1
|
Li H, Song Q, Su X, Shen Y, Yan H, Yu Z, Li Z, Yuan J, Huang J, Ni Z, Gu L, Fang W. Serum angiopoietin-2/angiopoietin-1 ratio is associated with cardiovascular and all-cause mortality in peritoneal dialysis patients: a prospective cohort study. Ren Fail 2024; 46:2380037. [PMID: 39082686 PMCID: PMC11293270 DOI: 10.1080/0886022x.2024.2380037] [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: 08/17/2023] [Revised: 04/22/2024] [Accepted: 07/10/2024] [Indexed: 08/03/2024] Open
Abstract
INTRODUCTION Our objective was to examine the factors associated with the serum angiopoietin-2/angiopoietin-1 (Angpt-2/Angpt-1) ratio in peritoneal dialysis (PD) patients and to investigate the association between Angpt-2/Angpt-1 ratio and cardiovascular and all-cause mortality. METHODS Patients on PD who were prevalent between January 2014 and April 2015 in the center of Renji Hospital were enrolled. At the time of enrollment, serum and dialysate samples were collected to detect biochemical parameters, serum angiopoietin-2 and angiopoietin-1 levels. Patients were dichotomized into two groups according to a median of Angpt-2/Angpt-1 ratio and followed up prospectively until the end of the study. RESULTS A total of 325 patients were enrolled, including 168 males (51.7%) with a mean age of 56.9 ± 14.2 years and a median PD duration of 32.4 (9.8-55.9) months. Multiple linear regression showed pulse pressure (β = 0.206, p < .001) and high-sensitivity C-reactive protein (hs-CRP) (β = 0.149, p = .011) were positively correlated with serum Angpt-2/Angpt-1 ratio, while residual renal function (RRF) (β= -0.219, p < .001) was negatively correlated with serum Angpt-2/Angpt-1 ratio. Multivariate Cox regression analysis showed the high serum Angpt-2/Angpt-1 ratio was an independent predictor of cardiovascular mortality (hazard ratio (HR)=2.467, 95% confidence interval (CI) 1.243-4.895, p = .010) and all-cause mortality (HR = 1.486, 95%CI 1.038-2.127, p = .031). In further subgroup analysis by gender, a significant association was shown in high Angpt-2/Angpt-1 ratio with all-cause mortality in male (p < .05), but not in female patients (p>.05). CONCLUSIONS High Angpt-2/Angpt-1 ratio is an independent risk factor for cardiovascular and all-cause mortality in PD patients.
Collapse
Affiliation(s)
- Han Li
- Department of Nephrology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Shanghai Center for Peritoneal Dialysis Research, Shanghai, China
| | - Qianhui Song
- Department of Nephrology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Shanghai Center for Peritoneal Dialysis Research, Shanghai, China
| | - Xinyu Su
- Department of Nephrology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Shanghai Center for Peritoneal Dialysis Research, Shanghai, China
| | - Yiwei Shen
- Department of Nephrology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Shanghai Center for Peritoneal Dialysis Research, Shanghai, China
| | - Hao Yan
- Department of Nephrology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Shanghai Center for Peritoneal Dialysis Research, Shanghai, China
| | - Zanzhe Yu
- Department of Nephrology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Shanghai Center for Peritoneal Dialysis Research, Shanghai, China
| | - Zhenyuan Li
- Department of Nephrology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Shanghai Center for Peritoneal Dialysis Research, Shanghai, China
| | - Jiangzi Yuan
- Department of Nephrology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Shanghai Center for Peritoneal Dialysis Research, Shanghai, China
| | - Jiaying Huang
- Department of Nephrology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Shanghai Center for Peritoneal Dialysis Research, Shanghai, China
| | - Zhaohui Ni
- Department of Nephrology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Shanghai Center for Peritoneal Dialysis Research, Shanghai, China
| | - Leyi Gu
- Department of Nephrology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Shanghai Center for Peritoneal Dialysis Research, Shanghai, China
| | - Wei Fang
- Department of Nephrology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Shanghai Center for Peritoneal Dialysis Research, Shanghai, China
| |
Collapse
|
2
|
Ishiyama A, Suda K, Rao X, Sun M, Lane GJ, Yamataka A, Koga H. Angiopoietin-1 attenuates lipopolysaccharide-induced endotoxemia in a Hirschsprung's disease murine model by improving intestinal vascular integrity: implications for treating postoperative Hirschsprung-associated enterocolitis. Pediatr Surg Int 2024; 40:277. [PMID: 39466437 DOI: 10.1007/s00383-024-05867-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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 10/18/2024] [Indexed: 10/30/2024]
Abstract
PURPOSE Angiopoietin-1 (Ang1) mitigates inflammation as a proangiogenic growth factor. Action of Ang1 on lipopolysaccharide (LPS)-induced endotoxemic inflammation was investigated in endothelin receptor-B null Hirschsprung's disease mice (KO). METHODS LPS or saline was injected intraperitoneally in KO (KO-LPS; n = 9, KO-sal; n = 5) and wild-type (WT) (WT-LPS; n = 6, WT-sal; n = 6) pups obtained within 24 h of birth. Normoganglionic terminal ileum harvested 6 h after LPS was used for RNA extraction and histology. IL-1β, SELE, VEGFA, Ang1, Angiopoietin-2 (Ang2), and TIE2 expression analyzed by quantitative polymerase chain reaction (qPCR), vascular permeability assessed by the Miles assay, severity of inflammation, and immunofluorescence for phospho-TIE2 and VE-cadherin were used to assess endothelial cell contact integrity and compared with KO pups pretreated with intraperitoneal Ang1 [Ang1(KO-LPS); n = 5] or saline [sal(KO-LPS); n = 6] 2 h before LPS. RESULTS KO-LPS pups showed significantly increased inflammation (p < 0.05) and expression of IL-1β, SELE, VEGFA, and Ang2 (p = 0.019, 0.003, 0.008 and < 0.0001, respectively); expression of Ang1 and TIE2 remained unchanged when compared with KO-saline. In Ang1(KO-LPS) ileum, changes seen in sal(KO-LPS) were eliminated and phospho-TIE2 and VE-cadherin fluorescence increased. CONCLUSION Ang1 successfully attenuated LPS-induced normoganglionic intestinal inflammation, downregulated pro-inflammatory genes, and improved vascular barrier integrity in KO pups.
Collapse
Affiliation(s)
- Asuka Ishiyama
- Department of Pediatric General and Urogenital Surgery, Juntendo University School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo, 113-8421, Japan
| | - Kazuto Suda
- Department of Pediatric General and Urogenital Surgery, Juntendo University School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo, 113-8421, Japan.
| | - Xuxuan Rao
- Department of Pediatric General and Urogenital Surgery, Juntendo University School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo, 113-8421, Japan
| | - Minhua Sun
- Department of Pediatric General and Urogenital Surgery, Juntendo University School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo, 113-8421, Japan
| | - Geoffrey J Lane
- Department of Pediatric General and Urogenital Surgery, Juntendo University School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo, 113-8421, Japan
| | - Atsuyuki Yamataka
- Department of Pediatric General and Urogenital Surgery, Juntendo University School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo, 113-8421, Japan
| | - Hiroyuki Koga
- Department of Pediatric General and Urogenital Surgery, Juntendo University School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo, 113-8421, Japan
| |
Collapse
|
3
|
Dodani K, Pinilla L, Sánchez-de-la-Torre M. Predictors and markers of the cardiovascular impact of obstructive sleep apnoea. Curr Opin Pulm Med 2024:00063198-990000000-00196. [PMID: 39189145 DOI: 10.1097/mcp.0000000000001118] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/28/2024]
Abstract
PURPOSE OF REVIEW Obstructive sleep apnoea (OSA) is the most common form of sleep-disordered breathing and has been linked to cardiovascular health. However, some of the findings supporting this are controversial. These discrepancies might be a result of heterogeneity among OSA patients, and thus, additional information would be required to better stratify OSA patients according to cardiovascular risk. In this review, we aim to discuss the potential of biomarkers to fulfil this role. RECENT FINDINGS Randomized controlled trials have been unable to confirm whether OSA treatment with continuous positive airway pressure (CPAP) has a positive effect on cardiovascular outcomes. Emerging physiology-based metrics of OSA seem to be more suitable for identifying patients at higher risk of cardiovascular disease and predicting the effects of CPAP outcomes on cardiovascular health. Similarly, blood-based molecular markers have gained attention in this context over the last few years. SUMMARY Accurate cardiovascular risk stratification and appropriate treatment allocation for OSA patients remain challenging. However, significant efforts are being made to develop novel tools to address these important issues.
Collapse
Affiliation(s)
- Krish Dodani
- Group of Precision Medicine in Chronic Diseases, University Hospital Arnau de Vilanova and Santa María, University of Lleida; Institut de Recerca Biomèdica de Lleida (IRBLleida), Lleida
| | - Lucía Pinilla
- Centro de Investigación Biomédica en Red de Enfermedades Respiratorias (CIBERES), Madrid, Spain
- Adelaide Institute for Sleep Health and FHMRI Sleep Health, College of Medicine and Public Health, Flinders University, Adelaide, Australia
| | - Manuel Sánchez-de-la-Torre
- Centro de Investigación Biomédica en Red de Enfermedades Respiratorias (CIBERES), Madrid, Spain
- Group of Precision Medicine in Chronic Diseases, Hospital Nacional de Parapléjicos, IDISCAM, Department of Nursing, Physiotherapy and Occupational Therapy, Faculty of Physiotherapy and Nursing, University of Castilla-La Mancha, Toledo, Spain
| |
Collapse
|
4
|
Lu F, Wu B, Dong L, Shu X, Wang Y. Pro-angiogenic cytokine features of left ventricular remodeling in patients with bicuspid aortic valve. Hellenic J Cardiol 2024:S1109-9666(24)00161-1. [PMID: 39038608 DOI: 10.1016/j.hjc.2024.07.006] [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: 12/16/2023] [Revised: 05/15/2024] [Accepted: 07/16/2024] [Indexed: 07/24/2024] Open
Abstract
OBJECTIVE Bicuspid aortic valve (BAV) is prone to promote left ventricular remodeling (LVR), which is associated with adverse clinical outcomes. Although the association between angiogenic activity and LVR has been established, pro-angiogenic cytokine features and potential biomarker candidates for LVR in patients with BAV remain to be clarified. METHODS From November 2018 to May 2019, patients with BAV diagnosed by transthoracic echocardiography at our institution were included. LVR was diagnosed on the basis of echocardiographic calculations of relative wall thickness (RWT) and left ventricular mass index (LVMI). A multiplex ELISA array was used to measure the plasma levels of 60 angiogenesis-related cytokines. RESULTS Among 103 patients with BAV, 71 were categorized into the LVR group and 32 into the normal left ventricular (LV) geometry group. BAV patients with LVR demonstrated increased LVMI, elevated prevalence of moderate to severe aortic stenosis and aortic regurgitation, and decreased LV ejection fraction (LVEF). Plasma levels of angiopoietin-1 were elevated in BAV patients with or without LVR compared with healthy controls (P = 0.001, P < 0.001, respectively), and were negatively correlated with RWT (r = -0.222, P = 0.027). Plasma levels of angiopoietin-2 were elevated in the LVR group (P = 0.001) compared with the normal LV geometry group, and were negatively correlated with LVEF (r = -0.330, P = 0.002). CONCLUSION Decreased angiogenesis plays a crucial role in the occurrence and progression of LVR in patients with BAV. Disturbance in the pro- and anti-angiogenesis equilibrium in BAV patients with LVR may reflect the aggravation of endothelial injury and dysfunction.
Collapse
Affiliation(s)
- Feiwei Lu
- Department of Echocardiography, Zhongshan Hospital, Fudan University, Shanghai, China; Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Boting Wu
- Department of Transfusion, Zhongshan Hospital, Fudan University, Shanghai, China.
| | - Lili Dong
- Department of Echocardiography, Zhongshan Hospital, Fudan University, Shanghai, China; Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Xianhong Shu
- Department of Echocardiography, Zhongshan Hospital, Fudan University, Shanghai, China; Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Yongshi Wang
- Department of Echocardiography, Zhongshan Hospital, Fudan University, Shanghai, China; Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai, China.
| |
Collapse
|
5
|
Alshawaf E, Abu-Farha M, Mohammad A, Devarajan S, Al-Khairi I, Cherian P, Ali H, Al-Matrouk H, Al-Mulla F, Al Attar A, Abubaker J. Angiopoietin-2 and Angiopoietin-like Proteins with a Prospective Role in Predicting Diabetic Nephropathy. Biomedicines 2024; 12:949. [PMID: 38790911 PMCID: PMC11118931 DOI: 10.3390/biomedicines12050949] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Revised: 04/14/2024] [Accepted: 04/20/2024] [Indexed: 05/26/2024] Open
Abstract
Angiopoietins are crucial growth factors for maintaining a healthy, functional endothelium. Patients with type 2 diabetes (T2D) exhibit significant levels of angiogenic markers, particularly Angiopoietin-2, which compromises endothelial integrity and is connected to symptoms of endothelial injury and failure. This report examines the levels of circulating angiopoietins in people with T2D and diabetic nephropathy (DN) and explores its link with ANGPTL proteins. We quantified circulating ANGPTL3, ANGPTL4, ANGPTL8, Ang1, and Ang2 in the fasting plasma of 117 Kuwaiti participants, of which 50 had T2D and 67 participants had DN. The Ang2 levels increased with DN (4.34 ± 0.32 ng/mL) compared with T2D (3.42 ± 0.29 ng/mL). This increase correlated with clinical parameters including the albumin-to-creatinine ratio (ACR) (r = 0.244, p = 0.047), eGFR (r = -0.282, p = 0.021), and SBP (r = -0.28, p = 0.024). Furthermore, Ang2 correlated positively to both ANGPTL4 (r = 0.541, p < 0.001) and ANGPTL8 (r = 0.41, p = 0.001). Multiple regression analysis presented elevated ANGPTL8 and ACRs as predictors for Ang2's increase in people with DN. In people with T2D, ANGPTL4 positively predicted an Ang2 increase. The area under the curve (AUC) in receiver operating characteristic (ROC) analysis of the combination of Ang2 and ANGPTL8 was 0.77 with 80.7% specificity. In conclusion, significantly elevated Ang2 in people with DN correlated with clinical markers such as the ACR, eGFR, and SBP, ANGPTL4, and ANGPTL8 levels. Collectively, this study highlights a close association between Ang2 and ANGPTL8 in a population with DN, suggesting them as DN risk predictors.
Collapse
Affiliation(s)
- Eman Alshawaf
- Department of Biochemistry and Molecular Biology, Dasman Diabetes Institute, Dasman 15462, Kuwait; (E.A.); (M.A.-F.); (A.M.); (I.A.-K.); (P.C.)
| | - Mohamed Abu-Farha
- Department of Biochemistry and Molecular Biology, Dasman Diabetes Institute, Dasman 15462, Kuwait; (E.A.); (M.A.-F.); (A.M.); (I.A.-K.); (P.C.)
- Diabetology Unit, Amiri Hospital, Dasman Diabetes Institute, Dasman 15462, Kuwait
| | - Anwar Mohammad
- Department of Biochemistry and Molecular Biology, Dasman Diabetes Institute, Dasman 15462, Kuwait; (E.A.); (M.A.-F.); (A.M.); (I.A.-K.); (P.C.)
| | - Sriraman Devarajan
- National Dasman Diabetes Biobank, Dasman Diabetes Institute, Dasman 15462, Kuwait;
| | - Irina Al-Khairi
- Department of Biochemistry and Molecular Biology, Dasman Diabetes Institute, Dasman 15462, Kuwait; (E.A.); (M.A.-F.); (A.M.); (I.A.-K.); (P.C.)
| | - Preethi Cherian
- Department of Biochemistry and Molecular Biology, Dasman Diabetes Institute, Dasman 15462, Kuwait; (E.A.); (M.A.-F.); (A.M.); (I.A.-K.); (P.C.)
| | - Hamad Ali
- Functional Genomic Unit, Dasman Diabetes Institute, Dasman 15462, Kuwait; (H.A.); (F.A.-M.)
- Department of Medical Laboratory Sciences, Faculty of Allied Health Sciences, Health Sciences Center, Kuwait University, Kuwait 15462, Kuwait
| | - Hawra Al-Matrouk
- Medical Department, Amiri Hospital, Ministry of Health, Kuwait 15462, Kuwait;
| | - Fahd Al-Mulla
- Functional Genomic Unit, Dasman Diabetes Institute, Dasman 15462, Kuwait; (H.A.); (F.A.-M.)
- Department of Translational Research, Dasman Diabetes Institute, Dasman 15462, Kuwait
| | - Abdulnabi Al Attar
- Diabetology Unit, Amiri Hospital, Dasman Diabetes Institute, Dasman 15462, Kuwait
| | - Jehad Abubaker
- National Dasman Diabetes Biobank, Dasman Diabetes Institute, Dasman 15462, Kuwait;
| |
Collapse
|
6
|
Mohebi R, Liu Y, Hansen MK, Yavin Y, Sattar N, Pollock CA, Butler J, Jardine M, Masson S, Heerspink HJ, Januzzi JL. Associations of Angiopoietin 2 and Vascular Endothelial Growth Factor-A Concentrations with Clinical End Points. Clin J Am Soc Nephrol 2024; 19:429-437. [PMID: 38099944 PMCID: PMC11020427 DOI: 10.2215/cjn.0000000000000389] [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: 08/28/2023] [Accepted: 12/11/2023] [Indexed: 01/09/2024]
Abstract
BACKGROUND Angiopoietin 2 regulates endothelial function partially mediated by vascular endothelial growth factor-A (VEGF-A) and may play a role in diabetic kidney disease (DKD). We assessed the association of angiopoietin 2 and VEGF-A with cardiorenal outcomes and investigated the effect of canagliflozin on angiopoietin 2 and VEGF-A concentrations. METHODS Two thousand five hundred sixty-five study participants with DKD and available plasma samples treated with canagliflozin or placebo in the Canagliflozin and Kidney Events in Diabetes with Established Nephropathy Clinical Evaluation (CREDENCE) trial were included. Angiopoietin 2 and VEGF-A concentrations were measured at baseline, year 1, and year 3. The primary composite end point of the trial was a composite of kidney failure, doubling of the serum creatinine level, and kidney or cardiovascular death. RESULTS Patients with the highest baseline quartile of angiopoietin 2, but not VEGF-A, concentration had the highest risk clinical profile. Treatment with canagliflozin significantly lowered concentrations of angiopoietin 2 (adjusted geometric mean ratio: 0.94; 95% confidence interval, 0.92 to 0.95; P < 0.001), but not VEGF-A. In multivariable-adjusted modeling, each 50% increment in log baseline angiopoietin 2 concentrations was associated with a higher risk of primary composite outcome (hazard ratio, 1.27; 95% confidence interval, 1.13 to 1.43). Angiopoietin 2 change at year 1 compared with baseline explained 10% of the effect of canagliflozin on the primary composite outcome. VEGF-A concentrations were not associated with outcomes, alone or in combination with angiopoietin 2. CONCLUSIONS Higher angiopoietin 2 levels were associated with cardiorenal risk among individuals with DKD independent of VEGF-A. Canagliflozin lowered angiopoietin 2 concentrations. CLINICAL TRIAL REGISTRY NAME AND REGISTRATION NUMBER Evaluation of the Effects of Canagliflozin on Renal and Cardiovascular Outcomes in Participants With Diabetic Nephropathy, NCT02065791 .
Collapse
Affiliation(s)
- Reza Mohebi
- Cardiology Division, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
| | - Yuxi Liu
- Cardiology Division, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
| | | | - Yshai Yavin
- Janssen Research & Development, LLC, Spring House, Pennsylvania
| | - Naveed Sattar
- BHF Glasgow Cardiovascular Research Centre, University of Glasgow, Glasgow, United Kingdom
| | - Carol A. Pollock
- Kolling Institute, Royal North Shore Hospital University of Sydney, Sydney, New South Wales, Australia
| | - Javed Butler
- Department of Medicine, University of Mississippi Medical Center, Jackson, Mississippi
- Baylor Scott & White Institute, Dallas, Texas
| | - Meg Jardine
- The George Institute for Global Health, UNSW Sydney, Sydney, New South Wales, Australia
- NHMRC Clinical Trials Centre, University of Sydney, Sydney, New South Wales, Australia
- Concord Repatriation General Hospital, Sydney, New South Wales, Australia
| | - Serge Masson
- Roche Diagnostics International, Rotkreuz, Switzerland
| | - Hiddo J.L. Heerspink
- Department Clinical Pharmacy and Pharmacology, University of Groningen, Groningen, The Netherlands
| | - James L. Januzzi
- Cardiology Division, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
- Heart Failure and Biomarker Trials, Baim Institute for Clinical Research, Boston, Massachusetts
| |
Collapse
|
7
|
Dattani A, Brady EM, Kanagala P, Stoma S, Parke KS, Marsh AM, Singh A, Arnold JR, Moss AJ, Zhao L, Cvijic ME, Fronheiser M, Du S, Costet P, Schafer P, Carayannopoulos L, Chang CP, Gordon D, Ramirez-Valle F, Jerosch-Herold M, Nelson CP, Squire IB, Ng LL, Gulsin GS, McCann GP. Is atrial fibrillation in HFpEF a distinct phenotype? Insights from multiparametric MRI and circulating biomarkers. BMC Cardiovasc Disord 2024; 24:94. [PMID: 38326736 PMCID: PMC10848361 DOI: 10.1186/s12872-024-03734-0] [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: 01/17/2024] [Indexed: 02/09/2024] Open
Abstract
BACKGROUND Heart failure with preserved ejection fraction (HFpEF) and atrial fibrillation (AF) frequently co-exist. There is a limited understanding on whether this coexistence is associated with distinct alterations in myocardial remodelling and mechanics. We aimed to determine if patients with atrial fibrillation (AF) and heart failure with preserved ejection fraction (HFpEF) represent a distinct phenotype. METHODS In this secondary analysis of adults with HFpEF (NCT03050593), participants were comprehensively phenotyped with stress cardiac MRI, echocardiography and plasma fibroinflammatory biomarkers, and were followed for the composite endpoint (HF hospitalisation or death) at a median of 8.5 years. Those with AF were compared to sinus rhythm (SR) and unsupervised cluster analysis was performed to explore possible phenotypes. RESULTS 136 subjects were included (SR = 75, AF = 61). The AF group was older (76 ± 8 vs. 70 ± 10 years) with less diabetes (36% vs. 61%) compared to the SR group and had higher left atrial (LA) volumes (61 ± 30 vs. 39 ± 15 mL/m2, p < 0.001), lower LA ejection fraction (EF) (31 ± 15 vs. 51 ± 12%, p < 0.001), worse left ventricular (LV) systolic function (LVEF 63 ± 8 vs. 68 ± 8%, p = 0.002; global longitudinal strain 13.6 ± 2.9 vs. 14.7 ± 2.4%, p = 0.003) but higher LV peak early diastolic strain rates (0.73 ± 0.28 vs. 0.53 ± 0.17 1/s, p < 0.001). The AF group had higher levels of syndecan-1, matrix metalloproteinase-2, proBNP, angiopoietin-2 and pentraxin-3, but lower level of interleukin-8. No difference in clinical outcomes was observed between the groups. Three distinct clusters were identified with the poorest outcomes (Log-rank p = 0.029) in cluster 2 (hypertensive and fibroinflammatory) which had equal representation of SR and AF. CONCLUSIONS Presence of AF in HFpEF is associated with cardiac structural and functional changes together with altered expression of several fibro-inflammatory biomarkers. Distinct phenotypes exist in HFpEF which may have differing clinical outcomes.
Collapse
Affiliation(s)
- Abhishek Dattani
- Department of Cardiovascular Sciences, University of Leicester and the National Institute for Health Research Leicester Biomedical Research Centre, Glenfield Hospital, Leicester, UK.
| | - Emer M Brady
- Department of Cardiovascular Sciences, University of Leicester and the National Institute for Health Research Leicester Biomedical Research Centre, Glenfield Hospital, Leicester, UK
| | | | - Svetlana Stoma
- Department of Cardiovascular Sciences, University of Leicester and the National Institute for Health Research Leicester Biomedical Research Centre, Glenfield Hospital, Leicester, UK
| | - Kelly S Parke
- Department of Cardiovascular Sciences, University of Leicester and the National Institute for Health Research Leicester Biomedical Research Centre, Glenfield Hospital, Leicester, UK
| | - Anna-Marie Marsh
- Department of Cardiovascular Sciences, University of Leicester and the National Institute for Health Research Leicester Biomedical Research Centre, Glenfield Hospital, Leicester, UK
| | - Anvesha Singh
- Department of Cardiovascular Sciences, University of Leicester and the National Institute for Health Research Leicester Biomedical Research Centre, Glenfield Hospital, Leicester, UK
| | - Jayanth R Arnold
- Department of Cardiovascular Sciences, University of Leicester and the National Institute for Health Research Leicester Biomedical Research Centre, Glenfield Hospital, Leicester, UK
| | - Alastair J Moss
- Department of Cardiovascular Sciences, University of Leicester and the National Institute for Health Research Leicester Biomedical Research Centre, Glenfield Hospital, Leicester, UK
| | - Lei Zhao
- Bristol Myers Squibb, Princeton, NJ, USA
| | | | | | - Shuyan Du
- Bristol Myers Squibb, Princeton, NJ, USA
| | | | | | | | | | | | | | | | - Christopher P Nelson
- Department of Cardiovascular Sciences, University of Leicester and the National Institute for Health Research Leicester Biomedical Research Centre, Glenfield Hospital, Leicester, UK
| | - Iain B Squire
- Department of Cardiovascular Sciences, University of Leicester and the National Institute for Health Research Leicester Biomedical Research Centre, Glenfield Hospital, Leicester, UK
| | - Leong L Ng
- Department of Cardiovascular Sciences, University of Leicester and the National Institute for Health Research Leicester Biomedical Research Centre, Glenfield Hospital, Leicester, UK
| | - Gaurav S Gulsin
- Department of Cardiovascular Sciences, University of Leicester and the National Institute for Health Research Leicester Biomedical Research Centre, Glenfield Hospital, Leicester, UK
| | - Gerry P McCann
- Department of Cardiovascular Sciences, University of Leicester and the National Institute for Health Research Leicester Biomedical Research Centre, Glenfield Hospital, Leicester, UK
| |
Collapse
|
8
|
Szymczak A, Kusztal M, Gołębiowski T, Letachowicz K, Goździk A, Kościelska-Kasprzak K, Tukiendorf A, Krajewska M. High Plasma Angiopoietin-2 Levels Predict the Need to Initiate Dialysis within Two Years in Patients with Chronic Kidney Disease. Int J Mol Sci 2023; 24:10036. [PMID: 37373181 DOI: 10.3390/ijms241210036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Revised: 06/04/2023] [Accepted: 06/05/2023] [Indexed: 06/29/2023] Open
Abstract
Volume status, congestion, endothelial activation, and injury all play roles in glomerular filtration rate (GFR) decline. In this study, we aimed to determine whether the plasma endothelial and overhydration markers could serve as independent predictors for dialysis initiation in patients with chronic kidney disease (CKD) 3b-5 (GFR < 45 mL/min/1.72 m2) and preserved ejection fraction. A prospective, observational study in a single academic center was conducted from March 2019 to March 2022. Plasma levels of angiopoietin (Ang)-2, Vascular Endothelial Growth Factor-C (VEGF-C), Vascular Cell Adhesion Molecule-1 (VCAM-1), Copeptin (CPP), beta-trace protein (BTP), brain natriuretic peptide (BNP), and cardiac troponin I (cTnI) were all measured. Lung ultrasound (US) B-lines, bioimpedance, and echocardiography with global longitudinal strain (GLS) were recorded. The study outcome was the initiation of chronic dialysis (renal replacement therapy) during 24 months of follow-up. A total of 105 consecutive patients with a mean eGFR of 21.3 mL/min/1.73 m were recruited and finally analyzed. A positive correlation between Ang-2 and VCAM-1 and BTP was observed. Ang-2 correlated positively with BNP, cTnI, sCr, E/e', and the extracellular water (ECW)/intracellular water (ICW) ratio (ECW/ICW). After 24 months, a deterioration in renal function was observed in 47 patients (58%). In multivariate regression analysis, both VCAM-1 and Ang-2 showed independent influences on risk of renal replacement therapy initiation. In a Kaplan-Meier analysis, 72% of patients with Ang-2 concentrations below the median (3.15 ng/mL) survived without dialysis for two years. Such an impact was not observed for GFR, VCAM, CCP, VEGF-C, or BTP. Endothelial activation, quantified by plasma levels of Ang-2, may play a key role in GFR decline and the need for dialysis initiation in patients with CKD 3b, 4, and 5.
Collapse
Affiliation(s)
- Anna Szymczak
- Department of Nephrology and Transplantation Medicine, Wroclaw Medical University, 50-556 Wroclaw, Poland
| | - Mariusz Kusztal
- Department of Nephrology and Transplantation Medicine, Wroclaw Medical University, 50-556 Wroclaw, Poland
| | - Tomasz Gołębiowski
- Department of Nephrology and Transplantation Medicine, Wroclaw Medical University, 50-556 Wroclaw, Poland
| | - Krzysztof Letachowicz
- Department of Nephrology and Transplantation Medicine, Wroclaw Medical University, 50-556 Wroclaw, Poland
| | - Anna Goździk
- Institute of Cardiology, Wroclaw Medical University, 50-556 Wroclaw, Poland
| | | | | | - Magdalena Krajewska
- Department of Nephrology and Transplantation Medicine, Wroclaw Medical University, 50-556 Wroclaw, Poland
| |
Collapse
|
9
|
Gregorczyk-Maga I, Szustkiewicz-Karoń A, Gajda M, Kapusta M, Maga W, Schönborn M. The Concentration of Pro- and Antiangiogenic Factors in Saliva and Gingival Crevicular Fluid Compared to Plasma in Patients with Peripheral Artery Disease and Type 2 Diabetes. Biomedicines 2023; 11:1596. [PMID: 37371691 DOI: 10.3390/biomedicines11061596] [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: 04/24/2023] [Revised: 05/25/2023] [Accepted: 05/29/2023] [Indexed: 06/29/2023] Open
Abstract
Several studies have investigated various biomarkers in relation to peripheral artery disease (PAD) for disease stratification and early-onset detection. In PAD, angiogenesis is required for tissue restoration and tissue perfusion. Considering changes in angiogenesis in patients with PAD, angiogenic factors could be explored as one of the new prognostic molecules. In recent studies, saliva and gingival crevicular fluid (GCF) have gained recognition as new, easily obtained diagnostic materials. This study aimed to compare the levels of selected circulating angiogenic factors (VEGF-A, PDGF-BB, and ANG-1) in unstimulated whole saliva (WS) and GCF versus plasma at three points in time to find possible correlations between their concentrations among patients with PAD and diabetes type 2 in 32 patients with Rutherford stages 5 and 6. A significant positive correlation has been demonstrated between circulating PDGF-BB levels in GCF and plasma. In most cases, comorbidities do not have an impact on the change in general correlation for the whole group. Our results clearly showed that GCF could be a good source for PDGF assessment. However, future studies with a larger number of subjects are warranted to confirm this finding and identify the most accurate angiogenic biomarkers in saliva or GCF that could be applied in clinical practice.
Collapse
Affiliation(s)
- Iwona Gregorczyk-Maga
- Faculty of Medicine, Institute of Dentistry, Jagiellonian University Medical College, 31-155 Krakow, Poland
| | | | - Mateusz Gajda
- Department of Angiology, Faculty of Medicine, Jagiellonian University Medical College, 31-121 Krakow, Poland
- Doctoral School of Medicine and Health Sciences, Jagiellonian University Medical College, 31-121 Krakow, Poland
| | - Maria Kapusta
- Department of Clinical Biochemistry, Jagiellonian University Medical College, 30-688 Krakow, Poland
| | - Wojciech Maga
- Faculty of Medicine, Institute of Dentistry, Jagiellonian University Medical College, 31-155 Krakow, Poland
| | - Martyna Schönborn
- Department of Angiology, Faculty of Medicine, Jagiellonian University Medical College, 31-121 Krakow, Poland
- Doctoral School of Medicine and Health Sciences, Jagiellonian University Medical College, 31-121 Krakow, Poland
| |
Collapse
|
10
|
Li M, Popovic Z, Chu C, Reichetzeder C, Pommer W, Krämer BK, Hocher B. Impact of Angiopoietin-2 on Kidney Diseases. KIDNEY DISEASES (BASEL, SWITZERLAND) 2023; 9:0. [PMID: 38306230 PMCID: PMC10826602 DOI: 10.1159/000529774] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/08/2022] [Accepted: 02/14/2023] [Indexed: 02/04/2024]
Abstract
Background Angiopoietins (Ang) are essential angiogenic factors involved in angiogenesis, vascular maturation, and inflammation. The most studied angiopoietins, angiopoietin-1 (Ang-1) and angiopoietin-2 (Ang-2), behave antagonistically to each other in vivo to sustain vascular endothelium homeostasis. While Ang-1 typically acts as the endothelium-protective mediator, its context-dependent antagonist Ang-2 can promote endothelium permeability and vascular destabilization, hence contributing to a poor outcome in vascular diseases via endothelial injury, vascular dysfunction, and microinflammation. The pathogenesis of kidney diseases is associated with endothelial dysfunction and chronic inflammation in renal diseases. Summary Several preclinical studies report overexpression of Ang-2 in renal tissues of certain kidney disease models; additionally, clinical studies show increased levels of circulating Ang-2 in the course of chronic kidney disease, implying that Ang-2 may serve as a useful biomarker in these patients. However, the exact mechanisms of Ang-2 action in renal diseases remain unclear. Key Messages We summarized the recent findings on Ang-2 in kidney diseases, including preclinical studies and clinical studies, aiming to provide a systematic understanding of the role of Ang-2 in these diseases.
Collapse
Affiliation(s)
- Mei Li
- Fifth Department of Medicine (Nephrology/Endocrinology/Rheumatology), University Medical Centre Mannheim, University of Heidelberg, Heidelberg, Germany
| | - Zoran Popovic
- Institute of Pathology, University Medical Centre Mannheim, University of Heidelberg, Heidelberg, Germany
| | - Chang Chu
- Fifth Department of Medicine (Nephrology/Endocrinology/Rheumatology), University Medical Centre Mannheim, University of Heidelberg, Heidelberg, Germany
- Department of Nephrology, Charité, Universitätsmedizin Berlin, Berlin, Germany
| | | | - Wolfgang Pommer
- Charité University Hospital Department of Nephrology and Internal Intensive Care Medicine, Berlin, Germany
| | - Bernhard K. Krämer
- Fifth Department of Medicine (Nephrology/Endocrinology/Rheumatology), University Medical Centre Mannheim, University of Heidelberg, Heidelberg, Germany
- European Center for Angioscience, Medical Faculty Mannheim of the University of Heidelberg, Mannheim, Germany
- Center for Innate Immunoscience, Medical Faculty Mannheim of the University of Heidelberg, Mannheim, Germany
| | - Berthold Hocher
- Fifth Department of Medicine (Nephrology/Endocrinology/Rheumatology), University Medical Centre Mannheim, University of Heidelberg, Heidelberg, Germany
- Reproductive and Genetic Hospital of CITIC-Xiangya, Changsha, China
- Institute of Medical Diagnostics, IMD Berlin, Berlin, Germany
| |
Collapse
|
11
|
Effects of Chronic Kidney Disease on Nanomechanics of the Endothelial Glycocalyx Are Mediated by the Mineralocorticoid Receptor. Int J Mol Sci 2022; 23:ijms231810659. [PMID: 36142571 PMCID: PMC9503126 DOI: 10.3390/ijms231810659] [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: 07/15/2022] [Revised: 08/31/2022] [Accepted: 09/03/2022] [Indexed: 02/03/2023] Open
Abstract
Endothelial mechanics control vascular reactivity and are regulated by the mineralocorticoid receptor (MR) and its downstream target, the epithelial Na+ channel (ENaC). Endothelial dysfunction is a hallmark of chronic kidney disease (CKD), but its mechanisms are poorly understood. We hypothesized that CKD disrupts endothelial mechanics in an MR/ENaC-dependent process. METHODS Primary human endothelial cells were cultured with uremic serum derived from children with stage 3-5 (predialysis) CKD or adult hemodialysis (HD) patients or healthy controls. The height and stiffness of the endothelial glycocalyx (eGC) and cortex were monitored by atomic force microscopy (AFM) using an ultrasensitive mechanical nanosensor. RESULTS In a stage-dependent manner, sera from children with CKD induced a significant increase in eGC and cortex stiffness and an incremental reduction of the eGC height. AFM measurements were significantly associated with individual pulse wave velocity and serum concentrations of gut-derived uremic toxins. Serum from HD patients increased MR expression and mechanical stiffness of the endothelial cortex, an effect reversed by MR and ENaC antagonists, decreased eNOS expression and NO bioavailability, and augmented monocyte adhesion. CONCLUSION These data indicate progressive structural damage of the endothelial surface with diminishing kidney function and identify the MR as a mediator of CKD-induced endothelial dysfunction.
Collapse
|
12
|
Cholecalciferol supplementation and angiogenic markers in chronic kidney disease. PLoS One 2022; 17:e0268946. [PMID: 35657784 PMCID: PMC9165782 DOI: 10.1371/journal.pone.0268946] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2021] [Accepted: 05/11/2022] [Indexed: 11/19/2022] Open
Abstract
Vitamin D plays an important role in proliferation and differentiation of cells and deficiency of vitamin D disturbs angiogenic balance. Previous studies in animal models have reported an association between serum levels of vitamin D and balance between pro- and anti-angiogenic factors. There is insufficient evidence about the effect of vitamin D on mediators of angiogenesis in patients with CKD. We investigated the effect of cholecalciferol supplementation on serum levels of angiogenic markers in non-diabetic patients with CKD stage 3–4. In this secondary analysis on stored samples of our previously published randomized, double-blind, placebo-controlled trial, stable patients of either sex, aged 18–70 years, with non-diabetic CKD stage 3–4 and vitamin D deficiency (serum 25-hydroxyvitamin D ≤20 ng/ml) were randomized to receive either two directly observed oral doses of cholecalciferol (300,000 IU) or matching placebo at baseline and 8 weeks. The primary outcome was change in brachial artery flow-mediated dilatation at 16 weeks. Changes in levels of serum angiogenesis markers (angiopoietin-1, angiopoietin-2, VEGF-A, VEGEF-R, and Tie-2) between groups over 16 weeks were compared. A total 120 patients were enrolled. Supplementation with cholecalciferol led to significant improvement in FMD. Serum 25(OH)D levels were similar in both groups at baseline (13.21±4.78 ng/ml and 13.40±4.42 ng/ml; p = 0.888). At 16 weeks, the serum 25(OH)D levels increased in the cholecalciferol group but not in the placebo group (between-group difference in mean change:23.40 ng/ml; 95% CI, 19.76 to 27.06; p<0.001). Serum levels of angiogenic markers were similar at baseline. At 16 weeks, angiopoietin-2 level decreased in cholecalciferol group (mean difference:-0.73 ng/ml, 95%CI, -1.25 to -0.20, p = 0.002) but not in placebo group (mean difference -0.46 ng/ml, 95%CI, -1.09 to 0.17, p = 0.154), however there was no between-group difference at 16 weeks (between-group difference in mean change: -0.27 ng/ml, 95%CI, -1.09 to 0.55, p = 0.624). Serum angiopoietin-1 level increased [mean change: 5.63 (0.51 to 10.75), p = 0.018] and VEGF-R level decreased [mean change: -87.16 (-131.89 to -42.44), p<0.001] in placebo group but did not show any change in cholecalciferol group. Our data shows the changes in Ang-1, Ang-2 and Ang-1/Ang-2 ratio after high dose oral cholecalciferol supplementation in patients with non-diabetic G3-4 CKD. The data suggests changes in circulating levels of angiogenic markers which needs to be confirmed through an adequately powered study.
Collapse
|
13
|
Hsu PC, Huang JC, Tsai WC, Hung WW, Chang WA, Wu LY, Chang CY, Tsai YC, Hsu YL. Tumor Necrosis Factor Receptor Superfamily Member 21 Induces Endothelial-Mesenchymal Transition in Coronary Artery Endothelium of Type 2 Diabetes Mellitus. Biomedicines 2022; 10:biomedicines10061282. [PMID: 35740304 PMCID: PMC9220259 DOI: 10.3390/biomedicines10061282] [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: 04/05/2022] [Revised: 05/24/2022] [Accepted: 05/27/2022] [Indexed: 12/02/2022] Open
Abstract
Diabetes mellitus (DM) is an increasing threat to human health and regarded as an important public issue. Coronary artery disease is one of the main causes of death in type 2 DM patients. However, the effect of hyperglycemia on coronary artery endothelial cells (CAECs) and the pathophysiologic mechanisms are still not well-explored. This study aims to explore the signal pathway and novel biomarkers of injury of CAECs in DM in understanding the microenvironment changes and mechanisms of diabetic heart disease. Next-generation sequence (NGS) and bioinformatics analysis to analyze the CAECs of one type 2 DM patient and one normal individual was performed, and it was found that tumor necrosis factor receptor superfamily member 21 (TNFRSF21) was a soluble factor in circulating system. Further experiments confirmed that advanced glycation end products (AGEs), the metabolite derived by hyperglycemia, increased the expression of TNFRSF21 in CAECs. TNFRSF21 induced endothelial–mesenchymal transition (EndoMT) in CAECs, resulting in increased permeability of CAECs. In addition, levels of serum TNFRSF21 were higher in type 2 DM patients with left ventricular hypertrophy (LVH) than those without LVH. Serum TNFRSF21 levels were also positively correlated with the LV mass index and negatively with LV systolic function. Serum TNFRSF21 levels were associated with changes in cardiac structure and function in patients with type 2 DM. In conclusion, TNFRSF21 plays a pathogenic role in heart disease of type 2 DM, and can be used as a biomarker of the impairment of cardiac structure and function in type 2 DM patients.
Collapse
Affiliation(s)
- Po-Chao Hsu
- Division of Cardiology, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 807, Taiwan; (P.-C.H.); (W.-C.T.)
- School of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan; (J.-C.H.); (W.-A.C.)
| | - Jiun-Chi Huang
- School of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan; (J.-C.H.); (W.-A.C.)
- Division of Nephrology, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 807, Taiwan
| | - Wei-Chung Tsai
- Division of Cardiology, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 807, Taiwan; (P.-C.H.); (W.-C.T.)
- School of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan; (J.-C.H.); (W.-A.C.)
| | - Wei-Wen Hung
- Division of Endocrinology and Metabolism, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 807, Taiwan;
| | - Wei-An Chang
- School of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan; (J.-C.H.); (W.-A.C.)
- Division of Pulmonary and Critical Care Medicine, Kaohsiung Medical University Hospital, Medical University, Kaohsiung 807, Taiwan
| | - Ling-Yu Wu
- Graduate Institute of Clinical Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan;
| | - Chao-Yuan Chang
- Department of Anatomy, Kaohsiung Medical University, Kaohsiung 807, Taiwan;
| | - Yi-Chun Tsai
- School of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan; (J.-C.H.); (W.-A.C.)
- Division of Nephrology, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 807, Taiwan
- Division of General Medicine, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 807, Taiwan
- Liquid Biopsy and Cohort Research, Kaohsiung Medical University, Kaohsiung 807, Taiwan
- Drug Development and Value Creation Research Center, Kaohsiung Medical University, Kaohsiung 807, Taiwan
- Correspondence: (Y.-C.T.); (Y.-L.H.); Tel.: +886-7-312-1101-5029 (Y.-C.T.); +886-7-312-1101-2015 (Y.-L.H.)
| | - Ya-Ling Hsu
- Drug Development and Value Creation Research Center, Kaohsiung Medical University, Kaohsiung 807, Taiwan
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan
- Correspondence: (Y.-C.T.); (Y.-L.H.); Tel.: +886-7-312-1101-5029 (Y.-C.T.); +886-7-312-1101-2015 (Y.-L.H.)
| |
Collapse
|
14
|
Zhong Z, Li HY, Zhong H, Lin W, Lin S, Zhou T. All-trans retinoic acid regulating angiopoietins-1 and alleviating extracellular matrix accumulation in interstitial fibrosis rats. Ren Fail 2021; 43:658-663. [PMID: 33820492 PMCID: PMC8032328 DOI: 10.1080/0886022x.2021.1910046] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2020] [Revised: 03/02/2021] [Accepted: 03/11/2021] [Indexed: 02/08/2023] Open
Abstract
All-trans retinoic acid (ATRA) is one of essentially active metabolite of vitamin A, and plays an important role in diverse physiological processes, such as cellular growth and function. Renal interstitial fibrosis (RIF) is a common pathological characteristic of chronic renal disease causing end-stage renal disease currently lacking effective treatment. Low level of Angiopoietins-1 (Angpt-1) is associated with extracellular matrix accumulation and fibrosis diseases. This study was performed to assess the association of ATRA with Angpt-1 in RIF disease. Rats were divided into three groups: group of sham (SHO group), group of unilateral ureteral obstruction group (UUO group), UUO mice administrated daily at the dose of ATRA (ATRA group). Masson-staining was used to detect the histologic lesion. Immunohistochemistry and Western-blot were applied to determine the targeted proteins. RIF score was significantly increased in UUO rats when compared with that of SHO group, and the fibrosis score was notably reduced in ATRA group. Transforming growth factor-β1 (TGF-β1), collagen IV (Col-IV) and fibronectin (FN) expressions in UUO group were significantly up-regulated, whereas Angpt-1 expression was significantly down-regulated compared with the SHO group. ATRA treatment reduced TGF-β1, Col-IV and FN expressions and improved Angpt-1 expression compared with the UUO group. The protein expression of Angpt-1 in kidney tissue of UUO group was negatively correlated with RIF index and protein expressions of Col-IV, FN and TGF-β1. In conclusion, low expression of Angpt-1 was associated with the RIF disease and ATRA treatment can increase the Angpt-1 and alleviate the RIF lesion in UUO rats.
Collapse
Affiliation(s)
- Zhiqing Zhong
- Department of Nephrology, the Second Affiliated Hospital, Shantou University Medical College, Shantou, China
| | - Hong-Yan Li
- Department of Nephrology, Huadu District People's Hospital of Guangzhou, Southern Medical University, Guangzhou, China
| | - Hongzhen Zhong
- Department of Nephrology, the Second Affiliated Hospital, Shantou University Medical College, Shantou, China
| | - Wenshan Lin
- Department of Nephrology, the Second Affiliated Hospital, Shantou University Medical College, Shantou, China
| | - Shujun Lin
- Department of Nephrology, the Second Affiliated Hospital, Shantou University Medical College, Shantou, China
| | - Tianbiao Zhou
- Department of Nephrology, the Second Affiliated Hospital, Shantou University Medical College, Shantou, China
- CONTACT Tianbiao Zhou Department of Nephrology, the Second Affiliated Hospital, Shantou University Medical College, No. 69 Dongxia Road, Shantou, 515041, China
| |
Collapse
|
15
|
Chu C, Chen X, Hasan AA, Szakallova A, Krämer BK, Tepel M, Hocher B. Angiopoietin-2 predicts all-cause mortality in male but not female end-stage kidney disease patients on hemodialysis. Nephrol Dial Transplant 2021; 37:1348-1356. [PMID: 34792167 PMCID: PMC9217660 DOI: 10.1093/ndt/gfab332] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2021] [Indexed: 11/29/2022] Open
Abstract
Background Angiopoietin-2 (Ang-2) plays a pivotal role in pathological vascular remodeling and angiogenesis. Both vascular mechanisms are active in patients with end-stage renal disease (ESRD) and may contribute to the high mortality in these patients. The aim of this multicenter prospective cohort study was to investigate baseline serum Ang-2 concentrations in ESRD patients on hemodialysis (HD) for their ability to predict all-cause mortality. Methods We conducted a prospective cohort study in 340 stable HD patients from different chronic dialysis centers in Berlin, Germany. The primary endpoint was all-cause mortality during a 5-year follow-up period. Blood samples and clinical data were collected at baseline. Serum Ang-2 was measured with a validated enzyme-linked immunosorbent assay (Biomedica, Vienna, Austria). Results A total of 313 HD patients (206 men and 107 women) were finally included in the study. Receiver operating characteristic (ROC) analysis of Ang-2 concentrations yielded an area under the curve (AUC) of 0.65 (P < 0.0001) for predicting all-cause mortality in the entire study population and was used to determine the optimal cut-off (111.0 pmol/L) for all-cause mortality. Kaplan–Meier survival analysis indicated that male but not female end-stage kidney disease patients on HD with higher Ang-2 concentrations had a significantly lower survival (log-rank test, P < 0.0001 and P = 0.380 for male and female patients, respectively). Multivariable Cox regression analyses adjusted for age, comorbidity, smoking, dialysis vintage, serum creatinine, hemoglobin, C-reactive protein, serum albumin, intact parathyroid hormone (iPTH), low-density lipoprotein (LDL) and Kt/V likewise indicated that elevated Ang-2 concentrations are associated with all-cause mortality in male {hazard ratio [HR] 3.294 [95% confidence interval (CI) 1.768–6.138]; P = 0.0002} but not in female end-stage kidney disease patients on HD [HR 1.084 (95% CI 0.476–2.467); P = 0.847]. Conclusion Ang-2 at baseline is independently associated with all-cause mortality in male ESRD patients on HD.
Collapse
Affiliation(s)
- Chang Chu
- Fifth Department of Medicine (Nephrology/Endocrinology/Rheumatology), University Medical Centre Mannheim, University of Heidelberg, Mannheim, Germany.,Department of Nephrology, Charité - Universitätsmedizin Berlin, Campus Mitte, Berlin, Germany
| | - Xin Chen
- Fifth Department of Medicine (Nephrology/Endocrinology/Rheumatology), University Medical Centre Mannheim, University of Heidelberg, Mannheim, Germany.,Department of Nephrology, Charité - Universitätsmedizin Berlin, Campus Mitte, Berlin, Germany
| | - Ahmed A Hasan
- Fifth Department of Medicine (Nephrology/Endocrinology/Rheumatology), University Medical Centre Mannheim, University of Heidelberg, Mannheim, Germany
| | | | - Bernhard K Krämer
- Fifth Department of Medicine (Nephrology/Endocrinology/Rheumatology), University Medical Centre Mannheim, University of Heidelberg, Mannheim, Germany.,European Center for Angioscience ECAS, Medical Faculty Mannheim of the University of Heidelberg, Mannheim, Germany
| | - Martin Tepel
- Department of Nephrology, Charité - Universitätsmedizin Berlin, Campus Mitte, Berlin, Germany.,Department of Nephrology, Odense University Hospital, Odense, Denmark
| | - Berthold Hocher
- Fifth Department of Medicine (Nephrology/Endocrinology/Rheumatology), University Medical Centre Mannheim, University of Heidelberg, Mannheim, Germany.,Institute of Medical Diagnostics, IMD Berlin-Potsdam, Berlin, Germany.,Key Laboratory of Study and Discovery of Small Targeted Molecules of Hunan Province, School of Medicine, Hunan Normal University, Changsha, China.,Reproductive and Genetic Hospital of CITIC-Xiangya, Changsha, China
| |
Collapse
|
16
|
Thakkar AB, Ma Y, Dela Cruz M, Wu Y, Arechiga V, Swaminathan S, Ganz P, Wu AHB, Scherzer R, Deeks S, Hsue PY. Effect of HIV-1 Infection on Angiopoietin 1 and 2 Levels and Measures of Microvascular and Macrovascular Endothelial Dysfunction. J Am Heart Assoc 2021; 10:e021397. [PMID: 34726064 PMCID: PMC8751943 DOI: 10.1161/jaha.121.021397] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Background Individuals infected with HIV have an increased risk of developing cardiovascular disease; yet, the underlying mechanisms remain unknown. Recent evidence has implicated the Tie-2 tyrosine kinase receptor system and its associated ligands ANG1 (angiopoietin 1) and ANG2 (angiopoietin 2) in maintaining vascular homeostasis. In the general population, lower ANG1 levels and higher ANG2 levels are strongly correlated with the development of cardiovascular disease. In this study, we aim to investigate the associations of HIV infection with angiopoietin levels and endothelial dysfunction. Methods and Results In this cross-sectional study, we compared measures of ANG1, ANG2, and endothelial dysfunction using flow-mediated vasodilation of the brachial artery in 39 untreated subjects infected with HIV, 47 treated subjects infected with HIV, and 46 uninfected subjects from the SCOPE (Observational Study of the Consequences of the Protease Inhibitor Era) cohort. Compared with uninfected controls, treated individuals infected with HIV had 53.1% lower mean ANG1 levels (P<0.01) and similar ANG2 levels. On the other hand, untreated individuals infected with HIV had similar ANG1 levels, and 29.2% had higher ANG2 levels (P<0.01) compared with uninfected controls. When compared with individuals with untreated HIV infection, those with treated HIV infection had 56% lower ANG1 levels (P<0.01) and 22% lower ANG2 levels (P<0.01).Both treated and untreated HIV infection were associated with significant impairment in hyperemic velocity, a key measure of microvascular dysfunction (median 61 versus 72 cm/s, P<0.01), compared with uninfected controls (median 73 cm/s). This difference persisted after adjustment for ANG1 and ANG2 levels. Interestingly, when compared with untreated individuals infected with HIV, treated individuals infected with HIV had worse hyperemic velocity (-12.35 cm/s, P=0.05). In contrast, HIV status, ANG1 levels, and ANG2 levels were not associated with macrovascular dysfunction as measured by flow-mediated dilatation and brachial artery diameter, 2 other measures of vascular homeostasis. Conclusions HIV infection affects the balance between levels of ANG1 and ANG2 and may disturb endothelial homeostasis through disruption of vascular homeostasis. Individuals with treated HIV had decreased ANG1 levels and similar ANG2 levels, whereas individuals with untreated HIV had similar ANG1 levels and increased ANG2 levels, suggesting that treatment status may alter the balance between ANG1 and ANG2. HIV also promotes endothelial dysfunction via impairment of microvascular dysfunction, independent of the Tie-2 receptor system; the finding of worse microvascular dysfunction in the setting of treated HIV infection may reflect the impact of viral persistence on the microvasculature or toxicities of specific antiretroviral regimens. Further research to clarify the mechanism of HIV-mediated endothelial dysfunction is necessary to advance treatment of cardiovascular complications of HIV infection.
Collapse
Affiliation(s)
- Anjali B Thakkar
- Division of Cardiology Department of Medicine Zuckerberg San Francisco General HospitalUniversity of California San Francisco San Francisco CA
| | - Yifei Ma
- Division of Cardiology Department of Medicine Zuckerberg San Francisco General HospitalUniversity of California San Francisco San Francisco CA
| | - Mark Dela Cruz
- Section of Cardiology Department of Medicine University of Chicago Medical Center Chicago IL
| | - Yuaner Wu
- Division of Cardiology Department of Medicine Zuckerberg San Francisco General HospitalUniversity of California San Francisco San Francisco CA
| | - Victor Arechiga
- Division of Cardiology Department of Medicine Zuckerberg San Francisco General HospitalUniversity of California San Francisco San Francisco CA
| | - Shreya Swaminathan
- Division of Cardiology Department of Medicine Zuckerberg San Francisco General HospitalUniversity of California San Francisco San Francisco CA
| | - Peter Ganz
- Division of Cardiology Department of Medicine Zuckerberg San Francisco General HospitalUniversity of California San Francisco San Francisco CA
| | - Alan H B Wu
- Division of Clinical Chemistry Department of Laboratory Medicine Zuckerberg San Francisco General HospitalUniversity of California San Francisco San Francisco CA
| | - Rebecca Scherzer
- San Francisco Veteran's Affairs Medical Center San Francisco CA.,Department of Medicine University of California San Francisco CA
| | - Steven Deeks
- Positive Health Program Zuckerberg San Francisco General HospitalUniversity of California San Francisco CA
| | - Priscilla Y Hsue
- Division of Cardiology Department of Medicine Zuckerberg San Francisco General HospitalUniversity of California San Francisco San Francisco CA
| |
Collapse
|
17
|
Exosome-eluting stents for vascular healing after ischaemic injury. Nat Biomed Eng 2021; 5:1174-1188. [PMID: 33820981 PMCID: PMC8490494 DOI: 10.1038/s41551-021-00705-0] [Citation(s) in RCA: 97] [Impact Index Per Article: 32.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2019] [Accepted: 02/19/2021] [Indexed: 02/07/2023]
Abstract
Drug-eluting stents implanted after ischaemic injury reduce the proliferation of endothelial cells and vascular smooth muscle cells and thus neointimal hyperplasia. However, the eluted drug also slows down the re-endothelialization process, delays arterial healing and can increase the risk of late restenosis. Here we show that stents releasing exosomes derived from mesenchymal stem cells in the presence of reactive oxygen species enhance vascular healing in rats with renal ischaemia-reperfusion injury, promoting endothelial cell tube formation and proliferation, and impairing the migration of smooth muscle cells. Compared with drug-eluting stents and bare-metal stents, the exosome-coated stents accelerated re-endothelialization and decreased in-stent restenosis 28 days after implantation. We also show that exosome-eluting stents implanted in the abdominal aorta of rats with unilateral hindlimb ischaemia regulated macrophage polarization, reduced local vascular and systemic inflammation, and promoted muscle tissue repair.
Collapse
|
18
|
Ricciardi CA, Gnudi L. Vascular growth factors as potential new treatment in cardiorenal syndrome in diabetes. Eur J Clin Invest 2021; 51:e13579. [PMID: 33942293 DOI: 10.1111/eci.13579] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Revised: 04/11/2021] [Accepted: 04/21/2021] [Indexed: 02/06/2023]
Abstract
BACKGROUND Cardiorenal syndrome in diabetes is characterised by alterations of the cardiovascular system paralleled by kidney disease with progressive renal function decline. In diabetes, chronic metabolic and haemodynamic perturbations drive endothelial dysfunction, inflammation, oxidative stress and progressive tissue fibrosis which, in turn, lead to heart and renal anatomo-functional damage. In physiology, vascular growth factors have been implicated in vascular homeostasis; their imbalance, in disease setting such as diabetes, leads to vascular dysfunction and cardiorenal damage. AIMS To define the role of vascular growth factors and angiopoietins in cardiorenal syndrome. MATERIAL AND METHODS We will focus on the two most studied vascular growth factors, vascular endothelial growth factor (VEGF) and angiopoietins (Angpt). The balance and crosstalk between these growth factors are important in organ development and in the maintenance of a healthy vasculature, heart and kidney. The observed alterations in expression/function of these vascular growth factors, as seen in diabetes, are a protective response against external perturbations. RESULTS The chronic insults driving diabetes-mediated cardiorenal damage results in a paradoxical situation, whereby the vascular growth factors imbalance becomes a mechanism of disease. Studies have explored the possibility of modulating the expression/action of vascular growth factors to improve disease outcome. Experimental work has been conducted in animals and has been gradually translated in humans. DISCUSSION Difficulties have been encountered especially when considering the magnitude, timing and duration of interventions targeting a selective vascular growth factor. Targeting VEGF in cardiovascular disease has been challenging, while modulation of the Angpt system seems more promising. CONCLUSION Future studies will establish the translatability of therapies targeting vascular growth factors for heart and kidney disease in patients with diabetes.
Collapse
Affiliation(s)
- Carlo Alberto Ricciardi
- Section Vascular Biology and Inflammation, School of Cardiovascular Medicine & Sciences, British Heart Foundation Centre for Research Excellence, Faculty of Life Sciences & Medicine, King's College London, London, UK
| | - Luigi Gnudi
- Section Vascular Biology and Inflammation, School of Cardiovascular Medicine & Sciences, British Heart Foundation Centre for Research Excellence, Faculty of Life Sciences & Medicine, King's College London, London, UK
| |
Collapse
|
19
|
Tsai HJ, Tsai WC, Hung WC, Hung WW, Chang CC, Dai CY, Tsai YC. Gut Microbiota and Subclinical Cardiovascular Disease in Patients with Type 2 Diabetes Mellitus. Nutrients 2021; 13:nu13082679. [PMID: 34444839 PMCID: PMC8397936 DOI: 10.3390/nu13082679] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2021] [Revised: 07/29/2021] [Accepted: 07/30/2021] [Indexed: 01/14/2023] Open
Abstract
Type 2 diabetes (T2D) is associated with an increased risk of cardiovascular disease (CVD). The gut microbiota may contribute to the onset and progression of T2D and CVD. The aim of this study was to evaluate the relationship between the gut microbiota and subclinical CVD in T2D patients. This cross-sectional study used echocardiographic data to evaluate the cardiac structure and function in T2D patients. We used a quantitative polymerase chain reaction to measure the abundances of targeted fecal bacterial species that have been associated with T2D, including Bacteroidetes, Firmicutes, Clostridium leptum group, Faecalibacterium prausnitzii, Bacteroides, Bifidobacterium, Akkermansia muciniphila, and Escherichia coli. A total of 155 subjects were enrolled (mean age 62.9 ± 10.1 years; 57.4% male and 42.6% female). Phyla Bacteroidetes and Firmicutes and genera Bacteroides were positively correlated with the left ventricular ejection fraction. Low levels of phylum Firmicutes were associated with an increased risk of left ventricular hypertrophy. High levels of both phylum Bacteroidetes and genera Bacteroides were negatively associated with diastolic dysfunction. A high phylum Firmicutes/Bacteroidetes (F/B) ratio and low level of genera Bacteroides were correlated with an increased left atrial diameter. Phyla Firmicutes and Bacteroidetes, the F/B ratio, and the genera Bacteroides were associated with variations in the cardiac structure and systolic and diastolic dysfunction in T2D patients. These findings suggest that changes in the gut microbiome may be the potential marker of the development of subclinical CVD in T2D patients.
Collapse
Affiliation(s)
- Hui-Ju Tsai
- Department of Family Medicine, Kaohsiung Municipal Ta-Tung Hospital, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 801, Taiwan;
- Department of Family Medicine, School of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan
- Research Center for Environmental Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan
| | - Wei-Chung Tsai
- Division of Cardiology, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 807, Taiwan;
| | - Wei-Chun Hung
- Department of Microbiology and Immunology, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan; (W.-C.H.); (C.-C.C.)
| | - Wei-Wen Hung
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 807, Taiwan;
| | - Chen-Chia Chang
- Department of Microbiology and Immunology, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan; (W.-C.H.); (C.-C.C.)
| | - Chia-Yen Dai
- School of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan;
- Division of Hepatobiliary, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 807, Taiwan
| | - Yi-Chun Tsai
- School of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan;
- Division of General Medicine, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 807, Taiwan
- Liquid Biopsy and Cohort Research Center, Kaohsiung Medical University, Kaohsiung 807, Taiwan
- Correspondence: ; Tel.: +886-7-3121101-5029; Fax: +886-7-3122810
| |
Collapse
|
20
|
Glorieux G, Vanholder R, Van Biesen W, Pletinck A, Schepers E, Neirynck N, Speeckaert M, De Bacquer D, Verbeke F. Free p-cresyl sulfate shows the highest association with cardiovascular outcome in chronic kidney disease. Nephrol Dial Transplant 2021; 36:998-1005. [DOI: 10.1093/ndt/gfab004] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2020] [Indexed: 12/13/2022] Open
Abstract
Abstract
Background
Several protein-bound uraemic toxins (PBUTs) have been associated with cardiovascular (CV) and all-cause mortality in chronic kidney disease (CKD) but the degree to which this is the case per individual PBUT and the pathophysiological mechanism have only partially been unraveled.
Methods
We compared the prognostic value of both total and free concentrations of five PBUTs [p-cresyl sulfate (pCS), p-cresyl glucuronide, indoxyl sulfate, indole acetic acid and hippuric acid] in a cohort of 523 patients with non-dialysis CKD Stages G1–G5. Patients were followed prospectively for the occurrence of a fatal or non-fatal CV event as the primary endpoint and a number of other major complications as secondary endpoints. In addition, association with and the prognostic value of nine markers of endothelial activation/damage was compared.
Results
After a median follow-up of 5.5 years, 149 patients developed the primary endpoint. In multivariate Cox regression models adjusted for age, sex, systolic blood pressure, diabetes mellitus and estimated glomerular filtration rate, and corrected for multiple testing, only free pCS was associated with the primary endpoint {hazard ratio [HR]1.39 [95% confidence interval (CI) 1.14–1.71]; P = 0.0014}. Free pCS also correlated with a disintegrin and metalloproteinase with a thrombospondin type 1 motif, member 13 (r = −0.114, P < 0.05), angiopoietin-2 (ANGPT2) (r = 0.194, P < 0.001), matrix metallopeptidase 7 (MMP-7; (r = 0.238, P < 0.001) and syndecan 1 (r = 0.235, P < 0.001). Of these markers of endothelial activation/damage, ANGPT2 [HR 1.46 (95% CI 1.25–1.70); P < 0.0001] and MMP-7 [HR 1.31 (95% CI 1.08–1.59); P = 0.0056] were also predictive of the primary outcome.
Conclusions
Among PBUTs, free pCS shows the highest association with CV outcome in non-dialysed patients with CKD. Two markers of endothelial activation/damage that were significantly correlated with free pCS, ANGPT2 and MMP-7 were also associated with CV outcome. The hypothesis that free pCS exerts its CV toxic effects by an adverse effect on endothelial function deserves further exploration.
Collapse
Affiliation(s)
- Griet Glorieux
- Department of Internal Medicine and Pediatrics, Nephrology Section, Ghent University Hospital, Ghent, Belgium
| | - Raymond Vanholder
- Department of Internal Medicine and Pediatrics, Nephrology Section, Ghent University Hospital, Ghent, Belgium
| | - Wim Van Biesen
- Department of Internal Medicine and Pediatrics, Nephrology Section, Ghent University Hospital, Ghent, Belgium
| | - Anneleen Pletinck
- Department of Internal Medicine and Pediatrics, Nephrology Section, Ghent University Hospital, Ghent, Belgium
| | - Eva Schepers
- Department of Internal Medicine and Pediatrics, Nephrology Section, Ghent University Hospital, Ghent, Belgium
| | - Nathalie Neirynck
- Department of Internal Medicine and Pediatrics, Nephrology Section, Ghent University Hospital, Ghent, Belgium
| | - Marijn Speeckaert
- Department of Internal Medicine and Pediatrics, Nephrology Section, Ghent University Hospital, Ghent, Belgium
| | - Dirk De Bacquer
- Department of Public Health and Primary Care, Ghent University, Ghent, Belgium
| | - Francis Verbeke
- Department of Internal Medicine and Pediatrics, Nephrology Section, Ghent University Hospital, Ghent, Belgium
| |
Collapse
|
21
|
Nicolini G, Forini F, Kusmic C, Iervasi G, Balzan S. Angiopoietin 2 signal complexity in cardiovascular disease and cancer. Life Sci 2019; 239:117080. [PMID: 31756341 DOI: 10.1016/j.lfs.2019.117080] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2019] [Revised: 11/12/2019] [Accepted: 11/13/2019] [Indexed: 12/12/2022]
Abstract
The angiopoietin signal transduction system is a complex of vascular-specific kinase pathways that plays a crucial role in angiogenesis and maintenance of vascular homeostasis. Angiopoietin1 (Ang1) and 2 (Ang2), the ligand proteins of the pathway, belong to a family of glycoproteins that signal primarily through the transmembrane Tyrosine-kinase-2 receptor. Despite a considerable sequence homology, Ang1 and Ang2 manifest antagonistic effects in pathophysiological conditions. While Ang1 promotes the activation of survival pathways and the stabilization of the normal mature vessels, Ang2 can either favor vessel destabilization and leakage or promote abnormal EC proliferation in a context-dependent manner. Altered Ang1/Ang2 balance has been reported in various pathological conditions in association with inflammation and deregulated angiogenesis. In particular, increased Ang2 levels have been documented in human cancer and cardiovascular disease (CVD), including ischemic myocardial injury, heart failure and other cardiovascular complications secondary to diabetes, chronic renal damage and hypertension. Despite the obvious phenotypic differences, CVD and cancer share some common Ang2-dependent etiopathological mechanisms such as inflammation, epithelial (or endothelial) to mesenchymal transition, and adverse vascular network remodeling. Interestingly, both cancer and CVD are negatively affected by thyroid hormone dyshomeostasis. This review provides an overview of the complex Ang2-dependent signaling involved in CVD and cancer, as well as a survey of the related clinical literature. Moreover, on the basis of recent molecular acquisitions in an experimental model of post ischemic cardiac disease, the putative novel role of the thyroid hormone in the regulation of Ang1/Ang2 balance is also briefly discussed.
Collapse
Affiliation(s)
| | - Francesca Forini
- Institute of Clinical Physiology, CNR, Via G.Moruzzi 1, 56124 Pisa, Italy.
| | - Claudia Kusmic
- Institute of Clinical Physiology, CNR, Via G.Moruzzi 1, 56124 Pisa, Italy.
| | - Giorgio Iervasi
- Institute of Clinical Physiology, CNR, Via G.Moruzzi 1, 56124 Pisa, Italy.
| | - Silvana Balzan
- Institute of Clinical Physiology, CNR, Via G.Moruzzi 1, 56124 Pisa, Italy.
| |
Collapse
|
22
|
Ou X, Gao JH, He LH, Yu XH, Wang G, Zou J, Zhao ZW, Zhang DW, Zhou ZJ, Tang CK. Angiopoietin-1 aggravates atherosclerosis by inhibiting cholesterol efflux and promoting inflammatory response. Biochim Biophys Acta Mol Cell Biol Lipids 2019; 1865:158535. [PMID: 31678621 DOI: 10.1016/j.bbalip.2019.158535] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2019] [Revised: 10/21/2019] [Accepted: 10/25/2019] [Indexed: 01/14/2023]
Abstract
OBJECTIVE Angiopoietin-1 (Ang-1), a secreted protein, mainly regulates angiogenesis. Ang-1 has been shown to promote the development of atherosclerosis, whereas little is known about its effects on lipid metabolism and inflammation in this process. METHOD Ang-1 was transfected into ApoE-/- mice via lentiviral vector or incubated with THP-1 derived macrophages. Oil red O and HE staining were performed to measure the size of atherosclerotic plaques in ApoE-/- mice. Immunofluorescence was employed to show the expression of target proteins in aorta. [3H] labeled cholesterol was performed to examine the efficiency of cholesterol efflux and reverse cholesterol transport (RCT) both in vivo and vitro. Western blot and qPCR were used to quantify target proteins both in vivo and vitro. ELISA detected the levels of pro-inflammatory cytokines in mouse peritoneal macrophage. RESULTS Our data showed that Ang-1 augmented atherosclerotic plaques formation and inhibited cholesterol efflux. The binding of Ang-1 to Tie2 resulted in downregulation of LXRα, ABCA1 and ABCG1 expression via inhibiting the translocation of TFE3 into nucleus. In addition, Ang-1 decreased serum HDL-C levels and reduced reverse cholesterol transport (RCT) in ApoE-/- mice. Furthermore, Ang-1 induced lipid accumulation followed by increasing TNF-α, IL-6, IL-1β,and MCP-1 produced by MPMs, as well as inducing M1 phenotype macrophage marker iNOS and CD86 expression in aorta of ApoE-/- mice. CONCLUSION Ang-1 has an adverse effect on cholesterol efflux by decreasing the expression of ABCA1 and ABCG1 via Tie2/TFE3/LXRα pathway, thereby promoting inflammation and accelerating atherosclerosis progression.
Collapse
Affiliation(s)
- Xiang Ou
- Institute of Cardiovascular Disease, Key Laboratory for Arteriosclerology of Hunan Province, Medical Research Experiment Center, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, University of South China, Hengyang, Hunan 421001, China; Department of Endocrinology, The First Hospital of Changsha, Changsha, Hunan 410005, China
| | - Jia-Hui Gao
- Institute of Cardiovascular Disease, Key Laboratory for Arteriosclerology of Hunan Province, Medical Research Experiment Center, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, University of South China, Hengyang, Hunan 421001, China
| | - Lin-Hao He
- School of Pharmacy and Life Science College, Medical Research Center, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, University of South China, Hengyang, Hunan 421001, China
| | - Xiao-Hua Yu
- Institute of Cardiovascular Disease, Key Laboratory for Arteriosclerology of Hunan Province, Medical Research Experiment Center, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, University of South China, Hengyang, Hunan 421001, China
| | - Gang Wang
- Institute of Cardiovascular Disease, Key Laboratory for Arteriosclerology of Hunan Province, Medical Research Experiment Center, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, University of South China, Hengyang, Hunan 421001, China
| | - Jin Zou
- Institute of Cardiovascular Disease, Key Laboratory for Arteriosclerology of Hunan Province, Medical Research Experiment Center, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, University of South China, Hengyang, Hunan 421001, China
| | - Zhen-Wang Zhao
- Institute of Cardiovascular Disease, Key Laboratory for Arteriosclerology of Hunan Province, Medical Research Experiment Center, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, University of South China, Hengyang, Hunan 421001, China
| | - Da-Wei Zhang
- Department of Pediatrics and Group on the Molecular and Cell Biology of Lipids, University of Alberta, Edmonton, Alberta T6G 2S2, Canada
| | - Zhi-Jiao Zhou
- Department of Pathology, Third Xiangya Hospital, Central South University, Changsha, Hunan, China.
| | - Chao-Ke Tang
- Institute of Cardiovascular Disease, Key Laboratory for Arteriosclerology of Hunan Province, Medical Research Experiment Center, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, University of South China, Hengyang, Hunan 421001, China.
| |
Collapse
|
23
|
Scurt FG, Menne J, Brandt S, Bernhardt A, Mertens PR, Haller H, Chatzikyrkou C. Systemic Inflammation Precedes Microalbuminuria in Diabetes. Kidney Int Rep 2019; 4:1373-1386. [PMID: 31701047 PMCID: PMC6829192 DOI: 10.1016/j.ekir.2019.06.005] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2019] [Revised: 05/22/2019] [Accepted: 06/03/2019] [Indexed: 12/13/2022] Open
Abstract
Aim The aim of the case-control study was to investigate if serum biomarkers indicative of vascular inflammation and endothelial dysfunction can predict the development of microalbuminuria in patients with diabetes mellitus type 2. Methods Among participants enrolled in the ROADMAP (Randomized Olmesartan And Diabetes MicroAlbuminuria Prevention) and observational follow-up (OFU) studies, a panel of 15 serum biomarkers was quantified from samples obtained at initiation of the study and tested for associations with the development of new-onset microalbuminuria during follow-up. A case-control study was conducted with inclusion of 172 patients with microalbuminuria and 188 matched controls. Nonparametric inferential, nonlinear regression, mediation, and bootstrapping statistical methods were used for the analysis. Results The median follow-up time was 37 months. At baseline, mean concentrations of C-X-C motif chemokine ligand 16 (CXCL-16), transforming growth factor (TGF)–β1 and angiopoietin-2 were higher in patients with subsequent microalbuminuria. In the multivariate analysis, after adjustment for age, sex, body mass index, glycated hemoglobin, duration of diabetes, low-density lipoprotein (LDL), smoking status, blood pressure, baseline urine albumin-to-creatinine ratio (UACR), estimated glomerular filtration rate (eGFR), time of follow-up and cardiovascular disease, CXCL-16 (odds ratio [OR] 2.60, 95% confidence interval [CI] 1.71–3.96), angiopoietin-2 (OR 1.50, 95% CI 1.14–1.98) and TGF-β1 (OR 1.03, 95% CI 1.01–1.04) remained significant predictors of new-onset microalbuminuria (P < 0.001). Inclusion of these biomarkers in conventional clinical risk models for prediction of microalbuminuria increased the area under the curve (AUC) from 0.638 to 0.760 (P < 0.001). Conclusion In patients with type 2 diabetes, elevated plasma levels of CXCL-16, angiopoietin-2, and TGF-β1 are independently predictive of microalbuminuria. Thus, these serum markers improve renal risk models beyond established clinical risk factors.
Collapse
Affiliation(s)
- Florian G Scurt
- Clinic of Nephrology, Hypertension, Diabetes and Endocrinology, Health Campus Immunology, Infectiology, and Inflammation, Otto-von-Guericke University, Magdeburg, Germany
| | - Jan Menne
- Nephrology Section, Hanover Medical School, Hanover, Germany
| | - Sabine Brandt
- Clinic of Nephrology, Hypertension, Diabetes and Endocrinology, Health Campus Immunology, Infectiology, and Inflammation, Otto-von-Guericke University, Magdeburg, Germany
| | - Anja Bernhardt
- Clinic of Nephrology, Hypertension, Diabetes and Endocrinology, Health Campus Immunology, Infectiology, and Inflammation, Otto-von-Guericke University, Magdeburg, Germany
| | - Peter R Mertens
- Clinic of Nephrology, Hypertension, Diabetes and Endocrinology, Health Campus Immunology, Infectiology, and Inflammation, Otto-von-Guericke University, Magdeburg, Germany
| | - Hermann Haller
- Nephrology Section, Hanover Medical School, Hanover, Germany
| | - Christos Chatzikyrkou
- Clinic of Nephrology, Hypertension, Diabetes and Endocrinology, Health Campus Immunology, Infectiology, and Inflammation, Otto-von-Guericke University, Magdeburg, Germany.,Nephrology Section, Hanover Medical School, Hanover, Germany
| | | |
Collapse
|
24
|
Akwii RG, Sajib MS, Zahra FT, Mikelis CM. Role of Angiopoietin-2 in Vascular Physiology and Pathophysiology. Cells 2019; 8:cells8050471. [PMID: 31108880 PMCID: PMC6562915 DOI: 10.3390/cells8050471] [Citation(s) in RCA: 302] [Impact Index Per Article: 60.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2019] [Revised: 05/06/2019] [Accepted: 05/16/2019] [Indexed: 12/18/2022] Open
Abstract
Angiopoietins 1–4 (Ang1–4) represent an important family of growth factors, whose activities are mediated through the tyrosine kinase receptors, Tie1 and Tie2. The best characterized are angiopoietin-1 (Ang1) and angiopoietin-2 (Ang2). Ang1 is a potent angiogenic growth factor signaling through Tie2, whereas Ang2 was initially identified as a vascular disruptive agent with antagonistic activity through the same receptor. Recent data demonstrates that Ang2 has context-dependent agonist activities. Ang2 plays important roles in physiological processes and the deregulation of its expression is characteristic of several diseases. In this review, we summarize the activity of Ang2 on blood and lymphatic endothelial cells, its significance in human physiology and disease, and provide a current view of the molecular signaling pathways regulated by Ang2 in endothelial cells.
Collapse
Affiliation(s)
- Racheal G Akwii
- Department of Pharmaceutical Sciences, School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, TX 79106, USA.
| | - Md S Sajib
- Department of Pharmaceutical Sciences, School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, TX 79106, USA.
| | - Fatema T Zahra
- Department of Pharmaceutical Sciences, School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, TX 79106, USA.
| | - Constantinos M Mikelis
- Department of Pharmaceutical Sciences, School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, TX 79106, USA.
| |
Collapse
|
25
|
Asl ER, Rasmi Y, Khadem-Ansari MH, Seyed-Mohammadzad M, Rostamzadeh A, Ghaffari F, Mokarizadeh N. Increased levels of angiogenic factors in microvascular angina. Med Pharm Rep 2019; 92:31-35. [PMID: 30957084 PMCID: PMC6448487 DOI: 10.15386/cjmed-1101] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2018] [Revised: 09/16/2018] [Accepted: 09/27/2018] [Indexed: 11/23/2022] Open
Abstract
Background Recent studies have suggested that angiogenic factors may affect vascular endothelial integrity. On the other hand, endothelial dysfunction is the main pathological mechanism in microvascular angina (MVA) or cardiac syndrome X. Therefore, we aimed to determine the levels of angiogenic factors in MVA patients. In addition, we investigated the effects of metoprolol, as a beta blocker agent, on the serum levels of these factors. Methods Thirty patients with MVA (17 female/13 male; mean age: 55.53±9.18 years) and twenty healthy controls (14 female/6 male; mean age: 51.40±9.16 years) were enrolled. The serum amounts of angiopoietin-1 (Ang-1), angiopoietin-2 (Ang-2) and tyrosine kinase-2 receptor (Tie-2) were measured in healthy controls, MVA patients at baseline and after metoprolol therapy (25 mg for one month) by enzyme-linked immunosorbent assay. Results The levels of Ang-2 and Tie-2 were significantly higher in MVA patients at baseline in comparison with controls (Ang-2: 277.02±186.08 vs.164.46±49.83 ng/l, P=0.011; Tie-2: 28.97±18.85 vs. 14.90±4.05 ng/ml, P=0.002; respectively). But this difference in the Ang-1 levels was not significant (P=0.829). Additionally, the levels of angiogenic factors in MVA patients after metoprolol therapy were not significantly changed in comparison with the baseline status (P>0.05). Conclusion Our results considered a possible role for angiogenic factors in the pathophysiology of MVA, which need further investigation for elucidation. In addition, this study has not showed an effective role for metoprolol in changing the angiogenic factors levels as a therapeutic agent in MVA.
Collapse
Affiliation(s)
- Elmira Roshani Asl
- Department of Biochemistry, School of Medicine, Urmia University of Medical Sciences, Urmia, Iran
| | - Yousef Rasmi
- Department of Biochemistry, School of Medicine, Urmia University of Medical Sciences, Urmia, Iran.,Cellular and Molecular Research Center, Urmia University of Medical Sciences, Urmia, Iran
| | | | | | - Alireza Rostamzadeh
- Department of Cardiology, School of Medicine, Urmia University of Medical Sciences, Urmia, Iran
| | - Fereshteh Ghaffari
- Department of Biochemistry, School of Medicine, Urmia University of Medical Sciences, Urmia, Iran
| | - Narmin Mokarizadeh
- Department of Biochemistry, School of Medicine, Urmia University of Medical Sciences, Urmia, Iran
| |
Collapse
|
26
|
Jian W, Li L, Wei XM, Guan JH, Yang GL, Gui C. Serum angiopoietin-2 concentrations of post-PCI are correlated with the parameters of renal function in patients with coronary artery disease. Medicine (Baltimore) 2019; 98:e13960. [PMID: 30608432 PMCID: PMC6344115 DOI: 10.1097/md.0000000000013960] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
Patients with coronary artery disease (CAD) frequently have comorbidity of chronic kidney disease (CKD). Their renal function may deteriorate because of the use of contrast agent after percutaneous coronary intervention (PCI). Angiopoietin-2 (Ang-2), which is highly expressed in the site of angiogenesis, plays an important role in both CAD and CKD. This study aimed to investigate the relation of serum Ang-2 concentrations with the renal function after PCI.This study enrolled 57 patients with CAD undergoing PCI. Blood samples for Ang-2 were collected in the first morning after admission and within 24 to 48 h after PCI. The parameters of renal function (serum creatinine, cystatin C and eGFR) were tested on the first day after admission and within 72 h after PCI.Overall, serum Ang-2 levels of post-PCI were significantly lower than those of pre-PCI [median, 1733 (IQR, 1100-2568) vs median, 2523 (IQR, 1702-3640) pg/mL; P < .001]. However, in patients with CKD (eGFR < 60 mL/min/1.73 m), there was no significant difference between serum Ang-2 levels of post-PCI and those of pre-PCI [median, 2851 (IQR, 1720-4286) vs. median, 2492 (IQR, 1434-4994) pg/mL; P = .925]. In addition, serum Ang-2 levels of post-PCI, but not pre-PCI, were significantly correlated with the post-PCI parameters of renal function.Serum Ang-2 concentrations of post-PCI are closely related to renal function in patients with CAD. It may have potential to be the early biomarker of contrast-induced nephropathy in the future.
Collapse
Affiliation(s)
- Wen Jian
- Department of Cardiology, The First Affiliated Hospital of Guangxi Medical University
- Guangxi Key Laboratory Base of Precision Medicine in Cardio-Cerebrovascular Diseases Control and Prevention
- Guangxi Clinical Research Center for Cardio-Cerebrovascular Diseases, Nanning
| | - Lang Li
- Department of Cardiology, The First Affiliated Hospital of Guangxi Medical University
- Guangxi Key Laboratory Base of Precision Medicine in Cardio-Cerebrovascular Diseases Control and Prevention
- Guangxi Clinical Research Center for Cardio-Cerebrovascular Diseases, Nanning
| | - Xiao-Min Wei
- Department of Cardiology, Gongren Hospital of Wuzhou, Wuzhou
| | - Jia-Hui Guan
- Department of Respiratory Medicine, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, People's Republic of China
| | - Guo-Liang Yang
- Department of Cardiology, The First Affiliated Hospital of Guangxi Medical University
- Guangxi Key Laboratory Base of Precision Medicine in Cardio-Cerebrovascular Diseases Control and Prevention
- Guangxi Clinical Research Center for Cardio-Cerebrovascular Diseases, Nanning
| | - Chun Gui
- Department of Cardiology, The First Affiliated Hospital of Guangxi Medical University
- Guangxi Key Laboratory Base of Precision Medicine in Cardio-Cerebrovascular Diseases Control and Prevention
- Guangxi Clinical Research Center for Cardio-Cerebrovascular Diseases, Nanning
| |
Collapse
|
27
|
Yang X, Zhang H, Shi Y, Yu Z, Yan H, Ni Z, Qian J, Fang W. Association of serum angiopoietin-2 with malnutrition, inflammation, atherosclerosis and valvular calcification syndrome and outcome in peritoneal dialysis patients: a prospective cohort study. J Transl Med 2018; 16:312. [PMID: 30445969 PMCID: PMC6240212 DOI: 10.1186/s12967-018-1687-0] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2018] [Accepted: 11/09/2018] [Indexed: 01/20/2023] Open
Abstract
Background To examine serum angiopoietin-2 (Angpt-2) in relation to malnutrition, inflammation, atherosclerosis and cardiac valvular calcification, so-called MIAC syndrome and its predictive role in outcomes of peritoneal dialysis (PD) patients. Methods A prospective observational study was conducted in 324 chronic PD patients. Biochemical analysis was performed at baseline for serum angiopoietins, albumin and high sensitive C-reactive protein (hs-CRP) and echocardiography was done to detect cardiac valvular calcification. Primary study end points were fatal or nonfatal cardiovascular events and mortality. Results The median of serum Angpt-2 levels was 5.44 ng/mL (interquartile range, 3.41–7.85). Across the three tertiles of serum Angpt-2, a significant trend effect was observed for body mass index, normalized protein catabolic rate, calcium × phosphorus product, hs-CRP, brain natriuretic peptide, lower-density lipoprotein cholesterol, left ventricular ejection fraction, total weekly urea clearance and residual renal function (all p < 0.05). Serum Angpt-2 showed a significant increase across the four groups of patients with increasing components of MIAC syndrome (p < 0.001). There were 77 deaths and 57 cardiovascular events. High serum Angpt-2 was an independent predictor of fatal and nonfatal cardiovascular events in PD patients (p = 0.02), however serum Angpt-2 was not an independent predictor of all-cause mortality (p = 0.3). Conclusions Serum Angpt-2 showed close association with valvular calcification, atherosclerosis, inflammation and malnutrition, having significant independent prognostic value and is useful for cardiovascular event stratification in chronic PD patients. Angpt-2 might be a potential mediator of increased cardiovascular risk in patients undergoing PD treatment. Electronic supplementary material The online version of this article (10.1186/s12967-018-1687-0) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Xiaoxiao Yang
- Department of Nephrology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, No. 160, Pujian Road, Pudong District, Shanghai, 200127, People's Republic of China.,Shanghai Center for Peritoneal Dialysis Research, Shanghai, People's Republic of China
| | - He Zhang
- Department of Nephrology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, No. 160, Pujian Road, Pudong District, Shanghai, 200127, People's Republic of China.,Shanghai Center for Peritoneal Dialysis Research, Shanghai, People's Republic of China
| | - Yuanyuan Shi
- Department of Nephrology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, No. 160, Pujian Road, Pudong District, Shanghai, 200127, People's Republic of China.,Shanghai Center for Peritoneal Dialysis Research, Shanghai, People's Republic of China
| | - Zanzhe Yu
- Department of Nephrology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, No. 160, Pujian Road, Pudong District, Shanghai, 200127, People's Republic of China.,Shanghai Center for Peritoneal Dialysis Research, Shanghai, People's Republic of China
| | - Hao Yan
- Department of Nephrology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, No. 160, Pujian Road, Pudong District, Shanghai, 200127, People's Republic of China.,Shanghai Center for Peritoneal Dialysis Research, Shanghai, People's Republic of China
| | - Zhaohui Ni
- Department of Nephrology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, No. 160, Pujian Road, Pudong District, Shanghai, 200127, People's Republic of China.,Shanghai Center for Peritoneal Dialysis Research, Shanghai, People's Republic of China
| | - Jiaqi Qian
- Department of Nephrology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, No. 160, Pujian Road, Pudong District, Shanghai, 200127, People's Republic of China.,Shanghai Center for Peritoneal Dialysis Research, Shanghai, People's Republic of China
| | - Wei Fang
- Department of Nephrology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, No. 160, Pujian Road, Pudong District, Shanghai, 200127, People's Republic of China. .,Shanghai Center for Peritoneal Dialysis Research, Shanghai, People's Republic of China.
| |
Collapse
|
28
|
Anderson CE, Hamm LL, Batuman G, Kumbala DR, Chen CS, Kallu SG, Siriki R, Gadde S, Kleinpeter MA, Krane NK, Simon EE, He J, Chen J. The association of angiogenic factors and chronic kidney disease. BMC Nephrol 2018; 19:117. [PMID: 29783932 PMCID: PMC5963107 DOI: 10.1186/s12882-018-0909-2] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2017] [Accepted: 04/27/2018] [Indexed: 01/19/2023] Open
Abstract
BACKGROUND There are limited data on the associations of circulating angiogenic factors with chronic kidney disease (CKD). We investigate the associations of circulating vascular endothelial growth factor (VEGF)-A, angiopoietin-1, angiopoietin-1/VEGF-A ratio, VEGF receptor 1 (VEGFR-1), VEGFR-2, and pentraxin-3 with CKD. METHODS We recruited 201 patients with CKD and 201 community controls without CKD from the greater New Orleans area. CKD was defined as estimated glomerular filtration rate (eGFR) < 60 mL/min/1.73 m2 or presence of albuminuria. Multivariable quantile and logistic regression models were used to examine the relationship between angiogenesis-related factors and CKD adjusting for confounding factors. RESULTS After adjusting for covariables including traditional cardiovascular disease (CVD) risk factors, C-reactive protein, and history of CVD, the medians (interquartile range) were 133.08 (90.39, 204.15) in patients with CKD vs. 114.17 (72.45, 170.32) pg/mL in controls without CKD (p = 0.002 for group difference) for VEGF-A; 3951.2 (2471.9, 6656.6) vs. 4270.5 (2763.7, 6537.2) pg/mL (p = 0.70) for angiopoietin-1; 25.87 (18.09, 47.90) vs. 36.55 (25.71, 61.10) (p = 0.0001) for angiopoietin-1/VEGF-A ratio; 147.81 (122.94, 168.79) vs. 144.16 (123.74, 168.05) ng/mL (p = 0.25) for VEGFR-1; 26.20 (22.67, 29.92) vs. 26.28 (23.10, 29.69) ng/mL (p = 0.31) for VEGFR-2; and 1.01 (0.79, 1.49)vs. 0.89 (0.58, 1.18) ng/mL (p = 0.01) for pentraxin-3, respectively. In addition, an elevated VEGF-A level and decreased angiopoietin-1/VEGF-A ratio were associated with increased odds of CKD. CONCLUSIONS These data indicate that plasma VEGF-A and pentraxin-3 levels were increased and the angiopoietin-1/VEGF-A ratio was decreased in patients with CKD. Future prospective studies are warranted to examine whether angiogenic factors play a role in progression of CKD.
Collapse
Affiliation(s)
- Christopher E. Anderson
- 0000 0001 2217 8588grid.265219.bDepartment of Epidemiology, Tulane University School of Public Health and Tropical Medicine, 1440 Canal Street, room 1504, New Orleans, LA 70112 USA
| | - L. Lee Hamm
- 0000 0001 2217 8588grid.265219.bDepartment of Medicine, Tulane University School of Medicine, 1430 Tulane Avenue SL45, New Orleans, LA 70112 USA ,0000 0001 2217 8588grid.265219.bTulane Hypertension and Renal Center of Excellence, Tulane University School of Medicine, New Orleans, LA USA
| | - Gem Batuman
- 0000 0001 2217 8588grid.265219.bDepartment of Medicine, Tulane University School of Medicine, 1430 Tulane Avenue SL45, New Orleans, LA 70112 USA
| | - Damodar R. Kumbala
- 0000 0004 0608 1972grid.240416.5Department of Nephrology, Ochsner Health System, Ochsner Medical Center, 1514 Jefferson Highway, New Orleans, LA 70121 USA
| | - Chung-Shiuan Chen
- 0000 0001 2217 8588grid.265219.bDepartment of Epidemiology, Tulane University School of Public Health and Tropical Medicine, 1440 Canal Street, room 1504, New Orleans, LA 70112 USA
| | - Swapna G. Kallu
- 0000 0001 2217 8588grid.265219.bDepartment of Medicine, Tulane University School of Medicine, 1430 Tulane Avenue SL45, New Orleans, LA 70112 USA
| | - Ravi Siriki
- 0000 0001 2217 8588grid.265219.bDepartment of Medicine, Tulane University School of Medicine, 1430 Tulane Avenue SL45, New Orleans, LA 70112 USA
| | - Shilpa Gadde
- 0000 0001 2217 8588grid.265219.bDepartment of Medicine, Tulane University School of Medicine, 1430 Tulane Avenue SL45, New Orleans, LA 70112 USA
| | - Myra A. Kleinpeter
- 0000 0001 2217 8588grid.265219.bDepartment of Medicine, Tulane University School of Medicine, 1430 Tulane Avenue SL45, New Orleans, LA 70112 USA
| | - N. Kevin Krane
- 0000 0001 2217 8588grid.265219.bDepartment of Medicine, Tulane University School of Medicine, 1430 Tulane Avenue SL45, New Orleans, LA 70112 USA
| | - Eric E. Simon
- 0000 0001 2217 8588grid.265219.bDepartment of Medicine, Tulane University School of Medicine, 1430 Tulane Avenue SL45, New Orleans, LA 70112 USA
| | - Jiang He
- 0000 0001 2217 8588grid.265219.bDepartment of Epidemiology, Tulane University School of Public Health and Tropical Medicine, 1440 Canal Street, room 1504, New Orleans, LA 70112 USA ,0000 0001 2217 8588grid.265219.bDepartment of Medicine, Tulane University School of Medicine, 1430 Tulane Avenue SL45, New Orleans, LA 70112 USA ,0000 0001 2217 8588grid.265219.bTulane Hypertension and Renal Center of Excellence, Tulane University School of Medicine, New Orleans, LA USA
| | - Jing Chen
- 0000 0001 2217 8588grid.265219.bDepartment of Epidemiology, Tulane University School of Public Health and Tropical Medicine, 1440 Canal Street, room 1504, New Orleans, LA 70112 USA ,0000 0001 2217 8588grid.265219.bDepartment of Medicine, Tulane University School of Medicine, 1430 Tulane Avenue SL45, New Orleans, LA 70112 USA ,0000 0001 2217 8588grid.265219.bTulane Hypertension and Renal Center of Excellence, Tulane University School of Medicine, New Orleans, LA USA
| |
Collapse
|
29
|
Tsai YC, Lee CS, Chiu YW, Lee JJ, Lee SC, Hsu YL, Kuo MC. Angiopoietin-2, Renal Deterioration, Major Adverse Cardiovascular Events and All-Cause Mortality in Patients with Diabetic Nephropathy. Kidney Blood Press Res 2018; 43:545-554. [PMID: 29642068 DOI: 10.1159/000488826] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2018] [Accepted: 03/28/2018] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND/AIMS Diabetic nephropathy is the leading cause of end-stage renal disease and accounts for 30∼40% of patients requiring maintenance dialysis, thereby increasing the burden on health insurance programs. Diabetic nephropathy is also the strongest predictor of cardiovascular morbidity and mortality. The aim of this study was to examine whether angiopoietin-2 (Angpt2), a modulator of endothelial function, affects the clinical outcomes of diabetic patients. METHODS This study enrolled 236 patients with diabetes mellitus with estimated glomerular filtration rate (eGFR) < 60ml/min/1.73m2 from January 2006 to December 2011, who were followed until June 2017. Clinical outcomes included renal outcomes (commencing dialysis and rapid decline in renal function (eGFR decline > 3 ml/min per 1.73 m2/year)), major adverse cardiovascular events (MACEs), and all-cause mortality. RESULTS Over a mean follow-up period of 3.9±2.7 years, 135 (57.2%) patients commenced dialysis, 106 (44.9%) had rapid decline in renal function, and 50 (21.2%) had MACEs or died from all-causes. Log-formed Angpt2 was significantly associated with increased risks of commencing dialysis (HR: 3.91, 95% CI: 1.56-9.76), rapid renal function decline (OR: 6.81, 95% CI: 1.06-43.88), and MACEs or all-cause mortality (HR: 6.34, 95% CI: 1.18-33.97) in the adjusted analysis. Patients in the highest quartile had hazard ratios of 2.90 and 3.11 for commencing dialysis and rapid renal function decline, respectively, compared to those in the lowest quartile after adjustments. Similar significant dose-response results were found in composite outcomes of either MACEs or all-cause mortality. CONCLUSION Angpt2 is an independent predictor of adverse clinical outcomes in diabetic patients. Further studies are needed to identify the pathogenic role of Angpt2 in renal deterioration and cardiovascular complications of diabetes mellitus.
Collapse
Affiliation(s)
- Yi-Chun Tsai
- Graduate Institute of Clinical Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan.,Division of General Medicine, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan.,Division of Nephrology, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan.,Faculty of Renal Care, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Chee-Siong Lee
- Division of General Medicine, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan.,Division of Cardiology, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan
| | - Yi-Wen Chiu
- Division of Nephrology, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan.,Faculty of Renal Care, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Jia-Jung Lee
- Division of Nephrology, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan.,Faculty of Renal Care, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Su-Chu Lee
- Division of Nephrology, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan
| | - Ya-Ling Hsu
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Mei-Chuan Kuo
- Graduate Institute of Clinical Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan.,Division of Nephrology, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan.,Faculty of Renal Care, Kaohsiung Medical University, Kaohsiung, Taiwan
| |
Collapse
|
30
|
Campos NG, Marizeiro DF, Florêncio ACL, Silva ÍC, Meneses GC, Bezerra GF, Martins AMC, Libório AB. Effects of respiratory muscle training on endothelium and oxidative stress biomarkers in hemodialysis patients: A randomized clinical trial. Respir Med 2018; 134:103-109. [DOI: 10.1016/j.rmed.2017.12.005] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/25/2017] [Revised: 12/04/2017] [Accepted: 12/06/2017] [Indexed: 11/29/2022]
|