1
|
Lee JJ, Arpino JM, Yin H, Nong Z, Szpakowski A, Hashi AA, Chevalier J, O'Neil C, Pickering JG. Systematic Interrogation of Angiogenesis in the Ischemic Mouse Hind Limb: Vulnerabilities and Quality Assurance. Arterioscler Thromb Vasc Biol 2020; 40:2454-2467. [PMID: 32787524 PMCID: PMC7505144 DOI: 10.1161/atvbaha.120.315028] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Supplemental Digital Content is available in the text. Objective: There has been little success in translating preclinical studies of mouse hind limb ischemia into benefit for patients with peripheral artery disease. Using systematic strategies, we sought to define the injury and angiogenesis landscapes in mice subjected to hind limb ischemia and ascertain whether published studies to date have used an analysis strategy concordant with these data. Approach and Results: Maps of ischemic injury were generated from 22 different hind limb muscles and 33 muscle territories in 12-week-old C57BL/6 mice, based on loss or centralization of myofiber nuclei. Angiogenesis was similarly mapped based on CD (cluster of differentiation) 31–positive capillary content. Only 10 of 33 muscle territories displayed consistent muscle injury, with the distal anterior hind limb muscles most reliably injured. Angiogenesis was patchy and exclusively associated with zones of regenerated muscle (central nuclei). Angiogenesis was not observed in normal appearing muscle, necrotic muscle, or injury border zones. Systematic review of mouse hind limb angiogenesis studies identified 5147 unique publications, of which 509 met eligibility criteria for analysis. Only 7% of these analyzed manuscripts evaluated angiogenesis in distal anterior hind limb muscles and only 15% consistently examined for angiogenesis in zones of muscle regeneration. Conclusions: In 12-week C57BL/6 mice, angiogenesis postfemoral artery excision proceeds exclusively in zones of muscle regeneration. Only a minority of studies to date have analyzed angiogenesis in regions of demonstrably regenerating muscle or in high-likelihood territories. Quality assurance standards, informed by the atlas and mapping data herein, could augment data reliability and potentially help translate mouse hind limb ischemia studies to patient care.
Collapse
Affiliation(s)
- Jason J Lee
- Robarts Research Institute (J.J.L., J.-M.A., H.Y., Z.N., A.S., J.C., C.O., J.G.P.), Western University, London, Ontario, Canada.,Department of Medicine (J.J.L., A.A.H., J.G.P.), Western University, London, Ontario, Canada.,Department of Medical Biophysics (J.J.L., J.-M.A., J.C., J.G.P.), Western University, London, Ontario, Canada
| | - John-Michael Arpino
- Robarts Research Institute (J.J.L., J.-M.A., H.Y., Z.N., A.S., J.C., C.O., J.G.P.), Western University, London, Ontario, Canada.,Department of Medical Biophysics (J.J.L., J.-M.A., J.C., J.G.P.), Western University, London, Ontario, Canada
| | - Hao Yin
- Robarts Research Institute (J.J.L., J.-M.A., H.Y., Z.N., A.S., J.C., C.O., J.G.P.), Western University, London, Ontario, Canada
| | - Zengxuan Nong
- Robarts Research Institute (J.J.L., J.-M.A., H.Y., Z.N., A.S., J.C., C.O., J.G.P.), Western University, London, Ontario, Canada
| | - Alexis Szpakowski
- Robarts Research Institute (J.J.L., J.-M.A., H.Y., Z.N., A.S., J.C., C.O., J.G.P.), Western University, London, Ontario, Canada
| | - Abdulaziz A Hashi
- Department of Medicine (J.J.L., A.A.H., J.G.P.), Western University, London, Ontario, Canada
| | - Jacqueline Chevalier
- Robarts Research Institute (J.J.L., J.-M.A., H.Y., Z.N., A.S., J.C., C.O., J.G.P.), Western University, London, Ontario, Canada.,Department of Medical Biophysics (J.J.L., J.-M.A., J.C., J.G.P.), Western University, London, Ontario, Canada
| | - Caroline O'Neil
- Robarts Research Institute (J.J.L., J.-M.A., H.Y., Z.N., A.S., J.C., C.O., J.G.P.), Western University, London, Ontario, Canada
| | - J Geoffrey Pickering
- Robarts Research Institute (J.J.L., J.-M.A., H.Y., Z.N., A.S., J.C., C.O., J.G.P.), Western University, London, Ontario, Canada.,Department of Medicine (J.J.L., A.A.H., J.G.P.), Western University, London, Ontario, Canada.,Department of Medical Biophysics (J.J.L., J.-M.A., J.C., J.G.P.), Western University, London, Ontario, Canada.,Department of Biochemistry (J.G.P.), Western University, London, Ontario, Canada
| |
Collapse
|
2
|
Abstract
Along with the progress of global aging, the prognosis of severe ischemic heart disease (IHD) remains poor, and thus the development of effective angiogenic therapy remains an important clinical unmet need. We have developed low-energy extracorporeal cardiac shock wave therapy as an innovative minimally invasive angiogenic therapy and confirmed its efficacy in a porcine chronic myocardial ischemia model in animal experiments as well as in patients with refractory angina. Since ultrasound is more advantageous for clinical application than shock waves, we then aimed to develop ultrasound therapy for IHD. We demonstrated that specific conditions of low-intensity pulsed ultrasound (LIPUS) therapy improve myocardial ischemia in animal models through the enhancement of angiogenesis mediated by endothelial mechanotransduction. To examine the effectiveness of our LIPUS therapy in patients with severe angina pectoris, we are now conducting a prospective multicenter clinical trial in Japan. Furthermore, to overcome the current serious situation of dementia pandemic but with no effective treatments worldwide, we have recently demonstrated that our LIPUS therapy also improves cognitive impairment in mouse models of Alzheimer's disease and vascular dementia. Here, we summarize the progress in our studies to develop angiogenic therapies with sound waves.
Collapse
Affiliation(s)
- Tomohiko Shindo
- Department of Cardiovascular Medicine, Tohoku University Graduate School of Medicine, Sendai, Miyagi, Japan
| | - Hiroaki Shimokawa
- Department of Cardiovascular Medicine, Tohoku University Graduate School of Medicine, Sendai, Miyagi, Japan
| |
Collapse
|
3
|
Gastrointestinal cancer cells treatment with bevacizumab activates a VEGF autoregulatory mechanism involving telomerase catalytic subunit hTERT via PI3K-AKT, HIF-1α and VEGF receptors. PLoS One 2017; 12:e0179202. [PMID: 28594907 PMCID: PMC5466359 DOI: 10.1371/journal.pone.0179202] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2016] [Accepted: 05/25/2017] [Indexed: 11/29/2022] Open
Abstract
Background Targeting angiogenesis has been considered a promising treatment of choice for a large number of malignancies, including gastrointestinal cancers. Bevacizumab is an anti-vascular endothelial growth factor (anti-VEGF) being used for this purpose. However, treatment efficacy is largely questioned. Telomerase activity, responsible for cancer cell immortality, is detected in 85–95% of human cancers and is considered a potential regulator of VEGF. The aim of our study was to investigate the interrelationship between VEGF and hTERT in gastrointestinal cancers and to explore cell response to a combined inhibition of telomerase and VEGF. Methods AGS (gastric cancer), Caco-2 (colorectal cancer) and HepG2/C3A (hepatocellular carcinoma), were treated with telomerase inhibitors BIBR-1232 (10μM) and costunolide (10μM), with bevacizumab (Avastin® at 5 ng/ml or 100μg/ml) or with a combination of both types of inhibitors. VEGF and hTERT mRNA levels, and telomerase activity were detected by RT-PCR. VEGF levels were quantified by ELISA. Telomerase was knocked down using hTERT siRNA and hTERT was overexpressed in the telomerase negative cell line, Saos-2 (osteosarcoma), using constructs expressing either wild type hTERT (hTERT-WT) or dominant negative hTERT (hTERT-DN). Tube formation by HUVECs was assessed using ECMatrix™ (EMD Millipore). Results Our results showed that telomerase regulates VEGF expression and secretion through its catalytic subunit hTERT in AGS, Caco2, and HepG2/C3A, independent of its catalytic activity. Interestingly, VEGF inhibition with bevacizumab (100μg/ml) increased hTERT expression 42.3% in AGS, 94.1% in Caco2, and 52.5% in HepG2/C3A, and increased telomerase activity 30-fold in AGS, 10.3-fold in Caco2 and 8-fold in HepG2/C3A. A further investigation showed that VEGF upregulates hTERT expression in a mechanism that implicates the PI3K/AKT/mTOR pathway and HIF-1α. Moreover, bevacizumab treatment increased VEGFR1 and VEGFR2 expression in cancer cells and human umbilical vein endothelial cells (HUVECs) through hTERT. Thus, the combination of bevacizumab with telomerase inhibitors decreased VEGF expression and secretion by cancer cells, inhibited VEGFR1 and VEGFR2 upregulation, and reduced tube formation by HUVECs. Conclusions Taken together, our results suggest that bevacizumab treatment activates a VEGF autoregulatory mechanism involving hTERT and VEGF receptors and that an inhibition of this pathway could improve tumor cell response to anti-VEGF treatment.
Collapse
|
4
|
Zaporozhets T, Besednova N. Prospects for the therapeutic application of sulfated polysaccharides of brown algae in diseases of the cardiovascular system: review. PHARMACEUTICAL BIOLOGY 2016; 54:3126-3135. [PMID: 27252012 DOI: 10.1080/13880209.2016.1185444] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/02/2015] [Revised: 12/22/2015] [Accepted: 04/27/2016] [Indexed: 06/05/2023]
Abstract
CONTEXT Fucoidans are water-soluble, highly sulfated, branched homo- and hetero-polysaccharides derived from the fibrillar cell walls and intercellular spaces of brown seaweeds of the class Phaeophyceae. Fucoidans possess mimetic properties of the natural ligands of protein receptors and regulate functions of biological systems via key signaling molecules. OBJECTIVES The aim of this review was to collect and combine all available scientific literature about the potential use of the fucoidans for diseases of cardiovascular system. MATERIALS AND METHODS The review has been compiled using references from major databases such as Web of Science, PubMed, Scopus, Elsevier, Springer and Google Scholar (up to September 2015). After obtaining all reports from database (a total number is about 580), the papers were carefully analyzed in order to find data related to the topic of this review (129 references). RESULTS An exhaustive survey of literature revealed that fucoidans possess a broad spectrum of biological activity, including anti-coagulant, hypolipidemic, anti-thrombotic, anti-inflammatory, immunomodulatory, anti-tumor, anti-adhesive and anti-hypertensive properties. Numerous investigations of fucoidans in diseases of the cardiovascular system mainly focus on pleiotropic anti-inflammatory effects. Fucoidans also possess pro-angiogenic and pro-vasculogenic properties. CONCLUSION A great number of investigations in the past years have demonstrated that fucoidans has great potential for in-depth investigation of their effects on cardiovascular system. Through this review, the authors hope to attract the attention of researchers to use fucoidan as mimetic of natural ligand receptor protein with the view of developing new formulations with an improved therapeutic value.
Collapse
Affiliation(s)
- Tatyana Zaporozhets
- a Somov Institute of Epidemiology and Microbiology , Vladivostok , Russian Federation
| | - Natalia Besednova
- a Somov Institute of Epidemiology and Microbiology , Vladivostok , Russian Federation
| |
Collapse
|
5
|
Powell RJ, Dormandy J, Simons M, Morishita R, Annex BH. Therapeutic angiogenesis for critical limb ischemia: design of the hepatocyte growth factor therapeutic angiogenesis clinical trial. Vasc Med 2016; 9:193-8. [PMID: 15675184 DOI: 10.1191/1358863x04vm557oa] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
The objective of the HGF-STAT clinical trial is to determine whether perfusion can be improved by gene transfer with a plasmid DNA containing hepatocyte growth factor (HGF) in the affected limb of patients with unreconstructable critical limb ischemia (CLI). CLI results in a high rate of limb loss and impaired quality of life. The current therapeutic strategies, including bypass surgery and percutaneous interventions, are only successful in treating a subset of patients. Therapeutic angiogenesis is an investigational method that seeks to favorably impact tissue per-fusion in CLI. HGF-STAT is a double-blind, parallel-group, placebo-controlled, dose response study in 100 patients with unreconstructable CLI. Eligible subjects will be randomized 1:1:1:1 to receive saline placebo or one of three dose/regimens of HGF plasmid DNA. The selection of outcome measures, including the primary endpoint, and changes in transcutaneous oxygen pressure (TcPO2) from baseline to 3 months will be discussed. In conclusion, this study will help to determine whether therapeutic angiogenesis with HGF is a viable option in the treatment of patients with CLI.
Collapse
Affiliation(s)
- Richard J Powell
- Division of Vascular Surgery, Department of Surgery, Dartmouth-Hitchcock Medical Center, Lebanon, NH, USA.
| | | | | | | | | |
Collapse
|
6
|
Katz MG, Fargnoli AS, Kendle AP, Hajjar RJ, Bridges CR. Gene Therapy in Cardiac Surgery: Clinical Trials, Challenges, and Perspectives. Ann Thorac Surg 2016; 101:2407-16. [PMID: 26801060 PMCID: PMC4987708 DOI: 10.1016/j.athoracsur.2015.12.004] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/01/2015] [Revised: 10/24/2015] [Accepted: 12/07/2015] [Indexed: 12/28/2022]
Abstract
The concept of gene therapy was introduced in the 1970s after the development of recombinant DNA technology. Despite the initial great expectations, this field experienced early setbacks. Recent years have seen a revival of clinical programs of gene therapy in different fields of medicine. There are many promising targets for genetic therapy as an adjunct to cardiac surgery. The first positive long-term results were published for adenoviral administration of vascular endothelial growth factor with coronary artery bypass grafting. In this review we analyze the past, present, and future of gene therapy in cardiac surgery. The articles discussed were collected through PubMed and from author experience. The clinical trials referenced were found through the Wiley clinical trial database (http://www.wiley.com/legacy/wileychi/genmed/clinical/) as well as the National Institutes of Health clinical trial database (Clinicaltrials.gov).
Collapse
Affiliation(s)
- Michael G Katz
- Sanger Heart and Vascular Institute, Charlotte, North Carolina; Mount Sinai School of Medicine, New York, New York
| | - Anthony S Fargnoli
- Sanger Heart and Vascular Institute, Charlotte, North Carolina; Mount Sinai School of Medicine, New York, New York
| | - Andrew P Kendle
- Sanger Heart and Vascular Institute, Charlotte, North Carolina
| | | | | |
Collapse
|
7
|
Allipour Birgani S, Mailänder M, Wasle I, Dietrich H, Gruber J, Distler O, Sgonc R. Efficient therapy of ischaemic lesions with VEGF121-fibrin in an animal model of systemic sclerosis. Ann Rheum Dis 2015; 75:1399-406. [PMID: 26362758 PMCID: PMC4766736 DOI: 10.1136/annrheumdis-2015-207548] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2015] [Accepted: 08/01/2015] [Indexed: 11/17/2022]
Abstract
Background In systemic sclerosis (SSc), chronic and uncontrolled overexpression of vascular endothelial growth factor (VEGF) results in chaotic vessels, and intractable fingertip ulcers. Vice versa, VEGF is a potent mediator of angiogenesis if temporally and spatially controlled. We have addressed this therapeutic dilemma in SSc by a novel approach using a VEGF121 variant that covalently binds to fibrin and gets released on demand by cellular enzymatic activity. Using University of California at Davis (UCD)-206 chickens, we tested the hypothesis that cell-demanded release of fibrin-bound VEGF121 leads to the formation of stable blood vessels, and clinical improvement of ischaemic lesions. Methods Ninety-one early and late ischaemic comb and neck skin lesions of UCD-206 chickens were treated locally with VEGF121-fibrin, fibrin alone, or left untreated. After 1 week of treatment the clinical outcome was assessed. Angiogenesis was studied by immunofluorescence staining of vascular markers quantitatively analysed using TissueQuest. Results Overall, 79.3% of the lesions treated with VEGF121-fibrin showed clinical improvement, whereas 71.0% of fibrin treated controls, and 93.1% of untreated lesions deteriorated. This was accompanied by significantly increased growth of stable microvessels, upregulation of the proangiogenic VEGFR-2 and its regulator TAL-1, and increase of endogenous endothelial VEGF expression. Conclusions Our findings in the avian model of SSc suggest that cell-demanded release of VEGF121 from fibrin matrix induces controlled angiogenesis by differential regulation of VEGFR-1 and VEGFR-2 expression, shifting the balance towards the proangiogenic VEGFR-2. The study shows the potential of covalently conjugated VEGF-fibrin matrices for the therapy of ischaemic lesions such as fingertip ulcers.
Collapse
Affiliation(s)
- Shadab Allipour Birgani
- Division of Experimental Pathophysiology and Immunology, Biocenter, Medical University of Innsbruck, Innsbruck, Austria
| | - Marion Mailänder
- Division of Experimental Pathophysiology and Immunology, Biocenter, Medical University of Innsbruck, Innsbruck, Austria
| | - Ines Wasle
- Division of Experimental Pathophysiology and Immunology, Biocenter, Medical University of Innsbruck, Innsbruck, Austria
| | - Hermann Dietrich
- Central Laboratory Animal Facilities, Medical University of Innsbruck, Innsbruck, Austria
| | - Johann Gruber
- Department of Internal Medicine VI, Medical University of Innsbruck, Innsbruck, Austria
| | - Oliver Distler
- Department of Rheumatology, Center of Experimental Rheumatology, University Hospital Zurich, Zurich, Switzerland
| | - Roswitha Sgonc
- Division of Experimental Pathophysiology and Immunology, Biocenter, Medical University of Innsbruck, Innsbruck, Austria
| |
Collapse
|
8
|
Rege A, Seifert AC, Schlattman D, Ouyang Y, Li KW, Basaldella L, Brem H, Tyler BM, Thakor NV. Longitudinal in vivo monitoring of rodent glioma models through thinned skull using laser speckle contrast imaging. JOURNAL OF BIOMEDICAL OPTICS 2012; 17:126017. [PMID: 23235836 PMCID: PMC3519490 DOI: 10.1117/1.jbo.17.12.126017] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/07/2012] [Revised: 11/08/2012] [Accepted: 11/09/2012] [Indexed: 06/01/2023]
Abstract
Laser speckle contrast imaging (LSCI) is a contrast agent free imaging technique suited for longitudinal assessment of vascular remodeling that accompanies brain tumor growth. We report the use of LSCI to monitor vascular changes in a rodent glioma model. Ten rats are inoculated with 9L gliosarcoma cells, and the angiogenic response is monitored five times over two weeks through a thinned skull imaging window. We are able to visualize neovascularization and measure the number of vessels per unit area to assess quantitatively the microvessel density (MVD). Spatial spread of MVD reveals regions of high MVD that may correspond to tumor location. Whole-field average MVD values increase with time in the tumor group but are fairly stable in the control groups. Statistical analysis shows significant differences in MVD values between the tumor group and both saline-receiving and unperturbed control groups over the two-week period (p<0.05). In conclusion, LSCI is suitable for investigation of tumor angiogenesis in rodent models. In addition, the statistical difference (p<0.02) between MVD values of the tumor (24.40 ± 1.41) and control groups (15.40 ± 1.60) on the 14th day after inoculation suggests a potential use of LSCI in the clinic in distinguishing tumor environments from normal vasculature.
Collapse
Affiliation(s)
- Abhishek Rege
- Johns Hopkins University, Department of Biomedical Engineering, Baltimore, Maryland
| | | | - Dan Schlattman
- Infinite Biomedical Technologies LLC, Baltimore, Maryland
| | - Yu Ouyang
- Infinite Biomedical Technologies LLC, Baltimore, Maryland
| | - Khan W. Li
- Johns Hopkins University, Department of Neurosurgery, Baltimore, Maryland
| | - Luca Basaldella
- Johns Hopkins University, Department of Neurosurgery, Baltimore, Maryland
| | - Henry Brem
- Johns Hopkins University, Department of Biomedical Engineering, Baltimore, Maryland
- Johns Hopkins University, Department of Neurosurgery, Baltimore, Maryland
- Johns Hopkins University, Departments of Oncology and Ophthalmology, Baltimore, Maryland
| | - Betty M. Tyler
- Johns Hopkins University, Department of Neurosurgery, Baltimore, Maryland
| | - Nitish V. Thakor
- Johns Hopkins University, Department of Biomedical Engineering, Baltimore, Maryland
- Infinite Biomedical Technologies LLC, Baltimore, Maryland
| |
Collapse
|
9
|
Araki S, Izumiya Y, Hanatani S, Rokutanda T, Usuku H, Akasaki Y, Takeo T, Nakagata N, Walsh K, Ogawa H. Akt1-mediated skeletal muscle growth attenuates cardiac dysfunction and remodeling after experimental myocardial infarction. Circ Heart Fail 2011; 5:116-25. [PMID: 22135402 DOI: 10.1161/circheartfailure.111.964783] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND It is appreciated that aerobic endurance exercise can attenuate unfavorable myocardial remodeling following myocardial infarction. In contrast, little is known about the effects of increasing skeletal muscle mass, typically achieved through resistance training, on this process. Here, we utilized transgenic (TG) mice that can induce the growth of functional skeletal muscle by switching Akt1 signaling in muscle fibers to assess the impact of glycolytic muscle growth on post-myocardial infarction cardiac remodeling. METHODS AND RESULTS Male-noninduced TG mice and their nontransgenic littermates (control) were subjected to left anterior coronary artery ligation. Two days after surgery, mice were provided doxycycline in their drinking water to activate Akt1 transgene expression in a skeletal muscle-specific manner. Myogenic Akt1 activation led to diminished left ventricular dilation and reduced contractile dysfunction compared with control mice. Improved cardiac function in Akt1 TG mice was coupled to diminished myocyte hypertrophy, decreased interstitial fibrosis, and increased capillary density. ELISA and protein array analyses demonstrated that serum levels of proangiogenic growth factors were upregulated in Akt1 TG mice compared with control mice. Cardiac eNOS was activated in Akt1 TG mice after myocardial infarction. The protective effect of skeletal muscle Akt activation on cardiac remodeling and systolic function was abolished by treatment with the eNOS inhibitor l-NAME. CONCLUSIONS Akt1-mediated skeletal muscle growth attenuates cardiac remodeling after myocardial infarction and is associated with an increased capillary density in the heart. This improvement appears to be mediated by skeletal muscle to cardiac communication, leading to activation of eNOS-signaling in the heart.
Collapse
Affiliation(s)
- Satoshi Araki
- Department of Cardiovascular Medicine, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
10
|
Powerski M, Henrich D, Sander A, Teiler A, Marzi I. In vitro manipulation of endothelial progenitor cell adhesion to vascular endothelium and extracellular matrix by the phorbol ester PMA. ARTIFICIAL CELLS, BLOOD SUBSTITUTES, AND IMMOBILIZATION BIOTECHNOLOGY 2011; 39:214-22. [PMID: 21314293 DOI: 10.3109/10731199.2011.555838] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Injection of endothelial progenitor cells (EPCs) into arteries for cell therapy is a promising field in regenerative medicine. However, adhesion of EPCs during capillary passage is restricted, and non-adhering cells are lost into circulation. Here we demonstrate that it is possible to achieve a three- to sevenfold higher rate of EPC adhesion to endothelium and extracellular matrix molecules after short-term activation with phorbol myristate acetate (PMA). In addition, differentiation and toxicity analyses of PMA activated EPCs showed no impact on cell differentiation and negligible impact on cell survival.
Collapse
Affiliation(s)
- Maciej Powerski
- Department of Trauma Surgery, Johann Wolfgang Goethe University, Frankfurt/Main, Germany.
| | | | | | | | | |
Collapse
|
11
|
Layman H, Li X, Nagar E, Vial X, Pham SM, Andreopoulos FM. Enhanced angiogenic efficacy through controlled and sustained delivery of FGF-2 and G-CSF from fibrin hydrogels containing ionic-albumin microspheres. JOURNAL OF BIOMATERIALS SCIENCE-POLYMER EDITION 2010; 23:185-206. [PMID: 21192837 DOI: 10.1163/092050610x546417] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Neo-vessel formation in ischemic tissues relies on numerous growth factors and cell fractions for the formation of mature, stable, functional vasculature. However, the efforts to regenerate tissues typically rely on the administration of a single growth factor or cells alone. Conversely, polymeric matrices have been investigated extensively to deliver multiple growth factors at pre-determined rates to form stable blood vessels in ischemic tissues. We report on a novel sequential delivery system of a fibrin hydrogel containing ionic-albumin microspheres that allows for the controlled release of two growth factors. The use of this system was investigated in the context of therapeutic angiogenesis. Material properties were determined based on degree of swelling measurements and degradation characteristics. Release kinetics of model angiogenic polypeptides FGF-2 and G-CSF were determined using ELISA and the bioactivity of released protein was evaluated in human endothelial cell cultures. The release of growth factors from ionic-albumin microspheres was significantly delayed compared to the growth factor released from fibrin matrices in the absence of spheres. The scaffolds were implanted in a murine critical limb ischemia model at two concentrations, 40 ng (low) and 400 ng (high), restoring 92% of the blood flow in a normally perfused limb using a fibrin hydrogel releasing FGF-2 containing albumin-PLL microspheres releasing G-CSF (measured by LDPI at the high concentration), a 3.2-fold increase compared to untreated limbs. The extent of neo-vessel formation was delineated by immunohistochemical staining for capillary density (CD-31+) and mature vessel formation (α-SMA+). In conclusion, our study demonstrated that the release kinetics from our scaffold have distinct kinetics previously unpublished and the delivery of these factors resulted in hindlimb reperfusion, and robust capillary and mature vessel formation after 8 weeks compared to either growth factor alone or bolus administration of growth factor.
Collapse
Affiliation(s)
- Hans Layman
- Department of Surgery, University of California at San Francisco, San Francisco, CA, USA
| | | | | | | | | | | |
Collapse
|
12
|
Powerski MJ, Henrich D, Sander A, Wastl D, Ludwig K, Marzi I. CD133+CD34+ stem cells are mobilized after musculoskeletal surgery and target endothelium activated by surgical wound fluid. Langenbecks Arch Surg 2010; 396:379-87. [PMID: 20213459 DOI: 10.1007/s00423-010-0626-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2010] [Accepted: 02/17/2010] [Indexed: 12/31/2022]
Abstract
PURPOSE CD133+CD34+ hematopoietic stem cells (HSCs) have been shown to differentiate into cell types of nonhematopoietic lineage. It is unclear whether HSCs target and repair damaged musculoskeletal tissue. We aimed to analyze if HSCs are mobilized after musculoskeletal surgery to circulation, home to surgical wound fluid (SWF)-activated endothelium, and are chemoattracted by SWF under in vitro conditions. METHODS Circulating HSC levels were measured at t = 3, 8, 24, 48 h postoperatively using fluorescence-activated cell sorting (FACS) and compared with preoperative levels (t = 0) and normal volunteers. For adhesion experiments, HSCs were incubated on SWF-activated human umbilical vein endothelial cells (HUVECs) and HSC/HUVEC ratios determined by FACS. Adhesion receptor expression on HSC (L-selectin, lymphocyte function-associated antigen 1 (LFA-1), very late antigen-4) and SWF-activated HUVECs (P-selectin, E-selectin, V-cell adhesion molecules (CAM), I-CAM) was determined and HSC adhesion measured again after blocking upregulated receptors. Using a modified Boyden chamber, HSC chemotaxis was analyzed for an SWF and cytokine-neutralized SWF (vascular endothelial growth factor (VEGF), stromal-derived factor-1, interleukin-8) gradient. RESULTS Circulating HSCs were significantly increased 8 h after surgery. Increasing HSC adhesion to HUVECs was shown for SWF isolated at any postoperative time point, and chemoattraction was significantly induced in an SWF gradient with SWF isolated 8 and 24 h postoperatively. Receptor and cytokine blockade experiments with monoclonal antibodies revealed decreased HSC adhesion to SWF-activated endothelium and showed lower chemotaxis after blocking the LFA-1-I-CAM-1 receptor axis (adhesion) and neutralizing VEGF-165 (chemotaxis). CONCLUSIONS Our data demonstrate that HSCs are mobilized after trauma, target to wound-associated endothelium via the LFA-1-I-CAM-1 axis, and are chemoattracted by VEGF-165 under in vitro conditions.
Collapse
Affiliation(s)
- Maciej Janusz Powerski
- Department of Trauma Surgery, Johann Wolfgang Goethe University, Frankfurt am Main, Germany.
| | | | | | | | | | | |
Collapse
|
13
|
Lara-Hernández R, Lozano-Vilardell P, Cordobés-Gual J. [Novel therapies of non-revascularizing peripheral arterial occlusive disease: therapeutic angiogenesis]. Med Clin (Barc) 2009; 131:665-9. [PMID: 19087795 DOI: 10.1157/13128727] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Critical limb ischemia is the end stage of peripheral arterial occlusive disease, with a deep impact in patient's quality of life. In some patients, there is no revascularizing treatment options, that determines major limb amputation in a high percentage of patients, not only for uncontrolled limb pain but also for complications of the trophic lesions. In the last years, several studies have shown the possibility to increase the perfusion in the ischemic tissue, by recombinant proteins, gene therapy or cellular therapy, all of them known as therapeutic angiogenesis. Several good results have been published but the way of treatment, doses and possible adverse effects still lack definitive conclusions. Randomized comparative studies should be carried out to determine the best treatment option.
Collapse
Affiliation(s)
- Raúl Lara-Hernández
- Servicio de Angiología y Cirugía Vascular. Hospital Universitario Arnau de Vilanova. Institut de Recerca Biomédica de Catalunya. Lleida. España.
| | | | | |
Collapse
|
14
|
Ouchi N, Oshima Y, Ohashi K, Higuchi A, Ikegami C, Izumiya Y, Walsh K. Follistatin-like 1, a secreted muscle protein, promotes endothelial cell function and revascularization in ischemic tissue through a nitric-oxide synthase-dependent mechanism. J Biol Chem 2008; 283:32802-11. [PMID: 18718903 DOI: 10.1074/jbc.m803440200] [Citation(s) in RCA: 239] [Impact Index Per Article: 14.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Myogenic Akt signaling coordinates blood vessel recruitment with normal tissue growth. Here, we investigated the role of Follistatin-like 1 (Fstl1) in the regulation of endothelial cell function and blood vessel growth in muscle. Transgenic Akt1 overexpression in skeletal muscle led to myofiber growth that was coupled to an increase in muscle capillary density. Myogenic Akt signaling or ischemic hind limb surgery led to the induction of Fstl1 in muscle and increased circulating levels of Fstl1. Intramuscular administration of an adenoviral vector expressing Fstl1 (Ad-Fstl1) accelerated flow recovery and increased capillary density in the ischemic hind limbs of wild-type mice, and this was associated with an increase in endothelial nitric oxide synthase (eNOS) phosphorylation at residue Ser-1179. In cultured endothelial cells, Ad-Fstl1 stimulated migration and differentiation into network structures and inhibited apoptosis under conditions of serum deprivation. These cell responses were associated with the activating phosphorylation of Akt and eNOS. Conversely, transduction with dominant-negative Akt or LY294002 blocked Fstl1-stimulated eNOS phosphorylation and inhibited Fstl1-stimulated cellular responses. Treatment with the eNOS inhibitor N(G)-nitro-L-arginine methyl ester also reduced endothelial cell migration and differentiation induced by Ad-Fstl1. The stimulatory effect of Ad-Fstl1 on ischemic limb reperfusion was abolished in mice lacking eNOS. These data indicate that Fstl1 is a secreted muscle protein or myokine that can function to promote endothelial cell function and stimulates revascularization in response to ischemic insult through its ability to activate Akt-eNOS signaling.
Collapse
Affiliation(s)
- Noriyuki Ouchi
- Molecular Cardiology/Whitaker Cardiovascular Institute, Boston University School of Medicine, Boston, Massachusetts 02118, USA.
| | | | | | | | | | | | | |
Collapse
|
15
|
Powell RJ, Simons M, Mendelsohn FO, Daniel G, Henry TD, Koga M, Morishita R, Annex BH. Results of a Double-Blind, Placebo-Controlled Study to Assess the Safety of Intramuscular Injection of Hepatocyte Growth Factor Plasmid to Improve Limb Perfusion in Patients With Critical Limb Ischemia. Circulation 2008; 118:58-65. [DOI: 10.1161/circulationaha.107.727347] [Citation(s) in RCA: 231] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Background—
The Study to Assess the Safety of Intramuscular Injection of Hepatocyte Growth Factor Plasmid to Improve Limb Perfusion in Patients With Critical Limb Ischemia (HGF-STAT trial) determined the effect of hepatocyte growth factor (HGF) plasmid on safety and limb tissue perfusion as measured by transcutaneous oxygen tension (TcP
o
2
) in patients with critical limb ischemia (CLI).
Methods and Results—
Randomized patients with rest pain or ischemic ulcers and TcP
o
2
<40 mm Hg and/or toe pressure <50 mm Hg received placebo or HGF-plasmid intramuscular injection as follows: 0.4 mg at days 0, 14, and 28 (low dose); 4.0 mg at days 0 and 28 (middle dose); or 4.0 mg at days 0, 14, and 28 (high dose). Patients were evaluated for safety, changes in TcP
o
2
and ankle and toe pressure, amputation, and wound healing. Ninety-three of 104 treated patients were evaluated for safety (mean age 70 years, 63% male, 53% diabetic, 64% with tissue loss, mean ankle-brachial index 0.41, and mean toe pressure 26 mm Hg). Adverse events occurred in 86% of the patients, most of which were related to CLI or comorbid conditions and were not different between groups. TcP
o
2
(mean±SE) increased at 6 months in the high-dose group (24.0±4.2 mm Hg, 95% CI 15.5 to 32.4 mm Hg) compared with the placebo (9.4±4.2 mm Hg, 95% CI 0.9 to 17.8), low-dose (11.1±3.7 mm Hg, CI 3.7 to 18.7 mm Hg), and middle-dose (7.3±4.8 mm Hg, CI −2.2 to 17.0 mm Hg) groups (ANCOVA
P
=0.0015). There was no difference between groups in secondary end points, including ankle-brachial index, toe-brachial index, pain relief, wound healing, or major amputation.
Conclusions—
Intramuscular injection of HGF plasmid was safe and well tolerated. Larger studies to determine whether HGF plasmid can improve wound healing and limb salvage in patients with CLI are warranted.
Collapse
Affiliation(s)
- Richard J. Powell
- From Department of Surgery, Section of Vascular Surgery (R.J.P.) and Department of Medicine, Section of Cardiology (M.S.), Dartmouth-Hitchcock Medical Center, Lebanon, NH; Princeton Heart and Vascular Center (F.O.M.), Birmingham, Ala; John F. Kennedy Medical Center (G.D.), Atlantis, Fla; Minneapolis Heart Institute Foundation (T.D.H.), Minneapolis, Minn; AnGes Inc (M.K.), Gaithersburg, Md; Osaka University (R.M.), Osaka, Japan; and Duke University Medical Center (B.H.A.), Durham, NC
| | - Michael Simons
- From Department of Surgery, Section of Vascular Surgery (R.J.P.) and Department of Medicine, Section of Cardiology (M.S.), Dartmouth-Hitchcock Medical Center, Lebanon, NH; Princeton Heart and Vascular Center (F.O.M.), Birmingham, Ala; John F. Kennedy Medical Center (G.D.), Atlantis, Fla; Minneapolis Heart Institute Foundation (T.D.H.), Minneapolis, Minn; AnGes Inc (M.K.), Gaithersburg, Md; Osaka University (R.M.), Osaka, Japan; and Duke University Medical Center (B.H.A.), Durham, NC
| | - Farrel O. Mendelsohn
- From Department of Surgery, Section of Vascular Surgery (R.J.P.) and Department of Medicine, Section of Cardiology (M.S.), Dartmouth-Hitchcock Medical Center, Lebanon, NH; Princeton Heart and Vascular Center (F.O.M.), Birmingham, Ala; John F. Kennedy Medical Center (G.D.), Atlantis, Fla; Minneapolis Heart Institute Foundation (T.D.H.), Minneapolis, Minn; AnGes Inc (M.K.), Gaithersburg, Md; Osaka University (R.M.), Osaka, Japan; and Duke University Medical Center (B.H.A.), Durham, NC
| | - George Daniel
- From Department of Surgery, Section of Vascular Surgery (R.J.P.) and Department of Medicine, Section of Cardiology (M.S.), Dartmouth-Hitchcock Medical Center, Lebanon, NH; Princeton Heart and Vascular Center (F.O.M.), Birmingham, Ala; John F. Kennedy Medical Center (G.D.), Atlantis, Fla; Minneapolis Heart Institute Foundation (T.D.H.), Minneapolis, Minn; AnGes Inc (M.K.), Gaithersburg, Md; Osaka University (R.M.), Osaka, Japan; and Duke University Medical Center (B.H.A.), Durham, NC
| | - Timothy D. Henry
- From Department of Surgery, Section of Vascular Surgery (R.J.P.) and Department of Medicine, Section of Cardiology (M.S.), Dartmouth-Hitchcock Medical Center, Lebanon, NH; Princeton Heart and Vascular Center (F.O.M.), Birmingham, Ala; John F. Kennedy Medical Center (G.D.), Atlantis, Fla; Minneapolis Heart Institute Foundation (T.D.H.), Minneapolis, Minn; AnGes Inc (M.K.), Gaithersburg, Md; Osaka University (R.M.), Osaka, Japan; and Duke University Medical Center (B.H.A.), Durham, NC
| | - Minako Koga
- From Department of Surgery, Section of Vascular Surgery (R.J.P.) and Department of Medicine, Section of Cardiology (M.S.), Dartmouth-Hitchcock Medical Center, Lebanon, NH; Princeton Heart and Vascular Center (F.O.M.), Birmingham, Ala; John F. Kennedy Medical Center (G.D.), Atlantis, Fla; Minneapolis Heart Institute Foundation (T.D.H.), Minneapolis, Minn; AnGes Inc (M.K.), Gaithersburg, Md; Osaka University (R.M.), Osaka, Japan; and Duke University Medical Center (B.H.A.), Durham, NC
| | - Ryuichi Morishita
- From Department of Surgery, Section of Vascular Surgery (R.J.P.) and Department of Medicine, Section of Cardiology (M.S.), Dartmouth-Hitchcock Medical Center, Lebanon, NH; Princeton Heart and Vascular Center (F.O.M.), Birmingham, Ala; John F. Kennedy Medical Center (G.D.), Atlantis, Fla; Minneapolis Heart Institute Foundation (T.D.H.), Minneapolis, Minn; AnGes Inc (M.K.), Gaithersburg, Md; Osaka University (R.M.), Osaka, Japan; and Duke University Medical Center (B.H.A.), Durham, NC
| | - Brian H. Annex
- From Department of Surgery, Section of Vascular Surgery (R.J.P.) and Department of Medicine, Section of Cardiology (M.S.), Dartmouth-Hitchcock Medical Center, Lebanon, NH; Princeton Heart and Vascular Center (F.O.M.), Birmingham, Ala; John F. Kennedy Medical Center (G.D.), Atlantis, Fla; Minneapolis Heart Institute Foundation (T.D.H.), Minneapolis, Minn; AnGes Inc (M.K.), Gaithersburg, Md; Osaka University (R.M.), Osaka, Japan; and Duke University Medical Center (B.H.A.), Durham, NC
| |
Collapse
|
16
|
Kalka C, Baumgartner I. Gene and stem cell therapy in peripheral arterial occlusive disease. Vasc Med 2008; 13:157-72. [DOI: 10.1177/1358863x08088616] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Abstract Peripheral arterial occlusive disease (PAOD) is a manifestation of systemic atherosclerosis strongly associated with a high risk of cardiovascular morbidity and mortality. In a considerable proportion of patients with PAOD, revascularization either by endovascular means or by open surgery combined with best possible risk factor modification does not achieve limb salvage or relief of ischaemic rest pain. As a consequence, novel therapeutic strategies have been developed over the last two decades aiming to promote neovascularization and remodelling of collaterals. Gene and stem cell therapy are the main directions for clinical investigation concepts. For both, preclinical studies have shown promising results using a wide variety of genes encoding for growth factors and populations of adult stem cells, respectively. As a consequence, clinical trials have been performed applying gene and stem cell-based concepts. However, it has become apparent that a straightforward translation into humans is not possible. While several trials reported relief of symptoms and functional improvement, other trials did not confirm this early promise of efficacy. Ongoing clinical trials with an improved study design are needed to confirm the potential that gene and cell therapy may have and to prevent the gaps in our scientific knowledge that will jeopardize the establishment of angiogenic therapy as an additional medical treatment of PAOD. This review summarizes the experimental background and presents the current status of clinical applications and future perspectives of the therapeutic use of gene and cell therapy strategies for PAOD.
Collapse
Affiliation(s)
- C Kalka
- Division of Vascular Medicine, Swiss Cardiovascular Center, University Hospital of Bern, Switzerland
| | - Iris Baumgartner
- Division of Vascular Medicine, Swiss Cardiovascular Center, University Hospital of Bern, Switzerland
| |
Collapse
|
17
|
Xu H, Shi BM, Lu XF, Liang F, Jin X, Wu TH, Xu J. Vascular endothelial growth factor attenuates hepatic sinusoidal capillarization in thioacetamide-induced cirrhotic rats. World J Gastroenterol 2008; 14:2349-57. [PMID: 18416461 PMCID: PMC2705089 DOI: 10.3748/wjg.14.2349] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
AIM: To investigate the effect of vascular endothelial growth factor (VEGF) transfection on hepatic sinusoidal capillarization.
METHODS: Enhanced green fluorescent protein (EGFP)/VEGF transfection was confirmed by immunofluorescence microscopy and immunohistoche-mistry both in primary hepatocytes and in normal liver. Cirrhotic rats were generated by thioacetamide (TAA) administration and then divided into a treatment group, which received injections of 400 &mgr;g of plasmid DNA encoding an EGFP-VEGF fusion protein, and a blank group, which received an equal amount of normal saline through the portal vein. The portal vein pressure was measured in the normal and cirrhotic state, in treated and blank groups. The average number of fenestrae per hepatic sinusoid was determined using transmission electron microscopy (TEM), while the relative abundance of VEGF transcripts was examined by Gene array.
RESULTS: Green fluorescent protein was observed in the cytoplasms of liver cells under immunofluorescence microscopy 24 h after transfection with EGFP/VEGF plasmid in vitro. Staining with polyclonal antibodies against VEGF illustrated that hepatocytes expressed immunodetectable VEGF both in vitro and in vitro. There were significant differences in the number of fenestrae and portal vein pressures between normal and cirrhotic rats (7.40 ± 1.71 vs 2.30 ± 1.16 and 9.32 ± 0.85 cmH2O vs 17.92 ± 0.90 cmH2O, P < 0.01), between cirrhotic and treated rats (2.30 ± 1.16 cmH2O vs 4.60 ± 1.65 and 17.92 ± 0.90 cmH2O vs 15.52 ± 0.93 cmH2O, P < 0.05) and between the treatment group and the blank group (4.60 ± 1.65 cmH2O vs 2.10 ± 1.10 cmH2O and 15.52 ± 0.93 cmH2O vs 17.26 ± 1.80 cmH2O, P < 0.05). Gene-array analysis revealed that the relative abundance of transcripts of VEGF family members decreased in the cirrhotic state and increased after transfection.
CONCLUSION: Injection of a plasmid encoding VEGF through the portal vein is an effective method to induce the formation of fenestrae and decrease portal vein pressure in cirrhotic rats. Therefore, it may be a good choice for treating hepatic cirrhosis and portal hypertension.
Collapse
|
18
|
Ponce ML, Kleinmann HK. The chick chorioallantoic membrane as an in vivo angiogenesis model. ACTA ACUST UNITED AC 2008; Chapter 19:Unit 19.5. [PMID: 18228425 DOI: 10.1002/0471143030.cb1905s18] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
The chick chorioallantoic membrane (CAM) assay for angiogenic activity is a model originally developed to study the angiogenic activity of tumor samples. It is an in vivo assay that can be readily performed in any laboratory setting. The effects of a test compound on angiogenesis are tested by exposing day 10 embryos to the compound and following the patterns of blood vessel development, scoring the appearance of the CAM at day 12 or 13.
Collapse
|
19
|
Arom KV, Ruengsakulrach P, Jotisakulratana V. Efficacy of Intramyocardial Injection of Angiogenic Cell Precursors for Ischemic Cardiomyopathy. INNOVATIONS-TECHNOLOGY AND TECHNIQUES IN CARDIOTHORACIC AND VASCULAR SURGERY 2008. [DOI: 10.1177/155698450800300109] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Affiliation(s)
- Kitipan V. Arom
- Division of Cardiovascular Surgery, Bangkok Heart Hospital, Bangkok, Thailand
| | | | | |
Collapse
|
20
|
Efficacy of intramyocardial injection of angiogenic cell precursors for ischemic cardiomyopathy: a case match study. INNOVATIONS-TECHNOLOGY AND TECHNIQUES IN CARDIOTHORACIC AND VASCULAR SURGERY 2008; 3:38-45. [PMID: 22436722 DOI: 10.1097/imi.0b013e31816755dd] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
INTRODUCTION : The objective of this study is to determine the efficacy of intramyocardial angiogenic cell precursors (ACPs) injection in ischemic cardiomyopathy (ICM). METHODS : Twenty-five ICM patients (cell group) underwent intramyocardial ACPs injection. Seventeen ICM patients (control group) treated by medical means were matched with cell group. There was no statistically significant difference between cell and control groups in relation to left ventricular ejection fraction (LVEF) and comorbidities. In the cell group, mean age was 58.4 ± 13.7 years. Mean LVEF was 26.1% ± 7.4%. New York Heart Association (NYHA) class was 2.9 ± 0.6. The ACPs were derived and expanded from autologous blood. The number of cells before injection was 27.4 ± 18.8 million cells. The cells were injected into the nonviable myocardium and hypokinetic segments in the cell group. RESULTS : There was no new ventricular arrhythmia. NYHA was improved by 0.9 ± 1.0 (P < 0.001) at 229.9 ± 98.8 days. Six-minute walk test and quality of life assessed by short form-36 improved in the cell group. LVEF was improved in 72% of patients (18 of 25). LVEF improved by 6.4 ± 9.9 points % (P = 0.003) at 290.4 ± 210.3 days. The percentage of infarction area decreased 21.9 ± 17.4 points % at 159 ± 54 days postoperatively. There was no significant improvement of NYHA and LVEF in the control group. CONCLUSIONS : For this efficacy study, the NYHA class, quality of life, and six-minute walk test were improved after cell transplantation. The LVEF was also significantly improved in the cell treated group.
Collapse
|
21
|
Awad HA, Zhang X, Reynolds DG, Guldberg RE, O'Keefe RJ, Schwarz EM. Recent Advances in Gene Delivery for Structural Bone Allografts. ACTA ACUST UNITED AC 2007; 13:1973-85. [PMID: 17518728 DOI: 10.1089/ten.2006.0107] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
In this paper, we review the progress toward developing strategies to engineer improved structural grafting of bone. Three strategies are typically used to augment massive bone defect repair. The first is to engraft mesenchymal stem cells (MSCs) onto a graft or a biosynthetic matrix to provide a viable osteoinductive scaffold material for segmental defect repair. The second strategy is to introduce critical factor(s), for example, bone morphogenetic proteins (BMPs), in the form of bone-derived or recombinant proteins onto the graft or matrix directly. The third strategy uses targeted delivery of therapeutic genes (using viral and nonviral vectors) that either transduce host cells in vivo or stably transduce cells in vitro for subsequent implantation in vivo. We developed a murine femoral model in which allografts can be revitalized via recombinant adeno-associated virus (rAAV) gene transfer. Specifically, allografts coated with rAAV expressing either the constitutively active BMP type I receptor Alk2 (caAlk2), or the angiogenic factor vascular endothelial growth factor (VEGF) combined with the osteoclastogenic factor receptor activator of NF-kappa B ligand (RANKL) have remarkable osteogenic, angiogenic, and remodeling effects that have not been previously documented in healing allografts. Using histomorphometric and micro computed tomography (muCT) imaging we show that rAAV-mediated delivery of caAlk2 induces significant osteoinduction manifested by a mineralized callus on the surface of the allograft, which resembles the healing response of an autograft. We also demonstrate that the rAAV-mediated gene transfer of the combination of VEGF and RANKL can induce significant vascularization and remodeling of processed structural allografts. By contrast, rAAV-LacZ coated allograft controls appeared similar to necrotic allografts and lacked significant mineralized callus, neovascularization, and remodeling. Therefore, innovations in gene delivery offer promising therapeutic approaches for tissue engineering of structural bone substitutes that can potentially have clinical applications in challenging indications.
Collapse
Affiliation(s)
- Hani A Awad
- Department of Biomedical Engineering, The Center for Musculoskeletal Research, University of Rochester, Rochester, New York 14620, USA.
| | | | | | | | | | | |
Collapse
|
22
|
|
23
|
Gosain A, Matthies AM, Dovi JV, Barbul A, Gamelli RL, DiPietro LA. Exogenous pro-angiogenic stimuli cannot prevent physiologic vessel regression. J Surg Res 2006; 135:218-25. [PMID: 16904692 DOI: 10.1016/j.jss.2006.04.006] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2006] [Revised: 04/06/2006] [Accepted: 04/07/2006] [Indexed: 10/24/2022]
Abstract
BACKGROUND In healing wounds, rising levels of vascular endothelial growth factor (VEGF) induce a period of robust angiogenesis. The levels of pro-angiogenic factors in the wound begin to decline just before a period of vascular regression, suggesting that these mediators are necessary to sustain vessel density. The purpose of this study was to determine if the maintenance of pro-angiogenic stimuli in the wound would prevent physiological vessel regression. MATERIALS AND METHODS A standard subcutaneous sponge wound model was modified by the addition of a mini-osmotic pump, allowing manipulation of the wound milieu by the addition of exogenous growth factors. After initial characterization of this model, exogenous VEGF (10 microg/mL), FGF (10 microg/mL), PDGF (10 microg/mL), or VEGF (10 microg/mL) plus FGF (10 microg/mL) were delivered to wounds and blood vessel density analyzed by immunohistochemistry. RESULTS VEGF administration resulted in a transient increase in wound vessel density (P < 0.05). None of the pro-angiogenic growth factors (VEGF, FGF, PDGF, VEGF/FGF) were able to prevent vascular regression (P = NS). CONCLUSIONS These findings suggest that the anti-angiogenic signals that mediate physiological vascular regression in wounds are strongly dominant over pro-angiogenic stimuli during the later phases of wound healing. Clinical manipulation of anti-angiogenic signals in addition to the currently used pro-angiogenic targets may be needed to achieve therapeutic modulation of blood vessel density.
Collapse
Affiliation(s)
- Ankush Gosain
- Department of Cell Biology, Neurobiology and Anatomy, Loyola University Medical Center, Burn and Shock Trauma Institute, Maywood, Illinois, USA
| | | | | | | | | | | |
Collapse
|
24
|
Ismail M, Henklein P, Huang X, Braumann C, Rückert RI, Dubiel W. Identification of HIV-1 Tat peptides for future therapeutic angiogenesis. Eur J Haematol 2006; 77:157-65. [PMID: 16800839 DOI: 10.1111/j.1600-0609.2006.00682.x] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Therapeutic angiogenesis represents a novel approach to treat critical limb ischemia when revascularization is no more an option. The clinical use of the vascular endothelial growth factor is questioned, because of its side effects. This study was designed to identify and characterize human immunodeficiency virus type 1 (HIV-1) Tat-derived peptides based on their pro-angiogenic properties. A series of Tat-derived peptides were synthesized containing mutations in the basic domain. To minimize side effects Tat peptides were selected exerting no effects on the proteasome and on the viability of human umbilical vein endothelial cells (HUVEC). Tatpep5, 15, and 16 increased the endogenous levels of the pro-angiogenic transcription factors c-Jun and SP-1 as well as the production of the plasminogen activator inhibitor-1 (PAI-1) by HUVEC. A significant induction of endothelial cell invasion was observed upon treatment of HUVEC with Tat peptides. In addition, selected Tat peptides induced tube formation by HUVEC as visualized and quantified in a Matrigel matrix. Our data demonstrate that the selected Tat peptides fulfill essential criteria for pro-angiogenic substances. They represent the basis for the development of novel pro-angiogenic drugs for future therapeutic angiogenesis, which might be applied for treatment of unreconstructible critical limb ischemia.
Collapse
Affiliation(s)
- Mahmoud Ismail
- Department of Surgery, Universitatsmedizin Berlin, Berlin, Germany
| | | | | | | | | | | |
Collapse
|
25
|
Hirsch AT, Haskal ZJ, Hertzer NR, Bakal CW, Creager MA, Halperin JL, Hiratzka LF, Murphy WRC, Olin JW, Puschett JB, Rosenfield KA, Sacks D, Stanley JC, Taylor LM, White CJ, White J, White RA, Antman EM, Smith SC, Adams CD, Anderson JL, Faxon DP, Fuster V, Gibbons RJ, Hunt SA, Jacobs AK, Nishimura R, Ornato JP, Page RL, Riegel B. ACC/AHA 2005 Practice Guidelines for the management of patients with peripheral arterial disease (lower extremity, renal, mesenteric, and abdominal aortic): a collaborative report from the American Association for Vascular Surgery/Society for Vascular Surgery, Society for Cardiovascular Angiography and Interventions, Society for Vascular Medicine and Biology, Society of Interventional Radiology, and the ACC/AHA Task Force on Practice Guidelines (Writing Committee to Develop Guidelines for the Management of Patients With Peripheral Arterial Disease): endorsed by the American Association of Cardiovascular and Pulmonary Rehabilitation; National Heart, Lung, and Blood Institute; Society for Vascular Nursing; TransAtlantic Inter-Society Consensus; and Vascular Disease Foundation. Circulation 2006; 113:e463-654. [PMID: 16549646 DOI: 10.1161/circulationaha.106.174526] [Citation(s) in RCA: 2189] [Impact Index Per Article: 121.6] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
26
|
Hirsch AT, Haskal ZJ, Hertzer NR, Bakal CW, Creager MA, Halperin JL, Hiratzka LF, Murphy WR, Olin JW, Puschett JB, Rosenfield KA, Sacks D, Stanley JC, Taylor LM, White CJ, White J, White RA, Antman EM, Smith SC, Adams CD, Anderson JL, Faxon DP, Fuster V, Gibbons RJ, Halperin JL, Hiratzka LF, Hunt SA, Jacobs AK, Nishimura R, Ornato JP, Page RL, Riegel B. ACC/AHA 2005 Guidelines for the Management of Patients With Peripheral Arterial Disease (Lower Extremity, Renal, Mesenteric, and Abdominal Aortic): A Collaborative Report from the American Association for Vascular Surgery/Society for Vascular Surgery,⁎Society for Cardiovascular Angiography and Interventions, Society for Vascular Medicine and Biology, Society of Interventional Radiology, and the ACC/AHA Task Force on Practice Guidelines (Writing Committee to Develop Guidelines for the Management of Patients With Peripheral Arterial Disease). J Am Coll Cardiol 2006. [DOI: 10.1016/j.jacc.2006.02.024] [Citation(s) in RCA: 100] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
|
27
|
Miyajima S, Shirai A, Yamamoto S, Okada N, Matsushita T. Risk factors for major limb amputations in diabetic foot gangrene patients. Diabetes Res Clin Pract 2006; 71:272-9. [PMID: 16139385 DOI: 10.1016/j.diabres.2005.07.005] [Citation(s) in RCA: 103] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/14/2004] [Revised: 05/06/2005] [Accepted: 07/05/2005] [Indexed: 11/25/2022]
Abstract
We analyzed the clinical picture of diabetic foot lesion patients to investigate the risk factors for major limb amputations. The subjects were 210 diabetic foot lesion patients treated at our department over the past 9 years. The mean follow-up period was 604.5 [standard deviation (S.D.) 451.2] days with a median value of 492 days. There were 113 men and 97 women. By the final follow-up day, 18 underwent bypass surgeries (9%) and 13 skin grafts (6%), while 110 patients (52%) finally required limb amputation. The breakdown was 45 major amputations above or below the knee and 65 minor amputations of the toes or metatarsals. The outcomes of the major amputations were retrospectively analyzed by group. The blood glucose control was poor in all 45 major amputees, and their mean HbA1c (8.80%) was higher than that in the minor or non-amputation group (7.79%, P = 0.035). In the major amputation group, two patients had loss of vision due to retinopathy, and 30 patients received long-term hemodialysis due to nephropathy. The rate of arteriosclerosis obliterans (ASO) was higher in the major amputation group than in the minor or non-amputation group, and arteriography showed significantly high rates of multiple stenosis. Multivariate analysis of these results by the proportional hazard model showed that ASO with multiple stenosis (hazard ratio 3.23, 95% CI: 1.12-5.10), hemodialysis (2.14, 95% CI: 1.17-3.44), and HbA1c (1.20, 95% CI: 1.03-1.41) were independent risk factors for major amputation. The 3-year survival rate was 24.1% in the major amputation group and 93.0% in the minor or non-amputation group, and the life expectancy was significantly lower for the major amputees than the minor or non-amputees (P<0.0001). Together with early detection and treatment of foot lesions, good blood glucose control and early management of systemic complications such as nephropathy and arteriosclerosis are considered important to avoid major amputations.
Collapse
Affiliation(s)
- Susumu Miyajima
- Department of Dermatology, Osaka Koseinenkin Hospital, 4-2-78, Fukushima-ku, Fukushima, Osaka 553-0003, Japan.
| | | | | | | | | |
Collapse
|
28
|
Valable S, Montaner J, Bellail A, Berezowski V, Brillault J, Cecchelli R, Divoux D, Mackenzie ET, Bernaudin M, Roussel S, Petit E. VEGF-induced BBB permeability is associated with an MMP-9 activity increase in cerebral ischemia: both effects decreased by Ang-1. J Cereb Blood Flow Metab 2005; 25:1491-504. [PMID: 15902195 DOI: 10.1038/sj.jcbfm.9600148] [Citation(s) in RCA: 158] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
After cerebral ischemia, angiogenesis, by supplying for the deficient perfusion, may be a beneficial process for limiting neuronal death and promoting tissue repair. In this study, we showed that the combination of Ang-1 and vascular endothelial growth factor (VEGF) provides a more adapted therapeutic strategy than the use of VEGF alone. Indeed, we showed on a focal ischemia model that an early administration of VEGF exacerbates ischemic damage, because of its effects on blood-brain barrier (BBB) permeability. In contrast, a coapplication of Ang-1 and VEGF leads to a significant reduction of the ischemic and edema volumes by 50% and 42%, respectively, in comparison with VEGF-treated mice. We proposed that Ang-1 blocks the BBB permeability effect of VEGF in association with a modulation of matrix metalloproteinase (MMP) activity. Indeed, we showed on both ischemic in vivo and BBB in vitro models that VEGF enhances BBB damage and MMP-9 activity and that Ang-1 counteracts both effects. However, we also showed a synergic angiogenic effect of Ang-1 and VEGF in the brain. Taken together, these results allow to propose that, in cerebral ischemia, the combination of Ang-1 and VEGF could be used early to promote the formation of mature neovessels without inducing side effects on BBB permeability.
Collapse
Affiliation(s)
- Samuel Valable
- UMR-CNRS 6185, Centre Cyceron, University of Caen, Caen, France
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Christman KL, Fang Q, Yee MS, Johnson KR, Sievers RE, Lee RJ. Enhanced neovasculature formation in ischemic myocardium following delivery of pleiotrophin plasmid in a biopolymer. Biomaterials 2005; 26:1139-44. [PMID: 15451633 DOI: 10.1016/j.biomaterials.2004.04.025] [Citation(s) in RCA: 76] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2004] [Accepted: 04/08/2004] [Indexed: 11/18/2022]
Abstract
Coronary heart disease is currently the leading killer in the western world. Therapeutic angiogenic agents are currently being examined for treatment of this disease. We have recently demonstrated the effective use of Pleiotrophin (PTN) as a therapeutic agent for treatment of ischemic myocardium. We have also shown that injection of the biopolymer fibrin glue preserves left ventricular geometry and prevents a deterioration of cardiac function following myocardial infarction. Due to the low transfection efficiency of naked plasmid injections, we examined the use of PTN plasmid and the biopolymer as a gene-activated matrix in the myocardium. In this study, we demonstrate that delivery of PTN plasmid in fibrin glue increases neovasculature formation compared to injection of the naked plasmid in saline.
Collapse
Affiliation(s)
- Karen L Christman
- University of California Berkeley and San Francisco Joint Bioengineering Graduate Group, San Francisco, CA 94143, USA.
| | | | | | | | | | | |
Collapse
|
30
|
Mukhopadhyay D, Zeng H, Bhattacharya R. Complexity in the vascular permeability factor/vascular endothelial growth factor (VPF/VEGF)-receptors signaling. Mol Cell Biochem 2005; 264:51-61. [PMID: 15544035 DOI: 10.1023/b:mcbi.0000044374.85095.df] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
The adult vasculature results from a network of vessels that is originally derived in the embryo by vasculogenesis, a process whereby vessels are formed de novo from endothelial cell (EC) precursors, known as angioblasts. During vasculogenesis, angioblasts proliferate and come together to form an initial network of vessels, also known as the primary capillary plexus. Sprouting and branching of new vessels from the preexisting vessels in the process of angiogenesis remodel the capillary plexus. Normal angiogenesis, a well-balanced process, is important in the embryo to promote primary vascular tree as well as an adequate vasculature from developing organs. On the other hand, pathological angiogenesis which frequently occurs in tumors, rheumatoid arthritis, diabetic retinopathy and other circumstances can induce their own blood supply from the preexisting vasculature in a route that is close to normal angiogenesis. Vascular permeability factor/vascular endothelial growth factor (VPF/VEGF) is perhaps the most important of pro-angiogenic cytokine because of its ability to regulate most of the steps in the angiogenic cascade. The main goal of this review article is to discuss the complex nature of the mode of action of VPF/VEGF on vascular endothelium. To this end, we conclude that more research needs to be done for completely understanding the VPF/VEGF biology with relation to angiogenesis.
Collapse
Affiliation(s)
- Debabrata Mukhopadhyay
- Mayo Clinic Cancer Center, Department of Biochemistry and Molecular Biology, Mayo Clinic Foundation, Gugg, Rochester, MN 55905, USA.
| | | | | |
Collapse
|
31
|
Hagstrom JE, Hegge J, Zhang G, Noble M, Budker V, Lewis DL, Herweijer H, Wolff JA. A facile nonviral method for delivering genes and siRNAs to skeletal muscle of mammalian limbs. Mol Ther 2005; 10:386-98. [PMID: 15294185 DOI: 10.1016/j.ymthe.2004.05.004] [Citation(s) in RCA: 119] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2004] [Accepted: 05/03/2004] [Indexed: 10/26/2022] Open
Abstract
Delivery is increasingly being recognized as the critical hurdle holding back the tremendous promise of nucleic acid-based therapies that include gene therapy and more recently siRNA-based therapeutics. While numerous candidate genes (and siRNA sequences) with therapeutic potential have been identified, their utility has not yet been realized because of inefficient and/or unsafe delivery technologies. We now describe an intravascular, nonviral methodology that enables efficient and repeatable delivery of nucleic acids to muscle cells (myofibers) throughout the limb muscles of mammals. The procedure involves the injection of naked plasmid DNA or siRNA into a distal vein of a limb that is transiently isolated by a tourniquet or blood pressure cuff. Nucleic acid delivery to myofibers is facilitated by its rapid injection in sufficient volume to enable extravasation of the nucleic acid solution into muscle tissue. High levels of transgene expression in skeletal muscle were achieved in both small and large animals with minimal toxicity. Evidence of siRNA delivery to limb muscle was also obtained. The simplicity, effectiveness, and safety of the procedure make this methodology well suited to limb muscle gene therapy applications.
Collapse
|
32
|
Zaccagnini G, Gaetano C, Della Pietra L, Nanni S, Grasselli A, Mangoni A, Benvenuto R, Fabrizi M, Truffa S, Germani A, Moretti F, Pontecorvi A, Sacchi A, Bacchetti S, Capogrossi MC, Farsetti A. Telomerase mediates vascular endothelial growth factor-dependent responsiveness in a rat model of hind limb ischemia. J Biol Chem 2005; 280:14790-8. [PMID: 15687494 DOI: 10.1074/jbc.m414644200] [Citation(s) in RCA: 63] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Telomere dysfunction contributes to reduced cell viability, altered differentiation, and impaired regenerative/proliferative responses. Recent advances indicate that telomerase activity confers a pro-angiogenic phenotype to endothelial cells and their precursors. We have investigated whether telomerase contributes to tissue regeneration following hind limb ischemia and vascular endothelial growth factor 165 (VEGF(165)) treatment. VEGF delivery induced angiogenesis and increased expression of the telomerase reverse transcriptase (TERT) and telomerase activity in skeletal muscles and satellite and endothelial cells. Adenovirus-mediated transfer of wild type TERT but not of a dominant negative mutant, TERTdn, significantly induced capillary but not arteriole formation. However, when co-delivered with VEGF, TERTdn abrogated VEGF-dependent angiogenesis, arteriogenesis, and blood flow increase. This effect was paralleled by in vitro evidence that telomerase inhibition by 3'-azido-3'-deoxythymidine in VEGF-treated endothelial cells strongly reduced capillary density and promoted apoptosis in the absence of serum. Similar results were obtained with adenovirus-mediated expression of TERTdn and AKTdn, both reducing endogenous TERT activity and angiogenesis on Matrigel. Mechanistically, neo-angiogenesis in our system involved: (i) VEGF-dependent activation of telomerase through the nitric oxide pathway and (ii) telomerase-dependent activation of endothelial cell differentiation and protection from apoptosis. Furthermore, detection of TERT in activated satellite cells identified them as VEGF targets during muscle regeneration. Because TERT behaves as an angiogenic factor and a downstream effector of VEGF signaling, telomerase activity appears required for VEGF-dependent remodeling of ischemic tissue at the capillaries and arterioles level.
Collapse
Affiliation(s)
- Germana Zaccagnini
- Laboratory of Vascular Biology and Gene Therapy, Cardiology Center I. Monzino, Milan 20138, Italy
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Collinson DJ, Donnelly R. Therapeutic Angiogenesis in Peripheral Arterial Disease: Can Biotechnology Produce an Effective Collateral Circulation? Eur J Vasc Endovasc Surg 2004; 28:9-23. [PMID: 15177227 DOI: 10.1016/j.ejvs.2004.03.021] [Citation(s) in RCA: 101] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/30/2004] [Indexed: 11/23/2022]
Abstract
The physiological processes of angiogenesis, vasculogenesis and arteriogenesis contribute to the growth of collateral vessels in response to obstructive arterial disease causing lower limb or myocardial ischaemia, but in clinical practice the endogenous angiogenic response is often suboptimal or impaired, e.g. by factors such as ageing, diabetes or drug therapies. Therapeutic angiogenesis is an application of biotechnology to stimulate new vessel formation via local administration of pro-angiogenic growth factors in the form of recombinant protein or gene therapy, or by implantation of endothelial progenitor cells that will synthesize multiple angiogenic cytokines. Numerous experimental and clinical studies have sought to establish 'proof of concept' for therapeutic angiogenesis in PAD and myocardial ischaemia using different treatment modalities, but the results have been inconsistent. This review summarises the mechanisms of angiogenesis and the results of recent trials evaluating the efficacy and safety of different gene therapy, recombinant protein and cellular-based treatment approaches to enhance collateral vessel formation.
Collapse
Affiliation(s)
- D J Collinson
- Centre for Integrated Systems Biology and Medicine, School of Medical and Surgical Sciences University of Nottingham, Nottingham, UK
| | | |
Collapse
|
34
|
Ziv K, Nevo N, Dafni H, Israely T, Granot D, Brenner O, Neeman M. Longitudinal MRI tracking of the angiogenic response to hind limb ischemic injury in the mouse. Magn Reson Med 2004; 51:304-11. [PMID: 14755656 DOI: 10.1002/mrm.10687] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
Ischemic injury and revascularization are frequently associated with hyperpermeability. Although extravasation of plasma proteins may promote tissue recovery through the generation of the provisional matrix that supports angiogenesis, edema may also result in progressive damage to the muscle. The aim of this research was to determine the time course of hyperpermeability associated with the angiogenic response induced by ligation of the femoral artery at the right posterior limb in mice. Hyperpermeability was followed noninvasively by MRI using an in-house-built permanent polyethylene catheter that enabled daily intravenous administration of biotin-BSA-Gd-DTPA. The mice were scanned once prior to ligation and five times during the week post-ligation. The MRI data, along with histopathology, indicated that the early hemodynamic compensation over loss of arterial blood supply occurred by angiogenesis and dilation of vessels in the skin and subcutaneous fat, and was accompanied by vascular hyperpermeability around the site of ligation. Functional recovery of the ischemic limb (i.e., regaining the ability to step on the limb), and the color and shape of the toes correlated with regeneration as shown by histopathology and MRI analysis. Thus, MRI provided valuable information on the transient hyperpermeability induced during the early stages of angiogenesis, and its subsequent resolution along with functional recovery from acute hind limb ischemia in mice.
Collapse
Affiliation(s)
- Keren Ziv
- Department of Biological Regulation, Weizmann Institute of Science, Rehovot, Israel
| | | | | | | | | | | | | |
Collapse
|
35
|
Weis S, Shintani S, Weber A, Kirchmair R, Wood M, Cravens A, McSharry H, Iwakura A, Yoon YS, Himes N, Burstein D, Doukas J, Soll R, Losordo D, Cheresh D. Src blockade stabilizes a Flk/cadherin complex, reducing edema and tissue injury following myocardial infarction. J Clin Invest 2004; 113:885-94. [PMID: 15067321 PMCID: PMC362122 DOI: 10.1172/jci20702] [Citation(s) in RCA: 117] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2003] [Accepted: 01/06/2004] [Indexed: 01/13/2023] Open
Abstract
Ischemia resulting from myocardial infarction (MI) promotes VEGF expression, leading to vascular permeability (VP) and edema, a process that we show here contributes to tissue injury throughout the ventricle. This permeability/edema can be assessed noninvasively by MRI and can be observed at the ultrastructural level as gaps between adjacent endothelial cells. Many of these gaps contain activated platelets adhering to exposed basement membrane, reducing vessel patency. Following MI, genetic or pharmacological blockade of Src preserves endothelial cell barrier function, suppressing VP and infarct volume, providing long-term improvement in cardiac function, fibrosis, and survival. To our surprise, an intravascular injection of VEGF into healthy animals, but not those deficient in Src, induced similar endothelial gaps, VP, platelet plugs, and some myocyte damage. Mechanistically, we show that quiescent blood vessels contain a complex involving Flk, VE-cadherin, and beta-catenin that is transiently disrupted by VEGF injection. Blockade of Src prevents disassociation of this complex with the same kinetics with which it prevents VEGF-mediated VP/edema. These findings define a molecular mechanism to account for the Src requirement in VEGF-mediated permeability and provide a basis for Src inhibition as a therapeutic option for patients with acute MI.
Collapse
Affiliation(s)
- Sara Weis
- Department of Immunology, The Scripps Research Institute, La Jolla, California 92037, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Ehrbar M, Djonov VG, Schnell C, Tschanz SA, Martiny-Baron G, Schenk U, Wood J, Burri PH, Hubbell JA, Zisch AH. Cell-Demanded Liberation of VEGF
121
From Fibrin Implants Induces Local and Controlled Blood Vessel Growth. Circ Res 2004; 94:1124-32. [PMID: 15044320 DOI: 10.1161/01.res.0000126411.29641.08] [Citation(s) in RCA: 311] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Although vascular endothelial growth factor (VEGF) has been described as a potent angiogenic stimulus, its application in therapy remains difficult: blood vessels formed by exposure to VEGF tend to be malformed and leaky. In nature, the principal form of VEGF possesses a binding site for ECM components that maintain it in the immobilized state until released by local cellular enzymatic activity. In this study, we present an engineered variant form of VEGF, α
2
PI
1–8
-VEGF
121
, that mimics this concept of matrix-binding and cell-mediated release by local cell-associated enzymatic activity, working in the surgically-relevant biological matrix fibrin. We show that matrix-conjugated α
2
PI
1–8
-VEGF
121
is protected from clearance, contrary to native VEGF
121
mixed into fibrin, which was completely released as a passive diffusive burst. Grafting studies on the embryonic chicken chorioallantoic membrane (CAM) and in adult mice were performed to assess and compare the quantity and quality of neovasculature induced in response to fibrin implants formulated with matrix-bound α
2
PI
1–8
-VEGF
121
or native diffusible VEGF
121
. Our CAM measurements demonstrated that cell-demanded release of α
2
PI
1–8
-VEGF
121
increases the formation of new arterial and venous branches, whereas exposure to passively released wild-type VEGF
121
primarily induced chaotic changes within the capillary plexus. Specifically, our analyses at several levels, from endothelial cell morphology and endothelial interactions with periendothelial cells, to vessel branching and network organization, revealed that α
2
PI
1–8
-VEGF
121
induces vessel formation more potently than native VEGF
121
and that those vessels possess more normal morphologies at the light microscopic and ultrastructural level. Permeability studies in mice validated that vessels induced by α
2
PI
1–8
-VEGF
121
do not leak. In conclusion, cell-demanded release of engineered VEGF
121
from fibrin implants may present a therapeutically safe and practical modality to induce local angiogenesis.
Collapse
Affiliation(s)
- Martin Ehrbar
- Institute for Biomedical Engineering, and University of Zurich, Zurich, Switzerland
| | | | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Weis S, Shintani S, Weber A, Kirchmair R, Wood M, Cravens A, McSharry H, Iwakura A, Yoon YS, Himes N, Burstein D, Doukas J, Soll R, Losordo D, Cheresh D. Src blockade stabilizes a Flk/cadherin complex, reducing edema and tissue injury following myocardial infarction. J Clin Invest 2004. [DOI: 10.1172/jci200420702] [Citation(s) in RCA: 272] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
|
38
|
Huang J, Ito Y, Kobune M, Sasaki K, Nakamura K, Dehari H, Takahashi K, Ikeda K, Uchida H, Kato K, Hamada H. Myocardial injection of CA promoter-based plasmid mediates efficient transgene expression in rat heart. J Gene Med 2003; 5:900-908. [PMID: 14533199 DOI: 10.1002/jgm.429] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
Abstract
BACKGROUND Although naked plasmid injection is the safest and most convenient method for gene delivery, a major limitation of this approach is currently poor transgene expression. The CA promoter (chicken beta-actin promoter with cytomegalovirus, CMV, enhancer) is one of the strongest transcriptional control modules found; however, it is uncertain whether a CA promoter-based vector is efficient enough for naked gene therapy in a cardiovascular context. METHODS The beta-galactosidase (LacZ) expression provided by CA promoter plasmid (pCAZ2) injection into the skeletal muscle or the heart of Lewis rats was compared with CMV promoter plasmid or adenoviral vector (AxCAZ3). The effect of Simian virus 40 of the replication origin (SV40ori) deletion from pCAZ2 on transgene expression was also evaluated. RESULTS pCAZ2 showed the highest LacZ expression in both skeletal muscle and heart in comparison with the CMV promoter-based vector 5 days after naked plasmid injection. LacZ expression in the heart obtained using 20 micro g of pCAZ2 was almost equivalent to that shown with AxCAZ3 at 6.0 x 10(9) optical particle units. The time course of transgene expression driven by CMV and CA promoters in the heart were similar, with the CA promoter providing significantly higher gene expression than the CMV promoter across all time points examined. SV40ori deletion from pCAZ2 did not affect transgene expression in either skeletal muscle or heart. CONCLUSIONS Transgene expression mediated by naked CA promoter-based plasmid injection was shown to be quite efficient in the heart. We propose that the CA promoter vector is suitable for myocardial gene therapy.
Collapse
Affiliation(s)
- Jianhua Huang
- Department of Molecular Medicine, Sapporo Medical University, South-1, West-17, Chuo-ku, Sapporo 0608556, Japan
| | - Yoshinori Ito
- Department of Molecular Medicine, Sapporo Medical University, South-1, West-17, Chuo-ku, Sapporo 0608556, Japan
- Division of Gene Therapy, Sapporo Medical University, South-1, West-17, Chuo-ku, Sapporo 0608556, Japan
| | - Masayoshi Kobune
- Department of Molecular Medicine, Sapporo Medical University, South-1, West-17, Chuo-ku, Sapporo 0608556, Japan
- 4th Department of Internal Medicine, Sapporo Medical University, South-1, West-17, Chuo-ku, Sapporo 0608556, Japan
| | - Katsunori Sasaki
- Department of Molecular Medicine, Sapporo Medical University, South-1, West-17, Chuo-ku, Sapporo 0608556, Japan
| | - Kiminori Nakamura
- Department of Molecular Medicine, Sapporo Medical University, South-1, West-17, Chuo-ku, Sapporo 0608556, Japan
| | - Hironari Dehari
- Department of Molecular Medicine, Sapporo Medical University, South-1, West-17, Chuo-ku, Sapporo 0608556, Japan
| | - Kazuhiro Takahashi
- Department of Molecular Medicine, Sapporo Medical University, South-1, West-17, Chuo-ku, Sapporo 0608556, Japan
- Department of Thoracic and Cardiovascular Surgery, Sapporo Medical University, South-1, West-17, Chuo-ku, Sapporo 0608556, Japan
| | - Katsuya Ikeda
- Department of Molecular Medicine, Sapporo Medical University, South-1, West-17, Chuo-ku, Sapporo 0608556, Japan
- Department of Thoracic and Cardiovascular Surgery, Sapporo Medical University, South-1, West-17, Chuo-ku, Sapporo 0608556, Japan
| | - Hiroaki Uchida
- Department of Molecular Medicine, Sapporo Medical University, South-1, West-17, Chuo-ku, Sapporo 0608556, Japan
| | - Kazunori Kato
- Department of Molecular Medicine, Sapporo Medical University, South-1, West-17, Chuo-ku, Sapporo 0608556, Japan
| | - Hirofumi Hamada
- Department of Molecular Medicine, Sapporo Medical University, South-1, West-17, Chuo-ku, Sapporo 0608556, Japan
- Division of Gene Therapy, Sapporo Medical University, South-1, West-17, Chuo-ku, Sapporo 0608556, Japan
| |
Collapse
|
39
|
Abstract
Advances in imaging are transforming our understanding of angiogenesis and the evaluation of drugs that stimulate or inhibit angiogenesis in preclinical models and human disease. Vascular imaging makes it possible to quantify the number and spacing of blood vessels, measure blood flow and vascular permeability, and analyze cellular and molecular abnormalities in blood vessel walls. Microscopic methods ranging from fluorescence, confocal and multiphoton microscopy to electron microscopic imaging are particularly useful for elucidating structural and functional abnormalities of angiogenic blood vessels. Magnetic resonance imaging (MRI), computed tomography (CT), positron emission tomography (PET), ultrasonography and optical imaging provide noninvasive, functionally relevant images of angiogenesis in animals and humans. An ongoing dilemma is, however, that microscopic methods provide their highest resolution on preserved tissue specimens, whereas clinical methods give images of living tissues deep within the body but at much lower resolution and specificity and generally cannot resolve vessels of the microcirculation. Future challenges include developing new imaging methods that can bridge this resolution gap and specifically identify angiogenic vessels. Another goal is to determine which microscopic techniques are the best benchmarks for interpreting clinical images. The importance of angiogenesis in cancer, chronic inflammatory diseases, age-related macular degeneration and reversal of ischemic heart and limb disease provides incentive for meeting these challenges.
Collapse
Affiliation(s)
- Donald M McDonald
- Cardiovascular Research Institute, Comprehensive Cancer Center, and Department of Anatomy, University of California, 513 Parnassus Avenue, San Francisco, California 94143-0452, USA.
| | | |
Collapse
|
40
|
Conti E, Pitocco D, Capoluongo E, Zuppi C, Ghirlanda G, Crea F, Andreotti F. IGF-1 and macrovascular complications of diabetes: alternative interpretations of recently published data. Diabetes Care 2003; 26:1653-4; author reply 1654-5. [PMID: 12716855 DOI: 10.2337/diacare.26.5.1653] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
|
41
|
Shi BM, Wang XY, Mu QL, Wu TH, Liu HJ, Yang Z. Angiogenesis effect on rat liver after administration of expression vector encoding vascular endothelial growth factor D. World J Gastroenterol 2003; 9:312-5. [PMID: 12532456 PMCID: PMC4611336 DOI: 10.3748/wjg.v9.i2.312] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/12/2002] [Revised: 08/04/2002] [Accepted: 08/23/2002] [Indexed: 02/06/2023] Open
Abstract
AIM To verify the expressing efficiency and angiogenesis effect after administration of expression vector encoding for vascular endothelial growth factor D in normal and ischemic rat liver. METHODS Ten female S-D rats were administrated with liver tissue dot injection of naked PCHO/hVEGF-D, 50 microg/dot, three dots for each. The same amount of physiological saline was used as control in the neighboring lobe. Fourteen S-D rats, using inflow occlusion of left lateral lobe, were divided into two groups, seven rats in each group. One was ischemic plasmid group, which received naked plasmid PCHO/hVEGF-D injection of 150 microg. The other received the equal amount of natural saline injection and designed as control. The expressions of hVEGF-D in mRNA and protein levels were identified by in situ hybridization and immunohistochemistry, respectively. Endothelial cells were labeled by the factor VIII immunohistochemistrically. The average number of peri-sinusoidal capillaries of each group was calculated and compared statistically 8 days after injection. RESULTS A large amount of hVEGF-D in mRNA level was found in both normal and ischemic plasmid groups and but none in their corresponding control groups. The protein of hVEGF was also highly expressed in both normal and ischemic plasmid groups than in the controls. The mean number of capillaries under microscopy (X200) of the plasmid group and control was 10.2+/-2.78 vs 7.1+/-2.02 (P<0.05), and those of ischemic plasmid group and ischemic control were 7.43+/-1.72 vs 4.71+/-1.11 with statistical difference (P<0.05). CONCLUSION The naked PCHO/hVEGF-D dot injection to normal, ischemic rat liver can produce comparatively high expression of hVEGF in both protein and mRNA levels, and prominently increase the number of new capillaries around hepatic sinuses. Therefore, it could be another ideal choice for the treatment of ischemic liver diseases.
Collapse
Affiliation(s)
- Bao-Min Shi
- Department of General Surgery, Shandong Provincial Hospital, Clinical College of Shandong University, 324 Jingwu Road, Jinan 250021, Shandong Province, China.
| | | | | | | | | | | |
Collapse
|
42
|
Abstract
PURPOSE OF REVIEW To provide an update on clinical trials of gene therapy for atherosclerotic cardiovascular disease published since 1 August 2001 and summarize the general advantages and potential problems of gene transfer in these disorders. RECENT FINDINGS There are two major areas in which gene therapy has entered clinical trials. The first is angiogenesis for coronary and peripheral arterial disease. Two relatively small placebo-controlled trials for coronary disease were reported, one using intramyocardial plasmid VEGF-2 gene, the other using intracoronary adenoviral FGF-4 gene. The VEGF-2 study in no-option patients showed reduced angina, and significant improvement in perfusion and function, whereas the FGF-4 study in less severely affected patients showed promising results in some subsets. In peripheral artery disease two phase 1 studies of adenoviral NV1FGF and VEGF showed some objective improvement in pain, ulcer size and ankle:brachial index in one study and endothelial function in the other. Both adenoviral and plasmid VEGF gene transfer at angioplasty increased vascularity in a phase 2 double-blind study. The other major area is the prevention of graft disease and restenosis using antisense oligodeoxynucleotides. E2F decoy led to a significant reduction in venous graft complications after ex-vivo transfection at the time of coronary bypass surgery, whereas the c-Myc oligodeoxynucleotide was ineffective in preventing in-stent coronary restenosis. SUMMARY There are more reviews of gene therapy for atherosclerosis in the literature than publications with original data or trials, but in the past year the imbalance is being redressed, with some promising results from controlled studies.
Collapse
Affiliation(s)
- Saul Benedict Freedman
- Department of Cardiology, University of Sydney, Concord Repatriation General Hospital, Australia.
| |
Collapse
|