1
|
van Leent MMT, Duivenvoorden R. TGF-β: A Wrench in the Gears of Arteriovenous Fistula Maturation. Arterioscler Thromb Vasc Biol 2024; 44:2527-2529. [PMID: 39445425 DOI: 10.1161/atvbaha.124.321827] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2024]
Affiliation(s)
- Mandy M T van Leent
- BioMedical Engineering and Imaging Institute (M.M.T.v.L., R.D.), Icahn School of Medicine at Mount Sinai, New York
- Cardiovascular Research Institute (M.M.T.v.L.), Icahn School of Medicine at Mount Sinai, New York
- Department of Radiology (M.M.T.v.L), Icahn School of Medicine at Mount Sinai, New York
| | - Raphaël Duivenvoorden
- BioMedical Engineering and Imaging Institute (M.M.T.v.L., R.D.), Icahn School of Medicine at Mount Sinai, New York
- Department of Nephrology, Radboud University Medical Center, Nijmegen, the Netherlands (R.D.)
| |
Collapse
|
2
|
Battistella A, Linger M, Nguyen AT, Madukwe D, Roy-Chaudhury P, Tan W. Rebuilding vascular access: from the viewpoint of mechanics and materials. Front Bioeng Biotechnol 2024; 12:1448186. [PMID: 39295847 PMCID: PMC11409097 DOI: 10.3389/fbioe.2024.1448186] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Accepted: 08/22/2024] [Indexed: 09/21/2024] Open
Abstract
This review presents a comprehensive analysis of vascular access in hemodialysis, focusing on the current modalities, their associated challenges, and recent technological advancements. It closely examines the status of three primary types of vascular access: arteriovenous fistulas, arteriovenous grafts, and central venous catheters. The review delves into the complications and pathologies associated with these access types, emphasizing the mechanobiology-related pathogenesis of arteriovenous access. Furthermore, it explores recent clinical trials, biomaterials, and device innovations, highlighting novel pharmaceutical approaches, advanced materials, device designs, and cutting-edge technologies aimed at enhancing the efficacy, safety, and longevity of vascular access in hemodialysis. This synthesis of current knowledge and emerging trends underscores the dynamic evolution of vascular access strategies and their critical role in improving patient care in hemodialysis.
Collapse
Affiliation(s)
- Aurora Battistella
- Paul M. Rady Mechanical Engineering, University of Colorado at Boulder, Boulder, CO, United States
| | - Morgan Linger
- Paul M. Rady Mechanical Engineering, University of Colorado at Boulder, Boulder, CO, United States
- Department of Biomedical Engineering, University of Colorado at Boulder, Boulder, CO, United States
| | - Anh Thy Nguyen
- Paul M. Rady Mechanical Engineering, University of Colorado at Boulder, Boulder, CO, United States
| | - David Madukwe
- Paul M. Rady Mechanical Engineering, University of Colorado at Boulder, Boulder, CO, United States
| | - Prabir Roy-Chaudhury
- Department of Medicine, University of North Carolina Kidney Center, Chapel Hill, NC, United States
- WG (Bill) Hefner VA Medical Center, Salisbury, NC, United States
| | - Wei Tan
- Paul M. Rady Mechanical Engineering, University of Colorado at Boulder, Boulder, CO, United States
- Department of Biomedical Engineering, University of Colorado at Boulder, Boulder, CO, United States
| |
Collapse
|
3
|
Yan R, Song A, Zhang C. The Pathological Mechanisms and Therapeutic Molecular Targets in Arteriovenous Fistula Dysfunction. Int J Mol Sci 2024; 25:9519. [PMID: 39273465 PMCID: PMC11395150 DOI: 10.3390/ijms25179519] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2024] [Revised: 08/28/2024] [Accepted: 08/30/2024] [Indexed: 09/15/2024] Open
Abstract
The number of patients with end-stage renal disease (ESRD) requiring hemodialysis is increasing worldwide. Although arteriovenous fistula (AVF) is the best and most important vascular access (VA) for hemodialysis, its primary maturation failure rate is as high as 60%, which seriously endangers the prognosis of hemodialysis patients. After AVF establishment, the venous outflow tract undergoes hemodynamic changes, which are translated into intracellular signaling pathway cascades, resulting in an outward and inward remodeling of the vessel wall. Outward remodeling refers to the thickening of the vessel wall and the dilation of the lumen to accommodate the high blood flow in the AVF, while inward remodeling is mainly characterized by intimal hyperplasia. More and more studies have shown that the two types of remodeling are closely related in the occurrence and development of, and jointly determining the final fate of, AVF. Therefore, it is essential to investigate the underlying mechanisms involved in outward and inward remodeling for identifying the key targets in alleviating AVF dysfunction. In this review, we summarize the current clinical diagnosis, monitoring, and treatment techniques for AVF dysfunction and discuss the possible pathological mechanisms related to improper outward and inward remodeling in AVF dysfunction, as well as summarize the similarities and differences between the two remodeling types in molecular mechanisms. Finally, the representative therapeutic targets of potential clinical values are summarized.
Collapse
Affiliation(s)
- Ruiwei Yan
- Department of Nephrology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Anni Song
- Department of Nephrology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Chun Zhang
- Department of Nephrology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| |
Collapse
|
4
|
Zhui L, Yuling C, Hansheng W, Xiangjie L. Omentin reduces venous neointimal hyperplasia in arteriovenous fistula through hypoxia-inducible factor-1 alpha inhibition. Microvasc Res 2024; 154:104688. [PMID: 38640999 DOI: 10.1016/j.mvr.2024.104688] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Revised: 03/22/2024] [Accepted: 04/14/2024] [Indexed: 04/21/2024]
Abstract
Arteriovenous fistula (AVF) failure often involves venous neointimal hyperplasia (VNH) driven by elevated hypoxia-inducible factor-1 alpha (HIF-1α) in the venous wall. Omentin, known for its anti-inflammatory and anti-hyperplasia properties, has an uncertain role in early AVF failure. This study investigates omentin's impact on VNH using a chronic renal failure (CRF) rabbit model. The CRF rabbit model of AVF received omentin-expressing adenoviral vector or control β-gal vector to assess omentin's effects on VNH. Human vascular smooth muscle cells (HVSMCs), stimulated with tumor necrosis factor-α (TNF-α), were exposed to recombinant human omentin (Rh-OMT) to study its influence on cell proliferation and migration. The AMP-activated protein kinase (AMPK) inhibitor compound C and the mammalian target of rapamycin (mTOR) activator MHY1485 were employed to explore omentin's mechanisms in VNH reduction through HIF-1α inhibition. Omentin treatment reduced VNH in CRF rabbits, concomitant with HIF-1α down-regulation and the suppression of downstream factors, including vascular endothelial growth factor and matrix metalloproteinases. Rh-OMT inhibited TNF-α-induced HVSMC proliferation and migration by modulating both cell cycle and cell adhesion proteins. Additionally, omentin reduced HIF-1α expression through the AMPK/mTOR pathway activation. Notably, the blockade of AMPK/mTOR signaling reversed omentin-mediated inhibition of VNH, cell proliferation, and migration, both in vivo and in vitro. In conclusion, omentin mitigates VNH post-AVF creation by restraining HIF-1α via AMPK/mTOR signaling. Strategies boosting circulating omentin levels may offer promise in averting AVF failure.
Collapse
MESH Headings
- Animals
- Hyperplasia
- Neointima
- Hypoxia-Inducible Factor 1, alpha Subunit/metabolism
- Hypoxia-Inducible Factor 1, alpha Subunit/genetics
- Cytokines/metabolism
- Rabbits
- Humans
- GPI-Linked Proteins/metabolism
- GPI-Linked Proteins/pharmacology
- GPI-Linked Proteins/genetics
- Disease Models, Animal
- Cell Proliferation/drug effects
- Signal Transduction
- Myocytes, Smooth Muscle/pathology
- Myocytes, Smooth Muscle/metabolism
- Myocytes, Smooth Muscle/drug effects
- Lectins/pharmacology
- Lectins/metabolism
- Cell Movement/drug effects
- Muscle, Smooth, Vascular/pathology
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/drug effects
- AMP-Activated Protein Kinases/metabolism
- Cells, Cultured
- Arteriovenous Shunt, Surgical/adverse effects
- Male
- Kidney Failure, Chronic/pathology
- TOR Serine-Threonine Kinases/metabolism
- Graft Occlusion, Vascular/pathology
- Graft Occlusion, Vascular/prevention & control
- Graft Occlusion, Vascular/metabolism
- Graft Occlusion, Vascular/physiopathology
- Jugular Veins/pathology
- Jugular Veins/metabolism
- Jugular Veins/transplantation
Collapse
Affiliation(s)
- Li Zhui
- Department of Vascular Surgery, the First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China.
| | - Chen Yuling
- Department of Vascular Surgery, the First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Wang Hansheng
- Department of Vascular Surgery, the First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Li Xiangjie
- Department of Vascular Surgery, the First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| |
Collapse
|
5
|
Ohashi Y, Protack CD, Aoyagi Y, Gonzalez L, Thaxton C, Zhang W, Kano M, Bai H, Yatsula B, Alves R, Hoshina K, Schneider EB, Long X, Perry RJ, Dardik A. Heterogeneous gene expression during early arteriovenous fistula remodeling suggests that downregulation of metabolism predicts adaptive venous remodeling. Sci Rep 2024; 14:13287. [PMID: 38858395 PMCID: PMC11164895 DOI: 10.1038/s41598-024-64075-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Accepted: 06/05/2024] [Indexed: 06/12/2024] Open
Abstract
Clinical outcomes of arteriovenous fistulae (AVF) for hemodialysis remain inadequate since biological mechanisms of AVF maturation and failure are still poorly understood. Aortocaval fistula creation (AVF group) or a sham operation (sham group) was performed in C57BL/6 mice. Venous limbs were collected on postoperative day 7 and total RNA was extracted for high throughput RNA sequencing and bioinformatic analysis. Genes in metabolic pathways were significantly downregulated in the AVF, whereas significant sex differences were not detected. Since gene expression patterns among the AVF group were heterogenous, the AVF group was divided into a 'normal' AVF (nAVF) group and an 'outliers' (OUT) group. The gene expression patterns of the nAVF and OUT groups were consistent with previously published data showing venous adaptive remodeling, whereas enrichment analyses showed significant upregulation of metabolism, inflammation and coagulation in the OUT group compared to the nAVF group, suggesting the heterogeneity during venous remodeling reflects early gene expression changes that may correlate with AVF maturation or failure. Early detection of these processes may be a translational strategy to predict fistula failure and reduce patient morbidity.
Collapse
Affiliation(s)
- Yuichi Ohashi
- Vascular Biology and Therapeutics Program, Yale School of Medicine, New Haven, CT, USA
- Department of Surgery, Yale School of Medicine, New Haven, CT, USA
- Division of Vascular Surgery, Department of Surgery, The University of Tokyo, Tokyo, Japan
| | - Clinton D Protack
- Vascular Biology and Therapeutics Program, Yale School of Medicine, New Haven, CT, USA
- Department of Surgery, Yale School of Medicine, New Haven, CT, USA
| | - Yukihiko Aoyagi
- Vascular Biology and Therapeutics Program, Yale School of Medicine, New Haven, CT, USA
- Department of Surgery, Yale School of Medicine, New Haven, CT, USA
- Department of Surgery and Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Luis Gonzalez
- Vascular Biology and Therapeutics Program, Yale School of Medicine, New Haven, CT, USA
- Department of Surgery, Yale School of Medicine, New Haven, CT, USA
| | - Carly Thaxton
- Vascular Biology and Therapeutics Program, Yale School of Medicine, New Haven, CT, USA
- Department of Surgery, Yale School of Medicine, New Haven, CT, USA
| | - Weichang Zhang
- Vascular Biology and Therapeutics Program, Yale School of Medicine, New Haven, CT, USA
- Department of Surgery, Yale School of Medicine, New Haven, CT, USA
| | - Masaki Kano
- Vascular Biology and Therapeutics Program, Yale School of Medicine, New Haven, CT, USA
- Department of Surgery, Yale School of Medicine, New Haven, CT, USA
- Department of Cardiovascular Surgery, Tokyo Medical University, Tokyo, Japan
| | - Hualong Bai
- Vascular Biology and Therapeutics Program, Yale School of Medicine, New Haven, CT, USA
- Department of Surgery, Yale School of Medicine, New Haven, CT, USA
| | - Bogdan Yatsula
- Vascular Biology and Therapeutics Program, Yale School of Medicine, New Haven, CT, USA
- Department of Surgery, Yale School of Medicine, New Haven, CT, USA
| | - Rafael Alves
- Vascular Biology and Therapeutics Program, Yale School of Medicine, New Haven, CT, USA
- Department of Surgery, Yale School of Medicine, New Haven, CT, USA
| | - Katsuyuki Hoshina
- Division of Vascular Surgery, Department of Surgery, The University of Tokyo, Tokyo, Japan
| | - Eric B Schneider
- Department of Surgery, Center for Health Services and Outcomes Research, Yale School of Medicine, New Haven, CT, USA
| | - Xiaochun Long
- Vascular Biology Center, Medical College of Georgia at Augusta University, Augusta, GA, USA
| | - Rachel J Perry
- Department of Internal Medicine, Yale School of Medicine, New Haven, CT, USA
- Department of Cellular and Molecular Physiology, Yale School of Medicine, New Haven, CT, USA
| | - Alan Dardik
- Vascular Biology and Therapeutics Program, Yale School of Medicine, New Haven, CT, USA.
- Department of Surgery, Yale School of Medicine, New Haven, CT, USA.
- Department of Cellular and Molecular Physiology, Yale School of Medicine, New Haven, CT, USA.
- Surgical Service, Veterans Affairs Connecticut Healthcare System, West Haven, CT, USA.
- Yale School of Medicine, 10 Amistad Street, Room 437, PO Box 208089, New Haven, CT, 06520-8089, USA.
| |
Collapse
|
6
|
Bai H, Varsanik MA, Thaxton C, Ohashi Y, Gonzalez L, Zhang W, Aoyagi Y, Kano M, Yatsula B, Li Z, Pocivavsek L, Dardik A. Disturbed flow in the juxta-anastomotic area of an arteriovenous fistula correlates with endothelial loss, acute thrombus formation, and neointimal hyperplasia. Am J Physiol Heart Circ Physiol 2024; 326:H1446-H1461. [PMID: 38578237 PMCID: PMC11380968 DOI: 10.1152/ajpheart.00054.2024] [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: 01/31/2024] [Revised: 02/27/2024] [Accepted: 03/28/2024] [Indexed: 04/06/2024]
Abstract
Clinical failure of arteriovenous neointimal hyperplasia (NIH) fistulae (AVF) is frequently due to juxta-anastomotic NIH (JANIH). Although the mouse AVF model recapitulates human AVF maturation, previous studies focused on the outflow vein distal to the anastomosis. We hypothesized that the juxta-anastomotic area (JAA) has increased NIH compared with the outflow vein. AVF was created in C57BL/6 mice without or with chronic kidney disease (CKD). Temporal and spatial changes of the JAA were examined using histology and immunofluorescence. Computational techniques were used to model the AVF. RNA-seq and bioinformatic analyses were performed to compare the JAA with the outflow vein. The jugular vein to carotid artery AVF model was created in Wistar rats. The neointima in the JAA shows increased volume compared with the outflow vein. Computational modeling shows an increased volume of disturbed flow at the JAA compared with the outflow vein. Endothelial cells are immediately lost from the wall contralateral to the fistula exit, followed by thrombus formation and JANIH. Gene Ontology (GO) enrichment analysis of the 1,862 differentially expressed genes (DEG) between the JANIH and the outflow vein identified 525 overexpressed genes. The rat jugular vein to carotid artery AVF showed changes similar to the mouse AVF. Disturbed flow through the JAA correlates with rapid endothelial cell loss, thrombus formation, and JANIH; late endothelialization of the JAA channel correlates with late AVF patency. Early thrombus formation in the JAA may influence the later development of JANIH.NEW & NOTEWORTHY Disturbed flow and focal endothelial cell loss in the juxta-anastomotic area of the mouse AVF colocalizes with acute thrombus formation followed by late neointimal hyperplasia. Differential flow patterns between the juxta-anastomotic area and the outflow vein correlate with differential expression of genes regulating coagulation, proliferation, collagen metabolism, and the immune response. The rat jugular vein to carotid artery AVF model shows changes similar to the mouse AVF model.
Collapse
MESH Headings
- Animals
- Neointima
- Hyperplasia
- Arteriovenous Shunt, Surgical
- Thrombosis/physiopathology
- Thrombosis/pathology
- Thrombosis/genetics
- Thrombosis/etiology
- Thrombosis/metabolism
- Mice, Inbred C57BL
- Rats, Wistar
- Male
- Jugular Veins/metabolism
- Jugular Veins/pathology
- Jugular Veins/physiopathology
- Disease Models, Animal
- Carotid Arteries/pathology
- Carotid Arteries/physiopathology
- Carotid Arteries/metabolism
- Carotid Arteries/surgery
- Mice
- Rats
- Regional Blood Flow
- Endothelium, Vascular/metabolism
- Endothelium, Vascular/physiopathology
- Endothelium, Vascular/pathology
- Renal Insufficiency, Chronic/pathology
- Renal Insufficiency, Chronic/physiopathology
- Renal Insufficiency, Chronic/genetics
- Renal Insufficiency, Chronic/metabolism
- Endothelial Cells/metabolism
- Endothelial Cells/pathology
Collapse
Affiliation(s)
- Hualong Bai
- Vascular Biology and Therapeutics Program, Yale School of Medicine, New Haven, Connecticut, United States
- Department of Surgery, Yale School of Medicine, New Haven, Connecticut, United States
| | - M Alyssa Varsanik
- Section of Vascular Surgery, Department of Surgery, University of Chicago Medicine, Chicago, Illinois, United States
| | - Carly Thaxton
- Vascular Biology and Therapeutics Program, Yale School of Medicine, New Haven, Connecticut, United States
- Department of Surgery, Yale School of Medicine, New Haven, Connecticut, United States
| | - Yuichi Ohashi
- Vascular Biology and Therapeutics Program, Yale School of Medicine, New Haven, Connecticut, United States
- Department of Surgery, Yale School of Medicine, New Haven, Connecticut, United States
| | - Luis Gonzalez
- Vascular Biology and Therapeutics Program, Yale School of Medicine, New Haven, Connecticut, United States
- Department of Surgery, Yale School of Medicine, New Haven, Connecticut, United States
| | - Weichang Zhang
- Vascular Biology and Therapeutics Program, Yale School of Medicine, New Haven, Connecticut, United States
- Department of Surgery, Yale School of Medicine, New Haven, Connecticut, United States
| | - Yukihiko Aoyagi
- Vascular Biology and Therapeutics Program, Yale School of Medicine, New Haven, Connecticut, United States
- Department of Surgery, Yale School of Medicine, New Haven, Connecticut, United States
| | - Masaki Kano
- Vascular Biology and Therapeutics Program, Yale School of Medicine, New Haven, Connecticut, United States
- Department of Surgery, Yale School of Medicine, New Haven, Connecticut, United States
| | - Bogdan Yatsula
- Vascular Biology and Therapeutics Program, Yale School of Medicine, New Haven, Connecticut, United States
- Department of Surgery, Yale School of Medicine, New Haven, Connecticut, United States
| | - Zhuo Li
- Vascular Biology and Therapeutics Program, Yale School of Medicine, New Haven, Connecticut, United States
- Department of Surgery, Yale School of Medicine, New Haven, Connecticut, United States
| | - Luka Pocivavsek
- Section of Vascular Surgery, Department of Surgery, University of Chicago Medicine, Chicago, Illinois, United States
| | - Alan Dardik
- Vascular Biology and Therapeutics Program, Yale School of Medicine, New Haven, Connecticut, United States
- Department of Surgery, Yale School of Medicine, New Haven, Connecticut, United States
- Department of Cellular and Molecular Physiology, Yale University; New Haven, Connecticut, United States
- Department of Surgery, VA Connecticut Healthcare Systems, West Haven, Connecticut, United States
| |
Collapse
|
7
|
Dardik A. A surgeon-scientist's approach to improving arteriovenous fistula patency. JVS Vasc Sci 2024; 5:100207. [PMID: 38975292 PMCID: PMC11225657 DOI: 10.1016/j.jvssci.2024.100207] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Accepted: 05/06/2024] [Indexed: 07/09/2024] Open
Affiliation(s)
- Alan Dardik
- Vascular Biology and Therapeutics Program, Division of Vascular Surgery and Endovascular Therapy, Yale School of Medicine, New Haven, CT
- Department of Surgery, Yale School of Medicine, New Haven, CT
- Department of Cellular and Molecular Physiology, Yale School of Medicine, New Haven, CT
- Department of Surgery, VA Connecticut Healthcare System, West Haven, CT
| |
Collapse
|
8
|
Liu SJ, Cao YL, Zhang C. Hirudin in the Treatment of Chronic Kidney Disease. Molecules 2024; 29:1029. [PMID: 38474541 DOI: 10.3390/molecules29051029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Revised: 02/13/2024] [Accepted: 02/26/2024] [Indexed: 03/14/2024] Open
Abstract
Chronic kidney disease (CKD) is a common public health concern. The global burden of CKD is increasing due to the high morbidity and mortality associated with it, indicating the shortcomings of therapeutic drugs at present. Renal fibrosis is the common pathology of CKD, which is characterized by glomerulosclerosis, renal tubular atrophy, and renal interstitial fibrosis. Natural hirudin is an active ingredient extracted from Hirudo medicinalis, which has been found to be the strongest natural specific inhibitor of thrombin. Evidence based on pharmacological data has shown that hirudin has important protective effects in CKD against diabetic nephrology, nephrotic syndrome, and renal interstitial fibrosis. The mechanisms of hirudin in treating CKD are mainly related to inhibiting the inflammatory response, preventing apoptosis of intrinsic renal cells, and inhibiting the interactions between thrombin and protease-activated receptors. In this review, we summarize the function and beneficial properties of hirudin for the treatment of CKD, and its underlying mechanisms.
Collapse
Affiliation(s)
- Sai-Ji Liu
- Department of Nephrology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Yi-Ling Cao
- Department of Nephrology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Chun Zhang
- Department of Nephrology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| |
Collapse
|
9
|
Liu J, Zhang D, Brahmandam A, Matsubara Y, Gao M, Tian J, Liu B, Shu C, Dardik A. Bioinformatics identifies predictors of arteriovenous fistula maturation. J Vasc Access 2024; 25:172-186. [PMID: 35686495 PMCID: PMC9734286 DOI: 10.1177/11297298221102298] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Arteriovenous fistulae (AVF) are the preferred access for hemodialysis but still have poor rates of maturation and patency limiting their clinical use. The underlying mechanisms of venous remodeling remain poorly understood, and only limited numbers of unbiased approaches have been reported. METHODS Biological Gene Ontology (GO) term enrichment analysis and differentially expressed genes (DEG) analysis were performed for three AVF datasets. A microRNA enrichment analysis and L1000CDS2 query were performed to identify factors predicting AVF patency. RESULTS The inflammatory and immune responses were activated during both early and late phases of AVF maturation, with upregulation of neutrophil and leukocyte regulation, cytokine production, and cytokine-mediated signaling. In men with failed AVF, negative regulation of myeloid-leukocyte differentiation and regulation of macrophage activation were significantly upregulated. Compared to non-diabetic patients, diabetic patients had significantly reduced immune response-related enrichment such as cell activation in immune response, regulation of immune-effector process, and positive regulation of defense response; in addition, diabetic patients showed no enrichment of the immune response-regulating signaling pathway. CONCLUSIONS These data show coordinated, and differential regulation of genes associated with AVF maturation, and different patterns of several pathways are associated with sex differences in AVF failure. Inflammatory and immune responses are activated during AVF maturation and diabetes may impair AVF maturation by altering these responses. These findings suggest several novel molecular targets to improve sex specific AVF maturation.
Collapse
Affiliation(s)
- Jia Liu
- Department of Vascular Surgery, the Second Xiangya Hospital, Central South University, Changsha, Hunan, People’s Republic of China
- The Vascular Biology and Therapeutics Program, Yale School of Medicine, New Haven, CT, USA
- Division of Vascular Surgery, Department of Surgery, Yale School of Medicine, New Haven, CT, USA
| | - Dingyao Zhang
- The Stem Cell Center, Yale School of Medicine, New Haven, CT, USA
| | - Anand Brahmandam
- The Vascular Biology and Therapeutics Program, Yale School of Medicine, New Haven, CT, USA
- Division of Vascular Surgery, Department of Surgery, Yale School of Medicine, New Haven, CT, USA
| | - Yutaka Matsubara
- The Vascular Biology and Therapeutics Program, Yale School of Medicine, New Haven, CT, USA
- Division of Vascular Surgery, Department of Surgery, Yale School of Medicine, New Haven, CT, USA
- The Department of Surgery and Sciences, Kyushu University, Fukuoka, Japan
| | - Mingjie Gao
- The Vascular Biology and Therapeutics Program, Yale School of Medicine, New Haven, CT, USA
- Division of Vascular Surgery, Department of Surgery, Yale School of Medicine, New Haven, CT, USA
| | - Jingru Tian
- The Stem Cell Center, Yale School of Medicine, New Haven, CT, USA
| | - Bing Liu
- The Vascular Biology and Therapeutics Program, Yale School of Medicine, New Haven, CT, USA
| | - Chang Shu
- Department of Vascular Surgery, the Second Xiangya Hospital, Central South University, Changsha, Hunan, People’s Republic of China
- State Key Laboratory of Cardiovascular Disease, Center of Vascular Surgery, Fuwai Hospital, National Center for Cardiovascular Disease, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Alan Dardik
- The Vascular Biology and Therapeutics Program, Yale School of Medicine, New Haven, CT, USA
- Division of Vascular Surgery, Department of Surgery, Yale School of Medicine, New Haven, CT, USA
- Department of Cellular and Molecular Physiology, Yale School of Medicine, New Haven, CT, USA
| |
Collapse
|
10
|
Liu L, Gao J, Tang Y, Guo G, Gan H. Increased expression of the P2Y 12 receptor is involved in the failure of autogenous arteriovenous fistula caused by stenosis. Ren Fail 2023; 45:2278314. [PMID: 38532720 PMCID: PMC11073481 DOI: 10.1080/0886022x.2023.2278314] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2023] [Accepted: 10/27/2023] [Indexed: 03/28/2024] Open
Abstract
OBJECTIVE This study investigated the role of the P2Y12 receptor in autogenous arteriovenous fistula (AVF) failure resulting from stenosis. METHODS Stenotic venous tissues and blood samples were obtained from patients with end-stage renal disease (ESRD) together with AVF stenosis, while venous tissues and blood samples were collected from patients with ESRD undergoing initial AVF surgery as controls. Immunohistochemistry and/or immunofluorescence techniques were utilized to assess the expression of P2Y12, transforming growth factor-β1 (TGF-β1), monocyte chemotactic protein 1 (MCP-1), and CD68 in the venous tissues. The expression levels of P2Y12, TGFβ1, and MCP-1 were quantified using quantitative reverse transcription-polymerase chain reaction and western blot analyses. Double and triple immunofluorescence staining was performed to precisely localize the cellular localization of P2Y12 expression. RESULTS Expression levels of P2Y12, TGFβ1, MCP-1, and CD68 were significantly higher in stenotic AVF venous tissues than in the control group tissues. Double and triple immunofluorescence staining of stenotic AVF venous tissues indicated that P2Y12 was predominantly expressed in α-SMA-positive vascular smooth muscle cells (VSMCs) and, to a lesser extent, in CD68-positive macrophages, with limited expression in CD31-positive endothelial cells. Moreover, a subset of macrophage-like VSMCs expressing P2Y12 were observed in both stenotic AVF venous tissues and control venous tissues. Additionally, a higher number of P2Y12+/TGF-β1+ double-positive cells were identified in stenotic AVF venous tissues than in the control group tissues. CONCLUSION Increased expression of P2Y12 in stenotic AVF venous tissues of patients with ESRD suggests its potential involvement in the pathogenesis of venous stenosis within AVFs.
Collapse
Affiliation(s)
- Lei Liu
- Department of Nephrology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Department of Nephrology, Chongqing University Three Gorges Hospital, Chongqing, China
- Department of Nephrology, Chongqing Three Gorges Central Hospital, Chongqing, China
| | - Jianya Gao
- Department of Nephrology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Department of Nephrology, Chongqing University Three Gorges Hospital, Chongqing, China
- Department of Nephrology, Chongqing Three Gorges Central Hospital, Chongqing, China
| | - Yuewu Tang
- Department of Nephrology, Chongqing University Three Gorges Hospital, Chongqing, China
- Department of Nephrology, Chongqing Three Gorges Central Hospital, Chongqing, China
| | - Guangfeng Guo
- Department of Nephrology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Hua Gan
- Department of Nephrology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| |
Collapse
|
11
|
Satam K, Ohashi Y, Thaxton C, Gonzalez L, Setia O, Bai H, Aoyagi Y, Xie Y, Zhang W, Yatsula B, Martin KA, Cai Y, Dardik A. Sex hormones impact early maturation and immune response in the arteriovenous fistula mouse model. Am J Physiol Heart Circ Physiol 2023; 325:H77-H88. [PMID: 37145957 PMCID: PMC10243550 DOI: 10.1152/ajpheart.00049.2023] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/26/2023] [Revised: 04/19/2023] [Accepted: 05/04/2023] [Indexed: 05/07/2023]
Abstract
Arteriovenous fistulae (AVF) fail to mature more frequently in female patients compared with male patients, leading to inferior outcomes and decreased utilization. Since our mouse AVF model recapitulates sex differences in human AVF maturation, we hypothesized that sex hormones mediate these differences during AVF maturation. C57BL/6 mice (9-11 wk) were treated with aortocaval AVF surgery and/or gonadectomy. AVF hemodynamics were measured via ultrasound (days 0-21). Blood was collected for FACS and tissue for immunofluorescence and ELISA (days 3 and 7); wall thickness was assessed by histology (day 21). Inferior vena cava shear stress was higher in male mice (P = 0.0028) after gonadectomy, and they had increased wall thickness (22.0 ± 1.8 vs. 12.7 ± 1.2 µm; P < 0.0001). Conversely, female mice had decreased wall thickness (6.8 ± 0.6 vs. 15.3 ± 0.9 µm; P = 0.0002). Intact female mice had higher proportions of circulating CD3+ T cells on day 3 (P = 0.0043), CD4+ (P = 0.0003) and CD8+ T cells (P = 0.005) on day 7, and CD11b+ monocytes on day 3 (P = 0.0046). After gonadectomy, these differences disappeared. In intact female mice, CD3+ T cells (P = 0.025), CD4+ T cells (P = 0.0178), CD8+ T cells (P = 0.0571), and CD68+ macrophages (P = 0.0078) increased in the fistula wall on days 3 and 7. This disappeared after gonadectomy. Furthermore, female mice had higher IL-10 (P = 0.0217) and TNF-α (P = 0.0417) levels in their AVF walls than male mice. Sex hormones mediate AVF maturation, suggesting that hormone receptor signaling may be a target to improve AVF maturation.NEW & NOTEWORTHY After arteriovenous fistula creation, females have lower rates of maturation and higher rates of failure than males. In a mouse model of venous adaptation that recapitulates human fistula maturation, sex hormones may be mechanisms of the sexual dimorphism: testosterone is associated with reduced shear stress, whereas estrogen is associated with increased immune cell recruitment. Modulating sex hormones or downstream effectors suggests sex-specific therapies and could address disparities in sex differences in clinical outcomes.
Collapse
Affiliation(s)
- Keyuree Satam
- Yale School of Medicine, New Haven, Connecticut, United States
- Vascular Biology and Therapeutics Program, Yale School of Medicine, New Haven, Connecticut, United States
| | - Yuichi Ohashi
- Vascular Biology and Therapeutics Program, Yale School of Medicine, New Haven, Connecticut, United States
| | - Carly Thaxton
- Yale School of Medicine, New Haven, Connecticut, United States
- Vascular Biology and Therapeutics Program, Yale School of Medicine, New Haven, Connecticut, United States
- Department of Surgery, Department of Veterans Affairs Connecticut Healthcare System, West Haven, Connecticut, United States
| | - Luis Gonzalez
- Yale School of Medicine, New Haven, Connecticut, United States
- Vascular Biology and Therapeutics Program, Yale School of Medicine, New Haven, Connecticut, United States
| | - Ocean Setia
- Vascular Biology and Therapeutics Program, Yale School of Medicine, New Haven, Connecticut, United States
- Division of Vascular Surgery and Endovascular Therapy, Department of Surgery, Yale School of Medicine, New Haven, Connecticut, United States
| | - Hualong Bai
- Vascular Biology and Therapeutics Program, Yale School of Medicine, New Haven, Connecticut, United States
| | - Yukihiko Aoyagi
- Vascular Biology and Therapeutics Program, Yale School of Medicine, New Haven, Connecticut, United States
| | - Yangzhouyun Xie
- Division of Vascular Surgery and Endovascular Therapy, Department of Surgery, Yale School of Medicine, New Haven, Connecticut, United States
| | - Weichang Zhang
- Vascular Biology and Therapeutics Program, Yale School of Medicine, New Haven, Connecticut, United States
| | - Bogdan Yatsula
- Vascular Biology and Therapeutics Program, Yale School of Medicine, New Haven, Connecticut, United States
| | - Kathleen A Martin
- Section of Cardiovascular Medicine, Yale School of Medicine, New Haven, Connecticut, United States
| | - Yujun Cai
- Division of Vascular Surgery and Endovascular Therapy, Department of Surgery, Yale School of Medicine, New Haven, Connecticut, United States
| | - Alan Dardik
- Vascular Biology and Therapeutics Program, Yale School of Medicine, New Haven, Connecticut, United States
- Department of Surgery, Department of Veterans Affairs Connecticut Healthcare System, West Haven, Connecticut, United States
- Division of Vascular Surgery and Endovascular Therapy, Department of Surgery, Yale School of Medicine, New Haven, Connecticut, United States
| |
Collapse
|
12
|
Sex Differences in Arteriovenous Fistula Failure: Insights from Bioinformatics Analysis. J Cardiovasc Dev Dis 2022; 10:jcdd10010003. [PMID: 36661898 PMCID: PMC9862581 DOI: 10.3390/jcdd10010003] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2022] [Revised: 12/06/2022] [Accepted: 12/14/2022] [Indexed: 12/25/2022] Open
Abstract
(1) Background: Arteriovenous fistulas (AVFs) are the preferred access for hemodialysis. Unfortunately, about 60% of patients, especially female patients, fail to receive normal dialysis within one year after surgery because of AVF failure. However, the underlying mechanisms caused by sex differences in AVF failure remain unclear. (2) Methods: We performed analysis of DEGs and functional analysis with the dataset GSE119296 to reveal the biology underlying AVF failure. Immune responses were calculated using CIBERSORT. A protein-protein interaction network and hub gene were constructed using STRING and stepwise identification of potential drugs was performed online. (3) Results: Functional analysis showed that extracellular matrix reprogramming and PI3K-AKT pathway enrichment were significant in both male and female patients. COL1A1 was the hub gene in male patients, whereas CDK1 was the hub gene in female patients. Immune responses including γδ-T cells and mast cells are activated in female patients while no significant differences were noted in the male group. (4) Conclusions: In this study, we used a series of mature and recognized bioinformatic strategies to determine the following items: (1) Reveal the pathogenesis of AVF failure through HUB genes and signaling pathways between the different sexes. (2) Determine the relationship between sex differences in AVF failure and immune abnormalities. (3) Search for relevant sex-specific drugs targeting AVF failure.
Collapse
|
13
|
Gao M, Gao X, Taniguchi R, Brahmandam A, Matsubara Y, Liu J, Liu H, Zhang W, Dardik A. Sex differences in arterial identity correlate with neointimal hyperplasia after balloon injury. Mol Biol Rep 2022; 49:8301-8315. [PMID: 35715609 PMCID: PMC9463237 DOI: 10.1007/s11033-022-07644-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Accepted: 05/25/2022] [Indexed: 11/25/2022]
Abstract
BACKGROUND Endovascular treatment of atherosclerotic arterial disease exhibits sex differences in clinical outcomes including restenosis. However, sex-specific differences in arterial identity during arterial remodeling have not been described. We hypothesized that sex differences in expression of the arterial determinant erythropoietin-producing hepatocellular receptor interacting protein (Ephrin)-B2 occur during neointimal proliferation and arterial remodeling. METHODS AND RESULTS Carotid balloon injury was performed in female and male Sprague-Dawley rats without or 14 days after gonadectomy; the left common carotid artery was injured and the right carotid artery in the same animal was used as an uninjured control. Arterial hemodynamics were evaluated in vivo using ultrasonography pre-procedure and post-procedure at 7 and 14 days and wall composition examined using histology, immunofluorescence and Western blot at 14 days after balloon injury. There were no significant baseline sex differences. 14 days after balloon injury, there was decreased neointimal thickness in female rats with decreased smooth muscle cell proliferation and decreased type I and III collagen deposition, as well as decreased TNFα- or iNOS-positive CD68+ cells and increased CD206- or TGM2-positive CD68+ cells. Female rats also showed less immunoreactivity of VEGF-A, NRP1, phosphorylated EphrinB2, and increased Notch1, as well as decreased phosphorylated Akt1, p38 and ERK1/2. These differences were not present in rats pretreated with gonadectomy. CONCLUSIONS Decreased neointimal thickness in female rats after carotid balloon injury is associated with altered arterial identity that is dependent on intact sex hormones. Alteration of arterial identity may be a mechanism of sex differences in neointimal proliferation after arterial injury.
Collapse
Affiliation(s)
- Mingjie Gao
- Department of Vascular Ultrasonography, Xuanwu Hospital, Capital Medical University, Beijing, China
- Vascular Biology and Therapeutics Program, Yale School of Medicine, New Haven, CT, USA
- Division of Vascular and Endovascular Surgery, Department of Surgery, Yale School of Medicine, New Haven, CT, USA
| | - Xixiang Gao
- Vascular Biology and Therapeutics Program, Yale School of Medicine, New Haven, CT, USA
- Division of Vascular and Endovascular Surgery, Department of Surgery, Yale School of Medicine, New Haven, CT, USA
- Department of Vascular Surgery, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Ryosuke Taniguchi
- Vascular Biology and Therapeutics Program, Yale School of Medicine, New Haven, CT, USA
- Division of Vascular and Endovascular Surgery, Department of Surgery, Yale School of Medicine, New Haven, CT, USA
- Division of Vascular Surgery, The University of Tokyo, Tokyo, Japan
| | - Anand Brahmandam
- Vascular Biology and Therapeutics Program, Yale School of Medicine, New Haven, CT, USA
- Division of Vascular and Endovascular Surgery, Department of Surgery, Yale School of Medicine, New Haven, CT, USA
| | - Yutaka Matsubara
- Vascular Biology and Therapeutics Program, Yale School of Medicine, New Haven, CT, USA
- Division of Vascular and Endovascular Surgery, Department of Surgery, Yale School of Medicine, New Haven, CT, USA
- Department of Surgery and Sciences, Kyushu University, Fukuoka, Japan
| | - Jia Liu
- Vascular Biology and Therapeutics Program, Yale School of Medicine, New Haven, CT, USA
- Division of Vascular and Endovascular Surgery, Department of Surgery, Yale School of Medicine, New Haven, CT, USA
| | - Hao Liu
- Vascular Biology and Therapeutics Program, Yale School of Medicine, New Haven, CT, USA
- Division of Vascular and Endovascular Surgery, Department of Surgery, Yale School of Medicine, New Haven, CT, USA
- Division of Vascular and Interventional Radiology, Department of General Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Weichang Zhang
- Vascular Biology and Therapeutics Program, Yale School of Medicine, New Haven, CT, USA
- Division of Vascular and Endovascular Surgery, Department of Surgery, Yale School of Medicine, New Haven, CT, USA
| | - Alan Dardik
- Vascular Biology and Therapeutics Program, Yale School of Medicine, New Haven, CT, USA.
- Division of Vascular and Endovascular Surgery, Department of Surgery, Yale School of Medicine, New Haven, CT, USA.
- Department of Surgery, VA Connecticut Healthcare Systems, West Haven, CT, USA.
- Yale School of Medicine, 10 Amistad Street, Room 437, PO Box 208089, New Haven, CT, 06520-8089, USA.
| |
Collapse
|
14
|
Laboyrie SL, de Vries MR, de Jong A, de Boer HC, Lalai RA, Martinez L, Vazquez-Padron RI, Rotmans JI. von Willebrand Factor: A Central Regulator of Arteriovenous Fistula Maturation Through Smooth Muscle Cell Proliferation and Outward Remodeling. J Am Heart Assoc 2022; 11:e024581. [PMID: 35929448 PMCID: PMC9496319 DOI: 10.1161/jaha.121.024581] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Background Arteriovenous fistula (AVF) maturation failure is a main limitation of vascular access. Maturation is determined by the intricate balance between outward remodeling and intimal hyperplasia, whereby endothelial cell dysfunction, platelet aggregation, and vascular smooth muscle cell (VSMC) proliferation play a crucial role. von Willebrand Factor (vWF) is an endothelial cell-derived protein involved in platelet aggregation and VSMC proliferation. We investigated AVF vascular remodeling in vWF-deficient mice and vWF expression in failed and matured human AVFs. Methods and Results Jugular-carotid AVFs were created in wild-type and vWF-/- mice. AVF flow was determined longitudinally using ultrasonography, whereupon AVFs were harvested 14 days after surgery. VSMCs were isolated from vena cavae to study the effect of vWF on VSMC proliferation. Patient-matched samples of the basilic vein were obtained before brachio-basilic AVF construction and during superficialization or salvage procedure 6 weeks after AVF creation. vWF deficiency reduced VSMC proliferation and macrophage infiltration in the intimal hyperplasia. vWF-/- mice showed reduced outward remodeling (1.5-fold, P=0.002) and intimal hyperplasia (10.2-fold, P<0.0001). AVF flow in wild-type mice was incremental over 2 weeks, whereas flow in vWF-/- mice did not increase, resulting in a two-fold lower flow at 14 days compared with wild-type mice (P=0.016). Outward remodeling in matured patient AVFs coincided with increased local vWF expression in the media of the venous outflow tract. Absence of vWF in the intimal layer correlated with an increase in the intima-media ratio. Conclusions vWF enhances AVF maturation because its positive effect on outward remodeling outweighs its stimulating effect on intimal hyperplasia.
Collapse
Affiliation(s)
- Suzanne L Laboyrie
- Internal Medicine Leiden University Medical Centre Leiden The Netherlands
| | | | - Alwin de Jong
- Surgery Leiden University Medical Centre Leiden The Netherlands
| | - Hetty C de Boer
- Internal Medicine Leiden University Medical Centre Leiden The Netherlands
| | - Reshma A Lalai
- Internal Medicine Leiden University Medical Centre Leiden The Netherlands
| | | | | | - Joris I Rotmans
- Internal Medicine Leiden University Medical Centre Leiden The Netherlands
| |
Collapse
|
15
|
Taniguchi R, Ohashi Y, Lee JS, Hu H, Gonzalez L, Zhang W, Langford J, Matsubara Y, Yatsula B, Tellides G, Fahmy TM, Hoshina K, Dardik A. Endothelial Cell TGF-β (Transforming Growth Factor-Beta) Signaling Regulates Venous Adaptive Remodeling to Improve Arteriovenous Fistula Patency. Arterioscler Thromb Vasc Biol 2022; 42:868-883. [PMID: 35510552 DOI: 10.1161/atvbaha.122.317676] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
BACKGROUND Arteriovenous fistulae (AVF) are the gold standard for vascular access for hemodialysis. Although the vein must thicken and dilate for successful hemodialysis, excessive wall thickness leads to stenosis causing AVF failure. Since TGF-β (transforming growth factor-beta) regulates ECM (extracellular matrix) deposition and smooth muscle cell (SMC) proliferation-critical components of wall thickness-we hypothesized that disruption of TGF-β signaling prevents excessive wall thickening during venous remodeling. METHODS A mouse aortocaval fistula model was used. SB431542-an inhibitor of TGF-β receptor I-was encapsulated in nanoparticles and applied to the AVF adventitia in C57BL/6J mice. Alternatively, AVFs were created in mice with conditional disruption of TGF-β receptors in either SMCs or endothelial cells. Doppler ultrasound was performed serially to confirm patency and to measure vessel diameters. AVFs were harvested at predetermined time points for histological and immunofluorescence analyses. RESULTS Inhibition of TGF-β signaling with SB431542-containing nanoparticles significantly reduced p-Smad2-positive cells in the AVF wall during the early maturation phase (days 7-21) and was associated with decreased AVF wall thickness that showed both decreased collagen density and decreased SMC proliferation. SMC-specific TGF-β signaling disruption decreased collagen density but not SMC proliferation or wall thickness. Endothelial cell-specific TGF-β signaling disruption decreased both collagen density and SMC proliferation in the AVF wall and was associated with reduced wall thickness, increased outward remodeling, and improved AVF patency. CONCLUSIONS Endothelial cell-targeted TGF-β inhibition may be a translational strategy to improve AVF patency.
Collapse
Affiliation(s)
- Ryosuke Taniguchi
- Vascular Biology and Therapeutics Program (R.T., Y.O., H.H., L.G., W.Z., J.L., Y.M., B.Y., G.T., A.D.), Yale School of Medicine, New Haven, CT.,Division of Vascular Surgery, The University of Tokyo, Japan (R.T., Y.O., K.H.)
| | - Yuichi Ohashi
- Vascular Biology and Therapeutics Program (R.T., Y.O., H.H., L.G., W.Z., J.L., Y.M., B.Y., G.T., A.D.), Yale School of Medicine, New Haven, CT.,Division of Vascular Surgery, The University of Tokyo, Japan (R.T., Y.O., K.H.)
| | - Jung Seok Lee
- Department of Biomedical Engineering, Yale University, New Haven, CT (J.S.L., T.M.F.)
| | - Haidi Hu
- Vascular Biology and Therapeutics Program (R.T., Y.O., H.H., L.G., W.Z., J.L., Y.M., B.Y., G.T., A.D.), Yale School of Medicine, New Haven, CT.,Department of Vascular and Thyroid Surgery, The First Hospital of China Medical University, Shenyang (H.H.)
| | - Luis Gonzalez
- Vascular Biology and Therapeutics Program (R.T., Y.O., H.H., L.G., W.Z., J.L., Y.M., B.Y., G.T., A.D.), Yale School of Medicine, New Haven, CT
| | - Weichang Zhang
- Vascular Biology and Therapeutics Program (R.T., Y.O., H.H., L.G., W.Z., J.L., Y.M., B.Y., G.T., A.D.), Yale School of Medicine, New Haven, CT
| | - John Langford
- Vascular Biology and Therapeutics Program (R.T., Y.O., H.H., L.G., W.Z., J.L., Y.M., B.Y., G.T., A.D.), Yale School of Medicine, New Haven, CT
| | - Yutaka Matsubara
- Vascular Biology and Therapeutics Program (R.T., Y.O., H.H., L.G., W.Z., J.L., Y.M., B.Y., G.T., A.D.), Yale School of Medicine, New Haven, CT.,Department of Surgery and Sciences, Kyushu University, Fukuoka, Japan (Y.M.)
| | - Bogdan Yatsula
- Vascular Biology and Therapeutics Program (R.T., Y.O., H.H., L.G., W.Z., J.L., Y.M., B.Y., G.T., A.D.), Yale School of Medicine, New Haven, CT
| | - George Tellides
- Vascular Biology and Therapeutics Program (R.T., Y.O., H.H., L.G., W.Z., J.L., Y.M., B.Y., G.T., A.D.), Yale School of Medicine, New Haven, CT.,Division of Cardiac Surgery, Department of Surgery (G.T.), Yale School of Medicine, New Haven, CT.,Department of Surgery, VA Connecticut Healthcare Systems, West Haven, CT (G.T., A.D.)
| | - Tarek M Fahmy
- Department of Biomedical Engineering, Yale University, New Haven, CT (J.S.L., T.M.F.)
| | - Katsuyuki Hoshina
- Division of Vascular Surgery, The University of Tokyo, Japan (R.T., Y.O., K.H.)
| | - Alan Dardik
- Vascular Biology and Therapeutics Program (R.T., Y.O., H.H., L.G., W.Z., J.L., Y.M., B.Y., G.T., A.D.), Yale School of Medicine, New Haven, CT.,Division of Vascular and Endovascular Surgery, Department of Surgery (A.D.), Yale School of Medicine, New Haven, CT.,Department of Surgery, VA Connecticut Healthcare Systems, West Haven, CT (G.T., A.D.)
| |
Collapse
|
16
|
Samra G, Rai V, Agrawal DK. Innate and Adaptive Immune Cells Associates with Arteriovenous Fistula Maturation and Failure. Can J Physiol Pharmacol 2022; 100:716-727. [PMID: 35671528 DOI: 10.1139/cjpp-2021-0731] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Creation of arteriovenous fistula (AVF) causes local injury resulting in immune response of the body and infiltration of immune cells. Acute inflammation is favorable to control inflammation and proceed AVF towards maturation while chronic inflammation in AVF lead to AVF maturation failure. Chronic inflammation in AVF is due to chronic infiltration of immune cells and secretion of inflammatory cytokines. A balance between pro-inflammatory and anti-inflammatory response is must for AVF maturation and an overwhelmed proinflammatory infiltrate endue chronic inflammation and AVF failure. Since immune cell infiltration plays a critical role in maturation and failure of AVF, it is important to investigate the role of immune cells as well as their density in early and late phase of AVF maturation. The role of inflammation has been discussed in the literature and this review article focuses on the role of pro- and anti-inflammatory immune cells including macrophages, dendritic cells, T-cells, and T-regulatory cells in AVF maturation and maturation failure.
Collapse
Affiliation(s)
- Gunimat Samra
- Western University of Health Sciences, 6645, Translational Research, Pomona, California, United States;
| | - Vikrant Rai
- Western University of Health Sciences, 6645, Translational Research, Pomona, California, United States;
| | - Devendra K Agrawal
- Western University of Health Sciences, 6645, Department of Translational Research, Pomona, United States, 91766-1854;
| |
Collapse
|
17
|
Hirudin Regulates Vascular Function in Chronic Renal Failure through Modulating Macrophage Polarization. BIOMED RESEARCH INTERNATIONAL 2022; 2022:6043698. [PMID: 35496058 PMCID: PMC9042600 DOI: 10.1155/2022/6043698] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/29/2021] [Revised: 03/18/2022] [Accepted: 03/21/2022] [Indexed: 11/30/2022]
Abstract
Excessive inflammation is responsible for arteriovenous fistula (AVF) failure, which determines the therapeutic effect of chronic renal failure (CRF). Macrophage polarization is of great significance in the inflammatory response. Hirudin (Hiru) has been reported to possess a definite anti-inflammatory effect. This study is to uncover the impacts of Hiru on classically (M1)/alternatively (M2) macrophage polarization in the CRF rat model and rat vascular smooth muscle cells (VSMCs). After the CRF rat model was administrated with different concentrations of Hiru, blood urea nitrogen (BUN) and serum creatinine (Scr) levels were tested. H&E staining was to detect vascular injury, and IHC assay was to analyze inducible nitric oxide synthase (iNOS) and arginase-1 (Arg-1) expressions in vascular tissues. Levels of inflammatory factors were examined by ELISA. Besides, western blot was to estimate the levels of marker proteins related to macrophage, proliferation, and apoptosis. CCK-8 was to measure cell viability. We discovered that Hiru alleviated renal function injury and vascular injury, exacerbated VSMC hyperplasia, and stimulated the differentiation and activation of M1 macrophage towards M2 macrophage in vivo. Moreover, after treatment with lipopolysaccharide (LPS)/IFN-gamma (IFN-γ), the increased M1/M2 ratio and enhanced levels of inflammatory factors were observed. Furthermore, Hiru boosted the proliferation and ameliorated the inflammatory response and apoptosis of rat VSMCs during the process of coincubation of M1-conditioned medium (CM). Collectively, Hiru played a protective role against vascular injury in CRF directly or through its influence on M1 macrophage polarization and inflammation.
Collapse
|
18
|
Ratti S, Mauro R, Rocchi C, Mongiorgi S, Ramazzotti G, Gargiulo M, Manzoli L, Cocco L, Fiume R. Roles of PI3K/AKT/mTOR Axis in Arteriovenous Fistula. Biomolecules 2022; 12:biom12030350. [PMID: 35327539 PMCID: PMC8945685 DOI: 10.3390/biom12030350] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Revised: 01/28/2022] [Accepted: 02/14/2022] [Indexed: 11/22/2022] Open
Abstract
Renal failure is a worldwide disease with a continuously increasing prevalence and involving a rising need for long-term treatment, mainly by haemodialysis. Arteriovenous fistula (AVF) is the favourite type of vascular access for haemodialysis; however, the lasting success of this therapy depends on its maturation, which is directly influenced by many concomitant processes such as vein wall thickening or inflammation. Understanding the molecular mechanisms that drive AVF maturation and failure can highlight new or combinatorial drugs for more personalized therapy. In this review we analysed the relevance of critical enzymes such as PI3K, AKT and mTOR in processes such as wall thickening remodelling, immune system activation and inflammation reduction. We focused on these enzymes due to their involvement in the modulation of numerous cellular activities such as proliferation, differentiation and motility, and their impairment is related to many diseases such as cancer, metabolic syndrome and neurodegenerative disorders. In addition, these enzymes are highly druggable targets, with several inhibitors already being used in patient treatment for cancer and with encouraging results for AVF. Finally, we delineate how these enzymes may be targeted to control specific aspects of AVF in an effort to propose a more specialized therapy with fewer side effects.
Collapse
Affiliation(s)
- Stefano Ratti
- Cellular Signalling Laboratory, Department of Biomedical and Neuromotor Sciences, University of Bologna, 40126 Bologna, Italy; (S.R.); (S.M.); (G.R.); (L.M.); (R.F.)
| | - Raffaella Mauro
- Vascular Surgery Unit, IRCCS University Hospital, Policlinico S. Orsola-Malpighi, 40126 Bologna, Italy; (R.M.); (C.R.); (M.G.)
| | - Cristina Rocchi
- Vascular Surgery Unit, IRCCS University Hospital, Policlinico S. Orsola-Malpighi, 40126 Bologna, Italy; (R.M.); (C.R.); (M.G.)
| | - Sara Mongiorgi
- Cellular Signalling Laboratory, Department of Biomedical and Neuromotor Sciences, University of Bologna, 40126 Bologna, Italy; (S.R.); (S.M.); (G.R.); (L.M.); (R.F.)
| | - Giulia Ramazzotti
- Cellular Signalling Laboratory, Department of Biomedical and Neuromotor Sciences, University of Bologna, 40126 Bologna, Italy; (S.R.); (S.M.); (G.R.); (L.M.); (R.F.)
| | - Mauro Gargiulo
- Vascular Surgery Unit, IRCCS University Hospital, Policlinico S. Orsola-Malpighi, 40126 Bologna, Italy; (R.M.); (C.R.); (M.G.)
| | - Lucia Manzoli
- Cellular Signalling Laboratory, Department of Biomedical and Neuromotor Sciences, University of Bologna, 40126 Bologna, Italy; (S.R.); (S.M.); (G.R.); (L.M.); (R.F.)
| | - Lucio Cocco
- Cellular Signalling Laboratory, Department of Biomedical and Neuromotor Sciences, University of Bologna, 40126 Bologna, Italy; (S.R.); (S.M.); (G.R.); (L.M.); (R.F.)
- Correspondence: ; Tel.: +39-051-209-1639
| | - Roberta Fiume
- Cellular Signalling Laboratory, Department of Biomedical and Neuromotor Sciences, University of Bologna, 40126 Bologna, Italy; (S.R.); (S.M.); (G.R.); (L.M.); (R.F.)
| |
Collapse
|
19
|
Matsubara Y, Gonzalez L, Kiwan G, Liu J, Langford J, Gao M, Gao X, Taniguchi R, Yatsula B, Furuyama T, Matsumoto T, Komori K, Mori M, Dardik A. PD-L1 (Programmed Death Ligand 1) Regulates T-Cell Differentiation to Control Adaptive Venous Remodeling. Arterioscler Thromb Vasc Biol 2021; 41:2909-2922. [PMID: 34670406 PMCID: PMC8664128 DOI: 10.1161/atvbaha.121.316380] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
OBJECTIVE Patients with end-stage renal disease depend on hemodialysis for survival. Although arteriovenous fistulae (AVF) are the preferred vascular access for hemodialysis, the primary success rate of AVF is only 30% to 50% within 6 months, showing an urgent need for improvement. PD-L1 (programmed death ligand 1) is a ligand that regulates T-cell activity. Since T cells have an important role during AVF maturation, we hypothesized that PD-L1 regulates T cells to control venous remodeling that occurs during AVF maturation. Approach and results: In the mouse aortocaval fistula model, anti-PD-L1 antibody (200 mg, 3×/wk intraperitoneal) was given to inhibit PD-L1 activity during AVF maturation. Inhibition of PD-L1 increased T-helper type 1 cells and T-helper type 2 cells but reduced regulatory T cells to increase M1-type macrophages and reduce M2-type macrophages; these changes were associated with reduced vascular wall thickening and reduced AVF patency. Inhibition of PD-L1 also inhibited smooth muscle cell proliferation and increased endothelial dysfunction. The effects of anti-PD-L1 antibody on adaptive venous remodeling were diminished in nude mice; however, they were restored after T-cell transfer into nude mice, indicating the effects of anti-PD-L1 antibody on venous remodeling were dependent on T cells. CONCLUSIONS Regulation of PD-L1 activity may be a potential therapeutic target for clinical translation to improve AVF maturation.
Collapse
Affiliation(s)
- Yutaka Matsubara
- Vascular Biology and Therapeutics Program, Yale School of Medicine, New Haven, CT
- Department of Surgery and Sciences, Kyushu University, Fukuoka, Japan
| | - Luis Gonzalez
- Vascular Biology and Therapeutics Program, Yale School of Medicine, New Haven, CT
| | - Gathe Kiwan
- Vascular Biology and Therapeutics Program, Yale School of Medicine, New Haven, CT
| | - Jia Liu
- Vascular Biology and Therapeutics Program, Yale School of Medicine, New Haven, CT
| | - John Langford
- Vascular Biology and Therapeutics Program, Yale School of Medicine, New Haven, CT
| | - Mingjie Gao
- Vascular Biology and Therapeutics Program, Yale School of Medicine, New Haven, CT
- Department of Vascular Ultrasonography, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Xixiang Gao
- Vascular Biology and Therapeutics Program, Yale School of Medicine, New Haven, CT
- Department of Vascular Surgery, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Ryosuke Taniguchi
- Vascular Biology and Therapeutics Program, Yale School of Medicine, New Haven, CT
- Division of Vascular Surgery, The University of Tokyo, Tokyo, Japan
| | - Bogdan Yatsula
- Vascular Biology and Therapeutics Program, Yale School of Medicine, New Haven, CT
| | - Tadashi Furuyama
- Department of Surgery and Sciences, Kyushu University, Fukuoka, Japan
| | - Takuya Matsumoto
- Department of Vascular Surgery, Kyushu Central Hospital, Fukuoka, Japan
| | - Kimihiro Komori
- Division of Vascular Surgery, Department of Surgery, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Masaki Mori
- Department of Surgery and Sciences, Kyushu University, Fukuoka, Japan
| | - Alan Dardik
- Vascular Biology and Therapeutics Program, Yale School of Medicine, New Haven, CT
- Division of Vascular and Endovascular Surgery, Department of Surgery, Yale School of Medicine, New Haven, CT
- Department of Surgery, VA Connecticut Healthcare Systems, West Haven, CT
| |
Collapse
|
20
|
Rodriguez M, Chen J, Jain PP, Babicheva A, Xiong M, Li J, Lai N, Zhao T, Hernandez M, Balistrieri A, Parmisano S, Simonson T, Breen E, Valdez-Jasso D, Thistlethwaite PA, Shyy JYJ, Wang J, Garcia JGN, Makino A, Yuan JXJ. Upregulation of Calcium Homeostasis Modulators in Contractile-To-Proliferative Phenotypical Transition of Pulmonary Arterial Smooth Muscle Cells. Front Physiol 2021; 12:714785. [PMID: 34408668 PMCID: PMC8364962 DOI: 10.3389/fphys.2021.714785] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2021] [Accepted: 07/13/2021] [Indexed: 12/14/2022] Open
Abstract
Excessive pulmonary artery (PA) smooth muscle cell (PASMC) proliferation and migration are implicated in the development of pathogenic pulmonary vascular remodeling characterized by concentric arterial wall thickening and arteriole muscularization in patients with pulmonary arterial hypertension (PAH). Pulmonary artery smooth muscle cell contractile-to-proliferative phenotypical transition is a process that promotes pulmonary vascular remodeling. A rise in cytosolic Ca2+ concentration [(Ca2+) cyt ] in PASMCs is a trigger for pulmonary vasoconstriction and a stimulus for pulmonary vascular remodeling. Here, we report that the calcium homeostasis modulator (CALHM), a Ca2+ (and ATP) channel that is allosterically regulated by voltage and extracellular Ca2+, is upregulated during the PASMC contractile-to-proliferative phenotypical transition. Protein expression of CALHM1/2 in primary cultured PASMCs in media containing serum and growth factors (proliferative PASMC) was significantly greater than in freshly isolated PA (contractile PASMC) from the same rat. Upregulated CALHM1/2 in proliferative PASMCs were associated with an increased ratio of pAKT/AKT and pmTOR/mTOR and an increased expression of the cell proliferation marker PCNA, whereas serum starvation and rapamycin significantly downregulated CALHM1/2. Furthermore, CALHM1/2 were upregulated in freshly isolated PA from rats with monocrotaline (MCT)-induced PH and in primary cultured PASMC from patients with PAH in comparison to normal controls. Intraperitoneal injection of CGP 37157 (0.6 mg/kg, q8H), a non-selective blocker of CALHM channels, partially reversed established experimental PH. These data suggest that CALHM upregulation is involved in PASMC contractile-to-proliferative phenotypical transition. Ca2+ influx through upregulated CALHM1/2 may play an important role in the transition of sustained vasoconstriction to excessive vascular remodeling in PAH or precapillary PH. Calcium homeostasis modulator could potentially be a target to develop novel therapies for PAH.
Collapse
Affiliation(s)
- Marisela Rodriguez
- Section of Physiology, Division of Pulmonary, Critical Care and Sleep Medicine, La Jolla, CA, United States
- Department of Pediatrics, Tucson, AZ, United States
| | - Jiyuan Chen
- Section of Physiology, Division of Pulmonary, Critical Care and Sleep Medicine, La Jolla, CA, United States
- State Key Laboratory of Respiratory Diseases, First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Pritesh P. Jain
- Section of Physiology, Division of Pulmonary, Critical Care and Sleep Medicine, La Jolla, CA, United States
| | - Aleksandra Babicheva
- Section of Physiology, Division of Pulmonary, Critical Care and Sleep Medicine, La Jolla, CA, United States
| | - Mingmei Xiong
- Section of Physiology, Division of Pulmonary, Critical Care and Sleep Medicine, La Jolla, CA, United States
- State Key Laboratory of Respiratory Diseases, First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Jifeng Li
- Section of Physiology, Division of Pulmonary, Critical Care and Sleep Medicine, La Jolla, CA, United States
- Beijing Chaoyang Hospital, Capital Medical University, Beijing, China
| | - Ning Lai
- Section of Physiology, Division of Pulmonary, Critical Care and Sleep Medicine, La Jolla, CA, United States
- State Key Laboratory of Respiratory Diseases, First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Tengteng Zhao
- Section of Physiology, Division of Pulmonary, Critical Care and Sleep Medicine, La Jolla, CA, United States
| | - Moises Hernandez
- Division of Cardiothoracic Surgery, Department of Surgery, La Jolla, CA, United States
| | - Angela Balistrieri
- Section of Physiology, Division of Pulmonary, Critical Care and Sleep Medicine, La Jolla, CA, United States
| | - Sophia Parmisano
- Section of Physiology, Division of Pulmonary, Critical Care and Sleep Medicine, La Jolla, CA, United States
| | - Tatum Simonson
- Section of Physiology, Division of Pulmonary, Critical Care and Sleep Medicine, La Jolla, CA, United States
| | - Ellen Breen
- Section of Physiology, Division of Pulmonary, Critical Care and Sleep Medicine, La Jolla, CA, United States
| | - Daniela Valdez-Jasso
- Department of Bioengineering, University of California, San Diego, La Jolla, CA, United States
| | | | - John Y. -J. Shyy
- Division of Cardiovascular Medicine, Department of Medicine, La Jolla, CA, United States
| | - Jian Wang
- Section of Physiology, Division of Pulmonary, Critical Care and Sleep Medicine, La Jolla, CA, United States
- State Key Laboratory of Respiratory Diseases, First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Joe G. N. Garcia
- Department of Medicine, The University of Arizona, Tucson, AZ, United States
| | - Ayako Makino
- Division of Endocrinology and Metabolism, La Jolla, CA, United States
| | - Jason X. -J. Yuan
- Section of Physiology, Division of Pulmonary, Critical Care and Sleep Medicine, La Jolla, CA, United States
| |
Collapse
|
21
|
Vazquez-Padron RI, Duque JC, Tabbara M, Salman LH, Martinez L. Intimal Hyperplasia and Arteriovenous Fistula Failure: Looking Beyond Size Differences. KIDNEY360 2021; 2:1360-1372. [PMID: 34765989 PMCID: PMC8579754 DOI: 10.34067/kid.0002022021] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
The development of venous intimal hyperplasia (IH) has been historically associated with failure of arteriovenous fistulas (AVF) used for hemodialysis. This long-standing assumption, based on histological observations, has been recently challenged by clinical studies indicating that the size of the intima by itself is not enough to explain stenosis or AVF maturation failure. Irrespective of this lack of association, IH is present in most native veins and fistulas, is prominent in many cases, and suggests a role in the vein that may not be reflected by its dimensions. Therefore, the contribution of IH to AVF dysfunction remains controversial. Using only clinical data and avoiding extrapolations from animal models, we critically discuss the biological significance of IH in vein remodeling, vascular access function, and the response of the venous wall to repeated trauma in hemodialysis patients. We address questions and pose new ones such as: What are the factors that contribute to IH in pre-access veins and AVFs? Do cellular phenotypes and composition of the intima influence AVF function? Are there protective roles of the venous intima? This review explores these possibilities, with hopes of rekindling a critical discussion about venous IH that goes beyond thickness and AVF outcomes.
Collapse
Affiliation(s)
- Roberto I Vazquez-Padron
- DeWitt Daughtry Family Department of Surgery, Leonard M. Miller School of Medicine, University of Miami, Miami, Florida
| | - Juan C Duque
- Katz Family Division of Nephrology, Department of Medicine, Leonard M. Miller School of Medicine, University of Miami, Miami, Florida
| | - Marwan Tabbara
- DeWitt Daughtry Family Department of Surgery, Leonard M. Miller School of Medicine, University of Miami, Miami, Florida
| | - Loay H Salman
- Division of Nephrology, Albany Medical College, Albany, New York
| | - Laisel Martinez
- DeWitt Daughtry Family Department of Surgery, Leonard M. Miller School of Medicine, University of Miami, Miami, Florida
| |
Collapse
|
22
|
Chan SM, Weininger G, Langford J, Jane-Wit D, Dardik A. Sex Differences in Inflammation During Venous Remodeling of Arteriovenous Fistulae. Front Cardiovasc Med 2021; 8:715114. [PMID: 34368264 PMCID: PMC8335484 DOI: 10.3389/fcvm.2021.715114] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Accepted: 06/24/2021] [Indexed: 12/18/2022] Open
Abstract
Vascular disorders frequently have differing clinical presentations among women and men. Sex differences exist in vascular access for hemodialysis; women have reduced rates of arteriovenous fistula (AVF) maturation as well as fistula utilization compared with men. Inflammation is increasingly implicated in both clinical studies and animal models as a potent mechanism driving AVF maturation, especially in vessel dilation and wall thickening, that allows venous remodeling to the fistula environment to support hemodialysis. Sex differences have long been recognized in arterial remodeling and diseases, with men having increased cardiovascular events compared with pre-menopausal women. Many of these arterial diseases are driven by inflammation that is similar to the inflammation during AVF maturation. Improved understanding of sex differences in inflammation during vascular remodeling may suggest sex-specific vascular therapies to improve AVF success.
Collapse
Affiliation(s)
- Shin Mei Chan
- Vascular Biology and Therapeutics Program, Yale School of Medicine, New Haven, CT, United States
| | - Gabe Weininger
- Vascular Biology and Therapeutics Program, Yale School of Medicine, New Haven, CT, United States
| | - John Langford
- Vascular Biology and Therapeutics Program, Yale School of Medicine, New Haven, CT, United States.,Department of Surgery, Yale School of Medicine, New Haven, CT, United States
| | - Daniel Jane-Wit
- Vascular Biology and Therapeutics Program, Yale School of Medicine, New Haven, CT, United States.,Division of Cardiovascular Medicine, Department of Internal Medicine, Yale School of Medicine, New Haven, CT, United States.,Department of Immunobiology, Yale School of Medicine, New Haven, CT, United States
| | - Alan Dardik
- Vascular Biology and Therapeutics Program, Yale School of Medicine, New Haven, CT, United States.,Department of Surgery, Yale School of Medicine, New Haven, CT, United States.,Department of Surgery, Veterans Affairs (VA) Connecticut Healthcare System, West Haven, CT, United States
| |
Collapse
|
23
|
Wang T, Liu J, Liu H, Lee SR, Gonzalez L, Gorecka J, Shu C, Dardik A. Activation of EphrinB2 Signaling Promotes Adaptive Venous Remodeling in Murine Arteriovenous Fistulae. J Surg Res 2021; 262:224-239. [PMID: 33039109 PMCID: PMC8024410 DOI: 10.1016/j.jss.2020.08.071] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2020] [Revised: 08/26/2020] [Accepted: 08/30/2020] [Indexed: 12/25/2022]
Abstract
BACKGROUND Arteriovenous fistulae (AVF) are the preferred mode of vascular access for hemodialysis. Before use, AVF remodel by thickening and dilating to achieve a functional conduit via an adaptive process characterized by expression of molecular markers characteristic of both venous and arterial identity. Although signaling via EphB4, a determinant of venous identity, mediates AVF maturation, the role of its counterpart EphrinB2, a determinant of arterial identity, remains unclear. We hypothesize that EphrinB2 signaling is active during AVF maturation and may be a mechanism of venous remodeling. METHODS Aortocaval fistulae were created or sham laparotomy was performed in C57Bl/6 mice, and specimens were examined on Days 7 or 21. EphrinB2 reverse signaling was activated with EphB4-Fc applied periadventitially in vivo and in endothelial cell culture medium in vitro. Downstream signaling was assessed using immunoblotting and immunofluorescence. RESULTS Venous remodeling during AVF maturation was characterized by increased expression of EphrinB2 as well as Akt1, extracellular signal-regulated kinases 1/2 (ERK1/2), and p38. Activation of EphrinB2 with EphB4-Fc increased phosphorylation of EphrinB2, endothelial nitric oxide synthase, Akt1, ERK1/2, and p38 and was associated with increased diameter and wall thickness in the AVF. Both mouse and human endothelial cells treated with EphB4-Fc increased phosphorylation of EphrinB2, endothelial nitric oxide synthase, Akt1, ERK1/2, and p38 and increased endothelial cell tube formation and migration. CONCLUSIONS Activation of EphrinB2 signaling by EphB4-Fc was associated with adaptive venous remodeling in vivo while activating endothelial cell function in vitro. Regulation of EphrinB2 signaling may be a new strategy to improve AVF maturation and patency.
Collapse
Affiliation(s)
- Tun Wang
- Department of Vascular Surgery, The Second Xiangya Hospital, Central South University, Changsha, Hunan, People's Republic of China; The Vascular Biology and Therapeutics Program, Yale School of Medicine, New Haven, Connecticut; Department of Surgery, Yale School of Medicine, New Haven, Connecticut; Department of Cellular and Molecular Physiology, Yale School of Medicine, New Haven, Connecticut
| | - Jia Liu
- Department of Vascular Surgery, The Second Xiangya Hospital, Central South University, Changsha, Hunan, People's Republic of China; The Vascular Biology and Therapeutics Program, Yale School of Medicine, New Haven, Connecticut; Department of Surgery, Yale School of Medicine, New Haven, Connecticut; Department of Cellular and Molecular Physiology, Yale School of Medicine, New Haven, Connecticut
| | - Haiyang Liu
- The Vascular Biology and Therapeutics Program, Yale School of Medicine, New Haven, Connecticut; Department of Surgery, Yale School of Medicine, New Haven, Connecticut; Department of Cellular and Molecular Physiology, Yale School of Medicine, New Haven, Connecticut
| | - Shin-Rong Lee
- The Vascular Biology and Therapeutics Program, Yale School of Medicine, New Haven, Connecticut; Department of Surgery, Yale School of Medicine, New Haven, Connecticut; Department of Cellular and Molecular Physiology, Yale School of Medicine, New Haven, Connecticut
| | - Luis Gonzalez
- The Vascular Biology and Therapeutics Program, Yale School of Medicine, New Haven, Connecticut; Department of Surgery, Yale School of Medicine, New Haven, Connecticut; Department of Cellular and Molecular Physiology, Yale School of Medicine, New Haven, Connecticut
| | - Jolanta Gorecka
- The Vascular Biology and Therapeutics Program, Yale School of Medicine, New Haven, Connecticut; Department of Surgery, Yale School of Medicine, New Haven, Connecticut; Department of Cellular and Molecular Physiology, Yale School of Medicine, New Haven, Connecticut
| | - Chang Shu
- Department of Vascular Surgery, The Second Xiangya Hospital, Central South University, Changsha, Hunan, People's Republic of China; State Key Laboratory of Cardiovascular Disease, Center of Vascular Surgery, Fuwai Hospital, National Center for Cardiovascular Disease, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Alan Dardik
- The Vascular Biology and Therapeutics Program, Yale School of Medicine, New Haven, Connecticut; Department of Surgery, Yale School of Medicine, New Haven, Connecticut; Department of Cellular and Molecular Physiology, Yale School of Medicine, New Haven, Connecticut; Department of Surgery, VA Connecticut Healthcare System, West Haven, Connecticut.
| |
Collapse
|
24
|
Shih YC, Chen PY, Ko TM, Huang PH, Ma H, Tarng DC. MMP-9 Deletion Attenuates Arteriovenous Fistula Neointima through Reduced Perioperative Vascular Inflammation. Int J Mol Sci 2021; 22:ijms22115448. [PMID: 34064140 PMCID: PMC8196691 DOI: 10.3390/ijms22115448] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Revised: 05/15/2021] [Accepted: 05/18/2021] [Indexed: 01/05/2023] Open
Abstract
Matrix metalloproteinase 9 (MMP-9) expression is upregulated in vascular inflammation and participates in vascular remodeling, including aneurysm dilatation and arterial neointima development. Neointima at the arteriovenous (AV) fistula anastomosis site primarily causes AV fistula stenosis and failure; however, the effects of MMP-9 on perioperative AV fistula remodeling remain unknown. Therefore, we created AV fistulas (end-to-side anastomosis) in wild-type (WT) and MMP-9 knockout mice with chronic kidney disease to further clarify this. Neointima progressively developed in the AV fistula venous segment of WT mice during the four-week postoperative course, and MMP-9 knockout increased the lumen area and attenuated neointima size by reducing smooth muscle cell and collagen components. Early perioperative AV fistula mRNA sequencing data revealed that inflammation-related gene sets were negatively enriched in AV fistula of MMP-9 knockout mice compared to that in WT mice. qPCR results also showed that inflammatory genes, including tumor necrosis factor-α (TNF-α), monocyte chemoattractant protein-1 (MCP-1), interleukin-6 (IL-6), intercellular adhesion molecule-1 (ICAM-1), and vascular cell adhesion molecule-1 (VCAM-1), were downregulated. In addition, Western blot results showed that MMP-9 knockout reduced CD44 and RAC-alpha serine/threonine-protein kinase (Akt) and extracellular signal-regulated kinases (ERK) phosphorylation. In vitro, MMP-9 addition enhanced IL-6 and MCP-1 expression in vascular smooth muscle cells, as well as cell migration, which was reversed by an MMP-9 inhibitor. In conclusion, MMP-9 knockout attenuated AV fistula stenosis by reducing perioperative vascular inflammation.
Collapse
Affiliation(s)
- Yu-Chung Shih
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Taipei Veterans General Hospital, Taipei 11217, Taiwan; (Y.-C.S.); (H.M.)
- Institute of Clinical Medicine, National Yang Ming Chiao Tung University, Taipei 11221, Taiwan;
- Institute of Clinical Medicine, National Yang Ming University, Taipei 11221, Taiwan
- Department of Surgery, School of Medicine, National Yang Ming Chiao Tung University, Taipei 112, Taiwan
| | - Po-Yuan Chen
- Bioinformatics Program, Taiwan International Graduate Program, Academia Sinica, Taipei 115, Taiwan; (P.-Y.C.); (T.-M.K.)
- Institute of Bioinformatics and Systems Biology, National Yang Ming Chiao Tung University, Hsinchu 300, Taiwan
- Institute of Information Science, Academia Sinica, Taipei 115, Taiwan
| | - Tai-Ming Ko
- Bioinformatics Program, Taiwan International Graduate Program, Academia Sinica, Taipei 115, Taiwan; (P.-Y.C.); (T.-M.K.)
- Institute of Bioinformatics and Systems Biology, National Yang Ming Chiao Tung University, Hsinchu 300, Taiwan
- Department of Biological Science and Technology, National Yang Ming Chiao Tung University, Hsinchu 300, Taiwan
- Institute of Biomedical Sciences, Academia Sinica, Taipei 115, Taiwan
- Center of Intelligent Drug System and Smart Bio-devices (IDS2B), National Yang Ming Chiao Tung University, Hsinchu 300, Taiwan
| | - Po-Hsun Huang
- Institute of Clinical Medicine, National Yang Ming Chiao Tung University, Taipei 11221, Taiwan;
- Institute of Clinical Medicine, National Yang Ming University, Taipei 11221, Taiwan
- Department of Critical Care Medicine, Taipei Veterans General Hospital, Taipei 11217, Taiwan
| | - Hsu Ma
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Taipei Veterans General Hospital, Taipei 11217, Taiwan; (Y.-C.S.); (H.M.)
- Department of Surgery, School of Medicine, National Yang Ming Chiao Tung University, Taipei 112, Taiwan
- Department of Surgery, School of Medicine, National Defense Medical Center, Taipei 11490, Taiwan
| | - Der-Cherng Tarng
- Institute of Clinical Medicine, National Yang Ming Chiao Tung University, Taipei 11221, Taiwan;
- Institute of Clinical Medicine, National Yang Ming University, Taipei 11221, Taiwan
- Division of Nephrology, Department of Medicine, Taipei Veterans General Hospital, Taipei 11217, Taiwan
- Institute of Physiology, National Yang Ming Chiao Tung University, Taipei 11221, Taiwan
- Correspondence:
| |
Collapse
|
25
|
Matsubara (松原裕) Y, Kiwan G, Liu (刘佳) J, Gonzalez L, Langford J, Gao (高明杰) M, Gao (高喜翔) X, Taniguchi (谷口良輔) R, Yatsula B, Furuyama (古山正) T, Matsumoto (松本拓也) T, Komori (古森公浩) K, Dardik A. Inhibition of T-Cells by Cyclosporine A Reduces Macrophage Accumulation to Regulate Venous Adaptive Remodeling and Increase Arteriovenous Fistula Maturation. Arterioscler Thromb Vasc Biol 2021; 41:e160-e174. [PMID: 33472405 PMCID: PMC7904667 DOI: 10.1161/atvbaha.120.315875] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
OBJECTIVE Arteriovenous fistulae (AVF) are the preferred vascular access for hemodialysis, but the primary success rate of AVF remains poor. Successful AVF maturation requires vascular wall thickening and outward remodeling. A key factor determining successful AVF maturation is inflammation that is characterized by accumulation of both T-cells and macrophages. We have previously shown that anti-inflammatory (M2) macrophages are critically important for vascular wall thickening during venous remodeling; therefore, regulation of macrophage accumulation may be an important mechanism promoting AVF maturation. Since CD4+ T-cells such as T-helper type 1 cells, T-helper type 2 cells, and regulatory T-cells can induce macrophage migration, proliferation, and polarization, we hypothesized that CD4+ T-cells regulate macrophage accumulation to promote AVF maturation. Approach and Results: In a mouse aortocaval fistula model, T-cells temporally precede macrophages in the remodeling AVF wall. CsA (cyclosporine A; 5 mg/kg, sq, daily) or vehicle (5% dimethyl sulfoxide) was administered to inhibit T-cell function during venous remodeling. CsA reduced the numbers of T-helper type 1 cells, T-helper type 2, and regulatory T-cells, as well as M1- and M2-macrophage accumulation in the wall of the remodeling fistula; these effects were associated with reduced vascular wall thickening and increased outward remodeling in wild-type mice. However, these effects were eliminated in nude mice, showing that the effects of CsA on macrophage accumulation and adaptive venous remodeling are T-cell-dependent. CONCLUSIONS T-cells regulate macrophage accumulation in the maturing venous wall to control adaptive remodeling. Regulation of T-cells during AVF maturation may be a strategy that can improve AVF maturation. Graphic Abstract: A graphic abstract is available for this article.
Collapse
Affiliation(s)
- Yutaka Matsubara (松原裕)
- Vascular Biology and Therapeutics Program (Y.M., G.K., J. Liu, L.G., J. Langford, M.G., X.G., R.T., B.Y., A.D.), Yale School of Medicine, New Haven, CT
- Department of Surgery and Sciences, Kyushu University, Fukuoka, Japan (Y.M., T.F.)
| | - Gathe Kiwan
- Vascular Biology and Therapeutics Program (Y.M., G.K., J. Liu, L.G., J. Langford, M.G., X.G., R.T., B.Y., A.D.), Yale School of Medicine, New Haven, CT
| | - Jia Liu (刘佳)
- Vascular Biology and Therapeutics Program (Y.M., G.K., J. Liu, L.G., J. Langford, M.G., X.G., R.T., B.Y., A.D.), Yale School of Medicine, New Haven, CT
| | - Luis Gonzalez
- Vascular Biology and Therapeutics Program (Y.M., G.K., J. Liu, L.G., J. Langford, M.G., X.G., R.T., B.Y., A.D.), Yale School of Medicine, New Haven, CT
| | - John Langford
- Vascular Biology and Therapeutics Program (Y.M., G.K., J. Liu, L.G., J. Langford, M.G., X.G., R.T., B.Y., A.D.), Yale School of Medicine, New Haven, CT
| | - Mingjie Gao (高明杰)
- Vascular Biology and Therapeutics Program (Y.M., G.K., J. Liu, L.G., J. Langford, M.G., X.G., R.T., B.Y., A.D.), Yale School of Medicine, New Haven, CT
| | - Xixiang Gao (高喜翔)
- Vascular Biology and Therapeutics Program (Y.M., G.K., J. Liu, L.G., J. Langford, M.G., X.G., R.T., B.Y., A.D.), Yale School of Medicine, New Haven, CT
| | - Ryosuke Taniguchi (谷口良輔)
- Vascular Biology and Therapeutics Program (Y.M., G.K., J. Liu, L.G., J. Langford, M.G., X.G., R.T., B.Y., A.D.), Yale School of Medicine, New Haven, CT
| | - Bogdan Yatsula
- Vascular Biology and Therapeutics Program (Y.M., G.K., J. Liu, L.G., J. Langford, M.G., X.G., R.T., B.Y., A.D.), Yale School of Medicine, New Haven, CT
| | | | | | - Kimihiro Komori (古森公浩)
- Division of Vascular Surgery, Department of Surgery, Nagoya University Graduate School of Medicine, Japan (K.K.)
| | - Alan Dardik
- Vascular Biology and Therapeutics Program (Y.M., G.K., J. Liu, L.G., J. Langford, M.G., X.G., R.T., B.Y., A.D.), Yale School of Medicine, New Haven, CT
- Division of Vascular and Endovascular Surgery, Department of Surgery (A.D.), Yale School of Medicine, New Haven, CT
- Department of Surgery, VA Connecticut Healthcare Systems, West Haven, CT (A.D.)
| |
Collapse
|
26
|
Matsubara Y, Kiwan G, Fereydooni A, Langford J, Dardik A. Distinct subsets of T cells and macrophages impact venous remodeling during arteriovenous fistula maturation. JVS Vasc Sci 2020; 1:207-218. [PMID: 33748787 PMCID: PMC7971420 DOI: 10.1016/j.jvssci.2020.07.005] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Patients with end-stage renal failure depend on hemodialysis indefinitely without renal transplantation, requiring a long-term patent vascular access. While the arteriovenous fistula (AVF) remains the preferred vascular access for hemodialysis because of its longer patency and fewer complications compared with other vascular accesses, the primary patency of AVF is only 50-60%, presenting a clinical need for improvement. AVF mature by developing a thickened vascular wall and increased diameter to adapt to arterial blood pressure and flow volume. Inflammation plays a critical role during vascular remodeling and fistula maturation; increased shear stress triggers infiltration of T-cells and macrophages that initiate inflammation, with involvement of several different subsets of T-cells and macrophages. We review the literature describing distinct roles of the various subsets of T-cells and macrophages during vascular remodeling. Immunosuppression with sirolimus or prednisolone reduces neointimal hyperplasia during AVF maturation, suggesting novel approaches to enhance vascular remodeling. However, M2 macrophages and CD4+ T-cells play essential roles during AVF maturation, suggesting that total immunosuppression may suppress adaptive vascular remodeling. Therefore it is likely that regulation of inflammation during fistula maturation will require a balanced approach to coordinate the various inflammatory cell subsets. Advances in immunosuppressive drug development and delivery systems may allow for more targeted regulation of inflammation to improve vascular remodeling and enhance AVF maturation.
Collapse
Affiliation(s)
- Yutaka Matsubara
- Vascular Biology and Therapeutics Program, Yale School of Medicine, New Haven, CT.,Department of Surgery and Sciences, Kyushu University, Fukuoka, Japan
| | - Gathe Kiwan
- Vascular Biology and Therapeutics Program, Yale School of Medicine, New Haven, CT
| | - Arash Fereydooni
- Vascular Biology and Therapeutics Program, Yale School of Medicine, New Haven, CT
| | - John Langford
- Vascular Biology and Therapeutics Program, Yale School of Medicine, New Haven, CT
| | - Alan Dardik
- Vascular Biology and Therapeutics Program, Yale School of Medicine, New Haven, CT.,Division of Vascular and Endovascular Surgery, Department of Surgery, Yale School of Medicine, New Haven, CT.,Department of Surgery, VA Connecticut Healthcare Systems, West Haven, CT
| |
Collapse
|
27
|
Taniguchi R, Ono S, Isaji T, Gorecka J, Lee SR, Matsubara Y, Yatsula B, Koizumi J, Nishibe T, Hoshina K, Dardik A. A mouse model of stenosis distal to an arteriovenous fistula recapitulates human central venous stenosis. JVS Vasc Sci 2020; 1:109-122. [PMID: 33543148 PMCID: PMC7857464 DOI: 10.1016/j.jvssci.2020.07.003] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/02/2022] Open
Abstract
Objective Central venous stenosis (CVS) is a major cause of arteriovenous fistula (AVF) failure. However, central veins are relatively inaccessible to study with conventional Doppler ultrasound methods. To understand mechanisms underlying AVF failure owing to CVS, an animal model was established that creates a stenosis distal to an AVF. We hypothesized that this mouse model will show comparable morphology and physiology to human CVS. Methods An aortocaval fistula was created between the distal aorta and inferior vena cava (IVC); a stenosis was then created distal to the fistula by partial IVC ligation. Sham-operated animals, AVF without venous stenosis, and venous stenosis without AVF were used as controls. Physiologic properties of the IVC, both upstream and downstream of the stenosis, or the corresponding sites in models without stenosis, were assessed with ultrasound examination on days 0 to 21. The spectral broadening index was measured to assess the degree of disturbed shear stress. The IVC was harvested at day 21 and specimens were analyzed with immunofluorescence. Results The IVC diameter of mice with an AVF and stenosis showed increased upstream (P = .013), but decreased downstream diameter (P = .001) compared with mice with an AVF but without a stenosis, at all postoperative times (days 3-21). IVC wall thickness increased in mice with an AVF, compared with IVC without an AVF (upstream of stenosis: 13.9 μm vs 11.0 μm vs 4.5 μm vs 3.9 μm; P = .020; downstream of stenosis: 6.0 μm vs 6.6 μm vs μm 4.5 μm vs 3.8 μm; P = .002; AVF with stenosis, AVF, stenosis, sham, respectively). AVF patency significantly decreased in mice with an AVF and stenosis by day 21 (50% vs 90%; P = .048). The IVC of mice with AVF and stenosis showed a venous waveform with pulsatility as well as enhanced velocity at and downstream of the stenosis; similar waveforms were observed in a human case of CVS. Downstream to the stenosis, the spectral broadening index was significantly higher compared with mice with AVF alone (1.06 vs 0.78; P = .011; day 21), and there was a trend towards less immunoreactivity of both Krüppel-like factor 2 and phosphorylated-endothelial nitric oxide synthase compared with mice with an AVF alone. Conclusions Partial IVC ligation distal to a mouse aortocaval fistula alters the fistula diameter and wall thickness, decreases patency, and increases distal disturbed flow compared with fistulae without a distal stenosis. Our mouse model of stenosis distal to an AVF may be a faithful representation of human CVS that shows similar morphology and physiology, including disturbed shear stress. A mouse model of venous stenosis distal to an arteriovenous fistula shows similar Doppler waveforms as those observed in a human case of central venous stenosis. These mice retain disturbed shear stress in the vein distal to the fistula, characterized by a sustained increase of the spectral broadening index and diminished expression of proteins upregulated by laminar shear stress. This novel mouse model will enable investigation of the physiology and downstream molecular pathways involved in central venous stenosis in humans.
Collapse
Affiliation(s)
- Ryosuke Taniguchi
- Vascular Biology and Therapeutics Program, Yale School of Medicine, New Haven.,Division of Vascular Surgery, The University of Tokyo, Bunkyo-ku, Tokyo
| | - Shun Ono
- Vascular Biology and Therapeutics Program, Yale School of Medicine, New Haven.,Department of Diagnostic Radiology, Tokai University School of Medicine, Isehara, Kanagawa
| | - Toshihiko Isaji
- Vascular Biology and Therapeutics Program, Yale School of Medicine, New Haven.,Division of Vascular Surgery, The University of Tokyo, Bunkyo-ku, Tokyo
| | - Jolanta Gorecka
- Vascular Biology and Therapeutics Program, Yale School of Medicine, New Haven
| | - Shin-Rong Lee
- Vascular Biology and Therapeutics Program, Yale School of Medicine, New Haven
| | - Yutaka Matsubara
- Vascular Biology and Therapeutics Program, Yale School of Medicine, New Haven.,Department of Surgery and Sciences, Kyushu University, Fukuoka
| | - Bogdan Yatsula
- Vascular Biology and Therapeutics Program, Yale School of Medicine, New Haven
| | - Jun Koizumi
- Department of Diagnostic Radiology, Tokai University School of Medicine, Isehara, Kanagawa
| | - Toshiya Nishibe
- Department of Cardiovascular Surgery, Tokyo Medical University, Shinjuku-ku, Tokyo
| | - Katsuyuki Hoshina
- Division of Vascular Surgery, The University of Tokyo, Bunkyo-ku, Tokyo
| | - Alan Dardik
- Vascular Biology and Therapeutics Program, Yale School of Medicine, New Haven.,Division of Vascular and Endovascular Surgery, Department of Surgery, Yale School of Medicine, New Haven.,Department of Surgery, VA Connecticut Healthcare Systems, West Haven
| |
Collapse
|
28
|
Gorecka J, Fereydooni A, Gonzalez L, Lee SR, Liu S, Ono S, Xu J, Liu J, Taniguchi R, Matsubara Y, Gao X, Gao M, Langford J, Yatsula B, Dardik A. Molecular Targets for Improving Arteriovenous Fistula Maturation and Patency. VASCULAR INVESTIGATION AND THERAPY 2019; 2:33-41. [PMID: 31608322 DOI: 10.4103/vit.vit_9_19] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
The increasing prevalence of chronic and end-stage renal disease creates an increased need for reliable vascular access, and although arteriovenous fistulae (AVF) are the preferred mode of hemodialysis access, 60% fail to mature and only 50% remain patent at one year. Fistulae mature by diameter expansion and wall thickening; this outward remodeling of the venous wall in the fistula environment relies on a delicate balance of extracellular matrix (ECM) remodeling, inflammation, growth factor secretion, and cell adhesion molecule upregulation in the venous wall. AVF failure occurs via two distinct mechanisms with early failure secondary to lack of outward remodeling, that is insufficient diameter expansion or wall thickening, whereas late failure occurs with excessive wall thickening due to neointimal hyperplasia (NIH) and insufficient diameter expansion in a previously functional fistula. In recent years, the molecular basis of AVF maturation and failure are becoming understood in order to develop potential therapeutic targets to aide maturation and prevent access loss. Erythropoietin-producing hepatocellular carcinoma (Eph) receptors, along with their ligands, ephrins, determine vascular identity and are critical for vascular remodeling in the embryo. Manipulation of Eph receptor signaling in adults, as well as downstream pathways, is a potential treatment strategy to improve the rates of AVF maturation and patency. This review examines our current understanding of molecular changes occurring following fistula creation, factors predictive of fistula success, and potential areas of intervention to decrease AVF failure.
Collapse
Affiliation(s)
- Jolanta Gorecka
- Vascular Biology and Therapeutics Program and the Department of Surgery, Yale School of Medicine, Yale University, New Haven, USA
| | - Arash Fereydooni
- Vascular Biology and Therapeutics Program and the Department of Surgery, Yale School of Medicine, Yale University, New Haven, USA
| | - Luis Gonzalez
- Vascular Biology and Therapeutics Program and the Department of Surgery, Yale School of Medicine, Yale University, New Haven, USA
| | - Shin Rong Lee
- Vascular Biology and Therapeutics Program and the Department of Surgery, Yale School of Medicine, Yale University, New Haven, USA
| | - Shirley Liu
- Vascular Biology and Therapeutics Program and the Department of Surgery, Yale School of Medicine, Yale University, New Haven, USA
| | - Shun Ono
- Vascular Biology and Therapeutics Program and the Department of Surgery, Yale School of Medicine, Yale University, New Haven, USA
| | - Jianbiao Xu
- Vascular Biology and Therapeutics Program and the Department of Surgery, Yale School of Medicine, Yale University, New Haven, USA
| | - Jia Liu
- Vascular Biology and Therapeutics Program and the Department of Surgery, Yale School of Medicine, Yale University, New Haven, USA.,The Third Xiangya Hospital, Central South University, Changsha, Hunan, China.,Department of Vascular Surgery, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Ryosuke Taniguchi
- Vascular Biology and Therapeutics Program and the Department of Surgery, Yale School of Medicine, Yale University, New Haven, USA
| | - Yutaka Matsubara
- Vascular Biology and Therapeutics Program and the Department of Surgery, Yale School of Medicine, Yale University, New Haven, USA
| | - Xixiang Gao
- Vascular Biology and Therapeutics Program and the Department of Surgery, Yale School of Medicine, Yale University, New Haven, USA.,Department of Vascular Surgery, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Mingjie Gao
- Vascular Biology and Therapeutics Program and the Department of Surgery, Yale School of Medicine, Yale University, New Haven, USA.,Department of Vascular Ultrasonography, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - John Langford
- Vascular Biology and Therapeutics Program and the Department of Surgery, Yale School of Medicine, Yale University, New Haven, USA
| | - Bogdan Yatsula
- Vascular Biology and Therapeutics Program and the Department of Surgery, Yale School of Medicine, Yale University, New Haven, USA
| | - Alan Dardik
- Vascular Biology and Therapeutics Program and the Department of Surgery, Yale School of Medicine, Yale University, New Haven, USA.,Section of Vascular and Endovascular Surgery, VA Connecticut Healthcare System, West Haven, USA
| |
Collapse
|