1
|
Mavaddatiyan L, Naeini S, Khodabandeh S, Hosseini F, Skelton RP, Azizi V, Talkhabi M. Exploring the association between aging, ferroptosis, and common age-related diseases. Arch Gerontol Geriatr 2025; 135:105877. [PMID: 40339241 DOI: 10.1016/j.archger.2025.105877] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2024] [Revised: 04/14/2025] [Accepted: 04/26/2025] [Indexed: 05/10/2025]
Abstract
Aging is a natural biological process that is characterized by the progressive decline in physiological functions and an increased vulnerability to age-related diseases. The aging process is driven by different cell and molecular mechanisms. It has recently been shown that aging is associated with heightened vulnerability to ferroptosis (an intracellular iron-dependent form of programmed cell death). This susceptibility arises from various factors including oxidative stress, impaired antioxidant defences, and dysregulated iron homeostasis. The progressive decline in cellular antioxidant capacity and the accumulation of damaged components contribute to the increased susceptibility of aging cells to ferroptosis. Dysregulation of key regulators involved in ferroptosis, such as glutathione peroxidase 4 (GPX4), iron regulatory proteins, and lipid metabolism enzymes, further exacerbates this vulnerability. The decline in cellular defence mechanisms against ferroptosis during aging contributes to the accumulation of damaged cells and tissues, ultimately resulting in the manifestation of age-related diseases. Understanding the intricate relevance between aging and ferroptosis holds significant potential for developing strategies to counteract the detrimental effects of aging and age-related diseases. This will subsequently act to mitigate the negative consequences of aging and improving overall health in the elderly population. This review aims to clarify the relationship between aging and ferroptosis, and explores the underlying mechanisms and implications for age-related disorders, including neurodegenerative, cardiovascular, and neoplastic diseases. We also discuss the accumulating evidence suggesting that the imbalance of redox homeostasis and perturbations in iron metabolism contribute to the age-associated vulnerability to ferroptosis.
Collapse
Affiliation(s)
- Laleh Mavaddatiyan
- Department of Animal Sciences and Marine Biology, Faculty of Life Sciences and Biotechnology, Shahid Beheshti University, Tehran, Iran
| | - SaghiHakimi Naeini
- Department of Animal Sciences and Marine Biology, Faculty of Life Sciences and Biotechnology, Shahid Beheshti University, Tehran, Iran
| | - Sara Khodabandeh
- Department of Animal Sciences and Marine Biology, Faculty of Life Sciences and Biotechnology, Shahid Beheshti University, Tehran, Iran
| | - Fatemeh Hosseini
- Department of Animal Sciences and Marine Biology, Faculty of Life Sciences and Biotechnology, Shahid Beheshti University, Tehran, Iran
| | - RhysJ P Skelton
- Flinders Medical Centre, Department of Ophthalmology, Bedford Park, Australia
| | - Vahid Azizi
- Department of Animal Sciences and Marine Biology, Faculty of Life Sciences and Biotechnology, Shahid Beheshti University, Tehran, Iran
| | - Mahmood Talkhabi
- Department of Animal Sciences and Marine Biology, Faculty of Life Sciences and Biotechnology, Shahid Beheshti University, Tehran, Iran.
| |
Collapse
|
2
|
Tsai MC, Fan HY, Hsu HY, Tseng PJ, Chuang SM, Yeh TL, Lee CC, Chien MN, Chien KL. Association of Serum 25-Hydroxyvitamin D With Stroke: Observational Mediation and Mendelian Randomization Study. J Clin Endocrinol Metab 2025; 110:e1420-e1429. [PMID: 39138829 DOI: 10.1210/clinem/dgae561] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Revised: 08/04/2024] [Accepted: 08/13/2024] [Indexed: 08/15/2024]
Abstract
CONTEXT The causal association and biological mechanism linking serum 25-hydroxyvitamin D (25(OH)D) to stroke risk lacks epidemiological evidence. OBJECTIVE This study aimed to investigate the association between 25(OH)D concentration and stroke risk as well as the potential mediating factors. DESIGN The community-based prospective community-based cohort study, the Chin-Shan Community Cardiovascular Cohort, was conducted from 1990 to December 2011, with external validation using a 2-sample Mendelian randomization (MR) study. PATIENTS A total of 1778 participants with serum 25(OH)D data were enrolled. METHODS In the Chin-Shan Community Cardiovascular Cohort observational study, the outcome was ascertained as stroke, while in the 2-sample MR study, it was defined as ischemic stroke. Causal effects were estimated using restricted cubic spline analysis, COX proportional hazard ratios, mediation analysis, and 2-sample MR. RESULTS Over 12 years (21 598 person-years) of follow-up, 163 participants (9.17%) developed stroke. Higher 25(OH)D concentrations were associated with lower stroke risk (hazard ratio: 0.64; 95% confidence interval, 0.43-0.96) after full-model adjustments. Mediation analysis showed a significant association between 25(OH)D concentration and stroke risk mediated by hypertension in unadjusted models (mediation percentage 23.3%, P = .008) that became nonsignificant in full models (mediation percentage, 15.5%; P = .072). Two-sample MR confirmed a significant inverse association between genetically determined 25(OH)D and stroke risk (inverse variance weighted method odds ratio 0.92; 95% confidence interval: 0.85-0.99; P = .036). However, hypertension had an insignificant mediating role in the MR study. CONCLUSION Higher 25(OH)D levels are linked to reduced stroke risk, potentially mediated by hypertension. Prioritizing blood pressure management may improve stroke prevention in 25(OH)D-deficient patients.
Collapse
Affiliation(s)
- Ming-Chieh Tsai
- Institute of Epidemiology and Preventive Medicine, College of Public Health, National Taiwan University, Taipei 10055, Taiwan
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Mackay Memorial Hospital, Taipei 10449, Taiwan
- Department of Medicine, Mackay Medical Collage, New Taipei City 25245, Taiwan
| | - Hsien-Yu Fan
- Institute of Epidemiology and Preventive Medicine, College of Public Health, National Taiwan University, Taipei 10055, Taiwan
- Center for Neuropsychiatric Research, National Health Research Institutes, Miaoli 35053, Taiwan
| | - Hsin-Yin Hsu
- Institute of Epidemiology and Preventive Medicine, College of Public Health, National Taiwan University, Taipei 10055, Taiwan
- Department of Medicine, Mackay Medical Collage, New Taipei City 25245, Taiwan
- Department of Family Medicine, MacKay Memorial Hospital, Taipei City 104217, Taiwan
| | - Po-Jung Tseng
- Division of Cardiovascular Surgery, Department of Surgery, Hsin Chu Armed Force Hospital, Hsinchu 30054, Taiwan
| | - Shih-Ming Chuang
- Institute of Epidemiology and Preventive Medicine, College of Public Health, National Taiwan University, Taipei 10055, Taiwan
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Mackay Memorial Hospital, Taipei 10449, Taiwan
- Department of Medicine, Mackay Medical Collage, New Taipei City 25245, Taiwan
| | - Tzu-Lin Yeh
- Institute of Epidemiology and Preventive Medicine, College of Public Health, National Taiwan University, Taipei 10055, Taiwan
- Department of Medicine, Mackay Medical Collage, New Taipei City 25245, Taiwan
- Department of Family Medicine, Hsinchu MacKay Memorial Hospital, Hsinchu City 30071, Taiwan
| | - Chun-Chuan Lee
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Mackay Memorial Hospital, Taipei 10449, Taiwan
- Department of Medicine, Mackay Medical Collage, New Taipei City 25245, Taiwan
| | - Ming-Nan Chien
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Mackay Memorial Hospital, Taipei 10449, Taiwan
- Department of Medicine, Mackay Medical Collage, New Taipei City 25245, Taiwan
| | - Kuo-Liong Chien
- Institute of Epidemiology and Preventive Medicine, College of Public Health, National Taiwan University, Taipei 10055, Taiwan
- Department of Internal Medicine, National Taiwan University Hospital, Taipei 10022, Taiwan
- Population Health Research Center, National Taiwan University Hospital, Taipei 10055, Taiwan
| |
Collapse
|
3
|
Kobayashi M, Fujii Y, Oda H, Hatori A, Chikazu D. Buccal Abscess With Dystrophic Calcification: A Case Report. Cureus 2025; 17:e81272. [PMID: 40291261 PMCID: PMC12032624 DOI: 10.7759/cureus.81272] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/27/2025] [Indexed: 04/30/2025] Open
Abstract
Dystrophic calcification, a type of ectopic calcification, refers to the pathological deposition of calcium ions in soft tissues. While cases have been reported in various regions of the body, its occurrence in the head and neck is rare. Here, we present a case of dystrophic calcification in the buccal mucosa. The patient, a 54-year-old man, initially visited his local dentist with complaints of swelling and pain in the right buccal mucosa, but the cause remained unidentified. He was subsequently referred to our department for further evaluation. At the initial visit, he exhibited swelling extending from the right buccal region to the mandible, along with redness and swelling in the right buccal mucosa near the premolars. A blood test revealed an elevated WBC count and increased CRP levels. USG showed hypoechoic areas in the right buccal region, raising suspicion of an abscess caused by infection or the presence of foreign material. The patient underwent an incision and drainage procedure, during which three tissue fragments were isolated. Pathological analysis confirmed that these were hard tissue structures distinct from teeth and bone, indicating dystrophic calcifications. Given that calcification can occur in the buccal region, it is essential to differentiate it from other conditions such as calcifying lymph nodes, phleboliths, sialolithiasis, and myositis ossificans. Accurate diagnosis requires a combination of radiological imaging, USG, clinical examination, and thorough medical history assessment.
Collapse
Affiliation(s)
- Mai Kobayashi
- Oral and Maxillofacial Surgery, Tokyo Medical University, Tokyo, JPN
| | - Yasuyuki Fujii
- Oral and Maxillofacial Surgery, Tokyo Medical University, Tokyo, JPN
| | - Hinano Oda
- Oral and Maxillofacial Surgery, Tokyo Medical University, Tokyo, JPN
| | - Ayano Hatori
- Oral and Maxillofacial Surgery, Tokyo Medical University, Tokyo, JPN
| | - Daichi Chikazu
- Oral and Maxillofacial Surgery, Tokyo Medical University, Tokyo, JPN
| |
Collapse
|
4
|
Rodelo-Haad C, Rodríguez-Ortiz ME, Garcia-Sáez R, Rivas-Domínguez A, Jurado-Montoya D, Martín-Malo A, Rodríguez M, Pendón-Ruiz de Mier MV, Muñoz-Castañeda JR. The true cost of phosphate control in chronic kidney disease. Clin Kidney J 2025; 18:i46-i60. [PMID: 40083951 PMCID: PMC11903093 DOI: 10.1093/ckj/sfae434] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2024] [Indexed: 03/16/2025] Open
Abstract
The loss of kidney function entails the development of a positive phosphate balance. The burden of addressing elevated phosphate levels is high. Both parathyroid hormone (PTH) and fibroblast growth factor 23 (FGF23) are increased to promote phosphaturia, thereby preventing the rise in serum phosphate. However, if the phosphate load is excessive, the corresponding phosphaturia is maximal, kidney function deteriorates and hyperphosphataemia becomes clinically evident in advanced stages of chronic kidney disease (CKD). In addition to its role in CKD progression, hyperphosphataemia has been linked to a multitude of adverse outcomes, including overt inflammation, vascular calcifications, endothelial dysfunction, cardiovascular disease, renal osteodystrophy and secondary hyperparathyroidism. Collectively, these factors contribute to the markedly elevated mortality rates observed among individuals with CKD. Furthermore, hyperphosphataemia has been identified as a significant contributor to the development of inflammatory processes, oxidative stress and fibrosis, which underlie the aetiology of numerous comorbidities. Additionally, elevated levels of PTH and FGF23 have been demonstrated to independently induce organ and tissue injury, which is associated with poor outcomes in CKD. This article provides a concise overview of the current understanding of phosphate handling by the kidney in the context of CKD. It outlines the detrimental effects of phosphate on various organs and the mechanisms through which it contributes to CKD progression. Additionally, we discuss the tools available for clinicians to identify patients at risk of an excessive phosphate load.
Collapse
Affiliation(s)
- Cristian Rodelo-Haad
- GC13, Mineral Metabolism, Vascular Calcification, Maimonides Biomedical Research Institute of Cordoba (IMIBIC), Cordoba, Spain
- University of Cordoba, Cordoba, Spain
- Nephrology Service, Reina Sofia University Hospital, Cordoba, Spain
- Redes de Investigación Cooperativa Orientadas a Resultados en Salud, RICORS2040, Institute of Health Carlos III, Madrid, Spain
| | - María E Rodríguez-Ortiz
- GC13, Mineral Metabolism, Vascular Calcification, Maimonides Biomedical Research Institute of Cordoba (IMIBIC), Cordoba, Spain
- University of Cordoba, Cordoba, Spain
- Nephrology Service, Reina Sofia University Hospital, Cordoba, Spain
- Redes de Investigación Cooperativa Orientadas a Resultados en Salud, RICORS2040, Institute of Health Carlos III, Madrid, Spain
| | - Raquel Garcia-Sáez
- GC13, Mineral Metabolism, Vascular Calcification, Maimonides Biomedical Research Institute of Cordoba (IMIBIC), Cordoba, Spain
- University of Cordoba, Cordoba, Spain
- Nephrology Service, Reina Sofia University Hospital, Cordoba, Spain
| | - Antonio Rivas-Domínguez
- GC13, Mineral Metabolism, Vascular Calcification, Maimonides Biomedical Research Institute of Cordoba (IMIBIC), Cordoba, Spain
- University of Cordoba, Cordoba, Spain
- Nephrology Service, Reina Sofia University Hospital, Cordoba, Spain
| | - Daniel Jurado-Montoya
- GC13, Mineral Metabolism, Vascular Calcification, Maimonides Biomedical Research Institute of Cordoba (IMIBIC), Cordoba, Spain
- University of Cordoba, Cordoba, Spain
- Nephrology Service, Reina Sofia University Hospital, Cordoba, Spain
| | - Alejandro Martín-Malo
- GC13, Mineral Metabolism, Vascular Calcification, Maimonides Biomedical Research Institute of Cordoba (IMIBIC), Cordoba, Spain
- University of Cordoba, Cordoba, Spain
- Nephrology Service, Reina Sofia University Hospital, Cordoba, Spain
- Redes de Investigación Cooperativa Orientadas a Resultados en Salud, RICORS2040, Institute of Health Carlos III, Madrid, Spain
| | - Mariano Rodríguez
- GC13, Mineral Metabolism, Vascular Calcification, Maimonides Biomedical Research Institute of Cordoba (IMIBIC), Cordoba, Spain
- University of Cordoba, Cordoba, Spain
- Nephrology Service, Reina Sofia University Hospital, Cordoba, Spain
- Redes de Investigación Cooperativa Orientadas a Resultados en Salud, RICORS2040, Institute of Health Carlos III, Madrid, Spain
- European Uremic Toxins Group (EUTOx)
- COST Action CA21165 – Personalized medicine in chronic kidney disease: improved outcome based on Big Data (PerMediK)
| | - M Victoria Pendón-Ruiz de Mier
- GC13, Mineral Metabolism, Vascular Calcification, Maimonides Biomedical Research Institute of Cordoba (IMIBIC), Cordoba, Spain
- University of Cordoba, Cordoba, Spain
- Nephrology Service, Reina Sofia University Hospital, Cordoba, Spain
- Redes de Investigación Cooperativa Orientadas a Resultados en Salud, RICORS2040, Institute of Health Carlos III, Madrid, Spain
| | - Juan Rafael Muñoz-Castañeda
- GC13, Mineral Metabolism, Vascular Calcification, Maimonides Biomedical Research Institute of Cordoba (IMIBIC), Cordoba, Spain
- University of Cordoba, Cordoba, Spain
- Nephrology Service, Reina Sofia University Hospital, Cordoba, Spain
- Redes de Investigación Cooperativa Orientadas a Resultados en Salud, RICORS2040, Institute of Health Carlos III, Madrid, Spain
| |
Collapse
|
5
|
Moldovan D. The Severity of Carotid Calcifications, but Not Fibroblast Growth Factor 23, Is Associated with Mortality in Hemodialysis: A Single Center Experience. Diseases 2025; 13:73. [PMID: 40136613 PMCID: PMC11941726 DOI: 10.3390/diseases13030073] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2025] [Revised: 02/21/2025] [Accepted: 02/25/2025] [Indexed: 03/27/2025] Open
Abstract
BACKGROUND The study goal was to assess the mortality effect of carotid vascular calcifications (VC), of fibroblast growth factor 23 (FGF-23), mineral markers, and comorbidities in hemodialysis (HD) patients. METHODS The influence of carotid VC severity, FGF-23, laboratory markers, clinical features, and comorbidities on mortality was analyzed in a cohort of 88 HD patients. The follow-up period lasted 8 years. The cut-off value for carotid VC was 4 for all-cause and cardiovascular mortality. RESULTS Carotid VC, diabetes, low serum albumin, high serum C-reactive protein (CRP), and the presence of cardiovascular diseases are associated with all-cause and cardiovascular mortality. Carotid VC score over 4 was an independent predictor of all-cause and cardiovascular mortality, along with diabetes, low albumin, and high CRP. FGF-23 was not found to be predictable for the study outcomes. CONCLUSIONS The study documented in a cohort of patients prevalent in chronic HD that carotid VC predicts all-cause and cardiovascular mortality at 8 years and improves risk stratification, but FGF-23 is not associated with mortality. Other risk factors for all-cause and cardiovascular mortality were diabetes, inflammation, and malnutrition. However, future efforts are needed to assess whether a risk-based approach, including VC screening, improves survival.
Collapse
Affiliation(s)
- Diana Moldovan
- Department of Nephrology, "Iuliu Hatieganu" University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania
| |
Collapse
|
6
|
Wang X, Ren J, Fang F, Wang E, Li J, He W, Zhang Z, Shen Y, Liu X. Matrix vesicles from osteoblasts promote atherosclerotic calcification. Matrix Biol 2024; 134:79-92. [PMID: 39580186 DOI: 10.1016/j.matbio.2024.09.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Revised: 09/20/2024] [Accepted: 09/20/2024] [Indexed: 11/25/2024]
Abstract
Atherosclerotic calcification often coincides with osteoporosis, suggesting a potential interplay between bone and vascular mineralization. Osteoblast-derived matrix vesicles (Ost-MVs), pivotal in bone mineralization, have emerged as potential contributors to ectopic vascular calcification. However, the precise role of Ost-MVs in vascular calcification and the underlying mechanisms remain elusive. In this study, we observed a concomitant increase in atherosclerotic calcification and bone loss, accompanied by elevated release of Ost-MVs into circulation. We demonstrate that circulating Ost-MVs target plaque lesions in the setting of atherosclerosis. Mechanistically, vascular injury facilitates transendothelial transport of Ost-MVs, collagen І remodeling promotes Ost-MVs aggregation, and vascular smooth muscle cell (VSMC) phenotypic switching enhances MV uptake. These pathological changes during atherosclerosis collectively contribute to Ost-MVs recruitment into the vasculature. Furthermore, Ost-MVs and VSMC-derived matrix vesicles (VSMC-MVs) exacerbate calcification via the Ras-Raf-ERK pathway. Our findings unveil a novel Ost-MVs-mediated mechanism participating in vascular calcification and enriching our understanding of bone-vascular crosstalk.
Collapse
Affiliation(s)
- Xiaoli Wang
- Institute of Biomedical Engineering, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, PR China
| | - Jie Ren
- Institute of Biomedical Engineering, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, PR China
| | - Fei Fang
- Institute of Biomedical Engineering, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, PR China
| | - Erxiang Wang
- Institute of Biomedical Engineering, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, PR China
| | - Jianwei Li
- Department of Endocrinology and Metabolism, West China Hospital, Sichuan University, Chengdu, PR China
| | - Weihong He
- Department of Physiology, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, PR China
| | - Zhen Zhang
- Department of Cardiology, The Third People's Hospital of Chengdu, Affiliated Hospital of Southwest Jiaotong University, Chengdu, PR China.
| | - Yang Shen
- Institute of Biomedical Engineering, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, PR China.
| | - Xiaoheng Liu
- Institute of Biomedical Engineering, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, PR China.
| |
Collapse
|
7
|
Marti-Garcia D, Martinez-Martinez A, Sanz FJ, Devesa-Peiro A, Sebastian-Leon P, Del Aguila N, Pellicer A, Diaz-Gimeno P. Age-related uterine changes and its association with poor reproductive outcomes: a systematic review and meta-analysis. Reprod Biol Endocrinol 2024; 22:152. [PMID: 39616336 PMCID: PMC11607893 DOI: 10.1186/s12958-024-01323-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/13/2024] [Accepted: 11/14/2024] [Indexed: 12/18/2024] Open
Abstract
BACKGROUND The decline in women's fertility becomes clinically relevant between 35-40 years old, when there is insufficient ovarian activity, and it becomes more difficult to achieve pregnancy naturally and through artificial reproductive technologies. A competent endometrium is required for establishing and maintaining a pregnancy to term, however, experts in the field underestimate the contribution of endometrial age and its impact on reproductive outcomes remains unclear. STUDY DESIGN A systematic search of full-text articles available in PubMed was conducted to retrieve relevant studies published until March 2023. Search terms included: endometrium, uterus, age, aging, pregnancy, and oocyte donation. Terms related to reproductive pathologies were excluded. Eligibility criteria included original, rigorous, and accessible peer-reviewed work, published in English on the effect of age on the uterus and endometrium. RESULTS From 11,354 records identified, 142 studies were included for systematic review, and 59 were eligible for meta-analysis of endometrial thickness (n = 7), pregnancy rate (n = 22), implantation rate (n = 10), live birth rate (n = 10) and pregnancy loss rate (n = 11). Studies for the meta-analysis of reproductive outcomes only included transfers of embryos from ovum donation (ovum donors < 36 years old). Age shrinks the uterus; depletes endometrial blood supply through narrow uterine veins and a progressive loss of uterine spiral arteries; disrupts endometrial architecture and cellular composition; alters hormone production, shortening menstrual cycle length and impeding endometrial progression to the secretory stage; and dysregulates key endometrial functions such as adhesion, proliferation, apoptosis, and receptivity, among others. Women over 35-40 years old had significantly thinner endometrium (MD 0.52 mm). Advanced maternal age is associated with lower odds of achieving implantation (27%) and clinical pregnancy (20%), or higher odds of experiencing pregnancy loss (44%). CONCLUSION Due to the effect of age on endometrium reported in this review, managing patients with advanced maternal age may require considering the endometrial factor as a potential tissue to treat with anti-aging strategies. This review provides researchers and clinicians with an updated and in-depth summary of this topic, encouraging the development of new tailored anti-aging and preventive strategies for precision medicine in endometrial factor in infertility. TRIAL REGISTRATION PROSPERO 2023 (CRD42023416947).
Collapse
Affiliation(s)
- Diana Marti-Garcia
- IVI-RMA Global Research Alliance, IVI Foundation, Instituto de Investigación Sanitaria La Fe (IIS La Fe), Av. Fernando Abril Martorell 106, Torre A, Planta 1ª, Valencia, 46026, Spain
| | - Asunta Martinez-Martinez
- IVI-RMA Global Research Alliance, IVI Foundation, Instituto de Investigación Sanitaria La Fe (IIS La Fe), Av. Fernando Abril Martorell 106, Torre A, Planta 1ª, Valencia, 46026, Spain
| | - Francisco Jose Sanz
- IVI-RMA Global Research Alliance, IVI Foundation, Instituto de Investigación Sanitaria La Fe (IIS La Fe), Av. Fernando Abril Martorell 106, Torre A, Planta 1ª, Valencia, 46026, Spain
| | - Almudena Devesa-Peiro
- IVI-RMA Global Research Alliance, IVI Foundation, Instituto de Investigación Sanitaria La Fe (IIS La Fe), Av. Fernando Abril Martorell 106, Torre A, Planta 1ª, Valencia, 46026, Spain
| | - Patricia Sebastian-Leon
- IVI-RMA Global Research Alliance, IVI Foundation, Instituto de Investigación Sanitaria La Fe (IIS La Fe), Av. Fernando Abril Martorell 106, Torre A, Planta 1ª, Valencia, 46026, Spain
| | - Nataly Del Aguila
- IVI-RMA Global Research Alliance, IVI Foundation, Instituto de Investigación Sanitaria La Fe (IIS La Fe), Av. Fernando Abril Martorell 106, Torre A, Planta 1ª, Valencia, 46026, Spain
| | | | - Patricia Diaz-Gimeno
- IVI-RMA Global Research Alliance, IVI Foundation, Instituto de Investigación Sanitaria La Fe (IIS La Fe), Av. Fernando Abril Martorell 106, Torre A, Planta 1ª, Valencia, 46026, Spain.
| |
Collapse
|
8
|
Morena-Carrere M, Jaussent I, Chenine L, Dupuy AM, Bargnoux AS, Leray-Moragues H, Klouche K, Vernhet H, Canaud B, Cristol JP. Severe Coronary Artery Calcifications in Chronic Kidney Disease Patients, Coupled with Inflammation and Bone Mineral Disease Derangement, Promote Major Adverse Cardiovascular Events through Vascular Remodeling. Kidney Blood Press Res 2024; 50:33-45. [PMID: 39602894 PMCID: PMC11844697 DOI: 10.1159/000542418] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Accepted: 10/28/2024] [Indexed: 11/29/2024] Open
Abstract
INTRODUCTION Cardiovascular (CV) diseases persist as the foremost cause of morbidity/mortality among chronic kidney disease (CKD) patients. This paper examines the values of coronary artery calcification (CAC) and biomarkers of CV on major adverse CV events (MACE)/CV death in a sample of 425 non-dialysis CKD patients. METHODS At inclusion, patients underwent chest multidetector computed tomography for CAC scoring and biomarkers of CV risk including CRP, mineral metabolism markers, fibroblast growth factor-23 (FGF-23), α-Klotho, osteoprotegerin, tartrate-resistant acid phosphatase 5b (TRAP5b), sclerostin, matrix gla protein (both dephosphorylated uncarboxylated [dp-ucMGP] and total uncarboxylated), and growth differentiation factor-15 (GDF-15) were measured. Patients were followed for a median of 3.61 years (25th-75th percentiles = 1.92-6.70). RESULTS Our results reported that CAC was a major independent factor of MACE/CV mortality showing a hazard ratio of 1.71 95% (confidence interval = 1.00-2.93) after adjustment for age, gender, diabetes, and history of CV events for patients with CAC >300. Interestingly, CAC effect was further enhanced in the presence of low levels of 25(OH) vitamin D3 or α-Klotho and high levels of intact parathyroid hormone (PTH), high-sensitive C reactive protein, FGF-23, osteoprotegerin, sclerostin, dp-ucMGP, or GDF-15. CONCLUSION CAC constitutes a significant CV risk, further exacerbated by inflammation, hyperparathyroidism, and regulation of bone molecules implicated in calcification progression. This finding aligns with the original concept of multiple hits. Consequently, addressing the detrimental environment that fosters plaque vulnerability, reducing chronic low-grade inflammation, and normalizing mineral metabolism markers (such as vitamin D and PTH) and bone-regulating molecules may emerge as a viable therapeutic strategy.
Collapse
Affiliation(s)
- Marion Morena-Carrere
- PhyMedExp, University of Montpellier, INSERM, CNRS, Department of Biochemistry and Hormonology, University Hospital Center of Montpellier, Montpellier, France
| | | | - Leila Chenine
- Department of Nephrology, University Hospital Center of Montpellier, Montpellier, France
| | - Anne-Marie Dupuy
- Department of Biochemistry and Hormonology, University Hospital Center of Montpellier, Montpellier, France
| | - Anne-Sophie Bargnoux
- PhyMedExp, University of Montpellier, INSERM, CNRS, Department of Biochemistry and Hormonology, University Hospital Center of Montpellier, Montpellier, France
| | | | - Kada Klouche
- PhyMedExp, University of Montpellier, INSERM, CNRS, Department of Intensive Care Medicine, University Hospital Center of Montpellier, Montpellier, France
| | - Hélène Vernhet
- Department of Radiology, University Hospital Center of Montpellier, Montpellier, France
| | - Bernard Canaud
- University of Montpellier, Nephrology, Montpellier, France
| | - Jean-Paul Cristol
- PhyMedExp, University of Montpellier, INSERM, CNRS, Department of Biochemistry and Hormonology, University Hospital Center of Montpellier, Montpellier, France
- Charles Mion Foundation, AIDER-Santé, Montpellier, France
| |
Collapse
|
9
|
Köktürk U, Güdül NE, Erbay İ, Doğan PE, Hazinedar E, Kısa F, Koca R, Avcı A. A marker of systemic inflammation in hidradenitis suppurativa patients without cardiovascular disease: aortic arch calcification. Arch Dermatol Res 2024; 316:709. [PMID: 39460803 DOI: 10.1007/s00403-024-03461-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2024] [Revised: 10/01/2024] [Accepted: 10/16/2024] [Indexed: 10/28/2024]
Abstract
BACKGROUND In this study, we aimed to investigate whether there is a relationship between aortic arch calcification (AAC) and hidradenitis suppurativa (HS) in HS patients without cardiovascular disease. METHODS In this study, patients over 18 years of age who applied to the dermatology outpatient clinic between January 2023 and February 2024 were followed up with the diagnosis of HS without cardiovascular disease, and a healthy control group matched in terms of age and gender were included retrospectively. RESULTS In total, 130 patients with HS without cardiovascular disease and 130 control patients were included in the study. AAC was significantly higher in the HS group compared to the control group (p = 0.028). In the multivariate analysis, we found that age and HS were independent predictors of AAC (OR: 1.048 (1.009-1.089); p = 0.015, OR: 3.158 (1.181-8.445); p = 0.022, respectively). When we divided the groups as having AAC (grade 1-3) and not having AAC (grade 0), the rate of HS disease was significantly higher in the group with AAC compared to the group without AAC (75.0% vs. 47.5% p = 0.010). CONCLUSIONS AAC is observed more frequently in patients with HS without cardiovascular disease than in healthy individuals. Moreover, HS can be considered as an independent predictor of AAC. AAC may contribute to developing treatment strategies in HS patients without cardiovascular disease.
Collapse
Affiliation(s)
- Uğur Köktürk
- Department of Cardiology, Faculty of Medicine, Zonguldak Bülent Ecevit University, Esenköy, Kozlu, Zonguldak, 67000, Turkey.
| | - Naile Eriş Güdül
- Department of Cardiology, Faculty of Medicine, Zonguldak Bülent Ecevit University, Esenköy, Kozlu, Zonguldak, 67000, Turkey
| | - İlke Erbay
- Department of Cardiology, Faculty of Medicine, Zonguldak Bülent Ecevit University, Esenköy, Kozlu, Zonguldak, 67000, Turkey
| | - Pelin Ertop Doğan
- Department of Dermatology, Faculty of Medicine, Zonguldak Bülent Ecevit University, Zonguldak, Turkey
| | - Emel Hazinedar
- Department of Dermatology, Faculty of Medicine, Zonguldak Bülent Ecevit University, Zonguldak, Turkey
| | - Furkan Kısa
- Department of Cardiology, Faculty of Medicine, Zonguldak Bülent Ecevit University, Esenköy, Kozlu, Zonguldak, 67000, Turkey
| | - Rafet Koca
- Department of Dermatology, Faculty of Medicine, Zonguldak Bülent Ecevit University, Zonguldak, Turkey
| | - Ahmet Avcı
- Department of Cardiology, Faculty of Medicine, Zonguldak Bülent Ecevit University, Esenköy, Kozlu, Zonguldak, 67000, Turkey
| |
Collapse
|
10
|
Ahmed B, Rahman AA, Lee S, Malhotra R. The Implications of Aging on Vascular Health. Int J Mol Sci 2024; 25:11188. [PMID: 39456971 PMCID: PMC11508873 DOI: 10.3390/ijms252011188] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2024] [Revised: 10/07/2024] [Accepted: 10/11/2024] [Indexed: 10/28/2024] Open
Abstract
Vascular aging encompasses structural and functional changes in the vasculature, significantly contributing to cardiovascular diseases, which are the leading cause of death globally. The incidence and prevalence of these diseases increase with age, with most morbidity and mortality attributed to myocardial infarction and stroke. Diagnosing and intervening in vascular aging while understanding the mechanisms behind age-induced vascular phenotypic and pathophysiological alterations offers the potential for delaying and preventing cardiovascular mortality in an aging population. This review delves into various aspects of vascular aging by examining age-related changes in arterial health at the cellular level, including endothelial dysfunction, cellular senescence, and vascular smooth muscle cell transdifferentiation, as well as at the structural level, including arterial stiffness and changes in wall thickness and diameter. We also explore aging-related changes in perivascular adipose tissue deposition, arterial collateralization, and calcification, providing insights into the physiological and pathological implications. Overall, aging induces phenotypic changes that augment the vascular system's susceptibility to disease, even in the absence of traditional risk factors, such as hypertension, diabetes, obesity, and smoking. Overall, age-related modifications in cellular phenotype and molecular homeostasis increase the vulnerability of the arterial vasculature to structural and functional alterations, thereby accelerating cardiovascular risk. Increasing our understanding of these modifications is crucial for success in delaying or preventing cardiovascular diseases. Non-invasive techniques, such as measuring carotid intima-media thickness, pulse wave velocity, and flow-mediated dilation, as well as detecting vascular calcifications, can be used for the early detection of vascular aging. Targeting specific pathological mechanisms, such as cellular senescence and enhancing angiogenesis, holds promise for innovative therapeutic approaches.
Collapse
Affiliation(s)
- Bulbul Ahmed
- Evans Department of Medicine and Whitaker Cardiovascular Institute, Boston University School of Medicine, Boston, MA 02118, USA;
| | - Ahmed A. Rahman
- Department of Pediatric Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Sujin Lee
- Division of Vascular Surgery, Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA;
| | - Rajeev Malhotra
- Division of Cardiology, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| |
Collapse
|
11
|
Wang Y, Liu X, Zhang J, Zhou B, Yue W, Hu K. Long sleep duration is associated with abdominal aortic calcification among male adults with chronic kidney disease: NHANES 2013-2014. Sci Rep 2024; 14:22076. [PMID: 39333665 PMCID: PMC11436971 DOI: 10.1038/s41598-024-72879-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2024] [Accepted: 09/11/2024] [Indexed: 09/29/2024] Open
Abstract
There are no studies exploring the correlation between sleep duration and abdominal aortic calcification (AAC). This study aims to investigate this relationship and its significance. Additionally, given the higher prevalence of sleep disorders and AAC in patients with chronic kidney disease (CKD), we conducted further studies in this population. We analyzed data from the National Health and Nutrition Examination Survey (NHANES) 2013-2014. Sleep duration was assessed by a sleep questionnaire and categorized into 2-5, 6-8, and ≥ 9 h. The AAC-24 score is determined using the Kauppila scoring system and used for AAC assessment. Multivariable linear and logistic regression analysis were used to explore the relationship between sleep duration and AAC. Among the 2,996 participants, 14.29% reported nightly short sleep (2-5 h), 77.64% reported intermediate sleep (6-8 h), and 8.08% reported long sleep (≥ 9 h). After adjusting for potential confounding factors, among male participants with CKD, long sleep (≥ 9 h) significantly increased AAC-24 scores compared with intermediate sleep (6-8 h) (β: 2.12; 95% CI: 0.75, 3.50), and the risk of severe AAC (SAAC) was increased by 1.55 times (OR: 2.55; 95% CI: 1.02, 6.36). And among female CKD and non-CKD participants, sleep duration was not associated with AAC. Long sleep duration increases the risk of AAC among male adults with CKD. Prospective studies are needed to confirm this finding.
Collapse
Affiliation(s)
- Yuhan Wang
- Department of Respiratory and Critical Care Medicine, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Xu Liu
- Department of Respiratory and Critical Care Medicine, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Jingyi Zhang
- Department of Respiratory and Critical Care Medicine, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Beini Zhou
- Department of Respiratory and Critical Care Medicine, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Wuriliga Yue
- Department of Respiratory and Critical Care Medicine, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Ke Hu
- Department of Respiratory and Critical Care Medicine, Renmin Hospital of Wuhan University, Wuhan, 430060, China.
| |
Collapse
|
12
|
Chen S, Tang R, Liu B. Current Understanding of Cardiovascular Calcification in Patients with Chronic Kidney Disease. Int J Mol Sci 2024; 25:10225. [PMID: 39337709 PMCID: PMC11432307 DOI: 10.3390/ijms251810225] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2024] [Revised: 09/20/2024] [Accepted: 09/21/2024] [Indexed: 09/30/2024] Open
Abstract
The burden of chronic kidney disease (CKD) is increasing, posing a serious threat to human health. Cardiovascular calcification (CVC) is one of the most common manifestations of CKD, which significantly influences the morbidity and mortality of patients. The manifestation of CVC is an unusual accumulation of mineral substances containing calcium and phosphate. The main component is hydroxyapatite. Many cells are involved in this process, such as smooth muscle cells (SMCs) and endothelial cells. CVC is an osteogenic process initiated by complex mechanisms such as metabolic disorders of calcium and phosphorus minerals, inflammation, extracellular vesicles, autophagy, and micro-RNAs with a variety of signaling pathways like Notch, STAT, and JAK. Although drug therapy and dialysis technology continue to advance, the survival time and quality of life of CVC patients still face challenges. Therefore, early diagnosis and prevention of CKD-related CVC, reducing its mortality rate, and improving patients' quality of life have become urgent issues in the field of public health. In this review, we try to summarize the state-of-the-art understanding of the progression of CVC and hope that it will help in the prevention and treatment of CVC in CKD.
Collapse
Affiliation(s)
| | | | - Bicheng Liu
- Institute of Nephrology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing 210009, China; (S.C.); (R.T.)
| |
Collapse
|
13
|
Xu N, Xie Q, Chen Y, Li J, Zhang X, Zheng H, Cheng Y, Wu M, Shen A, Wei L, Yao M, Yang Y, Sferra TJ, Jafri A, Fang Y, Peng J. Gastrodin Alleviates Angiotensin II-Induced Hypertension and Myocardial Apoptosis via Inhibition of the PRDX2/p53 Pathway In Vivo and In Vitro. Pharmaceuticals (Basel) 2024; 17:1200. [PMID: 39338362 PMCID: PMC11434704 DOI: 10.3390/ph17091200] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2024] [Revised: 09/02/2024] [Accepted: 09/05/2024] [Indexed: 09/30/2024] Open
Abstract
Gastrodin, a highly potent compound found in the traditional Chinese medicine Gastrodia elata Blume, exhibits significant antihypertensive properties. However, its role and the mechanism behind its protective effects on hypertensive cardiac conditions are not well understood. This study aims to investigate the cardiac protective effects and underlying mechanisms of gastrodin in angiotensin II (Ang II)-induced hypertensive models, both in vivo and in vitro. Treatment with gastrodin significantly decreased blood pressure and the heart weight/tibial length (HW/TL) ratio and attenuated cardiac dysfunction and pathological damage in Ang II-infused C57BL/6 mice. RNA sequencing analysis (RNA-seq) revealed 697 up-regulated and 714 down-regulated transcripts, along with 1105 signaling pathways, in Ang II-infused C57BL/6 mice following gastrodin treatment, compared to Ang II-induced hypertensive mice. Furthermore, the analyses of the top 30 Gene Ontology (GO) terms and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway indicated significant enrichment in apoptosis and the peroxiredoxin 2 (PRDX2)/p53 pathway. Consistently, gastrodin treatment significantly reduced myocardial apoptosis in both the cardiac tissues of Ang II-induced hypertensive mice and Ang II-stimulated H9c2 cells. Additionally, gastrodin treatment significantly decreased the protein levels of PRDX2, p53, cleaved caspase-3, cleaved caspase-9, and Bax/Bcl-2 ratio in the cardiac tissues of Ang II-infused mice and H9c2 cells stimulated with Ang II. In conclusion, gastrodin treatment can mitigate hypertension-induced myocardial apoptosis in hypertensive mice by inhibiting the PRDX2/p53 pathway.
Collapse
Affiliation(s)
- Nanhui Xu
- Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou 350122, China; (N.X.); (Q.X.); (X.Z.); (H.Z.); (Y.C.); (M.W.); (A.S.); (L.W.); (M.Y.); (Y.Y.)
- Fujian Key Laboratory of Integrative Medicine on Geriatrics, Fujian University of Traditional Chinese Medicine, Fuzhou 350122, China
- Fujian Collaborative Innovation Center for Integrative Medicine in Prevention and Treatment of Major Chronic Cardiovascular Diseases, Fuzhou 350122, China
| | - Qiurong Xie
- Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou 350122, China; (N.X.); (Q.X.); (X.Z.); (H.Z.); (Y.C.); (M.W.); (A.S.); (L.W.); (M.Y.); (Y.Y.)
- Fujian Key Laboratory of Integrative Medicine on Geriatrics, Fujian University of Traditional Chinese Medicine, Fuzhou 350122, China
- Fujian Collaborative Innovation Center for Integrative Medicine in Prevention and Treatment of Major Chronic Cardiovascular Diseases, Fuzhou 350122, China
- Innovation and Transformation Center, Fujian University of Traditional Chinese Medicine, Fuzhou 350122, China
| | - Youqin Chen
- Department of Pediatrics, Rainbow Babies and Children’s Hospital, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA; (Y.C.); (T.J.S.); (A.J.)
| | - Jiapeng Li
- Department of Physical Education, Fujian University of Traditional Chinese Medicine, Fuzhou 350122, China;
| | - Xiuli Zhang
- Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou 350122, China; (N.X.); (Q.X.); (X.Z.); (H.Z.); (Y.C.); (M.W.); (A.S.); (L.W.); (M.Y.); (Y.Y.)
- Fujian Key Laboratory of Integrative Medicine on Geriatrics, Fujian University of Traditional Chinese Medicine, Fuzhou 350122, China
- Fujian Collaborative Innovation Center for Integrative Medicine in Prevention and Treatment of Major Chronic Cardiovascular Diseases, Fuzhou 350122, China
| | - Huifang Zheng
- Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou 350122, China; (N.X.); (Q.X.); (X.Z.); (H.Z.); (Y.C.); (M.W.); (A.S.); (L.W.); (M.Y.); (Y.Y.)
- Fujian Key Laboratory of Integrative Medicine on Geriatrics, Fujian University of Traditional Chinese Medicine, Fuzhou 350122, China
- Fujian Collaborative Innovation Center for Integrative Medicine in Prevention and Treatment of Major Chronic Cardiovascular Diseases, Fuzhou 350122, China
| | - Ying Cheng
- Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou 350122, China; (N.X.); (Q.X.); (X.Z.); (H.Z.); (Y.C.); (M.W.); (A.S.); (L.W.); (M.Y.); (Y.Y.)
- Fujian Key Laboratory of Integrative Medicine on Geriatrics, Fujian University of Traditional Chinese Medicine, Fuzhou 350122, China
- Fujian Collaborative Innovation Center for Integrative Medicine in Prevention and Treatment of Major Chronic Cardiovascular Diseases, Fuzhou 350122, China
| | - Meizhu Wu
- Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou 350122, China; (N.X.); (Q.X.); (X.Z.); (H.Z.); (Y.C.); (M.W.); (A.S.); (L.W.); (M.Y.); (Y.Y.)
- Fujian Key Laboratory of Integrative Medicine on Geriatrics, Fujian University of Traditional Chinese Medicine, Fuzhou 350122, China
- Fujian Collaborative Innovation Center for Integrative Medicine in Prevention and Treatment of Major Chronic Cardiovascular Diseases, Fuzhou 350122, China
| | - Aling Shen
- Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou 350122, China; (N.X.); (Q.X.); (X.Z.); (H.Z.); (Y.C.); (M.W.); (A.S.); (L.W.); (M.Y.); (Y.Y.)
- Fujian Key Laboratory of Integrative Medicine on Geriatrics, Fujian University of Traditional Chinese Medicine, Fuzhou 350122, China
- Fujian Collaborative Innovation Center for Integrative Medicine in Prevention and Treatment of Major Chronic Cardiovascular Diseases, Fuzhou 350122, China
| | - Lihui Wei
- Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou 350122, China; (N.X.); (Q.X.); (X.Z.); (H.Z.); (Y.C.); (M.W.); (A.S.); (L.W.); (M.Y.); (Y.Y.)
- Fujian Key Laboratory of Integrative Medicine on Geriatrics, Fujian University of Traditional Chinese Medicine, Fuzhou 350122, China
- Fujian Collaborative Innovation Center for Integrative Medicine in Prevention and Treatment of Major Chronic Cardiovascular Diseases, Fuzhou 350122, China
- Innovation and Transformation Center, Fujian University of Traditional Chinese Medicine, Fuzhou 350122, China
| | - Mengying Yao
- Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou 350122, China; (N.X.); (Q.X.); (X.Z.); (H.Z.); (Y.C.); (M.W.); (A.S.); (L.W.); (M.Y.); (Y.Y.)
- Fujian Key Laboratory of Integrative Medicine on Geriatrics, Fujian University of Traditional Chinese Medicine, Fuzhou 350122, China
- Fujian Collaborative Innovation Center for Integrative Medicine in Prevention and Treatment of Major Chronic Cardiovascular Diseases, Fuzhou 350122, China
| | - Yanyan Yang
- Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou 350122, China; (N.X.); (Q.X.); (X.Z.); (H.Z.); (Y.C.); (M.W.); (A.S.); (L.W.); (M.Y.); (Y.Y.)
- Fujian Key Laboratory of Integrative Medicine on Geriatrics, Fujian University of Traditional Chinese Medicine, Fuzhou 350122, China
- Fujian Collaborative Innovation Center for Integrative Medicine in Prevention and Treatment of Major Chronic Cardiovascular Diseases, Fuzhou 350122, China
- Innovation and Transformation Center, Fujian University of Traditional Chinese Medicine, Fuzhou 350122, China
| | - Thomas J. Sferra
- Department of Pediatrics, Rainbow Babies and Children’s Hospital, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA; (Y.C.); (T.J.S.); (A.J.)
| | - Anjum Jafri
- Department of Pediatrics, Rainbow Babies and Children’s Hospital, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA; (Y.C.); (T.J.S.); (A.J.)
| | - Yi Fang
- Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou 350122, China; (N.X.); (Q.X.); (X.Z.); (H.Z.); (Y.C.); (M.W.); (A.S.); (L.W.); (M.Y.); (Y.Y.)
- Fujian Key Laboratory of Integrative Medicine on Geriatrics, Fujian University of Traditional Chinese Medicine, Fuzhou 350122, China
- Fujian Collaborative Innovation Center for Integrative Medicine in Prevention and Treatment of Major Chronic Cardiovascular Diseases, Fuzhou 350122, China
- Innovation and Transformation Center, Fujian University of Traditional Chinese Medicine, Fuzhou 350122, China
| | - Jun Peng
- Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou 350122, China; (N.X.); (Q.X.); (X.Z.); (H.Z.); (Y.C.); (M.W.); (A.S.); (L.W.); (M.Y.); (Y.Y.)
- Fujian Key Laboratory of Integrative Medicine on Geriatrics, Fujian University of Traditional Chinese Medicine, Fuzhou 350122, China
- Fujian Collaborative Innovation Center for Integrative Medicine in Prevention and Treatment of Major Chronic Cardiovascular Diseases, Fuzhou 350122, China
- Innovation and Transformation Center, Fujian University of Traditional Chinese Medicine, Fuzhou 350122, China
| |
Collapse
|
14
|
Srinivas B, Alluri K, Rhaleb NE, Belmadani S, Matrougui K. Role of plasmacytoid dendritic cells in vascular dysfunction in mice with renovascular hypertension. Heliyon 2024; 10:e31799. [PMID: 38882290 PMCID: PMC11176769 DOI: 10.1016/j.heliyon.2024.e31799] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2023] [Revised: 05/21/2024] [Accepted: 05/22/2024] [Indexed: 06/18/2024] Open
Abstract
Endothelial dysfunction and inflammation are clinically significant risk factors for cardiovascular diseases in hypertension. Although immune cells play a role in hypertension, the impact of plasmacytoid dendritic cells in established renovascular hypertension-induced cardiovascular complications is not fully understood. We investigated plasmacytoid dendritic cells' contribution to arterial endothelial dysfunction and inflammation in renovascular hypertension. A two-kidney one-clip (2K1C) model for four weeks in both male and female mice was used to induce renovascular hypertension. We treated mice with or without anti-PDCA-1 antibodies for one week to deplete the plasmacytoid dendritic cells. Renovascular hypertension causes cardiac hypertrophy, lung edema, and microvascular endothelial dysfunction associated with inflammation induction in mice. Moreover, renovascular hypertension affects the profile of immune cells, including dendritic cells and macrophages, with variations between male and female mice. Interestingly, the depletion of plasmacytoid dendritic cells significantly reduces blood pressure, cardiac hypertrophy, lung edema, inflammation, and oxidative stress and improves microvascular endothelial function via the endoplasmic reticulum (ER) stress, autophagy, and mTOR-dependent mechanisms. Plasmacytoid dendritic cells significantly contribute to the development of cardiovascular complications in renovascular hypertension by modulating immune cells, inflammation, oxidative stress, and ER stress.
Collapse
Affiliation(s)
- Balaji Srinivas
- Eastern Virginia Medical School, Department of Physiological Sciences, 800 W Olney Rd, Norfolk, VA 23501, USA
| | - Kiran Alluri
- Eastern Virginia Medical School, Department of Physiological Sciences, 800 W Olney Rd, Norfolk, VA 23501, USA
| | - Nour-Eddine Rhaleb
- Department of Internal Medicine, Hypertension and Vascular Research Division, Henry Ford Hospital, Detroit, MI 48202, USA
- Department of Physiology, Wayne State University, Detroit, MI 48201, USA
| | - Souad Belmadani
- Eastern Virginia Medical School, Department of Physiological Sciences, 800 W Olney Rd, Norfolk, VA 23501, USA
| | - Khalid Matrougui
- Eastern Virginia Medical School, Department of Physiological Sciences, 800 W Olney Rd, Norfolk, VA 23501, USA
| |
Collapse
|
15
|
Neofytou IE, Stamou A, Demopoulos A, Roumeliotis S, Zebekakis P, Liakopoulos V, Stamellou E, Dounousi E. Vitamin K for Vascular Calcification in Kidney Patients: Still Alive and Kicking, but Still a Lot to Learn. Nutrients 2024; 16:1798. [PMID: 38931153 PMCID: PMC11206649 DOI: 10.3390/nu16121798] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2024] [Revised: 06/03/2024] [Accepted: 06/05/2024] [Indexed: 06/28/2024] Open
Abstract
Patients with chronic kidney disease (CKD) suffer disproportionately from a high burden of cardiovascular disease, which, despite recent scientific advances, remains partly understood. Vascular calcification (VC) is the result of an ongoing process of misplaced calcium in the inner and medial layers of the arteries, which has emerged as a critical contributor to cardiovascular events in CKD. Beyond its established role in blood clotting and bone health, vitamin K appears crucial in regulating VC via vitamin K-dependent proteins (VKDPs). Among these, the matrix Gla protein (MGP) serves as both a potent inhibitor of VC and a valuable biomarker (in its inactive form) for reflecting circulating vitamin K levels. CKD patients, especially in advanced stages, often present with vitamin K deficiency due to dietary restrictions, medications, and impaired intestinal absorption in the uremic environment. Epidemiological studies confirm a strong association between vitamin K levels, inactive MGP, and increased CVD risk across CKD stages. Based on the promising results of pre-clinical data, an increasing number of clinical trials have investigated the potential benefits of vitamin K supplementation to prevent, delay, or even reverse VC, but the results have remained inconsistent.
Collapse
Affiliation(s)
- Ioannis Eleftherios Neofytou
- 2nd Department of Nephrology, AHEPA Hospital, Medical School, Aristotle University of Thessaloniki, 54636 Thessaloniki, Greece; (I.E.N.); (A.S.); (A.D.); (V.L.)
| | - Aikaterini Stamou
- 2nd Department of Nephrology, AHEPA Hospital, Medical School, Aristotle University of Thessaloniki, 54636 Thessaloniki, Greece; (I.E.N.); (A.S.); (A.D.); (V.L.)
| | - Antonia Demopoulos
- 2nd Department of Nephrology, AHEPA Hospital, Medical School, Aristotle University of Thessaloniki, 54636 Thessaloniki, Greece; (I.E.N.); (A.S.); (A.D.); (V.L.)
| | - Stefanos Roumeliotis
- 2nd Department of Nephrology, AHEPA Hospital, Medical School, Aristotle University of Thessaloniki, 54636 Thessaloniki, Greece; (I.E.N.); (A.S.); (A.D.); (V.L.)
| | - Pantelis Zebekakis
- 1st Department of Internal Medicine, AHEPA Hospital, Medical School, Aristotle University of Thessaloniki, 54636 Thessaloniki, Greece;
| | - Vassilios Liakopoulos
- 2nd Department of Nephrology, AHEPA Hospital, Medical School, Aristotle University of Thessaloniki, 54636 Thessaloniki, Greece; (I.E.N.); (A.S.); (A.D.); (V.L.)
| | - Eleni Stamellou
- Department of Nephrology, Faculty of Medicine, School of Health Sciences, University of Ioannina, 45110 Ioannina, Greece; (E.S.); (E.D.)
- Division of Nephrology and Clinical Immunology, RWTH Aachen University, 52062 Aachen, Germany
| | - Evangelia Dounousi
- Department of Nephrology, Faculty of Medicine, School of Health Sciences, University of Ioannina, 45110 Ioannina, Greece; (E.S.); (E.D.)
| |
Collapse
|
16
|
Chen L, Chen D, Gong H, Wang C, Gao Y, Li Y, Tang W, Zha P, Ran X. Pedal medial arterial calcification in diabetic foot ulcers: A significant risk factor of amputation and mortality. J Diabetes 2024; 16:e13527. [PMID: 38584152 PMCID: PMC10999494 DOI: 10.1111/1753-0407.13527] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/06/2023] [Revised: 11/30/2023] [Accepted: 12/25/2023] [Indexed: 04/09/2024] Open
Abstract
AIMS Pedal medial arterial calcification (MAC) is frequently observed in individuals with diabetic foot ulcers (DFUs). However, the impact of pedal MAC on individuals with DFUs remains uncertain. The main aim of this study was to evaluate the association between pedal MAC with amputation and mortality outcomes. METHODS A prospective, observational cohort study was conducted at West China Hospital from January 2012 to December 2021. Logistic regression analyses, Kaplan-Meier survival method, and Cox proportional hazards models were employed to evaluate the relationship between pedal MAC and amputation as well as mortality. RESULTS A total of 979 patients were enrolled in the study. Peripheral artery disease (PAD) was observed in 53% of patients with DFUs, and pedal MAC was found in 8%. Over a median follow-up of 46 (23-72) months, foot amputation was performed on 190 patients, and mortality occurred in 246 patients. Pedal MAC showed a significant association with amputation both in unadjusted analysis (odds ratio [OR] = 2.98, 95% confidence interval [CI] = 1.86-4.76, p < .001) and after adjusting sex, age, albumin levels, hemoglobin levels, and diabetic retinopathy status (OR 2.29, 95% CI 1.33-3.93, p = .003). The risk of amputation was found to be twofold higher in individuals with PAD and pedal MAC compared to those with PAD alone (OR 2.05, 95% CI 1.10-3.82, p = .024). Furthermore, the presence of pedal MAC was significantly associated with an increased risk of mortality (p = .005), particularly among individuals with DFUs but without PAD (HR 4.26, 95% CI 1.90-9.52, p < .001), rather than in individuals presenting with both DFUs and PAD. CONCLUSION The presence of pedal MAC is significantly associated with both amputation and mortality in individuals with DFUs. Moreover, pedal MAC could provide additional value to predict amputation other than PAD.
Collapse
Affiliation(s)
- Lihong Chen
- Department of Endocrinology & MetabolismWest China Hospital, Sichuan UniversityChengduChina
- Innovation Center for Wound Repair, Diabetic Foot Care CenterWest China Hospital, Sichuan UniversityChengduChina
| | - Dawei Chen
- Department of Endocrinology & MetabolismWest China Hospital, Sichuan UniversityChengduChina
| | - Hongping Gong
- International Medical Center Ward, Department of General PracticeWest China Hospital, Sichuan UniversityChengduChina
| | - Chun Wang
- Department of Endocrinology & MetabolismWest China Hospital, Sichuan UniversityChengduChina
| | - Yun Gao
- Department of Endocrinology & MetabolismWest China Hospital, Sichuan UniversityChengduChina
| | - Yan Li
- Department of Clinical Research ManagementWest China Hospital, Sichuan UniversityChengduChina
| | - Weiwei Tang
- Department of Endocrinology & MetabolismWest China Hospital, Sichuan UniversityChengduChina
- Innovation Center for Wound Repair, Diabetic Foot Care CenterWest China Hospital, Sichuan UniversityChengduChina
| | - Panpan Zha
- Department of Endocrinology & MetabolismWest China Hospital, Sichuan UniversityChengduChina
- Innovation Center for Wound Repair, Diabetic Foot Care CenterWest China Hospital, Sichuan UniversityChengduChina
| | - Xingwu Ran
- Department of Endocrinology & MetabolismWest China Hospital, Sichuan UniversityChengduChina
- Innovation Center for Wound Repair, Diabetic Foot Care CenterWest China Hospital, Sichuan UniversityChengduChina
| |
Collapse
|
17
|
Lalayiannis AD, Soeiro EMD, Moysés RMA, Shroff R. Chronic kidney disease mineral bone disorder in childhood and young adulthood: a 'growing' understanding. Pediatr Nephrol 2024; 39:723-739. [PMID: 37624528 PMCID: PMC10817832 DOI: 10.1007/s00467-023-06109-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/19/2023] [Revised: 07/06/2023] [Accepted: 07/19/2023] [Indexed: 08/26/2023]
Abstract
Chronic kidney disease (CKD) mineral and bone disorder (MBD) comprises a triad of biochemical abnormalities (of calcium, phosphate, parathyroid hormone and vitamin D), bone abnormalities (turnover, mineralization and growth) and extra-skeletal calcification. Mineral dysregulation leads to bone demineralization causing bone pain and an increased fracture risk compared to healthy peers. Vascular calcification, with hydroxyapatite deposition in the vessel wall, is a part of the CKD-MBD spectrum and, in turn, leads to vascular stiffness, left ventricular hypertrophy and a very high cardiovascular mortality risk. While the growing bone requires calcium, excess calcium can deposit in the vessels, such that the intake of calcium, calcium- containing medications and high calcium dialysate need to be carefully regulated. Normal physiological bone mineralization continues into the third decade of life, many years beyond the rapid growth in childhood and adolescence, implying that skeletal calcium requirements are much higher in younger people compared to the elderly. Much of the research into the link between bone (de)mineralization and vascular calcification in CKD has been performed in older adults and these data must not be extrapolated to children or younger adults. In this article, we explore the physiological changes in bone turnover and mineralization in children and young adults, the pathophysiology of mineral bone disease in CKD and a potential link between bone demineralization and vascular calcification.
Collapse
Affiliation(s)
- Alexander D Lalayiannis
- Birmingham Women's and Children's NHS Foundation Trust, Birmingham, UK.
- University College London Great Ormond Street Hospital Institute of Child Health, London, UK.
| | | | - Rosa M A Moysés
- Sao Paulo University Faculty of Medicine, Universidade de Sao Paulo Faculdade de Medicina, São Paulo, Brazil
| | - Rukshana Shroff
- University College London Great Ormond Street Hospital Institute of Child Health, London, UK
| |
Collapse
|
18
|
Li X, Lindholm B. The role of irisin in kidney diseases. Clin Chim Acta 2024; 554:117756. [PMID: 38218331 DOI: 10.1016/j.cca.2023.117756] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Revised: 12/31/2023] [Accepted: 12/31/2023] [Indexed: 01/15/2024]
Abstract
Irisin is a hormone that is produced mainly by skeletal muscles in response to exercise. It has been found to have a close correlation with obesity and diabetes mellitus for its energy expenditure and metabolic properties. Recent research has revealed that irisin also possesses anti-inflammatory, anti-oxidative and anti-apoptotic properties, which make it associated with major chronic diseases, such as chronic kidney disease (CKD), liver diseases, osteoporosis, atherosclerosis and Alzheimer s disease. The identification of irisin has not only opened up new possibilities for monitoring metabolic and non-metabolic diseases but also presents a promising therapeutic target due to its multiple biological functions. Studies have shown that circulating irisin levels are lower in CKD patients than in non-CKD patients and decrease with increasing CKD stage. Furthermore, irisin also plays a role in many CKD-related complications like protein energy wasting (PEW), cardiovascular disease (CVD) and chronic kidney disease-mineral and bone disorder (CKD-MBD). In this review, we present the current knowledge on the role of irisin in kidney diseases and their complications.
Collapse
Affiliation(s)
- Xiejia Li
- Department of Nephrology, The 2nd Xiangya Hospital, Central South University, Changsha, Hunan, China; Renal Medicine and Baxter Novum, Department of Clinical Science, Intervention and Technology, Karolinska Institutet, Stockholm, Sweden.
| | - Bengt Lindholm
- Renal Medicine and Baxter Novum, Department of Clinical Science, Intervention and Technology, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
19
|
He L, Li Y, Jin J, Cheng M, Bai Y, Xu J. Comparative efficacy of sodium thiosulfate, bisphosphonates, and cinacalcet for the treatment of vascular calcification in patients with haemodialysis: a systematic review and network meta-analysis. BMC Nephrol 2024; 25:26. [PMID: 38254024 PMCID: PMC10804723 DOI: 10.1186/s12882-024-03460-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Accepted: 01/06/2024] [Indexed: 01/24/2024] Open
Abstract
BACKGROUND Up to now, there is no unequivocal intervention to mitigate vascular calcification (VC) in patients with hemodialysis. This network meta-analysis aimed to systematically evaluate the clinical efficacy of sodium thiosulfate, bisphosphonates, and cinacalcet in treating vascular calcification. METHODS A comprehensive study search was performed using PubMed, Web of Science, the Cochrane Library, EMBASE and China National Knowledge Internet (CNKI) to collect randomized controlled trials (RCTs) of sodium thiosulfate, bisphosphonates, and cinacalcet for vascular calcification among hemodialysis patients. Then, network meta-analysis was conducted using Stata 17.0 software. RESULTS In total, eleven RCTs including 1083 patients were qualified for this meta-analysis. We found that cinacalcet (SMD - 0.59; 95% CI [-0.95, -0.24]) had significant benefit on vascular calcification compared with conventional therapy, while sodium thiosulfate or bisphosphonates did not show such efficiency. Furthermore, as for ranking the efficacy assessment, cinacalcet possessed the highest surface under the cumulative ranking curve (SUCRA) value (88.5%) of lessening vascular calcification and was superior to sodium thiosulfate (50.4%) and bisphosphonates (55.4%). Thus, above results suggested that cinacalcet might be the most promising drug for vascular calcification treatment in hemodialysis patients. Mechanistically, our findings illustrated that cinacalcet reduced serum calcium (SMD - 1.20; 95% CI [-2.08, - 0.33]) and showed the tendency in maintaining the balance of intact Parathyroid Hormone (iPTH) level. CONCLUSIONS This network meta-analysis indicated that cinacalcet appear to be more effective than sodium thiosulfate and bisphosphonates in mitigating vascular calcification through decreasing serum calcium and iPTH. And cinacalcet might be a reasonable option for hemodialysis patients with VC in clinical practice. SYSTEMATIC REVIEW REGISTRATION [ http://www.crd.york.ac.uk/PROSPERO ], identifier [CRD42022379965].
Collapse
Affiliation(s)
- Lei He
- Departments of Nephrology, Hebei Key Laboratory of vascular calcification in kidney disease; Hebei Clinical Research Center for Chronic Kidney Disease, The Fourth Hospital of Hebei Medical University, 12 Jiankang Road, 050011, Shijiazhuang, China
| | - Yuzhe Li
- Departments of Nephrology, Hebei Key Laboratory of vascular calcification in kidney disease; Hebei Clinical Research Center for Chronic Kidney Disease, The Fourth Hospital of Hebei Medical University, 12 Jiankang Road, 050011, Shijiazhuang, China
| | - Jingjing Jin
- Departments of Nephrology, Hebei Key Laboratory of vascular calcification in kidney disease; Hebei Clinical Research Center for Chronic Kidney Disease, The Fourth Hospital of Hebei Medical University, 12 Jiankang Road, 050011, Shijiazhuang, China
| | - Meijuan Cheng
- Departments of Nephrology, Hebei Key Laboratory of vascular calcification in kidney disease; Hebei Clinical Research Center for Chronic Kidney Disease, The Fourth Hospital of Hebei Medical University, 12 Jiankang Road, 050011, Shijiazhuang, China
| | - Yaling Bai
- Departments of Nephrology, Hebei Key Laboratory of vascular calcification in kidney disease; Hebei Clinical Research Center for Chronic Kidney Disease, The Fourth Hospital of Hebei Medical University, 12 Jiankang Road, 050011, Shijiazhuang, China
| | - Jinsheng Xu
- Departments of Nephrology, Hebei Key Laboratory of vascular calcification in kidney disease; Hebei Clinical Research Center for Chronic Kidney Disease, The Fourth Hospital of Hebei Medical University, 12 Jiankang Road, 050011, Shijiazhuang, China.
| |
Collapse
|
20
|
Kang JH, Kawano T, Murata M, Toita R. Vascular calcification and cellular signaling pathways as potential therapeutic targets. Life Sci 2024; 336:122309. [PMID: 38042282 DOI: 10.1016/j.lfs.2023.122309] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2023] [Revised: 11/21/2023] [Accepted: 11/24/2023] [Indexed: 12/04/2023]
Abstract
Increased vascular calcification (VC) is observed in patients with cardiovascular diseases such as atherosclerosis, diabetes, and chronic kidney disease. VC is divided into three types according to its location: intimal, medial, and valvular. Various cellular signaling pathways are associated with VC, including the Wnt, mitogen-activated protein kinase, phosphatidylinositol-3 kinase/Akt, cyclic nucleotide-dependent protein kinase, protein kinase C, calcium/calmodulin-dependent kinase II, adenosine monophosphate-activated protein kinase/mammalian target of rapamycin, Ras homologous GTPase, apoptosis, Notch, and cytokine signaling pathways. In this review, we discuss the literature concerning the key cellular signaling pathways associated with VC and their role as potential therapeutic targets. Inhibitors to these pathways represent good candidates for use as potential therapeutic agents for the prevention and treatment of VC.
Collapse
Affiliation(s)
- Jeong-Hun Kang
- National Cerebral and Cardiovascular Center Research Institute, 6-1 Shinmachi, Kishibe, Suita, Osaka 564-8565, Japan.
| | - Takahito Kawano
- Center for Advanced Medical Innovation, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan
| | - Masaharu Murata
- Center for Advanced Medical Innovation, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan
| | - Riki Toita
- Biomedical Research Institute, National Institute of Advanced Industrial Science and Technology (AIST), 1-8-31 Midorigaoka, Ikeda, Osaka, 563-8577, Japan; AIST-Osaka University Advanced Photonics and Biosensing Open Innovation Laboratory, AIST, 2-1 Yamadaoka, Suita, Osaka 565-0871, Japan
| |
Collapse
|
21
|
Wei X, Shen Z, Zhu M, Fang M, Wang S, Zhang T, Zhang B, Yang X, Lv Z, Duan Y, Jiang M, Ma C, Li Q, Chen Y. The pterostilbene-dihydropyrazole derivative Ptd-1 ameliorates vascular calcification by regulating inflammation. Int Immunopharmacol 2023; 125:111198. [PMID: 37952482 DOI: 10.1016/j.intimp.2023.111198] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Revised: 10/21/2023] [Accepted: 11/05/2023] [Indexed: 11/14/2023]
Abstract
Vascular calcification is an independent risk factor for cardiovascular disease. However, there is still a lack of adequate treatment. This study aimed to examine the potential of (E)-1-(5-(2-(4-fluorobenzyloxy)Styryl)-4,6-dimethoxyphenyl)-3-methyl-4,5-dihydro-1H-pyrazole-1-yl) ethyl ketone (Ptd-1) to alleviate vascular calcification. ApoE-deficient mice were fed a high-fat diet for 12/16 weeks to induce intimal calcification, and wild-type mice were induced with a combination of nicotine and vitamin D3 to induce medial calcification. Human aortic smooth muscle cells (HASMCs) and aortic osteogenic differentiation were induced in vitro with phosphate. In the mouse model of atherosclerosis, Ptd-1 significantly ameliorated the progression of atherosclerosis and intimal calcification, and there were significant reductions in lipid deposition and calcium salt deposition in the aorta and aortic root. In addition, Ptd-1 significantly improved medial calcification in vivo and osteogenic differentiation in vitro. Mechanistically, Ptd-1 reduced the levels of the inflammatory factors IL-1β, TNFα and IL-6 in vivo and in vitro. Furthermore, we demonstrated that Ptd-1 could attenuate the expression of p-ERK1/2 and β-catenin, and that the levels of inflammatory factors were elevated in the presence of ERK1/2 and β-catenin agonists. Interestingly, we determined that activation of the ERK1/2 pathway promoted β-catenin expression, which further regulated the IL-6/STAT3 signaling pathway. Ptd-1 blocked ERK1/2 signaling, leading to decreased expression of inflammatory factors, which in turn improved vascular calcification. Taken together, our study reveals that Ptd-1 ameliorates vascular calcification by regulating the production of inflammatory factors, providing new ideas for the treatment of vascular calcification.
Collapse
Affiliation(s)
- Xiaoning Wei
- Key Laboratory of Metabolism and Regulation for Major Diseases of Anhui Higher Education Institutes, School of Food and Biological Engineering, Hefei University of Technology, Hefei, China
| | - Zhenbao Shen
- Key Laboratory of Metabolism and Regulation for Major Diseases of Anhui Higher Education Institutes, School of Food and Biological Engineering, Hefei University of Technology, Hefei, China
| | - Mengmeng Zhu
- Key Laboratory of Metabolism and Regulation for Major Diseases of Anhui Higher Education Institutes, School of Food and Biological Engineering, Hefei University of Technology, Hefei, China
| | - Mengyuan Fang
- Key Laboratory of Metabolism and Regulation for Major Diseases of Anhui Higher Education Institutes, School of Food and Biological Engineering, Hefei University of Technology, Hefei, China
| | - Shengnan Wang
- Key Laboratory of Metabolism and Regulation for Major Diseases of Anhui Higher Education Institutes, School of Food and Biological Engineering, Hefei University of Technology, Hefei, China
| | - Tingting Zhang
- Key Laboratory of Metabolism and Regulation for Major Diseases of Anhui Higher Education Institutes, School of Food and Biological Engineering, Hefei University of Technology, Hefei, China
| | - Baotong Zhang
- Department of Human Cell Biology and Genetics, School of Medicine, Southern University of Science and Technology, Shenzhen 518055, China
| | - Xiaoxiao Yang
- Key Laboratory of Metabolism and Regulation for Major Diseases of Anhui Higher Education Institutes, School of Food and Biological Engineering, Hefei University of Technology, Hefei, China
| | - Zhilin Lv
- Key Laboratory of Metabolism and Regulation for Major Diseases of Anhui Higher Education Institutes, School of Food and Biological Engineering, Hefei University of Technology, Hefei, China
| | - Yajun Duan
- Department of Cardiology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Meixiu Jiang
- The Institute of Translational Medicine, the National Engineering Research Center for Bioengineering Drugs and the Technologies, Nanchang University, Nanchang, China
| | - Chuanrui Ma
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China.
| | - Qingshan Li
- Key Laboratory of Metabolism and Regulation for Major Diseases of Anhui Higher Education Institutes, School of Food and Biological Engineering, Hefei University of Technology, Hefei, China.
| | - Yuanli Chen
- Key Laboratory of Metabolism and Regulation for Major Diseases of Anhui Higher Education Institutes, School of Food and Biological Engineering, Hefei University of Technology, Hefei, China.
| |
Collapse
|
22
|
Wang C, Ma Q, Yu X. Bile Acid Network and Vascular Calcification-Associated Diseases: Unraveling the Intricate Connections and Therapeutic Potential. Clin Interv Aging 2023; 18:1749-1767. [PMID: 37885621 PMCID: PMC10599251 DOI: 10.2147/cia.s431220] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Accepted: 10/10/2023] [Indexed: 10/28/2023] Open
Abstract
Bile acids play a crucial role in promoting intestinal nutrient absorption and biliary cholesterol excretion, thereby protecting the liver from cholesterol accumulation and bile acid toxicity. Additionally, bile acids can bind to specific nuclear and membrane receptors to regulate energy expenditure and specific functions of particular tissues. Vascular calcification refers to the pathological process of calcium-phosphate deposition in blood vessel walls, which serves as an independent predictor for cardiovascular adverse events. In addition to aging, this pathological change is associated with aging-related diseases such as atherosclerosis, hypertension, chronic kidney disease, diabetes mellitus, and osteoporosis. Emerging evidence suggests a close association between the bile acid network and these aforementioned vascular calcification-associated conditions. Several bile acids have been proven to participate in calcium-phosphate metabolism, affecting the transdifferentiation of vascular smooth muscle cells and thus influencing vascular calcification. Targeting the bile acid network shows potential for ameliorating these diseases and their concomitant vascular calcification by regulating pathways such as energy metabolism, inflammatory response, oxidative stress, and cell differentiation. Here, we present a summary of the metabolism and functions of the bile acid network and aim to provide insights into the current research on the profound connections between the bile acid network and these vascular calcification-associated diseases, as well as the therapeutic potential.
Collapse
Affiliation(s)
- Cui Wang
- Laboratory of Endocrinology & Metabolism/Department of Endocrinology & Metabolism, Rare Disease Center, West China Hospital, Sichuan University, Chengdu, Sichuan Province, 610041, People’s Republic of China
| | - Qing Ma
- General Practice Ward/International Medical Center Ward, General Practice Medical Center, West China Hospital, Sichuan University/West China School of Nursing, Sichuan University, Chengdu, Sichuan Province, 610041, People’s Republic of China
| | - Xijie Yu
- Laboratory of Endocrinology & Metabolism/Department of Endocrinology & Metabolism, Rare Disease Center, West China Hospital, Sichuan University, Chengdu, Sichuan Province, 610041, People’s Republic of China
| |
Collapse
|
23
|
Yang S, Zeng Z, Yuan Q, Chen Q, Wang Z, Xie H, Liu J. Vascular calcification: from the perspective of crosstalk. MOLECULAR BIOMEDICINE 2023; 4:35. [PMID: 37851172 PMCID: PMC10584806 DOI: 10.1186/s43556-023-00146-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Accepted: 09/20/2023] [Indexed: 10/19/2023] Open
Abstract
Vascular calcification (VC) is highly correlated with cardiovascular disease morbidity and mortality, but anti-VC treatment remains an area to be tackled due to the ill-defined molecular mechanisms. Regardless of the type of VC, it does not depend on a single cell but involves multi-cells/organs to form a complex cellular communication network through the vascular microenvironment to participate in the occurrence and development of VC. Therefore, focusing only on the direct effect of pathological factors on vascular smooth muscle cells (VSMCs) tends to overlook the combined effect of other cells and VSMCs, including VSMCs-VSMCs, ECs-VMSCs, Macrophages-VSMCs, etc. Extracellular vesicles (EVs) are a collective term for tiny vesicles with a membrane structure that are actively secreted by cells, and almost all cells secrete EVs. EVs docked on the surface of receptor cells can directly mediate signal transduction or transfer their contents into the cell to elicit a functional response from the receptor cells. They have been proven to participate in the VC process and have also shown attractive therapeutic prospects. Based on the advantages of EVs and the ability to be detected in body fluids, they may become a novel therapeutic agent, drug delivery vehicle, diagnostic and prognostic biomarker, and potential therapeutic target in the future. This review focuses on the new insight into VC molecular mechanisms from the perspective of crosstalk, summarizes how multi-cells/organs interactions communicate via EVs to regulate VC and the emerging potential of EVs as therapeutic methods in VC. We also summarize preclinical experiments on crosstalk-based and the current state of clinical studies on VC-related measures.
Collapse
Affiliation(s)
- Shiqi Yang
- Department of Metabolism and Endocrinology, Hengyang Medical School, The First Affiliated Hospital, University of South China, Hengyang, 421001, Hunan, China
- Department of Clinical Laboratory Medicine, Hengyang Medical School, The First Affiliated Hospital, University of South China, Hengyang, 421001, Hunan, China
| | - Zhaolin Zeng
- Department of Metabolism and Endocrinology, Hengyang Medical School, The First Affiliated Hospital, University of South China, Hengyang, 421001, Hunan, China
| | - Qing Yuan
- Department of Metabolism and Endocrinology, Hengyang Medical School, The First Affiliated Hospital, University of South China, Hengyang, 421001, Hunan, China
- Department of Clinical Laboratory Medicine, Hengyang Medical School, The First Affiliated Hospital, University of South China, Hengyang, 421001, Hunan, China
| | - Qian Chen
- Department of Metabolism and Endocrinology, Hengyang Medical School, The First Affiliated Hospital, University of South China, Hengyang, 421001, Hunan, China
| | - Zuo Wang
- Institute of Cardiovascular Disease, Key Lab for Arteriosclerology of Hunan Province, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China
| | - Hui Xie
- Department of Orthopaedics, Movement System Injury and Repair Research Centre, Xiangya Hospital, Central South University, Changsha, Hunan Province, China.
| | - Jianghua Liu
- Department of Metabolism and Endocrinology, Hengyang Medical School, The First Affiliated Hospital, University of South China, Hengyang, 421001, Hunan, China.
| |
Collapse
|
24
|
Huang C, Duan Z, Xu C, Chen Y. Influence of sodium thiosulfate on coronary artery calcification of patients on dialysis: a meta-analysis. Ren Fail 2023; 45:2254569. [PMID: 37755153 PMCID: PMC10538455 DOI: 10.1080/0886022x.2023.2254569] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2023] [Accepted: 08/29/2023] [Indexed: 09/28/2023] Open
Abstract
Coronary artery calcification (CAC) is common in dialysis patients and is associated with a higher risk of future cardiovascular events. Sodium thiosulfate (STS) is effective for calciphylaxis in dialysis patients; however, the influence of STS on CAC in dialysis patients remains unclear. This systematic review and meta-analysis were conducted to evaluate the effects of STS on CAC in patients undergoing dialysis. PubMed, Embase, Cochrane Library, CNKI, and Wanfang databases were searched from inception to 22 March 2023 for controlled studies comparing the influence of STS versus usual care without STS on CAC scores in dialysis patients. A random effects model incorporating the potential influence of heterogeneity was used to pool the results. Nine studies, including two non-randomized studies and seven randomized controlled trials, were included in the meta-analysis. Among these, 365 patients on dialysis were included in the study. Compared with usual care without STS, intravenous STS for 3-6 months was associated with significantly reduced CAC scores (mean difference [MD] = -180.17, 95% confidence interval [CI]: -276.64 to -83.70, p < 0.001, I2 = 0%). Sensitivity analysis limited to studies of patients on hemodialysis showed similar results (MD: -167.33, 95% CI: -266.57 to -68.09, p = 0.001; I2 = 0%). Subgroup analyses according to study design, sample size, mean age, sex, dialysis vintage of the patients, and treatment duration of STS also showed consistent results (p for subgroup differences all > 0.05). In conclusion, intravenous STS may be effective in attenuating CAC in dialysis patients.
Collapse
Affiliation(s)
- Chong Huang
- Department of Nephrology, the Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Zhibing Duan
- Department of Nephrology, the Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Chengyun Xu
- Department of Nephrology, the Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Yan Chen
- Department of Nephrology, the Second Affiliated Hospital of Nanchang University, Nanchang, China
| |
Collapse
|
25
|
Cozzolino M, Maffei Faccioli F, Cara A, Boni Brivio G, Rivela F, Ciceri P, Magagnoli L, Galassi A, Barbuto S, Speciale S, Minicucci C, Cianciolo G. Future treatment of vascular calcification in chronic kidney disease. Expert Opin Pharmacother 2023; 24:2041-2057. [PMID: 37776230 DOI: 10.1080/14656566.2023.2266381] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Accepted: 09/29/2023] [Indexed: 10/02/2023]
Abstract
INTRODUCTION Cardiovascular disease (CVD) is one of the global leading causes of morbidity and mortality in chronic kidney disease (CKD) patients. Vascular calcification (VC) is a major cause of CVD in this population and is the consequence of complex interactions between inhibitor and promoter factors leading to pathological deposition of calcium and phosphate in soft tissues. Different pathological landscapes are associated with the development of VC, such as endothelial dysfunction, oxidative stress, chronic inflammation, loss of mineralization inhibitors, release of calcifying extracellular vesicles (cEVs) and circulating calcifying cells. AREAS COVERED In this review, we examined the literature and summarized the pathophysiology, biomarkers and focused on the treatments of VC. EXPERT OPINION Even though there is no consensus regarding specific treatment options, we provide the currently available treatment strategies that focus on phosphate balance, correction of vitamin D and vitamin K deficiencies, avoidance of both extremes of bone turnover, normalizing calcium levels and reduction of inflammatory response and the potential and promising therapeutic approaches liketargeting cellular mechanisms of calcification (e.g. SNF472, TNAP inhibitors).Creating novel scores to detect in advance VC and implementing targeted therapies is crucial to treat them and improve the future management of these patients.
Collapse
Affiliation(s)
- Mario Cozzolino
- Renal Division, Department of Health Sciences, ASST Santi Paolo e Carlo, University of Milan, Milan, Italy
| | - Federico Maffei Faccioli
- Renal Division, Department of Health Sciences, ASST Santi Paolo e Carlo, University of Milan, Milan, Italy
| | - Anila Cara
- Renal Division, Department of Health Sciences, ASST Santi Paolo e Carlo, University of Milan, Milan, Italy
| | - Giulia Boni Brivio
- Renal Division, Department of Health Sciences, ASST Santi Paolo e Carlo, University of Milan, Milan, Italy
| | - Francesca Rivela
- Renal Division, Department of Health Sciences, ASST Santi Paolo e Carlo, University of Milan, Milan, Italy
| | - Paola Ciceri
- Renal Division, Department of Health Sciences, ASST Santi Paolo e Carlo, University of Milan, Milan, Italy
| | - Lorenza Magagnoli
- Renal Division, Department of Health Sciences, ASST Santi Paolo e Carlo, University of Milan, Milan, Italy
| | - Andrea Galassi
- Renal Division, Department of Health Sciences, ASST Santi Paolo e Carlo, University of Milan, Milan, Italy
| | - Simona Barbuto
- Nephrology, Dialysis and Renal Transplant Unit, IRCCS-Azienda Ospedaliero-Universitaria di Bologna, Alma Mater Studiorum University of Bologna, Bologna, Italy
| | - Serena Speciale
- Nephrology, Dialysis and Renal Transplant Unit, IRCCS-Azienda Ospedaliero-Universitaria di Bologna, Alma Mater Studiorum University of Bologna, Bologna, Italy
| | - Carlo Minicucci
- Nephrology, Dialysis and Renal Transplant Unit, IRCCS-Azienda Ospedaliero-Universitaria di Bologna, Alma Mater Studiorum University of Bologna, Bologna, Italy
| | - Giuseppe Cianciolo
- Nephrology, Dialysis and Renal Transplant Unit, IRCCS-Azienda Ospedaliero-Universitaria di Bologna, Alma Mater Studiorum University of Bologna, Bologna, Italy
| |
Collapse
|
26
|
Lu D, Jiang H, Zou T, Jia Y, Zhao Y, Wang Z. Endothelial-to-mesenchymal transition: New insights into vascular calcification. Biochem Pharmacol 2023; 213:115579. [PMID: 37589048 DOI: 10.1016/j.bcp.2023.115579] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Revised: 04/23/2023] [Accepted: 04/25/2023] [Indexed: 08/18/2023]
Abstract
With the continuous progress of atherosclerosis research, the significant pathological change of it--vascular calcification (VC), gains increasing attention. In recent years, numerous studies have demonstrated that it is an independent predictor of death risk of cardiovascular disease, and it has a strong correlation with poor clinical prognosis. As the world's population continues to age, the occurrence of VC is expected to reach its highest point in the near future. Therefore, it is essential to investigate ways to prevent or even reverse this process for clinical purposes. Endothelial-to-mesenchymal transition (EndMT) describes the progressive differentiation of endothelial cells into mesenchymal stem cells (MSCs) under various stimuli and acquisition of pluripotent cell characteristics. More and more studies show that EndMT plays a vital role in various cardiovascular diseases, including atherosclerosis, vascular calcification and heart valvular disease. EndMT is also involved in the formation and progression of VC. This review vividly describes the history, characteristics of EndMT and how it affects the endothelial cell process, then focuses on the relationship between vascular endothelium, EndMT, amino acid metabolism, and vascular calcification. Finally, it overviews the signal pathway of EndMT and drugs targeting EndMT, hoping to provide new ideas and a theoretical basis for studying potential therapeutic targets of VC.
Collapse
Affiliation(s)
- Dingkun Lu
- Department of Cardiology, Affiliated Hospital of Jiangsu University, Zhenjiang 212001, China
| | - Han Jiang
- Department of Cardiology, Affiliated Hospital of Jiangsu University, Zhenjiang 212001, China
| | - Ting Zou
- Department of Cardiology, Affiliated Hospital of Jiangsu University, Zhenjiang 212001, China
| | - Yuanwang Jia
- Department of Cardiology, Affiliated Hospital of Jiangsu University, Zhenjiang 212001, China
| | - Yunyun Zhao
- Department of Cardiology, Affiliated Hospital of Jiangsu University, Zhenjiang 212001, China
| | - Zhongqun Wang
- Department of Cardiology, Affiliated Hospital of Jiangsu University, Zhenjiang 212001, China.
| |
Collapse
|
27
|
Fularski P, Krzemińska J, Lewandowska N, Młynarska E, Saar M, Wronka M, Rysz J, Franczyk B. Statins in Chronic Kidney Disease-Effects on Atherosclerosis and Cellular Senescence. Cells 2023; 12:1679. [PMID: 37443712 PMCID: PMC10340582 DOI: 10.3390/cells12131679] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2023] [Revised: 06/19/2023] [Accepted: 06/20/2023] [Indexed: 07/15/2023] Open
Abstract
Chronic kidney disease (CKD) is a serious health problem that can affect various systems in the human body. Renal failure promotes mechanisms of premature cellular aging and also features of generalized inflammation in the body, which translates into a close relationship between kidney dysfunction and cardiovascular disease (CVD). As kidney function deteriorates, cardiovascular risk and mortality increase in this group of patients. Oxidative stress and inflammation are two closely related processes that initiate a vicious cycle by activating each other. Together with aging, they represent the key factors that cause and exacerbate CVD in CKD. Patients with CKD are particularly vulnerable to the accumulation of aging endothelial cells, vascular smooth muscle and macrophages, increasing the risk of atherosclerosis. Several mechanisms are known that can lead to the progression of the aforementioned problems, such as the accumulation of uremic toxins, persistent inflammation, impaired lipid and electrolyte metabolism, nitric oxide (NO) deficiency, the increased production of reactive oxygen species (ROS) and damage to deoxyribonucleic acid (DNA) and mitochondria. According to research, we can distinguish a group of drugs that effectively counteract the negative effects of CKD-statins. This is a group of drugs that inhibit 3-hydroxy-3-methylglutaryl-coenzyme-A (HMG-CoA) reductase and affect a number of cellular processes and pathways, resulting in the overall slowing of atherosclerosis and cellular aging.
Collapse
Affiliation(s)
- Piotr Fularski
- Department of Nephrocardiology, Medical University of Lodz, ul. Zeromskiego 113, 90-549 Lodz, Poland; (P.F.); (J.K.); (N.L.); (M.S.); (M.W.); (B.F.)
| | - Julia Krzemińska
- Department of Nephrocardiology, Medical University of Lodz, ul. Zeromskiego 113, 90-549 Lodz, Poland; (P.F.); (J.K.); (N.L.); (M.S.); (M.W.); (B.F.)
| | - Natalia Lewandowska
- Department of Nephrocardiology, Medical University of Lodz, ul. Zeromskiego 113, 90-549 Lodz, Poland; (P.F.); (J.K.); (N.L.); (M.S.); (M.W.); (B.F.)
| | - Ewelina Młynarska
- Department of Nephrocardiology, Medical University of Lodz, ul. Zeromskiego 113, 90-549 Lodz, Poland; (P.F.); (J.K.); (N.L.); (M.S.); (M.W.); (B.F.)
| | - Maciej Saar
- Department of Nephrocardiology, Medical University of Lodz, ul. Zeromskiego 113, 90-549 Lodz, Poland; (P.F.); (J.K.); (N.L.); (M.S.); (M.W.); (B.F.)
| | - Magdalena Wronka
- Department of Nephrocardiology, Medical University of Lodz, ul. Zeromskiego 113, 90-549 Lodz, Poland; (P.F.); (J.K.); (N.L.); (M.S.); (M.W.); (B.F.)
| | - Jacek Rysz
- Department of Nephrology, Hypertension and Family Medicine, Medical University of Lodz, ul. Zeromskiego 113, 90-549 Lodz, Poland;
| | - Beata Franczyk
- Department of Nephrocardiology, Medical University of Lodz, ul. Zeromskiego 113, 90-549 Lodz, Poland; (P.F.); (J.K.); (N.L.); (M.S.); (M.W.); (B.F.)
| |
Collapse
|
28
|
Bechelli C, Macabrey D, Deglise S, Allagnat F. Clinical Potential of Hydrogen Sulfide in Peripheral Arterial Disease. Int J Mol Sci 2023; 24:9955. [PMID: 37373103 DOI: 10.3390/ijms24129955] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Revised: 06/01/2023] [Accepted: 06/05/2023] [Indexed: 06/29/2023] Open
Abstract
Peripheral artery disease (PAD) affects more than 230 million people worldwide. PAD patients suffer from reduced quality of life and are at increased risk of vascular complications and all-cause mortality. Despite its prevalence, impact on quality of life and poor long-term clinical outcomes, PAD remains underdiagnosed and undertreated compared to myocardial infarction and stroke. PAD is due to a combination of macrovascular atherosclerosis and calcification, combined with microvascular rarefaction, leading to chronic peripheral ischemia. Novel therapies are needed to address the increasing incidence of PAD and its difficult long-term pharmacological and surgical management. The cysteine-derived gasotransmitter hydrogen sulfide (H2S) has interesting vasorelaxant, cytoprotective, antioxidant and anti-inflammatory properties. In this review, we describe the current understanding of PAD pathophysiology and the remarkable benefits of H2S against atherosclerosis, inflammation, vascular calcification, and other vasculo-protective effects.
Collapse
Affiliation(s)
- Clémence Bechelli
- Department of Vascular Surgery, Lausanne University Hospital, 1005 Lausanne, Switzerland
| | - Diane Macabrey
- Department of Vascular Surgery, Lausanne University Hospital, 1005 Lausanne, Switzerland
| | - Sebastien Deglise
- Department of Vascular Surgery, Lausanne University Hospital, 1005 Lausanne, Switzerland
| | - Florent Allagnat
- Department of Vascular Surgery, Lausanne University Hospital, 1005 Lausanne, Switzerland
| |
Collapse
|
29
|
Snijders BMG, Peters MJL, Koek HL. Ectopic Calcification: What Do We Know and What Is the Way Forward? J Clin Med 2023; 12:jcm12113687. [PMID: 37297880 DOI: 10.3390/jcm12113687] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Accepted: 05/12/2023] [Indexed: 06/12/2023] Open
Abstract
Ectopic calcification, or ectopic mineralization, is a pathologic condition in which calcifications develop in soft tissues [...].
Collapse
Affiliation(s)
- Birgitta M G Snijders
- Department of Geriatrics, University Medical Center Utrecht, Utrecht University, 3584 CX Utrecht, The Netherlands
| | - Mike J L Peters
- Department of Geriatrics, University Medical Center Utrecht, Utrecht University, 3584 CX Utrecht, The Netherlands
- Department of Internal Medicine, University Medical Center Utrecht, Utrecht University, 3584 CX Utrecht, The Netherlands
| | - Huiberdina L Koek
- Department of Geriatrics, University Medical Center Utrecht, Utrecht University, 3584 CX Utrecht, The Netherlands
| |
Collapse
|
30
|
De Marzo V, Viglino U, Zecchino S, Matos JG, Piredda E, Pigati M, Vercellino M, Crimi G, Balbi M, Seitun S, Porto I. Supra-renal aortic atheroma extent and composition predict acute kidney injury after transcatheter aortic valve replacement: A three-dimensional computed tomography study. Int J Cardiol 2023; 381:8-15. [PMID: 37001646 DOI: 10.1016/j.ijcard.2023.03.053] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Revised: 03/21/2023] [Accepted: 03/24/2023] [Indexed: 04/09/2023]
Abstract
OBJECTIVE Acute kidney injury (AKI) may complicate transcatheter aortic valve replacement (TAVR) and could be linked to atheroembolization associated with catheter manipulation in the supra-renal (SR) aorta. We sought to determine the impact of SR aortic atheroma burden (SR-AAB) and composition, as well as of the aortic valve calcium score (AV-CS), measured at pre-operative multislice computed tomography (PO-MSCT), on AKI-TAVR. METHODS All TAVR-patients 3 January-2018 to December-2020 were included. A three-dimensional analysis of PO-MSCT was performed, calculating percentage SR-AAB (%SR-AAB) as [(absolute SR-AAB volume)*100/vessel volume]. Types of plaque were defined according to Hounsfield unit (HU) intensity ranges. Calcified plaque was subcategorized into 3 strata: low- (351-700 HU), mid- (701-1000 HU), and high‑calcium (>1000 HU, termed 1 K-plaque). RESULTS The study population included 222 patients [mean age 83.3 ± 5.7 years, 95 (42.8%) males], AKI-TAVR occurred in 67/222 (30.2%). Absolute SR-AAB (41.3 ± 16.4 cm3 vs. 32.5 ± 10.7 cm3,p < 0.001) and %SR-AAB (17.6 ± 5.1% vs. 13.9 ± 4.3%,p < 0.001) were significantly higher in patients developing AKI-TAVR. Patients who developed AKI-TAVR had higher mid‑calcium (6.9 ± 3.8% vs. 4.2 ± 3.5%,p < 0.001) and 1 K-plaque (5.4 ± 3.7% vs. 2.4 ± 2.4%,p < 0.001) with no difference in AV-CS (p = 0.691). Adjusted multivariable logistic regression analysis showed that %SR-AAB [OR (x%increase): 1.12, 95%CI: 1.04-1.22,p = 0.006] and %SR-calcified plaque [OR (x%increase): 5.60, 95%CI: 2.50-13.36,p < 0.001] were associated with AKI-TAVR. Finally, 3-knots spline analyses identified %SR-AAB >15.0% and %SR-calcified plaque >7.0% as optimal thresholds to predict an increased risk of AKI-TAVR. CONCLUSIONS Suprarenal aortic atheroma, when highly calcified, is associated with AKI-TAVR. Perioperative-MSCT assessment of aortic atherosclerosis may help in identification of patients at high-risk for AKI-TAVR, who could benefit from higher peri-operative surveillance.
Collapse
Affiliation(s)
- Vincenzo De Marzo
- Chair of Cardiovascular Disease, Department of Internal Medicine and Specialties (Di.M.I.), University of Genoa, Italy
| | - Umberto Viglino
- Radiology Unit, IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Simone Zecchino
- Chair of Cardiovascular Disease, Department of Internal Medicine and Specialties (Di.M.I.), University of Genoa, Italy
| | - Joao Gavina Matos
- Radiology Unit, IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Elisa Piredda
- Chair of Cardiovascular Disease, Department of Internal Medicine and Specialties (Di.M.I.), University of Genoa, Italy
| | - Maria Pigati
- Radiology Unit, IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Matteo Vercellino
- Chair of Cardiovascular Disease, Department of Internal Medicine and Specialties (Di.M.I.), University of Genoa, Italy
| | - Gabriele Crimi
- Chair of Cardiovascular Disease, Department of Internal Medicine and Specialties (Di.M.I.), University of Genoa, Italy
| | - Manrico Balbi
- Chair of Cardiovascular Disease, Department of Internal Medicine and Specialties (Di.M.I.), University of Genoa, Italy; Cardiology Unit, Cardiothoracic and Vascular Department, IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Sara Seitun
- Radiology Unit, IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Italo Porto
- Chair of Cardiovascular Disease, Department of Internal Medicine and Specialties (Di.M.I.), University of Genoa, Italy; Cardiology Unit, Cardiothoracic and Vascular Department, IRCCS Ospedale Policlinico San Martino, Genoa, Italy.
| |
Collapse
|
31
|
Park HJ, Kim Y, Kim MK, Kim HJ, Bae SK, Bae MK. Inhibition of the Semaphorin 4D-Plexin-B1 axis prevents calcification in vascular smooth muscle cells. BMB Rep 2023; 56:160-165. [PMID: 36443004 PMCID: PMC10068346 DOI: 10.5483/bmbrep.2022-0165] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Revised: 11/01/2022] [Accepted: 11/26/2022] [Indexed: 04/14/2024] Open
Abstract
Vascular calcification is common in cardiovascular diseases including atherosclerosis, and is associated with an increased risk of pathological events and mortality. Some semaphorin family members play an important role in atherosclerosis. In the present study, we show that Semaphorin 4D/Sema4D and its Plexin-B1 receptor were significantly upregulated in calcified aorta of a rat chronic kidney disease model. Significantly higher Sema4D and Plexin-B1 expression was also observed during inorganic phosphate-induced calcification of vascular smooth muscle cells. Knockdown of Sema4D or Plexin-B1 genes attenuated both the phosphate-induced osteogenic phenotype of vascular smooth muscle cells, through regulation of SMAD1/5 signaling, as well as apoptosis of vascular smooth muscle cells, through modulation of the Gas6/Axl/Akt survival pathway. Taken together, our results offer new insights on the role of Sema4D and Plexin-B1 as potential therapeutic targets against vascular calcification. [BMB Reports 2023; 56(3): 160-165].
Collapse
Affiliation(s)
- Hyun-Joo Park
- Department of Oral Physiology, School of Dentistry, Pusan National University, Yangsan 50612, Korea
- Periodontal Disease Signaling Network Research Center (MRC), School of Dentistry, Pusan National University, Yangsan 50612, Korea
- Dental and Life Science Institute, School of Dentistry, Pusan National University, Yangsan 50612, Korea
| | - Yeon Kim
- Department of Oral Physiology, School of Dentistry, Pusan National University, Yangsan 50612, Korea
- Periodontal Disease Signaling Network Research Center (MRC), School of Dentistry, Pusan National University, Yangsan 50612, Korea
- Dental and Life Science Institute, School of Dentistry, Pusan National University, Yangsan 50612, Korea
| | - Mi-Kyoung Kim
- Department of Oral Physiology, School of Dentistry, Pusan National University, Yangsan 50612, Korea
- Periodontal Disease Signaling Network Research Center (MRC), School of Dentistry, Pusan National University, Yangsan 50612, Korea
| | - Hyung Joon Kim
- Department of Oral Physiology, School of Dentistry, Pusan National University, Yangsan 50612, Korea
- Periodontal Disease Signaling Network Research Center (MRC), School of Dentistry, Pusan National University, Yangsan 50612, Korea
- Dental and Life Science Institute, School of Dentistry, Pusan National University, Yangsan 50612, Korea
| | - Soo-Kyung Bae
- Periodontal Disease Signaling Network Research Center (MRC), School of Dentistry, Pusan National University, Yangsan 50612, Korea
- Dental and Life Science Institute, School of Dentistry, Pusan National University, Yangsan 50612, Korea
- Department of Dental Pharmacology, School of Dentistry, Pusan National University, Yangsan 50612, Korea
| | - Moon-Kyoung Bae
- Department of Oral Physiology, School of Dentistry, Pusan National University, Yangsan 50612, Korea
- Periodontal Disease Signaling Network Research Center (MRC), School of Dentistry, Pusan National University, Yangsan 50612, Korea
- Dental and Life Science Institute, School of Dentistry, Pusan National University, Yangsan 50612, Korea
| |
Collapse
|
32
|
Wen W, Portales-Castillo I, Seethapathy R, Krinsky S, Kroshinsky D, Kalim S, Goverman J, Nazarian RM, Chitalia V, Malhotra R, Kramann R, Malhotra CK, Nigwekar SU. Intravenous sodium thiosulphate for vascular calcification of hemodialysis patients-a systematic review and meta-analysis. Nephrol Dial Transplant 2023; 38:733-745. [PMID: 35521751 PMCID: PMC10111152 DOI: 10.1093/ndt/gfac171] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2022] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND Vascular calcification (VC) is a common comorbidity among patients with chronic kidney disease (CKD), indicating major cardiovascular events. This study aimed to evaluate the effects and safety of intravenous sodium thiosulphate (STS) for VC in CKD patients. METHODS Electronic databases were searched for clinical trials that provided data comparing outcomes among patients treated with and without STS. The PRISMA guidelines were followed. Efficacy was assessed using calcification scores and arterial stiffness. Safety was examined by analyzing adverse symptoms, electrolytes and bone mineral density (BMD). Random-effects models were performed. Meta-regression and sensitivity analysis were done. The risk of bias was assessed using the Cochrane tools. RESULTS Among the 5601 publications, 6 studies involving 305 participants (mean age: 56 years, male: 56.6%) with all participants on maintenance hemodialysis met eligibility criteria. For efficacy, the progression in Agatston scores in the coronary arteries [107 patients, mean difference (MD): -241.27, 95% confidence interval (95% CI): -421.50 to -61.03] and iliac arteries (55 patients, MD: -382.00, 95% CI: -751.07 to -12.93) was lower in the STS treated group compared with controls. The increase in pulse wave velocity was lower in the STS group (104 patients, MD: -1.29 m/s, 95% CI: -2.24 to -0.34 m/s). No association was found between the change in calcification scores and STS regimen. For safety, gastrointestinal symptoms (e.g. nausea) and increased anion gap acidosis were noted. No reduction in BMD by STS was observed. CONCLUSIONS Intravenous STS may attenuate the progression of VC and arterial stiffness in hemodialysis patients. Large and well-designed randomized controlled trials are warranted.
Collapse
Affiliation(s)
- Wen Wen
- Department of Nephrology, Beijing Tsinghua Changgung Hospital, School of Clinical Medicine, Tsinghua University, Beijing, China
- Division of Nephrology, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
| | | | | | - Scott Krinsky
- Division of Nephrology, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Daniela Kroshinsky
- Department of Dermatology, Massachusetts General Hospital, Boston, MA, USA
| | - Sahir Kalim
- Division of Nephrology, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Jeremy Goverman
- Sumner Redstone Burn Center, Massachusetts General Hospital, Boston, MA, USA
| | | | - Vipul Chitalia
- Renal Section, Department of Medicine, Boston University Medical Center, Boston, MA, USA
| | - Rajeev Malhotra
- Cardiovascular Research Center and the Cardiology Division of the Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Rafael Kramann
- Division of Nephrology and Clinical Immunology, Medical Faculty RWTH Aachen University, Aachen, Germany
- Institute of Experimental Medicine and Systems Biology, Medical Faculty RWTH Aachen University, Aachen, Germany
- Department of Internal Medicine, Nephrology and Transplantation, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Cindy K Malhotra
- Department of Pharmacy, Massachusetts General Hospital, Boston, MA, USA
| | - Sagar U Nigwekar
- Division of Nephrology, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
| |
Collapse
|
33
|
Ma K, Zheng ZR, Meng Y. Pathogenesis of Chronic Kidney Disease Is Closely Bound up with Alzheimer's Disease, Especially via the Renin-Angiotensin System. J Clin Med 2023; 12:jcm12041459. [PMID: 36835994 PMCID: PMC9966558 DOI: 10.3390/jcm12041459] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Revised: 02/01/2023] [Accepted: 02/07/2023] [Indexed: 02/15/2023] Open
Abstract
Chronic kidney disease (CKD) is a clinical syndrome secondary to the definitive change in function and structure of the kidney, which is characterized by its irreversibility and slow and progressive evolution. Alzheimer's disease (AD) is characterized by the extracellular accumulation of misfolded β-amyloid (Aβ) proteins into senile plaques and the formation of neurofibrillary tangles (NFTs) containing hyperphosphorylated tau. In the aging population, CKD and AD are growing problems. CKD patients are prone to cognitive decline and AD. However, the connection between CKD and AD is still unclear. In this review, we take the lead in showing that the development of the pathophysiology of CKD may also cause or exacerbate AD, especially the renin-angiotensin system (RAS). In vivo studies had already shown that the increased expression of angiotensin-converting enzyme (ACE) produces a positive effect in aggravating AD, but ACE inhibitors (ACEIs) have protective effects against AD. Among the possible association of risk factors in CKD and AD, we mainly discuss the RAS in the systemic circulation and the brain.
Collapse
Affiliation(s)
- Ke Ma
- The First Affiliated Hospital of Jinan University, Guangzhou 510000, China
| | - Zi-Run Zheng
- The First Affiliated Hospital of Jinan University, Guangzhou 510000, China
| | - Yu Meng
- The First Affiliated Hospital of Jinan University, Guangzhou 510000, China
- Central Laboratory, The Fifth Affiliated Hospital of Jinan University, Heyuan 517000, China
- Institute of Nephrology, Jinan University, Guangzhou 510000, China
- Correspondence:
| |
Collapse
|
34
|
Tan JL, Yi J, Cao XY, Wang FY, Xie SL, Zhou LL, Qin L, Dai AG. Celastrol: The new dawn in the treatment of vascular remodeling diseases. Biomed Pharmacother 2023; 158:114177. [PMID: 36809293 DOI: 10.1016/j.biopha.2022.114177] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Revised: 12/16/2022] [Accepted: 12/28/2022] [Indexed: 01/05/2023] Open
Abstract
Evidence is mounting that abnormal vascular remodeling leads to many cardiovascular diseases (CVDs). This suggests that vascular remodeling can be a crucial target for the prevention and treatment of CVDs. Recently, celastrol, an active ingredient of the broadly used Chinese herb Tripterygium wilfordii Hook F, has attracted extensive interest for its proven potential to improve vascular remodeling. Substantial evidence has shown that celastrol improves vascular remodeling by ameliorating inflammation, hyperproliferation, and migration of vascular smooth muscle cells, vascular calcification, endothelial dysfunction, extracellular matrix remodeling, and angiogenesis. Moreover, numerous reports have proven the positive effects of celastrol and its therapeutic promise in treating vascular remodeling diseases such as hypertension, atherosclerosis, and pulmonary artery hypertension. The present review summarizes and discusses the molecular mechanism of celastrol regulating vascular remodeling and provides preclinical proof for future clinical applications of celastrol.
Collapse
Affiliation(s)
- Jun-Lan Tan
- Department of Respiratory Diseases, School of Medicine, Hunan University of Chinese Medicine, Changsha 410208, Hunan, China; Hunan Provincial Key Laboratory of Vascular Biology and Translational Medicine, Changsha 410208, Hunan, China
| | - Jian Yi
- The First Affiliated Hospital of Hunan University of Chinese Medicine, Changsha 410021, Hunan, China
| | - Xian-Ya Cao
- Department of Respiratory Diseases, School of Medicine, Hunan University of Chinese Medicine, Changsha 410208, Hunan, China; Hunan Provincial Key Laboratory of Vascular Biology and Translational Medicine, Changsha 410208, Hunan, China
| | - Fei-Ying Wang
- Department of Respiratory Diseases, School of Medicine, Hunan University of Chinese Medicine, Changsha 410208, Hunan, China; Hunan Provincial Key Laboratory of Vascular Biology and Translational Medicine, Changsha 410208, Hunan, China
| | - Si-Lin Xie
- Department of Respiratory Diseases, School of Medicine, Hunan University of Chinese Medicine, Changsha 410208, Hunan, China; Hunan Provincial Key Laboratory of Vascular Biology and Translational Medicine, Changsha 410208, Hunan, China
| | - Ling-Ling Zhou
- Department of Respiratory Diseases, School of Medicine, Hunan University of Chinese Medicine, Changsha 410208, Hunan, China; Hunan Provincial Key Laboratory of Vascular Biology and Translational Medicine, Changsha 410208, Hunan, China
| | - Li Qin
- Hunan Provincial Key Laboratory of Vascular Biology and Translational Medicine, Changsha 410208, Hunan, China; Laboratory of Stem Cell Regulation with Chinese Medicine and Its Application, School of Pharmacy, Hunan University of Chinese Medicine, Changsha 410208, Hunan, China.
| | - Ai-Guo Dai
- Department of Respiratory Diseases, School of Medicine, Hunan University of Chinese Medicine, Changsha 410208, Hunan, China; Hunan Provincial Key Laboratory of Vascular Biology and Translational Medicine, Changsha 410208, Hunan, China; Department of Respiratory Medicine, The First Affiliated Hospital of Hunan University of Chinese Medicine, Changsha 410021, Hunan, China.
| |
Collapse
|
35
|
Ni X, Liu L, Yao Y, Zhang C, Su H, Lv Y, Li R, Sun L, Zhou Q, Zhu X, Yang Z, Chen Z, He W, Zhu H, Zhang S, Hu C, Yuan H. The genetic correlation and causal association between key factors that influence vascular calcification and cardiovascular disease incidence. Front Cardiovasc Med 2023; 10:1096662. [PMID: 36776247 PMCID: PMC9908996 DOI: 10.3389/fcvm.2023.1096662] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2022] [Accepted: 01/13/2023] [Indexed: 01/27/2023] Open
Abstract
Background Serum calcium (Ca), vitamin D (VD), and vitamin K (VK) levels are key determinants of vascular calcification, which itself impacts cardiovascular disease (CVD) risk. The specific relationships between the levels of these different compounds and particular forms of CVD, however, remain to be fully defined. Objective This study was designed to explore the associations between these serum levels and CVDs with the goal of identifying natural interventions capable of controlling vascular calcification and thereby protecting against CVD pathogenesis, extending the healthy lifespan of at-risk individuals. Methods Linkage disequilibrium score (LDSC) regression and a two-sample Mendelian randomization (MR) framework were leveraged to systematically examine the causal interplay between these serum levels and nine forms of CVD, as well as longevity through the use of large publically accessible Genome-Wide Association Studies (GWAS) datasets. The optimal concentrations of serum Ca and VD to lower CVD risk were examined through a restrictive cubic spline (RCS) approach. Results After Bonferroni correction, the positive genetic correlations were observed between serum Ca levels and myocardial infarction (MI) (p = 1.356E-04), as well as coronary artery disease (CAD) (p = 3.601E-04). Negative genetic correlations were detected between levels of VD and CAD (p = 0.035), while elevated VK1 concentrations were causally associated with heart failure (HF) [odds ratios (OR) per 1-standard deviation (SD) increase: 1.044], large artery stroke (LAS) (OR per 1-SD increase: 1.172), and all stroke (AS) (OR per 1-SD increase: 1.041). Higher serum Ca concentrations (OR per 1-SD increase: 0.865) and VD levels (OR per 1-SD increase: 0.777) were causally associated with reduced odds of longevity. These findings remained consistent in sensitivity analyses, and serum Ca and VD concentrations of 2.376 mmol/L and 46.8 nmol/L, respectively, were associated with a lower CVD risk (p < 0.001). Conclusion Our findings support a genetic correlation between serum Ca and VD and CVD risk, and a causal relationship between VK1 levels and CVD risk. The optimal serum Ca (2.376 mmol/L) and VD levels (46.8 nmol/L) can reduce cardiovascular risk.
Collapse
Affiliation(s)
- Xiaolin Ni
- The Key Laboratory of Geriatrics, Beijing Institute of Geriatrics, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing Hospital, National Center of Gerontology of National Health Commission, Beijing, China,State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China,Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China
| | - Lei Liu
- Department of Medical Microbiology and Infection Prevention, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Yao Yao
- China Center for Health Development Studies, Peking University, Beijing, China
| | - Chi Zhang
- The Key Laboratory of Geriatrics, Beijing Institute of Geriatrics, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing Hospital, National Center of Gerontology of National Health Commission, Beijing, China
| | | | - Yuan Lv
- Jiangbin Hospital, Zhenjiang, China
| | | | - Liang Sun
- The Key Laboratory of Geriatrics, Beijing Institute of Geriatrics, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing Hospital, National Center of Gerontology of National Health Commission, Beijing, China
| | - Qi Zhou
- The Key Laboratory of Geriatrics, Beijing Institute of Geriatrics, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing Hospital, National Center of Gerontology of National Health Commission, Beijing, China
| | - Xiaoquan Zhu
- The Key Laboratory of Geriatrics, Beijing Institute of Geriatrics, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing Hospital, National Center of Gerontology of National Health Commission, Beijing, China
| | - Ze Yang
- The Key Laboratory of Geriatrics, Beijing Institute of Geriatrics, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing Hospital, National Center of Gerontology of National Health Commission, Beijing, China
| | - Zuoguan Chen
- Department of Vascular Surgery, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences Peking Union Medical College, Beijing, China
| | - Wei He
- Department of Cardiology, Beijing Hospital, National Center of Gerontology, Beijing, China
| | - Huolan Zhu
- Department of Geriatrics, Shaanxi Provincial Clinical Research Center for Geriatric Medicine, Shaanxi Provincial People’s Hospital, Xi’an, China
| | - Shenqi Zhang
- Department of Joint and Sports Medicine, Zaozhuang Municipal Hospital Affiliated to Jining Medical University, Zaozhuang, Shandong, China
| | - Caiyou Hu
- Jiangbin Hospital, Zhenjiang, China,Caiyou Hu,
| | - Huiping Yuan
- The Key Laboratory of Geriatrics, Beijing Institute of Geriatrics, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing Hospital, National Center of Gerontology of National Health Commission, Beijing, China,*Correspondence: Huiping Yuan,
| |
Collapse
|
36
|
Guo L, Wang Y, Li S, Zhou L, Li D. GALNT3 protects against phosphate-induced calcification in vascular smooth muscle cells by enhancing active FGF23 and inhibiting the wnt/β-catenin signaling pathway. Cell Signal 2022; 100:110477. [PMID: 36162588 DOI: 10.1016/j.cellsig.2022.110477] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2022] [Revised: 09/12/2022] [Accepted: 09/20/2022] [Indexed: 11/03/2022]
Abstract
Vascular calcification (VC) acts as a notable risk factor in the cardiovascular system. Disorder of phosphorus (Pi) metabolism promotes VC. Recent findings show that polypeptide N-acetylgalactosaminyltransferase 3(GALNT3) is Pi responsive and with potent effects on Pi homeostasis. However, whether GALNT3 is involved in high Pi-induced VC remains unclear. The present study investigated the potential role of GALNT3 as a novel regulator of VC. In vitro, human aortic smooth muscle cells (HASMCs) calcification was induced by inorganic Pi, while in vivo, C57BL/6 J mice were used to determine the effects of GALNT3 on Vitamin D3-induced medial arterial calcification. Alizarin red staining, Von Kossa staining, calcium and alkaline phosphatase (ALP) activity were performed to test VC. We showed that expression of GALNT3 was increased in the calcified HASMCs and aortas of the calcified mice.In vitro, overexpression of GALNT3 increased the levels of active full-length FGF23, accompanied by suppression of the osteoblast-related factors (Runx2 and BMP2), and further inhibited the formation of calcified nodules. Moreover, the protein levels of Wnt3a and active β-catenin were determined and it was found that GALNT3 significantly inhibited their expression. LiCl, a Wnt/β-catenin signaling activator, was observed to reverse the protective effect of GALNT3 overexpression. The opposite results were observed in the GALNT3 knockdown cells. In vivo, overexpression of GALNT3 by adeno-associated virus decreased the serum Pi and slowed the formation of aortic calcification in the calcified mice. In conclusion, our results indicate that GALNT3 counteracts high Pi-induced osteoblastic differentiation of VSMCs and protects against the initiation and progression of VC by inhibiting the Wnt/β-catenin signaling pathway.
Collapse
Affiliation(s)
- Liwei Guo
- School of Forensic Medicine, Xinxiang Medical University, Xinxiang, Henan, China; Xinxiang Key Laboratory of Metabolism and Integrative Physiology, Xinxiang, Henan, China.
| | - Yikai Wang
- School of Forensic Medicine, Xinxiang Medical University, Xinxiang, Henan, China; Xinxiang Key Laboratory of Metabolism and Integrative Physiology, Xinxiang, Henan, China
| | - Shijie Li
- School of Forensic Medicine, Xinxiang Medical University, Xinxiang, Henan, China; Xinxiang Key Laboratory of Metabolism and Integrative Physiology, Xinxiang, Henan, China
| | - Lulu Zhou
- School of Forensic Medicine, Xinxiang Medical University, Xinxiang, Henan, China; Xinxiang Key Laboratory of Metabolism and Integrative Physiology, Xinxiang, Henan, China
| | - Duan Li
- School of Basic Medicine, Xinxiang Medical University, Xinxiang, Henan, China.
| |
Collapse
|
37
|
Wang L, Jin F, Wang P, Hou S, Jin T, Chang X, Zhao L. Adropin Inhibits Vascular Smooth Muscle Cell Osteogenic Differentiation to Alleviate Vascular Calcification via the JAK2/STAT3 Signaling Pathway. BIOMED RESEARCH INTERNATIONAL 2022; 2022:9122264. [PMID: 35937397 PMCID: PMC9348938 DOI: 10.1155/2022/9122264] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/12/2022] [Revised: 07/01/2022] [Accepted: 07/04/2022] [Indexed: 11/17/2022]
Abstract
Vascular calcification is characterized as the deposition of hydroxyapatite mineral in the form of calcium-phosphate complexes in the vasculature. Transdifferentiation between vascular smooth muscle cells (VSMCs) and osteoblast-like cells is considered essential in the progression of vascular calcification. The pathophysiological mechanisms underlying vascular calcification and VSMC osteogenic differentiation remain to be fully elucidated, and the development of novel therapies is required. In the present study, PCR and western blot analysis were conducted to quantify the mRNA and protein expression levels of calcification-associated markers (bone morphogenetic protein 2, alkaline phosphatase, osteoprotegerin, osteocalcin, and runt-related transcription factor 2) and adropin in VSMCs and rat vascular tissues. The calcification of VSMCs was assessed using alizarin red staining. Moreover, adropin expression levels in VSMCs were analyzed using immunofluorescence. Lentiviral transfection and small interfering RNA were used for overexpression and knockdown of adropin in VSMCs, respectively. The results demonstrated that adropin alleviated vascular calcification in vivo. Moreover, adropin also inhibited osteogenic differentiation and the calcification of VSMCs in vitro. Notably, results of the present study revealed that the tyrosine protein kinase JAK2 (JAK2)/signal transducer and activator of transcription 3 (STAT3) signaling pathway played a key role in the aforementioned inhibition. In conclusion, the results of the present study demonstrated that adropin inhibited VSMC osteogenic differentiation to alleviate vascular calcification via the JAK2/STAT3 signaling pathway.
Collapse
Affiliation(s)
- Li Wang
- Department of Cardiology, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China 215004
- Emergency Department, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China 215004
| | - Fulu Jin
- Department of Cardiology, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China 215004
| | - Peiyu Wang
- Department of Cardiology, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China 215004
| | - Shiqiang Hou
- Department of Cardiology, Shanghai Institute of Cardiovascular Disease, Zhongshan Hospital, Fudan University, Shanghai, China 200032
| | - Tao Jin
- Department of Cardiology, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China 215004
| | - Xiansong Chang
- Department of Cardiology, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China 215004
- Emergency Department, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China 215004
| | - Liangping Zhao
- Department of Cardiology, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China 215004
| |
Collapse
|
38
|
Cardiovascular Risk after Kidney Transplantation: Causes and Current Approaches to a Relevant Burden. J Pers Med 2022; 12:jpm12081200. [PMID: 35893294 PMCID: PMC9329988 DOI: 10.3390/jpm12081200] [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: 06/29/2022] [Revised: 07/11/2022] [Accepted: 07/20/2022] [Indexed: 11/17/2022] Open
Abstract
Background. Cardiovascular disease is a frequent complication after kidney transplantation and represents the leading cause of mortality in this population. Material and Methods. We searched for the relevant articles in the National Institutes of Health library of medicine, transplant, cardiologic and nephrological journals. Results. The pathogenesis of cardiovascular disease in kidney transplant is multifactorial. Apart from non-modifiable risk factors, such as age, gender, genetic predisposition and ethnicity, several traditional and non-traditional modifiable risk factors contribute to its development. Traditional factors, such as diabetes, hypertension and dyslipidemia, may be present before and may worsen after transplantation. Immunosuppressants and impaired graft function may strongly influence the exacerbation of these comorbidities. However, in the last years, several studies showed that many other cardiovascular risk factors may be involved in kidney transplantation, including hyperuricemia, inflammation, low klotho and elevated Fibroblast Growth Factor 23 levels, deficient levels of vitamin D, vascular calcifications, anemia and poor physical activity and quality of life. Conclusions. The timely and effective treatment of time-honored and recently discovered modifiable risk factors represent the basis of the prevention of cardiovascular complications in kidney transplantation. Reduction of cardiovascular risk can improve the life expectancy, the quality of life and the allograft function and survival.
Collapse
|
39
|
Exogenous BMP7 administration attenuated vascular calcification and improved bone disorders in chronic uremic rats. Biochem Biophys Res Commun 2022; 621:8-13. [DOI: 10.1016/j.bbrc.2022.06.101] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2022] [Revised: 06/13/2022] [Accepted: 06/30/2022] [Indexed: 11/18/2022]
|
40
|
Merino-Ribas A, Araujo R, Pereira L, Campos J, Barreiros L, Segundo MA, Silva N, Costa CFFA, Quelhas-Santos J, Trindade F, Falcão-Pires I, Alencastre I, Dumitrescu IB, Sampaio-Maia B. Vascular Calcification and the Gut and Blood Microbiome in Chronic Kidney Disease Patients on Peritoneal Dialysis: A Pilot Study. Biomolecules 2022; 12:biom12070867. [PMID: 35883423 PMCID: PMC9313079 DOI: 10.3390/biom12070867] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Revised: 06/08/2022] [Accepted: 06/16/2022] [Indexed: 12/13/2022] Open
Abstract
Vascular calcification (VC) is a frequent condition in chronic kidney disease (CKD) and a well-established risk factor for the development of cardiovascular disease (CVD). Gut dysbiosis may contribute to CVD and inflammation in CKD patients. Nonetheless, the role of gut and blood microbiomes in CKD-associated VC remains unknown. Therefore, this pilot study aimed to explore the link between gut and blood microbiomes and VC in CKD patients on peritoneal dialysis (CKD-PD). Our results showed relative changes in specific taxa between CKD-PD patients with and without VC, namely Coprobacter, Coprococcus 3, Lactobacillus, and Eubacterium eligens group in the gut, and Cutibacterium, Pajaroellobacter, Devosia, Hyphomicrobium, and Pelomonas in the blood. An association between VC and all-cause mortality risk in CKD-PD patients was also observed, and patients with higher mortality risk corroborate the changes of Eubacterium eligens in the gut and Devosia genus in the blood. Although we did not find differences in uremic toxins, intestinal translocation markers, and inflammatory parameters among CKD-PD patients with and without VC, soluble CD14 (sCD14), a nonspecific marker of monocyte activation, positively correlated with VC severity. Therefore, gut Eubacterium eligens group, blood Devosia, and circulating sCD14 should be further explored as biomarkers for VC, CVD, and mortality risk in CKD.
Collapse
Affiliation(s)
- Ana Merino-Ribas
- Nephrology & Infectious Diseases R & D Group, i3S—Instituto de Investigação e Inovação em Saúde, INEB—Instituto de Engenharia Biomédica, Universidade do Porto, 4200-135 Porto, Portugal; (A.M.-R.); (R.A.); (L.P.); (J.C.); (C.F.F.A.C.); (I.A.)
- Departament de Medicina, Universitat Autonoma de Barcelona, 08035 Barcelona, Spain;
- Nephrology Department, Hospital Universitari de Girona Doctor Josep Trueta, 17007 Girona, Spain
| | - Ricardo Araujo
- Nephrology & Infectious Diseases R & D Group, i3S—Instituto de Investigação e Inovação em Saúde, INEB—Instituto de Engenharia Biomédica, Universidade do Porto, 4200-135 Porto, Portugal; (A.M.-R.); (R.A.); (L.P.); (J.C.); (C.F.F.A.C.); (I.A.)
| | - Luciano Pereira
- Nephrology & Infectious Diseases R & D Group, i3S—Instituto de Investigação e Inovação em Saúde, INEB—Instituto de Engenharia Biomédica, Universidade do Porto, 4200-135 Porto, Portugal; (A.M.-R.); (R.A.); (L.P.); (J.C.); (C.F.F.A.C.); (I.A.)
- Nephrology Department, Centro Hospitalar Universitário de São João, 4200-319 Porto, Portugal;
| | - Joana Campos
- Nephrology & Infectious Diseases R & D Group, i3S—Instituto de Investigação e Inovação em Saúde, INEB—Instituto de Engenharia Biomédica, Universidade do Porto, 4200-135 Porto, Portugal; (A.M.-R.); (R.A.); (L.P.); (J.C.); (C.F.F.A.C.); (I.A.)
| | - Luísa Barreiros
- LAQV, REQUIMTE, Departamento de Ciências Químicas, Faculdade de Farmácia, Universidade do Porto, 4050-313 Porto, Portugal; (L.B.); (M.A.S.)
| | - Marcela A. Segundo
- LAQV, REQUIMTE, Departamento de Ciências Químicas, Faculdade de Farmácia, Universidade do Porto, 4050-313 Porto, Portugal; (L.B.); (M.A.S.)
| | - Nádia Silva
- Nephrology Department, Centro Hospitalar Universitário de São João, 4200-319 Porto, Portugal;
| | - Carolina F. F. A. Costa
- Nephrology & Infectious Diseases R & D Group, i3S—Instituto de Investigação e Inovação em Saúde, INEB—Instituto de Engenharia Biomédica, Universidade do Porto, 4200-135 Porto, Portugal; (A.M.-R.); (R.A.); (L.P.); (J.C.); (C.F.F.A.C.); (I.A.)
- Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, 4050-313 Porto, Portugal
| | - Janete Quelhas-Santos
- UnIC@RISE- Cardiovascular Research and Development Centre, Department of Surgery and Physiology, Faculty of Medicine, University of Porto, 4200-319 Porto, Portugal; (J.Q.-S.); (F.T.); (I.F.-P.)
| | - Fábio Trindade
- UnIC@RISE- Cardiovascular Research and Development Centre, Department of Surgery and Physiology, Faculty of Medicine, University of Porto, 4200-319 Porto, Portugal; (J.Q.-S.); (F.T.); (I.F.-P.)
| | - Inês Falcão-Pires
- UnIC@RISE- Cardiovascular Research and Development Centre, Department of Surgery and Physiology, Faculty of Medicine, University of Porto, 4200-319 Porto, Portugal; (J.Q.-S.); (F.T.); (I.F.-P.)
| | - Ines Alencastre
- Nephrology & Infectious Diseases R & D Group, i3S—Instituto de Investigação e Inovação em Saúde, INEB—Instituto de Engenharia Biomédica, Universidade do Porto, 4200-135 Porto, Portugal; (A.M.-R.); (R.A.); (L.P.); (J.C.); (C.F.F.A.C.); (I.A.)
| | - Ioana Bancu Dumitrescu
- Departament de Medicina, Universitat Autonoma de Barcelona, 08035 Barcelona, Spain;
- Fresenius Nephrocare, 110372 Pitesti, Romania
| | - Benedita Sampaio-Maia
- Nephrology & Infectious Diseases R & D Group, i3S—Instituto de Investigação e Inovação em Saúde, INEB—Instituto de Engenharia Biomédica, Universidade do Porto, 4200-135 Porto, Portugal; (A.M.-R.); (R.A.); (L.P.); (J.C.); (C.F.F.A.C.); (I.A.)
- Faculdade de Medicina Dentária, Universidade do Porto, 4200-393 Porto, Portugal
- Correspondence: ; Tel.: +351-220-901-100
| |
Collapse
|
41
|
Pan X, Pi C, Ruan X, Zheng H, Zhang D, Liu X. Mammalian Sirtuins and Their Relevance in Vascular Calcification. Front Pharmacol 2022; 13:907835. [PMID: 35677446 PMCID: PMC9168231 DOI: 10.3389/fphar.2022.907835] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Accepted: 05/09/2022] [Indexed: 12/11/2022] Open
Abstract
Cardiovascular diseases are a group of diseases with high morbidity and mortality that affect millions of people each year. Vascular calcification (VC) is an active process that involves the mineral deposition of calcium-phosphate complexes. VC is closely related to cardiovascular diseases, such as hypertension, heart failure, and calcific aortic stenosis, and is a type of ectopic calcification that occurs in the vessel walls. The sirtuins (silent mating-type information regulation 2; SIRTs), are a family of histone deacetylases whose function relies on nicotinamide adenine dinucleotide (NAD+). They have non-negligible functions in the regulation of energy metabolism, senescence, apoptosis, and other biological processes. Sirtuins have important effects on bone homeostasis and VC processes that share many similarities with bone formation. Sirtuins have been confirmed to deacetylate a variety of target proteins related to the occurrence and development of VC, thereby affecting the process of VC and providing new possibilities for the prevention and treatment of cardiovascular diseases. To facilitate the understanding of vascular calcification and accelerate the development of cardiovascular drugs, we reviewed and summarized recent research progress on the relationship between different types of sirtuins and VC.
Collapse
Affiliation(s)
- Xinyue Pan
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Caixia Pi
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Xianchun Ruan
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
- West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu, China
| | - Hanhua Zheng
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
- West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu, China
| | - Demao Zhang
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
- West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu, China
- *Correspondence: Demao Zhang, ; Xiaoheng Liu,
| | - Xiaoheng Liu
- West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu, China
- *Correspondence: Demao Zhang, ; Xiaoheng Liu,
| |
Collapse
|
42
|
Sun Y, Zhang H, Tian W. Dietary fiber and prevalence of abdominal aortic calcification in the United States (from the national health and nutrition examination survey data [2013-2014]). Nutr J 2022; 21:25. [PMID: 35524312 PMCID: PMC9074363 DOI: 10.1186/s12937-022-00782-0] [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: 01/14/2022] [Accepted: 04/29/2022] [Indexed: 11/29/2022] Open
Abstract
Background Abdominal aortic calcification (AAC) is recognized as a valuable predictor of cardiovascular diseases (CVDs). Dietary fiber is strongly correlated with CVDs. However, the effect of dietary fiber on AAC in the population is not well understood. Objective To assess the relationship between dietary fiber intake and AAC in the US adult population. Methods A total of 2671 individuals with both dietary fiber intake and AAC score data were enrolled from the 2013–2014 National Health and Nutrition Examination Survey (NHANES), a cross-sectional health examination in the US. Multinomial logistic regression was used to calculate the odds ratio (OR), with 95% confidence interval (CI). To reveal the relationship between dietary fiber intake and AAC, restricted cubic spline was also applied. Results Out of the total participants, 241 (9%) had severe AAC and 550 (20%) had mild-moderate AAC. Multinomial logistic regression indicated that higher intake of dietary fiber was associated with lower risk of severe AAC, but not with lower risk of mild-moderate AAC. For every one standard deviation increase (9.4 g/day) in dietary fiber intake, the odds of severe AAC were reduced by 28% [OR 0.72 (95% CI, 0.57–0.90), p = 0.004], after adjusting for confounding factors. Dose–response relationship revealed that dietary fiber intake was negatively correlated with severe AAC (p for linear < 0.001, p for nonlinear = 0.695). Conclusions Dietary fiber intake was negatively associated with severe AAC, and showed a dose–response relationship in US adults.
Collapse
Affiliation(s)
- YuJiao Sun
- Department of Geriatrics, The First Affiliated Hospital of China Medical University, No.155 Nanjing North Street, Shenyang, 110001, China
| | - HuanRui Zhang
- Department of Geriatrics, The First Affiliated Hospital of China Medical University, No.155 Nanjing North Street, Shenyang, 110001, China
| | - Wen Tian
- Department of Geriatrics, The First Affiliated Hospital of China Medical University, No.155 Nanjing North Street, Shenyang, 110001, China.
| |
Collapse
|
43
|
Zhang F, Hao N, Wang L, Sun G, Feng X, Li C, Tan W, Wang F. Association of serum creatinine with aortic arch calcification in middle-aged and elderly adults: an observational cross-sectional study from China. BMC Cardiovasc Disord 2022; 22:167. [PMID: 35413799 PMCID: PMC9004043 DOI: 10.1186/s12872-022-02617-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2021] [Accepted: 04/04/2022] [Indexed: 11/10/2022] Open
Abstract
Background and aims Vascular calcification (VC) is a strong predictor of cardiovascular events and all-cause mortality in cardiovascular diseases (CVD). Renal dysfunction is closely related to VC. Serum creatinine, as an important indicator of renal function in chronic kidney disease (CKD), is closely associated with increased VC. Here, to explore the potential role of serum creatinine in CVD, we examined the association between serum creatinine level and aortic arch calcification (AAC) presence in a larger general population. Methods A total of 9067 participants aged > 45 years were included in this study. All participants underwent postero-anterior chest X-ray examination to diagnose AAC. According to the distribution characteristics, serum creatinine levels in male and female were divided into tertiles respectively. Univariate and multivariate logistic regression analysis were used to analyze the association between aortic calcification and serum creatinine. Results Participants included 3776 men and 5291 women, and 611 and 990 AAC were detected, respectively. Serum creatinine level in the female AAC group was significantly higher than that in the non-AAC group (p < 0.001), while there was no significant difference in male serum creatinine between the two groups (p = 0.241). After logistic regression analysis excluded confounding factors, with the first tertile of serum creatinine as the reference, multivariable-adjusted ORs and 95% CIs of the second and the highest tertile of female and male were 1.045 (0.856–1.276), 1.263 (1.036–1.539); 0.953 (0.761–1.193), 0.948 (0.741–1.198), respectively. Conclusion Elevated serum creatinine levels are independently associated with higher AAC incidence in female aged > 45 years old. Measuring serum creatinine levels may assist the early screening individuals at high risk of developing CVD. And higher attention should be given to female's serum creatinine levels in daily clinical practice. Supplementary Information The online version contains supplementary material available at 10.1186/s12872-022-02617-6.
Collapse
Affiliation(s)
- Feifei Zhang
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, Jiangsu, China
| | - Nannan Hao
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, Jiangsu, China
| | - Lei Wang
- Department of Rheumatology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, Jiangsu, China
| | - Guoming Sun
- Division of Rheumatology, The Affiliated Changzhou NO.2 People's Hospital of Nanjing Medical University, Changzhou, 213004, Jiangsu, China
| | - Xiaoke Feng
- Integrated Traditional Chinese and Western Medicine Institute of Nanjing Medical University, Nanjing, 210029, Jiangsu, China
| | - Chunjian Li
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, Jiangsu, China
| | - Wenfeng Tan
- Department of Rheumatology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, Jiangsu, China.
| | - Fang Wang
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, Jiangsu, China.
| |
Collapse
|
44
|
New Therapeutics Targeting Arterial Media Calcification: Friend or Foe for Bone Mineralization? Metabolites 2022; 12:metabo12040327. [PMID: 35448514 PMCID: PMC9027727 DOI: 10.3390/metabo12040327] [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: 03/01/2022] [Revised: 03/31/2022] [Accepted: 04/03/2022] [Indexed: 01/27/2023] Open
Abstract
The presence of arterial media calcification, a highly complex and multifactorial disease, puts patients at high risk for developing serious cardiovascular consequences and mortality. Despite the numerous insights into the mechanisms underlying this pathological mineralization process, there is still a lack of effective treatment therapies interfering with the calcification process in the vessel wall. Current anti-calcifying therapeutics may induce detrimental side effects at the level of the bone, as arterial media calcification is regulated in a molecular and cellular similar way as physiological bone mineralization. This especially is a complication in patients with chronic kidney disease and diabetes, who are the prime targets of this pathology, as they already suffer from a disturbed mineral and bone metabolism. This review outlines recent treatment strategies tackling arterial calcification, underlining their potential to influence the bone mineralization process, including targeting vascular cell transdifferentiation, calcification inhibitors and stimulators, vascular smooth muscle cell (VSMC) death and oxidative stress: are they a friend or foe? Furthermore, this review highlights nutritional additives and a targeted, local approach as alternative strategies to combat arterial media calcification. Paving a way for the development of effective and more precise therapeutic approaches without inducing osseous side effects is crucial for this highly prevalent and mortal disease.
Collapse
|
45
|
Chen C, Wu Y, Lu HL, Liu K, Qin X. Identification of potential biomarkers of vascular calcification using bioinformatics analysis and validation in vivo. PeerJ 2022; 10:e13138. [PMID: 35313524 PMCID: PMC8934046 DOI: 10.7717/peerj.13138] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Accepted: 02/28/2022] [Indexed: 01/12/2023] Open
Abstract
Background Vascular calcification (VC) is the most widespread pathological change in diseases of the vascular system. However, we know poorly about the molecular mechanisms and effective therapeutic approaches of VC. Methods The VC dataset, GSE146638, was downloaded from the Gene Expression Omnibus (GEO) database. Using the edgeR package to screen Differentially expressed genes (DEGs). Gene set enrichment analysis (GSEA) and gene set variation analysis (GSVA) were used to find pathways affecting VC. Gene ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) were performed on the DEGs. Meanwhile, using the String database and Cytoscape software to construct protein-protein interaction (PPI) networks and identify hub genes with the highest module scores. Correlation analysis was performed for hub genes. Receiver operating characteristic (ROC) curves, expression level analysis, GSEA, and subcellular localization were performed for each hub gene. Expression of hub genes in normal and calcified vascular tissues was verified by quantitative reverse transcription PCR (RT-qPCR) and immunohistochemistry (IHC) experiments. The hub gene-related miRNA-mRNA and TF-mRNA networks were constructed and functionally enriched for analysis. Finally, the DGIdb database was utilized to search for alternative drugs targeting VC hub genes. Results By comparing the genes with normal vessels, there were 64 DEGs in mildly calcified vessels and 650 DEGs in severely calcified vessels. Spp1, Sost, Col1a1, Fn1, and Ibsp were central in the progression of the entire VC by the MCODE plug-in. These hub genes are primarily enriched in ossification, extracellular matrix, and ECM-receptor interactions. Expression level results showed that Spp1, Sost, Ibsp, and Fn1 were significantly highly expressed in VC, and Col1a1 was incredibly low. RT-qPCR and IHC validation results were consistent with bioinformatic analysis. We found multiple pathways of hub genes acting in VC and identified 16 targeting drugs. Conclusions This study perfected the molecular regulatory mechanism of VC. Our results indicated that Spp1, Sost, Col1a1, Fn1, and Ibsp could be potential novel biomarkers for VC and promising therapeutic targets.
Collapse
Affiliation(s)
- Chuanzhen Chen
- Department of Vascular Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Province, China
| | - Yinteng Wu
- Department of Orthopedic and Trauma Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Province, China
| | - Hai-lin Lu
- Department of Vascular Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Province, China
| | - Kai Liu
- Department of Vascular Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Province, China
| | - Xiao Qin
- Department of Vascular Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Province, China
| |
Collapse
|
46
|
Golledge J. Update on the pathophysiology and medical treatment of peripheral artery disease. Nat Rev Cardiol 2022; 19:456-474. [PMID: 34997200 DOI: 10.1038/s41569-021-00663-9] [Citation(s) in RCA: 88] [Impact Index Per Article: 29.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 12/07/2021] [Indexed: 12/15/2022]
Abstract
Approximately 6% of adults worldwide have atherosclerosis and thrombosis of the lower limb arteries (peripheral artery disease (PAD)) and the prevalence is rising. PAD causes leg pain, impaired health-related quality of life, immobility, tissue loss and a high risk of major adverse events, including myocardial infarction, stroke, revascularization, amputation and death. In this Review, I describe the pathophysiology, presentation, outcome, preclinical research and medical management of PAD. Established treatments for PAD include antithrombotic drugs, such as aspirin and clopidogrel, and medications to treat dyslipidaemia, hypertension and diabetes mellitus. Randomized controlled trials have demonstrated that these treatments reduce the risk of major adverse events. The drug cilostazol, exercise therapy and revascularization are the current treatment options for the limb symptoms of PAD, but each has limitations. Novel therapies to promote collateral and new capillary growth and treat PAD-related myopathy are under investigation. Methods to improve the implementation of evidence-based medical management, novel drug therapies and rehabilitation programmes for PAD-related pain, functional impairment and ischaemic foot disease are important areas for future research.
Collapse
Affiliation(s)
- Jonathan Golledge
- Queensland Research Centre for Peripheral Vascular Disease, College of Medicine and Dentistry, James Cook University, Townsville, Queensland, Australia. .,The Department of Vascular and Endovascular Surgery, Townsville University Hospital, Townsville, Queensland, Australia. .,The Australian Institute of Tropical Health and Medicine, James Cook University, Townsville, Queensland, Australia.
| |
Collapse
|
47
|
Park HJ, Kim MK, Kim Y, Kim HJ, Bae SK, Bae MK. Neuromedin B modulates phosphate-induced vascular calcification. BMB Rep 2021. [PMID: 34674793 PMCID: PMC8633520 DOI: 10.5483/bmbrep.2021.54.11.089] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Affiliation(s)
- Hyun-Joo Park
- Department of Oral Physiology, Yangsan 50612, Korea
- Periodontal Disease Signaling Network Research Center (MRC), Yangsan 50612, Korea
- Dental and Life Science Institute, Yangsan 50612, Korea
| | - Mi-Kyoung Kim
- Department of Oral Physiology, Yangsan 50612, Korea
- Periodontal Disease Signaling Network Research Center (MRC), Yangsan 50612, Korea
| | - Yeon Kim
- Department of Oral Physiology, Yangsan 50612, Korea
- Periodontal Disease Signaling Network Research Center (MRC), Yangsan 50612, Korea
- Dental and Life Science Institute, Yangsan 50612, Korea
| | - Hyung Joon Kim
- Department of Oral Physiology, Yangsan 50612, Korea
- Periodontal Disease Signaling Network Research Center (MRC), Yangsan 50612, Korea
- Dental and Life Science Institute, Yangsan 50612, Korea
| | - Soo-Kyung Bae
- Periodontal Disease Signaling Network Research Center (MRC), Yangsan 50612, Korea
- Dental and Life Science Institute, Yangsan 50612, Korea
- Department of Dental Pharmacology, School of Dentistry, Pusan National University, Yangsan 50612, Korea
| | - Moon-Kyoung Bae
- Department of Oral Physiology, Yangsan 50612, Korea
- Periodontal Disease Signaling Network Research Center (MRC), Yangsan 50612, Korea
- Dental and Life Science Institute, Yangsan 50612, Korea
| |
Collapse
|
48
|
Endothelial Contribution to Warfarin-Induced Arterial Media Calcification in Mice. Int J Mol Sci 2021; 22:ijms222111615. [PMID: 34769044 PMCID: PMC8583869 DOI: 10.3390/ijms222111615] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Revised: 10/22/2021] [Accepted: 10/25/2021] [Indexed: 12/12/2022] Open
Abstract
Arterial media calcification (AMC) is predominantly regulated by vascular smooth muscle cells (VSMCs), which transdifferentiate into pro-calcifying cells. In contrast, there is little evidence for endothelial cells playing a role in the disease. The current study investigates cellular functioning and molecular pathways underlying AMC, respectively by, an ex vivo isometric organ bath set-up to explore the interaction between VSMCs and ECs and quantitative proteomics followed by functional pathway interpretation. AMC development, which was induced in mice by dietary warfarin administration, was proved by positive Von Kossa staining and a significantly increased calcium content in the aorta compared to that of control mice. The ex vivo organ bath set-up showed calcified aortic segments to be significantly more sensitive to phenylephrine induced contraction, compared to control segments. This, together with the fact that calcified segments as compared to control segments, showed a significantly smaller contraction in the absence of extracellular calcium, argues for a reduced basal NO production in the calcified segments. Moreover, proteomic data revealed a reduced eNOS activation to be part of the vascular calcification process. In summary, this study identifies a poor endothelial function, next to classic pro-calcifying stimuli, as a possible initiator of arterial calcification.
Collapse
|
49
|
Arterial calcifications and osteoprotegerin in chronic hemodialysis patients: impact on 6-year survival. Int Urol Nephrol 2021; 54:1135-1143. [PMID: 34505226 DOI: 10.1007/s11255-021-02988-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2021] [Accepted: 08/21/2021] [Indexed: 10/20/2022]
Abstract
AIM The association between end-stage renal disease and cardiovascular mortality may be influenced through vascular alterations, in particular atherosclerosis and vascular calcification. The study goal was to assess the impact of each type of arterial intimal calcifications (AIC) and arterial medial calcifications (AMC), of osteoprotegerin (OPG), mineral metabolism markers and other features on all-cause and cardiovascular mortality in chronic hemodialysis patients. METHODS Ultrasound was performed in 87 patients on the carotid and femoral arteries, and the severity of AIC and AMC was assessed calculating a score according to the extension of calcification. We analyzed the link between AIC, AMC, OPG, mineral markers and mortality after 6 years of follow-up. RESULTS The cutoff value for OPG determined using ROC was 4.9 pmol/l for all-cause and cardiovascular mortality. Patients with higher serum OPG levels presented higher mortality rates. Our study revealed that AIC, high OPG, low ankle-arm index, presence of diabetes, smoking status, and lack of arteriovenous fistula are associated with all-cause and cardiovascular mortality in univariate regression analysis. Multivariate analysis identified AIC scoring based on the segmentation method as an independent predictor of all-cause and cardiovascular mortality, along with increased OPG levels. AMC scoring was not a predictor of mortality. CONCLUSIONS Identifying and scoring AIC on ultrasound and measuring OPG levels, as a basis of the HD patient assessment may become valuable tools in clinical work, as these have an impact on death toll.
Collapse
|
50
|
Hammad SK, Eissa RG, Shaheen MA, Younis NN. Resveratrol Ameliorates Aortic Calcification in Ovariectomized Rats via SIRT1 Signaling. Curr Issues Mol Biol 2021; 43:1057-1071. [PMID: 34563044 PMCID: PMC8928980 DOI: 10.3390/cimb43020075] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Revised: 09/01/2021] [Accepted: 09/02/2021] [Indexed: 12/26/2022] Open
Abstract
Postmenopausal women are at an increased risk of vascular calcification which is defined as the pathological deposition of minerals in the vasculature, and is strongly linked with increased cardiovascular disease risk. Since estrogen-replacement therapy is associated with increased cancer risk, there is a strong need for safer therapeutic approaches. In this study we aimed to investigate the protective and therapeutic effects of the phytoestrogen resveratrol against vascular calcification in ovariectomized rats, a preclinical model of postmenopause. Furthermore, we aimed to compare the effects of resveratrol to those of estrogen and to explore the mechanisms underpinning those effects. Treatment with resveratrol or estrogen ameliorated aortic calcification in ovariectomized rats, as shown by reduced calcium deposition in the arterial wall. Mechanistically, the effects of resveratrol and estrogen were mediated via the activation of SIRT1 signaling. SIRT1 protein expression was downregulated in the aortas of ovariectomized rats, and upregulated in rats treated with resveratrol or estrogen. Moreover, resveratrol and estrogen reduced the levels of the osteogenic markers: runt-related transcription factor 2 (RUNX2), osteocalcin and alkaline phosphatase (ALP) which have been shown to play a role during vascular calcification. Additionally, the senescence markers (p53, p16 and p21) which were also reported to play a role in the pathogenesis of vascular calcification, were reduced upon treatment with resveratrol and estrogen. In conclusion, the phytoestrogen resveratrol may be a safer alternative to estrogen, as a therapeutic approach against the progression of vascular calcification during postmenopause.
Collapse
Affiliation(s)
- Sally K. Hammad
- Department of Biochemistry, Faculty of Pharmacy, Zagazig University, Zagazig 44519, Egypt; (R.G.E.); (N.N.Y.)
| | - Rana G. Eissa
- Department of Biochemistry, Faculty of Pharmacy, Zagazig University, Zagazig 44519, Egypt; (R.G.E.); (N.N.Y.)
| | - Mohamed A. Shaheen
- Department of Histology and Cell Biology, Faculty of Medicine, Zagazig University, Zagazig 44519, Egypt;
| | - Nahla N. Younis
- Department of Biochemistry, Faculty of Pharmacy, Zagazig University, Zagazig 44519, Egypt; (R.G.E.); (N.N.Y.)
| |
Collapse
|