Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 37
Filter
1.
Int J Oral Sci ; 14(1): 23, 2022 04 27.
Article in English | MEDLINE | ID: mdl-35477924

ABSTRACT

The significant clinical feature of bisphosphonate-related osteonecrosis of the jaw (BRONJ) is the exposure of the necrotic jaw. Other clinical manifestations include jaw pain, swelling, abscess, and skin fistula, which seriously affect the patients' life, and there is no radical cure. Thus, new methods need to be found to prevent the occurrence of BRONJ. Here, a novel nanoparticle, tFNA-KLT, was successfully synthesized by us, in which the nanoparticle tetrahedral framework nucleic acid (tFNA) was used for carrying angiogenic peptide, KLT, and then further enhanced angiogenesis. TFNA-KLT possessed the same characteristics as tFNA, such as simple synthesis, stable structure, and good biocompatibility. Meanwhile, tFNA enhanced the stability of KLT and carried more KLT to interact with endothelial cells. First, it was confirmed that tFNA-KLT had the superior angiogenic ability to tFNA and KLT both in vitro and in vivo. Then we apply tFNA-KLT to the prevention of BRONJ. The results showed that tFNA-KLT can effectively prevent the occurrence of BRONJ by accelerating angiogenesis. In summary, the prepared novel nanoparticle, tFNA-KLT, was firstly synthesized by us. It was also firstly confirmed by us that tFNA-KLT significantly enhanced angiogenesis and can effectively prevent the occurrence of BRONJ by accelerating angiogenesis, thus providing a new avenue for the prevention of BRONJ and a new choice for therapeutic angiogenesis.


Subject(s)
Bisphosphonate-Associated Osteonecrosis of the Jaw , Nanoparticles , Nucleic Acids , Angiogenic Proteins/therapeutic use , Bisphosphonate-Associated Osteonecrosis of the Jaw/drug therapy , Bisphosphonate-Associated Osteonecrosis of the Jaw/prevention & control , Endothelial Cells , Humans , Nucleic Acids/therapeutic use
2.
Article in English | WPRIM (Western Pacific) | ID: wpr-929151

ABSTRACT

The significant clinical feature of bisphosphonate-related osteonecrosis of the jaw (BRONJ) is the exposure of the necrotic jaw. Other clinical manifestations include jaw pain, swelling, abscess, and skin fistula, which seriously affect the patients' life, and there is no radical cure. Thus, new methods need to be found to prevent the occurrence of BRONJ. Here, a novel nanoparticle, tFNA-KLT, was successfully synthesized by us, in which the nanoparticle tetrahedral framework nucleic acid (tFNA) was used for carrying angiogenic peptide, KLT, and then further enhanced angiogenesis. TFNA-KLT possessed the same characteristics as tFNA, such as simple synthesis, stable structure, and good biocompatibility. Meanwhile, tFNA enhanced the stability of KLT and carried more KLT to interact with endothelial cells. First, it was confirmed that tFNA-KLT had the superior angiogenic ability to tFNA and KLT both in vitro and in vivo. Then we apply tFNA-KLT to the prevention of BRONJ. The results showed that tFNA-KLT can effectively prevent the occurrence of BRONJ by accelerating angiogenesis. In summary, the prepared novel nanoparticle, tFNA-KLT, was firstly synthesized by us. It was also firstly confirmed by us that tFNA-KLT significantly enhanced angiogenesis and can effectively prevent the occurrence of BRONJ by accelerating angiogenesis, thus providing a new avenue for the prevention of BRONJ and a new choice for therapeutic angiogenesis.


Subject(s)
Humans , Angiogenic Proteins/therapeutic use , Bisphosphonate-Associated Osteonecrosis of the Jaw/prevention & control , Endothelial Cells , Nanoparticles , Nucleic Acids/therapeutic use
4.
J Biomed Mater Res A ; 108(9): 1881-1889, 2020 09.
Article in English | MEDLINE | ID: mdl-32314537

ABSTRACT

Restoring blood supply is an effective way for the therapy of myocardial infarction (MI). It was reported a specific angiogenic peptide (VMP) derived from vascular endothelial growth factor (VEGF) could activate its receptor to mimic the biological activity of VEGF. In this study, in order to improve the local concentration in infarction region, a collagen-binding domain was synthesized with VMP to construct collagen binding domain (CBD)-VMP peptides. The fused CBD-VMP could bind specifically to collagen which was rich in cardiac extracellular matrix (c-ECM), without impacting the biological activity of VMP peptides. When the CBD-VMP peptides loaded on collagen scaffold and implanted into the rats subcutaneously, significant vascularization was observed. Then, CBD-VMP peptides binding with injectable c-ECM injected into the MI rat by intramuscular administration, significant blood vessels regeneration, and decrease of cell apoptosis were observed, that corelated with the recovery of cardiac function. It might be an alternative promising strategy for the clinical application of MI.


Subject(s)
Angiogenic Proteins/therapeutic use , Collagen/therapeutic use , Myocardial Infarction/drug therapy , Angiogenic Proteins/administration & dosage , Animals , Collagen/administration & dosage , Male , Myocardial Infarction/pathology , Neovascularization, Physiologic/drug effects , Rats , Rats, Sprague-Dawley
5.
Toxicon ; 165: 78-82, 2019 Jul.
Article in English | MEDLINE | ID: mdl-31054289

ABSTRACT

The cutaneous wound healing property of a pro-angiogenic venom peptide (RVVAP) in a cream-based formulation was evaluated using the excision wound healing model on Wistar strain rats. The wound healing potency and modest antibacterial activity of RVVAP was enhanced significantly (p < 0.05) when combined with Aloe vera extract. RVVAP was also found to be non-toxic at the tested dose of 1.0 mg/kg. Nevertheless, the release of inflammatory cytokines such as IL-1, IL-6, IL-10, and TNF-α in RVVAP-treated mice was suppressed, compared to the untreated controls. This is the first report assessing the wound healing potential of a low-molecular mass, non-enzymatic, pro-angiogenic peptide purified from snake venom.


Subject(s)
Angiogenic Proteins/therapeutic use , Daboia , Viper Venoms/chemistry , Wound Healing/drug effects , Angiogenic Proteins/isolation & purification , Animals , Rats , Rats, Wistar
6.
Molecules ; 24(7)2019 Mar 28.
Article in English | MEDLINE | ID: mdl-30925755

ABSTRACT

We describe progress and obstacles in the development of novel peptide-hydrogel therapeutics for unmet medical needs in ischemia treatment, focusing on the development and translation of therapies specifically in peripheral artery disease (PAD). Ischemia is a potentially life-threatening complication in PAD, which affects a significant percentage of the elderly population. While studies on inducing angiogenesis to treat PAD were started two decades ago, early results from animal models as well as clinical trials have not yet been translated into clinical practice. We examine some of the challenges encountered during such translation. We further note the need for sustained angiogenic effect involving whole growth factor, gene therapy and synthetic growth factor strategies. Finally, we discuss the need for tissue depots for de novo formation of microvasculature. These scaffolds can act as templates for neovasculature development to improve circulation and healing at the preferred anatomical location.


Subject(s)
Angiogenic Proteins/therapeutic use , Ischemia/drug therapy , Translational Research, Biomedical , Animals , Disease Models, Animal , Drug Discovery , Humans , Neovascularization, Physiologic/drug effects
7.
Arch Biochem Biophys ; 660: 72-86, 2018 12 15.
Article in English | MEDLINE | ID: mdl-30336134

ABSTRACT

Pro-angiogenic therapy provides a promising new perspective in tackling of many common and severe pathological conditions, such as central and peripheral vascular diseases. Pro-angiogenic therapy also finds interesting applications in the regenerative medicine for the treatment of chronic wounds and in tissue engineering. However, clinical studies on therapeutic angiogenesis, mainly performed by administrating growth factors, have not led to convincing results until now, mainly due to the unfavorable pharmacokinetic and to safety concerns. Thus, the research of new pro-angiogenic molecules endowed of improved pharmacological profile is strongly encouraged. This review focuses on Vascular Endothelial Growth Factor (VEGF) mimetic peptides exerting a pro-angiogenic activity, which are considered among the most promising alternatives to the VEGF based therapy. Peptides show a great potential in drug discovery, as they feature straightforward development approaches, robust and cheap synthetic methodologies for their preparation and functionalization, improved safety and efficacy profiles. Thus, pro-angiogenic peptides represent a valuable alternative to traditional drugs for biomedical applications in cardiovascular diseases and regenerative medicine.


Subject(s)
Angiogenic Proteins/therapeutic use , Biomimetic Materials/therapeutic use , Angiogenic Proteins/chemistry , Animals , Biomimetic Materials/chemistry , Disease , Humans , Signal Transduction , Vascular Endothelial Growth Factor A/metabolism
8.
J Neuroinflammation ; 15(1): 178, 2018 Jun 09.
Article in English | MEDLINE | ID: mdl-29885663

ABSTRACT

BACKGROUND: Neuroinflammation and blood-brain barrier (BBB) disruption are two critical mechanisms of subarachnoid hemorrhage (SAH)-induced brain injury, which are closely related to patient prognosis. Recently, angiogenic factor with G-patch and FHA domain 1 (Aggf1) was shown to inhibit inflammatory effect and preserve vascular integrity in non-nervous system diseases. This study aimed to determine whether Aggf1 could attenuate neuroinflammation and preserve BBB integrity after experimental SAH, as well as the underlying mechanisms of its protective roles. METHODS: Two hundred forty-nine male Sprague-Dawley rats were subjected to the endovascular perforation model of SAH. Recombinant human Aggf1 (rh-Aggf1) was administered intravenously via tail vein injection at 1 h after SAH induction. To investigate the underlying neuroprotection mechanism, Aggf1 small interfering RNA (Aggf1 siRNA) and PI3K-specific inhibitor LY294002 were administered through intracerebroventricular (i.c.v.) before SAH induction. SAH grade, neurological score, brain water content, BBB permeability, Western blot, and immunohistochemistry were performed. RESULTS: Expression of endogenous Aggf1 was markedly increased after SAH. Aggf1 was primarily expressed in endothelial cells and astrocytes, as well as microglia after SAH. Administration of rh-Aggf1 significantly reduced brain water content and BBB permeability, decreased the numbers of infiltrating neutrophils, and activated microglia in the ipsilateral cerebral cortex following SAH. Furthermore, rh-Aggf1 treatment improved both short- and long-term neurological functions after SAH. Meanwhile, exogenous rh-Aggf1 significantly increased the expression of PI3K, p-Akt, VE-cadherin, Occludin, and Claudin-5, as well as decreased the expression of p-NF-κB p65, albumin, myeloperoxidase (MPO), TNF-α, and IL-1ß. Conversely, knockdown of endogenous Aggf1 aggravated BBB breakdown, inflammatory response and neurological impairments at 24 h after SAH. Additionally, the protective roles of rh-Aggf1 were abolished by LY294002. CONCLUSIONS: Taken together, exogenous Aggf1 treatment attenuated neuroinflammation and BBB disruption, improved neurological deficits after SAH in rats, at least in part through the PI3K/Akt/NF-κB pathway.


Subject(s)
Angiogenic Proteins/therapeutic use , Blood-Brain Barrier/drug effects , Encephalitis/drug therapy , Encephalitis/etiology , Signal Transduction/drug effects , Subarachnoid Hemorrhage/complications , Angiogenic Proteins/genetics , Animals , Blood-Brain Barrier/physiology , Brain Edema/etiology , Capillary Permeability/drug effects , Chromones/therapeutic use , Disease Models, Animal , Enzyme Inhibitors/therapeutic use , Humans , Injections, Intraventricular , Male , Maze Learning/drug effects , Morpholines/therapeutic use , Nerve Tissue Proteins/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , RNA, Small Interfering/physiology , Rats , Rats, Sprague-Dawley , Subarachnoid Hemorrhage/mortality , Subarachnoid Hemorrhage/pathology
9.
J Cardiovasc Pharmacol Ther ; 23(2): 130-141, 2018 03.
Article in English | MEDLINE | ID: mdl-29025278

ABSTRACT

Although there have been advances in coronary artery bypass grafting and percutaneous coronary intervention, some patients who have ischemic coronary artery disease (CAD) are ineligible for revascularization due to suboptimal anatomy. Cardiac angiogenesis is not only a physiological response to ischemia or hypoxia but also a potential target of therapeutic strategies. Preclinical studies have shown a great enthusiasm on therapeutic angiogenesis for ischemic CAD. However, the latest trials provided the limited evidence on its efficacy. This article aims to discuss the physiological process of angiogenesis, the characteristic of angiogenic growth factors, delivery system, and clinical and preclinical studies, which can provide a novel insight into the therapeutic angiogenesis for CAD.


Subject(s)
Angiogenesis Inducing Agents/therapeutic use , Angiogenic Proteins/therapeutic use , Coronary Artery Disease/therapy , Coronary Vessels/drug effects , Neovascularization, Physiologic/drug effects , Angiogenesis Inducing Agents/adverse effects , Angiogenic Proteins/biosynthesis , Angiogenic Proteins/genetics , Animals , Coronary Artery Disease/genetics , Coronary Artery Disease/metabolism , Coronary Artery Disease/physiopathology , Coronary Vessels/physiopathology , Genetic Therapy/adverse effects , Genetic Therapy/methods , Humans , Neovascularization, Physiologic/genetics , Treatment Outcome
10.
Geriatr Gerontol Int ; 17(11): 2150-2156, 2017 Nov.
Article in English | MEDLINE | ID: mdl-28488306

ABSTRACT

AIM: In patients with diabetes or ischemia, angiogenesis and infection control are required for chronic leg ulcers, which substantially impair patients' quality of life. We developed a novel functional peptide, named AG30/5C, with angiogenic and anti-microbial properties. Treatment with AG30/5C significantly accelerated the wound healing of full-thickness defects in mice. To evaluate the safety of AG30/5C in the treatment of leg ulcers, a physician-initiated clinical study was carried out. METHODS: The first-in-human trial was designed as an open-label treatment with AG30/5C (0.1 mg/mL) given twice per day for 11 days, and with a follow-up period of 17 days. The inclusion criteria for severe skin ulcers were: (i) diabetes or critical limb ischemia; (ii) resistance to standard therapy for 1 month; and (iii) detection of methicillin-resistant Staphylococcus aureus in the skin ulcer. RESULTS: Four patients were enrolled in this study, and two patients met these criteria. For the evaluation of safety, three adverse effects were reported as possibly related to AG30/5C treatment; however, these adverse effects were not severe and resolved during or after treatment. Thus, there were no safety concerns. In both patients, the size of the ulcer decreased after treatment (44.62% and 10.23% decrease), and further decreased after the follow-up period (73.85% and 10.23% decrease). The former patient was diagnosed as Werner syndrome and the skin ulcer was resistant to standard therapy; however, it was sensitive to AG30/5C treatment. CONCLUSIONS: Topical treatment with AG30/5C for severe leg ulcers was safe, well tolerated and effective. Geriatr Gerontol Int 2017; 17: 2150-2156.


Subject(s)
Angiogenic Proteins/therapeutic use , Leg Ulcer/therapy , Peptides/therapeutic use , Angiogenic Proteins/adverse effects , Humans , Peptides/adverse effects , Physicians , Severity of Illness Index , Treatment Outcome
11.
Nucl Med Biol ; 43(9): 552-558, 2016 09.
Article in English | MEDLINE | ID: mdl-27395415

ABSTRACT

INTRODUCTION: Although liposomes hold promise for cancer therapy, the effectiveness of treating myocardial ischemia by promoting angiogenesis has yet to be proved. Nanoliposomes loaded with therapeutic agents can effectively target ischemic myocardium via enhanced permeability and retention. Surface polyethylene glycol (PEG) modification can further facilitate effective targeting by prolonging liposomal circulation. This study aimed to determine whether PEGylated nanoliposomes are effective in facilitating targeted drug delivery and treating myocardial ischemia. METHODS: Rats subjected to 30min of myocardial ischemia were given (99m)Tc-hexamethylpropyleneamine oxime- or (99m)Tc-diethylenetriamine pentaacetate-labeled liposomes with mean diameters of ~100nm or ~600nm with or without PEG modifications to determine the extent of myocardial uptake in the different conditions. Therapeutic effectiveness was assessed by studying changes in myocardial perfusion defects with (99m)Tc-tetrofosmin autoradiography and vascular density with immunohistochemistry at 7days post-treatment. RESULTS: The liver and spleen showed the largest capacity for liposome uptake. Uptake by the liver and spleen was more pronounced when the liposomes were larger. Conversely, myocardial liposome uptake was significantly greater when the liposomes were ~100nm rather than ~600nm in diameter. Surface modification with PEG significantly augmented myocardial uptake of ~100nm liposomes. PEG modification did not affect the size dependence. To investigate therapeutic efficacy, hearts subjected to ischemia received PEGylated nanoliposomes encapsulated with angiogenic peptides. Our data demonstrated that PEGylated nanoliposomes loaded with angiogenic peptides improved myocardial perfusion defects and increased vascular density. A 10-fold increase in liposomal concentration did not further benefit myocardial ischemia. CONCLUSIONS: Liposomal angiogenic formulation with size control and PEG modification may be effective treatment strategy for myocardial ischemia. Increasing the concentration of liposomes does not necessarily benefit myocardial ischemia.


Subject(s)
Angiogenic Proteins/administration & dosage , Angiogenic Proteins/pharmacology , Coronary Circulation/drug effects , Myocardial Ischemia/diagnostic imaging , Myocardial Ischemia/physiopathology , Polyethylene Glycols/chemistry , Angiogenic Proteins/therapeutic use , Animals , Capsules , Dose-Response Relationship, Drug , Liposomes , Male , Myocardial Ischemia/drug therapy , Myocardial Ischemia/metabolism , Neovascularization, Physiologic/drug effects , Organophosphorus Compounds/metabolism , Organophosphorus Compounds/pharmacokinetics , Organotechnetium Compounds/metabolism , Organotechnetium Compounds/pharmacokinetics , Radionuclide Imaging , Rats , Rats, Sprague-Dawley , Tissue Distribution
12.
J Endod ; 40(11): 1784-90, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25155266

ABSTRACT

INTRODUCTION: Prostacyclin (PGI2), a member of the prostaglandin family, can promote angiogenesis and cell proliferation. METHODS: In this study, the effect of the application of a PGI2 analog (iloprost) on dentin repair was examined in vitro and in vivo. RESULTS: Iloprost significantly stimulated the expression of vascular endothelial growth factor and osteo-/odontogenic marker messenger RNA in human dental pulp cells (HDPCs) under osteoinductive conditions in vitro. In addition, iloprost enhanced HDPC alkaline phosphatase enzymatic activity and mineral deposition. An in vivo study was performed using a rat molar mechanical pulp exposure model. After 30 days, histologic analysis revealed that there was a dramatic tertiary dentin formation in the iloprost-treated group compared with the calcium hydroxide and the untreated control groups. Furthermore, vascular endothelial growth factor protein expression in dental pulp tissue was increased in the iloprost-treated group as determined by immunohistochemical staining. CONCLUSIONS: Taken together, the present study, for the first time, shows that iloprost induces the expression of osteo-/odontogenic markers in vitro and promotes angiogenic factor expression and enhances tertiary dentin formation in vivo. This implies the potential clinical usefulness of iloprost in vital pulp therapy.


Subject(s)
Dentin, Secondary/drug effects , Iloprost/pharmacology , Adult , Alkaline Phosphatase/drug effects , Angiogenic Proteins/pharmacology , Angiogenic Proteins/therapeutic use , Animals , Bone Morphogenetic Protein 2/drug effects , Bone Morphogenetic Protein 4/drug effects , Calcification, Physiologic/drug effects , Calcium Hydroxide/therapeutic use , Cells, Cultured , Core Binding Factor Alpha 1 Subunit/drug effects , Dental Pulp/cytology , Dental Pulp/drug effects , Dental Pulp/injuries , Dental Pulp Exposure/drug therapy , Disease Models, Animal , Humans , Iloprost/therapeutic use , Male , Odontogenesis/drug effects , Osteogenesis/drug effects , Rats , Rats, Wistar , Transcription Factors/drug effects , Vascular Endothelial Growth Factor A/drug effects
13.
Circulation ; 124(11 Suppl): S18-26, 2011 Sep 13.
Article in English | MEDLINE | ID: mdl-21911811

ABSTRACT

BACKGROUND: Experimentally, exogenous administration of recombinant stromal cell-derived factor-1α (SDF) enhances neovasculogenesis and cardiac function after myocardial infarction. Smaller analogs of SDF may provide translational advantages including enhanced stability and function, ease of synthesis, lower cost, and potential modulated delivery via engineered biomaterials. In this study, computational protein design was used to create a more efficient evolution of the native SDF protein. METHODS AND RESULTS: Protein structure modeling was used to engineer an SDF polypeptide analog (engineered SDF analog [ESA]) that splices the N-terminus (activation and binding) and C-terminus (extracellular stabilization) with a diproline segment designed to limit the conformational flexibility of the peptide backbone and retain the relative orientation of these segments observed in the native structure of SDF. Endothelial progenitor cells (EPCs) in ESA gradient, assayed by Boyden chamber, showed significantly increased migration compared with both SDF and control gradients. EPC receptor activation was evaluated by quantification of phosphorylated AKT, and cells treated with ESA yielded significantly greater phosphorylated AKT levels than SDF and control cells. Angiogenic growth factor assays revealed a distinct increase in angiopoietin-1 expression in the ESA- and SDF-treated hearts. In addition, CD-1 mice (n=30) underwent ligation of the left anterior descending coronary artery and peri-infarct intramyocardial injection of ESA, SDF-1α, or saline. At 2 weeks, echocardiography demonstrated a significant gain in ejection fraction, cardiac output, stroke volume, and fractional area change in mice treated with ESA compared with controls. CONCLUSIONS: Compared with native SDF, a novel engineered SDF polypeptide analog (ESA) more efficiently induces EPC migration and improves post-myocardial infarction cardiac function and thus offers a more clinically translatable neovasculogenic therapy.


Subject(s)
Angiogenic Proteins/chemistry , Angiogenic Proteins/pharmacology , Chemokine CXCL12/chemistry , Chemokine CXCL12/pharmacology , Computational Biology/methods , Neovascularization, Physiologic/drug effects , Protein Engineering/methods , Angiogenic Proteins/therapeutic use , Animals , Cardiac Output/drug effects , Cardiac Output/physiology , Cell Movement/drug effects , Cell Movement/physiology , Cells, Cultured , Chemokine CXCL12/therapeutic use , Coronary Vessels/drug effects , Coronary Vessels/physiology , Endothelial Cells/cytology , Endothelial Cells/drug effects , Male , Mice , Mice, Inbred Strains , Models, Animal , Myocardial Infarction/physiopathology , Myocardial Infarction/prevention & control , Neovascularization, Physiologic/physiology , Rats , Rats, Wistar , Stem Cells/cytology , Stem Cells/drug effects , Stroke Volume/drug effects , Stroke Volume/physiology
14.
Proc Natl Acad Sci U S A ; 108(33): 13438-43, 2011 Aug 16.
Article in English | MEDLINE | ID: mdl-21808036

ABSTRACT

There is great demand for the development of novel therapies for ischemic cardiovascular disease, a leading cause of morbidity and mortality worldwide. We report here on the development of a completely synthetic cell-free therapy based on peptide amphiphile nanostructures designed to mimic the activity of VEGF, one of the most potent angiogenic signaling proteins. Following self-assembly of peptide amphiphiles, nanoscale filaments form that display on their surfaces a VEGF-mimetic peptide at high density. The VEGF-mimetic filaments were found to induce phosphorylation of VEGF receptors and promote proangiogenic behavior in endothelial cells, indicated by an enhancement in proliferation, survival, and migration in vitro. In a chicken embryo assay, these nanostructures elicited an angiogenic response in the host vasculature. When evaluated in a mouse hind-limb ischemia model, the nanofibers increased tissue perfusion, functional recovery, limb salvage, and treadmill endurance compared to controls, which included the VEGF-mimetic peptide alone. Immunohistological evidence also demonstrated an increase in the density of microcirculation in the ischemic hind limb, suggesting the mechanism of efficacy of this promising potential therapy is linked to the enhanced microcirculatory angiogenesis that results from treatment with these polyvalent VEGF-mimetic nanofibers.


Subject(s)
Angiogenic Proteins/therapeutic use , Ischemia/drug therapy , Nanostructures/chemistry , Vascular Endothelial Growth Factor A/physiology , Wound Healing/drug effects , Angiogenic Proteins/chemistry , Animals , Cell Line , Chick Embryo , Endothelium, Vascular , Humans , Mice , Molecular Mimicry , Nanostructures/therapeutic use , Neovascularization, Physiologic/drug effects
15.
Neurosurgery ; 68(5): 1372-9; discussion 1379, 2011 May.
Article in English | MEDLINE | ID: mdl-21273924

ABSTRACT

BACKGROUND: Granulocyte colony-stimulating factor (G-CSF) mobilizes hematopoietic bone marrow cells into systemic circulation and has been used clinically to treat chemotherapy-induced neutropenia. Recently, G-CSF was shown to have neuroprotective and angiogenetic effects in acute cerebral infarction. OBJECTIVE: To evaluate the effects of G-CSF for angiogenesis after indirect bypass surgery. METHODS: : Chronic cerebral hypoperfusions were induced in male Wistar rats by permanent bilateral internal carotid artery occlusion (BICAO). After BICAO, unilateral indirect bypass and encephalogaleosynangiosis (EGS) were performed, and human recombinant G-CSF (10 µg/kg) or saline was injected intramuscularly for 5 consecutive days. We measured regional cerebral blood flow (rCBF) by laser Doppler flowmetry and performed immunohistochemical analysis 21 days after BICAO. RESULTS: BICAO decreased rCBF to 62.52% ± 5.8% of control (P < .01). The rCBF increased significantly 21 days after BICAO in all treatment groups (n = 10; P < .05) except the G-E- group. The rCBF increase observed in the G+E+ group was significantly higher than that observed in other groups. Both G-CSF and EGS treatments significantly increased the number of small vessels (P < .01), and G-CSF and EGS showed additive effect in increasing the number of small vessels. CONCLUSION: Combined use of G-CSF and indirect bypass surgery induces an increase in rCBF and angiogenesis under conditions of cerebral chronic hypoperfusion. This is the first report to demonstrate that G-CSF can enhance angiogenesis induced by indirect bypass surgery, and that this combined therapy is a safe and easy method of treatment.


Subject(s)
Angiogenic Proteins/pharmacology , Cerebral Infarction/drug therapy , Cerebral Infarction/surgery , Cerebral Revascularization/methods , Disease Models, Animal , Granulocyte Colony-Stimulating Factor/pharmacology , Angiogenic Proteins/therapeutic use , Animals , Cerebral Infarction/physiopathology , Combined Modality Therapy , Filgrastim , Granulocyte Colony-Stimulating Factor/therapeutic use , Humans , Male , Neovascularization, Physiologic/drug effects , Neovascularization, Physiologic/physiology , Random Allocation , Rats , Rats, Wistar , Recombinant Proteins/pharmacology , Recombinant Proteins/therapeutic use
16.
Heart ; 97(3): 181-9, 2011 Feb.
Article in English | MEDLINE | ID: mdl-20884790

ABSTRACT

Since the discovery of vascular endothelial growth factor (VEGF), therapeutic angiogenesis has attracted interest as an alternative treatment for ischaemic heart and peripheral disease. In parallel, the view has also gained ground that angiogenesis has an important role in the pathogenesis of atherosclerotic disease and its clinical sequelae. These conflicting perspectives have both been based on a large quantity of preclinical data obtained from mainly small rodent models of disease. However, in recent years further research and the results of clinical trials of pro-angiogenic and anti-angiogenic treatments have provided new insights into the impact of VEGF and other angiogenesis-based approaches on human health and disease. This review discusses therapeutic angiogenesis in the light of recent scientific advances and clinical evidence, and considers the future challenges and prospects for therapeutic angiogenesis.


Subject(s)
Cardiovascular Diseases/therapy , Neovascularization, Physiologic/physiology , Angiogenic Proteins/therapeutic use , Cardiovascular Diseases/physiopathology , Genetic Therapy/methods , Humans , Neovascularization, Physiologic/drug effects , Randomized Controlled Trials as Topic , Vascular Endothelial Growth Factor A/physiology , Vascular Endothelial Growth Factor A/therapeutic use
17.
J Cardiovasc Transl Res ; 3(5): 469-77, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20607468

ABSTRACT

Although most medicines have historically been small molecules, many newly approved drugs are derived from proteins. Protein therapies have been developed for treatment of diseases in almost every organ system, including the heart. Great excitement has now arisen in the field of regenerative medicine, particularly for cardiac regeneration after myocardial infarction. Every year, millions of people suffer from acute myocardial infarction, but the adult mammalian myocardium has limited regeneration potential. Regeneration of the heart after myocardium infarction is therefore an exciting target for protein therapeutics. In this review, we discuss different classes of proteins that have therapeutic potential to regenerate the heart after myocardial infarction. Protein candidates have been described that induce angiogenesis, including fibroblast growth factors and vascular endothelial growth factors, although thus far clinical development has been disappointing. Chemotactic factors that attract stem cells, e.g., hepatocyte growth factor and stromal cell-derived factor-1, may also be useful. Finally, neuregulins and periostin are proteins that induce cell-cycle reentry of cardiomyocytes, and growth factors like IGF-1 can induce growth and differentiation of stem cells. As our knowledge of the biology of regenerative processes and the role of specific proteins in these processes increases, the use of proteins as regenerative drugs could develop as a cardiac therapy.


Subject(s)
Cardiovascular Agents/therapeutic use , Myocardial Infarction/drug therapy , Myocardium/pathology , Proteins/therapeutic use , Regeneration/drug effects , Regenerative Medicine , Angiogenic Proteins/therapeutic use , Animals , Cell Differentiation/drug effects , Cell Movement/drug effects , Cell Proliferation/drug effects , Humans , Myocardial Infarction/pathology , Myocardial Infarction/physiopathology , Neovascularization, Physiologic/drug effects , Stem Cells/drug effects , Stem Cells/pathology , Translational Research, Biomedical
19.
J Neuroimmune Pharmacol ; 2(3): 284-9, 2007 Sep.
Article in English | MEDLINE | ID: mdl-18040863

ABSTRACT

In the normal mature brain, blood vessel formation is tightly downregulated. However, pathologic processes such as ischemia can induce cerebral vascular regeneration. Angiogenesis is one of the major styles of new vessel formation. In this article, we summarize the major angiogenic factors in the brain, discuss the significant changes of angiogenic factors and endothelial progenitor cells (EPCs) in response to brain ischemia, and finally, review the therapeutic potential of angiogenic factors and EPCs in experimental cerebral ischemia based on the concept of neurovascular unit.


Subject(s)
Angiogenic Proteins/therapeutic use , Brain Ischemia/therapy , Neovascularization, Physiologic/physiology , Angiogenesis Inducing Agents/therapeutic use , Angiogenic Proteins/physiology , Animals , Brain Ischemia/physiopathology , Humans , Stem Cells/cytology , Stem Cells/physiology
20.
Curr Vasc Pharmacol ; 5(3): 195-209, 2007 Jul.
Article in English | MEDLINE | ID: mdl-17627563

ABSTRACT

Peripheral arterial disease (PAD) remains a major cause of morbidity. Despite advances in revascularisation procedures and medical treatment, limb salvage and relief of pain are still not satisfactory in patients with severe disease. This has prompted the exploration of alternative modes of treatment including enhancement of new vessel formation (angiogenesis). Angiogenic Growth Factors (AGF), mainly Vascular Endothelial Growth Factor (VEGF), basic Fibroblast Growth Factor (bFGF) and Hepatocyte Growth Factor (HGF) have emerged as exciting therapeutic modalities. Both experimental and clinical studies have demonstrated that topical (mainly intramuscular) AGF gene therapy results in improved peripheral vasculature and alleviation of symptoms. However, most clinical work is limited to small patient series and the long-term safety and efficacy are still unclear. Clinical benefit must be balanced against potential untoward effects, such as tumour growth and atherosclerotic plaque angiogenesis leading to plaque instability. VEGF is important in the pathogenesis of diabetic microvascular disease. Further studies are required before implementation of AGF therapy in clinical practice.


Subject(s)
Angiogenesis Inducing Agents/therapeutic use , Angiogenic Proteins/therapeutic use , Genetic Therapy , Neovascularization, Physiologic/drug effects , Peripheral Vascular Diseases/therapy , Angiogenesis Inducing Agents/metabolism , Angiogenesis Inducing Agents/pharmacology , Angiogenic Proteins/genetics , Angiogenic Proteins/metabolism , Angiogenic Proteins/pharmacology , Animals , Disease Models, Animal , Fibroblast Growth Factor 2/therapeutic use , Gene Transfer Techniques , Genetic Therapy/adverse effects , Hepatocyte Growth Factor/therapeutic use , Humans , Peripheral Vascular Diseases/drug therapy , Peripheral Vascular Diseases/genetics , Peripheral Vascular Diseases/metabolism , Peripheral Vascular Diseases/physiopathology , Recombinant Proteins/therapeutic use , Regional Blood Flow , Treatment Outcome , Vascular Endothelial Growth Factor A/therapeutic use
SELECTION OF CITATIONS
SEARCH DETAIL
...