1
|
Nishiyama A, Shigematsu K, Koyama H, Hoshina K, Miyata T. Anatomical Courses of Collateral Circulations in Patients with Infrainguinal Chronic Lower-Extremity Arterial Occlusive Disease. Ann Vasc Surg 2025; 117:36-43. [PMID: 40216018 DOI: 10.1016/j.avsg.2025.03.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2025] [Revised: 03/30/2025] [Accepted: 03/31/2025] [Indexed: 05/10/2025]
Abstract
BACKGROUND This study aimed to identify the anatomical course of collateral vessels in chronic lower-extremity arterial occlusive disease to optimize intramuscular injection sites for angiogenesis therapy. METHODS We retrospectively analyzed 35 limbs with superficial femoral artery (SFA) occlusion and 17 limbs with 3 crural artery occlusion using 1-mm slice contrast-enhanced computed tomography. Collateral vessels (≥1 mm) connecting the common femoral to popliteal arteries, and popliteal to foot arteries were identified. Donor and recipient arteries, and vessel courses were documented. RESULTS In SFA occlusion, 49 collateral vessels were identified. The deep femoral artery was the sole donor. Recipient arteries were predominantly the lateral (94%) and medial (6%) superior genicular arteries. Seventy-one percent (35 of 49) of collaterals ran within the short head of the biceps femoris. In crural artery occlusion, 17 collaterals were found. Donor arteries included the peroneal (29%), posterior tibial (24%), and combinations thereof. Recipient arteries were the anterior tibial (53%), plantar (29%), and dorsalis pedis (18%). All collaterals coursed through the soleus muscle, with 35% traversing the posterior tibial muscle. CONCLUSION Collateral vessels in chronic lower-extremity arterial occlusive disease exhibit preferential development within specific muscles. In SFA occlusion, collaterals develop predominantly within the short head of the biceps femoris, while in crural artery occlusion, collaterals develop within the soleus muscle. These findings suggest that targeted intramuscular injections, guided by anatomical knowledge of collateral pathways, may enhance angiogenesis therapy efficacy.
Collapse
Affiliation(s)
- Ayako Nishiyama
- Department of Vascular Surgery, The University of Tokyo, Tokyo, Japan; Department of Vascular Surgery, Saiseikai Kawaguchi General Hospital, Saitama, Japan.
| | - Kunihiro Shigematsu
- Department of Vascular Surgery, The University of Tokyo, Tokyo, Japan; Department of Vascular Surgery, International University of Health and Welfare, Mita General Hospital, Tokyo, Japan
| | - Hiroyuki Koyama
- Department of Vascular Surgery, The University of Tokyo, Tokyo, Japan; Department of Surgery, Airi Hospital, Tokyo, Japan
| | - Katsuyuki Hoshina
- Department of Vascular Surgery, The University of Tokyo, Tokyo, Japan
| | - Tetsuro Miyata
- Department of Vascular Surgery, The University of Tokyo, Tokyo, Japan; Japan Medical Safety Research Organization, Tokyo, Japan
| |
Collapse
|
2
|
Webster KA. Translational Relevance of Advanced Age and Atherosclerosis in Preclinical Trials of Biotherapies for Peripheral Artery Disease. Genes (Basel) 2024; 15:135. [PMID: 38275616 PMCID: PMC10815340 DOI: 10.3390/genes15010135] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2023] [Revised: 01/08/2024] [Accepted: 01/17/2024] [Indexed: 01/27/2024] Open
Abstract
Approximately 6% of adults worldwide suffer from peripheral artery disease (PAD), primarily caused by atherosclerosis of lower limb arteries. Despite optimal medical care and revascularization, many PAD patients remain symptomatic and progress to critical limb ischemia (CLI) and risk major amputation. Delivery of pro-angiogenic factors as proteins or DNA, stem, or progenitor cells confers vascular regeneration and functional recovery in animal models of CLI, but the effects are not well replicated in patients and no pro-angiogenic biopharmacological procedures are approved in the US, EU, or China. The reasons are unclear, but animal models that do not represent clinical PAD/CLI are implicated. Consequently, it is unclear whether the obstacles to clinical success lie in the toxic biochemical milieu of human CLI, or in procedures that were optimized on inappropriate models. The question is significant because the former case requires abandonment of current strategies, while the latter encourages continued optimization. These issues are discussed in the context of relevant preclinical and clinical data, and it is concluded that preclinical mouse models that include age and atherosclerosis as the only comorbidities that are consistently present and active in clinical trial patients are necessary to predict clinical success. Of the reviewed materials, no biopharmacological procedure that failed in clinical trials had been tested in animal models that included advanced age and atherosclerosis relevant to PAD/CLI.
Collapse
Affiliation(s)
- Keith A. Webster
- Vascular Biology Institute, University of Miami, Miami, FL 33146, USA;
- Department of Ophthalmology, Baylor College of Medicine, Houston, TX 77030, USA
| |
Collapse
|
3
|
Shah AJ, Pavlatos N, Kalra DK. Preventive Therapies in Peripheral Arterial Disease. Biomedicines 2023; 11:3157. [PMID: 38137379 PMCID: PMC10741180 DOI: 10.3390/biomedicines11123157] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Revised: 11/16/2023] [Accepted: 11/21/2023] [Indexed: 12/24/2023] Open
Abstract
Atherosclerosis, while initially deemed a bland proliferative process, is now recognized as a multifactorial-lipoprotein-mediated inflammation-driven pathway. With the rising incidence of atherosclerotic disease of the lower extremity arteries, the healthcare burden and clinical morbidity and mortality due to peripheral artery disease (PAD) are currently escalating. With a healthcare cost burden of over 21 billion USD and 200 million patients afflicted worldwide, accurate knowledge regarding the pathophysiology, presentation, and diagnosis of the disease is crucial. The role of lipoproteins and their remnants in atherosclerotic vessel occlusion and plaque formation and progression has been long established. This review paper discusses the epidemiology, pathophysiology, and presentation of PAD. PAD has been repeatedly noted to portend to poor cardiovascular and limb outcomes. We discuss major therapeutic avenues for the prevention of major cardiovascular adverse events and major limb adverse events in patients with PAD.
Collapse
Affiliation(s)
- Aangi J. Shah
- Department of Internal Medicine, University of Louisville School of Medicine, Louisville, KY 40202, USA; (A.J.S.); (N.P.)
| | - Nicholas Pavlatos
- Department of Internal Medicine, University of Louisville School of Medicine, Louisville, KY 40202, USA; (A.J.S.); (N.P.)
| | - Dinesh K. Kalra
- Division of Cardiology, University of Louisville School of Medicine, Louisville, KY 40202, USA
| |
Collapse
|
4
|
Morishita R, Shimamura M, Takeya Y, Nakagami H, Chujo M, Ishihama T, Yamada E, Rakugi H. Combined Analysis of Clinical Data on HGF Gene Therapy to Treat Critical Limb Ischemia in Japan. Curr Gene Ther 2021; 20:25-35. [PMID: 32416690 DOI: 10.2174/1566523220666200516171447] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2020] [Revised: 04/15/2020] [Accepted: 04/16/2020] [Indexed: 01/28/2023]
Abstract
OBJECTIVE The objective of this combined analysis of data from clinical trials in Japan, using naked plasmid DNA encoding hepatocyte growth factor (HGF), was to document the safety and efficacy of intramuscular HGF gene therapy in patients with critical limb ischemia (CLI). METHODS HGF gene transfer was performed in 22 patients with CLI in a single-center open trial at Osaka University; 39 patients in a randomized, placebo-controlled, multi-center phase III trial, 10 patients with Buerger's disease in a multi-center open trial; and 6 patients with CLI in a multi-center open trial using 2 or 3 intramuscular injections of naked HGF plasmid at 2 or 4 mg. Resting pain on a visual analogue scale (VAS) and wound healing as primary endpoints were evaluated at 12 weeks after the initial injection. Serious adverse events caused by gene transfer were detected in 7 out of 77 patients (9.09%). Only one patient experienced peripheral edema (1.30%), in contrast to those who had undergone treatment with VEGF. At 12 weeks after gene transfer, combined evaluation of VAS and ischemic ulcer size demonstrated a significant improvement in HGF gene therapy group as compared to the placebo group (P=0.020). RESULTS The long-term analysis revealed a sustained decrease in the size of ischemic ulcer in HGF gene therapy group. In addition, VAS score over 50 mm at baseline (total 27 patients) demonstrated a tendency (P=0.059), but not significant enough, to improve VAS score in HGF gene therapy as compared to the placebo group. CONCLUSION The findings indicated that intramuscular injection of naked HGF plasmid tended to improve the resting pain and significantly decreased the size of the ischemic ulcer in the patients with CLI who did not have any alternative therapy, such as endovascular treatment (EVT) or bypass graft surgery. An HGF gene therapy product, CollategeneTM, was recently launched with conditional and time-limited approval in Japan to treat ischemic ulcer in patients with CLI. Further clinical trials would provide new therapeutic options for patients with CLI.
Collapse
Affiliation(s)
- Ryuichi Morishita
- Department of Clinical Gene Therapy, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan
| | - Munehisa Shimamura
- Department of Health Development and Medicine, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan
| | - Yasushi Takeya
- Department of Geriatric Medicine, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan
| | - Hironori Nakagami
- Department of Health Development and Medicine, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan
| | | | | | | | - Hiromi Rakugi
- Department of Geriatric Medicine, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan
| |
Collapse
|
5
|
Nakae T, Obana M, Maeda T, Ikeda A, Miyazaki H, Tanaka S, Maeda M, Yamashita K, Terai K, Obika S, Fujio Y. Title: Gene transfer by pyro-drive jet injector is a novel therapeutic approach for muscle diseases. Gene 2021; 788:145664. [PMID: 33887371 DOI: 10.1016/j.gene.2021.145664] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Revised: 03/19/2021] [Accepted: 04/15/2021] [Indexed: 01/23/2023]
Abstract
The angiogenic gene therapy is an attractive approach for the treatment of ischemic muscle diseases, including peripheral arterial disease and ischemic heart diseases. Although a variety of gene transfer methods have been developed, the efficiency of gene transfer is still limited. We have been developing the needleless high-energy bioinjector device, Pyro-drive Jet Injector (PJI), based on pyrotechnics using a combination of ignition powder and gunpowder, however, the utility of PJI in gene transfer into muscle tissues remains unclear. pcDNA3.1 plasmid containing Flag was injected to the thigh muscles of C57BL/6J mice using PJI or needle, as a control. Histological analysis demonstrated that the protein expression of Flag was observed in a wider range in PJI group than in needle group. To assess the validity of PJI for gene therapy, pcDNA3.1-human fibroblast growth factor 2 (FGF2), which has angiogenic activity and tissue protective properties, was injected into the ischemic thigh muscles with PJI or needle. ELISA assay revealed that the protein expression of FGF2 was increased in the thigh muscle tissues by PJI-mediated gene delivery. Significantly, histological analyses revealed that muscle fiber cross-sectional area and the number of endothelial marker CD31 (+) cells was increased in ischemic hind-limb tissues of the PJI-FGF2 group but not in those of needle-FGF2 group. To expand the applicability of the PJI-mediated gene transfer, pcDNA3.1-venus plasmid was injected into murine hearts with PJI or needle. PJI method was successful in gene transfer into murine hearts, especially into cardiomyocytes, with high efficiency when compared to needle method. Collectively, the non-needle, non-liposomal and non-viral gene transfer by PJI could be a novel therapeutic approach for muscle diseases.
Collapse
Affiliation(s)
- Takafumi Nakae
- Laboratory of Clinical Science and Biomedicine, Graduate School of Pharmaceutical Sciences, Osaka University, Japan
| | - Masanori Obana
- Laboratory of Clinical Science and Biomedicine, Graduate School of Pharmaceutical Sciences, Osaka University, Japan; Radioisotope Research Center, Institute for Radiation Sciences, Osaka University, Japan; Integrated Frontier Research for Medical Science Division, Institute for Open and Transdisciplinary Research Initiatives (OTRI), Osaka University, Japan; Global Center for Medical Engineering and Informatics (MEI), Osaka University, Japan.
| | - Takahiro Maeda
- Laboratory of Clinical Science and Biomedicine, Graduate School of Pharmaceutical Sciences, Osaka University, Japan
| | - Akari Ikeda
- Laboratory of Clinical Science and Biomedicine, Graduate School of Pharmaceutical Sciences, Osaka University, Japan
| | - Hiroshi Miyazaki
- Medical Device Research, Industry Business Unit, Daicel Corporation, Japan
| | - Shota Tanaka
- Laboratory of Clinical Science and Biomedicine, Graduate School of Pharmaceutical Sciences, Osaka University, Japan
| | - Makiko Maeda
- Laboratory of Clinical Pharmacology and Therapeutics, Graduate School of Pharmaceutical Sciences, Osaka University, Japan.
| | - Kunihiko Yamashita
- Medical Device Research, Industry Business Unit, Daicel Corporation, Japan; Department of Device Application for Molecular Therapeutics, Graduate School of Medicine, Osaka University, Japan
| | - Kazuhiro Terai
- Medical Device Research, Industry Business Unit, Daicel Corporation, Japan
| | - Satoshi Obika
- Laboratory of Bioorganic Chemistry, Graduate School of Pharmaceutical Sciences, Osaka University, Japan
| | - Yasushi Fujio
- Laboratory of Clinical Science and Biomedicine, Graduate School of Pharmaceutical Sciences, Osaka University, Japan; Integrated Frontier Research for Medical Science Division, Institute for Open and Transdisciplinary Research Initiatives (OTRI), Osaka University, Japan; Laboratory of Clinical Pharmacology and Therapeutics, Graduate School of Pharmaceutical Sciences, Osaka University, Japan.
| |
Collapse
|
6
|
Forster R, Liew A, Bhattacharya V, Shaw J, Stansby G, Cochrane Vascular Group. Gene therapy for peripheral arterial disease. Cochrane Database Syst Rev 2018; 10:CD012058. [PMID: 30380135 PMCID: PMC6517203 DOI: 10.1002/14651858.cd012058.pub2] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
BACKGROUND Peripheral arterial disease (PAD), caused by narrowing of the arteries in the limbs, is increasing in incidence and prevalence as our population is ageing and as diabetes is becoming more prevalent. PAD can cause pain in the limbs while walking, known as intermittent claudication, or can be more severe and cause pain while at rest, ulceration, and ultimately gangrene and limb loss. This more severe stage of PAD is known as 'critical limb ischaemia'. Treatments for PAD include medications that help to reduce the increased risk of cardiovascular events and help improve blood flow, as well as endovascular or surgical repair or bypass of the blocked arteries. However, many people are unresponsive to medications and are not suited to surgical or endovascular treatment, leaving amputation as the last option. Gene therapy is a novel approach in which genetic material encoding for proteins that may help increase revascularisation is injected into the affected limbs of patients. This type of treatment has been shown to be safe, but its efficacy, especially regarding ulcer healing, effects on quality of life, and other symptomatic outcomes remain unknown. OBJECTIVES To assess the effects of gene therapy for symptomatic peripheral arterial disease. SEARCH METHODS The Cochrane Vascular Information Specialist searched Cochrane CENTRAL, the Cochrane Vascular Specialised Register, MEDLINE Ovid, Embase Ovid, CINAHL, and AMED, along with trials registries (all searched 27 November 2017). We also checked reference lists of included studies and systematic reviews for further studies. SELECTION CRITERIA We included randomised and quasi-randomised studies that evaluated gene therapy versus no gene therapy in people with PAD. We excluded studies that evaluated direct growth hormone treatment or cell-based treatments. DATA COLLECTION AND ANALYSIS Two review authors independently selected studies, performed quality assessment, and extracted data from the included studies. We collected pertinent information on each study, as well as data for the outcomes of amputation-free survival, ulcer healing, quality of life, amputation, all-cause mortality, ankle brachial index, symptom scores, and claudication distance. MAIN RESULTS We included in this review a total of 17 studies with 1988 participants (evidence current until November 2017). Three studies limited their inclusion to people with intermittent claudication, 12 limited inclusion to people with varying levels of critical limb ischaemia, and two included people with either condition. Study investigators evaluated many different types of gene therapies, using different protocols. Most studies evaluated growth factor-encoding gene therapy, with six studies using vascular endothelial growth factor (VEGF)-encoding genes, four using hepatocyte growth factor (HGF)-encoding genes, and three using fibroblast growth factor (FGF)-encoded genes. Two studies evaluated hypoxia-inducible factor 1-alpha (HIF-1α) gene therapy, one study used a developmental endothelial locus-1 gene therapy, and the final study evaluated a stromal cell-derived factor-1 (SDF-1) gene therapy. Most studies reported outcomes after 12 months of follow-up, but follow-up ranged from three months to two years.Overall risk of bias varied between studies, with many studies not providing sufficient detail for adequate determination of low risk of bias for many domains. Two studies did not utilise a placebo control, leading to risk of performance bias. Several studies reported in previous protocols or in their Methods sections that they would report on certain outcomes for which no data were then reported, increasing risk of reporting bias. All included studies reported sponsorships from corporate entities that led to unclear risk of other bias. The overall quality of evidence ranged from moderate to very low, generally as the result of heterogeneity and imprecision, with few or no studies reporting on outcomes.Evidence suggests no clear differences for the outcomes of amputation-free survival, major amputation, and all-cause mortality between those treated with gene therapy and those not receiving this treatment (all moderate-quality evidence). Low-quality evidence suggests improvement in complete ulcer healing with gene therapy (odds ratio (OR) 2.16, 95% confidence interval (CI) 1.02 to 4.59; P = 0.04). We could not combine data on quality of life and can draw no conclusions at this time regarding this outcome (very low-quality evidence). We included one study in the meta-analysis for ankle brachial index, which showed no clear differences between treatments, but we can draw no overall association (low-quality evidence). We combined in a meta-analysis pain symptom scores as assessed by visual analogue scales from two studies and found no clear differences between treatment groups (very low-quality evidence). We carried out extensive subgroup analyses by PAD classification, dosage schedule, vector type, and gene used but identified no substantial differences. AUTHORS' CONCLUSIONS Moderate-quality evidence shows no clear differences in amputation-free survival, major amputation, and all-cause mortality between those treated with gene therapy and those not receiving gene therapy. Some evidence suggests that gene therapy may lead to improved complete ulcer healing, but this outcome needs to be explored with improved reporting of the measure, such as decreased ulcer area in cm², and better description of ulcer types and healing. Further standardised data that are amenable to meta-analysis are needed to evaluate other outcomes such as quality of life, ankle brachial index, symptom scores, and claudication distance.
Collapse
Affiliation(s)
- Rachel Forster
- University of EdinburghUsher Institute of Population Health Sciences and InformaticsEdinburghUKEH8 9AG
| | - Aaron Liew
- Newcastle UniversityInstitute of Cellular Medicine4th Floor, William Leech BuildingFramlington PlaceNewcastle upon TyneUKNE2 4HH
- National University of Ireland Galway (NUIG), Portiuncula University Hospital & Galway University Hospital, Saolta University Health Care GroupGalwayIreland
| | - Vish Bhattacharya
- Queen Elizabeth HospitalDepartment of General and Vascular SurgeryQueen Elizabeth AvenueSheriff HillGatesheadTyne and WearUKNE9 6SX
| | - James Shaw
- Newcastle UniversityInstitute of Cellular Medicine4th Floor, William Leech BuildingFramlington PlaceNewcastle upon TyneUKNE2 4HH
| | - Gerard Stansby
- Freeman HospitalNorthern Vascular CentreNewcastle upon TyneUKNE7 7DN
| | | |
Collapse
|
7
|
Makarevich PI, Dergilev KV, Tsokolaeva ZI, Boldyreva MA, Shevchenko EK, Gluhanyuk EV, Gallinger JO, Menshikov MY, Parfyonova YV. Angiogenic and pleiotropic effects of VEGF165 and HGF combined gene therapy in a rat model of myocardial infarction. PLoS One 2018; 13:e0197566. [PMID: 29787588 PMCID: PMC5963747 DOI: 10.1371/journal.pone.0197566] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2017] [Accepted: 05/05/2018] [Indexed: 12/31/2022] Open
Abstract
Since development of plasmid gene therapy for therapeutic angiogenesis by J. Isner this approach was an attractive option for ischemic diseases affecting large cohorts of patients. However, first placebo-controlled clinical trials showed its limited efficacy questioning further advance to practice. Thus, combined methods using delivery of several angiogenic factors got into spotlight as a way to improve outcomes. This study provides experimental proof of concept for a combined approach using simultaneous delivery of VEGF165 and HGF genes to alleviate consequences of myocardial infarction (MI). However, recent studies suggested that angiogenic growth factors have pleiotropic effects that may contribute to outcome so we expanded focus of our work to investigate potential mechanisms underlying action of VEGF165, HGF and their combination in MI. Briefly, Wistar rats underwent coronary artery ligation followed by injection of plasmid bearing VEGF165 or HGF or mixture of these. Histological assessment showed decreased size of post-MI fibrosis in both—VEGF165- or HGF-treated animals yet most prominent reduction of collagen deposition was observed in VEGF165+HGF group. Combined delivery group rats were the only to show significant increase of left ventricle (LV) wall thickness. We also found dilatation index improved in HGF or VEGF165+HGF treated animals. These effects were partially supported by our findings of c-kit+ cardiac stem cell number increase in all treated animals compared to negative control. Sporadic Ki-67+ mature cardiomyocytes were found in peri-infarct area throughout study groups with comparable effects of VEGF165, HGF and their combination. Assessment of vascular density in peri-infarct area showed efficacy of both–VEGF165 and HGF while combination of growth factors showed maximum increase of CD31+ capillary density. To our surprise arteriogenic response was limited in HGF-treated animals while VEGF165 showed potent positive influence on a-SMA+ blood vessel density. The latter hinted to evaluate infiltration of monocytes as they are known to modulate arteriogenic response in myocardium. We found that monocyte infiltration was driven by VEGF165 and reduced by HGF resulting in alleviation of VEGF-stimulated monocyte taxis after combined delivery of these 2 factors. Changes of monocyte infiltration were concordant with a-SMA+ arteriole density so we tested influence of VEGF165 or HGF on endothelial cells (EC) that mediate angiogenesis and inflammatory response. In a series of in vitro experiments we found that VEGF165 and HGF regulate production of inflammatory chemokines by human EC. In particular MCP-1 levels changed after treatment by recombinant VEGF, HGF or their combination and were concordant with NF-κB activation and monocyte infiltration in corresponding groups in vivo. We also found that both–VEGF165 and HGF upregulated IL-8 production by EC while their combination showed additive type of response reaching peak values. These changes were HIF-2 dependent and siRNA-mediated knockdown of HIF-2α abolished effects of VEGF165 and HGF on IL-8 production. To conclude, our study supports combined gene therapy by VEGF165 and HGF to treat MI and highlights neglected role of pleiotropic effects of angiogenic growth factors that may define efficacy via regulation of inflammatory response and endothelial function.
Collapse
Affiliation(s)
- Pavel I. Makarevich
- Laboratory of Angiogenesis, National Medical Research Center of Cardiology, Moscow, Russia
- Laboratory of Gene and Cell Therapy, Institute of Regenerative Medicine, Lomonosov Moscow State University, Moscow, Russia
- * E-mail:
| | - Konstantin V. Dergilev
- Laboratory of Angiogenesis, National Medical Research Center of Cardiology, Moscow, Russia
| | - Zoya I. Tsokolaeva
- Laboratory of Angiogenesis, National Medical Research Center of Cardiology, Moscow, Russia
- Laboratory of Gene and Cell Technology, Faculty of Medicine, Lomonosov Moscow State University, Moscow, Russia
| | - Maria A. Boldyreva
- Laboratory of Angiogenesis, National Medical Research Center of Cardiology, Moscow, Russia
- Laboratory of Gene and Cell Technology, Faculty of Medicine, Lomonosov Moscow State University, Moscow, Russia
| | - Evgeniy K. Shevchenko
- Laboratory of Angiogenesis, National Medical Research Center of Cardiology, Moscow, Russia
- Laboratory of Gene and Cell Technology, Faculty of Medicine, Lomonosov Moscow State University, Moscow, Russia
| | - Evgeny V. Gluhanyuk
- Laboratory of Angiogenesis, National Medical Research Center of Cardiology, Moscow, Russia
| | - Julia O. Gallinger
- Laboratory of Angiogenesis, National Medical Research Center of Cardiology, Moscow, Russia
| | - Mikhail Yu. Menshikov
- Laboratory of Angiogenesis, National Medical Research Center of Cardiology, Moscow, Russia
- Laboratory of Gene and Cell Technology, Faculty of Medicine, Lomonosov Moscow State University, Moscow, Russia
| | - Yelena V. Parfyonova
- Laboratory of Angiogenesis, National Medical Research Center of Cardiology, Moscow, Russia
- Laboratory of Gene and Cell Technology, Faculty of Medicine, Lomonosov Moscow State University, Moscow, Russia
| |
Collapse
|
8
|
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
|
9
|
Zhou HM, Liu F, Yang AG, Guo YQ, Zhou YR, Gu YQ, Yan BY, Li QH. Efficacy, safety and influencing factors of intra-calf muscular injection of bone marrow mononuclear cells in the treatment of type 2 diabetes mellitus-induced lower extremity vascular disease. Exp Ther Med 2017; 14:5177-5185. [PMID: 29201234 DOI: 10.3892/etm.2017.5193] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2016] [Accepted: 03/17/2017] [Indexed: 11/06/2022] Open
Abstract
The efficacy, safety and impact of vascular endothelial growth factor (VEGF) and basic fibroblast growth factor (bFGF) associated with the intra-calf muscular injection of bone marrow mononuclear cells (BMMCs) in the treatment of type 2 diabetes mellitus (T2DM)-induced lower extremity vascular disease (LEVD) were evaluated. Patients with T2DM-LEVD were randomly divided into a control group and BMMCs group to assess the efficacy and safety of the treatment; serum VEGF and bFGF levels were detected. The BMMCs group was divided into superior genicular artery (SGA) and inferior genicular artery (IGA) subgroups as well as low-dose and high-dose subgroups for the comparison of efficacy indices. The BMMCs group exhibited significantly improved indices (P<0.05) compared with the control group and no fatalities or cancer occurred. There were no significant changes in serum VEGF and bFGF levels (P>0.05). The claudication distance in the IGA subgroup was significantly greater that in the SGA subgroup (P<0.05); the low-dose subgroup and the high-dose subgroup did not demonstrate any significant differences in each index (P>0.05). BMMC treatment for T2DM-LEVD was found to be safe and effective and had no significant impact on serum VEGF and bFGF levels in the short term; However, the degree of LEVD may affect its efficacy.
Collapse
Affiliation(s)
- Hui-Min Zhou
- Department of Endocrinology, The First Hospital of Hebei Medical University, Shijiazhuang, Hebei 050031, P.R. China
| | - Fan Liu
- Department of Endocrinology, The First Hospital of Hebei Medical University, Shijiazhuang, Hebei 050031, P.R. China
| | - Ai-Ge Yang
- Department of Endocrinology, The First Hospital of Hebei Medical University, Shijiazhuang, Hebei 050031, P.R. China
| | - Yu-Qing Guo
- Department of Endocrinology, The First Hospital of Hebei Medical University, Shijiazhuang, Hebei 050031, P.R. China
| | - Ya-Ru Zhou
- Department of Endocrinology, The Third Hospital of Hebei Medical University, Shijiazhuang, Hebei 050000, P.R. China
| | - Yong-Quan Gu
- Department of Vascular Surgery, Xuanwu Hospital, Capital Medical University, Beijing 100053, P.R. China
| | - Bao-Yong Yan
- Cell Therapy Laboratory, The First Hospital of Hebei Medical University, Shijiazhuang, Hebei 050031, P.R. China
| | - Quan-Hai Li
- Cell Therapy Laboratory, The First Hospital of Hebei Medical University, Shijiazhuang, Hebei 050031, P.R. China
| |
Collapse
|
10
|
Nishijima A, Yamamoto N, Yoshida R, Yanagibayashi S, Takikawa M, Hayasaka R, Maruyama E, Nishijima J, Sekido M. Maggot debridement therapy with a direct dressing can cause compression injuries in patients with chronic limb ischemia. CASE REPORTS IN PLASTIC SURGERY AND HAND SURGERY 2017; 4:84-88. [PMID: 28971112 PMCID: PMC5613910 DOI: 10.1080/23320885.2017.1373596] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/26/2017] [Accepted: 08/28/2017] [Indexed: 11/02/2022]
Abstract
While there are no reports regarding dressing-associated iatrogenic skin ulcer as an adverse event of maggot debridement therapy (MDT), MDT is clinically used on patients with critical limb ischaemia with dermal fragility. Herein we report causes and counter measures for a case of iatrogenic skin ulcer induced by MDT dressing.
Collapse
Affiliation(s)
- Akio Nishijima
- Department of Plastic and Reconstructive Surgery, New Tokyo HospitalMatsudoChibaJapan
| | - Naoto Yamamoto
- Department of Plastic and Reconstructive Surgery, New Tokyo HospitalMatsudoChibaJapan
| | - Ryuichi Yoshida
- Department of Plastic and Reconstructive Surgery, New Tokyo HospitalMatsudoChibaJapan
| | - Satoshi Yanagibayashi
- Department of Plastic and Reconstructive Surgery, New Tokyo HospitalMatsudoChibaJapan
| | - Megumi Takikawa
- Department of Plastic and Reconstructive Surgery, New Tokyo HospitalMatsudoChibaJapan
| | - Rie Hayasaka
- Department of Plastic and Reconstructive Surgery, New Tokyo HospitalMatsudoChibaJapan
| | - Eri Maruyama
- Department of Plastic and Reconstructive Surgery, New Tokyo HospitalMatsudoChibaJapan
| | - Junko Nishijima
- Department of Plastic and Reconstructive Surgery, New Tokyo HospitalMatsudoChibaJapan
| | - Mitsuru Sekido
- Department of Plastic and Reconstructive Surgery, University of TsukubaTsukubaIbarakiJapan
| |
Collapse
|
11
|
Kanayasu-Toyoda T, Ishii-Watabe A, Kikuchi Y, Kitagawa H, Suzuki H, Tamura H, Tada M, Suzuki T, Mizuguchi H, Yamaguchi T. Occludin as a functional marker of vascular endothelial cells on tube-forming activity. J Cell Physiol 2017; 233:1700-1711. [PMID: 28681912 DOI: 10.1002/jcp.26082] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2017] [Accepted: 07/05/2017] [Indexed: 01/03/2023]
Abstract
Cell therapy using endothelial progenitor cells (EPCs) is a promising strategy for the treatment of ischemic diseases. Two types of EPCs have been identified: early EPCs and late EPCs. Late EPCs are able to form tube structure by themselves, and have a high proliferative ability. The functional marker(s) of late EPCs, which relate to their therapeutic potential, have not been fully elucidated. Here we compared the gene expression profiles of several human cord blood derived late EPC lines which exhibit different tube formation activity, and we observed that the expression of occludin (OCLN) in these lines correlated with the tube formation ability, suggesting that OCLN is a candidate functional marker of late EPCs. When OCLN was knocked down by transfecting siRNA, the tube formation on Matrigel, the S phase + G2 /M phase in the cell cycle, and the spheroid-based sprouting of late EPCs were markedly reduced, suggesting the critical role of OCLN in tube formation, sprouting, and proliferation. These results indicated that OCLN plays a novel role in neovascularization and angiogenesis.
Collapse
Affiliation(s)
- Toshie Kanayasu-Toyoda
- Department of Pharmacology, Nihon Pharmaceutical University, Komuro, Inamachi, Kita-adachigun, Saitama, Japan.,Division of Microbiology, National Institute of Health Sciences, Setagaya-ku, Tokyo, Japan
| | - Akiko Ishii-Watabe
- Division of Biological Chemistry and Biologicals, National Institute of Health Sciences, Setagaya-ku, Tokyo, Japan
| | - Yutaka Kikuchi
- Division of Microbiology, National Institute of Health Sciences, Setagaya-ku, Tokyo, Japan
| | - Hiroko Kitagawa
- Division of Biological Chemistry and Biologicals, National Institute of Health Sciences, Setagaya-ku, Tokyo, Japan
| | - Hiroko Suzuki
- The Graduate School of Pharmaceutical Sciences, Keio University, Minato-ku, Tokyo, Japan
| | - Hiroomi Tamura
- The Graduate School of Pharmaceutical Sciences, Keio University, Minato-ku, Tokyo, Japan
| | - Minoru Tada
- Division of Biological Chemistry and Biologicals, National Institute of Health Sciences, Setagaya-ku, Tokyo, Japan
| | - Takuo Suzuki
- Division of Biological Chemistry and Biologicals, National Institute of Health Sciences, Setagaya-ku, Tokyo, Japan
| | - Hiroyuki Mizuguchi
- Laboratory of Biochemistry and Molecular Biology, Graduate School of Pharmaceutical Sciences, Osaka University, Suita, Osaka, Japan
| | - Teruhide Yamaguchi
- Department of Pharmacology, Nihon Pharmaceutical University, Komuro, Inamachi, Kita-adachigun, Saitama, Japan.,Division of Microbiology, National Institute of Health Sciences, Setagaya-ku, Tokyo, Japan
| |
Collapse
|
12
|
Gorenoi V, Brehm MU, Koch A, Hagen A, Cochrane Vascular Group. Growth factors for angiogenesis in peripheral arterial disease. Cochrane Database Syst Rev 2017; 6:CD011741. [PMID: 28594443 PMCID: PMC6481523 DOI: 10.1002/14651858.cd011741.pub2] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
BACKGROUND Peripheral artery disease (PAD) is associated with a high clinical and socioeconomic burden. Treatments to alleviate the symptoms of PAD and decrease the risks of amputation and death are a high societal priority. A number of growth factors have shown a potential to stimulate angiogenesis. Growth factors delivered directly (as recombinant proteins), or indirectly (e.g. by viral vectors or DNA plasmids encoding these factors), have emerged as a promising strategy to treat patients with PAD. OBJECTIVES To assess the effects of growth factors that promote angiogenesis for treating people with PAD of the lower extremities. SEARCH METHODS The Cochrane Vascular Information Specialist searched the Specialised Register (June 2016) and CENTRAL (2016, Issue 5). We searched trial registries for details of ongoing or unpublished studies. We also checked the reference lists of relevant publications and, if necessary, tried to contact the trialists for details of the studies. SELECTION CRITERIA We included randomised controlled trials comparing growth factors (delivered directly or indirectly) with no intervention, placebo or any other intervention not based on the growth factor's action in patients with PAD of the lower extremities. The primary outcomes were limb amputation, death and adverse events. The secondary outcomes comprised walking ability, haemodynamic measures, ulceration and rest pain. DATA COLLECTION AND ANALYSIS Two review authors independently selected trials and assessed the risk of bias. We used outcomes of the studies at low risk of bias for the main analysis and of all studies in the sensitivity analyses. We calculated odds ratios (OR) for dichotomous outcomes and mean differences for continuous outcomes with 95% confidence intervals (CI). We evaluated statistical heterogeneity using the I2 statistic and Cochrane's Q test. We conducted meta-analysis for the overall effect and for each growth factor as a subgroup analysis using OR in a fixed-effect model. We evaluated the robustness of the results in a sensitivity analysis using risk ratio (RR) and/or a random-effects model. We also assessed the quality of the evidence for each outcome. MAIN RESULTS We included 20 trials in the review and used 14 studies (on approximately 1400 participants) with published results in the analyses. Six published studies compared fibroblast growth factors (FGF), four studies hepatocyte growth factors (HGF) and another four studies vascular endothelial growth factors (VEGF), versus placebo or no therapy. Six of these studies exclusively or mainly investigated participants with intermittent claudication and eight studies exclusively participants with critical limb ischaemia. Follow-up generally ranged from three months to one year. Two small studies provided some data at 2 years and one of them also at 10 years.The direction and size of effects for growth factors on major limb amputations (OR 0.99, 95% CI 0.71 to 1.38; 10 studies, N = 1075) and death (OR 0.99, 95% CI 0.69 to 1.41; 12 studies, N = 1371) at up to two years are uncertain. The quality of the evidence is low due to risk of bias and imprecision (at one year, moderate-quality evidence due to imprecision). However, growth factors may decrease the rate of any limb amputations (OR 0.56, 95% CI 0.31 to 0.99; 6 studies, N = 415). The quality of the evidence is low due to risk of bias and selective reporting.The direction and size of effects for growth factors on serious adverse events (OR 1.09, 95% CI 0.79 to 1.50; 13 studies, N = 1411) and on any adverse events (OR 1.10, 95% CI 0.73 to 1.64; 4 studies, N = 709) at up to two years are also uncertain. The quality of the evidence is low due to risk of bias and imprecision (for serious adverse events at one year, moderate-quality evidence due to imprecision).Growth factors may improve haemodynamic measures (low-quality evidence), ulceration (very low-quality evidence) and rest pain (very low-quality evidence) up to one year, but they have little or no effect on walking ability (low-quality evidence). We did not identify any relevant differences in effects between growth factors (FGF, HGF and VEGF). AUTHORS' CONCLUSIONS The results of this review do not support the use of therapy with the growth factors FGF, HGF or VEGF in people with PAD of the lower extremities to prevent death or major limb amputation or to improve walking ability. However, the use of these growth factors may improve haemodynamic measures and decrease the rate of any limb amputations (probably due to preventing minor amputations) with an uncertain effect on adverse events; an improvement of ulceration and rest pain is very uncertain. New trials at low risk of bias are needed to generate evidence with more certainty.
Collapse
Affiliation(s)
- Vitali Gorenoi
- Hannover Medical SchoolEvidence based Medicine & Health Technology Assessment Working Group, Institute for Epidemiology, Social Medicine and Health Systems ResearchCarl‐Neuberg‐Str. 1HannoverGermany30625
| | - Michael U Brehm
- Hannover Medical SchoolDepartment for Cardiology and AngiologyCarl‐Neuberg‐Str. 1HannoverGermany30265
| | - Armin Koch
- Institute for Biometry, Hannover Medical SchoolCarl‐Neuberg‐Str. 1HannoverGermany30625
| | - Anja Hagen
- Hannover Medical SchoolEvidence based Medicine & Health Technology Assessment Working Group, Institute for Epidemiology, Social Medicine and Health Systems ResearchCarl‐Neuberg‐Str. 1HannoverGermany30625
| | | |
Collapse
|
13
|
Nishijima A, Yamamoto N, Yoshida R, Hozawa K, Yanagibayashi S, Takikawa M, Hayasaka R, Nishijima J, Okada T, Sekido M. Maggot debridement therapy for a patient with critical limb ischaemia and severe cardiac dysfunction: possibility of limb salvage. CASE REPORTS IN PLASTIC SURGERY AND HAND SURGERY 2017; 4:42-47. [PMID: 28567433 PMCID: PMC5439397 DOI: 10.1080/23320885.2017.1327322] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/06/2017] [Accepted: 05/02/2017] [Indexed: 12/02/2022]
Abstract
Ischaemic skin ulcer occurred on the foot of a 73-year-old man who had a history of fulminant myocarditis with severe cardiac dysfunction. We attempted wound bed preparation by maggot debridement therapy and salvaged his limb. It can be one of the adjuvant treatment strategies for critical limb ischaemia.
Collapse
Affiliation(s)
- Akio Nishijima
- Department of Plastic and Reconstructive Surgery, New Tokyo Hospital, Matsudo, Chiba, Japan
| | - Naoto Yamamoto
- Department of Plastic and Reconstructive Surgery, New Tokyo Hospital, Matsudo, Chiba, Japan
| | - Ryuichi Yoshida
- Department of Plastic and Reconstructive Surgery, New Tokyo Hospital, Matsudo, Chiba, Japan
| | - Koji Hozawa
- Department of Cardiovascular Medicine, New Tokyo Hospital, Matsudo, Chiba, Japan
| | - Satoshi Yanagibayashi
- Department of Plastic and Reconstructive Surgery, New Tokyo Hospital, Matsudo, Chiba, Japan
| | - Megumi Takikawa
- Department of Plastic and Reconstructive Surgery, New Tokyo Hospital, Matsudo, Chiba, Japan
| | - Rie Hayasaka
- Department of Plastic and Reconstructive Surgery, New Tokyo Hospital, Matsudo, Chiba, Japan
| | - Junko Nishijima
- Department of Plastic and Reconstructive Surgery, New Tokyo Hospital, Matsudo, Chiba, Japan
| | - Tadasu Okada
- Department of Dermatology, Mami Dermatology Clinic, Koshiba, Nara, Japan
| | - Mitsuru Sekido
- Department of Plastic and Reconstructive Surgery, University of Tsukuba, Tsukuba, Ibaraki, Japan
| |
Collapse
|
14
|
Aoyama N, Nishinari M, Ohtani S, Kanai A, Noda C, Hirata M, Miyamoto A, Watanabe M, Minamino T, Izumi T, Ako J. Clinical features and predictors of patients with critical limb ischemia who responded to autologous mononuclear cell transplantation for therapeutic angiogenesis. Heart Vessels 2017; 32:1099-1108. [DOI: 10.1007/s00380-017-0968-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/17/2016] [Accepted: 03/10/2017] [Indexed: 01/11/2023]
|
15
|
He S, Gleason J, Fik-Rymarkiewicz E, DiFiglia A, Bharathan M, Morschauser A, Djuretic I, Xu Y, Krakovsky M, Jankovic V, Buensuceso C, Edinger J, Herzberg U, Hofgartner W, Hariri R. Human Placenta-Derived Mesenchymal Stromal-Like Cells Enhance Angiogenesis via T Cell-Dependent Reprogramming of Macrophage Differentiation. Stem Cells 2017; 35:1603-1613. [PMID: 28233380 DOI: 10.1002/stem.2598] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2016] [Revised: 01/20/2017] [Accepted: 02/11/2017] [Indexed: 12/11/2022]
Abstract
Peripheral arterial disease (PAD) is a leading cause of limb loss and mortality worldwide with limited treatment options. Mesenchymal stromal cell (MSC) therapy has demonstrated positive effects on angiogenesis in preclinical models and promising therapeutic efficacy signals in early stage clinical studies; however, the mechanisms underlying MSC-mediated angiogenesis remain largely undefined. Here, we investigated the mechanism of action of human placenta-derived MSC-like cells (PDA-002) in inducing angiogenesis using mice hind limb ischemia model. We showed that PDA-002 improved blood flow and promoted collateral vessel formation in the injured limb. Histological analysis demonstrated that PDA-002 increased M2-like macrophages in ischemic tissue. Analysis of the changes in functional T cell phenotype in the draining lymph nodes revealed that PDA-002 treatment was associated with the induction of cytokine and gene expression signatures of Th2 response. Angiogenic effect of PDA-002 was markedly reduced in Balb/c nude mice compared with wild type. This reduction in efficacy was reversed by T cell reconstitution, suggesting T cells are essential for PDA-002-mediated angiogenesis. Furthermore, effect of PDA-002 on macrophage differentiation was also T cell-dependent as a PDA-002-mediated M2-like macrophage skewing was only observed in wild type and T cell reconstituted nude mice, but not in nude mice. Finally, we showed that PDA-002-treated animals had enhanced angiogenic recovery in response to the second injury when PDA-002 no longer persisted in vivo. These results suggest that PDA-002 enhances angiogenesis through an immunomodulatory mechanism involving T cell-dependent reprogramming of macrophage differentiation toward M2-like phenotype. Stem Cells 2017;35:1603-1613.
Collapse
Affiliation(s)
- Shuyang He
- Celgene Cellular Therapeutics, Warren, New Jersey, USA
| | | | | | | | | | | | | | - Yan Xu
- Invivotek, Hamilton, New Jersey, USA
| | | | | | | | - James Edinger
- Celgene Cellular Therapeutics, Warren, New Jersey, USA
| | - Uri Herzberg
- Celgene Cellular Therapeutics, Warren, New Jersey, USA
| | | | - Robert Hariri
- Celgene Cellular Therapeutics, Warren, New Jersey, USA
| |
Collapse
|
16
|
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]
|
17
|
Hou L, Kim JJ, Woo YJ, Huang NF. Stem cell-based therapies to promote angiogenesis in ischemic cardiovascular disease. Am J Physiol Heart Circ Physiol 2015; 310:H455-65. [PMID: 26683902 DOI: 10.1152/ajpheart.00726.2015] [Citation(s) in RCA: 78] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/16/2015] [Accepted: 12/09/2015] [Indexed: 12/30/2022]
Abstract
Stem cell therapy is a promising approach for the treatment of tissue ischemia associated with myocardial infarction and peripheral arterial disease. Stem and progenitor cells derived from bone marrow or from pluripotent stem cells have shown therapeutic benefit in boosting angiogenesis as well as restoring tissue function. Notably, adult stem and progenitor cells including mononuclear cells, endothelial progenitor cells, and mesenchymal stem cells have progressed into clinical trials and have shown positive benefits. In this review, we overview the major classes of stem and progenitor cells, including pluripotent stem cells, and summarize the state of the art in applying these cell types for treating myocardial infarction and peripheral arterial disease.
Collapse
Affiliation(s)
- Luqia Hou
- Veterans Affairs Palo Alto Health Care System, Palo Alto, California; Stanford Cardiovascular Institute, Stanford University, Stanford, California; and
| | - Joseph J Kim
- Veterans Affairs Palo Alto Health Care System, Palo Alto, California; Stanford Cardiovascular Institute, Stanford University, Stanford, California; and
| | - Y Joseph Woo
- Stanford Cardiovascular Institute, Stanford University, Stanford, California; and Department of Cardiothoracic Surgery, Stanford University, Stanford, California
| | - Ngan F Huang
- Veterans Affairs Palo Alto Health Care System, Palo Alto, California; Stanford Cardiovascular Institute, Stanford University, Stanford, California; and Department of Cardiothoracic Surgery, Stanford University, Stanford, California
| |
Collapse
|
18
|
Abu Dabrh AM, Steffen MW, Asi N, Undavalli C, Wang Z, Elamin MB, Conte MS, Murad MH. Nonrevascularization-based treatments in patients with severe or critical limb ischemia. J Vasc Surg 2015; 62:1330-9.e13. [DOI: 10.1016/j.jvs.2015.07.069] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2015] [Accepted: 07/09/2015] [Indexed: 11/17/2022]
|
19
|
Litwin M, Radwańska A, Paprocka M, Kieda C, Dobosz T, Witkiewicz W, Baczyńska D. The role of FGF2 in migration and tubulogenesis of endothelial progenitor cells in relation to pro-angiogenic growth factor production. Mol Cell Biochem 2015; 410:131-42. [PMID: 26314253 DOI: 10.1007/s11010-015-2545-5] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2015] [Accepted: 08/18/2015] [Indexed: 01/23/2023]
Abstract
In recent years, special attention has been paid to finding new pro-angiogenic factors which could be used in gene therapy of vascular diseases such as critical limb ischaemia (CLI). Angiogenesis, the formation of new blood vessels, is a complex process dependent on different cytokines, matrix proteins, growth factors and other pro- or anti-angiogenic stimuli. Numerous lines of evidence suggest that key mediators of angiogenesis, vascular endothelial growth factor (VEGF) and hepatocyte growth factor (HGF) together with fibroblast growth factor2 (FGF2) are involved in regulation of the normal and pathological process of angiogenesis. However, less information is available on the complex interactions between these and other angiogenic factors. The aim of this study was to characterise the effect of fibroblast growth factor2 on biological properties of human endothelial progenitor cells with respect to the expression level of other regulatory cytokines. Ectopic expression of FGF2 in EP cells stimulates their pro-angiogenic behaviour, leading to increased proliferation, migration and tube formation abilities. Moreover, we show that the expression profile of VEGF and other pro-angiogenic cytokines, such as HGF, MCP2, and interleukins, is affected differently by FGF2 in EPC. In conclusion, we provide evidence that FGF2 directly affects not only the biological properties of EP cells but also the expression pattern and secretion of numerous chemocytokines. Our results suggest that FGF2 could be applied in therapeutic approaches for CLI and other ischaemic diseases of the vascular system in vivo.
Collapse
Affiliation(s)
- Monika Litwin
- WroVasc - Integrated Cardiovascular Centre, Regional Specialist Hospital, Research and Development Centre, Kamienskiego 73a, Wrocław, Poland.
| | - Agata Radwańska
- WroVasc - Integrated Cardiovascular Centre, Regional Specialist Hospital, Research and Development Centre, Kamienskiego 73a, Wrocław, Poland
- Institut de Biologie Valrose, CNRS UMR 7277, Faculte des Sciences, Universite Nice-Sophia Antipolis, ParcValrose, 28 Avenue Valrose, 06108, Nice cedex, France
| | - Maria Paprocka
- WroVasc - Integrated Cardiovascular Centre, Regional Specialist Hospital, Research and Development Centre, Kamienskiego 73a, Wrocław, Poland
- Ludwik Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, Wrocław, Poland
| | - Claudine Kieda
- Centre de Biophysique Moléculaire, CNRS, 45071, Orléans Cedex 2, France
| | - Tadeusz Dobosz
- WroVasc - Integrated Cardiovascular Centre, Regional Specialist Hospital, Research and Development Centre, Kamienskiego 73a, Wrocław, Poland
- Laboratory of Molecular Technique, Institute of Forensic Medicine, Medical University of Wrocław, Wrocław, Poland
| | - Wojciech Witkiewicz
- WroVasc - Integrated Cardiovascular Centre, Regional Specialist Hospital, Research and Development Centre, Kamienskiego 73a, Wrocław, Poland
| | - Dagmara Baczyńska
- WroVasc - Integrated Cardiovascular Centre, Regional Specialist Hospital, Research and Development Centre, Kamienskiego 73a, Wrocław, Poland
- Laboratory of Molecular Technique, Institute of Forensic Medicine, Medical University of Wrocław, Wrocław, Poland
| |
Collapse
|
20
|
Agrawal K, Eberhardt RT. Contemporary medical management of peripheral arterial disease: a focus on risk reduction and symptom relief for intermittent claudication. Cardiol Clin 2015; 33:111-37. [PMID: 25439335 DOI: 10.1016/j.ccl.2014.09.010] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Peripheral arterial disease (PAD) is primarily caused by progressive systemic atherosclerosis manifesting in the lower extremities. This review addresses the epidemiology, clinical presentation and evaluation, and medical management of PAD, with a focus on intermittent claudication. Key advances in the recognition of cardiovascular risk in asymptomatic individuals with mildly abnormal ankle-brachial index, newer reflections on exercise therapy, and a review of established and investigational agents for the treatment of symptomatic PAD, such as cilostazol, statins, and angiotensin-converting enzyme inhibitors, are highlighted.
Collapse
Affiliation(s)
- Kush Agrawal
- Cardiovascular and Endovascular Intervention, Section of Cardiovascular Medicine, Boston Medical Center, Boston, MA 02118, USA
| | - Robert T Eberhardt
- Vascular Medicine Program, Section of Cardiovascular Medicine, Boston Medical Center, Boston University School of Medicine, 88 East Newton Street, Boston MA 02118, USA.
| |
Collapse
|
21
|
A review of the pathophysiology and potential biomarkers for peripheral artery disease. Int J Mol Sci 2015; 16:11294-322. [PMID: 25993296 PMCID: PMC4463701 DOI: 10.3390/ijms160511294] [Citation(s) in RCA: 114] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2015] [Revised: 03/29/2015] [Accepted: 04/08/2015] [Indexed: 12/12/2022] Open
Abstract
Peripheral artery disease (PAD) is due to the blockage of the arteries supplying blood to the lower limbs usually secondary to atherosclerosis. The most severe clinical manifestation of PAD is critical limb ischemia (CLI), which is associated with a risk of limb loss and mortality due to cardiovascular events. Currently CLI is mainly treated by surgical or endovascular revascularization, with few other treatments in routine clinical practice. There are a number of problems with current PAD management strategies, such as the difficulty in selecting the appropriate treatments for individual patients. Many patients undergo repeated attempts at revascularization surgery, but ultimately require an amputation. There is great interest in developing new methods to identify patients who are unlikely to benefit from revascularization and to improve management of patients unsuitable for surgery. Circulating biomarkers that predict the progression of PAD and the response to therapies could assist in the management of patients. This review provides an overview of the pathophysiology of PAD and examines the association between circulating biomarkers and PAD presence, severity and prognosis. While some currently identified circulating markers show promise, further larger studies focused on the clinical value of the biomarkers over existing risk predictors are needed.
Collapse
|
22
|
Choi D, Son B, Park TH, Hong J. Controlled surface functionality of magnetic nanoparticles by layer-by-layer assembled nano-films. NANOSCALE 2015; 7:6703-6711. [PMID: 25798789 DOI: 10.1039/c4nr07373h] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/04/2023]
Abstract
Over the past several years, the preparation of functionalized nanoparticles has been aggressively pursued in order to develop desired structures, compositions, and structural order. Among the various nanoparticles, iron oxide magnetic nanoparticles (MNPs) have shown great promise because the material generated using these MNPs can be used in a variety of biomedical applications and possible bioactive functionalities. In this study, we report the development of various functionalized MNPs (F-MNPs) generated using the layer-by-layer (LbL) self-assembly method. To provide broad functional opportunities, we fabricated F-MNP bio-toolbox by using three different materials: synthetic polymers, natural polymers, and carbon materials. Each of these F-MNPs displays distinct properties, such as enhanced thickness or unique morphologies. In an effort to explore their biomedical applications, we generated basic fibroblast growth factor (bFGF)-loaded F-MNPs. The bFGF-loaded F-MNPs exhibited different release mechanisms and loading amounts, depending on the film material and composition order. Moreover, bFGF-loaded F-MNPs displayed higher biocompatibility and possessed superior proliferation properties than the bare MNPs and pure bFGF, respectively. We conclude that by simply optimizing the building materials and the nanoparticle's film composition, MNPs exhibiting various bioactive properties can be generated.
Collapse
Affiliation(s)
- Daheui Choi
- School of Chemical Engineering & Material Science, Chung-Ang University, 47 Heukseok-ro, Dongjak-gu, Seoul 156-756, Republic of Korea.
| | | | | | | |
Collapse
|
23
|
Deev RV, Kalinin RE, Chervyakov YV, Staroverov IN, Plaksa IL, Mzavanadze ND, Pozdeev DV, Dalgatov GD, Isaev AA. Long-term results of pl-VEGF165 intramuscular gene transfer in patients with atherosclerotic chronic lower limb ischemia. ACTA ACUST UNITED AC 2015. [DOI: 10.17116/kardio20158443-49] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
|
24
|
Bura A, Planat-Benard V, Bourin P, Silvestre JS, Gross F, Grolleau JL, Saint-Lebese B, Peyrafitte JA, Fleury S, Gadelorge M, Taurand M, Dupuis-Coronas S, Leobon B, Casteilla L. Phase I trial: the use of autologous cultured adipose-derived stroma/stem cells to treat patients with non-revascularizable critical limb ischemia. Cytotherapy 2014; 16:245-57. [PMID: 24438903 DOI: 10.1016/j.jcyt.2013.11.011] [Citation(s) in RCA: 216] [Impact Index Per Article: 19.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2013] [Revised: 11/23/2013] [Accepted: 11/26/2013] [Indexed: 12/14/2022]
Abstract
BACKGROUND AIMS Non-revascularizable critical limb ischemia (CLI) is the most severe stage of peripheral arterial disease, with no therapeutic option. Extensive preclinical studies have demonstrated that adipose-derived stroma cell (ASC) transplantation strongly improves revascularization and tissue perfusion in ischemic limbs. This study, named ACellDREAM, is the first phase I trial to evaluate the feasibility and safety of intramuscular injections of autologous ASC in non-revascularizable CLI patients. METHODS Seven patients were consecutively enrolled, on the basis of the following criteria: (i) lower-limb rest pain or ulcer; (ii) ankle systolic oxygen pressure <50 or 70 mm Hg for non-diabetic and diabetic patients, respectively, or first-toe systolic oxygen pressure <30 mm Hg or 50 mm Hg for non-diabetic and diabetic patients, respectively; (iii) not suitable for revascularization. ASCs from abdominal fat were grown for 2 weeks and were then characterized. RESULTS More than 200 million cells were obtained, with almost total homogeneity and no karyotype abnormality. The expressions of stemness markers Oct4 and Nanog were very low, whereas expression of telomerase was undetectable in human ASCs compared with human embryonic stem cells. ASCs (10(8)) were then intramuscularly injected into the ischemic leg of patients, with no complication, as judged by an independent committee. Trans-cutaneous oxygen pressure tended to increase in most patients. Ulcer evolution and wound healing showed improvement. CONCLUSIONS These data demonstrate the feasibility and safety of autologous ASC transplantation in patients with objectively proven CLI not suitable for revascularization. The improved wound healing also supports a putative functional efficiency.
Collapse
Affiliation(s)
- Alessandra Bura
- Service de médecine vasculaire, Pôle cardiovasculaire et métabolique, Centre hospitalo-universitaire de Toulouse, Toulouse, France; Université de Toulouse III, Paul Sabatier, Toulouse, France
| | - Valerie Planat-Benard
- CNRS, Université Toulouse III, UPS UMR5273 STROMAlab, Toulouse, France; EFS (Etablissement Français du Sang), STROMAlab, Toulouse, France; Inserm U1031 STROMAlab, Toulouse, France; Université Toulouse III, UPS UMR5273 STROMAlab, Toulouse, France
| | - Philippe Bourin
- EFS (Etablissement Français du Sang), STROMAlab, Toulouse, France; CSA21, 7 chemin des silos, Toulouse, France
| | - Jean-Sebastien Silvestre
- INSERM UMRS 970, Paris Cardiovascular Research Center, Université Paris Descartes, Sorbonne Paris cité, Paris, France
| | - Fabian Gross
- Centre d'Investigation Clinique de Biothérapies de Toulouse, Toulouse cedex 9, France; Direction de la Recherche Médicale et Innovation (DRMI) du CHU de Toulouse, Toulouse cedex 9, France
| | - Jean-Louis Grolleau
- Université de Toulouse III, Paul Sabatier, Toulouse, France; Service de chirurgie plastique, reconstructrice et esthétique, Centre hospitalo-universitaire de Toulouse, Toulouse, France
| | - Bertrand Saint-Lebese
- Service de chirurgie vasculaire, Pôle cardiovasculaire et métabolique, Centre hospitalo-universitaire de Toulouse, Toulouse, France
| | - Julie-Anne Peyrafitte
- CNRS, Université Toulouse III, UPS UMR5273 STROMAlab, Toulouse, France; EFS (Etablissement Français du Sang), STROMAlab, Toulouse, France; Inserm U1031 STROMAlab, Toulouse, France; Université Toulouse III, UPS UMR5273 STROMAlab, Toulouse, France
| | - Sandrine Fleury
- CNRS, Université Toulouse III, UPS UMR5273 STROMAlab, Toulouse, France; EFS (Etablissement Français du Sang), STROMAlab, Toulouse, France; Inserm U1031 STROMAlab, Toulouse, France; Université Toulouse III, UPS UMR5273 STROMAlab, Toulouse, France
| | - Melanie Gadelorge
- CNRS, Université Toulouse III, UPS UMR5273 STROMAlab, Toulouse, France; EFS (Etablissement Français du Sang), STROMAlab, Toulouse, France; Inserm U1031 STROMAlab, Toulouse, France; Université Toulouse III, UPS UMR5273 STROMAlab, Toulouse, France
| | - Marion Taurand
- CNRS, Université Toulouse III, UPS UMR5273 STROMAlab, Toulouse, France; EFS (Etablissement Français du Sang), STROMAlab, Toulouse, France; Inserm U1031 STROMAlab, Toulouse, France; Université Toulouse III, UPS UMR5273 STROMAlab, Toulouse, France
| | - Sophie Dupuis-Coronas
- CNRS, Université Toulouse III, UPS UMR5273 STROMAlab, Toulouse, France; EFS (Etablissement Français du Sang), STROMAlab, Toulouse, France; Inserm U1031 STROMAlab, Toulouse, France; Université Toulouse III, UPS UMR5273 STROMAlab, Toulouse, France
| | - Bertrand Leobon
- Université de Toulouse III, Paul Sabatier, Toulouse, France; Service de chirurgie cardiovasculaire, Pôle cardiovasculaire et métabolique, Centre hospitalo-universitaire de Toulouse, Toulouse, France
| | - Louis Casteilla
- CNRS, Université Toulouse III, UPS UMR5273 STROMAlab, Toulouse, France; EFS (Etablissement Français du Sang), STROMAlab, Toulouse, France; Inserm U1031 STROMAlab, Toulouse, France; Université Toulouse III, UPS UMR5273 STROMAlab, Toulouse, France.
| |
Collapse
|
25
|
Ahimastos AA, Latouche C, Natoli AK, Reddy-luthmoodoo M, Golledge J, Kingwell BA. Potential Vascular Mechanisms of Ramipril Induced Increases in Walking Ability in Patients With Intermittent Claudication. Circ Res 2014; 114:1144-55. [DOI: 10.1161/circresaha.114.302420] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Rationale:
We recently reported that ramipril more than doubled maximum walking times in patients with peripheral artery disease with intermittent claudication.
Objective:
Our aim was to conduct exploratory analyses of the effects of ramipril therapy on circulating biomarkers of angiogenesis/arteriogenesis, thrombosis, inflammation, and leukocyte adhesion in patients with intermittent claudication.
Methods and Results:
One hundred sixty-five patients with intermittent claudication (mean, 65.3 [SD, 6.7] years) were administered ramipril 10 mg per day (n=82) or matching placebo (n=83) for 24 weeks in a randomized, double-blind study. Plasma biomarkers of angiogenesis/arteriogenesis (vascular endothelial growth factor-A, fibroblast growth factor-2), thrombosis (D-dimer, von Willebrand factor, thrombin-antithrombin III), inflammation (high-sensitivity C-reactive protein, osteopontin), and leukocyte adhesion (soluble vascular cell adhesion molecule-1, soluble intracellular adhesion molecule-1) were measured at baseline and 24 weeks. Relative to placebo, ramipril was associated with increases in vascular endothelial growth factor-A by 38% (95% confidence interval [CI], 34%–42%) and fibroblast growth factor-2 by 64% (95% CI, 44–85%;
P
<0.001 for both), and reductions in D-dimer by 24% (95% CI, −30% to −18%), von Willebrand factor by 22% (95% CI, −35% to −9%), thrombin-antithrombin III by 16% (95% CI, −19% to −13%), high-sensitivity C-reactive protein by 13% (95% CI, −14% to −9%), osteopontin by 12% (95% CI, −14% to −10%), soluble vascular cell adhesion molecule-1 by 14% (95% CI, −18% to −10%), and soluble intracellular adhesion molecule-1 by 15% (95% CI, −17% to −13%; all
P
<0.001). With the exception of von Willebrand factor, all the above changes correlated significantly with the change in maximum walking time (
P
=0.02−0.001) in the group treated with ramipril.
Conclusions:
Ramipril is associated with an increase in the biomarkers of angiogenesis/arteriogenesis and reduction in the markers of thrombosis, inflammation, and leukocyte adhesion. This study informs strategies to improve mobility in patients with intermittent claudication.
Clinical Trial Registration Information:
URL:
http://clinicaltrials.gov
. Unique identifier: NCT00681226.
Collapse
Affiliation(s)
- Anna A. Ahimastos
- From Baker IDI Heart and Diabetes Institute and Department of Cardiovascular Medicine, Alfred Hospital, Melbourne, Australia (A.A.A., C.L., A.K.N., M.R., B.A.K.); Queensland Research Centre for Peripheral Vascular Disease, James Cook University, Townsville, Queensland, Australia (J.G.); and Department of Vascular and Endovascular Surgery, Townsville Hospital, Queensland, Australia (J.G.)
| | - Celine Latouche
- From Baker IDI Heart and Diabetes Institute and Department of Cardiovascular Medicine, Alfred Hospital, Melbourne, Australia (A.A.A., C.L., A.K.N., M.R., B.A.K.); Queensland Research Centre for Peripheral Vascular Disease, James Cook University, Townsville, Queensland, Australia (J.G.); and Department of Vascular and Endovascular Surgery, Townsville Hospital, Queensland, Australia (J.G.)
| | - Alaina K. Natoli
- From Baker IDI Heart and Diabetes Institute and Department of Cardiovascular Medicine, Alfred Hospital, Melbourne, Australia (A.A.A., C.L., A.K.N., M.R., B.A.K.); Queensland Research Centre for Peripheral Vascular Disease, James Cook University, Townsville, Queensland, Australia (J.G.); and Department of Vascular and Endovascular Surgery, Townsville Hospital, Queensland, Australia (J.G.)
| | - Medini Reddy-luthmoodoo
- From Baker IDI Heart and Diabetes Institute and Department of Cardiovascular Medicine, Alfred Hospital, Melbourne, Australia (A.A.A., C.L., A.K.N., M.R., B.A.K.); Queensland Research Centre for Peripheral Vascular Disease, James Cook University, Townsville, Queensland, Australia (J.G.); and Department of Vascular and Endovascular Surgery, Townsville Hospital, Queensland, Australia (J.G.)
| | - Jonathan Golledge
- From Baker IDI Heart and Diabetes Institute and Department of Cardiovascular Medicine, Alfred Hospital, Melbourne, Australia (A.A.A., C.L., A.K.N., M.R., B.A.K.); Queensland Research Centre for Peripheral Vascular Disease, James Cook University, Townsville, Queensland, Australia (J.G.); and Department of Vascular and Endovascular Surgery, Townsville Hospital, Queensland, Australia (J.G.)
| | - Bronwyn A. Kingwell
- From Baker IDI Heart and Diabetes Institute and Department of Cardiovascular Medicine, Alfred Hospital, Melbourne, Australia (A.A.A., C.L., A.K.N., M.R., B.A.K.); Queensland Research Centre for Peripheral Vascular Disease, James Cook University, Townsville, Queensland, Australia (J.G.); and Department of Vascular and Endovascular Surgery, Townsville Hospital, Queensland, Australia (J.G.)
| |
Collapse
|
26
|
Therapeutic site selection is important for the successful development of collateral vessels. J Vasc Surg 2014; 62:190-9. [PMID: 24630870 DOI: 10.1016/j.jvs.2014.02.004] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2013] [Revised: 02/05/2014] [Accepted: 02/06/2014] [Indexed: 12/18/2022]
Abstract
BACKGROUND Induction of collateral development to improve tissue perfusion is a promising approach for the treatment of arterial occlusive diseases. Several growth factors and cells have been reported to increase collateral circulation; however, the appropriate site for the delivery of these factors and cells is unclear. In this study, we identified the delivery site for growth factor in a rabbit model of limb ischemia and evaluated whether specific delivery of basic fibroblast growth factor (bFGF) to this site enhanced collateral augmentation. METHODS The left femoral artery of Japanese white rabbits was excised to induce limb ischemia. Twenty-eight days thereafter, angiograms were obtained to identify the typical pattern of collateral development in this model. Subsequently, bFGF (100 μg) was selectively injected into the left coccygeofemoral muscle (coccygeo group) or adductor muscle (adductor group), major thigh muscles in proximity. Collateral development was evaluated at 28 days after injection, and its mechanism was assessed by immunologic and morphometric analyses of muscle samples. RESULTS Angiographic evaluation of this model revealed that after femoral artery excision, collateral vessels generally developed in the left coccygeofemoral muscle, whereas few collateral vessels were detected in the left adductor muscle. At 28 days after injection, calf blood pressure ratio, defined as left pressure to right pressure, was significantly higher in the coccygeo group than in the adductor group (0.85 ± 0.05 vs 0.69 ± 0.05, respectively; P < .01). Similar results were observed in blood flow through the internal iliac artery (resting: 24.6 ± 6.1 vs 17.4 ± 8.0 mL/min, P < .01; maximum: 47.4 ± 12.3 vs 33.2 ± 10.7 mL/min, P < .01) and in the angiographic score (0.67 ± 0.13 vs 0.39 ± 0.11; P < .01). Immunologic analyses of the coccygeofemoral muscle at day 3 showed marked expressions of Ki-67, monocyte chemotactic protein 1, and FGF receptor 1 in the coccygeo group compared with the adductor group. Morphometric analyses of the same muscle at day 14 also revealed that collateral vessel density and wall thickness were significantly increased in the coccygeal group compared with the adductor group. CONCLUSIONS These findings demonstrated that selective bFGF delivery to the coccygeofemoral muscle markedly improved collateral development and limb perfusion compared with delivery to the adductor muscle, suggesting that site selection is important in increasing therapeutic efficacy.
Collapse
|
27
|
Takagi G, Miyamoto M, Tara S, Kirinoki-Ichikawa S, Kubota Y, Hada T, Takagi I, Mizuno K. Therapeutic vascular angiogenesis for intractable macroangiopathy-related digital ulcer in patients with systemic sclerosis: a pilot study. Rheumatology (Oxford) 2014; 53:854-9. [PMID: 24390937 DOI: 10.1093/rheumatology/ket432] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
OBJECTIVE SSc causes intractable ischaemic ulcers. To avoid major amputation, we examined the safety and efficacy of therapeutic vascular angiogenesis for digital ulcers due to SSc. METHODS A single-centre, open-label pilot study was conducted in patients with an ischaemic digital ulcer [n = 40, mean age 65 years (s.d. 8), Rutherford class III-5 or III-6) due to lcSSc (n = 11) or arteriosclerosis obliterans (ASO; n = 29). Bone marrow mononuclear cells (0.4-5.1 × 10(10) cells in total) were administered into the ischaemic limbs. We evaluated short-term safety and efficacy by means of a pain scale, (99m)Tc-tetrofosmin scintigraphy and transcutaneous oxygen tension (TcPO2) before and 4 weeks after treatment. Also, the 2-year outcome was compared. RESULTS There was a case of amputation in each group within 4 weeks after therapy. The pain scale significantly decreased in both groups [lcSSc 93 mm (s.d. 9) to 11 (s.d. 16), P < 0.01; ASO 77 mm (s.d. 22) to 16 (s.d. 13), P < 0.01] and TcPO2 significantly improved [lcSSc 9.0 mmHg (s.d. 9) to 35 (s.d. 14), P < 0.01; ASO 18 mmHg (s.d. 10) to 29 (s.d. 21), P < 0.05). At the 2-year follow-up, the limb amputation rate was 9.1% in lcSSc and 20.7% in ASO (P = 0.36), while the recurrence rate was 18.2% in lcSSc and 17.2% in ASO (P = 0.95). All-cause mortality was 27.3% in lcSSc and 17.2% in ASO (P = 0.65). CONCLUSION In patients with lcSSc, bone marrow mononuclear cell implantation provides clinical benefit and is safe, without major adverse reactions, and may become an effective strategy. TRIAL REGISTRATION UMIN-CTR, http://www.umin.ac.jp/ctr/index-j.htm, no. UMIN000004112.
Collapse
Affiliation(s)
- Gen Takagi
- Department of Internal Medicine, Division of Cardiovascular and Regenerative Medicine, Nippon Medical School, 1-1-5 Sendagi, Bunkyo-ku, Tokyo 113-8603, Japan.
| | | | | | | | | | | | | | | |
Collapse
|
28
|
Knepper JP, Henke PK. Diagnosis, Prevention, and Treatment of Claudication. Surg Clin North Am 2013; 93:779-88, vii. [DOI: 10.1016/j.suc.2013.04.005] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
|
29
|
Kinlay S. Outcomes for clinical studies assessing drug and revascularization therapies for claudication and critical limb ischemia in peripheral artery disease. Circulation 2013; 127:1241-50. [PMID: 23509032 PMCID: PMC4507406 DOI: 10.1161/circulationaha.112.001232] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Affiliation(s)
- Scott Kinlay
- MBBS, Cardiovascular Division, VA Boston Healthcare System, 1400 VFW Pkwy, West Roxbury, MA 02132, USA.
| |
Collapse
|
30
|
Affiliation(s)
- Jeffrey S Berger
- Divisions of Cardiology and Vascular Surgery, New York University School of Medicine, New York, NY, USA
| | | |
Collapse
|
31
|
Bleda S, De Haro J, Varela C, Esparza L, De Maturana IL, Acin F. Impact of VEGF polymorphisms on the severity of peripheral artery disease in diabetic patients. Growth Factors 2012; 30:277-82. [PMID: 22762535 DOI: 10.3109/08977194.2012.703664] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
OBJECTIVE To determine the potential genotype vascular endothelial growth factor (VEGF) gene differences in diabetic patients with peripheral arterial disease (PAD), which might be associated with different stages of the vascular disease. METHODS A study was conducted with type 2 diabetic patients with PAD [n = 70; 32 intermittent claudication and 38 critical limb ischaemia (CLI)]. Genotyping of the VEGF gene insertion/deletion - 2549, - 2578 C/A and +405 G/C polymorphisms was done in both groups and correlated them with the severity of PAD. We compared serum VEGF levels in both groups. RESULTS There was a higher frequency of +405 CC and - 2578 CC genotypes in claudication group [(31.3% vs. 5.4%, p = 0.01) and (37.5% vs. 15.8%, p = 0.05), respectively]. The presence of +405 GG and - 2578 AA genotypes was more common among CLI patients [(57.8% vs. 37.5%, p = 0.01) and (42.1% vs. 18.8%, p = 0.05), respectively]. There were higher serum VEGF levels in patients with CLI (p = 0.029). CONCLUSIONS We found preliminary evidence regarding the association between VEGF polymorphisms and different stages of PAD in diabetic patients.
Collapse
Affiliation(s)
- Silvia Bleda
- Angiology and Vascular Surgery Department of Hospital Universitario de Getafe, Madrid, Spain.
| | | | | | | | | | | |
Collapse
|
32
|
Davies MG. Critical limb ischemia: cell and molecular therapies for limb salvage. Methodist Debakey Cardiovasc J 2012; 8:20-7. [PMID: 23342184 PMCID: PMC3549646 DOI: 10.14797/mdcj-8-4-20] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
There is a growing interest in developing new limb salvage therapies for patients with severe peripheral artery disease who have no alternative to amputation. Cell and gene therapy studies are showing promise in controlling pain and minor ulceration in patients with significant critical limb ischemia. Among cardiovascular cell and molecular therapy programs, The Methodist Hospital is one of the leading centers in both gene and cell therapy for critical limb ischemia. Randomized controlled trials continue to be performed, and these experimental therapies will move from research to pharmacy within the decade. In conjunction with aggressive medical and surgical management, these emergent therapies may help patients with critical limb ischemia avoid a major amputation and are one of the foundations of any advanced limb salvage program.
Collapse
Affiliation(s)
- Mark G Davies
- Methodist DeBakey Heart & Vascular Center, The Methodist Hospital, Houston, Texas, USA
| |
Collapse
|
33
|
Hastings CL, Kelly HM, Murphy MJ, Barry FP, O'Brien FJ, Duffy GP. Development of a thermoresponsive chitosan gel combined with human mesenchymal stem cells and desferrioxamine as a multimodal pro-angiogenic therapeutic for the treatment of critical limb ischaemia. J Control Release 2012; 161:73-80. [DOI: 10.1016/j.jconrel.2012.04.033] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2012] [Revised: 04/16/2012] [Accepted: 04/20/2012] [Indexed: 12/01/2022]
|
34
|
Enhanced vascular endothelial growth factor gene expression in ischaemic skin of critical limb ischaemia patients. Int J Vasc Med 2012; 2012:691528. [PMID: 22536509 PMCID: PMC3321296 DOI: 10.1155/2012/691528] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2011] [Accepted: 01/25/2012] [Indexed: 11/25/2022] Open
Abstract
Objectives. To perform a quantitative analysis of the vascular endothelial growth factor (VEGF) gene transcription in the skin of ischemic legs and provide information for VEGF in the pathogenesis in critical limb ischemia (CLI). Methods. Skin biopsies were obtained from 40 patients with CLI. Control samples came from 44 patients with chronic venous disease. VEGF gene expression was analysed using quantitative polymerase chain reaction. Results. Patients with CLI had higher skin VEGF expression than control group (RQ: 1.3 ± 0.1 versus 1, P = 0.04). Conclusions. We found an association between ischemic skin and an elevated VEGF expression in legs from patients with CLI. These data support that the mechanism for VEGF upregulation in hypoxia conditions is intact and acts appropriately in the ischaemic limbs from patients with CLI.
Collapse
|
35
|
Bleda S, De Haro J, Varela C, Esparza L, Ferruelo A, Acin F. Vascular endothelial growth factor polymorphisms are involved in the late vascular complications in Type II diabetic patients. Diab Vasc Dis Res 2012; 9:68-74. [PMID: 22064697 DOI: 10.1177/1479164111426162] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
AIM To determine the potential genotype differences in the vascular endothelial growth factor (VEGF) gene in diabetic patients, which might explain the difference in terms of the development of clinical vascular complications: great vessels atherosclerosis vs. retinopathy. METHODS Genotyping of the VEFG gene insertion/deletion -2549, the C-2578A and the G+405C polymorphisms was done in 40 diabetic patients (26 with peripheral artery disease (PAD) and 14 with diabetic retinopathy (DR)). RESULTS There was a significant increase in the frequency of the VEGF -2549 DD genotype in PAD patients compared with the DR group (34.6 vs. 0; p = 0.016), as well as in the distribution of the VEGF -2549 ID genotype in DR compared with PAD patients (85.7 vs. 38.4; p = 0.005). There was a significant increase in the frequency of the VEGF -2578 CC genotype in the PAD group compared with DR (34.6 vs. 0; p = 0.016), as well as in the VEGF -2578 CA genotype in DR patients compared with PAD (85.7 vs. 34.6; p = 0.002). The VEGF +405 genotype was not associated with diabetic vascular complications. CONCLUSION This study provides preliminary evidence that VEGF polymorphisms are associated with a differential presentation of diabetic vascular complications.
Collapse
Affiliation(s)
- Silvia Bleda
- Vascular Surgery and Angiology Department, Hospital Universitario Getafe, Carretera de Toledo Km 12.5, 28905 Madrid, Spain.
| | | | | | | | | | | |
Collapse
|
36
|
Exploiting extracellular matrix-stem cell interactions: A review of natural materials for therapeutic muscle regeneration. Biomaterials 2012; 33:428-43. [DOI: 10.1016/j.biomaterials.2011.09.078] [Citation(s) in RCA: 76] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2011] [Accepted: 09/28/2011] [Indexed: 02/07/2023]
|
37
|
|
38
|
Effects of erythropoietin on angiogenesis after myocardial infarction in porcine. Heart Vessels 2011; 27:79-88. [DOI: 10.1007/s00380-011-0197-2] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/27/2010] [Accepted: 11/19/2010] [Indexed: 10/15/2022]
|
39
|
Bronas UG, Treat-Jacobson D. Peripheral Artery Disease in the Elderly: Prevalence, Clinical Implications, and Therapy. CURRENT CARDIOVASCULAR RISK REPORTS 2011. [DOI: 10.1007/s12170-011-0185-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
|
40
|
Yuan QY, Zhu ZW, Wang Z, Wang XM, Li XS, Huang J, Si LY. A novel method of augmenting gene expression and angiogenesis in the normal and ischemic canine myocardium. Heart Vessels 2011; 27:316-26. [PMID: 21688013 DOI: 10.1007/s00380-011-0165-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/31/2010] [Accepted: 04/22/2011] [Indexed: 02/07/2023]
Abstract
This study presents a novel method that direct intramyocardial injection of low-dose plasmid DNA and microbubbles combined with insonation could further augment gene expression in normal and ischemic canine myocardium. Plasmids encoding enhanced green fluorescent protein (pEGFP) and hepatocyte growth factor (pHGF) (500 μg) were individually mixed with 0.5 ml of microbubble solution (MB) and injected into the normal or acute ischemic canine myocardium. The dogs in the plasmid + MB/US group underwent insonation (US). Other dogs were randomly divided into three treatment groups: plasmid and insonation, plasmid and MB injection, and plasmid injection only. The EGFP and HGF mRNA expressions were assessed in the myocardium at the injection site and at sites 0.5 and 1 cm remote from the injection site. Compared to plasmid transfer alone, a mean 13.4-fold enhancement of gene expression was achieved in the EGFP + MB/US group at 48 h (p < 0.01). HGF mRNA expression in ischemic zones was markedly elevated after 28 days, with a mean 9.0-fold enhancement in the HGF + MB/US group (p < 0.01). EGFP protein expression was detected in the normal myocardium at 1 cm remote from the injection site in the EGFP + MB/US group. Similarly, HGF protein expression was detected in the ischemic myocardium at 0.5 cm remote from the injection site in the HGF + MB/US group. These findings indicate that the radius of gene expression was partly extended in the two plasmid + MB/US groups. The capillary density increased from 20.9 ± 5.3/mm(2) in control myocardial infarction dogs without treatment to 126.7 ± 38.2/mm(2) in the HGF + MB/US group (p < 0.01). Taken together, the present data demonstrate that direct intramyocardial injection of an angiogenic gene and microbubbles combined with insonation can augment gene expression and angiogenesis. Consequently, this strategy may be a useful tool for gene therapy of ischemic heart disease.
Collapse
Affiliation(s)
- Qiao-Ying Yuan
- Department of Geriatrics, Southwest Hospital, Third Military Medical University, Gao Tan Yan Street, Sha Ping Ba District, Chongqing, 400038, People's Republic of China
| | | | | | | | | | | | | |
Collapse
|
41
|
Belch J, Hiatt WR, Baumgartner I, Driver IV, Nikol S, Norgren L, Van Belle E. Effect of fibroblast growth factor NV1FGF on amputation and death: a randomised placebo-controlled trial of gene therapy in critical limb ischaemia. Lancet 2011; 377:1929-37. [PMID: 21621834 DOI: 10.1016/s0140-6736(11)60394-2] [Citation(s) in RCA: 245] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
BACKGROUND Patients with critical limb ischaemia have a high rate of amputation and mortality. We tested the hypothesis that non-viral 1 fibroblast growth factor (NV1FGF) would improve amputation-free survival. METHODS In this phase 3 trial (EFC6145/TAMARIS), 525 patients with critical limb ischaemia unsuitable for revascularisation were enrolled from 171 sites in 30 countries. All had ischaemic ulcer in legs or minor skin gangrene and met haemodynamic criteria (ankle pressure <70 mm Hg or a toe pressure <50 mm Hg, or both, or a transcutaneous oxygen pressure <30 mm Hg on the treated leg). Patients were randomly assigned to either NV1FGF at 0·2 mg/mL or matching placebo (visually identical) in a 1:1 ratio. Randomisation was done with a central interactive voice response system by block size 4 and was stratified by diabetes status and country. Investigators, patients, and study teams were masked to treatment. Patients received eight intramuscular injections of their assigned treatment in the index leg on days 1, 15, 29, and 43. The primary endpoint was time to major amputation or death at 1 year analysed by intention to treat with a log-rank test using a multivariate Cox proportional hazard model. This trial is registered with ClinicalTrials.gov, number NCT00566657. FINDINGS 259 patients were assigned to NV1FGF and 266 to placebo. All 525 patients were analysed. The mean age was 70 years (range 50-92), 365 (70%) were men, 280 (53%) had diabetes, and 248 (47%) had a history of coronary artery disease. The primary endpoint or components of the primary did not differ between treatment groups, with major amputation or death in 86 patients (33%) in the placebo group, and 96 (36%) in the active group (hazard ratio 1·11, 95% CI 0·83-1·49; p=0·48). No significant safety issues were recorded. INTERPRETATION TAMARIS provided no evidence that NV1FGF is effective in reduction of amputation or death in patients with critical limb ischaemia. Thus, this group of patients remains a major therapeutic challenge for the clinician. FUNDING Sanofi-Aventis, Paris, France.
Collapse
Affiliation(s)
- Jill Belch
- The Institute of Cardiovascular Research, Vascular and Inflammatory Diseases Research Unit, Ninewells Hospital and Medical School, Dundee, UK.
| | | | | | | | | | | | | |
Collapse
|
42
|
Affiliation(s)
- F Gerry R Fowkes
- Wolfson Unit for Prevention of Peripheral Vascular Diseases, Centre for Population Health Sciences, University of Edinburgh, Edinburgh EH8 9AG, UK.
| | | |
Collapse
|
43
|
Oda M, Toba K, Kato K, Ozawa T, Yanagawa T, Ikarashi N, Takayama T, Suzuki T, Hanawa H, Masuko M, Kobayashi H, Aizawa Y. Hypocellularity and insufficient expression of angiogenic factors in implanted autologous bone marrow in patients with chronic critical limb ischemia. Heart Vessels 2011; 27:38-45. [PMID: 21384270 DOI: 10.1007/s00380-011-0125-5] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/20/2010] [Accepted: 01/28/2011] [Indexed: 01/30/2023]
Abstract
The aim of this study was to identify the clinical parameters of absolutely poor-prognosis patients with chronic critical limb ischemia (AP-CLI). Sixteen no-option CLI patients with arteriosclerosis obliterans: ASO (nine) and non-ASO patients (seven) treated with bone marrow-mononuclear cell implantation (BMI) were analyzed. There were three AP-CLI patients (all ASO). The mRNA expression of several angiogenic factors in the implanted cells was analyzed in comparison with normal donor bone marrow. To observe the response of bone marrow components to hypoxia, normal bone marrow cells were cultured for 24 h in 2.5% O(2), and mRNA expression of angiogenic factors were measured. AP-CLI patients exhibited extraordinary low bone marrow cellularity as well as the percentage of CD34-positive cells. Among angiogenic factors, only VEGF expression was maintained in response to HIF-1, while other factors such as HGF, Ang-1, PLGF, and SDF-1 decreased in the implanted bone marrow cells of the patients with CLI compared to normal bone marrow cells. HIF-1 and all of the five angiogenic factors increased in vitro in response to hypoxia. Thus it is highly likely that angiogenic factors except VEGF do not respond to chronic ischemia in bone marrow in vivo. An organ-protection system against tissue ischemia may be applied for acute hypoxia, but it may be insufficient for chronic ischemia.
Collapse
Affiliation(s)
- Masato Oda
- First Department of Internal Medicine, Niigata University Medical and Dental Hospital, Niigata, Japan
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Stimulation of angiogenesis in rat ischemic limbby intramuscular implantation of mononuclear fraction cells from autologous bone marrow. Bull Exp Biol Med 2011; 150:530-4. [DOI: 10.1007/s10517-011-1183-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
|
45
|
Idei N, Soga J, Hata T, Fujii Y, Fujimura N, Mikami S, Maruhashi T, Nishioka K, Hidaka T, Kihara Y, Chowdhury M, Noma K, Taguchi A, Chayama K, Sueda T, Higashi Y. Autologous bone-marrow mononuclear cell implantation reduces long-term major amputation risk in patients with critical limb ischemia: a comparison of atherosclerotic peripheral arterial disease and Buerger disease. Circ Cardiovasc Interv 2011; 4:15-25. [PMID: 21205941 DOI: 10.1161/circinterventions.110.955724] [Citation(s) in RCA: 92] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
BACKGROUND Bone-marrow mononuclear cell (BM-MNC) implantation improves ischemic symptoms in patients with critical limb ischemia (CLI). The purpose of this study was to evaluate long-term clinical outcomes after autologous BM-MNC implantation in patients with CLI. METHODS AND RESULTS We assessed long-term clinical outcomes after BM-MNC implantation in 51 patients with CLI, including 25 patients with peripheral arterial disease (PAD) and 26 patients with Buerger disease. Forty-six CLI patients who had no BM-MNC implantation served as control subjects. Median follow-up period was 4.8 years. The 4-year amputation-free rates after BM-MNC implantation were 48% in PAD patients and 95% in Buerger disease, and they were 0% in control PAD patients and 6% in control Buerger disease. The 4-year overall survival rates after BM-MNC implantation were 76% in PAD patients and 100% in Buerger disease, and they were 67% in control PAD patients and 100% in control Buerger disease. Multivariable Cox proportional hazards analysis revealed that BM-MNC implantation correlated with prevention of major amputation and that hemodialysis and diabetes mellitus correlated with major amputation. In Buerger disease, ankle brachial pressure index and transcutaneous oxygen pressure were significantly increased after 1 month and remained high during 3-year follow-up. However, in patients with PAD, ankle brachial pressure index and transcutaneous oxygen pressure significantly increased after 1 month and gradually decreased during 3-year follow-up and returned to baseline levels. CONCLUSIONS These findings suggest that BM-MNC implantation is safe and effective in patients with CLI, especially in patients with Buerger disease. Clinical Trial Registration- URL: http://home.hiroshima-u.ac.jp/angio/. Unique identifier: 001769.
Collapse
Affiliation(s)
- Naomi Idei
- Department of Cardiovascular Medicine, Graduate School of Biomedical Sciences, Hiroshima University, Hiroshima, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Kim HG, Choi OH. Neovascularization in a mouse model via stem cells derived from human fetal amniotic membranes. Heart Vessels 2010; 26:196-205. [PMID: 21188388 DOI: 10.1007/s00380-010-0064-6] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/12/2009] [Accepted: 05/28/2010] [Indexed: 01/16/2023]
Abstract
In this study, we evaluated the effect of culture-expanded mesenchymal stem cells (MSCs), derived from amniotic membranes, on neovascularization and blood flow, in an animal model of limb ischemia in immune-deficient mice. MSCs were cultured from human amniotic membranes by collagenase digestion. Human amniotic mesenchymal stem cells (hAMSCs) were administered intramuscularly at three different sites of the ischemic leg whose femoral vessels were ligated. After 4 weeks of culture, a population of homogeneous mesenchymal cells was isolated from the human amniotic membranes after confluence was reached. We performed three different groups of mice model [controls, hAMSCs, conditioned media from the hAMSCs (hAMSCs-CM)]. The blood flow recovery in the hindlimb ischemia model was significantly higher in the hAMSC-transplanted group than in the control group. Moreover, hAMSCs-CM significantly improved the cutaneous blood flow. The histological examination showed that red fluorescence (CM-DiI)-labeled hAMSCs was detected in the interstitial tissues between the muscle fibers 2 weeks after transplantation. The results of this study showed that hAMSCs may be an attractive, alternative source of progenitor or stem cells for basic research as well as clinical applications.
Collapse
Affiliation(s)
- Hwi Gon Kim
- Department of Obstetrics and Gynecology, Pusan National University Yangsan Hospital, Beomeo-ri, Mulgeum-eup, Yangsan, Gyeongnam, 626-770, Korea
| | | |
Collapse
|
47
|
AHR, a novel acute hypoxia-response sequence, drives reporter gene expression under hypoxiain vitroandin vivo. Cell Biol Int 2010; 35:1-8. [DOI: 10.1042/cbi20100290] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
|
48
|
Pattillo CB, Bir S, Rajaram V, Kevil CG. Inorganic nitrite and chronic tissue ischaemia: a novel therapeutic modality for peripheral vascular diseases. Cardiovasc Res 2010; 89:533-41. [PMID: 20851809 DOI: 10.1093/cvr/cvq297] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
Ischaemic tissue damage represents the ultimate form of tissue pathophysiology due to cardiovascular disease, which is the leading cause of morbidity and mortality across the globe. A significant amount of basic research and clinical investigation has been focused on identifying cellular and molecular pathways to alleviate tissue damage and dysfunction due to ischaemia and subsequent reperfusion. Over many years, the gaseous molecule nitric oxide (NO) has emerged as an important regulator of cardiovascular health as well as protector against tissue ischaemia and reperfusion injury. However, clinical translation of NO therapy for these pathophysiological conditions has not been realized for various reasons. Work from our laboratory and several others suggests that a new form of NO-associated therapy may be possible through the use of nitrite anion (sodium nitrite), a prodrug which can be reduced to NO in ischaemic tissues. In this manner, nitrite anion serves as a highly selective NO donor in ischaemic tissues without substantially altering otherwise normal tissue. This surprising and novel discovery has reinvigorated hopes for effectively restoring NO bioavailability in vulnerable tissues while continuing to reveal the complexity of NO biology and metabolism within the cardiovascular system. However, some concerns may exist regarding the effect of nitrite on carcinogenesis. This review highlights the emergence of nitrite anion as a selective NO prodrug for ischaemic tissue disorders and discusses the potential therapeutic utility of this agent for peripheral vascular disease.
Collapse
Affiliation(s)
- Christopher B Pattillo
- Department of Pathology and Cardiology, LSU Health Sciences Center-Shreveport, 1501 Kings Hwy, Shreveport, LA 71130, USA
| | | | | | | |
Collapse
|
49
|
Gene delivery of soluble vascular endothelial growth factor receptor-1 (sFlt-1) inhibits intra-plaque angiogenesis and suppresses development of atherosclerotic plaque. Clin Exp Med 2010; 11:113-21. [DOI: 10.1007/s10238-010-0112-7] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2010] [Accepted: 08/25/2010] [Indexed: 02/05/2023]
|
50
|
The Next 10 years in the Management of Peripheral Artery Disease: Perspectives from The ‘PAD 2009’ Conference. Eur J Vasc Endovasc Surg 2010; 40:375-80. [DOI: 10.1016/j.ejvs.2010.05.005] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2010] [Accepted: 05/12/2010] [Indexed: 11/22/2022]
|