1
|
Shi W, Xie X, Zhao Y, Liu Y, Zhang X. Characteristics and prognostic values of abdominal aortic branches calcification in hemodialysis patients. Ren Fail 2025; 47:2432538. [PMID: 39763079 PMCID: PMC11721613 DOI: 10.1080/0886022x.2024.2432538] [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: 05/12/2024] [Revised: 10/16/2024] [Accepted: 11/17/2024] [Indexed: 01/12/2025] Open
Abstract
BACKGROUND Vascular calcification is highly prevalent and associated with mortality in hemodialysis patients. However, extreme splanchnic arterial calcification in calciphylaxis with poor prognosis raises questions regarding the reliability of previous vascular calcification scoring methods. Therefore, this study aimed to examine the distribution characteristics of abdominal aortic branch calcification and identify a more reliable predictor of mortality in hemodialysis patients. METHODS The cohort study included 237 hemodialysis patients. The distribution characteristics of abdominal aortic branch calcification were determined by quantifying the calcification volumes. The primary and secondary outcomes were all-cause mortality and new-onset cardiovascular events, respectively. We compared the prognostic values of abdominal aortic branch calcification and constructed a predictive nomogram model. RESULTS The prevalence of abdominal vascular calcification in hemodialysis patients was 95.36%, with the highest prevalence in the abdominal aorta (88.61%) and internal iliac artery (85.65%). During a median follow-up period of 3.92 years, 137 patients died. Internal iliac artery and mesenteric artery calcification showed the greatest predictive values for mortality. Internal iliac artery calcification and serum albumin level were independently associated with mortality in hemodialysis patients (p < .001). The nomogram model constructed with internal iliac artery calcification, serum albumin level, age, and comorbid cardiovascular disease was well discriminative, calibrated, and clinically applicable for predicting 3-year survival. CONCLUSION Abdominal aortic branch calcification, particularly internal iliac artery calcification, is a preferable prognostic predictor than abdominal aorta or coronary artery calcification in hemodialysis patients.
Collapse
Affiliation(s)
- Wen Shi
- Institute of Nephrology, Zhong Da Hospital, Southeast University School of Medicine, Nanjing, Jiangsu, China
| | - Xiaotong Xie
- Institute of Nephrology, Zhong Da Hospital, Southeast University School of Medicine, Nanjing, Jiangsu, China
| | - Yu Zhao
- Institute of Nephrology, Zhong Da Hospital, Southeast University School of Medicine, Nanjing, Jiangsu, China
| | - Yuqiu Liu
- Institute of Nephrology, Zhong Da Hospital, Southeast University School of Medicine, Nanjing, Jiangsu, China
| | - Xiaoliang Zhang
- Institute of Nephrology, Zhong Da Hospital, Southeast University School of Medicine, Nanjing, Jiangsu, China
| |
Collapse
|
2
|
Cosenso-Martin LN, Souza RDM, Uyemura JRR, da Silva Lopes V, Fernandes LAB, de Oliveira KA, Spaziani AO, Yugar-Toledo JC, Silva MAV, Vilela-Martin JF. Increased arterial stiffness in normotensive individuals with hypoparathyroidism. Sci Rep 2025; 15:8817. [PMID: 40087339 PMCID: PMC11909248 DOI: 10.1038/s41598-025-92708-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2024] [Accepted: 03/03/2025] [Indexed: 03/17/2025] Open
Abstract
Hypoparathyroidism is the inability of parathyroid hormone (PTH) to maintain calcium homeostasis. Patients with post-surgical hypoparathyroidism may have an increased risk of mortality; there is clinical and molecular evidence of the effects of this condition on the cardiovascular system. The aim of this study was to evaluate arterial stiffness by measuring the carotid-femoral pulse wave velocity (PWV) in post-surgical hypoparathyroidism patients. A cross-sectional study was conducted with 30 post-surgical hypoparathyroidism patients and 25 volunteers from the Endocrinology Outpatient Clinic of the Medical School. The SphygmoCor system was used to evaluate arterial stiffness by analyzing the PWV. The mean ages of the hypoparathyroidism (50.4 years) and control individuals (49.6 years) were similar. The mean PWVs were 8.7 and 7.5 m/s in the Hypoparathyroidism and Control groups, respectively (p-value = 0.084). Considering only normotensive patients, PWV was statistically higher in the Hypoparathyroidism Group (7.6 versus 6.5 m/s; p-value = 0.039). For this group, serum ionized calcium, phosphorus, and the calcium x phosphorus product levels were positively associated to PWV. Hypoparathyroidism increases arterial stiffness as assessed by PWV. Serum ionized calcium, phosphorus, and the calcium x phosphorus product are affected. A more effective investigative and therapeutic approach for patients with hypoparathyroidism can help control cardiovascular risk.
Collapse
Affiliation(s)
- Luciana N Cosenso-Martin
- Internal Medicine Department, Faculdade de Medicina de São José do Rio Preto (FAMERP), Ave Brig Farila Lima 5416, São José do Rio Preto, São Paulo, 15090-000, Brazil
| | - Rodrigo Duart Martins Souza
- Internal Medicine Department, Faculdade de Medicina de São José do Rio Preto (FAMERP), Ave Brig Farila Lima 5416, São José do Rio Preto, São Paulo, 15090-000, Brazil
| | - Jessica R Roma Uyemura
- Internal Medicine Department, Faculdade de Medicina de São José do Rio Preto (FAMERP), Ave Brig Farila Lima 5416, São José do Rio Preto, São Paulo, 15090-000, Brazil
| | - Valquíria da Silva Lopes
- Internal Medicine Department, Faculdade de Medicina de São José do Rio Preto (FAMERP), Ave Brig Farila Lima 5416, São José do Rio Preto, São Paulo, 15090-000, Brazil
| | - Letícia A Barufi Fernandes
- Internal Medicine Department, Faculdade de Medicina de São José do Rio Preto (FAMERP), Ave Brig Farila Lima 5416, São José do Rio Preto, São Paulo, 15090-000, Brazil
| | - Kleber Aparecido de Oliveira
- Internal Medicine Department, Faculdade de Medicina de São José do Rio Preto (FAMERP), Ave Brig Farila Lima 5416, São José do Rio Preto, São Paulo, 15090-000, Brazil
| | - Amanda Oliva Spaziani
- Internal Medicine Department, Faculdade de Medicina de São José do Rio Preto (FAMERP), Ave Brig Farila Lima 5416, São José do Rio Preto, São Paulo, 15090-000, Brazil
| | - Juan Carlos Yugar-Toledo
- Internal Medicine Department, Faculdade de Medicina de São José do Rio Preto (FAMERP), Ave Brig Farila Lima 5416, São José do Rio Preto, São Paulo, 15090-000, Brazil
| | - Marco Antonio Vieira Silva
- Internal Medicine Department, Faculdade de Medicina de São José do Rio Preto (FAMERP), Ave Brig Farila Lima 5416, São José do Rio Preto, São Paulo, 15090-000, Brazil
| | - José Fernando Vilela-Martin
- Internal Medicine Department, Faculdade de Medicina de São José do Rio Preto (FAMERP), Ave Brig Farila Lima 5416, São José do Rio Preto, São Paulo, 15090-000, Brazil.
| |
Collapse
|
3
|
Salera D, Merkel N, Bellasi A, de Borst MH. Pathophysiology of chronic kidney disease-mineral bone disorder (CKD-MBD): from adaptive to maladaptive mineral homeostasis. Clin Kidney J 2025; 18:i3-i14. [PMID: 40083952 PMCID: PMC11903091 DOI: 10.1093/ckj/sfae431] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Indexed: 03/16/2025] Open
Abstract
Chronic kidney disease-mineral bone disorder (CKD-MBD) is a multifaceted condition commonly seen in people with reduced kidney function. It involves a range of interconnected issues in mineral metabolism, bone health and cardiovascular calcification, which are linked to a lower quality of life and shorter life expectancy. Although various epidemiological studies show that the laboratory changes defining CKD-MBD become more common as the glomerular filtration rate declines, the pathophysiology of CKD-MBD is still largely unexplained. We herein review the current understanding of CKD-MBD, provide a conceptual framework to understand this syndrome, and review the genetic and environmental factors that may influence the clinical manifestation of CKD-MBD. However, a deeper understanding of the pathophysiology of CKD-MBD is needed to understand the phenotype variability and the relative contribution to organ damage of factors involved in CKD-MBD to develop more effective interventions to improve outcomes in patients with CKD.
Collapse
Affiliation(s)
- Davide Salera
- Service of Nephrology, Ospedale Regionale di Lugano, Ospedale Civico, Ente Ospedaliero Cantonale, Lugano, Switzerland
| | - Nathalie Merkel
- Service of Nephrology, Ospedale Regionale di Lugano, Ospedale Civico, Ente Ospedaliero Cantonale, Lugano, Switzerland
| | - Antonio Bellasi
- Service of Nephrology, Ospedale Regionale di Lugano, Ospedale Civico, Ente Ospedaliero Cantonale, Lugano, Switzerland
- Università della Svizzera italiana (USi), Faculty of Biomedical Sciences, Lugano, Switzerland
| | - Martin H de Borst
- Department of Internal Medicine, Division of Nephrology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| |
Collapse
|
4
|
Iseri K, Hida N. Risk factors for osteonecrosis of the jaw in patients with chronic kidney disease: a nested case-control study. J Bone Miner Res 2025; 40:262-269. [PMID: 39656914 DOI: 10.1093/jbmr/zjae193] [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: 08/14/2024] [Revised: 11/11/2024] [Accepted: 12/02/2024] [Indexed: 12/17/2024]
Abstract
Osteonecrosis of the jaw (ONJ) is a severe disease leading to decreased quality of life, but risk factors for ONJ in chronic kidney disease (CKD) patients remain unclear. We conducted a nested case-control study using a large Japanese administrative database to investigate. CKD patients were identified based on estimated glomerular filtration rate (eGFR) measurements, and ONJ cases were identified using ICD-10 codes and diagnostic terms. Controls were matched 1:4 by age and sex. Among 597 026 CKD patients, 75 ONJ cases were identified during a median follow-up of 2.9 yr (incidence rate: 3.27 per 100 000 patient-years). A total of 375 patients (250 males, 66.7%) with a median age of 72 yr (interquartile range (IQR), 64-78) were included after matching controls. The use of bisphosphonates and denosumab for tumor-related disorders in the case group was significantly higher compared to the control group. There was no significant association between kidney functions and the ONJ risk. Multivariate analysis revealed that anti-resorptive drugs for tumor-related disorders [odds ratio (OR): 74.74, 95% confidence interval (CI): 8.81-634.39, p<.001] and oral corticosteroids (OR: 13.23, 95% CI: 3.34-52.33, p<.001) were significantly associated with increased ONJ risk, while anti-resorptive drugs for osteoporosis and injectable corticosteroid use were not. Other relevant factors such as diabetes, liver disease, anabolic drugs, and radiation therapy did not have a significant association with ONJ risk. When stratified by indications for bisphosphonate use (known to be eliminated by renal excretion), bisphosphonate use for tumor-related disorders showed a significant association with ONJ risk (OR: 27.80, 95% CI: 2.47-313.29, p<.01), while bisphosphonates use for osteoporosis did not (OR: 0.74, 95% CI: 0.19-2.92, p=.67). These findings suggest that anti-resorptive drugs for tumor-related disorders and oral corticosteroids are associated with ONJ risk in CKD patients. Heightened surveillance may be necessary for CKD patients receiving these treatments to prevent or detect ONJ early.
Collapse
Affiliation(s)
- Ken Iseri
- Division of Clinical Research and Development, Department of Clinical Pharmacy, School of Pharmacy, Showa University, Tokyo 142-8666, Japan
- Jinsei-kai Kasai Dialysis Clinic, Tokyo 134-0083, Japan
| | - Noriko Hida
- Division of Clinical Research and Development, Department of Clinical Pharmacy, School of Pharmacy, Showa University, Tokyo 142-8666, Japan
| |
Collapse
|
5
|
Shen Y, Huang H, Shen L, Yao W, Wang R, Kang M, Huang J, Xie Y, Yang H. ZBTB16 DRIVES VASCULAR CALCIFICATION THROUGH ACCELERATING VSMCS OSTEOBLASTIC TRANSITION IN CHRONIC KIDNEY DISEASE VIA WNT/Β-CATENIN PATHWAY. Shock 2025; 63:312-319. [PMID: 39450908 DOI: 10.1097/shk.0000000000002488] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2024]
Abstract
ABSTRACT Chronic kidney disease (CKD)-related vascular calcification (VC) is a common degenerative phenomenon of the vessel wall and its pathological basis is the phenotypic transformation of vascular smooth muscle cells (VSMCs). Zinc finger and BR-C (Broad-Complex), ttk (tramtrack), and bab (bric à brac) (BTB) domain containing 16 (ZBTB16) have been reported to be expressed in the aortic tissues in a rat model of VC. This work is conducted to reveal the functions of ZBTB16 on VC in CKD and to probe its involved reaction mechanisms. In vivo CKD rat models were established by adenine and VSMC calcification were stimulated with high phosphate (Pi) in vitro . Renal function indexes were estimated with relevant assay kits. Renal tissues were histologically examined with hematoxylin and eosin staining. Alizarin red and von kossa staining were used to measure arterial calcification. Reverse transcription-quantitative PCR and western blot were used to detect ZBTB16 expression. Western blot, immunohistochemistry, and immunofluorescence staining were used to detect osteogenic markers and smooth muscle cell markers. Western blot was used to measure the expressions of proteins implicated in Wnt/β-catenin pathway. In the blood samples of CKD patients with VC, aortic tissues of CKD rats, and Pi-treated VSMCs, ZBTB16 expression was significantly increased. ZBTB16 knockdown reduced renal dysfunction, calcium deposition and inhibited VSMCs osteoblast differentiation both in vitro and in vivo . Moreover, silencing with ZBTB16 inactivated Wingless-related integration site (Wnt)/β-catenin pathway. LiCl (Wnt/β-catenin agonist) reversed the protective effects of ZBTB16 knockdown on the calcification and osteoblastic transformation in vitro . Together, ZBTB16 silencing may downregulate Wnt/β-catenin pathway to protect against CKD-associated VC via repressing the osteoblastic transformation of VSMCs.
Collapse
Affiliation(s)
| | - Huaxing Huang
- Department of Nephrology, The Second Affiliated Hospital of Nantong University, Nantong, China
| | - Lianglan Shen
- Department of Nephrology, The Second Affiliated Hospital of Nantong University, Nantong, China
| | - Wubin Yao
- Department of Nephrology, The Second Affiliated Hospital of Nantong University, Nantong, China
| | - Rong Wang
- Department of Nephrology, The Second Affiliated Hospital of Nantong University, Nantong, China
| | - Meizi Kang
- Department of Nephrology, The Second Affiliated Hospital of Nantong University, Nantong, China
| | - Jiashan Huang
- Department of Radiology, The Second Affiliated Hospital of Nantong University, Nantong, China
| | - Yan Xie
- Department of Geriatrics, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Hongli Yang
- Department of Nephrology, The Second Affiliated Hospital of Nantong University, Nantong, China
| |
Collapse
|
6
|
Wu M, Li H, Sun X, Zhong R, Cai L, Chen R, Madeniyet M, Ren K, Peng Z, Yang Y, Chen W, Tu Y, Lai M, Deng J, Wu Y, Zhao S, Ruan Q, Rao M, Xie S, Ye Y, Wan J. Aerobic exercise prevents renal osteodystrophy via irisin-activated osteoblasts. JCI Insight 2025; 10:e184468. [PMID: 39883525 PMCID: PMC11949034 DOI: 10.1172/jci.insight.184468] [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: 07/03/2024] [Accepted: 01/16/2025] [Indexed: 01/31/2025] Open
Abstract
Renal osteodystrophy is commonly seen in patients with chronic kidney disease (CKD) due to disrupted mineral homeostasis. Given the impaired renal function in these patients, common antiresorptive agents, including bisphosphonates, must be used with caution or even contraindicated. Therefore, an alternative therapy without renal burden to combat renal osteodystrophy is urgently needed. Here, we report that clinically relevant aerobic exercise significantly prevents high-turnover renal osteodystrophy in CKD mice and patients with CKD without compromising renal function. Mechanistically, 4-week aerobic exercise in CKD mice increased expression of skeletal muscle PPARγ coactivator-1α (PGC-1α) and circulating irisin. Both exercise and irisin administration significantly activated osteoblasts, but not osteoclasts, via integrin αvβ5, thereby conferring bone quality benefits. Removal of irisin-influenced thermogenic adipose tissues or genetic ablation of uncoupling protein 1 did not alter the irisin-conferred antiosteodystrophy effect. Importantly, in a pilot clinical study, 12-week aerobic exercise in patients with high-grade CKD significantly increased circulating irisin and prevented osteodystrophy progression, without detectable renal burden. The combination of irisin and current antiresorptive agents effectively rescued renal osteodystrophy in mice. Our work provides mechanistic insights into the role of exercise and irisin in renal osteodystrophy, and it highlights a clinically relevant, low-cost, kidney-friendly therapy for patients with this devastating disease.
Collapse
Affiliation(s)
- Meng Wu
- Department of Nephrology, Blood Purification Research Center, the First Affiliated Hospital, Fujian Medical University, Fuzhou, China
- Department of Nephrology, Longyan First Affiliated Hospital of Fujian Medical University, Longyan, China
| | - Huilan Li
- Department of Nephrology, Longyan First Affiliated Hospital of Fujian Medical University, Longyan, China
- Department of Nephrology, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Xiaoting Sun
- School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, China
| | - Rongrong Zhong
- Department of Cardiology, Basic Scientific Research Center, Longyan First Affiliated Hospital of Fujian Medical University, Longyan, China
| | - Linli Cai
- Department of Cardiology, Basic Scientific Research Center, Longyan First Affiliated Hospital of Fujian Medical University, Longyan, China
| | - Ruibo Chen
- Department of Cellular and Genetic Medicine, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Madiya Madeniyet
- Department of Cellular and Genetic Medicine, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Kana Ren
- Department of Cellular and Genetic Medicine, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Zhen Peng
- Department of Cellular and Genetic Medicine, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Yujie Yang
- Fundamental Research Center, Shanghai Yangzhi Rehabilitation Hospital (Shanghai Sunshine Rehabilitation Center), School of Medicine, Tongji University, Shanghai, China
| | - Weiqin Chen
- Department of Nephrology, Longyan First Affiliated Hospital of Fujian Medical University, Longyan, China
| | - Yanling Tu
- Department of Nephrology, Longyan First Affiliated Hospital of Fujian Medical University, Longyan, China
| | - Miaoxin Lai
- Department of Nephrology, Longyan First Affiliated Hospital of Fujian Medical University, Longyan, China
| | - Jinxiu Deng
- Department of Nephrology, Longyan First Affiliated Hospital of Fujian Medical University, Longyan, China
| | - Yuting Wu
- Department of Cardiology, Basic Scientific Research Center, Longyan First Affiliated Hospital of Fujian Medical University, Longyan, China
| | - Shumin Zhao
- Department of Cardiology, Basic Scientific Research Center, Longyan First Affiliated Hospital of Fujian Medical University, Longyan, China
| | - Qingyan Ruan
- Department of Cardiology, Basic Scientific Research Center, Longyan First Affiliated Hospital of Fujian Medical University, Longyan, China
| | - Mei Rao
- Department of Cardiology, Basic Scientific Research Center, Longyan First Affiliated Hospital of Fujian Medical University, Longyan, China
| | - Sisi Xie
- Department of Cardiology, Basic Scientific Research Center, Longyan First Affiliated Hospital of Fujian Medical University, Longyan, China
| | - Ying Ye
- Department of Oral Implantology, Stomatological Hospital and Dental School of Tongji University, Shanghai Engineering Research Center of Tooth Restoration and Regeneration, Shanghai, China
| | - Jianxin Wan
- Department of Nephrology, Blood Purification Research Center, the First Affiliated Hospital, Fujian Medical University, Fuzhou, China
- Fujian Clinical Research Center for Metabolic Chronic Kidney Disease, the First Affiliated Hospital, and
- Department of Nephrology, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, China
| |
Collapse
|
7
|
He S, Li X, He Y, Guo L, Dong Y, Wang L, Yang L, Li L, Huang S, Fu J, Lin Q, Zhang Z, Zhang L. High-density lipoprotein nanoparticles spontaneously target to damaged renal tubules and alleviate renal fibrosis by remodeling the fibrotic niches. Nat Commun 2025; 16:1061. [PMID: 39870661 PMCID: PMC11772610 DOI: 10.1038/s41467-025-56223-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Accepted: 01/10/2025] [Indexed: 01/30/2025] Open
Abstract
Chronic kidney disease (CKD) ultimately causes renal fibrosis and end-stage renal disease, thus seriously threatens human health. However, current medications for CKD and fibrosis are inefficient, which is often due to poor targeting capability to renal tubule. In this study, we discover that biomimetic high-density lipoprotein (bHDL) lipid nanoparticles possess excellent targeting ability to injured tubular epithelial cells by kidney injury molecule-1(KIM-1) mediated internalization. Thus, we co-load anti-inflammatory drug triptolide (TP) and anti-fibrotic drug nintedanib (BIBF) on bHDL nanoparticles to treat CKD. Based on the targeted delivery and mutual enhancement of the efficacy of co-delivered drugs, the bHDL-based system effectively reduces kidney injury and alleviates renal fibrosis in different CKD mouse models. The mechanistic study shows that BIBF and TP synergistically remodel the fibrotic niches by decreasing inflammatory cytokines, limiting immune cell infiltration and inhibiting the activation of myofibroblasts. The bHDL vehicle also possesses high manufacturability, good safety and adequately reduces the toxicity of TP. Thus, this system is promising for the treatment of CKD and bHDL has good potential for delivering agents to damaged renal tubular epithelial cells.
Collapse
Affiliation(s)
- Shanshan He
- Key Laboratory of Drug Targeting and Drug Delivery Systems, Ministry of Education, West China School of Pharmacy, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Xiaoyang Li
- Key Laboratory of Drug Targeting and Drug Delivery Systems, Ministry of Education, West China School of Pharmacy, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Yuanyuan He
- College of Polymer Science and Engineering, West China School of Public Health, Med-X center of materials, Sichuan University, Chengdu, Sichuan, 610065, China
| | - Ling Guo
- National Engineering Technology Research Center for Miao Medicine, Guizhou Engineering Technology Research Center for Processing and Preparation of Traditional Chinese Medicine and Ethnic Medicine, College of Pharmaceutical Sciences, Guizhou University of Traditional Chinese Medicine, Guiyang, 550025, P. R. China
| | - Yunzhou Dong
- Key Laboratory of Drug Targeting and Drug Delivery Systems, Ministry of Education, West China School of Pharmacy, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Leilei Wang
- Key Laboratory of Drug Targeting and Drug Delivery Systems, Ministry of Education, West China School of Pharmacy, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Lan Yang
- College of Polymer Science and Engineering, West China School of Public Health, Med-X center of materials, Sichuan University, Chengdu, Sichuan, 610065, China
| | - Lin Li
- College of Polymer Science and Engineering, West China School of Public Health, Med-X center of materials, Sichuan University, Chengdu, Sichuan, 610065, China
| | - Shiyun Huang
- Key Laboratory of Drug Targeting and Drug Delivery Systems, Ministry of Education, West China School of Pharmacy, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Jiali Fu
- Key Laboratory of Drug Targeting and Drug Delivery Systems, Ministry of Education, West China School of Pharmacy, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Qing Lin
- Key Laboratory of Drug Targeting and Drug Delivery Systems, Ministry of Education, West China School of Pharmacy, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Zhirong Zhang
- Key Laboratory of Drug Targeting and Drug Delivery Systems, Ministry of Education, West China School of Pharmacy, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Ling Zhang
- College of Polymer Science and Engineering, West China School of Public Health, Med-X center of materials, Sichuan University, Chengdu, Sichuan, 610065, China.
| |
Collapse
|
8
|
Hamidizad Z, Kadkhodaee M, Kianian F, Ranjbaran M, Heidari F, Seifi B. Neuroprotective Effects of Sodium Nitroprusside on CKD-Induced Cognitive Dysfunction in Rats: Role of CBS and Nrf2/HO-1 Pathway. Neuromolecular Med 2025; 27:8. [PMID: 39775152 DOI: 10.1007/s12017-024-08828-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2024] [Accepted: 12/26/2024] [Indexed: 01/11/2025]
Abstract
Chronic kidney disease (CKD) is a conceivable new risk factor for cognitive disorder and dementia. Uremic toxicity, oxidative stress, and peripheral-central inflammation have been considered important mediators of CKD-induced nervous disorders. Nitric oxide (NO) is a retrograde neurotransmitter in synapses, and has vital roles in intracellular signaling in neurons. This research aims to determine the effectiveness of NO in CKD-induced cognitive deficits by considering the nuclear factor-erythroid factor 2-related factor 2 (Nrf2)/ heme oxygenase-1 (HO-1) signaling pathway and the important roles of cystathionine beta-synthase (CBS, H2S producing enzyme). Forty rats were divided into four experimental groups: sham, five-sixth (5/6) nephrectomy (5/6Nx, CKD), CKD + NO donor (Sodium nitroprusside, SNP), CKD + SNP and a CBS inhibitor (amino-oxy acetic acid, AOAA). To assess the neurocognitive abilities, eleven weeks after 5/6Nx, behavioral tests (Novel object recognition test, Passive avoidance test, and Barnes maze test) were done. Twelfth week after 5/6Nx, blood urea nitrogen (BUN) and serum creatinine (sCr) levels, as well as the nuclear factor-erythroid factor 2-related factor 2 (Nrf2), heme oxygenase-1 (HO-1) expression levels and neuronal injury in the hippocampus and prefrontal cortex were assessed. As predicted, the levels of BUN and sCr (both P < 0.001) and neuronal injury in the hippocampus (P < 0.001 for CA1; CA3; DG) and prefrontal cortex (P < 0.001) increased in CKD rats as well as 5/6Nx induced reduction of Nrf2 (both P < 0.001) /HO-1(P < 0.001; P < 0.01 respectively) pathway activity in the hippocampus and prefrontal cortex in CKD rats. Moreover, CKD leads to cognitive disorder and memory loss (Novel object recognition test (NOR) (P < 0.001), Passive avoidance test (PA) (P < 0.001) and Barnes maze (BA) (Escape latency (P < 0.001); Error (P < 0.001)). SNP treatment significantly improved Nrf2 (both P < 0.001) /HO-1 (P < 0.001; P < 0.05 respectively) pathways and neuronal injury (P < 0.001 for CA1; CA3; DG) in the hippocampus and prefrontal cortex in CKD rats as well as enhanced learning and memory ability in CKD rats. However, ameliorating effects of SNP on cognitive disorder (NOR (P < 0.05), PA (P < 0.001) and BA (Escape latency (P < 0.05); Error (P < 0.001)) and Nrf2 (P < 0.01; P < 0.001 in the hippocampus and prefrontal cortex respectively) /HO-1 (P < 0.05 in both) signaling pathway activity were nullified by CBS inhibitor and H2S reduction. In conclusion, this study demonstrated that NO improved CKD-induced cognitive impairment and neuronal death which is may be depended to CBS activity and endogenous H2S levels.
Collapse
Affiliation(s)
- Zeinab Hamidizad
- Electrophysiology Research Center, Neuroscience Institute, Tehran University of Medical Sciences, Tehran, Iran
- Department of Physiology, Faculty of Medicine, Qom University of Medical Sciences, Qom, Iran
| | - Mehri Kadkhodaee
- Department of Physiology, Faculty of Medicine, Tehran University of Medical Sciences, Poorsina Ave, Tehran, Iran
| | - Farzaneh Kianian
- Department of Physiology, Faculty of Medicine, Tehran University of Medical Sciences, Poorsina Ave, Tehran, Iran
| | - Mina Ranjbaran
- Department of Physiology, Faculty of Medicine, Tehran University of Medical Sciences, Poorsina Ave, Tehran, Iran
| | - Fatemeh Heidari
- Department of Anatomy, School of Medicine, Qom University of Medical Sciences, Qom, Iran
| | - Behjat Seifi
- Electrophysiology Research Center, Neuroscience Institute, Tehran University of Medical Sciences, Tehran, Iran.
- Department of Physiology, Faculty of Medicine, Tehran University of Medical Sciences, Poorsina Ave, Tehran, Iran.
| |
Collapse
|
9
|
Liu Y, Li Z, Xu Y, Mao H, Huang N. Uric Acid and Atherosclerosis in Patients with Chronic Kidney Disease: Recent Progress, Mechanisms, and Prospect. KIDNEY DISEASES (BASEL, SWITZERLAND) 2025; 11:112-127. [PMID: 40124130 PMCID: PMC11928073 DOI: 10.1159/000543781] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/04/2024] [Accepted: 01/16/2025] [Indexed: 03/25/2025]
Abstract
Background Chronic kidney disease (CKD) is a prevalent global health concern, significantly linked to increased cardiovascular morbidity and mortality. Among various risk factors, uric acid (UA) has emerged as a potentially modifiable contributor to cardiovascular complications in CKD patients. Summary Elevated serum uric acid levels frequently occur in individuals with CKD and are associated with the development of atherosclerosis (AS). Uric acid has been demonstrated to exacerbate inflammatory processes, promote oxidative stress, and cause endothelial dysfunction, which are critical factors that drive the formation of atherosclerotic plaques. Furthermore, high uric acid levels can worsen renal function, establishing a detrimental cycle that amplifies cardiovascular risk. Key Messages This review investigates the complex interconnection between UA and AS in patients with CKD, highlighting the underlying mechanisms and therapeutic considerations. A more profound comprehension of this relationship is essential for enhancing cardiovascular health and outcomes in this vulnerable population.
Collapse
Affiliation(s)
- Yuchu Liu
- Department of Nephrology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
- Key Laboratory of Nephrology, Ministry of Health of China, Guangdong Provincial Key Laboratory of Nephrology, Guangzhou, China
| | - Zeyu Li
- Department of Nephrology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
- Key Laboratory of Nephrology, Ministry of Health of China, Guangdong Provincial Key Laboratory of Nephrology, Guangzhou, China
| | - Yuanwen Xu
- Department of Nephrology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
- Key Laboratory of Nephrology, Ministry of Health of China, Guangdong Provincial Key Laboratory of Nephrology, Guangzhou, China
| | - Haiping Mao
- Department of Nephrology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
- Key Laboratory of Nephrology, Ministry of Health of China, Guangdong Provincial Key Laboratory of Nephrology, Guangzhou, China
| | - Naya Huang
- Department of Nephrology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
- Key Laboratory of Nephrology, Ministry of Health of China, Guangdong Provincial Key Laboratory of Nephrology, Guangzhou, China
| |
Collapse
|
10
|
Sun CC, Xiao JL, Sun C, Tang CF. Ferroptosis and Its Potential Role in the Physiopathology of Skeletal Muscle Atrophy. Int J Mol Sci 2024; 25:12463. [PMID: 39596528 PMCID: PMC11595065 DOI: 10.3390/ijms252212463] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Revised: 11/13/2024] [Accepted: 11/18/2024] [Indexed: 11/28/2024] Open
Abstract
Skeletal muscle atrophy is a major health concern, severely affecting the patient's mobility and life quality. In the pathological process of skeletal muscle atrophy, with the progressive decline in muscle quality, strength, and function, the incidence of falling, fracture, and death is greatly increased. Unfortunately, there are no effective treatments for this devastating disease. Thus, it is imperative to investigate the exact pathological molecular mechanisms underlying the development of skeletal muscle atrophy and to identify new therapeutic targets. Decreased muscle mass, strength, and muscle fiber cross-sectional area are typical pathological features and manifestations of skeletal muscle atrophy. Ferroptosis, an emerging type of programmed cell death, is characterized by iron-dependent oxidative damage, lipid peroxidation, and reactive oxygen species accumulation. Notably, the understanding of its role in skeletal muscle atrophy is emerging. Ferroptosis has been found to play an important role in the intricate interplay between the pathological mechanisms of skeletal muscle atrophy and its progression caused by multiple factors. This provides new opportunities and challenges in the treatment of skeletal muscle atrophy. Therefore, we systematically elucidated the ferroptosis mechanism and its progress in skeletal muscle atrophy, aiming to provide a comprehensive insight into the intricate relationship between ferroptosis and skeletal muscle atrophy from the perspectives of iron metabolism and lipid peroxidation and to provide new insights for targeting the pathways related to ferroptosis and the treatment of skeletal muscle atrophy.
Collapse
Affiliation(s)
- Chen-Chen Sun
- School of Physical Education, Hunan First Normal University, Changsha 410205, China;
| | - Jiang-Ling Xiao
- College of Physical Education, Hunan Normal University, Changsha, 410012, China; (J.-L.X.); (C.S.)
| | - Chen Sun
- College of Physical Education, Hunan Normal University, Changsha, 410012, China; (J.-L.X.); (C.S.)
| | - Chang-Fa Tang
- College of Physical Education, Hunan Normal University, Changsha, 410012, China; (J.-L.X.); (C.S.)
| |
Collapse
|
11
|
Shimamoto S, Nakahara T, Yamada S, Nagasu H, Kishi S, Nakashima N, Tsuruya K, Okada H, Tamura K, Narita I, Maruyama S, Yano Y, Yokoo T, Wada T, Wada J, Kanda E, Kataoka H, Nangaku M, Kashihara N, Nakano T. Association between proteinuria and mineral metabolism disorders in chronic kidney disease: the Japan chronic kidney disease database extension (J-CKD-DB-Ex). Sci Rep 2024; 14:27481. [PMID: 39523380 PMCID: PMC11551138 DOI: 10.1038/s41598-024-79291-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2024] [Accepted: 11/07/2024] [Indexed: 11/16/2024] Open
Abstract
Chronic kidney disease-mineral and bone disorder (CKD-MBD) are recognized as a systemic disease affecting the prognosis of patients with CKD. Proper management of CKD-MBD is important to improve the prognosis of patients with CKD. Although proteinuria is recognized as a poor prognostic factor in these patients, few reports have examined its association with CKD-MBD. We examined the association between proteinuria and CKD-MBD using data from the Japan Chronic Kidney Disease Database Extension (J-CKD-DB-Ex). Among the patients registered in the J-CKD-DB-Ex, 30,977 with CKD stages G2-G5 who had serum creatinine, albumin, calcium, and phosphate concentrations measured at least once and urinalysis performed were included. The patients were divided into four groups (negative, 1+, 2+, and 3+) according to the degree of proteinuria. The association between proteinuria and CKD-MBD was examined by a logistic regression analysis. In a model adjusted for age, sex, diabetes, and the estimated glomerular filtration rate (eGFR), the odds ratio of the 3 + group compared with the negative group significantly increased to 2.67 (95% confidence interval, 2.29-3.13) for hyperphosphatemia, 2.68 (1.94-3.71) for hypocalcemia, and 1.56 (1.24-1.98) for hypomagnesemia. Proteinuria is associated with hyperphosphatemia, hypocalcemia, and hypomagnesemia in patients with CKD independently of eGFR.
Collapse
Affiliation(s)
- Sho Shimamoto
- Department of Medicine and Clinical Science, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 8128582, Japan
| | - Takako Nakahara
- Department of Medical Technology, Faculty of Health Science and Technology, Kawasaki University of Medical Welfare, Okayama, Japan
| | - Shunsuke Yamada
- Department of Medicine and Clinical Science, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 8128582, Japan
| | - Hajime Nagasu
- Department of Nephrology and Hypertension, Kawasaki Medical School, Okayama, Japan
| | - Seiji Kishi
- Department of Nephrology and Hypertension, Kawasaki Medical School, Okayama, Japan
| | - Naoki Nakashima
- Department of Medical Informatics, Graduate School of Medical Science, Kyushu University, Fukuoka, Japan
| | | | - Hirokazu Okada
- Department of Nephrology, Faculty of Medicine, Saitama Medical University, Saitama, Japan
| | - Kouichi Tamura
- Department of Medical Science and Cardiorenal Medicine, Graduate School of Medicine, Yokohama City University, Kanagawa, Japan
| | - Ichiei Narita
- Division of Clinical Nephrology and Rheumatology, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| | - Shoichi Maruyama
- Department of Nephrology, Nagoya University Graduate School of Medicine, Aichi, Japan
| | - Yuichiro Yano
- Department of General Medicine, Juntendo University Faculty of Medicine, Tokyo, Japan
| | - Takashi Yokoo
- Division of Kidney and Hypertension, Department of Internal Medicine, Jikei University School of Medicine, Tokyo, Japan
| | - Takashi Wada
- Department of Nephrology and Rheumatology, Kanazawa University, Kanazawa, Japan
| | - Jun Wada
- Department of Nephrology, Rheumatology, Endocrinology and Metabolism, Dentistry and Pharmaceutical Sciences, Okayama University Graduate School of Medicine, Okayama, Japan
| | - Eiichiro Kanda
- Department of Health Data Science, Kawasaki Medical School, Okayama, Japan
| | - Hiromi Kataoka
- Department of Medical Technology, Faculty of Health Science and Technology, Kawasaki University of Medical Welfare, Okayama, Japan
| | - Masaomi Nangaku
- Division of Nephrology and Endocrinology, University of Tokyo Graduate School of Medicine, Tokyo, Japan
| | - Naoki Kashihara
- Department of Nephrology and Hypertension, Kawasaki Medical School, Okayama, Japan
| | - Toshiaki Nakano
- Department of Medicine and Clinical Science, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 8128582, Japan.
| |
Collapse
|
12
|
Liu SF, Kucherenko MM, Sang P, Li Q, Yao J, Nambiar Veetil N, Gransar T, Alesutan I, Voelkl J, Salinas G, Grune J, Simmons S, Knosalla C, Kuebler WM. RUNX2 is stabilised by TAZ and drives pulmonary artery calcification and lung vascular remodelling in pulmonary hypertension due to left heart disease. Eur Respir J 2024; 64:2300844. [PMID: 39542509 DOI: 10.1183/13993003.00844-2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2023] [Accepted: 07/13/2024] [Indexed: 11/17/2024]
Abstract
BACKGROUND Calcification is common in chronic vascular disease, yet its role in pulmonary hypertension due to left heart disease is unknown. Here, we probed for the role of runt-related transcription factor-2 (RUNX2), a master transcription factor in osteogenesis, and its regulation by the HIPPO pathway transcriptional coactivator with PDZ-binding motif (TAZ) in the osteogenic reprogramming of pulmonary artery smooth muscle cells and vascular calcification in patients with pulmonary hypertension due to left heart disease. We similarly examined its role using an established rat model of pulmonary hypertension due to left heart disease induced by supracoronary aortic banding. METHODS Pulmonary artery samples were collected from patients and rats with pulmonary hypertension due to left heart disease. Genome-wide RNA sequencing was performed, and pulmonary artery calcification assessed. Osteogenic signalling via TAZ and RUNX2 was delineated by protein biochemistry. In vivo, the therapeutic potential of RUNX2 or TAZ inhibition by CADD522 or verteporfin was tested in the rat model. RESULTS Gene ontology term analysis identified significant enrichment in ossification and osteoblast differentiation genes, including RUNX2, in pulmonary arteries of patients and lungs of rats with pulmonary hypertension due to left heart disease. Pulmonary artery calcification was evident in both patients and rats. Both TAZ and RUNX2 were upregulated and activated in pulmonary artery smooth muscle cells of patients and rats. Co-immunoprecipitation revealed a direct interaction of RUNX2 with TAZ in pulmonary artery smooth muscle cells. TAZ inhibition or knockdown decreased RUNX2 abundance due to accelerated RUNX2 protein degradation rather than reduced de novo synthesis. Inhibition of either TAZ or RUNX2 attenuated pulmonary artery calcification, distal lung vascular remodelling and pulmonary hypertension development in the rat model. CONCLUSION Pulmonary hypertension due to left heart disease is associated with pulmonary artery calcification that is driven by TAZ-dependent stabilisation of RUNX2, causing osteogenic reprogramming of pulmonary artery smooth muscle cells. The TAZ-RUNX2 axis may present a therapeutic target in pulmonary hypertension due to left heart disease.
Collapse
Affiliation(s)
- Shao-Fei Liu
- Institute for Physiology, Charité - Universitätsmedizin Berlin, corporate member of the Free University Berlin and the Humboldt University Berlin, Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), partner site Berlin, Berlin, Germany
- These authors contributed equally to the study
| | - Mariya M Kucherenko
- Institute for Physiology, Charité - Universitätsmedizin Berlin, corporate member of the Free University Berlin and the Humboldt University Berlin, Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), partner site Berlin, Berlin, Germany
- Department of Cardiothoracic and Vascular Surgery, Deutsches Herzzentrum der Charité (DHZC), Berlin, Germany
- Charité - Universitätsmedizin Berlin, corporate member of the Free University Berlin and the Humboldt University Berlin, Berlin, Germany
- These authors contributed equally to the study
| | - Pengchao Sang
- Institute for Physiology, Charité - Universitätsmedizin Berlin, corporate member of the Free University Berlin and the Humboldt University Berlin, Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), partner site Berlin, Berlin, Germany
- Department of Cardiothoracic and Vascular Surgery, Deutsches Herzzentrum der Charité (DHZC), Berlin, Germany
| | - Qiuhua Li
- Institute for Physiology, Charité - Universitätsmedizin Berlin, corporate member of the Free University Berlin and the Humboldt University Berlin, Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), partner site Berlin, Berlin, Germany
| | - Juquan Yao
- Institute for Physiology, Charité - Universitätsmedizin Berlin, corporate member of the Free University Berlin and the Humboldt University Berlin, Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), partner site Berlin, Berlin, Germany
| | - Netra Nambiar Veetil
- Institute for Physiology, Charité - Universitätsmedizin Berlin, corporate member of the Free University Berlin and the Humboldt University Berlin, Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), partner site Berlin, Berlin, Germany
- Department of Cardiothoracic and Vascular Surgery, Deutsches Herzzentrum der Charité (DHZC), Berlin, Germany
- Charité - Universitätsmedizin Berlin, corporate member of the Free University Berlin and the Humboldt University Berlin, Berlin, Germany
| | - Tara Gransar
- Institute for Physiology, Charité - Universitätsmedizin Berlin, corporate member of the Free University Berlin and the Humboldt University Berlin, Berlin, Germany
- Department of Cardiothoracic and Vascular Surgery, Deutsches Herzzentrum der Charité (DHZC), Berlin, Germany
| | - Ioana Alesutan
- Institute of Physiology and Pathophysiology, Johannes Kepler University Linz, Linz, Austria
| | - Jakob Voelkl
- DZHK (German Centre for Cardiovascular Research), partner site Berlin, Berlin, Germany
- Institute of Physiology and Pathophysiology, Johannes Kepler University Linz, Linz, Austria
- Department of Nephrology and Medical Intensive Care, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Gabriela Salinas
- NGS - Integrative Genomics Core Unit (NIG), Institute of Pathology, University Medical Center Göttingen (UMG), Göttingen, Germany
| | - Jana Grune
- Institute for Physiology, Charité - Universitätsmedizin Berlin, corporate member of the Free University Berlin and the Humboldt University Berlin, Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), partner site Berlin, Berlin, Germany
| | - Szandor Simmons
- Institute for Physiology, Charité - Universitätsmedizin Berlin, corporate member of the Free University Berlin and the Humboldt University Berlin, Berlin, Germany
| | - Christoph Knosalla
- DZHK (German Centre for Cardiovascular Research), partner site Berlin, Berlin, Germany
- Department of Cardiothoracic and Vascular Surgery, Deutsches Herzzentrum der Charité (DHZC), Berlin, Germany
- Charité - Universitätsmedizin Berlin, corporate member of the Free University Berlin and the Humboldt University Berlin, Berlin, Germany
- These authors share the last authorship
| | - Wolfgang M Kuebler
- Institute for Physiology, Charité - Universitätsmedizin Berlin, corporate member of the Free University Berlin and the Humboldt University Berlin, Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), partner site Berlin, Berlin, Germany
- DZL (German Centre for Lung Research), partner site Berlin, Berlin, Germany
- The Keenan Research Centre for Biomedical Science at St. Michael's, Toronto, ON, Canada
- Departements of Surgery and Physiology, University of Toronto, Toronto, ON, Canada
- These authors share the last authorship
| |
Collapse
|
13
|
Zaimi M, Grapsa E. Current therapeutic approach of chronic kidney disease-mineral and bone disorder. Ther Apher Dial 2024; 28:671-689. [PMID: 38898685 DOI: 10.1111/1744-9987.14177] [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: 02/29/2024] [Revised: 05/14/2024] [Accepted: 05/31/2024] [Indexed: 06/21/2024]
Abstract
Chronic kidney disease (CKD) has emerged as one of the leading noncommunicable diseases affecting >10% of the population worldwide. Bone and mineral disorders are a common complication among patients with CKD resulting in a poor life quality, high fracture risk, increased morbidity and cardiovascular mortality. According to Kidney Disease: Improving Global Outcomes, renal osteodystrophy refers to changes in bone morphology found in bone biopsy, whereas CKD-mineral and bone disorder (CKD-MBD) defines a complex of disturbances including biochemical and hormonal alterations, disorders of bone and mineral metabolism and extraskeletal calcification. As a result, the management of CKD-MBD should focus on the aforementioned parameters, including the treatment of hyperphosphatemia, hypocalcemia, abnormal PTH and vitamin D levels. Regarding the bone fragility fractures, osteoporosis and renal osteodystrophy, which constitute the bone component of CKD-MBD, anti-osteoporotic agents constitute the mainstay of treatment. However, a thorough elucidation of the CKD-MBD pathogenesis is crucial for the ideal personalized treatment approach. In this paper, we review the pathology and management of CKD-MBD based on the current literature with special attention to recent advances.
Collapse
Affiliation(s)
- Maria Zaimi
- National and Kapodistrian University of Athens, Aretaieio Hospital, Athens, Greece
| | - Eirini Grapsa
- National and Kapodistrian University of Athens, Aretaieio Hospital, Athens, Greece
| |
Collapse
|
14
|
Yang S, Chen Q, Fan Y, Zhang C, Cao M. The essential role of dual-energy x-ray absorptiometry in the prediction of subclinical cardiovascular disease. Front Cardiovasc Med 2024; 11:1377299. [PMID: 39280034 PMCID: PMC11393745 DOI: 10.3389/fcvm.2024.1377299] [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: 01/27/2024] [Accepted: 08/15/2024] [Indexed: 09/18/2024] Open
Abstract
Subclinical cardiovascular disease (Sub-CVD) is an early stage of cardiovascular disease and is often asymptomatic. Risk factors, including hypertension, diabetes, obesity, and lifestyle, significantly affect Sub-CVD. Progress in imaging technology has facilitated the timely identification of disease phenotypes and risk categorization. The critical function of dual-energy x-ray absorptiometry (DXA) in predicting Sub-CVD was the subject of this research. Initially used to evaluate bone mineral density, DXA has now evolved into an indispensable tool for assessing body composition, which is a pivotal determinant in estimating cardiovascular risk. DXA offers precise measurements of body fat, lean muscle mass, bone density, and abdominal aortic calcification, rendering it an essential tool for Sub-CVD evaluation. This study examined the efficacy of DXA in integrating various risk factors into a comprehensive assessment and how the application of machine learning could enhance the early discovery and control of cardiovascular risks. DXA exhibits distinct advantages and constraints compared to alternative imaging modalities such as ultrasound, computed tomography, magnetic resonance imaging, and positron emission tomography. This review advocates DXA incorporation into cardiovascular health assessments, emphasizing its crucial role in the early identification and management of Sub-CVD.
Collapse
Affiliation(s)
- Sisi Yang
- Department of Geriatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Qin Chen
- Department of Geriatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yang Fan
- Department of Geriatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Cuntai Zhang
- Department of Geriatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ming Cao
- Department of Geriatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
15
|
Yoshiko Y, Vucenik I. Inositol Hexaphosphate in Bone Health and Disease. Biomolecules 2024; 14:1072. [PMID: 39334839 PMCID: PMC11430719 DOI: 10.3390/biom14091072] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2024] [Revised: 08/23/2024] [Accepted: 08/24/2024] [Indexed: 09/30/2024] Open
Abstract
Dietary phytic acid/phytate/myo-inositol hexaphosphate (IP6), a phosphate reservoir in plants, was viewed as antinutrient, caused by an influence on the bioavailability of minerals through its chelating activity. However, there is a growing body of evidence indicating that IP6 has beneficial (e.g., antiinflammatory, antibacterial, and anticancer) effects on multiple biological processes. Also, IP6 and its metabolites are known to exist in mammalian cells, including human cells, and the role of IP6 as a functional molecule is attracting attention. IP6 can bind to the growth sites of hydroxy-apatite (HA) and calcium oxalate crystals to prevent their growth and hence inhibit pathological calcification. SNF472, hexasodium IP6, is currently being evaluated in clinical studies as a treatment for vascular calcification and calciphylaxis. However, since HA crystal growth within bone matrix is an essential process in bone formation, it is possible that IP6 intake may inhibit physiological mineralization and bone formation, although currently more published studies suggest that IP6 may contribute to bone health rather than inhibit bone formation. Given that IP6 and its metabolites are thought to have diverse activities and many health benefits, it remains important to consider the range of effects of IP6 on bone.
Collapse
Affiliation(s)
- Yuji Yoshiko
- Pi Skovy, 1-15-31-9, Mukainadahonmachi, Minami-ku, Hiroshima 734-0062, Japan
| | - Ivana Vucenik
- Department of Medical and Research Technology, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| |
Collapse
|
16
|
Fu K, Cai Q, Jin X, Chen L, Oo WM, Duong V, Li G, Zhu Z, Ding C, Zhang C, Gao Y, Hunter DJ. Association of serum calcium, vitamin D, and C-reactive protein with all-cause and cause-specific mortality in an osteoarthritis population in the UK: a prospective cohort study. BMC Public Health 2024; 24:2286. [PMID: 39175018 PMCID: PMC11342510 DOI: 10.1186/s12889-024-19825-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Accepted: 08/19/2024] [Indexed: 08/24/2024] Open
Abstract
BACKGROUND Osteoarthritis is a prevalent musculoskeletal condition, but the role of specific serum biomarkers, such as calcium, vitamin D, and C-reactive protein (CRP), in predicting mortality among individuals with osteoarthritis remains unclear. METHODS This observational study analyzed longitudinal data from over 500,000 participants in the UK Biobank, identifying those with osteoarthritis using ICD-9/10 codes or self-reported history. We performed multivariable cox-regression and flexible parametric survival model (FPSM) for survival analysis, with adjustments made through the inverse probability of treatment weight (IPTW) for baseline covariates identified by directed acyclic graphs (DAGs). RESULTS Of the 49,082 osteoarthritis population, the average age was 60.69 years, with 58.7% being female. During the follow-up period exceeding 15 years, a total of 5,522 people with osteoarthritis died. High serum calcium levels, compared to normal serum calcium levels, were significantly associated with all-cause mortality (hazard ratio (HR) 1.33, 95% confidence interval (CI) 1.11, 1.59), cardiovascular diseases (CVD)-related deaths (HR 1.55, 95% CI 1.05, 2.29), and other deaths (HR 1.59, 95% CI 1.20, 2.11). Low serum calcium levels, compared to normal serum calcium levels, was linked with CVD-related deaths (HR 2.06, 95% CI 1.02, 4.14). Vitamin D insufficiency, compared to sufficient vitamin D levels, was correlated with all-cause mortality (HR 1.22, 95% CI 1.13, 1.33), CVD-related deaths (HR 1.43, 95% CI 1.20, 1.72), and other deaths (HR 1.26, 95% CI 1.09, 1.45) but not with cancer-related deaths. High serum CRP levels, compared to normal CRP levels, were associated with all outcomes (all-cause mortality: HR 1.22, 95% CI 1.12, 1.33; CVD-related death: HR 1.24, 95%CI 1.03, 1.49; cancer-related death: HR 1.23, 95% CI 1.09, 1.40; other deaths: HR 1.19, 95%CI 1.03, 1.38). CONCLUSIONS Both high and low serum calcium levels, elevated CRP, and vitamin D insufficiency are potential predictors of increased mortality risk in the osteoarthritis population. These findings emphasize the importance of monitoring and possibly addressing these serum biomarkers in osteoarthritis populations to improve long-term outcomes. Further studies are needed to understand the underlying mechanisms and to propose therapeutic interventions.
Collapse
Affiliation(s)
- Kai Fu
- Department of Orthopedic Surgery, Shanghai Sixth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
- Kolling Institute, Sydney Musculoskeletal Health, The University of Sydney, Sydney, Australia.
| | - Qianying Cai
- Department of Orthopedic Surgery, Shanghai Sixth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Institute of Microsurgery on Extremities, Shanghai Sixth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xinzhong Jin
- Centre for Big Data Research in Health, University of New South Wales, Sydney, Australia
| | - Lingxiao Chen
- Department of Orthopaedics, Centre for Orthopaedics, Advanced Medical Research Institute, Cheeloo College of Medicine, Qilu Hospital of Shandong University, Shandong University, Shandong University, Jinan, Shandong, China
- Department of Biostatistics, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Win Min Oo
- Kolling Institute, Sydney Musculoskeletal Health, The University of Sydney, Sydney, Australia
- Department of Physical Medicine and Rehabilitation, Mandalay General Hospital, University of Medicine Mandalay, Mandalay, Myanmar
| | - Vicky Duong
- Kolling Institute, Sydney Musculoskeletal Health, The University of Sydney, Sydney, Australia
| | - Guangyi Li
- Department of Orthopedic Surgery, Shanghai Sixth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zhaohua Zhu
- Clinical Research Centre, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China
- Clinical Research Centre, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, China
- Menzies Institute for Medical Research, University of Tasmania, Hobart, TAS, Australia
| | - Changhai Ding
- Clinical Research Centre, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China
- Clinical Research Centre, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, China
- Menzies Institute for Medical Research, University of Tasmania, Hobart, TAS, Australia
| | - Changqing Zhang
- Department of Orthopedic Surgery, Shanghai Sixth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Youshui Gao
- Department of Orthopedic Surgery, Shanghai Sixth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - David J Hunter
- Kolling Institute, Sydney Musculoskeletal Health, The University of Sydney, Sydney, Australia
| |
Collapse
|
17
|
Rizzo M, Pennisi M, Macrì F, Falcone A, Di Pietro S, Mhalhel K, Giudice E. Bilateral Global Nephrocalcinosis in a Uremic Puppy. Vet Sci 2024; 11:338. [PMID: 39195792 PMCID: PMC11359828 DOI: 10.3390/vetsci11080338] [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: 06/06/2024] [Revised: 07/24/2024] [Accepted: 07/24/2024] [Indexed: 08/29/2024] Open
Abstract
This study explores kidney disease in young dogs, focusing on early diagnosis, management, and the importance of staging for effective treatment. Highlighting mineral metabolism imbalances and complications such as nephrocalcinosis, the study presents a case of severe renal failure with uremic syndrome and bilateral nephrocalcinosis in a 50-day-old puppy. Despite intensive care, the puppy's condition deteriorated rapidly, leading to euthanasia. The study underscores the challenges in diagnosing and managing canine nephrocalcinosis in young animals. It emphasizes the need for further research to improve the understanding and treatment outcomes in such cases, ultimately enhancing the quality of life for animals suffering from this rare condition.
Collapse
Affiliation(s)
- Maria Rizzo
- Department of Veterinary Sciences, University of Messina, Polo Universitario Annunziata, 98168 Messina, Italy; (M.R.); (M.P.); (F.M.); (K.M.); (E.G.)
| | - Melissa Pennisi
- Department of Veterinary Sciences, University of Messina, Polo Universitario Annunziata, 98168 Messina, Italy; (M.R.); (M.P.); (F.M.); (K.M.); (E.G.)
| | - Francesco Macrì
- Department of Veterinary Sciences, University of Messina, Polo Universitario Annunziata, 98168 Messina, Italy; (M.R.); (M.P.); (F.M.); (K.M.); (E.G.)
| | - Annastella Falcone
- Veterinary Teaching Hospital, University of Messina, Polo Universitario Annunziata, 98168 Messina, Italy;
| | - Simona Di Pietro
- Department of Veterinary Sciences, University of Messina, Polo Universitario Annunziata, 98168 Messina, Italy; (M.R.); (M.P.); (F.M.); (K.M.); (E.G.)
| | - Kamel Mhalhel
- Department of Veterinary Sciences, University of Messina, Polo Universitario Annunziata, 98168 Messina, Italy; (M.R.); (M.P.); (F.M.); (K.M.); (E.G.)
| | - Elisabetta Giudice
- Department of Veterinary Sciences, University of Messina, Polo Universitario Annunziata, 98168 Messina, Italy; (M.R.); (M.P.); (F.M.); (K.M.); (E.G.)
| |
Collapse
|
18
|
Fisher A, Fisher L, Srikusalanukul W. Prediction of Osteoporotic Hip Fracture Outcome: Comparative Accuracy of 27 Immune-Inflammatory-Metabolic Markers and Related Conceptual Issues. J Clin Med 2024; 13:3969. [PMID: 38999533 PMCID: PMC11242639 DOI: 10.3390/jcm13133969] [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: 06/11/2024] [Revised: 06/26/2024] [Accepted: 07/03/2024] [Indexed: 07/14/2024] Open
Abstract
Objectives: This study, based on the concept of immuno-inflammatory-metabolic (IIM) dysregulation, investigated and compared the prognostic impact of 27 indices at admission for prediction of postoperative myocardial injury (PMI) and/or hospital death in hip fracture (HF) patients. Methods: In consecutive HF patient (n = 1273, mean age 82.9 ± 8.7 years, 73.5% females) demographics, medical history, laboratory parameters, and outcomes were recorded prospectively. Multiple logistic regression and receiver-operating characteristic analyses (the area under the curve, AUC) were used to establish the predictive role for each biomarker. Results: Among 27 IIM biomarkers, 10 indices were significantly associated with development of PMI and 16 were indicative of a fatal outcome; in the subset of patients aged >80 years with ischaemic heart disease (IHD, the highest risk group: 90.2% of all deaths), the corresponding figures were 26 and 20. In the latter group, the five strongest preoperative predictors for PMI were anaemia (AUC 0.7879), monocyte/eosinophil ratio > 13.0 (AUC 0.7814), neutrophil/lymphocyte ratio > 7.5 (AUC 0.7784), eosinophil count < 1.1 × 109/L (AUC 0.7780), and neutrophil/albumin × 10 > 2.4 (AUC 0.7732); additionally, sensitivity was 83.1-75.4% and specificity was 82.1-75.0%. The highest predictors of in-hospital death were platelet/lymphocyte ratio > 280.0 (AUC 0.8390), lymphocyte/monocyte ratio < 1.1 (AUC 0.8375), albumin < 33 g/L (AUC 0.7889), red cell distribution width > 14.5% (AUC 0.7739), and anaemia (AUC 0.7604), sensitivity 88.2% and above, and specificity 85.1-79.3%. Internal validation confirmed the predictive value of the models. Conclusions: Comparison of 27 IIM indices in HF patients identified several simple, widely available, and inexpensive parameters highly predictive for PMI and/or in-hospital death. The applicability of IIM biomarkers to diagnose and predict risks for chronic diseases, including OP/OF, in the preclinical stages is discussed.
Collapse
Affiliation(s)
- Alexander Fisher
- Department of Geriatric Medicine, The Canberra Hospital, ACT Health, Canberra 2605, Australia
- Department of Orthopaedic Surgery, The Canberra Hospital, ACT Health, Canberra 2605, Australia
- Medical School, Australian National University, Canberra 2601, Australia
| | - Leon Fisher
- Frankston Hospital, Peninsula Health, Melbourne 3199, Australia
| | - Wichat Srikusalanukul
- Department of Geriatric Medicine, The Canberra Hospital, ACT Health, Canberra 2605, Australia
| |
Collapse
|
19
|
Zittermann A, Zelzer S, Herrmann M, Gummert JF, Kleber M, Trummer C, Theiler-Schwetz V, Keppel MH, Maerz W, Pilz S. Determinants of circulating calcitriol in cardiovascular disease. J Steroid Biochem Mol Biol 2024; 241:106528. [PMID: 38677380 DOI: 10.1016/j.jsbmb.2024.106528] [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: 01/05/2024] [Revised: 03/28/2024] [Accepted: 04/23/2024] [Indexed: 04/29/2024]
Abstract
Circulating calcitriol may contribute to the risk of cardiovascular disease (CVD), but its regulation in patients with CVD is poorly characterized. We therefore aimed to assess determinants of circulating calcitriol in these patients. We analyzed 2183 independent samples from a large cohort of patients scheduled for coronary angiography and 1727 independent samples from different other cohorts from patients with a wide range of CVDs, including heart transplant candidates, to quantify the association of different parameters with circulating calcitriol. We performed univariable and multivariable linear regression analyses using the mathematical function that fitted best with circulating calcitriol. In the multivariable analysis of the large single cohort, nine parameters remained significant, explaining 30.0 % (32.4 % after exclusion of 22 potential outliers) of the variation in circulating calcitriol (r=0.548). Log-transformed 25-hydroxyvitamin D [25(OH)D] and log-transformed glomerular filtration rate were the strongest predictors, explaining 17.6 % and 6.6 %, respectively, of the variation in calcitriol. In the analysis of the combined other cohorts, including heart transplant candidates, the multivariable model explained a total of 42.6 % (46.1 % after exclusion of 21 potential outliers) of the variation in calcitriol (r=0.653) with log-transformed fibroblast growth factor-23 and log-transformed 25(OH)D explaining 29.0 % and 6.2 %, respectively. Circulating 25(OH)D was positively and FGF-23 inversely associated with circulating calcitriol. Although significant, PTH was only a weak predictor of calcitriol in both analyses (<2.5 %). In patients with CVD, FGF-23 and 25(OH)D are important independent determinants of circulating calcitriol. The relative importance of these two parameters may vary according to CVD severity. Future studies should focus on the clinical importance of regulating circulating calcitriol by different parameters.
Collapse
Affiliation(s)
- A Zittermann
- Clinic for Thoracic and Cardiovascular Surgery, Herz, und Diabeteszentrum NRW, Ruhr-University Bochum, Bad Oeynhausen, 32545, Germany.
| | - S Zelzer
- Clinical Institute of Medical and Chemical Laboratory Diagnostics, Medical University of Graz, Graz 8036, Austria
| | - M Herrmann
- Clinical Institute of Medical and Chemical Laboratory Diagnostics, Medical University of Graz, Graz 8036, Austria
| | - J F Gummert
- Clinic for Thoracic and Cardiovascular Surgery, Herz, und Diabeteszentrum NRW, Ruhr-University Bochum, Bad Oeynhausen, 32545, Germany
| | - M Kleber
- Vth Department of Medicine (Nephrology, Hypertensiology, Rheumatology, Endocrinology, Diabetology, Lipidology), Medical Faculty Mannheim, University of Heidelberg, Mannheim 68167, Germany; SYNLAB MVZ Humangenetik Mannheim, Mannheim 68163, Germany
| | - C Trummer
- Division of Endocrinology and Diabetology, Department of Internal Medicine, Medical University of Graz, Graz 8036, Austria
| | - V Theiler-Schwetz
- Division of Endocrinology and Diabetology, Department of Internal Medicine, Medical University of Graz, Graz 8036, Austria
| | - M H Keppel
- Clinical Institute of Medical and Chemical Laboratory Diagnostics, Medical University of Graz, Graz 8036, Austria
| | - W Maerz
- Vth Department of Medicine (Nephrology, Hypertensiology, Rheumatology, Endocrinology, Diabetology, Lipidology), Medical Faculty Mannheim, University of Heidelberg, Mannheim 68167, Germany; SYNLAB MVZ Humangenetik Mannheim, Mannheim 68163, Germany; SYNLAB Holding, Deutschland GmbH, Mannheim, Augsburg 68159, Germany
| | - S Pilz
- Division of Endocrinology and Diabetology, Department of Internal Medicine, Medical University of Graz, Graz 8036, Austria
| |
Collapse
|
20
|
Morisi N, Montani M, Ehode EN, Virzì GM, Perrone S, Malaguti V, Ferrarini M, Donati G. Evaluating Short-Term Outcomes of Tunneled and Non-Tunneled Central Venous Catheters in Hemodialysis. J Clin Med 2024; 13:3664. [PMID: 38999230 PMCID: PMC11242506 DOI: 10.3390/jcm13133664] [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: 05/30/2024] [Revised: 06/14/2024] [Accepted: 06/19/2024] [Indexed: 07/14/2024] Open
Abstract
Background: The necessity of using central venous catheters (CVCs) in hemodialysis, coupled with their associated complications, remains a critical concern in nephrology. This study aims to compare the short-term prognosis of tunneled (T-CVC) and non-tunneled (NT-CVC) CVCs in acute hemodialysis patients, specifically focusing on infection rates, malpositioning, and lumen thrombosis within the first three weeks post-insertion. Methods: A retrospective analysis was conducted on 176 CVCs placed between January and December 2023 at the Policlinico di Modena and the Ospedale Civile di Baggiovara. Patient demographics, CHA2DS2-VASc scores, and comorbid conditions were recorded at the time of catheter placement. Outcomes assessed included catheter-related infections, malpositioning, and lumen thrombosis. Statistical analyses, including Chi-square tests, Fisher's exact tests, and Kaplan-Meier survival analysis, were performed to evaluate differences between T-CVCs and NT-CVCs. Results: The sample comprised 43% females with a mean age of 69.3 years (SD 13.9) and a mean CHADS-VASC score of 3.72 (SD 1.4). Hypertension (90%) was the most prevalent comorbidity. Of the 176 CVCs, 127 were T-CVCs and 49 were NT-CVCs. Infection rates were 3.15% for T-CVCs and 8.16% for NT-CVCs (p = 0.07). Malpositioning occurred in 0.79% of T-CVCs and 4.08% of NT-CVCs (p = 0.47). There was one case of lumen thrombosis in the NT-CVC group. Kaplan-Meier analysis indicated a significant divergence in infection-related catheter survival favoring T-CVCs after ten days (p = 0.034). Conclusions: While non-tunneled CVCs do not significantly alter short-term prognosis compared to tunneled CVCs, the latter show a better infection-related survival rate beyond ten days. Therefore, primary insertion of T-CVCs may be preferable when resources and clinical conditions permit, although NT-CVCs remain a viable option when immediate T-CVC insertion is challenging.
Collapse
Affiliation(s)
- Niccolò Morisi
- Surgical, Medical, Dental and Morphological Sciences Department (CHIMOMO), University of Modena and Reggio Emilia, 41126 Modena, Italy
- Nephrology, Dialysis and Kidney Transplant Unit, Azienda Ospedaliero-Universitaria di Modena, 41121 Modena, Italy
| | - Martina Montani
- Surgical, Medical, Dental and Morphological Sciences Department (CHIMOMO), University of Modena and Reggio Emilia, 41126 Modena, Italy
- Nephrology, Dialysis and Kidney Transplant Unit, Azienda Ospedaliero-Universitaria di Modena, 41121 Modena, Italy
| | - Edwidge Ntouba Ehode
- Surgical, Medical, Dental and Morphological Sciences Department (CHIMOMO), University of Modena and Reggio Emilia, 41126 Modena, Italy
- Nephrology, Dialysis and Kidney Transplant Unit, Azienda Ospedaliero-Universitaria di Modena, 41121 Modena, Italy
| | - Grazia Maria Virzì
- IRRIV-International Renal Research Institute Vicenza Foundation, 36100 Vicenza, Italy
- Department of Nephrology, Dialysis and Transplantation, San Bortolo Hospital, 36100 Vicenza, Italy
| | - Salvatore Perrone
- Nephrology, Dialysis and Kidney Transplant Unit, Azienda Ospedaliero-Universitaria di Modena, 41121 Modena, Italy
| | - Vittoria Malaguti
- Nephrology, Dialysis and Kidney Transplant Unit, Azienda Ospedaliero-Universitaria di Modena, 41121 Modena, Italy
| | - Marco Ferrarini
- Surgical, Medical, Dental and Morphological Sciences Department (CHIMOMO), University of Modena and Reggio Emilia, 41126 Modena, Italy
| | - Gabriele Donati
- Surgical, Medical, Dental and Morphological Sciences Department (CHIMOMO), University of Modena and Reggio Emilia, 41126 Modena, Italy
- Nephrology, Dialysis and Kidney Transplant Unit, Azienda Ospedaliero-Universitaria di Modena, 41121 Modena, Italy
| |
Collapse
|
21
|
Secondulfo C, Visco V, Virtuoso N, Fortunato M, Migliarino S, Rispoli A, La Mura L, Stellato A, Caliendo G, Settembre E, Galluccio F, Hamzeh S, Bilancio G. Vitamin D: A Bridge between Kidney and Heart. Life (Basel) 2024; 14:617. [PMID: 38792638 PMCID: PMC11123235 DOI: 10.3390/life14050617] [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: 04/02/2024] [Revised: 04/30/2024] [Accepted: 05/09/2024] [Indexed: 05/26/2024] Open
Abstract
Chronic kidney disease (CKD) and cardiovascular disease (CVD) are highly prevalent conditions, each significantly contributing to the global burden of morbidity and mortality. CVD and CKD share a great number of common risk factors, such as hypertension, diabetes, obesity, and smoking, among others. Their relationship extends beyond these factors, encompassing intricate interplay between the two systems. Within this complex network of pathophysiological processes, vitamin D has emerged as a potential linchpin, exerting influence over diverse physiological pathways implicated in both CKD and CVD. In recent years, scientific exploration has unveiled a close connection between these two prevalent conditions and vitamin D, a crucial hormone traditionally recognized for its role in bone health. This article aims to provide an extensive review of vitamin D's multifaceted and expanding actions concerning its involvement in CKD and CVD.
Collapse
Affiliation(s)
- Carmine Secondulfo
- Department “Scuola Medica Salernitana”, University of Salerno, 84081 Baronissi, Italy
| | - Valeria Visco
- Department “Scuola Medica Salernitana”, University of Salerno, 84081 Baronissi, Italy
| | - Nicola Virtuoso
- Cardiology Unit, Salerno University Hospital “San Giovanni di Dio e Ruggi d’Aragona”, 84131 Salerno, Italy
| | - Martino Fortunato
- Cardiology Unit, Salerno University Hospital “San Giovanni di Dio e Ruggi d’Aragona”, 84131 Salerno, Italy
| | - Serena Migliarino
- Cardiology Unit, Salerno University Hospital “San Giovanni di Dio e Ruggi d’Aragona”, 84131 Salerno, Italy
| | - Antonella Rispoli
- Cardiology Unit, Salerno University Hospital “San Giovanni di Dio e Ruggi d’Aragona”, 84131 Salerno, Italy
| | - Lucia La Mura
- Centro Medico Ascione Srl, 80059 Torre del Greco, Italy
| | - Adolfo Stellato
- Department “Scuola Medica Salernitana”, University of Salerno, 84081 Baronissi, Italy
| | - Giuseppe Caliendo
- Department “Scuola Medica Salernitana”, University of Salerno, 84081 Baronissi, Italy
| | - Emanuela Settembre
- Department “Scuola Medica Salernitana”, University of Salerno, 84081 Baronissi, Italy
| | - Fabiana Galluccio
- Department of Medicine and Surgery, University of Naples “Federico II”, 80138 Napoli, Italy
| | - Sarah Hamzeh
- Department of Medicine and Surgery, University of Naples “Federico II”, 80138 Napoli, Italy
| | - Giancarlo Bilancio
- Department “Scuola Medica Salernitana”, University of Salerno, 84081 Baronissi, Italy
- Nephrology Unit, Salerno University Hospital “San Giovanni di Dio e Ruggi d’Aragona”, 84131 Salerno, Italy
| |
Collapse
|
22
|
Mohamed ON, Mohamed MRM, Hassan IG, Alakkad AF, Othman A, Setouhi A, Issa AS. The Relationship of Fetuin-A with Coronary Calcification, Carotid Atherosclerosis, and Mortality Risk in Non-Dialysis Chronic Kidney Disease. J Lipid Atheroscler 2024; 13:194-211. [PMID: 38826181 PMCID: PMC11140250 DOI: 10.12997/jla.2024.13.2.194] [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: 11/23/2023] [Revised: 12/24/2023] [Accepted: 01/09/2024] [Indexed: 06/04/2024] Open
Abstract
Objective This study investigated the relationship of fetuin-A with coronary calcification, carotid atherosclerosis, and mortality risk in non-dialysis chronic kidney disease (CKD). Methods The study included 135 adult patients with CKD at stages 3-5, who were divided into coronary artery calcification (CAC) and non-CAC groups. We excluded current smokers and individuals with diabetes mellitus, inflammatory conditions, liver diseases, acute kidney failure, chronic hemodialysis, and cancer. We conducted kidney function tests, complete blood counts, and measured serum levels of fetuin-A, tumor necrosis factor-alpha (TNF-α), high-sensitivity C-reactive protein (hs-CRP), interleukin-6 (IL-6), total cholesterol (TC), total triglycerides (TG), high-density lipoprotein cholesterol, and low-density lipoprotein cholesterol. Cardiac spiral computed tomography was used to calculate the CAC score, employing the Agatston method. Carotid ultrasonography was performed to assess carotid intima-media thickness (CIMT) and to detect the presence of plaques. Results CAC patients had considerably higher levels of TNF-α (p<0.001), IL-6 (p<0.001), hs-CRP (p=0.006), TC, TG, parathyroid hormone (PTH) (p<0.001) and phosphorus (p<0.001) than non-CAC patients. They also had significantly lower levels of fetuin-A (p<0.001). Fetuin-A was considerably lower in CKD subgroups as CKD progressed. Fetuin-A (p=0.046), age (p=0.009), TNF-α (p=0.027), IL-6 (p=0.005), TG (p=0.002), PTH (p=0.002), and phosphorus (p=0.004) were significant predictors of CAC. CAC and fetuin-A were strong predictors of all-cause mortality and cardiovascular (CV) mortality. Fetuin-A was a significant predictor of CIMT (p=0.045). Conclusion Fetuin-A reliably predicted CAC and CIMT. Fetuin-A and CAC emerged as significant risk factors for all-cause and CV mortality in non-dialysis CKD.
Collapse
Affiliation(s)
- Osama Nady Mohamed
- Department of Internal Medicine, Faculty of Medicine, Minia University, Minya, Egypt
| | | | - Israa Gamal Hassan
- Department of Internal Medicine, Faculty of Medicine, Minia University, Minya, Egypt
| | - Atef Farouk Alakkad
- Department of Internal Medicine, Faculty of Medicine, Minia University, Minya, Egypt
| | - Ashraf Othman
- Department of Clinical Pathology, Faculty of Medicine, Minia University, Minya, Egypt
| | - Amr Setouhi
- Department of Cardiology, Faculty of Medicine, Minia University, Minya, Egypt
| | - Ahmed S. Issa
- Department of Radiology, Faculty of Medicine, Minia University, Minya, Egypt
| |
Collapse
|
23
|
Huang Y, Feng X, Fan H, Luo J, Wang Z, Yang Y, Yang W, Zhang W, Zhou J, Yuan Z, Xiong Y. Circulating miR-423-5p levels are associated with carotid atherosclerosis in patients with chronic kidney disease. Nutr Metab Cardiovasc Dis 2024; 34:1146-1156. [PMID: 38220508 DOI: 10.1016/j.numecd.2023.12.018] [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: 08/23/2023] [Revised: 12/12/2023] [Accepted: 12/20/2023] [Indexed: 01/16/2024]
Abstract
BACKGROUND AND AIMS Carotid atherosclerosis is associated with an elevated risk of stroke in patients with chronic kidney disease. However, the molecular basis for the incidence of carotid atherosclerosis in patients with CKD is poorly understood. Here, we investigated whether circulating miR-423-5p is a crucial link between CKD and carotid atherosclerosis. METHODS AND RESULTS We recruited 375 participants for a cross-sectional study to examine the occurrence of carotid plaque and plaque thicknesses. Levels of miR-423-5p were determined by qPCR analysis. We found that non-dialysis CKD patients had higher circulating exosomal and plasma miR-423-5p levels, and dialysis-dependent patients had lower miR-423-5p levels than non-dialysis CKD patients. After excluding for the influence of dialysis patients, linear regression analysis indicated that levels of circulating miR-423-5p are negatively correlated with eGFR (P < 0.001). Higher plasma miR-423-5p levels were associated with the incidence and severity of carotid plaques. In parallel, we constructed a murine model of CKD with a 5/6 nephrectomy protocol and performed RNA sequencing studies of aortic tissues. Consistent with these findings in CKD patients, circulating exosomal miR-423-5p levels in CKD mice were elevated. Furthermore, our RNA-seq studies indicated that the putative target genes of miR-423-5p were related to oxidative stress functions for aorta of CKD mice. CONCLUSION Levels of miR-423-5p are associated with the presence and severity of carotid plaque in CKD. Data from our mouse model suggests that miR-423-5p likely influences gene expression programs related to oxidative stress in aorta of CKD mice.
Collapse
Affiliation(s)
- Yuzhi Huang
- Cardiovascular Department, First Affiliated Hospital of Xi'an Jiao Tong University, Xi'an, 710061, China; Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi'an, 710061, China
| | - Xueying Feng
- Cardiovascular Department, First Affiliated Hospital of Xi'an Jiao Tong University, Xi'an, 710061, China; Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi'an, 710061, China
| | - Heze Fan
- Cardiovascular Department, First Affiliated Hospital of Xi'an Jiao Tong University, Xi'an, 710061, China; Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi'an, 710061, China
| | - Jian Luo
- Health Management Center, Xi'an People's Hospital (Xi'an Fourth Hospital), Xi'an, Shaanxi, China
| | - Zihao Wang
- Cardiovascular Department, First Affiliated Hospital of Xi'an Jiao Tong University, Xi'an, 710061, China; Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi'an, 710061, China
| | - Yuxuan Yang
- Cardiovascular Department, First Affiliated Hospital of Xi'an Jiao Tong University, Xi'an, 710061, China; Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi'an, 710061, China
| | - Wenbo Yang
- Cardiovascular Department, First Affiliated Hospital of Xi'an Jiao Tong University, Xi'an, 710061, China; Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi'an, 710061, China
| | - Wenjiao Zhang
- Cardiovascular Department, First Affiliated Hospital of Xi'an Jiao Tong University, Xi'an, 710061, China; Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi'an, 710061, China
| | - Juan Zhou
- Cardiovascular Department, First Affiliated Hospital of Xi'an Jiao Tong University, Xi'an, 710061, China; Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi'an, 710061, China
| | - Zuyi Yuan
- Cardiovascular Department, First Affiliated Hospital of Xi'an Jiao Tong University, Xi'an, 710061, China; Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi'an, 710061, China.
| | - Ying Xiong
- Cardiovascular Department, First Affiliated Hospital of Xi'an Jiao Tong University, Xi'an, 710061, China; Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi'an, 710061, China.
| |
Collapse
|
24
|
Ahn SY, Ko GJ, Hwang HS, Jeong KH, Jin K, Kim YG, Moon JY, Lee SH, Lee SY, Yang DH, Jung JY, Oh KH, Lee YK, Kim GH, Kim SW, Kim YH, Lee DY, Hong YA, Park HC, Yoon SA, Choi BS, Ban TH, Kim HJ, Kwon YJ. Understanding the Korean Dialysis Cohort for Mineral, Vascular Calcification, and Fracture (ORCHESTRA) Study: Design, Method, and Baseline Characteristics. Kidney Blood Press Res 2024; 49:326-335. [PMID: 38657581 DOI: 10.1159/000539030] [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: 07/09/2023] [Accepted: 03/23/2024] [Indexed: 04/26/2024] Open
Abstract
INTRODUCTION End-stage renal disease (ESRD) is a growing disease worldwide, including Korea. This is an important condition that affects patient outcome. To provide optimal management for mineral disturbance, vascular calcification, and bone disease in ESRD patients, the Korean dialysis cohort for mineral, vascular calcification, and fracture (ORCHESTRA) study was conducted by enrolling Korean dialysis patients. METHODS Sixteen university-affiliated hospitals and one Veterans' Health Service Medical Center participated in this study. This prospective cohort study enrolled approximately 900 consecutive patients on dialysis between May 2019 and January 2021. Enrolled subjects were evaluated at baseline for demographic information, laboratory tests, radiologic imaging, and bone mineral densitometry (BMD) scans. After enrollment, regular assessments of the patients were performed, and their biospecimens were collected according to the study protocol. The primary outcomes were the occurrence of major adverse cardiovascular events, invasive treatment for peripheral artery disease, and osteoporotic fractures. The secondary outcomes were hospitalization for cerebrovascular disease or progression of abdominal aortic calcification. Participants will be assessed for up to 3 years to determine whether primary or secondary outcomes occur. RESULTS Between May 2019 and January 2021, all participating centers recruited 900 consecutive dialysis patients, including 786 undergoing hemodialysis (HD) and 114 undergoing peritoneal dialysis (PD). The mean age of the subjects was 60.4 ± 12.3 years. Males accounted for 57.7% of the total population. The mean dialysis vintage was 6.1 ± 6.0 years. The HD group was significantly older, had a longer dialysis vintage, and more comorbidities. Overall, the severity of vascular calcification was higher and the level of BMD was lower in the HD group than in the PD group. CONCLUSION This nationwide, multicenter, prospective cohort study focused on chronic kidney disease-mineral and bone disorder and aimed to provide clinical evidence to establish optimal treatment guidelines for Asian dialysis patients.
Collapse
Affiliation(s)
- Shin Young Ahn
- Department of Internal Medicine, Korea University Guro Hospital, Seoul, Republic of Korea,
| | - Gang Jee Ko
- Department of Internal Medicine, Korea University Guro Hospital, Seoul, Republic of Korea
| | - Hyeon Seok Hwang
- Department of Internal Medicine, Kyung Hee University Medical Center, Seoul, Republic of Korea
| | - Kyung Hwan Jeong
- Department of Internal Medicine, Kyung Hee University Medical Center, Seoul, Republic of Korea
| | - Kyubok Jin
- Department of Internal Medicine, Keimyung University Dongsan Medical Center, Daegu, Republic of Korea
| | - Yang Gyun Kim
- Department of Internal Medicine, Kyung Hee University East-West Neo Medical Center, Seoul, Republic of Korea
| | - Ju-Young Moon
- Department of Internal Medicine, Kyung Hee University East-West Neo Medical Center, Seoul, Republic of Korea
| | - Sang Ho Lee
- Department of Internal Medicine, Kyung Hee University East-West Neo Medical Center, Seoul, Republic of Korea
| | - So-Young Lee
- Department of Internal Medicine, CHA Medical School Bundang CHA Medical Center, Seongnam, Republic of Korea
| | - Dong-Ho Yang
- Department of Internal Medicine, CHA Medical School Bundang CHA Medical Center, Seongnam, Republic of Korea
| | - Ji Yong Jung
- Department of Internal Medicine, Gachon University Gil Medical Center, Incheon, Republic of Korea
| | - Kook-Hwan Oh
- Department of Internal Medicine, Seoul National University Hospital, Seoul, Republic of Korea
| | - Young-Ki Lee
- Department of Internal Medicine, Hallym University Kangnam Sacred Heart Hospital, Seoul, Republic of Korea
| | - Gheun-Ho Kim
- Department of Internal Medicine, Hanyang University Seoul Hospital, Seoul, Republic of Korea
| | - Soo Wan Kim
- Department of Internal Medicine, Chonnam National University Medical School and Hospital, Gwangju, Republic of Korea
| | - Yeong Hoon Kim
- Department of Internal Medicine, Inje University Busan Paik Hospital, Busan, Republic of Korea
| | - Dong-Young Lee
- Department of Internal Medicine, Veterans Health Service Medical Center, Seoul, Republic of Korea
| | - Yu Ah Hong
- Department of Internal Medicine, The Catholic University of Korea Daejeon St. Mary's Hospital, Daejeon, Republic of Korea
| | - Hyeong Cheon Park
- Department of Internal Medicine, Gangnam Severance Hospital, Seoul, Republic of Korea
| | - Sun Ae Yoon
- Department of Internal Medicine, The Catholic University of Korea Uijeongbu St. Mary's Hospital, Uijeongbu, Republic of Korea
| | - Bum Soon Choi
- Department of Internal Medicine, The Catholic University of Korea Eunpyeong St. Mary's Hospital, Seoul, Republic of Korea
| | - Tae Hyun Ban
- Department of Internal Medicine, The Catholic University of Korea Eunpyeong St. Mary's Hospital, Seoul, Republic of Korea
| | - Hyo Jin Kim
- Department of Internal Medicine, Pusan National University Hospital, Busan, Republic of Korea
| | - Young Joo Kwon
- Department of Internal Medicine, Korea University Guro Hospital, Seoul, Republic of Korea
| |
Collapse
|
25
|
Gu L, Xia Z, Qing B, Wang W, Chen H, Wang J, Chen Y, Gai Z, Hu R, Yuan Y. Systemic Inflammatory Response Index (SIRI) is associated with all-cause mortality and cardiovascular mortality in population with chronic kidney disease: evidence from NHANES (2001-2018). Front Immunol 2024; 15:1338025. [PMID: 38558798 PMCID: PMC10978803 DOI: 10.3389/fimmu.2024.1338025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Accepted: 02/19/2024] [Indexed: 04/04/2024] Open
Abstract
Objective To examine the correlation between SIRI and the probability of cardiovascular mortality as well as all-cause mortality in individuals with chronic kidney disease. Methods A cohort of 3,262 participants from the US National Health and Nutrition Examination Survey (NHANES) database were included in the study. We categorized participants into five groups based on the stage of chronic kidney disease. A weighted Cox regression model was applied to assess the relationship between SIRI and mortality. Subgroup analyses, Kaplan-Meier survival curves, and ROC curves were conducted. Additionally, restricted cubic spline analysis was employed to elucidate the detailed association between SIRI and hazard ratio (HR). Results This study included a cohort of 3,262 individuals, of whom 1,535 were male (weighted proportion: 42%), and 2,216 were aged 60 or above (weighted proportion: 59%). Following adjustments for covariates like age, sex, race, and education, elevated SIRI remained a significant independent risk factor for cardiovascular mortality (HR=2.50, 95%CI: 1.62-3.84, p<0.001) and all-cause mortality (HR=3.02, 95%CI: 2.03-4.51, p<0.001) in CKD patients. The restricted cubic spline analysis indicated a nonlinear relationship between SIRI and cardiovascular mortality, with SIRI>1.2 identified as an independent risk factor for cardiovascular mortality in CKD patients. Conclusion Heightened SIRI independently poses a risk for both all-cause and cardiovascular mortality in chronic kidney disease patients, with potentially heightened significance in the early stages (Stage I to Stage III) of chronic kidney disease.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | - Yunchang Yuan
- Department of Thoracic Surgery, the Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| |
Collapse
|
26
|
Qi H, Wang B, Zhu L. Independent associations of serum calcium with or without albumin adjustment and serum phosphorus with nonalcoholic fatty liver disease: results from NHANES 1999-2018. Front Endocrinol (Lausanne) 2024; 15:1323990. [PMID: 38505748 PMCID: PMC10948406 DOI: 10.3389/fendo.2024.1323990] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/18/2023] [Accepted: 02/21/2024] [Indexed: 03/21/2024] Open
Abstract
Background The associations of serum calcium and phosphorus with nonalcoholic fatty liver disease (NAFLD) remain unclear. In addition, there may be an effect of albumin correction on the association between serum calcium and NAFLD. We aimed to explore these relationships in the National Health and Nutrition Examination Survey (NHANES). Methods Eligible adult individuals from NHANES 1999-2018 were recruited for the study. We explored the associations of serum calcium, albumin-adjusted serum calcium, and serum phosphorus with NAFLD in multivariable-adjusted regression models. In addition, restricted cubic spline (RCS), stratified analysis, and multiple sensitivity analyses were used for further elaboration. Results The study sample consisted of 20,900 participants, with an observed NAFLD prevalence of 44.65%. Fully adjusted models indicated that serum calcium was inversely associated with NAFLD (odds ratio [OR] and 95% confidence interval [CI] = 0.70 (0.62, 0.78), p<0.0001), whereas albumin-adjusted serum calcium was positively associated with NAFLD (OR and 95% CI=1.59 (1.41, 1.79), p<0.0001). RCS modeling indicated that serum calcium without and with albumin adjustment was linearly(p nonlinear = 0.083) and nonlinearly (p nonlinear < 0.0001) associated with NAFLD, respectively, whereas serum phosphorus showed a U-shaped relationship with NAFLD(p nonlinear < 0.0001). Gender is a significant influence in all associations, and other variables may also have an effect. Sensitivity analyses indicated that these associations were independent of additional significant confounders. Conclusion Serum calcium and phosphorus were significantly associated with the development of NAFLD. These findings suggest the potential clinical significance of serum calcium/phosphorus and albumin levels in individuals at high risk for NAFLD. Our study supports the potential role of serum calcium/phosphorus homeostasis in the pathophysiology of NAFLD and could serve as NAFLD-related biomarkers.
Collapse
Affiliation(s)
| | | | - Lei Zhu
- Department of Hepatobiliary Surgery, Renmin Hospital of Wuhan University, Wuhan, China
| |
Collapse
|
27
|
Jin J, Cheng M, Wu X, Zhang H, Zhang D, Liang X, Qian Y, Guo L, Zhang S, Bai Y, Xu J. Circulating miR-129-3p in combination with clinical factors predicts vascular calcification in hemodialysis patients. Clin Kidney J 2024; 17:sfae038. [PMID: 38524234 PMCID: PMC10960567 DOI: 10.1093/ckj/sfae038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Indexed: 03/26/2024] Open
Abstract
Background Vascular calcification (VC) commonly occurs and seriously increases the risk of cardiovascular events and mortality in patients with hemodialysis. For optimizing individual management, we will develop a diagnostic multivariable prediction model for evaluating the probability of VC. Methods The study was conducted in four steps. First, identification of miRNAs regulating osteogenic differentiation of vascular smooth muscle cells (VSMCs) in calcified condition. Second, observing the role of miR-129-3p on VC in vitro and the association between circulating miR-129-3p and VC in hemodialysis patients. Third, collecting all indicators related to VC as candidate variables, screening predictors from the candidate variables by Lasso regression, developing the prediction model by logistic regression and showing it as a nomogram in training cohort. Last, verifying predictive performance of the model in validation cohort. Results In cell experiments, miR-129-3p was found to attenuate vascular calcification, and in human, serum miR-129-3p exhibited a negative correlation with vascular calcification, suggesting that miR-129-3p could be one of the candidate predictor variables. Regression analysis demonstrated that miR-129-3p, age, dialysis duration and smoking were valid factors to establish the prediction model and nomogram for VC. The area under receiver operating characteristic curve of the model was 0.8698. The calibration curve showed that predicted probability of the model was in good agreement with actual probability and decision curve analysis indicated better net benefit of the model. Furthermore, internal validation through bootstrap process and external validation by another independent cohort confirmed the stability of the model. Conclusion We build a diagnostic prediction model and present it as an intuitive tool based on miR-129-3p and clinical indicators to evaluate the probability of VC in hemodialysis patients, facilitating risk stratification and effective decision, which may be of great importance for reducing the risk of serious cardiovascular events.
Collapse
Affiliation(s)
- Jingjing Jin
- Departments of Nephrology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, PR China
- Hebei Key Laboratory of Vascular Calcification in Kidney Disease, Shijiazhuang, PR China
- Hebei Clinical Research Center for Chronic Kidney Disease, Shijiazhuang, PR China
| | - Meijuan Cheng
- Departments of Nephrology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, PR China
- Hebei Key Laboratory of Vascular Calcification in Kidney Disease, Shijiazhuang, PR China
- Hebei Clinical Research Center for Chronic Kidney Disease, Shijiazhuang, PR China
| | - Xueying Wu
- Departments of Nephrology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, PR China
- Hebei Key Laboratory of Vascular Calcification in Kidney Disease, Shijiazhuang, PR China
- Hebei Clinical Research Center for Chronic Kidney Disease, Shijiazhuang, PR China
| | - Haixia Zhang
- Departments of Nephrology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, PR China
- Hebei Key Laboratory of Vascular Calcification in Kidney Disease, Shijiazhuang, PR China
- Hebei Clinical Research Center for Chronic Kidney Disease, Shijiazhuang, PR China
| | - Dongxue Zhang
- Departments of Nephrology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, PR China
- Hebei Key Laboratory of Vascular Calcification in Kidney Disease, Shijiazhuang, PR China
- Hebei Clinical Research Center for Chronic Kidney Disease, Shijiazhuang, PR China
| | - Xiangnan Liang
- Departments of Nephrology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, PR China
- Hebei Key Laboratory of Vascular Calcification in Kidney Disease, Shijiazhuang, PR China
- Hebei Clinical Research Center for Chronic Kidney Disease, Shijiazhuang, PR China
| | - Yuetong Qian
- Departments of Nephrology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, PR China
- Hebei Key Laboratory of Vascular Calcification in Kidney Disease, Shijiazhuang, PR China
- Hebei Clinical Research Center for Chronic Kidney Disease, Shijiazhuang, PR China
| | - Liping Guo
- Departments of Nephrology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, PR China
- Hebei Key Laboratory of Vascular Calcification in Kidney Disease, Shijiazhuang, PR China
- Hebei Clinical Research Center for Chronic Kidney Disease, Shijiazhuang, PR China
| | - Shenglei Zhang
- Departments of Nephrology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, PR China
- Hebei Key Laboratory of Vascular Calcification in Kidney Disease, Shijiazhuang, PR China
- Hebei Clinical Research Center for Chronic Kidney Disease, Shijiazhuang, PR China
| | - Yaling Bai
- Departments of Nephrology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, PR China
- Hebei Key Laboratory of Vascular Calcification in Kidney Disease, Shijiazhuang, PR China
- Hebei Clinical Research Center for Chronic Kidney Disease, Shijiazhuang, PR China
| | - Jinsheng Xu
- Departments of Nephrology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, PR China
- Hebei Key Laboratory of Vascular Calcification in Kidney Disease, Shijiazhuang, PR China
- Hebei Clinical Research Center for Chronic Kidney Disease, Shijiazhuang, PR China
| |
Collapse
|
28
|
Kofotolios I, Bonios MJ, Adamopoulos M, Mourouzis I, Filippatos G, Boletis JN, Marinaki S, Mavroidis M. The Han:SPRD Rat: A Preclinical Model of Polycystic Kidney Disease. Biomedicines 2024; 12:362. [PMID: 38397964 PMCID: PMC10887417 DOI: 10.3390/biomedicines12020362] [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: 12/29/2023] [Revised: 01/27/2024] [Accepted: 01/30/2024] [Indexed: 02/25/2024] Open
Abstract
Autosomal Dominant Polycystic Kidney Disease (ADPKD) stands as the most prevalent hereditary renal disorder in humans, ultimately culminating in end-stage kidney disease. Animal models carrying mutations associated with polycystic kidney disease have played an important role in the advancement of ADPKD research. The Han:SPRD rat model, carrying an R823W mutation in the Anks6 gene, is characterized by cyst formation and kidney enlargement. The mutated protein, named Samcystin, is localized in cilia of tubular epithelial cells and seems to be involved in cystogenesis. The homozygous Anks6 mutation leads to end-stage renal disease and death, making it a critical factor in kidney development and function. This review explores the utility of the Han:SPRD rat model, highlighting its phenotypic similarity to human ADPKD. Specifically, we discuss its role in preclinical trials and its importance for investigating the pathogenesis of the disease and developing new therapeutic approaches.
Collapse
Affiliation(s)
- Ioannis Kofotolios
- Clinic of Nephrology and Renal Tranplantation, Laiko Hospital, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece
- Center of Basic Research, Biomedical Research Foundation, Academy of Athens, 11527 Athens, Greece (M.M.)
| | - Michael J. Bonios
- Heart Failure and Transplant Unit, Onassis Cardiac Surgery Center, 17674 Athens, Greece;
| | - Markos Adamopoulos
- Center of Basic Research, Biomedical Research Foundation, Academy of Athens, 11527 Athens, Greece (M.M.)
| | - Iordanis Mourouzis
- Department of Pharmacology, National and Kapodistrian University of Athens, 11527 Athens, Greece;
| | - Gerasimos Filippatos
- Department of Cardiology, Attikon University Hospital, Medical School, National and Kapodistrian University of Athens, 12462 Athens, Greece
| | - John N. Boletis
- Clinic of Nephrology and Renal Tranplantation, Laiko Hospital, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece
| | - Smaragdi Marinaki
- Clinic of Nephrology and Renal Tranplantation, Laiko Hospital, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece
| | - Manolis Mavroidis
- Center of Basic Research, Biomedical Research Foundation, Academy of Athens, 11527 Athens, Greece (M.M.)
| |
Collapse
|
29
|
Nakayama M, Kobayashi S, Kusakabe M, Ohara M, Nakanishi K, Akizawa T, Fukagawa M. Tenapanor for peritoneal dialysis patients with hyperphosphatemia: a phase 3 trial. Clin Exp Nephrol 2024; 28:153-164. [PMID: 37910313 PMCID: PMC10808471 DOI: 10.1007/s10157-023-02406-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Accepted: 09/10/2023] [Indexed: 11/03/2023]
Abstract
BACKGROUND Tenapanor is a novel selective inhibitor of intestinal sodium/hydrogen exchanger 3 transporter. This is the first trial to assess the efficacy and safety of tenapanor in Japanese patients with hyperphosphatemia who are undergoing peritoneal dialysis. METHODS This phase 3, open-label, multicenter, single-arm clinical trial targeted patients whose serum phosphorus was within 3.5-7.0 mg/dL with phosphate binders at screening. After phosphate binder washout, tenapanor was orally administered twice-daily, stepwise from 5 to 30 mg/dose for 16 weeks. The primary endpoint, mean change in serum phosphorus level, was evaluated at week 8. The 16-week treatment period was completed with tenapanor alone, and only one phosphate binder type was allowed for combined use after the primary endpoint. RESULTS Of the 54 patients enrolled, 34 completed the study. At week 8, the primary endpoint, mean change in serum phosphorus level (last observation carried forward), was - 1.18 mg/dL (95% confidence interval: - 1.54, - 0.81 mg/dL) with tenapanor. From a baseline value of 7.65 mg/dL, serum phosphorus decreased to 6.14 and 5.44 mg/dL at weeks 8 and 16, respectively, and 46.3% and 76.5% of patients achieved serum phosphorus within 3.5-6.0 mg/dL at week 8 and week 16, respectively. The most common adverse event, diarrhea, occurred in 74.1% of patients; the severity of diarrhea was mild or moderate. Thus, the discontinuation percentage due to diarrhea was low at 5.6%. CONCLUSIONS Administration of tenapanor resulted in a sufficient reduction in serum phosphorus level at week 8 and was considered safe and tolerable. TRIAL REGISTRATION NCT04766385.
Collapse
Affiliation(s)
- Masaaki Nakayama
- Kidney Center, St. Luke's International Hospital, 9-1 Akashi-cho, Chuo-ku, Tokyo, 104-8560, Japan.
| | | | | | - Meiko Ohara
- R&D Division, Kyowa Kirin Co., Ltd., Tokyo, Japan
| | | | - Tadao Akizawa
- Division of Nephrology, Department of Medicine, Showa University School of Medicine, Tokyo, Japan
| | - Masafumi Fukagawa
- Division of Nephrology, Endocrinology, and Metabolism, Department of Internal Medicine, Tokai University School of Medicine, Kanagawa, Japan
| |
Collapse
|
30
|
Yan P, Li J, Zhang Y, Dan X, Wu X, Zhang X, Yang Y, Chen X, Li S, Chen P, Wan Q, Xu Y. Association of Circulating Carbohydrate Antigen 19-9 Level with Type 2 Diabetic Kidney Disease in Chinese Adults: A Cross-Sectional Study. Diabetes Metab Syndr Obes 2024; 17:467-477. [PMID: 38312210 PMCID: PMC10838495 DOI: 10.2147/dmso.s434972] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Accepted: 01/16/2024] [Indexed: 02/06/2024] Open
Abstract
Objective Very few and conflicting data are available regarding the correlation between circulating carbohydrate antigen 19-9 (CA19-9) levels and diabetic kidney disease (DKD) and its components including albuminuria and a low estimated glomerular filtration rate (eGFR). This study aimed to examine the association of circulating CA19-9 and DKD in Chinese patients with type 2 diabetes mellitus (T2DM). Methods A total of 402 hospitalized T2DM patients between September 2017 and December 2021 were included in this cross-sectional study. There were 224 and 178 subjects in non-DKD and DKD groups, respectively. Serum CA19-9 was measured by chemiluminescence method, and its potential relationship with DKD was evaluated by multivariate logistic regression and correlation analyses, and receiver operating characteristic (ROC) curve analysis. Results T2DM patients with DKD had significantly higher serum CA19-9 levels than those without, and serum CA19-9 levels were positively related to urinary albumin-to-creatinine ratio and negatively to eGFR (P<0.01). Multivariate regression analysis revealed that serum CA 19-9 was an independent factor of DKD [odds ratio (OR), 1.018; 95% confidence interval (CI), 1.002-1.035; P<0.05]. Moreover, an increased progressively risk of DKD with an increase in serum CA19-9 quartiles was observed (P for trend <0.001), and T2DM patients in the highest serum CA19-9 quartile were associated with an increased likelihood of DKD when compared to those in the lowest quartile (OR: 2.936, 95% CI 1.129-7.633, P<0.05). Last, the analysis of ROC curves suggested that serum CA 19-9 at a cut of 25.09 U/mL resulted in the highest Youden index with sensitivity 43.8% and 75.4% specificity to predict the presence of DKD. Conclusion These results showed that high circulating CA19-9 was related to DKD and may serve as a useful biomarker of DKD in hospitalized Chinese T2DM patients.
Collapse
Affiliation(s)
- Pijun Yan
- Department of Endocrinology and Metabolism, the Affiliated Hospital of Southwest Medical University, Luzhou, People's Republic of China
- Metabolic Vascular Disease Key Laboratory of Sichuan Province, Luzhou, People's Republic of China
- Sichuan Clinical Research Center for Diabetes and Metabolism, Luzhou, People's Republic of China
- Sichuan Clinical Research Center for Nephropathy, Luzhou, People's Republic of China
- Cardiovascular and Metabolic Diseases Key Laboratory of Luzhou, Luzhou, People's Republic of China
| | - Jia Li
- Department of Endocrinology and Metabolism, the Affiliated Hospital of Southwest Medical University, Luzhou, People's Republic of China
- Metabolic Vascular Disease Key Laboratory of Sichuan Province, Luzhou, People's Republic of China
- Sichuan Clinical Research Center for Diabetes and Metabolism, Luzhou, People's Republic of China
- Sichuan Clinical Research Center for Nephropathy, Luzhou, People's Republic of China
- Cardiovascular and Metabolic Diseases Key Laboratory of Luzhou, Luzhou, People's Republic of China
| | - Yi Zhang
- Department of Endocrinology and Metabolism, the Affiliated Hospital of Southwest Medical University, Luzhou, People's Republic of China
- Metabolic Vascular Disease Key Laboratory of Sichuan Province, Luzhou, People's Republic of China
- Sichuan Clinical Research Center for Diabetes and Metabolism, Luzhou, People's Republic of China
- Sichuan Clinical Research Center for Nephropathy, Luzhou, People's Republic of China
- Cardiovascular and Metabolic Diseases Key Laboratory of Luzhou, Luzhou, People's Republic of China
| | - Xiaofang Dan
- Department of Endocrinology and Metabolism, the Affiliated Hospital of Southwest Medical University, Luzhou, People's Republic of China
- Metabolic Vascular Disease Key Laboratory of Sichuan Province, Luzhou, People's Republic of China
- Sichuan Clinical Research Center for Diabetes and Metabolism, Luzhou, People's Republic of China
- Sichuan Clinical Research Center for Nephropathy, Luzhou, People's Republic of China
- Cardiovascular and Metabolic Diseases Key Laboratory of Luzhou, Luzhou, People's Republic of China
| | - Xian Wu
- Department of Endocrinology and Metabolism, the Affiliated Hospital of Southwest Medical University, Luzhou, People's Republic of China
- Metabolic Vascular Disease Key Laboratory of Sichuan Province, Luzhou, People's Republic of China
- Sichuan Clinical Research Center for Diabetes and Metabolism, Luzhou, People's Republic of China
- Sichuan Clinical Research Center for Nephropathy, Luzhou, People's Republic of China
- Cardiovascular and Metabolic Diseases Key Laboratory of Luzhou, Luzhou, People's Republic of China
| | - Xing Zhang
- Department of Endocrinology and Metabolism, the Affiliated Hospital of Southwest Medical University, Luzhou, People's Republic of China
- Metabolic Vascular Disease Key Laboratory of Sichuan Province, Luzhou, People's Republic of China
- Sichuan Clinical Research Center for Diabetes and Metabolism, Luzhou, People's Republic of China
- Sichuan Clinical Research Center for Nephropathy, Luzhou, People's Republic of China
- Cardiovascular and Metabolic Diseases Key Laboratory of Luzhou, Luzhou, People's Republic of China
| | - Yuxia Yang
- Department of Endocrinology and Metabolism, the Affiliated Hospital of Southwest Medical University, Luzhou, People's Republic of China
- Metabolic Vascular Disease Key Laboratory of Sichuan Province, Luzhou, People's Republic of China
- Sichuan Clinical Research Center for Diabetes and Metabolism, Luzhou, People's Republic of China
- Sichuan Clinical Research Center for Nephropathy, Luzhou, People's Republic of China
- Cardiovascular and Metabolic Diseases Key Laboratory of Luzhou, Luzhou, People's Republic of China
| | - Xiping Chen
- Clinical medical College, Southwest Medical University, Luzhou, People's Republic of China
| | - Shengxi Li
- Clinical medical College, Southwest Medical University, Luzhou, People's Republic of China
| | - Pan Chen
- Department of Endocrinology and Metabolism, the Affiliated Hospital of Southwest Medical University, Luzhou, People's Republic of China
- Metabolic Vascular Disease Key Laboratory of Sichuan Province, Luzhou, People's Republic of China
- Sichuan Clinical Research Center for Diabetes and Metabolism, Luzhou, People's Republic of China
- Sichuan Clinical Research Center for Nephropathy, Luzhou, People's Republic of China
- Cardiovascular and Metabolic Diseases Key Laboratory of Luzhou, Luzhou, People's Republic of China
| | - Qin Wan
- Department of Endocrinology and Metabolism, the Affiliated Hospital of Southwest Medical University, Luzhou, People's Republic of China
- Metabolic Vascular Disease Key Laboratory of Sichuan Province, Luzhou, People's Republic of China
- Sichuan Clinical Research Center for Diabetes and Metabolism, Luzhou, People's Republic of China
- Sichuan Clinical Research Center for Nephropathy, Luzhou, People's Republic of China
- Cardiovascular and Metabolic Diseases Key Laboratory of Luzhou, Luzhou, People's Republic of China
| | - Yong Xu
- Department of Endocrinology and Metabolism, the Affiliated Hospital of Southwest Medical University, Luzhou, People's Republic of China
- Metabolic Vascular Disease Key Laboratory of Sichuan Province, Luzhou, People's Republic of China
- Sichuan Clinical Research Center for Diabetes and Metabolism, Luzhou, People's Republic of China
- Sichuan Clinical Research Center for Nephropathy, Luzhou, People's Republic of China
- Cardiovascular and Metabolic Diseases Key Laboratory of Luzhou, Luzhou, People's Republic of China
| |
Collapse
|
31
|
He L, Li Y, Jin J, Cheng M, Bai Y, Xu J. Comparative efficacy of sodium thiosulfate, bisphosphonates, and cinacalcet for the treatment of vascular calcification in patients with haemodialysis: a systematic review and network meta-analysis. BMC Nephrol 2024; 25:26. [PMID: 38254024 PMCID: PMC10804723 DOI: 10.1186/s12882-024-03460-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Accepted: 01/06/2024] [Indexed: 01/24/2024] Open
Abstract
BACKGROUND Up to now, there is no unequivocal intervention to mitigate vascular calcification (VC) in patients with hemodialysis. This network meta-analysis aimed to systematically evaluate the clinical efficacy of sodium thiosulfate, bisphosphonates, and cinacalcet in treating vascular calcification. METHODS A comprehensive study search was performed using PubMed, Web of Science, the Cochrane Library, EMBASE and China National Knowledge Internet (CNKI) to collect randomized controlled trials (RCTs) of sodium thiosulfate, bisphosphonates, and cinacalcet for vascular calcification among hemodialysis patients. Then, network meta-analysis was conducted using Stata 17.0 software. RESULTS In total, eleven RCTs including 1083 patients were qualified for this meta-analysis. We found that cinacalcet (SMD - 0.59; 95% CI [-0.95, -0.24]) had significant benefit on vascular calcification compared with conventional therapy, while sodium thiosulfate or bisphosphonates did not show such efficiency. Furthermore, as for ranking the efficacy assessment, cinacalcet possessed the highest surface under the cumulative ranking curve (SUCRA) value (88.5%) of lessening vascular calcification and was superior to sodium thiosulfate (50.4%) and bisphosphonates (55.4%). Thus, above results suggested that cinacalcet might be the most promising drug for vascular calcification treatment in hemodialysis patients. Mechanistically, our findings illustrated that cinacalcet reduced serum calcium (SMD - 1.20; 95% CI [-2.08, - 0.33]) and showed the tendency in maintaining the balance of intact Parathyroid Hormone (iPTH) level. CONCLUSIONS This network meta-analysis indicated that cinacalcet appear to be more effective than sodium thiosulfate and bisphosphonates in mitigating vascular calcification through decreasing serum calcium and iPTH. And cinacalcet might be a reasonable option for hemodialysis patients with VC in clinical practice. SYSTEMATIC REVIEW REGISTRATION [ http://www.crd.york.ac.uk/PROSPERO ], identifier [CRD42022379965].
Collapse
Affiliation(s)
- Lei He
- Departments of Nephrology, Hebei Key Laboratory of vascular calcification in kidney disease; Hebei Clinical Research Center for Chronic Kidney Disease, The Fourth Hospital of Hebei Medical University, 12 Jiankang Road, 050011, Shijiazhuang, China
| | - Yuzhe Li
- Departments of Nephrology, Hebei Key Laboratory of vascular calcification in kidney disease; Hebei Clinical Research Center for Chronic Kidney Disease, The Fourth Hospital of Hebei Medical University, 12 Jiankang Road, 050011, Shijiazhuang, China
| | - Jingjing Jin
- Departments of Nephrology, Hebei Key Laboratory of vascular calcification in kidney disease; Hebei Clinical Research Center for Chronic Kidney Disease, The Fourth Hospital of Hebei Medical University, 12 Jiankang Road, 050011, Shijiazhuang, China
| | - Meijuan Cheng
- Departments of Nephrology, Hebei Key Laboratory of vascular calcification in kidney disease; Hebei Clinical Research Center for Chronic Kidney Disease, The Fourth Hospital of Hebei Medical University, 12 Jiankang Road, 050011, Shijiazhuang, China
| | - Yaling Bai
- Departments of Nephrology, Hebei Key Laboratory of vascular calcification in kidney disease; Hebei Clinical Research Center for Chronic Kidney Disease, The Fourth Hospital of Hebei Medical University, 12 Jiankang Road, 050011, Shijiazhuang, China
| | - Jinsheng Xu
- Departments of Nephrology, Hebei Key Laboratory of vascular calcification in kidney disease; Hebei Clinical Research Center for Chronic Kidney Disease, The Fourth Hospital of Hebei Medical University, 12 Jiankang Road, 050011, Shijiazhuang, China.
| |
Collapse
|
32
|
Hao QY, Yan J, Wei JT, Zeng YH, Feng LY, Que DD, Li SC, Guo JB, Fan Y, Ding YF, Zhang XL, Yang PZ, Gao JW, Li ZH. Prevotella copri promotes vascular calcification via lipopolysaccharide through activation of NF-κB signaling pathway. Gut Microbes 2024; 16:2351532. [PMID: 38727248 PMCID: PMC11093026 DOI: 10.1080/19490976.2024.2351532] [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: 10/23/2023] [Accepted: 05/01/2024] [Indexed: 05/16/2024] Open
Abstract
Emerging evidence indicates that alteration of gut microbiota plays an important role in chronic kidney disease (CKD)-related vascular calcification (VC). We aimed to investigate the specific gut microbiota and the underlying mechanism involved in CKD-VC. We identified an increased abundance of Prevotella copri (P. copri) in the feces of CKD rats (induced by using 5/6 nephrectomy followed by a high calcium and phosphate diet) with aortic calcification via amplicon sequencing of 16S rRNA genes. In patients with CKD, we further confirmed a positive correlation between abundance of P. copri and aortic calcification scores. Moreover, oral administration of live P. copri aggravated CKD-related VC and osteogenic differentiation of vascular smooth muscle cells in vivo, accompanied by intestinal destruction, enhanced expression of Toll-like receptor-4 (TLR4), and elevated lipopolysaccharide (LPS) levels. In vitro and ex vivo experiments consistently demonstrated that P. copri-derived LPS (Pc-LPS) accelerated high phosphate-induced VC and VSMC osteogenic differentiation. Mechanistically, Pc-LPS bound to TLR4, then activated the nuclear factor κB (NF-κB) and nucleotide-binding domain, leucine-rich-containing family, pyrin domain-containing-3 (NLRP3) inflammasome signals during VC. Inhibition of NF-κB reduced NLRP3 inflammasome and attenuated Pc-LPS-induced VSMC calcification. Our study clarifies a novel role of P. copri in CKD-related VC, by the mechanisms involving increased inflammation-regulating metabolites including Pc-LPS, and activation of the NF-κB/NLRP3 signaling pathway. These findings highlight P. copri and its-derived LPS as potential therapeutic targets for VC in CKD.
Collapse
MESH Headings
- Animals
- Humans
- Male
- Rats
- Feces/microbiology
- Gastrointestinal Microbiome
- Inflammasomes/metabolism
- Lipopolysaccharides/metabolism
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/pathology
- Myocytes, Smooth Muscle/metabolism
- NF-kappa B/metabolism
- NLR Family, Pyrin Domain-Containing 3 Protein/metabolism
- NLR Family, Pyrin Domain-Containing 3 Protein/genetics
- Osteogenesis/drug effects
- Prevotella/metabolism
- Rats, Sprague-Dawley
- Renal Insufficiency, Chronic/complications
- Renal Insufficiency, Chronic/microbiology
- Renal Insufficiency, Chronic/pathology
- Signal Transduction
- Toll-Like Receptor 4/metabolism
- Toll-Like Receptor 4/genetics
- Vascular Calcification/metabolism
- Vascular Calcification/microbiology
- Vascular Calcification/pathology
Collapse
Affiliation(s)
- Qing-Yun Hao
- Department of Cardiology, Laboratory of Heart Center, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Jing Yan
- Department of Cardiology, Laboratory of Heart Center, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Jin-Tao Wei
- Department of Cardiology, Dongguan Hospital of Southern Medical University, Southern Medical University, Dongguan, China
| | - Yu-Hong Zeng
- Medical Apparatus and Equipment Deployment, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Li-Yun Feng
- Department of Cardiology, Laboratory of Heart Center, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Dong-Dong Que
- Department of Cardiology, Laboratory of Heart Center, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Shi-Chao Li
- Department of Organ Transplantation, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Jing-Bin Guo
- Department of Cardiology, Laboratory of Heart Center, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Ying Fan
- Department of Cardiology, Dongguan Hospital of Southern Medical University, Southern Medical University, Dongguan, China
| | - Yun-Fa Ding
- Department of General Surgery, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Xiu-Li Zhang
- Department of Cardiology, Laboratory of Heart Center, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Ping-Zhen Yang
- Department of Cardiology, Laboratory of Heart Center, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Jing-Wei Gao
- Department of Cardiology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Ze-Hua Li
- Department of Cardiology, Laboratory of Heart Center, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| |
Collapse
|
33
|
Ogata H, Sugawara H, Yamamoto M, Ito H. Phosphate and Coronary Artery Disease in Patients with Chronic Kidney Disease. J Atheroscler Thromb 2024; 31:1-14. [PMID: 37766573 PMCID: PMC10776333 DOI: 10.5551/jat.rv22012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Accepted: 08/07/2023] [Indexed: 09/29/2023] Open
Abstract
Cardiovascular disease (CVD) is the leading cause of death in patients with chronic kidney disease (CKD). Both traditional and CKD-related factors are associated with CVD in CKD patients. Traditional factors that play an important role in the atherosclerotic process directly contribute to a higher risk of coronary artery disease in patients with early-stage CKD. Among CKD-related factors, CKD-mineral and bone disorder plays a critical role in the pathomechanism of nonatherosclerotic diseases, which increases the risk of cardiovascular morbidity and mortality in patients with advanced CKD. Higher serum phosphate levels were significantly associated with cardiovascular events and all-cause mortality in patients with or without CKD. An increased phosphate load, directly and indirectly, promotes arterial medial calcification and left ventricular hypertrophy, both of which predispose patients to coronary artery disease. Calciprotein particles that form in a hyperphosphatemic state promote the transformation of vascular smooth muscle cells (VSMCs) into osteoblastic cells, thereby providing a scaffold for medial calcification in the artery. Increases in fibroblast growth factor-23 and disturbed vitamin D metabolism induced by an excessive phosphate load play a significant role in the development of cardiomyocyte hypertrophy and cardiac fibrosis. Recently, hyperphosphatemia was reported to promote de novo cholesterol synthesis in VSMCs and macrophages, which is likely to contribute to statin resistance in patients with end-stage kidney disease. This review outlines the association between increased phosphate load and coronary artery disease in patients with CKD.
Collapse
Affiliation(s)
- Hiroaki Ogata
- Division of Nephrology, Department of Internal Medicine, Showa University Northern Yokohama Hospital, Yokohama, Japan
- Department of Medical Education, Showa University School of Medicine, Tokyo, Japan
| | - Hirohito Sugawara
- Division of Nephrology, Department of Internal Medicine, Showa University Northern Yokohama Hospital, Yokohama, Japan
| | - Masahiro Yamamoto
- Division of Nephrology, Department of Internal Medicine, Showa University Northern Yokohama Hospital, Yokohama, Japan
| | - Hidetoshi Ito
- Division of Nephrology, Department of Internal Medicine, Showa University Northern Yokohama Hospital, Yokohama, Japan
| |
Collapse
|
34
|
Yang X, Liu Y, Zhu X, Chen P, Xie X, Xu T, Zhang X, Zhao Y. Vascular, valvular and kidney calcification manifested in mouse models of adenine-induced chronic kidney disease. Ren Fail 2023; 45:2228920. [PMID: 37369635 DOI: 10.1080/0886022x.2023.2228920] [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: 03/23/2023] [Revised: 06/19/2023] [Accepted: 06/19/2023] [Indexed: 06/29/2023] Open
Abstract
BACKGROUND Ectopic calcification (EC) involves multiple organ systems in chronic kidney disease (CKD). Previous CKD-animal models primarily focused on a certain histological abnormality but did not show the correlation with calcified development among various tissues. This study compared calcified deposition in various tissues during CKD progression in mice. METHODS Male 8-week-old C57BL/6J mice were randomly allocated to the seven groups: a basic, adenine, high-phosphorus, or adenine and high-phosphorus diet for 12-16 weeks (Ctl16, A12, P16, or AP16, respectively); an adenine diet for 4-6 weeks; and a high-phosphorus or adenine and high-phosphorus diet for 10-12 weeks (A6 + P10, A4 + P12, or A4 + AP12, respectively). RESULTS Compared to the Ctl16 mice, the P16 mice only displayed a slight abnormality in serum calcium and phosphorus; the A12 mice had the most serious kidney impairment; the A4 + P12 and A6 + P10 mice had similar conditions of CKD, mineral abnormalities, and mild calcification in the kidney and aortic valves; the A4 + AP12 and AP16 groups had severe kidney impairment, mineral abnormalities and calcification in the kidneys, aortic valves and aortas. Furthermore, calcium-phosphate particles were deposited not only in the tubulointerstitial compartment but in the glomerular and tubular basement membrane. The elemental composition of EC in various tissues matched the calcification of human cardiovascular tissue as determined by energy dispersive spectroscopy. CONCLUSIONS The severity of CKD was unparalleled with the progression of mineral metabolism disorder and EC. Calcification was closely related in different tissues and observed in the glomerular and tubular basement membranes.
Collapse
Affiliation(s)
- Xin Yang
- Department of Nephrology, Zhong Da Hospital, Southeast University School of Medicine, Nanjing, China
| | - Yuqiu Liu
- Department of Nephrology, Zhong Da Hospital, Southeast University School of Medicine, Nanjing, China
| | - Xiaodong Zhu
- Department of Nephrology, Zhong Da Hospital, Southeast University School of Medicine, Nanjing, China
| | - Pingsheng Chen
- Department of Nephrology, Zhong Da Hospital, Southeast University School of Medicine, Nanjing, China
| | - Xiaotong Xie
- Department of Nephrology, Zhong Da Hospital, Southeast University School of Medicine, Nanjing, China
| | - Tian Xu
- Department of Nephrology, Zhong Da Hospital, Southeast University School of Medicine, Nanjing, China
| | - Xiaoliang Zhang
- Department of Nephrology, Zhong Da Hospital, Southeast University School of Medicine, Nanjing, China
| | - Yu Zhao
- Department of Nephrology, Zhong Da Hospital, Southeast University School of Medicine, Nanjing, China
| |
Collapse
|
35
|
Ding H, Zhu J, Tian Y, Xu L, Song L, Shi Y, Mu D, Chen R, Liu H, Liu B. Relationship between the triglyceride-glucose index and coronary artery calcification in asymptomatic, non-diabetic patients undergoing maintenance hemodialysis. Ren Fail 2023; 45:2200849. [PMID: 37133817 PMCID: PMC10158539 DOI: 10.1080/0886022x.2023.2200849] [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] [Indexed: 05/04/2023] Open
Abstract
OBJECTIVE Coronary artery calcification (CAC) is positively and independently associated with cardiovascular disease (CVD) in patients undergoing maintenance hemodialysis (MHD). Insulin resistance is independently associated with CAC and is an important risk factor for CVD. The triglyceride-glucose (TyG) index is a reliable biomarker of insulin resistance. This cross-sectional, observational study aimed to investigate the relationship between the TyG index and CAC in asymptomatic non-diabetic patients undergoing MHD. METHODS The quantitative coronary artery calcification score (CACS) was calculated and expressed using the Agatston score. The TyG index was calculated as ln [fasting triglyceride (mg/dL) × fasting glucose (mg/dL)/2]. Multiple Poisson regression analysis, Spearman correlation analysis, and receiver operating characteristic (ROC) curves were used to investigate the relationship between the TyG index and CAC. RESULTS The 151 patients were divided into three groups according to the tertiles of the TyG index. With an increase in the TyG index, the CACS significantly increased (Spearman's rho = 0.414, p < 0.001). Poisson regression analysis indicated that the TyG index was independently related to the presence of CAC (prevalence ratio, 1.281 [95% confidence interval, 1.121-1.465], p < 0.001). Furthermore, ROC curve analysis showed that the TyG index was of value in predicting the CAC in asymptomatic non-diabetic patients undergoing MHD, with an area under the curve of 0.667 (p = 0.010). CONCLUSION The TyG index is independently related to the presence of CAC in asymptomatic, non-diabetic patients undergoing MHD.
Collapse
Affiliation(s)
- Hong Ding
- Institute of Nephrology, People's Hospital of Yangzhong city, Zhenjiang, Jiangsu Province, China
| | - Jinhua Zhu
- Institute of Nephrology, People's Hospital of Yangzhong city, Zhenjiang, Jiangsu Province, China
| | - Ying Tian
- Institute of Nephrology, People's Hospital of Yangzhong city, Zhenjiang, Jiangsu Province, China
| | - Li Xu
- Institute of Nephrology, People's Hospital of Yangzhong city, Zhenjiang, Jiangsu Province, China
| | - Lei Song
- Institute of Nephrology, People's Hospital of Yangzhong city, Zhenjiang, Jiangsu Province, China
| | - Ying Shi
- Institute of Nephrology, People's Hospital of Yangzhong city, Zhenjiang, Jiangsu Province, China
| | - Dongxing Mu
- Institute of Nephrology, People's Hospital of Yangzhong city, Zhenjiang, Jiangsu Province, China
| | - Ruoxin Chen
- Institute of Nephrology, Zhongda Hospital, Southeast University School of Medicine, Nanjing, Jiangsu Province, China
| | - Hong Liu
- Institute of Nephrology, Zhongda Hospital, Southeast University School of Medicine, Nanjing, Jiangsu Province, China
| | - Bicheng Liu
- Institute of Nephrology, Zhongda Hospital, Southeast University School of Medicine, Nanjing, Jiangsu Province, China
| |
Collapse
|
36
|
Sommerer C, Müller-Krebs S, Nadal J, Schultheiss UT, Friedrich N, Nauck M, Schmid M, Nußhag C, Reiser J, Eckardt KU, Zeier M, Hayek SS. Prospective Cohort Study of Soluble Urokinase Plasminogen Activation Receptor and Cardiovascular Events in Patients With CKD. Kidney Int Rep 2023; 8:2265-2275. [PMID: 38025216 PMCID: PMC10658273 DOI: 10.1016/j.ekir.2023.08.038] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Accepted: 08/28/2023] [Indexed: 12/01/2023] Open
Abstract
Introduction Soluble urokinase plasminogen activation receptor (suPAR) is an immune-derived pathogenic factor for kidney and atherosclerotic disease. Whether the association between suPAR and cardiovascular (CV) outcomes is dependent on the severity of underlying kidney disease is unclear. Methods We measured serum suPAR levels in 4994 participants (mean age 60 years; 60% men; 36% with diabetes mellitus; mean estimated glomerular filtration rate (eGFR) 49 ml/min per 1.73 m2, SD 18) of the German Chronic Kidney Disease (GCKD) cohort and examined its association with all-cause death, CV death, and major CV events (MACE) across the range of eGFR and urine albumin-to-creatinine ratio (UACR). Results The median suPAR level was 1771 pg/ml (interquartile range [IQR] 1447-2254 pg/ml). SuPAR levels were positively and independently correlated with age, eGFR, UACR, and parathyroid hormone levels. There were 573 deaths, including 190 CV deaths and 683 MACE events at a follow-up time of 6.5 years. In multivariable analyses, suPAR levels (log2) were associated with all-cause death (hazard ratio [HR] 1.36, 95% confidence interval [CI] 1.21-1.53), CV death (HR 1.27, 95% CI 1.03-1.57), and MACE (HR 1.13, 95% CI 1.00-1.28), and were not found to differ according to diabetes mellitus status, baseline eGFR, UACR, or parathyroid hormone levels. In mediation analysis, suPAR's direct effect on all-cause death, CV death, and MACE accounted for 77%, 67%, and 60% of the total effect, respectively; whereas the effect mediated through eGFR accounted for 23%, 34%, and 40%, respectively. Conclusion In a large cohort of individuals with chronic kidney disease (CKD), suPAR levels were associated with mortality and CV outcomes independently of indices of kidney function, consistent with its independent role in the pathogenesis of atherosclerosis.
Collapse
Affiliation(s)
- Claudia Sommerer
- Department of Nephrology, University Hospital Heidelberg, Renal Center, Heidelberg, Germany
| | - Sandra Müller-Krebs
- Department of Nephrology, University Hospital Heidelberg, Renal Center, Heidelberg, Germany
| | - Jennifer Nadal
- Department of Medical Biometry, Informatics, and Epidemiology (IMBIE), University Hospital Bonn, Bonn, Germany
| | - Ulla T. Schultheiss
- Institute of Genetic Epidemiology, Faculty of Medicine and Medical Center, University of Freiburg, Freiburg, Germany
- Department of Medicine IV, Nephrology and Primary Care, Faculty of Medicine and Medical Center - University of Freiburg, Freiburg, Germany
| | - Nele Friedrich
- Institute of Clinical Chemistry and Laboratory Medicine, University Medicine Greifswald, Greifswald, Germany
- DZHK (German Center for Cardiovascular Research), Partner Site Greifswald, University Medicine, Greifswald, Germany
| | - Matthias Nauck
- Institute of Clinical Chemistry and Laboratory Medicine, University Medicine Greifswald, Greifswald, Germany
- DZHK (German Center for Cardiovascular Research), Partner Site Greifswald, University Medicine, Greifswald, Germany
| | - Matthias Schmid
- Department of Medical Biometry, Informatics, and Epidemiology (IMBIE), University Hospital Bonn, Bonn, Germany
| | - Christian Nußhag
- Department of Nephrology, University Hospital Heidelberg, Renal Center, Heidelberg, Germany
| | - Jochen Reiser
- Department of Medicine, Rush University Medical Center, Chicago, Illinois, USA
| | - Kai-Uwe Eckardt
- Department of Nephrology and Medical Intensive Care, Charité, Universitätsmedizin Berlin, Berlin, Germany
- Department of Nephrology and Hypertension, Universitätsklinikum Erlangen, Friedrich-Alexander University Erlangen-Nürnberg, Erlangen, Germany
| | - Martin Zeier
- Department of Nephrology, University Hospital Heidelberg, Renal Center, Heidelberg, Germany
| | - Salim S. Hayek
- Department of Medicine, Division of Cardiology, University of Michigan, Michigan, USA
| |
Collapse
|
37
|
Navab F, Rouhani MH, Moeinzadeh F, Clark CCT, Ziaei R. The effects of oral sodium bicarbonate supplementation on anthropometric measures in patients with chronic kidney disease: A systematic review and meta-analysis of randomized clinical trials. Food Sci Nutr 2023; 11:6749-6760. [PMID: 37970385 PMCID: PMC10630834 DOI: 10.1002/fsn3.3627] [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: 03/09/2023] [Revised: 08/01/2023] [Accepted: 08/04/2023] [Indexed: 11/17/2023] Open
Abstract
Metabolic acidosis (MA) may play a key role in the pathogenesis of protein-energy wasting (PEW) in patients with chronic kidney disease (CKD). To present a comprehensive synthesis of the effect of oral sodium bicarbonate (SB) supplementation on anthropometric measures in patients with CKD, a systematic review was undertaken in PubMed/MEDLINE, Web of Science, Cochrane CENTRAL, and Google Scholar, of relevant articles published prior to September 2022. The summary statistics of effect size, nonstandardized weighted mean difference (WMD), and 95% confidence interval (CI) were used to compare the effects of SB supplementation on anthropometric parameters vs. control group. To detect probable sources of heterogeneity, a series of predefined subgroup analyses were conducted. In total, 17 studies with 21 treatment arms, including 2203 participants (1149 cases, 1054 controls), met our inclusion criteria and were included in the meta-analysis. SB supplementation had no significant effect on body weight (BW), midarm muscle circumference (MAMC), or lean body mass (LBM) in patients with CKD. There was a significant increase in body mass index (BMI) (MD: 0.59 kg/m2, 95% CI: 0.25 to 0.93, p = 0.001) after SB supplementation in the overall analysis. In subgroup analysis, LBM was increased in studies that were ≥ 24-week duration (MD: 1.81 kg, 95% CI: 0.81 to 2.81) and in participants with BMI lower than 27 kg/m2 (MD: 1.81 mg/L, 95% CI: 0.81 to 2.81). SB supplementation may yield increases in BMI in predialysis CKD patients. However, our findings did not support the beneficial effects of SB supplementation on other anthropometric outcomes. There is an evident need for long-term high-quality interventions to confirm these findings.
Collapse
Affiliation(s)
- Fatemeh Navab
- Student Research Committee, Nutrition and Food Security Research Center and Department of Community Nutrition, School of Nutrition and Food ScienceIsfahan University of Medical SciencesIsfahanIran
| | - Mohammad Hossein Rouhani
- Nutrition and Food Security Research Center, Department of Community Nutrition, School of Nutrition and Food ScienceIsfahan University of Medical SciencesIsfahanIran
| | - Firouzeh Moeinzadeh
- Isfahan Kidney Diseases Research CenterIsfahan University of Medical SciencesIsfahanIran
| | | | - Rahele Ziaei
- Nutrition and Food Security Research Center, Department of Community Nutrition, School of Nutrition and Food ScienceIsfahan University of Medical SciencesIsfahanIran
| |
Collapse
|
38
|
Reimer KC, Nadal J, Meiselbach H, Schmid M, Schultheiss UT, Kotsis F, Stockmann H, Friedrich N, Nauck M, Krane V, Eckardt KU, Schneider MP, Kramann R, Floege J, Saritas T. Association of mineral and bone biomarkers with adverse cardiovascular outcomes and mortality in the German Chronic Kidney Disease (GCKD) cohort. Bone Res 2023; 11:52. [PMID: 37857629 PMCID: PMC10587182 DOI: 10.1038/s41413-023-00291-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Accepted: 09/07/2023] [Indexed: 10/21/2023] Open
Abstract
Mineral and bone disorder (MBD) in chronic kidney disease (CKD) is tightly linked to cardiovascular disease (CVD). In this study, we aimed to compare the prognostic value of nine MBD biomarkers to determine those associated best with adverse cardiovascular (CV) outcomes and mortality. In 5 217 participants of the German CKD (GCKD) study enrolled with an estimated glomerular filtration rate (eGFR) between 30-60 mL·min-1 per 1.73 m2 or overt proteinuria, serum osteoprotegerin (OPG), C-terminal fibroblast growth factor-23 (FGF23), intact parathyroid hormone (iPTH), bone alkaline phosphatase (BAP), cross-linked C-telopeptide of type 1 collagen (CTX1), procollagen 1 intact N-terminal propeptide (P1NP), phosphate, calcium, and 25-OH vitamin D were measured at baseline. Participants with missing values among these parameters (n = 971) were excluded, leaving a total of 4 246 participants for analysis. During a median follow-up of 6.5 years, 387 non-CV deaths, 173 CV deaths, 645 nonfatal major adverse CV events (MACEs) and 368 hospitalizations for congestive heart failure (CHF) were observed. OPG and FGF23 were associated with all outcomes, with the highest hazard ratios (HRs) for OPG. In the final Cox regression model, adjusted for CV risk factors, including kidney function and all other investigated biomarkers, each standard deviation increase in OPG was associated with non-CV death (HR 1.76, 95% CI: 1.35-2.30), CV death (HR 2.18, 95% CI: 1.50-3.16), MACE (HR 1.38, 95% CI: 1.12-1.71) and hospitalization for CHF (HR 2.05, 95% CI: 1.56-2.69). Out of the nine biomarkers examined, stratification based on serum OPG best identified the CKD patients who were at the highest risk for any adverse CV outcome and mortality.
Collapse
Affiliation(s)
- Katharina Charlotte Reimer
- Department of Nephrology, Rheumatology, and Clinical Immunology, University Hospital RWTH Aachen, Aachen, Germany
- Institute of Experimental Medicine and Systems Biology, RWTH Aachen University, Aachen, Germany
- Institute for Cell and Tumor Biology, RWTH Aachen University, Aachen, Germany
| | - Jennifer Nadal
- Institute of Medical Biometry, Informatics and Epidemiology, University Hospital of Bonn, Bonn, Germany
| | - Heike Meiselbach
- Department of Nephrology and Hypertension, University of Erlangen-Nürnberg, Erlangen, Germany
| | - Matthias Schmid
- Institute of Medical Biometry, Informatics and Epidemiology, University Hospital of Bonn, Bonn, Germany
| | - Ulla T Schultheiss
- Institute of Genetic Epidemiology, Faculty of Medicine and Medical Center - University of Freiburg, Freiburg, Germany
- Department of Medicine IV - Nephrology and Primary Care, Faculty of Medicine and Medical Center - University of Freiburg, Freiburg, Germany
| | - Fruzsina Kotsis
- Institute of Genetic Epidemiology, Faculty of Medicine and Medical Center - University of Freiburg, Freiburg, Germany
- Department of Medicine IV - Nephrology and Primary Care, Faculty of Medicine and Medical Center - University of Freiburg, Freiburg, Germany
| | - Helena Stockmann
- Department of Nephrology and Medical Intensive Care, Charité-Universitätsmedizin Berlin, Berlin, Germany
- Department of Nephrology, University Medical Center Regensburg, Regensburg, Germany
| | - Nele Friedrich
- Institute of Clinical Chemistry and Laboratory Medicine, University Medicine Greifswald, Greifswald, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Greifswald, University Medicine, Greifswald, Germany
| | - Matthias Nauck
- Institute of Clinical Chemistry and Laboratory Medicine, University Medicine Greifswald, Greifswald, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Greifswald, University Medicine, Greifswald, Germany
| | - Vera Krane
- Department of Medicine I, Division of Nephrology, University Hospital Würzburg, Würzburg, Germany
| | - Kai-Uwe Eckardt
- Department of Nephrology and Hypertension, University of Erlangen-Nürnberg, Erlangen, Germany
- Department of Nephrology and Medical Intensive Care, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Markus P Schneider
- Department of Nephrology and Hypertension, University of Erlangen-Nürnberg, Erlangen, Germany
| | - Rafael Kramann
- Department of Nephrology, Rheumatology, and Clinical Immunology, University Hospital RWTH Aachen, Aachen, Germany
- Institute of Experimental Medicine and Systems Biology, RWTH Aachen University, Aachen, Germany
| | - Jürgen Floege
- Department of Nephrology, Rheumatology, and Clinical Immunology, University Hospital RWTH Aachen, Aachen, Germany
| | - Turgay Saritas
- Department of Nephrology, Rheumatology, and Clinical Immunology, University Hospital RWTH Aachen, Aachen, Germany.
- Institute of Experimental Medicine and Systems Biology, RWTH Aachen University, Aachen, Germany.
| |
Collapse
|
39
|
Martínez-Hernández SL, Muñoz-Ortega MH, Ávila-Blanco ME, Medina-Pizaño MY, Ventura-Juárez J. Novel Approaches in Chronic Renal Failure without Renal Replacement Therapy: A Review. Biomedicines 2023; 11:2828. [PMID: 37893201 PMCID: PMC10604533 DOI: 10.3390/biomedicines11102828] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2023] [Revised: 09/28/2023] [Accepted: 10/12/2023] [Indexed: 10/29/2023] Open
Abstract
Chronic kidney disease (CKD) is characterized by renal parenchymal damage leading to a reduction in the glomerular filtration rate. The inflammatory response plays a pivotal role in the tissue damage contributing to renal failure. Current therapeutic options encompass dietary control, mineral salt regulation, and management of blood pressure, blood glucose, and fatty acid levels. However, they do not effectively halt the progression of renal damage. This review critically examines novel therapeutic avenues aimed at ameliorating inflammation, mitigating extracellular matrix accumulation, and fostering renal tissue regeneration in the context of CKD. Understanding the mechanisms sustaining a proinflammatory and profibrotic state may offer the potential for targeted pharmacological interventions. This, in turn, could pave the way for combination therapies capable of reversing renal damage in CKD. The non-replacement phase of CKD currently faces a dearth of efficacious therapeutic options. Future directions encompass exploring vaptans as diuretics to inhibit water absorption, investigating antifibrotic agents, antioxidants, and exploring regenerative treatment modalities, such as stem cell therapy and novel probiotics. Moreover, this review identifies pharmaceutical agents capable of mitigating renal parenchymal damage attributed to CKD, targeting molecular-level signaling pathways (TGF-β, Smad, and Nrf2) that predominate in the inflammatory processes of renal fibrogenic cells.
Collapse
Affiliation(s)
- Sandra Luz Martínez-Hernández
- Departamento de Microbiología, Centro de Ciencias Básicas, Universidad Autónoma de Aguascalientes, Aguascalientes 20100, Ags, Mexico
| | - Martín Humberto Muñoz-Ortega
- Departamento de Química, Centro de Ciencias Básicas, Universidad Autónoma de Aguascalientes, Aguascalientes 20100, Ags, Mexico
| | - Manuel Enrique Ávila-Blanco
- Departamento de Morfología, Centro de Ciencias Básicas, Universidad Autónoma de Aguascalientes, Aguascalientes 20100, Ags, Mexico
| | - Mariana Yazmin Medina-Pizaño
- Departamento de Morfología, Centro de Ciencias Básicas, Universidad Autónoma de Aguascalientes, Aguascalientes 20100, Ags, Mexico
| | - Javier Ventura-Juárez
- Departamento de Morfología, Centro de Ciencias Básicas, Universidad Autónoma de Aguascalientes, Aguascalientes 20100, Ags, Mexico
| |
Collapse
|
40
|
Watanabe Y, Mitomo S, Naganuma T, Nakajima A, Matsuoka S, Tahara S, Okutsu M, Nakamura S, Nakamura S. Impact of Stent Expansion Index on Stent Failure After Left Main Stenting. Am J Cardiol 2023; 205:164-172. [PMID: 37598602 DOI: 10.1016/j.amjcard.2023.07.157] [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: 04/19/2023] [Revised: 07/20/2023] [Accepted: 07/26/2023] [Indexed: 08/22/2023]
Abstract
Impact of the stent expansion index (EXPI) in percutaneous coronary intervention (PCI) for unprotected left main distal bifurcation lesions (ULMD) has been not completely understood especially in current-generation drug-eluting stent (cDES) era. We evaluated the impact of EXPI on clinical outcomes after PCI with cDES for ULMD. We identified 342 patients treated with cDES for ULMD and postintervention intravascular ultrasound between January 2010 and December 2019. In this study, the ratio of minimum stent area (MSA) to reference vessel area at the MSA site was adopted to assess the stent expansion. We defined the patients with the first and second tertile as low-intermediate EXPI group and those with the third tertile as high EXPI group and compared the clinical outcomes between both groups. The primary end point was target lesion failure (TLF). TLF was defined as a composite of cardiac death, target lesion revascularization (TLR) ,and myocardial infarction. The MSA was located in the ostium of left anterior descending coronary artery in most cases (318 of 342 patients; 93.0%). There were no significant differences between both groups in the baseline clinical, lesion, and procedural characteristics. The high EXPI group had lower TLF rate than the low-intermediate EXPI group (10.2% vs 19.9%, log-rank p = 0.033). In conclusion, this is the first report that the higher ratio of MSA to reference vessel area at the MSA site, which was defined as stent EXPI, was associated with more favorable clinical outcomes after PCI for ULMD.
Collapse
Affiliation(s)
- Yusuke Watanabe
- Department of interventional cardiology, Interventional Cardiology Unit, New Tokyo Hospital, Chiba, Japan.
| | - Satoru Mitomo
- Department of interventional cardiology, Interventional Cardiology Unit, New Tokyo Hospital, Chiba, Japan
| | - Toru Naganuma
- Department of interventional cardiology, Interventional Cardiology Unit, New Tokyo Hospital, Chiba, Japan
| | - Akihiro Nakajima
- Department of interventional cardiology, Interventional Cardiology Unit, New Tokyo Hospital, Chiba, Japan
| | - Satoshi Matsuoka
- Department of interventional cardiology, Interventional Cardiology Unit, New Tokyo Hospital, Chiba, Japan
| | - Satoko Tahara
- Department of interventional cardiology, Interventional Cardiology Unit, New Tokyo Hospital, Chiba, Japan
| | - Masaaki Okutsu
- Department of interventional cardiology, Interventional Cardiology Unit, New Tokyo Hospital, Chiba, Japan
| | - Shotaro Nakamura
- Department of interventional cardiology, Interventional Cardiology Unit, New Tokyo Hospital, Chiba, Japan
| | - Sunao Nakamura
- Department of interventional cardiology, Interventional Cardiology Unit, New Tokyo Hospital, Chiba, Japan
| |
Collapse
|
41
|
Luo H, Feng J, Zhang Y, Wang J, Xue G, Huang X, You S, Dong H, Li L, Li J, Xiao H, Ai X, Li X, Huang B. Efficacy and safety of tenapanor in hemodialysis patients with hyperphosphatemia: A systematic review and meta-analysis of randomized placebo-controlled trials. Ther Apher Dial 2023; 27:839-847. [PMID: 37349983 DOI: 10.1111/1744-9987.14028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Revised: 05/30/2023] [Accepted: 06/13/2023] [Indexed: 06/24/2023]
Abstract
BACKGROUND The effects of tenapanor in reducing serum phosphorus in hemodialysis patients with hyperphosphatemia are uncertain and no relevant meta-analysis has been conducted. We performed a meta-analysis of randomized placebo-controlled trials to evaluate the efficacy and safety of tenapanor. METHODS All randomized controlled trials of tenapanor were searched up to 1 August 2022. The primary endpoint was the change in serum phosphorus level from baseline with tenapanor and placebo. Data on drug-related adverse events (AEs), gastrointestinal AEs and diarrhea were collected to determine the safety of tenapanor. RESULTS There were 533 patients throughout five trials that were eligible. Tenapanor significantly lowered blood phosphorus level by 1.79 mg/dl in the mean difference than the placebo. Diarrhea, gastrointestinal AEs, and drug-related AEs were more severe than placebo. CONCLUSIONS This meta-analysis showed that although drug side effects were common, tenapanor significantly reduced serum phosphorus level in hemodialysis patients.
Collapse
Affiliation(s)
- Houli Luo
- Department of Radiology, Cheng du First People's Hospital, Chengdu, Sichuan, China
| | - Jian Feng
- Department of Critical Care Medicine, The General Hospital of Western Theater Command, Chengdu, Sichuan, China
| | - Yanbiao Zhang
- Department of Burn and Plastic, The General Hospital of Western Theater Command, Chengdu, Sichuan, China
| | - Jie Wang
- Department of Critical Care Medicine, The General Hospital of Western Theater Command, Chengdu, Sichuan, China
| | - Gang Xue
- Department of Burn and Plastic, The General Hospital of Western Theater Command, Chengdu, Sichuan, China
| | - Xi Huang
- Department of Burn and Plastic, The General Hospital of Western Theater Command, Chengdu, Sichuan, China
| | - Shuang You
- Department of Burn and Plastic, The General Hospital of Western Theater Command, Chengdu, Sichuan, China
| | - Hongfei Dong
- Department of Burn and Plastic, The General Hospital of Western Theater Command, Chengdu, Sichuan, China
| | - Lingfan Li
- Department of Burn and Plastic, The General Hospital of Western Theater Command, Chengdu, Sichuan, China
| | - Juncheng Li
- Department of Burn and Plastic, The General Hospital of Western Theater Command, Chengdu, Sichuan, China
| | - Hualin Xiao
- Department of Burn and Plastic, The General Hospital of Western Theater Command, Chengdu, Sichuan, China
| | - Xiang Ai
- Department of Burn and Plastic, The General Hospital of Western Theater Command, Chengdu, Sichuan, China
| | - Xianhui Li
- Department of Burn and Plastic, The General Hospital of Western Theater Command, Chengdu, Sichuan, China
| | - Bo Huang
- Department of Burn and Plastic, The General Hospital of Western Theater Command, Chengdu, Sichuan, China
| |
Collapse
|
42
|
Yang J, Pei T, Su G, Duan P, Liu X. AnnexinA6: a potential therapeutic target gene for extracellular matrix mineralization. Front Cell Dev Biol 2023; 11:1201200. [PMID: 37727505 PMCID: PMC10506415 DOI: 10.3389/fcell.2023.1201200] [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: 04/06/2023] [Accepted: 08/10/2023] [Indexed: 09/21/2023] Open
Abstract
The mineralization of the extracellular matrix (ECM) is an essential and crucial process for physiological bone formation and pathological calcification. The abnormal function of ECM mineralization contributes to the worldwide risk of developing mineralization-related diseases; for instance, vascular calcification is attributed to the hyperfunction of ECM mineralization, while osteoporosis is due to hypofunction. AnnexinA6 (AnxA6), a Ca2+-dependent phospholipid-binding protein, has been extensively reported as an essential target in mineralization-related diseases such as osteoporosis, osteoarthritis, atherosclerosis, osteosarcoma, and calcific aortic valve disease. To date, AnxA6, as the largest member of the Annexin family, has attracted much attention due to its significant contribution to matrix vesicles (MVs) production and release, MVs-ECM interaction, cytoplasmic Ca2+ influx, and maturation of hydroxyapatite, making it an essential target in ECM mineralization. In this review, we outlined the recent advancements in the role of AnxA6 in mineralization-related diseases and the potential mechanisms of AnxA6 under normal and mineralization-related pathological conditions. AnxA6 could promote ECM mineralization for bone regeneration in the manner described previously. Therefore, AnxA6 may be a potential osteogenic target for ECM mineralization.
Collapse
Affiliation(s)
| | | | | | | | - Xiaoheng Liu
- Institute of Biomedical Engineering, West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu, China
| |
Collapse
|
43
|
Cozzolino M, Maffei Faccioli F, Cara A, Boni Brivio G, Rivela F, Ciceri P, Magagnoli L, Galassi A, Barbuto S, Speciale S, Minicucci C, Cianciolo G. Future treatment of vascular calcification in chronic kidney disease. Expert Opin Pharmacother 2023; 24:2041-2057. [PMID: 37776230 DOI: 10.1080/14656566.2023.2266381] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Accepted: 09/29/2023] [Indexed: 10/02/2023]
Abstract
INTRODUCTION Cardiovascular disease (CVD) is one of the global leading causes of morbidity and mortality in chronic kidney disease (CKD) patients. Vascular calcification (VC) is a major cause of CVD in this population and is the consequence of complex interactions between inhibitor and promoter factors leading to pathological deposition of calcium and phosphate in soft tissues. Different pathological landscapes are associated with the development of VC, such as endothelial dysfunction, oxidative stress, chronic inflammation, loss of mineralization inhibitors, release of calcifying extracellular vesicles (cEVs) and circulating calcifying cells. AREAS COVERED In this review, we examined the literature and summarized the pathophysiology, biomarkers and focused on the treatments of VC. EXPERT OPINION Even though there is no consensus regarding specific treatment options, we provide the currently available treatment strategies that focus on phosphate balance, correction of vitamin D and vitamin K deficiencies, avoidance of both extremes of bone turnover, normalizing calcium levels and reduction of inflammatory response and the potential and promising therapeutic approaches liketargeting cellular mechanisms of calcification (e.g. SNF472, TNAP inhibitors).Creating novel scores to detect in advance VC and implementing targeted therapies is crucial to treat them and improve the future management of these patients.
Collapse
Affiliation(s)
- Mario Cozzolino
- Renal Division, Department of Health Sciences, ASST Santi Paolo e Carlo, University of Milan, Milan, Italy
| | - Federico Maffei Faccioli
- Renal Division, Department of Health Sciences, ASST Santi Paolo e Carlo, University of Milan, Milan, Italy
| | - Anila Cara
- Renal Division, Department of Health Sciences, ASST Santi Paolo e Carlo, University of Milan, Milan, Italy
| | - Giulia Boni Brivio
- Renal Division, Department of Health Sciences, ASST Santi Paolo e Carlo, University of Milan, Milan, Italy
| | - Francesca Rivela
- Renal Division, Department of Health Sciences, ASST Santi Paolo e Carlo, University of Milan, Milan, Italy
| | - Paola Ciceri
- Renal Division, Department of Health Sciences, ASST Santi Paolo e Carlo, University of Milan, Milan, Italy
| | - Lorenza Magagnoli
- Renal Division, Department of Health Sciences, ASST Santi Paolo e Carlo, University of Milan, Milan, Italy
| | - Andrea Galassi
- Renal Division, Department of Health Sciences, ASST Santi Paolo e Carlo, University of Milan, Milan, Italy
| | - Simona Barbuto
- Nephrology, Dialysis and Renal Transplant Unit, IRCCS-Azienda Ospedaliero-Universitaria di Bologna, Alma Mater Studiorum University of Bologna, Bologna, Italy
| | - Serena Speciale
- Nephrology, Dialysis and Renal Transplant Unit, IRCCS-Azienda Ospedaliero-Universitaria di Bologna, Alma Mater Studiorum University of Bologna, Bologna, Italy
| | - Carlo Minicucci
- Nephrology, Dialysis and Renal Transplant Unit, IRCCS-Azienda Ospedaliero-Universitaria di Bologna, Alma Mater Studiorum University of Bologna, Bologna, Italy
| | - Giuseppe Cianciolo
- Nephrology, Dialysis and Renal Transplant Unit, IRCCS-Azienda Ospedaliero-Universitaria di Bologna, Alma Mater Studiorum University of Bologna, Bologna, Italy
| |
Collapse
|
44
|
Luo H, Feng J, Xue G, Zhang Y, Li Y, Huang X, Chen X, You S, Dong H, Li L, Li J, Xiao H, Ai X, Li X, Huang B. Comparative Efficacy and Acceptability of 12 Phosphorus-Lowering Drugs in Adults with Hyperphosphatemia and Chronic Kidney Disease: A Systematic Review and Network Meta-Analysis. Blood Purif 2023; 52:609-620. [PMID: 37591223 DOI: 10.1159/000531577] [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: 11/20/2022] [Accepted: 06/09/2023] [Indexed: 08/19/2023]
Abstract
BACKGROUND Hyperphosphatemia is associated with cardiovascular morbidity and mortality in adults with chronic kidney disease (CKD). Drug therapy has an irreplaceable role in the management of hyperphosphatemia. OBJECTIVES We aimed to compare and rank phosphorus-lowering drugs, including phosphate binder and nonphosphate binder, in hyperphosphatemia adults with CKD. METHODS We did a systematic review and frequentist random-effect network meta-analysis. We searched in PubMed, Cochrane Library, Web of Science, and Embase from inception to February 1, 2023, for randomized controlled trials of 12 phosphorus-lowering drugs in adults with hyperphosphatemia and CKD. Primary outcomes were efficacy (changes in serum phosphorus) and acceptability (treatment withdrawals due to any cause). We ranked each drug according to the value of surface under the cumulative ranking curve. We applied the Confidence in Network Meta-Analysis frameworks to rate the certainty of evidence. This study was registered with PROSPERO, number CRD42022322270. RESULTS We identified 2,174 citations, and of these, we included 94 trials comprising 14,459 participants and comparing 13 drugs or placebo. In terms of efficacy, except for niacinamide, all drugs lowered the level of serum phosphorus compared with placebo, with mean difference ranging between -1.61 (95% credible interval [CrI], -2.60 to -0.62) mg/dL for magnesium carbonate and -0.85 (-1.66 to -0.05) mg/dL for bixalomer. Only ferric citrate with odds ratios 0.56 (95% CrI: 0.36-0.89) was significantly associated with fewer dropouts for acceptability. Of the 94 trials, 43 (46%), 7 (7%), and 44 (47%) trials were rated as high, moderate, and low risk of bias, respectively, the certainty of the evidence was moderate to very low. CONCLUSIONS Magnesium carbonate has the best phosphorus-lowering effect in hyperphosphatemia adults with CKD; considering efficacy and acceptability, ferric citrate shows evidence to be the most appropriate drug with or without dialysis.
Collapse
Affiliation(s)
- Houli Luo
- Department of Radiology, Chengdu First People's Hospital, Chengdu, China
| | - Jian Feng
- Department of Critical Care Medicine, The General Hospital of Western Theater Command, Chengdu, China
| | - Gang Xue
- Department of Burn and Plastic, The General Hospital of Western Theater Command, Chengdu, China
| | - Yanbiao Zhang
- Department of Burn and Plastic, The General Hospital of Western Theater Command, Chengdu, China
| | - Yunming Li
- Department of Information, The General Hospital of Western Theater Command, Chengdu, China
| | - Xi Huang
- Department of Burn and Plastic, The General Hospital of Western Theater Command, Chengdu, China
| | - Xin Chen
- Department of Neurosurgery, The General Hospital of Western Theater Command, Chengdu, China
| | - Shuang You
- Department of Burn and Plastic, The General Hospital of Western Theater Command, Chengdu, China
| | - Hongfei Dong
- Department of Burn and Plastic, The General Hospital of Western Theater Command, Chengdu, China
| | - Lingfan Li
- Department of Burn and Plastic, The General Hospital of Western Theater Command, Chengdu, China
| | - Juncheng Li
- Department of Burn and Plastic, The General Hospital of Western Theater Command, Chengdu, China
| | - Hualin Xiao
- Department of Burn and Plastic, The General Hospital of Western Theater Command, Chengdu, China
| | - Xiang Ai
- Department of Burn and Plastic, The General Hospital of Western Theater Command, Chengdu, China
| | - Xianhui Li
- Department of Burn and Plastic, The General Hospital of Western Theater Command, Chengdu, China
| | - Bo Huang
- Department of Burn and Plastic, The General Hospital of Western Theater Command, Chengdu, China
| |
Collapse
|
45
|
Xu S, Wang F, Mai P, Peng Y, Shu X, Nie R, Zhang H. Mechanism Analysis of Vascular Calcification Based on Fluid Dynamics. Diagnostics (Basel) 2023; 13:2632. [PMID: 37627891 PMCID: PMC10453151 DOI: 10.3390/diagnostics13162632] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2023] [Revised: 08/05/2023] [Accepted: 08/07/2023] [Indexed: 08/27/2023] Open
Abstract
Vascular calcification is the abnormal deposition of calcium phosphate complexes in blood vessels, which is regarded as the pathological basis of multiple cardiovascular diseases. The flowing blood exerts a frictional force called shear stress on the vascular wall. Blood vessels have different hydrodynamic properties due to discrepancies in geometric and mechanical properties. The disturbance of the blood flow in the bending area and the branch point of the arterial tree produces a shear stress lower than the physiological magnitude of the laminar shear stress, which can induce the occurrence of vascular calcification. Endothelial cells sense the fluid dynamics of blood and transmit electrical and chemical signals to the full-thickness of blood vessels. Through crosstalk with endothelial cells, smooth muscle cells trigger osteogenic transformation, involved in mediating vascular intima and media calcification. In addition, based on the detection of fluid dynamics parameters, emerging imaging technologies such as 4D Flow MRI and computational fluid dynamics have greatly improved the early diagnosis ability of cardiovascular diseases, showing extremely high clinical application prospects.
Collapse
Affiliation(s)
- Shuwan Xu
- Department of Cardiology, The Eighth Affiliated Hospital of Sun Yat-Sen University, Shenzhen 518033, China; (S.X.); (F.W.); (P.M.)
| | - Feng Wang
- Department of Cardiology, The Eighth Affiliated Hospital of Sun Yat-Sen University, Shenzhen 518033, China; (S.X.); (F.W.); (P.M.)
| | - Peibiao Mai
- Department of Cardiology, The Eighth Affiliated Hospital of Sun Yat-Sen University, Shenzhen 518033, China; (S.X.); (F.W.); (P.M.)
| | - Yanren Peng
- Department of Cardiology, Sun Yat-Sen Memorial Hospital of Sun Yat-Sen University, Guangzhou 510120, China; (Y.P.); (X.S.)
| | - Xiaorong Shu
- Department of Cardiology, Sun Yat-Sen Memorial Hospital of Sun Yat-Sen University, Guangzhou 510120, China; (Y.P.); (X.S.)
| | - Ruqiong Nie
- Department of Cardiology, Sun Yat-Sen Memorial Hospital of Sun Yat-Sen University, Guangzhou 510120, China; (Y.P.); (X.S.)
| | - Huanji Zhang
- Department of Cardiology, The Eighth Affiliated Hospital of Sun Yat-Sen University, Shenzhen 518033, China; (S.X.); (F.W.); (P.M.)
| |
Collapse
|
46
|
Liu C, Zhang Y, Guo J, Sun W, Ji Y, Wang Y, Liu J, Kong X. Overexpression of microRNA-93-5p and microRNA-374a-5p Suppresses the Osteogenic Differentiation and Mineralization of Human Aortic Valvular Interstitial Cells Through the BMP2/Smad1/5/RUNX2 Signaling Pathway. J Cardiovasc Pharmacol 2023; 82:138-147. [PMID: 37232560 DOI: 10.1097/fjc.0000000000001440] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Accepted: 05/14/2023] [Indexed: 05/27/2023]
Abstract
ABSTRACT Aortic valve calcification commonly occurs in patients with chronic kidney disease (CKD). However, the regulatory functions of microRNAs (miRNAs/miRs) in the osteogenic differentiation of human aortic valvular interstitial cells (hAVICs) in patients with CKD remain largely unknown. This study aimed to explore the functional role and underlying mechanisms of miR-93-5p and miR-374a-5p in the osteogenic differentiation of hAVICs. For this purpose, hAVICs calcification was induced with high-calcium/high-phosphate medium and the expression levels of miR-93-5p and miR-374a-5p were determined using bioinformatics assay. Alizarin red staining, intracellular calcium content, and alkaline phosphatase activity were used to evaluate calcification. The expression levels of bone morphogenetic protein-2 (BMP2), runt-related transcription factor 2 (Runx2), and phosphorylated (p)-Smad1/5 were detected by luciferase reporter assay, reverse transcription-quantitative polymerase chain reaction (RT-qPCR), and western blot analysis. The results revealed that the expression levels of miR-93-5p and miR-374a-5p were significantly decreased in hAVICs in response to high-calcium/high-phosphate medium. The overexpression of miR-93-5p and miR-374a-5p effectively suppressed the high-calcium/high-phosphate-induced calcification and osteogenic differentiation makers. Mechanistically, the overexpression of miR-93-5p and miR-374a-5p inhibits osteogenic differentiation by regulating the BMP2/Smad1/5/Runx2 signaling pathway. Taken together, this study indicates that miR-93-5p and miR-374a-5p suppress the osteogenic differentiation of hAVICs associated with calcium-phosphate metabolic dyshomeostasis through the inhibition of the BMP2/Smad1/5/Runx2 signaling pathway.
Collapse
Affiliation(s)
- Cuiying Liu
- Department of Cardiovascular Medicine, Medical School of Southeast University, Nanjing, China
| | - Yajie Zhang
- Central Laboratory, Nanjing Hospital of Traditional Chinese Medicine, Nanjing, China
| | - Jing Guo
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China; and
| | - Wei Sun
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China; and
| | - Yue Ji
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China; and
| | - Yaqing Wang
- Department of Cardiology, Geriatric Hospital of Nanjing Medical University, Nanjing, China
| | - Jing Liu
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China; and
| | - Xiangqing Kong
- Department of Cardiovascular Medicine, Medical School of Southeast University, Nanjing, China
| |
Collapse
|
47
|
Alhuarrat MAD, Alhuarrat MR, Varrias D, Patel SR, Sims DB, Latib A, Jorde UP, Saeed O. Outcomes of Non-ST-Segment Myocardial Infarction During Chronic Heart Failure and End-Stage Renal Disease. Am J Cardiol 2023; 200:1-7. [PMID: 37269688 DOI: 10.1016/j.amjcard.2023.05.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Revised: 04/24/2023] [Accepted: 05/07/2023] [Indexed: 06/05/2023]
Abstract
Non-ST-segment myocardial infarction (NSTEMI) occurs frequently in a growing population of patients with chronic heart failure (HF) and end-stage renal disease (ESRD) but outcomes with invasive management approaches are unknown. We sought to determine in-hospital outcomes with percutaneous coronary intervention (PCI) in comparison with medical management only. The National Inpatient Sample was used to capture hospitalizations in the United States from 2006 to 2019. Admissions for NSTEMI in patients with chronic HF and ESRD were identified by International Classification of Diseases codes. The cohort was divided into those that received PCI or medical management only. In-hospital outcomes were compared by multivariable logistic regression and propensity matching. In 27,433 hospitalizations, 8,004 patients (29%) underwent PCI, and 19,429 (71%) were managed with medications only. PCI was associated with lower adjusted odds of death during hospitalization (adjusted odds ratio 0.59, 95% confidence interval 0.52 to 0.66, p <0.01). This association remained consistent after propensity matching (adjusted odds ratio 0.56, 95% confidence interval 0.49 to 0.64, p <0.01) and was apparent across all subtypes of HF. Patients with PCI had greater duration (5, 3, to 9 vs, 5, 3 to 8 days, p <0.01) and cost of hospitalization ($107,942, 70,230 to $173,182 vs, $44,156, 24,409 to $80,810, p <0.01). In conclusion, patients with HF and ESRD admitted for NSTEMI experienced lower in-hospital mortality with PCI in comparison with medical therapy only. Invasive percutaneous revascularization may be reasonable for appropriately selected patients with HF and ESRD but randomized controlled trials are needed to determine its safety and efficacy in this high-risk population.
Collapse
Affiliation(s)
- Majd Al Deen Alhuarrat
- Division of Internal Medicine, Jacobi Medical Center, Albert Einstein College of Medicine, Bronx, New York
| | | | - Dimitrios Varrias
- Division of Internal Medicine, Jacobi Medical Center, Albert Einstein College of Medicine, Bronx, New York
| | - Snehal R Patel
- Division of Cardiology, Montefiore Medical Center, Albert Einstein College of Medicine, Bronx, New York
| | - Daniel B Sims
- Division of Cardiology, Montefiore Medical Center, Albert Einstein College of Medicine, Bronx, New York
| | - Azeem Latib
- Division of Cardiology, Montefiore Medical Center, Albert Einstein College of Medicine, Bronx, New York
| | - Ulrich P Jorde
- Division of Cardiology, Montefiore Medical Center, Albert Einstein College of Medicine, Bronx, New York
| | - Omar Saeed
- Division of Cardiology, Montefiore Medical Center, Albert Einstein College of Medicine, Bronx, New York.
| |
Collapse
|
48
|
Hung KC, Yao WC, Liu YL, Yang HJ, Liao MT, Chong K, Peng CH, Lu KC. The Potential Influence of Uremic Toxins on the Homeostasis of Bones and Muscles in Chronic Kidney Disease. Biomedicines 2023; 11:2076. [PMID: 37509715 PMCID: PMC10377042 DOI: 10.3390/biomedicines11072076] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2023] [Revised: 07/21/2023] [Accepted: 07/21/2023] [Indexed: 07/30/2023] Open
Abstract
Patients with chronic kidney disease (CKD) often experience a high accumulation of protein-bound uremic toxins (PBUTs), specifically indoxyl sulfate (IS) and p-cresyl sulfate (pCS). In the early stages of CKD, the buildup of PBUTs inhibits bone and muscle function. As CKD progresses, elevated PBUT levels further hinder bone turnover and exacerbate muscle wasting. In the late stage of CKD, hyperparathyroidism worsens PBUT-induced muscle damage but can improve low bone turnover. PBUTs play a significant role in reducing both the quantity and quality of bone by affecting osteoblast and osteoclast lineage. IS, in particular, interferes with osteoblastogenesis by activating aryl hydrocarbon receptor (AhR) signaling, which reduces the expression of Runx2 and impedes osteoblast differentiation. High PBUT levels can also reduce calcitriol production, increase the expression of Wnt antagonists (SOST, DKK1), and decrease klotho expression, all of which contribute to low bone turnover disorders. Furthermore, PBUT accumulation leads to continuous muscle protein breakdown through the excessive production of reactive oxygen species (ROS) and inflammatory cytokines. Interactions between muscles and bones, mediated by various factors released from individual tissues, play a crucial role in the mutual modulation of bone and muscle in CKD. Exercise and nutritional therapy have the potential to yield favorable outcomes. Understanding the underlying mechanisms of bone and muscle loss in CKD can aid in developing new therapies for musculoskeletal diseases, particularly those related to bone loss and muscle wasting.
Collapse
Affiliation(s)
- Kuo-Chin Hung
- Division of Nephrology, Department of Medicine, Min-Sheng General Hospital, Taoyuan City 330, Taiwan
- Department of Pharmacy, Tajen University, Pingtung 907, Taiwan
| | - Wei-Cheng Yao
- Department of Anesthesiology, Min-Sheng General Hospital, Taoyuan City 330, Taiwan
- Department of Medical Education and Clinical Research, Min-Sheng General Hospital, Taoyuan City 330, Taiwan
| | - Yi-Lien Liu
- Department of Family Medicine, Min-Sheng General Hospital, Taoyuan City 330, Taiwan
| | - Hung-Jen Yang
- Department of General Medicine, Min-Sheng General Hospital, Taoyuan City 330, Taiwan
| | - Min-Tser Liao
- Department of Pediatrics, Taoyuan Armed Forces General Hospital Hsinchu Branch, Hsinchu City 300, Taiwan
- Department of Pediatrics, Taoyuan Armed Forces General Hospital, Taoyuan 325, Taiwan
- Department of Pediatrics, Tri-Service General Hospital, National Defense Medical Center, Taipei 114, Taiwan
| | - Keong Chong
- Division of Endocrinology and Metabolism, Department of Medicine, Min-Sheng General Hospital, Taoyuan City 330, Taiwan
| | - Ching-Hsiu Peng
- Division of Nephrology, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, and School of Medicine, Tzu Chi University, Hualien 970, Taiwan
| | - Kuo-Cheng Lu
- Division of Nephrology, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, and School of Medicine, Tzu Chi University, Hualien 970, Taiwan
- Division of Nephrology, Department of Medicine, Fu-Jen Catholic University Hospital, School of Medicine, Fu-Jen Catholic University, New Taipei City 242, Taiwan
- Division of Nephrology, Department of Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei 114, Taiwan
| |
Collapse
|
49
|
Shen A, Shi J, Wang Y, Zhang Q, Chen J. Identification of key biomarkers based on the proliferation of secondary hyperparathyroidism by bioinformatics analysis and machine learning. PeerJ 2023; 11:e15633. [PMID: 37456892 PMCID: PMC10340109 DOI: 10.7717/peerj.15633] [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: 02/13/2023] [Accepted: 06/03/2023] [Indexed: 07/18/2023] Open
Abstract
Objective Secondary hyperparathyroidism (SHPT) is a frequent complication of chronic kidney disease (CKD) associated with morbidity and mortality. This study aims to identify potential biomarkers that may be used to predict the progression of SHPT and to elucidate the molecular mechanisms of SHPT pathogenesis at the transcriptome level. Methods We analyzed differentially expressed genes (DEGs) between diffuse and nodular parathyroid hyperplasia of SHPT patients from the GSE75886 dataset, and then verified DEG levels with the GSE83421 data file of primary hyperparathyroidism (PHPT) patients. Candidate gene sets were selected by machine learning screens of differential genes and immune cell infiltration was explored with the CIBERSORT algorithm. RcisTarget was used to predict transcription factors, and Cytoscape was used to construct a lncRNA-miRNA-mRNA network to identify possible molecular mechanisms. Immunohistochemistry (IHC) staining and quantitative real-time polymerase chain reaction (qRT-PCR) were used to verify the expression of screened genes in parathyroid tissues of SHPT patients and animal models. Results A total of 614 DEGs in GSE75886 were obtained as candidate gene sets for further analysis. Five key genes (USP12, CIDEA, PCOLCE2, CAPZA1, and ACCN2) had significant expression differences between groups and were screened with the best ranking in the machine learning process. These genes were shown to be closely related to immune cell infiltration levels and play important roles in the immune microenvironment. Transcription factor ZBTB6 was identified as the master regulator, alongside multiple other transcription factors. Combined with qPCR and IHC assay of hyperplastic parathyroid tissues from SHPT patients and rats confirm differential expression of USP12, CIDEA, PCOLCE2, CAPZA1, and ACCN2, suggesting that they may play important roles in the proliferation and progression of SHPT. Conclusion USP12, CIDEA, PCOLCE2, CAPZA1, and ACCN2 have great potential both as biomarkers and as therapeutic targets in the proliferation of SHPT. These findings suggest novel potential targets and future directions for SHPT research.
Collapse
Affiliation(s)
- Aiwen Shen
- Nephrology, Huashan Hospital, Fudan University, Shanghai, China
- National Clinical Research Center for Aging and Medicine, Huashan Hospital, Fudan University, Shanghai, China
| | - Jialin Shi
- Nephrology, Huashan Hospital, Fudan University, Shanghai, China
- National Clinical Research Center for Aging and Medicine, Huashan Hospital, Fudan University, Shanghai, China
| | - Yu Wang
- Nephrology, Huashan Hospital, Fudan University, Shanghai, China
- National Clinical Research Center for Aging and Medicine, Huashan Hospital, Fudan University, Shanghai, China
| | - Qian Zhang
- Nephrology, Huashan Hospital, Fudan University, Shanghai, China
- National Clinical Research Center for Aging and Medicine, Huashan Hospital, Fudan University, Shanghai, China
| | - Jing Chen
- Nephrology, Huashan Hospital, Fudan University, Shanghai, China
- National Clinical Research Center for Aging and Medicine, Huashan Hospital, Fudan University, Shanghai, China
| |
Collapse
|
50
|
Zhao D, Wang W, Tang T, Zhang YY, Yu C. Current progress in artificial intelligence-assisted medical image analysis for chronic kidney disease: A literature review. Comput Struct Biotechnol J 2023; 21:3315-3326. [PMID: 37333860 PMCID: PMC10275698 DOI: 10.1016/j.csbj.2023.05.029] [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: 10/28/2022] [Revised: 05/28/2023] [Accepted: 05/28/2023] [Indexed: 06/20/2023] Open
Abstract
Chronic kidney disease (CKD) causes irreversible damage to kidney structure and function. Arising from various etiologies, risk factors for CKD include hypertension and diabetes. With a progressively increasing global prevalence, CKD is an important public health problem worldwide. Medical imaging has become an important diagnostic tool for CKD through the non-invasive identification of macroscopic renal structural abnormalities. Artificial intelligence (AI)-assisted medical imaging techniques aid clinicians in the analysis of characteristics that cannot be easily discriminated by the naked eye, providing valuable information for the identification and management of CKD. Recent studies have demonstrated the effectiveness of AI-assisted medical image analysis as a clinical support tool using radiomics- and deep learning-based AI algorithms for improving the early detection, pathological assessment, and prognostic evaluation of various forms of CKD, including autosomal dominant polycystic kidney disease. Herein, we provide an overview of the potential roles of AI-assisted medical image analysis for the diagnosis and management of CKD.
Collapse
Affiliation(s)
- Dan Zhao
- Department of Nephrology, Tongji Hospital, School of Medicine, Tongji University, Shanghai 200065, China
| | - Wei Wang
- Department of Radiology, Tongji Hospital, School of Medicine, Tongji University, Shanghai 200065, China
| | - Tian Tang
- Department of Nephrology, Tongji Hospital, School of Medicine, Tongji University, Shanghai 200065, China
| | - Ying-Ying Zhang
- Department of Nephrology, Tongji Hospital, School of Medicine, Tongji University, Shanghai 200065, China
| | - Chen Yu
- Department of Nephrology, Tongji Hospital, School of Medicine, Tongji University, Shanghai 200065, China
| |
Collapse
|