1
|
Xu Z, Jiang F, Wu X, Ren B, Zhang C, Lin L, Li S. ACAA2 Protects Against Cardiac Dysfunction and Lipid Peroxidation in Renal Insufficiency with the Treatment of S-Nitroso-L-Cysteine. Biomolecules 2025; 15:364. [PMID: 40149900 PMCID: PMC11940541 DOI: 10.3390/biom15030364] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2025] [Revised: 02/16/2025] [Accepted: 02/22/2025] [Indexed: 03/29/2025] Open
Abstract
The key fatty acid β-oxidation protein acetyl-CoA acyltransferase 2 (ACAA2) plays a significant role in myocardial lipid peroxidation and cardiac dysfunction induced by renal insufficiency. However, the mechanisms of lipid metabolism related to renal insufficiency-associated cardiac dysfunction remain poorly understood, and current clinical treatments have been largely ineffective. Through analysis of the Gene Expression Omnibus (GEO) database, we identified that the cardiac functional changes caused by renal insufficiency were primarily centered around the fatty acid β-oxidation signaling pathway, where ACAA2 plays a pivotal role in fatty acid β-oxidation, the tricarboxylic acid cycle, and ketone body metabolism. In an adenine-induced renal insufficiency mouse model, further examination with hematoxylin-eosin staining, Masson staining, and Oil Red O staining revealed alterations in the heart and kidney as well as the accumulation of lipid. Non-invasive blood pressure measurements and ultrasound images demonstrated improvements of peripheral vascular and right ventricular hemodynamic parameters with S-nitroso-L-cysteine (CSNO) inhalation therapy. In cell experiments, knocking down ACAA2 led to accumulation of lipid droplets and exacerbation of oxidative stress in cardiomyocytes, while overexpression of ACAA2 reversed these effects. The transcription factor FOXO4 was found to regulate lipid peroxidation by modulating ACAA2, and knocking down FOXO4 partially restored the expression of ACAA2, reducing oxidative stress in cardiomyocytes. Furthermore, exogenous CSNO effectively restored the expression of ACAA2 and reduced the level of FOXO4, thereby mitigating lipid peroxidation and improving cardiac function. Therefore, in the context of renal insufficiency, regulating the FOXO4-ACAA2 axis through CSNO inhalation therapy may provide a novel therapeutic strategy for alleviating myocardial lipid peroxidation and improving cardiac function.
Collapse
Affiliation(s)
- Zhengqi Xu
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095# Jiefang Ave., Wuhan 430030, China; (Z.X.); (F.J.); (X.W.); (B.R.); (L.L.)
| | - Feng Jiang
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095# Jiefang Ave., Wuhan 430030, China; (Z.X.); (F.J.); (X.W.); (B.R.); (L.L.)
| | - Xiaofan Wu
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095# Jiefang Ave., Wuhan 430030, China; (Z.X.); (F.J.); (X.W.); (B.R.); (L.L.)
| | - Bowen Ren
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095# Jiefang Ave., Wuhan 430030, China; (Z.X.); (F.J.); (X.W.); (B.R.); (L.L.)
| | - Cuntai Zhang
- Key Laboratory of Vascular Aging, Ministry of Education, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095# Jiefang Ave., Wuhan 430030, China;
| | - Li Lin
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095# Jiefang Ave., Wuhan 430030, China; (Z.X.); (F.J.); (X.W.); (B.R.); (L.L.)
- Key Laboratory of Vascular Aging, Ministry of Education, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095# Jiefang Ave., Wuhan 430030, China;
| | - Sheng Li
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095# Jiefang Ave., Wuhan 430030, China; (Z.X.); (F.J.); (X.W.); (B.R.); (L.L.)
- Key Laboratory of Vascular Aging, Ministry of Education, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095# Jiefang Ave., Wuhan 430030, China;
| |
Collapse
|
2
|
Zhao Z, Zhang C, Li Y, Liu J, Wang L, Wang X, Wang Y, Liu M, Yue X, Wang X, Wang Y, Ji L, Zhao X, Li D. Association between exposure to brominated flame retardants and atherosclerosis: Evidence for inflammatory status as a potential mediator. THE SCIENCE OF THE TOTAL ENVIRONMENT 2025; 967:178822. [PMID: 39952214 DOI: 10.1016/j.scitotenv.2025.178822] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/21/2024] [Revised: 12/29/2024] [Accepted: 02/09/2025] [Indexed: 02/17/2025]
Abstract
Brominated flame retardants (BFRs) are a widely used category of environmentally persistent, bioaccumulative chemicals. However, research focusing on the potential adverse effects of these chemicals on human health, particularly on cardiovascular issues, remains limited. This study aimed to investigate the association between BFR exposure and the development of atherosclerosis with a particular focus on the potential role of inflammatory indicators as mediators. Six typical BFRs (BB-153, BDE-28, BDE-47, BDE-99, BDE-100, and BDE-153) and six inflammatory indicators (white blood cells [WBC], neutrophils, mononuclear cells, lymphocytes, blood platelet [PLT], and C-reactive protein) were examined using data from 1654 participants in the National Health and Nutrition Examination Survey 2003-2004. Statistical analysis revealed that the levels of serum BFR, including BB-153, BDE-28, BDE-47, BDE-99, and BDE-100, were markedly elevated in the atherosclerosis group compared with those in the non-atherosclerosis group (all P <0.05). Restricted cubic splines demonstrated a nonlinear relationship between BB-153 and BDE-100 and atherosclerosis, with BB-153 exhibiting a significant correlation with atherosclerosis in an inverted U-shape (P = 0.001). In the Binary logistic regression model, BB-153, BDE-28, BDE-47, and BDE-100 were significantly correlated with atherosclerosis, with BB-153 exhibiting the strongest association [OR = 2.059, 95 % CI: (1.540-2.754), P = 0.001]. BFRs were re-analyzed after being divided into four quartiles, revealing a dose-response relationship in which the risk of atherosclerosis increased with higher serum BFRs levels. Bayesian kernel machine regression model and quantile-based G-computation model analyses also demonstrated consistent correlations with mixed BFR exposures, with BB-153 having the greatest contribution. Moreover, mediation analysis demonstrated that WBC count and PLT levels were the primary mediators (proportion mediated: 5.10 % and 4.20 %, respectively) in the link between of BDE-153, BDE-100, and atherosclerosis. Thus, the inflammatory status may serve as a mediating factor in the relationship between BFR exposure and atherosclerosis.
Collapse
Affiliation(s)
- Zihui Zhao
- School of Public Health, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan 250117, China
| | - Chi Zhang
- NHC Key Laboratory of Food Safety Risk Assessment, Chinese Academy of Medical Science Research Unit (No. 2019RU014), China National Center for Food Safety Risk Assessment, Beijing 100021, China
| | - Yuanyuan Li
- Binzhou Center for Disease Control and Prevention, Binzhou Institute of Preventive Medicine, Binzhou 256602, China
| | - Junli Liu
- Binzhou Center for Disease Control and Prevention, Binzhou Institute of Preventive Medicine, Binzhou 256602, China
| | - Liangao Wang
- School of Public Health, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan 250117, China
| | - Xianhao Wang
- School of Public Health, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan 250117, China
| | - Yiqian Wang
- School of Public Health, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan 250117, China
| | - Meng Liu
- School of Public Health, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan 250117, China
| | - Xianfeng Yue
- School of Public Health, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan 250117, China
| | - Xiaoyan Wang
- School of Basic Medical Sciences & Institute of Basic Medical Research, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan 250117, China
| | - Yuting Wang
- School of Basic Medical Sciences & Institute of Basic Medical Research, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan 250117, China
| | - Long Ji
- School of Public Health, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan 250117, China; School of Sports Medicine and Rehabilitation, Shandong First Medical University & Shandong Academy of Medical Sciences, Taian 271016, China.
| | - Xuezhen Zhao
- School of Public Health, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan 250117, China
| | - Dong Li
- School of Public Health, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan 250117, China; School of Public Health, Jining Medical University, Jining 272067, China
| |
Collapse
|
3
|
Gao Y, Chen S, Fu J, Wang C, Tang Y, Luo Y, Zhuo X, Chen X, Shen Y. Factors associated with risk analysis for asymptomatic left ventricular diastolic dysfunction in nondialysis patients with chronic kidney disease. Ren Fail 2024; 46:2353334. [PMID: 38785296 PMCID: PMC11133225 DOI: 10.1080/0886022x.2024.2353334] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2023] [Accepted: 05/06/2024] [Indexed: 05/25/2024] Open
Abstract
Heart failure (HF) constitutes a major determinant of outcome in chronic kidney disease (CKD) patients. The main pattern of HF in CKD patients is preserved ejection fraction (HFpEF), and left ventricular diastolic dysfunction (LVDD) is a frequent pathophysiological mechanism and specific preclinical manifestation of HFpEF. Therefore, exploring and intervention of the factors associated with risk for LVDD is of great importance in reducing the morbidity and mortality of cardiovascular disease (CVD) complications in CKD patients. We designed this retrospective cross-sectional study to collect clinical and echocardiographic data from 339 nondialysis CKD patients without obvious symptoms of HF to analyze the proportion of asymptomatic left ventricular diastolic dysfunction (ALVDD) and its related factors associated with risk by multivariate logistic regression analysis. Among the 339 nondialysis CKD patients, 92.04% had ALVDD. With the progression of CKD stage, the proportion of ALVDD gradually increased. The multivariate logistic regression analysis revealed that increased age (OR 1.237; 95% confidence interval (CI) 1.108-1.381, per year), diabetic nephropathy (DN) and hypertensive nephropathy (HTN) (OR 25.000; 95% CI 1.355-48.645, DN and HTN vs chronic interstitial nephritis), progression of CKD stage (OR 2.785; 95% CI 1.228-6.315, per stage), increased mean arterial pressure (OR 1.154; 95% CI 1.051-1.268, per mmHg), increased urinary protein (OR 2.825; 95% CI 1.484-5.405, per g/24 h), and low blood calcium (OR 0.072; 95% CI 0.006-0.859, per mmol/L) were factors associated with risk for ALVDD in nondialysis CKD patients after adjusting for other confounding factors. Therefore, dynamic monitoring of these factors associated with risk, timely diagnosis and treatment of ALVDD can delay the progression to symptomatic HF, which is of great importance for reducing CVD mortality, and improving the prognosis and quality of life in CKD patients.
Collapse
Affiliation(s)
- Yajuan Gao
- Department of Nephrology, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Shengnan Chen
- Department of Nephrology, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Jiani Fu
- Department of Nephrology, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Cui Wang
- Department of Nephrology, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Yali Tang
- Department of Nephrology, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Yongbai Luo
- Department of Cardiovascular Medicine, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Xiaozhen Zhuo
- Department of Cardiovascular Medicine, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Xueying Chen
- Department of Nephrology, Shan Yang County People’s Hospital, Shangluo City, China
| | - Yan Shen
- Department of Nephrology, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| |
Collapse
|
4
|
Liu W, Weng S, Cao C, Yi Y, Wu Y, Peng D. Association between monocyte-lymphocyte ratio and all-cause and cardiovascular mortality in patients with chronic kidney diseases: A data analysis from national health and nutrition examination survey (NHANES) 2003-2010. Ren Fail 2024; 46:2352126. [PMID: 38832474 DOI: 10.1080/0886022x.2024.2352126] [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: 01/05/2024] [Accepted: 05/01/2024] [Indexed: 06/05/2024] Open
Abstract
BACKGROUND The relationship between monocyte-to-lymphocyte ratio (MLR) and prognosis in patients with chronic kidney disease (CKD) remains unclear. The aim of this study was to investigate the association between MLR and both all-cause mortality and cardiovascular disease (CVD) mortality in patients with CKD. METHODS This study analyzed data from National Health and Nutrition Examination Survey 2003-2010. This study included 11262 eligible subjects, and 3015 of them were with CKD. We first compared the differences in clinical characteristics between individuals with and without CKD, and then grouped the CKD population based on quartiles of MLR. The partial correlation analysis was conducted to assess the relationships between MLR and some important clinical features. Cox proportional hazards models were used to investigate the associations between MLR and mortality from all-cause and cardiovascular disease. Restricted cubic spline (RCS) was used to investigate the dose-response relationship between MLR and mortality, the receiver operating characteristic (ROC) curves is used to compare the efficacy of MLR with different clinical biological indicators in assessing the risk of death. RESULTS During a median follow-up of 10.3 years in CKD population, 1398 (43%) all-cause deaths and 526 (16%) CVD deaths occurred. It has been found that individuals with CKD have higher MLR level. The partial correlation analysis results showed that even after adjusting for age, sex, and race, MLR is still correlated with blood glucose, lipid levels, and kidney function indicators. The results of the cox proportional hazards regression model and Kaplan-Meier curve shown after adjusting for covariates, higher MLR was significantly associated with an increased risk of mortality. Consistent results were also observed when MLR was examined as categorical variable (quartiles). The RCS demonstrated a positive association between MLR and the risk of all-cause mortality and cardiovascular mortality. The ROC results indicate that the predictive efficacy of MLR for all-cause mortality risk is comparable to eGFR, higher than NLR and CRP. The predictive efficacy of MLR for cardiovascular mortality risk is higher than these three indicators. CONCLUSION Compared to non-CKD population, the CKD population has higher levels of MLR. In the CKD population, MLR is positively correlated with the risk of death. Furthermore, the predictive efficacy of MLR for mortality risk is higher than other clinical indicators. This suggests that MLR can serve as a simple and effective clinical indicator for predicting mortality risk in CKD patients.
Collapse
Affiliation(s)
- Wenwu Liu
- Department of Cardiovascular Medicine, Research Institute of Blood Lipids and Atherosclerosis, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Shuwei Weng
- Department of Cardiovascular Medicine, Research Institute of Blood Lipids and Atherosclerosis, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Chenghui Cao
- Department of Cardiovascular Medicine, Research Institute of Blood Lipids and Atherosclerosis, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Yuting Yi
- Department of Cardiovascular Medicine, Research Institute of Blood Lipids and Atherosclerosis, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Yue Wu
- Department of Cardiovascular Medicine, Research Institute of Blood Lipids and Atherosclerosis, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Daoquan Peng
- Department of Cardiovascular Medicine, Research Institute of Blood Lipids and Atherosclerosis, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| |
Collapse
|
5
|
Duni A, Kitsos A, Bechlioulis A, Lakkas L, Markopoulos G, Tatsis V, Koutlas V, Tzalavra E, Baxevanos G, Vartholomatos G, Mitsis M, Naka KK, Dounousi E. Identification of Novel Independent Correlations between Cellular Components of the Immune System and Strain-Related Indices of Myocardial Dysfunction in CKD Patients and Kidney Transplant Recipients without Established Cardiovascular Disease. Int J Mol Sci 2024; 25:9162. [PMID: 39273110 PMCID: PMC11395156 DOI: 10.3390/ijms25179162] [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: 06/25/2024] [Revised: 08/15/2024] [Accepted: 08/21/2024] [Indexed: 09/15/2024] Open
Abstract
The role of immune system components in the development of myocardial remodeling in chronic kidney disease (CKD) and kidney transplantation remains an open question. Our aim was to investigate the associations between immune cell subpopulations in the circulation of CKD patients and kidney transplant recipients (KTRs) with subclinical indices of myocardial performance. We enrolled 44 CKD patients and 38 KTRs without established cardiovascular disease. A selected panel of immune cells was measured by flow cytometry. Classical and novel strain-related indices of ventricular function were measured by speckle-tracking echocardiography at baseline and following dipyridamole infusion. In CKD patients, the left ventricular (LV) relative wall thickness correlated with the CD14++CD16- monocytes (β = 0.447, p = 0.004), while the CD14++CD16+ monocytes were independent correlates of the global radial strain (β = 0.351, p = 0.04). In KTRs, dipyridamole induced changes in global longitudinal strain correlated with CD14++CD16+ monocytes (β = 0.423, p = 0.009) and CD4+ T-cells (β = 0.403, p = 0.01). LV twist and untwist were independently correlated with the CD8+ T-cells (β = 0.405, p = 0.02 and β = -0.367, p = 0.03, respectively) in CKD patients, whereas the CD14++CD16+ monocytes were independent correlates of LV twist and untwist in KTRs (β = 0.405, p = 0.02 and β = -0.367, p = 0.03, respectively). Immune cell subsets independently correlate with left ventricular strain and torsion-related indices in CKD patients and KTRs without established CVD.
Collapse
Affiliation(s)
- Anila Duni
- Department of Nephrology, Faculty of Medicine, School of Health Sciences, University Hospital of Ioannina, University of Ioannina, 455 00 Ioannina, Greece
- Kidney Transplant Unit, Department of Surgery, Faculty of Medicine, School of Health Sciences, University Hospital of Ioannina, University of Ioannina, 455 00 Ioannina, Greece
| | - Athanasios Kitsos
- Department of Nephrology, Faculty of Medicine, School of Health Sciences, University Hospital of Ioannina, University of Ioannina, 455 00 Ioannina, Greece
- Kidney Transplant Unit, Department of Surgery, Faculty of Medicine, School of Health Sciences, University Hospital of Ioannina, University of Ioannina, 455 00 Ioannina, Greece
| | - Aris Bechlioulis
- Second Department of Cardiology, Faculty of Medicine, School of Health Sciences, University Hospital of Ioannina, University of Ioannina, 455 00 Ioannina, Greece
| | - Lampros Lakkas
- Second Department of Cardiology, Faculty of Medicine, School of Health Sciences, University Hospital of Ioannina, University of Ioannina, 455 00 Ioannina, Greece
| | - Georgios Markopoulos
- Laboratory of Haematology-Unit of Molecular Biology and Translational Flow Cytometry, University Hospital of Ioannina, 455 00 Ioannina, Greece
| | - Vasileios Tatsis
- Kidney Transplant Unit, Department of Surgery, Faculty of Medicine, School of Health Sciences, University Hospital of Ioannina, University of Ioannina, 455 00 Ioannina, Greece
| | - Vasileios Koutlas
- Kidney Transplant Unit, Department of Surgery, Faculty of Medicine, School of Health Sciences, University Hospital of Ioannina, University of Ioannina, 455 00 Ioannina, Greece
| | - Eirini Tzalavra
- Kidney Transplant Unit, Department of Surgery, Faculty of Medicine, School of Health Sciences, University Hospital of Ioannina, University of Ioannina, 455 00 Ioannina, Greece
| | - Gerasimos Baxevanos
- Laboratory of Haematology-Unit of Molecular Biology and Translational Flow Cytometry, University Hospital of Ioannina, 455 00 Ioannina, Greece
- Department of Internal Medicine, General Hospital of Ioannina, G. Chatzikosta, 454 45 Ioannina, Greece
| | - Georgios Vartholomatos
- Laboratory of Haematology-Unit of Molecular Biology and Translational Flow Cytometry, University Hospital of Ioannina, 455 00 Ioannina, Greece
| | - Michail Mitsis
- Kidney Transplant Unit, Department of Surgery, Faculty of Medicine, School of Health Sciences, University Hospital of Ioannina, University of Ioannina, 455 00 Ioannina, Greece
| | - Katerina K Naka
- Second Department of Cardiology, Faculty of Medicine, School of Health Sciences, University Hospital of Ioannina, University of Ioannina, 455 00 Ioannina, Greece
| | - Evangelia Dounousi
- Department of Nephrology, Faculty of Medicine, School of Health Sciences, University Hospital of Ioannina, University of Ioannina, 455 00 Ioannina, Greece
- Kidney Transplant Unit, Department of Surgery, Faculty of Medicine, School of Health Sciences, University Hospital of Ioannina, University of Ioannina, 455 00 Ioannina, Greece
| |
Collapse
|
6
|
Kane J, Vos WG, Bosmans LA, van Os BW, den Toom M, Hoeksema‐Hackmann S, Moen‐de Wit D, Gijbels MJ, Beckers L, Grefhorst A, Levels JHM, Jakulj L, Vervloet MG, Lutgens E, Eringa EC. Peritoneal Dialysis Aggravates and Accelerates Atherosclerosis in Uremic ApoE-/- Mice. J Am Heart Assoc 2024; 13:e034066. [PMID: 38979792 PMCID: PMC11292770 DOI: 10.1161/jaha.123.034066] [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: 02/28/2024] [Accepted: 04/29/2024] [Indexed: 07/10/2024]
Abstract
BACKGROUND Atherosclerosis is highly prevalent in people with chronic kidney disease (CKD), including those receiving peritoneal dialysis (PD). Although it is lifesaving, PD induces profound systemic inflammation, which may aggravate atherosclerosis. Therefore, the hypothesis is that this PD-induced inflammation aggravates atherosclerosis via immune cell activation. METHODS AND RESULTS ApoE-/- mice were subjected to a 5/6 nephrectomy to induce CKD. Three weeks later, mice were fed a high-cholesterol diet. Half of the nephrectomized mice then received daily peritoneal infusions of 3.86% Physioneal for 67 further days (CKD+PD) until the end of the experiment, and were compared with mice without CKD. Sham operated and PD-only mice were additional controls. CKD+PD mice displayed more severe atherosclerotic disease than control mice. Plaque area increased, and plaques were more advanced with a vulnerable phenotype typified by decreased collagen content and decreased fibrous cap thickness. Increased CD3+ T-cell numbers were present in plaques and perivascular adipose tissue of CKD and CKD+PD mice. Plaques of CKD+PD mice contained more iNOS+ immune cells. Spleens of CKD+PD mice showed more CD4+ central memory, terminally differentiated type 1 T-helper (Th1), Th17, and CX3C motif chemokine receptor 1+ (CX3CR1) CD4+ T-cells with less regulatory and effector T-cells. CONCLUSIONS PD-fluid exposure in uremic mice potentiates systemic and vascular T-cell-driven inflammation and aggravates atherosclerosis. PD polarized CD4+ T-cells toward an inflammatory Th1/Th17 phenotype, and increased CX3CR1+ CD4+ T-cells, which are associated with vascular homing in CKD-associated atherosclerosis. Targeting CD4+ T-cell activation and CX3CR1+ polarization has the potential to attenuate atherosclerosis in PD patients.
Collapse
Affiliation(s)
- Jamie Kane
- Department of Nephrology, Amsterdam Cardiovascular SciencesAmsterdam University Medical CentreAmsterdamthe Netherlands
- Department of Physiology, Amsterdam Cardiovascular SciencesAmsterdam University Medical CentreAmsterdamthe Netherlands
- Department of Medical Biochemistry, Amsterdam Cardiovascular Sciences, Amsterdam Immunity and InfectionAmsterdam University Medical CentreAmsterdamthe Netherlands
| | - Winnie G. Vos
- Department of Medical Biochemistry, Amsterdam Cardiovascular Sciences, Amsterdam Immunity and InfectionAmsterdam University Medical CentreAmsterdamthe Netherlands
| | - Laura A. Bosmans
- Department of Medical Biochemistry, Amsterdam Cardiovascular Sciences, Amsterdam Immunity and InfectionAmsterdam University Medical CentreAmsterdamthe Netherlands
- Department of Experimental Vascular Medicine, Amsterdam Cardiovascular SciencesAmsterdam University Medical CentreAmsterdamthe Netherlands
| | - Bram W. van Os
- Department of Medical Biochemistry, Amsterdam Cardiovascular Sciences, Amsterdam Immunity and InfectionAmsterdam University Medical CentreAmsterdamthe Netherlands
| | - Myrthe den Toom
- Department of Medical Biochemistry, Amsterdam Cardiovascular Sciences, Amsterdam Immunity and InfectionAmsterdam University Medical CentreAmsterdamthe Netherlands
| | | | - Denise Moen‐de Wit
- Animal Research Institute AMCAmsterdam University Medical CentreAmsterdamthe Netherlands
| | - Marion J. Gijbels
- Department of Medical Biochemistry, Amsterdam Cardiovascular Sciences, Amsterdam Immunity and InfectionAmsterdam University Medical CentreAmsterdamthe Netherlands
- Department of Pathology, Cardiovascular Research Institute Maastricht (CARIM)Maastricht University Medical CentreMaastrichtthe Netherlands
| | - Linda Beckers
- Department of Medical Biochemistry, Amsterdam Cardiovascular Sciences, Amsterdam Immunity and InfectionAmsterdam University Medical CentreAmsterdamthe Netherlands
| | - Aldo Grefhorst
- Department of Experimental Vascular Medicine, Amsterdam Cardiovascular SciencesAmsterdam University Medical CentreAmsterdamthe Netherlands
| | - Johannes H. M. Levels
- Department of Experimental Vascular Medicine, Amsterdam Cardiovascular SciencesAmsterdam University Medical CentreAmsterdamthe Netherlands
| | - Lily Jakulj
- Department of Nephrology, Amsterdam Cardiovascular SciencesAmsterdam University Medical CentreAmsterdamthe Netherlands
- Dianet Dialysis Centre AmsterdamAmsterdamthe Netherlands
| | - Marc G. Vervloet
- Department of Nephrology, Amsterdam Cardiovascular SciencesAmsterdam University Medical CentreAmsterdamthe Netherlands
- Department of NephrologyRadboud University Medical CentreNijmegenthe Netherlands
| | - Esther Lutgens
- Department of Cardiovascular Medicine and ImmunologyMayo ClinicRochesterMN
| | - Etto C. Eringa
- Department of Physiology, Amsterdam Cardiovascular SciencesAmsterdam University Medical CentreAmsterdamthe Netherlands
- Department of PhysiologyMaastricht UniversityMaastrichtthe Netherlands
| |
Collapse
|
7
|
Panisset V, Girerd N, Bozec E, Lamiral Z, d'Hervé Q, Frimat L, Huttin O, Girerd S. Long-term changes in cardiac remodelling in prevalent kidney graft recipients. Int J Cardiol 2024; 403:131852. [PMID: 38360102 DOI: 10.1016/j.ijcard.2024.131852] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Revised: 01/26/2024] [Accepted: 02/10/2024] [Indexed: 02/17/2024]
Abstract
BACKGROUND Approximately 15% of kidney transplant (KT) recipients develop de novo heart failure after KT. There are scarce data reporting the long-term changes in cardiac structure and function among KT recipients. Despite the improvement in renal function, transplant-related complications as well as immunosuppressive therapy could have an impact on cardiac remodelling during follow-up. We aimed to describe the long-term changes in echocardiographic parameters in prevalent KT recipients and identify the clinical and laboratory factors associated with these changes. METHODS A centralised blinded review of two echocardiographic examinations after KT (on average after 17 and 39 months post-KT respectively) was performed among 80 patients (age 50.4 ± 16.2, diabetes 13.8% pre-KT), followed by linear regression to identify clinico-biological factors related to echocardiographic changes. RESULTS Left atrial volume index (LAVI) increased significantly (34.2 ± 10.8 mL/m2vs. 37.6 ± 15.0 mL/m2, annualised delta 3.1 ± 11.4 mL/m2/year; p = 0.034) while left ventricular ejection fraction (LVEF) decreased (62.1 ± 9.0% vs. 59.7 ± 9.9%, annualised delta -2.7 ± 13.6%/year; p = 0.04). Male sex (β = 8.112 ± 2.747; p < 0.01), pre-KT hypertension (β = 9.725 ± 4.156; p < 0.05), graft from expanded criteria donor (β = 3.791 ± 3.587; p < 0.05), and induction by anti-thymocyte globulin (β = 7.920 ± 2.974; p = 0.01) were associated with an increase in LAVI during follow-up. Higher haemoglobin (>12.9 g/dL) at the time of the first echocardiography (β = 6.029 ± 2.967; p < 0.05) and ACEi/ARB therapy (β = 8.306 ± 3.161; p < 0.05) were associated with an increase in LVEF during follow-up. CONCLUSION This study confirms the existence of long-term cardiac remodelling after KT despite dialysis cessation, characterised by an increase in LAVI and a decrease in LVEF. A better management of anaemia and using ACEi/ARB therapy may prevent such remodelling.
Collapse
Affiliation(s)
- Valentin Panisset
- Nephrology Department, University Hospital of Nancy, Vandoeuvre-lès-Nancy, France
| | - Nicolas Girerd
- Université de Lorraine, Inserm, Centre d'Investigations Cliniques-1433, and Inserm U1116; CHRU Nancy; F-CRIN INI-CRCT, Vandoeuvre-lès-Nancy, France
| | - Erwan Bozec
- Université de Lorraine, Inserm, Centre d'Investigations Cliniques-1433, and Inserm U1116; CHRU Nancy; F-CRIN INI-CRCT, Vandoeuvre-lès-Nancy, France
| | - Zohra Lamiral
- Université de Lorraine, Inserm, Centre d'Investigations Cliniques-1433, and Inserm U1116; CHRU Nancy; F-CRIN INI-CRCT, Vandoeuvre-lès-Nancy, France
| | - Quentin d'Hervé
- Nephrology Department, University Hospital of Nancy, Vandoeuvre-lès-Nancy, France
| | - Luc Frimat
- Nephrology Department, University Hospital of Nancy, Vandoeuvre-lès-Nancy, France
| | - Olivier Huttin
- Cardiology Department, University Hospital of Nancy, Vandoeuvre-lès- Nancy, France
| | - Sophie Girerd
- Nephrology Department, University Hospital of Nancy, Vandoeuvre-lès-Nancy, France; Université de Lorraine, Inserm, Centre d'Investigations Cliniques-1433, and Inserm U1116; CHRU Nancy; F-CRIN INI-CRCT, Vandoeuvre-lès-Nancy, France.
| |
Collapse
|
8
|
Behrens F, Bartolomaeus H, Wilck N, Holle J. Gut-immune axis and cardiovascular risk in chronic kidney disease. Clin Kidney J 2024; 17:sfad303. [PMID: 38229879 PMCID: PMC10790347 DOI: 10.1093/ckj/sfad303] [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: 08/02/2023] [Indexed: 01/18/2024] Open
Abstract
Patients with chronic kidney disease (CKD) suffer from marked cardiovascular morbidity and mortality, so lowering the cardiovascular risk is paramount to improve quality of life and survival in CKD. Manifold mechanisms are hold accountable for the development of cardiovascular disease (CVD), and recently inflammation arose as novel risk factor significantly contributing to progression of CVD. While the gut microbiome was identified as key regulator of immunity and inflammation in several disease, CKD-related microbiome-immune interaction gains increasing importance. Here, we summarize the latest knowledge on microbiome dysbiosis in CKD, subsequent changes in bacterial and host metabolism and how this drives inflammation and CVD in CKD. Moreover, we outline potential therapeutic targets along the gut-immune-cardiovascular axis that could aid the combat of CVD development and high mortality in CKD.
Collapse
Affiliation(s)
- Felix Behrens
- Department of Pediatric Gastroenterology, Nephrology and Metabolic Diseases, Charité – Universitätsmedizin Berlin, Berlin, Germany
- Experimental and Clinical Research Center (ECRC), a cooperation of Charité – Universitätsmedizin Berlin and Max Delbrück Center for Molecular Medicine (MDC), Berlin, Germany
- German Centre for Cardiovascular Research (DZHK), Partner Site Berlin, Berlin, Germany
- Institute of Physiology, Charité – Universitätsmedizin Berlin, Berlin, Germany
| | - Hendrik Bartolomaeus
- Experimental and Clinical Research Center (ECRC), a cooperation of Charité – Universitätsmedizin Berlin and Max Delbrück Center for Molecular Medicine (MDC), Berlin, Germany
- German Centre for Cardiovascular Research (DZHK), Partner Site Berlin, Berlin, Germany
- Max Delbrück Center for Molecular Medicine (MDC), Berlin, Germany
- Department of Nephrology und Intensive Medical Care, Charité – Universitätsmedizin Berlin, Berlin, Germany
| | - Nicola Wilck
- Experimental and Clinical Research Center (ECRC), a cooperation of Charité – Universitätsmedizin Berlin and Max Delbrück Center for Molecular Medicine (MDC), Berlin, Germany
- German Centre for Cardiovascular Research (DZHK), Partner Site Berlin, Berlin, Germany
- Max Delbrück Center for Molecular Medicine (MDC), Berlin, Germany
- Department of Nephrology und Intensive Medical Care, Charité – Universitätsmedizin Berlin, Berlin, Germany
| | - Johannes Holle
- Department of Pediatric Gastroenterology, Nephrology and Metabolic Diseases, Charité – Universitätsmedizin Berlin, Berlin, Germany
- Experimental and Clinical Research Center (ECRC), a cooperation of Charité – Universitätsmedizin Berlin and Max Delbrück Center for Molecular Medicine (MDC), Berlin, Germany
- German Centre for Cardiovascular Research (DZHK), Partner Site Berlin, Berlin, Germany
- Max Delbrück Center for Molecular Medicine (MDC), Berlin, Germany
| |
Collapse
|
9
|
Sárközy M, Watzinger S, Kovács ZZ, Acar E, Márványkövi F, Szűcs G, Lauber GY, Galla Z, Siska A, Földesi I, Fintha A, Kriston A, Kovács F, Horváth P, Kővári B, Cserni G, Krenács T, Szabó PL, Szabó GT, Monostori P, Zins K, Abraham D, Csont T, Pokreisz P, Podesser BK, Kiss A. Neuregulin-1β Improves Uremic Cardiomyopathy and Renal Dysfunction in Rats. JACC Basic Transl Sci 2023; 8:1160-1176. [PMID: 37791301 PMCID: PMC10543921 DOI: 10.1016/j.jacbts.2023.03.003] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/24/2023] [Revised: 03/01/2023] [Accepted: 03/01/2023] [Indexed: 10/05/2023]
Abstract
Chronic kidney disease is a global health problem affecting 10% to 12% of the population. Uremic cardiomyopathy is often characterized by left ventricular hypertrophy, fibrosis, and diastolic dysfunction. Dysregulation of neuregulin-1β signaling in the heart is a known contributor to heart failure. The systemically administered recombinant human neuregulin-1β for 10 days in our 5/6 nephrectomy-induced model of chronic kidney disease alleviated the progression of uremic cardiomyopathy and kidney dysfunction in type 4 cardiorenal syndrome. The currently presented positive preclinical data warrant clinical studies to confirm the beneficial effects of recombinant human neuregulin-1β in patients with chronic kidney disease.
Collapse
Affiliation(s)
- Márta Sárközy
- MEDICS Research Group, Department of Biochemistry, Albert Szent-Györgyi Medical School, University of Szeged, Szeged, Hungary
- Interdisciplinary Center of Excellence, University of Szeged, Szeged, Hungary
| | - Simon Watzinger
- Ludwig Boltzmann Institute for Cardiovascular Research at Center for Biomedical Research and Translational Surgery, Medical University of Vienna, Vienna, Austria
| | - Zsuzsanna Z.A. Kovács
- MEDICS Research Group, Department of Biochemistry, Albert Szent-Györgyi Medical School, University of Szeged, Szeged, Hungary
- Interdisciplinary Center of Excellence, University of Szeged, Szeged, Hungary
| | - Eylem Acar
- Ludwig Boltzmann Institute for Cardiovascular Research at Center for Biomedical Research and Translational Surgery, Medical University of Vienna, Vienna, Austria
| | - Fanni Márványkövi
- MEDICS Research Group, Department of Biochemistry, Albert Szent-Györgyi Medical School, University of Szeged, Szeged, Hungary
- Interdisciplinary Center of Excellence, University of Szeged, Szeged, Hungary
| | - Gergő Szűcs
- MEDICS Research Group, Department of Biochemistry, Albert Szent-Györgyi Medical School, University of Szeged, Szeged, Hungary
- Interdisciplinary Center of Excellence, University of Szeged, Szeged, Hungary
| | - Gülsüm Yilmaz Lauber
- Ludwig Boltzmann Institute for Cardiovascular Research at Center for Biomedical Research and Translational Surgery, Medical University of Vienna, Vienna, Austria
| | - Zsolt Galla
- Metabolic and Newborn Screening Laboratory, Department of Pediatrics, Albert Szent-Györgyi Medical School, University of Szeged, Hungary
| | - Andrea Siska
- Department of Laboratory Medicine, Albert Szent-Györgyi Medical School, University of Szeged, Szeged, Hungary
| | - Imre Földesi
- Department of Laboratory Medicine, Albert Szent-Györgyi Medical School, University of Szeged, Szeged, Hungary
| | - Attila Fintha
- Department of Pathology and Experimental Cancer Research, Semmelweis University, Budapest, Hungary
| | - András Kriston
- Synthetic and Systems Biology Unit, Biological Research Centre, Eötvös Loránd Research Network, Szeged, Hungary
- Single-Cell Technologies Ltd, Szeged, Hungary
- Institute for Molecular Medicine Finland (FIMM), University of Helsinki, Helsinki, Finland
| | - Ferenc Kovács
- Synthetic and Systems Biology Unit, Biological Research Centre, Eötvös Loránd Research Network, Szeged, Hungary
- Single-Cell Technologies Ltd, Szeged, Hungary
- Institute for Molecular Medicine Finland (FIMM), University of Helsinki, Helsinki, Finland
| | - Péter Horváth
- Synthetic and Systems Biology Unit, Biological Research Centre, Eötvös Loránd Research Network, Szeged, Hungary
- Single-Cell Technologies Ltd, Szeged, Hungary
- Institute for Molecular Medicine Finland (FIMM), University of Helsinki, Helsinki, Finland
| | - Bence Kővári
- Department of Pathology, Albert Szent-Györgyi Medical School, University of Szeged, Szeged, Hungary
| | - Gábor Cserni
- Department of Pathology, Albert Szent-Györgyi Medical School, University of Szeged, Szeged, Hungary
| | - Tibor Krenács
- Department of Pathology and Experimental Cancer Research, Semmelweis University, Budapest, Hungary
| | - Petra Lujza Szabó
- Ludwig Boltzmann Institute for Cardiovascular Research at Center for Biomedical Research and Translational Surgery, Medical University of Vienna, Vienna, Austria
| | - Gábor Tamás Szabó
- Ludwig Boltzmann Institute for Cardiovascular Research at Center for Biomedical Research and Translational Surgery, Medical University of Vienna, Vienna, Austria
| | - Péter Monostori
- Metabolic and Newborn Screening Laboratory, Department of Pediatrics, Albert Szent-Györgyi Medical School, University of Szeged, Hungary
| | - Karin Zins
- Center for Anatomy and Cell Biology, Medical University of Vienna, Vienna, Austria
| | - Dietmar Abraham
- Center for Anatomy and Cell Biology, Medical University of Vienna, Vienna, Austria
| | - Tamás Csont
- MEDICS Research Group, Department of Biochemistry, Albert Szent-Györgyi Medical School, University of Szeged, Szeged, Hungary
- Interdisciplinary Center of Excellence, University of Szeged, Szeged, Hungary
| | - Peter Pokreisz
- Ludwig Boltzmann Institute for Cardiovascular Research at Center for Biomedical Research and Translational Surgery, Medical University of Vienna, Vienna, Austria
| | - Bruno K. Podesser
- Ludwig Boltzmann Institute for Cardiovascular Research at Center for Biomedical Research and Translational Surgery, Medical University of Vienna, Vienna, Austria
| | - Attila Kiss
- Ludwig Boltzmann Institute for Cardiovascular Research at Center for Biomedical Research and Translational Surgery, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
10
|
Lu C, Wu L, Tang MY, Liu YF, Liu L, Liu XY, Zhang C, Huang L. Indoxyl sulfate in atherosclerosis. Toxicol Lett 2023:S0378-4274(23)00215-1. [PMID: 37414304 DOI: 10.1016/j.toxlet.2023.07.001] [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: 07/18/2022] [Revised: 06/19/2023] [Accepted: 07/03/2023] [Indexed: 07/08/2023]
Abstract
Atherosclerosis (AS), a chronic vascular inflammatory disease, has become a main focus of attention worldwide for its chronic progressing disease course and serious complications in the later period. Nevertheless, explanations for the exact molecular mechanisms of AS initiation and development remain to be an unsolved problem. The classic pathogenesis theories, such as lipid percolation and deposition, endothelium injury, inflammation and immune damage, provide the foundation for discovering the new key molecules or signaling mechanisms. Recently, indoxyl sulfate (IS), one of non-free uremia toxins, has been noticeable for its multiple atherogenic effects. IS exists at high concentration in plasma for its great albumin binding rate. Patients with uremia have markedly elevated serum levels of IS due both to the deterioration of renal function and to the high binding affinity of IS to albumin. Nowadays, elevated incidence of circulatory disease among patients with renal dysfunction indicates correlation of uremic toxins with cardiovascular damage. In this review, the atherogenic effects of IS and the underlying mechanisms are summarized with emphasis on several key pathological events associated with AS developments, such as vascular endothelium dysfunction, arterial medial lesions, vascular oxidative stress, excessive inflammatory responses, calcification, thrombosis and foam cell formation. Although recent studies have proved the great correlation between IS and AS, deciphering cellular and pathophysiological signaling by confirming key factors involved in IS-mediated atherosclerosis development may enable identification of novel therapeutic targets.
Collapse
Affiliation(s)
- Cong Lu
- Research Laboratory of Translational Medicine, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, People's Republic of China; Departments of Clinical Medicine, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, People's Republic of China
| | - Li Wu
- Research Laboratory of Translational Medicine, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, People's Republic of China; Departments of Clinical Medicine, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, People's Republic of China
| | - Mu-Yao Tang
- Research Laboratory of Translational Medicine, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, People's Republic of China; Departments of Clinical Medicine, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, People's Republic of China
| | - Yi-Fan Liu
- Research Laboratory of Translational Medicine, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, People's Republic of China
| | - Lei Liu
- Research Laboratory of Translational Medicine, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, People's Republic of China; Departments of Clinical Medicine, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, People's Republic of China
| | - Xi-Ya Liu
- Research Laboratory of Translational Medicine, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, People's Republic of China; Departments of Clinical Medicine, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, People's Republic of China
| | - Chun Zhang
- Research Laboratory of Translational Medicine, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, People's Republic of China; Departments of Clinical Medicine, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, People's Republic of China
| | - Liang Huang
- Research Laboratory of Translational Medicine, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, People's Republic of China.
| |
Collapse
|
11
|
Soppert J, Heussen NM, Noels H. The authors reply. Kidney Int 2023; 103:1199-1201. [PMID: 37210195 DOI: 10.1016/j.kint.2023.02.023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Revised: 02/20/2023] [Accepted: 02/27/2023] [Indexed: 05/22/2023]
Affiliation(s)
- Josefin Soppert
- Institute for Molecular Cardiovascular Research (IMCAR), University Hospital RWTH Aachen, Aachen, Germany; Department of Anesthesiology, University Hospital RWTH Aachen, Aachen, Germany
| | - Nicole Maria Heussen
- Department of Medical Statistics, RWTH Aachen University, Aachen, Germany; Center of Biostatistics and Epidemiology, Medical School, Sigmund Freud University, Vienna, Austria
| | - Heidi Noels
- Institute for Molecular Cardiovascular Research (IMCAR), University Hospital RWTH Aachen, Aachen, Germany; Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, the Netherlands.
| |
Collapse
|
12
|
Yang M, Peng B, Zhuang Q, Li J, Zhang P, Liu H, Zhu Y, Ming Y. Machine learning-based investigation of the relationship between immune status and left ventricular hypertrophy in patients with end-stage kidney disease. Front Cardiovasc Med 2023; 10:1187965. [PMID: 37273870 PMCID: PMC10233114 DOI: 10.3389/fcvm.2023.1187965] [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: 03/16/2023] [Accepted: 05/05/2023] [Indexed: 06/06/2023] Open
Abstract
Background Left ventricular hypertrophy (LVH) is the most frequent cardiac complication among end-stage kidney disease (ESKD) patients, which has been identified as predictive of adverse outcomes. Emerging evidence has suggested that immune system is implicated in the development of cardiac hypertrophy in multiple diseases. We applied machine learning models to exploring the relation between immune status and LVH in ESKD patients. Methods A cohort of 506 eligible patients undergoing immune status assessment and standard echocardiography simultaneously in our center were retrospectively analyzed. The association between immune parameters and the occurrence of LVH were evaluated through univariate and multivariate logistic analysis. To develop a predictive model, we utilized four distinct modeling approaches: support vector machine (SVM), logistic regression (LR), multi-layer perceptron (MLP), and random forest (RF). Results In comparison to the non-LVH group, ESKD patients with LVH exhibited significantly impaired immune function, as indicated by lower cell counts of CD3+ T cells, CD4+ T cells, CD8+ T cells, and B cells. Additionally, multivariable Cox regression analysis revealed that a decrease in CD3+ T cell count was an independent risk factor for LVH, while a decrease in NK cell count was associated with the severity of LVH. The RF model demonstrated superior performance, with an average area under the curve (AUC) of 0.942. Conclusion Our findings indicate a strong association between immune parameters and LVH in ESKD patients. Moreover, the RF model exhibits excellent predictive ability in identifying ESKD patients at risk of developing LVH. Based on these results, immunomodulation may represent a promising approach for preventing and treating this disease.
Collapse
Affiliation(s)
- Min Yang
- Transplantation Center, The Third Xiangya Hospital, Central South University, Changsha, China
- Engineering and Technology Research Center for Transplantation Medicine of National Health Commission, Changsha, China
| | - Bo Peng
- Transplantation Center, The Third Xiangya Hospital, Central South University, Changsha, China
- Engineering and Technology Research Center for Transplantation Medicine of National Health Commission, Changsha, China
| | - Quan Zhuang
- Transplantation Center, The Third Xiangya Hospital, Central South University, Changsha, China
- Engineering and Technology Research Center for Transplantation Medicine of National Health Commission, Changsha, China
| | - Junhui Li
- Transplantation Center, The Third Xiangya Hospital, Central South University, Changsha, China
- Engineering and Technology Research Center for Transplantation Medicine of National Health Commission, Changsha, China
| | - Pengpeng Zhang
- Transplantation Center, The Third Xiangya Hospital, Central South University, Changsha, China
- Engineering and Technology Research Center for Transplantation Medicine of National Health Commission, Changsha, China
| | - Hong Liu
- Transplantation Center, The Third Xiangya Hospital, Central South University, Changsha, China
- Engineering and Technology Research Center for Transplantation Medicine of National Health Commission, Changsha, China
| | - Yi Zhu
- Transplantation Center, The Third Xiangya Hospital, Central South University, Changsha, China
- Engineering and Technology Research Center for Transplantation Medicine of National Health Commission, Changsha, China
| | - Yingzi Ming
- Transplantation Center, The Third Xiangya Hospital, Central South University, Changsha, China
- Engineering and Technology Research Center for Transplantation Medicine of National Health Commission, Changsha, China
| |
Collapse
|
13
|
Abstract
Chronic kidney disease is associated with an increased risk for the development and progression of cardiovascular disorders including hypertension, dyslipidemia, and coronary artery disease. Chronic kidney disease may also affect the myocardium through complex systemic changes, resulting in structural remodeling such as hypertrophy and fibrosis, as well as impairments in both diastolic and systolic function. These cardiac changes in the setting of chronic kidney disease define a specific cardiomyopathic phenotype known as uremic cardiomyopathy. Cardiac function is tightly linked to its metabolism, and research over the past 3 decades has revealed significant metabolic remodeling in the myocardium during the development of heart failure. Because the concept of uremic cardiomyopathy has only been recognized in recent years, there are limited data on metabolism in the uremic heart. Nonetheless, recent findings suggest overlapping mechanisms with heart failure. This work reviews key features of metabolic remodeling in the failing heart in the general population and extends this to patients with chronic kidney disease. The knowledge of similarities and differences in cardiac metabolism between heart failure and uremic cardiomyopathy may help identify new targets for mechanistic and therapeutic research on uremic cardiomyopathy.
Collapse
Affiliation(s)
- T Dung Nguyen
- Department of Internal Medicine I, University Hospital Jena, Jena, Germany
| | | |
Collapse
|
14
|
Duni A, Kitsos A, Bechlioulis A, Markopoulos GS, Lakkas L, Baxevanos G, Mitsis M, Vartholomatos G, Naka KK, Dounousi E. Differences in the Profile of Circulating Immune Cell Subsets in Males with Type 2 Cardiorenal Syndrome versus CKD Patients without Established Cardiovascular Disease. Biomedicines 2023; 11:biomedicines11041029. [PMID: 37189647 DOI: 10.3390/biomedicines11041029] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Revised: 03/17/2023] [Accepted: 03/22/2023] [Indexed: 03/29/2023] Open
Abstract
Maladaptive activation of the immune system plays a key role in the pathogenesis of chronic kidney disease (CKD). Our aim was to investigate differences in circulating immune cells between type 2 cardiorenal syndrome (CRS-2) patients and CKD patients without cardiovascular disease (CVD). CRS-2 patients were prospectively followed up, with the primary endpoint being all-cause and cardiovascular mortality. Method: A total of 39 stable males with CRS-2 and 24 male CKD patients matched for eGFR (CKD-EPI) were enrolled. A selected panel of immune cell subsets was measured by flow cytometry. Results: Compared to CKD patients, CRS-2 patients displayed higher levels of proinflammatory CD14++CD16+ monocytes (p = 0.04) and T regulatory cells (Tregs) (p = 0.03), lower lymphocytes (p = 0.04), and lower natural killer cells (p = 0.001). Decreased lymphocytes, T-lymphocytes, CD4+ T-cells, CD8+ T-cells, Tregs, and increased CD14++CD16+ monocytes were associated with mortality at a median follow-up of 30 months (p < 0.05 for all). In a multivariate model including all six immune cell subsets, only CD4+ T-lymphocytes remained independent predictors of mortality (OR 0.66; 95% CI 0.50–0.87; p = 0.004). Conclusion: Patients with CRS-2 exhibit alterations in immune cell profile compared to CKD patients of similar kidney function but without CVD. In the CRS-2 cohort, CD4+ T-lymphocytes independently predicted fatal cardiovascular events.
Collapse
|
15
|
Han B, Zhang X, Wang L, Yuan W. Dysbiosis of Gut Microbiota Contributes to Uremic Cardiomyopathy via Induction of IFNγ-Producing CD4 + T Cells Expansion. Microbiol Spectr 2023; 11:e0310122. [PMID: 36788674 PMCID: PMC9927280 DOI: 10.1128/spectrum.03101-22] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2022] [Accepted: 11/18/2022] [Indexed: 12/23/2022] Open
Abstract
Uremic cardiomyopathy (UCM) correlates with chronic kidney disease (CKD)-induced morbidity and mortality. Gut microbiota has been involved in the pathogenesis of certain cardiovascular disease, but the role of gut microbiota in the pathogenesis of UCM remains unknown. Here, we performed a case-control study to compare the gut microbiota of patients with CKD and healthy controls by 16S rRNA (rRNA) gene sequencing. To test the causative relationship between gut microbiota and UCM, we performed fecal microbiota transplantation (FMT) in 5/6th nephrectomy model of CKD. We found that opportunistic pathogens, particularly Klebsiella pneumoniae (K. pneumoniae), are markedly enriched in patients with CKD. FMT from CKD patients aggravated diastolic dysfunction in the mouse model. The diastolic dysfunction was associated with microbiome-dependent increases in heart-infiltrating IFNγ+ CD4+ T cells. Monocolonization with K. pneumoniae increased cardiac IFNγ+ CD4+ T cells infiltration and promoted UCM development of the mouse model. A probiotic Bifidobacterium animalis decreased the relative abundance of K. pneumoniae, reduced levels of cardiac IFNγ+ CD4+ T cells and ameliorated the severity of diastolic dysfunction in the mice. Thus, the aberrant gut microbiota in CKD patients, especially K. pneumoniae, contributed to UCM pathogenesis through the induction of heart-infiltrating IFNγ+ CD4+ T cells expansion, proposing that a Gut Microbiota-Gut-Kidney-Heart axis could play a critical role in elucidating the etiology of UCM, and suggesting that modulation of the gut bacteria may serve as a promising target for the amelioration of UCM. IMPORTANCE Uremic cardiomyopathy (UCM) correlates tightly with increased mortality in patients with chronic kidney disease (CKD), yet the pathogenesis of UCM remains incompletely understood, limiting therapeutic approaches. Our study proposed that a Gut Microbiota-Gut-Kidney-Heart axis could play a critical role in understanding etiology of UCM. There is a major need in future clinical trials of patients with CKD to explore if modulation of gut microbiota by fecal microbiota transplantation (FMT), probiotics or antibiotics can alleviate cardiac dysfunction, reduce mortality, and improve life quality.
Collapse
Affiliation(s)
- Bin Han
- Department of Nephrology, Shanghai General Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Xiaoqian Zhang
- Department of Nephrology, Shanghai General Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Ling Wang
- Department of Nephrology, Shanghai General Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Weijie Yuan
- Department of Nephrology, Shanghai General Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| |
Collapse
|
16
|
Matsui M, Onoue K, Saito Y. sFlt-1 in Chronic Kidney Disease: Friend or Foe? Int J Mol Sci 2022; 23:ijms232214187. [PMID: 36430665 PMCID: PMC9697971 DOI: 10.3390/ijms232214187] [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: 10/03/2022] [Revised: 11/08/2022] [Accepted: 11/14/2022] [Indexed: 11/18/2022] Open
Abstract
Placental growth factor (PlGF) and its receptor, fms-like tyrosine kinase-1 (Flt-1), are important regulators involved in angiogenesis, atherogenesis, and inflammation. This review article focuses on the function of PlGF/Flt-1 signaling and its regulation by soluble Flt-1 (sFlt-1) in chronic kidney disease (CKD). Elevation of circulating sFlt-1 and downregulation of sFlt-1 in the vascular endothelium by uremic toxins and oxidative stress both exacerbate heart failure and atherosclerosis. Circulating sFlt-1 is inconsistent with sFlt-1 synthesis, because levels of matrix-bound sFlt-1 are much higher than those of circulating sFlt-1, as verified by a heparin loading test, and are drastically reduced in CKD.
Collapse
Affiliation(s)
- Masaru Matsui
- Department of Nephrology, Nara Prefecture General Medical Center, 2-897-5 Shichijo-Nishimachi, Nara 630-8581, Japan
- Department of Nephrology, Nara Medical University, 840 Shijo-Cho, Kashihara 634-8521, Japan
- Correspondence: ; Tel./Fax: +81-742-46-6001
| | - Kenji Onoue
- Department of Cardiology, Nara Medical University, 840 Shijo-Cho, Kashihara 634-8521, Japan
| | - Yoshihiko Saito
- Nara Prefecture Seiwa Medical Center, 1-14-16, Mimuro, Sango-Cho, Ikoma-Gun 636-0802, Japan
| |
Collapse
|
17
|
Xu Y, Cao L, Ji S, Shen W. LncRNA ANRIL-mediated miR-181b-5p/S1PR1 axis is involved in the progression of uremic cardiomyopathy through activating T cells. Sci Rep 2022; 12:18027. [PMID: 36302829 PMCID: PMC9613656 DOI: 10.1038/s41598-022-22955-x] [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: 06/08/2022] [Accepted: 10/21/2022] [Indexed: 01/24/2023] Open
Abstract
This study aimed to explore the regulatory role of lncRNA ANRIL/miR-181b-5p/S1PR1 in UC. UC mouse model was established by 5/6th nephrectomy. We detected body weight, serum levels of renal function and inflammatory factors (biochemical analyzer/ELISA), and cardiac parameters (echocardiography). HE and Masson staining showed the pathological changes and fibrosis in myocardial and nephridial tissues. The expression of ANRIL, miR-181b-5p, and S1PR1 were detected by qRT-PCR or Western blot/immunofluorescence. T cells activation was analyzed by Flow cytometry. ANRIL/S1PR1 were up-regulated and miR-181b-5p was down-regulated in UC mice. ANRIL silencing up-regulated miR-181b-5p and down-regulated S1PR1 (a target of miR-181b-5p). ANRIL silencing increased the body weight, recovered renal function [decreased blood urea nitrogen (BUN) and serum creatinine (Scr)] and cardiac function [decreased left ventricular end-diastolic diameter (LVEDD), LV end-systolic diameter (LVESD), LV systolic anterior wall thickness (LVAWS), LV end-diastolic anterior wall thickness (LVAWD), myocardial performance index (MPI), and isovolumic relaxation time (IVRT); increased LV ejection fraction (LVEF), LVEF/MPI, fractional shortening (FS), and E- and A-waves (E/A)], inhibited the inflammation [decreased interferon (IFN)-γ, interleukin (IL)-2, IL-10, and tumor necrosis factor (TNF)-α], and relieved pathological injuries and fibrosis. ANRIL silencing also recovered the viability and inhibited the inflammation of activated T cells in vitro, and inhibited T cell activation in UC mice in vivo. In addition, miR-181b-5p overexpression exhibited same effects with ANRIL silencing in UC. ANRIL silencing inhibited T cell activation through regulating miR-181b-5p/S1PR1, contributing to the remission of UC.
Collapse
Affiliation(s)
- Ying Xu
- grid.417401.70000 0004 1798 6507Urology and Nephrology Center, Department of Urology, Zhejiang Provincial People’s Hospital (Affiliated People’s Hospital, Hangzhou Medical College), Hangzhou, China
| | - Luxi Cao
- grid.417401.70000 0004 1798 6507Urology and Nephrology Center, Department of Nephrology, Zhejiang Provincial People’s Hospital (Affiliated People’s Hospital, Hangzhou Medical College), Hangzhou, China
| | - Shuiyu Ji
- grid.417401.70000 0004 1798 6507Urology and Nephrology Center, Department of Nephrology, Zhejiang Provincial People’s Hospital (Affiliated People’s Hospital, Hangzhou Medical College), Hangzhou, China
| | - Wei Shen
- grid.417401.70000 0004 1798 6507Urology and Nephrology Center, Department of Nephrology, Zhejiang Provincial People’s Hospital (Affiliated People’s Hospital, Hangzhou Medical College), Hangzhou, China
| |
Collapse
|
18
|
Mawhin MA, Bright RG, Fourre JD, Vloumidi EI, Tomlinson J, Sardini A, Pusey CD, Woollard KJ. Chronic kidney disease mediates cardiac dysfunction associated with increased resident cardiac macrophages. BMC Nephrol 2022; 23:47. [PMID: 35090403 PMCID: PMC8796634 DOI: 10.1186/s12882-021-02593-7] [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: 06/16/2021] [Accepted: 11/01/2021] [Indexed: 11/29/2022] Open
Abstract
BACKGROUND The leading cause of death in end-stage kidney disease is related to cardiovascular disease. Macrophages are known to be involved in both chronic kidney disease (CKD) and heart failure, however their role in the development of cardiorenal syndrome is less clear. We thus sought to investigate the role of macrophages in uremic cardiac disease. METHODS We assessed cardiac response in two experimental models of CKD and tested macrophage and chemokine implication in monocytopenic CCR2-/- and anti-CXCL10 treated mice. We quantified CXCL10 in human CKD plasma and tested the response of human iPSC-derived cardiomyocytes and primary cardiac fibroblasts to serum from CKD donors. RESULTS We found that reduced kidney function resulted in the expansion of cardiac macrophages, in particular through local proliferation of resident populations. Influx of circulating monocytes contributed to this increase. We identified CXCL10 as a crucial factor for cardiac macrophage expansion in uremic disease. In humans, we found increased plasma CXCL10 concentrations in advanced CKD, and identified the production of CXCL10 in cardiomyocytes and cardiac fibroblasts. CONCLUSIONS This study provides new insight into the role of the innate immune system in uremic cardiomyopathy.
Collapse
Affiliation(s)
- M A Mawhin
- Centre for Inflammatory Disease, Department of Immunology and Inflammation, Imperial College London, London, UK.
| | - R G Bright
- Centre for Inflammatory Disease, Department of Immunology and Inflammation, Imperial College London, London, UK
| | - J D Fourre
- Faculty of Medicine, National Heart & Lung Institute, Imperial College London, London, UK
| | - E I Vloumidi
- MRC Laboratory of Molecular Biology, Imperial College London, London, UK
| | - J Tomlinson
- Renal Directorate, Imperial College Healthcare NHS Trust, London, UK
| | - A Sardini
- MRC London Institute of Medical Sciences, Imperial College London, London, UK
| | - C D Pusey
- Centre for Inflammatory Disease, Department of Immunology and Inflammation, Imperial College London, London, UK
| | - K J Woollard
- Centre for Inflammatory Disease, Department of Immunology and Inflammation, Imperial College London, London, UK.
| |
Collapse
|
19
|
Huang XB, Ye SZ, Wu JW, Fu QS, Liu BH, Qiu HX, Cheng GQ. Diversity of the T cell receptor β chain complementarity-determining region 3 in peripheral blood of neonates with sepsis: an analysis based on immune repertoire sequencing. ZHONGGUO DANG DAI ER KE ZA ZHI = CHINESE JOURNAL OF CONTEMPORARY PEDIATRICS 2021; 23:1154-1160. [PMID: 34753548 DOI: 10.7499/j.issn.1008-8830.2106044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
OBJECTIVES To investigate the diversity of peripheral blood T cell receptor (TCR) β chain complementarity-determining region 3 (CDR3) based on immune repertoire sequencing in neonates with sepsis and the possible pathogenesis of neonatal sepsis. METHODS A total of 12 neonates with sepsis were enrolled as the case group, and 9 healthy full-term infants, matched for gestational age, birth weight, and age, were enrolled as the control group. Omega nucleic acid purification kit (SQ blood DNA Kit II) was used to extract DNA from peripheral blood samples, TCR β chain CDR3 was amplified by multiplex PCR, and then high-throughput sequencing was performed for the products to analyze the diversity of TCR β chain CDR3 and the difference in expression. RESULTS The length and type of TCR β chain CDR3 were similar between the case and control groups, and Gaussian distribution was observed in both groups. With D50 and Shannon-Wiener index as the evaluation indices for diversity, the case group had a significantly lower diversity of TCR β chain CDR3 than the control group (P<0.05). The frequency of 48 genes in TCR β chain V segment was compared, and the results showed that compared with the control group, the case group had significantly higher frequencies of TRBV10-3, TRBV2, and TRBV20-1 (P<0.05). The frequency of 13 genes in TCR β chain J segment were compared, and the results showed that compared with the control group, the case group had significantly higher frequencies of TRBJ2-3, TRBJ2-5, and TRBJ2-7 (P<0.05). CONCLUSIONS There is a significant change in the diversity of TCR β chain CDR3 in the peripheral blood of neonates with sepsis, suggesting that it might be associated with the immune pathogenesis of neonatal sepsis.
Collapse
Affiliation(s)
- Xun-Bin Huang
- Department of Neonatology, Longgang District Central Hospital of Shenzhen, Shenzhen, Guangdong 518116, China
| | - Shu-Zhen Ye
- Department of Neonatology, Longgang District Central Hospital of Shenzhen, Shenzhen, Guangdong 518116, China
| | - Ji-Wei Wu
- Department of Neonatology, Longgang District Central Hospital of Shenzhen, Shenzhen, Guangdong 518116, China
| | - Qing-Song Fu
- Department of Neonatology, Longgang District Central Hospital of Shenzhen, Shenzhen, Guangdong 518116, China
| | - Bi-Hua Liu
- Department of Neonatology, Longgang District Central Hospital of Shenzhen, Shenzhen, Guangdong 518116, China
| | - Hui-Xian Qiu
- Department of Neonatology, Longgang District Central Hospital of Shenzhen, Shenzhen, Guangdong 518116, China
| | | |
Collapse
|
20
|
Soppert J, Frisch J, Wirth J, Hemmers C, Boor P, Kramann R, Vondenhoff S, Moellmann J, Lehrke M, Hohl M, van der Vorst EPC, Werner C, Speer T, Maack C, Marx N, Jankowski J, Roma LP, Noels H. A systematic review and meta-analysis of murine models of uremic cardiomyopathy. Kidney Int 2021; 101:256-273. [PMID: 34774555 DOI: 10.1016/j.kint.2021.10.025] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2021] [Revised: 09/22/2021] [Accepted: 10/18/2021] [Indexed: 02/06/2023]
Abstract
Chronic kidney disease (CKD) triggers the risk of developing uremic cardiomyopathy as characterized by cardiac hypertrophy, fibrosis and functional impairment. Traditionally, animal studies are used to reveal the underlying pathological mechanism, although variable CKD models, mouse strains and readouts may reveal diverse results. Here, we systematically reviewed 88 studies and performed meta-analyses of 52 to support finding suitable animal models for future experimental studies on pathological kidney-heart crosstalk during uremic cardiomyopathy. We compared different mouse strains and the direct effect of CKD on cardiac hypertrophy, fibrosis and cardiac function in "single hit" strategies as well as cardiac effects of kidney injury combined with additional cardiovascular risk factors in "multifactorial hit" strategies. In C57BL/6 mice, CKD was associated with a mild increase in cardiac hypertrophy and fibrosis and marginal systolic dysfunction. Studies revealed high variability in results, especially regarding hypertrophy and systolic function. Cardiac hypertrophy in CKD was more consistently observed in 129/Sv mice, which express two instead of one renin gene and more consistently develop increased blood pressure upon CKD induction. Overall, "multifactorial hit" models more consistently induced cardiac hypertrophy and fibrosis compared to "single hit" kidney injury models. Thus, genetic factors and additional cardiovascular risk factors can "prime" for susceptibility to organ damage, with increased blood pressure, cardiac hypertrophy and early cardiac fibrosis more consistently observed in 129/Sv compared to C57BL/6 strains.
Collapse
Affiliation(s)
- Josefin Soppert
- Institute for Molecular Cardiovascular Research (IMCAR), University Hospital RWTH Aachen, Aachen, Germany
| | - Janina Frisch
- Department of Biophysics, Center for Human and Molecular Biology (ZHMB), Saarland University, Homburg, Germany
| | - Julia Wirth
- Institute for Molecular Cardiovascular Research (IMCAR), University Hospital RWTH Aachen, Aachen, Germany
| | - Christian Hemmers
- Institute for Molecular Cardiovascular Research (IMCAR), University Hospital RWTH Aachen, Aachen, Germany
| | - Peter Boor
- Institute of Pathology, University Hospital RWTH Aachen, Aachen, Germany; Department of Nephrology and Clinical Immunology, University Hospital RWTH Aachen, Aachen, Germany
| | - Rafael Kramann
- Department of Nephrology and Clinical Immunology, University Hospital RWTH Aachen, Aachen, Germany
| | - Sonja Vondenhoff
- Institute for Molecular Cardiovascular Research (IMCAR), University Hospital RWTH Aachen, Aachen, Germany
| | - Julia Moellmann
- Department of Internal Medicine I, Cardiology, University Hospital RWTH Aachen, Aachen, Germany
| | - Michael Lehrke
- Department of Internal Medicine I, Cardiology, University Hospital RWTH Aachen, Aachen, Germany
| | - Mathias Hohl
- Department of Internal Medicine III, Cardiology/Angiology, University of Homburg, Homburg/Saar, Germany
| | - Emiel P C van der Vorst
- Institute for Molecular Cardiovascular Research (IMCAR), University Hospital RWTH Aachen, Aachen, Germany; Department of Pathology, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University Medical Centre, Maastricht, The Netherlands; Interdisciplinary Centre for Clinical Research (IZKF), RWTH Aachen University, Aachen, Germany; Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximilians-University Munich, Munich, Germany; German Centre for Cardiovascular Research (DZHK), partner site Munich Heart Alliance, Munich, Germany
| | - Christian Werner
- Department of Internal Medicine III, Cardiology/Angiology, University of Homburg, Homburg/Saar, Germany
| | - Thimoteus Speer
- Translational Cardio-Renal Medicine, Saarland University, Homburg/Saar, Germany
| | - Christoph Maack
- Department of Translational Research, Comprehensive Heart Failure Center (CHFC), University Hospital Würzburg, Würzburg, Germany
| | - Nikolaus Marx
- Department of Internal Medicine I, Cardiology, University Hospital RWTH Aachen, Aachen, Germany
| | - Joachim Jankowski
- Institute for Molecular Cardiovascular Research (IMCAR), University Hospital RWTH Aachen, Aachen, Germany; Department of Pathology, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University Medical Centre, Maastricht, The Netherlands
| | - Leticia Prates Roma
- Department of Biophysics, Center for Human and Molecular Biology (ZHMB), Saarland University, Homburg, Germany
| | - Heidi Noels
- Institute for Molecular Cardiovascular Research (IMCAR), University Hospital RWTH Aachen, Aachen, Germany; Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, The Netherlands.
| |
Collapse
|
21
|
Amador-Martínez I, García-Ballhaus J, Buelna-Chontal M, Cortés-González C, Massó F, Jaisser F, Barrera-Chimal J. Early inflammatory changes and CC chemokine ligand-8 upregulation in the heart contribute to uremic cardiomyopathy. FASEB J 2021; 35:e21761. [PMID: 34245616 DOI: 10.1096/fj.202100746r] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2021] [Revised: 06/08/2021] [Accepted: 06/14/2021] [Indexed: 12/23/2022]
Abstract
Uremic cardiomyopathy is a common complication in chronic kidney disease (CKD) patients, accounting for a high mortality rate. Several mechanisms have been proposed to link CKD and cardiac alterations; however, the early cardiac modifications that occur in CKD that may trigger cardiac remodeling and dysfunction remain largely unexplored. Here, in a mouse model of CKD induced by 5/6 nephrectomy, we first analyzed the early transcriptional and inflammatory changes that occur in the heart. Five days after 5/6 nephrectomy, RNA-sequencing showed the upregulation of 54 genes in the cardiac tissue of CKD mice and the enrichment of biological processes related to immune system processes. Increased cardiac infiltration of T-CD4+ lymphocytes, myeloid cells, and macrophages during early CKD was observed. Next, since CC chemokine ligand-8 (CCL8) was one of the most upregulated genes in the heart of mice with early CKD, we investigated the effect of acute and transient CCL8 inhibition on uremic cardiomyopathy severity. An increase in CCL8 protein levels was confirmed in the heart of early CKD mice. CCL8 inhibition attenuated the early infiltration of T-CD4+ lymphocytes and macrophages to the cardiac tissue, leading to a protection against chronic cardiac fibrotic remodeling, inflammation and cardiac dysfunction induced by CKD. Altogether, our data show the occurrence of transcriptional and inflammatory changes in the heart during the early phases of CKD and identify CCL8 as a key contributor to the early cardiac inflammatory state that triggers further cardiac remodeling and dysfunction in uremic cardiomyopathy.
Collapse
Affiliation(s)
- Isabel Amador-Martínez
- Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Ciudad Universitaria, Mexico City, Mexico.,Laboratorio de Fisiología Cardiovascular y Trasplante Renal, Unidad de Investigación UNAM-INC, Instituto Nacional de Cardiología Ignacio Chávez, Mexico City, Mexico
| | - Johannes García-Ballhaus
- Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Ciudad Universitaria, Mexico City, Mexico.,Laboratorio de Fisiología Cardiovascular y Trasplante Renal, Unidad de Investigación UNAM-INC, Instituto Nacional de Cardiología Ignacio Chávez, Mexico City, Mexico
| | - Mabel Buelna-Chontal
- Departamento de Biomedicina Cardiovascular, Instituto Nacional de Cardiología Ignacio Chávez, Mexico City, Mexico
| | - César Cortés-González
- Unidad de Investigación Biomédica en Cáncer, Instituto Nacional de Cancerología, Mexico City, Mexico
| | - Felipe Massó
- Laboratorio de Medicina Traslacional, Unidad de Investigación UNAM-INC, Instituto Nacional de Cardiología Ignacio Chávez, Mexico City, Mexico
| | - Frédéric Jaisser
- INSERM, UMRS 1138, Centre de Recherche des Cordeliers, Sorbonne Université, Université de Paris, Paris, France.,French-Clinical Research Infrastructure Network (F-CRIN), INI-CRCT, INSERM Centre d'Investigations Cliniques-Plurithématique 1433, UMR 1116, CHRU de Nancy, Université de Lorraine, Nancy, France
| | - Jonatan Barrera-Chimal
- Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Ciudad Universitaria, Mexico City, Mexico.,Laboratorio de Fisiología Cardiovascular y Trasplante Renal, Unidad de Investigación UNAM-INC, Instituto Nacional de Cardiología Ignacio Chávez, Mexico City, Mexico
| |
Collapse
|
22
|
Huang J, Kong Y, Xie C, Zhou L. Stem/progenitor cell in kidney: characteristics, homing, coordination, and maintenance. Stem Cell Res Ther 2021; 12:197. [PMID: 33743826 PMCID: PMC7981824 DOI: 10.1186/s13287-021-02266-0] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Accepted: 03/02/2021] [Indexed: 02/06/2023] Open
Abstract
Renal failure has a high prevalence and is becoming a public health problem worldwide. However, the renal replacement therapies such as dialysis are not yet satisfactory for its multiple complications. While stem/progenitor cell-mediated tissue repair and regenerative medicine show there is light at the end of tunnel. Hence, a better understanding of the characteristics of stem/progenitor cells in kidney and their homing capacity would greatly promote the development of stem cell research and therapy in the kidney field and open a new route to explore new strategies of kidney protection. In this review, we generally summarize the main stem/progenitor cells derived from kidney in situ or originating from the circulation, especially bone marrow. We also elaborate on the kidney-specific microenvironment that allows stem/progenitor cell growth and chemotaxis, and comment on their interaction. Finally, we highlight potential strategies for improving the therapeutic effects of stem/progenitor cell-based therapy. Our review provides important clues to better understand and control the growth of stem cells in kidneys and develop new therapeutic strategies.
Collapse
Affiliation(s)
- Jiewu Huang
- State Key Laboratory of Organ Failure Research, National Clinical Research Center of Kidney Disease, Division of Nephrology, Nanfang Hospital, Southern Medical University, 1838 North Guangzhou Ave, Guangzhou, 510515, China
| | - Yaozhong Kong
- Department of Nephrology, the First People's Hospital of Foshan, Foshan, Guangdong, China
| | - Chao Xie
- Department of Nephrology, the First People's Hospital of Foshan, Foshan, Guangdong, China
| | - Lili Zhou
- State Key Laboratory of Organ Failure Research, National Clinical Research Center of Kidney Disease, Division of Nephrology, Nanfang Hospital, Southern Medical University, 1838 North Guangzhou Ave, Guangzhou, 510515, China. .,Bioland Laboratory (Guangzhou Regenerative Medicine and Health Guangdong Laboratory), Guangzhou, China.
| |
Collapse
|
23
|
Saito Y. The role of the PlGF/Flt-1 signaling pathway in the cardiorenal connection. J Mol Cell Cardiol 2020; 151:106-112. [PMID: 33045252 DOI: 10.1016/j.yjmcc.2020.10.001] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/14/2020] [Revised: 09/29/2020] [Accepted: 10/02/2020] [Indexed: 02/07/2023]
Abstract
Although the concept of the cardiorenal connection is widely accepted, athe underlying molecular mechanism has not been clearly defined. Nevertheless, accumulating evidence indicates that the nervous system and both the humoral and cellular immune systems are all involved. This review article focuses on the roles of the signaling pathway of placental growth factor (PlGF) and its receptor, fms-like tyrosine kinase-1 (Flt-1), in the development of the cardiorenal connection. PlGF, a member of the vascular endothelial cell growth factor family, is a specific ligand for Flt-1 and plays roles in the development of atherosclerosis, wound healing after ischemia injury, and angiogenesis through Flt-1 signaling. Flt-1, a tyrosine-kinase type receptor with a single transmembrane domain, has a soluble isoform (sFlt-1) consisting of only extracellular domains, and is an intrinsic antagonist of PlGF. In renal dysfunction, PlGF is upregulated and sFlt-1 is downregulated by oxidative stress or uremic toxins, leading to activation of the PlGF/Flt-1 signaling pathway, which in turn plays a role in the worsening of atherosclerosis and heart failure, both of which are frequently associated with renal dysfunction. Monocyte chemotactic protein-1 (MCP-1) is involved in the process downstream of the Flt-1 signaling pathway. Plasma levels of sFlt-1 correlate with the severity of renal dysfunction in patients with heart failure or myocardial infarction, and are associated with the incidence of cardiovascular events. This is inconsistent with the concept of relative activation of the PlGF/Flt-1 pathway in renal dysfunction. However, the level of circulating sFlt-1 does not always parallel sFlt-1 synthesis, probably because sFlt-1 is stored on cell surfaces through its heparin-binding domains and its quantity is regulated differently in renal dysfunction. This review summarizes a novel concept wherein noninfectious inflammation via PlGF/Flt-1 signaling is involved in the development of renal dysfunction-related cardiovascular complications.
Collapse
Affiliation(s)
- Yoshihiko Saito
- Department of Cardiovascular Medicine, Nara Medical University, 840 Shijo-cho, Kashihara 634-8522, Japan.
| |
Collapse
|
24
|
Manousek J, Felsoci M, Miklik R, Parenica J, Krejci J, Bjørklund G, Klanova J, Mlejnek D, Miklikova M, Lokaj P, Chirumbolo S, Spinar J. Delayed-type Hypersensitivity to Metals in Newly Diagnosed Patients with Nonischemic Dilated Cardiomyopathy. Cardiovasc Toxicol 2020; 20:571-580. [PMID: 32557318 DOI: 10.1007/s12012-020-09582-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
The causes of nonischemic dilated cardiomyopathy are classified as genetic or nongenetic, but environmental factors such as metal pollutants may interact with genetic susceptibility. The presence of metal particles has been detected in the myocardium, including in those patients with dilated cardiomyopathy. It is also known that hypersensitivity reactions can induce inflammation in tissue. The present study aimed to verify if metal-induced delayed-type hypersensitivity is present in patients with nonischemic dilated cardiomyopathy. The patient group consisted of 30 patients with newly diagnosed dilated cardiomyopathy; the control group comprised 41 healthy subjects. All patients and control subjects provided blood samples for lymphocyte transformation testing (MELISA®) to assess possible hypersensitivity to seven common metals. Specific exposure to metals was based on interview data. Results showed that exposure to cadmium and lead (p = 0.0002), aluminum (p = 0.0006), nickel (p = 0.0012), and chromium (p = 0.0065) was more often reported by patients than controls. The patients also had significantly more frequent hypersensitivity reactions to mercury (26.7% vs. 7.3%, p = 0.014624), nickel (40% vs. 12.2%, p = 0.02341), and silver (20% vs. 4.8%, p = 0.025468) than the control group. Patients with dilated cardiomyopathy had greater exposure to certain metals compared with healthy controls. Hypersensitivity to metals was more frequent in patients with dilated cardiomyopathy, suggesting a possible association that warrants further investigation.
Collapse
Affiliation(s)
- Jan Manousek
- Department of Internal Medicine and Cardiology, University Hospital Brno and Faculty of Medicine, Masaryk University, Brno, Czech Republic
| | - Marian Felsoci
- Department of Internal Medicine and Cardiology, University Hospital Brno and Faculty of Medicine, Masaryk University, Brno, Czech Republic
| | - Roman Miklik
- Department of Cardiology, University Hospital and Faculty of Medicine Pilsen, Charles University, Prague, Czech Republic.
| | - Jiri Parenica
- Department of Internal Medicine and Cardiology, University Hospital Brno and Faculty of Medicine, Masaryk University, Brno, Czech Republic
| | - Jan Krejci
- Department of Cardiovascular Diseases, St. Anne's University Hospital and Faculty of Medicine, Masaryk University, Brno, Czech Republic
| | - Geir Bjørklund
- Council for Nutritional and Environmental Medicine (CONEM), Toften 24, 8610, Mo i Rana, Norway.
| | - Jana Klanova
- Research Centre for Toxic Compounds in the Environment (RECETOX), Faculty of Science, Masaryk University, Brno, Czech Republic
| | - Dalibor Mlejnek
- Department of Cardiovascular Diseases, St. Anne's University Hospital and Faculty of Medicine, Masaryk University, Brno, Czech Republic
| | - Marie Miklikova
- Department of Internal Medicine and Cardiology, University Hospital Brno and Faculty of Medicine, Masaryk University, Brno, Czech Republic
| | - Petr Lokaj
- Department of Internal Medicine and Cardiology, University Hospital Brno and Faculty of Medicine, Masaryk University, Brno, Czech Republic
| | - Salvatore Chirumbolo
- Neurosciences, Biomedicine and Movement Sciences, University of Verona, Verona, Italy.,CONEM Scientific Secretary, Verona, Italy
| | - Jindrich Spinar
- Department of Internal Medicine and Cardiology, University Hospital Brno and Faculty of Medicine, Masaryk University, Brno, Czech Republic
| |
Collapse
|
25
|
Bansal N, Zelnick L, Go A, Anderson A, Christenson R, Deo R, Defilippi C, Lash J, He J, Ky B, Seliger S, Soliman E, Shlipak M. Cardiac Biomarkers and Risk of Incident Heart Failure in Chronic Kidney Disease: The CRIC (Chronic Renal Insufficiency Cohort) Study. J Am Heart Assoc 2019; 8:e012336. [PMID: 31645163 PMCID: PMC6898812 DOI: 10.1161/jaha.119.012336] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Background Cardiac biomarkers may signal mechanistic pathways involved in heart failure (HF), a leading complication in chronic kidney disease. We tested the associations of NT‐proBNP (N‐terminal pro‐B‐type natriuretic peptide), high‐sensitivity troponin T (hsTnT), galectin‐3, growth differentiation factor‐15 (GDF‐15), and soluble ST2 (sST2) with incident HF in chronic kidney disease. Methods and Results We examined adults with chronic kidney disease enrolled in a prospective, multicenter study. All biomarkers were measured at baseline. The primary outcome was incident HF. Secondary outcomes included HF with preserved ejection fraction (EF≥50%) and reduced ejection fraction (EF<50%). Cox models were used to test the association of each cardiac biomarker with HF, adjusting for demographics, kidney function, cardiovascular risk factors, and medication use. Among 3314 participants, all biomarkers, with the exception of galectin‐3, were significantly associated with increased risk of incident HF (hazard ratio per SD higher concentration of log‐transformed biomarker): NT‐proBNP (hazard ratio, 2.07; 95% CI, 1.79–2.39); hsTnT (hazard ratio, 1.38; 95% CI, 1.21–1.56); GDF‐15 (hazard ratio, 1.44; 95% CI, 1.26–1.66) and sST2 (hazard ratio, 1.19; 95% CI, 1.05–1.35). Higher NT‐proBNP, hsTnT, and GDF‐15 were also associated with a greater risk of HF with reduced EF; while higher NT‐proBNP GDF‐15 and sST2 were associated with HF with preserved EF. Galectin‐3 was not associated with either HF with reduced EF or HF with preserved EF. Conclusions In chronic kidney disease, elevations of NT‐proBNP, hsTnT, GDF‐15, sST2 were associated with incident HF. There was a borderline association of galectin‐3 with incident HF. NT‐proBNP and hsTnT were more strongly associated with HF with reduced EF, while the associations of the newer biomarkers GDF‐15 and sST2 were stronger for HF with preserved EF.
Collapse
Affiliation(s)
| | | | - Alan Go
- Division of Research Kaiser Permanente Northern California Oakland CA
| | | | | | - Rajat Deo
- University of Pennsylvania Philadelphia PA
| | | | | | - Jiang He
- Tulane University New Orleans LA
| | - Bonnie Ky
- University of Pennsylvania Philadelphia PA
| | | | | | | | | | | |
Collapse
|