1
|
Sun X, Li C. Neural repair function of osteopontin in stroke and stroke‑related diseases (Review). Exp Ther Med 2024; 28:459. [PMID: 39478739 PMCID: PMC11523235 DOI: 10.3892/etm.2024.12749] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2024] [Accepted: 09/06/2024] [Indexed: 11/02/2024] Open
Abstract
Stroke, including hemorrhagic stroke and ischemic stroke, is a common disease of the central nervous system. It is characterized by a high mortality and disability rate and is closely associated with atherosclerosis, hypertension hyperglycemia, atrial fibrillation and unhealthy living habits. The continuous development of surgery and medications has decreased the mortality rate of patients with stroke and has greatly improved the disease prognosis. At present, the direction of clinical treatment and research has gradually shifted to the repair of nerve function after stroke. Osteopontin (OPN) is a widely distributed extracellular matrix protein. Due to its structural characteristics, OPN can be cut and modified into terminal fragments with different functions, which play different roles in various pathophysiological processes, such as formation of tumors, inflammation and autoimmune diseases. It has also become a potential diagnostic and therapeutic marker. In order to comprehensively analyze the specific role of OPN in nerve repair and its relationship with stroke and stroke-related diseases, the following key words were used: 'Osteopontin, stroke, atherosis, neuroplasticity, neural repair'. PubMed, Web of Science and Cochrane articles related to OPN were searched and summarized. The present review describes the OPN structure, isoforms, functions and its neural repair mechanism, and its association with the occurrence and development of stroke and related diseases was explored.
Collapse
Affiliation(s)
- Xin Sun
- Department of Neurosurgery, Yanbian University Affiliated Hospital, Yanbian University, Yanji, Jilin 133000, P.R. China
| | - Chunhao Li
- Department of Neurosurgery, Yanbian University Affiliated Hospital, Yanbian University, Yanji, Jilin 133000, P.R. China
| |
Collapse
|
2
|
Zusi C, Bonetti S, Rinaldi E, Csermely A, Boselli ML, Travia D, Santi L, Bonora E, Bonadonna RC, Trombetta M. Interactions of the Osteokines, Glucose/Insulin System and Vascular Risk Networks in Patients With Newly Diagnosed Type 2 Diabetes (VNDS 15). Diabetes Metab Res Rev 2024; 40:e3847. [PMID: 39393014 DOI: 10.1002/dmrr.3847] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/05/2024] [Revised: 08/08/2024] [Accepted: 08/26/2024] [Indexed: 10/13/2024]
Abstract
BACKGROUND AND AIM Bone as an endocrine organ regulates metabolic processes independently of mineral metabolism through the production/release of proteins collectively named 'osteokines'. Relevant connections were reported between the insulin/glucose system, calcification of the atherosclerotic plaque, and several osteokines. We aimed to test the hypothesis that the osteokine network could be involved in beta-cell function, insulin sensitivity, and vascular damage in a cohort of people with newly diagnosed type 2 diabetes (T2D). SUBJECTS AND METHODS In 794 drug-naive, GADA-negative, newly-diagnosed T2D patients (mean ± SD age: 59 ± 9.8 years; BMI: 29.3 ± 5.3 kg/m2; HbA1c: 6.6 ± 1.3%) we assessed: plasma concentration of osteocalcin (OCN), osteopontin (OPN), RANKL, and its putative decoy receptor osteoprotegerin (OPG); insulin sensitivity (SI) by hyperinsulinemic euglycemic clamp; beta cell function (BCF), estimated by OGTT minimal modelling and expressed as derivative (DC) and proportional (PC) control. Echo-doppler of carotid and lower limb arteries were also performed in 708 and 701 subjects, respectively. RESULTS OCN, RANKL and OPG were significantly associated with PC (p < 0.02); OCN was positively related to DC (p = 0.018). OPG was associated with lower IS (p < 0.001). Finally, the higher RANKL levels, the greater was the severity of atherosclerosis in common carotid artery (p < 0.001). Increased OPG and OPN concentrations were related to subclinical atherosclerosis in peripheral arteries of lower limbs (p = 0.023 and p = 0.047, respectively). CONCLUSION These data suggest that, in patients with newly diagnosed T2D, the osteokine network crosstalks with the glucose/insulin system and may play a role in modulating the atherosclerotic process.
Collapse
Affiliation(s)
- Chiara Zusi
- Department of Medicine, University of Verona, Verona, Italy
| | - Sara Bonetti
- Department of Medicine, University of Verona, Verona, Italy
| | - Elisabetta Rinaldi
- Department of Medicine, University of Verona, Verona, Italy
- Department of Medicine, Division of Endocrinology, Diabetes and Metabolism, Hospital Trust of Verona, Verona, Italy
| | - Alessandro Csermely
- Department of Medicine, University of Verona, Verona, Italy
- Department of Medicine, Division of Endocrinology, Diabetes and Metabolism, Hospital Trust of Verona, Verona, Italy
| | | | - Daniela Travia
- Department of Medicine, Division of Endocrinology, Diabetes and Metabolism, Hospital Trust of Verona, Verona, Italy
| | - Lorenza Santi
- Department of Medicine, University of Verona, Verona, Italy
| | - Enzo Bonora
- Department of Medicine, University of Verona, Verona, Italy
- Department of Medicine, Division of Endocrinology, Diabetes and Metabolism, Hospital Trust of Verona, Verona, Italy
| | - Riccardo C Bonadonna
- Department of Medicine and Surgery, University of Parma, Parma, Italy
- Division of Endocrinology and Metabolic Diseases, Azienda Ospedaliera Universitaria di Parma, Parma, Italy
| | - Maddalena Trombetta
- Department of Medicine, University of Verona, Verona, Italy
- Department of Medicine, Division of Endocrinology, Diabetes and Metabolism, Hospital Trust of Verona, Verona, Italy
| |
Collapse
|
3
|
Lee DY. Emerging Circulating Biomarkers for Enhanced Cardiovascular Risk Prediction. J Lipid Atheroscler 2024; 13:262-279. [PMID: 39355403 PMCID: PMC11439747 DOI: 10.12997/jla.2024.13.3.262] [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: 01/04/2024] [Revised: 04/12/2024] [Accepted: 06/06/2024] [Indexed: 10/03/2024] Open
Abstract
Cardiovascular disease (CVD) continues to be the primary cause of mortality worldwide, underscoring the importance of identifying additional cardiovascular risk factors. The consensus is that lipid levels alone do not fully reflect the status of atherosclerosis, thus necessitating extensive research on cardiovascular biomarkers. This review encompasses a wide spectrum of methodologies for identifying novel risk factors or biomarkers for CVD. Inflammation, oxidative stress, plaque instability, cardiac remodeling, and fibrosis play pivotal roles in CVD pathogenesis. We introduce and discuss several promising biomarkers-namely, osteocalcin, angiogenin, lipoprotein-associated phospholipase A2, growth differentiation factor 15, galectin-3, growth stimulation expressed gene 2, and microRNAs, all of which have potential implications in the assessment and management of cardiovascular risk.
Collapse
Affiliation(s)
- Da Young Lee
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Korea University College of Medicine, Seoul, Korea
| |
Collapse
|
4
|
Lederer ED, Sobh MM, Brier ME, Gaweda AE. Application of artificial intelligence to chronic kidney disease mineral bone disorder. Clin Kidney J 2024; 17:sfae143. [PMID: 38899159 PMCID: PMC11184350 DOI: 10.1093/ckj/sfae143] [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: 12/29/2023] [Indexed: 06/21/2024] Open
Abstract
The global derangement of mineral metabolism that accompanies chronic kidney disease (CKD-MBD) is a major driver of the accelerated mortality for individuals with kidney disease. Advances in the delivery of dialysis, in the composition of phosphate binders, and in the therapies directed towards secondary hyperparathyroidism have failed to improve the cardiovascular event profile in this population. Many obstacles have prevented progress in this field including the incomplete understanding of pathophysiology, the lack of clinical targets for early stages of chronic kidney disease, and the remarkably wide diversity in clinical manifestations. We describe in this review a novel approach to CKD-MBD combining mathematical modelling of biologic processes with machine learning artificial intelligence techniques as a tool for the generation of new hypotheses and for the development of innovative therapeutic approaches to this syndrome. Clinicians need alternative targets of therapy, tools for risk profile assessment, and new therapies to address complications early in the course of disease and to personalize therapy to each individual. The complexity of CKD-MBD suggests that incorporating artificial intelligence techniques into the diagnostic, therapeutic, and research armamentarium could accelerate the achievement of these goals.
Collapse
Affiliation(s)
- Eleanor D Lederer
- VA North Texas Health Care Services, Dallas TX, USA
- Department of Medicine and Charles and Jane Pak Center for Mineral Metabolism and Clinical Research, UT Southwestern Medical Center, Dallas, TX, USA
- Department of Medicine, University of Louisville Health Sciences Center, Louisville, KY, USA
| | - Mahmoud M Sobh
- Nephrology and Internal Medicine, Mansoura University, Mansoura, Egypt
| | - Michael E Brier
- Department of Medicine, University of Louisville Health Sciences Center, Louisville, KY, USA
- Robley Rex VA Medical Center, Louisville, KY, USA
| | - Adam E Gaweda
- Department of Medicine, University of Louisville Health Sciences Center, Louisville, KY, USA
- Robley Rex VA Medical Center, Louisville, KY, USA
| |
Collapse
|
5
|
Wang YH, Hsiao CH, Wang JH, Hsu BG. Osteocalcin: A Potential Marker of Peripheral Arterial Stiffness in Hypertensive Patients. MEDICINA (KAUNAS, LITHUANIA) 2024; 60:835. [PMID: 38793018 PMCID: PMC11122903 DOI: 10.3390/medicina60050835] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Revised: 05/10/2024] [Accepted: 05/17/2024] [Indexed: 05/26/2024]
Abstract
Background and Objectives: Brachial-ankle pulse wave velocity (baPWV) is an established independent risk factor for cardiovascular events, cardiovascular mortality, and all-cause mortality. Osteocalcin (OC) is recognized to be associated with vascular function. The present study assessed the correlation between serum OC levels and peripheral arterial stiffness (PAS) measured through baPWV in hypertensive patients. Materials and Methods: Fasting blood samples were collected from 120 hypertensive participants. The serum total OC levels were measured using a commercial enzyme-linked immunosorbent assay kit, whereas the baPWV device was used to detect PAS. The PAS group had left or right baPWV > 18.0 m/s. Results: Among the hypertensive patients, 24 (20.0%) were classified into the PAS group. The PAS group exhibited a significantly older age (p = 0.011), higher prevalence of diabetes (p = 0.010), systolic blood pressure (p = 0.019), levels of serum fasting glucose (p = 0.003), blood urea nitrogen (p = 0.024), creatinine (p = 0.004), C-reactive protein (p = 0.007), OC (p = 0.002), and lower estimated glomerular filtration rate (p = 0.004) than the non-PAS group. Age (odds ratio [OR]: 1.076, 95% CI: 1.004-1.153, p = 0.037) and serum OC level (OR: 1.797, 95% confidence interval (CI): 1.077-3.000, p = 0.025) were independent factors linked to PAS in hypertensive patients in the multivariate logistic regression analysis. Conclusions: Serum OC levels and older age are positively associated with PAS in hypertensive patients.
Collapse
Affiliation(s)
- Yung-Hsuan Wang
- Division of Chest Medicine, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien 97004, Taiwan
| | - Chien-Hao Hsiao
- Division of Cardiology, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien 97004, Taiwan;
| | - Ji-Hung Wang
- Division of Cardiology, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien 97004, Taiwan;
- School of Medicine, Tzu Chi University, Hualien 97004, Taiwan
| | - Bang-Gee Hsu
- School of Medicine, Tzu Chi University, Hualien 97004, Taiwan
- Division of Nephrology, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien 97004, Taiwan
| |
Collapse
|
6
|
Karakose S, Bal Z, Sezer S. The comparison of paricalcitol and calcitriol effects on pulse wave velocity, osteocalcin, and fetuin-A in chronic hemodialysis patients. Semin Dial 2024; 37:131-137. [PMID: 37392044 DOI: 10.1111/sdi.13167] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2023] [Revised: 05/09/2023] [Accepted: 06/10/2023] [Indexed: 07/02/2023]
Abstract
INTRODUCTION Vascular calcification is an intervenable factor in the pathophysiology of cardiovascular disease. Treatment-related factors might worsen the arterial stiffness in chronic hemodialysis patients. The aim of the study is to compare the effects of 1-year treatment with paricalcitol or calcitriol on pulse wave velocity (PWV), which is an indicator of arterial stiffness and osteocalcin and fetuin-A levels. METHODS Seventy-six hemodialysis patients who had similar PWV1 at the beginning were evaluated after a 1-year treatment of paricalcitol or calcitriol. PWV2, serum osteocalcin, and fetuin-A levels were measured at the end of the study. RESULTS At the end of the study, PWV2 of paricalcitol group was statistically lower than the calcitriol group. Osteocalcin levels were statistically lower and fetuin-A levels were statistically higher in the paricalcitol group than the calcitriol group at the end of the study. The number of patients with PWV2 > 7 m/s and using paricalcitol was 16 (39%) but 25 (41%) patients were using calcitriol; the differences were statistically significant. CONCLUSIONS The long-term benefits of paricalcitol were superior to the benefits of calcitriol. Paricalcitol has protective effects from vascular calcification in chronic hemodialysis patients.
Collapse
Affiliation(s)
- Suleyman Karakose
- Nephrology Department, University of Health Sciences, Konya City Hospital, Konya, Turkey
| | - Zeynep Bal
- Nephrology Department, Baskent University Faculty of Medicine, Ankara, Turkey
| | - Siren Sezer
- Nephrology Department, Baskent University Faculty of Medicine, Ankara, Turkey
| |
Collapse
|
7
|
Fan Z, Wei X, Zhu X, Yang K, Tian L, Du Y, Yang L. Correlation between soluble klotho and chronic kidney disease-mineral and bone disorder in chronic kidney disease: a meta-analysis. Sci Rep 2024; 14:4477. [PMID: 38396063 PMCID: PMC10891172 DOI: 10.1038/s41598-024-54812-4] [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: 09/18/2023] [Accepted: 02/16/2024] [Indexed: 02/25/2024] Open
Abstract
We conducted a systematic search across medical databases, including PubMed, Web of Science, EMBASE, and Cochrane Library, up to March 2023. A total of 1944 subjects or individuals from 17 studies were included in our final analysis. The correlation coefficient (r) between sKlotho and calcium was [0.14, (0.02, 0.26)], and a moderate heterogeneity was observed (I2 = 66%, P < 0.05). The correlation coefficient (r) between Klotho and serum phosphate was [- 0.21, (- 0.37, - 0.04)], with apparent heterogeneity (I2 = 84%, P < 0.05). The correlation coefficient (r) between sKlotho and parathyroid hormone and vascular calcification was [- 0.23,(- 0.29, - 0.17); - 0.15, (- 0.23, - 0.08)], with no significant heterogeneity among the studies. (I2 = 40%, P < 0.05; I2 = 30%, P < 0.05). A significant correlation exists between low sKlotho levels and an increased risk of CKD-MBD in patients with CKD. According to the findings, sKlotho may play a role in alleviating CKD-MBD by lowering phosphorus and parathyroid hormone levels, regulating calcium levels, and suppressing vascular calcification. As analysis showed that sKlotho has an important impact on the pathogenesis and progression of CKD-MBD in CKD patients. Nonetheless, further comprehensive and high-quality studies are needed to validate our conclusions.
Collapse
Affiliation(s)
- Zhongyu Fan
- Department of Nephrology, The First Hospital of Jilin University, Changchun, China
| | - Xuejiao Wei
- Department of Nephrology, The First Hospital of Jilin University, Changchun, China
| | - Xiaoyu Zhu
- Department of Nephrology, The First Hospital of Jilin University, Changchun, China
| | - Kun Yang
- Department of Nephrology, The First Hospital of Jilin University, Changchun, China
| | - Ling Tian
- Department of Nephrology, The First Hospital of Jilin University, Changchun, China
| | - Yujun Du
- Department of Nephrology, The First Hospital of Jilin University, Changchun, China.
| | - Liming Yang
- Department of Nephrology, The First Hospital of Jilin University, Changchun, China.
| |
Collapse
|
8
|
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
|
9
|
Ballester-Servera C, Cañes L, Alonso J, Puertas-Umbert L, Vázquez-Sufuentes P, Taurón M, Roselló-Díez E, Marín F, Rodríguez C, Martínez-González J. Upregulation of NOR-1 in calcified human vascular tissues: impact on osteogenic differentiation and calcification. Transl Res 2024; 264:1-14. [PMID: 37690706 DOI: 10.1016/j.trsl.2023.09.004] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/12/2023] [Revised: 09/04/2023] [Accepted: 09/05/2023] [Indexed: 09/12/2023]
Abstract
Cardiovascular calcification is a significant public health issue whose pathophysiology is not fully understood. NOR-1 regulates critical processes in cardiovascular remodeling, but its contribution to ectopic calcification is unknown. NOR-1 was overexpressed in human calcific aortic valves and calcified atherosclerotic lesions colocalizing with RUNX2, a factor essential for osteochondrogenic differentiation and calcification. NOR-1 and osteogenic markers were upregulated in calcifying human valvular interstitial cells (VICs) and human vascular smooth muscle cells (VSMCs). Gain- and loss-of-function approaches demonstrated that NOR-1 negatively modulates the expression of osteogenic genes relevant for the osteogenic transdifferentiation (RUNX2, IL-6, BMP2, and ALPL) and calcification of VICs. VSMCs from transgenic mice overexpressing NOR-1 in these cells (TgNOR-1VSMC) expressed lower basal levels of osteogenic genes (IL-6, BMP2, ALPL, OPN) than cells from WT littermates, and their upregulation by a high-phosphate osteogenic medium (OM) was completely prevented by NOR-1 transgenesis. Consistently, this was associated with a dramatic reduction in the calcification of both transgenic VSMCs and aortic rings from TgNOR-1VSMC mice exposed to OM. Atherosclerosis and calcification were induce in mice by the administration of AAV-PCSK9D374Y and a high-fat/high-cholesterol diet. Challenged-TgNOR-1VSMC mice exhibited decreased vascular expression of osteogenic markers, and both less atherosclerotic burden (assessed in whole aorta and lesion size in aortic arch and brachiocephalic artery) and less vascular calcification (assessed either by near-infrared fluorescence imaging or histological analysis) than WT mice. Our data indicate that NOR-1 negatively modulates the expression of genes critically involved in the osteogenic differentiation of VICs and VSMCs, thereby restraining ectopic cardiovascular calcification.
Collapse
Affiliation(s)
- Carme Ballester-Servera
- Instituto de Investigaciones Biomédicas de Barcelona-Consejo Superior de Investigaciones Científicas (IIBB-CSIC), Barcelona, Spain; CIBER de Enfermedades Cardiovasculares, Instituto de Salud Carlos III, Madrid, Spain; Institut d'Investigació Biomèdica Sant Pau (IIB SANT PAU), Barcelona, Spain
| | - Laia Cañes
- Instituto de Investigaciones Biomédicas de Barcelona-Consejo Superior de Investigaciones Científicas (IIBB-CSIC), Barcelona, Spain; CIBER de Enfermedades Cardiovasculares, Instituto de Salud Carlos III, Madrid, Spain; Institut d'Investigació Biomèdica Sant Pau (IIB SANT PAU), Barcelona, Spain
| | - Judith Alonso
- Instituto de Investigaciones Biomédicas de Barcelona-Consejo Superior de Investigaciones Científicas (IIBB-CSIC), Barcelona, Spain; CIBER de Enfermedades Cardiovasculares, Instituto de Salud Carlos III, Madrid, Spain; Institut d'Investigació Biomèdica Sant Pau (IIB SANT PAU), Barcelona, Spain
| | - Lidia Puertas-Umbert
- CIBER de Enfermedades Cardiovasculares, Instituto de Salud Carlos III, Madrid, Spain; Institut d'Investigació Biomèdica Sant Pau (IIB SANT PAU), Barcelona, Spain; Institut de Recerca Hospital de la Santa Creu i Sant Pau (IRHSCSP), Barcelona, Spain
| | - Paula Vázquez-Sufuentes
- Instituto de Investigaciones Biomédicas de Barcelona-Consejo Superior de Investigaciones Científicas (IIBB-CSIC), Barcelona, Spain; Institut d'Investigació Biomèdica Sant Pau (IIB SANT PAU), Barcelona, Spain
| | - Manel Taurón
- CIBER de Enfermedades Cardiovasculares, Instituto de Salud Carlos III, Madrid, Spain; Institut d'Investigació Biomèdica Sant Pau (IIB SANT PAU), Barcelona, Spain; Departamento de Cirugía Cardíaca, Hospital de la Santa Creu i Sant Pau-Universitat Autònoma de Barcelona (HSCSP-UAB), Barcelona, Spain
| | - Elena Roselló-Díez
- Institut d'Investigació Biomèdica Sant Pau (IIB SANT PAU), Barcelona, Spain; Departamento de Cirugía Cardíaca, Hospital de la Santa Creu i Sant Pau-Universitat Autònoma de Barcelona (HSCSP-UAB), Barcelona, Spain
| | - Francisco Marín
- CIBER de Enfermedades Cardiovasculares, Instituto de Salud Carlos III, Madrid, Spain; Departamento de Cardiología, Hospital Clínico Universitario Virgen de la Arrixaca-Instituto Murciano de Investigación Biosanitaria (IMIB-Arrixaca), Murcia, Spain
| | - Cristina Rodríguez
- CIBER de Enfermedades Cardiovasculares, Instituto de Salud Carlos III, Madrid, Spain; Institut d'Investigació Biomèdica Sant Pau (IIB SANT PAU), Barcelona, Spain; Institut de Recerca Hospital de la Santa Creu i Sant Pau (IRHSCSP), Barcelona, Spain
| | - José Martínez-González
- Instituto de Investigaciones Biomédicas de Barcelona-Consejo Superior de Investigaciones Científicas (IIBB-CSIC), Barcelona, Spain; CIBER de Enfermedades Cardiovasculares, Instituto de Salud Carlos III, Madrid, Spain; Institut d'Investigació Biomèdica Sant Pau (IIB SANT PAU), Barcelona, Spain.
| |
Collapse
|
10
|
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: 4] [Impact Index Per Article: 4.0] [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
|
11
|
Akbari T, Kazemi Fard T, Fadaei R, Rostami R, Moradi N, Movahedi M, Fallah S. Evaluation of MMP-9, IL-6, TNF-α levels and peripheral blood mononuclear cells genes expression of MMP-9 and TIMP-1 in Iranian patients with coronary artery disease. J Cardiovasc Thorac Res 2023; 15:223-230. [PMID: 38357561 PMCID: PMC10862034 DOI: 10.34172/jcvtr.2023.31844] [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: 04/17/2023] [Accepted: 11/27/2023] [Indexed: 02/16/2024] Open
Abstract
Introduction Coronary artery disease (CAD) is the main cause of death and is characterized by atherosclerosis in coronary arteries. Inflammation plays a crucial role in the progression and development of atherosclerosis. Methods The present study consisted of 132 Iranian individuals who underwent coronary angiography, 65 patients with CAD, and 67 controls. The matrix metalloproteinase-9 (MMP-9), TNF-α, IL-6, and vitamin D serum levels were measured by the ELISA technique. The gene expression of MMP-9 and tissue inhibitors of metalloproteinase (TIMP-1) was estimated by real-time PCR assay. Results A considerable increase in levels and PBMC gene expression of MMP-9 and serum levels of IL-6 and TNF-α were found in CAD patients compared with controls. A significant decrease was detected in vitamin D levels of CAD patients in comparison with controls. A considerable direct correlation was found between MMP-9 levels and MMP-9 and TIMP1 gene expression in CAD patients. MMP-9 levels positively correlated with LDL-C in CAD patients. The correlation between TIMP1 gene expression and IL-6 levels was also negatively significant. There were positive correlations between MMP-9 levels with IL-6 and TNF-α serum levels in CAD patients. Conclusion This study showed that the interaction between MMPs, TIMP1, and cytokines could play a role in the pathogenesis of atherosclerosis. The present study suggested that high levels of TNF-α and IL-6 and vitamin D deficiency in our studied patients could disturb the MMP-9/TIMP-1 balance and lipid metabolism, leading to plaque formation/ rupture in predisposed CAD patients.
Collapse
Affiliation(s)
- Tooran Akbari
- Department of Biochemistry, Faculty of Biological Sciences, North Tehran Branch, Islamic Azad University, Tehran, Iran
| | - Toktam Kazemi Fard
- Department of Biochemistry, Faculty of Biological Sciences, North Tehran Branch, Islamic Azad University, Tehran, Iran
| | - Reza Fadaei
- Department of Pharmacology, Vanderbilt University, Nashville, TN, USA
| | - Rahim Rostami
- Department of Clinical Biochemistry, Faculty of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Nariman Moradi
- Department of Clinical Biochemistry, Faculty of Medicine, Kurdistan University of Medical Sciences, Sanandaj, Iran
| | - Monireh Movahedi
- Department of Biochemistry, Faculty of Biological Sciences, North Tehran Branch, Islamic Azad University, Tehran, Iran
| | - Soudabeh Fallah
- Department of Clinical Biochemistry, Faculty of Medicine, Iran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
12
|
Lin RA, Hsieh JT, Huang CC, Yang CY, Lin YP, Tarng DC. Circulating Osteocalcin Fractions are Associated with Vascular Calcification and Mortality in Chronic Hemodialysis Patients. Calcif Tissue Int 2023; 113:416-425. [PMID: 37665403 DOI: 10.1007/s00223-023-01122-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/02/2023] [Accepted: 07/31/2023] [Indexed: 09/05/2023]
Abstract
BACKGROUND Vascular calcification, a component of chronic kidney disease-mineral and bone disorder (CKD-MBD), is prevalent in patients with end-stage kidney disease (ESKD) and contributes to high mortality. However, the association between the blood level of total osteocalcin (OC) and vascular calcification and mortality remains inconclusive. We, therefore, investigated whether different OC fractions can serve as biomarkers of vascular calcification and mortality in the ESKD population. METHODS This observational cohort study enrolled patients on maintenance hemodialysis. Plasma carboxylated OC (cOC), uncarboxylated OC (ucOC), and intact parathyroid hormone (PTH) were measured. The percentage of carboxylated OC (%cOC) was calculated as dividing cOC by total OC. The vascular calcification severity was defined by an aortic calcification grade. The patients were followed for three years and one month. RESULTS A total of 184 patients were enrolled. In the multivariable logistic regression, plasma %cOC, but not cOC or ucOC, was independently associated with the severity of vascular calcification (OR 1.019, p = 0.036). A significant U-shaped correlation was found between plasma %cOC and PTH (p = 0.002). In the multivariable Cox regression, patients with higher plasma %cOC had a higher risk of mortality (quartiles Q4 versus Q1-Q3, HR 1.991 [95% CI: 1.036-3.824], p = 0.039). CONCLUSIONS In patients undergoing chronic hemodialysis, plasma %cOC positively correlated with vascular calcification and exhibited a U-shaped correlation with PTH. Furthermore, a higher plasma %cOC was associated with increased mortality. These findings suggest that plasma %cOC may serve as a biomarker for CKD-MBD and a predictor of clinical outcomes in chronic hemodialysis patients.
Collapse
Affiliation(s)
- Ruei-An Lin
- Faculty of Medicine, School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
- Department of Medical Education, National Taiwan University Hospital, Taipei, Taiwan
| | - Jyh-Tong Hsieh
- Institute of Clinical Medicine, School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
- Division of Nephrology, Department of Medicine, Taipei Veterans General Hospital and Institute of Clinical Medicine, School of Medicine, National Yang Ming Chiao Tung University, 201, Sec. 2, Shih-Pai Rd., Beitou Dist, Taipei, 11217, Taiwan
| | - Chin-Chou Huang
- Faculty of Medicine, School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
- Division of Cardiology, Department of Medicine, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Chih-Yu Yang
- Faculty of Medicine, School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan.
- Institute of Clinical Medicine, School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan.
- Division of Nephrology, Department of Medicine, Taipei Veterans General Hospital, Taipei, Taiwan.
- National Yang Ming Chiao Tung University, Hsinchu, Taiwan.
- Stem Cell Research Center, National Yang Ming Chiao Tung University, Taipei, Taiwan.
| | - Yao-Ping Lin
- Faculty of Medicine, School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
- Division of Nephrology, Department of Medicine, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Der-Cherng Tarng
- Faculty of Medicine, School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
- Institute of Clinical Medicine, School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
- Division of Nephrology, Department of Medicine, Taipei Veterans General Hospital, Taipei, Taiwan
- National Yang Ming Chiao Tung University, Hsinchu, Taiwan
| |
Collapse
|
13
|
Maier JA, Andrés V, Castiglioni S, Giudici A, Lau ES, Nemcsik J, Seta F, Zaninotto P, Catalano M, Hamburg NM. Aging and Vascular Disease: A Multidisciplinary Overview. J Clin Med 2023; 12:5512. [PMID: 37685580 PMCID: PMC10488447 DOI: 10.3390/jcm12175512] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Revised: 08/18/2023] [Accepted: 08/21/2023] [Indexed: 09/10/2023] Open
Abstract
Vascular aging, i.e., the deterioration of the structure and function of the arteries over the life course, predicts cardiovascular events and mortality. Vascular degeneration can be recognized before becoming clinically symptomatic; therefore, its assessment allows the early identification of individuals at risk. This opens the possibility of minimizing disease progression. To review these issues, a search was completed using PubMed, MEDLINE, and Google Scholar from 2000 to date. As a network of clinicians and scientists involved in vascular medicine, we here describe the structural and functional age-dependent alterations of the arteries, the clinical tools for an early diagnosis of vascular aging, and the cellular and molecular events implicated. It emerges that more studies are necessary to identify the best strategy to quantify vascular aging, and to design proper physical activity programs, nutritional and pharmacological strategies, as well as social interventions to prevent, delay, and eventually revert the disease.
Collapse
Affiliation(s)
- Jeanette A. Maier
- Department of Biomedical and Clinical Sciences, Università di Milano, 20157 Milano, Italy;
- VAS-European Independent foundation in Angiology/Vascular Medicine, 20157 Milano, Italy; (M.C.); (N.M.H.)
| | - Vicente Andrés
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), 28029 Madrid, Spain;
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), 28029 Madrid, Spain
| | - Sara Castiglioni
- Department of Biomedical and Clinical Sciences, Università di Milano, 20157 Milano, Italy;
| | - Alessandro Giudici
- Department of Biomedical Engineering, CARIM School for Cardiovascular Diseases, Maastricht University, 6229 ER Maastricht, The Netherlands;
- GROW School for Oncology and Reproduction, Maastricht University, 6229 ER Maastricht, The Netherlands
| | - Emily S. Lau
- Division of Cardiology Massachusetts General Hospital, Boston, MA 02114, USA;
| | - János Nemcsik
- Health Service of Zugló (ZESZ), Department of Family Medicine, Semmelweis University, Stáhly u. 7-9, 1085 Budapest, Hungary;
| | - Francesca Seta
- Vascular Biology Section, Boston University Chobanian & Avedisian School of Medicine, Boston, MA 02118, USA;
| | - Paola Zaninotto
- UCL Research Department of Epidemiology & Public Health, University College London, London WC1E 6BT, UK;
| | - Mariella Catalano
- VAS-European Independent foundation in Angiology/Vascular Medicine, 20157 Milano, Italy; (M.C.); (N.M.H.)
- Inter-University Research Center on Vascular Disease, Università di Milano, 20157 Milano, Italy
| | - Naomi M. Hamburg
- VAS-European Independent foundation in Angiology/Vascular Medicine, 20157 Milano, Italy; (M.C.); (N.M.H.)
- Vascular Biology Section, Boston University Chobanian & Avedisian School of Medicine, Boston, MA 02118, USA;
| |
Collapse
|
14
|
Nesci A, Carnuccio C, Ruggieri V, D'Alessandro A, Di Giorgio A, Santoro L, Gasbarrini A, Santoliquido A, Ponziani FR. Gut Microbiota and Cardiovascular Disease: Evidence on the Metabolic and Inflammatory Background of a Complex Relationship. Int J Mol Sci 2023; 24:ijms24109087. [PMID: 37240434 DOI: 10.3390/ijms24109087] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Revised: 05/09/2023] [Accepted: 05/12/2023] [Indexed: 05/28/2023] Open
Abstract
Several studies in recent years have demonstrated that gut microbiota-host interactions play an important role in human health and disease, including inflammatory and cardiovascular diseases. Dysbiosis has been linked to not only well-known inflammatory diseases, such as inflammatory bowel diseases, rheumatoid arthritis, and systemic lupus erythematous, but also to cardiovascular risk factors, such as atherosclerosis, hypertension, heart failure, chronic kidney disease, obesity, and type 2 diabetes mellitus. The ways the microbiota is involved in modulating cardiovascular risk are multiple and not only related to inflammatory mechanisms. Indeed, human and the gut microbiome cooperate as a metabolically active superorganism, and this affects host physiology through metabolic pathways. In turn, congestion of the splanchnic circulation associated with heart failure, edema of the intestinal wall, and altered function and permeability of the intestinal barrier result in the translocation of bacteria and their products into the systemic circulation, further enhancing the pro-inflammatory conditions underlying cardiovascular disorders. The aim of the present review is to describe the complex interplay between gut microbiota, its metabolites, and the development and evolution of cardiovascular diseases. We also discuss the possible interventions intended to modulate the gut microbiota to reduce cardiovascular risk.
Collapse
Affiliation(s)
- Antonio Nesci
- Angiology and Noninvasive Vascular Diagnostics Unit, Department of Cardiovascular Sciences, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy
| | - Claudia Carnuccio
- Angiology and Noninvasive Vascular Diagnostics Unit, Department of Cardiovascular Sciences, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy
| | - Vittorio Ruggieri
- Angiology and Noninvasive Vascular Diagnostics Unit, Department of Cardiovascular Sciences, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy
| | - Alessia D'Alessandro
- Angiology and Noninvasive Vascular Diagnostics Unit, Department of Cardiovascular Sciences, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy
| | - Angela Di Giorgio
- Angiology and Noninvasive Vascular Diagnostics Unit, Department of Cardiovascular Sciences, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy
| | - Luca Santoro
- Angiology and Noninvasive Vascular Diagnostics Unit, Department of Cardiovascular Sciences, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy
| | - Antonio Gasbarrini
- Digestive Disease Center (CEMAD), Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy
- Department of Translational Medicine and Surgery, Catholic University of the Sacred Heart, 00168 Rome, Italy
| | - Angelo Santoliquido
- Angiology and Noninvasive Vascular Diagnostics Unit, Department of Cardiovascular Sciences, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy
- Department of Translational Medicine and Surgery, Catholic University of the Sacred Heart, 00168 Rome, Italy
| | - Francesca Romana Ponziani
- Digestive Disease Center (CEMAD), Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy
- Department of Translational Medicine and Surgery, Catholic University of the Sacred Heart, 00168 Rome, Italy
| |
Collapse
|
15
|
Nattero-Chávez L, Bayona Cebada A, Fernández-Durán E, Quintero Tobar A, Dorado Avendaño B, Escobar-Morreale H, Luque-Ramírez M. "Arterial stiffness is not associated with changes in the circadian pattern of blood pressure in patients with type 1 diabetes mellitus and cardiovascular autonomic dysfunction". Diab Vasc Dis Res 2023; 20:14791641231173621. [PMID: 37184151 DOI: 10.1177/14791641231173621] [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] [Indexed: 05/16/2023] Open
Abstract
INTRODUCTION Cardiovascular autonomic neuropathy (CAN) associates an abnormal circadian pattern in blood pressure (BP) regulation that might be aggravated by the coexistence of arterial stiffness. We aimed to evaluate the effect of arterial stiffness in the circadian rhythm of BP in patients with type 1 diabetes and CAN. METHODS Cross-sectional study including 56 consecutive patients with type 1 diabetes and CAN, with (n = 28) or without (n = 24) arterial stiffness as defined by an ankle-brachial index above 1.2. CAN was diagnosed by BP and heart rate responses to active standing and cardiovascular autonomic reflex tests. Absence of nocturnal decrease in BP-"non-dipping" pattern- was defined by a daytime to nighttime decrease in mean BP smaller than 10%. RESULTS The study's subjects mean age was 40 ± 11 years-old, their mean duration of diabetes was 22 ± 10 years, and their mean A1c was 7.9 ± 1.5%. A "non-dipping" pattern was observed in 28 patients (54%) regardless of the presence or absence of arterial stiffness. Age, waist circumference, body mass index, and A1c, were introduced as independent variables into a multiple regression analysis. The stepwise model (R2: 0.113, p = 0.016) retained only A1c levels (β: ‒ 0.333, 95% confidence interval [CI]: -3.10 to -0.33) as significant predictor of the percentage of nighttime decrease in mean BP. CONCLUSIONS A non-dipping pattern in BP is very common in patients with type 1 diabetes presenting with subclinical CAN and is associated with a poorer metabolic control. On the contrary, coexistence of arterial stiffness is not associated with abnormalities in circadian BP regulation.
Collapse
Affiliation(s)
- Lía Nattero-Chávez
- Department of Endocrinology and Nutrition, Hospital Universitario Ramón y Cajal, Madrid, Spain
- Grupo de Investigación en Diabetes, Obesidad y Reproducción Humana, Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Instituto de Salud Carlos III, Madrid, Spain
- Universidad de Alcalá, Alcalá de Henares, Madrid, Spain
| | - Ane Bayona Cebada
- Department of Endocrinology and Nutrition, Hospital Universitario Ramón y Cajal, Madrid, Spain
- Grupo de Investigación en Diabetes, Obesidad y Reproducción Humana, Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Instituto de Salud Carlos III, Madrid, Spain
| | - Elena Fernández-Durán
- Grupo de Investigación en Diabetes, Obesidad y Reproducción Humana, Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Instituto de Salud Carlos III, Madrid, Spain
| | - Alejandra Quintero Tobar
- Grupo de Investigación en Diabetes, Obesidad y Reproducción Humana, Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Instituto de Salud Carlos III, Madrid, Spain
| | - Beatriz Dorado Avendaño
- Department of Endocrinology and Nutrition, Hospital Universitario Ramón y Cajal, Madrid, Spain
| | - Héctor Escobar-Morreale
- Department of Endocrinology and Nutrition, Hospital Universitario Ramón y Cajal, Madrid, Spain
- Grupo de Investigación en Diabetes, Obesidad y Reproducción Humana, Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Instituto de Salud Carlos III, Madrid, Spain
- Universidad de Alcalá, Alcalá de Henares, Madrid, Spain
| | - Manuel Luque-Ramírez
- Department of Endocrinology and Nutrition, Hospital Universitario Ramón y Cajal, Madrid, Spain
- Grupo de Investigación en Diabetes, Obesidad y Reproducción Humana, Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Instituto de Salud Carlos III, Madrid, Spain
- Universidad de Alcalá, Alcalá de Henares, Madrid, Spain
| |
Collapse
|
16
|
Lu X, Liu X, Liang E, Yang R, Liu Y, Liu X, Yan F, Xing Y. Panax quinquefolius saponin inhibits vascular smooth muscle cell calcification via activation of nuclear factor-erythroid 2-related factor 2. BMC Complement Med Ther 2023; 23:129. [PMID: 37085826 PMCID: PMC10120105 DOI: 10.1186/s12906-023-03961-6] [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: 12/26/2022] [Accepted: 04/14/2023] [Indexed: 04/23/2023] Open
Abstract
BACKGROUND Panax quinquefolius saponin (PQS) is the main active component of Panax quinquefolius. Emerging evidence suggests that PQS exerts beneficial effects against cardiovascular diseases. However, the role and mechanism of PQS in vascular calcification are not unclear. The present study investigated the effects of PQS on the calcification of vascular smooth muscle cell (VSMCs). METHODS The present study used calcification medium containing 3 mM inorganic phosphate (Pi) to induce rat VSMCs calcification. We investigated the effects of PQS on VSMCs calcification using alizarin red staining and alkaline phosphatase (ALP) activity assays. The intracellular reactive oxygen species (ROS) levels and the transcriptional activity of nuclear factor-erythroid 2-related factor 2 (Nrf2) were determined. The mRNA and protein expression levels of Nrf2, the antioxidant gene heme oxygenase-1 (HO-1), osteogenic markers, including runt-related transcription factor 2 (Runx2) and bone morphogenetic protein 2 (BMP2), and Kelch-like ECH-associated protein 1 (Keap1) were also measured. RESULTS Treatment with Pi significantly increased intracellular calcium deposition and ALP activity, which were suppressed by PQS in a concentration-dependent manner. During VSMCs calcification, PQS inhibited the mRNA and protein expression of Runx2 and BMP2. PQS treatment reduced intracellular ROS production and significantly upregulated Nrf2 transcriptional activity and the expression of Nrf2 and its target antioxidant gene HO-1. PQS suppressed the Pi-induced protein expression of Keap1, which is an endogenous inhibitor of Nrf2. Keap1 siRNA treatment induced Nrf2 expression and downregulated Runx2 expression in the presence of Pi and PQS. CONCLUSION Taken together, these findings suggest that PQS could effectively inhibit VSMCs calcification by ameliorating oxidative stress and regulating osteogenic genes via the promotion of Nrf2 expression.
Collapse
Affiliation(s)
- Xiaoting Lu
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Department of Cardiology, Qilu Hospital of Shandong University, Shandong, 250012, Jinan, China
| | - Xue Liu
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Department of Cardiology, Qilu Hospital of Shandong University, Shandong, 250012, Jinan, China
| | - Ershun Liang
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Department of Cardiology, Qilu Hospital of Shandong University, Shandong, 250012, Jinan, China
| | - Ruixue Yang
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Department of Cardiology, Qilu Hospital of Shandong University, Shandong, 250012, Jinan, China
| | - Yan Liu
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Department of Cardiology, Qilu Hospital of Shandong University, Shandong, 250012, Jinan, China
| | - Xiaoqiong Liu
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Department of Cardiology, Qilu Hospital of Shandong University, Shandong, 250012, Jinan, China
| | - Fangfang Yan
- Department of Traditional Chinese Medicine, Qilu Hospital of Shandong University, Jinan, 250012, Shandong, China
| | - Yifan Xing
- Department of Medical Insurance, Qilu Hospital of Shandong University, Jinan, 250012, Shandong, China.
| |
Collapse
|
17
|
Atteia HH, Alamri ES, Sirag N, Zidan NS, Aljohani RH, Alzahrani S, Arafa MH, Mohammad NS, Asker ME, Zaitone SA, Sakr AT. Soluble guanylate cyclase agonist, isoliquiritigenin attenuates renal damage and aortic calcification in a rat model of chronic kidney failure. Life Sci 2023; 317:121460. [PMID: 36716925 DOI: 10.1016/j.lfs.2023.121460] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2022] [Revised: 01/15/2023] [Accepted: 01/25/2023] [Indexed: 01/30/2023]
Abstract
AIMS Chronic kidney disease (CKD) is a growing fatal health problem worldwide associated with vascular calcification. Therapeutic approaches are limited with higher costs and poor outcomes. Adenine supplementation is one of the most relevant CKD models to human. Insufficient Nitric Oxide (NO)/ cyclic Guanosine Monophosphate (cGMP) signaling plays a key role in rapid development of renal fibrosis. Natural products display proven protection against CKD. Current study therefore explored isoliquiritigenin, a bioflavonoid extracted from licorice roots, potential as a natural activator for soluble Guanylate Cyclase (sGC) in a CKD rat model. MATERIALS AND METHODS 60 male Wistar rats were grouped into Control group (n = 10) and the remaining rats received adenine (200 mg/kg, p.o) for 2 wk to induce CKD. They were equally sub-grouped into: Adenine untreated group and 4 groups orally treated by isoliquiritigenin low or high dose (20 or 40 mg/kg) with/without a selective sGC inhibitor, ODQ (1-H(1,2,4)oxadiazolo(4,3-a)-quinoxalin-1-one, 2 mg/kg, i.p) for 8 wk. KEY FINDINGS Long-term treatment with isoliquiritigenin dose-dependently and effectively amended adenine-induced chronic renal and endothelial dysfunction. It not only alleviated renal fibrosis and apoptosis markers but also aortic calcification. Additionally, this chalcone neutralized renal inflammatory response and oxidative stress. Isoliquiritigenin beneficial effects were associated with up-regulation of serum NO, renal and aortic sGC, cGMP and its dependent protein kinase (PKG). However, co-treatment with ODQ antagonized isoliquiritigenin therapeutic impact. SIGNIFICANCE Isoliquiritigenin seems to exert protective effects against CKD and vascular calcification by activating sGC, increasing cGMP and its downstream PKG.
Collapse
Affiliation(s)
- Hebatallah Husseini Atteia
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, University of Tabuk, Tabuk, Saudi Arabia; Department of Biochemistry, Faculty of Pharmacy, Zagazig University, 44519 Zagazig, Sharkia Gov., Egypt.
| | - Eman Saad Alamri
- Department of Nutrition and Food Science, University of Tabuk, Tabuk, Saudi Arabia
| | - Nizar Sirag
- Department of Natural Products and Alternative Medicine, Faculty of Pharmacy, University of Tabuk, Tabuk, Saudi Arabia
| | - Nahla Salah Zidan
- Department of Nutrition and Food Science, University of Tabuk, Tabuk, Saudi Arabia; Department of Home Economics, Faculty of Specific Education, Kafr ElSheikh University, Kafr ElSheikh, Egypt
| | | | - Sharifa Alzahrani
- Pharmacology Department, Faculty of Medicine, University of Tabuk, Tabuk, Saudi Arabia
| | - Manar Hamed Arafa
- Department of Forensic Medicine and Clinical Toxicology, Faculty of Medicine, Zagazig University, Zagazig, Sharkia Gov., Egypt
| | - Nanies Sameeh Mohammad
- Department of Forensic Medicine and Clinical Toxicology, Faculty of Medicine, Zagazig University, Zagazig, Sharkia Gov., Egypt
| | - Mervat Elsayed Asker
- Department of Biochemistry, Faculty of Pharmacy, Zagazig University, 44519 Zagazig, Sharkia Gov., Egypt
| | - Sawsan A Zaitone
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Suez Canal University, Ismailia, Egypt; Department of Pharmacology and Toxicology, Faculty of Pharmacy, University of Tabuk, Tabuk, Saudi Arabia
| | - Amr Tawfik Sakr
- Department of Biochemistry, Faculty of Pharmacy, University of Sadat City (USC), Menoufia, Egypt
| |
Collapse
|
18
|
Serum OPG and RANKL Levels as Risk Factors for the Development of Cardiovascular Calcifications in End-Stage Renal Disease Patients in Hemodialysis. Life (Basel) 2023; 13:life13020454. [PMID: 36836810 PMCID: PMC9967106 DOI: 10.3390/life13020454] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Revised: 01/19/2023] [Accepted: 01/29/2023] [Indexed: 02/09/2023] Open
Abstract
Cardiovascular calcifications (CVC) are frequently observed in chronic kidney disease (CKD) patients and contribute to their cardiovascular mortality. The aim of the present study was to investigate the impact of osteoprotegerin (OPG)/Receptor Activator of NF-κΒ (RANK)/RANK ligand (RANKL) pathway in the development and evolution of CVCs in hemodialysis patients. In total, 80 hemodialysis patients were assessed for the presence of vascular (abdominal aorta and muscular arteries) calcifications and results were correlated to serum OPG and RANKL levels and the OPG/RANKL ratio. Traditional cardiovascular risk factors and mineral bone disease parameters were also estimated. The presence of VCs was also evaluated 5 years after the initiation of the study, and results were correlated to the initial serum OPG levels. Age, diabetes mellitus, coronary artery disease and OPG levels (p < 0.001) were associated with VCs, whereas RANKL levels were not. Multivariate analysis though revealed that only OPG levels were significantly associated with abdominal aorta calcifications (p = 0.026), but they were not correlated with the progression of VCs. Serum OPG levels are positively and independently associated with VCs in HD patients, but not with their progression. RANKL levels did not show any associations, whereas further studies are needed to establish the significance of OPG/RANKL ratio.
Collapse
|
19
|
Inflammatory, Metabolic, and Coagulation Effects on Medial Arterial Calcification in Patients with Peripheral Arterial Disease. Int J Mol Sci 2023; 24:ijms24043132. [PMID: 36834544 PMCID: PMC9962230 DOI: 10.3390/ijms24043132] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Revised: 02/01/2023] [Accepted: 02/03/2023] [Indexed: 02/09/2023] Open
Abstract
Calcium deposits in the vessel wall in the form of hydroxyapatite can accumulate in the intimal layer, as in atherosclerotic plaque, but also in the medial layer, as in medial arterial calcification (MAC) or medial Möenckeberg sclerosis. Once considered a passive, degenerative process, MAC has recently been shown to be an active process with a complex but tightly regulated pathophysiology. Atherosclerosis and MAC represent distinct clinical entities that correlate in different ways with conventional cardiovascular risk factors. As both entities coexist in the vast majority of patients, it is difficult to estimate the relative contribution of specific risk factors to their development. MAC is strongly associated with age, diabetes mellitus, and chronic kidney disease. Given the complexity of MAC pathophysiology, it is expected that a variety of different factors and signaling pathways may be involved in the development and progression of the disease. In this article, we focus on metabolic factors, primarily hyperphosphatemia and hyperglycemia, and a wide range of possible mechanisms by which they might contribute to the development and progression of MAC. In addition, we provide insight into possible mechanisms by which inflammatory and coagulation factors are involved in vascular calcification processes. A better understanding of the complexity of MAC and the mechanisms involved in its development is essential for the development of potential preventive and therapeutic strategies.
Collapse
|
20
|
Makris K, Mousa C, Cavalier E. Alkaline Phosphatases: Biochemistry, Functions, and Measurement. Calcif Tissue Int 2023; 112:233-242. [PMID: 36571614 DOI: 10.1007/s00223-022-01048-x] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Accepted: 12/01/2022] [Indexed: 12/27/2022]
Abstract
Alkaline phosphatases (ALPs) are a group of isoenzymes, situated on the external layer of the cell membrane; they catalyze the hydrolysis of organic phosphate esters present in the extracellular space. Zinc and magnesium are significant co-factors for the biological activity of these enzymes. Although ALPs are available in various body tissues and have distinct physiochemical properties, they are true isoenzymes since they catalyze a similar reaction. In the liver, ALP is cytosolic and present in the canalicular membrane of the hepatocytes. ALPs are available in placenta, ileal mucosa, kidney, bone, and liver. However, most of the ALPs in serum (over 80%) are delivered from liver and bone and in more modest quantities from the intestines. Despite the fact that alkaline phosphatases are found in numerous tissues all through the body, their exact physiological function remains largely unknown.
Collapse
Affiliation(s)
- Konstantinos Makris
- Clinical Biochemistry Department, KAT General Hospital, Kifissia, Athens, Greece
- Laboratory for the Research of Musculoskeletal System "Th. Garofalidis", School of Medicine, National and Kapodistrian, University of Athens, Athens, Greece
| | - Chagigia Mousa
- 6th Orthopedic Department, KAT General Hospital, Kifissia, Athens, Greece
| | - Etienne Cavalier
- Department of Clinical Chemistry, CIRM, University of Liege, CHU de Liège, CHU Sart-Tilman, B-4000, Liège, Belgium.
| |
Collapse
|
21
|
Kozák E, Bartstra JW, de Jong PA, Mali WPTM, Fülöp K, Tőkési N, Pomozi V, Risseeuw S, Norel JOV, van Leeuwen R, Váradi A, Spiering W. Plasma Level of Pyrophosphate Is Low in Pseudoxanthoma Elasticum Owing to Mutations in the ABCC6 Gene, but It Does Not Correlate with ABCC6 Genotype. J Clin Med 2023; 12:jcm12031047. [PMID: 36769695 PMCID: PMC9917606 DOI: 10.3390/jcm12031047] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2022] [Revised: 01/04/2023] [Accepted: 01/22/2023] [Indexed: 01/31/2023] Open
Abstract
BACKGROUND Pseudoxanthoma elasticum (PXE), a monogenic disorder resulting in calcification affecting the skin, eyes and peripheral arteries, is caused by mutations in the ABCC6 gene, and is associated with low plasma inorganic pyrophosphate (PPi). It is unknown how ABCC6 genotype affects plasma PPi. METHODS We studied the association of ABCC6 genotype (192 patients with biallelic pathogenic ABCC6 mutations) and PPi levels, and its association with the severity of arterial and ophthalmological phenotypes. ABCC6 variants were classified as truncating or non-truncating, and three groups of the 192 patients were formed: those with truncating mutations on both chromosomes (n = 121), those with two non-truncating mutations (n = 10), and a group who had one truncating and one non-truncating ABCC6 mutation (n = 61). The hypothesis formulated before this study was that there was a negative association between PPi level and disease severity. RESULTS Our findings confirm low PPi in PXE compared with healthy controls (0.53 ± 0.15 vs. 1.13 ± 0.29 µM, p < 0.01). The PPi of patients correlated with increasing age (β: 0.05 µM, 95% CI: 0.03-0.06 per 10 years) and was higher in females (0.55 ± 0.17 vs. 0.51 ± 0.13 µM in males, p = 0.03). However, no association between PPi and PXE phenotypes was found. When adjusted for age and sex, no association between PPi and ABCC6 genotype was found. CONCLUSIONS Our data suggest that the relationship between ABCC6 mutations and reduced plasma PPi may not be as direct as previously thought. PPi levels varied widely, even in patients with the same ABCC6 mutations, further suggesting a lack of direct correlation between them, even though the ABCC6 protein-mediated pathway is responsible for ~60% of this metabolite in the circulation. We discuss potential factors that may perturb the expected associations between ABCC6 genotype and PPi and between PPi and disease severity. Our findings support the argument that predictions of pathogenicity made on the basis of mutations (or on the structure of the mutated protein) could be misleading.
Collapse
Affiliation(s)
- Eszter Kozák
- Institute of Enzymology, Research Center for Natural Sciences, Hungarian Academy of Sciences Center of Excellence, 1117 Budapest, Hungary
| | - Jonas W. Bartstra
- Department of Radiology, University Medical Center Utrecht, Utrecht University, 3584 CX Utrecht, The Netherlands
| | - Pim A. de Jong
- Department of Radiology, University Medical Center Utrecht, Utrecht University, 3584 CX Utrecht, The Netherlands
| | - Willem P. T. M. Mali
- Department of Radiology, University Medical Center Utrecht, Utrecht University, 3584 CX Utrecht, The Netherlands
| | - Krisztina Fülöp
- Institute of Enzymology, Research Center for Natural Sciences, Hungarian Academy of Sciences Center of Excellence, 1117 Budapest, Hungary
| | - Natália Tőkési
- Institute of Enzymology, Research Center for Natural Sciences, Hungarian Academy of Sciences Center of Excellence, 1117 Budapest, Hungary
| | - Viola Pomozi
- Institute of Enzymology, Research Center for Natural Sciences, Hungarian Academy of Sciences Center of Excellence, 1117 Budapest, Hungary
| | - Sara Risseeuw
- Department of Ophthalmology, University Medical Center Utrecht, Utrecht University, 3584 CX Utrecht, The Netherlands
| | | | - Redmer van Leeuwen
- Department of Ophthalmology, University Medical Center Utrecht, Utrecht University, 3584 CX Utrecht, The Netherlands
| | - András Váradi
- Institute of Enzymology, Research Center for Natural Sciences, Hungarian Academy of Sciences Center of Excellence, 1117 Budapest, Hungary
| | - Wilko Spiering
- Department of Vascular Medicine, University Medical Center Utrecht, Utrecht University, 3584 CX Utrecht, The Netherlands
- Correspondence: ; Tel.: +31-88-7571188
| |
Collapse
|
22
|
Suh JW, Lee KM, Ko EA, Yoon DS, Park KH, Kim HS, Yook JI, Kim NH, Lee JW. Promoting angiogenesis and diabetic wound healing through delivery of protein transduction domain-BMP2 formulated nanoparticles with hydrogel. J Tissue Eng 2023; 14:20417314231190641. [PMID: 37601810 PMCID: PMC10434183 DOI: 10.1177/20417314231190641] [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: 04/25/2023] [Accepted: 07/12/2023] [Indexed: 08/22/2023] Open
Abstract
Decreased angiogenesis contributes to delayed wound healing in diabetic patients. Recombinant human bone morphogenetic protein-2 (rhBMP2) has also been demonstrated to promote angiogenesis. However, the short half-lives of soluble growth factors, including rhBMP2, limit their use in wound-healing applications. To address this limitation, we propose a novel delivery model using a protein transduction domain (PTD) formulated in a lipid nanoparticle (LNP). We aimed to determine whether a gelatin hydrogel dressing loaded with LNP-formulated PTD-BMP2 (LNP-PTD-BMP2) could enhance the angiogenic function of BMP2 and improve diabetic wound healing. In vitro, compared to the control and rhBMP2, LNP-PTD-BMP2 induced greater tube formation in human umbilical vein endothelial cells and increased the cell recruitment capacity of HaCaT cells. We inflicted large, full-thickness back skin wounds on streptozotocin-induced diabetic mice and applied gelatin hydrogel (GH) cross-linked by microbial transglutaminase containing rhBMP2, LNP-PTD-BMP2, or a control to these wounds. Wounds treated with LNP-PTD-BMP2-loaded GH exhibited enhanced wound closure, increased re-epithelialization rates, and higher collagen deposition than those with other treatments. Moreover, LNP-PTD-BMP2-loaded GH treatment resulted in more CD31- and α-SMA-positive cells, indicating greater neovascularization capacity than rhBMP2-loaded GH or GH treatments alone. Furthermore, in vivo near-infrared fluorescence revealed that LNP-PTD-BMP2 has a longer half-life than rhBMP2 and that BMP2 localizes around wounds. In conclusion, LNP-PTD-BMP2-loaded GH is a viable treatment option for diabetic wounds.
Collapse
Affiliation(s)
- Jae Wan Suh
- Department of Orthopaedic Surgery, Dankook University College of Medicine, Cheonan, South Korea
| | - Kyoung-Mi Lee
- Department of Orthopaedic Surgery, Yonsei University College of Medicine, Seoul, South Korea
| | - Eun Ae Ko
- Department of Orthopaedic Surgery, Yonsei University College of Medicine, Seoul, South Korea
| | - Dong Suk Yoon
- Department of Biomedical Science, Hwasung Medi-Science University, Hwaseong-Si, Gyeonggi-Do, South Korea
| | - Kwang Hwan Park
- Department of Orthopaedic Surgery, Yonsei University College of Medicine, Seoul, South Korea
| | - Hyun Sil Kim
- Department of Oral Pathology, Oral Cancer Research Institute, Yonsei University College of Dentistry, Seoul, South Korea
| | - Jong In Yook
- Department of Oral Pathology, Oral Cancer Research Institute, Yonsei University College of Dentistry, Seoul, South Korea
| | - Nam Hee Kim
- Department of Oral Pathology, Oral Cancer Research Institute, Yonsei University College of Dentistry, Seoul, South Korea
| | - Jin Woo Lee
- Department of Orthopaedic Surgery, Yonsei University College of Medicine, Seoul, South Korea
- Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul, South Korea
| |
Collapse
|
23
|
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: 0] [Impact Index Per Article: 0] [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
|
24
|
GDF11 Is a Novel Protective Factor Against Vascular Calcification. J Cardiovasc Pharmacol 2022; 80:852-860. [PMID: 36027600 DOI: 10.1097/fjc.0000000000001357] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/06/2022] [Accepted: 07/31/2022] [Indexed: 12/13/2022]
Abstract
ABSTRACT Vascular calcification (VC) occurs via an active cell-mediated process, which involves osteogenic differentiation, apoptosis, and phenotypic transformation of vascular smooth muscle cells (VSMCs). As a member of the transforming growth factor-β family, growth differentiation factor 11 (GDF11) can inhibit apoptosis and osteogenic differentiation and maintain the stability of atherosclerotic plaques. In this study, coronary artery calcium score (CACS) of participants with GDF11 measurements was measured using computed tomography angiography and was scored according to the Agatston score. β-glycerophosphate (10 mM), dexamethasone (100 nM), and l -ascorbic acid (50 µg/mL) [osteogenic medium (OM)] were used to induce calcification of human aortic smooth muscle cells. We found that CACS was negatively correlated with serum GDF11 levels in patients and GDF11 was a strong predictor of elevated CACS (OR = 0.967, 95% CI: 0.945-0.991; P = 0.006), followed by age (OR = 1.151, 95% CI: 1.029-1.286; P = 0.014), triglycerides (OR = 4.743, 95% CI: 1.170-19.236; P = 0.029), C-reactive protein (OR = 1.230, 95% CI: 1.010-1.498; P = 0.04), and hypertension (OR = 7.264, 95% CI: 1.099-48.002; P = 0.04). Furthermore, exogenous GDF11 inhibited OM-induced calcification by inhibiting osteogenic differentiation, the phenotypic transformation and apoptosis of human aortic smooth muscle cells. Our study demonstrates that GDF11 plays a crucial role in reducing vascular calcification and serves as a potential intervention target to vascular calcification.
Collapse
|
25
|
Vallée A. Arterial Stiffness and the Canonical WNT/β-catenin Pathway. Curr Hypertens Rep 2022; 24:499-507. [PMID: 35727523 DOI: 10.1007/s11906-022-01211-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/09/2022] [Indexed: 01/31/2023]
Abstract
PURPOSE OF REVIEW Arterial stiffness (AS) was mainly associated with cardiovascular morbidity and mortality in a hypertensive patient. Some risk factors contribute to the development of AS, such as aging, high blood pressure, vascular calcification, inflammation, and diabetes mellitus. The WNT/β-catenin pathway is implicated in numerous signaling and regulating pathways, including embryogenesis, cell proliferation, migration and polarity, apoptosis, and organogenesis. The activation of the WNT/β-catenin pathway is associated with the development of these risk factors. RECENT FINDINGS Aortic pulse wave velocity (PWV) is measured to determine AS, and in peripheral artery disease patients, PWV is higher than controls. An augmentation in PWV by 1 m/s has been shown to increase the risk of cardiovascular events by 14%. AS measured by PWV is characterized by the deregulation of the WNT/β-catenin pathway by the inactivation of its two inhibitors, i.e., DKK1 and sclerostin. Thus, this review focuses on the role of the WNT/β-catenin pathway which contributes to the development of arterial stiffness.
Collapse
Affiliation(s)
- Alexandre Vallée
- Department of Epidemiology - Data - Biostatistics, Delegation of Clinical Research and Innovation, Foch Hospital, 92150, Suresnes, France.
| |
Collapse
|
26
|
Attenuating effect of magnesium on pulmonary arterial calcification in rodent models of pulmonary hypertension. J Hypertens 2022; 40:1979-1993. [PMID: 36052522 DOI: 10.1097/hjh.0000000000003211] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
OBJECTIVE Vascular calcification has been considered as a potential therapeutic target in pulmonary hypertension. Mg2+ has a protective role against calcification. This study aimed to investigate whether Mg2+ could alleviate pulmonary hypertension by reducing medial calcification of pulmonary arteries. METHODS Monocrotaline (MCT)-induced and chronic hypoxia-induced pulmonary hypertension rats were given an oral administration of 10% MgSO4 (10 ml/kg per day). Additionally, we administered Mg2+ in calcified pulmonary artery smooth muscle cells (PASMCs) after incubating with β-glycerophosphate (β-GP, 10 mmol/l). RESULTS In vivo, MCT-induced and chronic hypoxia-induced pulmonary hypertension indexes, including right ventricular systolic pressure, right ventricular mass index, and arterial wall thickness, as well as Alizarin Red S (ARS) staining-visualized calcium deposition, high calcium levels, and osteochondrogenic differentiation in pulmonary arteries, were mitigated by dietary Mg2+ intake. In vitro, β-GP-induced calcium-rich deposits stained by ARS, calcium content, as well as the detrimental effects of calcification to proliferation, migration, and resistance to apoptosis of PASMCs were alleviated by high Mg2+ but exacerbated by low Mg2+. Expression levels of mRNA and protein of β-GP-induced osteochondrogenic markers, RUNX Family Transcription Factor 2, and Msh Homeobox 2 were decreased by high Mg2+ but increased by low Mg2+; however, Mg2+ did not affect β-GP-induced expression of SRY-Box Transcription Factor 9. Moreover, mRNA expression and protein levels of β-GP-reduced calcification inhibitor, Matrix GLA protein was increased by high Mg2+ but decreased by low Mg2+. CONCLUSION Mg2+ supplement is a powerful strategy to treat pulmonary hypertension by mitigating pulmonary arterial calcification as the calcification triggered physiological and pathological changes to PASMCs.
Collapse
|
27
|
Laurain A, Rubera I, Razzouk-Cadet M, Bonnafous S, Albuquerque M, Paradis V, Patouraux S, Duranton C, Lesaux O, Lefthériotis G, Tran A, Anty R, Gual P, Iannelli A, Favre G. Arterial Calcifications in Patients with Liver Cirrhosis Are Linked to Hepatic Deficiency of Pyrophosphate Production Restored by Liver Transplantation. Biomedicines 2022; 10:1496. [PMID: 35884801 PMCID: PMC9312703 DOI: 10.3390/biomedicines10071496] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Revised: 06/11/2022] [Accepted: 06/14/2022] [Indexed: 11/24/2022] Open
Abstract
Liver fibrosis is associated with arterial calcification (AC). Since the liver is a source of inorganic pyrophosphate (PPi), an anti-calcifying compound, we investigated the relationship between plasma PPi ([PPi]pl), liver fibrosis, liver function, AC, and the hepatic expression of genes regulating PPi homeostasis. To that aim, we compared [PPi]pl before liver transplantation (LT) and 3 months after LT. We also assessed the expression of four key regulators of PPi in liver tissues and established correlations between AC, and scores of liver fibrosis and liver failure in these patients. LT candidates with various liver diseases were included. AC scores were assessed in coronary arteries, abdominal aorta, and aortic valves. Liver fibrosis was evaluated on liver biopsies and from non-invasive tests (FIB-4 and APRI scores). Liver functions were assessed by measuring serum albumin, ALBI, MELD, and Pugh−Child scores. An enzymatic assay was used to dose [PPi]pl. A group of patients without liver alterations from a previous cohort provided a control group. Gene expression assays were performed with mRNA extracted from liver biopsies and compared between LT recipients and the control individuals. [PPi]pl negatively correlated with APRI (r = −0.57, p = 0.001, n = 29) and FIB-4 (r = −0.47, p = 0.006, n = 29) but not with interstitial fibrosis index from liver biopsies (r = 0.07, p = 0.40, n = 16). Serum albumin positively correlated with [PPi]pl (r = 0.71; p < 0.0001, n = 20). ALBI, MELD, and Pugh−Child scores correlated negatively with [PPi]pl (r = −0.60, p = 0.0005; r = −0.56, p = 0.002; r = −0.41, p = 0.02, respectively, with n = 20). Liver fibrosis assessed on liver biopsies by FIB-4 and by APRI positively correlated with coronary AC (r = 0.51, p = 0.02, n = 16; r = 0.58, p = 0.009, n = 20; r = 0.41, p = 0.04, n = 20, respectively) and with abdominal aorta AC (r = 0.50, p = 0.02, n = 16; r = 0.67, p = 0.002, n = 20; r = 0.61, p = 0.04, n = 20, respectively). FIB-4 also positively correlated with aortic valve calcification (r = 0.40, p = 0.046, n = 20). The key regulator genes of PPi production in liver were lower in patients undergoing liver transplantation as compared to controls. Three months after surgery, serum albumin levels were restored to physiological levels (40 [37−44] vs. 35 [30−40], p = 0.009) and [PPi]pl was normalized (1.40 [1.07−1.86] vs. 0.68 [0.53−0.80] µmol/L, p = 0.0005, n = 12). Liver failure and/or fibrosis correlated with AC in several arterial beds and were associated with low plasma PPi and dysregulation of key proteins involved in PPi homeostasis. Liver transplantation normalized these parameters.
Collapse
Affiliation(s)
- Audrey Laurain
- Department of Nephrology, Pasteur 1 University Hospital, 06001 Nice, France;
- Faculty of Medicine, Tour Pasteur, 28 Avenue de Valombrose, University of Côte d’Azur, 06000 Nice, France; (I.R.); (S.B.); (S.P.); (C.D.); (G.L.); (A.T.); (R.A.); (P.G.); (A.I.)
- LP2M CNRS UMR 7370, Tour Pasteur, 28 Avenue de Valombrose, 06000 Nice, France
| | - Isabelle Rubera
- Faculty of Medicine, Tour Pasteur, 28 Avenue de Valombrose, University of Côte d’Azur, 06000 Nice, France; (I.R.); (S.B.); (S.P.); (C.D.); (G.L.); (A.T.); (R.A.); (P.G.); (A.I.)
- LP2M CNRS UMR 7370, Tour Pasteur, 28 Avenue de Valombrose, 06000 Nice, France
| | | | - Stéphanie Bonnafous
- Faculty of Medicine, Tour Pasteur, 28 Avenue de Valombrose, University of Côte d’Azur, 06000 Nice, France; (I.R.); (S.B.); (S.P.); (C.D.); (G.L.); (A.T.); (R.A.); (P.G.); (A.I.)
- Team 8 “Chronic Liver Diseases Associated with Obesity and Alcohol” Inserm, U1065, Centre Méditerranéen de Médecine Moléculaire (C3M) Bâtiment Universitaire ARCHIMED? 151 Route Saint Antoine de Ginestière BP 2 3194, 06204 Nice, France
- Digestive Unit, Archet 2 University Hospital, 06200 Nice, France
| | - Miguel Albuquerque
- Pathology Department, Beaujon University Hospital, AP-HP, 92110 Clichy, France; (M.A.); (V.P.)
- Inserm U1149, Beaujon University Hospital, 92110 Clichy, France
| | - Valérie Paradis
- Pathology Department, Beaujon University Hospital, AP-HP, 92110 Clichy, France; (M.A.); (V.P.)
- Inserm U1149, Beaujon University Hospital, 92110 Clichy, France
| | - Stéphanie Patouraux
- Faculty of Medicine, Tour Pasteur, 28 Avenue de Valombrose, University of Côte d’Azur, 06000 Nice, France; (I.R.); (S.B.); (S.P.); (C.D.); (G.L.); (A.T.); (R.A.); (P.G.); (A.I.)
- Pathology Department, Pasteur 1 University Hospital, 06000 Nice, France
| | - Christophe Duranton
- Faculty of Medicine, Tour Pasteur, 28 Avenue de Valombrose, University of Côte d’Azur, 06000 Nice, France; (I.R.); (S.B.); (S.P.); (C.D.); (G.L.); (A.T.); (R.A.); (P.G.); (A.I.)
- LP2M CNRS UMR 7370, Tour Pasteur, 28 Avenue de Valombrose, 06000 Nice, France
| | - Olivier Lesaux
- Department Cell and Molecular Biology, John A. Burns School of Medicine, University of Hawaii, Honolulu, HI 96813-5534, USA;
| | - Georges Lefthériotis
- Faculty of Medicine, Tour Pasteur, 28 Avenue de Valombrose, University of Côte d’Azur, 06000 Nice, France; (I.R.); (S.B.); (S.P.); (C.D.); (G.L.); (A.T.); (R.A.); (P.G.); (A.I.)
- LP2M CNRS UMR 7370, Tour Pasteur, 28 Avenue de Valombrose, 06000 Nice, France
- Department of Vascular Medicine and Surgery, Pasteur 1 University Hospital, 06000 Nice, France
| | - Albert Tran
- Faculty of Medicine, Tour Pasteur, 28 Avenue de Valombrose, University of Côte d’Azur, 06000 Nice, France; (I.R.); (S.B.); (S.P.); (C.D.); (G.L.); (A.T.); (R.A.); (P.G.); (A.I.)
- Team 8 “Chronic Liver Diseases Associated with Obesity and Alcohol” Inserm, U1065, Centre Méditerranéen de Médecine Moléculaire (C3M) Bâtiment Universitaire ARCHIMED? 151 Route Saint Antoine de Ginestière BP 2 3194, 06204 Nice, France
- Digestive Unit, Archet 2 University Hospital, 06200 Nice, France
| | - Rodolphe Anty
- Faculty of Medicine, Tour Pasteur, 28 Avenue de Valombrose, University of Côte d’Azur, 06000 Nice, France; (I.R.); (S.B.); (S.P.); (C.D.); (G.L.); (A.T.); (R.A.); (P.G.); (A.I.)
- Team 8 “Chronic Liver Diseases Associated with Obesity and Alcohol” Inserm, U1065, Centre Méditerranéen de Médecine Moléculaire (C3M) Bâtiment Universitaire ARCHIMED? 151 Route Saint Antoine de Ginestière BP 2 3194, 06204 Nice, France
- Digestive Unit, Archet 2 University Hospital, 06200 Nice, France
| | - Philippe Gual
- Faculty of Medicine, Tour Pasteur, 28 Avenue de Valombrose, University of Côte d’Azur, 06000 Nice, France; (I.R.); (S.B.); (S.P.); (C.D.); (G.L.); (A.T.); (R.A.); (P.G.); (A.I.)
- Team 8 “Chronic Liver Diseases Associated with Obesity and Alcohol” Inserm, U1065, Centre Méditerranéen de Médecine Moléculaire (C3M) Bâtiment Universitaire ARCHIMED? 151 Route Saint Antoine de Ginestière BP 2 3194, 06204 Nice, France
| | - Antonio Iannelli
- Faculty of Medicine, Tour Pasteur, 28 Avenue de Valombrose, University of Côte d’Azur, 06000 Nice, France; (I.R.); (S.B.); (S.P.); (C.D.); (G.L.); (A.T.); (R.A.); (P.G.); (A.I.)
- Team 8 “Chronic Liver Diseases Associated with Obesity and Alcohol” Inserm, U1065, Centre Méditerranéen de Médecine Moléculaire (C3M) Bâtiment Universitaire ARCHIMED? 151 Route Saint Antoine de Ginestière BP 2 3194, 06204 Nice, France
- Digestive Unit, Archet 2 University Hospital, 06200 Nice, France
| | - Guillaume Favre
- Department of Nephrology, Pasteur 1 University Hospital, 06001 Nice, France;
- Faculty of Medicine, Tour Pasteur, 28 Avenue de Valombrose, University of Côte d’Azur, 06000 Nice, France; (I.R.); (S.B.); (S.P.); (C.D.); (G.L.); (A.T.); (R.A.); (P.G.); (A.I.)
- LP2M CNRS UMR 7370, Tour Pasteur, 28 Avenue de Valombrose, 06000 Nice, France
| |
Collapse
|
28
|
Sanabria-de la Torre R, García-Fontana C, González-Salvatierra S, Andújar-Vera F, Martínez-Heredia L, García-Fontana B, Muñoz-Torres M. The Contribution of Wnt Signaling to Vascular Complications in Type 2 Diabetes Mellitus. Int J Mol Sci 2022; 23:6995. [PMID: 35805996 PMCID: PMC9266892 DOI: 10.3390/ijms23136995] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Revised: 06/21/2022] [Accepted: 06/21/2022] [Indexed: 02/01/2023] Open
Abstract
Vascular complications are the leading cause of morbidity and mortality among patients with type 2 diabetes mellitus (T2DM). These vascular abnormalities result in a chronic hyperglycemic state, which influences many signaling molecular pathways that initially lead to increased oxidative stress, increased inflammation, and endothelial dysfunction, leading to both microvascular and macrovascular complications. Endothelial dysfunction represents the initial stage in both types of vascular complications; it represents "mandatory damage" in the development of microvascular complications and only "introductory damage" in the development of macrovascular complications. Increasing scientific evidence has revealed an important role of the Wnt pathway in the pathophysiology of the vascular wall. It is well known that the Wnt pathway is altered in patients with T2DM. This review aims to be an update of the current literature related to the Wnt pathway molecules that are altered in patients with T2DM, which may also be the cause of damage to the vasculature. Both microvascular complications (retinopathy, nephropathy, and neuropathy) and macrovascular complications (coronary artery disease, cerebrovascular disease, and peripheral arterial disease) are analyzed. This review aims to concisely concentrate all the evidence to facilitate the view on the vascular involvement of the Wnt pathway and its components by highlighting the importance of exploring possible therapeutic strategy for patients with T2DM who develop vascular pathologies.
Collapse
Affiliation(s)
- Raquel Sanabria-de la Torre
- Department of Medicine, University of Granada, 18016 Granada, Spain; (R.S.-d.l.T.); (S.G.-S.); (L.M.-H.); (M.M.-T.)
- Instituto de Investigación Biosanitaria de Granada (ibs.GRANADA), 18012 Granada, Spain;
| | - Cristina García-Fontana
- Instituto de Investigación Biosanitaria de Granada (ibs.GRANADA), 18012 Granada, Spain;
- Endocrinology and Nutrition Unit, University Hospital Clínico San Cecilio, 18016 Granada, Spain
- CIBER de Fragilidad y Envejecimiento Saludable (CIBERFES), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Sheila González-Salvatierra
- Department of Medicine, University of Granada, 18016 Granada, Spain; (R.S.-d.l.T.); (S.G.-S.); (L.M.-H.); (M.M.-T.)
- Instituto de Investigación Biosanitaria de Granada (ibs.GRANADA), 18012 Granada, Spain;
| | - Francisco Andújar-Vera
- Instituto de Investigación Biosanitaria de Granada (ibs.GRANADA), 18012 Granada, Spain;
- Department of Computer Science and Artificial Intelligence, University of Granada, 18071 Granada, Spain
- Andalusian Research Institute in Data Science and Computational Intelligence (DaSCI Institute), 18014 Granada, Spain
| | - Luis Martínez-Heredia
- Department of Medicine, University of Granada, 18016 Granada, Spain; (R.S.-d.l.T.); (S.G.-S.); (L.M.-H.); (M.M.-T.)
- Instituto de Investigación Biosanitaria de Granada (ibs.GRANADA), 18012 Granada, Spain;
| | - Beatriz García-Fontana
- Instituto de Investigación Biosanitaria de Granada (ibs.GRANADA), 18012 Granada, Spain;
- Endocrinology and Nutrition Unit, University Hospital Clínico San Cecilio, 18016 Granada, Spain
- CIBER de Fragilidad y Envejecimiento Saludable (CIBERFES), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Manuel Muñoz-Torres
- Department of Medicine, University of Granada, 18016 Granada, Spain; (R.S.-d.l.T.); (S.G.-S.); (L.M.-H.); (M.M.-T.)
- Instituto de Investigación Biosanitaria de Granada (ibs.GRANADA), 18012 Granada, Spain;
- Endocrinology and Nutrition Unit, University Hospital Clínico San Cecilio, 18016 Granada, Spain
- CIBER de Fragilidad y Envejecimiento Saludable (CIBERFES), Instituto de Salud Carlos III, 28029 Madrid, Spain
| |
Collapse
|
29
|
Hoogervorst R, van Overhagen H, de Jong PA, Spiering W, de Borst GJ, Veger HTC, Mairuhu ATA, Mali WPTM. Treatment of arterial calcification in patients with chronic limb threatening ischemia with etidronate: protocol of an investigator-initiated multicenter, double blind, placebo-controlled, randomized clinical trial. CVIR Endovasc 2022; 5:26. [PMID: 35666322 PMCID: PMC9170866 DOI: 10.1186/s42155-022-00298-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Accepted: 04/08/2022] [Indexed: 11/29/2022] Open
Abstract
Background Pathologic studies have shown that in patients with critical limb threatening ischaemia (CLTI) medial arterial calcifications are frequently found and may be responsible for aggravating the disease. These extensive calcifitcations are found not only in arteries of the leg but also in the coronary arteries and the aorta. The progression of these calcifications is fast and they stiffen the vessel wall and may thus increase the cardiovascular risk. Reduction of progression of calcification may not only reduce the burden of CLTI but may also reduce the high residual cardiovascular risk. Medial calcifications have been halted by etidronate in other trials. Its potential to reduce the burden from peripheral vascular disease in CLTI and residual cardiovascular risk remains to be established. Methods This is an investigator-initiated multicenter, double blind, placebo-controlled, randomized trial comparing the effects of etidronate versus placebo in patients with CLTI. Subjects will be randomized to either treatment with etidronate for 12 months (cyclical 20 mg/kg for 2 weeks on and 10 weeks off) orally or placebo for 12 months (in a similar routine). The primary endpoint is the change in arterial calcification as quantified by CT-scan. Secondary endpoints are the number of amputations above and below the ankle, mortality, number of vascular interventions and quality of life. Discussion Up to now, the inert end stage of vascular disease in patients with CLTI, has been considered calcification of vessel walls. We believe there is reason to reverse causation and hypothesize that calcification causes vascular disease. This reversal can be proven in a clinical trial if halting the calcification process improves the outcome of the patient. Therefore we use etidronate, a bisphosphate that has proven to stop the calcification in several rare monogenetic calcifying diseases. We aim to perform this mechanistic proof-of-concept study hopefully leading to a clinical outcome study later on.
Collapse
Affiliation(s)
- R Hoogervorst
- Haga Hospital, HagaZiekenhuis, The Hague, The Netherlands.
| | | | - P A de Jong
- Haga Hospital, HagaZiekenhuis, The Hague, The Netherlands.,UMC Utrecht, The Hague, The Netherlands
| | - W Spiering
- Haga Hospital, HagaZiekenhuis, The Hague, The Netherlands.,UMC Utrecht, The Hague, The Netherlands
| | - G J de Borst
- Haga Hospital, HagaZiekenhuis, The Hague, The Netherlands.,UMC Utrecht, The Hague, The Netherlands
| | - H T C Veger
- Haga Hospital, HagaZiekenhuis, The Hague, The Netherlands
| | - A T A Mairuhu
- Haga Hospital, HagaZiekenhuis, The Hague, The Netherlands
| | - W P T M Mali
- Haga Hospital, HagaZiekenhuis, The Hague, The Netherlands.,UMC Utrecht, The Hague, The Netherlands
| |
Collapse
|
30
|
Ren SC, Mao N, Yi S, Ma X, Zou JQ, Tang X, Fan JM. Vascular Calcification in Chronic Kidney Disease: An Update and Perspective. Aging Dis 2022; 13:673-697. [PMID: 35656113 PMCID: PMC9116919 DOI: 10.14336/ad.2021.1024] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Accepted: 10/24/2021] [Indexed: 12/13/2022] Open
Abstract
Chronic kidney disease is a devastating condition resulting from irreversible loss of nephron numbers and function and leading to end-stage renal disease and mineral disorders. Vascular calcification, an ectopic deposition of calcium-phosphate salts in blood vessel walls and heart valves, is an independent risk factor of cardiovascular morbidity and mortality in chronic kidney disease. Moreover, aging and related metabolic disorders are essential risk factors for chronic kidney disease and vascular calcification. Marked progress has been recently made in understanding and treating vascular calcification in chronic kidney disease. However, there is a paucity of systematic reviews summarizing this progress, and investigating unresolved issues is warranted. In this systematic review, we aimed to overview the underlying mechanisms of vascular calcification in chronic kidney diseases and discuss the impact of chronic kidney disease on the pathophysiology of vascular calcification. Additionally, we summarized potential clinical diagnostic biomarkers and therapeutic applications for vascular calcification with chronic kidney disease. This review may offer new insights into the pathogenesis, diagnosis, and therapeutic intervention of vascular calcification.
Collapse
Affiliation(s)
- Si-Chong Ren
- Chengdu Medical College, Chengdu, China.
- Department of Nephrology, First Affiliated Hospital of Chengdu Medical College, Chengdu, China.
- Center for Translational Medicine, Sichuan Academy of Traditional Chinese Medicine, Chengdu, China.
| | - Nan Mao
- Chengdu Medical College, Chengdu, China.
- Department of Nephrology, First Affiliated Hospital of Chengdu Medical College, Chengdu, China.
| | - Si Yi
- Chengdu Medical College, Chengdu, China.
- Clinical Research Center for Geriatrics of Sichuan Province, Chengdu, China.
| | - Xin Ma
- Chengdu Medical College, Chengdu, China.
- Department of Nephrology, First Affiliated Hospital of Chengdu Medical College, Chengdu, China.
| | - Jia-Qiong Zou
- Chengdu Medical College, Chengdu, China.
- Department of Nephrology, First Affiliated Hospital of Chengdu Medical College, Chengdu, China.
| | - Xiaoqiang Tang
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of Ministry of Education, State Key Laboratory of Biotherapy, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Jun-Ming Fan
- Chengdu Medical College, Chengdu, China.
- Clinical Research Center for Geriatrics of Sichuan Province, Chengdu, China.
| |
Collapse
|
31
|
Sulistyowati E, Hsu JH, Lee SJ, Huang SE, Sihotang WY, Wu BN, Dai ZK, Lin MC, Yeh JL. Potential Actions of Baicalein for Preventing Vascular Calcification of Smooth Muscle Cells In Vitro and In Vivo. Int J Mol Sci 2022; 23:ijms23105673. [PMID: 35628483 PMCID: PMC9143966 DOI: 10.3390/ijms23105673] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2022] [Revised: 05/12/2022] [Accepted: 05/14/2022] [Indexed: 12/16/2022] Open
Abstract
Vascular calcification (VC) is associated with cardiovascular disease. Baicalein, a natural flavonoid extract of Scutellaria baicalensis rhizome has several biological properties which may inhibit VC. We investigated whether baicalein suppresses Runt-related transcription factor 2 (Runx2) and bone morphogenetic protein 2 (BMP-2) and upregulates smooth muscle 22-alpha (SM22-α) and alpha-smooth muscle actin (α-SMA). In an in vitro experiment, primary rat aortic vascular smooth muscle cells (VSMCs) were pretreated with 0.1, 1, and 5 μM baicalein, followed by β-glycerophosphate (β-GP) to induce calcification. In an in vivo experiment, VC was generated by vitamin D3 plus nicotine (VDN) administration to male Sprague Dawley (SD) rats randomly assigned into a control group, a VC group, a VC group pretreated with baicalein, and a baicalein alone group. Each group comprised 10 rats. Left ventricular (LV) morphology, function and performance were assessed by echocardiography. Calcium content was measured by Alizarin red S staining and alkaline phosphatase (ALP) activity assays. Apoptotic VSMCs were detected by flow cytometry. Protein levels and superoxide changes were evaluated using Western blotting and immunofluorescence assays respectively. Plasma malondialdehyde (MDA) was assayed. Baicalein pretreatment significantly reduced calcium content in calcified VSMCs (p < 0.001) as well as in VC rat aortic smooth muscle (p < 0.001). Additionally, ALP activity was decreased in calcified VSMCs and VC rat aortic smooth muscle (p < 0.001). Apoptosis was significantly attenuated by 1 μM baicalein pretreatment in calcified VSMCs. Runx2 and BMP-2 expressions were downregulated by the baicalein in calcified VSMCs. Baicalein pretreatment increased typical VSMCs markers SM22-α and α-SMA in calcified VSMCs. Baicalein pretreatment was associated with adverse changes in LV morphometry. Markers of oxidative stress declined, and endogenous antioxidants increased in VC rats pretreated with baicalein. Baicalein mitigates VC through the inhibition of Runx2/BMP-2 signaling pathways, enhancement of vascular contractile phenotype and oxidative stress reduction. However, our study is of basic experimental design; more advanced investigations to identify other molecular regulators of VC and their mechanisms of action is required.
Collapse
Affiliation(s)
- Erna Sulistyowati
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan; (E.S.); (J.-H.H.); (S.-J.L.); (S.-E.H.); (W.Y.S.); (B.-N.W.); (Z.-K.D.)
- Faculty of Medicine, University of Islam Malang, Malang 65145, Indonesia
| | - Jong-Hau Hsu
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan; (E.S.); (J.-H.H.); (S.-J.L.); (S.-E.H.); (W.Y.S.); (B.-N.W.); (Z.-K.D.)
- Department of Pediatrics, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan
- Department of Pediatrics, Kaohsiung Medical University Hospital, Kaohsiung 807, Taiwan
| | - Szu-Jung Lee
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan; (E.S.); (J.-H.H.); (S.-J.L.); (S.-E.H.); (W.Y.S.); (B.-N.W.); (Z.-K.D.)
| | - Shang-En Huang
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan; (E.S.); (J.-H.H.); (S.-J.L.); (S.-E.H.); (W.Y.S.); (B.-N.W.); (Z.-K.D.)
| | - Widya Yanti Sihotang
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan; (E.S.); (J.-H.H.); (S.-J.L.); (S.-E.H.); (W.Y.S.); (B.-N.W.); (Z.-K.D.)
- Faculty of Public Health, Prima University of Indonesia, Medan 20118, Indonesia
| | - Bin-Nan Wu
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan; (E.S.); (J.-H.H.); (S.-J.L.); (S.-E.H.); (W.Y.S.); (B.-N.W.); (Z.-K.D.)
- Department of Pharmacology, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan
- Department of Medical Research, Kaohsiung Medical University Hospital, Kaohsiung 807, Taiwan
| | - Zen-Kong Dai
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan; (E.S.); (J.-H.H.); (S.-J.L.); (S.-E.H.); (W.Y.S.); (B.-N.W.); (Z.-K.D.)
- Department of Pediatrics, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan
- Department of Pediatrics, Kaohsiung Medical University Hospital, Kaohsiung 807, Taiwan
| | - Ming-Chung Lin
- Department of Anesthesiology, Chi Mei Medical Center, Tainan 710, Taiwan
- Correspondence: (M.-C.L.); (J.-L.Y.); Tel.: +886-7-3121101 (ext. 2139) (J.-L.Y.)
| | - Jwu-Lai Yeh
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan; (E.S.); (J.-H.H.); (S.-J.L.); (S.-E.H.); (W.Y.S.); (B.-N.W.); (Z.-K.D.)
- Department of Pharmacology, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan
- Department of Medical Research, Kaohsiung Medical University Hospital, Kaohsiung 807, Taiwan
- Department of Marine Biotechnology and Resources, National Sun Yat-sen University, Kaohsiung 804, Taiwan
- Correspondence: (M.-C.L.); (J.-L.Y.); Tel.: +886-7-3121101 (ext. 2139) (J.-L.Y.)
| |
Collapse
|
32
|
Niu Z, Su G, Li T, Yu H, Shen Y, Zhang D, Liu X. Vascular Calcification: New Insights Into BMP Type I Receptor A. Front Pharmacol 2022; 13:887253. [PMID: 35462911 PMCID: PMC9019578 DOI: 10.3389/fphar.2022.887253] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Accepted: 03/21/2022] [Indexed: 11/13/2022] Open
Abstract
Vascular calcification (VC) is a complex ectopic calcification process and an important indicator of increased risk for diabetes, atherosclerosis, chronic kidney disease, and other diseases. Therefore, clarifying the pathogenesis of VC is of great clinical significance. Numerous studies have shown that the onset and progression of VC are similar to bone formation. Members of the bone morphogenetic protein (BMP) family of proteins are considered key molecules in the progression of vascular calcification. BMP type I receptor A (BMPR1A) is a key receptor of BMP factors acting on the cell membrane, is widely expressed in various tissues and cells, and is an important “portal” for BMP to enter cells and exert their biological effect. In recent years, many discoveries have been made regarding the occurrence and treatment of ectopic ossification-related diseases involving BMP signaling targets. Studies have confirmed that BMPR1A is involved in osteogenic differentiation and that its high expression in vascular endothelial cells and smooth muscle cells can lead to vascular calcification. This article reviews the role of BMPR1A in vascular calcification and the possible underlying molecular mechanisms to provide clues for the clinical treatment of such diseases.
Collapse
Affiliation(s)
- Zhixing Niu
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Guanyue Su
- West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, China
| | - Tiantian Li
- West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, China
| | - Hongchi Yu
- West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, China
| | - Yang Shen
- West China School of Basic Medical Sciences & 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 & Forensic Medicine, Sichuan University, Chengdu, China
- *Correspondence: Demao Zhang, ; Xiaoheng Liu,
| | - Xiaoheng Liu
- West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, China
- *Correspondence: Demao Zhang, ; Xiaoheng Liu,
| |
Collapse
|
33
|
Peride I, Tiglis M, Neagu TP, Niculae A, Checherita IA. Magnesium—A More Important Role in CKD–MBD than We Thought. Diagnostics (Basel) 2022; 12:diagnostics12040880. [PMID: 35453928 PMCID: PMC9031465 DOI: 10.3390/diagnostics12040880] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2022] [Revised: 03/13/2022] [Accepted: 03/30/2022] [Indexed: 12/17/2022] Open
Abstract
Chronic kidney disease (CKD) is associated with different complications, including chronic kidney disease–mineral and bone disorder (CKD–MBD), which represents a systemic disorder that involves the presence of different mineral or bone structure abnormalities (i.e., modification of bone turnover, strength, volume, etc.), including even vascular calcification development. Even if, over the years, different pathophysiological theories have been developed to explain the onset and progression of CKD–MBD, the influence and importance of serum magnesium level on the evolution of CKD have only recently been highlighted. So far, data are inconclusive and conflicting; therefore, further studies are necessary to validate these findings, which could be useful in developing a better, more adequate, and personalized management of CKD patients.
Collapse
Affiliation(s)
- Ileana Peride
- Clinical Department No. 3, “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania;
- Correspondence: (I.P.); (A.N.)
| | - Mirela Tiglis
- Clinical Department No. 14, “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania;
| | - Tiberiu Paul Neagu
- Clinical Department No. 11, “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania;
| | - Andrei Niculae
- Clinical Department No. 3, “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania;
- Correspondence: (I.P.); (A.N.)
| | - Ionel Alexandru Checherita
- Clinical Department No. 3, “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania;
| |
Collapse
|
34
|
Shi C, Tan J, Lu J, Huang J, Li X, Xu J, Wang X. MicroRNA-17-5p promotes vascular calcification by targeting ANKH. Curr Neurovasc Res 2022; 19:108-116. [PMID: 35297350 DOI: 10.2174/1567202619666220316115425] [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: 02/02/2022] [Revised: 02/08/2022] [Accepted: 02/11/2022] [Indexed: 11/22/2022]
Abstract
BACKGROUND MicroRNAs (miRNAs) may participate in the process of vascular calcification. However, the role of microRNA-17-5p in vascular calcification has not been clarified. In this study, we showed the effects of microRNA-17-5p on vascular calcification. MATERIALS AND METHODS Vascular smooth muscle cells (VSMCs) were transfected with miR-17-5p mimics, an miR-17-5p inhibitor or a negative control (NC) using Lipofectamine 2000. Then the cells were induced by an osteogenic medium. Alkaline phosphatase (ALP) activity and mineralization were determined. Osteocalcin (OC), bone morphogenetic protein 2(BMP-2), Col1agren Ia (Colla), Runx2 and ankylosis protein homolog (ANKH) gene expressions were determined by reverse transcription-polymerase chain reaction. Vascular calcification was developed using a renal failure model. RESULTS The ALP activity was increased when miR-17-5p mimics were transfected, whereas the miR-17-5p inhibitor reduced ALP activity (p < 0.05). The number and average area of mineral node in miR-17-5p mimics group were larger than those in corresponding control and NC groups (p < 0.05). The number and average area of the mineral nodes in the miR-17-5p inhibitor group were smaller than those in corresponding control and NC groups (p < 0.05). Bmp2, OC, Col1a and Runx2 were higher in the miR-17-5p mimics group compared to those in the control and NC groups. ANKH expression was decreased in VSMCs with the miR-17-5p mimics and increased in VSMCs with miR-17-5p inhibitor. ANKH siRNA intervention also promoted mineralization. The miR-17-5p expression was upregulated and ANKH was down-regulated in the aortic arteries with calcification. CONCLUSION Our data showed that miR-17-5p may promote vascular calcification by inhibiting ANKH expression.
Collapse
Affiliation(s)
- Chao Shi
- Department of Endocrinology, Shanghai Gongli Hospital, the Second Military Medical University, Shanghai 200135,China
| | - Jiaorong Tan
- Department of Endocrinology, Shanghai Putuo People's Hospital, Shanghai 200060,China
| | - Jiancan Lu
- Department of Endocrinology, Shanghai Gongli Hospital, the Second Military Medical University, Shanghai 200135,China
| | - Junling Huang
- Emergency department of Internal Medicine, Shanghai Gongli Hospital, the Second Military Medical University, Shanghai 200135,China
| | - Xiangqi Li
- Department of Endocrinology, Shanghai Gongli Hospital, the Second Military Medical University, Shanghai 200135,China
| | - Jiahong Xu
- Department of Cardiology,Tongji Hospital,Tongji University School of Medicine, Shanghai 200065, China
| | - Xing Wang
- Department of Endocrinology, Shanghai Gongli Hospital, the Second Military Medical University, Shanghai 200135,China
| |
Collapse
|
35
|
Rahmani R, Baranoski JF, Albuquerque FC, Lawton MT, Hashimoto T. Intracranial aneurysm calcification – A narrative review. Exp Neurol 2022; 353:114052. [DOI: 10.1016/j.expneurol.2022.114052] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Revised: 03/14/2022] [Accepted: 03/17/2022] [Indexed: 11/16/2022]
|
36
|
Matikainen N, Pekkarinen T, Ryhänen EM, Schalin-Jäntti C. Physiology of Calcium Homeostasis: An Overview. Endocrinol Metab Clin North Am 2021; 50:575-590. [PMID: 34774235 DOI: 10.1016/j.ecl.2021.07.005] [Citation(s) in RCA: 49] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Calcium plays a key role in skeletal mineralization and several intracellular and extracellular homeostatic networks. It is an essential element that is only available to the body through dietary sources. Daily acquisition of calcium depends, in addition to the actual intake, on the hormonally regulated state of calcium homeostasis through three main mechanisms: bone turnover, intestinal absorption, and renal reabsorption. These procedures are regulated by a group of interacting circulating hormones and their key receptors. This includes parathyroid hormone (PTH), PTH-related peptide, 1,25-dihydroxyvitamin D, calcitonin, fibroblast growth factor 23, the prevailing calcium concentration itself, the calcium-sensing receptor, as well as local processes in the bones, gut, and kidneys.
Collapse
Affiliation(s)
- Niina Matikainen
- Endocrinology, Abdominal Center, Helsinki University Hospital and University of Helsinki, PB 340, 00029 HUS, Helsinki, Finland
| | - Tuula Pekkarinen
- Endocrinology, Abdominal Center, Helsinki University Hospital and University of Helsinki, PB 340, 00029 HUS, Helsinki, Finland
| | - Eeva M Ryhänen
- Endocrinology, Abdominal Center, Helsinki University Hospital and University of Helsinki, PB 340, 00029 HUS, Helsinki, Finland
| | - Camilla Schalin-Jäntti
- Endocrinology, Abdominal Center, Helsinki University Hospital and University of Helsinki, PB 340, 00029 HUS, Helsinki, Finland.
| |
Collapse
|
37
|
CTRP3 is a coronary artery calcification biomarker and protects against vascular calcification by inhibiting β-catenin nuclear translocation to prevent vascular smooth muscle cell osteogenic differentiation. J Cardiol 2021; 79:551-558. [PMID: 34774387 DOI: 10.1016/j.jjcc.2021.10.018] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/11/2021] [Revised: 09/21/2021] [Accepted: 10/19/2021] [Indexed: 11/23/2022]
Abstract
BACKGROUND Coronary artery calcification (CAC) is an important risk factor for cardiovascular events and has been shown to be correlated with serum adiponectin levels. However, it remains unknown whether C1 tumor necrosis factor-related protein 3 (CTRP3) (homologous to adiponectin) is associated with CAC, and whether CTRP3 affects the osteoblastic differentiation of vascular smooth muscle cells. Here, we analyzed the association between CTRP3 expression and CAC. METHODS A case-control study was conducted involving 119 patients with coronary heart disease to identify the predictive value of CTRP3 for CAC. Additionally, mouse aortic smooth muscle cells transfected for β-catenin overexpression were subjected to treatment with CTRP3 and the β-catenin inhibitor JW74. The calcium content in smooth muscle cells was determined. Western blotting was performed to measure the expression levels of different osteoblastogenic proteins in vascular smooth muscle cells obtained from different treatment groups. RESULTS The serum CTRP3 levels were significantly lower in patients with CAC than in those without CAC, and even lower in patients with both CAC and diabetes mellitus. CTRP3 played roles as a protective factor and potential predictor in CAC. CTRP3 inhibited the osteogenic differentiation of vascular smooth muscle cells induced under high glucose and lipid conditions by inhibiting the nuclear translocation of β-catenin. CONCLUSIONS CTRP3 may serve as a valuable screening biomarker and a novel therapeutic target in CAC, particularly in diabetes patients.
Collapse
|
38
|
Chudek J, Kolonko A, Ziaja J, Francuz T, Kamińska D, Owczarek AJ, Kuczera P, Kujawa-Szewieczek A, Kusztal M, Kowalik AP, Bożek-Pająk D, Kluz J, Choręza P, Król R, Krajewska M, Cierpka L, Więcek A. Beneficial Effect of Successful Simultaneous Pancreas-Kidney Transplantation on Plasma Profile of Metalloproteinases in Type 1 Diabetes Mellitus Patients. J Clin Med 2021; 10:jcm10173800. [PMID: 34501247 PMCID: PMC8432100 DOI: 10.3390/jcm10173800] [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: 08/03/2021] [Revised: 08/21/2021] [Accepted: 08/23/2021] [Indexed: 12/04/2022] Open
Abstract
It is not fully elucidated whether the restoring of normal glucose metabolism after successful simultaneous pancreas-kidney transplantation (SPK) improves vascular wall morphology and function in type 1 diabetic (T1D) patients. Therefore, we compared arterial stiffness, assessed by pulse wave velocity (PWV), carotid intima-media thickness (IMT), and biomarkers of arterial wall calcification in T1D patients after SPK or kidney transplantation alone (KTA). In 39 SPK and 39 KTA adult patients of similar age, PWV, IMT, circulating matrix metalloproteinases (MMPs) and calcification biomarkers were assessed at median 83 months post transplantation. Additionally, carotid plaques were visualized and semi-qualitatively classified. Although PWV and IMT values were similar, the occurrence of atherosclerotic plaques (51.3 vs. 70.3%, p < 0.01) and calcified lesions (35.9 vs. 64.9%, p < 0.05) was lower in SPK patients. There were significantly lower concentrations of MMP-1, MMP-2, MMP-3, and osteocalcin in SPK subjects. Among the analyzed biomarkers, only logMMP-1, logMMP-2, and logMMP-3 concentrations were associated with log HbA1c. Multivariate stepwise backward regression analysis revealed that MMP-1 and MMP-3 variability were explained only by log HbA1c. Normal glucose metabolism achieved by SPK is followed by the favorable profile of circulating matrix metalloproteinases, which may reflect the vasoprotective effect of pancreas transplantation.
Collapse
Affiliation(s)
- Jerzy Chudek
- Department of Internal Medicine and Oncological Chemotherapy, Medical University of Silesia, 40-027 Katowice, Poland;
| | - Aureliusz Kolonko
- Department of Nephrology, Transplantation and Internal Medicine, Medical University of Silesia, 40-027 Katowice, Poland; (P.K.); (A.K.-S.); (A.W.)
- Correspondence: ; Tel.: +48-32-259-1429
| | - Jacek Ziaja
- Department of General, Vascular and Transplant Surgery, Medical University of Silesia, 40-027 Katowice, Poland; (J.Z.); (A.P.K.); (D.B.-P.); (R.K.); (L.C.)
| | - Tomasz Francuz
- Department of Biochemistry, Medical University of Silesia, 40-752 Katowice, Poland;
| | - Dorota Kamińska
- Department of Nephrology and Transplantation Medicine, Wrocław Medical University, 50-556 Wrocław, Poland; (D.K.); (M.K.); (M.K.)
| | - Aleksander J. Owczarek
- Health Promotion and Obesity Management Unit, Department of Pathophysiology, Medical University of Silesia, 40-752 Katowice, Poland; (A.J.O.); (P.C.)
| | - Piotr Kuczera
- Department of Nephrology, Transplantation and Internal Medicine, Medical University of Silesia, 40-027 Katowice, Poland; (P.K.); (A.K.-S.); (A.W.)
| | - Agata Kujawa-Szewieczek
- Department of Nephrology, Transplantation and Internal Medicine, Medical University of Silesia, 40-027 Katowice, Poland; (P.K.); (A.K.-S.); (A.W.)
| | - Mariusz Kusztal
- Department of Nephrology and Transplantation Medicine, Wrocław Medical University, 50-556 Wrocław, Poland; (D.K.); (M.K.); (M.K.)
| | - Adrian P. Kowalik
- Department of General, Vascular and Transplant Surgery, Medical University of Silesia, 40-027 Katowice, Poland; (J.Z.); (A.P.K.); (D.B.-P.); (R.K.); (L.C.)
| | - Dominika Bożek-Pająk
- Department of General, Vascular and Transplant Surgery, Medical University of Silesia, 40-027 Katowice, Poland; (J.Z.); (A.P.K.); (D.B.-P.); (R.K.); (L.C.)
| | - Joanna Kluz
- Department of Angiology, Hypertension and Diabetology, University Clinical Hospital, 50-556 Wrocław, Poland;
| | - Piotr Choręza
- Health Promotion and Obesity Management Unit, Department of Pathophysiology, Medical University of Silesia, 40-752 Katowice, Poland; (A.J.O.); (P.C.)
| | - Robert Król
- Department of General, Vascular and Transplant Surgery, Medical University of Silesia, 40-027 Katowice, Poland; (J.Z.); (A.P.K.); (D.B.-P.); (R.K.); (L.C.)
| | - Magdalena Krajewska
- Department of Nephrology and Transplantation Medicine, Wrocław Medical University, 50-556 Wrocław, Poland; (D.K.); (M.K.); (M.K.)
| | - Lech Cierpka
- Department of General, Vascular and Transplant Surgery, Medical University of Silesia, 40-027 Katowice, Poland; (J.Z.); (A.P.K.); (D.B.-P.); (R.K.); (L.C.)
| | - Andrzej Więcek
- Department of Nephrology, Transplantation and Internal Medicine, Medical University of Silesia, 40-027 Katowice, Poland; (P.K.); (A.K.-S.); (A.W.)
| |
Collapse
|
39
|
Bargieł W, Cierpiszewska K, Maruszczak K, Pakuła A, Szwankowska D, Wrzesińska A, Gutowski Ł, Formanowicz D. Recognized and Potentially New Biomarkers-Their Role in Diagnosis and Prognosis of Cardiovascular Disease. MEDICINA (KAUNAS, LITHUANIA) 2021; 57:medicina57070701. [PMID: 34356982 PMCID: PMC8305174 DOI: 10.3390/medicina57070701] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Revised: 07/03/2021] [Accepted: 07/04/2021] [Indexed: 01/08/2023]
Abstract
Atherosclerosis and its consequences are the leading cause of mortality in the world. For this reason, we have reviewed atherosclerosis biomarkers and selected the most promising ones for review. We focused mainly on biomarkers related to inflammation and oxidative stress, such as the highly sensitive C-reactive protein (hs-CRP), interleukin 6 (IL-6), and lipoprotein-associated phospholipase A2 (Lp-PLA2). The microRNA (miRNA) and the usefulness of the bone mineralization, glucose, and lipid metabolism marker osteocalcin (OC) were also reviewed. The last biomarker we considered was angiogenin (ANG). Our review shows that due to the multifactorial nature of atherosclerosis, no single marker is known so far, the determination of which would unambiguously assess the severity of atherosclerosis and help without any doubt in the prognosis of cardiovascular risk.
Collapse
Affiliation(s)
- Weronika Bargieł
- Faculty of Medicine, Poznan University of Medical Sciences, 60-812 Poznan, Poland; (W.B.); (K.C.); (K.M.); (A.P.); (D.S.); (A.W.)
| | - Katarzyna Cierpiszewska
- Faculty of Medicine, Poznan University of Medical Sciences, 60-812 Poznan, Poland; (W.B.); (K.C.); (K.M.); (A.P.); (D.S.); (A.W.)
| | - Klara Maruszczak
- Faculty of Medicine, Poznan University of Medical Sciences, 60-812 Poznan, Poland; (W.B.); (K.C.); (K.M.); (A.P.); (D.S.); (A.W.)
| | - Anna Pakuła
- Faculty of Medicine, Poznan University of Medical Sciences, 60-812 Poznan, Poland; (W.B.); (K.C.); (K.M.); (A.P.); (D.S.); (A.W.)
| | - Dominika Szwankowska
- Faculty of Medicine, Poznan University of Medical Sciences, 60-812 Poznan, Poland; (W.B.); (K.C.); (K.M.); (A.P.); (D.S.); (A.W.)
| | - Aleksandra Wrzesińska
- Faculty of Medicine, Poznan University of Medical Sciences, 60-812 Poznan, Poland; (W.B.); (K.C.); (K.M.); (A.P.); (D.S.); (A.W.)
| | - Łukasz Gutowski
- Department of Medical Chemistry and Laboratory Medicine, Poznan University of Medical Sciences, Rokietnicka 8, 60-806 Poznan, Poland;
| | - Dorota Formanowicz
- Department of Medical Chemistry and Laboratory Medicine, Poznan University of Medical Sciences, Rokietnicka 8, 60-806 Poznan, Poland;
- Correspondence:
| |
Collapse
|
40
|
Zhelyazkova-Savova MD, Yotov YT, Nikolova MN, Nazifova-Tasinova NF, Vankova DG, Atanasov AA, Galunska BT. Statins, vascular calcification, and vitamin K-dependent proteins: Is there a relation? Kaohsiung J Med Sci 2021; 37:624-631. [PMID: 33634559 DOI: 10.1002/kjm2.12373] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Revised: 12/30/2020] [Accepted: 01/24/2021] [Indexed: 12/16/2022] Open
Abstract
The present cross-sectional clinical study aimed to examine the connection between statin exposure, coronary artery calcification (CAC), and vitamin K-dependent proteins (VKDPs) in patients with cardiovascular (CV) conditions. Two groups of patients were studied: patients with established CV disease (CVD) and healthy patients at moderate risk for CVD (a control group). The groups were also split into statin users and non-users. The following VKDPs were measured in plasma: uncarboxylated Matrix Gla-protein (ucMGP), undercarboxylated (ucOC), and carboxylated osteocalcin (cOC), Gla-rich protein (GRP). CAC score (CACS) was determined by multislice computed tomography. Among all the participants in the study, CACS was more pronounced in statin users compared to non-users; the same was found also among the CVD patients and among the controls. While the levels of ucMGP and GRP did not differ between statin users and non-users, ucOC and ucOC/cOC were significantly elevated in statin users, indicating vitamin K deficiency. There was a positive correlation between the levels of ucOC and CACS in the entire population and in the group of statin users, but not in statin non-users. No association was found between ucMGP or GRP and CACS. Statins had also an impact on the international normalized ratio and interacted with vitamin K antagonists (VKAs). Our results are in agreement with the existing evidence about positive association between statins and vascular calcification. They enlighten to a certain extent the possible mechanisms through which statins may enhance calcium accumulation in arterial wall, namely, by inhibition of vitamin K dependent proteins and functions involved in vascular protection.
Collapse
Affiliation(s)
- Maria D Zhelyazkova-Savova
- Department of Pharmacology, Clinical Pharmacology and Therapy, Varna Medical University, Varna, Bulgaria
| | - Yoto T Yotov
- First Department of Internal Diseases, Faculty of Medicine, Medical University of Varna, Varna, Bulgaria
| | - Miglena N Nikolova
- Department of Biochemistry, Molecular medicine and Nutrigenomics, Faculty of Pharmacy, Medical University of Varna, Varna, Bulgaria
| | - Neshe F Nazifova-Tasinova
- Department of Biochemistry, Molecular medicine and Nutrigenomics, Faculty of Pharmacy, Medical University of Varna, Varna, Bulgaria
| | - Deyana G Vankova
- Department of Biochemistry, Molecular medicine and Nutrigenomics, Faculty of Pharmacy, Medical University of Varna, Varna, Bulgaria
| | - Atanas A Atanasov
- First Department of Internal Diseases, Faculty of Medicine, Medical University of Varna, Varna, Bulgaria
| | - Bistra Tz Galunska
- Department of Biochemistry, Molecular medicine and Nutrigenomics, Faculty of Pharmacy, Medical University of Varna, Varna, Bulgaria
| |
Collapse
|
41
|
Osteoclast-Mediated Cell Therapy as an Attempt to Treat Elastin Specific Vascular Calcification. Molecules 2021; 26:molecules26123643. [PMID: 34203711 PMCID: PMC8232296 DOI: 10.3390/molecules26123643] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2021] [Revised: 06/08/2021] [Accepted: 06/09/2021] [Indexed: 01/03/2023] Open
Abstract
Inflammation and stiffness in the arteries is referred to as vascular calcification. This process is a prevalent yet poorly understood consequence of cardiovascular disease and diabetes mellitus, comorbidities with few treatments clinically available. Because this is an active process similar to bone formation, it is hypothesized that osteoclasts (OCs), bone-resorbing cells in the body, could potentially work to reverse existing calcification by resorbing bone material. The receptor activator of nuclear kappa B-ligand (RANKL) is a molecule responsible for triggering a response in monocytes and macrophages that allows them to differentiate into functional OCs. In this study, OC and RANKL delivery were employed to determine whether calcification could be attenuated. OCs were either delivered via direct injection, collagen/alginate microbeads, or collagen gel application, while RANKL was delivered via injection, through either a porcine subdermal model or aortic injury model. While in vitro results yielded a decrease in calcification using OC therapy, in vivo delivery mechanisms did not provide control or regulation to keep cells localized long enough to induce calcification reduction. However, these results do provide context and direction for the future of OC therapy, revealing necessary steps for this treatment to effectively reduce calcification in vivo. The discrepancy between in vivo and in vitro success for OC therapy points to the need for a more stable and time-controlled delivery mechanism that will allow OCs not only to remain at the site of calcification, but also to be regulated so that they are healthy and functioning normally when introduced to diseased tissue.
Collapse
|
42
|
Fehérvári L, Frigy A, Kocsis L, Szabó IA, Szabo TM, Urkon M, Jakó Z, Nagy EE. Serum Osteoprotegerin and Carotid Intima-Media Thickness Are Related to High Arterial Stiffness in Heart Failure with Reduced Ejection Fraction. Diagnostics (Basel) 2021; 11:diagnostics11050764. [PMID: 33923139 PMCID: PMC8145213 DOI: 10.3390/diagnostics11050764] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Revised: 04/15/2021] [Accepted: 04/20/2021] [Indexed: 12/11/2022] Open
Abstract
Arterial stiffness (AS) is a complex vascular phenomenon with consequences for central hemodynamics and left-ventricular performance. Circulating biomarkers have been associated with AS; however, their value in heart failure is poorly characterized. Our aim was to evaluate the clinical and biomarker correlates of AS in the setting of heart failure with reduced ejection fraction (HFrEF). In 78 hospitalized, hemodynamically stable patients (20 women, 58 men, mean age 65.8 ± 1.41 years) with HFrEF, AS was measured using aortic pulse wave velocity (PWV). Serum OPG, RANKL, sclerostin, and DKK-1 were determined, and the relationships between the clinical variables, vascular-calcification-related biomarkers, and PWV were evaluated by correlation analysis and linear and logistic regression models. OPG and the OPG/RANKL ratio were significantly higher in the group of patients (n = 37, 47.4%) with increased PWV (>10 m/s). PWV was positively correlated with age, left-ventricular ejection fraction, and carotid intima-media thickness (cIMT), and negatively correlated with the glomerular filtration rate. OPG and cIMT were significantly associated with PWV in the logistic regression models when adjusted for hypertension, EF, and the presence of atherosclerotic manifestations. Elevated serum OPG, together with cIMT, were significantly related to increased AS in the setting of HFrEF.
Collapse
Affiliation(s)
- Lajos Fehérvári
- Department of Cardiology, Clinical County Hospital Mures, 540103 Targu Mures, Romania; (L.F.); (A.F.); (L.K.); (I.A.S.); (T.M.S.)
- Department of Internal Medicine IV, George Emil Palade University of Medicine, Pharmacy, Sciences and Technology of Targu Mures, 540103 Targu Mures, Romania
| | - Attila Frigy
- Department of Cardiology, Clinical County Hospital Mures, 540103 Targu Mures, Romania; (L.F.); (A.F.); (L.K.); (I.A.S.); (T.M.S.)
- Department of Internal Medicine IV, George Emil Palade University of Medicine, Pharmacy, Sciences and Technology of Targu Mures, 540103 Targu Mures, Romania
| | - Lóránd Kocsis
- Department of Cardiology, Clinical County Hospital Mures, 540103 Targu Mures, Romania; (L.F.); (A.F.); (L.K.); (I.A.S.); (T.M.S.)
| | - István Adorján Szabó
- Department of Cardiology, Clinical County Hospital Mures, 540103 Targu Mures, Romania; (L.F.); (A.F.); (L.K.); (I.A.S.); (T.M.S.)
- Department of Internal Medicine IV, George Emil Palade University of Medicine, Pharmacy, Sciences and Technology of Targu Mures, 540103 Targu Mures, Romania
| | - Timea Magdolna Szabo
- Department of Cardiology, Clinical County Hospital Mures, 540103 Targu Mures, Romania; (L.F.); (A.F.); (L.K.); (I.A.S.); (T.M.S.)
- Department of Biochemistry and Environmental Chemistry, George Emil Palade University of Medicine, Pharmacy, Sciences and Technology of Targu Mures, 540142 Targu Mures, Romania;
| | - Melinda Urkon
- Department of Biochemistry and Environmental Chemistry, George Emil Palade University of Medicine, Pharmacy, Sciences and Technology of Targu Mures, 540142 Targu Mures, Romania;
| | - Zita Jakó
- Laboratory of Medical Analysis, Emergency Clinical County Hospital Targu Mures, 540136 Targu Mures, Romania;
| | - Előd Ernő Nagy
- Department of Biochemistry and Environmental Chemistry, George Emil Palade University of Medicine, Pharmacy, Sciences and Technology of Targu Mures, 540142 Targu Mures, Romania;
- Laboratory of Medical Analysis, Clinical County Hospital Mures, 540394 Targu Mures, Romania
- Correspondence: ; Tel.: +40-733-956-395
| |
Collapse
|
43
|
Hu CT, Shao YD, Liu YZ, Xiao X, Cheng ZB, Qu SL, Huang L, Zhang C. Oxidative stress in vascular calcification. Clin Chim Acta 2021; 519:101-110. [PMID: 33887264 DOI: 10.1016/j.cca.2021.04.012] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2020] [Revised: 04/15/2021] [Accepted: 04/16/2021] [Indexed: 12/28/2022]
Abstract
Vascular calcification (VC), which is closely associated with significant mortality in cardiovascular disease, chronic kidney disease (CKD), and/or diabetes mellitus, is characterized by abnormal deposits of hydroxyapatite minerals in the arterial wall. The impact of oxidative stress (OS) on the onset and progression of VC has not been well described. Nicotinamide adenine dinucleotide phosphate (NADPH) oxidases, xanthine oxidases, myeloperoxidase (MPO), nitric oxide synthases (NOSs), superoxide dismutase (SOD) and paraoxonases (PONs) are relevant factors that influence the production of reactive oxygen species (ROS). Furthermore, excess ROS-induced OS has emerged as a critical mediator promoting VC through several mechanisms, including phosphate balance, differentiation of vascular smooth muscle cells (VSMCs), inflammation, DNA damage, and extracellular matrix remodeling. Because OS is a significant regulator of VC, antioxidants may be considered as novel treatment options.
Collapse
Affiliation(s)
- Chu-Ting Hu
- Institute of Cardiovascular Disease, Key Lab for Arteriosclerology of Hunan Province, Hengyang Medical College, University of South China, Hengyang, Hunan 421001, People's Republic of China; Research Lab for Clinical & Translational Medicine, Hengyang Medical College, University of South China, Hengyang, Hunan 421001, People's Republic of China; Departments of Medical Laboratory, Hengyang Medical College, University of South China, Hengyang, Hunan 421001, People's Republic of China
| | - Yi-Duo Shao
- Institute of Cardiovascular Disease, Key Lab for Arteriosclerology of Hunan Province, Hengyang Medical College, University of South China, Hengyang, Hunan 421001, People's Republic of China; Research Lab for Clinical & Translational Medicine, Hengyang Medical College, University of South China, Hengyang, Hunan 421001, People's Republic of China; Departments of Stomatology, Hengyang Medical College, University of South China, Hengyang, Hunan 421001, People's Republic of China
| | - Yi-Zhang Liu
- Institute of Cardiovascular Disease, Key Lab for Arteriosclerology of Hunan Province, Hengyang Medical College, University of South China, Hengyang, Hunan 421001, People's Republic of China; Research Lab for Clinical & Translational Medicine, Hengyang Medical College, University of South China, Hengyang, Hunan 421001, People's Republic of China; Departments of Clinical Medicine, Hengyang Medical College, University of South China, Hengyang, Hunan 421001, People's Republic of China
| | - Xuan Xiao
- Institute of Cardiovascular Disease, Key Lab for Arteriosclerology of Hunan Province, Hengyang Medical College, University of South China, Hengyang, Hunan 421001, People's Republic of China; Research Lab for Clinical & Translational Medicine, Hengyang Medical College, University of South China, Hengyang, Hunan 421001, People's Republic of China; Departments of Clinical Medicine, Hengyang Medical College, University of South China, Hengyang, Hunan 421001, People's Republic of China
| | - Zhe-Bin Cheng
- Institute of Cardiovascular Disease, Key Lab for Arteriosclerology of Hunan Province, Hengyang Medical College, University of South China, Hengyang, Hunan 421001, People's Republic of China; Research Lab for Clinical & Translational Medicine, Hengyang Medical College, University of South China, Hengyang, Hunan 421001, People's Republic of China; Departments of Stomatology, Hengyang Medical College, University of South China, Hengyang, Hunan 421001, People's Republic of China
| | - Shun-Lin Qu
- Institute of Cardiovascular Disease, Key Lab for Arteriosclerology of Hunan Province, Hengyang Medical College, University of South China, Hengyang, Hunan 421001, People's Republic of China
| | - Liang Huang
- Research Lab for Clinical & Translational Medicine, Hengyang Medical College, University of South China, Hengyang, Hunan 421001, People's Republic of China.
| | - Chi Zhang
- Institute of Cardiovascular Disease, Key Lab for Arteriosclerology of Hunan Province, Hengyang Medical College, University of South China, Hengyang, Hunan 421001, People's Republic of China.
| |
Collapse
|
44
|
Prevalence and risk factors for the development of abdominal aortic calcification among the US population: NHANES study. ACTA ACUST UNITED AC 2021; 6:e95-e101. [PMID: 34027218 PMCID: PMC8117070 DOI: 10.5114/amsad.2021.105527] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Accepted: 01/07/2021] [Indexed: 11/19/2022]
Abstract
Introduction Abdominal aortic calcification (AAC) is an important marker of subclinical cardiovascular disease and its prognosis. Advanced age, hypertension, smoking, dyslipidemia, diabetes mellitus, and higher truncal fat are known markers of AAC in studies conducted around the world. However, literature for these risk factors and their co-occurrence is limited in the US. Material and methods We used data from dual energy X-ray absorptiometry (Hologic, v4.0) to detect the occurrence of AAC in a sample population (n = 3140) of the NHANES survey using a computer-assisted interviewing system to assess the risk factors for AAC. Results We found the national prevalence of AAC in the US to be 28.8%. After adjusting for confounders, persons with hypertension: OR = 1.66 (95% CI: 1.30–2.13) and smokers: OR = 1.63 (95% CI: 1.24–2.14) were more likely to have AAC compared to their respective counterparts. Increasing age was positively associated with AAC: OR = 1.06 (95% CI: 1.04–1.08). There was a statistically significant negative association between body mass index (BMI) and AAC, more so in smokers than in non-smokers: OR = 0.97 (95% CI: 0.94–0.97). We did not observe any statistically significant association between diabetes and AAC. Conclusions Advanced age, smoking, and hypertension was associated with increased occurrence of AAC. Paradoxically, increasing BMI was inversely associated with AAC and there was no statistically significant association between total body and trunk fat percentages and AAC. To the best of our knowledge, this is the first study to establish the nationwide prevalence and associated factors in the US.
Collapse
|
45
|
The Impact of Sclerostin Levels on Long-Term Prognosis in Patients Undergoing Coronary Angiography: A Personalized Approach with 9-Year Follow-Up. J Pers Med 2021; 11:jpm11030186. [PMID: 33800939 PMCID: PMC8001826 DOI: 10.3390/jpm11030186] [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: 02/02/2021] [Revised: 02/27/2021] [Accepted: 03/01/2021] [Indexed: 12/28/2022] Open
Abstract
Sclerostin might play a role in atherosclerosis development. This study aimed to analyze the impact of baseline sclerostin levels on 9-year outcomes in patients without significant renal function impairment and undergoing coronary angiography. The primary study endpoint was the rate of major cardiovascular events (MACE), defined as a combined rate of myocardial infarction (MI), stroke, or death at 9 years. We included 205 patients with a mean age of 62.9 ± 0.6 years and 70.2% male. Median serum sclerostin concentration was 133.22 pg/mL (IQR 64.0–276.17). At 9 years, in the whole population, the rate of MACE was 34.1% (n = 70), MI: 11.2% (n = 23), stroke: 2.4% (n = 5), and death: 20.5% (n = 42). In the high sclerostin (>median) group, we observed statistically significant higher rates of MACE and death: 25.2% vs. 43.1% (HR 1.75, 95% CI 1.1–2.10, p = 0.02) and 14.6% vs. 26.5% (HR 1.86, 95% CI 1.02–3.41, p = 0.049), respectively. Similar relationships were observed in patients with chronic coronary syndrome and SYNTAX 0–22 subgroups. Our results suggest that sclerostin assessment might be useful in risk stratification, and subjects with higher sclerostin levels might have a worse prognosis.
Collapse
|
46
|
Xiao H, Chen J, Duan L, Li S. Role of emerging vitamin K‑dependent proteins: Growth arrest‑specific protein 6, Gla‑rich protein and periostin (Review). Int J Mol Med 2021; 47:2. [PMID: 33448308 PMCID: PMC7834955 DOI: 10.3892/ijmm.2020.4835] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2020] [Accepted: 10/21/2020] [Indexed: 01/27/2023] Open
Abstract
Vitamin K‑dependent proteins (VKDPs) are a group of proteins that need vitamin K to conduct carboxylation. Thus far, scholars have identified a total of 17 VKDPs in the human body. In this review, we summarize three important emerging VKDPs: Growth arrest‑specific protein 6 (Gas 6), Gla‑rich protein (GRP) and periostin in terms of their functions in physiological and pathological conditions. As examples, carboxylated Gas 6 and GRP effectively protect blood vessels from calcification, Gas 6 protects from acute kidney injury and is involved in chronic kidney disease, GRP contributes to bone homeostasis and delays the progression of osteoarthritis, and periostin is involved in all phases of fracture healing and assists myocardial regeneration in the early stages of myocardial infarction. However, periostin participates in the progression of cardiac fibrosis, idiopathic pulmonary fibrosis and airway remodeling of asthma. In addition, we discuss the relationship between vitamin K, VKDPs and cancer, and particularly the carboxylation state of VKDPs in cancer.
Collapse
Affiliation(s)
- Huiyu Xiao
- Department of Physiology, Dalian Medical University, Dalian, Liaoning 116044
| | - Jiepeng Chen
- Sungen Bioscience Co., Ltd., Shantou, Guangdong 515071, P.R. China
| | - Lili Duan
- Sungen Bioscience Co., Ltd., Shantou, Guangdong 515071, P.R. China
| | - Shuzhuang Li
- Department of Physiology, Dalian Medical University, Dalian, Liaoning 116044
| |
Collapse
|
47
|
Ma WQ, Sun XJ, Zhu Y, Liu NF. Metformin attenuates hyperlipidaemia-associated vascular calcification through anti-ferroptotic effects. Free Radic Biol Med 2021; 165:229-242. [PMID: 33513420 DOI: 10.1016/j.freeradbiomed.2021.01.033] [Citation(s) in RCA: 88] [Impact Index Per Article: 29.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/21/2020] [Revised: 01/13/2021] [Accepted: 01/17/2021] [Indexed: 12/14/2022]
Abstract
Ferroptosis is a form of regulated cell death that involves metabolic dysfunction resulting from iron-dependent excessive lipid peroxidation. Elevated plasma levels of free fatty acids are tightly associated with cardiometabolic risk factors in patients with obesity, diabetes mellitus, and metabolic syndrome. Metformin (Met) is an antidiabetic drug with beneficial cardiovascular disease effects. The aim of this study was to determine the effects of Met on ferroptosis induced by lipid overload and the effects of these changes on vascular smooth muscle cells (VSMCs) calcification. We developed a hyperlipidaemia-related vascular calcification in vivo model with rats fed a high-fat diet combined with vitamin D3 plus nicotine, and palmitic acid (PA), the most abundant long-chain saturated fatty acid in plasma, was used to induce lipid overload and develop an oxidative stress-related calcification model in vitro. The results showed that Met inhibits hyperlipidaemia-associated calcium deposition in the rat aortic tissue. In vitro, treatment of VSMCs with PA stimulates ferroptosis concomitant with increased calcium deposition in VSMCs, while pretreatment with Met attenuates these effects. Furthermore, PA also promotes the protein expression of the extracellular matrix protein periostin (POSTN) and its secretion into the extracellular environment. More importantly, upregulation of POSTN increased the sensitivity of cells to ferroptosis. Mechanistically, upregulation of POSTN suppresses SLC7A11 expression through the inhibition of p53 in VSMCs, which contributes to a decrease in glutathione synthesis and therefore triggers ferroptosis. Interestingly, overexpression of p53 attenuates the inhibitory effect of POSTN on SLC7A11 expression, accompanied by increased Gpx4 expression. Furthermore, p53 knockdown suppresses Met-mediated anti-ferroptosis effects in PA-treated VSMCs, which may be related to the downregulation of SLC7A11 expression. In addition, supplementation of VSMCs with Met enhances the antioxidative capacity of VSMCs through Nrf2 signalling activation. Collectively, targeting POSTN in VSMCs may provide a new strategy for vascular calcification prevention or treatment.
Collapse
Affiliation(s)
- Wen-Qi Ma
- Department of Cardiology, Zhongda Hospital, School of Medicine, Southeast University 87 Dingjiaqiao, Nanjing, 210009, PR China
| | - Xue-Jiao Sun
- Department of Cardiology, Zhongda Hospital, School of Medicine, Southeast University 87 Dingjiaqiao, Nanjing, 210009, PR China
| | - Yi Zhu
- Department of Cardiology, Zhongda Hospital, School of Medicine, Southeast University 87 Dingjiaqiao, Nanjing, 210009, PR China
| | - Nai-Feng Liu
- Department of Cardiology, Zhongda Hospital, School of Medicine, Southeast University 87 Dingjiaqiao, Nanjing, 210009, PR China.
| |
Collapse
|
48
|
Alshahawey M, El Borolossy R, El Wakeel L, Elsaid T, Sabri NA. The impact of cholecalciferol on markers of vascular calcification in hemodialysis patients: A randomized placebo controlled study. Nutr Metab Cardiovasc Dis 2021; 31:626-633. [PMID: 33594986 DOI: 10.1016/j.numecd.2020.09.014] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Revised: 08/20/2020] [Accepted: 09/12/2020] [Indexed: 12/24/2022]
Abstract
BACKGROUND AND AIM Vascular calcification is an independent risk factor for cardiovascular diseases and all-cause mortality in end stage renal disease, and particularly in hemodialysis patients. Vitamin D deficiency has been shown to be associated with vascular calcification among this category of patients. Cholecalciferol or vitamin D3; the native inactivated 25-hydroxy vitamin D [25(OH)D], has been proposed to have a good impact on vascular calcification and vitamin D deficiency. However, clinical data is still limited. METHODS AND RESULTS A prospective, randomized, placebo-controlled study was carried out to evaluate the effect of oral cholecalciferol on vascular calcification and 25(OH)D levels in hemodialysis patients. A total of sixty eligible hemodialysis patients were randomly assigned to either a treatment group (Oral 200.000IU Cholecalciferol per month) or a placebo group, for 3 months. Serum 25-hydroxy vitamin D (25(OH)D), fetuin-A, fibroblast growth factor (FGF-23), osteoprotegerin (OPG), calcium, phosphorus, their product (CaXP) and intact parathyroid hormone (iPTH) levels, were all assessed at baseline and at the end of the study. ClinicalTrials.gov registration number: NCT03602430. Cholecalciferol significantly increased serum levels of 25(OH)D and fetuin-A in the treatment group (p-value < 0.001), while no significant difference was observed in the placebo group. Cholecalciferol administration showed no effect on either FGF-23 or OPG. None of the treatment group patients experienced any adverse effects. CONCLUSION Cholecalciferol was shown to be an effective, tolerable, inexpensive pharmacotherapeutic option to overcome vitamin D deficiency, with a possible modulating effect on fetuin-A, among hemodialysis patients. CLINICALTRIALS. GOV REGISTRATION NUMBER NCT03602430.
Collapse
Affiliation(s)
- Mona Alshahawey
- Department of Clinical Pharmacy, Faculty of Pharmacy, Ain Shams University, Cairo, Egypt.
| | - Radwa El Borolossy
- Department of Clinical Pharmacy, Faculty of Pharmacy, Ain Shams University, Cairo, Egypt.
| | - Lamia El Wakeel
- Department of Clinical Pharmacy, Faculty of Pharmacy, Ain Shams University, Cairo, Egypt.
| | - Tamer Elsaid
- Department of Nephrology, Faculty of Medicine, Ain Shams University, Cairo, Egypt.
| | - Nagwa Ali Sabri
- Department of Clinical Pharmacy, Faculty of Pharmacy, Ain Shams University, Cairo, Egypt.
| |
Collapse
|
49
|
Salam S, Gallagher O, Gossiel F, Paggiosi M, Eastell R, Khwaja A. Vascular calcification relationship to vascular biomarkers and bone metabolism in advanced chronic kidney disease. Bone 2021; 143:115699. [PMID: 33091638 DOI: 10.1016/j.bone.2020.115699] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/27/2020] [Revised: 10/09/2020] [Accepted: 10/15/2020] [Indexed: 12/15/2022]
Abstract
BACKGROUND Vascular calcification (VC) and renal osteodystrophy are important complications of advanced chronic kidney disease (CKD). High resolution peripheral quantitative computed tomography (HRpQCT) is able to assess bone microstructure in renal osteodystrophy and lower leg arterial calcification (LLAC) is usually seen as an incidental finding. LLAC can be a useful quantitative assessment of VC in CKD but the relationship between LLAC and vascular biomarkers and bone is unknown. We aimed to assess the relationship between LLAC and biomarkers, bone turnover and microstructure. METHODS In this cross-sectional study, fasting blood samples were taken from 69 CKD stages 4-5D patients and 68 healthy controls. HRpQCT of distal tibia and radius were performed. 43 CKD patients had trans-iliac bone biopsy after tetracycline labelling. RESULTS LLAC was more severe in CKD than controls (median [IQR] 1.043 [0.05-16.52] vs 0 [0-0.55] mgHA, p < 0.001). CKD patients with diabetes (28%) had significantly higher LLAC compared to non-diabetic CKD (median [IQR] 24.07 [3.42-61.30] vs 0.23 [0-3.78] mgHA, p < 0.001). LLAC mass in CKD correlated with serum phosphate (rho = 0.29, p < 0.05), calcium x phosphate product (rho = 0.31, p < 0.05), intact parathyroid hormone (rho = 0.38, p < 0.01), intact fibroblast growth factor-23 (iFGF23) (rho = 0.40, p = 0.001), total alkaline phosphatase (rho = 0.41, p < 0.001), bone alkaline phosphatase (rho = 0.29, p < 0.05), osteocalcin (rho = 0.32, p < 0.05), osteoprotegerin (rho = 0.40, p = 0.001) and dephosphorylated-uncarboxylated matrix Gla protein (rho = 0.31, p < 0.05). LLAC in CKD also correlated with worse distal tibia cortical bone mineral density, thickness and porosity. No association was found between LLAC and bone turnover, mineralization or volume on biopsy in CKD. In multivariate analysis, only age, diabetes, iPTH and iFGF23 were independently associated with LLAC in CKD. CONCLUSIONS High levels of PTH and FGF23, along with older age and the presence of diabetes may all play independent roles in the development of LLAC in advanced CKD.
Collapse
Affiliation(s)
- Syazrah Salam
- Sheffield Kidney Institute, Sheffield Teaching Hospitals NHS Foundation Trust, United Kingdom; Academic Unit of Bone Metabolism and Mellanby Centre for Bone Research, University of Sheffield, United Kingdom.
| | - Orla Gallagher
- Academic Unit of Bone Metabolism and Mellanby Centre for Bone Research, University of Sheffield, United Kingdom
| | - Fatma Gossiel
- Academic Unit of Bone Metabolism and Mellanby Centre for Bone Research, University of Sheffield, United Kingdom
| | - Margaret Paggiosi
- Academic Unit of Bone Metabolism and Mellanby Centre for Bone Research, University of Sheffield, United Kingdom
| | - Richard Eastell
- Academic Unit of Bone Metabolism and Mellanby Centre for Bone Research, University of Sheffield, United Kingdom
| | - Arif Khwaja
- Sheffield Kidney Institute, Sheffield Teaching Hospitals NHS Foundation Trust, United Kingdom
| |
Collapse
|
50
|
Vidavsky N, Kunitake JAMR, Estroff LA. Multiple Pathways for Pathological Calcification in the Human Body. Adv Healthc Mater 2021; 10:e2001271. [PMID: 33274854 PMCID: PMC8724004 DOI: 10.1002/adhm.202001271] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2020] [Revised: 10/16/2020] [Indexed: 12/12/2022]
Abstract
Biomineralization of skeletal components (e.g., bone and teeth) is generally accepted to occur under strict cellular regulation, leading to mineral-organic composites with hierarchical structures and properties optimized for their designated function. Such cellular regulation includes promoting mineralization at desired sites as well as inhibiting mineralization in soft tissues and other undesirable locations. In contrast, pathological mineralization, with potentially harmful health effects, can occur as a result of tissue or metabolic abnormalities, disease, or implantation of certain biomaterials. This progress report defines mineralization pathway components and identifies the commonalities (and differences) between physiological (e.g., bone remodeling) and pathological calcification formation pathways, based, in part, upon the extent of cellular control within the system. These concepts are discussed in representative examples of calcium phosphate-based pathological mineralization in cancer (breast, thyroid, ovarian, and meningioma) and in cardiovascular disease. In-depth mechanistic understanding of pathological mineralization requires utilizing state-of-the-art materials science imaging and characterization techniques, focusing not only on the final deposits, but also on the earlier stages of crystal nucleation, growth, and aggregation. Such mechanistic understanding will further enable the use of pathological calcifications in diagnosis and prognosis, as well as possibly provide insights into preventative treatments for detrimental mineralization in disease.
Collapse
Affiliation(s)
- Netta Vidavsky
- Department of Chemical Engineering, Ben-Gurion University of the Negev, Beer-Sheva, 8410501, Israel
| | - Jennie A M R Kunitake
- Department of Materials Science and Engineering, Cornell University, Ithaca, NY, 14853, USA
| | - Lara A Estroff
- Department of Materials Science and Engineering, Cornell University, Ithaca, NY, 14853, USA
- Kavli Institute at Cornell for Nanoscale Science, Ithaca, NY, 14853, USA
| |
Collapse
|