Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 988
Filter
1.
Exp Clin Transplant ; 22(8): 613-621, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39254073

ABSTRACT

OBJECTIVES: This study investigated whether kidney transplant donors experience increased arterial stiffness compared with the general population and how arterial stiffness changes over time. MATERIALS AND METHODS: Our study included 59 kidney transplant donors and 27 healthy volunteers. All subjects underwent cardio-ankle vascular index measurements. We studied fibroblast growth factor23, klotho, monocyte chemoattractant protein-1, N-terminal pro-B-type natriuretic peptide, indoxyl sulfate, and p-cresyl sulfate levels. RESULTS: Cardio-ankle vascular index level was higher in donors 6 to 11 years after donation (8.02 ± 0.24 m/s) than in donors 2 to 6 years after donation (7.02 ± 0.27 m/s) and healthy volunteers (6.65 ± 0.22 m/s). Cardioankle vascular index level was positively correlated with age (r = 0.382, P < .001) and levels of triglyceride (r = 0.213, P = .049), blood urea nitrogen (r = 0.263, P = .014), creatinine (r = 0.354, P = .001), calcium (r = 0.228, P = .035), indoxyl sulfate (r = 0.219, P = .042), p-cresyl sulfate (r = 0.676, P ≤ .001), and monocyte chemoattractant protein-1 (r = 0.451, P ≤ .001) and negatively correlated with estimated glomerular filtration rate (r = -0.383, P < .001). Multiple linear regression analysis revealed that age (P = .026, B = 0.244), mean arterial blood pressure (P < .001, B = 0.446), blood urea nitrogen (P = .006, B = 0.302), creatinine (P = .032, B = 0.236), estimated glomerular filtration rate (P = .003, B = -0.323), fibroblast growth factor-23 (P = .007, B = 0.294), N-terminal pro-B-type natriuretic peptide (P = .005, B = 0.304), and monocyte chemoattractant protein-1 (P ≤ .001, B = 0.434) independently predicted cardio-ankle vascular index levels. CONCLUSIONS: Even without additional risk factors, kidney donors should be followed closely for arterial stiffness and cardiovascular disease, especially in the long-term (>5 years) after kidney transplant.


Subject(s)
Biomarkers , Cardio Ankle Vascular Index , Inflammation Mediators , Kidney Transplantation , Predictive Value of Tests , Vascular Calcification , Vascular Stiffness , Humans , Male , Female , Biomarkers/blood , Middle Aged , Kidney Transplantation/adverse effects , Adult , Case-Control Studies , Vascular Calcification/blood , Vascular Calcification/physiopathology , Vascular Calcification/etiology , Vascular Calcification/diagnosis , Time Factors , Inflammation Mediators/blood , Risk Factors , Fibroblast Growth Factors/blood , Fibroblast Growth Factor-23 , Chemokine CCL2/blood , Uremia/blood , Uremia/diagnosis , Uremia/physiopathology , Indican/blood , Treatment Outcome , Living Donors
2.
Ren Fail ; 46(2): 2394633, 2024 Dec.
Article in English | MEDLINE | ID: mdl-39230198

ABSTRACT

OBJECTIVE: This study aimed to investigate the effect of the soluble Klotho (sKlotho)/Wnt/ß-catenin signaling pathway on vascular calcification in rat models of chronic kidney disease (CKD) and the intervention effect of Shenyuan granules. METHODS: Rats with 5/6 nephrectomy and high phosphorus feeding were used to establish the vascular calcification model. The rats were given gradient doses of Shenyuan granules aqueous solution and calcitriol solution by gavage for 8 weeks, which were divided into experimental group and positive control group. RESULTS: The 5/6 nephrectomy combined with high phosphorus feeding induced thoracic aortic calcification in rats. Shenyuan granules intervention increased the serum sKlotho level, inhibited the mRNA and protein expression of Wnt1, ß-catenin, and Runx2 in the thoracic aorta, and alleviated thoracic aortic media calcification in rats. CONCLUSION: Shenyuan granules may partially regulate the Wnt/ß-catenin signaling pathway via serum sKl to interfere with the expression of Runx2, thereby improving vascular calcification in CKD.


Subject(s)
Drugs, Chinese Herbal , Glucuronidase , Klotho Proteins , Renal Insufficiency, Chronic , Vascular Calcification , Wnt Signaling Pathway , beta Catenin , Animals , Male , Rats , Aorta, Thoracic/metabolism , Aorta, Thoracic/pathology , beta Catenin/metabolism , Core Binding Factor Alpha 1 Subunit/metabolism , Disease Models, Animal , Drugs, Chinese Herbal/pharmacology , Drugs, Chinese Herbal/therapeutic use , Glucuronidase/metabolism , Glucuronidase/genetics , Klotho Proteins/metabolism , Nephrectomy , Rats, Sprague-Dawley , Renal Insufficiency, Chronic/metabolism , Renal Insufficiency, Chronic/complications , Vascular Calcification/metabolism , Vascular Calcification/etiology , Vascular Calcification/pathology , Wnt Signaling Pathway/drug effects , Wnt1 Protein/metabolism , Wnt1 Protein/genetics
3.
Arch Pediatr ; 31(4S1): 4S3-4S12, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39343471

ABSTRACT

Bone has several crucial functions. It is essential for locomotion and allows our body to stand erect against gravity. A mismatch between the mechanical stresses applied to it and its mechanical resistance leads to fractures. Bone also has numerous endocrine functions. It acts as a reservoir for minerals such as calcium and phosphorus, making it the target of calciotropic hormones that mobilize these minerals, particularly calcium, according to the body's needs. Additionally, bone secretes hormones, notably fibroblast growth factor 23 (FGF23), which regulates urinary excretion of phosphate and the bioavailability of active vitamin D. Bone mineralization is the process that facilitates the organized deposition of minerals in the bone matrix, providing rigidity and appropriate mechanical resistance. This process is compromised in genetically related bone mineralization disorders, such as those causing hypophosphatemia or hypophosphatasia. Conversely, calcification can be pathological, affecting soft tissues like the blood vessels, as seen in generalized arterial calcification of infancy (GACI) or arterial calcification due to CD73 deficiency (ACDC). The aim of this article is to first present the composition and structure of the mineralized bone matrix, to review the current understanding of the molecular mechanisms of mineralization, and finally to discuss the conditions associated with ectopic calcification and the underlying mechanisms.


Subject(s)
Fibroblast Growth Factor-23 , Humans , Calcinosis/etiology , Calcification, Physiologic/physiology , Fibroblast Growth Factors/metabolism , Fibroblast Growth Factors/physiology , Vascular Calcification/metabolism , Vascular Calcification/etiology , Hypophosphatasia/physiopathology , Hypophosphatasia/metabolism , Hypophosphatasia/genetics , Hypophosphatemia/metabolism , 5'-Nucleotidase/metabolism , 5'-Nucleotidase/physiology , Bone and Bones/metabolism , GPI-Linked Proteins
4.
Clin Transl Sci ; 17(10): e70036, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39344403

ABSTRACT

Aortic calcification-a marker of advanced atherosclerosis in large arteries-associates with cardiovascular mortality and morbidity. Little is known about the soluble inflamJarmatory profiles involved in large artery atherosclerosis. We investigated the correlation between aortic calcification in the abdominal aorta and cytokine levels in a cohort of peripheral artery disease patients. Aortic calcification index was measured from computed tomography exams and circulating cytokine levels were analyzed from blood serum samples of 156 consecutive patients prior to invasive treatment of peripheral artery disease. The study included 156 patients (mean age 70.7 years, 64 (41.0%) women). The mean ankle-brachial index (ABI) was 0.64 and the mean aortic calcification index (ACI) was 52.3. ACI was associated with cytokines cutaneous T-cell-attracting chemokine CTACK (ß 23.08, SE 5.22, p < 0.001) and monokine induced by gamma-interferon MIG (ß 9.40, SE 2.82, p 0.001) in univariate linear regression. After adjustment with cardiovascular risk factors, CTACK and MIG were independently associated with ACI, ß 17.9 (SE 5.22, p < 0.001) for CTACK and ß 6.80 (SE 3.33, p 0.043) for MIG. CTACK was significantly higher in the patients representing the highest ACI tertile (highest vs. middle, 7.53 vs. 7.34 Tukeys HSD p-value 0.023 and highest vs. lowest tertile 7.53 vs. 7.29, Tukeys HSD p-value 0.002). MIG was significantly higher in the highest tertile versus lowest (7.65 vs. 7.30, Tukeys HSD p-value 0.027). Cytokines CTACK and MIG are associated with higher ACI, suggesting that CTACK and MIG reflect atherosclerotic disease burden of the aorta. This might further suggest the possible association with other cardiovascular morbidities.


Subject(s)
Ankle Brachial Index , Biomarkers , Cytokines , Peripheral Arterial Disease , Vascular Calcification , Humans , Female , Male , Aged , Peripheral Arterial Disease/blood , Peripheral Arterial Disease/diagnosis , Peripheral Arterial Disease/complications , Cytokines/blood , Vascular Calcification/blood , Vascular Calcification/diagnosis , Vascular Calcification/etiology , Middle Aged , Biomarkers/blood , Aorta, Abdominal/diagnostic imaging , Aged, 80 and over , Tomography, X-Ray Computed , Aortic Diseases/blood , Aortic Diseases/diagnosis , Aortic Diseases/complications , Risk Factors
5.
Int J Mol Sci ; 25(18)2024 Sep 23.
Article in English | MEDLINE | ID: mdl-39337709

ABSTRACT

The burden of chronic kidney disease (CKD) is increasing, posing a serious threat to human health. Cardiovascular calcification (CVC) is one of the most common manifestations of CKD, which significantly influences the morbidity and mortality of patients. The manifestation of CVC is an unusual accumulation of mineral substances containing calcium and phosphate. The main component is hydroxyapatite. Many cells are involved in this process, such as smooth muscle cells (SMCs) and endothelial cells. CVC is an osteogenic process initiated by complex mechanisms such as metabolic disorders of calcium and phosphorus minerals, inflammation, extracellular vesicles, autophagy, and micro-RNAs with a variety of signaling pathways like Notch, STAT, and JAK. Although drug therapy and dialysis technology continue to advance, the survival time and quality of life of CVC patients still face challenges. Therefore, early diagnosis and prevention of CKD-related CVC, reducing its mortality rate, and improving patients' quality of life have become urgent issues in the field of public health. In this review, we try to summarize the state-of-the-art understanding of the progression of CVC and hope that it will help in the prevention and treatment of CVC in CKD.


Subject(s)
Renal Insufficiency, Chronic , Vascular Calcification , Humans , Renal Insufficiency, Chronic/metabolism , Renal Insufficiency, Chronic/complications , Renal Insufficiency, Chronic/pathology , Vascular Calcification/etiology , Vascular Calcification/metabolism , Cardiovascular Diseases/etiology , Cardiovascular Diseases/metabolism , Quality of Life , Animals
6.
PeerJ ; 12: e18063, 2024.
Article in English | MEDLINE | ID: mdl-39308809

ABSTRACT

Vascular calcification is quite common in patients with end-stage chronic kidney disease and is a major trigger for cardiovascular complications in these patients. These complications significantly impact the survival rate and long-term prognosis of individuals with chronic kidney disease. Numerous studies have demonstrated that the development of vascular calcification involves various pathophysiological mechanisms, with the osteogenic transdifferentiation of vascular smooth muscle cells (VSMCs) being of utmost importance. High phosphate levels, bone morphogenetic protein 2 (BMP2), and runt-related transcription factor 2 (RUNX2) play crucial roles in the osteogenic transdifferentiation process of VSMCs. This article primarily reviews the molecular mechanisms by which high phosphate, BMP2, and RUNX2 regulate vascular calcification secondary to chronic kidney disease, and discusses the complex interactions among these factors and their impact on the progression of vascular calcification. The insights provided here aim to offer new perspectives for future research on the phenotypic switching and osteogenic transdifferentiation of VSMCs, as well as to aid in optimizing clinical treatment strategies for this condition, bearing significant clinical and scientific implications.


Subject(s)
Bone Morphogenetic Protein 2 , Core Binding Factor Alpha 1 Subunit , Hyperphosphatemia , Muscle, Smooth, Vascular , Renal Insufficiency, Chronic , Vascular Calcification , Humans , Core Binding Factor Alpha 1 Subunit/metabolism , Core Binding Factor Alpha 1 Subunit/genetics , Vascular Calcification/metabolism , Vascular Calcification/pathology , Vascular Calcification/etiology , Bone Morphogenetic Protein 2/metabolism , Renal Insufficiency, Chronic/metabolism , Renal Insufficiency, Chronic/pathology , Renal Insufficiency, Chronic/complications , Hyperphosphatemia/metabolism , Muscle, Smooth, Vascular/metabolism , Muscle, Smooth, Vascular/pathology , Cell Transdifferentiation , Osteogenesis/physiology , Myocytes, Smooth Muscle/metabolism , Myocytes, Smooth Muscle/pathology
7.
Sheng Li Xue Bao ; 76(4): 526-536, 2024 Aug 25.
Article in Chinese | MEDLINE | ID: mdl-39192786

ABSTRACT

The present study aimed to explore the effect of swimming exercise on vascular calcification in type 2 diabetic rats and its related molecular mechanism. Male Sprague Dawley (SD) rats were randomly divided into normal control (NC), diabetes control (DC) and diabetes+exercise (DE) groups. The DC and DE groups were intraperitoneally injected with streptozotocin (STZ) and fed with high-fat diet to establish type 2 diabetes mellitus model. The NC and DC groups did not exercise, and the DE group performed swimming exercise for 8 weeks. ELISA was used to detect the serum glycated hemoglobin A1c (HbA1c) level. The aortas of rats were taken as sample. Assay kits were used to detect vascular calcium content and alkaline phosphatase (ALP) activity. Von Kossa staining was used to detect calcium deposition. qRT-PCR was used detect the expression of microRNA-145 (miR-145). Western blot was used to detect the protein expression levels of smooth muscle contraction markers, calcification marker and related proteins. The results showed that, compared with the NC group, the blood glucose, serum HbA1c level, vascular calcium content and ALP activity in the DC group were significantly increased, the protein expression levels of smooth muscle contraction markers smooth muscle protein 22α (SM22α) and α-smooth muscle actin (α-SMA) were significantly down-regulated, and the protein expression level of calcification marker osteopontin (OPN) was significantly up-regulated; Compared with the DC group, the serum HbA1c level, vascular calcium content and ALP activity in the DE group were significantly decreased, the protein expression levels of SM22α and α-SMA were significantly up-regulated, and the protein expression level of OPN was significantly down-regulated; Compared with the NC group, the expression of miR-145-5p in the DC group was significantly down-regulated, and the protein expression levels of transforming growth factor-ß (TGF-ß), SMAD2, ERK1/2 and p-ERK1/2 were significantly up-regulated; Compared with the DC group, the expression of miR-145-5p was significantly up-regulated in the DE group, while the expressions of TGF-ß, ERK1/2 and p-ERK1/2 were significantly down-regulated. These results suggest that miR-145/TGF-ß signaling is involved in the improving effects of 8-week swimming exercise on glucose metabolism disorder, vascular smooth muscle cell phenotype switching and vascular calcification in type 2 diabetes mellitus.


Subject(s)
Diabetes Mellitus, Experimental , Diabetes Mellitus, Type 2 , MicroRNAs , Physical Conditioning, Animal , Rats, Sprague-Dawley , Swimming , Vascular Calcification , Animals , Male , Rats , MicroRNAs/metabolism , MicroRNAs/genetics , Vascular Calcification/metabolism , Vascular Calcification/etiology , Diabetes Mellitus, Type 2/metabolism , Diabetes Mellitus, Experimental/metabolism , Swimming/physiology , Transforming Growth Factor beta/metabolism
8.
Mol Med ; 30(1): 130, 2024 Aug 24.
Article in English | MEDLINE | ID: mdl-39182021

ABSTRACT

BACKGROUND: Vascular calcification is a common vascular lesion associated with high morbidity and mortality from cardiovascular events. Antibiotics can disrupt the gut microbiota (GM) and have been shown to exacerbate or attenuate several human diseases. However, whether antibiotic-induced GM disruption affects vascular calcification remains unclear. METHODS: Antibiotic cocktail (ABX) treatment was utilized to test the potential effects of antibiotics on vascular calcification. The effects of antibiotics on GM and serum short-chain fatty acids (SCFAs) in vascular calcification mice were analyzed using 16 S rRNA gene sequencing and targeted metabolomics, respectively. Further, the effects of acetate, propionate and butyrate on vascular calcification were evaluated. Finally, the potential mechanism by which acetate inhibits osteogenic transformation of VSMCs was explored by proteomics. RESULTS: ABX and vancomycin exacerbated vascular calcification. 16 S rRNA gene sequencing and targeted metabolomics analyses showed that ABX and vancomycin treatments resulted in decreased abundance of Bacteroidetes in the fecal microbiota of the mice and decreased serum levels of SCFAs. In addition, supplementation with acetate was found to reduce calcium salt deposition in the aorta of mice and inhibit osteogenic transformation in VSMCs. Finally, using proteomics, we found that the inhibition of osteogenic transformation of VSMCs by acetate may be related to glutathione metabolism and ubiquitin-mediated proteolysis. After adding the glutathione inhibitor Buthionine sulfoximine (BSO) and the ubiquitination inhibitor MG132, we found that the inhibitory effect of acetate on VSMC osteogenic differentiation was weakened by the intervention of BSO, but MG132 had no effect. CONCLUSION: ABX exacerbates vascular calcification, possibly by depleting the abundance of Bacteroidetes and SCFAs in the intestine. Supplementation with acetate has the potential to alleviate vascular calcification, which may be an important target for future treatment of vascular calcification.


Subject(s)
Acetates , Anti-Bacterial Agents , Fatty Acids, Volatile , Gastrointestinal Microbiome , Vascular Calcification , Animals , Gastrointestinal Microbiome/drug effects , Vascular Calcification/metabolism , Vascular Calcification/etiology , Vascular Calcification/drug therapy , Mice , Fatty Acids, Volatile/metabolism , Acetates/pharmacology , Anti-Bacterial Agents/adverse effects , Anti-Bacterial Agents/pharmacology , Male , Osteogenesis/drug effects , RNA, Ribosomal, 16S/genetics , Disease Models, Animal , Mice, Inbred C57BL , Vancomycin/adverse effects , Vancomycin/pharmacology , Muscle, Smooth, Vascular/metabolism , Muscle, Smooth, Vascular/drug effects
9.
J Mol Med (Berl) ; 102(10): 1217-1227, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39136767

ABSTRACT

One of the hallmarks of chronic kidney disease (CKD) is the development of vascular calcification. Inorganic pyrophosphate is a potent inhibitor of calcification, and previous studies have reported low plasma pyrophosphate levels in hemodialysis patients. A long-term mouse model of CKD-accelerated vascular calcification was developed to study pyrophosphate metabolism and to test whether oral pyrophosphate supplementation attenuates the propensity for arterial calcification. CKD was induced by repeated injections of aristolochic acid in wild-type and Abcc6-/- mice, which tend to develop vascular calcifications. CKD accelerated the development of vascular calcifications in Abcc6-/- mice, in the aorta and small renal arteries, and decreased circulating pyrophosphate levels. Oral pyrophosphate supplementation for 6 months attenuated CKD-induced vascular calcification in this model. These results show that oral pyrophosphate may be of interest in preventing vascular calcification in patients with CKD. KEY MESSAGES: Chronic kidney disease accelerates the development of vascular calcification in pyrophosphate-deficient mice. Oral pyrophosphate supplementation for 6 months attenuates chronic kidney disease-induced vascular calcification in a mouse model. Oral pyrophosphate may be of interest in preventing vascular calcification in patients with chronic kidney disease.


Subject(s)
Diphosphates , Disease Models, Animal , Mice, Knockout , Multidrug Resistance-Associated Proteins , Renal Insufficiency, Chronic , Vascular Calcification , Animals , Renal Insufficiency, Chronic/pathology , Renal Insufficiency, Chronic/etiology , Renal Insufficiency, Chronic/prevention & control , Vascular Calcification/etiology , Vascular Calcification/prevention & control , Multidrug Resistance-Associated Proteins/genetics , Multidrug Resistance-Associated Proteins/metabolism , Mice , Male , Administration, Oral , Mice, Inbred C57BL , Aorta/pathology , Aorta/metabolism
10.
Ter Arkh ; 96(6): 593-599, 2024 Jul 07.
Article in Russian | MEDLINE | ID: mdl-39106500

ABSTRACT

AIM: To clarify the role of advanced glycation end products (AGEs) and inflammation in the development of vascular calcification and cardiovascular complications at different stages of chronic kidney disease (CKD) G1-G5D. MATERIALS AND METHODS: We examined 105 patients aged 19 to 75 years with stage C1-C5D CKD, 77 (74%) of whom were patients with diabetic nephropathy. The concentration of AGEs, interleukin (IL)-1, IL-6 and tumor necrosis factor α (TNF-α), troponin I, parathyroid hormone was determined by enzyme-linked immunosorbent assay (ELISA) using kits from BluGene biotech (Shanghai, China), Cloud-Clone Corp. (USA), ELISA Kit (Biomedica, Austria). RESULTS: A high content of AGEs, IL-1, IL-6, TNF-α was established, which directly correlated with the increase in renal failure and changes in the morpho-functional parameters of the left ventricle and aorta. CONCLUSION: An increase in serum concentrations of AGEs and inflammatory mediators, correlating with a decrease in renal function and changes in the morpho- functional parameters of the left ventricle and aorta, indicate their significant role in the processes of damage to the cardiovascular system in CKD.


Subject(s)
Glycation End Products, Advanced , Inflammation , Renal Insufficiency, Chronic , Vascular Calcification , Humans , Glycation End Products, Advanced/blood , Middle Aged , Male , Female , Renal Insufficiency, Chronic/blood , Renal Insufficiency, Chronic/diagnosis , Renal Insufficiency, Chronic/complications , Vascular Calcification/etiology , Vascular Calcification/blood , Adult , Inflammation/blood , Cardiovascular Diseases/etiology , Aged
11.
Kidney Int ; 106(4): 611-624, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39084258

ABSTRACT

Medial vascular calcification in chronic kidney disease (CKD) involves pro-inflammatory pathways induced by hyperphosphatemia. Several interleukin 6 family members have been associated with pro-calcific effects in vascular smooth muscle cells (VSMCs) and are considered as therapeutic targets. Therefore, we investigated the role of leukemia inhibitory factor (LIF) during VSMC calcification. LIF expression was found to be increased following phosphate exposure of VSMCs. LIF supplementation aggravated, while silencing of endogenous LIF or LIF receptor (LIFR) ameliorated the pro-calcific effects of phosphate in VSMCs. The soluble LIFR mediated antagonistic effects towards LIF and reduced VSMC calcification. Mechanistically, LIF induced phosphorylation of the non-receptor tyrosine-protein kinase 2 (TYK2) and signal transducer and activator of transcription-3 (STAT3) in VSMCs. TYK2 inhibition by deucravacitinib, a selective, allosteric oral immunosuppressant used in psoriasis treatment, not only blunted the effects of LIF, but also interfered with the pro-calcific effects induced by phosphate. Conversely, TYK2 overexpression aggravated VSMC calcification. Ex vivo calcification of mouse aortic rings was ameliorated by Tyk2 pharmacological inhibition and genetic deficiency. Cholecalciferol-induced vascular calcification in mice was improved by Tyk2 inhibition and in the Tyk2-deficient mice. Similarly, calcification was ameliorated in Abcc6/Tyk2-deficient mice after adenine/high phosphorus-induced CKD. Thus, our observations indicate a role for LIF in CKD-associated vascular calcification. Hence, the effects of LIF identify a central pro-calcific role of TYK2 signaling, which may be a future target to reduce the burden of vascular calcification in CKD.


Subject(s)
Leukemia Inhibitory Factor , Muscle, Smooth, Vascular , Myocytes, Smooth Muscle , Renal Insufficiency, Chronic , Signal Transduction , TYK2 Kinase , Vascular Calcification , Animals , Humans , Male , Mice , Cells, Cultured , Disease Models, Animal , Leukemia Inhibitory Factor/metabolism , Leukemia Inhibitory Factor/genetics , Leukemia Inhibitory Factor Receptor alpha Subunit/metabolism , Leukemia Inhibitory Factor Receptor alpha Subunit/genetics , Mice, Inbred C57BL , Mice, Knockout , Muscle, Smooth, Vascular/pathology , Muscle, Smooth, Vascular/metabolism , Muscle, Smooth, Vascular/drug effects , Myocytes, Smooth Muscle/drug effects , Myocytes, Smooth Muscle/metabolism , Myocytes, Smooth Muscle/pathology , Phosphates/metabolism , Phosphorylation , Renal Insufficiency, Chronic/metabolism , Renal Insufficiency, Chronic/pathology , STAT3 Transcription Factor/metabolism , TYK2 Kinase/metabolism , TYK2 Kinase/genetics , Vascular Calcification/pathology , Vascular Calcification/metabolism , Vascular Calcification/etiology , Vascular Calcification/genetics
12.
Calcif Tissue Int ; 115(4): 339-361, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39078512

ABSTRACT

Kidney transplantation is the most effective treatment option for most patients with end-stage kidney disease due to reduced mortality, decreased cardiovascular events and increased quality of life compared to patients treated with dialysis. However, kidney transplantation is not devoid of both acute and chronic complications including mineral bone disorders (MBD) which are already present in patients with chronic kidney disease (CKD) before kidney transplantation. The natural history of MBD after kidney transplantation is variable and new markers are needed to define MBD after kidney transplantation. One of these promising molecules is sclerostin. The main action of sclerostin is to inhibit bone formation and mineralization by blocking osteoblast differentiation and function. In kidney transplant recipients (KTRs), various studies have shown that sclerostin is associated with graft function, bone parameters, vascular calcification, and arterial stiffness although non-uniformly. Furthermore, data for inhibition of sclerostin with monoclonal antibody romosozumab for treatment of osteoporosis is available for general population but not in KTRs which osteoporosis is highly prevalent. In this narrative review, we have summarized the studies investigating the change of sclerostin before and after kidney transplantation, the relationship between sclerostin and laboratory parameters, bone metabolism and vascular calcification in the context of kidney transplantation. We also pointed out the uncertainties, explained the causes of divergent findings and suggest further potential study topics regarding sclerostin in kidney transplantation.


Subject(s)
Adaptor Proteins, Signal Transducing , Kidney Transplantation , Humans , Bone Diseases, Metabolic/etiology , Genetic Markers , Vascular Calcification/etiology , Chronic Kidney Disease-Mineral and Bone Disorder/blood
13.
Ren Fail ; 46(2): 2368091, 2024 Dec.
Article in English | MEDLINE | ID: mdl-39049724

ABSTRACT

Recent studies have shown that microRNA-16-5p (miR-16-5p) plays a crucial role in the pathological mechanism of vascular calcification. Nevertheless, the expression profile of miR-16-5p in maintenance hemodialysis (MHD) patients who are predisposed to vascular calcification remains unknown. This study aims to investigate the potential associations between calcification risk and serum miR-16-5p expression among MHD patients. This cross-sectional study involved 132 MHD patients from the Dialysis Center of Beijing Friendship Hospital between 1 January 2019 and 31 December 2020. The degree of calcification in MHD patients was assessed using the Abdominal aortic calcification (AAC) score, and miR-16-5p expression was quantified using quantitative real-time polymerase chain reaction (qRT-PCR) with the 2-ΔΔCT method. Statistical analyses, including spearman correlation, linear regression and logistic regression analysis were used to explore the associations between laboratory parameters and AAC score. Calcifications were observed in 79(59.80%) patients. The linear regression showed a one-quartile decrease in miR-16-5p expression led to a significant increase in the AAC score by 5.336 (95% CI: 2.670-10.662, p = 0.000). Multivariate logistic regression analyses revealed that decreased miR-16-5p expression, reduced serum urea nitrogen, elevated white blood cell count, and longer dialysis vintage were significantly associated with an increased incidence of vascular calcification. The Area Under the Curve (AUC) of the Receiver Operating Characteristic (ROC) of the miR-16-5p-based logistic regression model was 0.842 (95% CI: 0.771-0.913, p = 0.000). There was an independent association between miR-16-5p expression and calcification degree. Lower miR-16-5p expression levels seem to be a potential risk factor of vascular calcification in MHD patients.


Subject(s)
Aorta, Abdominal , MicroRNAs , Renal Dialysis , Vascular Calcification , Humans , MicroRNAs/blood , Male , Female , Renal Dialysis/adverse effects , Vascular Calcification/blood , Vascular Calcification/etiology , Middle Aged , Aorta, Abdominal/pathology , Aorta, Abdominal/diagnostic imaging , Cross-Sectional Studies , Aged , Kidney Failure, Chronic/therapy , Kidney Failure, Chronic/blood , Kidney Failure, Chronic/complications , ROC Curve , Risk Factors , Logistic Models
14.
Gerontology ; 70(8): 858-875, 2024.
Article in English | MEDLINE | ID: mdl-38824923

ABSTRACT

INTRODUCTION: Hypertension can accelerate and aggravate the process of arterial ageing and calcification. However, the mechanism behind has yet to be well elucidated. METHODS: Here, we monitored the dynamic changes of fibronectin (FN)/α5 integrin, bone morphogenetic protein 2/matrix Gla protein (BMP2/MGP), and Runx2 in the aorta of spontaneously hypertensive rats (SHRs) and thoracic aortic vascular smooth muscle cells (VSMCs), also the phenotypic transformation of VSMCs during the process of arterial ageing and calcification. Further, study on arterial ageing and calcification through antagonist experiments at the molecular level was explored. RESULTS: We found extracellular FN and its α5 integrin receptor expressions were positively associated with arterial ageing and calcification in SHR during ageing, as well in VSMCs from SHR in vitro. Integrin receptor inhibitor of GRGDSP would delay this arterial ageing and calcification process. Moreover, the elevated FN and α5 integrin receptor expression evoked the disequilibrium of BMP2/MGP, where the expression of BMP2, a potent osteogenic inducer, increased while MGP, a calcification inhibitor, decreased. Furthermore, it was followed by the upregulation of Runx2 and the phenotypic transformation of VSMCs from the contractile phenotype into the osteoblast-like cells. Notably, BMP2 antagonist of rmNoggin was sufficient to ameliorate the ageing and calcification process of VSMCs and exogenous BMP2-adding accelerate and aggregate the process. CONCLUSION: Our study revealed that hypertension-associated arterial ageing and calcification might be a consequence that hypertension up-regulated FN and its high binding affinity integrin α5 receptor in the aortic wall, which in turn aggravated the imbalance of BMP2/MGP, promoted the transcription of Runx2, and induced the phenotypic transformation of VSMCs from the contractile phenotype into the osteoblast-like cells. Our study would provide insights into hypertension-associated arterial ageing and calcification and shed new light on the control of arterial calcification, especially for those with hypertension.


Subject(s)
Aging , Bone Morphogenetic Protein 2 , Core Binding Factor Alpha 1 Subunit , Fibronectins , Hypertension , Matrix Gla Protein , Muscle, Smooth, Vascular , Phenotype , Rats, Inbred SHR , Vascular Calcification , Bone Morphogenetic Protein 2/metabolism , Animals , Muscle, Smooth, Vascular/metabolism , Muscle, Smooth, Vascular/pathology , Hypertension/metabolism , Hypertension/physiopathology , Rats , Fibronectins/metabolism , Core Binding Factor Alpha 1 Subunit/metabolism , Aging/metabolism , Aging/physiology , Vascular Calcification/metabolism , Vascular Calcification/pathology , Vascular Calcification/etiology , Male , Extracellular Matrix Proteins/metabolism , Myocytes, Smooth Muscle/metabolism , Calcium-Binding Proteins/metabolism , Integrin alpha5/metabolism , Integrin alpha5/genetics , Cells, Cultured
15.
Diabetes Obes Metab ; 26(9): 3860-3867, 2024 Sep.
Article in English | MEDLINE | ID: mdl-38934214

ABSTRACT

AIM: The association of overweight/obesity and metabolic dysfunction-associated steatotic liver disease (MASLD) in young adulthood with subclinical atherosclerosis [coronary artery calcification (CAC) and abdominal aortic calcification (AAC)] by middle age is unknown. METHOD: In total, 2274 participants aged 28-39 years from the coronary artery risk development in young adults study at year 10 (1995-1996) who were re-examined 15 years later were included. CAC and AAC were measured at year 25 using computed tomography. We examined the utility of three young adult phenotypes (lean group; overweight/obese group; overweight/obese MASLD group) at year 10 in predicting CAC or AAC by middle age. Modified Poisson regression was used to estimate the association between groups and CAC, and AAC. Independent determinates of CAC and AAC were determined with linear regression models. RESULTS: Compared with individuals categorized as lean in young adulthood, the relative risk for CAC by middle age was 1.09 (95% confidence interval: 0.93-1.28) for those with overweight/obesity and 1.32 (95% confidence interval: 1.08-1.61) for those with overweight/obesity-related MASLD. For AAC, no difference was observed between these three groups. Group, systolic blood pressure and group × systolic blood pressure interaction were all the independent determinates for CAC. CONCLUSION: In this study, young adults with overweight/obesity-related MASLD have a higher risk of developing CAC by middle age. These abnormalities are only partially explained by traditional cardiovascular risk factors, and overweight/obesity-related MASLD has an independent impact on CAC. Our study provides evidence for identifying young adults at higher risk of developing subclinical atherosclerosis.


Subject(s)
Coronary Artery Disease , Obesity , Overweight , Vascular Calcification , Humans , Male , Female , Adult , Coronary Artery Disease/epidemiology , Coronary Artery Disease/etiology , Coronary Artery Disease/diagnostic imaging , Vascular Calcification/epidemiology , Vascular Calcification/etiology , Vascular Calcification/diagnostic imaging , Vascular Calcification/complications , Obesity/complications , Overweight/complications , Fatty Liver/complications , Fatty Liver/epidemiology , Risk Factors , Middle Aged , Aorta, Abdominal/diagnostic imaging , Aorta, Abdominal/pathology , Young Adult , Non-alcoholic Fatty Liver Disease/complications , Non-alcoholic Fatty Liver Disease/epidemiology
16.
Nephrology (Carlton) ; 29(10): 663-670, 2024 Oct.
Article in English | MEDLINE | ID: mdl-38866394

ABSTRACT

AIM: This research aimed to explore the serum levels of solute carrier family 7 member 11 (SLC7A11) in patients with maintenance peritoneal dialysis (MPD) and its correlation with vascular calcification (VC) and clinical results. METHODS: This present prospective observational cohort study enrolled 189 patients with MPD who were undergoing regular peritoneal dialysis for over 3 months in our hospital from February 2020 to July 2022. The abdominal aortic calcification score was used to assess the VC condition of MPD patients. The serum SLC7A11, interleukin (IL)-6, IL-1ß and C-reactive protein levels were measured by enzyme-linked immunosorbent assay (ELISA). Demographic and clinical statistics were collected. All patients were followed up for 1 year and the overall survival time (OS) of all patients were recorded. All data used SPSS 18.0 for statistical analyses. RESULTS: Patients with moderate/severe calcification in MPD had a longer duration of dialysis, higher serum levels of phosphate (P) and calcium (Ca) and lower serum levels of SLC7A11. Spearman's analysis revealed a negative correlation between serum SLC7A11 levels and the levels of P, Ca and IL-1ß. Additionally, we observed an association between serum SLC7A11 levels and clinical prognosis as well as the extent of VC in MPD patients. Multivariate logistic regression analysis indicated that dialysis duration, SLC7A11, and P were risk factors for VC in MPD patients. CONCLUSION: The serum SLC7A11 levels decreased remarkably in MPD patients with moderate/severe calcification. This study may provide new targets and comprehensive approach to cardiovascular protection in patients with chronic kidney disease.


Subject(s)
Peritoneal Dialysis , Vascular Calcification , Humans , Vascular Calcification/blood , Vascular Calcification/etiology , Vascular Calcification/diagnosis , Peritoneal Dialysis/adverse effects , Male , Female , Middle Aged , Prospective Studies , Biomarkers/blood , Aged , Prognosis
17.
Nutrients ; 16(12)2024 Jun 07.
Article in English | MEDLINE | ID: mdl-38931153

ABSTRACT

Patients with chronic kidney disease (CKD) suffer disproportionately from a high burden of cardiovascular disease, which, despite recent scientific advances, remains partly understood. Vascular calcification (VC) is the result of an ongoing process of misplaced calcium in the inner and medial layers of the arteries, which has emerged as a critical contributor to cardiovascular events in CKD. Beyond its established role in blood clotting and bone health, vitamin K appears crucial in regulating VC via vitamin K-dependent proteins (VKDPs). Among these, the matrix Gla protein (MGP) serves as both a potent inhibitor of VC and a valuable biomarker (in its inactive form) for reflecting circulating vitamin K levels. CKD patients, especially in advanced stages, often present with vitamin K deficiency due to dietary restrictions, medications, and impaired intestinal absorption in the uremic environment. Epidemiological studies confirm a strong association between vitamin K levels, inactive MGP, and increased CVD risk across CKD stages. Based on the promising results of pre-clinical data, an increasing number of clinical trials have investigated the potential benefits of vitamin K supplementation to prevent, delay, or even reverse VC, but the results have remained inconsistent.


Subject(s)
Extracellular Matrix Proteins , Matrix Gla Protein , Renal Insufficiency, Chronic , Vascular Calcification , Vitamin K Deficiency , Vitamin K , Humans , Vascular Calcification/etiology , Renal Insufficiency, Chronic/complications , Vitamin K Deficiency/complications , Extracellular Matrix Proteins/blood , Extracellular Matrix Proteins/metabolism , Calcium-Binding Proteins/blood , Dietary Supplements , Cardiovascular Diseases/etiology , Cardiovascular Diseases/prevention & control , Biomarkers/blood
18.
BMC Nephrol ; 25(1): 157, 2024 May 07.
Article in English | MEDLINE | ID: mdl-38714960

ABSTRACT

BACKGROUND: This study aims to investigate the influencing factors of vascular calcification in peritoneal dialysis (PD) patients and its relationship with long-term prognosis. METHODS: This retrospective cohort study included chronic kidney disease patients undergoing peritoneal dialysis at the Peritoneal Dialysis Center of Beijing Luhu Hospital, Capital Medical University, from January 2019 to March 2019. Demographic and clinical laboratory data, including serum sclerostin (SOST), calcium (Ca), phosphate (P), serum albumin (ALB), and intact parathyroid hormone (iPTH) levels, were collected. Abdominal aortic calcification (AAC) was assessed using abdominal lateral X-ray examination to determine the occurrence of vascular calcification, and patients were divided into the AAC group and Non-AAC group based on the results. RESULTS: A total of 91 patients were included in the study. The AAC group consisted of 46 patients, while the Non-AAC group consisted of 45 patients. The AAC group had significantly older patients compared to the non-AAC group (P < 0.001) and longer dialysis time (P = 0.004). Multivariable logistic regression analysis indicated that risk factors for vascular calcification in PD patients included dialysis time, diabetes, hypertension, and SOST. Kaplan-Meier survival analysis showed that the AAC group had a significantly higher mortality rate than the non-AAC group (χ2 = 35.993, P < 0.001). Multivariable Cox regression analysis revealed that dialysis time, diabetes and AAC were risk factors for all-cause mortality in peritoneal dialysis patients. CONCLUSION: Longer dialysis time, comorbid diabetes, comorbid hypertension, and SOST are risk factors for vascular calcification in PD patients. Additionally, AAC, longer dialysis time, and comorbid diabetes are associated with increased risk of all-cause mortality in peritoneal dialysis patients.


Subject(s)
Peritoneal Dialysis , Renal Insufficiency, Chronic , Vascular Calcification , Vascular Calcification/diagnosis , Vascular Calcification/etiology , Peritoneal Dialysis/adverse effects , Prognosis , Retrospective Studies , Renal Insufficiency, Chronic/therapy , Humans , Male , Female , Middle Aged , Aged
19.
Redox Biol ; 73: 103183, 2024 07.
Article in English | MEDLINE | ID: mdl-38759418

ABSTRACT

AIMS: Vascular calcification is strongly linked to the development of major adverse cardiovascular events, but effective treatments are lacking. Sodium-glucose cotransporter 2 (SGLT2) inhibitors are an emerging category of oral hypoglycemic drugs that have displayed marked effects on metabolic and cardiovascular diseases, including recently reported vascular medial calcification. However, the roles and underlying mechanisms of SGLT2 inhibitors in vascular calcification have not been fully elucidated. Thus, we aimed to further determine whether SGLT2 inhibitors protect against vascular calcification and to investigate the mechanisms involved. METHODS AND RESULTS: A computed tomography angiography investigation of coronary arteries from 1554 patients with type 2 diabetes revealed that SGLT2 inhibitor use was correlated with a lower Agatston calcification score. In the vitamin D3 overdose, 5/6 nephrectomy chronic kidney disease-induced medial calcification and Western diet-induced atherosclerotic intimal calcification models, dapagliflozin (DAPA) substantially alleviated vascular calcification in the aorta. Furthermore, we showed that DAPA reduced vascular calcification via Runx2-dependent osteogenic transdifferentiation in vascular smooth muscle cells (VSMCs). Transcriptome profiling revealed that thioredoxin domain containing 5 (TXNDC5) was involved in the attenuation of vascular calcification by DAPA. Rescue experiments showed that DAPA-induced TXNDC5 downregulation in VSMCs blocked the protective effect on vascular calcification. Furthermore, TXNDC5 downregulation disrupted protein folding-dependent Runx2 stability and promoted subsequent proteasomal degradation. Moreover, DAPA downregulated TXNDC5 expression via amelioration of oxidative stress and ATF6-dependent endoplasmic reticulum stress. Consistently, the class effects of SGLT2 inhibitors on vascular calcification were validated with empagliflozin in intimal and medial calcification models. CONCLUSIONS: SGLT2 inhibitors ameliorate vascular calcification through blocking endoplasmic reticulum stress-dependent TXNDC5 upregulation and promoting subsequent Runx2 proteasomal degradation, suggesting that SGLT2 inhibitors are potentially beneficial for vascular calcification treatment and prevention.


Subject(s)
Glucosides , Osteogenesis , Sodium-Glucose Transporter 2 Inhibitors , Vascular Calcification , Vascular Calcification/metabolism , Vascular Calcification/drug therapy , Vascular Calcification/pathology , Vascular Calcification/etiology , Sodium-Glucose Transporter 2 Inhibitors/pharmacology , Animals , Humans , Osteogenesis/drug effects , Mice , Glucosides/pharmacology , Male , Thioredoxins/metabolism , Thioredoxins/genetics , Benzhydryl Compounds/pharmacology , Diabetes Mellitus, Type 2/metabolism , Diabetes Mellitus, Type 2/complications , Diabetes Mellitus, Type 2/drug therapy , Endoplasmic Reticulum/metabolism , Endoplasmic Reticulum/drug effects , Rats , Core Binding Factor Alpha 1 Subunit/metabolism , Core Binding Factor Alpha 1 Subunit/genetics , Disease Models, Animal , Muscle, Smooth, Vascular/metabolism , Muscle, Smooth, Vascular/drug effects , Muscle, Smooth, Vascular/pathology , Muscle, Smooth, Vascular/cytology , Myocytes, Smooth Muscle/metabolism , Myocytes, Smooth Muscle/drug effects , Endoplasmic Reticulum Stress/drug effects , Female
20.
Mol Med ; 30(1): 58, 2024 May 08.
Article in English | MEDLINE | ID: mdl-38720283

ABSTRACT

BACKGROUND: Vascular calcification (VC) is a complication in diabetes mellitus (DM) patients. Osteogenic phenotype switching of vascular smooth muscle cells (VSMCs) plays a critical role in diabetes-related VC. Mitophagy can inhibit phenotype switching in VSMCs. This study aimed to investigate the role of the glucagon-like peptide-1 receptor (GLP-1R) agonist exendin 4 (EX4) in mitophagy-induced phenotype switching. MATERIALS AND METHODS: The status of VC in T2DM mice was monitored using Von Kossa and Alizarin Red S (ARS) staining in mouse aortic tissue. Human aortic smooth muscle cells were cultured in high glucose (HG) and ß-glycerophosphate (ß-GP) conditioned medium. Accumulation of LC3B and p62 was detected in the mitochondrial fraction. The effect of EX4 in vitro and in vivo was investigated by knocking down AMPKα1. RESULTS: In diabetic VC mice, EX4 decreased the percentage of von Kossa/ARS positive area. EX4 inhibited osteogenic differentiation of HG/ß-GP-induced VSMCs. In HG/ß-GP-induced VSMCs, the number of mitophagosomes was increased, whereas the addition of EX4 restored mitochondrial function, increased the number of mitophagosome-lysosome fusions, and reduced p62 in mitochondrial frictions. EX4 increased the phosphorylation of AMPKα (Thr172) and ULK1 (Ser555) in HG/ß-GP-induced VSMCs. After knockdown of AMPKα1, ULK1 could not be activated by EX4. The accumulation of LC3B and p62 could not be reduced after AMPKα1 knockdown. Knockdown of AMPKα1 negated the therapeutic effects of EX4 on VC of diabetic mice. CONCLUSION: EX4 could promote mitophagy by activating the AMPK signaling pathway, attenuate insufficient mitophagy, and thus inhibit the osteogenic phenotype switching of VSMCs.


Subject(s)
AMP-Activated Protein Kinases , Exenatide , Glucagon-Like Peptide-1 Receptor , Mitophagy , Signal Transduction , Vascular Calcification , Animals , Mitophagy/drug effects , Vascular Calcification/etiology , Vascular Calcification/metabolism , Vascular Calcification/drug therapy , Signal Transduction/drug effects , Mice , Glucagon-Like Peptide-1 Receptor/agonists , Glucagon-Like Peptide-1 Receptor/metabolism , Male , AMP-Activated Protein Kinases/metabolism , Humans , Exenatide/pharmacology , Exenatide/therapeutic use , Muscle, Smooth, Vascular/metabolism , Muscle, Smooth, Vascular/drug effects , Muscle, Smooth, Vascular/pathology , Myocytes, Smooth Muscle/metabolism , Myocytes, Smooth Muscle/drug effects , Diabetes Mellitus, Type 2/complications , Diabetes Mellitus, Type 2/metabolism , Diabetes Mellitus, Type 2/drug therapy , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Experimental/complications , Diabetes Mellitus, Experimental/drug therapy , Disease Models, Animal , Mice, Inbred C57BL
SELECTION OF CITATIONS
SEARCH DETAIL