1
|
Kim K, Thome T, Pass C, Stone L, Vugman N, Palzkill V, Yang Q, O’Malley KA, Anderson EM, Fazzone B, Yue F, Berceli SA, Scali ST, Ryan TE. Multiomic Analysis of Calf Muscle in Peripheral Artery Disease and Chronic Kidney Disease. Circ Res 2025; 136:688-703. [PMID: 39963788 PMCID: PMC11949227 DOI: 10.1161/circresaha.124.325642] [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: 10/01/2024] [Revised: 02/04/2025] [Accepted: 02/07/2025] [Indexed: 03/06/2025]
Abstract
BACKGROUND Chronic kidney disease (CKD) has emerged as a significant risk factor that accelerates atherosclerosis, decreases muscle function, and increases the risk of amputation or death in patients with peripheral artery disease (PAD). However, the modulators underlying this exacerbated pathobiology are ill-defined. Recent work has demonstrated that uremic toxins are associated with limb amputation in PAD and have pathological effects in both the limb muscle and vasculature. Herein, we use multiomics to identify novel modulators of disease pathobiology in patients with PAD and CKD. METHODS A cross-sectional study enrolled 4 groups of participants: controls without PAD or CKD (n=28), patients with PAD only (n=46), patients with CKD only (n=31), and patients with both PAD and CKD (n=18). Both targeted (uremic toxins) and nontargeted metabolomics in plasma were performed using mass spectrometry. Calf muscle biopsies were used to measure histopathology, perform bulk and single-nucleus RNA sequencing, and assess mitochondrial function. Differential gene and metabolite analyses, as well as pathway and gene set enrichment analyses, were performed. RESULTS Patients with both PAD and CKD exhibited significantly lower calf muscle strength and smaller muscle fiber areas compared with controls and those with only PAD. Compared with controls, mitochondrial function was impaired in patients with CKD, with or without PAD, but not in PAD patients without CKD. Plasma metabolomics revealed substantial alterations in the metabolome of patients with CKD, with significant correlations observed between uremic toxins (eg, kynurenine and indoxyl sulfate) and both muscle strength and mitochondrial function. RNA sequencing analyses identified downregulation of mitochondrial genes and pathways associated with protein translation in patients with both PAD and CKD. Single-nucleus RNA sequencing further highlighted a mitochondrial deficiency in muscle fibers along with unique remodeling of fibro-adipogenic progenitor cells in patients with both PAD and CKD, with an increase in adipogenic cell populations. CONCLUSIONS CKD significantly exacerbates ischemic muscle pathology in PAD, as evidenced by diminished muscle strength, reduced mitochondrial function, and altered transcriptome profiles. The correlation between uremic toxins and muscle dysfunction suggests that targeting these metabolites may offer therapeutic potential for improving muscle health in PAD patients with CKD.
Collapse
Affiliation(s)
- Kyoungrae Kim
- Department of Applied Physiology and Kinesiology (K.K., T.T., C.P., L.S., N.V., V.P., Q.Y., T.E.R.)
| | - Trace Thome
- Department of Applied Physiology and Kinesiology (K.K., T.T., C.P., L.S., N.V., V.P., Q.Y., T.E.R.)
| | - Caroline Pass
- Department of Applied Physiology and Kinesiology (K.K., T.T., C.P., L.S., N.V., V.P., Q.Y., T.E.R.)
| | - Lauren Stone
- Department of Applied Physiology and Kinesiology (K.K., T.T., C.P., L.S., N.V., V.P., Q.Y., T.E.R.)
| | - Nicholas Vugman
- Department of Applied Physiology and Kinesiology (K.K., T.T., C.P., L.S., N.V., V.P., Q.Y., T.E.R.)
| | - Victoria Palzkill
- Department of Applied Physiology and Kinesiology (K.K., T.T., C.P., L.S., N.V., V.P., Q.Y., T.E.R.)
| | - Qingping Yang
- Department of Applied Physiology and Kinesiology (K.K., T.T., C.P., L.S., N.V., V.P., Q.Y., T.E.R.)
| | | | | | - Brian Fazzone
- Department of Surgery (K.A.O., E.M.A., B.F., S.A.B., S.T.S.)
| | - Feng Yue
- Department of Animal Sciences (F.Y.)
- Myology Institute (F.Y., T.E.R.)
| | | | | | - Terence E. Ryan
- Department of Applied Physiology and Kinesiology (K.K., T.T., C.P., L.S., N.V., V.P., Q.Y., T.E.R.)
- Center for Exercise Science (T.E.R.)
- Myology Institute (F.Y., T.E.R.)
- University of Florida, Gainesville (T.E.R.)
| |
Collapse
|
2
|
Behrens F, Holle J, Chen C, Ginsbach LF, Krause BC, Bruning U, Kriegel FL, Kaiser T, Szijártó IA, Anandakumar H, Lehmann K, Schumacher F, Durek P, Heinrich FF, Lodka D, Hoffmann C, Borchardt AA, Peters L, Michalick L, Querfeld U, Bufler P, Luch A, Kleuser B, Kirwan JA, Forslund SK, Thumfahrt J, Müller D, Wilck N, Mashreghi M, Löber U, Bartolomaeus H, Kuebler WM, Simmons S. Circulating Extracellular Vesicles as Putative Mediators of Cardiovascular Disease in Paediatric Chronic Kidney Disease. J Extracell Vesicles 2025; 14:e70062. [PMID: 40116365 PMCID: PMC11926757 DOI: 10.1002/jev2.70062] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Revised: 01/07/2025] [Accepted: 02/11/2025] [Indexed: 03/23/2025] Open
Abstract
Cardiovascular disease (CVD) is the leading cause of mortality in chronic kidney disease (CKD). However, the pathogenesis of CVD in CKD remains incompletely understood. Endothelial extracellular vesicles (EC-EVs) have previously been associated with CVD. We hypothesized that CKD alters EV release and cargo, subsequently promoting vascular remodelling. We recruited 94 children with CKD, including patients after kidney transplantation and healthy donors, and performed EV phenotyping and functional EV analyses in the absence of age-related comorbidities. Plasma EC-EVs were increased in haemodialysis patients and decreased after kidney transplantation. Thirty microRNAs were less abundant in total CKD plasma EVs with predicted importance in angiogenesis and smooth muscle cell proliferation. In vitro, CKD plasma EVs induced transcriptomic changes in angiogenesis pathways and functionally impaired angiogenic properties, migration and proliferation in ECs. High shear stress, as generated by arterio-venous fistulas, and uremic toxins were considered as potential drivers of EV release, but only the combination increased EV generation from venous ECs. The resulting EVs recapitulated miRNA changes observed in CKD in vivo. In conclusion, CKD results in the release of EVs with altered miRNA profiles and anti-angiogenic properties, which may mediate vascular pathology in children with CKD. EVs and their miRNA cargo may represent future therapeutic targets to attenuate CVD in CKD.
Collapse
Affiliation(s)
- Felix Behrens
- Institute of PhysiologyCharité – Universitätsmedizin BerlinBerlinGermany
- Department of Pediatric Gastroenterology, Nephrology and Metabolic DiseasesCharité – Universitätsmedizin BerlinBerlinGermany
- Berlin Institute of Health (BIH) at Charité – Universitätsmedizin BerlinBerlinGermany
- DZHK (German Centre for Cardiovascular Research), Partner Site BerlinBerlinGermany
- Experimental and Clinical Research CenterA Cooperation of Charité – Universitätsmedizin Berlin and Max Delbrück Center for Molecular MedicineBerlinGermany
- Max Delbrück Center for Molecular Medicine in the Helmholtz AssociationBerlinGermany
| | - Johannes Holle
- Department of Pediatric Gastroenterology, Nephrology and Metabolic DiseasesCharité – Universitätsmedizin BerlinBerlinGermany
- DZHK (German Centre for Cardiovascular Research), Partner Site BerlinBerlinGermany
- Experimental and Clinical Research CenterA Cooperation of Charité – Universitätsmedizin Berlin and Max Delbrück Center for Molecular MedicineBerlinGermany
- Max Delbrück Center for Molecular Medicine in the Helmholtz AssociationBerlinGermany
| | - Chia‐Yu Chen
- DZHK (German Centre for Cardiovascular Research), Partner Site BerlinBerlinGermany
- Experimental and Clinical Research CenterA Cooperation of Charité – Universitätsmedizin Berlin and Max Delbrück Center for Molecular MedicineBerlinGermany
- Max Delbrück Center for Molecular Medicine in the Helmholtz AssociationBerlinGermany
| | - Laura F. Ginsbach
- Institute of PhysiologyCharité – Universitätsmedizin BerlinBerlinGermany
| | - Benjamin C. Krause
- Department of Chemical and Product SafetyGerman Federal Institute for Risk Assessment (BfR)BerlinGermany
| | - Ulrike Bruning
- Berlin Institute of Health (BIH) at Charité – Universitätsmedizin BerlinBerlinGermany
- Max Delbrück Center for Molecular Medicine in the Helmholtz AssociationBerlinGermany
| | - Fabian L. Kriegel
- Department of Chemical and Product SafetyGerman Federal Institute for Risk Assessment (BfR)BerlinGermany
- Carl Zeiss MeditechBerlinGermany
| | - Toralf Kaiser
- German Rheumatism Research Centre (DRFZ)BerlinGermany
| | - István A. Szijártó
- Experimental and Clinical Research CenterA Cooperation of Charité – Universitätsmedizin Berlin and Max Delbrück Center for Molecular MedicineBerlinGermany
- Department of Nephrology und Medical Intensive CareCharité – Universitätsmedizin BerlinBerlinGermany
| | - Harithaa Anandakumar
- DZHK (German Centre for Cardiovascular Research), Partner Site BerlinBerlinGermany
- Experimental and Clinical Research CenterA Cooperation of Charité – Universitätsmedizin Berlin and Max Delbrück Center for Molecular MedicineBerlinGermany
- Max Delbrück Center for Molecular Medicine in the Helmholtz AssociationBerlinGermany
- Department of Nephrology und Medical Intensive CareCharité – Universitätsmedizin BerlinBerlinGermany
| | | | | | - Pawel Durek
- German Rheumatism Research Centre (DRFZ)BerlinGermany
| | | | - Dörte Lodka
- Experimental and Clinical Research CenterA Cooperation of Charité – Universitätsmedizin Berlin and Max Delbrück Center for Molecular MedicineBerlinGermany
| | - Carina Hoffmann
- Experimental and Clinical Research CenterA Cooperation of Charité – Universitätsmedizin Berlin and Max Delbrück Center for Molecular MedicineBerlinGermany
| | - André A. Borchardt
- Institute of PhysiologyCharité – Universitätsmedizin BerlinBerlinGermany
| | - Lisa Peters
- Institute of PhysiologyCharité – Universitätsmedizin BerlinBerlinGermany
- DZHK (German Centre for Cardiovascular Research), Partner Site BerlinBerlinGermany
- Institute of BiologyFreie Universität BerlinBerlinGermany
| | - Laura Michalick
- Institute of PhysiologyCharité – Universitätsmedizin BerlinBerlinGermany
- Berlin Institute of Health (BIH) at Charité – Universitätsmedizin BerlinBerlinGermany
- DZHK (German Centre for Cardiovascular Research), Partner Site BerlinBerlinGermany
| | - Uwe Querfeld
- Department of Pediatric Gastroenterology, Nephrology and Metabolic DiseasesCharité – Universitätsmedizin BerlinBerlinGermany
| | - Philip Bufler
- Department of Pediatric Gastroenterology, Nephrology and Metabolic DiseasesCharité – Universitätsmedizin BerlinBerlinGermany
| | - Andreas Luch
- Department of Chemical and Product SafetyGerman Federal Institute for Risk Assessment (BfR)BerlinGermany
| | | | - Jennifer A. Kirwan
- Berlin Institute of Health (BIH) at Charité – Universitätsmedizin BerlinBerlinGermany
- Max Delbrück Center for Molecular Medicine in the Helmholtz AssociationBerlinGermany
| | - Sofia K. Forslund
- DZHK (German Centre for Cardiovascular Research), Partner Site BerlinBerlinGermany
- Experimental and Clinical Research CenterA Cooperation of Charité – Universitätsmedizin Berlin and Max Delbrück Center for Molecular MedicineBerlinGermany
- Max Delbrück Center for Molecular Medicine in the Helmholtz AssociationBerlinGermany
| | - Julia Thumfahrt
- Department of Pediatric Gastroenterology, Nephrology and Metabolic DiseasesCharité – Universitätsmedizin BerlinBerlinGermany
| | - Dominik Müller
- Department of Pediatric Gastroenterology, Nephrology and Metabolic DiseasesCharité – Universitätsmedizin BerlinBerlinGermany
| | - Nicola Wilck
- DZHK (German Centre for Cardiovascular Research), Partner Site BerlinBerlinGermany
- Experimental and Clinical Research CenterA Cooperation of Charité – Universitätsmedizin Berlin and Max Delbrück Center for Molecular MedicineBerlinGermany
- Max Delbrück Center for Molecular Medicine in the Helmholtz AssociationBerlinGermany
- Department of Nephrology und Medical Intensive CareCharité – Universitätsmedizin BerlinBerlinGermany
| | - Mir‐Farzin Mashreghi
- Berlin Institute of Health (BIH) at Charité – Universitätsmedizin BerlinBerlinGermany
- German Rheumatism Research Centre (DRFZ)BerlinGermany
| | - Ulrike Löber
- DZHK (German Centre for Cardiovascular Research), Partner Site BerlinBerlinGermany
- Experimental and Clinical Research CenterA Cooperation of Charité – Universitätsmedizin Berlin and Max Delbrück Center for Molecular MedicineBerlinGermany
- Max Delbrück Center for Molecular Medicine in the Helmholtz AssociationBerlinGermany
| | - Hendrik Bartolomaeus
- DZHK (German Centre for Cardiovascular Research), Partner Site BerlinBerlinGermany
- Experimental and Clinical Research CenterA Cooperation of Charité – Universitätsmedizin Berlin and Max Delbrück Center for Molecular MedicineBerlinGermany
- Max Delbrück Center for Molecular Medicine in the Helmholtz AssociationBerlinGermany
- Department of Nephrology und Medical Intensive CareCharité – Universitätsmedizin BerlinBerlinGermany
| | - Wolfgang M. Kuebler
- Institute of PhysiologyCharité – Universitätsmedizin BerlinBerlinGermany
- DZHK (German Centre for Cardiovascular Research), Partner Site BerlinBerlinGermany
- Departments of Physiology and SurgeryUniversity of TorontoTorontoONCanada
- Keenan Research Centre for Biomedical ScienceSt. Michael's HospitalTorontoONCanada
| | - Szandor Simmons
- Institute of PhysiologyCharité – Universitätsmedizin BerlinBerlinGermany
- DZHK (German Centre for Cardiovascular Research), Partner Site BerlinBerlinGermany
| |
Collapse
|
3
|
Soomro QH, Charytan DM. New Insights on Cardiac Arrhythmias in Patients With Kidney Disease. Semin Nephrol 2024; 44:151518. [PMID: 38772780 DOI: 10.1016/j.semnephrol.2024.151518] [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] [Indexed: 05/23/2024]
Abstract
The risk of arrhythmia and its management become increasingly complex as kidney disease progresses. This presents a multifaceted clinical challenge. Our discussion addresses these specific challenges relevant to patients as their kidney disease advances. We highlight numerous opportunities for enhancing the current standard of care within this realm. Additionally, this review delves into research concerning early detection, prevention, diagnosis, and treatment of various arrhythmias spanning the spectrum of kidney disease.
Collapse
|
4
|
Palzkill VR, Tan J, Yang Q, Morcos J, Laitano O, Ryan TE. Activation of the Aryl Hydrocarbon Receptor in Endothelial Cells Impairs Ischemic Angiogenesis in Chronic Kidney Disease. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.07.24.550410. [PMID: 37546909 PMCID: PMC10401998 DOI: 10.1101/2023.07.24.550410] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/08/2023]
Abstract
Rationale Chronic kidney disease (CKD) is a strong risk factor for peripheral artery disease (PAD) that is associated with worsened clinical outcomes. CKD leads to accumulation of tryptophan metabolites that associate with adverse limb events in PAD and are ligands of the aryl hydrocarbon receptor (AHR) which may regulate ischemic angiogenesis. Objectives To test if endothelial cell-specific deletion of the AHR (AHRecKO) alters ischemic angiogenesis and limb function in mice with CKD subjected to femoral artery ligation. Findings Male AHRecKO mice with CKD displayed better limb perfusion recovery and enhanced ischemic angiogenesis compared to wildtype mice with CKD. However, the improved limb perfusion did not result in better muscle performance. In contrast to male mice, deletion of the AHR in female mice with CKD had no impact on perfusion recovery or angiogenesis. Using primary endothelial cells from male and female mice, treatment with indoxyl sulfate uncovered sex-dependent differences in AHR activating potential and RNA sequencing revealed wide ranging sex-differences in angiogenic signaling pathways. Conclusion Endothelium-specific deletion of the AHR improved ischemic angiogenesis in male, but not female, mice with CKD. There are sex-dependent differences in Ahr activating potential within endothelial cells that are independent of sex hormones.
Collapse
Affiliation(s)
- Victoria R. Palzkill
- Department of Applied Physiology and Kinesiology, The University of Florida, Gainesville, FL, USA
| | - Jianna Tan
- Department of Applied Physiology and Kinesiology, The University of Florida, Gainesville, FL, USA
| | - Qingping Yang
- Department of Applied Physiology and Kinesiology, The University of Florida, Gainesville, FL, USA
| | - Juliana Morcos
- Department of Applied Physiology and Kinesiology, The University of Florida, Gainesville, FL, USA
| | - Orlando Laitano
- Department of Applied Physiology and Kinesiology, The University of Florida, Gainesville, FL, USA
- Center for Exercise Science, The University of Florida, Gainesville, FL, USA
- The Myology Institute, The University of Florida, Gainesville, FL, USA
| | - Terence E. Ryan
- Department of Applied Physiology and Kinesiology, The University of Florida, Gainesville, FL, USA
- Center for Exercise Science, The University of Florida, Gainesville, FL, USA
- The Myology Institute, The University of Florida, Gainesville, FL, USA
| |
Collapse
|
5
|
Balestrieri N, Palzkill V, Pass C, Tan J, Salyers ZR, Moparthy C, Murillo A, Kim K, Thome T, Yang Q, O’Malley KA, Berceli SA, Yue F, Scali ST, Ferreira LF, Ryan TE. Activation of the Aryl Hydrocarbon Receptor in Muscle Exacerbates Ischemic Pathology in Chronic Kidney Disease. Circ Res 2023; 133:158-176. [PMID: 37325935 PMCID: PMC10330629 DOI: 10.1161/circresaha.123.322875] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Accepted: 06/07/2023] [Indexed: 06/17/2023]
Abstract
BACKGROUND Chronic kidney disease (CKD) accelerates the development of atherosclerosis, decreases muscle function, and increases the risk of amputation or death in patients with peripheral artery disease (PAD). However, the mechanisms underlying this pathobiology are ill-defined. Recent work has indicated that tryptophan-derived uremic solutes, which are ligands for AHR (aryl hydrocarbon receptor), are associated with limb amputation in PAD. Herein, we examined the role of AHR activation in the myopathy of PAD and CKD. METHODS AHR-related gene expression was evaluated in skeletal muscle obtained from mice and human PAD patients with and without CKD. AHRmKO (skeletal muscle-specific AHR knockout) mice with and without CKD were subjected to femoral artery ligation, and a battery of assessments were performed to evaluate vascular, muscle, and mitochondrial health. Single-nuclei RNA sequencing was performed to explore intercellular communication. Expression of the constitutively active AHR was used to isolate the role of AHR in mice without CKD. RESULTS PAD patients and mice with CKD displayed significantly higher mRNA expression of classical AHR-dependent genes (Cyp1a1, Cyp1b1, and Aldh3a1) when compared with either muscle from the PAD condition with normal renal function (P<0.05 for all 3 genes) or nonischemic controls. AHRmKO significantly improved limb perfusion recovery and arteriogenesis, preserved vasculogenic paracrine signaling from myofibers, increased muscle mass and strength, as well as enhanced mitochondrial function in an experimental model of PAD/CKD. Moreover, viral-mediated skeletal muscle-specific expression of a constitutively active AHR in mice with normal kidney function exacerbated the ischemic myopathy evidenced by smaller muscle masses, reduced contractile function, histopathology, altered vasculogenic signaling, and lower mitochondrial respiratory function. CONCLUSIONS These findings establish AHR activation in muscle as a pivotal regulator of the ischemic limb pathology in CKD. Further, the totality of the results provides support for testing of clinical interventions that diminish AHR signaling in these conditions.
Collapse
Affiliation(s)
- Nicholas Balestrieri
- Department of Applied Physiology and Kinesiology, The University of Florida, Gainesville, FL, USA
| | - Victoria Palzkill
- Department of Applied Physiology and Kinesiology, The University of Florida, Gainesville, FL, USA
| | - Caroline Pass
- Department of Applied Physiology and Kinesiology, The University of Florida, Gainesville, FL, USA
| | - Jianna Tan
- Department of Applied Physiology and Kinesiology, The University of Florida, Gainesville, FL, USA
| | - Zachary R. Salyers
- Department of Applied Physiology and Kinesiology, The University of Florida, Gainesville, FL, USA
| | - Chatick Moparthy
- Department of Applied Physiology and Kinesiology, The University of Florida, Gainesville, FL, USA
| | - Ania Murillo
- Department of Applied Physiology and Kinesiology, The University of Florida, Gainesville, FL, USA
| | - Kyoungrae Kim
- Department of Applied Physiology and Kinesiology, The University of Florida, Gainesville, FL, USA
| | - Trace Thome
- Department of Applied Physiology and Kinesiology, The University of Florida, Gainesville, FL, USA
| | - Qingping Yang
- Department of Applied Physiology and Kinesiology, The University of Florida, Gainesville, FL, USA
| | - Kerri A. O’Malley
- Department of Surgery, The University of Florida, Gainesville, FL, USA
| | - Scott A. Berceli
- Department of Surgery, The University of Florida, Gainesville, FL, USA
| | - Feng Yue
- Department of Animal Sciences, The University of Florida, Gainesville, FL, USA
- Myology Institute, The University of Florida, Gainesville, FL, USA
| | | | - Leonardo F. Ferreira
- Department of Applied Physiology and Kinesiology, The University of Florida, Gainesville, FL, USA
- Center for Exercise Science, The University of Florida, Gainesville, FL, USA
- Myology Institute, The University of Florida, Gainesville, FL, USA
| | - Terence E. Ryan
- Department of Applied Physiology and Kinesiology, The University of Florida, Gainesville, FL, USA
- Center for Exercise Science, The University of Florida, Gainesville, FL, USA
- Myology Institute, The University of Florida, Gainesville, FL, USA
| |
Collapse
|
6
|
Balestrieri N, Palzkill V, Pass C, Tan J, Salyers ZR, Moparthy C, Murillo A, Kim K, Thome T, Yang Q, O'Malley KA, Berceli SA, Yue F, Scali ST, Ferreira LF, Ryan TE. Chronic activation of the aryl hydrocarbon receptor in muscle exacerbates ischemic pathology in chronic kidney disease. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.05.16.541060. [PMID: 37292677 PMCID: PMC10245783 DOI: 10.1101/2023.05.16.541060] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Chronic kidney disease (CKD) accelerates the development of atherosclerosis, decreases muscle function, and increases the risk of amputation or death in patients with peripheral artery disease (PAD). However, the cellular and physiological mechanisms underlying this pathobiology are ill-defined. Recent work has indicated that tryptophan-derived uremic toxins, many of which are ligands for the aryl hydrocarbon receptor (AHR), are associated with adverse limb outcomes in PAD. We hypothesized that chronic AHR activation, driven by the accumulation of tryptophan-derived uremic metabolites, may mediate the myopathic condition in the presence of CKD and PAD. Both PAD patients with CKD and mice with CKD subjected to femoral artery ligation (FAL) displayed significantly higher mRNA expression of classical AHR-dependent genes ( Cyp1a1 , Cyp1b1 , and Aldh3a1 ) when compared to either muscle from the PAD condition with normal renal function ( P <0.05 for all three genes) or non-ischemic controls. Skeletal-muscle-specific AHR deletion in mice (AHR mKO ) significantly improved limb muscle perfusion recovery and arteriogenesis, preserved vasculogenic paracrine signaling from myofibers, increased muscle mass and contractile function, as well as enhanced mitochondrial oxidative phosphorylation and respiratory capacity in an experimental model of PAD/CKD. Moreover, viral-mediated skeletal muscle-specific expression of a constitutively active AHR in mice with normal kidney function exacerbated the ischemic myopathy evidenced by smaller muscle masses, reduced contractile function, histopathology, altered vasculogenic signaling, and lower mitochondrial respiratory function. These findings establish chronic AHR activation in muscle as a pivotal regulator of the ischemic limb pathology in PAD. Further, the totality of the results provide support for testing of clinical interventions that diminish AHR signaling in these conditions.
Collapse
|
7
|
Park S, Lee SH, Shin D, Hong D, Joh HS, Choi KH, Kim HK, Ha SJ, Park TK, Yang JH, Song YB, Hahn JY, Choi SH, Gwon HC, Lee JM. Prognostic Impact of Coronary Flow Reserve in Patients With CKD. Kidney Int Rep 2022; 8:64-74. [PMID: 36644355 PMCID: PMC9832048 DOI: 10.1016/j.ekir.2022.10.003] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Revised: 08/24/2022] [Accepted: 10/03/2022] [Indexed: 11/27/2022] Open
Abstract
Introduction Both coronary flow reserve (CFR) and chronic kidney disease (CKD) are known to be associated with adverse cardiac events. However, it is unclear how these prognostic factors are interrelated. This study evaluated the association between intracoronary physiologic indexes and CKD and their prognostic implications. Methods A total of 351 patients without left ventricular systolic dysfunction (ejection fraction ≥ 40%) and not on dialysis whose revascularization was deferred based on fractional flow reserve (FFR) > 0.80 were analyzed. Depressed CFR was defined as CFR ≤ 2.0. The primary outcome was a composite of cardiac death or hospitalization for heart failure at 3 years. Results Patients with CKD showed lower CFR than the non-CKD population (3.28 ± 1.77 vs. 2.60 ± 1.09, P < 0.001), mainly driven by increased resting coronary flow. There was no significant difference in hyperemic coronary flow, FFR, and index of microvascular resistance between the 2 groups. CFR was significantly associated with estimated glomerular filtration rate (eGFR) (P = 0.045), and the proportion of depressed CFR was significantly increased with higher CKD stages (P = 0.011). The risk of cardiac death or hospitalization for heart failure was the lowest in the non-CKD and preserved CFR group (11.9%) and the highest in the CKD and depressed CFR group (60.0%, overall log rank P < 0.001). Both CKD (adjusted hazard ratio [HRadj] 2.614, 95% confidence interval [CI] 1.505-4.539, P < 0.001) and depressed CFR (HRadj 3.237, 95% CI 2.015-5.199, P < 0.001) were independently associated with the risk of the primary outcome. Conclusion There was a significant association between severity of CKD and CFR. Both CKD and depressed CFR showed independent association with higher risk of cardiac death or hospitalization for heart failure.
Collapse
Affiliation(s)
- Sugeon Park
- Division of Cardiology, Department of Internal Medicine, Heart Vascular Stroke Institute, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Seung Hun Lee
- Department of Internal Medicine and Cardiovascular Center, Chonnam National University Hospital, Chonnam National University Medical School, Gwangju, Republic of Korea
| | - Doosup Shin
- Division of Cardiology, Department of Internal Medicine, Duke University Medical Center, Durham, North Carolina, USA
| | - David Hong
- Division of Cardiology, Department of Internal Medicine, Heart Vascular Stroke Institute, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Hyun Sung Joh
- Department of Internal Medicine and Cardiovascular Center, Seoul National University Boramae Medical Center, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Ki Hong Choi
- Division of Cardiology, Department of Internal Medicine, Heart Vascular Stroke Institute, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Hyun Kuk Kim
- Department of Internal Medicine and Cardiovascular Center, Chosun University Hospital, University of Chosun College of Medicine, Gwangju, Republic of Korea
| | - Sang Jin Ha
- Division of Cardiology, Department of Internal Medicine, Gangneung Asan Hospital, University of Ulsan College of Medicine, Gangneung, Republic of Korea
| | - Taek Kyu Park
- Division of Cardiology, Department of Internal Medicine, Heart Vascular Stroke Institute, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Jeong Hoon Yang
- Division of Cardiology, Department of Internal Medicine, Heart Vascular Stroke Institute, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Young Bin Song
- Division of Cardiology, Department of Internal Medicine, Heart Vascular Stroke Institute, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Joo-Yong Hahn
- Division of Cardiology, Department of Internal Medicine, Heart Vascular Stroke Institute, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Seung-Hyuk Choi
- Division of Cardiology, Department of Internal Medicine, Heart Vascular Stroke Institute, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Hyeon-Cheol Gwon
- Division of Cardiology, Department of Internal Medicine, Heart Vascular Stroke Institute, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Joo Myung Lee
- Division of Cardiology, Department of Internal Medicine, Heart Vascular Stroke Institute, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea,Correspondence: Joo Myung Lee, Division of Cardiology, Department of Internal Medicine, Heart Vascular Stroke Institute, Samsung Medical Center, Sungkyunkwan University School of Medicine, 81 Irwon-ro, Gangnam-gu, Seoul 06351, Republic of Korea.
| |
Collapse
|
8
|
Adam RJ, Williams AC, Kriegel AJ. Comparison of the Surgical Resection and Infarct 5/6 Nephrectomy Rat Models of Chronic Kidney Disease. Am J Physiol Renal Physiol 2022; 322:F639-F654. [PMID: 35379002 DOI: 10.1152/ajprenal.00398.2021] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
The 5/6 nephrectomy rat remnant kidney model is commonly employed to study chronic kidney disease (CKD). This model requires removal of one whole kidney and two-thirds of the other. The two most common ways of producing the remnant kidney are surgical resection of poles, known as the polectomy (Pol) model, or ligation of upper and lower renal arterial branches, resulting in pole infarction (Inf). These models have much in common, but also major phenotypic differences, and thus respectively model unique aspects of human CKD. The purpose of this review is to summarize phenotypic similarities and differences between these two models and their relation to human CKD, while emphasizing their vascular phenotype. In this article we review studies that have evaluated arterial blood pressure, the renin-angiotensin-aldosterone-system (RAAS), autoregulation, nitric oxide, single nephron physiology, angiogenic and anti-angiogenic factors, and capillary rarefaction in these two models. Phenotypic similarities: both models spontaneously develop hallmarks of human CKD including uremia, fibrosis, capillary rarefaction, and progressive renal function decline. They both undergo whole-organ hypertrophy, hyperfiltration of functional nephrons, reduced renal expression of angiogenic factor VEGF, increased renal expression of the anti-angiogenic thrombospondin-1, impaired renal autoregulation, and abnormal vascular nitric oxide physiology. Key phenotypic differences: the Inf model develops rapid-onset, moderate-to-severe systemic hypertension, and the Pol model early normotension followed by mild-to-moderate hypertension. The Inf rat has a markedly more active renin-angiotensin-aldosterone-system. Comparison of these two models facilitates understanding of how they can be utilized for studying CKD pathophysiology (e.g., RAAS dependent or independent pathology).
Collapse
Affiliation(s)
- Ryan J Adam
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI, United States.,Cardiovascular Center, Medical College of Wisconsin, Milwaukee, WI, United States
| | - Adaysha C Williams
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI, United States
| | - Alison J Kriegel
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI, United States.,Department of Pediatrics, Medical College of Wisconsin, Milwaukee, WI, United States.,Cardiovascular Center, Medical College of Wisconsin, Milwaukee, WI, United States.,Center of Systems Molecular Medicine, Medical College of Wisconsin, Milwaukee, WI, United States
| |
Collapse
|
9
|
Zhao E, Barber J, Sen CK, Arciero J. Modeling acute and chronic vascular responses to a major arterial occlusion. Microcirculation 2021; 29:e12738. [PMID: 34779082 PMCID: PMC9128362 DOI: 10.1111/micc.12738] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Revised: 10/21/2021] [Accepted: 11/09/2021] [Indexed: 12/25/2022]
Abstract
OBJECTIVE To incorporate chronic vascular adaptations into a mathematical model of the rat hindlimb to simulate flow restoration following total occlusion of the femoral artery. METHODS A vascular wall mechanics model is used to simulate acute and chronic vascular adaptations in the collateral arteries and collateral-dependent arterioles of the rat hindlimb. On an acute timeframe, the vascular tone of collateral arteries and distal arterioles is determined by responses to pressure, shear stress, and metabolic demand. On a chronic timeframe, sustained dilation of arteries and arterioles induces outward vessel remodeling represented by increased passive vessel diameter (arteriogenesis), and low venous oxygen saturation levels induce the growth of new capillaries represented by increased capillary number (angiogenesis). RESULTS The model predicts that flow compensation to an occlusion is enhanced primarily by arteriogenesis of the collateral arteries on a chronic time frame. Blood flow autoregulation is predicted to be disrupted and to occur for higher pressure values following femoral arterial occlusion. CONCLUSIONS Structural adaptation of the vasculature allows for increased blood flow to the collateral-dependent region after occlusion. Although flow is still below pre-occlusion levels, model predictions indicate that interventions which enhance collateral arteriogenesis would have the greatest potential for restoring flow.
Collapse
Affiliation(s)
- Erin Zhao
- Department of Mathematical Sciences, Indiana University – Purdue University Indianapolis, Indianapolis, IN
| | - Jared Barber
- Department of Mathematical Sciences, Indiana University – Purdue University Indianapolis, Indianapolis, IN
| | - Chandan K. Sen
- Indiana Center for Regenerative Medicine and Engineering, Department of Surgery, Indiana University School of Medicine, Indianapolis, IN
| | - Julia Arciero
- Department of Mathematical Sciences, Indiana University – Purdue University Indianapolis, Indianapolis, IN
| |
Collapse
|
10
|
Dupont V, Al-Rifai R, Poitevin G, Ortillon J, Jayyosi L, Terryn C, Francois C, Rieu P, Fritz G, Boulagnon-Rombi C, Fichel C, Schmidt AM, Tournois C, Nguyen P, Touré F. AgeR deletion decreases soluble fms-like tyrosine kinase 1 production and improves post-ischemic angiogenesis in uremic mice. Angiogenesis 2021; 24:47-55. [PMID: 32989644 DOI: 10.1007/s10456-020-09747-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2020] [Accepted: 09/17/2020] [Indexed: 10/23/2022]
Abstract
Peripheral arterial disease occurs more frequently and has a worse prognosis in patients with chronic kidney disease (CKD). The receptor for advanced glycation end products (RAGE) is involved in multiple aspects of uremia-associated vasculopathy. Previous data suggest that the RAGE pathway may promote soluble fms-like tyrosine kinase 1 (sFlt1) production, an anti-angiogenic molecule. Thus, we tested the hypothesis that the deletion of AgeR would decrease sFlt1 production and improve post-ischemic revascularization in uremic condition. We used a well-established CKD model (5/6 nephrectomy) in WT and AgeR-/- C57/Bl6 mice. Hindlimb ischemia was induced by femoral artery ligation. Revascularization was evaluated by complementary approaches: ischemic limb retraction, LASCA imagery, and capillary density. The production of sFlt1 was assessed at both RNA and protein levels. After hindlimb ischemia, uremic mice showed slower functional recovery (p < 0.01), decreased reperfusion (p < 0.01), lower capillary density (p = 0.02), and increased circulating sFlt1 levels (p = 0.03). AgeR deletion restored post-ischemic angiogenesis and was protective from sFlt1 increase in uremic mice. These findings show the main role of RAGE in post-ischemic angiogenesis impairment associated with CKD. RAGE may represent a key target for building new therapeutic approaches to improve the outcome of CKD patients with PAD.
Collapse
Affiliation(s)
- Vincent Dupont
- Department of Nephrology, University Hospital of Reims, Reims, France.
- CNRS UMR 7369, Université de Reims Champagne-Ardenne, Reims, France.
- EA-3801, Université de Reims Champagne-Ardenne, Reims, France.
| | - Rida Al-Rifai
- EA-3801, Université de Reims Champagne-Ardenne, Reims, France
| | - Gael Poitevin
- EA-3801, Université de Reims Champagne-Ardenne, Reims, France
| | - Jeremy Ortillon
- CNRS UMR 7369, Université de Reims Champagne-Ardenne, Reims, France
| | - Laura Jayyosi
- EA-3801, Université de Reims Champagne-Ardenne, Reims, France
| | - Christine Terryn
- Plateforme PICT, Université de Reims Champagne-Ardenne, Reims, France
| | | | - Philippe Rieu
- Department of Nephrology, University Hospital of Reims, Reims, France
| | - Günter Fritz
- Institute of Neuropathology, University of Freiburg, Freiburg, Germany
| | - Camile Boulagnon-Rombi
- Laboratoire D'Anatomie Pathologique, Université de Reims Champagne-Ardenne, Reims, France
| | - Caroline Fichel
- Laboratoire D'Anatomie Pathologique, Université de Reims Champagne-Ardenne, Reims, France
| | | | - Claire Tournois
- EA-3801, Université de Reims Champagne-Ardenne, Reims, France
| | - Philippe Nguyen
- EA-3801, Université de Reims Champagne-Ardenne, Reims, France
| | - Fatouma Touré
- Department of Nephrology, University Hospital of Limoges, Limoges, France
- CNRS UMR 7276, INSERM U1262, Université de Limoges, Limoges, France
| |
Collapse
|
11
|
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
|
12
|
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: 64] [Impact Index Per Article: 12.8] [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
|
13
|
Hydralazine improves ischemia-induced neovasculogenesis via xanthine-oxidase inhibition in chronic renal insufficiency. Pharmacol Res 2019; 151:104509. [PMID: 31678640 DOI: 10.1016/j.phrs.2019.104509] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/06/2019] [Revised: 09/13/2019] [Accepted: 10/21/2019] [Indexed: 01/10/2023]
Abstract
Oxidative stress is related to the progression of renal diseases and modulation of oxidative stress can lead to a reduction in vascular events in patients with chronic renal insufficiency (CRI). Indoxyl sulfate (IS) and xanthine oxidase (XO) are related to impaired neovasculogenesis in CRI. Hydralazine is suggested for blood pressure control in CRI. This study aimed to investigate whether hydralazine could improve ischemia-induced neovasculogenesis in CRI animals by reducing reactive oxygen species (ROS) levels. Mice underwent subtotal nephrectomy or sham surgery. Nitrendipine, probenecid, and allopurinol were used to reduce blood pressure, uric acid (UA), and XO activity levels, respectively, for comparison. Blood pressure, XO activity and UA levels that were increased after subtotal nephrectomy were reduced by hydralazine treatment. Allopurinol decreased blood XO activity and UA levels. Only hydralazine and allopurinol increased the number of circulating endothelial progenitor cells (EPCs) and improved neovasculogenesis in CRI mice. IS activated XO mRNA and ROS and inhibited the functions of EPCs and endothelial cells, which could be reversed by hydralazine. However, no additional beneficial effects were observed when XO was inhibited with both hydralazine and siRNA. In conclusion, hydralazine, as a potential XO inhibitor, not only reduced blood pressure and UA levels but also increased the number of circulating EPCs and improved neovasculogenesis in CRI animals. Hydralazine directly inhibited IS-induced ROS and XO activation in EPCs and endothelial cells, and restored their functions in vitro. Future studies should evaluate whether hydralazine could provide additional vascular protection in patients with CRI.
Collapse
|
14
|
Berru FN, Gray SE, Thome T, Kumar RA, Salyers ZR, Coleman M, Dennis Le, O'Malley K, Ferreira LF, Berceli SA, Scali ST, Ryan TE. Chronic kidney disease exacerbates ischemic limb myopathy in mice via altered mitochondrial energetics. Sci Rep 2019; 9:15547. [PMID: 31664123 PMCID: PMC6820860 DOI: 10.1038/s41598-019-52107-7] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2019] [Accepted: 10/12/2019] [Indexed: 02/07/2023] Open
Abstract
Chronic kidney disease (CKD) substantially increases the severity of peripheral arterial disease (PAD) symptomology, however, the biological mechanisms remain unclear. The objective herein was to determine the impact of CKD on PAD pathology in mice. C57BL6/J mice were subjected to a diet-induced model of CKD by delivery of adenine for six weeks. CKD was confirmed by measurements of glomerular filtration rate, blood urea nitrogen, and kidney histopathology. Mice with CKD displayed lower muscle force production and greater ischemic lesions in the tibialis anterior muscle (78.1 ± 14.5% vs. 2.5 ± 0.5% in control mice, P < 0.0001, N = 5-10/group) and decreased myofiber size (1661 ± 134 μm2 vs. 2221 ± 100 μm2 in control mice, P < 0.01, N = 5-10/group). This skeletal myopathy occurred despite normal capillary density (516 ± 59 vs. 466 ± 45 capillaries/20x field of view) and limb perfusion. CKD mice displayed a ~50-65% reduction in muscle mitochondrial respiratory capacity in ischemic muscle, whereas control mice had normal mitochondrial function. Hydrogen peroxide emission was modestly higher in the ischemic muscle of CKD mice, which coincided with decreased oxidant buffering. Exposure of cultured myotubes to CKD serum resulted in myotube atrophy and elevated oxidative stress, which were attenuated by mitochondrial-targeted therapies. Taken together, these findings suggest that mitochondrial impairments caused by CKD contribute to the exacerbation of ischemic pathology.
Collapse
Affiliation(s)
- Fabian N Berru
- Department of Applied Physiology and Kinesiology, University of Florida, Gainesville, FL, USA
| | - Sarah E Gray
- Division of Vascular Surgery and Endovascular Therapy, University of Florida, Gainesville, FL, USA
- Malcolm Randall Veteran Affairs Medical Center, Gainesville, FL, USA
| | - Trace Thome
- Department of Applied Physiology and Kinesiology, University of Florida, Gainesville, FL, USA
| | - Ravi A Kumar
- Department of Applied Physiology and Kinesiology, University of Florida, Gainesville, FL, USA
| | - Zachary R Salyers
- Department of Applied Physiology and Kinesiology, University of Florida, Gainesville, FL, USA
| | - Madeline Coleman
- Department of Applied Physiology and Kinesiology, University of Florida, Gainesville, FL, USA
| | - Dennis Le
- Department of Applied Physiology and Kinesiology, University of Florida, Gainesville, FL, USA
| | - Kerri O'Malley
- Division of Vascular Surgery and Endovascular Therapy, University of Florida, Gainesville, FL, USA
- Malcolm Randall Veteran Affairs Medical Center, Gainesville, FL, USA
| | - Leonardo F Ferreira
- Department of Applied Physiology and Kinesiology, University of Florida, Gainesville, FL, USA
- Center for Exercise Science, University of Florida, Gainesville, FL, USA
| | - Scott A Berceli
- Division of Vascular Surgery and Endovascular Therapy, University of Florida, Gainesville, FL, USA
- Malcolm Randall Veteran Affairs Medical Center, Gainesville, FL, USA
| | - Salvatore T Scali
- Division of Vascular Surgery and Endovascular Therapy, University of Florida, Gainesville, FL, USA
- Malcolm Randall Veteran Affairs Medical Center, Gainesville, FL, USA
| | - Terence E Ryan
- Department of Applied Physiology and Kinesiology, University of Florida, Gainesville, FL, USA.
- Center for Exercise Science, University of Florida, Gainesville, FL, USA.
| |
Collapse
|
15
|
Charytan DM, Skali H, Shah NR, Veeranna V, Cheezum MK, Taqueti VR, Kato T, Bibbo CR, Hainer J, Dorbala S, Blankstein R, Di Carli MF. Coronary flow reserve is predictive of the risk of cardiovascular death regardless of chronic kidney disease stage. Kidney Int 2017; 93:501-509. [PMID: 29032954 DOI: 10.1016/j.kint.2017.07.025] [Citation(s) in RCA: 60] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2016] [Revised: 07/14/2017] [Accepted: 07/27/2017] [Indexed: 01/09/2023]
Abstract
Microvascular rarefaction is found in experimental uremia, but data from patients with chronic kidney disease (CKD) are limited. We therefore quantified absolute myocardial blood flow and coronary flow reserve (the ratio of peak to resting flow) from myocardial perfusion positron emission tomography scans at a single institution. Individuals were classified into standard CKD categories based on the estimated glomerular filtration rate. Associations of coronary flow reserve with CKD stage and cardiovascular mortality were analyzed in models adjusted for cardiovascular risk factors. The coronary flow reserve was significantly associated with CKD stage, declining in early CKD, but it did not differ significantly among individuals with stage 4, 5, and dialysis-dependent CKD. Flow reserve with preserved kidney function was 2.01, 2.06 in stage 1 CKD, 1.91 in stage 2, 1.68 in stage 3, 1.54 in stage 4, 1.66 in stage 5, and 1.55 in dialysis-dependent CKD. Coronary flow reserve was significantly associated with cardiovascular mortality in adjusted models (hazard ratio 0.76, 95% confidence interval: 0.63-0.92 per tertile of coronary flow reserve) without evidence of effect modification by CKD. Thus, coronary flow reserve is strongly associated with cardiovascular risk regardless of CKD severity and is low in early stage CKD without further decrement in stage 5 or dialysis-dependent CKD. This suggests that CKD physiology rather than the effects of dialysis is the primary driver of microvascular disease. Our findings highlight the potential contribution of microvascular dysfunction to cardiovascular risk in CKD and the need to define mechanisms linking low coronary flow reserve to mortality.
Collapse
Affiliation(s)
- David M Charytan
- Department of Medicine, Brigham & Women's Hospital, Boston, Massachusetts, USA; Renal Division, Brigham & Women's Hospital, Boston, Massachusetts, USA.
| | - Hicham Skali
- Department of Medicine, Brigham & Women's Hospital, Boston, Massachusetts, USA; Cardiovascular Medicine, Brigham & Women's Hospital, Boston, Massachusetts, USA
| | - Nishant R Shah
- Department of Medicine, Brigham & Women's Hospital, Boston, Massachusetts, USA; Cardiovascular Medicine, Brigham & Women's Hospital, Boston, Massachusetts, USA
| | - Vikas Veeranna
- Department of Medicine, Brigham & Women's Hospital, Boston, Massachusetts, USA; Cardiovascular Medicine, Brigham & Women's Hospital, Boston, Massachusetts, USA
| | - Michael K Cheezum
- Department of Medicine, Brigham & Women's Hospital, Boston, Massachusetts, USA; Cardiovascular Medicine, Brigham & Women's Hospital, Boston, Massachusetts, USA
| | - Viviany R Taqueti
- Department of Medicine, Brigham & Women's Hospital, Boston, Massachusetts, USA; Cardiovascular Medicine, Brigham & Women's Hospital, Boston, Massachusetts, USA
| | - Takashi Kato
- Cardiovascular Medicine, Brigham & Women's Hospital, Boston, Massachusetts, USA; Gifu Prefectural General Medical Center, Gifu City, Japan
| | - Courtney R Bibbo
- Department of Medicine, Brigham & Women's Hospital, Boston, Massachusetts, USA; Cardiovascular Medicine, Brigham & Women's Hospital, Boston, Massachusetts, USA
| | - Jon Hainer
- Department of Medicine, Brigham & Women's Hospital, Boston, Massachusetts, USA; Cardiovascular Medicine, Brigham & Women's Hospital, Boston, Massachusetts, USA
| | - Sharmila Dorbala
- Department of Medicine, Brigham & Women's Hospital, Boston, Massachusetts, USA; Cardiovascular Medicine, Brigham & Women's Hospital, Boston, Massachusetts, USA
| | - Ron Blankstein
- Department of Medicine, Brigham & Women's Hospital, Boston, Massachusetts, USA; Cardiovascular Medicine, Brigham & Women's Hospital, Boston, Massachusetts, USA
| | - Marcelo F Di Carli
- Department of Medicine, Brigham & Women's Hospital, Boston, Massachusetts, USA; Cardiovascular Medicine, Brigham & Women's Hospital, Boston, Massachusetts, USA
| |
Collapse
|
16
|
Bone marrow-derived cells and their conditioned medium induce microvascular repair in uremic rats by stimulation of endogenous repair mechanisms. Sci Rep 2017; 7:9444. [PMID: 28842629 PMCID: PMC5572734 DOI: 10.1038/s41598-017-09883-x] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2017] [Accepted: 08/01/2017] [Indexed: 01/11/2023] Open
Abstract
The reduced number of circulating stem/progenitor cells that is found in chronic kidney disease (CKD) patients may contribute to impaired angiogenic repair and decreased capillary density in the heart. Cell therapy with bone marrow-derived cells (BMDCs) has been shown to induce positive effects on the microvasculature and cardiac function, most likely due to secretion of growth factors and cytokines, all of which are present in the conditioned medium (CM); however, this is controversial. Here we showed that treatment with BMDC or CM restored vascular density and decreased the extent of fibrosis in a rat model of CKD, the 5/6 nephrectomy. Engraftment and differentiation of exogenous BMDCs could not be detected. Yet CM led to the mobilization and infiltration of endogenous circulating cells into the heart. Cell recruitment was facilitated by the local expression of pro-inflammatory factors such as the macrophage chemoattractant protein-1, interleukin-6, and endothelial adhesion molecules. Consistently, in vitro assays showed that CM increased endothelial adhesiveness to circulating cells by upregulating the expression of adhesion molecules, and stimulated angiogenesis/endothelial tube formation. Overall, our results suggest that both treatments exert vasculoprotective effects on the heart of uremic rats by stimulating endogenous repair mechanisms.
Collapse
|
17
|
Heiss RU, Fahlbusch FB, Jacobi J, Daniel C, Ekici AB, Cordasic N, Amann K, Hartner A, Hilgers KF. Blunted transcriptional response to skeletal muscle ischemia in rats with chronic kidney disease: potential role for impaired ischemia-induced angiogenesis. Physiol Genomics 2017; 49:230-237. [PMID: 28213570 DOI: 10.1152/physiolgenomics.00124.2016] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2016] [Accepted: 02/15/2017] [Indexed: 11/22/2022] Open
Abstract
Chronic kidney disease (CKD) is associated with increased cardiovascular morbidity and mortality. Previous studies indicated an impairment of ischemia-induced angiogenesis in skeletal muscle of rats with CKD. We performed a systematic comparison of early gene expression in response to ischemia in rats with or without CKD to identify potential molecular mechanisms underlying impaired angiogenesis in CKD. CKD was induced in male rats by 5/6 nephrectomy (SNX); control rats were sham operated (sham). Eight weeks later, ischemia of the right limb was induced by ligation and resection of the femoral artery. Rats were killed 24 h after the onset of ischemia, and RNA was extracted from the musculus soleus of the ischemic and the nonischemic hindlimb. To identify differentially expressed transcripts, we analyzed RNA with Affymetrix GeneChip Rat Genome 230 2.0 Arrays. RT-PCR analysis of selected genes was performed to validate observed changes. Hindlimb ischemia upregulated 239 genes in CKD and 299 genes in control rats (66% overlap), whereas only a few genes were downregulated (14 in CKD and 34 in controls) compared with the nonischemic limb of the same animals. Comparison between the ischemic limbs of CKD and controls revealed downregulation of 65 genes in CKD; 37 of these genes were also among the ischemia-induced genes in controls. Analysis of functional groups (other than angiogenesis) pointed to genes involved in leukocyte recruitment and fatty acid metabolism. Transcript expression profiling points to a relatively small number of differentially expressed genes that may underlie the impaired postischemic angiogenesis in CKD.
Collapse
Affiliation(s)
- Rafael U Heiss
- Department of Nephrology and Hypertension, Friedrich-Alexander University, Erlangen-Nuremberg (FAU), Germany.,Department of Radiology, Friedrich-Alexander University, Erlangen-Nuremberg (FAU), Germany
| | - Fabian B Fahlbusch
- Department of Pediatrics and Adolescent Medicine, Friedrich-Alexander University, Erlangen-Nuremberg (FAU), Germany
| | - Johannes Jacobi
- Department of Nephrology and Hypertension, Friedrich-Alexander University, Erlangen-Nuremberg (FAU), Germany
| | - Christoph Daniel
- Institute for Nephropathology, Friedrich-Alexander University, Erlangen-Nuremberg (FAU), Germany; and
| | - Arif B Ekici
- Institute of Human Genetics, Friedrich-Alexander University, Erlangen-Nuremberg (FAU), Germany
| | - Nada Cordasic
- Department of Nephrology and Hypertension, Friedrich-Alexander University, Erlangen-Nuremberg (FAU), Germany
| | - Kerstin Amann
- Institute for Nephropathology, Friedrich-Alexander University, Erlangen-Nuremberg (FAU), Germany; and
| | - Andrea Hartner
- Department of Pediatrics and Adolescent Medicine, Friedrich-Alexander University, Erlangen-Nuremberg (FAU), Germany
| | - Karl F Hilgers
- Department of Nephrology and Hypertension, Friedrich-Alexander University, Erlangen-Nuremberg (FAU), Germany;
| |
Collapse
|
18
|
Hung SC, Kuo KL, Wu CC, Tarng DC. Indoxyl Sulfate: A Novel Cardiovascular Risk Factor in Chronic Kidney Disease. J Am Heart Assoc 2017; 6:JAHA.116.005022. [PMID: 28174171 PMCID: PMC5523780 DOI: 10.1161/jaha.116.005022] [Citation(s) in RCA: 158] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Affiliation(s)
- Szu-Chun Hung
- Division of Nephrology, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien, Taiwan.,School of Medicine, Tzu Chi University, Hualien, Taiwan
| | - Ko-Lin Kuo
- Division of Nephrology, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien, Taiwan.,School of Medicine, Tzu Chi University, Hualien, Taiwan
| | - Chih-Cheng Wu
- Cardiovascular Center, National Taiwan University Hospital, Hsinchu Branch, Hsinchu, Taiwan.,Institute of Biomedical Engineering, National Tsing-Hua University, Hsinchu, Taiwan
| | - Der-Cherng Tarng
- Institutes of Physiology and Clinical Medicine, National Yang-Ming University, Taipei, Taiwan .,Division of Nephrology, Department of Medicine, Taipei Veterans General Hospital, Taipei, Taiwan
| |
Collapse
|
19
|
Wu CC, Hung SC, Kuo KL, Tarng DC. Impact of Indoxyl Sulfate on Progenitor Cell-Related Neovascularization of Peripheral Arterial Disease and Post-Angioplasty Thrombosis of Dialysis Vascular Access. Toxins (Basel) 2017; 9:E25. [PMID: 28067862 PMCID: PMC5308257 DOI: 10.3390/toxins9010025] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2016] [Revised: 12/28/2016] [Accepted: 01/04/2017] [Indexed: 01/09/2023] Open
Abstract
Patients with chronic kidney disease (CKD) have an increased risk of vascular disease, which is associated with considerable health care costs. Vascular disease in CKD differs clinically and pathobiologically from that in patients with normal renal function. Besides the traditional risk factors, retention of uremic toxins contributes to the pathogenesis of vascular disease in patients with CKD. Indoxyl sulfate is a protein-bound uremic toxin and is inefficiently removed by conventional dialysis. Accumulating evidence suggests that indoxyl sulfate is a vascular toxin involved in atherosclerosis, arteriosclerosis, vascular calcification and vascular repair. Clinically, indoxyl sulfate is associated with total and cardiovascular mortality in patients with CKD. Recent studies have indicated that in addition to coronary and cerebral arteries, indoxyl sulfate plays a role in peripheral artery disease (PAD) and dialysis graft thrombosis. Emerging evidence suggests that indoxyl sulfate is implicated via novel mechanisms, including progenitor cell-related neovascularization and tissue factor-related hypercoagulability. These findings raise the possibility that strategies targeting serum indoxyl sulfate may have the potential to improve the outcomes of PAD and dialysis vascular access in patients with CKD.
Collapse
Affiliation(s)
- Chih-Cheng Wu
- Cardiovascular Center, National Taiwan University Hospital, Hsinchu Branch, Hsinchu 30059, Taiwan.
- National Tsing-Hua University, Institute of Biomedical Engineering, Hsinchu 30013, Taiwan.
- School of Medicine, National Yang-Ming University, Taipei 11217, Taiwan.
| | - Szu-Chun Hung
- Division of Nephrology, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation and School of Medicine, Tzu Chi University, Hualien 97004, Taiwan.
| | - Ko-Lin Kuo
- Division of Nephrology, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation and School of Medicine, Tzu Chi University, Hualien 97004, Taiwan.
| | - Der-Cherng Tarng
- Institutes of Physiology and Clinical Medicine, National Yang-Ming University, Taipei 11217, Taiwan.
- Division of Nephrology, Department of Medicine, Taipei Veterans General Hospital, Taipei 11217, Taiwan.
| |
Collapse
|
20
|
Hypoxia inducible factor stabilization improves defective ischemia-induced angiogenesis in a rodent model of chronic kidney disease. Kidney Int 2016; 91:616-627. [PMID: 27927598 DOI: 10.1016/j.kint.2016.09.028] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2016] [Revised: 08/24/2016] [Accepted: 09/15/2016] [Indexed: 12/17/2022]
Abstract
Chronic kidney disease (CKD) is associated with increased risk and worse prognosis of cardiovascular disease, including peripheral artery disease. An impaired angiogenic response to ischemia may contribute to poor outcomes of peripheral artery disease in patients with CKD. Hypoxia inducible factors (HIF) are master regulators of angiogenesis and therefore represent a promising target for therapeutic intervention. To test this we induced hind-limb ischemia in rats with CKD caused by 5/6 nephrectomy and administered two different treatments known to stabilize HIF protein in vivo: carbon monoxide and a pharmacological inhibitor of prolyl hydroxylation 2-(1-chloro-4- hydroxyisoquinoline-3-carboxamido) acetate (ICA). Expression levels of pro-angiogenic HIF target genes (Vegf, Vegf-r1, Vegf-r2, Ho-1) were measured by qRT-PCR. Capillary density was measured by CD31 immunofluorescence staining and HIF expression was evaluated by immunohistochemistry. Capillary density in ischemic skeletal muscle was significantly lower in CKD animals compared to sham controls. Rats with CKD showed significantly lower expression of HIF and all measured pro-angiogenic HIF target genes, including VEGF. Both HIF stabilizing treatments rescued HIF target gene expression in animals with CKD and led to significantly higher ischemia-induced capillary sprouting compared to untreated controls. ICA was effective regardless of whether it was administered before or after induction of ischemia and led to a HIF expression in skeletal muscle. Thus, impaired ischemia-induced angiogenesis in rats with CKD can be improved by HIF stabilization, even if started after onset of ischemia.
Collapse
|
21
|
Burkhardt D, Bartosova M, Schaefer B, Grabe N, Lahrmann B, Nasser H, Freise C, Schneider A, Lingnau A, Degenhardt P, Ranchin B, Sallay P, Cerkauskiene R, Malina M, Ariceta G, Schmitt CP, Querfeld U. Reduced Microvascular Density in Omental Biopsies of Children with Chronic Kidney Disease. PLoS One 2016; 11:e0166050. [PMID: 27846250 PMCID: PMC5113061 DOI: 10.1371/journal.pone.0166050] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2016] [Accepted: 10/21/2016] [Indexed: 01/20/2023] Open
Abstract
BACKGROUND Endothelial dysfunction is an early manifestation of cardiovascular disease (CVD) and consistently observed in patients with chronic kidney disease (CKD). We hypothesized that CKD is associated with systemic damage to the microcirculation, preceding macrovascular pathology. To assess the degree of "uremic microangiopathy", we have measured microvascular density in biopsies of the omentum of children with CKD. PATIENTS AND METHODS Omental tissue was collected from 32 healthy children (0-18 years) undergoing elective abdominal surgery and from 23 age-matched cases with stage 5 CKD at the time of catheter insertion for initiation of peritoneal dialysis. Biopsies were analyzed by independent observers using either a manual or an automated imaging system for the assessment of microvascular density. Quantitative immunohistochemistry was performed for markers of autophagy and apoptosis, and for the abundance of the angiogenesis-regulating proteins VEGF-A, VEGF-R2, Angpt1 and Angpt2. RESULTS Microvascular density was significantly reduced in uremic children compared to healthy controls, both by manual imaging with a digital microscope (median surface area 0.61% vs. 0.95%, p<0.0021 and by automated quantification (total microvascular surface area 0.89% vs. 1.17% p = 0.01). Density measured by manual imaging was significantly associated with age, height, weight and body surface area in CKD patients and healthy controls. In multivariate analysis, age and serum creatinine level were the only independent, significant predictors of microvascular density (r2 = 0.73). There was no immunohistochemical evidence for apoptosis or autophagy. Quantitative staining showed similar expression levels of the angiogenesis regulators VEGF-A, VEGF-receptor 2 and Angpt1 (p = 0.11), but Angpt2 was significantly lower in CKD children (p = 0.01). CONCLUSIONS Microvascular density is profoundly reduced in omental biopsies of children with stage 5 CKD and associated with diminished Angpt2 signaling. Microvascular rarefaction could be an early systemic manifestation of CKD-induced cardiovascular disease.
Collapse
Affiliation(s)
- Dorothea Burkhardt
- Department of Pediatric Nephrology, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Maria Bartosova
- Center for Pediatric and Adolescent Medicine, University of Heidelberg, Heidelberg, Germany
| | - Betti Schaefer
- Center for Pediatric and Adolescent Medicine, University of Heidelberg, Heidelberg, Germany
| | - Niels Grabe
- Bioquant, Hamamatsu Tissue Imaging and Analysis (TIGA) Center, University of Heidelberg, Heidelberg, Germany
| | - Bernd Lahrmann
- Bioquant, Hamamatsu Tissue Imaging and Analysis (TIGA) Center, University of Heidelberg, Heidelberg, Germany
| | - Hamoud Nasser
- Center for Cardiovascular Research, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Christian Freise
- Center for Cardiovascular Research, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Axel Schneider
- Department of Pediatric Surgery, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Anja Lingnau
- Department of Pediatric Urology, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Petra Degenhardt
- Department of Pediatric Surgery, Charité-Universitätsmedizin Berlin, Berlin, Germany
- Department of Pediatric Surgery, Klinikum Ernst von Bergmann, Potsdam, Germany
| | - Bruno Ranchin
- Hospices Civils de Lyon, Service de Nephrologie Pediatrique and Epicime-Centre d’Investigation Clinique 1407, Hopital Femme Mere Enfant, Lyon, France
| | - Peter Sallay
- First Department of Pediatrics, Semmelweis University, Budapest, Hungary
| | - Rimante Cerkauskiene
- Coordinating Centre for Children’s Rare Diseases, Children´s Hospital, Affiliate of Vilnius University Hospital Santariskiu Klinikos, Vilnius, Lithuania
| | - Michal Malina
- Department of Pediatrics, Second Faculty of Medicine, Charles University-Prague, Prague 5, Czech Republic
| | - Gema Ariceta
- Servicio de Nefrología Pediátrica, Hospital Universitari Vall d’Hebron, Barcelona, Spain
| | - Claus Peter Schmitt
- Center for Pediatric and Adolescent Medicine, University of Heidelberg, Heidelberg, Germany
| | - Uwe Querfeld
- Department of Pediatric Nephrology, Charité-Universitätsmedizin Berlin, Berlin, Germany
- Center for Cardiovascular Research, Charité Universitätsmedizin Berlin, Berlin, Germany
- * E-mail:
| |
Collapse
|
22
|
Hartner A, Jagusch L, Cordasic N, Amann K, Veelken R, Jacobi J, Hilgers KF. Impaired Neovascularization and Reduced Capillary Supply in the Malignant vs. Non-malignant Course of Experimental Renovascular Hypertension. Front Physiol 2016; 7:370. [PMID: 27625610 PMCID: PMC5003830 DOI: 10.3389/fphys.2016.00370] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2016] [Accepted: 08/11/2016] [Indexed: 01/29/2023] Open
Abstract
Malignant hypertension develops in some cases of hypertension but not in others. We hypothesized that an impaired neovascularization and a reduced capillary supply characterizes the malignant course of experimental hypertension. Two-kidney, one-clip renovascular hypertension was induced in rats; controls (sham) were sham operated. To distinguish malignant hypertension from non-malignant hypertension, we considered two factors: weight loss, and the number of typical vascular lesions (onion skin lesions and fibrinoid necroses) per kidney section of the nonclipped kidney. Animals in the upper half for both criteria were defined as malignant hypertensives. After 5 weeks, mean arterial blood pressure was elevated to the same degree in malignant hypertension and non-malignant hypertension whereas plasma renin and aldosterone were significantly higher in malignant hypertensives. The expression of plasminogen activator inhibitor-1 was elevated (up to 14-fold) in non-malignant but significantly more increased (up to 36-fold) in malignant hypertensive rats, compared to sham. As a bioassay for neovascularization, the area of granulation tissue ingrowth in polyvinyl discs (implanted subcutaneously) was reduced in malignant hypertension compared to non-malignant hypertension and sham, while there was no difference between non-malignant hypertension and sham. The number of renal and left ventricular capillaries was significantly lower in malignant hypertension compared to non-malignant hypertension, as was the number of proliferating endothelial cells. We conclude that an impaired neovascularization and capillarization occurs in malignant renovascular hypertension but not in the non-malignant course of the disease despite comparable blood pressure levels. This might contribute to the unique vascular lesions and progressive target organ damage observed in malignant hypertension.
Collapse
Affiliation(s)
- Andrea Hartner
- Department of Pediatrics and Adolescent Medicine, University Hospital of Erlangen Erlangen, Germany
| | - Lisa Jagusch
- Department of Nephrology and Hypertension, University Hospital of Erlangen Erlangen, Germany
| | - Nada Cordasic
- Department of Nephrology and Hypertension, University Hospital of Erlangen Erlangen, Germany
| | - Kerstin Amann
- Department of Nephropathology, University Hospital of Erlangen Erlangen, Germany
| | - Roland Veelken
- Department of Nephrology and Hypertension, University Hospital of Erlangen Erlangen, Germany
| | - Johannes Jacobi
- Department of Nephrology and Hypertension, University Hospital of Erlangen Erlangen, Germany
| | - Karl F Hilgers
- Department of Nephrology and Hypertension, University Hospital of Erlangen Erlangen, Germany
| |
Collapse
|
23
|
Abstract
The heart and the vascular tree undergo major structural and functional changes when kidney function declines and renal replacement therapy is required. The many cardiovascular risk factors and adaptive changes the heart undergoes include left ventricular hypertrophy and dilatation with concomitant systolic and diastolic dysfunction. Myocardial fibrosis is the consequence of impaired angio-adaptation, reduced capillary angiogenesis, myocyte-capillary mismatch, and myocardial micro-arteriopathy. The vascular tree can be affected by both atherosclerosis and arteriosclerosis with both lipid rich plaques and abundant media calcification. Development of cardiac and vascular disease is rapid, especially in young patients, and the phenotype resembles all aspects of an accelerated ageing process and latent cardiac failure. The major cause of left ventricular hypertrophy and failure and the most common problem directly affecting myocardial function is fluid overload and, usually, hypertension. In situations of stress, such as intradialytic hypotension and hypoxaemia, the hearts of these patients are more vulnerable to developing cardiac arrest, especially when such episodes occur frequently. As a result, cardiac and vascular mortality are several times higher in dialysis patients than in the general population. Trials investigating one pharmacological intervention (eg, statins) have shown limitations. Pragmatic designs for large trials on cardio-active interventions are mandatory for adequate cardioprotective renal replacement therapy.
Collapse
Affiliation(s)
- Christoph Wanner
- Comprehensive Heart Failure Center and Renal Division, University Hospital of Würzburg, Würzburg, Germany.
| | - Kerstin Amann
- Department of Nephropathology at the Department of Pathology, University of Erlangen-Nürnberg, Erlangen, Germany
| | - Tetsuo Shoji
- Department of Geriatrics and Vascular Medicine, Osaka City University Graduate School of Medicine, Osaka, Japan
| |
Collapse
|
24
|
Indoxyl sulfate suppresses endothelial progenitor cell–mediated neovascularization. Kidney Int 2016; 89:574-85. [DOI: 10.1016/j.kint.2015.11.020] [Citation(s) in RCA: 65] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2015] [Revised: 10/29/2015] [Accepted: 11/12/2015] [Indexed: 11/18/2022]
|
25
|
Driesen T, Schuler D, Schmetter R, Heiss C, Kelm M, Fischer JW, Freudenberger T. A systematic approach to assess locoregional differences in angiogenesis. Histochem Cell Biol 2015; 145:213-25. [PMID: 26526138 DOI: 10.1007/s00418-015-1379-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/19/2015] [Indexed: 10/22/2022]
Abstract
Skeletal muscle tissue differs with regard to the abundance of glycolytic and oxidative fiber types. In this context, capillary density has been described to be higher in muscle tissue with more oxidative metabolism as compared to that one with more glycolytic metabolism, and the highest abundance of capillaries has been found in boneward-oriented moieties of skeletal muscle tissue. Importantly, capillary formation is often analyzed as a measure for angiogenesis, a process that describes neo-vessel formation emanating from preexisting vessels, occurring, i.e., after arterial occlusion. However, a standardized way for investigation of calf muscle capillarization after surgically induced unilateral hind limb ischemia in mice, especially considering these locoregional differences, has not been provided so far. In this manuscript, a novel, methodical approach for reliable analysis of capillary density was established using anatomic-morphological reference points, and a software-assisted way of capillary density analysis is described. Thus, the systematic approach provided conscientiously considers intra-layer differences in capillary formation and therefore guarantees for a robust, standardized analysis of capillary density as a measure for angiogenesis. The significance of the methodology is further supported by the observation that capillary density in the calf muscle layers analyzed negatively correlates with distal lower limb perfusion measured in vivo.
Collapse
Affiliation(s)
- T Driesen
- Institut für Pharmakologie und Klinische Pharmakologie, Universitätsklinikum Düsseldorf, Heinrich-Heine-Universität Düsseldorf, Universitätsstraße 1, 40225, Düsseldorf, Germany
| | - D Schuler
- Klinik für Kardiologie, Pneumologie und Angiologie, Universitätsklinikum Düsseldorf, Moorenstraße 5, 40225 Düsseldorf, Germany
| | - R Schmetter
- Institut für Pharmakologie und Klinische Pharmakologie, Universitätsklinikum Düsseldorf, Heinrich-Heine-Universität Düsseldorf, Universitätsstraße 1, 40225, Düsseldorf, Germany
| | - C Heiss
- Klinik für Kardiologie, Pneumologie und Angiologie, Universitätsklinikum Düsseldorf, Moorenstraße 5, 40225 Düsseldorf, Germany
| | - M Kelm
- Klinik für Kardiologie, Pneumologie und Angiologie, Universitätsklinikum Düsseldorf, Moorenstraße 5, 40225 Düsseldorf, Germany
| | - J W Fischer
- Institut für Pharmakologie und Klinische Pharmakologie, Universitätsklinikum Düsseldorf, Heinrich-Heine-Universität Düsseldorf, Universitätsstraße 1, 40225, Düsseldorf, Germany
| | - T Freudenberger
- Institut für Pharmakologie und Klinische Pharmakologie, Universitätsklinikum Düsseldorf, Heinrich-Heine-Universität Düsseldorf, Universitätsstraße 1, 40225, Düsseldorf, Germany.
| |
Collapse
|
26
|
Soluble Flt-1 links microvascular disease with heart failure in CKD. Basic Res Cardiol 2015; 110:30. [PMID: 25893874 DOI: 10.1007/s00395-015-0487-4] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/23/2015] [Revised: 03/29/2015] [Accepted: 04/13/2015] [Indexed: 01/17/2023]
Abstract
Chronic kidney disease (CKD) is associated with an increased risk of heart failure (HF). Elevated plasma concentrations of soluble Flt-1 (sFlt-1) have been linked to cardiovascular disease in CKD patients, but whether sFlt-1 contributes to HF in CKD is still unknown. To provide evidence that concludes a pathophysiological role of sFlt-1 in CKD-associated HF, we measured plasma sFlt-1 concentrations in 586 patients with angiographically documented coronary artery disease and renal function classified according to estimated glomerular filtration rate (eGFR). sFlt-1 concentrations correlated negatively with eGFR and were associated with signs of heart failure, based on New York Heart Association functional class and reduced left ventricular ejection fraction (LVEF), and early mortality. Additionally, rats treated with recombinant sFlt-1 showed a 15 % reduction in LVEF and a 29 % reduction in cardiac output compared with control rats. High sFlt-1 concentrations were associated with a 15 % reduction in heart capillary density (number of vessels/cardiomyocyte) and a 24 % reduction in myocardial blood volume. Electron microscopy and histological analysis revealed mitochondrial damage and interstitial fibrosis in the hearts of sFlt-1-treated, but not control rats. In 5/6-nephrectomised rats, an animal model of CKD, sFlt-1 antagonism with recombinant VEGF121 preserved heart microvasculature and significantly improved heart function. Overall, these findings suggest that a component of cardiovascular risk in CKD patients could be directly attributed to sFlt-1. Assessment of patients with CKD confirmed that sFlt-1 concentrations were inversely correlated with renal function, while studies in rats suggested that sFlt-1 may link microvascular disease with HF in CKD.
Collapse
|
27
|
Chin SH, Item F, Wueest S, Zhou Z, Wiedemann MSF, Gai Z, Schoenle EJ, Kullak-Ublick GA, Al-Hasani H, Konrad D. Opposing effects of reduced kidney mass on liver and skeletal muscle insulin sensitivity in obese mice. Diabetes 2015; 64:1131-41. [PMID: 25325737 DOI: 10.2337/db14-0779] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Reduced kidney mass and/or function may result in multiple metabolic derangements, including insulin resistance. However, underlying mechanisms are poorly understood. Herein, we aimed to determine the impact of reduced kidney mass on glucose metabolism in lean and obese mice. To that end, 7-week-old C57BL/6J mice underwent uninephrectomy (UniNx) or sham operation. After surgery, animals were fed either a chow (standard) diet or a high-fat diet (HFD), and glucose homeostasis was assessed 20 weeks after surgery. Intraperitoneal glucose tolerance was similar in sham-operated and UniNx mice. However, insulin-stimulated glucose disposal in vivo was significantly diminished in UniNx mice, whereas insulin-stimulated glucose uptake into isolated skeletal muscle was similar in sham-operated and UniNx mice. Of note, capillary density was significantly reduced in skeletal muscle of HFD-fed UniNx mice. In contrast, hepatic insulin sensitivity was improved in UniNx mice. Furthermore, adipose tissue hypoxia-inducible factor 1α expression and inflammation were reduced in HFD-fed UniNx mice. Treatment with the angiotensin II receptor blocker telmisartan improved glucose tolerance and hepatic insulin sensitivity in HFD-fed sham-operated but not UniNx mice. In conclusion, UniNx protects from obesity-induced adipose tissue inflammation and hepatic insulin resistance, but it reduces muscle capillary density and, thus, deteriorates HFD-induced skeletal muscle glucose disposal.
Collapse
Affiliation(s)
- Siew Hung Chin
- Division of Pediatric Endocrinology and Diabetology, University Children's Hospital, Zurich, Switzerland Children's Research Center, University Children's Hospital, Zurich, Switzerland Zurich Center for Integrative Human Physiology, University of Zurich, Zurich, Switzerland
| | - Flurin Item
- Division of Pediatric Endocrinology and Diabetology, University Children's Hospital, Zurich, Switzerland Children's Research Center, University Children's Hospital, Zurich, Switzerland
| | - Stephan Wueest
- Division of Pediatric Endocrinology and Diabetology, University Children's Hospital, Zurich, Switzerland Children's Research Center, University Children's Hospital, Zurich, Switzerland
| | - Zhou Zhou
- German Diabetes Center at Heinrich Heine University, Düsseldorf, Germany
| | - Michael S F Wiedemann
- Division of Pediatric Endocrinology and Diabetology, University Children's Hospital, Zurich, Switzerland Children's Research Center, University Children's Hospital, Zurich, Switzerland Zurich Center for Integrative Human Physiology, University of Zurich, Zurich, Switzerland
| | - Zhibo Gai
- Department of Clinical Pharmacology and Toxicology, University Hospital Zurich, Zurich, Switzerland
| | - Eugen J Schoenle
- Division of Pediatric Endocrinology and Diabetology, University Children's Hospital, Zurich, Switzerland Children's Research Center, University Children's Hospital, Zurich, Switzerland
| | - Gerd A Kullak-Ublick
- Department of Clinical Pharmacology and Toxicology, University Hospital Zurich, Zurich, Switzerland
| | - Hadi Al-Hasani
- German Diabetes Center at Heinrich Heine University, Düsseldorf, Germany German Center for Diabetes Research, Düsseldorf, Germany
| | - Daniel Konrad
- Division of Pediatric Endocrinology and Diabetology, University Children's Hospital, Zurich, Switzerland Children's Research Center, University Children's Hospital, Zurich, Switzerland Zurich Center for Integrative Human Physiology, University of Zurich, Zurich, Switzerland
| |
Collapse
|
28
|
Abstract
PURPOSE OF REVIEW Left ventricular hypertrophy (LVH) is common in end-stage renal disease (ESRD) and has been advocated as a therapeutic target. We review the considerations for targeting LVH as a modifiable risk factor in ESRD. RECENT FINDINGS Pathologic myocardial changes underlying LVH provide an ideal substrate for the spread of arrhythmia and may be key contributors to the occurrence of sudden death in ESRD. LVH is present in 68-89% of incident hemodialysis patients and is frequently progressive, although regression is observed in a minority of patients. Higher degrees of baseline LVH, as well as greater increases in left ventricular mass index over time, are associated with decreased survival, but whether these associations are causal remains uncertain. Several interventions, including angiotensin blockade and frequent dialysis, can reduce the left ventricular mass index, but whether this is associated with improved survival has not been definitively demonstrated. SUMMARY LVH is a highly prevalent and reversible risk factor, which holds promise as a novel therapeutic target in ESRD. Interventional trials are needed to provide additional evidence that LVH regression improves survival before prevention and reversal of LVH can be definitively adopted as a therapeutic paradigm in ESRD.
Collapse
MESH Headings
- Death, Sudden, Cardiac/etiology
- Death, Sudden, Cardiac/prevention & control
- Disease Progression
- Humans
- Hypertrophy, Left Ventricular/diagnosis
- Hypertrophy, Left Ventricular/etiology
- Hypertrophy, Left Ventricular/mortality
- Hypertrophy, Left Ventricular/therapy
- Kidney Failure, Chronic/complications
- Kidney Failure, Chronic/diagnosis
- Kidney Failure, Chronic/mortality
- Kidney Failure, Chronic/therapy
- Prevalence
- Prognosis
- Risk Assessment
- Risk Factors
Collapse
Affiliation(s)
- David Charytan
- Renal Division, Brigham and Women's Hospital, Boston, Massachusetts, USA
| |
Collapse
|
29
|
Bhatt H, Kochar S, Htun WW, Julliard K, Fernaine G. Coronary Collateral Circulation and Cardiovascular Risk Factors: Is There a Paradox? Angiology 2014; 66:588-94. [PMID: 25092680 DOI: 10.1177/0003319714545342] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
We sought to determine the association of major cardiovascular risk factors and other comorbidities with the presence or absence of coronary collateral (CC) circulation. All electronic medical records from 2010 to 2011 were retrospectively reviewed. A total of 563 patients were divided into 2 groups: CC present (180) and CC absent (383). Smoking (P = .012, odds ratio [OR] 1.58), hypercholesterolemia (P = .001, OR 2.21), and hypertension (P = .034, OR 1.75) were associated with the presence of CC. Increasing body mass index (BMI, P = .001) and decreasing estimated glomerular filtration rate (eGFR, P = .042) were associated with the absence of CC. On multivariable linear regression analysis, hypercholesterolemia (P = .001, OR 2.28), BMI (P = .012, OR 0.77), and eGFR (P = .001, OR 0.70) were found to be independently associated with CC. Our findings will help predict patient populations more likely to have presence or absence of CC circulation.
Collapse
Affiliation(s)
- Hemal Bhatt
- Department of Internal Medicine, Lutheran Medical Center, Brooklyn, NY, USA
| | - Suzi Kochar
- Department of Internal Medicine, Lutheran Medical Center, Brooklyn, NY, USA
| | - Wah Wah Htun
- Department of Internal Medicine, Lutheran Medical Center, Brooklyn, NY, USA
| | - Kell Julliard
- Department of Internal Medicine, Lutheran Medical Center, Brooklyn, NY, USA
| | - George Fernaine
- Department of Cardiology, Lutheran Medical Center, Brooklyn, NY, USA
| |
Collapse
|
30
|
Increased concentration of circulating angiogenesis and nitric oxide inhibitors induces endothelial to mesenchymal transition and myocardial fibrosis in patients with chronic kidney disease. Int J Cardiol 2014; 176:99-109. [PMID: 25049013 DOI: 10.1016/j.ijcard.2014.06.062] [Citation(s) in RCA: 90] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/09/2014] [Revised: 05/19/2014] [Accepted: 06/28/2014] [Indexed: 02/07/2023]
Abstract
BACKGROUND Sudden cardiovascular death is increased in chronic kidney disease (CKD). Experimental CKD models suggest that angiogenesis and nitric oxide (NO) inhibitors induce myocardial fibrosis and microvascular dropout thereby facilitating arrhythmogenesis. We undertook this study to characterize associations of CKD with human myocardial pathology, NO-related circulating angiogenesis inhibitors, and endothelial cell behavior. METHODS We compared heart (n=54) and serum (n=162) samples from individuals with and without CKD, and assessed effects of serum on human coronary artery endothelial cells (HCAECs) in vitro. Left ventricular fibrosis and capillary density were quantified in post-mortem samples. Endothelial to mesenchymal transition (EndMT) was assessed by immunostaining of post-mortem samples and RNA expression in heart tissue obtained during cardiac surgery. Circulating asymmetric dimethylarginine (ADMA), endostatin (END), angiopoietin-2 (ANG), and thrombospondin-2 (TSP) were measured, and the effect of these factors and of subject serum on proliferation, apoptosis, and EndMT of HCAEC was analyzed. RESULTS Cardiac fibrosis increased 12% and 77% in stage 3-4 CKD and ESRD and microvascular density decreased 12% and 16% vs. preserved renal function. EndMT-derived fibroblast proportion was 17% higher in stage 3-4 CKD and ESRD (P trend = 0.02). ADMA, ANG, TSP, and END concentrations increased in CKD. Both individual factors and CKD serum increased HCAEC apoptosis (P=0.02), decreased proliferation (P=0.03), and induced EndMT. CONCLUSIONS CKD is associated with an increase in circulating angiogenesis and NO inhibitors, which impact proliferation and apoptosis of cardiac endothelial cells and promote EndMT, leading to cardiac fibrosis and capillary rarefaction. These processes may play key roles in CKD-associated CV disease.
Collapse
|
31
|
Jin Q, Sun L, Li S, Yang X, Lu Y, Li Y. Number and function impairment of resident C-Kit+ cardiac stem cells in mice with renal dysfunction caused by 5/6 nephrectomy. Ren Fail 2013; 35:1136-41. [PMID: 23879535 DOI: 10.3109/0886022x.2013.815108] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND Cardiac stem cell (CSC) dysfunction exists in various kinds of cardiovascular diseases, and may be responsible for the insufficient regeneration of cardiac myocytes and coronary vessels. However, whether chronic renal failure (CRF) affected CSC is unknown. METHOD CRF was induced in adult male mice by 5/6 nephrectomy. The mice were killed at 12 weeks after operation. C-kit+ CSC numbers was evaluated by flow cytometer. Apoptosis and DNA damage of C-kit+ CSC in the control and CRF mice was analyzed by immunohistochemistry. In the in vitro study, normal medium, and medium with uremic rat serum were used for the CSC culture. RESULTS CSC counts attenuated significantly in the chronic renal failure model, whereas apoptosis cells and 8-OHdG-positive cells significantly increased. CSC derived form 5/6 nephrectomy mice showed an impaired anti-oxidant potential. In the cultured cells, CSCs subjected to uremic rat serum showed a higher frequency of TUNEL stain-positive and 8-OHdG-positive cells. The uremia rat serum reduced the expression of hepatocyte growth factor (HGF) and vascular endothelial growth factor (VEGF) in CSC. CONCLUSIONS The current study elucidated that CSC number and function disorders existed in mice with chronic renal insufficiency. Apoptosis, oxidative stress and reduced angiogenic factors secretion caused by uremic toxins in serum are contributors to CSC dysfunction.
Collapse
Affiliation(s)
- Qiu Jin
- Kidney Disease Center, The First Affiliated Hospital, Medical College of Zhejiang University, Hangzhou, Zhejiang, PR China
| | | | | | | | | | | |
Collapse
|
32
|
Antioxidant treatment reverts increased arterial Basal tone and oxidative stress in nephrectomized (5/6) hypertensive rats. Int J Hypertens 2013; 2013:863067. [PMID: 23573416 PMCID: PMC3610369 DOI: 10.1155/2013/863067] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2012] [Accepted: 02/10/2013] [Indexed: 12/17/2022] Open
Abstract
Nonischemic 5/6 nephrectomized rat (NefR) is a model of chronic kidney disease. However, little is known about vascular dysfunction and its relation with hypertension in NefR. Aims. To evaluate possible alterations of endothelial function, NO-bioavailability, and basal tone in aorta from NefR and the role of oxidative stress. Sprague Dawley rats were divided into sham rats (SR), NefR, and NefR treated with tempol (NefR-T). Mean arterial pressure (MAP) and renal function were determined. In isolated aortic rings the following was measured: 1-endothelial function, 2-basal tone, 3-NO levels, 4-membrane potential (MP), and 5-oxidative stress. NefR increased MAP (SR: 119 ± 4 mmHg; n = 7; NefR: 169 ± 6; n = 8; P < 0.001). Tempol did not modify MAP (NefR-T: 168 ± 10; n = 6; P < 0.001). NefR showed endothelial dysfunction, increased basal tone and decreased NO levels (SR: 32 ± 2 nA; n = 7, NefR: 10 ± 2; n = 8; P < 0.001). In both in vitro and in vivo tempol improves basal tone, NO levels, and MP. Oxidative stress in NefR was reverted in NefR-T. We described, for the first time, that aorta from NefR presented increased basal tone related to endothelial dysfunction and decreased NO-bioavailability. The fact that tempol improves NO-contents and basal tone, without decrease MAP, indicates that oxidative stress could be implicated early and independently to hypertension, in the vascular alterations.
Collapse
|
33
|
Di Marco GS, König M, Stock C, Wiesinger A, Hillebrand U, Reiermann S, Reuter S, Amler S, Köhler G, Buck F, Fobker M, Kümpers P, Oberleithner H, Hausberg M, Lang D, Pavenstädt H, Brand M. High phosphate directly affects endothelial function by downregulating annexin II. Kidney Int 2012; 83:213-22. [PMID: 22913982 DOI: 10.1038/ki.2012.300] [Citation(s) in RCA: 80] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Hyperphosphatemia is associated with increased cardiovascular risk in patients with renal disease and in healthy individuals. Here we tested whether high phosphate has a role in the pathophysiology of cardiovascular events by interfering with endothelial function, thereby impairing microvascular function and angiogenesis. Protein expression analysis found downregulation of annexin II in human coronary artery endothelial cells, an effect associated with exacerbated shedding of annexin II-positive microparticles by the cells exposed to high phosphate media. EAhy926 endothelial cells exposed to sera from hyperphosphatemic patients also display decreased annexin II, suggesting a negative correlation between serum phosphate and annexin II expression. By using endothelial cell-based assays in vitro and the chicken chorioallantoic membrane assay in vivo, we found that angiogenesis, vessel wall morphology, endothelial cell migration, capillary tube formation, and endothelial survival were impaired in a hyperphosphatemic milieu. Blockade of membrane-bound extracellular annexin II with a specific antibody mimicked the effects of high phosphate. In addition, high phosphate stiffened endothelial cells in vitro and in rats in vivo. Thus, our results link phosphate and adverse clinical outcomes involving the endothelium in both healthy individuals and patients with renal disease.
Collapse
|
34
|
Charytan DM, Stern NM, Mauri L. CKD and coronary collateral supply in individuals undergoing coronary angiography after myocardial infarction. Clin J Am Soc Nephrol 2012; 7:1079-86. [PMID: 22516292 DOI: 10.2215/cjn.11171111] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
BACKGROUND AND OBJECTIVES CKD patients have high mortality risk after myocardial infarction (MI). An adequate supply of coronary collaterals to the culprit vessel responsible for MI is associated with reduced risks of death and complications. Whether a diminished supply of collaterals contributes to the high risk in CKD patients is uncertain. DESIGN, SETTING, PARTICIPANTS, & MEASUREMENTS Quantitative coronary angiography was performed in a consecutive series of individuals with (n=58) and without (n=165) CKD (estimated GFR <60 ml/min per 1.73 m(2)) who underwent coronary angiography at the time of MI. Collateral supply was analyzed and candidate predictors were assessed in patient-level and individual artery-level models using logistic regression and ordered categorical regression, respectively. RESULTS There were no significant differences in collateral supply among 58 CKD patients and 165 individuals with preserved renal function. Culprit artery collaterals were present in 25.0% of CKD patients compared with 27.2% of individuals with preserved renal function (P=0.76). The odds of having an adequate supply of culprit vessel collaterals were also not significantly different in individuals with and without CKD, respectively. CKD patients were 2.22-fold more likely to have visible collaterals to the nonculprit vessels in unadjusted analyses. The difference was not significant after correction for percent stenosis and comorbid factors. CONCLUSIONS Our results do not support an independent association between CKD and diminished collateral supply to either the culprit or nonculprit vessels in MI. Additional studies are warranted to better define associations between myocardial capillary supply, collateral supply, and the full range of human CKD.
Collapse
Affiliation(s)
- David M Charytan
- Department of Medicine, Brigham and Women's Hospital, Boston, MA 02120, USA.
| | | | | |
Collapse
|
35
|
Flisinski M, Brymora A, Bartlomiejczyk I, Wisniewska E, Golda R, Stefanska A, Paczek L, Manitius J. Decreased Hypoxia-Inducible Factor-1a in Gastrocnemius Muscle in Rats with Chronic Kidney Disease. ACTA ACUST UNITED AC 2012; 35:608-18. [DOI: 10.1159/000339706] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2010] [Accepted: 05/23/2012] [Indexed: 01/13/2023]
|
36
|
Monge M, Massy ZA, Zonneveld AJV, Rabelink TJ. Cellules progénitrices endothéliales, de quoi parle-t-on ? Nephrol Ther 2011; 7:521-5. [DOI: 10.1016/j.nephro.2011.01.006] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2010] [Revised: 01/09/2011] [Accepted: 01/16/2011] [Indexed: 11/29/2022]
|
37
|
Mayer G. Capillary rarefaction, hypoxia, VEGF and angiogenesis in chronic renal disease. Nephrol Dial Transplant 2011; 26:1132-7. [PMID: 21330358 PMCID: PMC3070072 DOI: 10.1093/ndt/gfq832] [Citation(s) in RCA: 93] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
Tubulointerstitial hypoxia and peritubular capillary rarefaction are typical features of chronic progressive renal disease. In response to low oxygen supply, hypoxia-inducible factors (HIFs) are activated but until now, it is unclear if this increased expression leads to a stabilization of the disease process and thus is nephroprotective or contributes to interstitial fibrosis and/or tubular atrophy. This duality has also been described as far as vascular endothelial growth factor (VEGF), one of the major target genes of HIFs, is concerned. On the one hand, neoangiogenesis driven by VEGF, if intact, ameliorates hypoxia, on the other, VEGF is a potent pro-inflammatory mediator and neoangiogenesis, if defective because interference by other pathologies exaggerates injury. In summary, experimental data support the idea that dependent on timing and predominant pathology, hypoxia counter-regulatory factors exert beneficial or undesirable effects. Thus, before their therapeutic potential can be fully explored, a better way to characterize the clinical and pathophysiological situation in an individual patient is mandatory.
Collapse
Affiliation(s)
- Gert Mayer
- Department of Internal Medicine IV, Nephrology and Hypertension, Medical University Innsbruck, Innsbruck, Austria.
| |
Collapse
|
38
|
Amann K, Odoni G, Benz K, Campean V, Jacobi J, Hilgers KF, Hartner A, Veelken R, Orth SR. Sympathetic blockade prevents the decrease in cardiac VEGF expression and capillary supply in experimental renal failure. Am J Physiol Renal Physiol 2011; 300:F105-12. [DOI: 10.1152/ajprenal.00363.2010] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Uremic cardiomyopathy of men and rodents is characterized by lower myocardial capillary supply that in rats could be prevented by central and peripheral blockade of the sympathetic nervous system. The underlying pathomechanisms remain largely unknown. We investigated whether alterations of cardiac vascular endothelial growth factor (VEGF) gene and protein expression were involved. In our long-term experiment, we analyzed whether VEGF gene and protein expression was altered in the heart of male Sprague-Dawley rats with either sham operation (sham, n = 10) or subtotal nephrectomy (SNX, n = 10). In our short-term experiment (17 sham, 24 SNX), the effect of a putative downregulation of sympathetic nervous activity by surgical renal denervation (interruption of renal afferent pathways) on cardiac gene expression of VEGF, flt-1, and flk-1 and on myocardial capillary supply was analyzed. In the long-term study, cardiac capillary supply and vascular endothelial growth factor gene and protein expression were significantly lower in SNX than in sham. In the short-term experiment, cardiac VEGF mRNA expression was significantly lower in untreated SNX (4,258 ± 2,078 units) than in both sham groups (11,709 ± 4,169 and 8,998 ± 4,823 units); this decrease was significantly prevented by renal denervation (8,190 ± 3,889, P < 0.05). We conclude that cardiac VEGF gene and protein expression is reduced in experimental renal failure, and this may be considered as one potential reason for impaired myocardial adaptation under the situation of cardiac hypertrophy. The beneficial effect of sympathetic downregulation on cardiac structure and function in renal failure may be at least in part explained by increased cardiac VEGF gene expression.
Collapse
Affiliation(s)
| | - G. Odoni
- Ospedale Civile di Dolo, Venezia, Italia
| | | | | | - J. Jacobi
- Hypertensiology and Nephrology, University of Erlangen-Nürnberg, Erlangen-Nürnberg
| | - K. F. Hilgers
- Hypertensiology and Nephrology, University of Erlangen-Nürnberg, Erlangen-Nürnberg
| | | | - R. Veelken
- Hypertensiology and Nephrology, University of Erlangen-Nürnberg, Erlangen-Nürnberg
| | - S. R. Orth
- Department of Internal Medicine, University of Regensburg, Regensburg
- Dialysis Centre Bad Aibling, Bad Aibling, Germany; and
| |
Collapse
|
39
|
Di Marco GS, Reuter S, Kentrup D, Ting L, Ting L, Grabner A, Jacobi AM, Pavenstädt H, Baba HA, Tiemann K, Brand M. Cardioprotective effect of calcineurin inhibition in an animal model of renal disease. Eur Heart J 2010; 32:1935-45. [PMID: 21138940 DOI: 10.1093/eurheartj/ehq436] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
AIMS Chronic kidney disease is directly associated with cardiovascular complications. Heart remodelling, including fibrosis, hypertrophy, and decreased vascularization, is frequently present in renal diseases. Our objective was to investigate the impact of calcineurin inhibitors (CNI) on cardiac remodelling and function in a rat model of renal disease. METHODS AND RESULTS Male Sprague Dawley rats were divided into six groups: sham-operated rats, 5/6 nephrectomized rats (Nx) treated with vehicle, CNI (cyclosporine A 5.0 or 7.5, or tacrolimus 0.5 mg/kg/day) or hydralazine (20 mg/kg twice a day) for 14 days, starting on the day of surgery. Creatinine clearance was significantly lower and blood pressure significantly higher in Nx rats when compared with controls. Morphological and echocardiographic analyses revealed increased left ventricular hypertrophy and decreased number of capillaries in Nx rats. Treatment with CNI affected neither the renal function nor the blood pressure, but prevented the development of cardiac hypertrophy and improved vascularization. In addition, regional blood volume improved as confirmed by contrast agent-based echocardiography. Hydralazine treatment did not avoid heart remodelling in this model. Gene expression analysis verified a decrease in hypertrophic genes in the heart of CNI-treated rats, while pro-angiogenic and stem cell-related genes were upregulated. Moreover, mobilization of stem/progenitor cells was increased through manipulation of the CD26/SDF-1 system. CONCLUSION We conclude from our studies that CNI-treatment significantly prevented cardiac remodelling and improved heart function in Nx rats without affecting renal function and blood pressure. This sheds new light on possible therapeutic strategies for renal patients at high cardiovascular risk.
Collapse
Affiliation(s)
- Giovana S Di Marco
- Department of Internal Medicine D, University of Münster, 48149 Münster, Germany
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Lee CM, Kim EM, Cheong SJ, Kim DW, Lim ST, Sohn MH, Jeong HJ. Targeted molecular imaging of VEGF receptors overexpressed in ischemic microvasculature using chitosan-DC101 conjugates. J Biomed Mater Res A 2010; 92:1510-7. [PMID: 19425046 DOI: 10.1002/jbm.a.32470] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Expression of vascular endothelial growth factor receptors (VEGFRs) increases in ischemic muscles, and thus, VEGFR could potentially be used as marker to detect ischemia. Here, we investigated whether (99m)Tc or Cy5.5-labeled chitosan-DC101 conjugates could identify VEGFR-2 overexpressed in ischemia. To this end, chitosan was conjugated with the DC101 antibody and Cy5.5, FITC, or the HYNIC chelator for (99m)Tc-labeling. Targeting of the conjugate was evaluated in vitro and in vivo through cell-binding studies and gamma/optical imaging, respectively. A hindlimb ischemic mouse model was surgically created by femoral artery occlusion. The chitosan-DC101 conjugates exhibited VEGFR-selective cell binding properties as determined by both confocal microscopy and flow cytometry. At postoperative times of 2, 12, and 24 h, (99m)Tc or Cy5.5-labeled chitosan-DC101 conjugates were intravenously injected into the mice, and gamma/optical imaging studies were conducted at 1 or 3 h. Both the gamma and optical imaging results indicated a significantly higher uptake in ischemic muscles when compared with the contralateral nonischemic muscle. Further, semiquantitative analysis of scintigraphic imaging data revealed that the ischemic to contralateral limb ratio was 4.5 +/- 0.25 at 24 h postoperation. Western blotting analysis confirmed VEGFR expression in the ischemic muscle. In conclusion, we believe that (99m)Tc or Cy5.5-labeled chitosan-DC101 conjugates have the potential to be useful as VEGFR-2-targeted imaging agents for monitoring ischemia.
Collapse
Affiliation(s)
- Chang-Moon Lee
- Department of Nuclear Medicine, Chonbuk National University Medical School and Hospital, Jeonju, Republic of Korea
| | | | | | | | | | | | | |
Collapse
|
41
|
Daniel C, Wagner A, Hohenstein B, Hugo C. Thrombospondin-2 therapy ameliorates experimental glomerulonephritis via inhibition of cell proliferation, inflammation, and TGF-beta activation. Am J Physiol Renal Physiol 2009; 297:F1299-309. [PMID: 19726547 DOI: 10.1152/ajprenal.00254.2009] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
We recently identified thrombospondin-2 (TSP-2) as an endogenous regulator of matrix remodelling and inflammation in experimental kidney disease by studying TSP-2-deficient mice. In this study, we asked whether systemic TSP-2 overexpression via thigh muscle transfection is able to ameliorate the time course of the anti-Thy1 glomerulonephritis model. After induction of anti-Thy1 nephritis, rats were transfected either with an overexpression plasmid for TSP-2 or lacZ as a control. Biopsies, urine, and blood samples were taken on days 1, 3, and 6 after disease induction. Muscular overexpression of TSP-2 reduced glomerular transforming growth factor (TGF)-beta activation and glomerular extracellular matrix formation as determined by collagen IV and fibronectin. In addition, activation of mesangial cells to the myofibroblast-like phenotype was also significantly decreased in TSP-2-overexpressing animals. TSP-2 overexpression inhibited both glomerular endothelial and mesangial cell proliferation, resulting in a reduced glomerular cell number and glomerular tuft area. The inflammatory response, as monitored by T cells and antigen-presenting cells, was reduced significantly by TSP-2 overexpression, but influx of macrophages was unchanged. These data demonstrate TSP-2 as a potential therapeutic agent to inhibit the glomerular proliferative and inflammatory response as well as TGF-beta activation and extracellular matrix accumulation in experimental mesangial proliferative glomerulonephritis.
Collapse
Affiliation(s)
- Christoph Daniel
- Department of Nephrology and Hypertension, University of Erlangen-Nuremberg, Erlangen, Germany.
| | | | | | | |
Collapse
|
42
|
Di Marco GS, Reuter S, Hillebrand U, Amler S, König M, Larger E, Oberleithner H, Brand E, Pavenstädt H, Brand M. The soluble VEGF receptor sFlt1 contributes to endothelial dysfunction in CKD. J Am Soc Nephrol 2009; 20:2235-45. [PMID: 19608702 DOI: 10.1681/asn.2009010061] [Citation(s) in RCA: 146] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Endothelial dysfunction contributes to the increased cardiovascular risk that accompanies CKD. We hypothesized that the soluble VEGF receptor 1 (sFlt-1), a VEGF antagonist, plays a role in endothelial dysfunction and decreased angiogenesis in CKD. We enrolled 130 patients with CKD stages 3 to 5 and 56 age- and gender-matched control patients. Plasma sFlt-1 levels were higher in patients with CKD and, after multivariate regression analyses, exclusively associated with renal function and levels of vWF, a marker of endothelial dysfunction. Compared with serum from control patients, both recombinant sFlt-1 and serum from patients with CKD had antiangiogenic activity in the chick chorioallantoic membrane (CAM) assay, induced endothelial cell apoptosis in vitro, and decreased nitric oxide generation in two different endothelial cell lines. Pretreating the sera with an antibody against sFlt-1 abrogated all of these effects. Furthermore, we observed increased sFlt1 levels in 5/6-nephrectomized rats compared with sham-operated animals. Finally, using real-time PCR and ELISA, we identified monocytes as a possible source of increased sFlt-1 in patients with CKD. Our findings show that excess sFlt-1 associates with endothelial dysfunction in CKD and suggest that increased sFlt-1 may predict cardiovascular risk in CKD.
Collapse
Affiliation(s)
- Giovana S Di Marco
- Department of Internal Medicine D, University Clinics Münster, Münster, Germany
| | | | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Long-term treatment of sirolimus but not cyclosporine ameliorates diabetic nephropathy in the rat. Transplantation 2009; 87:1290-9. [PMID: 19424027 DOI: 10.1097/tp.0b013e3181a192bd] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
BACKGROUND Not just de novo induction of diabetes mellitus, but also the progression of diabetic nephropathy may be enhanced under immunosuppressive therapy after organ transplantation. We evaluated whether sirolimus (SRL) or cyclosporine A (CsA) therapy would be a superior immunosuppressant in streptozotocin-induced diabetic nephropathy. METHODS Diabetes was induced by intravenous injection of streptozotozin (60 mg/kg body weight) in 26 male Sprague-Dawley rats. Eight days after diabetes induction, animals were divided into three groups, which were treated with placebo (n=8), SRL (n=9), or CsA (n=9). Six nondiabetic placebo-treated rats were included as controls. RESULTS After 19 weeks of diabetes, SRL significantly decreased fibrosis as assessed by periodic acid Schiff staining and by specific extracellular matrix proteins such as fibronectin and laminin at messenger RNA and protein level compared with the diabetic placebo group. SRL ameliorated renal inflammation, glomerular hypertrophy, and podocyte loss as indicated by morphometric and immunohistological analysis. SRL lowered expression and activity of glomerular transforming growth factor-beta1/2 and vascular endothelial growth factor, all of which are considered central cytokines in the pathogenesis of diabetic nephropathy. In contrast, calcineurin phosphatase inhibition through CsA did not ameliorate any of the features of diabetic nephropathy compared with placebo treatment but slightly aggravated glomerular fibrosis without affecting transforming growth factor-beta1/2 or vascular endothelial growth factor. CONCLUSION Compared with CsA, SRL by anti-inflammatory, antifibrotic, and podocyte-protective effects clearly seems to be the superior treatment of prevention or amelioration of diabetic nephropathy in the rat.
Collapse
|
44
|
Flisiński M, Brymora A, Elminowska-Wenda G, Bogucka J, Walasik K, Stefańska A, Odrowąż;-Sypniewska G, Manitius J. Influence of Different Stages of Experimental Chronic Kidney Disease on Rats Locomotor and Postural Skeletal Muscles Microcirculation. Ren Fail 2009; 30:443-51. [DOI: 10.1080/08860220801985694] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022] Open
|
45
|
Günthner T, Jankowski V, Kretschmer A, Nierhaus M, van der Giet M, Zidek W, Jankowski J. Endothelium and Vascular Smooth Muscle Cells in the Context of Uremia. Semin Dial 2009; 22:428-32. [DOI: 10.1111/j.1525-139x.2009.00594.x] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
|
46
|
Doi K, Leelahavanichkul A, Hu X, Sidransky KL, Zhou H, Qin Y, Eisner C, Schnermann J, Yuen PST, Star RA. Pre-existing renal disease promotes sepsis-induced acute kidney injury and worsens outcome. Kidney Int 2008; 74:1017-25. [PMID: 18633340 PMCID: PMC2614371 DOI: 10.1038/ki.2008.346] [Citation(s) in RCA: 89] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
While it is known that risk of death from sepsis is higher in patients with pre-existing chronic kidney disease its mechanism is unknown. To study this we established a two-stage mouse model where renal disease was first induced by folic acid injection followed by sub-lethal cecal ligation and puncture to induce sepsis. Septic mice with pre-existing renal disease had significantly higher mortality, serum creatinine, vascular permeability, plasma vascular endothelial growth factor (VEGF) levels, bacteremia, serum IL-10, splenocyte apoptosis and more severe septic shock when compared to septic mice without pre-existing disease. To evaluate the contribution of vascular and immunological dysfunction, we treated the folate-septic mice with soluble Flt-1 to bind VEGF and chloroquine to reduce splenocyte apoptosis. These treatments together resulted in a significant improvement in kidney injury, hemodynamics and survival. Our study shows that the sequential mouse model mimics human sepsis frequently complicated by pre-existing renal disease and might be useful in evaluating preventive and therapeutic strategies.
Collapse
Affiliation(s)
- Kent Doi
- National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland 20892-1268, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Wittmann S, Daniel C, Braun A, Vogelbacher R, Shimizu F, Kawachi H, Hugo C. The mTOR Inhibitor Everolimus Attenuates the Time Course of Chronic Anti-Thy1 Nephritis in the Rat. ACTA ACUST UNITED AC 2008; 108:e45-56. [DOI: 10.1159/000116112] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2007] [Accepted: 10/24/2007] [Indexed: 12/15/2022]
|
48
|
Constantinides CA, Tyritzis SI, Evangelou C, Kyroudi A, Liatsikos E, Karamessinis P, Zervas A, Pavlakis K. Vascular endothelial growth factor protein expression in a renal ablation rabbit model under prolonged warm and cold ischemia. Am J Nephrol 2007; 28:438-45. [PMID: 18097136 DOI: 10.1159/000112809] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2007] [Accepted: 11/16/2007] [Indexed: 11/19/2022]
Abstract
BACKGROUND/AIMS To establish a potential correlation between renal and systemic production of vascular endothelial growth factor (VEGF) protein after prolonged ischemia in a renal ablation model under normothermic and hypothermic conditions. METHODS 38 uninephrectomized New Zealand rabbits were divided into 5 groups. The rabbits of each group underwent partial nephrectomy under 90 and 60 min of warm and 90 and 120 min of cold ischemia, except for the sham group (S), which served as control. Serum creatinine (SCr) and blood-urea-nitrogen (BUN) levels were assessed. On the 15th postoperative day (POD), the animals were euthanized and the remaining kidneys were evaluated. VEGF immunohistochemistry and serum Western blot analysis were performed. RESULTS In comparison to the control group, groups 60W, 90C and 120C showed 1.6-, 1.14- and 1.75-fold decreases, respectively, while the production of VEGF was significantly declined by 7.4-fold in group 90W (p < 0.05). Immunohistochemistry revealed prominent VEGF staining in the above-mentioned three groups, while in group 90W staining was negative. Serum biochemistry and microscopic evaluation verified the same differentiation. CONCLUSION Renal and serum VEGF seem to have an analogous expression under conditions of prolonged ischemia. VEGF is overexpressed in hypothermic conditions compared to warm ischemia exceeding 60 min. Hypothermia can be more advantageous in a procedure applying prolonged ischemia.
Collapse
|
49
|
The mTOR Inhibitor Everolimus Induces Proteinuria and Renal Deterioration in the Remnant Kidney Model in the Rat. Transplantation 2007; 84:1492-9. [DOI: 10.1097/01.tp.0000282866.92367.99] [Citation(s) in RCA: 67] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
50
|
Ditting T, Hilgers KF, Stetter A, Linz P, Schönweiss C, Veelken R. Renal sympathetic nerves modulate erythropoietin plasma levels after transient hemorrhage in rats. Am J Physiol Renal Physiol 2007; 293:F1099-106. [PMID: 17634394 DOI: 10.1152/ajprenal.00267.2007] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
In contrast to other sympathetic outflow tracts, renal sympathetic nerve activity (RSNA) decreases in response to hypotensive hemorrhage. The functional significance of this "paradox" is not known. We tested the hypothesis that RSNA modulates renal perfusion and thus erythropoietin (EPO) release after transient hypotensive hemorrhage in anesthetized rats. Plasma EPO was measured before and after 30 min of transient hypotensive hemorrhage (i.e., -40 mmHg from mean baseline blood pressure, followed by reinfusion of shed blood) and 120 min thereafter in sham-denervated rats, and after renal denervation (DNX) or bilateral cervical vagotomy (VX) to abolish/blunt the RSNA decrease mediated by a cardiopulmonary reflex. RSNA, renal Doppler flow, renal vascular resistance (RVR), resistance index, and oxygen delivery/uptake (Do(2)/Vo(2)) were measured. RSNA decreased in intact animals (-40 +/- 5% from baseline, P < 0.05). This was blunted by VX. With intact nerves, EPO level did not increase. In DNX rats, EPO was increased at minute 120 (49 +/- 3 vs. 74 +/- 2 mU/ml; P < 0.05), in VX rats this (47 +/- 2 vs. 62 +/- 4 mU/ml; P < 0.05) was less pronounced. Do(2) in DNX rats was lower compared with intact and VX rats (0.25 +/- 0.04 vs. 0.51 +/- 0.06 and 0.54 +/- 0.05 ml O(2)/min; P < 0.05) due to lower Doppler flow and increased RVR. RVR and Do(2) were similar in intact and VX rats, but resistance index differed between all groups (0.70 +/- 0.02 vs. 0.78 +/- 0.02 vs. 0.85 +/- 0.02; P < 0.05, intact vs. VX vs. DNX), indicating differential reactivity of renal vasculature. Vo(2) was unaffected by VX and DNX. Renal sympathoinhibition during hypotensive hemorrhage might help to preserve sufficient oxygenation of renal tissue by modulation of hemodynamic mechanisms that act to adapt renal oxygen availability to demand.
Collapse
Affiliation(s)
- Tilmann Ditting
- Department of Nephrology, Friedrich-Alexander-Univ. Erlangen-Nürnberg, Erlangen, Germany
| | | | | | | | | | | |
Collapse
|