1
|
Applewhite B, Andreopoulos F, Vazquez-Padron RI. Periadventitial biomaterials to improve arteriovenous fistula and graft outcomes. J Vasc Access 2024; 25:713-727. [PMID: 36349745 DOI: 10.1177/11297298221135621] [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: 02/17/2024] Open
Abstract
Periadventitial biomaterials have been employed for nearly three decades to promote adaptive venous remodeling following hemodialysis vascular access creation in preclinical models and clinical trials. These systems are predicated on the combination of scaffolds, hydrogels, and/or particles with therapeutics (small molecules, proteins, genes, and cells) to prevent venous stenosis and subsequent maturation failure. Periadventitial biomaterial therapies have evolved from simple drug delivery vehicles for traditional drugs to more thoughtful designs tailored to the pathophysiology of access failure. The emergence of tissue engineering strategies and gene therapies are another exciting new direction. Despite favorable results in experimental and preclinical studies, no periadventitial therapy has been clinically approved to improve vascular access outcomes. After conducting an exhaustive review of the literature, we identify the seminal studies and clinical trials that utilize periadventitial biomaterials and discuss the key features of each biomaterial format and their respective shortcomings as they pertain to access maturation. This review provides a foundation from which clinicians, surgeons, biologists, and engineers can refer to and will hopefully inspire thoughtful, translatable treatments to finally address access failure.
Collapse
Affiliation(s)
- Brandon Applewhite
- DeWitt Daughtry Family Department of Surgery, Leonard M. Miller School of Medicine, University of Miami, Miami, FL, USA
- Department of Biomedical Engineering, University of Miami College of Engineering, Coral Gables, FL, USA
| | - Fotios Andreopoulos
- DeWitt Daughtry Family Department of Surgery, Leonard M. Miller School of Medicine, University of Miami, Miami, FL, USA
- Department of Biomedical Engineering, University of Miami College of Engineering, Coral Gables, FL, USA
| | - Roberto I Vazquez-Padron
- Department of Biomedical Engineering, University of Miami College of Engineering, Coral Gables, FL, USA
| |
Collapse
|
2
|
Taurisano M, Mancini A, D'elia F. CEUS-guided PTA on stenotic AVF: Morphological and functional point of view. J Vasc Access 2024; 25:576-583. [PMID: 36217665 DOI: 10.1177/11297298221126289] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
BACKGROUND Stenosis represents the main cause of hemodialysis fistula malfunction. The ultrasound-guided angioplasty with ecographic contrast (CEUS) could provide further advantages to the classical ultrasound guided method improving the morphological characterization of the stenosis and providing quantitative data with the creation of time intensity curves (TIC) collecting functional data comparable between pre and post procedure. METHODS A total of 10 CEUS-guided angioplasties were performed on malfunctioning fistulas. The sonographic contrast medium was injected into the vascular tree trough the introducer. Morphological and functional data nature were collected. Were generated TIC curves, obtained by positioning a ROI in correspondence with the post-stenotic tract of the efferent vein. The data collected, regarding the peak intensity reached by the signal (PI) and the time to reach the peak signal intensity (TTP), were compared in the pre and post-procedural phase with flow of vascular access (Qa) and resistance indices (RI). RESULTS Statistically significant correlation (p < 0.05) was observed between Qa and TTP (r = 0.77; p = 0.009), RI and TTP (r = 0.71; p = 0.02), Qa and PI (r = 0.86; p = 0.0012), and between RI and PI (r = 0.88; p < 0.001). CONCLUSION In addition to the advantages associated with the use of ultrasound contrast medium in improving the visualization and characterization of the stenosis by facilitating the PTA procedure, the functional data deriving from the quantitative analysis provide new parameters for evaluating the success of the procedure which could also be used as predictive markers of stenosis recurrence together with the classical ones.
Collapse
Affiliation(s)
- Marco Taurisano
- Department of Nephrology, Hospital Di Venere and John XXIII, Bari, Italy
| | - Andrea Mancini
- Department of Nephrology, Hospital Di Venere and John XXIII, Bari, Italy
| | - Filomena D'elia
- Department of Nephrology, Hospital Di Venere and John XXIII, Bari, Italy
| |
Collapse
|
3
|
Xiao Y, Vazquez-Padron RI, Martinez L, Singer HA, Woltmann D, Salman LH. Role of platelet factor 4 in arteriovenous fistula maturation failure: What do we know so far? J Vasc Access 2024; 25:390-406. [PMID: 35751379 PMCID: PMC9974241 DOI: 10.1177/11297298221085458] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
The rate of arteriovenous fistula (AVF) maturation failure remains unacceptably high despite continuous efforts on technique improvement and careful pre-surgery planning. In fact, half of all newly created AVFs are unable to be used for hemodialysis (HD) without a salvage procedure. While vascular stenosis in the venous limb of the access is the culprit, the underlying factors leading to vascular narrowing and AVF maturation failure are yet to be determined. We have recently demonstrated that AVF non-maturation is associated with post-operative medial fibrosis and fibrotic stenosis, and post-operative intimal hyperplasia (IH) exacerbates the situation. Multiple pathological processes and signaling pathways are underlying the stenotic remodeling of the AVF. Our group has recently indicated that a pro-inflammatory cytokine platelet factor 4 (PF4/CXCL4) is upregulated in veins that fail to mature after AVF creation. Platelet factor 4 is a fibrosis marker and can be detected in vascular stenosis tissue, suggesting that it may contribute to AVF maturation failure through stimulation of fibrosis and development of fibrotic stenosis. Here, we present an overview of the how PF4-mediated fibrosis determines AVF maturation failure.
Collapse
Affiliation(s)
- Yuxuan Xiao
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, NY, USA
| | - Roberto I Vazquez-Padron
- DeWitt Daughtry Family Department of Surgery, Leonard M. Miller School of Medicine, University of Miami, Miami, FL, USA
| | - Laisel Martinez
- DeWitt Daughtry Family Department of Surgery, Leonard M. Miller School of Medicine, University of Miami, Miami, FL, USA
| | - Harold A Singer
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, NY, USA
| | - Daniel Woltmann
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, NY, USA
| | - Loay H Salman
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, NY, USA
- Division of Nephrology and Hypertension, Albany Medical College, Albany, NY, USA
| |
Collapse
|
4
|
Taurisano M, Mancini A, Cortese C, Napoli M. Endovascular tools for vascular access stenosis: Flow-chart proposal. J Vasc Access 2024:11297298241229166. [PMID: 38362739 DOI: 10.1177/11297298241229166] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/17/2024] Open
Abstract
Stenosis represents the most relevant arteriovenous fistula (AVF) pathology and can affects the entire conduit forming the fistula, from afferent artery to central venous vessels. Correction of vascular access stenosis significantly affects the survival and quality of life for end stage renal disease patients (ESRD) dependent on hemodialysis. Guidelines consider the procedure of percutaneous transluminal angioplasty (PTA) relevant for the primary treatment of these lesions with excellent results in restoring AVF immediately at the end of the procedure. From first AVF angioplasty in 1981 to now, wide scientific innovation has led to development of new devices, composed by different materials and technologies, specific for the site and the type of stenosis to be treated, able to manage resistant stenotic lesion and to reduce stenosis recurrences. International guidelines do not clearly specify all treatment possibilities in the individual case. In this review the authors want to provide specific information on most used devices for stenosis treatment based on literature evidence, showing when and where to use the various tools available with flow-chart treatment proposal.
Collapse
Affiliation(s)
- Marco Taurisano
- Department of Nephrology, Hospital "Di Venere," Bari (BA), Apulia, Italy
| | - Andrea Mancini
- Department of Nephrology, Hospital "Di Venere," Bari (BA), Apulia, Italy
| | - Cosma Cortese
- Department of Nephrology, AUOC Policlinico di Bari, Bari (BA), Apulia, Italy
| | - Marcello Napoli
- Department of Nephrology, Hospital "Vito Fazzi," Lecce (LE), Apulia, Italy
| |
Collapse
|
5
|
Barcena AJR, Perez JVD, Bernardino MR, Damasco JA, Cortes A, Del Mundo HC, San Valentin EMD, Klusman C, Canlas GM, Heralde FM, Avritscher R, Fowlkes N, Bouchard RR, Cheng J, Huang SY, Melancon MP. Bioresorbable Mesenchymal Stem Cell-Loaded Electrospun Polymeric Scaffold Inhibits Neointimal Hyperplasia Following Arteriovenous Fistula Formation in a Rat Model of Chronic Kidney Disease. Adv Healthc Mater 2023; 12:e2300960. [PMID: 37395729 PMCID: PMC10592251 DOI: 10.1002/adhm.202300960] [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: 03/27/2023] [Revised: 06/18/2023] [Accepted: 06/30/2023] [Indexed: 07/04/2023]
Abstract
Bioresorbable perivascular scaffolds loaded with antiproliferative agents have been shown to enhance arteriovenous fistula (AVF) maturation by inhibiting neointimal hyperplasia (NIH). These scaffolds, which can mimic the three-dimensional architecture of the vascular extracellular matrix, also have an untapped potential for the local delivery of cell therapies against NIH. Hence, an electrospun perivascular scaffold from polycaprolactone (PCL) to support mesenchymal stem cell (MSC) attachment and gradual elution at the AVF's outflow vein is fabricated. Chronic kidney disease (CKD) in Sprague-Dawley rats is induced by performing 5/6th nephrectomy, then AVFs for scaffold application are created. The following groups of CKD rats are compared: no perivascular scaffold (i.e., control), PCL alone, and PCL+MSC scaffold. PCL and PCL+MSC significantly improve ultrasonographic (i.e., luminal diameter, wall-to-lumen ratio, and flow rate) and histologic (i.e., neointima-to-lumen ratio, neointima-to-media ratio) parameters compared to control, with PCL+MSC demonstrating further improvement in these parameters compared to PCL alone. Moreover, only PCL+MSC significantly reduces 18 F-fluorodeoxyglucose uptake on positron emission tomography. These findings suggest that adding MSCs promotes greater luminal expansion and potentially reduces the inflammatory process underlying NIH. The results demonstrate the utility of mechanical support loaded with MSCs at the outflow vein immediately after AVF formation to support maturation by minimizing NIH.
Collapse
Affiliation(s)
- Allan John R Barcena
- Department of Interventional Radiology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
- College of Medicine, University of the Philippines Manila, Manila NCR, 1000, Philippines
| | - Joy Vanessa D Perez
- Department of Interventional Radiology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
- College of Medicine, University of the Philippines Manila, Manila NCR, 1000, Philippines
| | - Marvin R Bernardino
- Department of Interventional Radiology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Jossana A Damasco
- Department of Interventional Radiology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Andrea Cortes
- Department of Interventional Radiology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Huckie C Del Mundo
- Department of Interventional Radiology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Erin Marie D San Valentin
- Department of Interventional Radiology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Carleigh Klusman
- Department of Interventional Radiology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
- School of Medicine, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Gino Martin Canlas
- Department of Chemistry, Lamar University, P.O. Box 10009, Beaumont, TX, 77710, USA
| | - Francisco M Heralde
- College of Medicine, University of the Philippines Manila, Manila NCR, 1000, Philippines
| | - Rony Avritscher
- Department of Interventional Radiology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Natalie Fowlkes
- Department of Veterinary Medicine and Surgery, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Richard R Bouchard
- Department of Imaging Physics, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Jizhong Cheng
- Section of Nephrology, Department of Medicine, Selzman Institute for Kidney Health, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Steven Y Huang
- Department of Interventional Radiology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Marites P Melancon
- Department of Interventional Radiology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
- The University of Texas MD Anderson Cancer Center UTHealth Houston Graduate School of Biomedical Sciences, Houston, TX, 77030, USA
| |
Collapse
|
6
|
Yu T, Tang W, Hung W, Lee T, Tsai I, Hsuan C, Chen C, Chung F, Lee Y, Wu C. Elevated plasma leptin levels are associated with vascular access dysfunction in patients on maintenance hemodialysis. J Clin Lab Anal 2023; 37:e24974. [PMID: 37840358 PMCID: PMC10681410 DOI: 10.1002/jcla.24974] [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: 05/09/2023] [Revised: 09/16/2023] [Accepted: 10/03/2023] [Indexed: 10/17/2023] Open
Abstract
BACKGROUND Inflammation has been associated with vascular access (VA) dysfunction. The adipocytokine leptin can directly induce pro-inflammatory T helper 1 immune responses and the pathogenesis of chronic inflammation. We explored the association between plasma leptin and VA dysfunction in patients on maintenance hemodialysis (HEMO). METHODS A total of 344 consecutive patients who received anastomosis for VA at a single HEMO center between June 1, 2010 and December 31, 2021 were screened. Of these patients, 267 met the inclusion criteria and were included. ELISA was used to measure circulating levels of leptin. RESULTS The VA dysfunction group had a higher leptin level than the patent VA group. A higher concentration of leptin was independently and significantly associated with an elevated risk of VA dysfunction. Multiple logistic regression analysis showed that leptin, female sex, and hypertension were independently associated with VA dysfunction, even after adjusting for known biomarkers. We then evaluated the ability of leptin, female sex, and hypertension to predict the risk of VA dysfunction, and the area under the curve (AUC) for leptin was 0.626 (p = 0.0001). When leptin, female sex, and hypertension were added to this multivariate model, the AUC increased to 0.679 (p = 0.001) for leptin and hypertension, and 0.690 for leptin, hypertension, and female sex (p = 0.004). In addition, plasma leptin levels were associated with sex, body mass index, and hemoglobin. CONCLUSIONS In addition to the association between leptin and VA dysfunction, hypertension and female sex independently predicted VA dysfunction in patients with HEMO.
Collapse
Affiliation(s)
- Teng‐Hung Yu
- Division of Cardiology, Department of Internal MedicineE‐Da Hospital, I‐Shou UniversityKaohsiungTaiwan
- School of Medicine, College of MedicineI‐Shou UniversityKaohsiungTaiwan
| | - Wei‐Hua Tang
- Division of Cardiology, Department of Internal MedicineTaipei Veterans General Hospital, Yuli BranchHualienTaiwan
- Faculty of Medicine, School of MedicineNational Yang Ming Chiao Tung UniversityTaipeiTaiwan
| | - Wei‐Chin Hung
- Division of Cardiology, Department of Internal MedicineE‐Da Hospital, I‐Shou UniversityKaohsiungTaiwan
- School of Medicine, College of MedicineI‐Shou UniversityKaohsiungTaiwan
| | - Thung‐Lip Lee
- Division of Cardiology, Department of Internal MedicineE‐Da Hospital, I‐Shou UniversityKaohsiungTaiwan
- School of Medicine for International Students, College of MedicineI‐Shou UniversityKaohsiungTaiwan
| | - I‐Ting Tsai
- School of Medicine, College of MedicineI‐Shou UniversityKaohsiungTaiwan
- Department of EmergencyE‐Da Hospital, I‐Shou UniversityKaohsiungTaiwan
| | - Chin‐Feng Hsuan
- Division of Cardiology, Department of Internal MedicineE‐Da Hospital, I‐Shou UniversityKaohsiungTaiwan
- School of Medicine, College of MedicineI‐Shou UniversityKaohsiungTaiwan
- Division of Cardiology, Department of Internal MedicineE‐Da Dachang Hospital, I‐Shou UniversityKaohsiungTaiwan
| | - Chia‐Chi Chen
- School of Medicine, College of MedicineI‐Shou UniversityKaohsiungTaiwan
- Department of PathologyE‐Da Hospital, I‐Shou UniversityKaohsiungTaiwan
- Department of Physical TherapyI‐Shou UniversityKaohsiungTaiwan
- The School of Chinese Medicine for Post Baccalaureate, College of MedicineI‐Shou UniversityKaohsiungTaiwan
| | - Fu‐Mei Chung
- Division of Cardiology, Department of Internal MedicineE‐Da Hospital, I‐Shou UniversityKaohsiungTaiwan
| | | | - Cheng‐Ching Wu
- Division of Cardiology, Department of Internal MedicineE‐Da Hospital, I‐Shou UniversityKaohsiungTaiwan
- School of Medicine, College of MedicineI‐Shou UniversityKaohsiungTaiwan
- Division of Cardiology, Department of Internal MedicineE‐Da Cancer Hospital, I‐Shou UniversityKaohsiungTaiwan
| |
Collapse
|
7
|
Barcena AJR, Perez JVD, Damasco JA, Bernardino MR, San Valentin EMD, Klusman C, Martin B, Cortes A, Canlas GM, Del Mundo HC, Heralde FM, Avritscher R, Fowlkes N, Bouchard RR, Cheng J, Huang SY, Melancon MP. Gold Nanoparticles for Monitoring of Mesenchymal Stem-Cell-Loaded Bioresorbable Polymeric Wraps for Arteriovenous Fistula Maturation. Int J Mol Sci 2023; 24:11754. [PMID: 37511512 PMCID: PMC10380871 DOI: 10.3390/ijms241411754] [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: 06/13/2023] [Revised: 06/30/2023] [Accepted: 07/04/2023] [Indexed: 07/30/2023] Open
Abstract
Mesenchymal stem cell (MSC)-seeded polymeric perivascular wraps have been shown to enhance arteriovenous fistula (AVF) maturation. However, the wraps' radiolucency makes their placement and integrity difficult to monitor. Through electrospinning, we infused gold nanoparticles (AuNPs) into polycaprolactone (PCL) wraps to improve their radiopacity and tested whether infusion affects the previously reported beneficial effects of the wraps on the AVF's outflow vein. Sprague Dawley rat MSCs were seeded on the surface of the wraps. We then compared the effects of five AVF treatments-no perivascular wrap (i.e., control), PCL wrap, PCL + MSC wrap, PCL-Au wrap, and PCL-Au + MSC wrap-on AVF maturation in a Sprague Dawley rat model of chronic kidney disease (n = 3 per group). Via micro-CT, AuNP-infused wraps demonstrated a significantly higher radiopacity compared to that of the wraps without AuNPs. Wraps with and without AuNPs equally reduced vascular stenoses, as seen via ultrasonography and histomorphometry. In the immunofluorescence analysis, representative MSC-seeded wraps demonstrated reduced neointimal staining for markers of infiltration with smooth muscle cells (α-SMA), inflammatory cells (CD45), and fibroblasts (vimentin) compared to that of the control and wraps without MSCs. In conclusion, AuNP infusion allows in vivo monitoring via micro-CT of MSC-seeded polymeric wraps over time, without compromising the benefits of the wrap for AVF maturation.
Collapse
Affiliation(s)
- Allan John R. Barcena
- Department of Interventional Radiology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; (A.J.R.B.); (J.V.D.P.); (J.A.D.); (M.R.B.); (E.M.D.S.V.); (B.M.); (A.C.); (H.C.D.M.); (R.A.); (S.Y.H.)
- College of Medicine, University of the Philippines Manila, Manila 1000, Philippines;
| | - Joy Vanessa D. Perez
- Department of Interventional Radiology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; (A.J.R.B.); (J.V.D.P.); (J.A.D.); (M.R.B.); (E.M.D.S.V.); (B.M.); (A.C.); (H.C.D.M.); (R.A.); (S.Y.H.)
- College of Medicine, University of the Philippines Manila, Manila 1000, Philippines;
| | - Jossana A. Damasco
- Department of Interventional Radiology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; (A.J.R.B.); (J.V.D.P.); (J.A.D.); (M.R.B.); (E.M.D.S.V.); (B.M.); (A.C.); (H.C.D.M.); (R.A.); (S.Y.H.)
| | - Marvin R. Bernardino
- Department of Interventional Radiology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; (A.J.R.B.); (J.V.D.P.); (J.A.D.); (M.R.B.); (E.M.D.S.V.); (B.M.); (A.C.); (H.C.D.M.); (R.A.); (S.Y.H.)
| | - Erin Marie D. San Valentin
- Department of Interventional Radiology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; (A.J.R.B.); (J.V.D.P.); (J.A.D.); (M.R.B.); (E.M.D.S.V.); (B.M.); (A.C.); (H.C.D.M.); (R.A.); (S.Y.H.)
| | - Carleigh Klusman
- Department of Interventional Radiology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; (A.J.R.B.); (J.V.D.P.); (J.A.D.); (M.R.B.); (E.M.D.S.V.); (B.M.); (A.C.); (H.C.D.M.); (R.A.); (S.Y.H.)
- School of Medicine, Baylor College of Medicine, Houston, TX 77030, USA
| | - Benjamin Martin
- Department of Interventional Radiology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; (A.J.R.B.); (J.V.D.P.); (J.A.D.); (M.R.B.); (E.M.D.S.V.); (B.M.); (A.C.); (H.C.D.M.); (R.A.); (S.Y.H.)
- School of Medicine, Baylor College of Medicine, Houston, TX 77030, USA
| | - Andrea Cortes
- Department of Interventional Radiology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; (A.J.R.B.); (J.V.D.P.); (J.A.D.); (M.R.B.); (E.M.D.S.V.); (B.M.); (A.C.); (H.C.D.M.); (R.A.); (S.Y.H.)
| | | | - Huckie C. Del Mundo
- Department of Interventional Radiology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; (A.J.R.B.); (J.V.D.P.); (J.A.D.); (M.R.B.); (E.M.D.S.V.); (B.M.); (A.C.); (H.C.D.M.); (R.A.); (S.Y.H.)
| | - Francisco M. Heralde
- College of Medicine, University of the Philippines Manila, Manila 1000, Philippines;
| | - Rony Avritscher
- Department of Interventional Radiology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; (A.J.R.B.); (J.V.D.P.); (J.A.D.); (M.R.B.); (E.M.D.S.V.); (B.M.); (A.C.); (H.C.D.M.); (R.A.); (S.Y.H.)
| | - Natalie Fowlkes
- Department of Veterinary Medicine and Surgery, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA;
| | - Richard R. Bouchard
- Department of Imaging Physics, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA;
| | - Jizhong Cheng
- Section of Nephrology, Department of Medicine, Selzman Institute for Kidney Health, Baylor College of Medicine, Houston, TX 77030, USA;
| | - Steven Y. Huang
- Department of Interventional Radiology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; (A.J.R.B.); (J.V.D.P.); (J.A.D.); (M.R.B.); (E.M.D.S.V.); (B.M.); (A.C.); (H.C.D.M.); (R.A.); (S.Y.H.)
| | - Marites P. Melancon
- Department of Interventional Radiology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; (A.J.R.B.); (J.V.D.P.); (J.A.D.); (M.R.B.); (E.M.D.S.V.); (B.M.); (A.C.); (H.C.D.M.); (R.A.); (S.Y.H.)
- The University of Texas MD Anderson Cancer Center UTHealth Houston Graduate School of Biomedical Sciences, Houston, TX 77030, USA
| |
Collapse
|
8
|
Applewhite B, Gupta A, Wei Y, Yang X, Martinez L, Rojas MG, Andreopoulos F, Vazquez-Padron RI. Periadventitial β-aminopropionitrile-loaded nanofibers reduce fibrosis and improve arteriovenous fistula remodeling in rats. Front Cardiovasc Med 2023; 10:1124106. [PMID: 36926045 PMCID: PMC10011136 DOI: 10.3389/fcvm.2023.1124106] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Accepted: 02/07/2023] [Indexed: 03/04/2023] Open
Abstract
Background Arteriovenous fistula (AVF) postoperative stenosis is a persistent healthcare problem for hemodialysis patients. We have previously demonstrated that fibrotic remodeling contributes to AVF non-maturation and lysyl oxidase (LOX) is upregulated in failed AVFs compared to matured. Herein, we developed a nanofiber scaffold for the periadventitial delivery of β-aminopropionitrile (BAPN) to determine whether unidirectional periadventitial LOX inhibition is a suitable strategy to promote adaptive AVF remodeling in a rat model of AVF remodeling. Methods Bilayer poly (lactic acid) ([PLA)-]- poly (lactic-co-glycolic acid) ([PLGA)] scaffolds were fabricated with using a two-step electrospinning process to confer directionality. BAPN-loaded and vehicle control scaffolds were wrapped around the venous limb of a rat femoral-epigastric AVF during surgery. AVF patency and lumen diameter were followed monitored using Doppler ultrasound surveillance and flow was measured before euthanasia. AVFs were harvested after 21 days for histomorphometry and immunohistochemistry. AVF compliance was measured using pressure myography. RNA from AVF veins was sequenced to analyze changes in gene expression due to LOX inhibition. Results Bilayer periadventitial nanofiber scaffolds extended BAPN release compared to the monolayer design (p < 0.005) and only released BAPN in one direction. Periadventitial LOX inhibition led to significant increases in AVF dilation and flow after 21 days. Histologically, BAPN trended toward increased lumen and significantly reduced fibrosis compared to control scaffolds (p < 0.01). Periadventitial BAPN reduced downregulated markers associated with myofibroblast differentiation including SMA, FSP-1, LOX, and TGF-β while increasing the contractile marker MYH11. RNA sequencing revealed differential expression of matrisome genes. Conclusion Periadventitial BAPN treatment reduces fibrosis and promotes AVF compliance. Interestingly, the inhibition of LOX leads to increased accumulation of contractile VSMC while reducing myofibroblast-like cells. Periadventitial LOX inhibition alters the matrisome to improve AVF vascular remodeling.
Collapse
Affiliation(s)
- Brandon Applewhite
- Department of Biomedical Engineering, University of Miami, Coral Gables, FL, United States
| | - Aavni Gupta
- Department of Surgery, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Yuntao Wei
- Department of Surgery, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Xiaofeng Yang
- Lewis Katz School of Medicine, Temple University, Philadelphia, PA, United States
| | - Laisel Martinez
- Department of Surgery, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Miguel G. Rojas
- Department of Surgery, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Fotios Andreopoulos
- Department of Biomedical Engineering, University of Miami, Coral Gables, FL, United States
- Department of Surgery, University of Miami Miller School of Medicine, Miami, FL, United States
| | | |
Collapse
|
9
|
Barcena AJR, Perez JVD, Damasco JA, Bernardino MR, San Valentin EMD, Klusman C, Martin B, Cortes A, Canlas G, Del Mundo HC, Heralde FM, Avritscher R, Fowlkes N, Bouchard RR, Cheng J, Huang SY, Melancon MP. Gold Nanoparticles for Monitoring of Mesenchymal Stem Cell-Loaded Bioresorbable Polymeric Wraps for Arteriovenous Fistulas. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.02.01.526611. [PMID: 36778466 PMCID: PMC9915579 DOI: 10.1101/2023.02.01.526611] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/09/2023]
Abstract
Background To address high rates of arteriovenous fistula (AVF) failure, a mesenchymal stem cell (MSC)-seeded polymeric perivascular wrap has been developed to reduce neointimal hyperplasia (NIH) and enhance AVF maturation in a rat model. However, the wrap's radiolucency makes its placement and integrity difficult to monitor. Purpose In this study, we infused gold nanoparticles (AuNPs) into the polymeric perivascular wrap to improve its radiopacity and tested the effect of infusion on the previously reported beneficial effects of the polymeric wrap on the AVF outflow vein. Materials and Methods We fabricated a polymeric perivascular wrap made of polycaprolactone (PCL) infused with AuNPs via electrospinning. Sprague-Dawley rat mesenchymal stem cells (MSCs) were seeded on the surface of the wraps. We then compared the effect of five AVF treatments-no perivascular wrap (i.e., control), PCL wrap, PCL+MSC wrap, PCL-Au wrap, and PCL-Au+MSC wrap-on AVF maturation in a Sprague-Dawley rat model of chronic kidney disease (n=3 per group). Statistical significance was defined as p<.05, and one-way analysis of variance was performed using GraphPad Prism software. Results On micro-CT, AuNP-infused wraps demonstrated significantly higher radiopacity compared to wraps without AuNPs. On ultrasonography, wraps with and without AuNPs equally reduced the wall-to-lumen ratio of the outflow vein, a marker of vascular stenosis. On histomorphometric analysis, wraps with and without AuNPs equally reduced the neointima-to- lumen ratio of the outflow vein, a measure of NIH. On immunofluorescence analysis, representative MSC-seeded wraps demonstrated reduced neointimal staining for markers of smooth muscle cells (α-SMA), inflammatory cells (CD45), and fibroblasts (vimentin) infiltration when compared to control and wraps without MSCs. Conclusion Gold nanoparticle infusion allows the in vivo monitoring via micro-CT of a mesenchymal stem cell-seeded polymeric wrap over time without compromising the benefits of the wrap on arteriovenous fistula maturation. Summary Statement Gold nanoparticle infusion enables in vivo monitoring via micro-CT of the placement and integrity over time of mesenchymal stem cell-seeded polymeric wrap supporting arteriovenous fistula maturation. Key Results Gold nanoparticle (AuNP)-infused perivascular wraps demonstrated higher radiopacity on micro-CT compared with wraps without AuNPs after 8 weeks.AuNP-infused perivascular wraps equally improved the wall-to-lumen ratio of the outflow vein (a marker of vascular stenosis) when compared with wraps without AuNPs, as seen on US.AuNP-infused perivascular wraps equally reduced the neointima-to-lumen ratio of the outflow vein (a measure of neointimal hyperplasia) when compared with wraps without AuNPs, as seen on histomorphometry.
Collapse
|
10
|
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
|
11
|
Gao M, Wang J. Risk Factors of Arteriovenous Fistula Stenosis of Patients with Maintenance Hemodialysis. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE : ECAM 2022; 2022:2968122. [PMID: 36387371 PMCID: PMC9649314 DOI: 10.1155/2022/2968122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/03/2022] [Revised: 07/20/2022] [Accepted: 07/26/2022] [Indexed: 07/29/2023]
Abstract
Objective To investigate the risk factors of arteriovenous fistula stenosis in maintenance hemodialysis patients. Methods A total of 80 patients with maintenance hemodialysis in our hospital from January 2017 to January 2022 were included. According to the stenosis degree of the arteriovenous fistula, patients were divided into a stenosis group (n = 35) and an unobstructed group (n = 45). Laboratory examination data, such as the demographic characteristics of patients, past medical history, the use of traditional Chinese medicine (TCM), the TCM for improving blood circulation to promote blood circulation and remove blood stasis, and cellular immune function, were collected. Multivariate Logistic regression analysis was used to analyze the influencing factors. Results There was no significant difference in the age, sexuality, body mass index, proportion of patients with coronary heart disease, cephalic vein diameter, radial artery diameter, albumin, fibrinogen, and blood calcium levels between the two groups (P > 0.05). Compared with the unobstructed group, the stenosis group had higher rates of diabetes and hypertension as well as significantly higher levels of platelets, parathyroid hormone, and D-dimer (P < 0.05). The proportion of TCM application in the stenosis group was 28.6% (10/35), which was significantly lower than that in the unobstructed group 57.8% (26/45). The difference was statistically significant (χ 2 = 6.785, P=0.009). The levels of CD3+ (37.5 ± 5.1% vs. 42.0 ± 4.8%), CD4+ (35.7 ± 3.2% vs. 39.6 ± 3.1%) and CD4+/CD8+ (1.4 ± 0.3 vs. 2.4 ± 1.0) in the stenosis group were significantly lower, while CD8+ level was significantly higher (26.8 ± 5.1% vs. 18.5 ± 6.2%) than that in the unobstructed group (P < 0.001). Logistic regression analysis indicated that the usage of TCM was an independent risk factor for arteriovenous fistula stenosis after adjustment for other factors (odds ratio [OR] 0.302; 95% confidence interval [95% CI] 0.100-0.896; P=0.039). Conclusions Less use of traditional Chinese medicine for activating blood circulation and removing blood stasis and poor cellular immune function are important factors affecting the function of the arteriovenous fistula in patients with maintenance hemodialysis, which is worthy of clinical attention.
Collapse
Affiliation(s)
- Meiling Gao
- Blood Purification Center, Changzhou Second People's Hospital, Changzhou 213000, Jiangsu, China
| | - Jing Wang
- Blood Purification Center, Changzhou Second People's Hospital, Changzhou 213000, Jiangsu, China
| |
Collapse
|
12
|
Barcena AJR, Perez JVD, Liu O, Mu A, Heralde FM, Huang SY, Melancon MP. Localized Perivascular Therapeutic Approaches to Inhibit Venous Neointimal Hyperplasia in Arteriovenous Fistula Access for Hemodialysis Use. Biomolecules 2022; 12:biom12101367. [PMID: 36291576 PMCID: PMC9599524 DOI: 10.3390/biom12101367] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Accepted: 09/23/2022] [Indexed: 01/14/2023] Open
Abstract
An arteriovenous fistula (AVF) is the preferred vascular access for chronic hemodialysis, but high failure rates restrict its use. Optimizing patients' perioperative status and the surgical technique, among other methods for preventing primary AVF failure, continue to fall short in lowering failure rates in clinical practice. One of the predominant causes of AVF failure is neointimal hyperplasia (NIH), a process that results from the synergistic effects of inflammation, hypoxia, and hemodynamic shear stress on vascular tissue. Although several systemic therapies have aimed at suppressing NIH, none has shown a clear benefit towards this goal. Localized therapeutic approaches may improve rates of AVF maturation by providing direct structural and functional support to the maturating fistula, as well as by delivering higher doses of pharmacologic agents while avoiding the adverse effects associated with systemic administration of therapeutic agents. Novel materials-such as polymeric scaffolds and nanoparticles-have enabled the development of different perivascular therapies, such as supportive mechanical devices, targeted drug delivery, and cell-based therapeutics. In this review, we summarize various perivascular therapeutic approaches, available data on their effectiveness, and the outlook for localized therapies targeting NIH in the setting of AVF for hemodialysis use. Highlights: Most systemic therapies do not improve AVF patency outcomes; therefore, localized therapeutic approaches may be beneficial. Locally delivered drugs and medical devices may improve AVF patency outcomes by providing biological and mechanical support. Cell-based therapies have shown promise in suppressing NIH by delivering a more extensive array of bioactive substances in response to the biochemical changes in the AVF microenvironment.
Collapse
Affiliation(s)
- Allan John R. Barcena
- Department of Interventional Radiology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
- College of Medicine, University of the Philippines Manila, Manila 1000, Philippines
| | - Joy Vanessa D. Perez
- Department of Interventional Radiology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
- College of Medicine, University of the Philippines Manila, Manila 1000, Philippines
| | - Olivia Liu
- Department of Interventional Radiology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
- Grossman School of Medicine, New York University, New York, NY 10016, USA
| | - Amy Mu
- Department of Interventional Radiology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
- The University of Texas Southwestern Medical School, Dallas, TX 75390, USA
| | - Francisco M. Heralde
- College of Medicine, University of the Philippines Manila, Manila 1000, Philippines
| | - Steven Y. Huang
- Department of Interventional Radiology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Marites P. Melancon
- Department of Interventional Radiology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
- The University of Texas MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Houston, TX 77030, USA
- Correspondence:
| |
Collapse
|
13
|
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
|
14
|
Zhao Z, Fu Q, Hu L, Liu Y. Identification of the Crucial Gene in Overflow Arteriovenous Fistula by Bioinformatics Analysis. Front Physiol 2021; 12:621830. [PMID: 34421628 PMCID: PMC8371383 DOI: 10.3389/fphys.2021.621830] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2020] [Accepted: 06/25/2021] [Indexed: 11/13/2022] Open
Abstract
Objective: The aim was to study the preliminary screening of the crucial genes in intimal hyperplasia in the venous segment of arteriovenous (AV) fistula and the underlying potential molecular mechanisms of intimal hyperplasia with bioinformatics analysis. Methods: The gene expression profile data (GSE39488) was analyzed to identify differentially expressed genes (DEGs). We performed Gene Ontology and Kyoto Encyclopedia of Genes and Genomes pathway enrichment analysis of DEGs. Gene set enrichment analysis (GSEA) was used to understand the potential activated signaling pathway. The protein-protein interaction (PPI) network was constructed with the STRING database and Cytoscape software. The Venn diagram between 10 hub genes and gene sets of 4 crucial signaling pathways was used to obtain core genes and relevant potential pathways. Furthermore, GSEAs were performed to understand their biological functions. Results: A total of 185 DEGs were screened in this study. The main biological function of the 111 upregulated genes in AV fistula primarily concentrated on cell proliferation and vascular remodeling, and the 74 downregulated genes in AV fistula were enriched in the biological function mainly relevant to inflammation. GSEA found four signaling pathways crucial for intimal hyperplasia, namely, MAPK, NOD-like, Cell Cycle, and TGF-beta signaling pathway. A total of 10 hub genes were identified, namely, EGR1, EGR2, EGR3, NR4A1, NR4A2, DUSP1, CXCR4, ATF3, CCL4, and CYR61. Particularly, DUSP1 and NR4A1 were identified as core genes that potentially participate in the MAPK signaling pathway. In AV fistula, the biological processes and pathways were primarily involved with MAPK signaling pathway and MAPK-mediated pathway with the high expression of DUSP1 and were highly relevant to cell proliferation and inflammation with the low expression of DUSP1. Besides, the biological processes and pathways in AV fistula with the high expression of NR4A1 similarly included the MAPK signaling pathway and the pathway mediated by MAPK signaling, and it was mainly involved with inflammation in AV fistula with the low expression of NR4A1. Conclusion: We screened four potential signaling pathways relevant to intimal hyperplasia and identified 10 hub genes, including two core genes (i.e., DUSP1 and NR4A1). Two core genes potentially participate in the MAPK signaling pathway and might serve as the therapeutic targets of intimal hyperplasia to prevent stenosis after AV fistula creation.
Collapse
Affiliation(s)
- Zhengde Zhao
- First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Qining Fu
- First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Liangzhu Hu
- First Affiliated Hospital of Chongqing Medical University, Chongqing, China.,Department of Vascular Surgery, South China Hospital, Health Science Center, Shenzhen University, Shenzhen, China
| | - Yangdong Liu
- First Affiliated Hospital of Chongqing Medical University, Chongqing, China.,Department of Vascular Surgery, South China Hospital, Health Science Center, Shenzhen University, Shenzhen, China
| |
Collapse
|
15
|
Chen L, Zhang W, Tan J, Hu M, Shi W, Zhang M, Wang Y, Yu B, Chen J. Morphological Lesion Types Are Associated with Primary and Secondary Patency Rates after High-Pressure Balloon Angioplasty for Dysfunctional Arteriovenous Fistulas. Blood Purif 2021; 51:425-434. [PMID: 34320498 DOI: 10.1159/000516883] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2020] [Accepted: 04/07/2021] [Indexed: 11/19/2022]
Abstract
BACKGROUND Neointimal hyperplasia (NIH) is believed to be the main reason for arteriovenous fistula (AVF) dysfunction, but other mechanisms are also recognized to be involved in the pathophysiological process. This study investigated whether different morphological types of AVF lesions are associated with the patency rate after percutaneous transluminal angioplasty (PTA). METHODS This retrospective study included 120 patients who underwent PTA for autogenous AVF dysfunction. All the cases were evaluated under Doppler ultrasound (DU) before intervention and divided into 3 types: Type I (NIH type), Type II (non-NIH type), and Type III (mixed type). Prognostic and clinical data were analyzed by Kaplan-Meier analysis and the Cox proportional hazards model. RESULTS There was no statistical difference in baseline variables among groups, except for lumen diameter. The primary patency rates in Type I, Type II, and Type III groups were 78.4, 93.2, and 83.2% at 6 months and 59.5, 84.7, and 75.5% at 1 year, respectively. The secondary patency rates in Type I, Type II, and Type III groups were 94.4, 97.1, and 100% at 6 months and 90.5, 97.1, and 94.7% at 1 year, respectively. The Kaplan-Meier curve showed that the primary and secondary patency rates of Type I group were lower than those of Type II group. Multivariable Cox regression analysis demonstrated that postoperative primary patency was correlated with end-to-end anastomosis (hazard ratio [HR] = 2.997, p = 0.008, 95% confidence interval [CI]: 1.328-6.764) and Type I lesion (HR = 5.395, p = 0.004, 95% CI: 1.730-16.824). CONCLUSIONS NIH-dominant lesions of AVF evaluated by DU preoperatively were a risk factor for poor primary and secondary patency rate after PTA in hemodialysis patients.
Collapse
Affiliation(s)
- Li Chen
- Department of Ultrasound, Huashan Hospital, Fudan University, Shanghai, China
| | - Weichen Zhang
- Department of Nephrology, Huashan Hospital, Fudan University, Shanghai, China,
| | - Jinyun Tan
- Department of Vascular Surgery, Huashan Hospital, Fudan University, Shanghai, China
| | - Min Hu
- Department of Vascular Surgery, Huashan Hospital, Fudan University, Shanghai, China
| | - Weihao Shi
- Department of Vascular Surgery, Pudong Hospital, Fudan University, Shanghai, China
| | - Minmin Zhang
- Department of Nephrology, Huashan Hospital, Fudan University, Shanghai, China
| | - Yong Wang
- Department of Ultrasound, Huashan Hospital, Fudan University, Shanghai, China
| | - Bo Yu
- Department of Vascular Surgery, Huashan Hospital, Fudan University, Shanghai, China.,Department of Vascular Surgery, Pudong Hospital, Fudan University, Shanghai, China
| | - Jing Chen
- Department of Nephrology, Huashan Hospital, Fudan University, Shanghai, China
| |
Collapse
|
16
|
Mauro R, Rocchi C, Vasuri F, Pini A, Croci Chiocchini AL, Ciavarella C, La Manna G, Pasquinelli G, Faggioli G, Gargiulo M. Tissue Ki67 proliferative index expression and pathological changes in hemodialysis arteriovenous fistulae: Preliminary single-center results. J Vasc Access 2021; 24:391-396. [PMID: 34308698 DOI: 10.1177/11297298211015495] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
BACKGROUND Arteriovenous fistula (AVF) for hemodialysis integrates outward remodeling with vessel wall thickening in response to drastic hemodynamic changes. Aim of this study is to determine the role of Ki67, a well-established proliferative marker, related to AVF, and its relationship with time-dependent histological morphologic changes. MATERIALS AND METHODS All patients were enrolled in 1 year and stratified in two groups: (A) pre-dialysis patients submitted to first AVF and (B) patients submitted to revision of AVF. Morphological changes: neo-angiogenesis (NAG), myointimal thickening (MIT), inflammatory infiltrate (IT), and aneurysmatic fistula degeneration (AD). The time of AVF creation was recorded. A biopsy of native vein in Group A and of arterialized vein in Group B was submitted to histological and immunohistochemical (IHC) analysis. IHC for Ki67 was automatically performed in all specimens. Ki67 immunoreactivity was assessed as the mean number of positive cells on several high-power fields, counted in the hot spots. RESULTS A total of 138 patients were enrolled, 69 (50.0%) Group A and 69 (50.0%) Group B. No NAG or MIT were found in Group A. Seven (10.1%) Group A veins showed a mild MIT. Analyzing the Group B, a moderate-to-severe MIT was present in 35 (50.7%), IT in 19 (27.5%), NAG in 37 (53.6%); AD was present in 10 (14.5%). All AVF of Group B with the exception of one (1.4%) showed a positivity for Ki67, with a mean of 12.31 ± 13.79 positive cells/hot spot (range 0-65). Ki67-immunoreactive cells had a subendothelial localization in 23 (33.3%) cases, a myointimal localization in SMC in 35 (50.7%) cases. The number of positive cells was significantly correlated with subendothelial localization of Ki67 (p = 0.001) and with NA (p = 0.001). CONCLUSIONS Native veins do not contain cycling cells. In contrast, vascular cell proliferation starts immediately after AVF creation and persists independently of the time the fistula is set up. The amount of proliferating cells is significantly associated with MIT and subendothelial localization of Ki67-immunoreactive cells, thus suggesting a role of Ki-67 index in predicting AVF failure.
Collapse
Affiliation(s)
- Raffaella Mauro
- Department of Specialty, Diagnostic and Experimental Medicine, S. Orsola-Malpighi Hospital, University of Bologna, Bologna, Italy
| | - Cristina Rocchi
- Department of Specialty, Diagnostic and Experimental Medicine, S. Orsola-Malpighi Hospital, University of Bologna, Bologna, Italy
| | - Francesco Vasuri
- Department of Specialty, Diagnostic and Experimental Medicine, S. Orsola-Malpighi Hospital, University of Bologna, Bologna, Italy
| | - Alessia Pini
- Department of Specialty, Diagnostic and Experimental Medicine, S. Orsola-Malpighi Hospital, University of Bologna, Bologna, Italy
| | - Anna Laura Croci Chiocchini
- Department of Specialty, Diagnostic and Experimental Medicine, S. Orsola-Malpighi Hospital, University of Bologna, Bologna, Italy
| | - Carmen Ciavarella
- Department of Specialty, Diagnostic and Experimental Medicine, S. Orsola-Malpighi Hospital, University of Bologna, Bologna, Italy
| | - Gaetano La Manna
- Department of Specialty, Diagnostic and Experimental Medicine, S. Orsola-Malpighi Hospital, University of Bologna, Bologna, Italy
| | - Gianandrea Pasquinelli
- Department of Specialty, Diagnostic and Experimental Medicine, S. Orsola-Malpighi Hospital, University of Bologna, Bologna, Italy
| | - Gianluca Faggioli
- Department of Specialty, Diagnostic and Experimental Medicine, S. Orsola-Malpighi Hospital, University of Bologna, Bologna, Italy
| | - Mauro Gargiulo
- Department of Specialty, Diagnostic and Experimental Medicine, S. Orsola-Malpighi Hospital, University of Bologna, Bologna, Italy
| |
Collapse
|
17
|
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
|
18
|
Divella C, Stasi A, Franzin R, Rossini M, Pontrelli P, Sallustio F, Netti GS, Ranieri E, Lacitignola L, Staffieri F, Crovace AM, Lucarelli G, Ditonno P, Battaglia M, Daha MR, van der Pol P, van Kooten C, Grandaliano G, Gesualdo L, Stallone G, Castellano G. Pentraxin-3-mediated complement activation in a swine model of renal ischemia/reperfusion injury. Aging (Albany NY) 2021; 13:10920-10933. [PMID: 33875620 PMCID: PMC8109140 DOI: 10.18632/aging.202992] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2020] [Accepted: 03/26/2021] [Indexed: 11/30/2022]
Abstract
Pentraxins are a family of evolutionarily conserved pattern recognition molecules with pivotal roles in innate immunity and inflammation, such as opsonization of pathogens during bacterial and viral infections. In particular, the long Pentraxin 3 (PTX3) has been shown to regulate several aspects of vascular and tissue inflammation during solid organ transplantation. Our study investigated the role of PTX3 as possible modulator of Complement activation in a swine model of renal ischemia/reperfusion (I/R) injury. We demonstrated that I/R injury induced early PTX3 deposits at peritubular and glomerular capillary levels. Confocal laser scanning microscopy revealed PTX3 deposits co-localizing with CD31+ endothelial cells. In addition, PTX3 was associated with infiltrating macrophages (CD163), dendritic cells (SWC3a) and myofibroblasts (FSP1). In particular, we demonstrated a significant PTX3-mediated activation of classical (C1q-mediated) and lectin (MBL-mediated) pathways of Complement. Interestingly, PTX3 deposits co-localized with activation of the terminal Complement complex (C5b-9) on endothelial cells, indicating that PTX3-mediated Complement activation occurred mainly at the renal vascular level. In conclusion, these data indicate that PTX3 might be a potential therapeutic target to prevent Complement-induced I/R injury.
Collapse
Affiliation(s)
- Chiara Divella
- Renal, Dialysis and Transplantation Unit, Department of Emergency and Organ Transplantation, University of Bari, Bari, Italy
| | - Alessandra Stasi
- Renal, Dialysis and Transplantation Unit, Department of Emergency and Organ Transplantation, University of Bari, Bari, Italy
| | - Rossana Franzin
- Renal, Dialysis and Transplantation Unit, Department of Emergency and Organ Transplantation, University of Bari, Bari, Italy
| | - Michele Rossini
- Renal, Dialysis and Transplantation Unit, Department of Emergency and Organ Transplantation, University of Bari, Bari, Italy
| | - Paola Pontrelli
- Renal, Dialysis and Transplantation Unit, Department of Emergency and Organ Transplantation, University of Bari, Bari, Italy
| | - Fabio Sallustio
- Department of Interdisciplinary Medicine, University of Bari, Bari, Italy.,Department of Basic Medical Sciences, Neuroscience and Sense Organs, University of Bari, Bari, Italy
| | - Giuseppe Stefano Netti
- Clinical Pathology Unit, Department of Medical and Surgical Sciences, University of Foggia, Foggia, Italy
| | - Elena Ranieri
- Clinical Pathology Unit, Department of Medical and Surgical Sciences, University of Foggia, Foggia, Italy
| | - Luca Lacitignola
- Veterinary Surgery Unit, Department of Emergency and Organ Transplantation, University of Bari, Bari, Italy
| | - Francesco Staffieri
- Veterinary Surgery Unit, Department of Emergency and Organ Transplantation, University of Bari, Bari, Italy
| | - Alberto Maria Crovace
- Veterinary Surgery Unit, Department of Emergency and Organ Transplantation, University of Bari, Bari, Italy
| | - Giuseppe Lucarelli
- Urology, Andrology and Renal Transplantation Unit, Department of Emergency and Organ Transplantation, University of Bari, Bari, Italy
| | - Pasquale Ditonno
- Urology, Andrology and Renal Transplantation Unit, Department of Emergency and Organ Transplantation, University of Bari, Bari, Italy
| | - Michele Battaglia
- Urology, Andrology and Renal Transplantation Unit, Department of Emergency and Organ Transplantation, University of Bari, Bari, Italy
| | - Mohamed R Daha
- Department of Nephrology, University of Leiden, Leiden, The Netherlands
| | - Peter van der Pol
- Department of Nephrology, University of Leiden, Leiden, The Netherlands
| | - Cees van Kooten
- Department of Nephrology, University of Leiden, Leiden, The Netherlands
| | | | - Loreto Gesualdo
- Renal, Dialysis and Transplantation Unit, Department of Emergency and Organ Transplantation, University of Bari, Bari, Italy
| | - Giovanni Stallone
- Nephrology, Dialysis and Transplantation Unit, Advanced Research center on Kidney Aging (A.R.K.A.), Department of Medical and Surgical Science, University of Foggia, Foggia, Italy
| | - Giuseppe Castellano
- Nephrology, Dialysis and Transplantation Unit, Advanced Research center on Kidney Aging (A.R.K.A.), Department of Medical and Surgical Science, University of Foggia, Foggia, Italy
| |
Collapse
|
19
|
Infante B, Franzin R, Madio D, Calvaruso M, Maiorano A, Sangregorio F, Netti GS, Ranieri E, Gesualdo L, Castellano G, Stallone G. Molecular Mechanisms of AKI in the Elderly: From Animal Models to Therapeutic Intervention. J Clin Med 2020; 9:jcm9082574. [PMID: 32784471 PMCID: PMC7464895 DOI: 10.3390/jcm9082574] [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] [Received: 06/20/2020] [Revised: 07/21/2020] [Accepted: 08/05/2020] [Indexed: 12/17/2022] Open
Abstract
Acute kidney injury (AKI), a critical syndrome characterized by a sudden reduction of renal function, is a common disorder among elderly patients particularly in Intensive Care Unit (ICU). AKI is closely associated with both short- and long-term mortality and length of hospital stay and is considered a predictor of chronic kidney disease (CKD). Specific hemodynamic, metabolic, and molecular changes lead to increased susceptibility to injury in the aged kidney; therefore, certain causes of AKI such as the prerenal reduction in renal perfusion or vascular obstructive conditions are more common in the elderly; moreover, AKI is often multifactorial and iatrogenic. Older patients present several comorbidities (diabetes, hypertension, heart failure) and are exposed to multiple medical interventions such as the use of nephrotoxic contrasts media and medications, which can also trigger AKI. Considering the emerging relevance of this condition, prevention and treatment of AKI in the elderly should be crucial in the internist and emergency setting. This review article summarizes the incidence, the risk factors, the pathophysiology, the molecular mechanisms and the strategies of prevention and treatment of AKI in elderly patients.
Collapse
Affiliation(s)
- Barbara Infante
- Nephrology, Dialysis and Transplantation Unit, Department of Medical and Surgical Sciences, University of Foggia, Viale Pinto Luigi 251, 71122 Foggia, Italy; (B.I.); (D.M.); (A.M.); (F.S.); (G.S.)
| | - Rossana Franzin
- Nephrology, Dialysis and Transplantation Unit, Department of Emergency and Organ Transplantation, University of Bari, 70124 Bari, Italy; (R.F.); (L.G.)
| | - Desirèe Madio
- Nephrology, Dialysis and Transplantation Unit, Department of Medical and Surgical Sciences, University of Foggia, Viale Pinto Luigi 251, 71122 Foggia, Italy; (B.I.); (D.M.); (A.M.); (F.S.); (G.S.)
| | - Martina Calvaruso
- Nephrology, Dialysis and Transplantation Unit, Department of Biomedical Sciences, University of Foggia, 71122 Foggia, Italy;
| | - Annamaria Maiorano
- Nephrology, Dialysis and Transplantation Unit, Department of Medical and Surgical Sciences, University of Foggia, Viale Pinto Luigi 251, 71122 Foggia, Italy; (B.I.); (D.M.); (A.M.); (F.S.); (G.S.)
| | - Fabio Sangregorio
- Nephrology, Dialysis and Transplantation Unit, Department of Medical and Surgical Sciences, University of Foggia, Viale Pinto Luigi 251, 71122 Foggia, Italy; (B.I.); (D.M.); (A.M.); (F.S.); (G.S.)
| | - Giuseppe Stefano Netti
- Clinical Pathology, Department of Surgical and Medical Sciences, University of Foggia, Viale Pinto Luigi 251, 71122 Foggia, Italy; (G.S.N.); (E.R.)
| | - Elena Ranieri
- Clinical Pathology, Department of Surgical and Medical Sciences, University of Foggia, Viale Pinto Luigi 251, 71122 Foggia, Italy; (G.S.N.); (E.R.)
| | - Loreto Gesualdo
- Nephrology, Dialysis and Transplantation Unit, Department of Emergency and Organ Transplantation, University of Bari, 70124 Bari, Italy; (R.F.); (L.G.)
| | - Giuseppe Castellano
- Nephrology, Dialysis and Transplantation Unit, Department of Medical and Surgical Sciences, University of Foggia, Viale Pinto Luigi 251, 71122 Foggia, Italy; (B.I.); (D.M.); (A.M.); (F.S.); (G.S.)
- Correspondence: ; Tel.: +39-088-173-2610; Fax: +39-088-173-6001
| | - Giovanni Stallone
- Nephrology, Dialysis and Transplantation Unit, Department of Medical and Surgical Sciences, University of Foggia, Viale Pinto Luigi 251, 71122 Foggia, Italy; (B.I.); (D.M.); (A.M.); (F.S.); (G.S.)
| |
Collapse
|
20
|
Molecular Mechanisms of Premature Aging in Hemodialysis: The Complex Interplay Between Innate and Adaptive Immune Dysfunction. Int J Mol Sci 2020; 21:ijms21103422. [PMID: 32408613 PMCID: PMC7279398 DOI: 10.3390/ijms21103422] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2020] [Revised: 05/01/2020] [Accepted: 05/07/2020] [Indexed: 02/07/2023] Open
Abstract
Hemodialysis (HD) patient are known to be susceptible to a wide range of early and long-term complication such as chronic inflammation, infections, malnutrition, and cardiovascular disease that significantly affect the incidence of mortality. A large gap between the number of people with end-stage kidney disease (ESKD) and patients who received kidney transplantation has been identified. Therefore, there is a huge need to explore the underlying pathophysiology of HD complications in order to provide treatment guidelines. The immunological dysregulation, involving both the innate and adaptive response, plays a crucial role during the HD sessions and in chronic, maintenance treatments. Innate immune system mediators include the dysfunction of neutrophils, monocytes, and natural killer (NK) cells with signaling mediated by NOD-like receptor P3 (NLRP3) and Toll-like receptor 4 (TLR4); in addition, there is a significant activation of the complement system that is mediated by dialysis membrane-surfaces. These effectors induce a persistent, systemic, pro-inflammatory, and pro-coagulant milieu that has been described as inflammaging. The adaptive response, the imbalance in the CD4+/CD8+ T cell ratio, and the reduction of Th2 and regulatory T cells, together with an altered interaction with B lymphocyte by CD40/CD40L, have been mainly implicated in immune system dysfunction. Altogether, these observations suggest that intervention targeting the immune system in HD patients could improve morbidity and mortality. The purpose of this review is to expand our understanding on the role of immune dysfunction in both innate and adaptive response in patients undergoing hemodialysis treatment.
Collapse
|
21
|
Franzin R, Stasi A, Fiorentino M, Stallone G, Cantaluppi V, Gesualdo L, Castellano G. Inflammaging and Complement System: A Link Between Acute Kidney Injury and Chronic Graft Damage. Front Immunol 2020; 11:734. [PMID: 32457738 PMCID: PMC7221190 DOI: 10.3389/fimmu.2020.00734] [Citation(s) in RCA: 62] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2019] [Accepted: 03/31/2020] [Indexed: 12/13/2022] Open
Abstract
The aberrant activation of complement system in several kidney diseases suggests that this pillar of innate immunity has a critical role in the pathophysiology of renal damage of different etiologies. A growing body of experimental evidence indicates that complement activation contributes to the pathogenesis of acute kidney injury (AKI) such as delayed graft function (DGF) in transplant patients. AKI is characterized by the rapid loss of the kidney's excretory function and is a complex syndrome currently lacking a specific medical treatment to arrest or attenuate progression in chronic kidney disease (CKD). Recent evidence suggests that independently from the initial trigger (i.e., sepsis or ischemia/reperfusions injury), an episode of AKI is strongly associated with an increased risk of subsequent CKD. The AKI-to-CKD transition may involve a wide range of mechanisms including scar-forming myofibroblasts generated from different sources, microvascular rarefaction, mitochondrial dysfunction, or cell cycle arrest by the involvement of epigenetic, gene, and protein alterations leading to common final signaling pathways [i.e., transforming growth factor beta (TGF-β), p16 ink4a , Wnt/β-catenin pathway] involved in renal aging. Research in recent years has revealed that several stressors or complications such as rejection after renal transplantation can lead to accelerated renal aging with detrimental effects with the establishment of chronic proinflammatory cellular phenotypes within the kidney. Despite a greater understanding of these mechanisms, the role of complement system in the context of the AKI-to-CKD transition and renal inflammaging is still poorly explored. The purpose of this review is to summarize recent findings describing the role of complement in AKI-to-CKD transition. We will also address how and when complement inhibitors might be used to prevent AKI and CKD progression, therefore improving graft function.
Collapse
Affiliation(s)
- Rossana Franzin
- Nephrology, Dialysis and Transplantation Unit, Department of Emergency and Organ Transplantation, University of Bari Aldo Moro, Bari, Italy
- Department Translational Medicine, University of Piemonte Orientale, Novara, Italy
| | - Alessandra Stasi
- Nephrology, Dialysis and Transplantation Unit, Department of Emergency and Organ Transplantation, University of Bari Aldo Moro, Bari, Italy
| | - Marco Fiorentino
- Nephrology, Dialysis and Transplantation Unit, Department of Emergency and Organ Transplantation, University of Bari Aldo Moro, Bari, Italy
| | - Giovanni Stallone
- Nephrology, Dialysis and Transplantation Unit, Department of Medical and Surgical Sciences, University of Foggia, Foggia, Italy
| | - Vincenzo Cantaluppi
- Department Translational Medicine, University of Piemonte Orientale, Novara, Italy
| | - Loreto Gesualdo
- Nephrology, Dialysis and Transplantation Unit, Department of Emergency and Organ Transplantation, University of Bari Aldo Moro, Bari, Italy
| | - Giuseppe Castellano
- Nephrology, Dialysis and Transplantation Unit, Department of Emergency and Organ Transplantation, University of Bari Aldo Moro, Bari, Italy
- Nephrology, Dialysis and Transplantation Unit, Department of Medical and Surgical Sciences, University of Foggia, Foggia, Italy
| |
Collapse
|
22
|
Cui J, Kessinger CW, Jhajj HS, Grau MS, Misra S, Libby P, McCarthy JR, Jaffer FA. Atorvastatin Reduces In Vivo Fibrin Deposition and Macrophage Accumulation, and Improves Primary Patency Duration and Maturation of Murine Arteriovenous Fistula. J Am Soc Nephrol 2020; 31:931-945. [PMID: 32152232 DOI: 10.1681/asn.2019060612] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2019] [Accepted: 01/07/2020] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND Arteriovenous fistulas placed surgically for dialysis vascular access have a high primary failure rate resulting from excessive inward remodeling, medial fibrosis, and thrombosis. No clinically established pharmacologic or perisurgical therapies currently address this unmet need. Statins' induction of multiple anti-inflammatory and antithrombotic effects suggests that these drugs might reduce arteriovenous fistula failure. Yet, the in vivo physiologic and molecular effects of statins on fistula patency and maturation remain poorly understood. METHODS We randomized 108 C57Bl/6J mice to receive daily atorvastatin 1.14 mg/kg or PBS (control) starting 7 days before end-to-side carotid artery-jugular vein fistula creation and for up to 42 days after fistula creation. We then assessed longitudinally the effects of statin therapy on primary murine fistula patency and maturation. We concomitantly analyzed the in vivo arteriovenous fistula thrombogenic and inflammatory macrophage response to statin therapy, using the fibrin-targeted, near-infrared fluorescence molecular imaging agent FTP11-CyAm7 and dextranated, macrophage-avid nanoparticles CLIO-VT680. RESULTS In vivo molecular-structural imaging demonstrated that atorvastatin significantly reduced fibrin deposition at day 7 and macrophage accumulation at days 7 and 14, findings supported by histopathologic and gene-expression analyses. Structurally, atorvastatin promoted favorable venous limb outward remodeling, preserved arteriovenous fistula blood flow, and prolonged primary arteriovenous fistula patency through day 42 (P<0.05 versus control for all measures). CONCLUSIONS These findings provide new in vivo evidence that statins improve experimental arteriovenous fistula patency and maturation, indicating that additional clinical evaluation of statin therapy in patients on dialysis undergoing arteriovenous fistula placement is warranted.
Collapse
Affiliation(s)
- Jie Cui
- Division of Cardiology, Cardiovascular Research Center, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts.,Nephrology Division, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Chase W Kessinger
- Division of Cardiology, Cardiovascular Research Center, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Harkamal S Jhajj
- Division of Cardiology, Cardiovascular Research Center, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Madeleine S Grau
- Division of Cardiology, Cardiovascular Research Center, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Sanjay Misra
- Department of Radiology, Vascular and Interventional Radiology Translational Laboratory, Mayo Clinic, Rochester, Minnesota
| | - Peter Libby
- Cardiovascular Medicine Division, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Jason R McCarthy
- Center for Systems Biology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Farouc A Jaffer
- Division of Cardiology, Cardiovascular Research Center, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
23
|
Colucci M, Torreggiani M, Bernardi I, Stangalino S, Catucci D, Esposito V, Sileno G, Esposito C. Smart Flow for the evaluation of the hemodialysis arteriovenous fistula. J Vasc Access 2020; 21:652-657. [PMID: 31894718 DOI: 10.1177/1129729819897171] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
BACKGROUND Smart Flow is an innovative tool available on the Carestream Touch Prime Ultrasound machines, which provides automated blood flow measurement and shows the vectors that form the blood flow in the vessel. We compared the use of Smart Flow with traditional Duplex Doppler Ultrasound to evaluate blood flow of arteriovenous fistulas in prevalent hemodialysis patients. METHODS A total of 31 chronic patients on hemodialysis were enrolled. Blood flow was measured on the brachial artery with Smart Flow and duplex Doppler ultrasound. In a subset of 26 patients, a video of the juxta-anastomotic efferent vein was recorded and analyzed to calculate an index of flow turbulence. RESULTS We enrolled 21 males and 10 females aged 68.52 ± 11.64 years at the time of evaluation with an average arteriovenous fistulas vintage of 50.23 ± 47.42 months and followed them up for 18.03 ± 5.18 months. Smart Flow and Duplex Doppler Ultrasound blood flow measurements positively correlated (p < 0.0001) in the same patient but Smart Flow gave higher blood flow values (995.0 vs 730.3 mL/min, p < 0.0001), and the Duplex Doppler Ultrasound blood flow standard deviation was similar to Smart Flow (125.4 vs 114.4 mL/min, p < 0.0001). The time needed to evaluate arteriovenous fistulas with Smart Flow was significantly shorter than Duplex Doppler Ultrasound (67.58 ± 19.89 vs 146.3 ± 26.35 s, p < 0.0001). No correlation was found between blood flow turbulence and the subsequent access failure. CONCLUSION Smart Flow is reliable, reproducible, and faster than traditional duplex ultrasound. However, the additional information given by the Smart Flow technique does not seem to add any further benefits in terms of prediction of the access failure.
Collapse
Affiliation(s)
- Marco Colucci
- Unit of Nephrology and Dialysis, ICS Maugeri s.p.a. SB, Pavia, Italy
| | | | - Irene Bernardi
- Unit of Nephrology and Dialysis, ICS Maugeri s.p.a. SB, Pavia, Italy
| | - Simone Stangalino
- Unit of Nephrology and Dialysis, ICS Maugeri s.p.a. SB, Pavia, Italy
| | - Davide Catucci
- Unit of Nephrology and Dialysis, ICS Maugeri s.p.a. SB, Pavia, Italy
| | - Vittoria Esposito
- Unit of Nephrology and Dialysis, ICS Maugeri s.p.a. SB, Pavia, Italy
| | - Giuseppe Sileno
- Unit of Nephrology and Dialysis, ICS Maugeri s.p.a. SB, Pavia, Italy
| | - Ciro Esposito
- Unit of Nephrology and Dialysis, ICS Maugeri s.p.a. SB, Pavia, Italy.,University of Pavia, Pavia, Italy
| |
Collapse
|
24
|
Sadaghianloo N, Contenti J, Dardik A, Mazure NM. Role of Hypoxia and Metabolism in the Development of Neointimal Hyperplasia in Arteriovenous Fistulas. Int J Mol Sci 2019; 20:ijms20215387. [PMID: 31671790 PMCID: PMC6862436 DOI: 10.3390/ijms20215387] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2019] [Revised: 10/24/2019] [Accepted: 10/25/2019] [Indexed: 12/11/2022] Open
Abstract
For patients with end-stage renal disease requiring hemodialysis, their vascular access is both their lifeline and their Achilles heel. Despite being recommended as primary vascular access, the arteriovenous fistula (AVF) shows sub-optimal results, with about 50% of patients needing a revision during the year following creation. After the AVF is created, the venous wall must adapt to new environment. While hemodynamic changes are responsible for the adaptation of the extracellular matrix and activation of the endothelium, surgical dissection and mobilization of the vein disrupt the vasa vasorum, causing wall ischemia and oxidative stress. As a consequence, migration and proliferation of vascular cells participate in venous wall thickening by a mechanism of neointimal hyperplasia (NH). When aggressive, NH causes stenosis and AVF dysfunction. In this review we show how hypoxia, metabolism, and flow parameters are intricate mechanisms responsible for the development of NH and stenosis during AVF maturation.
Collapse
Affiliation(s)
- Nirvana Sadaghianloo
- Centre de Méditerranéen de Médecine Moléculaire (C3M), Université Côte d'Azur, INSERM U1065, 151 Route de St Antoine de Ginestière, BP2 3194, 06204 Nice CEDEX 03, France.
- Department of Vascular Surgery, Centre Hospitalier Universitaire de Nice, 06000 Nice, France.
| | - Julie Contenti
- Centre de Méditerranéen de Médecine Moléculaire (C3M), Université Côte d'Azur, INSERM U1065, 151 Route de St Antoine de Ginestière, BP2 3194, 06204 Nice CEDEX 03, France.
- Department of Emergency Medicine, Centre Hospitalier Universitaire de Nice, 06000 Nice, France.
| | - Alan Dardik
- Department of Surgery and the Vascular Biology and Therapeutics Program, Yale University, New Haven, CT 06520, USA.
- Department of Surgery, VA Connecticut Healthcare Systems, West Haven, CT 06516, USA.
| | - Nathalie M Mazure
- Centre de Méditerranéen de Médecine Moléculaire (C3M), Université Côte d'Azur, INSERM U1065, 151 Route de St Antoine de Ginestière, BP2 3194, 06204 Nice CEDEX 03, France.
- Department of Vascular Surgery, Centre Hospitalier Universitaire de Nice, 06000 Nice, France.
| |
Collapse
|
25
|
Satish M, Gunasekar P, Agrawal DK. Pro-inflammatory and pro-resolving mechanisms in the immunopathology of arteriovenous fistula maturation. Expert Rev Cardiovasc Ther 2019; 17:369-376. [PMID: 31056981 DOI: 10.1080/14779072.2019.1612745] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Introduction: With high rates of arteriovenous fistula (AVF) failure, there is a continued need to predict other factors and mechanisms associated with maturation deficits. Given the central association of inflammation with AVF failure, with neointimal hyperplasia (NIH) as one such mechanism, inflammation must be considered in two endogenous ways, either pro-inflammatory or pro-resolving, resulting in inward or outward vascular remodeling. Areas covered: This review summarizes and critically evaluates the preclinical and interventional data underlying AVF failure in attempts to elucidate the necessary balance between inflammation and its resolution. Expert opinion: Understanding the pro-inflammatory and pro-resolving mechanisms underlying inward and outward vascular remodeling and NIH prevention with AVF maturation is a necessary effort to develop key diagnostic and therapeutic interventions towards the ongoing issue of long-term AVF patency. The ability for clinical application has progressed but is limited to the identification of key targets and pathways with little understanding of how they are related synergistically or antagonistically. Likewise, the balance between acute inflammation and pro-resolution requires pertinent temporal considerations necessary for timely therapeutic application and predictive measurement.
Collapse
Affiliation(s)
- Mohan Satish
- a Department of Clinical and Translational Science , Creighton University School of Medicine , Omaha , NE , USA
| | - Palanikumar Gunasekar
- a Department of Clinical and Translational Science , Creighton University School of Medicine , Omaha , NE , USA
| | - Devendra K Agrawal
- a Department of Clinical and Translational Science , Creighton University School of Medicine , Omaha , NE , USA
| |
Collapse
|
26
|
Chen B, Wang P, Brem A, Dworkin L, Liu Z, Gong R. Mineralocorticoid receptor: A hidden culprit for hemodialysis vascular access dysfunction. EBioMedicine 2019; 39:621-627. [PMID: 30527626 PMCID: PMC6354623 DOI: 10.1016/j.ebiom.2018.11.054] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2018] [Revised: 11/25/2018] [Accepted: 11/27/2018] [Indexed: 02/08/2023] Open
Abstract
Hemodialysis vascular access dysfunction is a common and intractable problem in clinical practice with no definitive therapy yet available. As a key mediator of vascular and cardiac maladaptive remodeling, mineralocorticoid receptor (MR) plays a pivotal role in vascular fibrosis and intimal hyperplasia (IH) and is potentiated locally in hemodialysis vascular access following diverse injuries, like barotrauma, cannulation and shear stress. MR-related genomic and non-genomic pathways are responsible for triggering vascular smooth muscle cell activation, proliferation, migration and extracellular matrix overproduction. In endothelial cells, MR signaling diminishes nitric oxide production and its bioavailability, but amplifies reactive oxygen species, leading to an inflammatory state. Moreover, MR favors macrophage polarization towards a pro-inflammatory phenotype. In clinical settings like post-angioplasty or stenting restenosis, the beneficial effect of MR antagonists on vascular fibrosis and IH has been validated. In aggregate, therapeutic targeting of MR may provide a new avenue to prevent hemodialysis vascular access dysfunction.
Collapse
Affiliation(s)
- Bohan Chen
- Institute of Nephrology, Blood Purification Center, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, China; Division of Kidney Disease and Hypertension, Department of Medicine, Brown University School of Medicine, Providence, RI, United States; Division of Nephrology, Department of Medicine, University of Toledo College of Medicine, Toledo, OH, United States
| | - Pei Wang
- Institute of Nephrology, Blood Purification Center, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Andrew Brem
- Division of Kidney Disease and Hypertension, Department of Medicine, Brown University School of Medicine, Providence, RI, United States
| | - Lance Dworkin
- Division of Nephrology, Department of Medicine, University of Toledo College of Medicine, Toledo, OH, United States
| | - Zhangsuo Liu
- Institute of Nephrology, Blood Purification Center, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.
| | - Rujun Gong
- Division of Kidney Disease and Hypertension, Department of Medicine, Brown University School of Medicine, Providence, RI, United States; Division of Nephrology, Department of Medicine, University of Toledo College of Medicine, Toledo, OH, United States.
| |
Collapse
|
27
|
Hao YM, Yuan HQ, Ren Z, Qu SL, Liu LS, Dang-HengWei, Yin K, Fu M, Jiang ZS. Endothelial to mesenchymal transition in atherosclerotic vascular remodeling. Clin Chim Acta 2018; 490:34-38. [PMID: 30571947 DOI: 10.1016/j.cca.2018.12.018] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2018] [Revised: 12/14/2018] [Accepted: 12/14/2018] [Indexed: 12/12/2022]
Abstract
Endothelial cells are the main components of the heart, blood vessels, and lymphatic vessels, which play an important role in regulating the physiological functions of the cardiovascular system. Endothelial dysfunction is involved in a variety of acute and chronic cardiovascular diseases. As a special type of epithelial-mesenchymal transition (EMT), endothelium to mesenchymal transition (EndMT) regulates the transformation of endothelial cells into mesenchymal cells accompanied by changes in the expression of various transcription factors and cytokines, which is closely related to vascular endothelial injury, vascular remodeling, myocardial fibrosis and valvar disease. Endothelial cells undergoing EndMT lose their endothelial characteristics and undergo a transition toward a more mesenchymal-like phenotype. However, the molecular mechanism of EndMT remains unclear. EndMT, as a type of endothelial dysfunction, can cause vascular remodeling which is a major determinant of atherosclerotic luminal area. Therefore, exploring the important signaling pathways in the process of EndMT may provide novel therapeutic strategies for treating atherosclerotic diseases.
Collapse
Affiliation(s)
- Ya-Meng Hao
- Institute of Cardiovascular Disease, Key Laboratory for Arteriosclerosis of Hunan Province, University of South, Hengyang City, Hunan Province 421001, PR China
| | - Hou-Qin Yuan
- Institute of Cardiovascular Disease, Key Laboratory for Arteriosclerosis of Hunan Province, University of South, Hengyang City, Hunan Province 421001, PR China
| | - Zhong Ren
- Institute of Cardiovascular Disease, Key Laboratory for Arteriosclerosis of Hunan Province, University of South, Hengyang City, Hunan Province 421001, PR China
| | - Shun-Lin Qu
- Institute of Cardiovascular Disease, Key Laboratory for Arteriosclerosis of Hunan Province, University of South, Hengyang City, Hunan Province 421001, PR China
| | - Lu-Shan Liu
- Institute of Cardiovascular Disease, Key Laboratory for Arteriosclerosis of Hunan Province, University of South, Hengyang City, Hunan Province 421001, PR China
| | - Dang-HengWei
- Institute of Cardiovascular Disease, Key Laboratory for Arteriosclerosis of Hunan Province, University of South, Hengyang City, Hunan Province 421001, PR China
| | - Kai Yin
- Institute of Cardiovascular Disease, Key Laboratory for Arteriosclerosis of Hunan Province, University of South, Hengyang City, Hunan Province 421001, PR China; Research Lab of Translational Medicine, Medical School, University of South China, Hengyang 421001, PR China
| | - Mingui Fu
- Department of Biomedical Science, Shock/Trauma Research Center, School of Medicine, University of Missouri Kansas City, Kansas City, MO 64108, USA
| | - Zhi-Sheng Jiang
- Institute of Cardiovascular Disease, Key Laboratory for Arteriosclerosis of Hunan Province, University of South, Hengyang City, Hunan Province 421001, PR China.
| |
Collapse
|
28
|
Martinez L, Duque JC, Tabbara M, Paez A, Selman G, Hernandez DR, Sundberg CA, Tey JCS, Shiu YT, Cheung AK, Allon M, Velazquez OC, Salman LH, Vazquez-Padron RI. Fibrotic Venous Remodeling and Nonmaturation of Arteriovenous Fistulas. J Am Soc Nephrol 2018; 29:1030-1040. [PMID: 29295872 DOI: 10.1681/asn.2017050559] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2017] [Accepted: 10/16/2017] [Indexed: 02/03/2023] Open
Abstract
The frequency of primary failure in arteriovenous fistulas (AVFs) remains unacceptably high. This lack of improvement is due in part to a poor understanding of the pathobiology underlying AVF nonmaturation. This observational study quantified the progression of three vascular features, medial fibrosis, intimal hyperplasia (IH), and collagen fiber organization, during early AVF remodeling and evaluated the associations thereof with AVF nonmaturation. We obtained venous samples from patients undergoing two-stage upper-arm AVF surgeries at a single center, including intraoperative veins at the first-stage access creation surgery and AVFs at the second-stage transposition procedure. Paired venous samples from both stages were used to evaluate change in these vascular features after anastomosis. Anatomic nonmaturation (AVF diameter never ≥6 mm) occurred in 39 of 161 (24%) patients. Neither preexisting fibrosis nor IH predicted AVF outcomes. Postoperative medial fibrosis associated with nonmaturation (odds ratio [OR], 1.55; 95% confidence interval [95% CI], 1.05 to 2.30; P=0.03, per 10% absolute increase in fibrosis), whereas postoperative IH only associated with failure in those individuals with medial fibrosis over the population's median value (OR, 2.63; 95% CI, 1.07 to 6.46; P=0.04, per increase of 1 in the intima/media ratio). Analysis of postoperative medial collagen organization revealed that circumferential alignment of fibers around the lumen associated with AVF nonmaturation (OR, 1.38; 95% CI, 1.03 to 1.84; P=0.03, per 10° increase in angle). This study demonstrates that excessive fibrotic remodeling of the vein after AVF creation is an important risk factor for nonmaturation and that high medial fibrosis determines the stenotic potential of IH.
Collapse
Affiliation(s)
| | - Juan C Duque
- Division of Nephrology and Hypertension, Leonard M. Miller School of Medicine, University of Miami, Miami, Florida
| | | | - Angela Paez
- DeWitt Daughtry Family Department of Surgery and
| | - Guillermo Selman
- Division of Nephrology and Hypertension, Albany Medical College, Albany, New York
| | | | - Chad A Sundberg
- Division of Nephrology and Hypertension, University of Utah, Salt Lake City, Utah
| | | | - Yan-Ting Shiu
- Division of Nephrology and Hypertension, University of Utah, Salt Lake City, Utah
| | - Alfred K Cheung
- Division of Nephrology and Hypertension, University of Utah, Salt Lake City, Utah.,Medical Service, Veterans Affairs Salt Lake City Healthcare System, Salt Lake City, Utah.,Department of Nephrology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Michael Allon
- Division of Nephrology, University of Alabama at Birmingham, Birmingham, Alabama; and
| | | | - Loay H Salman
- Division of Nephrology and Hypertension, Albany Medical College, Albany, New York
| | | |
Collapse
|
29
|
Brahmbhatt A, Remuzzi A, Franzoni M, Misra S. The molecular mechanisms of hemodialysis vascular access failure. Kidney Int 2017; 89:303-316. [PMID: 26806833 PMCID: PMC4734360 DOI: 10.1016/j.kint.2015.12.019] [Citation(s) in RCA: 170] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2015] [Accepted: 08/20/2015] [Indexed: 01/01/2023]
Abstract
The arteriovenous fistula has been used for more than 50 years to provide vascular access for patients undergoing hemodialysis. More than 1.5 million patients worldwide have end stage renal disease and this population will continue to grow. The arteriovenous fistula is the preferred vascular access for patients, but its patency rate at 1 year is only 60%. The majority of arteriovenous fistulas fail because of intimal hyperplasia. In recent years, there have been many studies investigating the molecular mechanisms responsible for intimal hyperplasia and subsequent thrombosis. These studies have identified common pathways including inflammation, uremia, hypoxia, sheer stress, and increased thrombogenicity. These cellular mechanisms lead to increased proliferation, migration, and eventually stenosis. These pathways work synergistically through shared molecular messengers. In this review, we will examine the literature concerning the molecular basis of hemodialysis vascular access malfunction.
Collapse
Affiliation(s)
- Akshaar Brahmbhatt
- Vascular and Interventional Radiology Translational Laboratory, Department of Radiology, Mayo Clinic, Rochester, Minnesota, USA
| | - Andrea Remuzzi
- Biomedical Engineering Department, IRCCS—Istituto di Ricerche Farmacologiche Mario Negri, Bergamo, Italy
- Engineering Department, University of Bergamo, Dalmine, Italy
| | - Marco Franzoni
- Biomedical Engineering Department, IRCCS—Istituto di Ricerche Farmacologiche Mario Negri, Bergamo, Italy
| | - Sanjay Misra
- Vascular and Interventional Radiology Translational Laboratory, Department of Radiology, Mayo Clinic, Rochester, Minnesota, USA
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, Minnesota, USA
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, Minnesota, USA
| |
Collapse
|
30
|
Kitrou P, Spiliopoulos S, Karnabatidis D, Katsanos K. Cutting balloons, covered stents and paclitaxel-coated balloons for the treatment of dysfunctional dialysis access. Expert Rev Med Devices 2016; 13:1119-1126. [PMID: 27791450 DOI: 10.1080/17434440.2016.1254548] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Percutaneous transcatheter balloon angioplasty has evolved to the current mainstay treatment for salvage of dysfunctional dialysis access. Nonetheless, it is frequently associated with recurrent vessel restenosis and the need for multiple repeat treatments in order to maintain hemodynamic patency. Cutting-balloons, covered stents or stent-grafts, and paclitaxel-coated balloons have been extensively tested and investigated with the aim to improve immediate anatomical and long-term clinical results. Areas covered: In the present overview, we discuss the background and appraise relevant medical literature on the aforementioned technologies and provide a more in-depth synthesis of the results of different clinical studies for each device category. We will also discuss the limitations in the mode of action of each group of devices and envision what the future holds for the challenging field of dialysis access interventions. Expert commentary: We propose a good practice algorithm for the treatment of thrombosed or dysfunctional dialysis access.
Collapse
Affiliation(s)
- Panagiotis Kitrou
- a Department of Interventional Radiology , Patras University Hospital, School of Medicine , Rion , Greece
| | - Stavros Spiliopoulos
- b 2nd Department of Radiology, Interventional Radiology Unit , ATTIKO Athens University Hospital , Athens , Greece
| | - Dimitris Karnabatidis
- a Department of Interventional Radiology , Patras University Hospital, School of Medicine , Rion , Greece
| | - Konstantinos Katsanos
- a Department of Interventional Radiology , Patras University Hospital, School of Medicine , Rion , Greece
- c Department of Interventional Radiology , Guy's and St. Thomas' Hospitals, NHS Foundation Trust, King's Health Partners , London , UK
| |
Collapse
|
31
|
Hu H, Patel S, Hanisch JJ, Santana JM, Hashimoto T, Bai H, Kudze T, Foster TR, Guo J, Yatsula B, Tsui J, Dardik A. Future research directions to improve fistula maturation and reduce access failure. Semin Vasc Surg 2016; 29:153-171. [PMID: 28779782 DOI: 10.1053/j.semvascsurg.2016.08.005] [Citation(s) in RCA: 79] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
With the increasing prevalence of end-stage renal disease, there is a growing need for hemodialysis. Arteriovenous fistulae (AVF) are the preferred type of vascular access for hemodialysis, but maturation and failure continue to present significant barriers to successful fistula use. AVF maturation integrates outward remodeling with vessel wall thickening in response to drastic hemodynamic changes in the setting of uremia, systemic inflammation, oxidative stress, and pre-existent vascular pathology. AVF can fail due to both failure to mature adequately to support hemodialysis and development of neointimal hyperplasia that narrows the AVF lumen, typically near the fistula anastomosis. Failure due to neointimal hyperplasia involves vascular cell activation and migration and extracellular matrix remodeling with complex interactions of growth factors, adhesion molecules, inflammatory mediators, and chemokines, all of which result in maladaptive remodeling. Different strategies have been proposed to prevent and treat AVF failure based on current understanding of the modes and pathology of access failure; these approaches range from appropriate patient selection and use of alternative surgical strategies for fistula creation, to the use of novel interventional techniques or drugs to treat failing fistulae. Effective treatments to prevent or treat AVF failure require a multidisciplinary approach involving nephrologists, vascular surgeons, and interventional radiologists, careful patient selection, and the use of tailored systemic or localized interventions to improve patient-specific outcomes. This review provides contemporary information on the underlying mechanisms of AVF maturation and failure and discusses the broad spectrum of options that can be tailored for specific therapy.
Collapse
Affiliation(s)
- Haidi Hu
- Department of Surgery, Yale University School of Medicine, 10 Amistad Street, Room 437, PO Box 208089, New Haven, CT 06520-8089; Department of Vascular and Thyroid Surgery, the First Affiliated Hospital of China Medical University, Shenyang, China; Vascular Biology and Therapeutics Program, Yale University, New Haven, CT
| | - Sandeep Patel
- Department of Surgery, Yale University School of Medicine, 10 Amistad Street, Room 437, PO Box 208089, New Haven, CT 06520-8089; Vascular Biology and Therapeutics Program, Yale University, New Haven, CT; Royal Free Hospital, University College London, London, UK
| | - Jesse J Hanisch
- Department of Surgery, Yale University School of Medicine, 10 Amistad Street, Room 437, PO Box 208089, New Haven, CT 06520-8089; Vascular Biology and Therapeutics Program, Yale University, New Haven, CT
| | - Jeans M Santana
- Department of Surgery, Yale University School of Medicine, 10 Amistad Street, Room 437, PO Box 208089, New Haven, CT 06520-8089; Vascular Biology and Therapeutics Program, Yale University, New Haven, CT
| | - Takuya Hashimoto
- Department of Surgery, Yale University School of Medicine, 10 Amistad Street, Room 437, PO Box 208089, New Haven, CT 06520-8089; Vascular Biology and Therapeutics Program, Yale University, New Haven, CT
| | - Hualong Bai
- Department of Surgery, Yale University School of Medicine, 10 Amistad Street, Room 437, PO Box 208089, New Haven, CT 06520-8089; Vascular Biology and Therapeutics Program, Yale University, New Haven, CT
| | - Tambudzai Kudze
- Department of Surgery, Yale University School of Medicine, 10 Amistad Street, Room 437, PO Box 208089, New Haven, CT 06520-8089; Vascular Biology and Therapeutics Program, Yale University, New Haven, CT
| | - Trenton R Foster
- Department of Surgery, Yale University School of Medicine, 10 Amistad Street, Room 437, PO Box 208089, New Haven, CT 06520-8089; Vascular Biology and Therapeutics Program, Yale University, New Haven, CT
| | - Jianming Guo
- Department of Surgery, Yale University School of Medicine, 10 Amistad Street, Room 437, PO Box 208089, New Haven, CT 06520-8089; Vascular Biology and Therapeutics Program, Yale University, New Haven, CT
| | - Bogdan Yatsula
- Department of Surgery, Yale University School of Medicine, 10 Amistad Street, Room 437, PO Box 208089, New Haven, CT 06520-8089; Vascular Biology and Therapeutics Program, Yale University, New Haven, CT
| | - Janice Tsui
- Royal Free Hospital, University College London, London, UK
| | - Alan Dardik
- Department of Surgery, Yale University School of Medicine, 10 Amistad Street, Room 437, PO Box 208089, New Haven, CT 06520-8089; Vascular Biology and Therapeutics Program, Yale University, New Haven, CT; VA Connecticut Healthcare System, West Haven, CT.
| |
Collapse
|
32
|
Abstract
Arteriovenous fistulas (AVFs) are essential for patients and clinicians faced with end-stage renal disease (ESRD). While this method of vascular access for hemodialysis is preferred to others due to its reduced rate of infection and complications, they are plagued by intimal hyperplasia. The pathogenesis of intimal hyperplasia and subsequent thrombosis is brought on by uremia, hypoxia, and shear stress. These forces upregulate inflammatory and proliferative cytokines acting on leukocytes, fibroblasts, smooth muscle cells, and platelets. This activation begins initially with the progression of uremia, which induces platelet dysfunction and primes the body for an inflammatory response. The vasculature subsequently undergoes changes in oxygenation and shear stress during AVF creation. This propagates a strong inflammatory response in the vessel leading to cellular proliferation. This combined response is then further subjected to the stressors of cannulation and dialysis, eventually leading to stenosis and thrombosis. This review aims to help interventional radiologists understand the biological changes and pathogenesis of access failure.
Collapse
Affiliation(s)
- Akshaar Brahmbhatt
- Vascular and Interventional Radiology Translational Laboratory, Department of Radiology, Mayo Clinic, Rochester, Minnesota
| | - Sanjay Misra
- Vascular and Interventional Radiology Translational Laboratory, Department of Radiology, Mayo Clinic, Rochester, Minnesota; Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, Minnesota
| |
Collapse
|
33
|
Skartsis N, Martinez L, Duque JC, Tabbara M, Velazquez OC, Asif A, Andreopoulos F, Salman LH, Vazquez-Padron RI. c-Kit signaling determines neointimal hyperplasia in arteriovenous fistulae. Am J Physiol Renal Physiol 2014; 307:F1095-104. [PMID: 25186298 DOI: 10.1152/ajprenal.00292.2014] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Stenosis of arteriovenous (A-V) fistulae secondary to neointimal hyperplasia (NIH) compromises dialysis delivery, which worsens patients' quality of life and increases medical costs associated with the maintenance of vascular accesses. In the present study, we evaluated the role of the receptor tyrosine kinase c-Kit in A-V fistula neointima formation. Initially, c-Kit was found in the neointima and adventitia of human brachiobasilic fistulae, whereas it was barely detectable in control veins harvested at the time of access creation. Using the rat A-V fistula model to study venous vascular remodeling, we analyzed the spatial and temporal pattern of c-Kit expression in the fistula wall. Interestingly, c-Kit immunoreactivity increased with time after anastomosis, which concurred with the accumulation of cells in the venous intima. In addition, c-Kit expression in A-V fistulae was positively altered by chronic kidney failure conditions. Both blockade of c-Kit with imatinib mesylate (Gleevec) and inhibition of stem cell factor production with a specific short hairpin RNA prevented NIH in the outflow vein of experimental fistulae. In agreement with these data, impaired c-Kit activity compromised the development of NIH in A-V fistulae created in c-KitW/Wv mutant mice. These results suggest that targeting of the c-Kit signaling pathway may be an effective approach to prevent postoperative NIH in A-V fistulae.
Collapse
Affiliation(s)
- Nikolaos Skartsis
- Department of Surgery and Vascular Biology Institute, University of Miami Miller School of Medicine, Miami, Florida
| | - Laisel Martinez
- Department of Surgery and Vascular Biology Institute, University of Miami Miller School of Medicine, Miami, Florida
| | - Juan Camilo Duque
- Department of Surgery and Vascular Biology Institute, University of Miami Miller School of Medicine, Miami, Florida
| | - Marwan Tabbara
- Department of Surgery and Vascular Biology Institute, University of Miami Miller School of Medicine, Miami, Florida
| | - Omaida C Velazquez
- Department of Surgery and Vascular Biology Institute, University of Miami Miller School of Medicine, Miami, Florida
| | - Arif Asif
- Division of Nephrology and Hypertension, Albany Medical College, Albany, New York
| | - Fotios Andreopoulos
- Biomedical Engineering Department, University of Miami, Coral Gables, Florida; and
| | - Loay H Salman
- Interventional Nephrology, University of Miami Miller School of Medicine, Miami, Florida
| | - Roberto I Vazquez-Padron
- Department of Surgery and Vascular Biology Institute, University of Miami Miller School of Medicine, Miami, Florida;
| |
Collapse
|