1
|
Mansutti G, Villiger M, Bouma BE, Uribe-Patarroyo N. Full-field amplitude speckle decorrelation angiography. BIOMEDICAL OPTICS EXPRESS 2024; 15:5756-5772. [PMID: 39421771 PMCID: PMC11482163 DOI: 10.1364/boe.530993] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Revised: 07/27/2024] [Accepted: 07/27/2024] [Indexed: 10/19/2024]
Abstract
We propose a new simple and cost-effective optical imaging technique, full-field amplitude speckle decorrelation angiography (FASDA), capable of visualizing skin microvasculature with high resolution, and sensitive to small, superficial vessels with slow blood flow and larger, deeper vessels with faster blood flow. FASDA makes use of a laser source with limited temporal coherence, can be implemented with cameras with conventional frame rates, and does not require raster scanning. The proposed imaging technique is based on the simultaneous evaluation of two metrics: the blood flow index, a contrast-based metric used in laser speckle contrast imaging, and the adaptive speckle decorrelation index (ASDI), a new metric that we defined based on the second-order autocorrelation function that considers the limited speckle modulation that occurs in partially-coherent imaging. We demonstrate excellent delineation of small, superficial vessels with slow blood flow in skin nevi using ASDI and larger, deeper vessels with faster blood flow using BFI, providing a powerful new tool for the imaging of microvasculature with significantly lower hardware complexity and cost than other optical imaging techniques.
Collapse
Affiliation(s)
- Giulia Mansutti
- Wellman Center for Photomedicine, Massachusetts General Hospital, Boston, MA, USA
| | - Martin Villiger
- Wellman Center for Photomedicine, Massachusetts General Hospital, Boston, MA, USA
| | - Brett E. Bouma
- Wellman Center for Photomedicine, Massachusetts General Hospital, Boston, MA, USA
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA, USA
| | | |
Collapse
|
2
|
Lima Posada I, Soulié M, Stephan Y, Palacios Ramirez R, Bonnard B, Nicol L, Pitt B, Kolkhof P, Mulder P, Jaisser F. Nonsteroidal Mineralocorticoid Receptor Antagonist Finerenone Improves Diastolic Dysfunction in Preclinical Nondiabetic Chronic Kidney Disease. J Am Heart Assoc 2024; 13:e032971. [PMID: 38842271 PMCID: PMC11255738 DOI: 10.1161/jaha.123.032971] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/22/2023] [Accepted: 03/15/2024] [Indexed: 06/07/2024]
Abstract
BACKGROUND The mineralocorticoid receptor plays a significant role in the development of chronic kidney disease (CKD) and associated cardiovascular complications. Classic steroidal mineralocorticoid receptor antagonists are a therapeutic option, but their use in the clinic is limited due to the associated risk of hyperkalemia in patients with CKD. Finerenone is a nonsteroidal mineralocorticoid receptor antagonist that has been recently investigated in 2 large phase III clinical trials (FIDELIO-DKD [Finerenone in Reducing Kidney Failure and Disease Progression in Diabetic Kidney Disease] and FIGARO-DKD [Finerenone in Reducing Cardiovascular Mortality and Morbidity in Diabetic Kidney Disease]), showing reductions in kidney and cardiovascular outcomes. METHODS AND RESULTS We tested whether finerenone improves renal and cardiac function in a preclinical nondiabetic CKD model. Twelve weeks after 5/6 nephrectomy, the rats showed classic signs of CKD characterized by a reduced glomerular filtration rate and increased kidney weight, associated with left ventricular (LV) diastolic dysfunction and decreased LV perfusion. These changes were associated with increased cardiac fibrosis and reduced endothelial nitric oxide synthase activating phosphorylation (ser 1177). Treatment with finerenone prevented LV diastolic dysfunction and increased LV tissue perfusion associated with a reduction in cardiac fibrosis and increased endothelial nitric oxide synthase phosphorylation. Curative treatment with finerenone improves nondiabetic CKD-related LV diastolic function associated with a reduction in cardiac fibrosis and increased cardiac phosphorylated endothelial nitric oxide synthase independently from changes in kidney function. Short-term finerenone treatment decreased LV end-diastolic pressure volume relationship and increased phosphorylated endothelial nitric oxide synthase and nitric oxide synthase activity. CONCLUSIONS We showed that the nonsteroidal mineralocorticoid receptor antagonist finerenone reduces renal hypertrophy and albuminuria, attenuates cardiac diastolic dysfunction and cardiac fibrosis, and improves cardiac perfusion in a preclinical nondiabetic CKD model.
Collapse
MESH Headings
- Animals
- Mineralocorticoid Receptor Antagonists/pharmacology
- Mineralocorticoid Receptor Antagonists/therapeutic use
- Renal Insufficiency, Chronic/drug therapy
- Renal Insufficiency, Chronic/physiopathology
- Renal Insufficiency, Chronic/complications
- Renal Insufficiency, Chronic/metabolism
- Naphthyridines/pharmacology
- Naphthyridines/therapeutic use
- Ventricular Dysfunction, Left/physiopathology
- Ventricular Dysfunction, Left/drug therapy
- Ventricular Dysfunction, Left/etiology
- Ventricular Dysfunction, Left/metabolism
- Male
- Disease Models, Animal
- Fibrosis
- Nitric Oxide Synthase Type III/metabolism
- Glomerular Filtration Rate/drug effects
- Ventricular Function, Left/drug effects
- Diastole/drug effects
- Kidney/drug effects
- Kidney/physiopathology
- Kidney/metabolism
- Phosphorylation
- Myocardium/metabolism
- Myocardium/pathology
- Rats, Sprague-Dawley
- Rats
- Nephrectomy
Collapse
Affiliation(s)
- Ixchel Lima Posada
- INSERM, UMRS 1138, Centre de Recherche des Cordeliers, Sorbonne Université, Université Paris CitéParisFrance
| | - Matthieu Soulié
- INSERM, UMRS 1138, Centre de Recherche des Cordeliers, Sorbonne Université, Université Paris CitéParisFrance
- Univ Rouen Normandie, INSERM EnVI UMR 1096RouenFrance
| | - Yohan Stephan
- Univ Rouen Normandie, INSERM EnVI UMR 1096RouenFrance
| | - Roberto Palacios Ramirez
- INSERM, UMRS 1138, Centre de Recherche des Cordeliers, Sorbonne Université, Université Paris CitéParisFrance
| | - Benjamin Bonnard
- INSERM, UMRS 1138, Centre de Recherche des Cordeliers, Sorbonne Université, Université Paris CitéParisFrance
| | - Lionel Nicol
- Univ Rouen Normandie, INSERM EnVI UMR 1096RouenFrance
| | - Bertram Pitt
- Department of MedicineUniversity of Michigan MedicineAnn ArborMI
| | - Peter Kolkhof
- Cardiovascular Precision Medicines, Research and Early Development, Pharmaceuticals, Bayer AGWuppertalGermany
| | - Paul Mulder
- Univ Rouen Normandie, INSERM EnVI UMR 1096RouenFrance
| | - Frederic Jaisser
- INSERM, UMRS 1138, Centre de Recherche des Cordeliers, Sorbonne Université, Université Paris CitéParisFrance
- Université de Lorraine, INSERM Centre d’Investigations Cliniques‐Plurithématique 1433, UMR 1116, CHRU de Nancy, French‐Clinical Research Infrastructure Network (F‐CRIN) INI‐CRCTNancyFrance
| |
Collapse
|
3
|
Theodorakopoulou M, Boutou A, Sarafidis P. Skeletal muscle oxygenation and exercise intolerance in hemodialysis: Navigating toward promising horizons? Respir Physiol Neurobiol 2024; 323:104238. [PMID: 38382591 DOI: 10.1016/j.resp.2024.104238] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2024] [Accepted: 02/18/2024] [Indexed: 02/23/2024]
Affiliation(s)
- Marieta Theodorakopoulou
- First Department of Nephrology, Hippokration Hospital, Aristotle University of Thessaloniki, Greece
| | - Afroditi Boutou
- Department of Respiratory Medicine, Hippokration Hospital, Thessaloniki, Greece.
| | - Pantelis Sarafidis
- First Department of Nephrology, Hippokration Hospital, Aristotle University of Thessaloniki, Greece
| |
Collapse
|
4
|
Jiang L, Hu X, Feng Y, Wang Z, Tang H, Lin Q, Shen Y, Zhu Y, Xu Q, Li X. Reduction of renal interstitial fibrosis by targeting Tie2 in vascular endothelial cells. Pediatr Res 2024; 95:959-965. [PMID: 38012310 PMCID: PMC10920200 DOI: 10.1038/s41390-023-02893-8] [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: 01/18/2023] [Revised: 10/08/2023] [Accepted: 10/30/2023] [Indexed: 11/29/2023]
Abstract
BACKGROUND Tie2, a functional angiopoietin receptor, is expressed in vascular endothelial cells and plays an important role in angiogenesis and vascular stability. This study aimed to evaluate the effects of an agonistic Tie2 signal on renal interstitial fibrosis (RIF) and elucidate the underlying mechanisms. METHODS We established an in vivo mouse model of folic acid-induced nephropathy (FAN) and an in vitro model of lipopolysaccharide-stimulated endothelial cell injury, then an agonistic Tie2 monoclonal antibody (Tie2 mAb) was used to intervent these processes. The degree of tubulointerstitial lesions and related molecular mechanisms were determined by histological assessment, immunohistochemistry, western blotting, and qPCR. RESULTS Tie2 mAb attenuated RIF and reduced the level of fibroblast-specific protein 1 (FSP1). Further, it suppressed vascular cell adhesion molecule-1 (VCAM-1) and increased CD31 density in FAN. In the in vitro model, Tie2 mAb was found to decrease the expression of VCAM-1, Bax, and α-smooth muscle actin (α-SMA). CONCLUSIONS The present findings indicate that the agonistic Tie2 mAb exerted vascular protective effects and ameliorated RIF via inhibition of vascular inflammation, apoptosis, and fibrosis. Therefore, Tie2 may be a potential target for the treatment of this disease. IMPACT This is the first report to confirm that an agonistic Tie2 monoclonal antibody can reduce renal interstitial fibrosis in folic acid-induced nephropathy in mice. This mechanism possibly involves vascular protective effects brought about by inhibition of vascular inflammation, apoptosis and fibrosis. Our data show that Tie2 signal may be a novel, endothelium-specific target for the treatment of tubulointerstitial fibrosis.
Collapse
Affiliation(s)
- Lu Jiang
- Department of Nephrology and Immunology, Children's Hospital of Soochow University, Suzhou, 215003, China
| | - Xiaohan Hu
- Institute of Pediatrics, Children's Hospital of Soochow University, Suzhou, 215003, China
| | - Yajun Feng
- Department of Pediatrics, Jiangyin People's Hospital, Jiangyin, 214400, China
| | - Zhen Wang
- Department of Pediatrics, Zibo Maternal and Child Health Care Hospital, Zibo, 255000, China
| | - Hanyun Tang
- Department of Nephrology and Immunology, Children's Hospital of Soochow University, Suzhou, 215003, China
| | - Qiang Lin
- Department of Nephrology and Immunology, Children's Hospital of Soochow University, Suzhou, 215003, China
| | - Yunyan Shen
- Department of Nephrology and Immunology, Children's Hospital of Soochow University, Suzhou, 215003, China
| | - Yun Zhu
- Department of Nephrology and Immunology, Children's Hospital of Soochow University, Suzhou, 215003, China
| | - Qinying Xu
- Department of Nephrology and Immunology, Children's Hospital of Soochow University, Suzhou, 215003, China
| | - Xiaozhong Li
- Department of Nephrology and Immunology, Children's Hospital of Soochow University, Suzhou, 215003, China.
| |
Collapse
|
5
|
Yao J, Sprick JD, Jeong J, Park J, Reiter DA. Differences in peripheral microcirculatory blood flow regulation in chronic kidney disease based on wavelet analysis of resting near-infrared spectroscopy. Microvasc Res 2024; 151:104624. [PMID: 37926135 PMCID: PMC11018197 DOI: 10.1016/j.mvr.2023.104624] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2023] [Revised: 10/13/2023] [Accepted: 11/01/2023] [Indexed: 11/07/2023]
Abstract
Vascular impairment is closely related to increased mortality in chronic kidney disease (CKD). The objective of this study was to assess impairments in the regulation of peripheral microvascular perfusion in patients with CKD based on time-frequency spectral analysis of resting near-infrared spectroscopy (NIRS) signals. Total hemoglobin (tHb) concentration and tissue saturation index (TSI) signals were collected using NIRS for a continuous 5 mins at 10 Hz from the forearm of 55 participants (34 CKD including 5 with end-stage renal disease, and 21 age-matched control). Continuous wavelet transform-based spectral analysis was used to quantify the spectral amplitude within five pre-defined frequency intervals (I, 0.0095-0.021 Hz; II, 0.021-0.052 Hz; III, 0.052-0.145 Hz; IV, 0.145-0.6 Hz and V, 0.6-2.0 Hz), representing endothelial, neurogenic, myogenic, respiratory and heartbeat activity, respectively. CKD patients showed lower tHb average spectral amplitude within the neurogenic frequency interval compared with controls (p = 0.014), consistent with an increased sympathetic outflow observed in CKD. CKD patients also showed lower TSI average spectral amplitude within the endothelial frequency interval compared with controls (p = 0.046), consistent with a reduced endothelial function in CKD. These findings demonstrate the potential of wavelet analysis of NIRS to provide complementary information on peripheral microvascular regulation in CKD.
Collapse
Affiliation(s)
- Jingting Yao
- Department of Radiology and Imaging Science, Emory University, Atlanta, GA, United States; Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, United States
| | - Justin D Sprick
- Department of Kinesiology, Health Promotion, and Recreation, University of North Texas, Denton, TX, United States
| | - Jinhee Jeong
- Division of Renal Medicine, Emory University, Atlanta, GA, United States; Atlanta Veterans Affairs Medical Center, Decatur, GA, United States
| | - Jeanie Park
- Division of Renal Medicine, Emory University, Atlanta, GA, United States; Atlanta Veterans Affairs Medical Center, Decatur, GA, United States
| | - David A Reiter
- Department of Radiology and Imaging Science, Emory University, Atlanta, GA, United States; Department of Orthopedics, Emory University, Atlanta, GA, United States; Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, United States.
| |
Collapse
|
6
|
Kaesler N, Cheng M, Nagai J, O’Sullivan J, Peisker F, Bindels EM, Babler A, Moellmann J, Droste P, Franciosa G, Dugourd A, Saez-Rodriguez J, Neuss S, Lehrke M, Boor P, Goettsch C, Olsen JV, Speer T, Lu TS, Lim K, Floege J, Denby L, Costa I, Kramann R. Mapping cardiac remodeling in chronic kidney disease. SCIENCE ADVANCES 2023; 9:eadj4846. [PMID: 38000021 PMCID: PMC10672229 DOI: 10.1126/sciadv.adj4846] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Accepted: 10/24/2023] [Indexed: 11/26/2023]
Abstract
Patients with advanced chronic kidney disease (CKD) mostly die from sudden cardiac death and recurrent heart failure. The mechanisms of cardiac remodeling are largely unclear. To dissect molecular and cellular mechanisms of cardiac remodeling in CKD in an unbiased fashion, we performed left ventricular single-nuclear RNA sequencing in two mouse models of CKD. Our data showed a hypertrophic response trajectory of cardiomyocytes with stress signaling and metabolic changes driven by soluble uremia-related factors. We mapped fibroblast to myofibroblast differentiation in this process and identified notable changes in the cardiac vasculature, suggesting inflammation and dysfunction. An integrated analysis of cardiac cellular responses to uremic toxins pointed toward endothelin-1 and methylglyoxal being involved in capillary dysfunction and TNFα driving cardiomyocyte hypertrophy in CKD, which was validated in vitro and in vivo. TNFα inhibition in vivo ameliorated the cardiac phenotype in CKD. Thus, interventional approaches directed against uremic toxins, such as TNFα, hold promise to ameliorate cardiac remodeling in CKD.
Collapse
Affiliation(s)
- Nadine Kaesler
- Clinic for Renal and Hypertensive Disorders, Rheumatological and Immunological Disease, University Hospital of the RWTH Aachen, Aachen, Germany
- Institute of Experimental Medicine and Systems Biology, University Hospital of the RWTH Aachen, Aachen, Germany
| | - Mingbo Cheng
- Institute for Computational Genomics, University Hospital of the RWTH Aachen, Aachen, Germany
| | - James Nagai
- Institute for Computational Genomics, University Hospital of the RWTH Aachen, Aachen, Germany
| | - James O’Sullivan
- Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, UK
| | - Fabian Peisker
- Institute of Experimental Medicine and Systems Biology, University Hospital of the RWTH Aachen, Aachen, Germany
| | - Eric M. J. Bindels
- Department of Hematology, Erasmus Medical Center, Rotterdam, Netherlands
| | - Anne Babler
- Institute of Experimental Medicine and Systems Biology, University Hospital of the RWTH Aachen, Aachen, Germany
| | - Julia Moellmann
- Department of Internal Medicine I, University Hospital of the RWTH Aachen, Aachen, Germany
| | - Patrick Droste
- Clinic for Renal and Hypertensive Disorders, Rheumatological and Immunological Disease, University Hospital of the RWTH Aachen, Aachen, Germany
- Institute of Pathology, University Hospital of the RWTH Aachen, Aachen, Germany
| | - Giulia Franciosa
- Novo Nordisk Foundation Center for Protein Research, University of Copenhagen, Copenhagen, Denmark
| | - Aurelien Dugourd
- Heidelberg University, Faculty of Medicine, and Heidelberg University Hospital, Institute for Computational Biomedicine, Bioquant, Heidelberg, Germany
| | - Julio Saez-Rodriguez
- Heidelberg University, Faculty of Medicine, and Heidelberg University Hospital, Institute for Computational Biomedicine, Bioquant, Heidelberg, Germany
| | - Sabine Neuss
- Institute of Pathology, University Hospital of the RWTH Aachen, Aachen, Germany
- Helmholtz Institute for Biomedical Engineering, Biointerface Laboratory, RWTH Aachen University, Aachen, Germany
| | - Michael Lehrke
- Department of Internal Medicine I, University Hospital of the RWTH Aachen, Aachen, Germany
| | - Peter Boor
- Clinic for Renal and Hypertensive Disorders, Rheumatological and Immunological Disease, University Hospital of the RWTH Aachen, Aachen, Germany
- Institute of Pathology, University Hospital of the RWTH Aachen, Aachen, Germany
| | - Claudia Goettsch
- Department of Internal Medicine I, University Hospital of the RWTH Aachen, Aachen, Germany
| | - Jesper V. Olsen
- Novo Nordisk Foundation Center for Protein Research, University of Copenhagen, Copenhagen, Denmark
| | - Thimoteus Speer
- Department of Medicine (Nephrology), Goethe University Frankfurt, Frankfurt, Germany
| | - Tzong-Shi Lu
- Brigham and Women’s Hospital, Renal Division, Boston, MA, USA
| | - Kenneth Lim
- Division of Nephrology and Hypertension, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Jürgen Floege
- Clinic for Renal and Hypertensive Disorders, Rheumatological and Immunological Disease, University Hospital of the RWTH Aachen, Aachen, Germany
| | - Laura Denby
- Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, UK
| | - Ivan Costa
- Institute for Computational Genomics, University Hospital of the RWTH Aachen, Aachen, Germany
| | - Rafael Kramann
- Clinic for Renal and Hypertensive Disorders, Rheumatological and Immunological Disease, University Hospital of the RWTH Aachen, Aachen, Germany
- Institute of Experimental Medicine and Systems Biology, University Hospital of the RWTH Aachen, Aachen, Germany
- Department of Internal Medicine, Nephrology and Transplantation, Erasmus Medical Center, Rotterdam, Netherlands
| |
Collapse
|
7
|
Baaten CCFMJ, Vondenhoff S, Noels H. Endothelial Cell Dysfunction and Increased Cardiovascular Risk in Patients With Chronic Kidney Disease. Circ Res 2023; 132:970-992. [PMID: 37053275 PMCID: PMC10097498 DOI: 10.1161/circresaha.123.321752] [Citation(s) in RCA: 35] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 04/15/2023]
Abstract
The endothelium is considered to be the gatekeeper of the vessel wall, maintaining and regulating vascular integrity. In patients with chronic kidney disease, protective endothelial cell functions are impaired due to the proinflammatory, prothrombotic and uremic environment caused by the decline in kidney function, adding to the increase in cardiovascular complications in this vulnerable patient population. In this review, we discuss endothelial cell functioning in healthy conditions and the contribution of endothelial cell dysfunction to cardiovascular disease. Further, we summarize the phenotypic changes of the endothelium in chronic kidney disease patients and the relation of endothelial cell dysfunction to cardiovascular risk in chronic kidney disease. We also review the mechanisms that underlie endothelial changes in chronic kidney disease and consider potential pharmacological interventions that can ameliorate endothelial health.
Collapse
Affiliation(s)
- Constance C F M J Baaten
- Institute for Molecular Cardiovascular Research (IMCAR), University Hospital RWTH Aachen, Aachen, Germany (C.C.F.M.J.B., S.V., H.N.)
- Department of Biochemistry, Cardiovascular Research Institute Maastricht, Maastricht University, Maastricht, the Netherlands (C.C.F.M.J.B., H.N.)
| | - Sonja Vondenhoff
- Institute for Molecular Cardiovascular Research (IMCAR), University Hospital RWTH Aachen, Aachen, Germany (C.C.F.M.J.B., S.V., H.N.)
| | - Heidi Noels
- Institute for Molecular Cardiovascular Research (IMCAR), University Hospital RWTH Aachen, Aachen, Germany (C.C.F.M.J.B., S.V., H.N.)
- Department of Biochemistry, Cardiovascular Research Institute Maastricht, Maastricht University, Maastricht, the Netherlands (C.C.F.M.J.B., H.N.)
| |
Collapse
|
8
|
Abramowitz MK. Strengthening the Link Between Cardiovascular Disease and Sarcopenia in CKD. Am J Kidney Dis 2023; 81:632-634. [PMID: 36871840 DOI: 10.1053/j.ajkd.2023.01.440] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Accepted: 01/18/2023] [Indexed: 03/06/2023]
Affiliation(s)
- Matthew K Abramowitz
- Department of Medicine, Albert Einstein College of Medicine, Bronx, NY, USA; Institute for Aging Research, Albert Einstein College of Medicine, Bronx, NY, USA; Diabetes Research Center, Albert Einstein College of Medicine, Bronx, NY, USA; Fleischer Institute for Diabetes and Metabolism, Albert Einstein College of Medicine, Bronx, NY, USA.
| |
Collapse
|
9
|
Meariman JK, Zulli H, Perez A, Bajracharya S, Mohandas R. Small vessel disease: Connections between the kidney and the heart. AMERICAN HEART JOURNAL PLUS : CARDIOLOGY RESEARCH AND PRACTICE 2023; 26:100257. [PMID: 38510186 PMCID: PMC10946057 DOI: 10.1016/j.ahjo.2023.100257] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Revised: 01/10/2023] [Accepted: 01/13/2023] [Indexed: 03/22/2024]
Abstract
Small vessel disease is characterized by global dysfunction of the microvascular system leading to reduced perfusion of various organ systems. The kidney is significantly vulnerable for microvascular dysfunction given its intricate capillary network and extensive endocrine influence. Studies have demonstrated a relationship between impaired renal function and small vessel disease in other organ systems, particularly the heart. Here we discuss the relationship between the kidney and the heart in the setting of microvascular dysfunction and identify areas of future study to better understand this relationship and potentially identify novel therapeutic strategies.
Collapse
Affiliation(s)
- Jacob K. Meariman
- Section of Nephrology & Hypertension, Department of Medicine, LSU Health New Orleans School of Medicine, New Orleans, LA 70112, United States of America
| | - Hannah Zulli
- Section of Nephrology & Hypertension, Department of Medicine, LSU Health New Orleans School of Medicine, New Orleans, LA 70112, United States of America
| | - Annalisa Perez
- Section of Nephrology & Hypertension, Department of Medicine, LSU Health New Orleans School of Medicine, New Orleans, LA 70112, United States of America
| | - S.D. Bajracharya
- Section of Nephrology & Hypertension, Department of Medicine, LSU Health New Orleans School of Medicine, New Orleans, LA 70112, United States of America
| | - Rajesh Mohandas
- Section of Nephrology & Hypertension, Department of Medicine, LSU Health New Orleans School of Medicine, New Orleans, LA 70112, United States of America
| |
Collapse
|
10
|
Theodorakopoulou MP, Zafeiridis A, Dipla K, Faitatzidou D, Koutlas A, Alexandrou ME, Doumas M, Papagianni A, Sarafidis P. Muscle Oxygenation and Microvascular Reactivity Across Different Stages of CKD: A Near-Infrared Spectroscopy Study. Am J Kidney Dis 2023; 81:655-664.e1. [PMID: 36608922 DOI: 10.1053/j.ajkd.2022.11.013] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Accepted: 11/19/2022] [Indexed: 01/05/2023]
Abstract
RATIONALE & OBJECTIVE Previous studies in chronic kidney disease (CKD) showed that vascular dysfunction in different circulatory beds progressively deteriorates with worsening CKD severity. This study evaluated muscle oxygenation and microvascular reactivity at rest, during an occlusion-reperfusion maneuver, and during exercise in patients with different stages of CKD versus controls. STUDY DESIGN Observational controlled study. SETTING & PARTICIPANTS 90 participants (18 per CKD stage 2, 3a, 3b, and 4, as well as 18 controls). PREDICTOR CKD stage. OUTCOME The primary outcome was muscle oxygenation at rest. Secondary outcomes were muscle oxygenation during occlusion-reperfusion and exercise, and muscle microvascular reactivity (hyperemic response). ANALYTICAL APPROACH Continuous measurement of muscle oxygenation [tissue saturation index (TSI)] using near-infrared spectroscopy at rest, during occlusion-reperfusion, and during a 3-minute handgrip exercise (at 35% of maximal voluntary contraction). Aortic pulse wave velocity and carotid intima-media thickness were also recorded. RESULTS Resting muscle oxygenation did not differ across the study groups (controls: 64.3% ± 2.9%; CKD stage 2: 63.8% ± 4.2%; CKD stage 3a: 64.1% ± 4.1%; CKD stage 3b: 62.3% ± 3.3%; CKD stage 4: 62.7% ± 4.3%; P=0.6). During occlusion, no significant differences among groups were detected in the TSI occlusion magnitude and TSI occlusion slope. However, during reperfusion the maximum TSI value was significantly lower in groups of patients with more advanced CKD stages compared with controls, as was the hyperemic response (controls: 11.2%±3.7%; CKD stage 2: 8.3%±4.6%; CKD stage 3: 7.8%±5.5%; CKD stage 3b: 7.3%±4.4%; CKD stage 4: 7.2%±3.3%; P=0.04). During the handgrip exercise, the average decline in TSI was marginally lower in patients with CKD than controls, but no significant differences were detected across CKD stages. LIMITATIONS Moderate sample size, cross-sectional evaluation. CONCLUSIONS Although no differences were observed in muscle oxygenation at rest or during occlusion, the microvascular hyperemic response during reperfusion was significantly impaired in CKD and was most prominent in more advanced CKD stages. This impaired ability of microvasculature to respond to stimuli may be a crucial component of the adverse vascular profile of patients with CKD and may contribute to exercise intolerance.
Collapse
Affiliation(s)
- Marieta P Theodorakopoulou
- Department of Nephrology, Hippokration Hospital, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Andreas Zafeiridis
- Exercise Physiology & Biochemistry Laboratory, Department of Sport Sciences at Serres, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Konstantina Dipla
- Exercise Physiology & Biochemistry Laboratory, Department of Sport Sciences at Serres, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Danai Faitatzidou
- Department of Nephrology, Hippokration Hospital, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Angelos Koutlas
- Exercise Physiology & Biochemistry Laboratory, Department of Sport Sciences at Serres, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Maria-Eleni Alexandrou
- Department of Nephrology, Hippokration Hospital, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Michael Doumas
- Second Propedeutic Department of Internal Medicine, Hippokration Hospital, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Aikaterini Papagianni
- Department of Nephrology, Hippokration Hospital, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Pantelis Sarafidis
- Department of Nephrology, Hippokration Hospital, Aristotle University of Thessaloniki, Thessaloniki, Greece.
| |
Collapse
|
11
|
Molina P, Ojeda R, Blanco A, Alcalde G, Prieto-Velasco M, Aresté N, Buades JM, Simó VE, Goicoechea M, Pérez-Morales RE, Sánchez-Álvarez E, Sánchez-Villanueva R, Montesa M, Arenas MD. Etiopathogenesis of chronic kidney disease-associated pruritus: putting the pieces of the puzzle together. Nefrologia 2023; 43:48-62. [PMID: 37173258 DOI: 10.1016/j.nefroe.2023.03.015] [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: 03/29/2022] [Accepted: 06/28/2022] [Indexed: 05/15/2023] Open
Abstract
Defined as the unpleasant sensation that causes the desire to scratch, pruritus is the most common skin symptom associated with uremia and appears in almost half of patients with advanced chronic kidney disease (CKD). Beyond its direct impact on quality of life, CKD-associated pruritus (CKD-aP) is an independent predictor of mortality that also has a synergistic effect with other quality of life-related symptoms, such as insomnia, depression, and anxiety. Although different mechanisms have been proposed to explain the origin of Pa-ERC, its etiopathogenesis is still not fully understood. Since new therapeutic targets have been identified and several clinical trials have recently shown promising results, our current understanding of the interrelationships has expanded significantly and the pathophysiological mechanisms underlying CKD-aP are now considered to be multifactorial. The potential triggers of pruritus in patients with CKD are discussed in this review, including hypotheses about skin xerosis, accumulation of uremic toxins, dysregulation of the immune system and systemic inflammation, uremic neuropathy, and imbalances in the endogenous opioid system. Other non-uremic causes of pruritus are also discussed, with the aim of guiding the physicians to apply an adequate aetiopathogenic approach to CKD-aP in their day-to-day clinical practice.
Collapse
Affiliation(s)
- Pablo Molina
- Servicio de Nefrología, FISABIO, Hospital Universitari Dr Peset, Departamento de Medicina, Universitat de València, Valencia, Spain.
| | - Raquel Ojeda
- Servicio de Nefrología, Hospital Universitario Renia Sofía, Córdoba, Spain
| | - Ana Blanco
- Fresenius Medical Care, Centro de Diálisis Alcobendas, Madrid, Spain; Servicio de Nefrología, Hospital Quirón Ruber Juan Bravo, Madrid, Spain
| | - Guillermo Alcalde
- Servicio de Nefrología, BIOARABA, Hospital Universitario Araba, Osakidetza, Universidad del País Vasco, Vitoria-Gasteiz, Álava, Spain
| | | | - Nuria Aresté
- Servicio de Nefrología, Hospital Universitario Virgen Macarena, Sevilla, Spain
| | - Juan Manuel Buades
- Servicio de Nefrología, Hospital Universitario Son Llàtzer, Institut d'Investigació Sanitària de les Illes Balears (IdISBa), Palma, Spain
| | - Vicent Esteve Simó
- Servicio de Nefrologia, Hospital Terrassa, Consorci Sanitari Terrassa (CST), Terrassa, Barcelona, Spain
| | - Marian Goicoechea
- Servicio de Nefrología, Hospital General Universitario Gregorio Marañón, Madrid, Spain
| | - Rosa Elena Pérez-Morales
- Servicio de Nefrología. Hospital Universitario Nuestra Señora de Candelaria, Santa Cruz de Tenerife, Spain
| | - Emilio Sánchez-Álvarez
- Servicio de Nefrología, Hospital Universitario de Cabueñes, Red de Investigación Renal (REDINREN), Gijón, Asturias, Spain
| | | | - María Montesa
- Servicio de Nefrología, FISABIO, Hospital Universitari Dr Peset, Departamento de Medicina, Universitat de València, Valencia, Spain
| | | |
Collapse
|
12
|
Nguyen IT, Joles JA, Verhaar MC, Lamb HJ, Dekkers IA. Obesity in relation to cardiorenal function. VISCERAL AND ECTOPIC FAT 2023:243-264. [DOI: 10.1016/b978-0-12-822186-0.00006-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
13
|
Pruritus in Chronic Kidney Disease: An Update. ALLERGIES 2022. [DOI: 10.3390/allergies2030009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Chronic kidney disease-associated pruritus (CKDaP) is an often under-diagnosed and under-recognized condition, despite its considerable prevalence within the chronic kidney disease (CKD) population. Universally accepted guidelines are also lacking. The true prevalence of CKDaP worldwide therefore remains unknown, although its negative impact on mortality and health-related quality of life outcomes is very clear. The pathophysiological mechanisms leading to the onset of CKDaP are only partly understood. CKDaP is currently believed to be caused by a multifactorial process, from local skin changes, metabolic alterations, the development of neuropathy and dysregulation of opioid pathways, and psychological factors. Much work has been carried out towards a more systematic and structured approach to clinical diagnosis. Various tools are now available to assess the severity of CKDaP. Many of these tools require greater validation before they can be incorporated into the guidelines and into routine clinical practice. Further efforts are also needed in order to increase the awareness of clinicians and patients so that they can identify the CKDaP signs and symptoms in a timely manner. Currently established treatment options for CKDaP focus on the prevention of xerosis via topical emollients, the optimization of dialysis management, early referral to kidney transplantation if appropriate, oral antihistamine, and a variety of neuropathic agents. Other novel treatment options include the following: topical analgesics, topical tacrolimus, cannabinoid-containing compounds, antidepressants, oral leukotrienes, opioids, and non-pharmacological alternative therapies (i.e., phototherapy, dietary supplements, acupuncture/acupressure). We provide an updated review on the evidence relating to the epidemiology, the pathophysiology, the clinical assessment and diagnosis, and the management of CKDaP.
Collapse
|
14
|
Etiopatogenia del prurito asociado a la enfermedad renal crónica: recomponiendo las piezas del puzle. Nefrologia 2022. [DOI: 10.1016/j.nefro.2022.06.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
|
15
|
Ho JQ, Abramowitz MK. Clinical Consequences of Metabolic Acidosis-Muscle. Adv Chronic Kidney Dis 2022; 29:395-405. [PMID: 36175077 DOI: 10.1053/j.ackd.2022.04.010] [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: 02/20/2022] [Revised: 04/10/2022] [Accepted: 04/25/2022] [Indexed: 01/25/2023]
Abstract
Metabolic acidosis is common in people with chronic kidney disease and can contribute to functional decline, morbidity, and mortality. One avenue through which metabolic acidosis can result in these adverse clinical outcomes is by negatively impacting skeletal muscle; this can occur through several pathways. First, metabolic acidosis promotes protein degradation and impairs protein synthesis, which lead to muscle breakdown. Second, metabolic acidosis hinders mitochondrial function, which decreases oxidative phosphorylation and reduces energy production. Third, metabolic acidosis directly limits muscle contraction. The purpose of this review is to examine the specific mechanisms of each pathway through which metabolic acidosis affects muscle, the impact of metabolic acidosis on physical function, and the effect of treating metabolic acidosis on functional outcomes.
Collapse
Affiliation(s)
- Jim Q Ho
- Department of Medicine, Albert Einstein College of Medicine, Bronx, NY
| | - Matthew K Abramowitz
- Department of Medicine, Albert Einstein College of Medicine, Bronx, NY; Institute for Aging Research, Albert Einstein College of Medicine, Bronx, NY; Diabetes Research Center, Albert Einstein College of Medicine, Bronx, NY; Fleischer Institute for Diabetes and Metabolism, Albert Einstein College of Medicine, Bronx, NY.
| |
Collapse
|
16
|
Gökgöz G, Tortumlu G, Akça Bayar S, Yilmaz G, Haberal M. Peripapillary Vascular Density Measurement in Pediatric Renal and Liver Transplant. EXP CLIN TRANSPLANT 2022; 20:96-101. [PMID: 35570610 DOI: 10.6002/ect.pediatricsymp2022.o32] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
OBJECTIVES Noninvasive monitorization of retinal structures of the eye could be a predictor for systemic microvasculature dysfunction in transplant recipients. In this study, our purpose was to determine the optic disc and peripapillary microvascular changes in pediatric patients who had undergone liver or renal transplant surgery. MATERIALS AND METHODS The study was performed at Başkent University. The medical records were reviewed, and patients who had liver or renal transplant in the last 10 years and were between 4 and 18 years old were included in the study. The optic disc and peripapillary vascular density parameters were obtained by optical coherence tomography angiography (Avanti RTVue XR). The results were compared with the results from age-matched, sex-matched, and spherical equivalent-matched healthy subjects. RESULTS Our study included 32 eyes of 16 liver transplant patients, 20 eyes of 10 renal transplant patients, and 64 eyes of 32 healthy participants (control). Whole image peripapillary, inside disc, peripapillary, superior and inferior hemisphere, and superior, inferior, temporal, and nasal quadrant peripapillary vascular densities were evaluated. No statistically significant differences in any parameter were noted between the healthy control group and the patient groups (P > .05 for all parameters). CONCLUSIONS Peripapillary vascular density measurements were not affected in pediatric renal and liver transplant patients.
Collapse
Affiliation(s)
- Gülşah Gökgöz
- From the Department of Ophthalmology, Baskent University Faculty of Medicine, Ankara, Turkey
| | | | | | | | | |
Collapse
|
17
|
El Chamieh C, Liabeuf S, Massy Z. Uremic Toxins and Cardiovascular Risk in Chronic Kidney Disease: What Have We Learned Recently beyond the Past Findings? Toxins (Basel) 2022; 14:280. [PMID: 35448889 PMCID: PMC9028122 DOI: 10.3390/toxins14040280] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2022] [Revised: 04/10/2022] [Accepted: 04/11/2022] [Indexed: 12/13/2022] Open
Abstract
Patients with chronic kidney disease (CKD) have an elevated prevalence of atheromatous (ATH) and/or non-atheromatous (non-ATH) cardiovascular disease (CVD) due to an array of CKD-related risk factors, such as uremic toxins (UTs). Indeed, UTs have a major role in the emergence of a spectrum of CVDs, which constitute the leading cause of death in patients with end-stage renal disease. The European Uremic Toxin Work Group has identified over 100 UTs, more than 25 of which are dietary or gut-derived. Even though relationships between UTs and CVDs have been described in the literature, there are few reviews on the involvement of the most toxic compounds and the corresponding physiopathologic mechanisms. Here, we review the scientific literature on the dietary and gut-derived UTs with the greatest toxicity in vitro and in vivo. A better understanding of these toxins' roles in the elevated prevalence of CVDs among CKD patients might facilitate the development of targeted treatments. Hence, we review (i) ATH and non-ATH CVDs and the respective levels of risk in patients with CKD and (ii) the mechanisms that underlie the influence of dietary and gut-derived UTs on CVDs.
Collapse
Affiliation(s)
- Carolla El Chamieh
- Center for Research in Epidemiology and Population Health (CESP), Paris-Saclay University, Versailles-Saint-Quentin-en-Yvelines University (UVSQ), INSERM UMRS 1018, F-94807 Villejuif, France;
| | - Sophie Liabeuf
- Pharmacology Department, Amiens University Hospital, F-80000 Amiens, France
- MP3CV Laboratory, EA7517, Jules Verne University of Picardie, F-80000 Amiens, France
| | - Ziad Massy
- Nephrology Department, Ambroise Paré University Hospital, APHP, F-92100 Paris, France
| |
Collapse
|
18
|
István L, Czakó C, Benyó F, Élő Á, Mihály Z, Sótonyi P, Varga A, Nagy ZZ, Kovács I. The effect of systemic factors on retinal blood flow in patients with carotid stenosis: an optical coherence tomography angiography study. GeroScience 2022; 44:389-401. [PMID: 34837589 PMCID: PMC8810958 DOI: 10.1007/s11357-021-00492-1] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Accepted: 11/16/2021] [Indexed: 12/17/2022] Open
Abstract
Carotid artery stenosis (CAS) is among the leading causes of mortality and permanent disabilities in the Western world. CAS is a consequence of systemic atherosclerotic disease affecting the majority of the aging population. Optical coherence tomography angiography (OCTA) is a novel imaging technique for visualizing retinal blood flow. It is a noninvasive, fast method for qualitative and quantitative assessment of the microcirculation. Cerebral and retinal circulation share similar anatomy, physiology, and embryology; thus, retinal microvasculature provides a unique opportunity to study the pathogenesis of cerebral small vessel disease in vivo. In this study, we aimed to analyze the effect of systemic risk factors on retinal blood flow in the eyes of patients with significant carotid artery stenosis using OCT angiography. A total of 112 eyes of 56 patients with significant carotid stenosis were included in the study. We found that several systemic factors, such as decreased estimated glomerular filtration rate (eGFR), hypertension, and carotid occlusion have a significant negative effect on retinal blood flow, while statin use and carotid surgery substantially improve ocular microcirculation. Neither diabetes, clopidogrel or acetylsalicylic acid use, BMI, serum lipid level, nor thrombocyte count showed a significant effect on ocular blood flow. Our results demonstrate that a systematic connection does exist between certain systemic risk factors and retinal blood flow in this patient population. OCTA could help in the assessment of cerebral circulation of patients with CAS due to its ability to detect subtle changes in retinal microcirculation that is considered to represent changes in intracranial blood flow.
Collapse
Affiliation(s)
- Lilla István
- Department of Ophthalmology, Semmelweis University, 39 Mária Street, 1085, Budapest, Hungary
| | - Cecilia Czakó
- Department of Ophthalmology, Semmelweis University, 39 Mária Street, 1085, Budapest, Hungary
| | - Fruzsina Benyó
- Department of Ophthalmology, Semmelweis University, 39 Mária Street, 1085, Budapest, Hungary
| | - Ágnes Élő
- Department of Ophthalmology, Semmelweis University, 39 Mária Street, 1085, Budapest, Hungary
| | - Zsuzsa Mihály
- Department of Vascular & Endovascular Surgery, Semmelweis University, Budapest, Hungary
| | - Péter Sótonyi
- Department of Vascular & Endovascular Surgery, Semmelweis University, Budapest, Hungary
| | - Andrea Varga
- Department of Vascular & Endovascular Surgery, Semmelweis University, Budapest, Hungary
| | - Zoltán Zsolt Nagy
- Department of Ophthalmology, Semmelweis University, 39 Mária Street, 1085, Budapest, Hungary
| | - Illés Kovács
- Department of Ophthalmology, Semmelweis University, 39 Mária Street, 1085, Budapest, Hungary.
- Department of Ophthalmology, Weill Cornell Medical College, New York, USA.
- Department of Clinical Ophthalmology, Faculty of Health Sciences, Semmelweis University, Budapest, Hungary.
| |
Collapse
|
19
|
Schricker S, Kimmel M. Unravelling the pathophysiology of chronic kidney disease-associated pruritus. Clin Kidney J 2022; 14:i23-i31. [PMID: 34987780 PMCID: PMC8702819 DOI: 10.1093/ckj/sfab200] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2021] [Indexed: 12/12/2022] Open
Abstract
For decades, itch related to chronic kidney disease (CKDaP) has been a clinical problem, but the aetiology and pathophysiology of CKDaP are still not yet fully understood—currently the underlying pathophysiological mechanisms are thought to be multifactorial. As new therapeutic targets have recently been identified and clinical trials have shown promising results, our current understanding of the interrelationships has expanded significantly. Here we review the pathophysiology and recent findings on modulation and sensitization of itch contributing to the development of CKDaP, covering hypothesis regarding immune system dysfunction, metabolic changes, uremic toxin deposition, peripheral neuropathy and imbalances in the endogenous opioid system.
Collapse
Affiliation(s)
- Severin Schricker
- Department of General Internal Medicine and Nephrology, Robert-Bosch-Hospital, Stuttgart, Germany
| | - Martin Kimmel
- Department of Internal Medicine, Division of Nephrology, Hypertension and Autoimmune Disorders, Alb-Fils Kliniken, Göppingen, Germany
| |
Collapse
|
20
|
Kan C, Lu X, Zhang R. Effects of hypoxia on bone metabolism and anemia in patients with chronic kidney disease. World J Clin Cases 2021; 9:10616-10625. [PMID: 35004993 PMCID: PMC8686129 DOI: 10.12998/wjcc.v9.i34.10616] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Revised: 08/12/2021] [Accepted: 10/25/2021] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Abnormal bone metabolism and renal anemia seriously affect the prognosis of patients with chronic kidney disease (CKD). Existing studies have mostly addressed the pathogenesis and treatment of bone metabolism abnormality and anemia in patients with CKD, but few have evaluated their mutual connection. Administration of exogenous erythropoietin to CKD patients with anemia used to be the mainstay of therapeutic approaches; however, with the availability of hypoxia-inducible factor (HIF) stabilizers such as roxadustat, more therapeutic choices for renal anemia are expected in the future. However, the effects posed by the hypoxic environment on both CKD complications remain incompletely understood. AIM To summarize the relationship between renal anemia and abnormal bone metabolism, and to discuss the influence of hypoxia on bone metabolism. METHODS CNKI and PubMed searches were performed using the key words "chronic kidney disease," "abnormal bone metabolism," "anemia," "hypoxia," and "HIF" to identify relevant articles published in multiple languages and fields. Reference lists from identified articles were reviewed to extract additional pertinent articles. Then we retrieved the Abstract and Introduction and searched the results from the literature, classified the extracted information, and summarized important information. Finally, we made our own conclusions. RESULTS There is a bidirectional relationship between renal anemia and abnormal bone metabolism. Abnormal vitamin D metabolism and hyperparathyroidism can affect bone metabolism, blood cell production, and survival rates through multiple pathways. Anemia will further attenuate the normal bone growth. The hypoxic environment regulates bone morphogenetic protein, vascular endothelial growth factor, and neuropilin-1, and affects osteoblast/osteoclast maturation and differentiation through bone metabolic changes. Hypoxia preconditioning of mesenchymal stem cells (MSCs) can enhance their paracrine effects and promote fracture healing. Concurrently, hypoxia reduces the inhibitory effect on osteocyte differentiation by inhibiting the expression of fibroblast growth factor 23. Hypoxia potentially improves bone metabolism, but it still carries potential risks. The optimal concentration and duration of hypoxia remain unclear. CONCLUSION There is a bidirectional relationship between renal anemia and abnormal bone metabolism. Hypoxia may improve bone metabolism but the concentration and duration of hypoxia remain unclear and need further study.
Collapse
Affiliation(s)
- Chao Kan
- Department of Clinical Medicine, Changchun University of Chinese Medicine, Changchun 130000, Jilin Province, China
| | - Xu Lu
- Department of Clinical Medicine, Changchun University of Chinese Medicine, Changchun 130000, Jilin Province, China
| | - Rui Zhang
- Department of Nephrology, Zhuhai People’s Hospital (Zhuhai Hospital Affiliated with Jinan University), Zhuhai 519070, Guangdong Province, China
| |
Collapse
|
21
|
Brightwell CR, Kulkarni AS, Paredes W, Zhang K, Perkins JB, Gatlin KJ, Custodio M, Farooq H, Zaidi B, Pai R, Buttar RS, Tang Y, Melamed ML, Hostetter TH, Pessin JE, Hawkins M, Fry CS, Abramowitz MK. Muscle fibrosis and maladaptation occur progressively in CKD and are rescued by dialysis. JCI Insight 2021; 6:150112. [PMID: 34784301 PMCID: PMC8783691 DOI: 10.1172/jci.insight.150112] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Accepted: 11/11/2021] [Indexed: 11/17/2022] Open
Abstract
BACKGROUND Skeletal muscle maladaptation accompanies chronic kidney disease (CKD) and negatively impacts physical function. Emphasis in CKD has historically been placed on muscle fiber intrinsic deficits, such as altered protein metabolism and atrophy. However, targeted treatment of fiber intrinsic dysfunction has produced limited improvement, whereas alterations within the fiber extrinsic environment have scarcely been examined. METHODS We investigated alterations to the skeletal muscle interstitial environment with deep cellular phenotyping of biopsies from patients with CKD compared to age-matched control participants and performed transcriptome profiling to define the molecular underpinnings of CKD-associated muscle impairments. We further examined changes in the observed muscle maladaptation following initiation of dialysis therapy for kidney failure. RESULTS Patients with CKD exhibited a progressive fibrotic muscle phenotype, which was associated with impaired regenerative capacity and lower vascular density. The severity of these deficits was strongly associated with the degree of kidney dysfunction. Consistent with these profound deficits, CKD was associated with broad alterations to the muscle transcriptome, including altered extracellular matrix organization, downregulated angiogenesis, and altered expression of pathways related to stem cell self-renewal. Remarkably, despite the seemingly advanced nature of this fibrotic transformation, dialysis treatment rescued these deficits, restoring a healthier muscle phenotype. Furthermore, after accounting for muscle atrophy, strength and endurance improved after dialysis initiation. CONCLUSION These data identify a dialysis-responsive muscle fibrotic phenotype in CKD and suggest that the early dialysis window presents a unique opportunity of improved muscle regenerative capacity during which targeted interventions may achieve maximal impact. TRIAL REGISTRATION NCT01452412FUNDING. NIH.
Collapse
Affiliation(s)
- Camille R Brightwell
- Department of Athletic Training and Clinical Nutrition, University of Kentucky, Lexington, United States of America
| | - Ameya S Kulkarni
- Department of Medicine, Albert Einstein College of Medicine, Bronx, United States of America
| | - William Paredes
- Department of Medicine, Albert Einstein College of Medicine, Bronx, United States of America
| | - Kehao Zhang
- Department of Medicine, Albert Einstein College of Medicine, Bronx, United States of America
| | - Jaclyn B Perkins
- Department of Nutrition and Metabolism, The University of Texas Medical Branch, Galveston, United States of America
| | - Knubian J Gatlin
- Department of Nutrition and Metabolism, The University of Texas Medical Branch, Galveston, United States of America
| | - Matthew Custodio
- Department of Medicine, Albert Einstein College of Medicine, Bronx, United States of America
| | - Hina Farooq
- Department of Medicine, Albert Einstein College of Medicine, Bronx, United States of America
| | - Bushra Zaidi
- Department of Medicine, Albert Einstein College of Medicine, Bronx, United States of America
| | - Rima Pai
- Department of Medicine, Albert Einstein College of Medicine, Bronx, United States of America
| | - Rupinder S Buttar
- Department of Medicine, Albert Einstein College of Medicine, Bronx, United States of America
| | - Yan Tang
- Department of Medicine, Albert Einstein College of Medicine, Bronx, United States of America
| | - Michal L Melamed
- Department of Medicine, Albert Einstein College of Medicine, Bronx, United States of America
| | - Thomas H Hostetter
- Department of Medicine, University of North Carolina School of Medicine, Chapel Hill, United States of America
| | - Jeffrey E Pessin
- Department of Medicine, Albert Einstein College of Medicine, Bronx, United States of America
| | - Meredith Hawkins
- Department of Medicine, Albert Einstein College of Medicine, Bronx, United States of America
| | | | - Matthew K Abramowitz
- Department of Medicine, Albert Einstein College of Medicine, Bronx, United States of America
| |
Collapse
|
22
|
Ku TC, Wang PH, Huang JL, Chen HY, Fang JT, Hsieh HL, Chen JL. The survival outcome of nasopharyngeal cancer patients with traditional Chinese medicine external use: A hospital-based study. JOURNAL OF ETHNOPHARMACOLOGY 2021; 279:114380. [PMID: 34197958 DOI: 10.1016/j.jep.2021.114380] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/20/2021] [Revised: 06/13/2021] [Accepted: 06/26/2021] [Indexed: 06/13/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE External-use traditional Chinese medicine (TCM) agents are widely used to relieve the adverse effects of radiation therapy in nasopharyngeal cancer patients. AIM OF THE STUDY Our study aimed to evaluate the influence of external-use TCM agents to relieve radiotherapy-related adverse effects on the efficacy of radiation therapy and the prognosis of nasopharyngeal cancer patients. MATERIALS AND METHODS By using the Chang Gung Research Database (CGRD), we analyzed 1823 newly diagnosed nasopharyngeal cancer patients with radiotherapy-related adverse effects between 2001/01 and 2015/12. We used Kaplan-Meier analysis and a Cox regression model to estimate the differences in effects on survival outcomes between two groups, TCM external users and non-TCM external users. RESULTS We found that TCM external users had significantly better 3-year and 5-year overall survival rates (log-rank test, p = 0.0377 and p = 0.034, respectively) than non-TCM external users. The 3-year and 5-year disease-free survival rates were not statistically significantly different between the groups. We also found a trend of improved 3-year and 5-year overall survival rates in TCM external users with advanced-stage disease, without statistical significance (log-rank test, p = 0.10 and p = 0.089, respectively). The subgroup analysis revealed lower risks of mortality in TCM external users among the nonhypertension, nonhyperlipidemia, nonischemic heart disease, noncirrhosis, and nonchronic kidney disease groups. CONCLUSIONS Our study showed that TCM agents external use could significantly improve 3-year and 5-year overall survival rates in nasopharyngeal cancer patients with radiotherapy-related adverse effects.
Collapse
Affiliation(s)
- Te-Chien Ku
- Division of Chinese Internal Medicine, Department of Traditional Chinese Medicine, Chang Gung Memorial Hospital, Taoyuan, Taiwan.
| | - Pin-Han Wang
- Division of Chinese Internal Medicine, Department of Traditional Chinese Medicine, Chang Gung Memorial Hospital, Taoyuan, Taiwan.
| | - Jhen-Ling Huang
- Center for Big Data Analytics and Statistics, Chang Gung Memorial Hospital, Taoyuan, Taiwan.
| | - Hsing-Yu Chen
- Division of Chinese Internal Medicine, Department of Traditional Chinese Medicine, Chang Gung Memorial Hospital, Taoyuan, Taiwan.
| | - Ji-Tseng Fang
- Kidney Research Center, Department of Nephrology, Chang Gung Memorial Hospital, Taoyuan, Taiwan; School of Medicine, College of Medicine, Chang Gung University, Taoyuan, Taiwan.
| | - Hsi-Lung Hsieh
- Department of Nursing, Division of Basic Medical Sciences, Research Center for Chinese Herbal Medicine, And Graduate Institute of Health Industry Technology, Chang Gung University of Science and Technology, Taoyuan, Taiwan; Department of Neurology, Chang Gung Memorial Hospital, Taoyuan, Taiwan.
| | - Jiun-Liang Chen
- Division of Chinese Internal Medicine, Department of Traditional Chinese Medicine, Chang Gung Memorial Hospital, Taoyuan, Taiwan; School of Traditional Chinese Medicine, College of Medicine, Chang Gung University, Taoyuan, Taiwan.
| |
Collapse
|
23
|
Gastaldi G, Pannier F, Roztočil K, Lugli M, Mansilha A, Haller H, Rabe E, VAN Rijn MJ. Chronic venous disease and diabetic microangiopathy: pathophysiology and commonalities. INT ANGIOL 2021; 40:457-469. [PMID: 34547884 DOI: 10.23736/s0392-9590.21.04664-2] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Chronic venous disease and diabetes mellitus are highly prevalent and debilitating conditions affecting millions of individuals globally. Although these conditions are typically considered as separate entities, they often co-exist which may be important in both understanding their pathophysiology and determining the best treatment strategy. Diabetes mellitus is twice as common in patients with chronic venous disease compared with the general population. Notably, a large proportion of patients with diabetes mellitus present with venous disorders, although this is often overlooked. The etiology of chronic venous disease is multifactorial, involving hemodynamic, genetic, and environmental factors which result in changes to the venous endothelium and structural wall as well as inflammation. Inflammation, endothelial dysfunction and hyperfiltration or leakage, are commonly observed in diabetes mellitus and cause various diabetic microvascular complications. Both diseases are also influenced by the increased expression of adhesion molecules, chemokines, and cytokines, and are characterized by the presence of vessel hypertension. Consequently, despite differences in etiology, the pathophysiology of both chronic venous disease and diabetic microangiopathy appears to be driven by endothelial dysfunction and inflammation. Treatment strategies should take the co-existence of chronic venous disease and diabetic microangiopathy into account. Compression therapy is recommended in inflammatory conditions that have an edema component as seen in both chronic venous disease and diabetes mellitus. Lifestyle changes like weight loss and exercise, will improve metabolic state and lower inflammation and should be promoted in these patients. Additionally, both patient populations may benefit from venoactive drugs.
Collapse
Affiliation(s)
- Giacomo Gastaldi
- Division of Endocrinology Diabetology Nutrition and Patient Education, Geneva University Hospitals, Geneva, Switzerland
| | - Felizitas Pannier
- Private Clinic Phlebology and Dermatology, Bonn, Germany.,Department of Dermatology, University of Cologne, Cologne, Germany
| | - Karel Roztočil
- Department of Transplantational and Vascular Surgery, Institute for Clinical and Experimental Medicine, Prague, Czech Republic
| | - Marzia Lugli
- Unit of Vascular Surgery, Cardiovascular Department, Hesperia Hospital, Modena, Italy
| | - Armando Mansilha
- Faculty of Medicine, University of Porto, Porto, Portugal.,Department of Angiology and Vascular Surgery, Hospital de S. João, Porto, Portugal
| | - Hermann Haller
- Hannover Medical School, Department of Nephrology and Hypertension, Hannover, Germany
| | - Eberhard Rabe
- Department of Dermatology (Emeritus), University of Bonn, Bonn, Germany
| | - Marie Josee VAN Rijn
- Department of Vascular Surgery, Erasmus Medical Center, Rotterdam, the Netherlands -
| |
Collapse
|
24
|
Díez J, Navarro-González JF, Ortiz A, Santamaría R, de Sequera P. Developing the subspecialty of cardio-nephrology: The time has come. A position paper from the coordinating committee from the Working Group for Cardiorenal Medicine of the Spanish Society of Nephrology. Nefrologia 2021; 41:391-402. [PMID: 36165108 DOI: 10.1016/j.nefroe.2021.02.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Accepted: 02/21/2021] [Indexed: 06/16/2023] Open
Abstract
Patients with the dual burden of chronic kidney disease (CKD) and cardiovascular disease (CVD) experience unacceptably high rates of morbidity and mortality, which also entail unfavorable effects on healthcare systems. Currently, concerted efforts to identify, prevent and treat CVD in CKD patients are lacking at the institutional level, with emphasis still being placed on individual specialty views on this topic. The authors of this position paper endorse the need for a dedicated interdisciplinary team of subspecialists in cardio-nephrology that manages appropriate clinical interventions across the inpatient and outpatient settings. There is a critical need for training programs, guidelines and best clinical practice models, and research funding from nephrology, cardiology and other professional societies, to support the development of the subspecialty of cardio-nephrology. This position paper from the coordinating committee from the Working Group for Cardiorenal Medicine of the Spanish Society of Nephrology (S.E.N.) is intended to be the starting point to develop the subspecialty of cardio-nephrology within the S.E.N.. The implementation of the subspecialty in day-to-day nephrological practice will help to diagnose, treat, and prevent CVD in CKD patients in a precise, clinically effective, and health cost-favorable manner.
Collapse
Affiliation(s)
- Javier Díez
- Departments of Nephrology and Cardiology, University of Navarra Clinic, Pamplona, Spain; Program of Cardiovascular Diseases, Center of Applied Medical Research, University of Navarra, Pamplona, Spain.
| | - Juan F Navarro-González
- Division of Nephrology and Research Unit, University Hospital Nuestra Señora de Candelaria, and Universitary Institute of Biomedical Technologies, University of La Laguna, Santa Cruz de Tenerife, Spain; Red de Investigación Renal (REDINREN), Madrid, Spain
| | - Alberto Ortiz
- Red de Investigación Renal (REDINREN), Madrid, Spain; Division of Nephrology IIS-Fundacion Jimenez Diaz, University Autonoma of Madrid, Madrid, Spain
| | - Rafael Santamaría
- Red de Investigación Renal (REDINREN), Madrid, Spain; Division of Nephrology, University Hospital Reina Sofia, Cordoba, Spain; Maimonides Biomedical Research Institute of Cordoba (IMIBIC), Spain
| | - Patricia de Sequera
- Nephrology Department, Hospital Universitario Infanta Leonor, University Complutense of Madrid, Madrid, Spain
| |
Collapse
|
25
|
Saha M, Sun QJ, Hildreth CM, Burke PGR, Phillips JK. Augmented Respiratory-Sympathetic Coupling and Hemodynamic Response to Acute Mild Hypoxia in Female Rodents With Chronic Kidney Disease. Front Physiol 2021; 12:623599. [PMID: 34113258 PMCID: PMC8185289 DOI: 10.3389/fphys.2021.623599] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Accepted: 04/15/2021] [Indexed: 11/15/2022] Open
Abstract
Carotid body feedback and hypoxia may serve to enhance respiratory–sympathetic nerve coupling (respSNA) and act as a driver of increased blood pressure. Using the Lewis polycystic kidney (LPK) rat model of chronic kidney disease, we examined respSNA in adult female rodents with CKD and their response to acute hypoxia or hypercapnia compared to Lewis control animals. Under urethane anesthesia, phrenic nerve activity, splanchnic sympathetic nerve activity (sSNA), and renal sympathetic nerve activity (rSNA) were recorded under baseline conditions and during mild hypoxic or hypercapnic challenges. At baseline, tonic SNA and blood pressure were greater in female LPK rats versus Lewis rats (all P < 0.05) and respSNA was at least two-fold larger [area under the curve (AUC), sSNA: 7.8 ± 1.1 vs. 3.4 ± 0.7 μV s, rSNA: 11.5 ± 3 vs. 4.8 ± 0.7 μV s, LPK vs. Lewis, both P < 0.05]. Mild hypoxia produced a larger pressure response in LPK [Δ mean arterial pressure (MAP) 30 ± 6 vs. 12 ± 6 mmHg] and augmented respSNA (ΔAUC, sSNA: 8.9 ± 3.4 vs. 2 ± 0.7 μV s, rSNA: 6.1 ± 1.2 vs. 3.1 ± 0.7 μV s, LPK vs. Lewis, all P ≤ 0.05). In contrast, central chemoreceptor stimulation produced comparable changes in blood pressure and respSNA (ΔMAP 13 ± 3 vs. 9 ± 5 mmHg; respSNA ΔAUC, sSNA: 2.5 ± 1 vs. 1.3 ± 0.7 μV s, rSNA: 4.2 ± 0.9 vs. 3.5 ± 1.4 μV s, LPK vs. Lewis, all P > 0.05). These results demonstrate that female rats with CKD exhibit heightened respSNA coupling at baseline that is further augmented by mild hypoxia, and not by hypercapnia. This mechanism may be a contributing driver of hypertension in this animal model of CKD.
Collapse
Affiliation(s)
- Manash Saha
- Department of Biomedical Sciences, Macquarie University, Sydney, NSW, Australia.,Department of Nephrology, National Institute of Kidney Disease and Urology, Dhaka, Bangladesh.,Graduate School of Medicine, Wollongong University, Wollongong, NSW, Australia
| | - Qi-Jian Sun
- Department of Biomedical Sciences, Macquarie University, Sydney, NSW, Australia
| | - Cara M Hildreth
- Department of Biomedical Sciences, Macquarie University, Sydney, NSW, Australia
| | - Peter G R Burke
- Department of Biomedical Sciences, Macquarie University, Sydney, NSW, Australia.,Neuroscience Research Australia, Sydney, NSW, Australia
| | | |
Collapse
|
26
|
Li S, Wang F, Sun D. The renal microcirculation in chronic kidney disease: novel diagnostic methods and therapeutic perspectives. Cell Biosci 2021; 11:90. [PMID: 34001267 PMCID: PMC8130426 DOI: 10.1186/s13578-021-00606-4] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2020] [Accepted: 05/06/2021] [Indexed: 12/14/2022] Open
Abstract
Chronic kidney disease (CKD) affects 8–16% of the population worldwide and is characterized by fibrotic processes. Understanding the cellular and molecular mechanisms underpinning renal fibrosis is critical to the development of new therapeutics. Microvascular injury is considered an important contributor to renal progressive diseases. Vascular endothelium plays a significant role in responding to physical and chemical signals by generating factors that help maintain normal vascular tone, inhibit leukocyte adhesion and platelet aggregation, and suppress smooth muscle cell proliferation. Loss of the rich capillary network results in endothelial dysfunction, hypoxia, and inflammatory and oxidative effects and further leads to the imbalance of pro- and antiangiogenic factors, endothelial cell apoptosis and endothelial-mesenchymal transition. New techniques, including both invasive and noninvasive techniques, offer multiple methods to observe and monitor renal microcirculation and guide targeted therapeutic strategies. A better understanding of the role of endothelium in CKD will help in the development of effective interventions for renal microcirculation improvement. This review focuses on the role of microvascular injury in CKD, the methods to detect microvessels and the novel treatments to ameliorate renal fibrosis.
Collapse
Affiliation(s)
- Shulin Li
- Department of Nephrology, Affiliated Hospital of Xuzhou Medical University, 99 West Huai-hai Road, Xuzhou, 221002, Jiangsu, China
| | - Fei Wang
- Department of Nephrology, Affiliated Hospital of Xuzhou Medical University, 99 West Huai-hai Road, Xuzhou, 221002, Jiangsu, China
| | - Dong Sun
- Department of Nephrology, Affiliated Hospital of Xuzhou Medical University, 99 West Huai-hai Road, Xuzhou, 221002, Jiangsu, China. .,Department of Internal Medicine and Diagnostics, Xuzhou Medical University, Xuzhou, 221002, China.
| |
Collapse
|
27
|
Detection of pro angiogenic and inflammatory biomarkers in patients with CKD. Sci Rep 2021; 11:8786. [PMID: 33888746 PMCID: PMC8062467 DOI: 10.1038/s41598-021-87710-0] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2020] [Accepted: 03/30/2021] [Indexed: 11/08/2022] Open
Abstract
Cardiovascular disease (CVD) is the most common cause of death in patients with native and post-transplant chronic kidney disease (CKD). To identify new biomarkers of vascular injury and inflammation, we analyzed the proteome of plasma and circulating extracellular vesicles (EVs) in native and post-transplant CKD patients utilizing an aptamer-based assay. Proteins of angiogenesis were significantly higher in native and post-transplant CKD patients versus healthy controls. Ingenuity pathway analysis (IPA) indicated Ephrin receptor signaling, serine biosynthesis, and transforming growth factor-β as the top pathways activated in both CKD groups. Pro-inflammatory proteins were significantly higher only in the EVs of native CKD patients. IPA indicated acute phase response signaling, insulin-like growth factor-1, tumor necrosis factor-α, and interleukin-6 pathway activation. These data indicate that pathways of angiogenesis and inflammation are activated in CKD patients' plasma and EVs, respectively. The pathways common in both native and post-transplant CKD may signal similar mechanisms of CVD.
Collapse
|
28
|
Díez J, Navarro-González JF, Ortiz A, Santamaría R, de Sequera P. Developing the subspecialty of cardio-nephrology: The time has come. A position paper from the coordinating committee from the Working Group for Cardiorenal Medicine of the Spanish Society of Nephrology. Nefrologia 2021. [PMID: 33892978 DOI: 10.1016/j.nefro.2021.02.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022] Open
Abstract
Patients with the dual burden of chronic kidney disease (CKD) and cardiovascular disease (CVD) experience unacceptably high rates of morbidity and mortality, which also entail unfavorable effects on healthcare systems. Currently, concerted efforts to identify, prevent and treat CVD in CKD patients are lacking at the institutional level, with emphasis still being placed on individual specialty views on this topic. The authors of this position paper endorse the need for a dedicated interdisciplinary team of subspecialists in cardio-nephrology that manages appropriate clinical interventions across the inpatient and outpatient settings. There is a critical need for training programs, guidelines and best clinical practice models, and research funding from nephrology, cardiology and other professional societies, to support the development of the subspecialty of cardio-nephrology. This position paper from the coordinating committee from the Working Group for Cardiorenal Medicine of the Spanish Society of Nephrology (S.E.N.) is intended to be the starting point to develop the subspecialty of cardio-nephrology within the S.E.N.. The implementation of the subspecialty in day-to-day nephrological practice will help to diagnose, treat, and prevent CVD in CKD patients in a precise, clinically effective, and health cost-favorable manner.
Collapse
Affiliation(s)
- Javier Díez
- Departments of Nephrology and Cardiology, University of Navarra Clinic, Pamplona, Spain; Program of Cardiovascular Diseases, Center of Applied Medical Research, University of Navarra, Pamplona, Spain.
| | - Juan F Navarro-González
- Division of Nephrology and Research Unit, University Hospital Nuestra Señora de Candelaria, and Universitary Institute of Biomedical Technologies, University of La Laguna, Santa Cruz de Tenerife, Spain; Red de Investigación Renal (REDINREN), Madrid, Spain
| | - Alberto Ortiz
- Red de Investigación Renal (REDINREN), Madrid, Spain; Division of Nephrology IIS-Fundacion Jimenez Diaz, University Autonoma of Madrid, Madrid, Spain
| | - Rafael Santamaría
- Red de Investigación Renal (REDINREN), Madrid, Spain; Division of Nephrology, University Hospital Reina Sofia, Cordoba, Spain; Maimonides Biomedical Research Institute of Cordoba (IMIBIC), Spain
| | - Patricia de Sequera
- Nephrology Department, Hospital Universitario Infanta Leonor, University Complutense of Madrid, Madrid, Spain
| |
Collapse
|
29
|
Querfeld U. Cardiovascular disease in childhood and adolescence: Lessons from children with chronic kidney disease. Acta Paediatr 2021; 110:1125-1131. [PMID: 33080082 DOI: 10.1111/apa.15630] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/04/2020] [Revised: 10/14/2020] [Accepted: 10/16/2020] [Indexed: 11/26/2022]
Abstract
Children suffering from chronic kidney disease (CKD) have the apparent highest risk for the development of cardiovascular disease (CVD) at a young age. While symptoms of CVD are characteristically absent in childhood and adolescence, remodelling of the myocardium, medium and large-sized arteries and of the microcirculation is clinically significant and can be assessed with non-invasive technology. Kidney disease and its progression are the driver of CVD, mediated by an unparalleled accumulation of risk factors converging on several comorbid conditions including hypertension, anaemia, dyslipidaemia, disturbed mineral metabolism and chronic persistent inflammation. Large prospective paediatric cohorts studies have provided valuable insights into the pathogenesis and the progression of CKD-induced cardiovascular comorbidity and have characterised the cardiovascular phenotype in young patients. They have also provided the rationale for close monitoring of risk factors and have defined therapeutic targets. Recently discovered new biomarkers could help identify the individual risk for CVD. Prevention of CVD by aggressive therapy of modifiable risk factors is essential to enable long-term survival of young patients with CKD.
Collapse
Affiliation(s)
- Uwe Querfeld
- Department of Pediatric Gastroenterology, Nephrology and Metabolic Diseases Charité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt‐Universität zu Berlin, and Berlin Institute of Health Berlin Germany
| |
Collapse
|
30
|
Krishnan S, Suarez-Martinez AD, Bagher P, Gonzalez A, Liu R, Murfee WL, Mohandas R. Microvascular dysfunction and kidney disease: Challenges and opportunities? Microcirculation 2021; 28:e12661. [PMID: 33025626 PMCID: PMC9990864 DOI: 10.1111/micc.12661] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Revised: 09/18/2020] [Accepted: 09/29/2020] [Indexed: 12/29/2022]
Abstract
Kidneys are highly vascular organs that despite their relatively small size receive 20% of the cardiac output. The highly intricate, delicately organized structure of renal microcirculation is essential to enable renal function and glomerular filtration rate through the local modulation of renal blood flow and intraglomerular pressure. Not surprisingly, the dysregulation of blood flow within the microvessels (abnormal vasoreactivity), fibrosis driven by disordered vascular-renal cross talk, or the loss of renal microvasculature (rarefaction) is associated with kidney disease. In addition, kidney disease can cause microcirculatory dysfunction in distant organs such as the heart and brain, mediated by mechanisms that remain to be elucidated. The objective of this review is to highlight the role of renal microvasculature in kidney disease. The overview will outline the impetus to study renal microvasculature, the bidirectional relationship between kidney disease and microvascular dysfunction, the key pathways driving microvascular diseases such as vasoreactivity, the cell dynamics coordinating fibrosis, and vessel rarefaction. Finally, we will also briefly highlight new therapies targeting the renal microvasculature to improve renal function.
Collapse
Affiliation(s)
- Suraj Krishnan
- Division of Nephrology, Hypertension & Transplantation, University of Florida College of Medicine, Gainesville, FL, USA
| | - Ariana D Suarez-Martinez
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL, USA
| | - Pooneh Bagher
- Department of Medical Physiology, Texas A&M University Health Science Center, Bryan, TX, USA
| | - Anjelica Gonzalez
- Department of Biomedical Engineering, Yale University, New Haven, CT, USA
| | - Ruisheng Liu
- Department of Molecular Pharmacology and Physiology, College of Medicine, University of South Florida, Tampa, FL, USA
| | - Walter L Murfee
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL, USA
| | - Rajesh Mohandas
- Division of Nephrology, Hypertension & Transplantation, University of Florida College of Medicine, Gainesville, FL, USA
| |
Collapse
|
31
|
Salyers ZR, Coleman M, Balestrieri NP, Ryan TE. Indoxyl sulfate impairs angiogenesis via chronic aryl hydrocarbon receptor activation. Am J Physiol Cell Physiol 2021; 320:C240-C249. [PMID: 33406025 DOI: 10.1152/ajpcell.00262.2020] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Chronic kidney disease (CKD) is associated with a substantial increased risk of cardiovascular disease. There is growing evidence that uremic metabolites, which accumulate in the blood with CKD, have detrimental impacts on endothelial cell health and function. However, the molecular mechanisms by which uremic metabolites negatively impact endothelial cell biology are not fully understood. In this study, activation of the aryl hydrocarbon receptor (AHR) via indoxyl sulfate, a known uremic metabolite, was found to impair endothelial cell tube formation and proliferation but not migratory function. Moreover, aortic ring cultures treated with indoxyl sulfate also exhibited decreased sprouting and high AHR activation. Next, genetic knockdown of the AHR using shRNA was found to rescue endothelial cell tube formation, proliferation, and aortic ring sprouting. Similarly, pharmacological AHR antagonism using resveratrol and CH223191 were also found to rescue angiogenesis in cell and aortic ring cultures. Finally, a constitutively active AHR (CAAHR) vector was generated and used to confirm AHR-specific effects. Expression of the CAAHR recapitulated the impaired tube formation and proliferation in cultured endothelial cells and decreased sprouting in aortic ring cultures. Taken together, these data define the impact of AHR activation on angiogenesis and highlight the potential for therapeutic AHR antagonists, which may improve angiogenesis in the context of CKD and cardiovascular disease.
Collapse
Affiliation(s)
- Zachary R Salyers
- Department of Applied Physiology and Kinesiology, University of Florida, Gainesville, Florida
| | - Madeline Coleman
- Department of Applied Physiology and Kinesiology, University of Florida, Gainesville, Florida
| | - Nicholas P Balestrieri
- Department of Applied Physiology and Kinesiology, University of Florida, Gainesville, Florida
| | - Terence E Ryan
- Department of Applied Physiology and Kinesiology, University of Florida, Gainesville, Florida.,Center for Exercise Science, University of Florida, Gainesville, Florida
| |
Collapse
|
32
|
Eroglu E, Kocyiğit I, Karakukcu C, Tuncay A, Zararsiz G, Eren D, Kahriman G, Hayri Sipahioglu M, Tokgoz B, Tasdemir K, Oymak O. Hypoxia-inducible factors in arteriovenous fistula maturation: A prospective cohort study. Eur J Clin Invest 2020; 50:e13350. [PMID: 32652532 DOI: 10.1111/eci.13350] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/12/2020] [Revised: 06/11/2020] [Accepted: 07/07/2020] [Indexed: 01/17/2023]
Abstract
BACKGROUND Neointimal hyperplasia is the main cause of arteriovenous fistula (AVF) failure. Hypoxia-inducible factors (HIFs) factors are associated with neointimal hyperplasia. Thus, we investigated the association between HIF-2 alpha (HIF-2α) and AVF maturation in end-stage kidney disease (ESKD) patients. METHODS This prospective cohort study was conducted in 21 voluntary healthy subjects and 50 patients with ESKD who were eligible for AVF creation. Inclusion criteria were being ESKD patients without a history of AVF surgery and dialysis. Eight patients excluded from the study due to having unavailable veins six patients were excluded due to acute thrombosis after surgery. One patient lost to follow-up. A total of 35 patients were included in final analysis. The blood samples were collected a day before the AVF surgery for biochemical parameters and HIF-2α measurement. HIF-2α levels were measured by the ELISA method. RESULTS Compared with healthy subjects, ESKD patients had a significantly higher level of HIF-2α. [1.3 (1.0-1.9) vs 2.2 (1.6-3.0)] (P = .002). Patients were divided into two groups after the evaluation of AVF maturation, as the mature group (n = 19) and the failure group (n = 16). Serum HIF-2α level was 1.7 (1.1-1.8) in the mature group; however, it was 3.1 (2.8-3.3 in failure group (P < .001). Multiple logistic regression analyses showed that HIF-2α independently predicted AVF maturation. The ROC curve analysis showed that HIF-2α > 2.65 predicted AVF maturation failure with the 87% sensitivity and 94% specificity [AUC:0.947, 95% CI (0.815-0.994), P < .001]. CONCLUSIONS HIF-2-α levels were higher in ESKD patients than healthy subjects. HIF-2-α could be a marker of AVF maturation failure.
Collapse
Affiliation(s)
- Eray Eroglu
- Department of Internal Medicine, Division of Nephrology, Erciyes University School of Medicine, Kayseri, Turkey.,Division of Renal Medicine and Baxter Novum, Karolinska Institutet, Stockholm, Sweden
| | - Ismail Kocyiğit
- Department of Internal Medicine, Division of Nephrology, Erciyes University School of Medicine, Kayseri, Turkey
| | - Cigdem Karakukcu
- Department of Biochemistry, Kayseri City Hospital, Kayseri, Turkey
| | - Aydin Tuncay
- Department of Cardiovascular Surgery, Erciyes University School of Medicine, Kayseri, Turkey
| | - Gokmen Zararsiz
- Roche Diagnostics GmbH, Personalized Health Care, Munich, Germany
| | - Davut Eren
- Department of Internal Medicine, Erciyes University School of Medicine, Kayseri, Turkey
| | - Guven Kahriman
- Department of Radiology, Erciyes University School of Medicine, Kayseri, Turkey
| | - Murat Hayri Sipahioglu
- Department of Internal Medicine, Division of Nephrology, Erciyes University School of Medicine, Kayseri, Turkey
| | - Bulent Tokgoz
- Department of Internal Medicine, Division of Nephrology, Erciyes University School of Medicine, Kayseri, Turkey
| | - Kutay Tasdemir
- Department of Cardiovascular Surgery, Erciyes University School of Medicine, Kayseri, Turkey
| | - Oktay Oymak
- Department of Internal Medicine, Division of Nephrology, Erciyes University School of Medicine, Kayseri, Turkey
| |
Collapse
|
33
|
Konst RE, Guzik TJ, Kaski JC, Maas AHEM, Elias-Smale SE. The pathogenic role of coronary microvascular dysfunction in the setting of other cardiac or systemic conditions. Cardiovasc Res 2020; 116:817-828. [PMID: 31977015 PMCID: PMC7526753 DOI: 10.1093/cvr/cvaa009] [Citation(s) in RCA: 54] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/15/2019] [Revised: 12/09/2019] [Accepted: 01/21/2020] [Indexed: 12/13/2022] Open
Abstract
Coronary microvascular dysfunction (CMD) plays a pathogenic role in cardiac and systemic conditions other than microvascular angina. In this review, we provide an overview of the pathogenic role of CMD in the setting of diabetes mellitus, obesity, hypertensive pregnancy disorders, chronic inflammatory and autoimmune rheumatic disorders, chronic kidney disease, hypertrophic cardiomyopathy, and aortic valve stenosis. In these various conditions, CMD results from different structural, functional, and/or dynamic alterations in the coronary microcirculation associated with the primary disease process. CMD is often detectable very early in the course of the primary disease, before clinical symptoms or signs of myocardial ischaemia are present, and it portrays an increased risk for cardiovascular events.
Collapse
Affiliation(s)
- Regina E Konst
- Department of Cardiology, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Tomasz J Guzik
- Institute of Cardiovascular and Medical Sciences, British Heart Foundation Glasgow Cardiovascular Research Centre, Glasgow, UK
| | - Juan-Carlos Kaski
- The Queen Elizabeth Hospital Discipline of Medicine, University of Adelaide, Central Adelaide Local Health Network, Coronary Vasomotion Disorders International Study Group (COVADIS), Adelaide, Australia.,Molecular and Clinical Sciences Research Institute, St George's, University of London, London, UK
| | - Angela H E M Maas
- Department of Cardiology, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Suzette E Elias-Smale
- Department of Cardiology, Radboud University Medical Center, Nijmegen, the Netherlands
| |
Collapse
|
34
|
Microvascular disease in chronic kidney disease: the base of the iceberg in cardiovascular comorbidity. Clin Sci (Lond) 2020; 134:1333-1356. [PMID: 32542397 PMCID: PMC7298155 DOI: 10.1042/cs20200279] [Citation(s) in RCA: 54] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2020] [Revised: 05/29/2020] [Accepted: 06/08/2020] [Indexed: 02/07/2023]
Abstract
Chronic kidney disease (CKD) is a relentlessly progressive disease with a very high mortality mainly due to cardiovascular complications. Endothelial dysfunction is well documented in CKD and permanent loss of endothelial homeostasis leads to progressive organ damage. Most of the vast endothelial surface area is part of the microcirculation, but most research in CKD-related cardiovascular disease (CVD) has been devoted to macrovascular complications. We have reviewed all publications evaluating structure and function of the microcirculation in humans with CKD and animals with experimental CKD. Microvascular rarefaction, defined as a loss of perfused microvessels resulting in a significant decrease in microvascular density, is a quintessential finding in these studies. The median microvascular density was reduced by 29% in skeletal muscle and 24% in the heart in animal models of CKD and by 32% in human biopsy, autopsy and imaging studies. CKD induces rarefaction due to the loss of coherent vessel systems distal to the level of smaller arterioles, generating a typical heterogeneous pattern with avascular patches, resulting in a dysfunctional endothelium with diminished perfusion, shunting and tissue hypoxia. Endothelial cell apoptosis, hypertension, multiple metabolic, endocrine and immune disturbances of the uremic milieu and specifically, a dysregulated angiogenesis, all contribute to the multifactorial pathogenesis. By setting the stage for the development of tissue fibrosis and end organ failure, microvascular rarefaction is a principal pathogenic factor in the development of severe organ dysfunction in CKD patients, especially CVD, cerebrovascular dysfunction, muscular atrophy, cachexia, and progression of kidney disease. Treatment strategies for microvascular disease are urgently needed.
Collapse
|
35
|
Vancheri F, Longo G, Vancheri S, Henein M. Coronary Microvascular Dysfunction. J Clin Med 2020; 9:E2880. [PMID: 32899944 PMCID: PMC7563453 DOI: 10.3390/jcm9092880] [Citation(s) in RCA: 174] [Impact Index Per Article: 34.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2020] [Revised: 09/02/2020] [Accepted: 09/02/2020] [Indexed: 01/09/2023] Open
Abstract
Many patients with chest pain undergoing coronary angiography do not show significant obstructive coronary lesions. A substantial proportion of these patients have abnormalities in the function and structure of coronary microcirculation due to endothelial and smooth muscle cell dysfunction. The coronary microcirculation has a fundamental role in the regulation of coronary blood flow in response to cardiac oxygen requirements. Impairment of this mechanism, defined as coronary microvascular dysfunction (CMD), carries an increased risk of adverse cardiovascular clinical outcomes. Coronary endothelial dysfunction accounts for approximately two-thirds of clinical conditions presenting with symptoms and signs of myocardial ischemia without obstructive coronary disease, termed "ischemia with non-obstructive coronary artery disease" (INOCA) and for a small proportion of "myocardial infarction with non-obstructive coronary artery disease" (MINOCA). More frequently, the clinical presentation of INOCA is microvascular angina due to CMD, while some patients present vasospastic angina due to epicardial spasm, and mixed epicardial and microvascular forms. CMD may be associated with focal and diffuse epicardial coronary atherosclerosis, which may reinforce each other. Both INOCA and MINOCA are more common in females. Clinical classification of CMD includes the association with conditions in which atherosclerosis has limited relevance, with non-obstructive atherosclerosis, and with obstructive atherosclerosis. Several studies already exist which support the evidence that CMD is part of systemic microvascular disease involving multiple organs, such as brain and kidney. Moreover, CMD is strongly associated with the development of heart failure with preserved ejection fraction (HFpEF), diabetes, hypertensive heart disease, and also chronic inflammatory and autoimmune diseases. Since coronary microcirculation is not visible on invasive angiography or computed tomographic coronary angiography (CTCA), the diagnosis of CMD is usually based on functional assessment of microcirculation, which can be performed by both invasive and non-invasive methods, including the assessment of delayed flow of contrast during angiography, measurement of coronary flow reserve (CFR) and index of microvascular resistance (IMR), evaluation of angina induced by intracoronary acetylcholine infusion, and assessment of myocardial perfusion by positron emission tomography (PET) and magnetic resonance (CMR).
Collapse
Affiliation(s)
- Federico Vancheri
- Department of Internal Medicine, S.Elia Hospital, 93100 Caltanissetta, Italy
| | - Giovanni Longo
- Cardiovascular and Interventional Department, S.Elia Hospital, 93100 Caltanissetta, Italy;
| | - Sergio Vancheri
- Radiology Department, I.R.C.C.S. Policlinico San Matteo, 27100 Pavia, Italy;
| | - Michael Henein
- Institute of Public Health and Clinical Medicine, Umea University, SE-90187 Umea, Sweden;
- Department of Fluid Mechanics, Brunel University, Middlesex, London UB8 3PH, UK
- Molecular and Nuclear Research Institute, St George’s University, London SW17 0RE, UK
| |
Collapse
|
36
|
Kuo W, Le NA, Spingler B, Wenger RH, Kipar A, Hetzel U, Schulz G, Müller B, Kurtcuoglu V. Simultaneous Three-Dimensional Vascular and Tubular Imaging of Whole Mouse Kidneys With X-ray μCT. MICROSCOPY AND MICROANALYSIS : THE OFFICIAL JOURNAL OF MICROSCOPY SOCIETY OF AMERICA, MICROBEAM ANALYSIS SOCIETY, MICROSCOPICAL SOCIETY OF CANADA 2020; 26:731-740. [PMID: 32627730 DOI: 10.1017/s1431927620001725] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
Concurrent three-dimensional imaging of the renal vascular and tubular systems on the whole-kidney scale with capillary level resolution is labor-intensive and technically difficult. Approaches based on vascular corrosion casting and X-ray micro computed tomography (μCT), for example, suffer from vascular filling artifacts and necessitate imaging with an additional modality to acquire tubules. In this work, we report on a new sample preparation, image acquisition, and quantification protocol for simultaneous vascular and tubular μCT imaging of whole, uncorroded mouse kidneys. The protocol consists of vascular perfusion with the water-soluble, aldehyde-fixable, polymeric X-ray contrast agent XlinCA, followed by laboratory-source μCT imaging and structural analysis using the freely available Fiji/ImageJ software. We achieved consistent filling of the entire capillary bed and staining of the tubules in the cortex and outer medulla. After imaging at isotropic voxel sizes of 3.3 and 4.4 μm, we segmented vascular and tubular systems and quantified luminal volumes, surface areas, diffusion distances, and vessel path lengths. This protocol permits the analysis of vascular and tubular parameters with higher reliability than vascular corrosion casting, less labor than serial sectioning and leaves tissue intact for subsequent histological examination with light and electron microscopy.
Collapse
Affiliation(s)
- Willy Kuo
- University of Zurich, Institute of Physiology, Winterthurerstrasse 190, 8057Zurich, Switzerland
- University of Zurich, National Centre of Competence in Research, Kidney. CH, Winterthurerstrasse 190, 8057Zurich, Switzerland
- University of Basel, Biomaterials Science Center, Department of Biomedical Engineering, Gewerbestrasse 14, 4123Allschwil, Switzerland
| | - Ngoc An Le
- University of Zurich, Department of Chemistry, Winterthurerstrasse 190, 8057Zurich, Switzerland
| | - Bernhard Spingler
- University of Zurich, Department of Chemistry, Winterthurerstrasse 190, 8057Zurich, Switzerland
| | - Roland H Wenger
- University of Zurich, Institute of Physiology, Winterthurerstrasse 190, 8057Zurich, Switzerland
- University of Zurich, National Centre of Competence in Research, Kidney. CH, Winterthurerstrasse 190, 8057Zurich, Switzerland
| | - Anja Kipar
- University of Zurich, Laboratory for Animal Model Pathology (LAMP), Institute of Veterinary Pathology, Vetsuisse Faculty, Winterthurerstrasse 268, 8057Zurich, Switzerland
| | - Udo Hetzel
- University of Zurich, Electron Microscopy Unit, Institute of Veterinary Pathology, Vetsuisse Faculty, Winterthurerstrasse 268, 8057Zurich, Switzerland
| | - Georg Schulz
- University of Basel, Biomaterials Science Center, Department of Biomedical Engineering, Gewerbestrasse 14, 4123Allschwil, Switzerland
| | - Bert Müller
- University of Basel, Biomaterials Science Center, Department of Biomedical Engineering, Gewerbestrasse 14, 4123Allschwil, Switzerland
| | - Vartan Kurtcuoglu
- University of Zurich, Institute of Physiology, Winterthurerstrasse 190, 8057Zurich, Switzerland
- University of Zurich, National Centre of Competence in Research, Kidney. CH, Winterthurerstrasse 190, 8057Zurich, Switzerland
- University of Zurich, Zurich Center for Integrative Human Physiology, 8057Zurich, Switzerland
| |
Collapse
|
37
|
Kaesler N, Babler A, Floege J, Kramann R. Cardiac Remodeling in Chronic Kidney Disease. Toxins (Basel) 2020; 12:toxins12030161. [PMID: 32150864 PMCID: PMC7150902 DOI: 10.3390/toxins12030161] [Citation(s) in RCA: 76] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2020] [Revised: 03/02/2020] [Accepted: 03/03/2020] [Indexed: 12/19/2022] Open
Abstract
Cardiac remodeling occurs frequently in chronic kidney disease patients and affects quality of life and survival. Current treatment options are highly inadequate. As kidney function declines, numerous metabolic pathways are disturbed. Kidney and heart functions are highly connected by organ crosstalk. Among others, altered volume and pressure status, ischemia, accelerated atherosclerosis and arteriosclerosis, disturbed mineral metabolism, renal anemia, activation of the renin-angiotensin system, uremic toxins, oxidative stress and upregulation of cytokines stress the sensitive interplay between different cardiac cell types. The fatal consequences are left-ventricular hypertrophy, fibrosis and capillary rarefaction, which lead to systolic and/or diastolic left-ventricular failure. Furthermore, fibrosis triggers electric instability and sudden cardiac death. This review focuses on established and potential pathophysiological cardiorenal crosstalk mechanisms that drive uremia-induced senescence and disease progression, including potential known targets and animal models that might help us to better understand the disease and to identify novel therapeutics.
Collapse
Affiliation(s)
- Nadine Kaesler
- Clinic for Renal and Hypertensive Disorders, Rheumatological and Immunological Disease, University Hospital of the RWTH Aachen, 52074 Aachen, Germany
| | - Anne Babler
- Clinic for Renal and Hypertensive Disorders, Rheumatological and Immunological Disease, University Hospital of the RWTH Aachen, 52074 Aachen, Germany
| | - Jürgen Floege
- Clinic for Renal and Hypertensive Disorders, Rheumatological and Immunological Disease, University Hospital of the RWTH Aachen, 52074 Aachen, Germany
| | - Rafael Kramann
- Clinic for Renal and Hypertensive Disorders, Rheumatological and Immunological Disease, University Hospital of the RWTH Aachen, 52074 Aachen, Germany
- Department of Internal Medicine, Nephrology and Transplantation, Erasmus Medical Center, 3015 GD Rotterdam, The Netherlands
- Correspondence:
| |
Collapse
|
38
|
Querfeld U, Schaefer F. Cardiovascular risk factors in children on dialysis: an update. Pediatr Nephrol 2020; 35:41-57. [PMID: 30382333 DOI: 10.1007/s00467-018-4125-x] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/14/2018] [Revised: 10/16/2018] [Accepted: 10/19/2018] [Indexed: 02/07/2023]
Abstract
Cardiovascular disease (CVD) is a life-limiting comorbidity in patients with chronic kidney disease (CKD). In childhood, imaging studies have demonstrated early phenotypic characteristics including increases in left ventricular mass, carotid artery intima-media thickness, and pulse wave velocity, which occur even in young children with early stages of CKD. Vascular calcifications are the signature of an advanced phenotype and are mainly found in adolescents and young adults treated with dialysis. Association studies have provided valuable information regarding the significance of a multitude of risk factors in promoting CVD in children with CKD by using intermediate endpoints of measurements of surrogate parameters of CVD. Dialysis aggravates pre-existing risk factors and accelerates the progression of CVD with additional dialysis-related risk factors. Coronary artery calcifications in children and young adults with CKD accumulate in a time-dependent manner on dialysis. Identification of risk factors has led to improved understanding of principal mechanisms of CKD-induced damage to the cardiovascular system. Treatment strategies include assessment and monitoring of individual risk factor load, optimization of treatment of modifiable risk factors, and intensified hemodialysis if early transplantation is not possible.
Collapse
Affiliation(s)
- Uwe Querfeld
- Department of Pediatrics, Division of Gastroenterology, Nephrology and Metabolic Medicine, Charité Universitätsmedizin Berlin, Berlin, Germany.
| | - Franz Schaefer
- Pediatric Nephrology Division, Center for Pediatrics and Adolescent Medicine, University of Heidelberg, Heidelberg, Germany
| |
Collapse
|
39
|
Evans RG, Smith DW, Lee C, Ngo JP, Gardiner BS. What Makes the Kidney Susceptible to Hypoxia? Anat Rec (Hoboken) 2019; 303:2544-2552. [DOI: 10.1002/ar.24260] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2019] [Revised: 04/24/2019] [Accepted: 05/13/2019] [Indexed: 12/19/2022]
Affiliation(s)
- Roger G. Evans
- Cardiovascular Disease Program, Biomedicine Discovery Institute and Department of Physiology Monash University Melbourne Victoria Australia
| | - David W. Smith
- Faculty of Engineering and Mathematical Sciences The University of Western Australia Perth Western Australia Australia
| | - Chang‐Joon Lee
- Faculty of Engineering and Mathematical Sciences The University of Western Australia Perth Western Australia Australia
- College of Science, Health, Engineering and Education Murdoch University Perth Western Australia Australia
| | - Jennifer P. Ngo
- Cardiovascular Disease Program, Biomedicine Discovery Institute and Department of Physiology Monash University Melbourne Victoria Australia
| | - Bruce S. Gardiner
- Faculty of Engineering and Mathematical Sciences The University of Western Australia Perth Western Australia Australia
- College of Science, Health, Engineering and Education Murdoch University Perth Western Australia Australia
| |
Collapse
|
40
|
PBI-4050 via GPR40 activation improves adenine-induced kidney injury in mice. Clin Sci (Lond) 2019; 133:1587-1602. [PMID: 31308217 DOI: 10.1042/cs20190479] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2019] [Revised: 07/08/2019] [Accepted: 07/15/2019] [Indexed: 02/06/2023]
Abstract
PBI-4050 (3-pentylbenzenacetic acid sodium salt), a novel first-in-class orally active compound that has completed clinical Phases Ib and II in subjects with chronic kidney disease (CKD) and metabolic syndrome respectively, exerts antifibrotic effects in several organs via a novel mechanism of action, partly through activation of the G protein receptor 40 (GPR40) receptor. Here we evaluate the effects of PBI-4050 in both WT and Gpr40-/- mice on adenine-induced tubulointerstitial injury, anemia and activation of the unfolded protein response (UPR) pathway. Adenine-induced CKD was achieved in 8-week-old C57BL/6 mice fed a diet supplemented with 0.25% adenine. After 1 week, PBI-4050 or vehicle was administered daily by oral-gavage for 3 weeks. Gpr40-/- mice were also subjected to adenine-feeding, with or without PBI-4050 treatment. PBI-4050 improved renal function and urine concentrating ability. Anemia was present in adenine-fed mice, while PBI-4050 blunted these effects and led to significantly higher plasma erythropoietin (EPO) levels. Adenine-induced renal fibrosis, endoplasmic reticulum (ER) stress and apoptosis were significantly decreased by PBI-4050. In parallel, Gpr40-/- mice were more susceptible to adenine-induced fibrosis, renal function impairment, anemia and ER stress compared with WT mice. Importantly, PBI-4050 treatment in Gpr40-/- mice failed to reduce renal injury in this model. Taken together, PBI-4050 prevented adenine-induced renal injury while these beneficial effects were lost upon Gpr40 deletion. These data reinforce PBI-4050's use as a renoprotective therapy and identify GPR40 as a crucial mediator of its beneficial effects.
Collapse
|
41
|
Hirunpattarasilp C, Attwell D, Freitas F. The role of pericytes in brain disorders: from the periphery to the brain. J Neurochem 2019; 150:648-665. [PMID: 31106417 DOI: 10.1111/jnc.14725] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2019] [Revised: 03/15/2019] [Accepted: 05/15/2019] [Indexed: 12/13/2022]
Abstract
It is becoming increasingly apparent that disorders of the brain microvasculature contribute to many neurological disorders. In recent years it has become clear that a major player in these events is the capillary pericyte which, in the brain, is now known to control the blood-brain barrier, regulate blood flow, influence immune cell entry and be crucial for angiogenesis. In this review we consider the under-explored possibility that peripheral diseases which affect the microvasculature, such as hypertension, kidney disease and diabetes, produce central nervous system (CNS) dysfunction by mechanisms affecting capillary pericytes within the CNS. We highlight how cellular messengers produced peripherally can act via signalling pathways within CNS pericytes to reshape blood vessels, restrict blood flow or compromise blood-brain barrier function, thus causing neuronal dysfunction. Increased understanding of how renin-angiotensin, Rho-kinase and PDGFRβ signalling affect CNS pericytes may suggest novel therapeutic approaches to reducing the CNS effects of peripheral disorders.
Collapse
Affiliation(s)
- Chanawee Hirunpattarasilp
- Department of Neuroscience, Andrew Huxley Building, University College London, Physiology & Pharmacology, Gower Street, London, UK
| | - David Attwell
- Department of Neuroscience, Andrew Huxley Building, University College London, Physiology & Pharmacology, Gower Street, London, UK
| | - Felipe Freitas
- Department of Neuroscience, Andrew Huxley Building, University College London, Physiology & Pharmacology, Gower Street, London, UK
| |
Collapse
|
42
|
Bersani-Amado LE, da Rocha BA, Schneider LCL, Ames FQ, Breithaupt Faloppa AC, Araújo GB, Dantas JA, Bersani-Amado CA, Cuman RKN. Nephropathy induced by renal microembolism: a characterization of biochemical and histopathological changes in rats. INTERNATIONAL JOURNAL OF CLINICAL AND EXPERIMENTAL PATHOLOGY 2019; 12:2311-2323. [PMID: 31934059 PMCID: PMC6949639] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 03/08/2019] [Accepted: 03/27/2019] [Indexed: 06/10/2023]
Abstract
The aim of this study was to investigate some biochemical parameters of renal function and the vascular, glomerular, tubular, and interstitial manifestations in the progression of nephropathy induced by renal microembolism. Renal microembolism was induced by the arterial injection of polymethacrylate microspheres in the remnant kidney of nephrectomized rats. Animals 110-120 days old were randomly divided into three groups: the control group (C; normal), the nephrectomized group (S; nephrectomized that did not undergo renal microembolism), and the model group (M, nephrectomized animals that underwent renal arterial microembolism). The animals were evaluated 30, 60, and 90 days after the induction of a renal microembolism. Blood and urine samples were collected to determine serum creatinine (Cr) and urea (Ur) concentrations and urine total protein (Pt) concentrations. The kidneys were weighed and processed for histopathological analysis using hematoxylin and eosin (HE), periodic acid-Schiff (PAS), Mallory-Azan, and Picro-Sirius staining. The samples were also subjected to immunohistochemistry with a proliferating cell nuclear antigen (PCNA) and a vascular endothelial growth factor receptor (VEGFR). The data demonstrated evidence of the occurrence of vascular, glomerular, tubular, and interstitial abnormalities in the renal tissue, and changes in the biochemical parameters of renal function (serum Cr and Ur and of 24-h urine Pt) in this experimental model of nephropathy induced by renal microembolism, which may indicate the development of chronic kidney disease (CKD). Additionally, the findings indicate that this is a good reproducibility model that may be useful for studying the pathogenesis of CKD that is caused by atheroembolism and possible treatment alternatives.
Collapse
Affiliation(s)
| | | | | | | | - Ana Cristina Breithaupt Faloppa
- Laboratory of Cardiovascular Surgery and Circulation Pathophysiology (LIM-11), Heart Institute (InCor), University of Sao Paulo Medical SchoolSão Paulo, Brazil
| | | | - Jailson Araujo Dantas
- Department of Pharmacology and Therapeutics, State University of MaringáParana, Brazil
| | | | | |
Collapse
|
43
|
Arinze NV, Gregory A, Francis JM, Farber A, Chitalia VC. Unique aspects of peripheral artery disease in patients with chronic kidney disease. Vasc Med 2019; 24:251-260. [PMID: 30823859 DOI: 10.1177/1358863x18824654] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Peripheral artery disease (PAD) represents a major health care burden. Despite the advent of screening and interventional procedures, the long-term clinical outcomes remain suboptimal, especially in patients with chronic kidney disease (CKD). While CKD and PAD share common predisposing factors, emerging studies indicate that their co-existence is not merely an association; instead, CKD represents a strong, independent risk factor for PAD. These findings implicate CKD-specific mediators of PAD that remain incompletely understood. Moreover, there is a need to understand the mechanisms underlying poor outcomes after interventions for PAD in CKD. This review discusses unique clinical aspects of PAD in patients with CKD, including high prevalence and worse outcomes after vascular interventions and the influence of renal allograft transplantation. In doing so, it also highlights underappreciated aspects of PAD in patients with CKD, such as disparities in revascularization and higher peri-procedural mortality. While previous reviews have discussed general mechanisms of PAD pathogenesis, focusing on PAD in CKD, this review underscores a need to probe for CKD-specific pathogenic pathways that may unravel novel biomarkers and therapeutic targets in PAD and ultimately improve the risk stratification and management of patients with CKD and PAD.
Collapse
Affiliation(s)
- Nkiruka V Arinze
- 1 Division of Vascular and Endovascular Surgery, Boston Medical Center, Boston University School of Medicine, Boston, MA, USA.,2 Renal Section, Department of Medicine, Boston Medical Center, Boston University School of Medicine, Boston, MA, USA
| | | | - Jean M Francis
- 2 Renal Section, Department of Medicine, Boston Medical Center, Boston University School of Medicine, Boston, MA, USA
| | - Alik Farber
- 1 Division of Vascular and Endovascular Surgery, Boston Medical Center, Boston University School of Medicine, Boston, MA, USA
| | - Vipul C Chitalia
- 2 Renal Section, Department of Medicine, Boston Medical Center, Boston University School of Medicine, Boston, MA, USA.,4 Whitaker Cardiovascular Institute, Boston University School of Medicine, Boston, MA, USA.,5 Veterans Affairs Boston Healthcare System, Boston, MA, USA
| |
Collapse
|
44
|
Lai J, Wu Y, Hang L, Gael A, Deng T, Yan Q, Fu Q, Li Z. [Zhenwu Decoction delays ventricular hypertrophy in rats with uremic cardiomyopathy]. NAN FANG YI KE DA XUE XUE BAO = JOURNAL OF SOUTHERN MEDICAL UNIVERSITY 2019; 39:113-119. [PMID: 30692076 DOI: 10.12122/j.issn.1673-4254.2019.01.18] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
OBJECTIVE To investigate the inhibitory effect of Zhenwu Decoction on ventricular hypertrophy in rats with uremic cardiomyopathy and explore the mechanism. METHODS Cardiocytes isolated from suckling rats were divided into control group and indoxyl sulfate (IS) group, and the protein synthesis was assayed with [3H]- leucine incorporation and cellular protein expressions were detected using Western blotting. Fifty SD rats were randomly divided into sham operation group, model group, and low- and high-dose Zhenwu Decoction treatment groups, and except for those in the sham operation group, all the rats underwent 5/6 nephrectomy. Four weeks after the operation, the rats in low- and high-dose treatment groups were given Zhenwu Decoction via gavage at the dose of 4.5 g/kg and 13.5 g/kg, respectively; the rats in the sham-operated and model groups were given an equal volume of distilled water. After 4 weeks of treatment, serum levels of IS were determined, and cardiac and ventricular mass indexes were measured in the rats; cardiac ultrasound was performed and Western blotting was used to measure the expressions of BNP, p-ERK1/2, p-p38 and p-JNK in the myocardium. RESULTS Rat cardiomyocytes treated with IS showed significantly enhanced protein synthesis and increased expression levels of BNP, p-erk1/2, and p-p38 as compared with the control cells (P < 0.01), but the expression of p-jnk was comparable between the two groups. In the animal experiment, the rats in the model group showed significantly increased serum creatinine (SCr) and urea nitrogen (BUN) levels, 24-h urine protein (24 hUpro), plasma IS level, left ventricular mass index (LVMI) and whole heart mass index (HMI) compared with those in the sham group (P < 0.01); Both LVESD and LVEDD were significantly reduced and LVAWS, LVAWD, LVPWS and LVPWD were significantly increased in the model rat, which also presented with obvious cardiomyocyte hypertrophy and increased myocardial expressions of BNP, p-ERK1/2, p-p38 and p-jnk (P < 0.01). Compared with the rats in the model group, the rats treated with low-dose and high-dose Zhenwu Decoction had significantly lowered levels of SCr, BUN, 24 hUpro and IS (P < 0.05) and decreased LVMI and HMI; LVESD, LVEDD, LVPWS, LVAWS, and LVAWD were improved more obviously in the high-dose group, and the myocardial expressions of BNP, p-ERK1/2, p-p38 and p-JNK was significantly downregulated after the treatment. CONCLUSIONS Zhenwu Decoctin can reduce plasma IS levels and inhibit ventricular hypertrophy to delay ventricular remodeling in rats with uremic cardiomyopathy.
Collapse
Affiliation(s)
- Jun Lai
- Department of Cardiology, Heart Center, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China
| | - Yingzhi Wu
- Department of Cardiology, Heart Center, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China
| | - Liwei Hang
- Department of Cardiology, Heart Center, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China
| | - Akindavyi Gael
- Department of Cardiology, Heart Center, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China
| | - Ting Deng
- Department of Cardiology, Heart Center, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China
| | - Quanneng Yan
- Department of Cardiology, Heart Center, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China
| | - Qiang Fu
- Department of Cardiology, Heart Center, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China
| | - Zhiliang Li
- Department of Cardiology, Heart Center, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China
| |
Collapse
|