1
|
Robillard S, Trân K, Lachance MS, Brazeau T, Boisvert E, Lizotte F, Auger-Messier M, Boudreault PL, Marsault É, Geraldes P. Apelin prevents diabetes-induced poor collateral vessel formation and blood flow reperfusion in ischemic limb. Front Cardiovasc Med 2023; 10:1191891. [PMID: 37636297 PMCID: PMC10450936 DOI: 10.3389/fcvm.2023.1191891] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Accepted: 08/01/2023] [Indexed: 08/29/2023] Open
Abstract
Introduction Peripheral arterial disease (PAD) is a major risk factor for lower-extremity amputation in diabetic patients. Unfortunately, previous clinical studies investigating therapeutic angiogenesis using the vascular endothelial growth factor (VEGF) have shown disappointing results in diabetic patients, which evokes the necessity for novel therapeutic agents. The apelinergic system (APJ receptor/apelin) is highly upregulated under hypoxic condition and acts as an activator of angiogenesis. Apelin treatment improves revascularization in nondiabetic models of ischemia, however, its role on angiogenesis in diabetic conditions remains poorly investigated. This study explored the impact of Pyr-apelin-13 in endothelial cell function and diabetic mouse model of hindlimb ischemia. Methods Nondiabetic and diabetic mice underwent femoral artery ligation to induce limb ischemia. Diabetic mice were implanted subcutaneously with osmotic pumps delivering Pyr-apelin-13 for 28 days. Blood flow reperfusion was measured for 4 weeks post-surgery and exercise willingness was assessed with voluntary wheels. In vitro, bovine aortic endothelial cells (BAECs) were exposed to normal (NG) or high glucose (HG) levels and hypoxia. Cell migration, proliferation and tube formation assays were performed following either VEGF or Pyr-apelin-13 stimulation. Results and Discussion Following limb ischemia, blood flow reperfusion, functional recovery of the limb and vascular density were improved in diabetic mice receiving Pyr-apelin-13 compared to untreated diabetic mice. In cultured BAECs, exposure to HG concentrations and hypoxia reduced VEGF proangiogenic actions, whereas apelin proangiogenic effects remained unaltered. Pyr-apelin-13 induced its proangiogenic actions through Akt/AMPK/eNOS and RhoA/ROCK signaling pathways under both NG or HG concentrations and hypoxia exposure. Our results identified the apelinergic system as a potential therapeutic target for angiogenic therapy in diabetic patients with PAD.
Collapse
Affiliation(s)
- Stéphanie Robillard
- Research Center of the Centre Hospitalier Universitaire de Sherbrooke, Sherbrooke, QC, Canada
| | - Kien Trân
- Department of Pharmacology and Physiology, Université de Sherbrooke, Sherbrooke, QC, Canada
| | - Marie-Sophie Lachance
- Research Center of the Centre Hospitalier Universitaire de Sherbrooke, Sherbrooke, QC, Canada
| | - Tristan Brazeau
- Research Center of the Centre Hospitalier Universitaire de Sherbrooke, Sherbrooke, QC, Canada
| | - Elizabeth Boisvert
- Research Center of the Centre Hospitalier Universitaire de Sherbrooke, Sherbrooke, QC, Canada
| | - Farah Lizotte
- Research Center of the Centre Hospitalier Universitaire de Sherbrooke, Sherbrooke, QC, Canada
| | - Mannix Auger-Messier
- Division of Cardiology, Department of Medicine, Université de Sherbrooke, Sherbrooke, QC, Canada
| | - Pierre-Luc Boudreault
- Department of Pharmacology and Physiology, Université de Sherbrooke, Sherbrooke, QC, Canada
| | - Éric Marsault
- Department of Pharmacology and Physiology, Université de Sherbrooke, Sherbrooke, QC, Canada
| | - Pedro Geraldes
- Division of Endocrinology, Department of Medicine, Université de Sherbrooke, Sherbrooke, QC, Canada
| |
Collapse
|
2
|
Boiko AS, Mednova IA, Kornetova EG, Bokhan NA, Ivanova SA. Serum Growth Factors in Schizophrenia Patients. Curr Issues Mol Biol 2023; 45:3291-3301. [PMID: 37185739 PMCID: PMC10136551 DOI: 10.3390/cimb45040215] [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: 02/28/2023] [Revised: 04/01/2023] [Accepted: 04/03/2023] [Indexed: 05/17/2023] Open
Abstract
Some hypotheses include schizophrenia as a neurodevelopmental disorder, which indicates a special role in growth factors and neuroglia in the development of schizophrenia symptoms. Growth factors are cytokine molecules that play an important role in the regulation of tissue nucleation, cell development, survival, and migration of all tissues in organisms, including the brain and nervous system. The aim of the study was to determine the serum concentration of six growth factors (EGF, VEGF, FGF-2, TGF-α, PDGF-AA, PDGF-AB/BB) in schizophrenia patients and to identify the correlations with clinical characteristics. After signing an informed consent form, 236 schizophrenia patients (F20 according to the ICD-10) and 102 healthy people were recruited in the study. In patients with schizophrenia, we observed a significant elevation in the TGF-α and PDGF-AA serum levels. The duration of schizophrenia was significantly positively correlated with the FGF-2 level. The PANSS total score had a positive correlation with the FGF-2 level and a negative correlation with the TGF-α level. Our results and literature indicate the involvement of growth factors in the mechanisms of development of schizophrenia. Combined biomarker screening seems to be necessary to improve diagnosis and clinical follow-up of patients with severe mental illnesses.
Collapse
Affiliation(s)
- Anastasiia S Boiko
- Mental Health Research Institute, Tomsk National Research Medical Center of the Russian Academy of Sciences, 634014 Tomsk, Russia
| | - Irina A Mednova
- Mental Health Research Institute, Tomsk National Research Medical Center of the Russian Academy of Sciences, 634014 Tomsk, Russia
| | - Elena G Kornetova
- Mental Health Research Institute, Tomsk National Research Medical Center of the Russian Academy of Sciences, 634014 Tomsk, Russia
- University Hospital, Siberian State Medical University, 634050 Tomsk, Russia
| | - Nikolay A Bokhan
- Mental Health Research Institute, Tomsk National Research Medical Center of the Russian Academy of Sciences, 634014 Tomsk, Russia
- Department of Psychiatry, Addictology and Psychotherapy, Siberian State Medical University, 634050 Tomsk, Russia
| | - Svetlana A Ivanova
- Mental Health Research Institute, Tomsk National Research Medical Center of the Russian Academy of Sciences, 634014 Tomsk, Russia
- University Hospital, Siberian State Medical University, 634050 Tomsk, Russia
| |
Collapse
|
3
|
Abbas H, Olivere LA, Padgett ME, Schmidt CA, Gilmore BF, McCord TJ, Southerland KW, McClung JM, Kontos CD. Muscle progenitor cells are required for skeletal muscle regeneration and prevention of adipogenesis after limb ischemia. Front Cardiovasc Med 2023; 10:1118738. [PMID: 36937923 PMCID: PMC10017542 DOI: 10.3389/fcvm.2023.1118738] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Accepted: 02/08/2023] [Indexed: 03/06/2023] Open
Abstract
Skeletal muscle injury in peripheral artery disease (PAD) has been attributed to vascular insufficiency, however evidence has demonstrated that muscle cell responses play a role in determining outcomes in limb ischemia. Here, we demonstrate that genetic ablation of Pax7+ muscle progenitor cells (MPCs) in a model of hindlimb ischemia (HLI) inhibited muscle regeneration following ischemic injury, despite a lack of morphological or physiological changes in resting muscle. Compared to control mice (Pax7WT), the ischemic limb of Pax7-deficient mice (Pax7Δ) was unable to generate significant force 7 or 28 days after HLI. A significant increase in adipose was observed in the ischemic limb 28 days after HLI in Pax7Δ mice, which replaced functional muscle. Adipogenesis in Pax7Δ mice corresponded with a significant increase in PDGFRα+ fibro/adipogenic progenitors (FAPs). Inhibition of FAPs with batimastat decreased muscle adipose but increased fibrosis. In vitro, Pax7Δ MPCs failed to form myotubes but displayed increased adipogenesis. Skeletal muscle from patients with critical limb threatening ischemia displayed increased adipose in more ischemic regions of muscle, which corresponded with fewer satellite cells. Collectively, these data demonstrate that Pax7+ MPCs are required for muscle regeneration after ischemia and suggest that muscle regeneration may be an important therapeutic target in PAD.
Collapse
Affiliation(s)
- Hasan Abbas
- Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, NC, United States
- Duke-NUS Medical School, Singapore, Singapore
- Department of Medicine, Division of Cardiology, Duke University Medical Center, Durham, NC, United States
| | | | - Michael E. Padgett
- Department of Medicine, Division of Cardiology, Duke University Medical Center, Durham, NC, United States
| | - Cameron A. Schmidt
- Department of Physiology, Brody School of Medicine, East Carolina University, Greenville, NC, United States
- Brody School of Medicine, East Carolina Diabetes and Obesity Institute, East Carolina University, Greenville, NC, United States
| | - Brian F. Gilmore
- Department of Surgery, Duke University Medical Center, Durham, NC, United States
| | - Timothy J. McCord
- Department of Cell Biology, Duke University School of Medicine, Durham, NC, United States
| | - Kevin W. Southerland
- Department of Surgery, Duke University Medical Center, Durham, NC, United States
| | - Joseph M. McClung
- Department of Physiology, Brody School of Medicine, East Carolina University, Greenville, NC, United States
- Brody School of Medicine, East Carolina Diabetes and Obesity Institute, East Carolina University, Greenville, NC, United States
- Brody School of Medicine, East Carolina Heart Institute, East Carolina University, Greenville, NC, United States
| | - Christopher D. Kontos
- Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, NC, United States
- Department of Medicine, Division of Cardiology, Duke University Medical Center, Durham, NC, United States
- Duke University School of Medicine, Durham, NC, United States
| |
Collapse
|
4
|
Rustagi Y, Abouhashem AS, Verma P, Verma SS, Hernandez E, Liu S, Kumar M, Guda PR, Srivastava R, Mohanty SK, Kacar S, Mahajan S, Wanczyk KE, Khanna S, Murphy MP, Gordillo GM, Roy S, Wan J, Sen CK, Singh K. Endothelial Phospholipase Cγ2 Improves Outcomes of Diabetic Ischemic Limb Rescue Following VEGF Therapy. Diabetes 2022; 71:1149-1165. [PMID: 35192691 PMCID: PMC9044136 DOI: 10.2337/db21-0830] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Accepted: 02/15/2022] [Indexed: 11/13/2022]
Abstract
Therapeutic vascular endothelial growth factor (VEGF) replenishment has met with limited success for the management of critical limb-threatening ischemia. To improve outcomes of VEGF therapy, we applied single-cell RNA sequencing (scRNA-seq) technology to study the endothelial cells of the human diabetic skin. Single-cell suspensions were generated from the human skin followed by cDNA preparation using the Chromium Next GEM Single-cell 3' Kit v3.1. Using appropriate quality control measures, 36,487 cells were chosen for downstream analysis. scRNA-seq studies identified that although VEGF signaling was not significantly altered in diabetic versus nondiabetic skin, phospholipase Cγ2 (PLCγ2) was downregulated. The significance of PLCγ2 in VEGF-mediated increase in endothelial cell metabolism and function was assessed in cultured human microvascular endothelial cells. In these cells, VEGF enhanced mitochondrial function, as indicated by elevation in oxygen consumption rate and extracellular acidification rate. The VEGF-dependent increase in cell metabolism was blunted in response to PLCγ2 inhibition. Follow-up rescue studies therefore focused on understanding the significance of VEGF therapy in presence or absence of endothelial PLCγ2 in type 1 (streptozotocin-injected) and type 2 (db/db) diabetic ischemic tissue. Nonviral topical tissue nanotransfection technology (TNT) delivery of CDH5 promoter-driven PLCγ2 open reading frame promoted the rescue of hindlimb ischemia in diabetic mice. Improvement of blood flow was also associated with higher abundance of VWF+/CD31+ and VWF+/SMA+ immunohistochemical staining. TNT-based gene delivery was not associated with tissue edema, a commonly noted complication associated with proangiogenic gene therapies. Taken together, our study demonstrates that TNT-mediated delivery of endothelial PLCγ2, as part of combination gene therapy, is effective in diabetic ischemic limb rescue.
Collapse
Affiliation(s)
- Yashika Rustagi
- Indiana Center for Regenerative Medicine and Engineering, Indiana University Health Comprehensive Wound Center, Indiana University School of Medicine, Indianapolis, IN
| | - Ahmed S. Abouhashem
- Indiana Center for Regenerative Medicine and Engineering, Indiana University Health Comprehensive Wound Center, Indiana University School of Medicine, Indianapolis, IN
- Sharkia Clinical Research Department, Ministry of Health and Population, Cairo, Egypt
| | - Priyanka Verma
- Indiana Center for Regenerative Medicine and Engineering, Indiana University Health Comprehensive Wound Center, Indiana University School of Medicine, Indianapolis, IN
| | - Sumit S. Verma
- Indiana Center for Regenerative Medicine and Engineering, Indiana University Health Comprehensive Wound Center, Indiana University School of Medicine, Indianapolis, IN
| | - Edward Hernandez
- Indiana Center for Regenerative Medicine and Engineering, Indiana University Health Comprehensive Wound Center, Indiana University School of Medicine, Indianapolis, IN
| | - Sheng Liu
- Center for Computational Biology and Bioinformatics, Indiana University School of Medicine, Indianapolis, IN
| | - Manishekhar Kumar
- Indiana Center for Regenerative Medicine and Engineering, Indiana University Health Comprehensive Wound Center, Indiana University School of Medicine, Indianapolis, IN
| | - Poornachander R. Guda
- Indiana Center for Regenerative Medicine and Engineering, Indiana University Health Comprehensive Wound Center, Indiana University School of Medicine, Indianapolis, IN
| | - Rajneesh Srivastava
- Indiana Center for Regenerative Medicine and Engineering, Indiana University Health Comprehensive Wound Center, Indiana University School of Medicine, Indianapolis, IN
| | - Sujit K. Mohanty
- Indiana Center for Regenerative Medicine and Engineering, Indiana University Health Comprehensive Wound Center, Indiana University School of Medicine, Indianapolis, IN
| | - Sedat Kacar
- Indiana Center for Regenerative Medicine and Engineering, Indiana University Health Comprehensive Wound Center, Indiana University School of Medicine, Indianapolis, IN
| | - Sanskruti Mahajan
- Indiana Center for Regenerative Medicine and Engineering, Indiana University Health Comprehensive Wound Center, Indiana University School of Medicine, Indianapolis, IN
| | - Kristen E. Wanczyk
- Indiana Center for Regenerative Medicine and Engineering, Indiana University Health Comprehensive Wound Center, Indiana University School of Medicine, Indianapolis, IN
| | - Savita Khanna
- Indiana Center for Regenerative Medicine and Engineering, Indiana University Health Comprehensive Wound Center, Indiana University School of Medicine, Indianapolis, IN
| | - Michael P. Murphy
- Indiana Center for Regenerative Medicine and Engineering, Indiana University Health Comprehensive Wound Center, Indiana University School of Medicine, Indianapolis, IN
| | - Gayle M. Gordillo
- Indiana Center for Regenerative Medicine and Engineering, Indiana University Health Comprehensive Wound Center, Indiana University School of Medicine, Indianapolis, IN
| | - Sashwati Roy
- Indiana Center for Regenerative Medicine and Engineering, Indiana University Health Comprehensive Wound Center, Indiana University School of Medicine, Indianapolis, IN
| | - Jun Wan
- Center for Computational Biology and Bioinformatics, Indiana University School of Medicine, Indianapolis, IN
| | - Chandan K. Sen
- Indiana Center for Regenerative Medicine and Engineering, Indiana University Health Comprehensive Wound Center, Indiana University School of Medicine, Indianapolis, IN
| | - Kanhaiya Singh
- Indiana Center for Regenerative Medicine and Engineering, Indiana University Health Comprehensive Wound Center, Indiana University School of Medicine, Indianapolis, IN
| |
Collapse
|
5
|
Pro-Calcific Environment Impairs Ischaemia-Driven Angiogenesis. Int J Mol Sci 2022; 23:ijms23063363. [PMID: 35328786 PMCID: PMC8954938 DOI: 10.3390/ijms23063363] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Revised: 03/14/2022] [Accepted: 03/17/2022] [Indexed: 02/04/2023] Open
Abstract
Peripheral arterial disease (PAD) is characterised by accelerated arterial calcification and impairment in angiogenesis. Studies implicate vascular calcification as a contributor to PAD, but the mechanisms remain unclear. We aimed to determine the effect of calcification on ischaemia-driven angiogenesis. Human coronary artery endothelial cells (ECs) were treated with calcification medium (CM: CaCl2 2.7 mM, Na2PO4 2.0 mM) for 24 h and exposed to normoxia (5% CO2) or hypoxia (1.2% O2; 5% CO2 balanced with N2). In normoxia, CM significantly inhibited tubule formation and migration and upregulated calcification markers of ALP, BMP2, and Runx2. CM elevated levels of calcification-protective gene OPG, demonstrating a compensatory mechanism by ECs. CM failed to induce pro-angiogenic regulators VEGFA and HIF-1α in hypoxia and further suppressed the phosphorylation of endothelial nitric oxide synthase (eNOS) that is essential for vascular function. In vivo, osteoprotegerin-deficient mice (OPG−/−), a calcification model, were subjected to hind-limb ischaemia (HLI) surgery. OPG−/− mice displayed elevated serum alkaline phosphatase (ALP) activity compared to wild-type controls. OPG−/− mice experienced striking reductions in blood-flow reperfusion in both 8-week-old and 6-month-old mice post-HLI. This coincided with significant impairment in tissue ischaemia and reduced limb function as assessed by clinical scoring (Tarlov). This study demonstrated for the first time that a pro-calcific environment is detrimental to ischaemia-driven angiogenesis. The degree of calcification in patients with PAD can often be a limiting factor with the use of standard therapies. These highly novel findings require further studies for full elucidation of the mechanisms involved and have implications for the development of therapies to suppress calcification in PAD.
Collapse
|
6
|
Santos SS, Gonzaga RV, Scarim CB, Giarolla J, Primi MC, Chin CM, Ferreira EI. Drug/Lead Compound Hydroxymethylation as a Simple Approach to Enhance Pharmacodynamic and Pharmacokinetic Properties. Front Chem 2022; 9:734983. [PMID: 35237565 PMCID: PMC8883432 DOI: 10.3389/fchem.2021.734983] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Accepted: 10/20/2021] [Indexed: 11/17/2022] Open
Abstract
Hydroxymethylation is a simple chemical reaction, in which the introduction of the hydroxymethyl group can lead to physical–chemical property changes and offer several therapeutic advantages, contributing to the improved biological activity of drugs. There are many examples in the literature of the pharmaceutical, pharmacokinetic, and pharmacodynamic benefits, which the hydroxymethyl group can confer to drugs, prodrugs, drug metabolites, and other therapeutic compounds. It is worth noting that this group can enhance the drug’s interaction with the active site, and it can be employed as an intermediary in synthesizing other therapeutic agents. In addition, the hydroxymethyl derivative can result in more active compounds than the parent drug as well as increase the water solubility of poorly soluble drugs. Taking this into consideration, this review aims to discuss different applications of hydroxymethyl derived from biological agents and its influence on the pharmacological effects of drugs, prodrugs, active metabolites, and compounds of natural origin. Finally, we report a successful compound synthesized by our research group and used for the treatment of neglected diseases, which is created from the hydroxymethylation of its parent drug.
Collapse
Affiliation(s)
- Soraya S. Santos
- Laboratório de Planejamento e Síntese de Quimioterápicos Potencialmente Ativos Em Doenças Negligenciadas (LAPEN), Departamento de Farmácia, Faculdade de Ciências Farmacêuticas, Universidade de São Paulo – USP, São Paulo, Brazil
| | - Rodrigo V. Gonzaga
- Laboratório de Planejamento e Síntese de Quimioterápicos Potencialmente Ativos Em Doenças Negligenciadas (LAPEN), Departamento de Farmácia, Faculdade de Ciências Farmacêuticas, Universidade de São Paulo – USP, São Paulo, Brazil
| | - Cauê B. Scarim
- Laboratório de Pesquisa e Desenvolvimento de Fármacos (LAPDESF), Departamento de Fármacos e Medicamentos, Faculdade de Ciências Farmacêuticas, Universidade Estadual de São Paulo “Júlio de Mesquita Filho” (UNESP), Araraquara, Brazil
| | - Jeanine Giarolla
- Laboratório de Planejamento e Síntese de Quimioterápicos Potencialmente Ativos Em Doenças Negligenciadas (LAPEN), Departamento de Farmácia, Faculdade de Ciências Farmacêuticas, Universidade de São Paulo – USP, São Paulo, Brazil
| | | | - Chung M. Chin
- Laboratório de Pesquisa e Desenvolvimento de Fármacos (LAPDESF), Departamento de Fármacos e Medicamentos, Faculdade de Ciências Farmacêuticas, Universidade Estadual de São Paulo “Júlio de Mesquita Filho” (UNESP), Araraquara, Brazil
- Centro de Pesquisa Avançada Em Medicina (CEPAM), Faculdade de Medicina, União Das Faculdades Dos Grande Lagos (UNILAGO), São José Do Rio Preto, Brazil
| | - Elizabeth I. Ferreira
- Laboratório de Planejamento e Síntese de Quimioterápicos Potencialmente Ativos Em Doenças Negligenciadas (LAPEN), Departamento de Farmácia, Faculdade de Ciências Farmacêuticas, Universidade de São Paulo – USP, São Paulo, Brazil
- *Correspondence: Elizabeth I. Ferreira,
| |
Collapse
|
7
|
Hansen L, Joseph G, Valdivia A, Taylor WR. Satellite Cell Expression of RAGE (Receptor for Advanced Glycation end Products) Is Important for Collateral Vessel Formation. J Am Heart Assoc 2021; 10:e022127. [PMID: 34689598 PMCID: PMC8751830 DOI: 10.1161/jaha.120.022127] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Background The growth and remodeling of vascular networks is an important component of the prognosis for patients with peripheral artery disease. One protein that has been previously implicated to play a role in this process is RAGE (receptor for advanced glycation end products). This study sought to determine the cellular source of RAGE in the ischemic hind limb and the role of RAGE signaling in this cell type. Methods and Results Using a hind limb ischemia model of vascular growth, this study found skeletal muscle satellite cells to be a novel major cellular source of RAGE in ischemic tissue by both staining and cellular sorting. Although wild-type satellite cells increased tumor necrosis factor-α and monocyte chemoattractant protein-1 production in response to ischemia in vivo and a RAGE ligand in vitro, satellite cells from RAGE knockout mice lacked the increase in cytokine production both in vivo in response to ischemia and in vitro after stimuli with the RAGE ligand high-mobility group box 1. Furthermore, encapsulated wild-type satellite cells improved perfusion after hind limb ischemia surgery by both perfusion staining and vessel quantification, but RAGE knockout satellite cells provided no improvement over empty capsules. Conclusions Thus, RAGE expression and signaling in satellite cells is crucial for their response to stimuli and angiogenic and arteriogenic functions.
Collapse
Affiliation(s)
- Laura Hansen
- Division of Cardiology Department of Medicine Emory University Atlanta GA.,Division of Cardiology Atlanta Veterans Affairs Medical Center Decatur GA
| | - Giji Joseph
- Division of Cardiology Department of Medicine Emory University Atlanta GA
| | - Alejandra Valdivia
- Division of Cardiology Department of Medicine Emory University Atlanta GA
| | - W Robert Taylor
- Division of Cardiology Department of Medicine Emory University Atlanta GA.,Division of Cardiology Atlanta Veterans Affairs Medical Center Decatur GA.,The Wallace H. Coulter Department of Biomedical Engineering Georgia Institute of Technology and Emory University Atlanta GA
| |
Collapse
|
8
|
Solly EL, Psaltis PJ, Bursill CA, Tan JTM. The Role of miR-181c in Mechanisms of Diabetes-Impaired Angiogenesis: An Emerging Therapeutic Target for Diabetic Vascular Complications. Front Pharmacol 2021; 12:718679. [PMID: 34483928 PMCID: PMC8414254 DOI: 10.3389/fphar.2021.718679] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Accepted: 08/06/2021] [Indexed: 12/13/2022] Open
Abstract
Diabetes mellitus is estimated to affect up to 700 million people by the year 2045, contributing to an immense health and economic burden. People living with diabetes have a higher risk of developing numerous debilitating vascular complications, leading to an increased need for medical care, a reduced quality of life and increased risk of early death. Current treatments are not satisfactory for many patients who suffer from impaired angiogenesis in response to ischaemia, increasing their risk of ischaemic cardiovascular conditions. These vascular pathologies are characterised by endothelial dysfunction and abnormal angiogenesis, amongst a host of impaired signaling pathways. Therapeutic stimulation of angiogenesis holds promise for the treatment of diabetic vascular complications that stem from impaired ischaemic responses. However, despite significant effort and research, there are no established therapies that directly stimulate angiogenesis to improve ischaemic complications such as ischaemic heart disease and peripheral artery disease, highlighting the immense unmet need. However, despite significant effort and research, there are no established therapies that directly stimulate angiogenesis in a clinical setting, highlighting the immense unmet need. MicroRNAs (miRNAs) are emerging as powerful targets for multifaceted diseases including diabetes and cardiovascular disease. This review highlights the potential role of microRNAs as therapeutic targets for rescuing diabetes-impaired angiogenesis, with a specific focus on miR-181c, which we have previously identified as an important angiogenic regulator. Here we summarise the pathways currently known to be regulated by miR-181c, which include the classical angiogenesis pathways that are dysregulated in diabetes, mitochondrial function and axonal guidance, and describe how these relate both directly and indirectly to angiogenesis. The pleiotropic actions of miR-181c across multiple key angiogenic signaling pathways and critical cellular processes highlight its therapeutic potential as a novel target for treating diabetic vascular complications.
Collapse
Affiliation(s)
- Emma L Solly
- Vascular Research Centre, Lifelong Health Theme, South Australian Health and Medical Research Institute, Adelaide, SA, Australia.,Adelaide Medical School, The University of Adelaide, Adelaide, SA, Australia
| | - Peter J Psaltis
- Vascular Research Centre, Lifelong Health Theme, South Australian Health and Medical Research Institute, Adelaide, SA, Australia.,Adelaide Medical School, The University of Adelaide, Adelaide, SA, Australia
| | - Christina A Bursill
- Vascular Research Centre, Lifelong Health Theme, South Australian Health and Medical Research Institute, Adelaide, SA, Australia.,Adelaide Medical School, The University of Adelaide, Adelaide, SA, Australia.,ARC Centre of Excellence for Nanoscale BioPhotonics, The University of Adelaide, Adelaide, SA, Australia
| | - Joanne T M Tan
- Vascular Research Centre, Lifelong Health Theme, South Australian Health and Medical Research Institute, Adelaide, SA, Australia.,Adelaide Medical School, The University of Adelaide, Adelaide, SA, Australia
| |
Collapse
|
9
|
Functional Recovery after Intramyocardial Injection of Adipose-Derived Stromal Cells Assessed by Cardiac Magnetic Resonance Imaging. Stem Cells Int 2021; 2021:5556800. [PMID: 33976700 PMCID: PMC8087467 DOI: 10.1155/2021/5556800] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2021] [Revised: 04/02/2021] [Accepted: 04/15/2021] [Indexed: 12/22/2022] Open
Abstract
Aims A major clinical concern is the continuous increase in the number of patients diagnosed with advanced coronary artery disease, ischemic heart failure, and refractory angina, and one of the most promising treatment options for these conditions is stem cell-based therapy. The aim of this study was to assess the functional improvement following intramyocardial injection of adipose-derived stromal cells, using cardiac magnetic resonance. Methods and Results Thirteen patients with ischemic heart failure, reduced left ventricular ejection fraction, refractory angina, and who have been disqualified from any form of direct revascularization were enrolled in the study with transthoracic autologous adipose-derived stromal cell implantation. All patients underwent cardiac magnetic resonance prior to the procedure and after 12 months of follow-up. A significant increase in stroke volume (83.1 ± 8.5 mL vs 93.8 ± 13.8 mL, p = 0.025) and stroke volume index (43.3 ± 7.6 mL/m2 vs 48.7 ± 9.1 mL/m2, p = 0.019), a statistical trend toward an increase in left ventricle ejection fraction (36.7 ± 13.2 vs 39.7 ± 14.9, p = 0.052), and cardiac output improvement (5.0 ± 0.7 vs 5.5 ± 0.9, p = 0.073) was observed in the patient postprocedure. Enhanced relative regional thickening was noted in the segments with adipose-derived stromal cell implantation. Conclusions Intramyocardial adipose-derived stromal cell implantation is a promising therapeutic option for selected, symptomatic patients with ischemic heart failure, who have preserved myocardial viability despite being unsuitable for direct revascularization.
Collapse
|
10
|
Shi Y, Xu X, Luan P, Kou W, Li M, Yu Q, Zhuang J, Xu Y, Peng W, Jian W. miR‑124‑3p regulates angiogenesis in peripheral arterial disease by targeting STAT3. Mol Med Rep 2020; 22:4890-4898. [PMID: 33174610 PMCID: PMC7646933 DOI: 10.3892/mmr.2020.11538] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2020] [Accepted: 09/09/2020] [Indexed: 12/13/2022] Open
Abstract
Peripheral arterial disease (PAD) is the third leading cause of cardiovascular morbidity worldwide, after coronary artery disease and stroke. As endogenous regulators of gene expression, microRNAs (miRs) are implicated in the development and progression of various diseases, including types of cancer, autoimmune diseases and heart diseases. In the present study, the role of miR-124-3p in PAD was investigated. The reverse transcription-quantitative PCR results indicated that the expression levels of miR-124-3p were significantly increased in the ischemic tissue of the hindlimb ischemia (HLI) model and in hypoxic human umbilical vein endothelial cells compared with the corresponding control groups. Proliferation, wound healing and tube formation assays demonstrated the inhibition of miR-124-3p on angiogenesis in vitro and the HLI model indicated the same function of miR-124-3p in vivo. A dual-luciferase reporter revealed STAT3 as the target of miR-124-3p. The expression levels of miR-124-3p in human blood were negatively correlated with ankle-brachial index, which is an index for the evaluation of the severity of PAD. Collectively, the present study indicated that miR-124-3p was a critical regulator of angiogenesis in PAD, and a potential diagnostic, prognostic and therapeutic target for PAD.
Collapse
Affiliation(s)
- Yefei Shi
- Department of Cardiology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, P.R. China
| | - Xu Xu
- Department of Cardiology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, P.R. China
| | - Peipei Luan
- Department of Cardiology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, P.R. China
| | - Wenxin Kou
- Department of Cardiology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, P.R. China
| | - Mingjie Li
- Department of Endocrinology, Xinhua Hospital Affiliated to Shanghai Jiaotong University, School of Medicine, Shanghai 200092, P.R. China
| | - Qing Yu
- Department of Cardiology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, P.R. China
| | - Jianhui Zhuang
- Department of Cardiology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, P.R. China
| | - Yawei Xu
- Department of Cardiology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, P.R. China
| | - Wenhui Peng
- Department of Cardiology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, P.R. China
| | - Weixia Jian
- Department of Endocrinology, Xinhua Hospital Affiliated to Shanghai Jiaotong University, School of Medicine, Shanghai 200092, P.R. China
| |
Collapse
|
11
|
Baek J, Cho Y, Park HJ, Choi G, Lee JS, Lee M, Yu SJ, Cho SW, Lee E, Im SG. A Surface-Tailoring Method for Rapid Non-Thermosensitive Cell-Sheet Engineering via Functional Polymer Coatings. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2020; 32:e1907225. [PMID: 32157771 DOI: 10.1002/adma.201907225] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/03/2019] [Revised: 12/30/2019] [Accepted: 02/18/2020] [Indexed: 06/10/2023]
Abstract
Cell sheet engineering, a technique utilizing a monolayer cell sheet, has recently emerged as a promising technology for scaffold-free tissue engineering. In contrast to conventional tissue-engineering approaches, the cell sheet technology allows cell harvest as a continuous cell sheet with intact extracellular matrix proteins and cell-cell junction, which facilitates cell transplantation without any other artificial biomaterials. A facile, non-thermoresponsive method is demonstrated for a rapid but highly reliable platform for cell-sheet engineering. The developed method exploits the precise modulation of cell-substrate interactions by controlling the surface energy of the substrate via a series of functional polymer coatings to enable prompt cell sheet harvesting within 100 s. The engineered surface can trigger an intrinsic cellular response upon the depletion of divalent cations, leading to spontaneous cell sheet detachment under physiological conditions (pH 7.4 and 37 °C) in a non-thermoresponsive manner. Additionally, the therapeutic potential of the cell sheet is successfully demonstrated by the transplantation of multilayered cell sheets into mouse models of diabetic wounds and ischemia. These findings highlight the ability of the developed surface for non-thermoresponsive cell sheet engineering to serve as a robust platform for regenerative medicine and provide significant breakthroughs in cell sheet technology.
Collapse
Affiliation(s)
- Jieung Baek
- Department of Chemical and Biomolecular Engineering and KI for NanoCentury, Korea Advanced Institute of Science and Technology, 291 Daehak-ro, Yuseong-gu, Daejeon, 34141, Republic of Korea
| | - Younghak Cho
- Department of Chemical and Biomolecular Engineering and KI for NanoCentury, Korea Advanced Institute of Science and Technology, 291 Daehak-ro, Yuseong-gu, Daejeon, 34141, Republic of Korea
| | - Hyun-Ji Park
- Department of Biotechnology, Yonsei University, Seoul, 03722, Republic of Korea
| | - Goro Choi
- Department of Chemical and Biomolecular Engineering and KI for NanoCentury, Korea Advanced Institute of Science and Technology, 291 Daehak-ro, Yuseong-gu, Daejeon, 34141, Republic of Korea
| | - Jong Seung Lee
- Department of Biotechnology, Yonsei University, Seoul, 03722, Republic of Korea
| | - Minseok Lee
- Department of Chemical and Biomolecular Engineering and KI for NanoCentury, Korea Advanced Institute of Science and Technology, 291 Daehak-ro, Yuseong-gu, Daejeon, 34141, Republic of Korea
| | - Seung Jung Yu
- Department of Chemical and Biomolecular Engineering and KI for NanoCentury, Korea Advanced Institute of Science and Technology, 291 Daehak-ro, Yuseong-gu, Daejeon, 34141, Republic of Korea
| | - Seung-Woo Cho
- Department of Biotechnology, Yonsei University, Seoul, 03722, Republic of Korea
- Center for Nanomedicine, Institute for Basic Science (IBS), Seoul, 03772, Republic of Korea
- Yonsei-IBS Institute, Yonsei University, Seoul, 03722, Republic of Korea
| | - Eunjung Lee
- Department of Chemical and Biomolecular Engineering and KI for NanoCentury, Korea Advanced Institute of Science and Technology, 291 Daehak-ro, Yuseong-gu, Daejeon, 34141, Republic of Korea
| | - Sung Gap Im
- Department of Chemical and Biomolecular Engineering and KI for NanoCentury, Korea Advanced Institute of Science and Technology, 291 Daehak-ro, Yuseong-gu, Daejeon, 34141, Republic of Korea
| |
Collapse
|
12
|
Dubský M, Jirkovská A, Bem R, Němcová A, Fejfarová V, Hazdrová J, Sutoris K, Chlupáč J, Skibová J, Jude EB. Impact of severe diabetic kidney disease on the clinical outcome of autologous cell therapy in people with diabetes and critical limb ischaemia. Diabet Med 2019; 36:1133-1140. [PMID: 31077439 DOI: 10.1111/dme.13985] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 05/10/2019] [Indexed: 12/21/2022]
Abstract
AIM To assess the impact of autologous cell therapy on critical limb ischaemia in people with diabetes and diabetic kidney disease. METHODS A total of 59 people with diabetes (type 1 or type 2) and critical limb ischaemia, persisting after standard revascularization, were treated with cell therapy in our foot clinic over 7 years; this group comprised 17 people with and 42 without severe diabetic kidney disease. The control group had the same inclusion criteria, but was treated conservatively and comprised 21 people with and 23 without severe diabetic kidney disease. Severe diabetic kidney disease was defined as chronic kidney disease stages 4-5 (GFR <30 ml/min/1.73 m²). Death and amputation-free survival were assessed during the 18-month follow-up; changes in transcutaneous oxygen pressure were evaluated at 6 and 12 months after cell therapy. RESULTS Transcutaneous oxygen pressure increased significantly in both groups receiving cell therapy compared to baseline (both P<0.01); no significant change in either of the control groups was observed. The cell therapy severe diabetic kidney disease group had a significantly longer amputation-free survival time compared to the severe diabetic kidney disease control group (hazard ratio 0.36, 95% CI 0.14-0.91; P=0.042); there was no difference in the non-severe diabetic kidney disease groups. The severe diabetic kidney disease control group had a tendency to have higher mortality (hazard ratio 2.82, 95% CI 0.81-9.80; P=0.062) than the non-severe diabetic kidney disease control group, but there was no difference between the severe diabetic kidney disease and non-severe diabetic kidney disease cell therapy groups. CONCLUSIONS The present study shows that autologous cell therapy in people with severe diabetic kidney disease significantly improved critical limb ischaemia and lengthened amputation-free survival in comparison with conservative treatment; however, the treatment did not influence overall survival.
Collapse
Affiliation(s)
- M Dubský
- Institute for Clinical and Experimental Medicine, Prague, Czech Republic
| | - A Jirkovská
- Institute for Clinical and Experimental Medicine, Prague, Czech Republic
| | - R Bem
- Institute for Clinical and Experimental Medicine, Prague, Czech Republic
| | - A Němcová
- Institute for Clinical and Experimental Medicine, Prague, Czech Republic
| | - V Fejfarová
- Institute for Clinical and Experimental Medicine, Prague, Czech Republic
| | - J Hazdrová
- Institute for Clinical and Experimental Medicine, Prague, Czech Republic
| | - K Sutoris
- Institute for Clinical and Experimental Medicine, Prague, Czech Republic
| | - J Chlupáč
- Institute for Clinical and Experimental Medicine, Prague, Czech Republic
| | - J Skibová
- Institute for Clinical and Experimental Medicine, Prague, Czech Republic
| | - E B Jude
- Diabetes Centre, Tameside Hospital NHS Foundation Trust and University of Manchester, Manchester, UK
| |
Collapse
|
13
|
Moon KC, Chung HY, Han SK, Jeong SH, Dhong ES. Possibility of Injecting Adipose-Derived Stromal Vascular Fraction Cells to Accelerate Microcirculation in Ischemic Diabetic Feet: A Pilot Study. Int J Stem Cells 2019; 12:107-113. [PMID: 30836733 PMCID: PMC6457712 DOI: 10.15283/ijsc18101] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2018] [Revised: 01/04/2019] [Accepted: 01/14/2019] [Indexed: 12/25/2022] Open
Abstract
Background and Objectives Beneficial effects of human adipose-derived stromal vascular fraction (SVF) cell injection on microcirculation have been recently reported in in vitro and in vivo studies. However, no clinical studies have reported its effect in diabetic patients who commonly experience compromised tissue perfusion, regardless of the status of intravascular blood flow. The present piloting study was designed to clinically examine the possibility of SVF cell injection to accelerate microcirculation, particularly in ischemic diabetic feet. Methods Ten diabetic feet were included to receive subcutaneous injection of SVF cells around wounds. Transcutaneous partial oxygen pressure (TcPO2) and cutaneous microvascular blood flow were measured before and every four weeks after cell injection until the 12th week visit. Results TcPO2 values increased from 31.3±7.4 before injection to 46.4±8.2 mmHg at 12 weeks after SVF injection (1.5-fold, p<0.05). Cutaneous microvascular blood flow levels increased from 34.0±21.1 before injection to 76.1±32.5 perfusion unit at 12 weeks after SVF injection (2.2-fold, p<0.05). There were no adverse events related to SVF cell injection. Conclusions Results of this study demonstrate that adipose-derived SVF cell injection have the possibility to provide beneficial effects on microcirculation in ischemic diabetic feet.
Collapse
Affiliation(s)
- Kyung-Chul Moon
- Department of Plastic Surgery, Korea University Guro Hospital, Seoul, Korea
| | - Ha-Yoon Chung
- Department of Plastic Surgery, Korea University Guro Hospital, Seoul, Korea
| | - Seung-Kyu Han
- Department of Plastic Surgery, Korea University Guro Hospital, Seoul, Korea
| | - Seong-Ho Jeong
- Department of Plastic Surgery, Korea University Guro Hospital, Seoul, Korea
| | - Eun-Sang Dhong
- Department of Plastic Surgery, Korea University Guro Hospital, Seoul, Korea
| |
Collapse
|
14
|
Osteopontin isoforms differentially promote arteriogenesis in response to ischemia via macrophage accumulation and survival. J Transl Med 2019; 99:331-345. [PMID: 29959420 PMCID: PMC6311150 DOI: 10.1038/s41374-018-0094-8] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2018] [Revised: 05/16/2018] [Accepted: 05/29/2018] [Indexed: 01/13/2023] Open
Abstract
Osteopontin (OPN) is critical for ischemia-induced neovascularization. Unlike rodents, humans express three OPN isoforms (a, b, and c); however, the roles of these isoforms in post-ischemic neovascularization and cell migration remain undefined. Our objective was to determine if OPN isoforms differentially affect post-ischemic neovascularization and to elucidate the mechanisms underlying these differences. To investigate if human OPN isoforms exert divergent effects on post-ischemic neovascularization, we utilized OPN-/- mice and a loss-of-function/gain-of-function approach in vivo and in vitro. In this study OPN-/- mice underwent hindlimb ischemia surgery and 1.5 × 106 lentivirus particles were administered intramuscularly to overexpress OPNa, OPNb, or OPNc. OPNa and OPNc significantly improved limb perfusion 30.4% ± 0.8 and 70.9% ± 6.3, respectively, and this translated to improved functional limb use, as measured by voluntary running wheel utilization. OPNa- and OPNc-treated animals exhibited significant increases in arteriogenesis, defined here as the remodeling of existing arterioles into larger conductance arteries. Macrophages play a prominent role in the arteriogenesis process and OPNa- and OPNc-treated animals showed significant increases in macrophage accumulation in vivo. In vitro, OPN isoforms did not affect macrophage polarization, whereas all three isoforms increased macrophage survival and decreased macrophage apoptosis. However, OPN isoforms exert differential effects on macrophage migration, where OPNa and OPNc significantly increased macrophage migration, with OPNc serving as the most potent isoform. In conclusion, human OPN isoforms exert divergent effects on neovascularization through differential effects on arteriogenesis and macrophage accumulation in vivo and on macrophage migration and survival, but not polarization, in vitro. Altogether, these data support that human OPN isoforms may represent novel therapeutic targets to improve neovascualrization and preserve tissue function in patients with obstructive artery diseases.
Collapse
|
15
|
Wolf K, Hu H, Isaji T, Dardik A. Molecular identity of arteries, veins, and lymphatics. J Vasc Surg 2019; 69:253-262. [PMID: 30154011 PMCID: PMC6309638 DOI: 10.1016/j.jvs.2018.06.195] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2018] [Accepted: 06/25/2018] [Indexed: 12/13/2022]
Abstract
BACKGROUND Arteries, veins, and lymphatic vessels are distinguished by structural differences that correspond to their different functions. Each of these vessels is also defined by specific molecular markers that persist throughout adult life; these markers are some of the molecular determinants that control the differentiation of embryonic undifferentiated cells into arteries, veins, or lymphatics. METHODS This is a review of experimental literature. RESULTS The Eph-B4 receptor and its ligand, ephrin-B2, are critical molecular determinants of vessel identity, arising on endothelial cells early in embryonic development. Eph-B4 and ephrin-B2 continue to be expressed on adult vessels and mark vessel identity. However, after vascular surgery, vessel identity can change and is marked by altered Eph-B4 and ephrin-B2 expression. Vein grafts show loss of venous identity, with less Eph-B4 expression. Arteriovenous fistulas show gain of dual arterial-venous identity, with both Eph-B4 and ephrin-B2 expression, and manipulation of Eph-B4 improves arteriovenous fistula patency. Patches used to close arteries and veins exhibit context-dependent gain of identity, that is, patches in the arterial environment gain arterial identity, whereas patches in the venous environment gain venous identity; these results show the importance of the host infiltrating cells in determining vascular identity after vascular surgery. CONCLUSIONS Changes in the vessel's molecular identity after vascular surgery correspond to structural changes that depend on the host's postsurgical environment. Regulation of vascular identity and the underlying molecular mechanisms may allow new therapeutic approaches to improve vascular surgical procedures.
Collapse
Affiliation(s)
- Katharine Wolf
- Vascular Biology and Therapeutics Program and Department of Surgery, Yale University School of Medicine, New Haven, Conn
| | - Haidi Hu
- Vascular Biology and Therapeutics Program and Department of Surgery, Yale University School of Medicine, New Haven, Conn
| | - Toshihiko Isaji
- Vascular Biology and Therapeutics Program and Department of Surgery, Yale University School of Medicine, New Haven, Conn
| | - Alan Dardik
- Vascular Biology and Therapeutics Program and Department of Surgery, Yale University School of Medicine, New Haven, Conn; VA Connecticut Healthcare System, West Haven, Conn.
| |
Collapse
|
16
|
Hussein EA. Stem Cell Therapy for Vascular Disorders. VASCULAR AND ENDOVASCULAR REVIEW 2018. [DOI: 10.15420/ver.2018.3.1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
Abstract
Peripheral vascular disease results from narrowing of the peripheral arteries that supply oxygenated blood and nutrients to the legs and feet. This pathology causes symptoms such as intermittent claudication (pain with walking), painful ischaemic ulcerations, or even limbthreatening gangrene. It is generally believed that the vascular endothelium, a monolayer of endothelial cells (ECs) that lines the luminal surface of all blood and lymphatic vessels, plays a dominant role in vascular homeostasis and vascular regeneration. As a result, stem cell-based regeneration of the endothelium may be a promising approach for the treatment of PAD. Critical limb ischaemia (CLI) is an advanced form of peripheral artery disease which is responsible for about 100,000 amputations each year in the US. Trials to date have reported clinical improvement and reduced need for amputation in patients with CLI who receive autologous bone marrow or mobilised peripheral blood stem cells for stimulation of angiogenesis. There is no effective treatment for lower limb ischaemia caused by peripheral vascular disease and it is necessary to amputate the limb at the end stage. Therefore, the concept of effective therapeutic angiogenesis has become widely accepted during the past few years and it has emerged as a strategy to treat tissue ischaemia by promoting collateral growth using drug, gene or cell therapy. This article provides an overview of current therapeutic challenges for the treatment of critical limb ischaemia, the basic mechanisms of stem cell therapy, the most relevant clinical trials as well as future directions for translational research in this area.
Collapse
|
17
|
NEMCOVA A, JIRKOVSKA A, DUBSKY M, BEM R, FEJFAROVA V, WOSKOVA V, PYSNA A, BUNCOVA M. Perfusion Scintigraphy in the Assessment of Autologous Cell Therapy in Diabetic Patients With Critical Limb Ischemia. Physiol Res 2018; 67:583-589. [DOI: 10.33549/physiolres.933868] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Perfusion scintigraphy with technetium-99-methoxy-isobutyl-isonitrile (99mTc-MIBI) is often used for assessing myocardial function but the number of studies concerning lower limb perfusion is limited. The aim of our study was to assess whether 99mTc-MIBI was an eligible method for evaluation of the effect of cell therapy on critical limb ischemia (CLI) in diabetic patients. 99mTc-MIBI of calf muscles was performed before and 3 months after autologous cell therapy (ACT) in 24 diabetic patients with CLI. Scintigraphic parameters such as rest count and exercising count after a stress test were defined. These parameters and their ratios were compared between treated and untreated (control) limbs and with changes in transcutaneous oxygen pressure (TcPO2) that served as a reference method. The effect of ACT was confirmed by a significant increase in TcPO2 values (p˂0.001) at 3 months after ACT. We did not observe any significant changes of scintigraphic parameters both at rest and after stress 3 months after ACT, there were no differences between treated and control limbs and no association with TcPO2 changes. Results of our study showed no significant contribution of 99mTc-MIBI of calf muscles to the assessment of ACT in diabetic patients with CLI over a 3-month follow-up period.
Collapse
Affiliation(s)
- A. NEMCOVA
- Diabetes Centre, Institute for Clinical and Experimental Medicine, Prague, Czech Republic
| | | | | | | | | | | | | | | |
Collapse
|
18
|
Bazzazi H, Zhang Y, Jafarnejad M, Popel AS. Computational modeling of synergistic interaction between αVβ3 integrin and VEGFR2 in endothelial cells: Implications for the mechanism of action of angiogenesis-modulating integrin-binding peptides. J Theor Biol 2018; 455:212-221. [PMID: 30036530 DOI: 10.1016/j.jtbi.2018.06.029] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2017] [Revised: 06/13/2018] [Accepted: 06/25/2018] [Indexed: 01/06/2023]
Abstract
Cooperation between VEGFR2 and integrin αVβ3 is critical for neovascularization in wound healing, cardiovascular ischemic diseases, ocular diseases, and tumor angiogenesis. In the present study, we developed a rule-based computational model to investigate the potential mechanism by which the Src-induced integrin association with VEGFR2 enhances VEGFR2 activation. Simulations demonstrated that the main function of integrin is to reduce the degradation of VEGFR2 and hence stabilize the activation signal. In addition, receptor synthesis rate and recruitment from internal compartment were found to be sensitive determinants of the activation state of VEGFR2. The model was then applied to simulate the effect of integrin-binding peptides such as tumstatin and cilengitide on VEGFR2 signaling. Further, computational modeling proposed potential molecular mechanisms for the angiogenesis-modulating activity of other integrin-binding peptides. The model highlights the complexity of the crosstalk between αVβ3 integrin and VEGFR2 and the necessity of utilizing models to elucidate potential mechanisms in angiogenesis-modulating peptide therapy.
Collapse
Affiliation(s)
- Hojjat Bazzazi
- Department of Biomedical Engineering, School of Medicine, Johns Hopkins University, Baltimore, MD 21205, United States
| | - Yu Zhang
- Department of Biomedical Engineering, School of Medicine, Johns Hopkins University, Baltimore, MD 21205, United States.
| | - Mohammad Jafarnejad
- Department of Biomedical Engineering, School of Medicine, Johns Hopkins University, Baltimore, MD 21205, United States
| | - Aleksander S Popel
- Department of Biomedical Engineering, School of Medicine, Johns Hopkins University, Baltimore, MD 21205, United States
| |
Collapse
|
19
|
Abstract
Advancing age will affect every individual and its impact on cardiac health deserves significant attention. The age-related physiological changes occurring in the coronary vasculature, myocardium, and valves set the stage upon which cardiovascular disease can escalate in the elderly population. The overall focus of this review is to highlight new and noteworthy studies and to incorporate reviews related to cardiac senescence in the context of the current state of the field. Lastly, future directions in the field of cardiac aging and the development of novel therapeutics to treat pathophysiological conditions typically associated with advancing age will be discussed.
Collapse
Affiliation(s)
- Amanda J LeBlanc
- Cardiovascular Innovation Institute, Department of Physiology, 302 E. Muhammad Ali Blvd, University of Louisville, Louisville, KY 40202
| | - Natia Q Kelm
- Cardiovascular Innovation Institute, Department of Physiology, 302 E. Muhammad Ali Blvd, University of Louisville, Louisville, KY 40202
| | - Monika George
- Cardiovascular Innovation Institute, Department of Physiology, 302 E. Muhammad Ali Blvd, University of Louisville, Louisville, KY 40202
| |
Collapse
|
20
|
Abstract
Peripheral arterial disease (PAD) refers to narrowing of the peripheral arteries and atherosclerosis is the most important cause. In patients with PAD, revascularization is the preferred therapeutic strategy; nonetheless several patients are not deemed candidates for it due to advanced disease or several comorbidities. The main target of therapeutic angiogenesis is to promote development of new arterial vessels and improve perfusion of ischemic tissue. Angiogenic growth factors such as vascular endothelial growth factor (VEGF), fibroblast growth factor (FGF), hepatocyte growth factor (HGF), administered intramuscularly or intra-arterially, have been shown to promote angiogenesis and development of collateral vasculature in preclinical studies. However, clinical studies failed to confirm their efficacy in ulcer healing and prevention of amputation, among patients with claudication or critical limb ischemia (CLI). Autologous progenitor cell therapy with bone marrow or adipose-derived progenitor cells administered intra-arterially or intra-muscularly, was shown to improve claudication symptoms and ankle-brachial index in small studies. However, subsequent randomized controlled studies did not demonstrate any beneficial effects of stem cell therapy on amputation rates and survival. Although, therapeutic angiogenesis remains an area of interest in PAD with several ongoing studies of investigational therapies, so far the use of these strategies in clinical practice has not been successful.
Collapse
Affiliation(s)
- Chakradhari Inampudi
- Division of Cardiovascular Diseases, University of Iowa Hospitals and Clinics, Iowa City, IA, USA
| | | | - Tomo Ando
- Division of Cardiology, Wayne State University, Detroit, MI, USA
| | - Alexandros Briasoulis
- Division of Cardiovascular Diseases, University of Iowa Hospitals and Clinics, Iowa City, IA, USA.
| |
Collapse
|
21
|
Foster AA, Dewi RE, Cai L, Hou L, Strassberg Z, Alcazar C, Heilshorn SC, Huang NF. Protein-engineered hydrogels enhance the survival of induced pluripotent stem cell-derived endothelial cells for treatment of peripheral arterial disease. Biomater Sci 2018; 6:614-622. [PMID: 29406542 PMCID: PMC5829050 DOI: 10.1039/c7bm00883j] [Citation(s) in RCA: 54] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
A key feature of peripheral arterial disease (PAD) is damage to endothelial cells (ECs), resulting in lower limb pain and restricted blood flow. Recent preclinical studies demonstrate that the transplantation of ECs via direct injection into the affected limb can result in significantly improved blood circulation. Unfortunately, the clinical application of this therapy has been limited by low cell viability and poor cell function. To address these limitations we have developed an injectable, recombinant hydrogel, termed SHIELD (Shear-thinning Hydrogel for Injectable Encapsulation and Long-term Delivery) for cell transplantation. SHIELD provides mechanical protection from cell membrane damage during syringe flow. Additionally, secondary in situ crosslinking provides a reinforcing network to improve cell retention, thereby augmenting the therapeutic benefit of cell therapy. In this study, we demonstrate the improved acute viability of human induced pluripotent stem cell-derived endothelial cells (iPSC-ECs) following syringe injection delivery in SHIELD, compared to saline. Using a murine hind limb ischemia model of PAD, we demonstrate enhanced iPSC-EC retention in vivo and improved neovascularization of the ischemic limb based on arteriogenesis following transplantation of iPSC-ECs delivered in SHIELD.
Collapse
Affiliation(s)
- Abbygail A. Foster
- Department of Materials Science and Engineering, Stanford University, Stanford, CA, USA
- Stanford Cardiovascular Institute, Stanford University, Stanford, CA, USA
| | - Ruby E. Dewi
- Department of Materials Science and Engineering, Stanford University, Stanford, CA, USA
| | - Lei Cai
- Department of Materials Science and Engineering, Stanford University, Stanford, CA, USA
| | - Luqia Hou
- Stanford Cardiovascular Institute, Stanford University, Stanford, CA, USA
- Veterans Affairs Palo Alto Health Care System, Palo Alto, CA, USA
| | | | - Cynthia Alcazar
- Veterans Affairs Palo Alto Health Care System, Palo Alto, CA, USA
| | - Sarah C. Heilshorn
- Department of Materials Science and Engineering, Stanford University, Stanford, CA, USA
- Stanford Cardiovascular Institute, Stanford University, Stanford, CA, USA
| | - Ngan F. Huang
- Stanford Cardiovascular Institute, Stanford University, Stanford, CA, USA
- Veterans Affairs Palo Alto Health Care System, Palo Alto, CA, USA
- Department of Cardiothoracic Surgery, Stanford, CA, USA
| |
Collapse
|
22
|
Ministro A, de Oliveira P, Nunes RJ, Dos Santos Rocha A, Correia A, Carvalho T, Rino J, Faísca P, Becker JD, Goyri-O'Neill J, Pina F, Poli E, Silva-Santos B, Pinto F, Mareel M, Serre K, Constantino Rosa Santos S. Low-dose ionizing radiation induces therapeutic neovascularization in a pre-clinical model of hindlimb ischemia. Cardiovasc Res 2018; 113:783-794. [PMID: 28444128 DOI: 10.1093/cvr/cvx065] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/23/2016] [Accepted: 03/29/2017] [Indexed: 11/12/2022] Open
Abstract
Aims We have previously shown that low-dose ionizing radiation (LDIR) induces angiogenesis but there is no evidence that it induces neovascularization in the setting of peripheral arterial disease. Here, we investigated the use of LDIR as an innovative and non-invasive strategy to stimulate therapeutic neovascularization using a model of experimentally induced hindlimb ischemia (HLI). Methods and results After surgical induction of unilateral HLI, both hindlimbs of female C57BL/6 mice were sham-irradiated or irradiated with four daily fractions of 0.3 Gy, in consecutive days and allowed to recover. We demonstrate that LDIR, significantly improved blood perfusion in the murine ischemic limb by stimulating neovascularization, as assessed by laser Doppler flow, capillary density, and collateral vessel formation. LDIR significantly increased the circulating levels of VEGF, PlGF, and G-CSF, as well as the number of circulating endothelial progenitor cells (EPCs) mediating their incorporation to ischemic muscles. These effects were dependent upon LDIR exposition on the ischemic niche (thigh and shank regions). In irradiated ischemic muscles, these effects were independent of the recruitment of monocytes and macrophages. Importantly, LDIR induced a durable and simultaneous up-regulation of a repertoire of pro-angiogenic factors and their receptors in endothelial cells (ECs), as evident in ECs isolated from the irradiated gastrocnemius muscles by laser capture microdissection. This specific mechanism was mediated via vascular endothelial growth factor (VEGF) receptor signaling, since VEGF receptor inhibition abrogated the LDIR-mediated gene up-regulation and impeded the increase in capillary density. Finally, the vasculature in an irradiated non-ischemic bed was not affected and after 52 week of LDIR exposure no differences in the incidence of morbidity and mortality were seen. Conclusions These findings disclose an innovative, non-invasive strategy to induce therapeutic neovascularization in a mouse model of HLI, emerging as a novel approach in the treatment of critical limb ischemia patients.
Collapse
Affiliation(s)
- Augusto Ministro
- Centro Cardiovascular da Universidade de Lisboa, Faculdade de Medicina, Universidade de Lisboa, Avenida Prof. Egas Moniz, 1649-028 Lisbon, Portugal.,Centro Hospitalar Lisboa Norte, Avenida Prof. Egas Moniz, 1649-035 Lisbon, Portugal
| | - Paula de Oliveira
- Centro Cardiovascular da Universidade de Lisboa, Faculdade de Medicina, Universidade de Lisboa, Avenida Prof. Egas Moniz, 1649-028 Lisbon, Portugal
| | - Raquel J Nunes
- Centro Cardiovascular da Universidade de Lisboa, Faculdade de Medicina, Universidade de Lisboa, Avenida Prof. Egas Moniz, 1649-028 Lisbon, Portugal
| | - André Dos Santos Rocha
- Centro Cardiovascular da Universidade de Lisboa, Faculdade de Medicina, Universidade de Lisboa, Avenida Prof. Egas Moniz, 1649-028 Lisbon, Portugal
| | - Adriana Correia
- Centro Cardiovascular da Universidade de Lisboa, Faculdade de Medicina, Universidade de Lisboa, Avenida Prof. Egas Moniz, 1649-028 Lisbon, Portugal
| | - Tânia Carvalho
- Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, Avenida Prof. Egas Moniz, 1649-028 Lisbon, Portugal
| | - José Rino
- Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, Avenida Prof. Egas Moniz, 1649-028 Lisbon, Portugal
| | - Pedro Faísca
- Research Center for Biosciences & Health Technologies, Faculdade de Medicina Veterinária, Universidade Lusófona de Humanidades e Tecnologias. Campo Grande 376, 1749-024 Lisbon, Portugal
| | - Jorg D Becker
- Instituto Gulbenkian de Ciência, Rua Quinta Grande 6, 2780-156 Oeiras, Portugal
| | - João Goyri-O'Neill
- Nova Medical School/Faculdade de Ciências Médicas, Universidade Nova de Lisboa, Campo Mártires da Pátria 130, 1169-056 Lisbon, Portugal
| | - Filomena Pina
- Centro Hospitalar Lisboa Norte, Avenida Prof. Egas Moniz, 1649-035 Lisbon, Portugal
| | - Esmeralda Poli
- Centro Hospitalar Lisboa Norte, Avenida Prof. Egas Moniz, 1649-035 Lisbon, Portugal
| | - Bruno Silva-Santos
- Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, Avenida Prof. Egas Moniz, 1649-028 Lisbon, Portugal
| | - Fausto Pinto
- Centro Cardiovascular da Universidade de Lisboa, Faculdade de Medicina, Universidade de Lisboa, Avenida Prof. Egas Moniz, 1649-028 Lisbon, Portugal.,Centro Hospitalar Lisboa Norte, Avenida Prof. Egas Moniz, 1649-035 Lisbon, Portugal.,Faculdade de Medicina, Universidade de Lisboa, Avenida Prof. Egas Moniz, 1649-028 Lisbon, Portugal
| | - Marc Mareel
- University Hospital Ghent, De Pintelaan, 185, B-9000 Ghent, Belgium
| | - Karine Serre
- Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, Avenida Prof. Egas Moniz, 1649-028 Lisbon, Portugal
| | - Susana Constantino Rosa Santos
- Centro Cardiovascular da Universidade de Lisboa, Faculdade de Medicina, Universidade de Lisboa, Avenida Prof. Egas Moniz, 1649-028 Lisbon, Portugal.,Faculdade de Medicina, Universidade de Lisboa, Avenida Prof. Egas Moniz, 1649-028 Lisbon, Portugal
| |
Collapse
|
23
|
Caicedo D, Devesa P, Arce VM, Requena J, Devesa J. Chronic limb-threatening ischemia could benefit from growth hormone therapy for wound healing and limb salvage. Ther Adv Cardiovasc Dis 2018; 12:53-72. [PMID: 29271292 PMCID: PMC5772430 DOI: 10.1177/1753944717745494] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/02/2017] [Accepted: 10/12/2017] [Indexed: 01/20/2023] Open
Abstract
Revascularization for chronic limb-threatening ischemia (CLTI) is necessary to alleviate symptoms and wound healing. When it fails or is not possible, there are few alternatives to avoid limb amputation in these patients. Although experimental studies with stem cells and growth factors have shown promise, clinical trials have demonstrated inconsistent results because CLTI patients generally need arteriogenesis rather than angiogenesis. Moreover, in addition to the perfusion of the limb, there is the need to improve the neuropathic response for wound healing, especially in diabetic patients. Growth hormone (GH) is a pleiotropic hormone capable of boosting the aforementioned processes and adds special benefits for the redox balance. This hormone has the potential to mitigate symptoms in ischemic patients with no other options and improves the cardiovascular complications associated with the disease. Here, we discuss the pros and cons of using GH in such patients, focus on its effects on peripheral arteries, and analyze the possible benefits of treating CLTI with this hormone.
Collapse
Affiliation(s)
- Diego Caicedo
- Scientific Direction, Medical Center Foltra. Travesía Montouto, 24; 15710-Teo, A Coruña, 15886, Spain
| | - Pablo Devesa
- Scientific Direction, Medical Center Foltra. Travesía Montouto, 24; 15710-Teo, A Coruña, 15886, Spain
| | - Víctor M. Arce
- Scientific Direction, Medical Center Foltra. Travesía Montouto, 24; 15710-Teo, A Coruña, 15886, Spain
| | - Julia Requena
- Scientific Direction, Medical Center Foltra. Travesía Montouto, 24; 15710-Teo, A Coruña, 15886, Spain
| | - Jesús Devesa
- Scientific Direction, Medical Center Foltra. Travesía Montouto, 24; 15710-Teo, A Coruña, 15886, Spain
| |
Collapse
|
24
|
Clegg LE, Mac Gabhann F. A computational analysis of pro-angiogenic therapies for peripheral artery disease. Integr Biol (Camb) 2018; 10:18-33. [PMID: 29327758 PMCID: PMC7017937 DOI: 10.1039/c7ib00218a] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Inducing therapeutic angiogenesis to effectively form hierarchical, non-leaky networks of perfused vessels in tissue engineering applications and ischemic disease remains an unmet challenge, despite extensive research and multiple clinical trials. Here, we use a previously-developed, multi-scale, computational systems pharmacology model of human peripheral artery disease to screen a diverse array of promising pro-angiogenic strategies, including gene therapy, biomaterials, and antibodies. Our previously-validated model explicitly accounts for VEGF immobilization, Neuropilin-1 binding, and weak activation of VEGF receptor 2 (VEGFR2) by the "VEGFxxxb" isoforms. First, we examine biomaterial-based delivery of VEGF engineered for increased affinity to the extracellular matrix. We show that these constructs maintain VEGF close to physiological levels and extend the duration of VEGFR2 activation. We demonstrate the importance of sub-saturating VEGF dosing to prevent angioma formation. Second, we examine the potential of ligand- or receptor-based gene therapy to normalize VEGF receptor signaling. Third, we explore the potential for antibody-based pro-angiogenic therapy. Our model supports recent observations that improvement in perfusion following treatment with anti-VEGF165b in mice is mediated by VEGF-receptor 1, not VEGFR2. Surprisingly, the model predicts that the approved anti-VEGF cancer drug, bevacizumab, may actually improve signaling of both VEGFR1 and VEGFR2 via a novel 'antibody swapping' effect that we demonstrate here. Altogether, this model provides insight into the mechanisms of action of several classes of pro-angiogenic strategies within the context of the complex molecular and physiological processes occurring in vivo. We identify molecular signaling similarities between promising approaches and key differences between promising and ineffective strategies.
Collapse
Affiliation(s)
- Lindsay E Clegg
- Institute for Computational Medicine, Institute for NanoBioTechnology, and Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, USA.
| | | |
Collapse
|
25
|
Ranchoux B, Harvey LD, Ayon RJ, Babicheva A, Bonnet S, Chan SY, Yuan JXJ, Perez VDJ. Endothelial dysfunction in pulmonary arterial hypertension: an evolving landscape (2017 Grover Conference Series). Pulm Circ 2018; 8:2045893217752912. [PMID: 29283043 PMCID: PMC5798691 DOI: 10.1177/2045893217752912] [Citation(s) in RCA: 105] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/23/2017] [Accepted: 12/18/2017] [Indexed: 02/06/2023] Open
Abstract
Endothelial dysfunction is a major player in the development and progression of vascular pathology in pulmonary arterial hypertension (PAH), a disease associated with small vessel loss and obstructive vasculopathy that leads to increased pulmonary vascular resistance, subsequent right heart failure, and premature death. Over the past ten years, there has been tremendous progress in our understanding of pulmonary endothelial biology as it pertains to the genetic and molecular mechanisms that orchestrate the endothelial response to direct or indirect injury, and how their dysregulation can contribute to the pathogenesis of PAH. As one of the major topics included in the 2017 Grover Conference Series, discussion centered on recent developments in four areas of pulmonary endothelial biology: (1) angiogenesis; (2) endothelial-mesenchymal transition (EndMT); (3) epigenetics; and (4) biology of voltage-gated ion channels. The present review will summarize the content of these discussions and provide a perspective on the most promising aspects of endothelial dysfunction that may be amenable for therapeutic development.
Collapse
Affiliation(s)
| | - Lloyd D. Harvey
- University of Pittsburgh Vascular Medicine Institute Division of Cardiology, Pittsburgh, PA, USA
| | - Ramon J. Ayon
- Division of Translational and Regenerative Medicine, The University of Arizona College of Medicine, Tucson, AZ, USA
| | - Aleksandra Babicheva
- Division of Translational and Regenerative Medicine, The University of Arizona College of Medicine, Tucson, AZ, USA
| | | | - Stephen Y. Chan
- University of Pittsburgh Vascular Medicine Institute Division of Cardiology, Pittsburgh, PA, USA
| | - Jason X.-J. Yuan
- Division of Translational and Regenerative Medicine, The University of Arizona College of Medicine, Tucson, AZ, USA
| | - Vinicio de Jesus Perez
- Division of Pulmonary and Critical Care Medicine, Stanford University Medical Center, Stanford, CA, USA
- The Vera Moulton Wall Center for Pulmonary Vascular Medicine, Stanford University Medical Center, Stanford, CA, USA
- Stanford Cardiovascular Institute, Stanford University Medical Center, Stanford, CA, USA
| |
Collapse
|
26
|
Conte SM, Vale PR. Peripheral Arterial Disease. Heart Lung Circ 2017; 27:427-432. [PMID: 29150158 DOI: 10.1016/j.hlc.2017.10.014] [Citation(s) in RCA: 107] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2017] [Revised: 08/10/2017] [Accepted: 10/18/2017] [Indexed: 12/12/2022]
Abstract
Peripheral arterial disease (PAD) affects 15% of the Australian population and is a powerful and serious predictor of cardiovascular mortality yet continues to be under-recognised and undertreated. Diagnosis is simple and management is centred upon symptom relief and risk minimisation. While novel and specialised therapies play a role, the bulk of management is approachable and feasible. In this review, we cover the epidemiology, risk factors, associated conditions, classification, and natural history of PAD. We then discuss current diagnostic and therapeutic options as well as emerging therapies for this common condition.
Collapse
Affiliation(s)
- Sean M Conte
- St Vincent's Hospital, Melbourne, Vic, Australia.
| | - Peter R Vale
- Mater Hospital Sydney, University of Notre Dame Australia School of Medicine, Sydney, NSW, Australia
| |
Collapse
|
27
|
Friedrich CC, Lin Y, Krannich A, Wu Y, Vacanti JP, Neville CM. Enhancing engineered vascular networks in vitro and in vivo: The effects of IGF1 on vascular development and durability. Cell Prolif 2017; 51. [PMID: 29110360 DOI: 10.1111/cpr.12387] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2016] [Accepted: 07/21/2017] [Indexed: 12/15/2022] Open
Abstract
OBJECTIVES Creation of functional, durable vasculature remains an important goal within the field of regenerative medicine. Engineered biological vasculature has the potential to restore or improve human tissue function. We hypothesized that the pleotropic effects of insulin-like growth factor 1 (IGF1) would enhance the engineering of capillary-like vasculature. MATERIALS AND METHODS The impact of IGF1 upon vasculogenesis was examined in in vitro cultures for a period of up to 40 days and as subcutaneous implants within immunodeficient mice. Co-cultures of human umbilical vein endothelial cells and human bone marrow-derived mesenchymal stem cells in collagen-fibronectin hydrogels were supplemented with either recombinant IGF1 protein or genetically engineered cells to provide sustained IGF1. Morphometric analysis was performed on the vascular networks that formed in four concentrations of IGF1. RESULTS IGF1 supplementation significantly enhanced de novo vasculogenesis both in vitro and in vivo. Effects were long-term as they lasted the duration of the study period, and included network density, vessel length, and diameter. Bifurcation density was not affected. However, the highest concentrations of IGF1 tested were either ineffective or even deleterious. Sustained IGF1 delivery was required in vivo as the inclusion of recombinant IGF1 protein had minimal impact. CONCLUSION IGF1 supplementation can be used to produce neovasculature with significantly enhanced network density and durability. Its use is a promising methodology for engineering de novo vasculature to support regeneration of functional tissue.
Collapse
Affiliation(s)
- Claudia C Friedrich
- Center for Regenerative Medicine, Massachusetts General Hospital, Boston, MA, USA.,Department of Surgery, Massachusetts General Hospital, Boston, MA, USA.,Department of Anesthesiology and Intensive Care Medicine, Campus Virchow Klinikum and Campus Charité Mitte, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Yunfeng Lin
- Center for Regenerative Medicine, Massachusetts General Hospital, Boston, MA, USA.,Department of Orthopaedics, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA.,State Key Laboratory of Oral Diseases, West China College of Stomatology, Sichuan University, Chengdu, China
| | - Alexander Krannich
- Department of Biostatistics, Clinical Research Unit, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Yinan Wu
- Department of Biostatistics, Clinical Research Unit, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Joseph P Vacanti
- Center for Regenerative Medicine, Massachusetts General Hospital, Boston, MA, USA.,Department of Surgery, Massachusetts General Hospital, Boston, MA, USA
| | - Craig M Neville
- Center for Regenerative Medicine, Massachusetts General Hospital, Boston, MA, USA.,Department of Surgery, Massachusetts General Hospital, Boston, MA, USA.,Department of Orthopaedics, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
28
|
Frump AL, Bonnet S, de Jesus Perez VA, Lahm T. Emerging role of angiogenesis in adaptive and maladaptive right ventricular remodeling in pulmonary hypertension. Am J Physiol Lung Cell Mol Physiol 2017; 314:L443-L460. [PMID: 29097426 DOI: 10.1152/ajplung.00374.2017] [Citation(s) in RCA: 53] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Right ventricular (RV) function is the primary prognostic factor for both morbidity and mortality in pulmonary hypertension (PH). RV hypertrophy is initially an adaptive physiological response to increased overload; however, with persistent and/or progressive afterload increase, this response frequently transitions to more pathological maladaptive remodeling. The mechanisms and disease processes underlying this transition are mostly unknown. Angiogenesis has recently emerged as a major modifier of RV adaptation in the setting of pressure overload. A novel paradigm has emerged that suggests that angiogenesis and angiogenic signaling are required for RV adaptation to afterload increases and that impaired and/or insufficient angiogenesis is a major driver of RV decompensation. Here, we summarize our current understanding of the concepts of maladaptive and adaptive RV remodeling, discuss the current literature on angiogenesis in the adapted and failing RV, and identify potential therapeutic approaches targeting angiogenesis in RV failure.
Collapse
Affiliation(s)
- Andrea L Frump
- Division of Pulmonary, Critical Care, Sleep and Occupational Medicine, Department of Medicine, Indiana University School of Medicine , Indianapolis, Indiana
| | - Sébastien Bonnet
- Pulmonary Hypertension Research Group, Institut Universitaire de Cardiologie et de Pneumologie de Québec Research Center, Laval University , Quebec City, Quebec , Canada
| | - Vinicio A de Jesus Perez
- Division of Pulmonary/Critical Care, Stanford University School of Medicine , Stanford, California.,Vera Moulton Wall Center for Pulmonary Vascular Disease, Stanford University School of Medicine , Stanford, California
| | - Tim Lahm
- Division of Pulmonary, Critical Care, Sleep and Occupational Medicine, Department of Medicine, Indiana University School of Medicine , Indianapolis, Indiana.,Richard L. Roudebush Veterans Affairs Medical Center , Indianapolis, Indiana.,Department of Cellular and Integrative Physiology, Indiana University School of Medicine , Indianapolis, Indiana
| |
Collapse
|
29
|
Hybrid Nitric Oxide Donor and its Carrier for the Treatment of Peripheral Arterial Diseases. Sci Rep 2017; 7:8692. [PMID: 28821752 PMCID: PMC5562917 DOI: 10.1038/s41598-017-08441-9] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2017] [Accepted: 07/12/2017] [Indexed: 01/20/2023] Open
Abstract
Nitric oxide (NO) has been known to promote physiological angiogenesis to treat peripheral arterial diseases (PAD) by increasing the vascular endothelial growth factor (VEGF) level in endothelial cells (ECs) and preventing platelet adherence and leukocyte chemotaxis. However, the ongoing ischemic event during peripheral ischemia produces superoxide and diminishes the NO bioavailability by forming toxic peroxynitrite anion. Here we disclose an efficacious hybrid molecule 4-(5-Amino-1,2,3-oxadiazol-3-yl)-2,2,6,6-tetramethyl-1-piperidinol (SA-2) containing both antioxidant and NO donor functionalities that provide a therapeutic level of NO necessary to promote angiogenesis and to protect ECs against hydrogen peroxide-induced oxidative stress. Compound SA-2 scavenged reactive oxygen species, inhibited proliferation and migration of smooth muscle cells (SMCs) and promoted the tube formation from ECs. Copolymer poly(lactic-co-glycolic acid) (PLGA) nanoparticles loaded with SA-2 provided a sustained release of NO over days, improved aqueous stability in serum, protected ECs against oxidative stress, and enhanced angiogenesis under stress conditions as compared to that of the control in the in vitro matrigel tube formation assay. These results indicated the potential use of SA-2 nanoparticles as an alternative therapy to treat PAD.
Collapse
|
30
|
Mistry RK, Brewer AC. Redox regulation of gasotransmission in the vascular system: A focus on angiogenesis. Free Radic Biol Med 2017; 108:500-516. [PMID: 28433660 PMCID: PMC5698259 DOI: 10.1016/j.freeradbiomed.2017.04.025] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/03/2017] [Revised: 04/15/2017] [Accepted: 04/18/2017] [Indexed: 02/06/2023]
Abstract
Reactive oxygen species have emerged as key participants in a broad range of physiological and pathophysiological processes, not least within the vascular system. Diverse cellular functions which have been attributed to some of these pro-oxidants within the vasculature include the regulation of blood pressure, neovascularisation and vascular inflammation. We here highlight the emerging roles of the enzymatically-generated reaction oxygen species, O2- and H2O2, in the regulation of the functions of the gaseous signalling molecules: nitric oxide (NO), carbon monoxide (CO), and hydrogen sulphide (H2S). These gasotransmitters are produced on demand from distinct enzymatic sources and in recent years it has become apparent that they are capable of mediating a number of homeostatic processes within the cardiovascular system including enhanced vasodilation, angiogenesis, wound healing and improved cardiac function following myocardial infarction. In common with O2- and/or H2O2 they signal by altering the functions of target proteins, either by the covalent modification of thiol groups or by direct binding to metal centres within metalloproteins, most notably haem proteins. The regulation of the enzymes which generate NO, CO and H2S have been shown to be influenced at both the transcriptional and post-translational levels by redox-dependent mechanisms, while the activity and bioavailability of the gasotransmitters themselves are also subject to oxidative modification. Within vascular cells, the family of nicotinamide adenine dinucleotide phosphate oxidases (NAPDH oxidases/Noxs) have emerged as functionally significant sources of regulated O2- and H2O2 production and accordingly, direct associations between Nox-generated oxidants and the functions of specific gasotransmitters are beginning to be identified. This review focuses on the current knowledge of the redox-dependent mechanisms which regulate the generation and activity of these gases, with particular reference to their roles in angiogenesis.
Collapse
Affiliation(s)
- Rajesh K Mistry
- Cardiovascular Division, James Black Centre, King's College London BHF Centre of Excellence, 125 Coldharbour Lane, London SE5 9NU, UK
| | - Alison C Brewer
- Cardiovascular Division, James Black Centre, King's College London BHF Centre of Excellence, 125 Coldharbour Lane, London SE5 9NU, UK.
| |
Collapse
|
31
|
Dubský M, Jirkovská A, Bem R, Nemcová A, Fejfarová V, Jude EB. Cell therapy of critical limb ischemia in diabetic patients - State of art. Diabetes Res Clin Pract 2017; 126:263-271. [PMID: 28288436 DOI: 10.1016/j.diabres.2017.02.028] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/30/2016] [Revised: 12/19/2016] [Accepted: 02/22/2017] [Indexed: 01/15/2023]
Abstract
In this review we report on the state of cell therapy of critical limb ischemia (CLI) with respect to differences between diabetic and non-diabetic patients mainly from the clinical point of view. CLI is the most severe form of peripheral arterial disease and its diagnosis and treatment in diabetic patients is very difficult. The therapeutic effect of standard methods of CLI treatment is only partial - more than one third of diabetic patients are not eligible for standard revascularization; therefore, new therapeutic techniques such as cell therapy have been studied in clinical trials. Presence of CLI in patients with diabetic foot disease is associated with worse clinical outcomes such as lack of healing of foot ulcers, major amputations and premature mortality. A revascularization procedure cannot be successful as the only method in contrast to patients without diabetes, but it must always be part of a complex therapy focused not only on ischemia, but also on treatment of infection, off-loading, metabolic control of diabetes and nutrition, local therapy, etc. Therefore, the main criteria for cell therapy may vary in diabetic patients and non-diabetic persons and results of this treatment method should always be assessed in the context of ensuring comprehensive therapy. This review carries out an analysis of the source of precursor cells, route of administration and brings a brief report of published data with respect to diabetic and non-diabetic patients and our experience with autologous cell therapy of diabetic patients with CLI. Analysis of the studies in terms of diabetes is difficult, because in most of them sub-analysis for diabetic patients is not performed separately. The other problem is that it is not clear if diabetic patients received adequate complex treatment for their foot ulcers which can strongly affect the rate of major amputation as an outcome of CLI treatment.
Collapse
Affiliation(s)
- Michal Dubský
- Institute for Clinical and Experimental Medicine, Prague, Czech Republic.
| | | | - Robert Bem
- Institute for Clinical and Experimental Medicine, Prague, Czech Republic
| | - Andrea Nemcová
- Institute for Clinical and Experimental Medicine, Prague, Czech Republic
| | | | - Edward B Jude
- Diabetes Centre, Tameside Hospital NHS Foundation Trust and University of Manchester, Lancashire, UK
| |
Collapse
|
32
|
Thej C, Ramadasse B, Walvekar A, Majumdar AS, Balasubramanian S. Development of a surrogate potency assay to determine the angiogenic activity of Stempeucel®, a pooled, ex-vivo expanded, allogeneic human bone marrow mesenchymal stromal cell product. Stem Cell Res Ther 2017; 8:47. [PMID: 28245882 PMCID: PMC5331748 DOI: 10.1186/s13287-017-0488-3] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2016] [Revised: 12/07/2016] [Accepted: 01/24/2017] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Mesenchymal stromal cells (MSCs) have emerged as a more beneficial alternative to conventional therapy and may offer a potential cure for unmet medical needs. MSCs are known to possess strong immunomodulatory and anti-inflammatory properties. Moreover, they promote angiogenesis and tissue regeneration through the secretion of trophic factors. For these reasons, the past decade witnessed a sharp increase in the number of clinical trials conducted with stem cells for various vascular diseases requiring angiogenesis. In this study, we evaluated the in vitro angiogenic potency of Stempeucel®, which is an allogeneic pooled human bone marrow-derived mesenchymal stromal cell (phBMMSC) product. We previously established the safety of Stempeucel® in our pre-clinical studies, and clinical trials conducted for critical limb ischaemia and acute myocardial infarction. METHODS Because the proposed mechanism of action of phBMMSCs is mainly through the secretion of pro-angiogenic cytokines, we developed a surrogate potency assay by screening various batches of large-scale expanded phBMMSCs for the expression of angiogenic factors and cytokines through gene expression and growth factor analyses, followed by in vitro functional assays. RESULTS The well characterized angiogenic vascular endothelial growth factor (VEGF) was selected and quantified in twenty six manufactured batches of phBMMSCs to establish consistency following the United States Food and Drug Administration recommendations. According to recommendations 21 CFR 211.165(e) and 211.194(a)(2), we also established and documented the specificity and reproducibility of the test methods employed through validation. Moreover, we also attempted to elucidate the mechanism of action of the cell population to ensure appropriate biological activity. The functional role of VEGF has been established through in vitro angiogenic assays and a dose-dependent correlation was observed with in vitro functional results. CONCLUSIONS The data generated from this study suggest the selection of VEGF as a single surrogate marker to test the angiogenic potency of phBMMSCs. Our study reports the quantification of VEGF in twenty six batches of large-scale manufactured phBMMSCs, and a concentration-dependent correlation of secreted VEGF to endothelial cell functions of migration, proliferation and tube formation, in the conditioned medium obtained from nine phBMMSC batches. To our cognizance, this is the first study in which a single angiogenic factor (VEGF) has been qualified as a surrogate potency marker through all three in vitro functional assays to determine the angiogenic potency of the phBMMSC population.
Collapse
Affiliation(s)
- Charan Thej
- Stempeutics Research Pvt. Ltd, Akshay Tech Park, #72 & 73, 2nd Floor, EPIP Zone, Phase 1, Bangalore, Whitefield, 560066, India.,Manipal University, Manipal, Karnataka, India
| | - Balamurugan Ramadasse
- Stempeutics Research Pvt. Ltd, Akshay Tech Park, #72 & 73, 2nd Floor, EPIP Zone, Phase 1, Bangalore, Whitefield, 560066, India
| | - Ankita Walvekar
- Stempeutics Research Pvt. Ltd, Akshay Tech Park, #72 & 73, 2nd Floor, EPIP Zone, Phase 1, Bangalore, Whitefield, 560066, India
| | - Anish S Majumdar
- Stempeutics Research Pvt. Ltd, Akshay Tech Park, #72 & 73, 2nd Floor, EPIP Zone, Phase 1, Bangalore, Whitefield, 560066, India.
| | - Sudha Balasubramanian
- Stempeutics Research Pvt. Ltd, Akshay Tech Park, #72 & 73, 2nd Floor, EPIP Zone, Phase 1, Bangalore, Whitefield, 560066, India.
| |
Collapse
|
33
|
Fakoya AOJ. New Delivery Systems of Stem Cells for Vascular Regeneration in Ischemia. Front Cardiovasc Med 2017; 4:7. [PMID: 28286751 PMCID: PMC5323391 DOI: 10.3389/fcvm.2017.00007] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2016] [Accepted: 02/07/2017] [Indexed: 01/08/2023] Open
Abstract
The finances of patients and countries are increasingly overwhelmed with the plague of cardiovascular diseases as a result of having to chronically manage the associated complications of ischemia such as heart failures, neurological deficits, chronic limb ulcers, gangrenes, and amputations. Hence, scientific research has sought for alternate therapies since pharmacological and surgical treatments have fallen below expectations in providing the desired quality of life. The advent of stem cells research has raised expectations with respect to vascular regeneration and tissue remodeling, hence assuring the patients of the possibility of an improved quality of life. However, these supposed encouraging results have been short-lived as the retention, survival, and engraftment rates of these cells appear to be inadequate; hence, the long-term beneficial effects of these cells cannot be ascertained. These drawbacks have led to the relentless research into better ways to deliver stem cells or angiogenic factors (which mobilize stem cells) to the regions of interest to facilitate increased retention, survival, engraftment, and regeneration. This review considered methods, such as the use of scaffolds, retrograde coronary delivery, improved combinations, stem cell pretreatment, preconditioning, stem cell exosomes, mannitol, magnet, and ultrasound-enhanced delivery, homing techniques, and stem cell modulation. Furthermore, the study appraised the possibility of a combination therapy of stem cells and macrophages, considering the enormous role macrophages play in repair, remodeling, and angiogenesis.
Collapse
|
34
|
Rybalko V, Hsieh PL, Ricles LM, Chung E, Farrar RP, Suggs LJ. Therapeutic potential of adipose-derived stem cells and macrophages for ischemic skeletal muscle repair. Regen Med 2017; 12:153-167. [PMID: 28244825 DOI: 10.2217/rme-2016-0094] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
AIM Progressive ischemia due to peripheral artery disease causes muscle damage and reduced strength of the lower extremities. Autologous cell therapy is an attractive treatment to restore perfusion and improve muscle function. Adipose-derived stem cells (ASCs) have therapeutic potential in tissue repair, including polarizing effects on macrophages (MPs). MATERIALS & METHODS Co-culture systems of ASCs and MPs were analyzed for gene and protein expression modifications in ASC-conditioned MPs. Co-transplantation of MPs/ASCs in vivo led to improved skeletal muscle regeneration in a mouse model of peripheral artery disease. RESULTS ASCs/MPs therapy restored muscle function, increased perfusion and reduced inflammatory infiltrate. CONCLUSION Combined MPs/ASCs cell therapy is a promising approach to restore muscle function and stimulate local angiogenesis in the ischemic limb.
Collapse
Affiliation(s)
- Viktoriya Rybalko
- Biomedical Engineering, Cockrell School of Engineering, The University of Texas at Austin, 107 W Dean Keeton, Austin, TX 78712, USA
| | - Pei-Ling Hsieh
- Department of Kinesiology, The University of Texas at Austin, 1 University Station D3700, Austin, TX 78712, USA
| | - Laura M Ricles
- Biomedical Engineering, Cockrell School of Engineering, The University of Texas at Austin, 107 W Dean Keeton, Austin, TX 78712, USA
| | - Eunna Chung
- Biomedical Engineering, Cockrell School of Engineering, The University of Texas at Austin, 107 W Dean Keeton, Austin, TX 78712, USA
| | - Roger P Farrar
- Department of Kinesiology, The University of Texas at Austin, 1 University Station D3700, Austin, TX 78712, USA
| | - Laura J Suggs
- Biomedical Engineering, Cockrell School of Engineering, The University of Texas at Austin, 107 W Dean Keeton, Austin, TX 78712, USA
| |
Collapse
|
35
|
Heterodimerisation between VEGFR-1 and VEGFR-2 and not the homodimers of VEGFR-1 inhibit VEGFR-2 activity. Vascul Pharmacol 2017; 88:11-20. [DOI: 10.1016/j.vph.2016.11.007] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2016] [Accepted: 11/20/2016] [Indexed: 01/13/2023]
|
36
|
Sakakibara N, Igarashi J, Takata M, Konishi R, Kato Y, Tsukamoto I. Synthesis and Evaluation of Novel Cyclopropane Nucleoside as Potential Tube Formation Agents. Chem Pharm Bull (Tokyo) 2017; 65:504-510. [DOI: 10.1248/cpb.c17-00056] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Affiliation(s)
- Norikazu Sakakibara
- Faculty of Pharmaceutical Sciences at Kagawa Campus, Tokushima Bunri University
| | - Junsuke Igarashi
- Department of Cardiovascular Physiology, Faculty of Medicine, Kagawa University
| | - Maki Takata
- Department of Pharmaco-Bio-Informatics, Faculty of Medicine, Kagawa University
| | - Ryoji Konishi
- Department of Pharmaco-Bio-Informatics, Faculty of Medicine, Kagawa University
| | - Yoshihisa Kato
- Faculty of Pharmaceutical Sciences at Kagawa Campus, Tokushima Bunri University
| | - Ikuko Tsukamoto
- Department of Pharmaco-Bio-Informatics, Faculty of Medicine, Kagawa University
| |
Collapse
|
37
|
Thomas KN, van Rij AM, Lucas SJE, Cotter JD. Lower-limb hot-water immersion acutely induces beneficial hemodynamic and cardiovascular responses in peripheral arterial disease and healthy, elderly controls. Am J Physiol Regul Integr Comp Physiol 2016; 312:R281-R291. [PMID: 28003211 DOI: 10.1152/ajpregu.00404.2016] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2016] [Revised: 12/19/2016] [Accepted: 12/19/2016] [Indexed: 11/22/2022]
Abstract
Passive heat induces beneficial perfusion profiles, provides substantive cardiovascular strain, and reduces blood pressure, thereby holding potential for healthy and cardiovascular disease populations. The aim of this study was to assess acute responses to passive heat via lower-limb, hot-water immersion in patients with peripheral arterial disease (PAD) and healthy, elderly controls. Eleven patients with PAD (age 71 ± 6 yr, 7 male, 4 female) and 10 controls (age 72 ± 7 yr, 8 male, 2 female) underwent hot-water immersion (30-min waist-level immersion in 42.1 ± 0.6°C water). Before, during, and following immersion, brachial and popliteal artery diameter, blood flow, and shear stress were assessed using duplex ultrasound. Lower-limb perfusion was measured also using venous occlusion plethysmography and near-infrared spectroscopy. During immersion, shear rate increased (P < 0.0001) comparably between groups in the popliteal artery (controls: +183 ± 26%; PAD: +258 ± 54%) and brachial artery (controls: +117 ± 24%; PAD: +107 ± 32%). Lower-limb blood flow increased significantly in both groups, as measured from duplex ultrasound (>200%), plethysmography (>100%), and spectroscopy, while central and peripheral pulse-wave velocity decreased in both groups. Mean arterial blood pressure was reduced by 22 ± 9 mmHg (main effect P < 0.0001, interaction P = 0.60) during immersion, and remained 7 ± 7 mmHg lower 3 h afterward. In PAD, popliteal shear profiles and claudication both compared favorably with those measured immediately following symptom-limited walking. A 30-min hot-water immersion is a practical means of delivering heat therapy to PAD patients and healthy, elderly individuals to induce appreciable systemic (chronotropic and blood pressure lowering) and hemodynamic (upper and lower-limb perfusion and shear rate increases) responses.
Collapse
Affiliation(s)
- Kate N Thomas
- Department of Surgical Sciences, Dunedin School of Medicine, University of Otago, Dunedin, New Zealand; .,School of Physical Education, Sport and Exercise Sciences, University of Otago, Dunedin, New Zealand
| | - André M van Rij
- Department of Surgical Sciences, Dunedin School of Medicine, University of Otago, Dunedin, New Zealand
| | - Samuel J E Lucas
- Department of Physiology, University of Otago, Dunedin, New Zealand; and.,School of Sport, Exercise and Rehabilitation Sciences, University of Birmingham, Birmingham, United Kingdom
| | - James D Cotter
- School of Physical Education, Sport and Exercise Sciences, University of Otago, Dunedin, New Zealand
| |
Collapse
|
38
|
Sarkar C, Ganju RK, Pompili VJ, Chakroborty D. Enhanced peripheral dopamine impairs post-ischemic healing by suppressing angiotensin receptor type 1 expression in endothelial cells and inhibiting angiogenesis. Angiogenesis 2016; 20:97-107. [DOI: 10.1007/s10456-016-9531-8] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2016] [Accepted: 11/02/2016] [Indexed: 01/11/2023]
|
39
|
Henrique Rossi F, Puech-Leão P, Mitsuro Izukawa N, Pontes Junior SC, Massamitsu Kambara A, Mattos Barreto RB, Hassan Saleh M, Gomes Ferreira Petisco AC, Vasconcelos Oliveira LA. Color-Flow Duplex Hemodynamic Assessment of Runoff in Ischemic Lower Limb Revascularization. Vascular 2016; 14:149-55. [PMID: 16956487 DOI: 10.2310/6670.2006.00031] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
The objective of this study was to evaluate the existence of hemodynamic arterial flow correlation between preoperative duplex scanning (DS) and intraoperative direct outflow resistance (IDOR) measurements in ischemic lower limb revascularization. Sixty-eight ischemic lower limbs were submitted to preoperative DS. Anatomic and hemodynamic arterial characteristics of the outflow system were recorded, and the results were considered in the distal anastomosis placement site decision making. IDOR measurements were obtained at the same arterial segment, and Pearson's correlation coefficient test was performed to study the preoperative DS power in predicting the intraoperative outflow resistance. DS was technically satisfactory and helped define the distal anastomosis site in 93.2% of the cases (supragenicular popliteal artery, 19 [27.9%]; infragenicular popliteal artery, 10 [14.7%]; crural artery, 31 [57.4%]). A positive correlation could be found between preoperative DS and IDOR (0.450; p < .001). This correlation was particularly powerful in the crural artery (0.715; p < .001) when compared with the popliteal arterial segment (0.237; p = .192). Preoperative DS may help define the best distal arterial and outflow segment to be revascularized based on anatomic and hemodynamic parameters. There is a positive flow correlation between preoperative DS and IDOR that seems to be stronger in crural revascularization surgery.
Collapse
Affiliation(s)
- Fabio Henrique Rossi
- Department of Vascular Surgery, Dante Pazzanese Cardiovascular Institute, São Paulo, Brazil.
| | | | | | | | | | | | | | | | | |
Collapse
|
40
|
LeBlanc AJ, Hoying JB. Adaptation of the Coronary Microcirculation in Aging. Microcirculation 2016; 23:157-67. [DOI: 10.1111/micc.12264] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2015] [Accepted: 12/08/2015] [Indexed: 02/06/2023]
Affiliation(s)
- Amanda J. LeBlanc
- Department of Physiology; Cardiovascular Innovation Institute; University of Louisville; Louisville Kentucky USA
| | - James B. Hoying
- Department of Physiology; Cardiovascular Innovation Institute; University of Louisville; Louisville Kentucky USA
| |
Collapse
|
41
|
Miller V, Lin A, Kako F, Gabunia K, Kelemen S, Brettschneider J, Fridman G, Fridman A, Autieri M. Microsecond-pulsed dielectric barrier discharge plasma stimulation of tissue macrophages for treatment of peripheral vascular disease. PHYSICS OF PLASMAS 2015; 22:122005. [PMID: 26543345 PMCID: PMC4617731 DOI: 10.1063/1.4933403] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/13/2015] [Accepted: 06/17/2015] [Indexed: 05/19/2023]
Abstract
Angiogenesis is the formation of new blood vessels from pre-existing vessels and normally occurs during the process of inflammatory reactions, wound healing, tissue repair, and restoration of blood flow after injury or insult. Stimulation of angiogenesis is a promising and an important step in the treatment of peripheral artery disease. Reactive oxygen species have been shown to be involved in stimulation of this process. For this reason, we have developed and validated a non-equilibrium atmospheric temperature and pressure short-pulsed dielectric barrier discharge plasma system, which can non-destructively generate reactive oxygen species and other active species at the surface of the tissue being treated. We show that this plasma treatment stimulates the production of vascular endothelial growth factor, matrix metalloproteinase-9, and CXCL 1 that in turn induces angiogenesis in mouse aortic rings in vitro. This effect may be mediated by the direct effect of plasma generated reactive oxygen species on tissue.
Collapse
Affiliation(s)
- V Miller
- AJ Drexel Plasma Institute, Drexel University , Camden, New Jersey 08103, USA
| | - A Lin
- AJ Drexel Plasma Institute, Drexel University , Camden, New Jersey 08103, USA
| | - F Kako
- Department of Physiology, Independence Blue Cross Cardiovascular Research Center, Temple University School of Medicine , Philadelphia, Pennsylvania 19140, USA
| | - K Gabunia
- Department of Physiology, Independence Blue Cross Cardiovascular Research Center, Temple University School of Medicine , Philadelphia, Pennsylvania 19140, USA
| | - S Kelemen
- Department of Physiology, Independence Blue Cross Cardiovascular Research Center, Temple University School of Medicine , Philadelphia, Pennsylvania 19140, USA
| | - J Brettschneider
- AJ Drexel Plasma Institute, Drexel University , Camden, New Jersey 08103, USA
| | - G Fridman
- AJ Drexel Plasma Institute, Drexel University , Camden, New Jersey 08103, USA
| | - A Fridman
- AJ Drexel Plasma Institute, Drexel University , Camden, New Jersey 08103, USA
| | - M Autieri
- Department of Physiology, Independence Blue Cross Cardiovascular Research Center, Temple University School of Medicine , Philadelphia, Pennsylvania 19140, USA
| |
Collapse
|
42
|
Zhao C, Popel AS. Computational Model of MicroRNA Control of HIF-VEGF Pathway: Insights into the Pathophysiology of Ischemic Vascular Disease and Cancer. PLoS Comput Biol 2015; 11:e1004612. [PMID: 26588727 PMCID: PMC4654485 DOI: 10.1371/journal.pcbi.1004612] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2015] [Accepted: 10/15/2015] [Indexed: 01/08/2023] Open
Abstract
HRMs (hypoxia-responsive miRNAs) are a specific group of microRNAs that are regulated by hypoxia. Recent studies revealed that several HRMs including let-7 family miRNAs were highly induced in response to HIF (hypoxia-inducible factor) stabilization in hypoxia, and they potently participated in angiogenesis by targeting AGO1 (argonaute 1) and upregulating VEGF (vascular endothelial growth factor). Here we constructed a novel computational model of microRNA control of HIF-VEGF pathway in endothelial cells to quantitatively investigate the role of HRMs in modulating the cellular adaptation to hypoxia. The model parameters were optimized and the simulations based on these parameters were validated against several published in vitro experimental data. To advance the mechanistic understanding of oxygen sensing in hypoxia, we demonstrated that the rate of HIF-1α nuclear import substantially influences its stabilization and the formation of HIF-1 transcription factor complex. We described the biological feedback loops involving let-7 and AGO1 in which the impact of external perturbations were minimized; as a pair of master regulators when low oxygen tension was sensed, they coordinated the critical process of VEGF desuppression in a controlled manner. Prompted by the model-motivated discoveries, we proposed and assessed novel pathway-specific therapeutics that modulate angiogenesis by adjusting VEGF synthesis in tumor and ischemic cardiovascular disease. Through simulations that capture the complex interactions between miRNAs and miRNA-processing molecules, this model explores an innovative perspective about the distinctive yet integrated roles of different miRNAs in angiogenesis, and it will help future research to elucidate the dysregulated miRNA profiles found in cancer and various cardiovascular diseases. Cells living in a hypoxic environment secrete signals to stimulate new blood vessel growth, a process termed angiogenesis, to acquire more oxygen and nutrients. Hypoxia-inducible factor 1 (HIF-1) accumulates in hypoxia and expedites the release of pro-angiogenic cytokines such as vascular endothelial growth factor (VEGF), a prime inducer of angiogenesis. The intermediate signaling events connecting HIF-1 and VEGF are tightly controlled by microRNAs (miRs), which are endogenous, non-coding RNA molecules and powerful regulators in cancer and cardiovascular disease. Given the importance of angiogenesis in tumor development and post-ischemia reperfusion, it holds great basic research and therapeutic value to investigate how miRs modulate intracellular VEGF synthesis to control angiogenesis in hypoxia. We present a computational model that details the interactions between miRs and other key molecules which make up different hierarchies in HIF-miR-VEGF pathway. Based on simulation analysis, new potential therapies are introduced and tested in silico, from which the strategies that most effectively reduce VEGF synthesis in cancer, or enhance VEGF release in ischemic vascular disease are identified. We conclude that in hypoxia different miRs work consonantly to fine-tune the cellular adaptations; when a master miR alters its expression, dynamics of other miRs vary accordingly which together contribute to aberrant RNA/protein profiles observed in the pathophysiology of multiple diseases.
Collapse
Affiliation(s)
- Chen Zhao
- Department of Biomedical Engineering, School of Medicine, Johns Hopkins University, Baltimore, Maryland, United States of America
- * E-mail:
| | - Aleksander S. Popel
- Department of Biomedical Engineering, School of Medicine, Johns Hopkins University, Baltimore, Maryland, United States of America
| |
Collapse
|
43
|
Kang HM, Sohn I, Jung J, Jeong JW, Park C. Exendin-4 protects hindlimb ischemic injury by inducing angiogenesis. Biochem Biophys Res Commun 2015; 465:758-63. [PMID: 26299927 DOI: 10.1016/j.bbrc.2015.08.080] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2015] [Accepted: 08/18/2015] [Indexed: 11/19/2022]
Abstract
Exendin-4, an analog of glucagon-like peptide-1, has shown to have beneficial effects on endothelial function, and was recently approved for the treatment of diabetes. In previous studies, we showed that exendin-4 induces angiogenesis in in vitro and ex vivo assays; in this study, we assessed the proangiogenic effects of exendin-4 in vivo using a mouse hindlimb ischemia model. Treatment with exendin-4 for three days mitigated hindlimb and gastrocnemius muscle fiber necrosis. Hindlimb perfusion was determined using indocyanine green fluorescence dynamics that showed, significantly higher blood flow rate to the ischemic hindlimbs in an exendin-4-treated group. Immunohistochemistry assay showed that exendin-4 increased CD31-positive areas in the gastrocnemius muscle of ischemic limbs. Furthermore, treatment of the hindlimbs of ischemic mice with exendin-4 increased vascular endothelial growth factor (VEGF) and phospho-extracellular signal-related kinase (ERK) on western blot analysis. Our data demonstrate that exendin-4 prevents hindlimb ischemic injury by inducing vessels via VEGF angiogenic-related pathways. These findings suggest that exendin-4 has potential as a therapeutic agent for vascular diseases that stimulate angiogenesis.
Collapse
Affiliation(s)
- Hye-Min Kang
- Department of Anatomy and Neurobiology, Biomedical Science Institute, School of Medicine, Kyung Hee University, Republic of Korea
| | - Inkyung Sohn
- Department of Anatomy and Neurobiology, Biomedical Science Institute, School of Medicine, Kyung Hee University, Republic of Korea
| | - Junyang Jung
- Department of Anatomy and Neurobiology, Biomedical Science Institute, School of Medicine, Kyung Hee University, Republic of Korea
| | - Joo-Won Jeong
- Department of Anatomy and Neurobiology, Biomedical Science Institute, School of Medicine, Kyung Hee University, Republic of Korea
| | - Chan Park
- Department of Anatomy and Neurobiology, Biomedical Science Institute, School of Medicine, Kyung Hee University, Republic of Korea.
| |
Collapse
|
44
|
Hashimoto T, Chen L, Kimura H, Endler A, Koyama H, Miyata T, Shibasaki F, Watanabe T. Silencing of eIF3e promotes blood perfusion recovery after limb ischemia through stabilization of hypoxia-inducible factor 2α activity. J Vasc Surg 2015; 64:219-226.e3. [PMID: 25758454 DOI: 10.1016/j.jvs.2015.01.004] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2014] [Accepted: 01/09/2015] [Indexed: 10/23/2022]
Abstract
OBJECTIVE We previously observed that silencing of eukaryotic translation initiation factor 3 subunit e (eIF3e), a hypoxia-independent downregulator of hypoxia-inducible factor 2α (HIF-2α), led to neoangiogenesis by promoting HIF-2α activity under normoxic conditions. In the current study, we investigated whether temporary silencing of eIF3e in muscles affects blood flow recovery in a mouse ischemic limb model. METHODS eIF3e silencing was performed using small interfering RNA (siRNA), and changes in gene transcription and protein expression were analyzed in vitro using murine primary skeletal muscle myoblast and human primary skeletal muscle myoblast cell cultures. In unilateral femoral artery ligation experiments, eIF3e siRNA-expressing plasmids were injected into the muscles of BALB/c mice near the ligation sites, and tissue damage and loss of limb function were scored for 28 days while serial measurements of limb perfusions were performed with laser Doppler perfusion imaging. RESULTS Silencing of eIF3e in murine primary skeletal muscle myoblasts led to stabilization of HIF-2α and upregulation of angiogenic transcripts, including basic fibroblast growth factor and platelet-derived growth factor B (P < .05), and the supernatant of eIF3e-silenced human primary skeletal muscle myoblasts triggered the tube formation of human umbilical vein endothelial cells. The in vivo mouse model of hindlimb ischemia revealed that single intramuscular injections of eIF3e siRNA-expressing plasmids significantly enhanced perfusion of ischemia-damaged limbs (P < .05) at days 7 and 14 and functional recovery at days 7, 14, and 21 (P < .05). CONCLUSIONS eIF3e is an angiogenesis suppressor and may be a therapeutic target for promoting angiogenesis after ischemic injuries.
Collapse
Affiliation(s)
- Takuya Hashimoto
- Division of Vascular Surgery, Department of Surgery, Graduate School of Medicine, University of Tokyo, Tokyo, Japan
| | - Li Chen
- Department of Molecular Medical Research, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan
| | - Hideo Kimura
- Division of Vascular Surgery, Department of Surgery, Graduate School of Medicine, University of Tokyo, Tokyo, Japan
| | - Alexander Endler
- Department of Molecular Medical Research, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan
| | - Hiroyuki Koyama
- Division of Vascular Surgery, Department of Surgery, Graduate School of Medicine, University of Tokyo, Tokyo, Japan; Division of Tissue Engineering, University of Tokyo Hospital, Tokyo, Japan; Department of Vascular Surgery, Saitama Medical Center, Saitama Medical University, Saitama, Japan.
| | - Tetsuro Miyata
- Vascular Center, Sanno Hospital and Sanno Medical Center, Tokyo, Japan
| | - Futoshi Shibasaki
- Department of Molecular Medical Research, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan
| | - Toshiaki Watanabe
- Division of Vascular Surgery, Department of Surgery, Graduate School of Medicine, University of Tokyo, Tokyo, Japan
| |
Collapse
|
45
|
Aird AL, Nevitt CD, Christian K, Williams SK, Hoying JB, LeBlanc AJ. Adipose-derived stromal vascular fraction cells isolated from old animals exhibit reduced capacity to support the formation of microvascular networks. Exp Gerontol 2015; 63:18-26. [PMID: 25617825 DOI: 10.1016/j.exger.2015.01.044] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2014] [Revised: 01/14/2015] [Accepted: 01/20/2015] [Indexed: 01/30/2023]
Abstract
UNLABELLED Adipose-derived regenerative and stem cells, defined collectively as the stromal vascular fraction (SVF), support the formation of neovascular networks at the site of implantation. The effect of advancing age on SVF cell population effectiveness towards stimulated neovascularization was evaluated. METHODS SVF was enzymatically isolated from adipose of young (ySVF, 4 months) or old (oSVF, 24 months) Fisher-344 rats, combined with type I collagen and polymerized. Encapsulated SVF was implanted subcutaneously into young Rag1 mice for two or four weeks. Angiogenic function of age-dependent SVF was also extensively evaluated in vitro using standard assays. RESULTS In vitro studies indicated no difference in angiogenic function between ySVF and oSVF (viability, proliferation, migration, and tube-formation). At two weeks post-implantation, there was no age-related difference in percent apoptosis in explanted constructs. By four weeks post-implantation, oSVF implants displayed 36% less total vessels/mm(2), 43% less perfused vessels/mm(2), and exhibited greater percent apoptosis compared to ySVF (n ≥ 12). Blocking thrombospondin-1 (Thbs-1), a protein found to be highly expressed in oSVF but not ySVF, increased the percent of perfused vascular volume and vessel diameters in oSVF constructs after two weeks compared to oSVF implants treated with control antibody. CONCLUSIONS Advancing donor age reduces the potential of adipose-derived SVF to derive a mature microcirculation, but does not hinder initial angiogenesis. However, modulation of Thbs-1 may improve this outcome. This data suggests that greater pruning, dysfunctional structural adaptation and/or poor maturation with initiation of blood flow may occur in oSVF.
Collapse
Affiliation(s)
- Allison L Aird
- Cardiovascular Innovation Institute, Jewish Hospital and University of Louisville, Louisville, KY 40202, United States
| | - Christopher D Nevitt
- Cardiovascular Innovation Institute, Jewish Hospital and University of Louisville, Louisville, KY 40202, United States; Department of Biochemistry and Molecular Biology, Jewish Hospital and University of Louisville, Louisville, KY 40202, United States
| | - Katelyn Christian
- Cardiovascular Innovation Institute, Jewish Hospital and University of Louisville, Louisville, KY 40202, United States
| | - Stuart K Williams
- Cardiovascular Innovation Institute, Jewish Hospital and University of Louisville, Louisville, KY 40202, United States; Department of Physiology and Biophysics, Jewish Hospital and University of Louisville, Louisville, KY 40202, United States
| | - James B Hoying
- Cardiovascular Innovation Institute, Jewish Hospital and University of Louisville, Louisville, KY 40202, United States; Department of Physiology and Biophysics, Jewish Hospital and University of Louisville, Louisville, KY 40202, United States
| | - Amanda J LeBlanc
- Cardiovascular Innovation Institute, Jewish Hospital and University of Louisville, Louisville, KY 40202, United States; Department of Physiology and Biophysics, Jewish Hospital and University of Louisville, Louisville, KY 40202, United States.
| |
Collapse
|
46
|
Sakakibara N, Igarashi J, Takata M, Konishi R, Suzue N, Kato Y, Maruyama T, Tsukamoto I. Design, Synthesis, and Evaluation of Novel 2-Halogenated or Aminated Carbocyclic Oxetanocin a Analogs as Potential Angiogenic Agents. HETEROCYCLES 2015. [DOI: 10.3987/com-15-13293] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
|
47
|
Das A, Lenz SM, Awojoodu AO, Botchwey EA. Abluminal stimulation of sphingosine 1-phosphate receptors 1 and 3 promotes and stabilizes endothelial sprout formation. Tissue Eng Part A 2014; 21:202-13. [PMID: 25315888 DOI: 10.1089/ten.tea.2013.0744] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Local delivery of lipid mediators has become a promising new approach for therapeutic angiogenesis and regenerative medicine. In this study, we investigated how gradient stimulation (either abluminal/distal or luminal/proximal) of engineered microvessels with sphingosine 1-phosphate (S1P) receptor-subtype-targeted molecules affects endothelial sprout growth using a microfluidic device. Our studies show that distal stimulation of microvessels with FTY720, an S1P1/3 selective agonist, promotes both arterial and venular sprout growth, whereas proximal stimulation does not. Using novel pharmacological antagonists of S1P receptor subtypes, we further show that S1P3 functionality is necessary for VEGF-induced sprouting, and confirmed these findings ex vivo using a murine aortic ring assay from S1P3-deficient mice. S1P3 agonist stimulation enhanced vascular stability in both cell types via upregulation of the interendothelial junction protein VE-cadherin. Lastly, S1P3 activation under flow promoted endothelial sprouting and branching while decreasing migratory cell fate in the microfluidic device. We used an in vivo murine dorsal skinfold window chamber model to confirm S1P3's role in neovascular branching. Together, these data suggest that a distal transendothelial gradient of S1P1/3-targeted drugs is an effective technique for both enhancing and stabilizing capillary morphogenesis in angiogenic applications.
Collapse
Affiliation(s)
- Anusuya Das
- 1 Department of Orthopaedic Surgery, University of Virginia , Charlottesville, Virginia
| | | | | | | |
Collapse
|
48
|
Dubský M, Jirkovská A, Bem R, Fejfarová V, Pagacová L, Nemcová A, Sixta B, Chlupac J, Peregrin JH, Syková E, Jude EB. Comparison of the effect of stem cell therapy and percutaneous transluminal angioplasty on diabetic foot disease in patients with critical limb ischemia. Cytotherapy 2014; 16:1733-8. [DOI: 10.1016/j.jcyt.2014.08.010] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2014] [Revised: 08/12/2014] [Accepted: 08/18/2014] [Indexed: 01/26/2023]
|
49
|
Lee KB, Kim DI. Clinical application of stem cells for therapeutic angiogenesis in patients with peripheral arterial disease. Int J Stem Cells 2014; 2:11-7. [PMID: 24855515 DOI: 10.15283/ijsc.2009.2.1.11] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/08/2009] [Indexed: 12/16/2022] Open
Abstract
Peripheral arterial disease (PAD) may ultimately cause to the loss of the affected limb due to gangrene or infection. Some patients with PAD may have severe coexisting diseases and diffuse involvement of their distal arteries, and so they are poor candidates for revascularization procedures. Angiogenesis has recently been suggested to be a new emerging treatment strategy for patients with PAD. Angiogenesis is defined as the sprouting of new capillaries from pre-existing vascular structures; this process plays a major role in the development of collateral vessels in an ischemic limb. Yet, the exact mechanism of angiogenesis is currently poorly understood. It has been established that angiogenesis is initiated by hypoxia and it requires various pro-angiogenic factors such as vascular endothelial growth factor. Therapeutic angiogenesis is aimed at enhancing natural angiogenesis by the administration of the cells or genes that can trigger angiogenesis and this can lead to pain relief and wound healing by the development of collateral vessels. Most of the recent clinical trials have reported that stem cell therapy for promoting angiogenesis in patients with PAD improves the ischemic symptoms and enhances wound healing. However, there are several limitations to approve a standard treatment for PAD such as small sample size in several prevous studies, their diverse inclusion criteria and the lack of standard assessment methods for the safety and outcome. Therefore, multicenter, large-scale and randomized controlled studies are needed to prove the safety and efficacy of the clinically applying stem cells for therapeutic angiogenesis in patients with PAD.
Collapse
Affiliation(s)
- Kyung-Bok Lee
- Division of Vascular Surgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Dong-Ik Kim
- Division of Vascular Surgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| |
Collapse
|
50
|
Miao YL, Wu W, Li BW, Fang WW, Liu Y, Li L, Mi WD. Clinical Effectiveness of Gene Therapy on Critical Limb Ischemia. Vasc Endovascular Surg 2014; 48:372-7. [PMID: 24951292 DOI: 10.1177/1538574414539397] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Therapeutic angiogenesis using gene therapy is a novel strategy for the treatment of critical limb ischemia (CLI). We conducted a meta-analysis to evaluate the efficacy and safety of gene therapy for the treatment of CLI with no option of revascularization. Randomized placebo controlled trials of gene therapy on CLI were identified by searching PubMed (from 1990 to October 2013) and EMBASE (from 1990 to October 2013). Five eligible studies were selected for the meta-analysis. Among these studies, a total of 425 patients received gene therapy of either fibroblast growth factor 1 or hepatocyte growth factor, and 365 patients were given placebo. No statistical differences were observed between the 2 groups in major amputation or death at 1 year (risk ratio [RR], 0.83; 95% confidence interval [CI], 0.51-1.39; P = .48) and wound healing at 6 months (RR, 1.55; 95% CI, 0.73-3.28; P = .25). Gene therapy had similar occurrence of serious adverse events as control (RR, 1.05; 95% CI, 0.97-1.14; P = .23).
Collapse
Affiliation(s)
- Yu-Liang Miao
- Anesthesia and Operation Center, Chinese PLA General Hospital, Beijing 100853, China
- Department of Anesthesiology, PLA No. 306 Hospital, Beijing, China
| | - Wei Wu
- Department of Anesthesiology, PLA No. 306 Hospital, Beijing, China
| | - Bao-Wei Li
- Department of Anesthesiology, PLA No. 306 Hospital, Beijing, China
| | - Wei-Wu Fang
- Department of Anesthesiology, PLA No. 306 Hospital, Beijing, China
| | - Yan Liu
- Department of Anesthesiology, PLA No. 306 Hospital, Beijing, China
| | - Lu Li
- Department of Cardiothoracic Surgery, PLA No. 306 Hospital, Beijing, China
| | - Wei-Dong Mi
- Anesthesia and Operation Center, Chinese PLA General Hospital, Beijing 100853, China
| |
Collapse
|