1
|
Kruit N, Sluiter TJ, de Vries MR. Role of Perivascular Adipose Tissue in Vein Remodeling. Arterioscler Thromb Vasc Biol 2025; 45:576-584. [PMID: 40079141 PMCID: PMC12017597 DOI: 10.1161/atvbaha.124.321692] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/14/2025]
Abstract
Perivascular adipose tissue (PVAT) plays a crucial, yet underexplored, role in vein remodeling, which occurs after bypass surgery using a venous graft or creation of arteriovenous fistulae for hemodialysis access. PVAT exhibits significant heterogeneity in phenotype and tissue composition depending on the vascular bed, as well as its anatomic location within the vasculature. Through the excretion of adipokines, cytokines, and chemokines, PVAT can shape the vascular response to local and systemic perturbations. Moreover, the active exchange of cells reinforces the bidirectional cross talk between the vessel wall and PVAT. In this review, we describe the role of PVAT in relation to postinterventional vein remodeling, specifically focusing on the effect of surgery on the PVAT phenotype. Moreover, we discuss the pathophysiological mechanisms that ultimately affect clinical outcomes and highlight the therapeutic potential of PVAT to improve vein remodeling.
Collapse
Affiliation(s)
- Nicky Kruit
- Department of Surgery (N.K., T.J.S., M.R.d.V.), Leiden University Medical Center, the Netherlands
- Einthoven Laboratory for Experimental Vascular and Regenerative Medicine (N.K., T.J.S., M.R.d.V.), Leiden University Medical Center, the Netherlands
| | - Thijs J. Sluiter
- Department of Surgery (N.K., T.J.S., M.R.d.V.), Leiden University Medical Center, the Netherlands
- Einthoven Laboratory for Experimental Vascular and Regenerative Medicine (N.K., T.J.S., M.R.d.V.), Leiden University Medical Center, the Netherlands
| | - Margreet R. de Vries
- Department of Surgery (N.K., T.J.S., M.R.d.V.), Leiden University Medical Center, the Netherlands
- Einthoven Laboratory for Experimental Vascular and Regenerative Medicine (N.K., T.J.S., M.R.d.V.), Leiden University Medical Center, the Netherlands
- Department of Surgery, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA (M.R.d.V.)
| |
Collapse
|
2
|
Wu Y, Lee TH, Cheng OH, Peden EK, Li Q, Wang J, Huang F, Melancon MP, Sheikh-Hamad D, Wang T, Truong L, Mitch WE, Liang M, Cheng J. Interplay between Skeletal Muscle Catabolism and Remodeling of Arteriovenous Fistula by Yes-Associated Protein 1 (YAP1) Signaling. J Am Soc Nephrol 2025; 36:845-858. [PMID: 39883520 PMCID: PMC12059102 DOI: 10.1681/asn.0000000605] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2024] [Accepted: 01/13/2025] [Indexed: 01/31/2025] Open
Abstract
Key Points Atrophied muscle–derived myostatin stimulated mesenchymal stem cell differentiation and adverse arteriovenous (AV) fistula remodeling through yes-associated protein 1 (YAP1) activation. Treatment with myostatin peptibody inhibited muscle wasting and blocked mesenchymal stem cell activation and AV fistula fibrosis. A light-sensitive drug-release strategy was engineered for the periadventitial delivery of verteporfin to improve AV fistula patency. Background Arteriovenous (AV) fistulas are the preferred access for dialysis but have a high incidence of failure. The aim of this study was to understand the crosstalk between skeletal muscle catabolism and AV fistula maturation failure. Methods Skeletal muscle metabolism and AV fistula maturation were evaluated in mice with CKD. The roles of myostatin and yes-associated protein 1 (YAP1) in regulating the transdifferentiation of adventitial mesenchymal stem cells (MSCs) and intima hyperplasia in AV fistula were investigated. Nanoparticles carrying a YAP1 inhibitor, verteporfin, with light irradiation–controlled release were synthesized and applied to AV fistula. Results Increased trichrome signals and stenosis were observed in AV fistulas from mice treated with myostatin and from mice with CKD. By contrast, blocking myostatin function with an anti-myostatin peptibody not only improved body weight and muscle size in CKD mice but also decreased neointima formation in AV fistulas. In cultured MSCs, myostatin induced YAP1 expression, promoting the differentiation of MSCs into myofibroblasts and inducing extracellular matrix deposition. Red light irradiation–controlled release of verteporfin from nanoparticles blocked YAP1 activation and alleviated myostatin-induced MSC activation. Periadventitial application and red light irradiation of nanoparticles carrying verteporfin significantly suppressed stiffening and neointima formation in AV fistula. Conclusions CKD induced muscle wasting, leading to increased production of myostatin, which stimulated MSC activation and vascular fibrosis linked to AV fistula stenosis. YAP1 signaling was activated in these processes. Red light irradiation–controlled release of verteporfin offered a feasible approach for local vascular drug intervention to improve AV fistula maturation.
Collapse
Affiliation(s)
- Yongdong Wu
- Section of Nephrology, Department of Medicine, Selzman Institute for Kidney Health, Baylor College of Medicine, Houston, Texas
- Department of Nephrology, Guangzhou First People's Hospital, South China University of Technology, Guangzhou, China
| | - Tae Hoon Lee
- Section of Nephrology, Department of Medicine, Selzman Institute for Kidney Health, Baylor College of Medicine, Houston, Texas
| | - Owen H. Cheng
- Section of Nephrology, Department of Medicine, Selzman Institute for Kidney Health, Baylor College of Medicine, Houston, Texas
| | - Eric K. Peden
- Department of Surgery, Houston Methodist Hospital, Houston, Texas
| | - Qingtian Li
- Section of Nephrology, Department of Medicine, Selzman Institute for Kidney Health, Baylor College of Medicine, Houston, Texas
| | - Jun Wang
- Department of Pediatrics, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, Texas
| | - Fengzhang Huang
- Section of Nephrology, Department of Medicine, Selzman Institute for Kidney Health, Baylor College of Medicine, Houston, Texas
| | - Marites P. Melancon
- Department of Interventional Radiology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - David Sheikh-Hamad
- Section of Nephrology, Department of Medicine, Selzman Institute for Kidney Health, Baylor College of Medicine, Houston, Texas
| | - Tao Wang
- Section of Nephrology, Department of Medicine, Selzman Institute for Kidney Health, Baylor College of Medicine, Houston, Texas
| | - Luan Truong
- Department of Pathology and Genomic medicine, Houston Methodist Hospital, Houston, Texas
| | - William E. Mitch
- Section of Nephrology, Department of Medicine, Selzman Institute for Kidney Health, Baylor College of Medicine, Houston, Texas
| | - Ming Liang
- Department of Nephrology, Guangzhou First People's Hospital, South China University of Technology, Guangzhou, China
| | - Jizhong Cheng
- Section of Nephrology, Department of Medicine, Selzman Institute for Kidney Health, Baylor College of Medicine, Houston, Texas
| |
Collapse
|
3
|
Barcena AJR, Perez JVD, Bernardino MR, Damasco JA, San Valentin EMD, Klusman C, Martin B, Canlas GM, Heralde FM, Fowlkes N, Bouchard RR, Cheng J, Huang SY, Melancon MP. Bismuth-infused perivascular wrap facilitates delivery of mesenchymal stem cells and attenuation of neointimal hyperplasia in rat arteriovenous fistulas. BIOMATERIALS ADVANCES 2025; 166:214052. [PMID: 39341164 PMCID: PMC11725062 DOI: 10.1016/j.bioadv.2024.214052] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Revised: 09/10/2024] [Accepted: 09/19/2024] [Indexed: 09/30/2024]
Abstract
BACKGROUND Mesenchymal stem cells (MSCs) have emerged as novel therapies for supporting arteriovenous fistula (AVF) maturation, and bioresorbable polymeric scaffolds have enabled sustained MSC delivery into maturing AVFs. However, the radiolucency of biopolymeric wraps prevents in vivo monitoring of their integrity and location, hindering long-term preclinical investigations. METHODS We infused bismuth nanoparticles (BiNPs) into polycaprolactone (PCL) to fabricate an electrospun perivascular wrap capable of MSC delivery and conducive to longitudinal monitoring using conventional imaging. We tested the wraps' effects on the attenuation of markers of neointimal hyperplasia (i.e., endothelial dysfunction, hypoxia, and inflammation), the leading cause of AVF failure, in rats with induced chronic kidney disease (n = 3 per time point) for the following groups: control (no wrap), PCL wrap, PCL with MSCs, PCL-Bi (BiNP-infused wrap), and PCL-Bi with MSCs. RESULTS Physicochemical characterization and in vitro biocompatibility tests revealed that BiNP infusion did not alter the wrap's non-cytotoxicity toward vascular cells, hemocompatibility, and capacity for MSC loading but facilitated long-term monitoring via micro-computed tomography. After 8 weeks, all treatment groups demonstrated significant improvement in wall-to-lumen ratio on ultrasonography (P < 0.001), neointima-to-lumen ratio on histomorphometry (P < 0.001), and attenuation of neointimal hypoxia on immunohistochemistry (P < 0.05). Compared to non-MSC wraps, MSC-loaded wraps not only attenuated endothelial dysfunction and neointimal inflammation but also reduced hypoxia and inflammation across all vascular layers. CONCLUSION These results demonstrate that MSC delivery through a radiopaque polymeric wrap could enhance AVF patency outcomes through the inhibition of multiple pathways inducing AVF failure.
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
| | - 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
| | - 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; 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; Baylor College of Medicine, Houston, TX 77030, USA
| | | | - Francisco M Heralde
- College of Medicine, University of the Philippines Manila, Manila 1000, Philippines
| | - 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
- Division 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
|
4
|
Baranwal G, Mukhtar H, Kane J, Lemieux A, Misra S. Advancements in Mesenchymal Stem Cell-Based Therapy for Enhancing Arteriovenous Fistula Patency. Int J Mol Sci 2024; 25:12719. [PMID: 39684430 DOI: 10.3390/ijms252312719] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2024] [Revised: 11/20/2024] [Accepted: 11/22/2024] [Indexed: 12/18/2024] Open
Abstract
Chronic kidney disease (CKD) affects more than 10% of the world's population. Hemodialysis, along with peritoneal dialysis and renal transplant, is one of the renal replacement therapies offered to patients with CKD/end-stage renal disease (ESRD). To proceed with hemodialysis, vascular access is required. The two means of long-term access are arteriovenous fistula (AVF) and arteriovenous graft (AVG). Multiple therapies have been created to help the long-term patency of AVFs. These therapies are needed as 40% of AVFs fail within the first year and additional intervention is required. Much of the existing research has focused on biomarkers, immune cells, hypoxia, and cell-based therapies. Regeneration therapy using mesenchymal stem cells seeks to investigate other ways that we can treat AVF failure. Mesenchymal stem cells are harvested as two main types, fetal and adult. Fetal cells are harvested at different times in fetal gestation and from multiple sources, placental blood, Whartons jelly, and amniotic stem cell fluid. Taken together, this review summarizes the different preclinical/clinical studies conducted using different types of MSCs towards vascular regenerative medicine and further highlights its potential to be a suitable alternative approach to enhance AVF patency.
Collapse
Affiliation(s)
- Gaurav Baranwal
- Vascular and Interventional Radiology Translational Laboratory, Department of Radiology, Mayo Clinic, Rochester, MN 55905, USA
| | - Haseeb Mukhtar
- Vascular and Interventional Radiology Translational Laboratory, Department of Radiology, Mayo Clinic, Rochester, MN 55905, USA
| | - Jamie Kane
- Vascular and Interventional Radiology Translational Laboratory, Department of Radiology, Mayo Clinic, Rochester, MN 55905, USA
| | - Alaura Lemieux
- Vascular and Interventional Radiology Translational Laboratory, Department of Radiology, Mayo Clinic, Rochester, MN 55905, USA
| | - Sanjay Misra
- Vascular and Interventional Radiology Translational Laboratory, Department of Radiology, Mayo Clinic, Rochester, MN 55905, USA
| |
Collapse
|
5
|
Vachharajani TJ, Bhargava V, Sequeira A, Meena P. Existing and Evolving Therapies for Arteriovenous Fistula and Graft Dysfunction. Indian J Nephrol 2024; 34:552-560. [PMID: 39649313 PMCID: PMC11619062 DOI: 10.25259/ijn_528_23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2024] [Accepted: 03/03/2024] [Indexed: 12/10/2024] Open
Abstract
A functional vascular access (VA) is of paramount importance to patients on hemodialysis therapy. While arteriovenous fistula (AVF) remains the preferred VA, their long-term patency is unpredictable. A dysfunctional VA contributes to a high morbidity rate, an increased susceptibility to major adverse cardiovascular events, recurrent hospitalization, and a poor quality of life. The recent innovations in devices and technologies have significantly expanded our options to create and prolong VA patency. Endovascular devices such as WavelinQ and Ellipsys are recent additions to creating a VA. The endovascular creation of AVF helps reduce the wait time and potentially avoids or reduces the duration of catheter use. The bioengineered graft and immediate access arteriovenous graft offer reasonable alternatives in a select group of patients. There is growing evidence that covered stents and drug-coated balloons offer options to prolong the VA patency. Finally, the role of stem cell therapy in VA is currently being explored. This article presents a comprehensive review of the conventional and current developments in the management of a dysfunctional VA.
Collapse
Affiliation(s)
- Tushar J Vachharajani
- Department of Medicine, John D Dingell Veterans Affairs Medical Centre, Detroit, MI, and Wayne State University School of Medicine, Detroit, United States
| | - Vinant Bhargava
- Department of Nephrology, Sir Gangaram Hospital, New Delhi, India
| | - Adrian Sequeira
- Department of Nephrology and Hypertension, LSU Health Shreveport School of Medicine, Detroit, United States
| | - Priti Meena
- Department of Nephrology, All India Institute of Medical Sciences (AIIMS)Bhubaneswar, India
| |
Collapse
|
6
|
Kane J, Lemieux A, Baranwal G, Misra S. The Role of Cardio-Renal Inflammation in Deciding the Fate of the Arteriovenous Fistula in Haemodialysis Therapy. Cells 2024; 13:1637. [PMID: 39404400 PMCID: PMC11475948 DOI: 10.3390/cells13191637] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2024] [Revised: 09/12/2024] [Accepted: 09/27/2024] [Indexed: 10/19/2024] Open
Abstract
Vascular access is an indispensable component of haemodialysis therapy for end-stage kidney disease patients. The arteriovenous fistula (AVF) is most common, but importantly, two-year failure rates are greater than fifty percent. AVF failure can occur due to a lack of suitable vascular remodelling, and inappropriate inflammation preventing maturation, or alternatively neointimal hyperplasia and vascular stenosis preventing long-term use. A comprehensive mechanistic understanding of these processes is still lacking, but recent studies highlight an essential role for inflammation from uraemia and the AVF itself. Inflammation affects each cell in the cascade of AVF failure, the endothelium, the infiltrating immune cells, and the vascular smooth muscle cells. This review examines the role of inflammation in each cell step by step and the influence on AVF failure. Inflammation resulting in AVF failure occurs initially via changes in endothelial cell activation, permeability, and vasoprotective chemokine secretion. Resultingly, immune cells can extravasate into the subendothelial space to release inflammatory cytokines and cause other deleterious changes to the microenvironment. Finally, all these changes modify vascular smooth muscle cell function, resulting in excessive and unchecked hyperplasia and proliferation, eventually leading to stenosis and the failure of the AVF. Finally, the emerging therapeutic options based off these findings are discussed, including mesenchymal stem cells, small-molecule inhibitors, and far-infrared therapies. Recent years have clearly demonstrated a vital role for inflammation in deciding the fate of the AVF, and future works must be centred on this to develop therapies for a hitherto unacceptably underserved patient population.
Collapse
Affiliation(s)
| | | | | | - Sanjay Misra
- Vascular and Interventional Radiology Translational Laboratory, Department of Radiology, Mayo Clinic, 200 First Street SW, Rochester, MN 55905, USA; (J.K.); (A.L.); (G.B.)
| |
Collapse
|
7
|
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
|
8
|
Sharma S, Pandey MK. Radiometals in Imaging and Therapy: Highlighting Two Decades of Research. Pharmaceuticals (Basel) 2023; 16:1460. [PMID: 37895931 PMCID: PMC10610335 DOI: 10.3390/ph16101460] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2023] [Revised: 10/03/2023] [Accepted: 10/05/2023] [Indexed: 10/29/2023] Open
Abstract
The present article highlights the important progress made in the last two decades in the fields of molecular imaging and radionuclide therapy. Advancements in radiometal-based positron emission tomography, single photon emission computerized tomography, and radionuclide therapy are illustrated in terms of their production routes and ease of radiolabeling. Applications in clinical diagnostic and radionuclide therapy are considered, including human studies under clinical trials; their current stages of clinical translations and findings are summarized. Because the metalloid astatine is used for imaging and radionuclide therapy, it is included in this review. In regard to radionuclide therapy, both beta-minus (β-) and alpha (α)-emitting radionuclides are discussed by highlighting their production routes, targeted radiopharmaceuticals, and current clinical translation stage.
Collapse
Affiliation(s)
| | - Mukesh K. Pandey
- Division of Nuclear Medicine, Department of Radiology, Mayo Clinic, Rochester, MN 55905, USA;
| |
Collapse
|
9
|
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
|
10
|
Ning X, Liu N, Sun T, You Y, Luo Y, Kang E, Chen Z, Wang Y, Ren J. Promotion of adipose stem cell transplantation using GelMA hydrogel reinforced by PLCL/ADM short nanofibers. Biomed Mater 2023; 18:065003. [PMID: 37647920 DOI: 10.1088/1748-605x/acf551] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2023] [Accepted: 08/30/2023] [Indexed: 09/01/2023]
Abstract
Adipose-derived mesenchymal stem cells (ADSCs) show poor survival after transplantation, limiting their clinical application. In this study, a series of poly(l-lactide-co-ϵ-caprolactone) (PLCL)/acellular dermal matrix (ADM) nanofiber scaffolds with different proportions were prepared by electrospinning. By studying their morphology, hydrophilicity, tensile mechanics, and biocompatibility, PLCL/ADM nanofiber scaffolds with the best composition ratio (PLCL:ADM = 7:3) were selected to prepare short nanofibers. And based on this, injectable gelatin methacryloyl (GelMA) hydrogel loaded with PLCL/ADM short nanofibers (GelMA-Fibers) was constructed as a transplantation vector of ADSCs. ADSCs and GelMA-Fibers were co-cultured, and the optimal loading concentration of PLCL/ADM nanofibers was investigated by cell proliferation assay, live/dead cell staining, and cytoskeleton stainingin vitro. In vivoinvestigations were also performed by H&E staining, Oil red O staining, and TUNEL staining, and the survival and apoptosis rates of ADSCs transplantedin vivowere analyzed. It was demonstrated that GelMA-Fibers could effectively promote the proliferation of ADSCsin vitro. Most importantly, GelMA-Fibers increased the survival rate of ADSCs transplantation and decreased their apoptosis rate within 14 d. In conclusion, the constructed GelMA-Fibers would provide new ideas and options for stem cell tissue engineering and stem cell-based clinical therapies.
Collapse
Affiliation(s)
- Xuchao Ning
- Department of Cosmetic and Plastic Surgery, Affiliated Hospital of Qingdao University, Qingdao, People's Republic of China
- Department of Plastic Surgery, Qilu Hospital (Qingdao), Cheeloo College of Medicine, Shandong University, Qingdao, People's Republic of China
| | - Na Liu
- Institute of Neuroregeneration and Neurorehabilitation, Qingdao Medical College, Qingdao University, Qingdao, People's Republic of China
| | - Tiancai Sun
- Collaborative Innovation Center for Nanomaterials & Devices, College of Physics, Qingdao University, Qingdao, People's Republic of China
| | - Yong You
- Department of Cosmetic and Plastic Surgery, Affiliated Hospital of Qingdao University, Qingdao, People's Republic of China
| | - Yanan Luo
- Department of Cosmetic and Plastic Surgery, Affiliated Hospital of Qingdao University, Qingdao, People's Republic of China
| | - Enhao Kang
- Department of Pathology, Affiliated Hospital of Qingdao University, Qingdao, People's Republic of China
| | - Zhenyu Chen
- Department of Cosmetic and Plastic Surgery, Affiliated Hospital of Qingdao University, Qingdao, People's Republic of China
| | - Yuanfei Wang
- Central Laboratory, Qingdao Stomatological Hospital Affiliated to Qingdao University, Qingdao, People's Republic of China
| | - Jizhen Ren
- Department of Cosmetic and Plastic Surgery, Affiliated Hospital of Qingdao University, Qingdao, People's Republic of China
| |
Collapse
|
11
|
Hosseiniasl SM, Felgendreff P, Tharwat M, Amiot B, AbuRmilah A, Minshew AM, Bornschlegl AM, Jalan-Sakrikar N, Smart M, Dietz AB, Huebert RC, Nyberg SL. Biodegradable biliary stents coated with mesenchymal stromal cells in a porcine choledochojejunostomy model. Cytotherapy 2023; 25:483-489. [PMID: 36842850 PMCID: PMC10399303 DOI: 10.1016/j.jcyt.2023.01.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2022] [Revised: 01/25/2023] [Accepted: 01/27/2023] [Indexed: 02/27/2023]
Abstract
BACKGROUND AIMS Roux en y anastomosis is a preferred method of biliary reconstruction in liver transplantation that involves living donors or pediatric patients. However, biliary stricture is a frequent and serious complication, accounting for up to 40% of biliary complications in these patients. Previously, we demonstrated that extraluminal delivery of adipose-derived (AD) mesenchymal stromal cells (MSCs) decreased peri-biliary fibrosis and increased neo-angiogenesis in a porcine model of duct-to-duct biliary anastomosis. In this study, we used a porcine model of Roux en y anastomosis to evaluate the beneficial impact of a novel intraluminal MSC delivery system. METHODS Nine animals were divided into three groups: no stent (group 1), bare stent (group 2) and stent coated with AD-MSCs (group 3). All animals underwent cholecystectomy with roux en y choledochojejunostomy. Two animals per group were followed for 4 weeks and one animal per group was followed for 8 weeks. Cholangiograms and blood were sampled at baseline and the end of study. Biliary tissue was collected and examined by Masson trichrome staining and immunohistochemical staining for MSC markers (CD34 and CD44) and for neo-angiogenesis (CD31). RESULTS Two of three animals in group 1 developed an anastomotic site stricture. No strictures were observed in the animals of group 2 or group 3. CD34 and CD44 staining showed that AD-MSCs engrafted successfully at the anastomotic site by intraluminal delivery (group 3). Furthermore, biliary tissue from group 3 showed significantly less fibrosis and increased angiogenesis compared with the other groups. CONCLUSIONS Intraluminal delivery of AD-MSCs resulted in successful biliary engraftment of AD-MSCs as well as reduced peri-biliary fibrosis and increased neo-angiogenesis.
Collapse
Affiliation(s)
| | - Philipp Felgendreff
- Department of Surgery, Mayo Clinic, Rochester, Minnesota, USA; Department for General, Visceral and Vascular Surgery, University Hospital Jena, Jena, Germany
| | - Mohammad Tharwat
- General Surgery Department, Faculty of Medicine, Zagazig University, Zagazig, Egypt
| | - Bruce Amiot
- Department of Surgery, Mayo Clinic, Rochester, Minnesota, USA; William J. von Liebig Center for Transplantation and Clinical Regeneration, Mayo Clinic, Rochester, Minnesota, USA
| | - Anan AbuRmilah
- Department of Surgery, Mayo Clinic, Rochester, Minnesota, USA
| | - Anna M Minshew
- Department of Surgery, Mayo Clinic, Rochester, Minnesota, USA
| | - Alexander M Bornschlegl
- Department of Laboratory Medicine and Pathology, Mayo Clinic and Foundation, Rochester, Minnesota, USA
| | - Nidhi Jalan-Sakrikar
- Gastroenterology Research Unit, Mayo Clinic and Foundation, Rochester, Minnesota, USA
| | - Michele Smart
- Department of Laboratory Medicine and Pathology, Mayo Clinic and Foundation, Rochester, Minnesota, USA
| | - Allan B Dietz
- Department of Laboratory Medicine and Pathology, Mayo Clinic and Foundation, Rochester, Minnesota, USA
| | - Robert C Huebert
- William J. von Liebig Center for Transplantation and Clinical Regeneration, Mayo Clinic, Rochester, Minnesota, USA; Gastroenterology Research Unit, Mayo Clinic and Foundation, Rochester, Minnesota, USA; Division of Gastroenterology and Hepatology, Mayo Clinic and Foundation, Rochester, Minnesota, USA
| | - Scott L Nyberg
- Department of Surgery, Mayo Clinic, Rochester, Minnesota, USA; William J. von Liebig Center for Transplantation and Clinical Regeneration, Mayo Clinic, Rochester, Minnesota, USA.
| |
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
|
Wang S, Wang Y, Xu B, Qin T, Lv Y, Yan H, Shao Y, Fang Y, Zheng S, Qiu Y. Biodistribution of 89Zr-oxine-labeled human bone marrow-derived mesenchymal stem cells by micro-PET/computed tomography imaging in Sprague-Dawley rats. Nucl Med Commun 2022; 43:834-846. [PMID: 35438673 PMCID: PMC9177155 DOI: 10.1097/mnm.0000000000001562] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2022] [Accepted: 03/29/2022] [Indexed: 11/25/2022]
Abstract
PURPOSE To develop a method for labeling human bone marrow mesenchymal stem cells (hMSCs) with 89Zr-oxine to characterize the biodistribution characteristics of hMSCs in normal Sprague-Dawley (SD) rats in real-time by micro-PET-computed tomography (micro-PET/CT) imaging. METHODS 89Zr-oxine complex was synthesized from 89Zr-oxalate and 8-hydroxyquinoline (oxine). After hMSCs were labeled with the 89Zr-oxine complex, the radioactivity retention, viability, proliferation, apoptosis, differentiation, morphology, and phenotype of labeled cells were assessed. The biodistribution of 89Zr-oxine-labeled hMSCs in SD rats was tracked in real-time by micro-PET/CT imaging. RESULTS The cell labeling efficiency was 52.6 ± 0.01%, and 89Zr-oxine was stably retained in cells (66.7 ± 0.9% retention on 7 days after labeling). Compared with the unlabeled hMSCs, 89Zr-oxine labeling did not affect the biological characteristics of cells. Following intravenous administration in SD rats, labeled hMSCs mainly accumulated in the liver (7.35 ± 1.41% ID/g 10 days after labeling, n = 6) and spleen (8.48 ± 1.20% ID/g 10 days after labeling, n = 6), whereas intravenously injected 89Zr-oxalate mainly accumulated in the bone (4.47 ± 0.35% ID/g 10 days after labeling, n = 3). CONCLUSION 89Zr-oxine labeling and micro-PET/CT imaging provide a useful and non-invasive method of assessing the biodistribution of cell therapy products in SD rats. The platform provides a foundation for us to further understand the mechanism of action and migration dynamics of cell therapy products.
Collapse
Affiliation(s)
- Shuzhe Wang
- College of Chemistry and Chemical Engineering, Shanghai University of Engineering Science
- Toxicology Department of China Academy of Pharmaceutical Industry Shanghai InnoStar Biotechnology Co. Ltd, Shanghai
| | - Yan Wang
- Toxicology Department of China Academy of Pharmaceutical Industry Shanghai InnoStar Biotechnology Co. Ltd, Shanghai
| | - Bohua Xu
- Radiographic Evaluation Department of InnoStar Biotechnology Nantong Co. Ltd, Nantong
| | - Tian Qin
- Radiographic Evaluation Department of InnoStar Biotechnology Nantong Co. Ltd, Nantong
| | - Yupeng Lv
- Radiographic Evaluation Department of InnoStar Biotechnology Nantong Co. Ltd, Nantong
| | - Heng Yan
- Radiographic Evaluation Department of InnoStar Biotechnology Nantong Co. Ltd, Nantong
| | - Yifei Shao
- Radiographic Evaluation Department of InnoStar Biotechnology Nantong Co. Ltd, Nantong
| | - Yangyang Fang
- Radiographic Evaluation Department of InnoStar Biotechnology Nantong Co. Ltd, Nantong
| | - Shaoqiu Zheng
- Radiographic Evaluation Department of InnoStar Biotechnology Nantong Co. Ltd, Nantong
- Jiangxi University of Traditional Chinese Medicine, Nanchang
- Yangtze Delta Advanced Research Institute, Yangtze Delta Pharmaceutical College Nantong, Jiangsu, China
| | - Yunliang Qiu
- Toxicology Department of China Academy of Pharmaceutical Industry Shanghai InnoStar Biotechnology Co. Ltd, Shanghai
| |
Collapse
|
14
|
Piryani AK, Kilari S, Takahashi E, DeMartino RR, Mandrekar J, Dietz AB, Misra S. Rationale and Trial Design of MesEnchymal Stem Cell Trial in Preventing Venous Stenosis of Hemodialysis Vascular Access Arteriovenous Fistula (MEST AVF Trial). KIDNEY360 2021; 2:1945-1952. [PMID: 35419530 PMCID: PMC8986037 DOI: 10.34067/kid.0005182021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/10/2021] [Accepted: 09/10/2021] [Indexed: 02/04/2023]
Abstract
Background Hemodialysis arteriovenous fistulas (AVFs) are the preferred vascular access for patients on hemodialysis. In the Hemodialysis Fistula Maturation Study, 44% of the patients achieved unassisted maturation of their fistula without needing an intervention. Venous neointimal hyperplasia (VNH) and subsequent venous stenosis are responsible for lack of maturation. There are no therapies that can prevent VNH/VS formation. The goal of this paper is to present the background, rationale, and trial design of an innovative phase 1/2 clinical study that is investigating the safety of autologous adipose-derived mesenchymal stem cells delivered locally to the adventitia of newly created upper extremity radiocephalic (RCF) or brachiocephalic fistula (BCF). Methods The rationale and preclinical studies used to obtain a physician-sponsored investigational new drug trial are discussed. The trial design and end points are discussed. Results This is an ongoing trial that will complete this year. Conclusion This is a phase 1/2 single-center, randomized trial that will investigate the safety and efficacy of autologous AMSCs in promoting maturation in new upper-extremity AVFs.Clinical Trial registration number: NCT02808208.
Collapse
Affiliation(s)
| | | | | | | | - Jay Mandrekar
- Department of Biostatistics, Mayo Clinic, Rochester, Minnesota
| | - Allan B. Dietz
- Division of Transfusion Medicine and Laboratory Medicine, Mayo Clinic, Rochester, Minnesota
| | - Sanjay Misra
- Department of Radiology, Mayo Clinic, Rochester, Minnesota
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, Minnesota
| |
Collapse
|
15
|
Pandey MK, DeGrado TR. Cyclotron Production of PET Radiometals in Liquid Targets: Aspects and Prospects. Curr Radiopharm 2021; 14:325-339. [PMID: 32867656 PMCID: PMC9909776 DOI: 10.2174/1874471013999200820165734] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Revised: 07/11/2020] [Accepted: 07/23/2020] [Indexed: 11/22/2022]
Abstract
The present review describes the methodological aspects and prospects of the production of Positron Emission Tomography (PET) radiometals in a liquid target using low-medium energy medical cyclotrons. The main objective of this review is to delineate and discuss the critical factors involved in the liquid target production of radiometals, including type of salt solution, solution composition, beam energy, beam current, the effect of irradiation duration (length of irradiation) and challenges posed by in-target chemistry in relation with irradiation parameters. We also summarize the optimal parameters for the production of various radiometals in liquid targets. Additionally, we discuss the future prospects of PET radiometals production in the liquid targets for academic research and clinical applications. Significant emphasis has been given to the production of 68Ga using liquid targets due to the growing demand for 68Ga labeled PSMA vectors, [68Ga]- Ga-DOTATATE, [68Ga]Ga-DOTANOC and some upcoming 68Ga labeled radiopharmaceuticals. Other PET radiometals included in the discussion are 86Y, 63Zn and 89Zr.
Collapse
Affiliation(s)
- Mukesh K. Pandey
- Division of Nuclear Medicine, Department of Radiology, Mayo Clinic Rochester, Minneapolis, 55905, USA,Address correspondence to this author at the Division of Nuclear Medicine, Department of Radiology, Mayo Clinic Rochester, Minneapolis, 55905, USA; E-mail:
| | - Timothy R. DeGrado
- Division of Nuclear Medicine, Department of Radiology, Mayo Clinic Rochester, Minneapolis, 55905, USA
| |
Collapse
|
16
|
Yang K, Xie D, Lin W, Xiang P, Peng C. Adipose mesenchymal stem cells and gingival mesenchymal stem cells have a comparable effect in endothelium repair. Exp Ther Med 2021; 22:1415. [PMID: 34676008 PMCID: PMC8524764 DOI: 10.3892/etm.2021.10851] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2020] [Accepted: 07/14/2021] [Indexed: 12/14/2022] Open
Abstract
Restenosis is the major factor influencing the long-term success rate of angioplasty and stent implantation and effective strategies to prevent restenosis remain limited. Mesenchymal stem cells (MSCs) are pluripotent stem cells capable of self-renewal and multidirectional differentiation, which may be able to promote endothelium repair, thereby reducing restenosis. The present study aimed to evaluate the effects of adipose MSCs (AMSCs) and gingival MSCs (GMSCs) on endothelium repair. MSCs were isolated from two human tissue types, namely adipose tissue and gingival tissue, and the effects of AMSCs and GMSCs in ex vivo endothelium repair and on vascular smooth muscle cell (SMC) growth were examined. To compare the feasibility of using AMSCs and GMSCs for the repair of endothelium damage in endothelial cell (EC) damage and vasoproliferative disorders, an ex vivo model of endothelium repair in a co-culture system was developed. It was indicated that AMSCs and GMSCs expressed characteristic MSC markers (CD105 and CD166). 3H-thymidine incorporation in the co-culture group of AMSCs and SMCs in the presence of ECs was lower compared with that in the GMSC and SMC co-culture group. The protein expression level of proliferating cell nuclear antigen in the co-culture group of AMSCs and SMCs in the presence of ECs were lower compared with that in the GMSC and SMC co-culture group. After co-culture with ECs for 5 days, 25.71±3.08% of AMSCs began to express CD31 protein and 20.06±2.09% of GMSCs began to express CD31 protein. Furthermore, anti-VEGF antibody was able to inhibit MSC differentiation. Collectively, the present results suggested that seeding of AMSCs had a stronger effect to inhibit the proliferation and migration of SMCs compared with GMSCs.
Collapse
Affiliation(s)
- Ke Yang
- Department of Cardiology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong 510630, P.R. China
| | - Dongmei Xie
- Department of Cardiology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong 510630, P.R. China
| | - Wanwen Lin
- Department of Cardiology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong 510630, P.R. China
| | - Peng Xiang
- Key Laboratory for Stem Cells and Tissue Engineering, Center for Stem Cell Biology and Tissue Engineering, Ministry of Education, Sun Yat-sen University, Guangzhou, Guangdong 510600, P.R. China
| | - Chaoquan Peng
- Department of Cardiology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong 510630, P.R. China
| |
Collapse
|
17
|
Takahashi EA, Kilari S, Misra S. Novel Clinical Therapies and Technologies in Dialysis Vascular Access. KIDNEY360 2021; 2:1373-1379. [PMID: 35369655 PMCID: PMC8676382 DOI: 10.34067/kid.0002962021] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/03/2021] [Accepted: 06/10/2021] [Indexed: 02/08/2023]
Abstract
The hemodialysis population continues to grow. Although procedures for dialysis have existed for >60 years, significant challenges with vascular access to support hemodialysis persist. Failure of arteriovenous fistulas (AVFs) to mature, loss of AVF and graft patency, thrombosis, and infection hinder long-term access, and add extra health care costs and patient morbidity. There have been numerous innovations over the last decade aimed at addressing the issues. In this study, we review the literature and summarize the recent evolution of drug delivery, graft development, minimally invasive AVF creation, and stem-cell therapy for hemodialysis access.
Collapse
Affiliation(s)
| | | | - Sanjay Misra
- Department of Radiology, Mayo Clinic, Rochester, Minnesota
| |
Collapse
|
18
|
Misra S, Kilari S, Yang B, Sharma A, Wu CC, Vazquez-Padron RI, Broadwater J. Anti Human CX3CR1 VHH Molecule Attenuates Venous Neointimal Hyperplasia of Arteriovenous Fistula in Mouse Model. J Am Soc Nephrol 2021; 32:1630-1648. [PMID: 33893223 PMCID: PMC8425661 DOI: 10.1681/asn.2020101458] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2020] [Accepted: 02/17/2021] [Indexed: 02/04/2023] Open
Abstract
BACKGROUND Fractalkine receptor 1 (CX3CR1) mediates macrophage infiltration and accumulation, causing venous neointimal hyperplasia (VNH)/venous stenosis (VS) in arteriovenous fistula (AVF). The effect of blocking CX3CR1 using an anti-human variable VHH molecule (hCX3CR1 VHH, BI 655088) on VNH/VS was determined using a humanized mouse in which the human CX3CR1 (hCX3CR1) gene was knocked in (KI). METHODS Whole-transcriptomic RNA sequencing with bioinformatics analysis was used on human stenotic AVF samples, C57BL/6J, hCX3CR1 KI mice with AVF and CKD, and in in vitro experiments to identify the pathways involved in preventing VNH/VS formation after hCX3CR1 VHH administration. RESULTS Accumulation of CX3CR1 and CD68 was significantly increased in stenotic human AVFs. In C57BL/6J mice with AVF, there was increased Cx3cr1, Cx3cl1, Cd68, and Tnf-α gene expression, and increased immunostaining of CX3CR1 and CD68. In hCX3CR1-KI mice treated with hCX3CR1 VHH molecule (KI-A), compared with vehicle controls (KI-V), there was increased lumen vessel area and patency, and decreased neointima in the AVF outflow veins. RNA-seq analysis identified TNF-α and NF-κB as potential targets of CX3CR1 inhibition. In KI-A-treated vessels compared with KI-V, there was decreased gene expression of Tnf- α, Mcp-1, and Il-1 β; with reduction of Cx3cl1, NF-κB, and Cd68; decreased M1, Ly6C, smooth muscle cells, fibroblast-activated protein, fibronectin, and proliferation; and increased TUNEL and M2 staining. In cell culture, monocytes stimulated with PMA and treated with hCX3CR1 VHH had decreased TNF- α, CD68, proliferation, and migration. CONCLUSIONS CX3CR1 blockade reduces VNH/VS formation by decreasing proinflammatory cues.
Collapse
Affiliation(s)
- 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
| | - Sreenivasulu Kilari
- Vascular and Interventional Radiology Translational Laboratory, Department of Radiology, Mayo Clinic, Rochester, Minnesota
| | - Binxia Yang
- Vascular and Interventional Radiology Translational Laboratory, Department of Radiology, Mayo Clinic, Rochester, Minnesota
| | - Amit Sharma
- Vascular and Interventional Radiology Translational Laboratory, Department of Radiology, Mayo Clinic, Rochester, Minnesota
| | - Chih-Cheng Wu
- Cardiovascular Center, National Taiwan University Hospital, Hsin-chu, Taiwan
| | - Roberto I. Vazquez-Padron
- Division of Vascular Surgery, Department of Surgery, Leonard M. Miller School of Medicine, University of Miami, Miami, Florida
| | - John Broadwater
- CardioMetabolic Diseases Research, Boehringer Ingelheim Pharmaceuticals, Inc., Ridgefield, Connecticut
| |
Collapse
|
19
|
Brahmbhatt AN, Misra S. Stem Cell Delivery for the Treatment of Arteriovenous Fistula Failure. STEM CELL THERAPY FOR VASCULAR DISEASES 2021:281-297. [DOI: 10.1007/978-3-030-56954-9_13] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/07/2025]
|
20
|
Rajendran RL, Jogalekar MP, Gangadaran P, Ahn BC. Noninvasive in vivo cell tracking using molecular imaging: A useful tool for developing mesenchymal stem cell-based cancer treatment. World J Stem Cells 2020; 12:1492-1510. [PMID: 33505597 PMCID: PMC7789123 DOI: 10.4252/wjsc.v12.i12.1492] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Revised: 10/05/2020] [Accepted: 10/21/2020] [Indexed: 02/06/2023] Open
Abstract
Mounting evidence has emphasized the potential of cell therapies in treating various diseases by restoring damaged tissues or replacing defective cells in the body. Cell therapies have become a strong therapeutic modality by applying noninvasive in vivo molecular imaging for examining complex cellular processes, understanding pathophysiological mechanisms of diseases, and evaluating the kinetics/dynamics of cell therapies. In particular, mesenchymal stem cells (MSCs) have shown promise in recent years as drug carriers for cancer treatment. They can also be labeled with different probes and tracked in vivo to assess the in vivo effect of administered cells, and to optimize therapy. The exact role of MSCs in oncologic diseases is not clear as MSCs have been shown to be involved in tumor progression and inhibition, and the exact interactions between MSCs and specific cancer microenvironments are not clear. In this review, a multitude of labeling approaches, imaging modalities, and the merits/demerits of each strategy are outlined. In addition, specific examples of the use of MSCs and in vivo imaging in cancer therapy are provided. Finally, present limitations and future outlooks in terms of the translation of different imaging approaches in clinics are discussed.
Collapse
Affiliation(s)
| | | | - Prakash Gangadaran
- Department of Nuclear Medicine, School of Medicine, Kyungpook National University, Daegu 41944, South Korea
- BK21 Plus KNU Biomedical Convergence Program, Department of Biomedical Science, School of Medicine, Kyungpook National University, Daegu 41944, South Korea
| | - Byeong-Cheol Ahn
- BK21 Plus KNU Biomedical Convergence Program, Department of Biomedical Science, School of Medicine, Kyungpook National University, Daegu 41944, South Korea
- Department of Nuclear Medicine, School of Medicine, Kyungpook National University, Kyungpook National University Hospital, Daegu 41944, South Korea.
| |
Collapse
|
21
|
Che Man R, Sulaiman N, Ishak MF, Bt Hj Idrus R, Abdul Rahman MR, Yazid MD. The Effects of Pro-Inflammatory and Anti-Inflammatory Agents for the Suppression of Intimal Hyperplasia: An Evidence-Based Review. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2020; 17:ijerph17217825. [PMID: 33114632 PMCID: PMC7672569 DOI: 10.3390/ijerph17217825] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Revised: 09/30/2020] [Accepted: 10/07/2020] [Indexed: 12/11/2022]
Abstract
Anti-atherogenic therapy is crucial in halting the progression of inflammation-induced intimal hyperplasia. The aim of this concise review was to methodically assess the recent findings of the different approaches, mainly on the recruitment of chemokines and/or cytokine and its effects in combating the intimal hyperplasia caused by various risk factors. Pubmed and Scopus databases were searched, followed by article selection based on pre-set inclusion and exclusion criteria. The combination of keywords used were monocyte chemoattractant protein-1 OR MCP-1 OR TNF-alpha OR TNF-α AND hyperplasia OR intimal hyperplasia OR neointimal hyperplasia AND in vitro. These keywords combination was incorporated in the study and had successfully identified 77 articles, with 22 articles were acquired from Pubmed, whereas 55 articles were obtained from Scopus. However, after title screening, only twelve articles meet the requirements of defined inclusion criteria. We classified the data into 4 different approaches, i.e., utilisation of natural product, genetic manipulation and protein inhibition, targeted drugs in clinical setting, and chemokine and cytokines induction. Most of the articles are working on genetic manipulation targeted on specific pathway to inhibit the pro-inflammatory factors expression. We also found that the utilisation of chemokine- and cytokine-related treatments are emerging throughout the years. However, there is no study utilising the combination of approaches that might give a better outcome in combating intimal hyperplasia. Hopefully, this concise review will provide an insight regarding the usage of different novel approaches in halting the progression of intimal hyperplasia, which serves as a key factor for the development of atherosclerosis in cardiovascular disease.
Collapse
Affiliation(s)
- Rohaina Che Man
- Centre for Tissue Engineering & Regenerative Medicine, Universiti Kebangsaan Malaysia Medical Centre, Jalan Yaacob Latif, Cheras 56000, Kuala Lumpur, Malaysia; (R.C.M.); (N.S.); (M.F.I.); (R.B.H.I.)
| | - Nadiah Sulaiman
- Centre for Tissue Engineering & Regenerative Medicine, Universiti Kebangsaan Malaysia Medical Centre, Jalan Yaacob Latif, Cheras 56000, Kuala Lumpur, Malaysia; (R.C.M.); (N.S.); (M.F.I.); (R.B.H.I.)
| | - Mohamad Fikeri Ishak
- Centre for Tissue Engineering & Regenerative Medicine, Universiti Kebangsaan Malaysia Medical Centre, Jalan Yaacob Latif, Cheras 56000, Kuala Lumpur, Malaysia; (R.C.M.); (N.S.); (M.F.I.); (R.B.H.I.)
| | - Ruszymah Bt Hj Idrus
- Centre for Tissue Engineering & Regenerative Medicine, Universiti Kebangsaan Malaysia Medical Centre, Jalan Yaacob Latif, Cheras 56000, Kuala Lumpur, Malaysia; (R.C.M.); (N.S.); (M.F.I.); (R.B.H.I.)
- Department of Physiology, Faculty of Medicine, Universiti Kebangsaan Malaysia Medical Centre, Jalan Yaacob Latif, Cheras 56000, Kuala Lumpur, Malaysia
| | - Mohd Ramzisham Abdul Rahman
- Department of Surgery, Faculty of Medicine, Universiti Kebangsaan Malaysia Medical Centre, Jalan Yaacob Latif, Cheras 56000, Kuala Lumpur, Malaysia;
| | - Muhammad Dain Yazid
- Centre for Tissue Engineering & Regenerative Medicine, Universiti Kebangsaan Malaysia Medical Centre, Jalan Yaacob Latif, Cheras 56000, Kuala Lumpur, Malaysia; (R.C.M.); (N.S.); (M.F.I.); (R.B.H.I.)
- Correspondence: ; Tel.: +603-9145-6995
| |
Collapse
|
22
|
Cai C, Kilari S, Zhao C, Simeon ML, Misra A, Li Y, van Wijnen AJ, Mukhopadhyay D, Misra S. Therapeutic Effect of Adipose Derived Mesenchymal Stem Cell Transplantation in Reducing Restenosis in a Murine Angioplasty Model. J Am Soc Nephrol 2020; 31:1781-1795. [PMID: 32587073 DOI: 10.1681/asn.2019101042] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2019] [Accepted: 04/27/2020] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Percutaneous transluminal angioplasty (PTA) is the first line of treatment for stenosis in the arteriovenous fistula (AVF) created to provide access for hemodialysis, but resenosis still occurs. Transplants of adipose-derived mesenchymal stem cells (AMSCs) labeled with green fluorescent protein (GFP) to the adventitia could reduce pro-inflammatory gene expression, possibly restoring patency in a murine model of PTA for venous stenosis. METHODS Partial nephrectomy of male C57BL/6J mice induced CKD. Placement of the AVF was 28 days later and, 14 days after that, PTA of the stenotic outflow vein was performed with delivery of either vehicle control or AMSCs (5×105) to the adventitia of the vein. Mice were euthanized 3 days later and gene expression for interleukin-1 beta (IL-1β) and tumor necrosis factor-alpha TNF-α) analyzed, and histopathologic analysis performed on day 14 and 28. GFP (+) AMSCs were tracked after transplantation for up to 28 days and Doppler ultrasound performed weekly after AVF creation. RESULTS Gene and protein expression of IL-1β and TNF-α, fibrosis, proliferation, apoptosis and smooth muscle actin decreased, and the proportions of macrophage types (M2/M1) shifted in a manner consistent with less inflammation in AMSC-transplanted vessels compared to controls. After PTA, AMSC-treated vessels had significantly higher wall shear stress, average peak, and mean velocity, with increased lumen vessel area and decreased neointima/media area ratio compared to the control group. At 28 days after delivery, GFP (+) AMSC were present in the adventitia of the outflow vein. CONCLUSIONS AMSC-treated vessels had improved vascular remodeling with decreased proinflammatory gene expression, inflammation, and fibrotic staining compared to untreated vessels.
Collapse
Affiliation(s)
- Chuanqi Cai
- Department of Vascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Vascular and Interventional Radiology Translational Laboratory, Department of Radiology, Mayo Clinic, Rochester, Minnesota
| | - Sreenivasulu Kilari
- Vascular and Interventional Radiology Translational Laboratory, Department of Radiology, Mayo Clinic, Rochester, Minnesota
| | - Chenglei Zhao
- Vascular and Interventional Radiology Translational Laboratory, Department of Radiology, Mayo Clinic, Rochester, Minnesota.,Department of Vascular Surgery, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Michael L Simeon
- Vascular and Interventional Radiology Translational Laboratory, Department of Radiology, Mayo Clinic, Rochester, Minnesota
| | - Avanish Misra
- Vascular and Interventional Radiology Translational Laboratory, Department of Radiology, Mayo Clinic, Rochester, Minnesota
| | - Yiqing Li
- Department of Vascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Andre J van Wijnen
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, Minnesota.,Department of Biochemistry and Molecular Biology, 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.,Department of Radiology, Vascular and Interventional Radiology, Mayo Clinic, Rochester, Minnesota
| |
Collapse
|
23
|
Yoon JK, Park BN, Ryu EK, An YS, Lee SJ. Current Perspectives on 89Zr-PET Imaging. Int J Mol Sci 2020; 21:ijms21124309. [PMID: 32560337 PMCID: PMC7352467 DOI: 10.3390/ijms21124309] [Citation(s) in RCA: 88] [Impact Index Per Article: 17.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Revised: 06/13/2020] [Accepted: 06/15/2020] [Indexed: 12/14/2022] Open
Abstract
89Zr is an emerging radionuclide that plays an essential role in immuno-positron emission tomography (PET) imaging. The long half-life of 89Zr (t1/2 = 3.3 days) is favorable for evaluating the in vivo distribution of monoclonal antibodies. Thus, the use of 89Zr is promising for monitoring antibody-based cancer therapies. Immuno-PET combines the sensitivity of PET with the specificity of antibodies. A number of studies have been conducted to investigate the feasibility of 89Zr immuno-PET imaging for predicting the efficacy of radioimmunotherapy and antibody therapies, imaging target expression, detecting target-expressing tumors, and the monitoring of anti-cancer chemotherapies. In this review, we summarize the current status of PET imaging using 89Zr in both preclinical and clinical studies by highlighting the use of immuno-PET for the targets of high clinical relevance. We also present 89Zr-PET applications other than immuno-PET, such as nanoparticle imaging and cell tracking. Finally, we discuss the limitations and the ongoing research being performed to overcome the remaining hurdles.
Collapse
Affiliation(s)
- Joon-Kee Yoon
- Department of Nuclear Medicine & Molecular Imaging, Ajou University School of Medicine, Worldcup-ro 164, Suwon 16499, Korea; (B.-N.P.); (Y.-S.A.); (S.-J.L.)
- Correspondence: ; Tel.: +82-31-219-4303
| | - Bok-Nam Park
- Department of Nuclear Medicine & Molecular Imaging, Ajou University School of Medicine, Worldcup-ro 164, Suwon 16499, Korea; (B.-N.P.); (Y.-S.A.); (S.-J.L.)
| | - Eun-Kyoung Ryu
- Division of Magnetic Resonance, Korea Basic Science Institute, 162, Yeongudanji-ro, Cheongju 28119, Korea;
| | - Young-Sil An
- Department of Nuclear Medicine & Molecular Imaging, Ajou University School of Medicine, Worldcup-ro 164, Suwon 16499, Korea; (B.-N.P.); (Y.-S.A.); (S.-J.L.)
| | - Su-Jin Lee
- Department of Nuclear Medicine & Molecular Imaging, Ajou University School of Medicine, Worldcup-ro 164, Suwon 16499, Korea; (B.-N.P.); (Y.-S.A.); (S.-J.L.)
| |
Collapse
|
24
|
Zhang Y, Sharma A, Joo DJ, Nelson E, AbuRmilah A, Amiot BP, Boyer CJ, Alexander JS, Jalan-Sakrikar N, Martin J, Moreira R, Chowdhury SA, Smart M, Dietz AB, Nyberg SL, Heimbach JK, Huebert RC. Autologous Adipose Tissue-Derived Mesenchymal Stem Cells Introduced by Biliary Stents or Local Immersion in Porcine Bile Duct Anastomoses. Liver Transpl 2020; 26:100-112. [PMID: 31742878 PMCID: PMC7061488 DOI: 10.1002/lt.25682] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/16/2019] [Accepted: 10/29/2019] [Indexed: 12/12/2022]
Abstract
Biliary complications (strictures and leaks) represent major limitations in living donor liver transplantation. Mesenchymal stem cells (MSCs) are a promising modality to prevent biliary complications because of immunosuppressive and angiogenic properties. Our goal was to evaluate the safety of adipose-derived MSC delivery to biliary anastomoses in a porcine model. Secondary objectives were defining the optimal method of delivery (intraluminal versus extraluminal) and to investigate MSC engraftment, angiogenesis, and fibrosis. Pigs were divided into 3 groups. Animals underwent adipose collection, MSC isolation, and expansion. Two weeks later, animals underwent bile duct transection, reanastomosis, and stent insertion. Group 1 received plastic stents wrapped in unseeded Vicryl mesh. Group 2 received stents wrapped in MSC-seeded mesh. Group 3 received unwrapped stents with the anastomosis immersed in an MSC suspension. Animals were killed 1 month after stent insertion when cholangiograms and biliary tissue were obtained. Serum was collected for liver biochemistries. Tissue was used for hematoxylin-eosin and trichrome staining and immunohistochemistry for MSC markers (CD44 and CD34) and for a marker of neoangiogenesis (CD31). There were no intraoperative complications. One pig died on postoperative day 3 due to acute cholangitis. All others recovered without complications. Cholangiography demonstrated no biliary leaks and minimal luminal narrowing. Surviving animals exhibited no symptoms, abnormal liver biochemistries, or clinically significant biliary stricturing. Group 3 showed significantly greater CD44 and CD34 staining, indicating MSC engraftment. Fibrosis was reduced at the anastomotic site in group 3 based on trichrome stain. CD31 staining of group 3 was more pronounced, supporting enhanced neoangiogenesis. In conclusion, adipose-derived MSCs were safely applied to biliary anastomoses. MSCs were locally engrafted within the bile duct and may have beneficial effects in terms of fibrosis and angiogenesis.
Collapse
Affiliation(s)
- Y Zhang
- Department of Surgery, Mayo Clinic and Foundation, Rochester, MN;,Chongqing University Cancer Hospital, Chongqing, China
| | - A Sharma
- Department of Surgery, Mayo Clinic and Foundation, Rochester, MN
| | - DJ Joo
- Department of Surgery, Mayo Clinic and Foundation, Rochester, MN;,Department of Surgery, Yonsei University, Seoul, Korea
| | - E Nelson
- Department of Surgery, Mayo Clinic and Foundation, Rochester, MN
| | - A AbuRmilah
- Department of Surgery, Mayo Clinic and Foundation, Rochester, MN
| | - BP Amiot
- Department of Surgery, Mayo Clinic and Foundation, Rochester, MN
| | - CJ Boyer
- Department of Molecular and Cellular Physiology, Louisiana State University Health Sciences Center, Shreveport, LA
| | - JS Alexander
- Department of Molecular and Cellular Physiology, Louisiana State University Health Sciences Center, Shreveport, LA
| | - N Jalan-Sakrikar
- Gastroenterology Research Unit, Mayo Clinic and Foundation, Rochester, MN
| | - J Martin
- Division of Gastroenterology and Hepatology, Mayo Clinic and Foundation, Rochester, MN
| | - R Moreira
- Department of Laboratory Medicine and Pathology, Mayo Clinic and Foundation, Rochester, MN
| | - SA Chowdhury
- Gastroenterology Research Unit, Mayo Clinic and Foundation, Rochester, MN
| | - M Smart
- Department of Laboratory Medicine and Pathology, Mayo Clinic and Foundation, Rochester, MN
| | - AB Dietz
- Department of Laboratory Medicine and Pathology, Mayo Clinic and Foundation, Rochester, MN
| | - SL Nyberg
- Department of Surgery, Mayo Clinic and Foundation, Rochester, MN;,William J. von Liebig Center for Transplantation and Clinical Regeneration, Mayo Clinic and Foundation, Rochester, MN
| | - JK Heimbach
- Department of Surgery, Mayo Clinic and Foundation, Rochester, MN;,William J. von Liebig Center for Transplantation and Clinical Regeneration, Mayo Clinic and Foundation, Rochester, MN
| | - RC Huebert
- Gastroenterology Research Unit, Mayo Clinic and Foundation, Rochester, MN.,Division of Gastroenterology and Hepatology, Mayo Clinic and Foundation, Rochester, MN.,William J. von Liebig Center for Transplantation and Clinical Regeneration, Mayo Clinic and Foundation, Rochester, MN
| |
Collapse
|
25
|
Abstract
Regenerative medicine with the use of stem cells has appeared as a potential therapeutic alternative for many disease states. Despite initial enthusiasm, there has been relatively slow transition to clinical trials. In large part, numerous questions remain regarding the viability, biology and efficacy of transplanted stem cells in the living subject. The critical issues highlighted the importance of developing tools to assess these questions. Advances in molecular biology and imaging have allowed the successful non-invasive monitoring of transplanted stem cells in the living subject. Over the years these methodologies have been updated to assess not only the viability but also the biology of transplanted stem cells. In this review, different imaging strategies to study the viability and biology of transplanted stem cells are presented. Use of these strategies will be critical as the different regenerative therapies are being tested for clinical use.
Collapse
Affiliation(s)
- Fakhar Abbas
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN, USA
| | - Joseph C. Wu
- Molecular Imaging Program at Stanford, Stanford University, Stanford, CA, USA
- Department of Medicine (Cardiology), Stanford University, Stanford, CA, USA
| | - Sanjiv Sam Gambhir
- Molecular Imaging Program at Stanford, Stanford University, Stanford, CA, USA
- Department of Bio-Engineering, Stanford University, Stanford, CA, USA
| | | |
Collapse
|
26
|
Liang M, Guo Q, Huang F, Han G, Song K, Luo J, Cheng H, Hu H, Peden EK, Chen C, Mitch WE, Du J, Fu X, Truong L, Cheng J. Notch signaling in bone marrow-derived FSP-1 cells initiates neointima formation in arteriovenous fistulas. Kidney Int 2019; 95:1347-1358. [PMID: 30799025 PMCID: PMC6763204 DOI: 10.1016/j.kint.2018.11.027] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2018] [Revised: 11/02/2018] [Accepted: 11/08/2018] [Indexed: 11/16/2022]
Abstract
Neointima formation is a major contributor to arteriovenous fistula (AVF) failure. We have previously shown that activation of the Notch signaling pathway contributes to neointima formation by promoting the migration of vascular smooth muscle cells (VSMCs) into the venous anastomosis. In the current study we investigated the mechanisms underlying the dedifferentiation and migration of VSMCs, and in particular the role of bone marrow-derived fibroblast specific protein 1 (FSP-1)+ cells, another cell type found in models of vascular injury. Using VSMC-specific reporter mice, we found that most of the VSMCs participating in AVF neointima formation originated from dedifferentiated VSMCs. We also observed infiltration of bone marrow-derived FSP-1+ cells into the arterial anastomosis where they could interact with VSMCs. In vitro, conditioned media from FSP-1+ cells stimulated VSMC proliferation and phenotype switching. Activated Notch signaling transformed FSP-1+ cells into type I macrophages and stimulated secretion of cytokines and growth factors. Pretreatment with a Notch inhibitor or knockout of the canonical downstream factor RBP-Jκ in bone marrow-derived FSP1+ cells decreased FSP1+ cell infiltration into murine AVFs, attenuating VSMC dedifferentiation and neointima formation. Our results suggest that targeting Notch signaling could provide a new therapeutic strategy to improve AVF patency.
Collapse
MESH Headings
- Animals
- Arteriovenous Shunt, Surgical/adverse effects
- Cell Dedifferentiation/drug effects
- Cells, Cultured
- Disease Models, Animal
- Humans
- Immunoglobulin J Recombination Signal Sequence-Binding Protein/genetics
- Immunoglobulin J Recombination Signal Sequence-Binding Protein/metabolism
- Macrophages/drug effects
- Macrophages/metabolism
- Male
- Mice
- Mice, Knockout
- Muscle, Smooth, Vascular/cytology
- Myocytes, Smooth Muscle/drug effects
- Myocytes, Smooth Muscle/metabolism
- Myocytes, Smooth Muscle/pathology
- Neointima/etiology
- Neointima/pathology
- Neointima/prevention & control
- Primary Cell Culture
- Receptors, Notch/antagonists & inhibitors
- Receptors, Notch/metabolism
- Renal Dialysis/adverse effects
- Renal Dialysis/methods
- Renal Insufficiency, Chronic/therapy
- S100 Calcium-Binding Protein A4/metabolism
- Signal Transduction/drug effects
- Vascular Patency/drug effects
Collapse
Affiliation(s)
- Ming Liang
- Department of Nephrology, Guangzhou First People's Hospital, Guangzhou Medical University, Guangzhou, China; Selzman Institute for Kidney Health, Section of Nephrology, Department of Medicine, Baylor College of Medicine, Houston, Texas, USA
| | - Qunying Guo
- Selzman Institute for Kidney Health, Section of Nephrology, Department of Medicine, Baylor College of Medicine, Houston, Texas, USA
| | - Fengzhang Huang
- Selzman Institute for Kidney Health, Section of Nephrology, Department of Medicine, Baylor College of Medicine, Houston, Texas, USA
| | - Guofeng Han
- Selzman Institute for Kidney Health, Section of Nephrology, Department of Medicine, Baylor College of Medicine, Houston, Texas, USA
| | - Ke Song
- Selzman Institute for Kidney Health, Section of Nephrology, Department of Medicine, Baylor College of Medicine, Houston, Texas, USA
| | - Jinlong Luo
- Selzman Institute for Kidney Health, Section of Nephrology, Department of Medicine, Baylor College of Medicine, Houston, Texas, USA
| | - Hunter Cheng
- Selzman Institute for Kidney Health, Section of Nephrology, Department of Medicine, Baylor College of Medicine, Houston, Texas, USA
| | - Hongzhen Hu
- Pain Center, Washington University, Saint Louis, Missouri, USA
| | - Eric K Peden
- Department of Vascular Surgery, DeBakey Heart and Vascular Institute, Houston Methodist Hospital, Houston, Texas, USA
| | - Changyi Chen
- Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, Texas, USA
| | - William E Mitch
- Selzman Institute for Kidney Health, Section of Nephrology, Department of Medicine, Baylor College of Medicine, Houston, Texas, USA
| | - Jie Du
- Beijing AnZhen Hospital, Capital Medical University, The Key Laboratory of Remodeling-related Cardiovascular Diseases, Ministry of Education, Beijing Institute of Heart, Lung and Blood Vessel Diseases, Beijing, China
| | - Xiaodong Fu
- Department of Nephrology, Guangzhou First People's Hospital, Guangzhou Medical University, Guangzhou, China
| | - Luan Truong
- Department of Pathology and Genomic Medicine, Houston Methodist Hospital, Methodist Hospital Research Institute, Houston, Texas, USA
| | - Jizhong Cheng
- Selzman Institute for Kidney Health, Section of Nephrology, Department of Medicine, Baylor College of Medicine, Houston, Texas, USA.
| |
Collapse
|
27
|
Shiu YT, Rotmans JI, Geelhoed WJ, Pike DB, Lee T. Arteriovenous conduits for hemodialysis: how to better modulate the pathophysiological vascular response to optimize vascular access durability. Am J Physiol Renal Physiol 2019; 316:F794-F806. [PMID: 30785348 PMCID: PMC6580244 DOI: 10.1152/ajprenal.00440.2018] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2018] [Revised: 02/04/2019] [Accepted: 02/17/2019] [Indexed: 12/11/2022] Open
Abstract
Vascular access is the lifeline for patients on hemodialysis. Arteriovenous fistulas (AVFs) are the preferred vascular access, but AVF maturation failure remains a significant clinical problem. Currently, there are no effective therapies available to prevent or treat AVF maturation failure. AVF maturation failure frequently results from venous stenosis at the AVF anastomosis, which is secondary to poor outward vascular remodeling and excessive venous intimal hyperplasia that narrows the AVF lumen. Arteriovenous grafts (AVGs) are the next preferred vascular access when an AVF creation is not possible. AVG failure is primarily the result of venous stenosis at the vein-graft anastomosis, which originates from intimal hyperplasia development. Although there has been advancement in our knowledge of the pathophysiology of AVF maturation and AVG failure, this has not translated into effective therapies for these two important clinical problems. Further work will be required to dissect out the mechanisms of AVF maturation failure and AVG failure to develop more specific therapies. This review highlights the major recent advancements in AVF and AVG biology, reviews major clinical trials, and discusses new areas for future research.
Collapse
Affiliation(s)
- Yan-Ting Shiu
- Division of Nephrology, University of Utah , Salt Lake City, Utah
| | - Joris I Rotmans
- Department of Internal Medicine, Leiden University Medical Center , Leiden , The Netherlands
| | - Wouter Jan Geelhoed
- Department of Internal Medicine, Leiden University Medical Center , Leiden , The Netherlands
| | - Daniel B Pike
- Division of Nephrology, University of Utah , Salt Lake City, Utah
| | - Timmy Lee
- Department of Medicine and Division of Nephrology, University of Alabama at Birmingham , Birmingham, Alabama
- Veterans Affairs Medical Center , Birmingham, Alabama
| |
Collapse
|
28
|
Evaluation of Venous Stenosis Angioplasty in a Murine Arteriovenous Fistula Model. J Vasc Interv Radiol 2019; 30:1512-1521.e3. [PMID: 30902494 DOI: 10.1016/j.jvir.2018.11.032] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2018] [Revised: 11/19/2018] [Accepted: 11/23/2018] [Indexed: 12/14/2022] Open
Abstract
PURPOSE To develop a clinically relevant model of percutaneous transluminal angioplasty (PTA) of venous stenosis in mice with arteriovenous fistula (AVF); to test the hypothesis that there is increased wall shear stress (WSS) after PTA; and to histologically characterize the vessels. MATERIALS AND METHODS Thirteen C57BL/6J male mice, 6-8 weeks old, underwent partial nephrectomy to create chronic kidney disease. Twenty-eight days later, an AVF was created from the right external jugular vein to the left carotid artery. Fourteen days later, an angioplasty or sham procedure was performed, and the mice were sacrificed 14 days later for histologic evaluation to identify the cells contributing to the vascular remodeling (α-SMA, FSP-1, CD31, and CD68), proliferation (Ki-67), cell death (TUNEL), and hypoxia staining (HIF-1α). Histomorphometric analysis was performed to assess lumen area, neointima+media area, and cellular density. Ultrasound was performed weekly after creation of the AVF. RESULTS Venous stenosis occurred 14 days after the creation of an AVF. PTA-treated vessels had significantly higher WSS; average peak systolic velocity, with increased lumen vessel area; and decreased neointima + media area compared to sham controls. There was a significant decrease in the staining of smooth muscle cells, fibroblasts, macrophages, HIF-1α, proliferation, and apoptosis and an increase in CD31-(+) cells. CONCLUSIONS A clinically relevant model of PTA of venous stenosis in mice was created. PTA-treated vessels had increased lumen vessel area and WSS. The alterations in tissue markers of vascular remodeling, tissue hypoxia, proliferation, and cell death may be implications for future design of drug and device development.
Collapse
|
29
|
Stem Cells in Alzheimer’s Disease: Current Standing and Future Challenges. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2018; 1079:93-102. [DOI: 10.1007/5584_2018_214] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
30
|
Li M, Luo X, Lv X, Liu V, Zhao G, Zhang X, Cao W, Wang R, Wang W. In vivo human adipose-derived mesenchymal stem cell tracking after intra-articular delivery in a rat osteoarthritis model. Stem Cell Res Ther 2016; 7:160. [PMID: 27832815 PMCID: PMC5103374 DOI: 10.1186/s13287-016-0420-2] [Citation(s) in RCA: 73] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2016] [Accepted: 10/11/2016] [Indexed: 12/11/2022] Open
Abstract
Background Human adipose-derived mesenchymal stem cells (haMSCs) have shown efficacy in treating osteoarthritis (OA) both preclinically and clinically via intra-articular (IA) injection. However, understanding the mode of action of the cell therapy has been limited by cell tracking capability and correlation between the pharmacokinetics of the injected cells and the intended pharmacodynamics effect. This study aims to explore methodology and to understand in vivo biodistribution of clinical-grade haMSCs labeled with fluorescent dye and injected into an immunocompetent OA rat model. Methods haMSCs labeled with fluorescent dye were investigated for their proliferation and differentiation capabilities. Labeled cells were used to establish detection threshold of a noninvasive biofluorescent imaging system before the cells (2.5 × 106) were injected into a conventional rat OA model induced by medial meniscectomy for 8 weeks. We attempted to reveal the existence of labeled cells in vivo by imaging and a molecular biomarker approach, and to correlate with the in vivo efficacy and physical presence over a follow-up period up to 10 weeks. Results In vitro proliferation and differentiation of haMSCs were not affected by the labeling of DiD dye. Detection thresholds of the labeled cells in vitro and in vivo were determined to be 104 and 105 cells, respectively. When 2.5 × 106 haMSCs were injected into the joints of a rat OA model, fluorescent signals (or >105 cells) lasted for about 10 weeks in the surgical knee joint at the same time as efficacy was observed. Signals in nonsurgical rats only lasted for 4 weeks. The human MSCs were shown to engraft to the rat joint tissues and were proliferative. Human FOXP2 gene was only detected in the knee joint tissue, suggesting limited biodistribution locally to the joints. Conclusions The current study represents the first attempt to correlate cell therapy efficacy on OA with the physical presence of the injected haMSCs in the OA model, and demonstrates that human adipose-derived mesenchymal stem cells persisted for 10 weeks locally in the rat joint, coinciding with the efficacy observed. It is postulated that persistence and/or proliferation of the haMSCs in the joint is required in order to exert their functions on promoting joint regeneration and/or cartilage protection, further supporting the safety and feasibility of IA injection of MSCs for the treatment of OA patients.
Collapse
Affiliation(s)
- Meng Li
- Cellular Biomedicine Group, 333 Guiping Road, Shanghai, 200233, China.,Cellular Biomedicine Group, 19925 Stevens Creek Blvd, Suite 100, Cupertino, CA, 95014, USA
| | - Xuan Luo
- Cellular Biomedicine Group, 333 Guiping Road, Shanghai, 200233, China.,Cellular Biomedicine Group, 19925 Stevens Creek Blvd, Suite 100, Cupertino, CA, 95014, USA
| | - Xiaoteng Lv
- Cellular Biomedicine Group, 333 Guiping Road, Shanghai, 200233, China.,Cellular Biomedicine Group, 19925 Stevens Creek Blvd, Suite 100, Cupertino, CA, 95014, USA
| | - Victor Liu
- Cellular Biomedicine Group, 333 Guiping Road, Shanghai, 200233, China.,Cellular Biomedicine Group, 19925 Stevens Creek Blvd, Suite 100, Cupertino, CA, 95014, USA
| | - Guangyu Zhao
- Cellular Biomedicine Group, 333 Guiping Road, Shanghai, 200233, China.,Cellular Biomedicine Group, 19925 Stevens Creek Blvd, Suite 100, Cupertino, CA, 95014, USA
| | - Xue Zhang
- Plastic Surgery Hospital (Institute), Peking Union Medical College, Chinese Academy of Medical Sciences, 33 Badachu Road, Shijingshan District, Beijing, 100144, China
| | - Wei Cao
- Cellular Biomedicine Group, 333 Guiping Road, Shanghai, 200233, China.,Cellular Biomedicine Group, 19925 Stevens Creek Blvd, Suite 100, Cupertino, CA, 95014, USA
| | - Richard Wang
- Cellular Biomedicine Group, 333 Guiping Road, Shanghai, 200233, China.,Cellular Biomedicine Group, 19925 Stevens Creek Blvd, Suite 100, Cupertino, CA, 95014, USA
| | - Wen Wang
- Cellular Biomedicine Group, 333 Guiping Road, Shanghai, 200233, China. .,Cellular Biomedicine Group, 19925 Stevens Creek Blvd, Suite 100, Cupertino, CA, 95014, USA.
| |
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
|
Lee T, Misra S. New Insights into Dialysis Vascular Access: Molecular Targets in Arteriovenous Fistula and Arteriovenous Graft Failure and Their Potential to Improve Vascular Access Outcomes. Clin J Am Soc Nephrol 2016; 11:1504-1512. [PMID: 27401527 PMCID: PMC4974876 DOI: 10.2215/cjn.02030216] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Vascular access dysfunction remains a major cause of morbidity and mortality in hemodialysis patients. At present there are few effective therapies for this clinical problem. The poor understanding of the pathobiology that leads to arteriovenous fistula (AVF) and graft (AVG) dysfunction remains a critical barrier to development of novel and effective therapies. However, in recent years we have made substantial progress in our understanding of the mechanisms of vascular access dysfunction. This article presents recent advances and new insights into the pathobiology of AVF and AVG dysfunction and highlights potential therapeutic targets to improve vascular access outcomes.
Collapse
Affiliation(s)
- Timmy Lee
- Department of Medicine and Division of Nephrology, University of Alabama at Birmingham, Birmingham, Alabama
- Veterans Affairs Medical Center, Birmingham, Alabama; and
| | - Sanjay Misra
- Vascular and Interventional Radiology Translational Laboratory, Department of Radiology, Mayo Clinic, Rochester, Minnesota
| |
Collapse
|
33
|
Salem H, Rocha NP, Colpo GD, Teixeira AL. Moving from the Dish to the Clinical Practice: A Decade of Lessons and Perspectives from the Pre-Clinical and Clinical Stem Cell Studies for Alzheimer’s Disease. J Alzheimers Dis 2016; 53:1209-30. [DOI: 10.3233/jad-160250] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Affiliation(s)
- Haitham Salem
- Department of Psychiatry and Behavioral Sciences, Neuropsychiatry Program, McGovern Medical School, The University of Texas Health Science Center, Houston, TX, USA
- Regenerative Medicine Program, University of Lübeck, Schleswig-Holstein, Germany
| | - Natalia Pessoa Rocha
- Department of Psychiatry and Behavioral Sciences, Neuropsychiatry Program, McGovern Medical School, The University of Texas Health Science Center, Houston, TX, USA
| | - Gabriela Delevati Colpo
- Department of Psychiatry and Behavioral Sciences, Neuropsychiatry Program, McGovern Medical School, The University of Texas Health Science Center, Houston, TX, USA
| | - Antonio Lucio Teixeira
- Department of Psychiatry and Behavioral Sciences, Neuropsychiatry Program, McGovern Medical School, The University of Texas Health Science Center, Houston, TX, USA
| |
Collapse
|
34
|
Janardhanan R, Yang B, Kilari S, Leof EB, Mukhopadhyay D, Misra S. The Role of Repeat Administration of Adventitial Delivery of Lentivirus-shRNA-Vegf-A in Arteriovenous Fistula to Prevent Venous Stenosis Formation. J Vasc Interv Radiol 2016; 27:576-83. [PMID: 26948326 DOI: 10.1016/j.jvir.2015.12.751] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2015] [Revised: 12/21/2015] [Accepted: 12/22/2015] [Indexed: 12/13/2022] Open
Abstract
PURPOSE To determine if a second dose of a lentivirus mediated small hairpin RNA that inhibits Vegf-A gene expression (LV-shRNA-Vegf-A) can improve lumen vessel area (LVA) of the outflow vein of an arteriovenous fistula (AVF) and decrease venous neointimal hyperplasia. MATERIALS AND METHODS Chronic kidney disease was created in C57BL/6 mice; 28 days later, an AVF was created by connecting the right carotid artery to the ipsilateral jugular vein. Immediately after AVF creation, 5 × 10(6) plaque-forming units of LV-shRNA-Vegf-A or control shRNA was administered to the adventitia of the outflow vein, and a second dose of the same treatment was administered 14 days later. Animals were sacrificed at 21 days, 28 days, and 42 days after AVF creation for reverse transcription polymerase chain reaction and histomorphometric analyses. RESULTS By day 21, there was a 125% increase in the average LVA (day 21, P = .11), with a decrease in cell proliferation (day 21, P = .0079; day 28, P = .28; day 42, P = .5), decrease in α-smooth muscle cell actin staining (day 21, P < .0001; day 28, P < .05; day 42, P = .59), and decrease in hypoxic stress (day 21, P < .001; day 28, P = .28; day 42, P = .46) in LV versus control shRNA vessels. CONCLUSIONS A second dose of LV-shRNA-Vegf-A administration results in a moderate improvement in LVA at day 21.
Collapse
Affiliation(s)
- Rajiv Janardhanan
- Amity Institute of Public Health, Amity University Uttar Pradesh, Noida, Uttar Pradesh, India; Vascular and Interventional Radiology Translational Laboratory Mayo Clinic, 200 First Street SW, Rochester, MN 55905
| | - Binxia Yang
- Amity Institute of Public Health, Amity University Uttar Pradesh, Noida, Uttar Pradesh, India; Vascular and Interventional Radiology Translational Laboratory Mayo Clinic, 200 First Street SW, Rochester, MN 55905
| | - Sreenivasulu Kilari
- Amity Institute of Public Health, Amity University Uttar Pradesh, Noida, Uttar Pradesh, India; Vascular and Interventional Radiology Translational Laboratory Mayo Clinic, 200 First Street SW, Rochester, MN 55905
| | - Edward B Leof
- Department of Radiology, and Department of Biochemistry and Molecular Biology, Mayo Clinic, 200 First Street SW, Rochester, MN 55905
| | - Debabrata Mukhopadhyay
- Department of Radiology, and Department of Biochemistry and Molecular Biology, Mayo Clinic, 200 First Street SW, Rochester, MN 55905
| | - Sanjay Misra
- Amity Institute of Public Health, Amity University Uttar Pradesh, Noida, Uttar Pradesh, India; Vascular and Interventional Radiology Translational Laboratory Mayo Clinic, 200 First Street SW, Rochester, MN 55905; Department of Radiology, and Department of Biochemistry and Molecular Biology, Mayo Clinic, 200 First Street SW, Rochester, MN 55905.
| |
Collapse
|