1
|
Melamed N, Kingdom JC, Fu L, Yip PM, Arruda-Caycho I, Hui D, Hladunewich MA. Predictive and Diagnostic Value of the Angiogenic Proteins in Patients With Chronic Kidney Disease. Hypertension 2024; 81:2251-2262. [PMID: 39162032 DOI: 10.1161/hypertensionaha.124.23411] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Accepted: 07/30/2024] [Indexed: 08/21/2024]
Abstract
BACKGROUND Our objective was to investigate the predictive and diagnostic accuracy of the angiogenic proteins sFlt-1 (soluble fms-like tyrosine kinase-1) and PlGF (placental growth factor) for preterm preeclampsia and explore the relationship between renal function and these proteins. METHODS We completed a blinded, prospective, longitudinal, observational study of patients with chronic kidney disease followed at a tertiary center (2018-2023). Serum samples were obtained at 3 time points along gestation (planned sampling): 12-16, 18-22, and 28-32 weeks. In addition, samples were obtained whenever preeclampsia was suspected (indicated sampling). sFlt-1 and PlGF levels remained concealed until the study ended. The primary outcome was preterm preeclampsia. The planned and indicated samples were used to estimate the predictive and diagnostic accuracy of the angiogenic proteins, respectively. RESULTS Of the 97 participants, 21 (21.6%) experienced preterm preeclampsia. In asymptomatic patients with chronic kidney disease, the angiogenic proteins were predictive of preterm preeclampsia only when sampled in the third trimester, in which case the sFlt-1/PlGF ratio (false positive rate of 37% for a detection rate of 80%) was more predictive than either sFlt-1 or PlGF in isolation. In patients with suspected preeclampsia, the diagnostic accuracy of the sFlt-1/PlGF ratio (false positive rate of 26% for a detection rate of 80%) was higher than that of sFlt-1 and PlGF in isolation. Diminished renal function was associated with increased levels of PlGF. CONCLUSIONS sFlt-1 and PlGF can effectively predict and improve the diagnostic accuracy for preterm preeclampsia among patients with chronic kidney disease. The optimal sFlt-1/PlGF ratio cutoff to rule out preeclampsia may need to be lower in patients with impaired renal function.
Collapse
Affiliation(s)
- Nir Melamed
- Division of Maternal-Fetal Medicine, Department of Obstetrics and Gynecology, Sunnybrook Health Sciences Centre, Temerty Faculty of Medicine (N.M., I.A.-C., D.H.), University of Toronto, Ontario, Canada
| | - John C Kingdom
- Division of Maternal-Fetal Medicine, Department of Obstetrics and Gynecology, Mount Sinai Hospital, Temerty Faculty of Medicine (J.C.K.), University of Toronto, Ontario, Canada
| | - Lei Fu
- Department of Laboratory Medicine and Molecular Diagnostics, Sunnybrook Health Sciences Centre (L.F., P.M.Y.), University of Toronto, Ontario, Canada
- Department of Laboratory Medicine and Pathobiology (L.F., P.M.Y.), University of Toronto, Ontario, Canada
| | - Paul M Yip
- Department of Laboratory Medicine and Molecular Diagnostics, Sunnybrook Health Sciences Centre (L.F., P.M.Y.), University of Toronto, Ontario, Canada
- Department of Laboratory Medicine and Pathobiology (L.F., P.M.Y.), University of Toronto, Ontario, Canada
| | - Isabel Arruda-Caycho
- Division of Maternal-Fetal Medicine, Department of Obstetrics and Gynecology, Sunnybrook Health Sciences Centre, Temerty Faculty of Medicine (N.M., I.A.-C., D.H.), University of Toronto, Ontario, Canada
| | - Dini Hui
- Division of Maternal-Fetal Medicine, Department of Obstetrics and Gynecology, Sunnybrook Health Sciences Centre, Temerty Faculty of Medicine (N.M., I.A.-C., D.H.), University of Toronto, Ontario, Canada
| | - Michelle A Hladunewich
- Division of Nephrology and Obstetric Medicine, Department of Medicine, Sunnybrook Health Sciences Centre, Temerty Faculty of Medicine (M.A.H.), University of Toronto, Ontario, Canada
| |
Collapse
|
2
|
Graupner O, Verlohren S, Groten T, Schlembach D, Stepan H, Kuschel B, Karge A, Pecks U. Significance of the sFlt-1/PlGF Ratio in Certain Cohorts - What Needs to be Considered? Geburtshilfe Frauenheilkd 2024; 84:629-634. [PMID: 38993800 PMCID: PMC11233204 DOI: 10.1055/a-2320-5843] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2024] [Accepted: 05/05/2024] [Indexed: 07/13/2024] Open
Abstract
The sFlt-1/PlGF ratio is an established tool in clinical practice, where it is part of a diagnostic algorithm and informs the prognosis of preeclampsia (PE). Maternal and gestational comorbidities can affect the performance of the sFlt-1/PlGF ratio and its constituent elements, and a good understanding of the potential pitfalls is required. The objective of this paper was to provide a current narrative review of the literature on the diagnostic and predictive performance of the sFlt-1/PlGF ratio in specific patient cohorts. Potential factors which can negatively affect the clinical interpretability and applicability of the sFlt-1/PlGF ratio include chronic kidney disease, twin pregnancy, and maternal obesity. Pathophysiological mechanisms related to these factors and disorders can result in different concentrations of sFlt-1 and/or PlGF in maternal blood, meaning that the use of standard cut-off values in specific cohorts can lead to errors. To what extent the cut-off values should be adapted in certain patient cohorts can only be clarified in large prospective cohort studies. This applies to the use of the ratio both for diagnosis and prognosis.
Collapse
Affiliation(s)
- Oliver Graupner
- Klinik und Poliklinik für Frauenheilkunde, Universitätsklinikum rechts der Isar, Technische Universität München, München, Germany
| | - Stefan Verlohren
- Klinik für Geburtsmedizin, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Tanja Groten
- Klinik für Geburtsmedizin, Universitätsklinikum Jena, Jena, Germany
| | - Dietmar Schlembach
- Klinik für Geburtsmedizin, Klinikum Neukölln, Vivantes Netzwerk für Gesundheit GmbH, Berlin, Germany
| | - Holger Stepan
- Klinik für Geburtsmedizin, Universitätsklinikum Leipzig, Leipzig, Germany
| | - Bettina Kuschel
- Klinik und Poliklinik für Frauenheilkunde, Universitätsklinikum rechts der Isar, Technische Universität München, München, Germany
| | - Anne Karge
- Klinik und Poliklinik für Frauenheilkunde, Universitätsklinikum rechts der Isar, Technische Universität München, München, Germany
| | - Ulrich Pecks
- Klinik für Gynäkologie und Geburtshilfe, Universitätsklinikum Würzburg, Würzburg, Germany
| |
Collapse
|
3
|
Januzzi JL, Liu Y, Sattar N, Yavin Y, Pollock CA, Butler J, Jardine M, Heerspink HJL, Masson S, Breyer M, Hansen MK. Vascular endothelial growth factors and risk of cardio-renal events: Results from the CREDENCE trial. Am Heart J 2024; 271:38-47. [PMID: 38401646 DOI: 10.1016/j.ahj.2024.02.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/01/2023] [Revised: 01/30/2024] [Accepted: 02/19/2024] [Indexed: 02/26/2024]
Abstract
BACKGROUND Circulating concentrations of vascular endothelial growth factor (VEGF) family members may be abnormally elevated in type 2 diabetes (T2D). The roles of placental growth factor (PlGF), soluble fms-like tyrosine kinase-1 (sFLT-1), and VEGF-A in cardio-renal complications of T2D are not established. METHOD The 2602 individuals with diabetic kidney disease (DKD) from the Canagliflozin and Renal Events in Diabetes with Established Nephropathy Clinical Evaluation trial were randomized to receive canagliflozin or placebo and followed for incident cardio-renal outcomes. PlGF, sFLT-1, and VEGF-A were measured at baseline, year 1, and year 3. Primary outcome was a composite of end-stage kidney disease, doubling of the serum creatinine, or renal/cardiovascular death. Cox proportional hazard regression was used to investigate the association between biomarkers with adverse clinical events. RESULTS At baseline, individuals with higher PlGF levels had more prevalent cardiovascular disease compared to those with lower values. Treatment with canagliflozin did not meaningfully change PlGF, sFLT-1, and VEGF-A concentrations at years 1 and 3. In a multivariable model, 1 unit increases in baseline log PlGF (hazard ratio [HR]: 1.76, 95% confidence interval [CI]: 1.23, 2.54, P-value = .002), sFLT-1 (HR: 3.34, [95% CI: 1.71, 6.52], P-value < .001), and PlGF/sFLT-1 ratio (HR: 4.83, [95% CI: 0.86, 27.01], P-value = .07) were associated with primary composite outcome, while 1 unit increase in log VEGF-A did not increase the risk of primary outcome (HR: 0.96 [95% CI: 0.81, 1.07]). Change by 1 year of each biomarker was also assessed: HR (95% CI) of primary composite outcome was 2.45 (1.70, 3.54) for 1 unit increase in 1-year concentration of log PlGF, 4.19 (2.18, 8.03) for 1 unit increase in 1-year concentration of log sFLT-1, and 21.08 (3.79, 117.4) for 1 unit increase in 1-year concentration of log PlGF/sFLT-1. Increase in 1-year concentrations of log VEGF-A was not associated with primary composite outcome (HR: 1.08, [95% CI: 0.93, 1.24], P-value = .30). CONCLUSIONS People with T2D and DKD with elevated levels of PlGF, sFLT-1, and PlGF/sFLT-1 ratio were at a higher risk for cardiorenal events. Canagliflozin did not meaningfully decrease concentrations of PlGF, sFLT-1, and VEGF-A. CLINICAL TRIAL CREDENCE, https://clinicaltrials.gov/ct2/show/NCT02065791.
Collapse
Affiliation(s)
- James L Januzzi
- Cardiology Division, Massachusetts General Hospital and Harvard Medical School, Boston, MA; Heart Failure and Biomarker Trials, Baim Institute for Clinical Research, Boston, MA.
| | - Yuxi Liu
- Cardiology Division, Massachusetts General Hospital and Harvard Medical School, Boston, MA
| | - Naveed Sattar
- BHF Glasgow Cardiovascular Research Centre, University of Glasgow, Glasgow, UK
| | - Yshai Yavin
- Janssen Research & Development, LLC, Spring House, PA
| | - Carol A Pollock
- Kolling Institute, Royal North Shore Hospital University of Sydney, Sydney, NSW, Australia
| | - Javed Butler
- University of Mississippi Medical Center, Jackson, MS; Baylor Scott & White Institute, Dallas, TX
| | - Meg Jardine
- The George Institute for Global Health, UNSW Sydney, Sydney, NSW
| | - Hiddo J L Heerspink
- Department Clinical Pharmacy and Pharmacology, University of Groningen, Groningen, The Netherlands
| | - Serge Masson
- Roche Diagnostics International, Rotkreuz, Switzerland
| | | | | |
Collapse
|
4
|
Biwer LA, Man JJ, Camarda ND, Carvajal BV, Karumanchi SA, Jaffe IZ. Prior Exposure to Experimental Preeclampsia Increases Atherosclerotic Plaque Inflammation in Atherogenic Mice-Brief Report. Arterioscler Thromb Vasc Biol 2024; 44:946-953. [PMID: 38450510 PMCID: PMC10978246 DOI: 10.1161/atvbaha.123.320474] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2023] [Accepted: 02/21/2024] [Indexed: 03/08/2024]
Abstract
BACKGROUND Women with a history of preeclampsia have evidence of premature atherosclerosis and increased risk of myocardial infarction and stroke compared with women who had a normotensive pregnancy. Whether this is due to common risk factors or a direct impact of prior preeclampsia exposure has never been tested in a mouse atherosclerosis model. METHODS Pregnant LDLR-KO (low-density lipoprotein receptor knockout; n=35) female mice were randomized in midgestation to sFlt1 (soluble fms-like tyrosine kinase 1)-expressing adenovirus or identical control adenovirus. Postpartum, mice were fed high-fat diet for 8 weeks to induce atherogenesis. Comparison between the control and preeclampsia models was made for metabolic parameters, atherosclerosis burden and composition by histology, plaque inflammation by flow cytometry, and aortic cytokines and inflammatory markers using a cytokine array. RESULTS In pregnant LDLR-KO mice, sFlt1 adenovirus significantly induced serum sFlt1, blood pressure, renal endotheliosis, and decreased pup viability. After 8 weeks of postpartum high fat feeding, body weight, fasting glucose, plasma cholesterol, HDL (high-density lipoprotein), and LDL (low-density lipoprotein) were not significantly different between groups with no change in aortic root plaque size, lipid content, or necrotic core area. Flow cytometry demonstrated significantly increased CD45+ aortic arch leukocytes and CD3+T cells and aortic lysate contained more CCL (CC motif chemokine ligand) 22 and fetuin A and decreased expression of IGFBP6 (insulin-like growth factor-binding protein 6) and CCL21 in preeclampsia-exposed mice compared with controls. CONCLUSIONS In atherogenic LDLR-KO mice, exposure to sFlt1-induced preeclampsia during pregnancy increases future atherosclerotic plaque inflammation, supporting the concept that preeclampsia directly exacerbates atherosclerotic inflammation independent of preexisting risk factors. This mechanism may contribute to ischemic vascular disease in women after preeclampsia pregnancy.
Collapse
Affiliation(s)
- Lauren A. Biwer
- Molecular Cardiology Research Center, Tufts Medical Center, Boston MA
- Department of Comparative Medicine, Vascular Biology and Therapeutics Program, Yale School of Medicine, New Haven, CT
| | - Joshua J. Man
- Molecular Cardiology Research Center, Tufts Medical Center, Boston MA
| | | | | | | | - Iris Z. Jaffe
- Molecular Cardiology Research Center, Tufts Medical Center, Boston MA
| |
Collapse
|
5
|
Matsui M, Onoue K, Saito Y. sFlt-1 in Chronic Kidney Disease: Friend or Foe? Int J Mol Sci 2022; 23:ijms232214187. [PMID: 36430665 PMCID: PMC9697971 DOI: 10.3390/ijms232214187] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Revised: 11/08/2022] [Accepted: 11/14/2022] [Indexed: 11/18/2022] Open
Abstract
Placental growth factor (PlGF) and its receptor, fms-like tyrosine kinase-1 (Flt-1), are important regulators involved in angiogenesis, atherogenesis, and inflammation. This review article focuses on the function of PlGF/Flt-1 signaling and its regulation by soluble Flt-1 (sFlt-1) in chronic kidney disease (CKD). Elevation of circulating sFlt-1 and downregulation of sFlt-1 in the vascular endothelium by uremic toxins and oxidative stress both exacerbate heart failure and atherosclerosis. Circulating sFlt-1 is inconsistent with sFlt-1 synthesis, because levels of matrix-bound sFlt-1 are much higher than those of circulating sFlt-1, as verified by a heparin loading test, and are drastically reduced in CKD.
Collapse
Affiliation(s)
- Masaru Matsui
- Department of Nephrology, Nara Prefecture General Medical Center, 2-897-5 Shichijo-Nishimachi, Nara 630-8581, Japan
- Department of Nephrology, Nara Medical University, 840 Shijo-Cho, Kashihara 634-8521, Japan
- Correspondence: ; Tel./Fax: +81-742-46-6001
| | - Kenji Onoue
- Department of Cardiology, Nara Medical University, 840 Shijo-Cho, Kashihara 634-8521, Japan
| | - Yoshihiko Saito
- Nara Prefecture Seiwa Medical Center, 1-14-16, Mimuro, Sango-Cho, Ikoma-Gun 636-0802, Japan
| |
Collapse
|
6
|
The VEGF Inhibitor Soluble Fms-like Tyrosine Kinase 1 Does Not Promote AKI-to-CKD Transition. Int J Mol Sci 2022; 23:ijms23179660. [PMID: 36077058 PMCID: PMC9456014 DOI: 10.3390/ijms23179660] [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: 07/29/2022] [Revised: 08/19/2022] [Accepted: 08/22/2022] [Indexed: 11/17/2022] Open
Abstract
(1) Background: Soluble Fms-like tyrosine kinase 1 (sFLT1) is an endogenous VEGF inhibitor. sFLT1 has been described as an anti-inflammatory treatment for diabetic nephropathy and heart fibrosis. However, sFLT1 has also been related to peritubular capillary (PTC) loss, which promotes fibrogenesis. Here, we studied whether transfection with sFlt1 aggravates experimental AKI-to-CKD transition and whether sFLT1 is increased in human kidney fibrosis. (2) Methods: Mice were transfected via electroporation with sFlt1. After confirming transfection efficacy, mice underwent unilateral ischemia/reperfusion injury (IRI) and were sacrificed 28 days later. Kidney histology and RNA were analyzed to study renal fibrosis, PTC damage and inflammation. Renal sFLT1 mRNA expression was measured in CKD biopsies and control kidney tissue. (3) Results: sFlt1 transfection did not aggravate renal fibrosis, PTC loss or macrophage recruitment in IRI mice. In contrast, higher transfection efficiency was correlated with reduced expression of pro-fibrotic and pro-inflammatory markers. In the human samples, sFLT1 mRNA levels were similar in CKD and control kidneys and were not correlated with interstitial fibrosis or PTC loss. (4) Conclusion: As we previously found that sFLT1 has therapeutic potential in diabetic nephropathy, our findings indicate that sFLT1 can be administered at a dose that is therapeutically effective in reducing inflammation, without promoting maladaptive kidney damage.
Collapse
|
7
|
Nocturnal hypertension-solving the puzzle of preeclampsia risk. Hypertens Res 2021; 44:1681-1682. [PMID: 34690350 DOI: 10.1038/s41440-021-00770-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2021] [Revised: 09/06/2021] [Accepted: 09/07/2021] [Indexed: 11/08/2022]
|
8
|
Zhou YH, Tang YZ, Guo LY, Zheng LL, Zhang D, Yang CY, Wang W. Overexpression of sFlt-1 represses ox-LDL-induced injury of HUVECs by activating autophagy via PI3K/AKT/mTOR pathway. Microvasc Res 2021; 139:104252. [PMID: 34520772 DOI: 10.1016/j.mvr.2021.104252] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Revised: 08/26/2021] [Accepted: 09/06/2021] [Indexed: 10/20/2022]
Abstract
Soluble fms-like tyrosine kinase-1 (sFlt-1), a circulating antiangiogenic protein, is involved in the pathogenesis of atherosclerosis (AS), and the underlying mechanism is still unclear. Here, we attempted to investigate the mechanism of action of sFlt-1 in AS. Human umbilical vein endothelial cells (HUVECs) were treated with oxidized low density lipoprotein (ox-LDL) to induce cell injury. ox-LDL treatment increased LC3-II/LC3-I ratio, Beclin-1 expression and GFP-LC3 puncta in HUVECs, suggesting that ox-LDL may induce autophagic flux impairment in HUVECs. ox-LDL-treated HUVECs displayed a decrease of sFlt-1 levels. Moreover, ox-LDL treatment reduced cell proliferation and elevated apoptosis in HUVECs, which was abrogated by sFlt-1 overexpression. Up-regulation of sFlt-1 repressed the activity of PI3K/AKT/mTOR signaling pathway and enhanced autophagy in HUVECs following ox-LDL treatment. Additionally, sFlt-1 overexpression-mediated increase of autophagy in ox-LDL-treated HUVECs was abolished by 3-methyladenine (autophagy inhibitor). 3-methyladenine abrogated the impact of sFlt-1 overexpression on proliferation and apoptosis in ox-LDL-treated HUVECs. This work confirmed that overexpression of sFlt-1 activated autophagy by repressing PI3K/Akt/mTOR signaling pathway, and thus alleviated ox-LDL-induced injury of HUVECs. Therefore, this study suggests that sFlt-1 may be a potential target for AS treatment.
Collapse
Affiliation(s)
- Yi-Hua Zhou
- Department of ICU, The Second Affiliated Hospital of Nanchang University, No. 1 Minde Road, Nanchang, Jiangxi 330006, China
| | - Yu-Zhi Tang
- Department of Ultrasound, The Second Affiliated Hospital of Nanchang University, No. 1 Minde Road, Nanchang, Jiangxi 330006, China
| | - Liang-Yun Guo
- Department of Ultrasound, The Second Affiliated Hospital of Nanchang University, No. 1 Minde Road, Nanchang, Jiangxi 330006, China
| | - Li-Li Zheng
- Department of Pharmacy, Jiangxi Maternal and Child Health Hospital, Nanchang, Jiangxi 330006, China
| | - Dan Zhang
- Department of Ultrasound, The Second Affiliated Hospital of Nanchang University, No. 1 Minde Road, Nanchang, Jiangxi 330006, China
| | - Can-Ying Yang
- Department of Ultrasound, The Second Affiliated Hospital of Nanchang University, No. 1 Minde Road, Nanchang, Jiangxi 330006, China
| | - Wei Wang
- Department of Neurology, The Second Affiliated Hospital of Nanchang University, No. 1 Minde Road, Nanchang, Jiangxi 330006, China.
| |
Collapse
|
9
|
Wewers TM, Schulz A, Nolte I, Pavenstädt H, Brand M, Di Marco GS. Circulating Soluble Fms-like Tyrosine Kinase in Renal Diseases Other than Preeclampsia. J Am Soc Nephrol 2021; 32:1853-1863. [PMID: 34155060 PMCID: PMC8455271 DOI: 10.1681/asn.2020111579] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Accepted: 04/20/2021] [Indexed: 02/04/2023] Open
Abstract
Soluble Fms-like tyrosine kinase (sFlt-1/sVEGFR1) is a naturally occurring antagonist of vascular endothelial growth factor (VEGF). Despite being a secreted, soluble protein lacking cytoplasmic and transmembrane domains, sFlt-1 can act locally and be protective against excessive microenvironmental VEGF concentration or exert autocrine functions independently of VEGF. Circulating sFlt-1 may indiscriminately affect endothelial function and the microvasculature of distant target organs. The clinical significance of excess sFlt-1 in kidney disease was first shown in preeclampsia, a major renal complication of pregnancy. However, circulating sFlt-1 levels appear to be increased in various diseases with varying degrees of renal impairment. Relevant clinical associations between circulating sFlt-1 and severe outcomes (e.g., endothelial dysfunction, renal impairment, cardiovascular disease, and all-cause mortality) have been observed in patients with CKD and after kidney transplantation. However, sFlt-1 appears to be protective against renal dysfunction-associated aggravation of atherosclerosis and diabetic nephropathy. Therefore, in this study, we provide an update on sFlt-1 in several kidney diseases other than preeclampsia, discuss clinical findings and experimental studies, and briefly consider its use in clinical practice.
Collapse
Affiliation(s)
- Theresa M. Wewers
- Department of Internal Medicine D, University Hospital Muenster, Muenster, Germany,Small Animal Hospital, University of Veterinary Medicine Hannover, Hannover, Germany
| | - Annika Schulz
- Department of Internal Medicine D, University Hospital Muenster, Muenster, Germany
| | - Ingo Nolte
- Small Animal Hospital, University of Veterinary Medicine Hannover, Hannover, Germany
| | - Hermann Pavenstädt
- Department of Internal Medicine D, University Hospital Muenster, Muenster, Germany
| | - Marcus Brand
- Department of Internal Medicine D, University Hospital Muenster, Muenster, Germany
| | - Giovana S. Di Marco
- Department of Internal Medicine D, University Hospital Muenster, Muenster, Germany,Correspondence: Giovana S. Di Marco, Albert-Schweitzer-Campus 1, Building A14, 48149 Münster, Germany.
| |
Collapse
|
10
|
Nakano T, Onoue K, Seno A, Ishihara S, Nakada Y, Nakagawa H, Ueda T, Nishida T, Soeda T, Watanabe M, Kawakami R, Hatakeyama K, Sakaguchi Y, Ohbayashi C, Saito Y. Involvement of chronic inflammation via monocyte chemoattractant protein-1 in uraemic cardiomyopathy: a human biopsy study. ESC Heart Fail 2021; 8:3156-3167. [PMID: 33988313 PMCID: PMC8318461 DOI: 10.1002/ehf2.13423] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2020] [Revised: 04/12/2021] [Accepted: 05/02/2021] [Indexed: 12/19/2022] Open
Abstract
Aims Patients undergoing dialysis, even those without coronary artery disease or valvular abnormalities, sometimes present with reduced heart function, which resembles dilated cardiomyopathy (DCM). This condition is known as uraemic cardiomyopathy (UCM). The mechanisms of UCM development are not fully understood. Previous studies demonstrated that the balance between placental growth factor (PlGF) and fms‐like tyrosine kinase‐1 (Flt‐1) is correlated with renal function, and PlGF/Flt‐1 signalling is involved in the development of cardiovascular diseases in patients with chronic kidney disease. This study was conducted to evaluate the pathogenesis of UCM and clarify the differences in the mechanisms of UCM and DCM by using human endomyocardial biopsy and blood samples. Methods and results The clinical and pathological features of 30 patients on dialysis with reduced cardiac function [left ventricular ejection fraction (LVEF) ≤50%] (UCM group; mean age: 58.5 ± 9.4 years and LVEF: 39.1 ± 7.2%), 196 DCM patients (DCM group; mean age: 62.7 ± 14.0 years and LVEF: 33.5 ± 8.8%) as controls with reduced cardiac function (LVEF ≤ 45%), and 21 patients as controls with normal cardiac function (control group; mean age: 56.2 ± 19.3 years and LVEF: 67.5 ± 6.7%) were analysed. The percentage of the interstitial fibrosis area in the UCM group was greater than that in the DCM group (P = 0.045). In UCM patients, the percentage of the interstitial fibrosis area was positively correlated with the duration of renal replacement therapy (P < 0.001). The number of infiltrated CD68‐positive macrophages in the myocardium and expression of monocyte chemoattractant protein‐1 (MCP‐1) in cardiomyocytes were significantly greater in the UCM group than in the other groups (P < 0.001, respectively). Furthermore, while the serum level of soluble form of Flt‐1, an endogenous inhibitor of PlGF, in the UCM group was lower compared with that in the DCM group (P < 0.001), the serum levels of PlGF and PlGF/soluble form of Flt‐1 ratio and plasma level of MCP‐1 in the UCM group were higher than those in the DCM group (P < 0.001, respectively). Conclusions These results suggest that activated PlGF/Flt‐1 signalling and subsequent macrophage‐mediated chronic non‐infectious inflammation via MCP‐1 in the myocardium are involved in the pathogenesis of UCM.
Collapse
Affiliation(s)
- Tomoya Nakano
- Department of Cardiovascular Medicine, Nara Medical University, Kashihara, Nara, Japan.,Department of Internal Medicine, Yamato-Takada Municipal Hospital, Yamato-Takada, Nara, Japan
| | - Kenji Onoue
- Department of Cardiovascular Medicine, Nara Medical University, Kashihara, Nara, Japan
| | - Ayako Seno
- Department of Cardiovascular Medicine, Nara Medical University, Kashihara, Nara, Japan
| | - Satomi Ishihara
- Department of Cardiovascular Medicine, Nara Medical University, Kashihara, Nara, Japan
| | - Yasuki Nakada
- Department of Cardiovascular Medicine, Nara Medical University, Kashihara, Nara, Japan
| | - Hitoshi Nakagawa
- Department of Cardiovascular Medicine, Nara Medical University, Kashihara, Nara, Japan
| | - Tomoya Ueda
- Department of Cardiovascular Medicine, Nara Medical University, Kashihara, Nara, Japan
| | - Taku Nishida
- Department of Cardiovascular Medicine, Nara Medical University, Kashihara, Nara, Japan
| | - Tsunenari Soeda
- Department of Cardiovascular Medicine, Nara Medical University, Kashihara, Nara, Japan
| | - Makoto Watanabe
- Department of Cardiovascular Medicine, Nara Medical University, Kashihara, Nara, Japan
| | - Rika Kawakami
- Department of Cardiovascular Medicine, Nara Medical University, Kashihara, Nara, Japan
| | - Kinta Hatakeyama
- Department of Pathology, National Cerebral and Cardiovascular Center, Suita, Osaka, Japan.,Department of Diagnostic Pathology, Nara Medical University, Kashihara, Nara, Japan
| | - Yasuhiro Sakaguchi
- Department of Cardiovascular Medicine, Nara Medical University, Kashihara, Nara, Japan
| | - Chiho Ohbayashi
- Department of Diagnostic Pathology, Nara Medical University, Kashihara, Nara, Japan
| | - Yoshihiko Saito
- Department of Cardiovascular Medicine, Nara Medical University, Kashihara, Nara, Japan
| |
Collapse
|
11
|
Wiles K, Bramham K, Seed PT, Brockbank A, Nelson-Piercy C, Karumanchi SA, Lightstone L, Chappell LC. Placental and endothelial biomarkers for the prediction of superimposed pre-eclampsia in chronic kidney disease. Pregnancy Hypertens 2021; 24:58-64. [PMID: 33677420 DOI: 10.1016/j.preghy.2021.02.010] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Revised: 02/09/2021] [Accepted: 02/16/2021] [Indexed: 12/21/2022]
Abstract
OBJECTIVES To evaluate PlGF, sFlt-1, and novel endothelial biomarkers hyaluronan and vascular cell adhesion molecule (VCAM), for the prediction of superimposed pre-eclampsia in women with chronic kidney disease (CKD). STUDY DESIGN Prospective cohort study of pregnant women with CKD in UK. MAIN OUTCOME MEASURES Outcomes including superimposed pre-eclampsia were based on predetermined criteria. Test performances of plasma PlGF, serum sFlt-1:PlGF, hyaluronan and VCAM concentrations were evaluated as area under the receiver-operating curve and at established and exploratory threshold concentrations. RESULTS There were 232 pregnancies in 221 women with CKD. One third (76/232) developed superimposed pre-eclampsia. From 21 to 37 weeks' gestation, plasma PlGF was decreased among women that developed superimposed preeclampsia. Plasma PlGF levels < 150 pg/ml had the highest sensitivity (79% 95% CI: 58-91%) and negative predictive value (97%, 95% CI: 93-99%) for the prediction of delivery with superimposed pre-eclampsia within 14 days. Predictive performances of hyaluronan and VCAM were lower than for plasma PlGF. Low plasma PlGF, high hyaluronan and high VCAM concentrations had lower predictive performance in women with pre-pregnancy CKD stages 3-5 compared to stages 1-2. sFlt-1:PlGF > 38 did not usefully predict the need to deliver in women with CKD when measured in serum. CONCLUSIONS Increased surveillance for the need for delivery should take place in women with CKD and plasma PlGF below 150 pg/ml after 20 weeks' gestation, with awareness that predictive value is reduced as excretory kidney function declines. Maternal endothelial dysfunction may alter the PlGF threshold at which superimposed pre-eclampsia manifests in women with CKD.
Collapse
Affiliation(s)
- Kate Wiles
- Department of Women and Children's Health, King's College London, London, United Kingdom; Department of Renal Medicine, Barts Health NHS Trust, United Kingdom.
| | - Kate Bramham
- Department of Women and Children's Health, King's College London, London, United Kingdom; Department of Renal Medicine, King's College Hospital NHS Foundation Trust, London, United Kingdom
| | - Paul T Seed
- Department of Women and Children's Health, King's College London, London, United Kingdom
| | - Anna Brockbank
- Department of Women and Children's Health, King's College London, London, United Kingdom
| | - Catherine Nelson-Piercy
- Department of Women and Children's Health, King's College London, London, United Kingdom; Guy's and St. Thomas' NHS Foundation Trust, United Kingdom
| | - S Ananth Karumanchi
- Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, United States
| | - Liz Lightstone
- Imperial College London and Imperial College Healthcare NHS Trust, United Kingdom
| | - Lucy C Chappell
- Department of Women and Children's Health, King's College London, London, United Kingdom; Guy's and St. Thomas' NHS Foundation Trust, United Kingdom
| |
Collapse
|
12
|
Saito Y. The role of the PlGF/Flt-1 signaling pathway in the cardiorenal connection. J Mol Cell Cardiol 2020; 151:106-112. [PMID: 33045252 DOI: 10.1016/j.yjmcc.2020.10.001] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/14/2020] [Revised: 09/29/2020] [Accepted: 10/02/2020] [Indexed: 02/07/2023]
Abstract
Although the concept of the cardiorenal connection is widely accepted, athe underlying molecular mechanism has not been clearly defined. Nevertheless, accumulating evidence indicates that the nervous system and both the humoral and cellular immune systems are all involved. This review article focuses on the roles of the signaling pathway of placental growth factor (PlGF) and its receptor, fms-like tyrosine kinase-1 (Flt-1), in the development of the cardiorenal connection. PlGF, a member of the vascular endothelial cell growth factor family, is a specific ligand for Flt-1 and plays roles in the development of atherosclerosis, wound healing after ischemia injury, and angiogenesis through Flt-1 signaling. Flt-1, a tyrosine-kinase type receptor with a single transmembrane domain, has a soluble isoform (sFlt-1) consisting of only extracellular domains, and is an intrinsic antagonist of PlGF. In renal dysfunction, PlGF is upregulated and sFlt-1 is downregulated by oxidative stress or uremic toxins, leading to activation of the PlGF/Flt-1 signaling pathway, which in turn plays a role in the worsening of atherosclerosis and heart failure, both of which are frequently associated with renal dysfunction. Monocyte chemotactic protein-1 (MCP-1) is involved in the process downstream of the Flt-1 signaling pathway. Plasma levels of sFlt-1 correlate with the severity of renal dysfunction in patients with heart failure or myocardial infarction, and are associated with the incidence of cardiovascular events. This is inconsistent with the concept of relative activation of the PlGF/Flt-1 pathway in renal dysfunction. However, the level of circulating sFlt-1 does not always parallel sFlt-1 synthesis, probably because sFlt-1 is stored on cell surfaces through its heparin-binding domains and its quantity is regulated differently in renal dysfunction. This review summarizes a novel concept wherein noninfectious inflammation via PlGF/Flt-1 signaling is involved in the development of renal dysfunction-related cardiovascular complications.
Collapse
Affiliation(s)
- Yoshihiko Saito
- Department of Cardiovascular Medicine, Nara Medical University, 840 Shijo-cho, Kashihara 634-8522, Japan.
| |
Collapse
|
13
|
Genetic variants of VEGFR-1 gene promoter in acute myocardial infarction. Hum Genomics 2019; 13:56. [PMID: 31744542 PMCID: PMC6862733 DOI: 10.1186/s40246-019-0243-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2019] [Accepted: 10/09/2019] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND Coronary artery disease (CAD) including acute myocardial infarction (AMI) is a common complex disease caused by atherosclerosis. Vascular epithelial growth factor receptor-1 (VEGFR-1) stimulates angiogenesis and vascular permeability, and functions as a decoy to sequester VEGF and prevent initiation of intracellular signaling. VEGFR-1 knockout mice exhibit significantly higher mortality due to heart failure, cardiac hypertrophy, and cardiac dysfunction. An evident increase in macrophage infiltration and cardiac fibrosis are also observed after transverse aortic constriction. Therefore, VEGFR-1 gene variants may be involved in CAD. In this study, VEGFR-1 gene promoter was genetically and functionally analyzed in large cohorts of AMI patients and ethnic-matched controls. RESULTS A total of 16 DNA sequence variants (DSVs) including six single-nucleotide polymorphisms (SNPs) were found in the VEGFR-1 gene promoter and 5'-untranslated region. Five novel DSVs and one SNP were only identified in AMI patients group. These DSVs and SNP significantly altered the transcriptional activity of the VEGFR-1 gene promoter in both HEK-293 and H9c2 cells (P < 0.05). Further electrophoretic mobility shift assay indicated that the DSVs and SNPs evidently affected the binding of transcription factors. CONCLUSIONS The genetic variants in VEGFR-1 gene identified in AMI patients may alter the transcriptional activity of the VEGFR-1 gene promoter and change VEGFR-1 level, contributing to AMI development.
Collapse
|
14
|
Nakada Y, Onoue K, Nakano T, Ishihara S, Kumazawa T, Nakagawa H, Ueda T, Nishida T, Soeda T, Okayama S, Watanabe M, Kawakami R, Saito Y. AST-120, an Oral Carbon Absorbent, Protects against the Progression of Atherosclerosis in a Mouse Chronic Renal Failure Model by Preserving sFlt-1 Expression Levels. Sci Rep 2019; 9:15571. [PMID: 31666542 PMCID: PMC6821698 DOI: 10.1038/s41598-019-51292-9] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2019] [Accepted: 09/27/2019] [Indexed: 12/21/2022] Open
Abstract
Soluble Flt-1 (sFlt-1), an endogenous antagonist of the proatherogenic cytokine placental growth factor, is decreased in chronic kidney disease (CKD), leading to atherosclerotic progression. In this study, we investigated the effect of AST-120, an oral carbon adsorbent which can remove uremic toxins, on sFlt-1 expression levels and atherosclerosis progression. Atherosclerotic apolipoprotein E-deficient mice underwent a 5/6 nephrectomy (5/6 NR) or a sham operation (sham) at 8 weeks of age and were then treated or not with oral AST-120 for 12 weeks. sFlt-1 expression levels and the degree of atherosclerosis were assessed at 22 weeks of age in each of the four groups (sham; n = 7, 5/6 NR; n = 10, sham + AST-120: n = 8, 5/6 NR + AST-120; n = 8). The expression levels of sFlt-1 mRNA in the kidney were significantly lower in the 5/6 NR group than in the sham group, but AST-120 treatment prevented this decrease in sFlt-1 levels. Similarly, the atherosclerotic plaque area of the thoracoabdominal aorta was significantly larger in the 5/6 NR group than in the sham group, and AST-120 treatment prevented this increase in atherosclerosis. AST-120 could, therefore, be used as a therapeutic to treat atherosclerosis in patients with CKD.
Collapse
Affiliation(s)
- Yasuki Nakada
- Department of Cardiovascular Medicine, Nara Medical University, Kashihara, Nara, 634-8522, Japan
| | - Kenji Onoue
- Department of Cardiovascular Medicine, Nara Medical University, Kashihara, Nara, 634-8522, Japan.
| | - Tomoya Nakano
- Department of Cardiovascular Medicine, Nara Medical University, Kashihara, Nara, 634-8522, Japan
| | - Satomi Ishihara
- Department of Cardiovascular Medicine, Nara Medical University, Kashihara, Nara, 634-8522, Japan
| | - Takuya Kumazawa
- Department of Cardiovascular Medicine, Nara Medical University, Kashihara, Nara, 634-8522, Japan
| | - Hitoshi Nakagawa
- Department of Cardiovascular Medicine, Nara Medical University, Kashihara, Nara, 634-8522, Japan
| | - Tomoya Ueda
- Department of Cardiovascular Medicine, Nara Medical University, Kashihara, Nara, 634-8522, Japan
| | - Taku Nishida
- Department of Cardiovascular Medicine, Nara Medical University, Kashihara, Nara, 634-8522, Japan
| | - Tsunenari Soeda
- Department of Cardiovascular Medicine, Nara Medical University, Kashihara, Nara, 634-8522, Japan
| | - Satoshi Okayama
- Department of Cardiovascular Medicine, Nara Medical University, Kashihara, Nara, 634-8522, Japan
| | - Makoto Watanabe
- Department of Cardiovascular Medicine, Nara Medical University, Kashihara, Nara, 634-8522, Japan
| | - Rika Kawakami
- Department of Cardiovascular Medicine, Nara Medical University, Kashihara, Nara, 634-8522, Japan
| | - Yoshihiko Saito
- Department of Cardiovascular Medicine, Nara Medical University, Kashihara, Nara, 634-8522, Japan
| |
Collapse
|
15
|
Draker N, Torry DS, Torry RJ. Placenta growth factor and sFlt-1 as biomarkers in ischemic heart disease and heart failure: a review. Biomark Med 2019; 13:785-799. [DOI: 10.2217/bmm-2018-0492] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Coronary heart disease (CHD) and heart failure (HF) produce significant morbidity/mortality but identifying new biomarkers could help in the management of each. In this article, we summarize the molecular regulation and biomarker potential of PIGF and sFlt-1 in CHD and HF. PlGF is elevated during ischemia and some studies have shown PlGF, sFlt-1 or PlGF:sFlt-1 ratio, when used in combination with standard biomarkers, strengthens predictions of outcomes. sFlt-1 and PlGF are elevated in HF with sFlt-1 as a stronger predictor of outcomes. Although promising, we discuss additional study criteria needed to confirm the clinical usefulness of PlGF or sFlt-1 in the detection and management of CHD or HF.
Collapse
Affiliation(s)
- Nicole Draker
- Department of Pharmaceutical & Administrative Sciences, Ellis Pharmacogenomics Lab, College of Pharmacy & Health Sciences, Drake University, Des Moines, IA 50311, USA
| | - Donald S Torry
- Department of Medical Microbiology, Immunology, & Cell Biology, Department of OB/GYN, Southern Illinois University, School of Medicine, Springfield, IL 62702, USA
| | - Ronald J Torry
- Department of Pharmaceutical & Administrative Sciences, Ellis Pharmacogenomics Lab, College of Pharmacy & Health Sciences, Drake University, Des Moines, IA 50311, USA
| |
Collapse
|
16
|
Wewers TM, Mayer AB, Pfleiderer A, Beul K, Schmidt R, Heitplatz B, Van Marck V, Nolte I, Pavenstädt H, Reuter S, Brand M, Di Marco GS. Increased soluble fms-like tyrosine kinase 1 after ischemia reperfusion contributes to adverse clinical outcomes following kidney transplantation. Kidney Int 2019; 95:1091-1102. [PMID: 30824181 DOI: 10.1016/j.kint.2018.11.023] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2018] [Revised: 10/05/2018] [Accepted: 11/02/2018] [Indexed: 12/20/2022]
Abstract
Renal ischemia reperfusion injury (IRI) adversely affects clinical outcomes following kidney transplantation. Understanding the cellular mechanisms and the changes in gene/protein expression following IRI may help to improve these outcomes. Serum soluble fms-like tyrosine kinase 1 (sFlt-1), a circulating antiangiogenic protein, is increased in the first week following kidney transplantation. We evaluated the casual relationship of elevated sFlt-1 levels with renal microvascular dysfunction following IRI in a longitudinal study of 93 kidney transplant recipients and in several animal models. Transplant recipients with higher sFlt-1 levels had higher odds of delayed graft function, graft rejection, impaired graft function, and death. In a subgroup of 25 participants who underwent kidney biopsy within 4 months of kidney transplantation, peritubular capillary area was lower in those with elevated serum sFtl-1 levels. The administration of recombinant sFlt-1 into rodents resulted in significant structural and functional changes of the renal microvasculature, including reduced peritubular capillary density and intracapillary blood volume, and lead to increased expression of inflammatory genes and increased fibrosis. In a murine model of IRI, the kidney was a site of sFlt-1 production, and systemic neutralization of sFlt-1 preserved peritubular capillary density and alleviated renal fibrosis. Our data indicate that high sFlt-1 levels after IRI play an important role in the pathogenesis of microvascular dysfunction, thereby contributing to adverse clinical outcomes following kidney transplantation.
Collapse
Affiliation(s)
- Theresa M Wewers
- Department of Internal Medicine D, University Hospital Münster, Münster, Germany; Small Animal Hospital, University of Veterinary Medicine Hannover, Hannover, Germany
| | - Anna B Mayer
- Department of Internal Medicine D, University Hospital Münster, Münster, Germany
| | - Alexander Pfleiderer
- Department of Internal Medicine D, University Hospital Münster, Münster, Germany
| | - Katrin Beul
- Department of Internal Medicine D, University Hospital Münster, Münster, Germany
| | - Rene Schmidt
- Institute for Biostatistics and Clinical Research, University Hospital Münster, Münster, Germany
| | - Barbara Heitplatz
- Department of Pathology, University Hospital Münster, Münster, Germany
| | - Veerle Van Marck
- Department of Pathology, University Hospital Münster, Münster, Germany
| | - Ingo Nolte
- Small Animal Hospital, University of Veterinary Medicine Hannover, Hannover, Germany
| | - Hermann Pavenstädt
- Department of Internal Medicine D, University Hospital Münster, Münster, Germany
| | - Stefan Reuter
- Department of Internal Medicine D, University Hospital Münster, Münster, Germany
| | - Marcus Brand
- Department of Internal Medicine D, University Hospital Münster, Münster, Germany
| | - Giovana S Di Marco
- Department of Internal Medicine D, University Hospital Münster, Münster, Germany.
| |
Collapse
|
17
|
Nakada Y, Kawakami R, Matsui M, Ueda T, Nakano T, Nakagawa H, Nishida T, Onoue K, Soeda T, Okayama S, Watanabe M, Okura H, Saito Y. Value of Placental Growth Factor as a Predictor of Adverse Events During the Acute Phase of Acute Decompensated Heart Failure. Circ J 2019; 83:395-400. [DOI: 10.1253/circj.cj-18-0523] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Affiliation(s)
- Yasuki Nakada
- Department of Cardiovascular Medicine, Nara Medical University
| | - Rika Kawakami
- Department of Cardiovascular Medicine, Nara Medical University
| | - Masaru Matsui
- Department of Cardiovascular Medicine, Nara Medical University
| | - Tomoya Ueda
- Department of Cardiovascular Medicine, Nara Medical University
| | - Tomoya Nakano
- Department of Cardiovascular Medicine, Nara Medical University
| | | | - Taku Nishida
- Department of Cardiovascular Medicine, Nara Medical University
| | - Kenji Onoue
- Department of Cardiovascular Medicine, Nara Medical University
| | - Tsunenari Soeda
- Department of Cardiovascular Medicine, Nara Medical University
| | - Satoshi Okayama
- Department of Cardiovascular Medicine, Nara Medical University
| | - Makoto Watanabe
- Department of Cardiovascular Medicine, Nara Medical University
| | - Hiroyuki Okura
- Department of Cardiovascular Medicine, Nara Medical University
| | - Yoshihiko Saito
- Department of Cardiovascular Medicine, Nara Medical University
| |
Collapse
|
18
|
Endogenous Antiangiogenic Factors in Chronic Kidney Disease: Potential Biomarkers of Progression. Int J Mol Sci 2018; 19:ijms19071859. [PMID: 29937525 PMCID: PMC6073618 DOI: 10.3390/ijms19071859] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2018] [Revised: 06/17/2018] [Accepted: 06/22/2018] [Indexed: 12/17/2022] Open
Abstract
Chronic kidney disease (CKD) is a major global health problem. Unless intensive intervention is initiated, some patients can rapidly progress to end-stage kidney disease. However, it is often difficult to predict renal outcomes using conventional laboratory tests in individuals with CKD. Therefore, many researchers have been searching for novel biomarkers to predict the progression of CKD. Angiogenesis is involved in physiological and pathological processes in the kidney and is regulated by the balance between a proangiogenic factor, vascular endothelial growth factor (VEGF)-A, and various endogenous antiangiogenic factors. In recent reports using genetically engineered mice, the roles of these antiangiogenic factors in the pathogenesis of kidney disease have become increasingly clear. In addition, recent clinical studies have demonstrated associations between circulating levels of antiangiogenic factors and renal dysfunction in CKD patients. In this review, we summarize recent advances in the study of representative endogenous antiangiogenic factors, including soluble fms-related tyrosine kinase 1, soluble endoglin, pigment epithelium-derived factor, VEGF-A165b, endostatin, and vasohibin-1, in associations with kidney diseases and discuss their predictive potentials as biomarkers of progression of CKD.
Collapse
|
19
|
Failla CM, Carbo M, Morea V. Positive and Negative Regulation of Angiogenesis by Soluble Vascular Endothelial Growth Factor Receptor-1. Int J Mol Sci 2018; 19:ijms19051306. [PMID: 29702562 PMCID: PMC5983705 DOI: 10.3390/ijms19051306] [Citation(s) in RCA: 55] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2018] [Revised: 04/23/2018] [Accepted: 04/24/2018] [Indexed: 12/20/2022] Open
Abstract
Vascular endothelial growth factor receptor (VEGFR)-1 exists in different forms, derived from alternative splicing of the same gene. In addition to the transmembrane form, endothelial cells produce a soluble VEGFR-1 (sVEGFR-1) isoform, whereas non-endothelial cells produce both sVEGFR-1 and a different soluble molecule, known as soluble fms-like tyrosine kinase (sFlt)1-14. By binding members of the vascular endothelial growth factor (VEGF) family, the soluble forms reduce the amounts of VEGFs available for the interaction with their transmembrane receptors, thereby negatively regulating VEGFR-mediated signaling. In agreement with this activity, high levels of circulating sVEGFR-1 or sFlt1-14 are associated with different pathological conditions involving vascular dysfunction. Moreover, sVEGFR-1 and sFlt1-14 have an additional role in angiogenesis: they are deposited in the endothelial cell and pericyte extracellular matrix, and interact with cell membrane components. Interaction of sVEGFR-1 with α5β1 integrin on endothelial cell membranes regulates vessel growth, triggering a dynamic, pro-angiogenic phenotype. Interaction of sVEGFR-1/sFlt1-14 with cell membrane glycosphingolipids in lipid rafts controls kidney cell morphology and glomerular barrier functions. These cell⁻matrix contacts represent attractive novel targets for pharmacological intervention in addition to those addressing interactions between VEGFs and their receptors.
Collapse
Affiliation(s)
| | - Miriam Carbo
- Department of Biochemical Sciences "A. Rossi Fanelli", Sapienza University, 00185 Rome, Italy.
| | - Veronica Morea
- National Research Council of Italy (CNR), Department of Biochemical Sciences "A. Rossi Fanelli", Institute of Molecular Biology and Pathology c/o, Sapienza University, 00185 Rome, Italy.
| |
Collapse
|
20
|
Kelaidi C, Kattamis A, Apostolakou F, Poziopoulos C, Lazaropoulou C, Delaporta P, Kanavaki I, Papassotiriou I. PlGF and sFlt-1 levels in patients with non-transfusion-dependent thalassemia: Correlations with markers of iron burden and endothelial dysfunction. Eur J Haematol 2018. [PMID: 29543340 DOI: 10.1111/ejh.13061] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
BACKGROUND Levels of the angiogenic cytokines placental growth factor (PlGF) and soluble Fms-like tyrosine kinase-1 (sFlt-1) and the angiogenic balance, expressed by sFlt-1/PlGF ratio, are perturbed in sickle-cell disease and iron overload, but they have not been evaluated in non-transfusion-dependent thalassemia (NTDT). PATIENTS AND METHODS We measured levels of PlGF, sFlt-1 and vWF:antigen in patients with NTDT of beta-thalassemia genotype, and correlated them with erythrocytic indices and markers of iron overload, inflammation, and tissue hypoxia. Thirty-four NTDT patients with mean hemoglobin level of 8.4 g/dL were included in the study along with 20 apparently healthy individuals who served as controls. RESULTS Ferritin, LDH, and hs-CRP were higher in patients as compared to controls. We found significant differences between patients and controls in regard to levels of PlGF (52.2 vs 17.2 pg/mL, P < .001), sFlt-1/PlGF (2 vs 4.7, P < .001), and vWF:antigen (88 vs 77.1 IU/dL, P < .01). There was a strong correlation of ferritin with PlGF (r = .653, P < .001) and with vWF:antigen (r = .503, P = .003). CONCLUSIONS In this study, we demonstrated an association between increased PlGF and iron overload and the degree of tissue hypoxia in patients with NTDT. High vWF:antigen expressing endothelial damage may be associated with specific NTDT comorbidities.
Collapse
Affiliation(s)
- Charikleia Kelaidi
- Department of Pediatric Hematology-Oncology, "Aghia Sophia" Children's Hospital, Athens, Greece
| | - Antonis Kattamis
- First Department of Pediatrics, National and Kapodistrian University of Athens Medical School, Athens, Greece
| | - Filia Apostolakou
- Department of Clinical Biochemistry, "Aghia Sophia" Children's Hospital, Athens, Greece
| | - Christos Poziopoulos
- Department of Clinical Biochemistry, "Aghia Sophia" Children's Hospital, Athens, Greece
| | | | - Polyxeni Delaporta
- First Department of Pediatrics, National and Kapodistrian University of Athens Medical School, Athens, Greece
| | - Ino Kanavaki
- Department of Clinical Biochemistry, "Aghia Sophia" Children's Hospital, Athens, Greece
| | - Ioannis Papassotiriou
- Department of Clinical Biochemistry, "Aghia Sophia" Children's Hospital, Athens, Greece
| |
Collapse
|
21
|
Gonzales SK, Badell M, Cottrell H, Rimawi B, Deepak V, Sidell N, Rajakumar A. Villous explants from preeclamptic placentas induce sFlt1 in PBMCs: An ex vivo co-culture study. Pregnancy Hypertens 2018; 12:40-46. [PMID: 29674197 DOI: 10.1016/j.preghy.2018.02.006] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2017] [Revised: 02/13/2018] [Accepted: 02/15/2018] [Indexed: 12/18/2022]
Abstract
OBJECTIVES Soluble Flt1 (sFlt1) is an anti-angiogenic protein linked to the pathology of preeclampsia (PE). While the placenta serves as the major organ producing sFlt1 during normal pregnancy, peripheral blood mononuclear cells (PBMCs), endothelial cells, and stromal cells also produce sFlt1. The key question is 'what drives the overexpression of sFlt1 observed during PE?' In the present work we show evidence for sFlt1 over-expression in PBMCs due to interaction with placental villi from PE patients. STUDY DESIGN sFlt1 production by PBMCs is estimated by using two blood collection methods with different coagulation chemistry. PBMCs were then cultured with homologous villous explants and heterologous villous explants to determine the effects of the interaction between the two tissues. MAIN OUTCOME MEASURES sFlt1 levels were estimated using real time PCR, ELISA, and gel electrophoresis. RESULTS Plasma samples obtained using CTAD as anti-coagulant showed 16-23% less sFlt1 compared to plasma collected in EDTA. Preeclamptic PBMCs showed higher basal level of sFlt1 mRNA. In addition, we show evidence of placental interaction as a cause of sFlt1 overexpression in PBMCs using homologous and heterologous co-culture system. However, during co-culture, we observed that while the sFlt1 expression in PE PBMCs is increased, PE villous explants show reduced sFlt1 RNA expression. CONCLUSION sFlt1 was produced in significant amounts by preeclamptic PBMCs, and ex vivo studies show that the placenta induces this over-expression. In contrast, exposure to PBMCs appears to decrease sFlt1 production by preeclamptic placenta.
Collapse
Affiliation(s)
| | | | | | | | - Venkataraman Deepak
- Division of Research, Department of Gynecology and Obstetrics, Emory University School of Medicine, Atlanta, GA, USA
| | - Neil Sidell
- Division of Research, Department of Gynecology and Obstetrics, Emory University School of Medicine, Atlanta, GA, USA
| | - Augustine Rajakumar
- Division of Research, Department of Gynecology and Obstetrics, Emory University School of Medicine, Atlanta, GA, USA.
| |
Collapse
|
22
|
Mitsides N, Cornelis T, Broers NJH, Diederen NMP, Brenchley P, Heitink-Ter Braak N, van der Sande FM, Schalkwijk CG, Kooman JP, Mitra S. Inflammatory and Angiogenic Factors Linked to Longitudinal Microvascular Changes in Hemodialysis Patients Irrespective of Treatment Dose Intensity. Kidney Blood Press Res 2017; 42:905-918. [PMID: 29145197 DOI: 10.1159/000485048] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2017] [Accepted: 08/25/2017] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND Cardiovascular disease is a major contributor to the poor outcomes observed in hemodialysis. We investigated the relationship between hemodialysis intensity and vascular parameters in high-dose (HDHD; >12hrs/week) and Conventional (CHD; ≤12hrs/week) hemodialysis intensity over a 6-month period. METHODS We present the 6-month longitudinal analysis of a 2-year multicenter study investigating the effects of HDHD on cardiovascular parameters. We used pulse wave velocity, 24hr ambulatory blood pressure and sublingual dark field capillaroscopy measurements to assess macro- and microcirculation on 6-monthly basis. Pro-inflammatory and endothelial biomarkers were also measured at 6-monthly intervals. RESULTS 47 participants (21 HDHD, 26 CHD) were studied. CHD were older (63.5±14.2 vs 53.7±12.6 yr; p=0.018), with shorter dialysis vintage (median 23 vs 61 months; p=0.001). There was considerable variability in the degree and direction of change of circulatory measurements over a 6-month period. Hemodialysis intensity (hrs/week) did not correlate to these changes, when adjusted for age, dialysis vintage and comorbidity. Higher levels of Interleukin (IL)-8 measured at baseline independently predicted an increase in the Perfused Boundary Region (5-25μm) of the endothelial glycocalyx (p=0.010) whilst higher levels of soluble Flt-1 had a significant inverse effect (p=0.002) in an adjusted linear model. CONCLUSION Hemodialysis intensity did not predict changes in either macro- or microvascular parameters. Inflammation mediated through the IL-8 pathway predicted microvascular injury while Flt-1, a potential marker of angiogenesis and endothelial repair, might have a significant protective role. Further understanding of these pathways will be necessary to improve dialysis outcomes.
Collapse
Affiliation(s)
- Nicos Mitsides
- Division of Cardiovascular Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester Academic Health Science Centre, Manchester, United Kingdom.,Nephrology Department, Central Manchester University Hospital NHS Foundation Trust, Manchester Academic Health Science Centre, Manchester, United Kingdom.,NIHR Devices for Dignity Healthcare Technology Co-operative, Royal Hallamshire Hospital, Sheffield, United Kingdom
| | | | - Natascha J H Broers
- Department of Internal Medicine, Division of Nephrology, Maastricht University Medical Center, Maastricht, the Netherlands.,NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University, Maastricht, the Netherlands
| | - Nanada M P Diederen
- Department of Internal Medicine, Division of Nephrology, Maastricht University Medical Center, Maastricht, the Netherlands
| | - Paul Brenchley
- Division of Cardiovascular Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester Academic Health Science Centre, Manchester, United Kingdom.,Nephrology Department, Central Manchester University Hospital NHS Foundation Trust, Manchester Academic Health Science Centre, Manchester, United Kingdom
| | - Nicole Heitink-Ter Braak
- Department of Internal Medicine, Division of Nephrology, Zuyderland Medical Center, Heerlen-Geleen, the Netherlands
| | - Frank M van der Sande
- Department of Internal Medicine, Division of Nephrology, Maastricht University Medical Center, Maastricht, the Netherlands
| | - Casper G Schalkwijk
- Department of Internal Medicine, Division of Nephrology, Maastricht University Medical Center, Maastricht, the Netherlands.,CARIM School for Cardiovascular Diseases, Maastricht University, Maastricht, the Netherlands
| | - Jeroen P Kooman
- Department of Internal Medicine, Division of Nephrology, Maastricht University Medical Center, Maastricht, the Netherlands.,NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University, Maastricht, the Netherlands
| | - Sandip Mitra
- Division of Cardiovascular Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester Academic Health Science Centre, Manchester, United Kingdom.,Nephrology Department, Central Manchester University Hospital NHS Foundation Trust, Manchester Academic Health Science Centre, Manchester, United Kingdom.,NIHR Devices for Dignity Healthcare Technology Co-operative, Royal Hallamshire Hospital, Sheffield, United Kingdom
| |
Collapse
|
23
|
Akhter T, Wikström AK, Larsson M, Larsson A, Wikström G, Naessen T. Serum Pentraxin 3 is associated with signs of arterial alteration in women with preeclampsia. Int J Cardiol 2017; 241:417-422. [DOI: 10.1016/j.ijcard.2017.03.076] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/19/2016] [Revised: 03/07/2017] [Accepted: 03/10/2017] [Indexed: 10/19/2022]
|
24
|
Seno A, Takeda Y, Matsui M, Okuda A, Nakano T, Nakada Y, Kumazawa T, Nakagawa H, Nishida T, Onoue K, Somekawa S, Watanabe M, Kawata H, Kawakami R, Okura H, Uemura S, Saito Y. Suppressed Production of Soluble Fms-Like Tyrosine Kinase-1 Contributes to Myocardial Remodeling and Heart Failure. Hypertension 2016; 68:678-87. [PMID: 27480835 DOI: 10.1161/hypertensionaha.116.07371] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2016] [Accepted: 06/26/2016] [Indexed: 01/17/2023]
Abstract
Soluble fms-like tyrosine kinase-1 (sFlt-1), an endogenous inhibitor of vascular endothelial growth factor and placental growth factor, is involved in the pathogenesis of cardiovascular disease. However, the significance of sFlt-1 in heart failure has not been fully elucidated. We found that sFlt-1 is decreased in renal failure and serves as a key molecule in atherosclerosis. In this study, we aimed to investigate the role of the decreased sFlt-1 production in heart failure, using sFlt-1 knockout mice. sFlt-1 knockout mice and wild-type mice were subjected to transverse aortic constriction and evaluated after 7 days. The sFlt-1 knockout mice had significantly higher mortality (52% versus 15%; P=0.0002) attributable to heart failure and showed greater cardiac hypertrophy (heart weight to body weight ratio, 8.95±0.45 mg/g in sFlt-1 knockout mice versus 6.60±0.32 mg/g in wild-type mice; P<0.0001) and cardiac dysfunction, which was accompanied by a significant increase in macrophage infiltration and cardiac fibrosis, than wild-type mice after transverse aortic constriction. An anti-placental growth factor-neutralizing antibody prevented pressure overload-induced cardiac hypertrophy, fibrosis, and cardiac dysfunction. Moreover, monocyte chemoattractant protein-1 expression was significantly increased in the hypertrophied hearts of sFlt-1 knockout mice compared with wild-type mice. Monocyte chemoattractant protein-1 inhibition with neutralizing antibody ameliorated maladaptive cardiac remodeling in sFlt-1 knockout mice after transverse aortic constriction. In conclusion, decreased sFlt-1 production plays a key role in the aggravation of cardiac hypertrophy and heart failure through upregulation of monocyte chemoattractant protein-1 expression in pressure-overloaded heart.
Collapse
Affiliation(s)
- Ayako Seno
- From the First Department of Internal Medicine, Nara Medical University, Kashihara, Nara, Japan (A.S., Y.T., M.M., A.O., T. Nakano, Y.N., T.K., H.N., T. Nishida, K.O., S.S., M.W., H.K., R.K., H.O., S.U., Y.S.); and Department of Regulatory Medicine for Blood Pressure, Kashihara, Nara, Japan (T.K., Y.S.)
| | - Yukiji Takeda
- From the First Department of Internal Medicine, Nara Medical University, Kashihara, Nara, Japan (A.S., Y.T., M.M., A.O., T. Nakano, Y.N., T.K., H.N., T. Nishida, K.O., S.S., M.W., H.K., R.K., H.O., S.U., Y.S.); and Department of Regulatory Medicine for Blood Pressure, Kashihara, Nara, Japan (T.K., Y.S.)
| | - Masaru Matsui
- From the First Department of Internal Medicine, Nara Medical University, Kashihara, Nara, Japan (A.S., Y.T., M.M., A.O., T. Nakano, Y.N., T.K., H.N., T. Nishida, K.O., S.S., M.W., H.K., R.K., H.O., S.U., Y.S.); and Department of Regulatory Medicine for Blood Pressure, Kashihara, Nara, Japan (T.K., Y.S.)
| | - Aya Okuda
- From the First Department of Internal Medicine, Nara Medical University, Kashihara, Nara, Japan (A.S., Y.T., M.M., A.O., T. Nakano, Y.N., T.K., H.N., T. Nishida, K.O., S.S., M.W., H.K., R.K., H.O., S.U., Y.S.); and Department of Regulatory Medicine for Blood Pressure, Kashihara, Nara, Japan (T.K., Y.S.)
| | - Tomoya Nakano
- From the First Department of Internal Medicine, Nara Medical University, Kashihara, Nara, Japan (A.S., Y.T., M.M., A.O., T. Nakano, Y.N., T.K., H.N., T. Nishida, K.O., S.S., M.W., H.K., R.K., H.O., S.U., Y.S.); and Department of Regulatory Medicine for Blood Pressure, Kashihara, Nara, Japan (T.K., Y.S.)
| | - Yasuki Nakada
- From the First Department of Internal Medicine, Nara Medical University, Kashihara, Nara, Japan (A.S., Y.T., M.M., A.O., T. Nakano, Y.N., T.K., H.N., T. Nishida, K.O., S.S., M.W., H.K., R.K., H.O., S.U., Y.S.); and Department of Regulatory Medicine for Blood Pressure, Kashihara, Nara, Japan (T.K., Y.S.)
| | - Takuya Kumazawa
- From the First Department of Internal Medicine, Nara Medical University, Kashihara, Nara, Japan (A.S., Y.T., M.M., A.O., T. Nakano, Y.N., T.K., H.N., T. Nishida, K.O., S.S., M.W., H.K., R.K., H.O., S.U., Y.S.); and Department of Regulatory Medicine for Blood Pressure, Kashihara, Nara, Japan (T.K., Y.S.)
| | - Hitoshi Nakagawa
- From the First Department of Internal Medicine, Nara Medical University, Kashihara, Nara, Japan (A.S., Y.T., M.M., A.O., T. Nakano, Y.N., T.K., H.N., T. Nishida, K.O., S.S., M.W., H.K., R.K., H.O., S.U., Y.S.); and Department of Regulatory Medicine for Blood Pressure, Kashihara, Nara, Japan (T.K., Y.S.)
| | - Taku Nishida
- From the First Department of Internal Medicine, Nara Medical University, Kashihara, Nara, Japan (A.S., Y.T., M.M., A.O., T. Nakano, Y.N., T.K., H.N., T. Nishida, K.O., S.S., M.W., H.K., R.K., H.O., S.U., Y.S.); and Department of Regulatory Medicine for Blood Pressure, Kashihara, Nara, Japan (T.K., Y.S.)
| | - Kenji Onoue
- From the First Department of Internal Medicine, Nara Medical University, Kashihara, Nara, Japan (A.S., Y.T., M.M., A.O., T. Nakano, Y.N., T.K., H.N., T. Nishida, K.O., S.S., M.W., H.K., R.K., H.O., S.U., Y.S.); and Department of Regulatory Medicine for Blood Pressure, Kashihara, Nara, Japan (T.K., Y.S.)
| | - Satoshi Somekawa
- From the First Department of Internal Medicine, Nara Medical University, Kashihara, Nara, Japan (A.S., Y.T., M.M., A.O., T. Nakano, Y.N., T.K., H.N., T. Nishida, K.O., S.S., M.W., H.K., R.K., H.O., S.U., Y.S.); and Department of Regulatory Medicine for Blood Pressure, Kashihara, Nara, Japan (T.K., Y.S.)
| | - Makoto Watanabe
- From the First Department of Internal Medicine, Nara Medical University, Kashihara, Nara, Japan (A.S., Y.T., M.M., A.O., T. Nakano, Y.N., T.K., H.N., T. Nishida, K.O., S.S., M.W., H.K., R.K., H.O., S.U., Y.S.); and Department of Regulatory Medicine for Blood Pressure, Kashihara, Nara, Japan (T.K., Y.S.)
| | - Hiroyuki Kawata
- From the First Department of Internal Medicine, Nara Medical University, Kashihara, Nara, Japan (A.S., Y.T., M.M., A.O., T. Nakano, Y.N., T.K., H.N., T. Nishida, K.O., S.S., M.W., H.K., R.K., H.O., S.U., Y.S.); and Department of Regulatory Medicine for Blood Pressure, Kashihara, Nara, Japan (T.K., Y.S.)
| | - Rika Kawakami
- From the First Department of Internal Medicine, Nara Medical University, Kashihara, Nara, Japan (A.S., Y.T., M.M., A.O., T. Nakano, Y.N., T.K., H.N., T. Nishida, K.O., S.S., M.W., H.K., R.K., H.O., S.U., Y.S.); and Department of Regulatory Medicine for Blood Pressure, Kashihara, Nara, Japan (T.K., Y.S.)
| | - Hiroyuki Okura
- From the First Department of Internal Medicine, Nara Medical University, Kashihara, Nara, Japan (A.S., Y.T., M.M., A.O., T. Nakano, Y.N., T.K., H.N., T. Nishida, K.O., S.S., M.W., H.K., R.K., H.O., S.U., Y.S.); and Department of Regulatory Medicine for Blood Pressure, Kashihara, Nara, Japan (T.K., Y.S.)
| | - Shiro Uemura
- From the First Department of Internal Medicine, Nara Medical University, Kashihara, Nara, Japan (A.S., Y.T., M.M., A.O., T. Nakano, Y.N., T.K., H.N., T. Nishida, K.O., S.S., M.W., H.K., R.K., H.O., S.U., Y.S.); and Department of Regulatory Medicine for Blood Pressure, Kashihara, Nara, Japan (T.K., Y.S.)
| | - Yoshihiko Saito
- From the First Department of Internal Medicine, Nara Medical University, Kashihara, Nara, Japan (A.S., Y.T., M.M., A.O., T. Nakano, Y.N., T.K., H.N., T. Nishida, K.O., S.S., M.W., H.K., R.K., H.O., S.U., Y.S.); and Department of Regulatory Medicine for Blood Pressure, Kashihara, Nara, Japan (T.K., Y.S.).
| |
Collapse
|
25
|
Kirbas A, Kirbas O, Daglar K, Inal HA, Kurmus O, Kara O, Timur H, Gencosmanoglu G, Danisman N. Novel indexes of arrhythmogenesis in preeclampsia: QT dispersion, Tp-e interval, and Tp-e/QT ratio. Pregnancy Hypertens 2016; 6:38-41. [PMID: 26955770 DOI: 10.1016/j.preghy.2016.01.002] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2015] [Accepted: 01/19/2016] [Indexed: 01/13/2023]
Abstract
OBJECTIVE There is increasing evidence that preeclampsia (PE) may also be a risk factor for future cardiovascular diseases (CVDs), including arrhythmia. In this study we aimed to evaluate the association between PE and ventricular repolarization using novel electrocardiogram markers: QT interval duration, Tp-e interval, and Tp-e/QT ratio. MATERIALS AND METHODS In this controlled cross-sectional study sixty-four pregnant women with PE (31 with mild and 33 with severe disease) and 32 healthy women with uncomplicated pregnancies in the third trimester were compared by measuring QT parameters, Tp-e interval, and Tp-e/QT ratio. RESULTS Tp-e interval and Tp-e/QT ratio values were significantly higher in both the mild and severe PE groups compared to the healthy pregnant group. CONCLUSION Prevention of CVD requires that patients be aware of their risk factors, be educated about their risk, and perhaps most importantly perceive them to be at risk. In this study, we documented that PE has a significant effect on ventricular repolarization. This alteration could, in part, explain the increased cardiovascular risk in women with a history of PE. This important association can be used to screen women for increased risk in order to better target counseling regarding lifestyle modifications and to follow up and manage women with a history of hypertensive disease of pregnancy more closely.
Collapse
Affiliation(s)
- Ayse Kirbas
- Department of Perinatology, Zekai Tahir Burak Women's Health Education and Research Hospital, Ankara, Turkey.
| | - Ozgur Kirbas
- Department of Cardiology, Yuksek Ihtisas Education and Research Hospital, Ankara, Turkey.
| | - Korkut Daglar
- Department of Perinatology, Zekai Tahir Burak Women's Health Education and Research Hospital, Ankara, Turkey.
| | - Hasan Ali Inal
- Department of Obstetrics and Gynecology, Konya Education and Research Hospital, Konya, Turkey.
| | - Ozge Kurmus
- Department of Cardiology, Mersin State Hospital, Mersin, Turkey.
| | - Ozgur Kara
- Department of Perinatology, Zekai Tahir Burak Women's Health Education and Research Hospital, Ankara, Turkey.
| | - Hakan Timur
- Department of Perinatology, Zekai Tahir Burak Women's Health Education and Research Hospital, Ankara, Turkey.
| | - Gulenay Gencosmanoglu
- Department of Perinatology, Zekai Tahir Burak Women's Health Education and Research Hospital, Ankara, Turkey.
| | - Nuri Danisman
- Department of Perinatology, Zekai Tahir Burak Women's Health Education and Research Hospital, Ankara, Turkey.
| |
Collapse
|
26
|
Sinning C, Schnabel RB, Zeller T, Seiffert M, Rupprecht HJ, Lackner KJ, Blankenberg S, Bickel C, Westermann D. Prognostic use of soluble fms-like tyrosine kinase-1 and placental growth factor in patients with coronary artery disease. Biomark Med 2016; 10:95-106. [DOI: 10.2217/bmm.15.111] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Background: Intention of the study is to assess the cardiovascular mortality of patients with coronary artery disease (CAD) with the biomarkers of angiogenesis PlGF and its endogenous inhibitor sFlt-1. Methods: The cohort included n = 1848 patients with CAD and 282 subjects without CAD. In 85 patients cardiovascular mortality, as combination of fatal myocardial infarction or any cardiac death, during a median follow-up duration of 3.9 years was reported. Results: In Kaplan–Meier curve analysis PlGF in rising thirds was not predictive regarding outcome (p = 0.54), the same was shown for sFlt-1 (p = 0.44). Cox regression for the fully adjusted model provided a hazard ratio (HR) of 0.8 (p = 0.18) for PlGF and for sFlt-1 a HR = 1.0 (p = 0.8). Conclusion: Our results point out that these biomarkers reflecting angiogenesis might not be suited to establish prognosis in CAD.
Collapse
Affiliation(s)
- Christoph Sinning
- Department of General & Interventional Cardiology, University Heart Center Hamburg, Germany
| | - Renate B Schnabel
- Department of General & Interventional Cardiology, University Heart Center Hamburg, Germany
| | - Tanja Zeller
- Department of General & Interventional Cardiology, University Heart Center Hamburg, Germany
| | - Moritz Seiffert
- Department of General & Interventional Cardiology, University Heart Center Hamburg, Germany
| | - Hans J Rupprecht
- Department of Internal Medicine II, GPR Klinikum Rüsselsheim, Germany
| | - Karl J Lackner
- Department of Clinical Chemistry & Laboratory Medicine, Johannes Gu-tenberg-University Mainz, Germany
| | - Stefan Blankenberg
- Department of General & Interventional Cardiology, University Heart Center Hamburg, Germany
| | - Christoph Bickel
- Department of Internal Medicine, Federal Armed Forces Central Hospital, Koblenz, Germany
| | - Dirk Westermann
- Department of General & Interventional Cardiology, University Heart Center Hamburg, Germany
| |
Collapse
|
27
|
Matsui M, Uemura S, Takeda Y, Samejima KI, Matsumoto T, Hasegawa A, Tsushima H, Hoshino E, Ueda T, Morimoto K, Okamoto K, Okada S, Onoue K, Okayama S, Kawata H, Kawakami R, Maruyama N, Akai Y, Iwano M, Shiiki H, Saito Y. Placental Growth Factor as a Predictor of Cardiovascular Events in Patients with CKD from the NARA-CKD Study. J Am Soc Nephrol 2015; 26:2871-81. [PMID: 25788536 DOI: 10.1681/asn.2014080772] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2014] [Accepted: 01/20/2015] [Indexed: 01/04/2023] Open
Abstract
Placental growth factor (PlGF) contributes to atherogenesis through vascular inflammation and plaque destabilization. High levels of PlGF may be associated with mortality and cardiovascular disease, but the relationship between PlGF level and adverse outcomes in patients with CKD is unclear. We conducted a prospective cohort study of 1351 consecutive participants with CKD enrolled in the Novel Assessment of Risk management for Atherosclerotic diseases in CKD (NARA-CKD) study between April 1, 2004, and December 31, 2011. During a median follow-up of 3 years, 199 participants died and 383 had cardiovascular events, defined as atherosclerotic disease or heart failure requiring hospitalization. In adjusted analyses, mortality and cardiovascular risk increased in each successive quartile of serum PlGF level; hazard ratios (HRs) (95% confidence intervals [95% CIs]) for mortality and cardiovascular risk, respectively, were 1.59 (0.83 to 3.16) and 1.55 (0.92 to 2.66) for the second quartile, 2.97 (1.67 to 5.59) and 3.39 (2.20 to 5.41) for the third quartile, and 3.87 (2.24 to 7.08) and 8.42 (5.54 to 13.3) for the fourth quartile. The composite end point of mortality and cardiovascular events occurred during the study period in 76.4% of patients in both the highest PlGF quartile (≥19.6 pg/ml) and the lowest eGFR tertile (<30 ml/min per 1.73 m(2)). The association between PlGF and mortality or cardiovascular events was not attenuated when participants were stratified by age, sex, traditional risk factors, and eGFR. These data suggest elevated PlGF is an independent risk factor for all-cause mortality and cardiovascular events in patients with CKD.
Collapse
Affiliation(s)
| | | | | | - Ken-Ichi Samejima
- First Department of Internal Medicine and Department of Medicine, Ohyodo Town Hospital, Nara, Japan
| | | | | | - Hideo Tsushima
- Department of Medicine, Ohyodo Town Hospital, Nara, Japan
| | - Ei Hoshino
- Department of Medicine, Saisei-kai Nara City Hospital, Nara, Japan
| | | | | | | | | | | | | | | | | | - Naoki Maruyama
- Department of Cardiology, Nara City General Hospital, Nara, Japan
| | | | - Masayuki Iwano
- Department of Nephrology, Fukui University, Fukui, Japan; and
| | - Hideo Shiiki
- Department of Medicine, Uda City General Hospital, Nara, Japan
| | - Yoshihiko Saito
- First Department of Internal Medicine and Department of Regulatory Medicine for Blood Pressure, Nara Medical University, Nara, Japan;
| | | |
Collapse
|
28
|
Ratio between fms-like tyrosine kinase 1 and placental growth factor in children with congenital heart disease. Pediatr Cardiol 2015; 36:591-9. [PMID: 25388629 DOI: 10.1007/s00246-014-1054-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/11/2014] [Accepted: 10/31/2014] [Indexed: 12/30/2022]
Abstract
Serum levels of soluble fms-like tyrosine kinase 1 (sFlt-1), an antiangiogenic factor, and its binding protein, placental growth factor (PlGF), are altered in women with preeclampsia. Recently, the sFlt-1/PlGF ratio has been shown to predict acute coronary syndrome in adults. However, few reports have described the use of the sFlt-1/PlGF ratio for evaluating an abnormal hemodynamic load in children with congenital heart disease (CHD). The sFlt-1/PlGF ratio was determined in 20 children with atrial septal defects (ASD), 26 children with ventricular septal defects (VSD), 57 children with tetralogy of Fallot (ToF), 35 children who were Fontan candidates (Fontan), and 14 controls. The preoperative sFlt-1/PlGF ratios in the ASD, VSD, and Fontan were significantly higher than those in the controls and were significantly decreased after surgical repair in the ASD and VSD. In the ToF, the sFlt-1/PlGF ratio was highest after first-stage repair and second-highest after final-stage palliation compared with the preoperative levels. The sFlt-1/PlGF ratio was highest after first-stage repair and much lower after final-stage palliation in the Fontan. Furthermore, these ratios correlated with the degree of the ventricular volume overload and hypoxia. Our study clearly demonstrated that the sFlt-1/PlGF ratio increases with volume overload and persistent hypoxia after surgery with CHD. These findings may prove useful in the management of CHD in children.
Collapse
|
29
|
Rambod M, Heine GH, Seiler S, Dominic EA, Rogacev KS, Dwivedi R, Ramezani A, Wing MR, Amdur RL, Fliser D, Raj DS. Association of vascular endothelial factors with cardiovascular outcome and mortality in chronic kidney disease patients: a 4-year cohort study. Atherosclerosis 2014; 236:360-5. [PMID: 25128974 PMCID: PMC4327843 DOI: 10.1016/j.atherosclerosis.2014.07.026] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/11/2014] [Revised: 07/23/2014] [Accepted: 07/25/2014] [Indexed: 12/22/2022]
Abstract
BACKGROUND Angiogenic cytokines fms-like tyrosine kinase-1(sFlt-1) and placental growth factor (PlGF) are associated with increased risk for cardiovascular disease (CVD) in the general population. In this study we examine the association between these vascular endothelial factors and atherosclerosis, cardiovascular outcome, and mortality in chronic kidney disease (CKD) patients. METHODS Serum level of PlGF and sFlt-1 were measured in 301 patients with CKD, who were followed for up to 4 years. Primary outcomes were CV events and all-cause mortality. Carotid-intima media thickness (CIMT) was used as marker of atherosclerosis. Kaplan-Meier survival curves and the Cox proportional hazard model were used to assess the association of biomarkers and clinical outcomes. RESULTS Mean (SD) PlGF and sFlt-1 were 5.45 ng/ml (3.76) and 68.6 (28.0) pg/ml, respectively. During the follow up time, 60 patients (19.9%) experienced CV events and 22 patients (7.3%) died. Compared with low PlGF, patients with PlGF above median level had higher CV events (12.7% vs. 27.2%, p = 0.002) and mortality (2.0% vs. 12.6%, p < 0.001). The associations of PlGF and sFlt-1 with CV events were not statistically significant in the fully adjusted model. Higher PlGF was associated with greater death risk (HR = 5.22, 95% CI: 1.49-18.33, p = 0.01), which was robust to adjustment for sFlt-1 and other risk factors. Elevated sFlt-1 level was also an independent predictor of mortality (HR 3.41, 95% CI: 1.49-9.51, p = 0.019). CONCLUSION In CKD patients not yet on dialysis, higher serum level of PlGF and sFlt-1 are associated with increased mortality, but not CV events.
Collapse
Affiliation(s)
- Mehdi Rambod
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Paul L. Foster School of Medicine, El Paso, TX, USA
| | - Gunnar H. Heine
- Saarland University Medical Center and Saarland University Faculty of Medicine, Internal Medicine IV - Nephrology and Hypertension, Homburg/Saar, Germany
| | - Sarah Seiler
- Saarland University Medical Center and Saarland University Faculty of Medicine, Internal Medicine IV - Nephrology and Hypertension, Homburg/Saar, Germany
| | | | - Kyrill S. Rogacev
- Saarland University Medical Center and Saarland University Faculty of Medicine, Internal Medicine IV - Nephrology and Hypertension, Homburg/Saar, Germany
| | - Rama Dwivedi
- Division of Renal Diseases and Hypertension, The George Washington University School of Medicine, Washington, DC, USA
- Division of Cardiovascular and Renal Products, Office of New Drugs, US Food and Drug Administration, Silver Spring, MD, USA
| | - Ali Ramezani
- Division of Renal Diseases and Hypertension, The George Washington University School of Medicine, Washington, DC, USA
| | - Maria R. Wing
- Division of Renal Diseases and Hypertension, The George Washington University School of Medicine, Washington, DC, USA
| | | | - Danilo Fliser
- Saarland University Medical Center and Saarland University Faculty of Medicine, Internal Medicine IV - Nephrology and Hypertension, Homburg/Saar, Germany
| | - Dominic S Raj
- Division of Renal Diseases and Hypertension, The George Washington University School of Medicine, Washington, DC, USA
| |
Collapse
|
30
|
Koizumi M, Tatebe J, Watanabe I, Yamazaki J, Ikeda T, Morita T. Aryl hydrocarbon receptor mediates indoxyl sulfate-induced cellular senescence in human umbilical vein endothelial cells. J Atheroscler Thromb 2014; 21:904-16. [PMID: 24727683 DOI: 10.5551/jat.23663] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022] Open
Abstract
AIM Vascular senescence, which is accelerated in individuals with chronic kidney disease (CKD), contributes to the development of cardio-renal syndrome, and various uremic toxins may play important roles in the mechanisms underlying this phenomenon. We recently reported that indoxyl sulfate (IS), a uremic toxin, directly activates aryl hydrocarbon receptor (AhR) and generates oxidative stress through NADPH oxidase-4 in human umbilical vein endothelial cells (HUVECs). In the current study, we sought to examine whether IS regulates sirtuin 1 (Sirt1) and affects endothelial senescence via AhR activation. METHODS HUVECs were incubated with 500 μmol/L of IS for the indicated time periods. In order to evaluate changes in the senescence of the HUVECs, the number of senescence-associated β-galactosidase (SA β-gal)-positive cells was determined using an image analysis software program. The intracellular nicotinamide phosphoribosyltransferase (iNampt) activity, cellular NAD(+)/NADPH ratio and Sirt1 activity were analyzed according to a colorimetric assay to determine the mechanism of cellular senescence. Furthermore, we evaluated the involvement of AhR in the senescence-related changes induced by IS using AhR antagonists. RESULTS IS decreased the iNampt activity, NAD(+)/NADPH ratio and Sirt1 activity, resulting in an increase in the percentage of SA β-gal-positive cells. On the other hand, the AhR antagonists restored the IS-induced decrease in the NAD(+) content in association with an improvement in the iNampt activity and ameliorated the senescence-related changes. Taken together, these results indicate that IS impairs the iNampt-NAD(+)-Sirt1 system via AhR activation, which in turn promotes endothelial senescence. CONCLUSIONS The IS-AhR pathway induces endothelial senescence. Therefore, blocking the effects of AhR in the endothelium may provide a new therapeutic tool for treating cardio-renal syndrome.
Collapse
Affiliation(s)
- Masayuki Koizumi
- Department of Cardiovascular Medicine, Toho University Faculty of Medicine
| | | | | | | | | | | |
Collapse
|
31
|
Darbepoetin alpha reduces oxidative stress and chronic inflammation in atherosclerotic lesions of apo E deficient mice in experimental renal failure. PLoS One 2014; 9:e88601. [PMID: 24586350 PMCID: PMC3938414 DOI: 10.1371/journal.pone.0088601] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2013] [Accepted: 01/11/2014] [Indexed: 11/19/2022] Open
Abstract
Background Cardiovascular morbidity and mortality is very important in patients with chronic renal failure. This occurs even in mild impairment of renal function and may be related to oxidative stress and chronic inflammation. The nephrectomized apo E knockout mouse is an accepted model for evaluating atherosclerosis in renal dysfunction. Erythropoietin derivates showed anti-oxidative and anti-inflammatory effects. Therefore, this study evaluates the effects of Darbepoetin on markers of oxidative stress and chronic inflammation in atherosclerotic lesions in apo E knockout mice with renal dysfunction. Methods Apo E knockout mice underwent unilateral (Unx, n = 20) or subtotal (Snx, n = 26) nephrectomy or sham operation (Sham, n = 16). Mice of each group were either treated with Darbepoetin or saline solution, a part of Snx mice received a tenfold higher dose of Darbepoetin. The aortic plaques were measured and morphologically characterized. Additional immunhistochemical analyses were performed on tissue samples taken from the heart and the aorta. Results Both Unx and Snx mice showed increased expression of markers of oxidative stress and chronic inflammation. While aortic plaque size was not different, Snx mice showed advanced plaque stages when compared to Unx mice. Darbepoetin treatment elevated hematocrit and lowered Nitrotyrosin as one marker of oxidative stress, inflammation in heart and aorta, plaque stage and in the high dose even plaque cholesterol content. In contrast, there was no influence of Darbepoetin on aortic plaque size; high dose Darbepoetin treatment resulted in elevated renal serum parameters. Conclusion Darbepoetin showed some protective cardiovascular effects irrespective of renal function, i.e. it improved plaque structure and reduced some signs of oxidative stress and chronic inflammation without affecting plaque size. Nevertheless, the dose dependent adverse effects must be considered as high Darbepoetin treatment elevated serum urea. Elevation of hematocrit might be a favorable effect in anemic Snx animals but a thrombogenic risk in Sham animals.
Collapse
|
32
|
Soluble fms-like tyrosine kinase-1 and atherosclerosis in chronic kidney disease. Kidney Int 2014; 85:238-40. [DOI: 10.1038/ki.2013.402] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
|
33
|
Zakiyanov O, Kriha V, Vachek J, Zima T, Tesar V, Kalousova M. Placental growth factor, pregnancy-associated plasma protein-A, soluble receptor for advanced glycation end products, extracellular newly identified receptor for receptor for advanced glycation end products binding protein and high mobility group box 1 levels in patients with acute kidney injury: a cross sectional study. BMC Nephrol 2013; 14:245. [PMID: 24188108 PMCID: PMC4228333 DOI: 10.1186/1471-2369-14-245] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2013] [Accepted: 10/23/2013] [Indexed: 11/17/2022] Open
Abstract
BACKGROUND Placental growth factor (PlGF), pregnancy-associated plasma protein-A (PAPP-A), soluble receptor for advanced glycation end products (sRAGE), extracellular newly identified receptor for RAGE binding protein (EN-RAGE) and high mobility group box 1 (HMGB-1) are novel biomarkers in chronic kidney disease (CKD). However, their clinical significance in acute kidney injury (AKI) is unknown. The aim of this cross-sectional study was to determine whether selected biomarkers are changed in AKI patients. METHODS Serum PlGF, PAPP-A, sRAGE, EN-RAGE and HMGB-1 levels were assessed in 40 patients with AKI, 42 CKD 5 patients, 31 haemodialysis patients (HD) and 39 age-matched healthy controls. RESULTS PAPP-A was elevated in AKI (20.6 ± 16.9 mIU/L) compared with controls (9.1 ± 2.3 mIU/L, p < 0.001). PlGF was not increased in AKI (11.7 ± 7.4 pg/mL) versus controls (8.5 ± 2.4 pg/mL, n.s.), as well as sRAGE was not elevated in AKI (2400 ± 1400 pg/mL) compared with controls (1760 ± 730 pg/mL, n.s), but was lower compared with CKD 5 (3200 ± 1500 pg/mL, p < 0.05); EN-RAGE was elevated in AKI 480 ± 450 ng/mL in comparison with controls (60 ± 62 ng/mL), CKD 5 (190 ± 120 ng/mL), and HD (120 ± 100 ng/mL), all p < 0.001. Similarly, HMGB-1 was increased in AKI (5.8 ± 7.5 ng/mL) versus controls (1.7 ± 1.4 ng/mL), CKD 5 (3.2 ± 3.1 ng/mL) and HD (2.5 ± 2.1 ng/mL), all p < 0.001.In AKI group, in multivariate regression analysis: PAPP-A levels were associated with transferrin (p <0.001), negatively with albumin (p < 0.01) and prealbumin (p < 0.05); PlGF levels were associated with C--reactive protein (p < 0.001). EN-RAGE levels were associated with ferritin (p < 0.01) and orosomucoid (p = 0.02), and HMGB-1 levels with leukocyte count (p < 0.01) and negatively with proteinuria (p = 0.02). CONCLUSIONS In AKI patients, PAPP-A, EN-RAGE and HMGB1 are elevated, but sRAGE and PlGF are not increased. Whereas PAPP-A correlates with markers of nutrition; PlGF, EN-RAGE and HMGB-1 are related to inflammatory parameters.
Collapse
Affiliation(s)
- Oskar Zakiyanov
- Department of Nephrology, First Faculty of Medicine, Charles University in Prague and General University Hospital in Prague, Prague, Czech Republic
- Institute of Medical Biochemistry and Laboratory Diagnostics, First Faculty of Medicine, Charles University in Prague and General University Hospital in Prague, Prague, Czech Republic
| | - Vitezslav Kriha
- Department of Physics, Faculty of Electrical Engineering, Czech Technical University in Prague, Prague, Czech Republic
| | - Jan Vachek
- Department of Nephrology, First Faculty of Medicine, Charles University in Prague and General University Hospital in Prague, Prague, Czech Republic
- Institute of Pharmacology, First Faculty of Medicine, Charles University in Prague, Prague, Czech Republic
| | - Tomas Zima
- Institute of Medical Biochemistry and Laboratory Diagnostics, First Faculty of Medicine, Charles University in Prague and General University Hospital in Prague, Prague, Czech Republic
| | - Vladimir Tesar
- Department of Nephrology, First Faculty of Medicine, Charles University in Prague and General University Hospital in Prague, Prague, Czech Republic
| | - Marta Kalousova
- Institute of Medical Biochemistry and Laboratory Diagnostics, First Faculty of Medicine, Charles University in Prague and General University Hospital in Prague, Prague, Czech Republic
| |
Collapse
|
34
|
Increased Soluble Flt-1 Correlates With Delayed Graft Function and Early Loss of Peritubular Capillaries in the Kidney Graft. Transplantation 2013; 96:739-44. [DOI: 10.1097/tp.0b013e31829f4772] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
35
|
Abstract
At least 468 individual genes have been manipulated by molecular methods to study their effects on the initiation, promotion, and progression of atherosclerosis. Most clinicians and many investigators, even in related disciplines, find many of these genes and the related pathways entirely foreign. Medical schools generally do not attempt to incorporate the relevant molecular biology into their curriculum. A number of key signaling pathways are highly relevant to atherogenesis and are presented to provide a context for the gene manipulations summarized herein. The pathways include the following: the insulin receptor (and other receptor tyrosine kinases); Ras and MAPK activation; TNF-α and related family members leading to activation of NF-κB; effects of reactive oxygen species (ROS) on signaling; endothelial adaptations to flow including G protein-coupled receptor (GPCR) and integrin-related signaling; activation of endothelial and other cells by modified lipoproteins; purinergic signaling; control of leukocyte adhesion to endothelium, migration, and further activation; foam cell formation; and macrophage and vascular smooth muscle cell signaling related to proliferation, efferocytosis, and apoptosis. This review is intended primarily as an introduction to these key signaling pathways. They have become the focus of modern atherosclerosis research and will undoubtedly provide a rich resource for future innovation toward intervention and prevention of the number one cause of death in the modern world.
Collapse
Affiliation(s)
- Paul N Hopkins
- Cardiovascular Genetics, Department of Internal Medicine, University of Utah, Salt Lake City, Utah, USA.
| |
Collapse
|
36
|
Suppressed soluble Fms-like tyrosine kinase-1 production aggravates atherosclerosis in chronic kidney disease. Kidney Int 2013; 85:393-403. [PMID: 24048373 DOI: 10.1038/ki.2013.339] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2013] [Revised: 06/19/2013] [Accepted: 06/27/2013] [Indexed: 12/27/2022]
Abstract
Patients with chronic kidney disease (CKD) die of cardiovascular diseases for unknown reasons. Blood vessel formation in plaques and its relationship with plaque stability could be involved with signaling through the Flt-1 receptor and its ligands, vascular endothelial growth factor, and the closely related placental growth factor (PlGF). Flt-1 also exists as a circulating regulatory splice variant short-inhibitory form (sFlt-1) that serves as a decoy receptor, thereby inactivating PlGF. Heparin releases sFlt-1 by displacing the sFlt-1 heparin-binding site from heparin sulfate proteoglycans. Heparin could provide diagnostic inference or could also induce an antiangiogenic state. In the present study, postheparin sFlt-1 levels were lower in CKD patients than in control subjects. More importantly, sFlt-1 levels were inversely related to atherosclerosis in CKD patients, and this correlation was more robust after heparin injection, as verified by subsequent cardiovascular events. Knockout of apolipoprotein E (ApoE) and/or sFlt-1 showed that the absence of sFlt-1 worsened atherogenesis in ApoE-deficient mice. Thus, the relationship between atherosclerosis and PlGF signaling, as regulated by sFlt-1, underscores the underappreciated role of heparin in sFlt-1 release. These clinical and experimental data suggest that novel avenues into CKD-dependent atherosclerosis and its detection are warranted.
Collapse
|
37
|
Kameda R, Yamaoka-Tojo M, Makino A, Wakaume K, Nemoto S, Kitasato L, Shimohama T, Tojo T, Machida Y, Izumi T. Soluble Fms-like tyrosine kinase 1 is a novel predictor of brain natriuretic peptide elevation. Int Heart J 2013; 54:133-9. [PMID: 23774235 DOI: 10.1536/ihj.54.133] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Soluble fms-like tyrosine kinase 1 (sFlt-1) is an endogenous inhibitor of vascular endothelial growth factor, which is involved in cardiovascular remodeling and atherosclerosis development. To examine the predictive role of sFlt-1 levels in patients with asymptomatic heart failure, we measured circulating sFlt-1 in patients with or without coronary artery disease (CAD). We analyzed 88 Japanese patients with CAD or patients at high risk for atherosclerosis and who were undergoing total risk management for cardiovascular disease prevention. Circulating sFlt-1 levels correlated with the increase in plasma brain natriuretic peptide levels (ΔBNP) from baseline to the observed levels 5 years later in CAD patients, patients with previous myocardial infarction, and men. ΔBNP levels correlated with sFlt-1 levels in the high-sFlt-1 patients with CAD (r = 0.511, P < 0.01). In all patients, end-systolic volume index (ΔESVI) increased in correlation with a decrease in left ventricular ejection fraction (ΔEF) in the long-term observation, independent of their history of myocardial infarction (ΔESVI = 2.5 mL/m(2) increase/year). Baseline level of sFlt-1 was independent of ΔESVI or ΔEF. The present 5-year observational study demonstrated that high sFlt-1 levels predicted moderate increases in BNP levels in CAD patients. Moreover, ΔBNP was correlated with ΔESVI/year in CAD patients with high-sFlt-1 levels. These data suggest that high sFlt-1 levels may be an effective biomarker to predict the progression of heart failure in patients with CAD.
Collapse
Affiliation(s)
- Ryo Kameda
- Kitasato University Graduate School of Medical Sciences, Japan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Kawahito H, Yamada H, Irie D, Kato T, Akakabe Y, Kishida S, Takata H, Wakana N, Ogata T, Ikeda K, Ueyama T, Matoba S, Mori Y, Matsubara H. Periaortic adipose tissue-specific activation of the renin-angiotensin system contributes to atherosclerosis development in uninephrectomized apoE-/- mice. Am J Physiol Heart Circ Physiol 2013; 305:H667-75. [PMID: 23812390 DOI: 10.1152/ajpheart.00053.2013] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Chronic kidney disease (CKD) is an independent risk factor for the development of cardiovascular disease. The perivascular adipose tissue is closely implicated in the development of atherosclerosis; however, the contribution to CKD-associated atherogenesis remains undefined. Eight-week-old apoE-deficient mice were uninephrectomized and fed a high-cholesterol diet starting at 12 wk of age. The atherosclerotic lesion area in the thoracic aorta was comparable in 16-wk-old uninephrectomized (UNX) mice and sham control mice; however, the lesion area was markedly exaggerated in 20-wk-old UNX mice compared with the control (54%, P < 0.05). While the accumulation of monocytes/macrophages and the mRNA expression levels of inflammatory cytokines/chemokines in the thoracic periaortic adipose tissue (PAT) did not differ between the two groups, angiotensinogen (AGT) mRNA expression and the angiotensin II (ANG II) concentration in the PAT were significantly higher in 16-wk-old UNX mice than in the control (1.9- and 1.5-fold increases vs. control, respectively; P < 0.05). ANG II concentrations in both the plasma and epididymal white adipose tissue (WAT) were comparable between the two groups, suggesting that PAT-specific activation of the renin-angiotensin system (RAS) is primarily involved in CKD-associated atherogenesis. The homeostasis model assessment-insulin resistance (HOMA-IR) index and plasma insulin level after glucose loading were significantly elevated in 16-wk-old UNX mice. In vitro stimulation of preadipocytes with insulin exaggerated the AGT mRNA expression along with increased mRNA expression of PPARγ. These findings suggest that PAT-specific RAS activation probably primarily contributes in accelerating atherosclerotic development in UNX mice and could thus represent a therapeutic target for preventing CKD-associated atherogenesis.
Collapse
Affiliation(s)
- Hiroyuki Kawahito
- Department of Cardiovascular Medicine, Kyoto Prefectural University of Medicine, Kyoto, Japan; and
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Matsumoto T, Uemura S, Takeda Y, Matsui M, Okada S, Nishida T, Soeda T, Okayama S, Somekawa S, Ishigami KI, Onoue K, Kawata H, Kawakami R, Horii M, Saito Y. An elevated ratio of placental growth factor to soluble fms-like tyrosine kinase-1 predicts adverse outcomes in patients with stable coronary artery disease. Intern Med 2013; 52:1019-27. [PMID: 23676585 DOI: 10.2169/internalmedicine.52.9073] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
OBJECTIVE To investigate the predictive values of placental growth factor (PlGF) and its endogenous antagonist, soluble fms-like tyrosine kinase-1 (sFlt-1), for the long-term prognosis of patients with stable coronary artery disease (CAD). Both PlGF and sFlt-1 play important roles in the pathological mechanisms of atherosclerosis. We recently demonstrated that the plasma levels of these molecules are correlated with the severity of coronary atherosclerosis. METHODS We enrolled 464 patients with stable CAD who consecutively underwent coronary angiography. Baseline blood samples were collected from the femoral artery immediately before coronary angiography (after the administration of 20 units of heparin), and the plasma levels of PlGF and sFlt-1 were measured. A Cox proportional hazard regression analysis was performed to evaluate the relationship between these parameters and the occurrence of all-cause death (ACD) and total cardiovascular events (TCVE) during a median follow-up of 3.3 years. RESULTS A total of 31 ACDs and 51 TCVEs occurred. Patients with higher PlGF/sFlt-1 ratios (>4.22×10(-2)) had a significantly higher risk of both ACD and TCVE than patients with lower ratios (<4.22×10(-2)) (hazard ratio [HR]: 3.32, 95% confidence interval [CI]: 1.43 to 7.72, p=0.005, and HR: 2.23, 95% CI: 1.23 to 4.03, p=0.008, respectively). A multivariate analysis showed the PlGF/sFlt-1 ratio to be an independent predictor for ACD, but not TCVE. CONCLUSION The baseline PlGF/sFlt-1 ratio is an independent predictor of long-term adverse outcomes in patients with stable CAD.
Collapse
Affiliation(s)
- Takaki Matsumoto
- The First Department of Internal Medicine, Nara Medical University, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Dewerchin M, Carmeliet P. PlGF: a multitasking cytokine with disease-restricted activity. Cold Spring Harb Perspect Med 2012; 2:cshperspect.a011056. [PMID: 22908198 DOI: 10.1101/cshperspect.a011056] [Citation(s) in RCA: 161] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Placental growth factor (PlGF) is a member of the vascular endothelial growth factor (VEGF) family that also comprises VEGF-A (VEGF), VEGF-B, VEGF-C, and VEGF-D. Unlike VEGF, PlGF is dispensable for development and health but has diverse nonredundant roles in tissue ischemia, malignancy, inflammation, and multiple other diseases. Genetic and pharmacological gain-of-function and loss-of-function studies have identified molecular mechanisms of this multitasking cytokine and characterized the therapeutic potential of delivering or blocking PlGF for various disorders.
Collapse
Affiliation(s)
- Mieke Dewerchin
- Laboratory of Angiogenesis and Neurovascular Link, VIB Vesalius Research Center, K.U. Leuven, Leuven, Belgium
| | | |
Collapse
|
41
|
Ky B, French B, Ruparel K, Sweitzer NK, Fang JC, Levy WC, Sawyer DB, Cappola TP. The vascular marker soluble fms-like tyrosine kinase 1 is associated with disease severity and adverse outcomes in chronic heart failure. J Am Coll Cardiol 2011; 58:386-94. [PMID: 21757116 DOI: 10.1016/j.jacc.2011.03.032] [Citation(s) in RCA: 67] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/16/2011] [Revised: 02/16/2011] [Accepted: 03/07/2011] [Indexed: 12/19/2022]
Abstract
OBJECTIVES We sought to evaluate placental growth factor (PlGF) and soluble Fms-like tyrosine kinase 1 (sFlt-1) as clinical biomarkers in chronic heart failure (HF). BACKGROUND Vascular remodeling is a crucial compensatory mechanism in chronic HF. The angiogenic ligand PlGF and its target receptor fms-like tyrosine kinase 1 modulate vascular growth and function, but their relevance in human HF is undefined. METHODS We measured plasma PlGF and sFlt-1 in 1,403 patients from the Penn Heart Failure Study, a multicenter cohort of chronic systolic HF. Subjects were followed for death, cardiac transplantation, or ventricular assist device placement over a median follow-up of 2 years. RESULTS The sFlt-1 was independently associated with measures of HF severity, including New York Heart Association functional class (p < 0.01) and B-type natriuretic peptide (p < 0.01). Patients in the 4th quartile of sFlt-1 (>379 pg/ml) had a 6.17-fold increased risk of adverse outcomes (p < 0.01). This association was robust, even after adjustment for the Seattle Failure Model (hazard ratio: 2.54, 95% confidence interval [CI]: 1.76 to 2.27, p < 0.01) and clinical confounders including HF etiology (hazard ratio: 1.67, 95% CI: 1.06 to 2.63, p = 0.03). Combined assessment of sFlt-1 and B-type natriuretic peptide exhibited high predictive accuracy at 1 year (area under the receiver-operator characteristic curve: 0.791, 95% CI: 0.752 to 0.831) that was greater than either marker alone (p < 0.01 and p = 0.03, respectively). In contrast, PlGF was not an independent marker of disease severity or outcomes. CONCLUSIONS Our findings support a role for sFlt-1 in the biology of human HF. With additional study, circulating sFlt-1 might emerge as a clinically useful biomarker to assess the influence of vascular remodeling on clinical outcomes.
Collapse
Affiliation(s)
- Bonnie Ky
- Penn Cardiovascular Institute, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA.
| | | | | | | | | | | | | | | |
Collapse
|
42
|
Charytan DM, Helfand AM, MacDonald BA, Cinelli A, Kalluri R, Zeisberg EM. Circulating endoglin concentration is not elevated in chronic kidney disease. PLoS One 2011; 6:e23718. [PMID: 21886815 PMCID: PMC3158786 DOI: 10.1371/journal.pone.0023718] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2011] [Accepted: 07/25/2011] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND Soluble endoglin, a TGF-β receptor, plays a key role in cardiovascular physiology. Whether circulating concentrations of soluble endoglin are elevated in CKD or underlie the high risk of cardiovascular death associated with chronic kidney disease (CKD) is unknown. METHODS Individuals with and without CKD were recruited at a single center. Estimated glomerular filtration rate (eGFR) was estimated using the modified MDRD study equation and the serum creatinine at the time of recruitment, and patients were assigned to specific CKD stage according to usual guidelines. Serum endoglin concentration was measured by ELISA and univariate and multivariable regression was used to analyze the association between eGFR or CKD stage and the concentration of soluble endoglin. RESULTS Serum endoglin was measured in 216 patients including 118 with stage 3 or higher CKD and 9 individuals with end stage renal disease (ESRD). Serum endoglin concentration did not vary significantly with CKD stage (increase of 0.16 ng/mL per 1 stage increase in CKD, P = 0.09) or eGFR (decrease -0.06 ng/mL per 10 mL/min/1.73 m(2) increase in GFR, P = 0.12), and was not higher in individuals with ESRD than in individuals with preserved renal function (4.2±1.1 and 4.3±1.2 ng/mL, respectively). Endoglin concentration was also not significantly associated with urinary albumin excretion. CONCLUSIONS Renal function is not associated with the circulating concentration of soluble endoglin. Elevations in soluble endoglin concentration are unlikely to contribute to the progression of CKD or the predisposition of individuals with CKD to develop cardiovascular disease.
Collapse
Affiliation(s)
- David M Charytan
- Renal Division, Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts, United States of America.
| | | | | | | | | | | |
Collapse
|
43
|
Zakiyanov O, Kalousová M, Zima T, Tesař V. Placental growth factor in patients with decreased renal function. Ren Fail 2011; 33:291-7. [PMID: 21401353 DOI: 10.3109/0886022x.2011.560402] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
BACKGROUND Patients with decreased renal function are characterized by high cardiovascular morbidity and mortality due to complications of premature atherosclerosis. Placental growth factor (PlGF) is a proatherogenic cytokine and new biomarker of cardiovascular events. The aim of this study was to determine PlGF levels and describe their relationship to renal function and risk factors of atherogenesis in patients with decreased renal function. METHODS The study group consisted of 114 subjects: 45 patients with various degrees of decreased renal function (CHRI), 31 long-term hemodialysis (HD) patients, and 38 age-matched healthy control subjects. PlGF was assessed immunochemically (enzyme-linked immunosorbent assay) and routine biochemical parameters were measured using standard laboratory methods. RESULTS PlGF levels were significantly increased in CHRI and HD patients compared to controls (10.5 ± 3.3 pg/mL in CHRI patients and 11.5 ± 3.4 pg/mL HD patients vs. 8.1 ± 1.8 pg/mL in controls, both p < 0.0001). In CHRI patients, PlGF was detectable in the urine, and its urine concentration correlated with its serum levels. In HD patients, PlGF correlated with low-density lipoproteins (r = 0.36, p < 0.05), but was not related to C-reactive protein levels. Higher levels of PlGF were found in CHRI patients with cardiovascular disease, compared with those free of such complication. CONCLUSIONS PlGF levels are increased in patients with decreased kidney function. PlGF is detectable in the urine, and serum and urine levels of PlGF are significantly interrelated. It is higher in CHRI patients with cardiovascular disease. Further studies are required to demonstrate the usefulness and significance of PlGF in patients with chronic kidney disease.
Collapse
Affiliation(s)
- Oskar Zakiyanov
- Department of Nephrology, First Faculty of Medicine, Charles University in Prague and General University Hospital in Prague, Prague, Czech Republic
| | | | | | | |
Collapse
|
44
|
Shin S, Lee SH, Park S, Kang SM, Chung N, Shim WH, Cho SY, Manabe I, Jang Y. Soluble fms-like tyrosine kinase-1 and the progression of carotid intima-media thickness – 24-month follow-up study –. Circ J 2010; 74:2211-5. [PMID: 20689217 DOI: 10.1253/circj.cj-10-0432] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
BACKGROUND The relationship between fms-like tyrosine kinase-1 (sFlt-1), a soluble receptor for vascular endothelial growth factor (VEGF), and vascular disease has not been established, so this study aimed to elucidate the association between sFlt-1 and the progression of carotid intima - media thickness (IMT) in hypertensive patients. METHODS AND RESULTS The 120 hypertensive patients under medical control were enrolled and 112 completed the study (age 59 ± 9 years, 57 females). Plasma VEGF and sFlt-1 levels were measured at enrollment. At baseline and 24-month visit, carotid IMT was measured and the association between sFlt-1 and IMT progression was assessed by linear regression. At baseline, age (r=0.186) and low level of high-density lipoprotein-cholesterol (HDL-C <40 mg/dl, r=0.214) were significantly related to carotid IMT. Over the 24 months, carotid IMT increased from 0.670 ± 0.089 mm to 0.696 ± 0.095 mm. There was a positive correlation between sFlt-1 tertiles and IMT change (P=0.05 by ANOVA). Upon multivariate analysis, log-transformed sFlt-1 level (β=0.137, P=0.003) and low HDL-C (β=0.048, P=0.04) were identified as predictors of IMT progression, independent of other confounding variables. CONCLUSIONS High sFlt-1 level is predictive of carotid IMT progression in hypertensive patients. Low HDL-C level was also associated with IMT change. These observations support a high sFlt-1 level being indicative of progression of atherosclerosis.
Collapse
Affiliation(s)
- Sanghoon Shin
- Cardiology Division, Department of Internal Medicine, Yonsei University College of Medicine, Seoul, Republic of Korea
| | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Kim SY, Lee SH, Park S, Kang SM, Chung N, Shim WH, Cho SY, Sun Ha Jee, Manabe I, Jang Y. Vascular Endothelial Growth Factor, Soluble Fms-Like Tyrosine Kinase 1, and the Severity of Coronary Artery Disease. Angiology 2010; 62:176-83. [DOI: 10.1177/0003319710370963] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
Background: The association between vascular endothelial growth factor (VEGF), soluble fms-like tyrosine kinase 1 (sFlt-1), and coronary artery disease (CAD) was investigated. Methods: We enrolled 112 hypertensive patients with proven CAD and 112 hypertensive controls matched for age and gender. The severity of CAD was assessed by the most severe clinical presentation of CAD in patients’ history and by the number of diseased vessels. Results: Vascular endothelial growth factor level was lower, whereas sFlt-1 level was higher in the CAD group compared to the controls. Diabetes mellitus (P = .001), smoking (P = .004), and higher sFlt-1 level (P = .01) were independently associated with CAD. Younger age (P = .02), smoking (P = .049), and higher VEGF levels (P = .02) were independently associated with a history of myocardial infarction (MI), whereas higher sFlt-1 level (P = .01) was independently associated with multivessel disease. Conclusion: Plasma sFlt-1 levels are positively associated with the presence of CAD and are associated with angiographical severity of CAD.
Collapse
Affiliation(s)
- Soo-Young Kim
- Cardiology Division, Department of Internal Medicine, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Sang-Hak Lee
- Cardiology Division, Department of Internal Medicine, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Sungha Park
- Cardiology Division, Department of Internal Medicine, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Seok-Min Kang
- Cardiology Division, Department of Internal Medicine, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Namsik Chung
- Cardiology Division, Department of Internal Medicine, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Won-Heum Shim
- Cardiology Division, Department of Internal Medicine, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Seung-Yun Cho
- Cardiology Division, Department of Internal Medicine, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Sun Ha Jee
- Department of Epidemiology and Health Promotion, Graduate School of Public Health, Yonsei University, Seoul, Republic of Korea
| | - Ichiro Manabe
- Department of Cardiovascular Medicine and Nano-Bioengineering Education Program, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Yangsoo Jang
- Cardiology Division, Department of Internal Medicine, Yonsei University College of Medicine, Seoul, Republic of Korea,
| |
Collapse
|
46
|
Affiliation(s)
- Burak Pamukcu
- University of Birmingham Centre for Cardiovascular Sciences, City Hospital
| | - Eduard Shantsila
- University of Birmingham Centre for Cardiovascular Sciences, City Hospital
| | - Gregory Y.H. Lip
- University of Birmingham Centre for Cardiovascular Sciences, City Hospital
| |
Collapse
|