardiovasc Pathol 2022. [DOI: 10.1016/b978-0-12-822224-9.00004-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/17/2022] Open
|
397
|
Sanhueza-Olivares F, Troncoso MF, Pino-de la Fuente F, Martinez-Bilbao J, Riquelme JA, Norambuena-Soto I, Villa M, Lavandero S, Castro PF, Chiong M. A potential role of autophagy-mediated vascular senescence in the pathophysiology of HFpEF. Front Endocrinol (Lausanne) 2022; 13:1057349. [PMID: 36465616 PMCID: PMC9713703 DOI: 10.3389/fendo.2022.1057349] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Accepted: 10/26/2022] [Indexed: 11/18/2022] Open
Abstract
Heart failure with preserved ejection fraction (HFpEF) is one of the most complex and most prevalent cardiometabolic diseases in aging population. Age, obesity, diabetes, and hypertension are the main comorbidities of HFpEF. Microvascular dysfunction and vascular remodeling play a major role in its development. Among the many mechanisms involved in this process, vascular stiffening has been described as one the most prevalent during HFpEF, leading to ventricular-vascular uncoupling and mismatches in aged HFpEF patients. Aged blood vessels display an increased number of senescent endothelial cells (ECs) and vascular smooth muscle cells (VSMCs). This is consistent with the fact that EC and cardiomyocyte cell senescence has been reported during HFpEF. Autophagy plays a major role in VSMCs physiology, regulating phenotypic switch between contractile and synthetic phenotypes. It has also been described that autophagy can regulate arterial stiffening and EC and VSMC senescence. Many studies now support the notion that targeting autophagy would help with the treatment of many cardiovascular and metabolic diseases. In this review, we discuss the mechanisms involved in autophagy-mediated vascular senescence and whether this could be a driver in the development and progression of HFpEF.
Collapse
Affiliation(s)
- Fernanda Sanhueza-Olivares
- Advanced Center for Chronic Diseases (ACCDiS), Faculty of Chemical and Pharmaceutical Sciences, University of Chile, Santiago, Chile
| | - Mayarling F. Troncoso
- Advanced Center for Chronic Diseases (ACCDiS), Faculty of Chemical and Pharmaceutical Sciences, University of Chile, Santiago, Chile
| | - Francisco Pino-de la Fuente
- Advanced Center for Chronic Diseases (ACCDiS), Faculty of Chemical and Pharmaceutical Sciences, University of Chile, Santiago, Chile
| | - Javiera Martinez-Bilbao
- Advanced Center for Chronic Diseases (ACCDiS), Faculty of Chemical and Pharmaceutical Sciences, University of Chile, Santiago, Chile
| | - Jaime A. Riquelme
- Advanced Center for Chronic Diseases (ACCDiS), Faculty of Chemical and Pharmaceutical Sciences, University of Chile, Santiago, Chile
| | - Ignacio Norambuena-Soto
- Advanced Center for Chronic Diseases (ACCDiS), Faculty of Chemical and Pharmaceutical Sciences, University of Chile, Santiago, Chile
| | - Monica Villa
- Advanced Center for Chronic Diseases (ACCDiS), Faculty of Chemical and Pharmaceutical Sciences, University of Chile, Santiago, Chile
| | - Sergio Lavandero
- Advanced Center for Chronic Diseases (ACCDiS), Faculty of Chemical and Pharmaceutical Sciences, University of Chile, Santiago, Chile
- Division of Cardiology, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - Pablo F. Castro
- Advanced Center for Chronic Diseases, Faculty of Medicine, Pontifical University Catholic of Chile, Santiago, Chile
| | - Mario Chiong
- Advanced Center for Chronic Diseases (ACCDiS), Faculty of Chemical and Pharmaceutical Sciences, University of Chile, Santiago, Chile
- *Correspondence: Mario Chiong,
| |
Collapse
|
398
|
Abstract
Vascular senescence plays a vital role in cardiovascular diseases and it is closely related to cellular senescence. At the molecular level, aging begins with a single cell, and it is characterized by telomere shortening, mitochondrial dysfunction, stem cell exhaustion, epigenetic changes, and so on. Epigenetics is an independent discipline that modifies DNA activity without altering the DNA sequence. The application of epigenetics helps to alleviate the occurrence of human diseases, inhibit senescence, and even inhibit tumor occurrence. Epigenetics mainly includes the modification of DNA, histone, and noncoding RNA. Herein, the application of epigenetics in vascular senescence and aging has been reviewed to provide the prospects and innovative inspirations for future research.
Collapse
|
399
|
Acetylsalicylic Acid Reduces Passive Aortic Wall Stiffness and Cardiovascular Remodelling in a Mouse Model of Advanced Atherosclerosis. Int J Mol Sci 2021; 23:ijms23010404. [PMID: 35008828 PMCID: PMC8745264 DOI: 10.3390/ijms23010404] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 12/21/2021] [Accepted: 12/27/2021] [Indexed: 12/31/2022] Open
Abstract
Acetylsalicylic acid (ASA) is widely used in secondary prevention of cardiovascular (CV) disease, mainly because of its antithrombotic effects. Here, we investigated whether ASA can prevent the progression of vessel wall remodelling, atherosclerosis, and CV complications in apolipoprotein E deficient (ApoE-/-) mice, a model of stable atherosclerosis, and in ApoE-/- mice with a mutation in the fibrillin-1 gene (Fbn1C1039G+/-), which is a model of elastic fibre fragmentation, accompanied by exacerbated unstable atherosclerosis. Female ApoE-/- and ApoE-/-Fbn1C1039G+/- mice were fed a Western diet (WD). At 10 weeks of WD, the mice were randomly divided into four groups, receiving either ASA 5 mg/kg/day in the drinking water (ApoE-/- (n = 14), ApoE-/-Fbn1C1039G+/- (n = 19)) or plain drinking water (ApoE-/- (n = 15), ApoE-/-Fbn1C1039G+/- (n = 21)) for 15 weeks. ApoE-/-Fbn1C1039G+/- mice showed an increased neutrophil-lymphocyte ratio (NLR) compared to ApoE-/- mice, and this effect was normalised by ASA. In the proximal ascending aorta wall, ASA-treated ApoE-/-Fbn1C1039G+/- mice showed less p-SMAD2/3 positive nuclei, a lower collagen percentage and an increased elastin/collagen ratio, consistent with the values measured in ApoE-/- mice. ASA did not affect plaque progression, incidence of myocardial infarction and survival of ApoE-/-Fbn1C1039G+/- mice, but systolic blood pressure, cardiac fibrosis and hypertrophy were reduced. In conclusion, ASA normalises the NLR, passive wall stiffness and cardiac remodelling in ApoE-/-Fbn1C1039G+/- mice to levels observed in ApoE-/- mice, indicating additional therapeutic benefits of ASA beyond its classical use.
Collapse
|
400
|
Early Diagnosis of Alzheimer’s Disease Using Cerebral Catheter Angiogram Neuroimaging: A Novel Model Based on Deep Learning Approaches. BIG DATA AND COGNITIVE COMPUTING 2021. [DOI: 10.3390/bdcc6010002] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Neuroimaging refers to the techniques that provide efficient information about the neural structure of the human brain, which is utilized for diagnosis, treatment, and scientific research. The problem of classifying neuroimages is one of the most important steps that are needed by medical staff to diagnose their patients early by investigating the indicators of different neuroimaging types. Early diagnosis of Alzheimer’s disease is of great importance in preventing the deterioration of the patient’s situation. In this research, a novel approach was devised based on a digital subtracted angiogram scan that provides sufficient features of a new biomarker cerebral blood flow. The used dataset was acquired from the database of K.A.U.H hospital and contains digital subtracted angiograms of participants who were diagnosed with Alzheimer’s disease, besides samples of normal controls. Since each scan included multiple frames for the left and right ICA’s, pre-processing steps were applied to make the dataset prepared for the next stages of feature extraction and classification. The multiple frames of scans transformed from real space into DCT space and averaged to remove noises. Then, the averaged image was transformed back to the real space, and both sides filtered with Meijering and concatenated in a single image. The proposed model extracts the features using different pre-trained models: InceptionV3 and DenseNet201. Then, the PCA method was utilized to select the features with 0.99 explained variance ratio, where the combination of selected features from both pre-trained models is fed into machine learning classifiers. Overall, the obtained experimental results are at least as good as other state-of-the-art approaches in the literature and more efficient according to the recent medical standards with a 99.14% level of accuracy, considering the difference in dataset samples and the used cerebral blood flow biomarker.
Collapse
|