51
|
Adenovirus-mediated expression of keratinocyte growth factor promotes secondary flap necrotic wound healing in an extended animal model. Aesthetic Plast Surg 2013; 37:1023-33. [PMID: 23949128 DOI: 10.1007/s00266-013-0200-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2013] [Accepted: 07/16/2013] [Indexed: 01/03/2023]
Abstract
BACKGROUND No effective treatments have been found for flap necrosis. Animal models that focus on the initial flap viability are inappropriate for necrotic wound studies. Keratinocyte growth factor (KGF) promotes keratinocyte proliferation with stronger activity and fewer complications and thus may be useful for necrotic flap wound healing. METHODS Rats with modified flap necrosis were randomly divided into four groups. An adenoviral vector expressing KGF was injected subdermally in the back of the animals after necrosis began. The expression and effect of KGF was assessed by real-time polymerase chain reaction, enzyme-linked immunoassay, and transwell, and wound healing was monitored. RESULTS The plasmid and adenovirus were able to express KGF and stimulate epithelial cell growth (p = 0.029). Histology showed that the necrosis healed fastest in the KGF administration group than in the control groups (p < 0.01). The adenovirus-mediated KGF (Ad-KGF) group had the thickest epithelium on days 15 (p = 0.044) and 25 (p = 0.014). The KGF level in the blood serum soared 10 and 15 days postoperatively (p < 0.01) but returned to baseline by day 25 (p = 0.561). The KGF mRNA levels in vivo increased dramatically in the Ad-KGF group (p = 0.037). CONCLUSIONS The extended flap model is applicable in necrotic wound study. Keratinocyte growth factor can promote secondary necrotic flap wound healing, and administration of KGF can be achieved by an adenoviral vector.
Collapse
|
52
|
Ha X, Ren J, Bi J, Zhang J, Wu Z. Therapeutic angiogenesis induced by human hepatocyte growth factor gene in hindlimb ischemia of dogs. Sci Bull (Beijing) 2013. [DOI: 10.1007/bf03325654] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
53
|
Hazekawa M, Morihata K, Yoshida M, Sakai Y, Uchida T. The angiogenic effect of ONO-1301, a novel long-acting prostacyclin agonist loaded in PLGA microspheres prepared using different molecular weights of PLGA, in a murine sponge model. Drug Dev Ind Pharm 2013; 40:1435-42. [DOI: 10.3109/03639045.2013.828220] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
54
|
Wang Z, Si LY. Hypoxia-inducible factor-1α and vascular endothelial growth factor in the cardioprotective effects of intermittent hypoxia in rats. Ups J Med Sci 2013; 118:65-74. [PMID: 23441597 PMCID: PMC3633332 DOI: 10.3109/03009734.2013.766914] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/06/2012] [Accepted: 12/11/2012] [Indexed: 11/16/2022] Open
Abstract
OBJECTIVE This study investigated the effects of short-term intermittent hypoxia (IH) preconditioning on cardiac structure and function in rats and the influence of ischemia reperfusion (I/R) injury. Special attention was then paid to the involvement of hypoxia-inducible factor-1α (HIF-1α) and vascular endothelial growth factor (VEGF). METHODS Wistar rats were given IH treatment for 1, 7, 14, or 28 days. Some of them were thereafter subject to myocardial infarction surgery. Right ventricle systolic pressure (RVSP), myocardial capillary density (CD), and mRNA/protein expression of HIF-1α, VEGF, and Bcl-2 in rat myocardial tissue were determined. Apoptotic cell number was determined by TUNEL staining, and concentrations of malondialdehyde (MDA) and superoxide dismutase (SOD) were measured. RESULTS IH treatment for 1, 7, 14, and 28 days reduced the myocardial infarction size, whereas IH for 28 days increased the RVSP, ratio of right to left ventricle weight (RV/LV+S), and CD. IH up-regulated the mRNA and protein levels of HIF-1α, VEGF, and Bcl-2 both under normal and I/R conditions. The induced expression of HIF-1α and VEGF by IH reached a peak after 7 days of treatment. Moreover, IH for 28 days induced cardiomyocyte apoptosis, whereas prior treatment with IH for 1, 7, 14, and 28 days all markedly attenuated the apoptosis effected by the subsequent I/R injury. IH also decreased the concentrations of MDA but increased those of SOD in myocardial tissue of both in normal rats and following I/R. CONCLUSIONS The present study demonstrates that short-term IH protects the heart from I/R injury through inhibiting apoptosis and oxidative stress. The up-regulation of HIF-1α and VEGF by short-term IH may participate in the cardioprotective effect of IH.
Collapse
Affiliation(s)
- Zhang Wang
- Department of Geriatrics, The First Affiliated Hospital, Third Military Medical University, Chongqing, China
| | - Liang-Yi Si
- Department of Geriatrics, The First Affiliated Hospital, Third Military Medical University, Chongqing, China
| |
Collapse
|
55
|
Dragneva G, Korpisalo P, Ylä-Herttuala S. Promoting blood vessel growth in ischemic diseases: challenges in translating preclinical potential into clinical success. Dis Model Mech 2013; 6:312-22. [PMID: 23471910 PMCID: PMC3597014 DOI: 10.1242/dmm.010413] [Citation(s) in RCA: 66] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
Angiogenic therapy, which involves the use of an exogenous stimulus to promote blood vessel growth, is an attractive approach for the treatment of ischemic diseases. It has been shown in animal models that the stimulation of blood vessel growth leads to the growth of the whole vascular tree, improvement of ischemic tissue perfusion and improved muscle aerobic energy metabolism. However, very few positive results have been gained from Phase 2 and 3 clinical angiogenesis trials. Many reasons have been given for the failures of clinical trials, including poor transgene expression (in gene-therapy trials) and instability of the vessels induced by therapy. In this Review, we discuss the selection of preclinical models as one of the main reasons why clinical translation has been unsuccessful thus far. This issue has received little attention, but could have had dramatic implications on the expectations of clinical trials. We highlight crucial differences between human patients and animal models with regards to blood flow and pressure, as well as issues concerning the chronic nature of ischemic diseases in humans. We use these as examples to demonstrate why the results from preclinical trials might have overestimated the efficacy of angiogenic therapies developed to date. We also suggest ways in which currently available animal models of ischemic disease could be improved to better mimic human disease conditions, and offer advice on how to work with existing models to avoid overestimating the efficacy of new angiogenic therapies.
Collapse
Affiliation(s)
- Galina Dragneva
- Department of Biotechnology and Molecular Medicine, A. I. Virtanen Institute, University of Eastern Finland, FI-70211 Kuopio, Finland
| | | | | |
Collapse
|
56
|
Yoshimura N, Miyazato M, Sasaki K, Yokoyama H, Oguchi T, Chancellor MB, Funahashi Y. Gene therapy for lower urinary tract dysfunction. Int J Urol 2012; 20:56-63. [DOI: 10.1111/j.1442-2042.2012.03226.x] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2012] [Accepted: 10/05/2012] [Indexed: 11/28/2022]
Affiliation(s)
- Naoki Yoshimura
- Department of Urology; University of Pittsburgh School of Medicine; Pittsburgh; Pennsylvania; USA
| | - Minoru Miyazato
- Department of Urology; University of Pittsburgh School of Medicine; Pittsburgh; Pennsylvania; USA
| | - Katsumi Sasaki
- Department of Urology; University of Pittsburgh School of Medicine; Pittsburgh; Pennsylvania; USA
| | - Hitoshi Yokoyama
- Department of Urology; University of Pittsburgh School of Medicine; Pittsburgh; Pennsylvania; USA
| | - Tomohiko Oguchi
- Department of Urology; University of Pittsburgh School of Medicine; Pittsburgh; Pennsylvania; USA
| | - Michael B Chancellor
- Department of Urology; Oakland University William Beaumont School of Medicine; Royal Oak; Michigan; USA
| | | |
Collapse
|
57
|
Shimizu K, Taniyama Y, Sanada F, Azuma J, Iwabayashi M, Iekushi K, Rakugi H, Morishita R. Hepatocyte Growth Factor Inhibits Lipopolysaccharide-Induced Oxidative Stress via Epithelial Growth Factor Receptor Degradation. Arterioscler Thromb Vasc Biol 2012; 32:2687-93. [DOI: 10.1161/atvbaha.112.300041] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Objective—
Lipopolysaccharide (LPS) triggers sepsis and systemic inflammatory response syndrome, which results in multiple organ failure. Our recent reports demonstrated that hepatocyte growth factor (HGF) attenuated angiotensin II–induced oxidative stress via epithelial growth factor receptor (EGFR) degradation in vascular smooth muscle cells. Here, we examined whether HGF can protect against systemic inflammatory response syndrome induced by LPS and investigated the mechanism.
Methods and Results—
HGF inhibited the increase in the expression of vascular cell adhesion molecule-1 and EGFR by LPS in vitro. HGF inhibited colocalization of EGFR and Src homology domain 2–containing inositol 5′-phosphatase 2. Furthermore, HGF inhibited reactive oxygen species production. We also injected LPS into HGF transgenic mice with increased HGF serum concentration and their littermates. HGF transgenic mice reduced LPS-induced vascular cell adhesion molecule-1 and reactive oxygen species compared with control, accompanied by significant EGFR degradation. Furthermore, HGF transgenic mice significantly improved survival in the LPS injection model.
Conclusion—
The present study revealed inhibition of LPS-induced vascular cell adhesion molecule-1 expression by HGF via the degradation of EGFR. We demonstrated that HGF regulated Src homology domain 2–containing inositol 5′-phosphatase 2 recruitment to EGFR and inhibited LPS-induced inflammation via EGFR degradation. This effect of HGF may be useful for the treatment of inflammatory disease.
Collapse
Affiliation(s)
- Kazutaka Shimizu
- From the Departments of Clinical Gene Therapy (K.S., Y.T., F.S., J.A., M.I., K.I., R.M.) and Geriatric Medicine and Nephrology (Y.T., J.A., K.I., H.R.), Osaka University Graduate School of Medicine, Yamadaoka, Suita, Japan
| | - Yoshiaki Taniyama
- From the Departments of Clinical Gene Therapy (K.S., Y.T., F.S., J.A., M.I., K.I., R.M.) and Geriatric Medicine and Nephrology (Y.T., J.A., K.I., H.R.), Osaka University Graduate School of Medicine, Yamadaoka, Suita, Japan
| | - Fumihiro Sanada
- From the Departments of Clinical Gene Therapy (K.S., Y.T., F.S., J.A., M.I., K.I., R.M.) and Geriatric Medicine and Nephrology (Y.T., J.A., K.I., H.R.), Osaka University Graduate School of Medicine, Yamadaoka, Suita, Japan
| | - Junya Azuma
- From the Departments of Clinical Gene Therapy (K.S., Y.T., F.S., J.A., M.I., K.I., R.M.) and Geriatric Medicine and Nephrology (Y.T., J.A., K.I., H.R.), Osaka University Graduate School of Medicine, Yamadaoka, Suita, Japan
| | - Masaaki Iwabayashi
- From the Departments of Clinical Gene Therapy (K.S., Y.T., F.S., J.A., M.I., K.I., R.M.) and Geriatric Medicine and Nephrology (Y.T., J.A., K.I., H.R.), Osaka University Graduate School of Medicine, Yamadaoka, Suita, Japan
| | - Kazuma Iekushi
- From the Departments of Clinical Gene Therapy (K.S., Y.T., F.S., J.A., M.I., K.I., R.M.) and Geriatric Medicine and Nephrology (Y.T., J.A., K.I., H.R.), Osaka University Graduate School of Medicine, Yamadaoka, Suita, Japan
| | - Hiromi Rakugi
- From the Departments of Clinical Gene Therapy (K.S., Y.T., F.S., J.A., M.I., K.I., R.M.) and Geriatric Medicine and Nephrology (Y.T., J.A., K.I., H.R.), Osaka University Graduate School of Medicine, Yamadaoka, Suita, Japan
| | - Ryuichi Morishita
- From the Departments of Clinical Gene Therapy (K.S., Y.T., F.S., J.A., M.I., K.I., R.M.) and Geriatric Medicine and Nephrology (Y.T., J.A., K.I., H.R.), Osaka University Graduate School of Medicine, Yamadaoka, Suita, Japan
| |
Collapse
|
58
|
Lu Q, Yao Y, Yao Y, Liu S, Huang Y, Lu S, Bai Y, Zhou B, Xu Y, Li L, Wang N, Wang L, Zhang J, Cheng X, Qin G, Ma W, Xu C, Tu X, Wang Q. Angiogenic factor AGGF1 promotes therapeutic angiogenesis in a mouse limb ischemia model. PLoS One 2012; 7:e46998. [PMID: 23110058 PMCID: PMC3479102 DOI: 10.1371/journal.pone.0046998] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2012] [Accepted: 09/11/2012] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND Peripheral arterial disease (PAD) is a common disease accounting for about 12% of the adult population, and causes significant morbidity and mortality. Therapeutic angiogenesis using angiogenic factors has been considered to be a potential treatment option for PAD patients. In this study, we assessed the potential of a new angiogenic factor AGGF1 for therapeutic angiogenesis in a critical limb ischemia model in mice for PAD. METHODS AND RESULTS We generated a unilateral hindlimb ischemia model in mice by ligation of the right common iliac artery and femoral artery. Ischemic mice with intrasmuscular administration of DNA for an expression plasmid for human AGGF1 (AGGF1 group) resulted in increased expression of both AGGF1 mRNA and protein after the administration compared with control mice with injection of the empty vector (control group). Color PW Doppler echocardiography showed that the blood flow in ischemic hindlimbs was significantly increased in the AGGF1 group compared to control mice at time points of 7, 14, and 28 days after DNA administration (n = 9/group, P = 0.049, 0.001, and 0.001, respectively). Increased blood flow in the AGGF1 group was correlated to increased density of CD31-positive vessels and decreased necrosis in muscle tissues injected with AGGF1 DNA compared with the control tissue injected with the empty vector. Ambulatory impairment was significantly reduced in the AGGF1 group compared to the control group (P = 0.004). The effect of AGGF1 was dose-dependent. At day 28 after gene transfer, AGGF1 was significantly better in increasing blood flow than FGF-2 (P = 0.034), although no difference was found for tissue necrosis and ambulatory impairment. CONCLUSIONS These data establish AGGF1 as a candidate therapeutic agent for therapeutic angiogenesis to treat PAD.
Collapse
Affiliation(s)
- Qiulun Lu
- Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology, Center for Human Genome Research, Cardio-X Institute, Huazhong University of Science and Technology, Wuhan, People’s Republic of China
| | - Yihong Yao
- Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology, Center for Human Genome Research, Cardio-X Institute, Huazhong University of Science and Technology, Wuhan, People’s Republic of China
| | - Yufeng Yao
- Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology, Center for Human Genome Research, Cardio-X Institute, Huazhong University of Science and Technology, Wuhan, People’s Republic of China
| | - Shizhi Liu
- Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology, Center for Human Genome Research, Cardio-X Institute, Huazhong University of Science and Technology, Wuhan, People’s Republic of China
| | - Yuan Huang
- Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology, Center for Human Genome Research, Cardio-X Institute, Huazhong University of Science and Technology, Wuhan, People’s Republic of China
| | - Shan Lu
- Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology, Center for Human Genome Research, Cardio-X Institute, Huazhong University of Science and Technology, Wuhan, People’s Republic of China
| | - Ying Bai
- Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology, Center for Human Genome Research, Cardio-X Institute, Huazhong University of Science and Technology, Wuhan, People’s Republic of China
| | - Bisheng Zhou
- Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology, Center for Human Genome Research, Cardio-X Institute, Huazhong University of Science and Technology, Wuhan, People’s Republic of China
| | - Yan Xu
- Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology, Center for Human Genome Research, Cardio-X Institute, Huazhong University of Science and Technology, Wuhan, People’s Republic of China
| | - Lei Li
- Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology, Center for Human Genome Research, Cardio-X Institute, Huazhong University of Science and Technology, Wuhan, People’s Republic of China
| | - Nan Wang
- Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology, Center for Human Genome Research, Cardio-X Institute, Huazhong University of Science and Technology, Wuhan, People’s Republic of China
| | - Li Wang
- Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology, Center for Human Genome Research, Cardio-X Institute, Huazhong University of Science and Technology, Wuhan, People’s Republic of China
| | - Jie Zhang
- Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology, Center for Human Genome Research, Cardio-X Institute, Huazhong University of Science and Technology, Wuhan, People’s Republic of China
| | - Xiang Cheng
- Institute of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People’s Republic of China
| | - Gangjian Qin
- Feinberg Cardiovascular Research Institute, Northwestern University Feinberg School of Medicine, Chicago, Illinois, United States of America
| | - Wei Ma
- The First Hospital of Wuhan City, Wuhan, People’s Republic of China
| | - Chengqi Xu
- Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology, Center for Human Genome Research, Cardio-X Institute, Huazhong University of Science and Technology, Wuhan, People’s Republic of China
| | - Xin Tu
- Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology, Center for Human Genome Research, Cardio-X Institute, Huazhong University of Science and Technology, Wuhan, People’s Republic of China
| | - Qing Wang
- Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology, Center for Human Genome Research, Cardio-X Institute, Huazhong University of Science and Technology, Wuhan, People’s Republic of China
- Center for Cardiovascular Genetics, Department of Molecular Cardiology, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, United States of America
| |
Collapse
|
59
|
Abstract
Peripheral arterial disease (PAD) is a common vascular disease that reduces blood flow capacity to the legs of patients. PAD leads to exercise intolerance that can progress in severity to greatly limit mobility, and in advanced cases leads to frank ischemia with pain at rest. It is estimated that 12 to 15 million people in the United States are diagnosed with PAD, with a much larger population that is undiagnosed. The presence of PAD predicts a 50% to 1500% increase in morbidity and mortality, depending on severity. Treatment of patients with PAD is limited to modification of cardiovascular disease risk factors, pharmacological intervention, surgery, and exercise therapy. Extended exercise programs that involve walking approximately five times per week, at a significant intensity that requires frequent rest periods, are most significant. Preclinical studies and virtually all clinical trials demonstrate the benefits of exercise therapy, including improved walking tolerance, modified inflammatory/hemostatic markers, enhanced vasoresponsiveness, adaptations within the limb (angiogenesis, arteriogenesis, and mitochondrial synthesis) that enhance oxygen delivery and metabolic responses, potentially delayed progression of the disease, enhanced quality of life indices, and extended longevity. A synthesis is provided as to how these adaptations can develop in the context of our current state of knowledge and events known to be orchestrated by exercise. The benefits are so compelling that exercise prescription should be an essential option presented to patients with PAD in the absence of contraindications. Obviously, selecting for a lifestyle pattern that includes enhanced physical activity prior to the advance of PAD limitations is the most desirable and beneficial.
Collapse
Affiliation(s)
- Tara L Haas
- Angiogenesis Research Group, Muscle Health Research Centre, Faculty of Health, York University, Toronto, Ontario, Canada
| | | | | | | |
Collapse
|
60
|
Makino H, Aoki M, Hashiya N, Yamasaki K, Azuma J, Sawa Y, Kaneda Y, Ogihara T, Morishita R. Long-term follow-up evaluation of results from clinical trial using hepatocyte growth factor gene to treat severe peripheral arterial disease. Arterioscler Thromb Vasc Biol 2012; 32:2503-9. [PMID: 22904270 DOI: 10.1161/atvbaha.111.244632] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
OBJECTIVE As angiogenic growth factors can stimulate the development of collateral arteries, a concept called therapeutic angiogenesis, we performed a phase I/IIa open-label clinical trial using intramuscular injection of naked plasmid DNA encoding hepatocyte growth factor (HGF). We reported long-term evaluation of 2 years after HGF gene therapy in 22 patients with severe peripheral arterial disease. METHODS AND RESULTS Twenty-two patients with peripheral arterial disease or Buerger disease staged by Fontaine IIb (n=7), III (n=4), and IV (n=11) were treated with HGF plasmid, either 2 mg or 4 mg ×2. Increase in ankle-branchial pressure index >0.1 was observed in 11 of 14 patients (79 %) at 2 years after gene therapy and in 11 of the 17 patients (65%) at 2 months. Reduction in rest pain (>2 cm in visual analog scale) was observed in 9 of 9 patients (100%) at 2 years and in 8 of 13 (62%) patients at 2 months. At 2 years, 9 of 10 (90%) ischemic ulcers reduced by >25%, accompanied by a reduction in the size of ulcer. Severe complications and adverse effects caused by gene transfer were not detected in any patient throughout the period up to 2 years. CONCLUSIONS Overall, the present study demonstrated long-term efficacy of HGF gene therapy up to 2 years. These findings may be cautiously interpreted to indicate that intramuscular injection of naked HGF plasmid is safe, feasible, and can achieve successful improvement of ischemic limbs as sole therapy.
Collapse
Affiliation(s)
- Hirofumi Makino
- Department of Clinical Gene Therapy, Graduate School of Medicine, Osaka University, 2-2 Yamada-oka, Suita 565-0871, Japan
| | | | | | | | | | | | | | | | | |
Collapse
|
61
|
Nakamura S, Ishihara M, Takikawa M, Kishimoto S, Isoda S, Fujita M, Sato M, Maehara T. Attenuation of limb loss in an experimentally induced hindlimb ischemic model by fibroblast growth factor-2/fragmin/protamine microparticles as a delivery system. Tissue Eng Part A 2012; 18:2239-47. [PMID: 22655590 DOI: 10.1089/ten.tea.2011.0741] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Fibroblast growth factor-2 (FGF-2) is a well-characterized protein that is used in the treatment of healing-impaired wounds. We previously reported that fragmin/protamine microparticles (F/P MPs) are useful as biodegradable carriers for the controlled release of cytokines. We examined the ability of FGF-2-containing (FGF-2/) F/P MPs to prevent limb loss in an experimentally induced ischemic hindlimb model using adult Balb/c-nu/nu male mice. One day after inducing ischemia, intramuscular injections of 100 μL of FGF-2/F/P MPs turbid suspension (10 μg/mL FGF-2 and 6 mg/mL F/P MPs) were administered into eight sites of the ischemic hindlimb. A 100-μL suspension of each of the following-10 μg/mL FGF-2, 6 mg/mL F/P MPs, and phosphate-buffered saline (PBS; the control)-was similarly injected into the hindlimb. From 5 days onward after the injections, recovery from ischemia was observed in the FGF-2/F/P MP-treated group, but only partial recovery occurred in the FGF-2-treated group. The F/P MP-treated and PBS-treated groups (i.e., control) exhibited no recovery from the ischemia. The histological evaluations of the hindlimbs also confirmed that the capillary (i.e., mature vessels) density was significantly higher in the FGF-2/F/P MP-treated group than in the other groups. The mice injected with FGF-2/F/P MPs also recovered hindlimb blood flow, as reflected by oxygen saturation and surface temperature evaluation. Our present approach using FGF-2/F/P MPs could be considered a valuable option for the therapeutic treatment of peripheral ischemic diseases.
Collapse
Affiliation(s)
- Shingo Nakamura
- Department of Surgery, National Defense Medical College, Tokorozawa, Saitama, Japan.
| | | | | | | | | | | | | | | |
Collapse
|
62
|
Nakamura S, Takikawa M, Ishihara M, Nakayama T, Kishimoto S, Isoda S, Ozeki Y, Sato M, Maehara T. Delivery system for autologous growth factors fabricated with low-molecular-weight heparin and protamine to attenuate ischemic hind-limb loss in a mouse model. J Artif Organs 2012; 15:375-85. [PMID: 22890790 DOI: 10.1007/s10047-012-0658-0] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2012] [Accepted: 07/23/2012] [Indexed: 12/18/2022]
Abstract
Frozen and thawed platelet-rich plasma (PRP) contains high concentrations of various growth factors, such as fibroblast growth factor (FGF)-2, vascular endothelial growth factor, and hepatocyte growth factor. We previously reported that low-molecular-weight heparin/protamine microparticles (LH/P MPs) are useful as biodegradable carriers for the controlled release of FGF-2. In this study, we examined the ability of PRP/LH/P MPs to prevent limb loss in an induced ischemic hind-limb model that used adult BALB/c-nu/nu male mice. One day after inducing ischemia, intramuscular injections of a PRP/LH/P MPs solution were administered into several sites of the ischemic hind limb. Seven days and onward after the injections, the PRP/LH/P MPs-treated and PRP-treated groups recovered from ischemia, as reflected by the improved oxygen saturation. In the PRP-treated group, however, the level of recovery of oxygen saturation after ischemia decreased after 14 days. From the 21st day onward, there was a significant difference between those two groups. In the LH/P MPs-treated group, a partial recovery occurred only in the early period. The saline-treated group (i.e., the control) and the noninjection group (i.e., ischemia only) exhibited no recovery. The limb survival rate at 1 year in the ischemia-induced mice injected with PRP/LH/P MPs was approximately 25 % (two of eight mice) but was absent in the other groups.
Collapse
Affiliation(s)
- Shingo Nakamura
- Department of Surgery, National Defense Medical College, 3-2 Namiki, Tokorozawa, Saitama, 359-8513, Japan.
| | | | | | | | | | | | | | | | | |
Collapse
|
63
|
Khazaei M, Salehi E. Rosiglitazone enhances neovascularization in diabetic rat ischemic hindlimb model. Biomed Pap Med Fac Univ Palacky Olomouc Czech Repub 2012; 156:312-7. [PMID: 23104577 DOI: 10.5507/bp.2012.052] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2011] [Accepted: 04/25/2012] [Indexed: 11/23/2022] Open
Abstract
BACKGROUND There is increasing evidence that peroxisome proliferator-activated receptors (PPARs) may be involved in the regulation of angiogenesis. In this study, we examined whether rosiglitazone, a PPARγ agonist, can restore angiogenesis in a rat hindlimb ischemia model of diabetes. METHODS Male wistar rats were divided into four groups (n=6 each): control, diabetic and control and diabetic rats who received rosiglitazone (8 mg/kg/day). Diabetes was induced by streptozotocin (55 mg/kg; ip). After 21 days, serum concentrations of nitric oxide (NO), vascular endothelial growth factor (VEGF) and soluble VEGF receptor-2 (VEGFR-2) were measured and neovascularization in ischemic legs was evaluated by immunohistochemistry. RESULTS Capillary density and capillary/fiber ratio in hindlimb ischemia of diabetic animals were significantly lower than the control group (P<0.05). Rosiglitazone significantly restored neovascularization in diabetic animals (P<0.05). CONCLUSIONS rosiglitazone enhances neovascularization in diabetic ischemic skeletal muscle and could be considered for treatment of peripheral artery disease in diabetic subjects.
Collapse
Affiliation(s)
- Majid Khazaei
- Department of Physiology, Isfahan University of Medical Sciences, Isfahan, Iran.
| | | |
Collapse
|
64
|
Pan-PPAR Agonist, Bezafibrate, Restores Angiogenesis in Hindlimb Ischemia in Normal and Diabetic Rats. INTERNATIONAL JOURNAL OF PEPTIDES 2012; 2012:637212. [PMID: 22701496 PMCID: PMC3371757 DOI: 10.1155/2012/637212] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/04/2012] [Accepted: 04/16/2012] [Indexed: 01/09/2023]
Abstract
Introduction. The aim of this study was to investigate the effect of bezafibrate as a pan-PPAR agonist on angiogenesis and serum nitrite, the main metabolite of nitric oxide (NO), vascular endothelial growth factor (VEGF) and VEGF receptor-2 (VEGFR-2) concentrations in hindlimb ischemia model of normal and type I diabetic rats. Methods. 28 male Wistar rats were divided into control and diabetic groups. Then, all rats underwent unilateral hindlimb ischemia. After recovery, they were randomly assigned to one of the following experimental groups: (1) control; (2) control + bezafibrate (400 mg/kg/day); (3) diabetic; (4) diabetic + beztafibrate. After three weeks, blood samples were taken and capillary density was evaluated in the gasterocnemius muscle of ischemic limb. Results. Bezafibrate increased capillary density and capillary/fiber ratio in ischemic leg of diabetic and control rats (P < 0.05). Serum VEGF and VEGFR-2 concentrations did not alter after bezafibrate administration, however, serum nitrite concentration was significantly higher in bezafibrate-treated groups than non-treated groups (P < 0.05). Discussion. It seems that bezafibrate, as a pan PPAR agonist, restores angiogenesis in hindlimb ischemic diabetic animals and is useful for prevention and/or treatment of peripheral artery disease in diabetic subjects.
Collapse
|
65
|
Bassi R, Trevisani A, Tezza S, Ben Nasr M, Gatti F, Vergani A, Farina A, Fiorina P. Regenerative therapies for diabetic microangiopathy. EXPERIMENTAL DIABETES RESEARCH 2012; 2012:916560. [PMID: 22536216 PMCID: PMC3321284 DOI: 10.1155/2012/916560] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 11/17/2011] [Accepted: 01/18/2012] [Indexed: 12/16/2022]
Abstract
Hyperglycaemia occurring in diabetes is responsible for accelerated arterial remodeling and atherosclerosis, affecting the macro- and the microcirculatory system. Vessel injury is mainly related to deregulation of glucose homeostasis and insulin/insulin-precursors production, generation of advanced glycation end-products, reduction in nitric oxide synthesis, and oxidative and reductive stress. It occurs both at extracellular level with increased calcium and matrix proteins deposition and at intracellular level, with abnormalities of intracellular pathways and increased cell death. Peripheral arterial disease, coronary heart disease, and ischemic stroke are the main causes of morbidity/mortality in diabetic patients representing a major clinical and economic issue. Pharmacological therapies, administration of growth factors, and stem cellular strategies are the most effective approaches and will be discussed in depth in this comprehensive review covering the regenerative therapies of diabetic microangiopathy.
Collapse
Affiliation(s)
- Roberto Bassi
- Nephrology Division, Transplantation Research Center (TRC), Children's Hospital, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
- DiSTeBA, Università del Salento, 73100 Lecce, Italy
| | | | - Sara Tezza
- Nephrology Division, Transplantation Research Center (TRC), Children's Hospital, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Moufida Ben Nasr
- Department of Biophysical and Medical Science, Higher Institute of Medical Technology, 1006 Tunis, Tunisia
| | - Francesca Gatti
- Nephrology Division, Transplantation Research Center (TRC), Children's Hospital, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
- DiSTeBA, Università del Salento, 73100 Lecce, Italy
| | - Andrea Vergani
- Nephrology Division, Transplantation Research Center (TRC), Children's Hospital, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
- Department of Medicine, San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Antonio Farina
- Department of Obstetrics and Gynecology, University of Bologna, 40138 Bologna, Italy
| | - Paolo Fiorina
- Nephrology Division, Transplantation Research Center (TRC), Children's Hospital, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
- Department of Medicine, San Raffaele Scientific Institute, 20132 Milan, Italy
| |
Collapse
|
66
|
Haraguchi Y, Shimizu T, Yamato M, Okano T. Concise review: cell therapy and tissue engineering for cardiovascular disease. Stem Cells Transl Med 2012. [PMID: 23197760 DOI: 10.5966/sctm.2012-0030] [Citation(s) in RCA: 60] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Cardiovascular disease is a major cause of morbidity and mortality, especially in developed countries. Various therapies for cardiovascular disease are investigated actively and are performed clinically. Recently, cell-based regenerative medicine using several cell sources has appeared as an alternative therapy for curing cardiovascular diseases. Scaffold-based or cell sheet-based tissue engineering is focused as a new generational cell-based regenerative therapy, and the clinical trials have also been started. Cell-based regenerative therapies have an enormous potential for treating cardiovascular disease. This review summarizes the recent research of cell sources and cell-based-regenerative therapies for cardiovascular diseases.
Collapse
Affiliation(s)
- Yuji Haraguchi
- Institute of Advanced Biomedical Engineering and Science, TWIns, Tokyo Women's Medical University, Tokyo, Japan
| | | | | | | |
Collapse
|
67
|
Ozturk A, Kucukardali Y, Tangi F, Erikci A, Uzun G, Bashekim C, Sen H, Terekeci H, Narin Y, Ozyurt M, Ozkan S, Sayan O, Rodop O, Nalbant S, Sıldıroglu O, Yalnız FF, Senkal IV, Sabuncu H, Oktenli C. Therapeutical potential of autologous peripheral blood mononuclear cell transplantation in patients with type 2 diabetic critical limb ischemia. J Diabetes Complications 2012; 26:29-33. [PMID: 22240264 DOI: 10.1016/j.jdiacomp.2011.11.007] [Citation(s) in RCA: 66] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/15/2010] [Revised: 11/20/2011] [Accepted: 11/30/2011] [Indexed: 12/19/2022]
Abstract
AIM The aim was to evaluate the therapeutic effectiveness of granulocyte colony-stimulating factor (G-CSF) mobilized peripheral blood mononuclear cells (PBMNCs) in critical limb ischemia (CLI) of type 2 diabetic patients. METHOD Forty diabetic patients with CLI were enrolled and randomized to treatment and control groups. In the treatment group, the patients received subcutaneous injections of recombinant human G-CSF (30 MU/day) for 5 days to mobilize stem cells. PBMNCs were collected and transplanted by multiple intramuscular injections of 1 ml in 1-1.5-cm depth into ischemic limbs. RESULTS At the end of 12 weeks of follow-up, the baseline and end point results in transplant group were as follows: Fontaine score improved from 3.8±03 to 3±0.5 (P=.0001), ankle brachial pressure index increased from 0.68±0.24 to 0.87±024 (P=.001), transcutaneous oxygen increased from 33±14 mmHg to 44±10 mmHg (P=.0001), and 6-min walking distance improved from 280±82 m to 338±98 m (P=.0001). Pain score decreased from 8.2±1.3 to 5.63±1.6 (P=.001), and the number of patients with limb ulcers was reduced from 9/20 (45%) to 3/20 (15%) (P=.031). In the control group, Fontaine score, 6-min walking distance, and pain score were improved; ankle brachial pressure index and transcutaneous oxygen pressure were not improved. The number of patients with limb ulcers did not change in the control group. There are improvement in amputation rates, collateral vessel development, and number of limb ulcers healed. CONCLUSIONS These results indicate that the autologous transplantation of G-CSF that mobilized PBMNCs in CLI diabetic patients is safe and effective in patient compliant reduction and improved perfusion.
Collapse
Affiliation(s)
- Ahmet Ozturk
- Gulhane Military Medical Academy Hematology Section, Istanbul, Turkey
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
68
|
Nonviral delivery of genetic medicine for therapeutic angiogenesis. Adv Drug Deliv Rev 2012; 64:40-52. [PMID: 21971337 DOI: 10.1016/j.addr.2011.09.005] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2011] [Revised: 06/29/2011] [Accepted: 09/18/2011] [Indexed: 01/08/2023]
Abstract
Genetic medicines that induce angiogenesis represent a promising strategy for the treatment of ischemic diseases. Many types of nonviral delivery systems have been tested as therapeutic angiogenesis agents. However, their delivery efficiency, and consequently therapeutic efficacy, remains to be further improved, as few of these technologies are being used in clinical applications. This article reviews the diverse nonviral gene delivery approaches that have been applied to the field of therapeutic angiogenesis, including plasmids, cationic polymers/lipids, scaffolds, and stem cells. This article also reviews clinical trials employing nonviral gene therapy and discusses the limitations of current technologies. Finally, this article proposes a future strategy to efficiently develop delivery vehicles that might be feasible for clinically relevant nonviral gene therapy, such as high-throughput screening of combinatorial libraries of biomaterials.
Collapse
|
69
|
Balasubramanian S, Venugopal P, Sundarraj S, Zakaria Z, Majumdar AS, Ta M. Comparison of chemokine and receptor gene expression between Wharton's jelly and bone marrow-derived mesenchymal stromal cells. Cytotherapy 2011; 14:26-33. [PMID: 22091833 DOI: 10.3109/14653249.2011.605119] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
BACKGROUND AIMS Because of their multilineage differentiation capacity, immunomodulatory role and homing ability, mesenchymal stromal cells (MSC) are emerging as a new therapeutic strategy for treating a variety of disorders. Although bone marrow (BM) is the best characterized source of MSC, Wharton's jelly (WJ) of the umbilical cord holds great promise as an alternative. As delivery direct to the site of injury is not always feasible, efficient homing of MSC to the site of injury is critical for inducing tissue repair and regeneration. MSC express a wide variety of growth factors, chemokines and receptors that are important for cell migration, homing and re-establishment of blood supply for recovery of damaged tissues. METHODS Detailed chemokine and receptor gene expression profiles of WJ MSC were established, and subsequently compared with those of BM-derived MSC using a polymerase chain reaction (PCR) array. Secretion of growth factors was analyzed and evaluated using culture supernatant from WJ and BM MSC. RESULTS Our results revealed a differential expression pattern of the chemokines and their receptors between WJ- and BM-derived MSC. Several Glutamic acid-Leucine-Arginine; ELR-positive CXC chemokine genes and secretion of growth factors, which promote angiogenesis, were found to be up-regulated in WJ MSC. CONCLUSIONS To understand better the localization and mechanism of tissue repair by transplanted WJ MSC, we attempted chemokine and their receptor transcription profiling, followed by analysis of growth factors secreted by WJ MSC, and compared them against those of BM MSC. The data suggest that MSC from different sources can be explored for distinct therapeutic roles.
Collapse
|
70
|
Mughal NA, Russell DA, Ponnambalam S, Homer-Vanniasinkam S. Gene therapy in the treatment of peripheral arterial disease. Br J Surg 2011; 99:6-15. [PMID: 22068822 DOI: 10.1002/bjs.7743] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/08/2011] [Indexed: 12/13/2022]
Abstract
BACKGROUND Peripheral arterial disease remains a significant global health burden despite revolutionary improvements in endovascular techniques over the past decade. The durability of intervention for critical limb ischaemia is poor, and the condition is associated with high morbidity and mortality rates. To address this deficiency, alternative therapeutic options are being explored. Advances in the fields of gene therapy and therapeutic angiogenesis have led to these being advocated as potential future treatments. METHODS Relevant medical literature from PubMed, Embase, the Cochrane Library and Google Scholar from the inception of these databases to June 2011 was reviewed. RESULTS Encouraging outcomes in preclinical trials using a variety of proangiogenic growth factors have led to numerous efficacy and safety studies. However, no clinical study has shown significant benefit for gene therapy over placebo. CONCLUSION Identifying the optimal site for gene delivery, choice of vector and duration of treatment is needed if gene therapy is to become a credible therapeutic option for peripheral arterial disease.
Collapse
Affiliation(s)
- N A Mughal
- Leeds Vascular Institute, Leeds General Infirmary, University of Leeds, Leeds, UK.
| | | | | | | |
Collapse
|
71
|
Saito Y, Nakagami H, Azuma N, Hirata S, Sanada F, Taniyama Y, Morishita R, Kaneda Y, Sasajima T. Critical roles of cold shock domain protein A as an endogenous angiogenesis inhibitor in skeletal muscle. Antioxid Redox Signal 2011; 15:2109-20. [PMID: 21473684 DOI: 10.1089/ars.2010.3714] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
UNLABELLED Angiogenesis is regulated by the local balance between angiogenic stimulators and inhibitors and is maintained by muscle-derived angiogenic factors in ischemic tissues. AIMS Our objectives were to investigate the effect of cold shock domain protein A (CSDA) as an endogenous angiogenesis inhibitor and to develop a novel strategy of therapeutic angiogenesis by blocking CSDA expression. RESULTS In human skeletal muscle cells, CSDA was upregulated during hypoxia when cells were damaged and apoptosis was induced. CSDA expression could repress the activity of hypoxia inducible factor-1α and nuclear factor κB, because CSDA can competitively bind the hypoxia response element and the nuclear factor κB-binding element. As a result, vascular endothelial growth factor-A, interleukin-6, and interleukin-8 secretions from skeletal muscle cells were decreased. Further, CSDA depletion increased the secretion level of these angiogenic factors. In a hindlimb ischemia model, transfer of short-hairpin RNA targeting CSDA ameliorated ischemia without direct transfer of angiogenic factors. In this ischemic tissue, vascular endothelial growth factor-A, interleukin-6, and CXCL2 protein levels were increased. INNOVATION AND CONCLUSION CSDA appears to play a critical role as an endogenous angiogenesis inhibitor in skeletal muscle, and RNA interference targeting of CSDA is a promising gene therapy strategy for treating peripheral arterial disease.
Collapse
Affiliation(s)
- Yukihiro Saito
- Department of Surgery, Asahikawa Medical University, 2-1 Midorigaoka-Higashi, Asahikawa, Hokkaido, Japan.
| | | | | | | | | | | | | | | | | |
Collapse
|
72
|
Haraguchi Y, Shimizu T, Yamato M, Okano T. Regenerative therapies using cell sheet-based tissue engineering for cardiac disease. Cardiol Res Pract 2011; 2011:845170. [PMID: 22007333 PMCID: PMC3189561 DOI: 10.4061/2011/845170] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/01/2011] [Revised: 08/11/2011] [Accepted: 08/14/2011] [Indexed: 12/12/2022] Open
Abstract
At present, cardiac diseases are a major cause of morbidity and mortality in the world. Recently, a cell-based regenerative medicine has appeared as one of the most potential and promising therapies for improving cardiac diseases. As a new generational cell-based regenerative therapy, tissue engineering is focused. Our laboratory has originally developed cell sheet-based scaffold-free tissue engineering. Three-dimensional myocardial tissue fabricated by stacking cardiomyocyte sheets, which are tightly interconnected to each other through gap junctions, beats simultaneously and macroscopically and shows the characteristic structures of native heart tissue. Cell sheet-based therapy cures the damaged heart function of animal models and is clinically applied. Cell sheet-based tissue engineering has a promising and enormous potential in myocardial tissue regenerative medicine and will cure many patients suffering from severe cardiac disease. This paper summarizes cell sheet-based tissue engineering and its satisfactory therapeutic effects on cardiac disease.
Collapse
Affiliation(s)
- Yuji Haraguchi
- Institute of Advanced Biomedical Engineering and Science, TWIns, Tokyo Women's Medical University, 8-1 Kawada-cho, Shinjuku-ku, Tokyo 162-8666, Japan
| | | | | | | |
Collapse
|
73
|
Okamoto N, Tanaka A, Jung K, Karasawa K, Orito K, Matsuda A, Amagai Y, Oida K, Ohmori K, Matsuda H. Silencing of int6 gene restores function of the ischaemic hindlimb in a rat model of peripheral arterial disease. Cardiovasc Res 2011; 92:209-17. [DOI: 10.1093/cvr/cvr203] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
|
74
|
Morishita R, Makino H, Aoki M, Hashiya N, Yamasaki K, Azuma J, Taniyama Y, Sawa Y, Kaneda Y, Ogihara T. Phase I/IIa clinical trial of therapeutic angiogenesis using hepatocyte growth factor gene transfer to treat critical limb ischemia. Arterioscler Thromb Vasc Biol 2010; 31:713-20. [PMID: 21183732 DOI: 10.1161/atvbaha.110.219550] [Citation(s) in RCA: 96] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
OBJECTIVE To evaluate the safety and feasibility of intramuscular gene transfer using naked plasmid DNA-encoding hepatocyte growth factor (HGF) and to assess its potential therapeutic benefit in patients with critical limb ischemia. METHODS AND RESULTS Gene transfer was performed in 22 patients with critical limb ischemia by intramuscular injection of HGF plasmid, either 2 or 4 mg, 2 times. Safety, ankle-brachial index, resting pain on a 10-cm visual analog scale, wound healing, and walking distance were evaluated before treatment and at 2 months after injection. No serious adverse event caused by gene transfer was detected over a follow-up of 6 months. Of particular importance, no peripheral edema, in contrast to that seen after treatment with vascular endothelial growth factor, was observed. In addition, the systemic HGF protein level did not increase during the study. At 2 months after gene transfer, the mean ± SD ankle-brachial index increased from 0.46 ± 0.08 to 0.59 ± 0.13 (P<0.001), the mean ± SD size of the largest ischemic ulcers decreased from 3.08 ± 1.54 to 2.32 ± 1.88 cm (P=0.007), and the mean ± SD visual analog scale score decreased from 5.92 ± 1.67 to 3.04 ± 2.50 cm (P=0.001). An increase in ankle-brachial index by >0.1, a reduction in ulcer size by >25%, and a reduction in visual analog scale score by >2 cm at 2 months after gene transfer were observed in 11 (64.7%) of 17 limbs, 18 (72%) of 25 ulcers, and 8 (61.5%) of 13 limbs, respectively. CONCLUSIONS Intramuscular injection of naked HGF plasmid is safe and feasible and can achieve successful improvement of ischemic limbs as sole therapy.
Collapse
Affiliation(s)
- Ryuichi Morishita
- Department of Clinical Gene Therapy, Osaka University Graduate School of Medicine, 2-2 Yamada-oka, Suita 565-0871, Japan.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
75
|
Powell RJ, Goodney P, Mendelsohn FO, Moen EK, Annex BH. Safety and efficacy of patient specific intramuscular injection of HGF plasmid gene therapy on limb perfusion and wound healing in patients with ischemic lower extremity ulceration: results of the HGF-0205 trial. J Vasc Surg 2010; 52:1525-30. [PMID: 21146749 PMCID: PMC5292269 DOI: 10.1016/j.jvs.2010.07.044] [Citation(s) in RCA: 109] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2010] [Revised: 07/21/2010] [Accepted: 07/22/2010] [Indexed: 12/16/2022]
Abstract
OBJECTIVES We have previously reported the results of a dose-finding phase II trial showing that HGF angiogenic gene therapy can increase TcPO2 compared with placebo in patients with critical limb ischemia (CLI). The purpose of this randomized placebo controlled multi-center trial was to further assess the safety and clinical efficacy of a modified HGF gene delivery technique in patients with CLI and no revascularization options. METHODS Patients with lower extremity ischemic tissue loss (Rutherford 5 and 6) received three sets of eight intramuscular injections every 2 weeks of HGF plasmid under duplex ultrasound guidance. Injection locations were individualized for each patient based on arteriographically defined vascular anatomy. Primary safety end point was incidence of adverse events (AE) or serious adverse events (SAE). Clinical end points included change from baseline in toe brachial index (TBI), rest pain assessment by a 10 cm visual analogue scale (VAS) as well as wound healing, amputation, and survival at 3 and 6 months. RESULTS Randomization ratio was 3:1 HGF (n = 21) vs placebo (n = 6). Mean age was 76 ± 2 years, with 56% male and 59% diabetic. There was no difference in demographics between groups. There was no difference in AEs or SAEs, which consisted mostly of transient injection site discomfort, worsening of CLI, and intercurrent illnesses. Change in TBI significantly improved from baseline at 6 months in the HGF-treated group compared with placebo (0.05 ± 0.05 vs -0.17 ± 0.04; P = .047). Change in VAS from baseline at 6 months was also significantly improved in the HGF-treated group compared with placebo (-1.9 ± 1.3 vs +0.06 ± 0.2; P = .04). Complete ulcer healing at 12 months occurred in 31% of the HGF group and 0% of the placebo (P = .28) There was no difference in major amputation of the treated limb (HGF 29% vs placebo 33%) or mortality at 12 months (HGF 19% vs placebo 17%) between groups. CONCLUSION HGF gene therapy using a patient vascular anatomy specific delivery technique appears safe, maintained limb perfusion, and decreased rest pain in patients with CLI compared with placebo. A larger study to assess the efficacy of this therapy on more clinically relevant end points is warranted.
Collapse
Affiliation(s)
- Richard J Powell
- Section of Vascular Surgery, Dartmouth-Hitchcock Medical Center, Lebanon, NH 03756, USA.
| | | | | | | | | |
Collapse
|
76
|
Impaired myocardium regeneration with skeletal cell sheets--a preclinical trial for tissue-engineered regeneration therapy. Transplantation 2010; 90:364-72. [PMID: 20555308 DOI: 10.1097/tp.0b013e3181e6f201] [Citation(s) in RCA: 108] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
BACKGROUND We hypothesized that autologous skeletal cell (SC) sheets regenerate the infract myocardium in porcine heart as a preclinical trial. METHODS AND RESULTS The impaired heart was created by implantation of ameroid constrictor on left anterior descending for 4 weeks. SCs isolated from leg muscle were cultured and detached from the temperature-responsive domain-coated dishes as single monolayer cell sheet at 20 degrees C. The following therapies were conducted: SC sheets (SC group, n=5); sham (C group n=5). Echocardiography demonstrated that cardiac performance was significantly improved in the SC group 3 and 6 months after operation (fractional area shortening, 3 months; SC vs. C=49.5+/-2.8 vs. 24.6+/-2.0%, P<0.05) and left ventricle dilatation was well attenuated in the SC group. Color kinesis index showed that distressed regional diastolic and systolic function in infarcted anterior wall was significantly recovered (SC vs. C=57.4+/-8.6 vs. 30.2+/-4.7%, P<0.05, diastolic: 58.5+/-4.5 vs. 35.4+/-6.6%, P<0.05, systolic). Factor VIII immunostains demonstrated that vascular density was significantly higher in the SC group than the C group. And % fibrosis and cell diameter were significantly lower in the SC group. And hematoxylin-eosin staining depicted that skeletal origin cells and well-developed-layered smooth muscle cells were detected in the implanted area. Positron emission tomography showed better myocardial perfusion and more viable myocardial tissue in the distressed myocardium receiving SC sheets compared with the myocardium receiving no sheets. CONCLUSIONS SC sheet implantation improved cardiac function by attenuating the cardiac remodeling in the porcine ischemic myocardium, suggesting a promising strategy for myocardial regeneration therapy in the impaired myocardium.
Collapse
|
77
|
Pyun WB, Hahn W, Kim DS, Yoo WS, Lee SD, Won JH, Rho BS, Park ZY, Kim JM, Kim S. Naked DNA expressing two isoforms of hepatocyte growth factor induces collateral artery augmentation in a rabbit model of limb ischemia. Gene Ther 2010; 17:1442-52. [PMID: 20668482 DOI: 10.1038/gt.2010.101] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Hepatocyte growth factor (HGF) has been shown to induce angiogenesis in vivo and has potential as a candidate gene for 'therapeutic angiogenesis'. In vivo, two isoforms of HGF, HGF₇₂₃ and HGF₇₂₈, consisting of 723 and 728 amino acids, are generated through alternative splicing between exons 4 and 5, but the biological effects of their coexpression have not yet been elucidated. In this study, we generated a series of genomic-complementary DNA (cDNA) hybrids of the HGF gene by inserting various truncated intron 4 into the junction of exons 4 and 5 of HGF cDNA and analyzed the biological activities of these hybrid constructs. We showed that: (1) the hybrid called HGF-X7, which contained 1502 base pairs of intron 4, could drive a higher level of HGF expression than other hybrid constructs and cDNAs of each isoform alone; (2) the pCK vector was most efficient for the gene expression of HGF-X7; (3) coexpression of both isoforms of HGF could more efficiently induce the migration of human umbilical vein endothelial cell (HUVEC) and of the mouse myoblast cell line C₂C₁₂ myoblasts than a single isoform of HGF and human vascular endothelial growth factor (VEGF)₁₆₅ at a given concentration; (4) intramuscular administration of pCK-HGF-X7 resulted in transient and localized HGF expression in the injected muscle without an increase in the HGF protein levels in other tissues including serum; and (5) intramuscular injection of pCK-HGF-X7 could more efficiently increase the number of angiographically recognizable collateral vessels, as well as improve an intra-arterial Doppler wire-measured blood flow in the rabbit model of hindlimb ischemia when compared with the identical vector encoding VEGF₁₆₅ gene. These results showed that transfer of the genomic-cDNA hybrid of the HGF gene could be used as a potential therapeutic approach to human vascular diseases.
Collapse
Affiliation(s)
- W-B Pyun
- Department of Cardiology, Medical College of Ewha Womans University, Mokdong Hospital, Seoul, Korea
| | | | | | | | | | | | | | | | | | | |
Collapse
|
78
|
Shigematsu H, Yasuda K, Iwai T, Sasajima T, Ishimaru S, Ohashi Y, Yamaguchi T, Ogihara T, Morishita R. Randomized, double-blind, placebo-controlled clinical trial of hepatocyte growth factor plasmid for critical limb ischemia. Gene Ther 2010; 17:1152-61. [DOI: 10.1038/gt.2010.51] [Citation(s) in RCA: 144] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
|
79
|
Barbato JE, Kibbe MR, Tzeng E. The Emerging Role of Gene Therapy in the Treatment of Cardiovascular Diseases. Crit Rev Clin Lab Sci 2010. [DOI: 10.1080/10408360390250621] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
|
80
|
Germani A, Di Campli C, Pompilio G, Biglioli P, Capogrossi MC. Regenerative therapy in peripheral artery disease. Cardiovasc Ther 2010; 27:289-304. [PMID: 19903190 DOI: 10.1111/j.1755-5922.2009.00105.x] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Patients with peripheral artery disease (PAD) and critical limb ischemia are the main candidates for limb amputations and have a poor life expectancy. Frequently, these patients are not eligible for either surgical or percutaneous interventions aimed at mechanical revascularization. Therefore, new strategies need to be identified to offer these patients a viable therapeutic option. Gene and cell therapy hold great promise for the treatment of peripheral vascular diseases because, in animal models, local delivery of growth factors and endothelial progenitor cells result in new blood vessel formation and regeneration of ischemic tissues. In this article, are reviewed phase I and phase II gene, and cell therapy clinical trials in patients with PAD.
Collapse
|
81
|
Aizawa K, Sato S, Terakawa M, Saitoh D, Tsuda H, Ashida H, Obara M. Accelerated adhesion of grafted skin by laser-induced stress wave-based gene transfer of hepatocyte growth factor. JOURNAL OF BIOMEDICAL OPTICS 2009; 14:064043. [PMID: 20059281 DOI: 10.1117/1.3253325] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/28/2023]
Abstract
Gene therapy using wound healing-associated growth factor gene has received much attention as a new strategy for improving the outcome of tissue transplantation. We delivered plasmid DNA coding for human hepatocyte growth factor (hHGF) to rat free skin grafts by the use of laser-induced stress waves (LISWs); autografting was performed with the grafts. Systematic analysis was conducted to evaluate the adhesion properties of the grafted tissue; angiogenesis, cell proliferation, and reepithelialization were assessed by immunohistochemistry, and reperfusion was measured by laser Doppler imaging as a function of time after grafting. Both the level of angiogenesis on day 3 after grafting and the increased ratio of blood flow on day 4 to that on day 3 were significantly higher than those in five control groups: grafting with hHGF gene injection alone, grafting with control plasmid vector injection alone, grafting with LISW application alone, grafting with LISW application after control plasmid vector injection, and normal grafting. Reepithelialization was almost completed on day 7 even at the center of the graft with LISW application after hHGF gene injection, while it was not for the grafts of the five control groups. These findings demonstrate the validity of our LISW-based HGF gene transfection to accelerate the adhesion of grafted skins.
Collapse
Affiliation(s)
- Kazuya Aizawa
- Keio University, Department of Electronics and Electrical Engineering, 3-14-1 Hiyoshi, Kohoku-ku, Yokohama, Kanagawa 223-8522, Japan
| | | | | | | | | | | | | |
Collapse
|
82
|
Yamamoto Y, Matsuura T, Narazaki G, Sugitani M, Tanaka K, Maeda A, Shiota G, Sato K, Yoshida A, Hisatome I. Synergistic effects of autologous cell and hepatocyte growth factor gene therapy for neovascularization in a murine model of hindlimb ischemia. Am J Physiol Heart Circ Physiol 2009; 297:H1329-36. [DOI: 10.1152/ajpheart.00321.2009] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Autologous cell implantation and angiogenic gene therapy have been evaluated in critical limb ischemic patients. Here, we compared the features of these strategies individually and in combination. C57BL/6J mice with ischemic hindlimbs were injected with adherent mononuclear cells (aMNCs) from bone marrow or adenovirus encoding the hepatocyte growth factor (HGF) gene (Ad-HGF). Under comparable angiogenic conditions, 10 × 105 aMNCs produced significantly higher amounts of VEGF and FGF-2 and stimulated the number of arterioles in ischemic muscle compared with 1 × 108 plaque-forming units (pfu) of Ad-HGF. Ad-HGF produced 10 times more HGF in ischemic muscle compared with aMNCs. Injection of 0.3 × 105 aMNCs previously transfected with Ad-HGF (aMNC/Ad-HGF) increased blood flow and elevated the numbers of capillaries and arterioles to levels comparable with that seen with 10 × 105 aMNCs or 1 × 108 pfu of Ad-HGF. Hypoxic conditions induced the apoptotic death of aMNCs. However, coincubation with HGF or aMNC/Ad-HGF protected cells against apoptosis. HGF stimulated the migration of aMNCs, and the migration capacity of the aMNC/Ad-HGF group was significantly higher than that in the aMNC/Ad-LacZ group. In conclusion, cell-based HGF gene therapy decreased the number of cells required for neovascularization. This strategy can be an effective angiogenic therapy.
Collapse
Affiliation(s)
- Yasutaka Yamamoto
- Division of Regenerative Medicine and Therapeutics, Institute of Regenerative Medicine and Biofunction, Graduate School of Medical Sciences,
| | - Takashi Matsuura
- Division of Regenerative Medicine and Therapeutics, Institute of Regenerative Medicine and Biofunction, Graduate School of Medical Sciences,
| | - Genta Narazaki
- Division of Regenerative Medicine and Therapeutics, Institute of Regenerative Medicine and Biofunction, Graduate School of Medical Sciences,
| | - Miyoko Sugitani
- Division of Regenerative Medicine and Therapeutics, Institute of Regenerative Medicine and Biofunction, Graduate School of Medical Sciences,
| | - Kohei Tanaka
- Division of Regenerative Medicine and Therapeutics, Institute of Regenerative Medicine and Biofunction, Graduate School of Medical Sciences,
| | - Akihiro Maeda
- Division of Molecular Biology, Department of Molecular and Cellular Biology, School of Life Sciences, Faculty of Medicine, and
| | - Goshi Shiota
- Division of Molecular and Genetic Medicine, Institute of Regenerative Medicine and Biofunction, Graduate School of Medical Sciences, Tottori University, Tottori, Japan
| | - Kenzo Sato
- Division of Molecular Biology, Department of Molecular and Cellular Biology, School of Life Sciences, Faculty of Medicine, and
| | - Akio Yoshida
- Division of Regenerative Medicine and Therapeutics, Institute of Regenerative Medicine and Biofunction, Graduate School of Medical Sciences,
| | - Ichiro Hisatome
- Division of Regenerative Medicine and Therapeutics, Institute of Regenerative Medicine and Biofunction, Graduate School of Medical Sciences,
| |
Collapse
|
83
|
Yang ZJ, Xu SL, Chen B, Zhang SL, Zhang YL, Wei W, Ma DC, Wang LS, Zhu TB, Li CJ, Wang H, Cao KJ, Gao W, Huang J, Ma WZ, Wu ZZ. HEPATOCYTE GROWTH FACTOR PLAYS A CRITICAL ROLE IN THE REGULATION OF CYTOKINE PRODUCTION AND INDUCTION OF ENDOTHELIAL PROGENITOR CELL MOBILIZATION: A PILOT GENE THERAPY STUDY IN PATIENTS WITH CORONARY HEART DISEASE. Clin Exp Pharmacol Physiol 2009; 36:790-6. [DOI: 10.1111/j.1440-1681.2009.05151.x] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
84
|
Wang N, Tong G, Yang J, Zhou Z, Pan H, Huo Y, Xu J, Zhang X, Ling F, Li P. Effect of hepatocyte growth-promoting factors on myocardial ischemia during exercise in patients with severe coronary artery disease. Int Heart J 2009; 50:291-9. [PMID: 19506333 DOI: 10.1536/ihj.50.291] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Hepatocyte growth-promoting factor (pHGF) has the greatest potential as a therapeutic agent for vascular growth factor. The aim of this study was to investigate the effect of pHGF on myocardial ischemia and exercise capacity in patients with severe coronary artery disease (CAD). Forty-nine patients were enrolled for a two week treatment period. Treadmill graded exercise tests with gas analysis were conducted before and after therapy. Serum levels of HGF were significantly elevated after therapy. The degrees of exercise-induced ST segment depression were decreased more significantly in the pHGF group. Similar differences were also found in the maximum heart rate and the maximum heart rate when the ST segment was depressed 1 mm while undergoing the treadmill graded exercise test. Both were increased more significantly in the pHGF group. Total exercise time, systolic blood pressure in the peak of exercise, the length of time that ST segment depression of 1 mm is needed, and total work all were increased in both groups after intervention. Furthermore, total exercise time and total work were increased more significantly in the pHGF group. The levels of HGF increased significantly after pHGF treatment. pHGF could favorably improve exercise-induced myocardial ischemia and enhance exercise capacity in patients with severe CAD.
Collapse
Affiliation(s)
- Ningfu Wang
- Department of Cardiology, the First People's Hospital of Hangzhou and Affiliated Hangzhou Hospital of Nanjing Medical University, Hangzhou City, China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
85
|
Iwashima S, Ozaki T, Maruyama S, Saka Y, Kobori M, Omae K, Yamaguchi H, Niimi T, Toriyama K, Kamei Y, Torii S, Murohara T, Yuzawa Y, Kitagawa Y, Matsuo S. Novel culture system of mesenchymal stromal cells from human subcutaneous adipose tissue. Stem Cells Dev 2009; 18:533-43. [PMID: 19055360 DOI: 10.1089/scd.2008.0358] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Accumulating evidence suggests that the delivery of human adipose tissue-derived stromal cells (hASCs) has great potential as regenerative therapy. This was performed to develop a method for expanding hASCs by reducing the amount of serum required. We demonstrate that hASCs were able to expand efficiently in media containing 2% serum and fibroblast growth factor-2. These cells, or low serum cultured hASCs (hLASCs), expressed cell surface markers similar to those on bone marrow-derived mesenchymal stem cells, and could be differentiated into cells of mesenchymal lineage. Of interest, hLASCs secreted higher levels of vascular endothelial growth factor (VEGF) and hepatocyte growth factor (HGF) than hASCs cultured in 20% serum (hHASCs). Moreover, hLASC-conditioned media significantly increased endothelial cell (EC) proliferation and decreased EC apoptosis compared to that obtained from hHASCs or control media only. Antibodies against VEGF and HGF virtually negated these effects. When hASCs were administered into the ischemic hindlimbs of nude rats, hLASCs improved blood flow, increased capillary density, and raised the levels of VEGF and HGF in the muscles as compared with hHASCs. In conclusion, we demonstrate a novel low serum culture system for hASCs, which may have great potential in regenerative cell therapy for damaged organs in the clinical setting.
Collapse
Affiliation(s)
- Shigejiro Iwashima
- Department of Nephrology, Nagoya University Graduate School of Medicine, Showaku, Nagoya, Aichi, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
86
|
Ono I, Yamashita T, Hida T, Jin HY, Ito Y, Hamada H, Akasaka Y, Ishii T, Jimbow K. Combined administration of basic fibroblast growth factor protein and the hepatocyte growth factor gene enhances the regeneration of dermis in acute incisional wounds. Wound Repair Regen 2009. [DOI: 10.1111/j.1067-1927.2004.012113.x-1] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
|
87
|
Assessment of myocardial blood perfusion improved by CD151 in a pig myocardial infarction model. Acta Pharmacol Sin 2009; 30:70-7. [PMID: 19079294 DOI: 10.1038/aps.2008.10] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
AIM To appraise the efficacy of CD151-induced myocardial therapeutic angiogenesis in a pig myocardial infarction model. METHODS CD151 and anti-CD151 were constructed into the recombinant adeno-associated virus (rAAV) vector. All 26 pigs were subjected to coronary artery ligation or no surgery. Eight weeks after coronary artery ligation, the expression of CD151 was measured by Western blot and immunostaining. Capillary density was evaluated using immunostaining for von Willebrand factor (vWF). 13N-labeled NH3 positron emission computed tomography ([13N]NH3PET) was measured to assess regional myocardial perfusion and the defect area. RESULTS CD151 gene delivery could increase the expression of CD151 at protein level. Over-expression of CD151 increased the density of total capillaries in the ischemic myocardium, significantly improved the blood perfusion and reduced the defect area percentage. CONCLUSION This study demonstrated that the rAAV-mediated CD151 gene delivery promoted efficient neovascularization and increased the blood perfusion after myocardial infarction in pigs.
Collapse
|
88
|
Matsubara H, Tsuchiya H, Watanabe K, Takeuchi A, Tomita K. Percutaneous nonviral delivery of hepatocyte growth factor in an osteotomy gap promotes bone repair in rabbits: a preliminary study. Clin Orthop Relat Res 2008; 466:2962-72. [PMID: 18813894 PMCID: PMC2628245 DOI: 10.1007/s11999-008-0493-z] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/12/2008] [Accepted: 08/19/2008] [Indexed: 01/31/2023]
Abstract
Hepatocyte growth factor (HGF) was initially identified in cultured hepatocytes and subsequently reported to induce angiogenic, morphogenic, and antiapoptotic activity in various tissues. These properties suggest a potential influence of HGF on bone healing. We asked if gene transfer of human HGF (hHGF) into an osteotomy gap with a hemagglutinating virus of Japan-envelope (HVJ-E) vector promotes bone healing in rabbits. HVJ-E that contained either hHGF or control plasmid was percutaneously injected into the osteotomy gap of rabbit tibias on Day 14. The osteotomy gap was evaluated by radiography, pQCT, mechanical tests, and histology at Week 8. The expression of hHGF was evaluated by reverse transcriptase-polymerase chain reaction and immunohistochemistry at Week 3. Radiography, pQCT, and histology suggested the hHGF group had faster fracture healing. Mechanical tests demonstrated the hHGF group had greater mechanical strength. The injected tissues at 3 weeks expressed hHGF mRNA by reverse transcriptase-polymerase chain reaction. hHGF-positive immunohistochemical staining was observed in various cells at the osteotomy gap at Week 3. The data suggest delivery of hHGF plasmid into the osteotomy gap promotes fracture repair, and HGF could become a novel agent for fracture treatment.
Collapse
Affiliation(s)
- Hidenori Matsubara
- Department of Orthopaedic Surgery, Graduate School of Medical Science, Kanazawa University, 13-1 Takara-machi, Kanazawa, 920-8641 Japan
| | - Hiroyuki Tsuchiya
- Department of Orthopaedic Surgery, Graduate School of Medical Science, Kanazawa University, 13-1 Takara-machi, Kanazawa, 920-8641 Japan
| | - Koji Watanabe
- Department of Orthopaedic Surgery, Graduate School of Medical Science, Kanazawa University, 13-1 Takara-machi, Kanazawa, 920-8641 Japan
| | - Akihiko Takeuchi
- Department of Orthopaedic Surgery, Graduate School of Medical Science, Kanazawa University, 13-1 Takara-machi, Kanazawa, 920-8641 Japan
| | - Katsuro Tomita
- Department of Orthopaedic Surgery, Graduate School of Medical Science, Kanazawa University, 13-1 Takara-machi, Kanazawa, 920-8641 Japan
| |
Collapse
|
89
|
|
90
|
Sugano M, Iwasaki Y, Abe M, Maeda T, Tsuchida K, Makino N. TNF-alpha employs a protein-tyrosine phosphatase to inhibit activation of hepatocyte growth factor receptor and hepatocyte growth factor-induced endothelial cell proliferation. Mol Cell Biochem 2008; 322:113-7. [DOI: 10.1007/s11010-008-9946-7] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2008] [Accepted: 10/22/2008] [Indexed: 01/22/2023]
|
91
|
Hu W, Criswell MH, Fong SL, Temm CJ, Rajashekhar G, Cornell TL, Clauss MA. Differences in the temporal expression of regulatory growth factors during choroidal neovascular development. Exp Eye Res 2008; 88:79-91. [PMID: 19013152 DOI: 10.1016/j.exer.2008.10.014] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2008] [Revised: 10/13/2008] [Accepted: 10/14/2008] [Indexed: 11/29/2022]
Abstract
Although the roles of vascular endothelial growth factor (VEGF), basic fibroblast growth factor (bFGF), and hepatocyte growth factor (HGF) in pathologic neovascularization have been well characterized in certain tissues, their particular functions and expression patterns in choroidal neovascularization (CNV) have not been clearly established. After localized laser trauma to Bruch's membrane to induce CNV development, the temporal changes in mRNA and protein expression of these 3 cytokines were documented and compared histologically to areas of immunofluorescence, the proliferation of endothelial cells, neovascular development, and temporal changes in vascular permeability. Changes in mRNA and protein levels of bFGF and HGF occurred quickly and reached peak expression within hours. This activity corresponded in time to intense and localized immunofluorescence for these cytokines within the choriocapillaris within laser lesion sites. During this same initial time period, mRNA upregulation of VEGF occurred, primarily within the neural retina and this expression corresponded to intense immunolabeling of Müller cells immediately adjacent to the lesion sites. By 3 days after lasering, increased VEGF(164) protein expression was measurable, whereas early neovascular development histologically corresponded to HGF and bFGF mRNA expansion into the developing choroidal neovascular membrane (CNVM). At 7 days, CNV expansion, maturation, and increased vascular permeability corresponded to peak VEGF mRNA and protein expression and to immunofluorescence of the CNVM. Differences also occurred in the expression of precursor and activated isoforms of these cytokines in the retinal pigment epithelium/choroid as compared to those in the retina. These molecular and immunocytochemical results suggest that bFGF and HGF may be important as initial regulators neovascularization in this CNV model; whereas VEGF may be important during later phases of angiogenesis and neovascular hyperpermeability.
Collapse
Affiliation(s)
- Wenzheng Hu
- Department of Ophthalmology, Indiana University School of Medicine, Indianapolis, IN 46202-5175, USA
| | | | | | | | | | | | | |
Collapse
|
92
|
Phase I clinical trial on intracoronary administration of Ad-hHGF treating severe coronary artery disease. Mol Biol Rep 2008; 36:1323-9. [DOI: 10.1007/s11033-008-9315-3] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2008] [Accepted: 07/03/2008] [Indexed: 10/21/2022]
|
93
|
Yasuda Y, Koyama H, Tabata Y, Fujihara Y, Oba M, Uchinuma E, Takato T. Controlled Delivery of bFGF Remodeled Vascular Network in Muscle Flap and Increased Perfusion Capacity Via Minor Pedicle. J Surg Res 2008; 147:132-7. [DOI: 10.1016/j.jss.2007.10.009] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2007] [Revised: 09/05/2007] [Accepted: 10/05/2007] [Indexed: 12/25/2022]
|
94
|
van Weel V, van Tongeren RB, van Hinsbergh VWM, van Bockel JH, Quax PHA. Vascular growth in ischemic limbs: a review of mechanisms and possible therapeutic stimulation. Ann Vasc Surg 2008; 22:582-97. [PMID: 18504100 DOI: 10.1016/j.avsg.2008.02.017] [Citation(s) in RCA: 63] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2007] [Revised: 01/15/2008] [Accepted: 02/29/2008] [Indexed: 01/13/2023]
Abstract
Stimulation of vascular growth to treat limb ischemia is promising, and early results obtained from uncontrolled clinical trials using angiogenic agents, e.g., vascular endothelial growth factor, led to high expectations. However, negative results from recent placebo-controlled trials warrant further research. Here, current insights into mechanisms of vascular growth in the adult, in particular the role of angiogenic factors, the immune system, and bone marrow, were reviewed, together with modes of its therapeutic stimulation and results from recent clinical trials. Three concepts of vascular growth have been described to date-angiogenesis, vasculogenesis, and arteriogenesis (collateral artery growth)-which represent different aspects of an integrated process. Stimulation of arteriogenesis seems clinically most relevant and has most recently been attempted using autologous bone marrow transplantation with some beneficial results, although the mechanism of action is not completely understood. Better understanding of the highly complex molecular and cellular mechanisms of vascular growth may yet lead to meaningful clinical applications.
Collapse
Affiliation(s)
- V van Weel
- Department of Surgery, Leiden University Medical Center, Leiden, the Netherlands
| | | | | | | | | |
Collapse
|
95
|
Wester T, Jørgensen JJ, Stranden E, Sandbæk G, Tjønnfjord G, Bay D, Kollerøs D, Kroese AJ, Brinchmann JE. Treatment with Autologous Bone Marrow Mononuclear Cells in Patients with Critical Lower Limb Ischaemia. A Pilot Study. Scand J Surg 2008; 97:56-62. [DOI: 10.1177/145749690809700108] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Background and Aims: Treatment with autologous, bone marrow mononuclear stem cells has shown effects in patients with chronic limb ischaemia in one randomized clinical study. The aim of the study was to test the potential effect of stem cell treatment in a strict defined group of patients with stable critical limb ischaemia (CLI). Design: A prospective, combined-centre pilot study. Material: Eight patients with CLI of the lower extremities, and without any other treatment options. Methods: Bone marrow cells were harvested from the patient's iliac crest and, after separation, injected into the calf muscles of the affected leg. Outcome was evaluated by digital subtraction angiography (DSA), visual analogue scale (VAS) and several non-invasive circulatory physiological tests. Results: There were no complications from the procedures. Two patients were amputated two months after cell injection. Five patients reported pain relief after four months. Five patients could be evaluated at eight months. According to VAS and physiological tests, they were all either stable or showed improvement. Conclusion: This method seems to be a safe option for treating patients with CLI. Inclusion of patients took a long time, mainly because many patients with CLI are offered endovascular treatment in our institution. While symptomatic improvement was found in individual patients, larger trials are required to investigate efficacy. This will probably require multi-centre participation.
Collapse
Affiliation(s)
- T. Wester
- Department of Vascular Surgery, Aker University Hospital
| | | | - E. Stranden
- Department of Vascular Diagnosis and Research, Aker University Hospital
| | - G. Sandbæk
- Department of Radiology, Oslo Vascular Centre, Aker University Hospital
| | - G. Tjønnfjord
- Department of Immunology, Rikshospitalet-Radiumhospitalet Medical Centre, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - D. Bay
- Department of Radiology, Oslo Vascular Centre, Aker University Hospital
| | - D. Kollerøs
- Department of Anaesthesiology, Aker University Hospital
| | - A. J. Kroese
- Department of Vascular Surgery, Aker University Hospital
| | - J. E. Brinchmann
- Department of Immunology, Rikshospitalet-Radiumhospitalet Medical Centre, Faculty of Medicine, University of Oslo, Oslo, Norway
| |
Collapse
|
96
|
Chang CW, Christensen LV, Lee M, Kim SW. Efficient expression of vascular endothelial growth factor using minicircle DNA for angiogenic gene therapy. J Control Release 2008; 125:155-63. [PMID: 18063165 PMCID: PMC2677388 DOI: 10.1016/j.jconrel.2007.10.014] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2007] [Revised: 10/05/2007] [Accepted: 10/17/2007] [Indexed: 10/22/2022]
Abstract
The application of plasmid DNA (pDNA)-based gene therapy is limited by its inefficient transgene expression. In this study, minicircle DNA was evaluated for efficient vascular endothelial growth factor (VEGF) expression in skeletal muscle cells. Production of minicircle DNA encoding VEGF was studied by a semi-quantitative electrophoresis method leading to optimized bacterial culture conditions and producing high purity (86.6%) minicircle DNA. The VEGF minicircle DNA under control of the SV40 promoter (pMini-SV-VEGF) showed an increased amount of VEGF mRNA and up to 8 times more VEGF expression than a conventional plasmid (pSV-VEGF) in two different skeletal muscle cell lines (C2C12 and L8). Minicircle DNA with different promoters, including the SV40, CMV and chicken beta-actin, was tested for VEGF expression in a C2C12 skeletal muscle cell line. The high VEGF expression generated by minicircle DNA stimulated efficient endothelial cell growth in vitro. Furthermore, minicircle DNA expressed higher VEGF compared to conventional plasmid in the tibialis anterior (TA) muscle of mice. Taken together, the results suggest that minicircle DNA is an efficacious gene vector for angiogenic VEGF expression in skeletal muscle and may be useful for treating peripheral arterial disease (PAD).
Collapse
Affiliation(s)
- Chien-Wen Chang
- Department of Pharmaceutics and Pharmaceutical Chemistry, University of Utah, Salt Lake City, UT 84112-5820, USA
| | - Lane V. Christensen
- Department of Pharmaceutics and Pharmaceutical Chemistry, University of Utah, Salt Lake City, UT 84112-5820, USA
| | - Minhyung Lee
- Department of Bioengineering, College of Engineering, Hanyang University, Seoul, Korea
| | - Sung Wan Kim
- Department of Pharmaceutics and Pharmaceutical Chemistry, University of Utah, Salt Lake City, UT 84112-5820, USA
| |
Collapse
|
97
|
Moreschi D, Fagundes DJ, Hernandes L, Haapalainen EF. Effects of Prostaglandin E1 in the Genesis of Blood Capillaries in the Ischemic Skeletal Muscle of Rats: Ultrastructural Analysis. Ann Vasc Surg 2008; 22:121-6. [DOI: 10.1016/j.avsg.2007.07.035] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2007] [Revised: 06/11/2007] [Accepted: 07/16/2007] [Indexed: 10/21/2022]
|
98
|
Moreschi Jr. D, Fagundes DJ, Amado LEB, Hernandes L, Moreschi HK. Efeitos da prostaglandina E1 (PGE1) na gênese de capilares sanguíneos em músculo esquelético isquêmico de ratos: estudo histológico. J Vasc Bras 2007. [DOI: 10.1590/s1677-54492007000400004] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
CONTEXTO: A angiogênese terapêutica é uma modalidade de tratamento para pacientes com insuficiência arterial crônica que não têm indicação para revascularização direta ou angioplastia e que não tiveram uma resposta satisfatória ao tratamento clínico. Entre as drogas utilizadas para essa finalidade está a prostaglandina E1 (PGE1). OBJETIVO: Estudar os aspectos morfológicos na gênese de capilares sanguíneos em músculo esquelético do membro caudal de ratos submetidos à isquemia sob a ação da PGE1, administrada por via intramuscular (IM) ou endovenosa (EV). MÉTODOS: Foram utilizados 48 ratos, linhagem Wistar-UEM, distribuídos aleatoriamente em três grupos de 16, redistribuídos igualmente em dois subgrupos, observados no 7º e 14º dias, sendo um grupo controle onde apenas foi provocada a isquemia no membro, outro com a isquemia e a injeção da PGE1 via IM e outro com a isquemia e a injeção da PGE1 EV. Para análise dos resultados, foram realizadas a coloração com hematoxilina e eosina (HE) e coloração imuno-histoquímica. RESULTADOS: Constatou-se um aumento estatisticamente significativo no número de capilares nos subgrupos com o uso da PGE1 IM e EV, através da contagem nos cortes corados com HE. A imunomarcação não foi eficiente para a quantificação dos capilares. CONCLUSÕES: A PGE1, administrada por via IM ou EV, promoveu, após 14 dias de observação, um aumento no número de capilares no músculo esquelético de ratos submetido à isquemia, identificáveis histologicamente com a coloração em HE. A imunocoloração não permitiu estabelecer uma correlação com o aumento de vasos encontrados na coloração com HE.
Collapse
|
99
|
Takahashi K. [Basic research and drug development for arteriosclerosis obliterans]. Nihon Yakurigaku Zasshi 2007; 130:393-397. [PMID: 18000354 DOI: 10.1254/fpj.130.393] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/25/2023]
|
100
|
Saito Y, Nakagami H, Kurooka M, Takami Y, Kikuchi Y, Hayashi H, Nishikawa T, Tamai K, Morishita R, Azuma N, Sasajima T, Kaneda Y. Cold shock domain protein A represses angiogenesis and lymphangiogenesis via inhibition of serum response element. Oncogene 2007; 27:1821-33. [DOI: 10.1038/sj.onc.1210824] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
|