Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 142
Filtrar
1.
Stem Cells Dev ; 30(21): 1070-1081, 2021 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-34514810

RESUMO

Studies have shown that human umbilical cord mesenchymal stem cells (hUCMSCs) could ameliorate liver fibrosis (LF) through inhibiting the activation of hepatic stellate cells (HSCs). However, the specific mechanisms have not been studied clearly. The purpose of this study was to explore the possible mechanism of hepatocyte growth factor (HGF)-transfected hUCMSCs in inhibiting the proliferation and activation of HSCs-T6. The upper and lower double-cell coculture system was established among HGF-hUCMSCs, LV5-NC-hUCMSCs, hUCMSCs, and HSCs-T6 in experimental groups; HSCs-T6 were cultured alone as control group. After coculturing for 1, 2, and 3 days, results showed that HGF-transfected hUCMSCs could decrease cell viability of HSCs-T6 and promote apoptosis; inhibit their activation and reduce the expression of Collagen I, Collagen III, TGF-ß1, Smad2 and Smad3, which may be related to inhibiting the activation of TGF-ß1/Smads signaling pathway. These findings suggested that HGF-transfected hUCMSCs may be used as an alternative and novel therapeutic approach for the treatment of LF.


Assuntos
Células Estreladas do Fígado , Fator de Crescimento de Hepatócito , Transplante de Células-Tronco Mesenquimais , Células-Tronco Mesenquimais , Proteínas Smad , Fator de Crescimento Transformador beta1 , Proliferação de Células , Células Estreladas do Fígado/metabolismo , Fator de Crescimento de Hepatócito/administração & dosagem , Fator de Crescimento de Hepatócito/metabolismo , Humanos , Cirrose Hepática/metabolismo , Cirrose Hepática/terapia , Transdução de Sinais , Proteínas Smad/metabolismo , Fator de Crescimento Transformador beta1/metabolismo , Cordão Umbilical
2.
Int J Mol Sci ; 22(9)2021 Apr 27.
Artigo em Inglês | MEDLINE | ID: mdl-33925510

RESUMO

BACKGROUND: Hepatocyte growth factor (HGF) is an endogenously induced bioactive molecule that has strong anti-apoptotic and tissue repair activities. In this research, we identified APOA4 as a novel pharmacodynamic (PD) marker of the recombinant human HGF (rh-HGF), E3112. METHODS: rh-HGF was administered to mice, and their livers were investigated for the PD marker. Candidates were identified from soluble proteins and validated by using human hepatocytes in vitro and an animal disease model in vivo, in which its c-Met dependency was also ensured. RESULTS: Among the genes induced or highly enhanced after rh-HGF exposure in vivo, a soluble apolipoprotein, Apoa4, was found to be induced by rh-HGF in the murine liver. By using primary cultured human hepatocytes, the significant induction of human APOA4 was observed at the mRNA and protein levels, and it was inhibited in the presence of a c-Met inhibitor. Although mice constitutively expressed Apoa4 mRNA in the small intestine and the liver, the liver was the primary organ affected by administered rh-HGF to strongly induce APOA4 in a dose- and c-Met-dependent manner. Serum APOA4 levels were increased after rh-HGF administration, not only in normal mice but also in anti-Fas-induced murine acute liver failure (ALF), which confirmed the pharmacodynamic nature of APOA4. CONCLUSIONS: APOA4 was identified as a soluble PD marker of rh-HGF with c-Met dependency. It should be worthwhile to clinically validate its utility through clinical trials with healthy subjects and ALF patients.


Assuntos
Apolipoproteínas A/sangue , Biomarcadores Farmacológicos/sangue , Fator de Crescimento de Hepatócito/farmacocinética , Hepatócitos/efeitos dos fármacos , Fígado/efeitos dos fármacos , Animais , Apolipoproteínas A/genética , Apolipoproteínas A/metabolismo , Células Cultivadas , Regulação da Expressão Gênica/efeitos dos fármacos , Fator de Crescimento de Hepatócito/administração & dosagem , Hepatócitos/metabolismo , Humanos , Fígado/fisiologia , Falência Hepática Aguda/sangue , Falência Hepática Aguda/etiologia , Masculino , Camundongos Endogâmicos BALB C , Proteínas Proto-Oncogênicas c-met/metabolismo , Proteínas Recombinantes/administração & dosagem , Proteínas Recombinantes/farmacocinética
3.
Life Sci ; 268: 119014, 2021 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-33412216

RESUMO

AIMS: Hepatocyte growth factor (HGF) is a multifunctional cytokine that plays important roles in pancreatic physiology. Approvals of gene therapy drugs have highlighted gene therapy as an innovative new drug modality, but the very recent reports of deaths in clinical trials have provided a warning that high-dose gene therapy can cause dangerous liver toxicity. The present study aimed to develop a safe and low-dose but therapeutically effective adenovirus-mediated HGF gene therapy for streptozotocin (STZ)-induced type 1 diabetes (T1D) in mice. MAIN METHODS: A single intravenous injection of a low dose (3 × 108 plaque forming units) of adenoviral vector expressing the HGF gene under the transcriptional control of a strong promoter, i.e., the cytomegalovirus immediate-early enhancer and a modified chicken ß-actin promoter (Ad.CA-HGF), was given to T1D mice. KEY FINDINGS: Low-dose HGF gene therapy significantly attenuated the elevation of blood glucose concentrations at the acute phase of T1D, and this effect persisted for several weeks. Temporal upregulation of plasma insulin at the acute phase was maintained at a normal level in Ad.CA-HGF-treated mice, suggesting that the therapeutic mechanism may involve protection of the remaining ß-cells by HGF. Liver enzymes in plasma were not elevated in any of the mice, including the Ad.CA-HGF-treated animals, all of which looked healthy, suggesting the absence of lethal adverse effects observed in patients receiving high intravenous doses of viral vectors. SIGNIFICANCE: A low dose of intravenous Ad-mediated HGF gene therapy is clinically feasible and safe, and thus represents a new therapeutic strategy for treating T1D.


Assuntos
Adenoviridae/genética , Diabetes Mellitus Tipo 1/terapia , Terapia Genética/métodos , Vetores Genéticos/administração & dosagem , Fator de Crescimento de Hepatócito/genética , Animais , Diabetes Mellitus Experimental/genética , Diabetes Mellitus Experimental/terapia , Diabetes Mellitus Tipo 1/genética , Vetores Genéticos/genética , Vetores Genéticos/farmacologia , Fator de Crescimento de Hepatócito/administração & dosagem , Hiperglicemia/genética , Hiperglicemia/terapia , Injeções Intravenosas , Insulina/sangue , Fígado/enzimologia , Masculino , Camundongos Endogâmicos C57BL , Regiões Promotoras Genéticas
4.
Mol Ther ; 27(12): 2158-2165, 2019 12 04.
Artigo em Inglês | MEDLINE | ID: mdl-31805256

RESUMO

NL003 is a plasmid engineered to simultaneously express two isoforms of hepatocyte growth factor. This phase II study was performed to assess the clinical safety and efficacy of intramuscular injection of NL003 in critical limb ischemia (CLI) patients for 6 months. Two hundred patients (Rutherford scale 4-5) were randomly assigned: placebo (n = 50), low-dose NL003 (n = 50), middle-dose NL003 (n = 50), or high-dose NL003 (n = 50). The drug was administered in the affected limb of 197 patients on days 0, 14, and 28. No significant differences in the incidence of adverse events (AEs) or serious AEs were found among the groups. At 6 months, pain severity was significantly reduced in all NL003 groups, but not in the placebo group (p < 0.05). The proportion of patients with complete ulcer healing in the high-dose group was significantly higher than that of the placebo group (p = 0.0095). There were no statistically significant differences in transcutaneous oxygen pressure (TcPO2), ankle-brachial index (ABI), or toe-brachial index (TBI) value among the four groups throughout the study period. These results provide the first effective evidence of significant improvements in total healing of ulcers in treated legs, complete pain relief without analgesics, and safety for NL003 in patients with Rutherford stage 4-5.


Assuntos
Terapia Genética/métodos , Fator de Crescimento de Hepatócito/administração & dosagem , Isquemia/terapia , Perna (Membro)/irrigação sanguínea , Plasmídeos/administração & dosagem , Método Duplo-Cego , Feminino , Seguimentos , Fator de Crescimento de Hepatócito/genética , Humanos , Injeções Intramusculares , Isquemia/genética , Isquemia/patologia , Masculino , Pessoa de Meia-Idade , Plasmídeos/genética , Prognóstico
5.
J Exp Clin Cancer Res ; 38(1): 362, 2019 Aug 19.
Artigo em Inglês | MEDLINE | ID: mdl-31426831

RESUMO

BACKGROUND: Tumor targeting small molecular inhibitors are the most popular treatments for many malignant diseases, including cancer. However, the lower clinical response and drug resistance still limit their clinical efficacies. HGFK1, the first kringle domain of hepatocyte growth factor, has been defined as a potent anti-angiogenic factor. Here, we aimed to develop and identify novel nanoparticles-PH1/pHGFK1 as potential therapeutic agents for the treatment of renal cell carcinoma (RCC). METHODS: We produced a novel cationic polymer-PH1 and investigated the anti-tumor activity of PH1/pHGFK1 nanoparticle alone and its combination therapy with sorafenib in RCC cell line xenografted mice model. Then, we figured out its molecular mechanisms in human RCC cell lines in vitro. RESULTS: We firstly demonstrated that intravenous injection of PH1/pHGFK1 nanoparticles significantly inhibited tumor growth and prolonged the survival time of tumor-bearing mice, as well as synergistically enhanced anti-tumor activities of sorafenib. Furthermore, we elucidated that recombinant HGFK1 improved sorafenib-induced cell apoptosis and arrested cell cycle. In addition, HGFK1 could also decrease sorafenib-induced autophagy and stemness via blockading NF-κB signaling pathway in RCC both in vitro and in vivo. CONCLUSIONS: HGFK1 could inhibit tumor growth, synergistically enhance anti-tumor activities of sorafenib and reverse its drug resistance evolution in RCC. Our results provide rational basis for clinical application of sorafenib and HGFK1 combination therapy in RCC patients.


Assuntos
Autofagia , Carcinoma de Células Renais/patologia , Sinergismo Farmacológico , Fator de Crescimento de Hepatócito/administração & dosagem , Nanopartículas/administração & dosagem , Células-Tronco Neoplásicas/patologia , Sorafenibe/farmacologia , Animais , Antineoplásicos/farmacologia , Apoptose , Carcinoma de Células Renais/metabolismo , Carcinoma de Células Renais/terapia , Ciclo Celular , Movimento Celular , Proliferação de Células , Feminino , Ácido Fólico/química , Humanos , Neoplasias Renais/metabolismo , Neoplasias Renais/patologia , Neoplasias Renais/terapia , Kringles , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Nanopartículas/química , Células-Tronco Neoplásicas/efeitos dos fármacos , Células-Tronco Neoplásicas/metabolismo , Polietilenoglicóis/química , Polietilenoimina/química , Células Tumorais Cultivadas , Ensaios Antitumorais Modelo de Xenoenxerto , beta-Ciclodextrinas/química
6.
Acta Neuropathol Commun ; 7(1): 96, 2019 06 12.
Artigo em Inglês | MEDLINE | ID: mdl-31189468

RESUMO

Amyotrophic lateral sclerosis (ALS) is a fatal neuromuscular disease resulting from motor neuron degeneration that causes muscle weakness, paralysis, and eventually respiratory failure. We investigated whether recombinant adeno-associated virus encoding human hepatocyte growth factor (rAAV-HGF) could generate beneficial effects in two mouse models with neuromuscular problems when intrathecally delivered to the subarachnoid space. We chose AAV serotype 1 (rAAV1) based on the expression levels and distribution of HGF protein in the lumbar spinal cord (LSC). After a single intrathecal (IT) injection of rAAV1-HGF, the protein level of HGF in the LSC peaked on day 14 and thereafter gradually decreased over the next 14 weeks. rAAV1-HGF was initially tested in the mouse nerve crush model. IT injection of rAAV1-HGF improved mouse hindlimb strength and rotarod performance, while histological analyses showed that the length of regenerated axons was increased and the structure of the neuromuscular junction (NMJ) was restored. rAAV1-HGF was also evaluated in the SOD1-G93A transgenic (TG) mouse model. Again, rAAV1-HGF not only improved motor performance but also increased the survival rate. Moreover, the number and diameter of spinal motor neurons (SMNs) were increased, and the shape of the NMJs restored. Data from in vitro motor cortical culture experiments indicated that treatment with recombinant HGF protein (rHGF) increased the axon length of corticospinal motor neurons (CSMNs). When cultures were treated with an ERK inhibitor, the effects of HGF on axon elongation, protein aggregation, and oxidative stress were suppressed, indicating that ERK phosphorylation played an important role(s). Taken together, our results suggested that HGF might play an important role(s) in delaying disease progression in the SOD1-G93A TG mouse model by reducing oxidative stress through the control of ERK phosphorylation.


Assuntos
Dependovirus/genética , Fator de Crescimento de Hepatócito/genética , Destreza Motora/fisiologia , Junção Neuromuscular/fisiologia , Neuropatia Ciática/genética , Superóxido Dismutase/genética , Animais , Células Cultivadas , Células HEK293 , Fator de Crescimento de Hepatócito/administração & dosagem , Humanos , Injeções Espinhais , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Destreza Motora/efeitos dos fármacos , Compressão Nervosa/métodos , Junção Neuromuscular/efeitos dos fármacos , Proteínas Recombinantes/administração & dosagem , Proteínas Recombinantes/genética , Neuropatia Ciática/tratamento farmacológico , Neuropatia Ciática/fisiopatologia
7.
Int J Mol Sci ; 20(5)2019 Feb 28.
Artigo em Inglês | MEDLINE | ID: mdl-30823442

RESUMO

Hepatocyte growth factor (HGF) was first identified as a potent mitogen for mature hepatocytes, and has also gained attention as a strong neurotrophic factor in the central nervous system. We found that during the acute phase of spinal cord injury (SCI) in rats, c-Met, the specific receptor for HGF, increases sharply, while the endogenous HGF up-regulation is relatively weak. Introducing exogenous HGF into the spinal cord by injecting an HGF-expressing viral vector significantly increased the neuron and oligodendrocyte survival, angiogenesis, and axonal regeneration, to reduce the area of damage and to promote functional recovery in rats after SCI. Other recent studies in rodents have shown that exogenously administered HGF during the acute phase of SCI reduces astrocyte activation to decrease glial scar formation, and exerts anti-inflammatory effects to reduce leukocyte infiltration. We also reported that the intrathecal infusion of recombinant human HGF (intrathecal rhHGF) improves neurological hand function after cervical contusive SCI in the common marmoset, a non-human primate. Based on these collective results, we conducted a phase I/II clinical trial of intrathecal rhHGF for patients with acute cervical SCI who showed a modified Frankel grade of A/B1/B2 72 h after injury onset, from June 2014 to May 2018.


Assuntos
Fator de Crescimento de Hepatócito/uso terapêutico , Traumatismos da Medula Espinal/tratamento farmacológico , Animais , Ensaios Clínicos Fase II como Assunto , Fator de Crescimento de Hepatócito/administração & dosagem , Fator de Crescimento de Hepatócito/efeitos adversos , Humanos
8.
Sci Rep ; 8(1): 13026, 2018 08 29.
Artigo em Inglês | MEDLINE | ID: mdl-30158635

RESUMO

Cystic fibrosis (CF), the most common inherited disease in Caucasians, is caused by mutations in the CFTR chloride channel, the most frequent of which is Phe508del. Phe508del causes not only intracellular retention and premature degradation of the mutant CFTR protein, but also defective channel gating and decreased half-life when experimentally rescued to the plasma membrane (PM). Despite recent successes in the functional rescue of several CFTR mutations with small-molecule drugs, the folding-corrector/gating-potentiator drug combinations approved for Phe508del-CFTR homozygous patients have shown only modest benefit. Several factors have been shown to contribute to this outcome, including an unexpected intensification of corrector-rescued Phe508del-CFTR PM instability after persistent co-treatment with potentiator drugs. We have previously shown that acute co-treatment with hepatocyte growth factor (HGF) can significantly enhance the chemical correction of Phe508del-CFTR. HGF coaxes the anchoring of rescued channels to the actin cytoskeleton via induction of RAC1 GTPase signalling. Here, we demonstrate that a prolonged, 15-day HGF treatment also significantly improves the functional rescue of Phe508del-CFTR by the VX-809 corrector/VX-770 potentiator combination, in polarized bronchial epithelial monolayers. Importantly, we found that HGF treatment also prevented VX-770-mediated destabilization of rescued Phe508del-CFTR and enabled further potentiation of the rescued channels. Most strikingly, prolonged HGF treatment prevented previously unrecognized epithelial dedifferentiation effects of sustained exposure to VX-809. This was observed in epithelium-like monolayers from both lung and intestinal origin, representing the two systems most affected by adverse symptoms in patients treated with VX-809 or the VX-809/VX-770 combination. Taken together, our findings strongly suggest that co-administration of HGF with corrector/potentiator drugs could be beneficial for CF patients.


Assuntos
Aminofenóis/administração & dosagem , Aminopiridinas/administração & dosagem , Benzodioxóis/administração & dosagem , Agonistas dos Canais de Cloreto/administração & dosagem , Regulador de Condutância Transmembrana em Fibrose Cística/deficiência , Fibrose Cística/tratamento farmacológico , Fator de Crescimento de Hepatócito/administração & dosagem , Quinolonas/administração & dosagem , Linhagem Celular , Regulador de Condutância Transmembrana em Fibrose Cística/genética , Humanos , Modelos Biológicos , Proteínas Mutantes/genética , Resultado do Tratamento
9.
Hum Gene Ther ; 29(7): 816-827, 2018 07.
Artigo em Inglês | MEDLINE | ID: mdl-29382231

RESUMO

Hepatic growth factor (HGF) has been widely used in studies on arterial remodeling after injury, and results turn out to be inconsistent. The changes of endogenous HGF expression after injury also remain controversial. This study clarified the role of exogenous human HGF (hHGF) gene transfer in neointimal hyperplasia and investigated the associated alterations of endogenous HGF and c-Met expressions under endothelial denudation with or without hHGF gene transfer using a balloon-injured rabbit aorta model. Sixty-one rabbits were randomly divided into normal controls, endothelial injury, endothelial injury with hHGF, or the control vector gene transfer groups. On weeks 1, 2, 4, and 8 after injury, neointimal hyperplasia and endothelialization were evaluated by the ratio of neointimal area to medial area (N/M ratio), CD31-positive staining, α-smooth muscle actin, and endothelial nitric oxide synthase expressions using histological analysis, immunohistochemistry staining, or real-time quantitative reverse transcriptase polymerase chain reaction. Endogenous rabbit HGF (rHGF) and c-Met expressions were detected with immunohistochemistry staining and quantitative reverse transcriptase polymerase chain reaction. It was found that expressions of endogeneous rHGF and c-Met in endothelial injury upregulated with peak levels on week 2 or week 4 after injury (p < 0.01). On week 1 after hHGF transfer, neointimal hyperplasia was significantly inhibited (p < 0.001), with decreased α-smooth muscle actin expression (p < 0.05) and improved endothelial cells regeneration and function (p < 0.01). More remarkable overexpression of endogenous rHGF and c-Met mRNAs were detected, and lowered positive staining of rHGF and c-Met was shown in the neointima (p < 0.05). These results demonstrated hHGF gene transfer induced further overexpression of endogenous rHGF and c-Met mRNAs but lowered immunoreactivities of rHGF and c-Met in the neointima, thus leading to significant attenuation of neointimal hyperplasia.


Assuntos
Terapia Genética , Fator de Crescimento de Hepatócito/administração & dosagem , Hiperplasia/terapia , Neointima/terapia , Proteínas Proto-Oncogênicas c-met/genética , Actinas/genética , Animais , Células Endoteliais/efeitos dos fármacos , Células Endoteliais/metabolismo , Regulação da Expressão Gênica/genética , Vetores Genéticos/administração & dosagem , Fator de Crescimento de Hepatócito/genética , Humanos , Hiperplasia/genética , Hiperplasia/patologia , Neointima/genética , Neointima/patologia , RNA Mensageiro/genética , Coelhos
10.
Hum Gene Ther ; 29(2): 271-282, 2018 02.
Artigo em Inglês | MEDLINE | ID: mdl-28950723

RESUMO

Investigations based on mesenchymal stem cells (MSCs) for osteoporosis have attracted attention recently. MSCs can be derived from various tissues, such as bone marrow, adipose, umbilical cord, placenta, and dental pulp. Among these, dental pulp-derived MSCs (DPSCs) and hepatocyte growth factor (HGF)-modified DPSCs (DPSCs-HGF) highly express osteogenic-related genes and have stronger osteogenic differentiation capacities. DPSCs have more benefits in treating osteoporosis. The purpose of this study was to investigate the roles of HGF gene-modified DPSCs in bone regeneration using a mouse model of ovariectomy (OVX)-induced bone loss. The HGF and luciferase genes were transferred into human DPSCs using recombinant adenovirus. These transduced cells were assayed for distribution or bone regeneration assay by transplantation into an OVX-induced osteoporosis model. By using bioluminogenic imaging, it was determined that some DPSCs could survive for >1 month in vivo. The DPSCs were mainly distributed to the lung in the early stage and to the liver in the late stage of OVX osteoporosis after administration, but they were scarcely distributed to the bone. The homing efficiency of DPSCs is higher when administrated in the early stage of a mouse OVX model. Micro-computed tomography indicated that DPSCs-Null or DPSCs-HGF transplantation significantly reduces OVX-induced bone loss in the trabecular bone of the distal femur metaphysis, and DPSCs-HGF show a stronger capacity to reduce bone loss. The data suggest that systemic infusion of DPSCs-HGF is a potential therapeutic approach for OVX-induced bone loss, which might be mediated by paracrine mechanisms.


Assuntos
Regeneração Óssea/genética , Reabsorção Óssea/terapia , Fator de Crescimento de Hepatócito/genética , Osteoporose/terapia , Animais , Regeneração Óssea/efeitos dos fármacos , Reabsorção Óssea/genética , Reabsorção Óssea/patologia , Diferenciação Celular/efeitos dos fármacos , Polpa Dentária/citologia , Polpa Dentária/transplante , Fator de Crescimento de Hepatócito/administração & dosagem , Humanos , Transplante de Células-Tronco Mesenquimais , Células-Tronco Mesenquimais/citologia , Camundongos , Osteogênese/genética , Osteoporose/fisiopatologia , Ovariectomia
11.
Kidney Blood Press Res ; 42(3): 495-508, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28854424

RESUMO

BACKGROUND/AIMS: The aim of this study was to assess the effect of human hepatocyte growth factor (hHGF)-loaded poly (ethylene glycol)-b-brush poly (l-lysine) (PEG-b-P(ELG-g-PLL)) copolymer on ischemia/reperfusion (I/R) injury to different organs. METHODS: The isoelectric point (pI) of hHGF is 5.5, and hHGF combined with PEG-b-P(ELG-g-PLL) copolymer via electrostatic interactions at pH 7.4. The synthesized PEG-b-P(ELG-g-PLL) copolymer was analyzed using 1H nuclear magnetic resonance (1H NMR) and gel permeation chromatography (GPC). The hHGF/PEG-b-P(ELG-g-PLL) complex was evaluated using a nanoparticle size instrument and transmission electron microscopy (TEM). In addition, vivo performance of hHGF/PEG-b-P(ELG-g-PLL) complex was evaluated using plasma hHGF concentration and different organs ischemia reperfusion injury in rats. RESULTS: An in vitro investigation showed that PEG-b-P(ELG-g-PLL) could serve as a potential hHGF nanocarrier with efficient encapsulation and sustained release. An additional in vivo investigation revealed that the hHGF/PEG-b-P(ELG-g-PLL) complex could prolong increases in plasma hHGF concentration and protect different organs (the brain, heart and kidney) against I/R injury. CONCLUSION: Poly (ethylene glycol)-block-brush poly (l-lysine) copolymer as an efficient nanocarrier for human hepatocyte growth factor with enhanced bioavailability and anti-ischemia reperfusion injury efficacy.


Assuntos
Portadores de Fármacos/química , Fator de Crescimento de Hepatócito/administração & dosagem , Polietilenoglicóis/uso terapêutico , Polilisina/análogos & derivados , Traumatismo por Reperfusão/tratamento farmacológico , Animais , Disponibilidade Biológica , Portadores de Fármacos/uso terapêutico , Fator de Crescimento de Hepatócito/farmacocinética , Humanos , Polilisina/uso terapêutico , Ratos
12.
Anticancer Res ; 37(8): 4255-4268, 2017 08.
Artigo em Inglês | MEDLINE | ID: mdl-28739717

RESUMO

BACKGROUND: Bone metastases are a common feature of advanced prostatic malignancies. They are characterised by a unique prevalence of osteoblastic phenotype and a poor prognosis. Paxillin is a 68-kDa signal transduction adaptor and scaffold protein that contains motifs involved in the mediation of protein-protein interactions. The state of paxillin phosphorylation is central to determining a cell's ability to adhere, detach and migrate and hence has been linked to processes such as wound repair and tumour metastasis. The current study explored the impact of paxillin suppression on prostate and breast cancer cell function and their responsiveness to hepatocyte growth factor (HGF) and bone matrix extract (BME) in order to assess its potential to influence bone colonization and homing. MATERIALS AND METHODS: Hammerhead ribozyme transgenes were used to knockdown the expression of paxillin in breast and prostate cancer cell lines. The impact on the cell growth, migration, adhesion and invasion was assessed using in vitro functional assays. In order to explore potential mechanisms, focal adhesion kinase (FAK) inhibitor was also used. RESULTS: Knockdown of paxillin expression was observed in all tested cell lines following transfection with the ribozyme transgene. The knockdown of paxillin increased proliferation and invasiveness of LNCaP cells, with no effect on their attachment abilities. The opposite, however, is true for PC-3 cells where, following knockdown, cellular attachment was significantly reduced, while no significant changes in growth and invasiveness were detected. In the MDA-MB-231 breast cancer knockdown model, cells had little difference in proliferative rates and generally increased attachment and reduced invasive abilities. Treatments with HGF and BME had differential effects on targeted cells when compared to controls. CONCLUSION: These data suggest that paxillin appears to influence major cell functions in a diverse range of prostate and breast cancer models. The responsiveness of cells to environmental factors such as HGF or BME may be influenced by paxillin status, although this seems to be dependent on cell type.


Assuntos
Neoplasias Ósseas/tratamento farmacológico , Extratos Celulares/administração & dosagem , Fator de Crescimento de Hepatócito/administração & dosagem , Paxilina/genética , Matriz Óssea/química , Neoplasias Ósseas/genética , Neoplasias Ósseas/patologia , Neoplasias Ósseas/secundário , Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/genética , Neoplasias da Mama/patologia , Extratos Celulares/química , Linhagem Celular Tumoral , Movimento Celular/genética , Proliferação de Células/efeitos dos fármacos , Feminino , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Técnicas de Silenciamento de Genes , Humanos , Masculino , Invasividade Neoplásica/genética , Paxilina/antagonistas & inibidores , Neoplasias da Próstata/tratamento farmacológico , Neoplasias da Próstata/genética , Neoplasias da Próstata/patologia
13.
Biotechnol Bioeng ; 114(10): 2379-2389, 2017 10.
Artigo em Inglês | MEDLINE | ID: mdl-28574594

RESUMO

In the last decade, numerous growth factors and biomaterials have been explored for the treatment of myocardial infarction (MI). While pre-clinical studies have demonstrated promising results, clinical trials have been disappointing and inconsistent, likely due to poor translatability. In the present study, we investigate a potential myocardial regenerative therapy consisting of a protein-engineered dimeric fragment of hepatocyte growth factor (HGFdf) encapsulated in a shear-thinning, self-healing, bioengineered hydrogel (SHIELD). We hypothesized that SHIELD would facilitate targeted, sustained intramyocardial delivery of HGFdf thereby attenuating myocardial injury and post-infarction remodeling. Adult male Wistar rats (n = 45) underwent sham surgery or induction of MI followed by injection of phosphate buffered saline (PBS), 10 µg HGFdf alone, SHIELD alone, or SHIELD encapsulating 10 µg HGFdf. Ventricular function, infarct size, and angiogenic response were assessed 4 weeks post-infarction. Treatment with SHIELD + HGFdf significantly reduced infarct size and increased both ejection fraction and borderzone arteriole density compared to the controls. Thus, sustained delivery of HGFdf via SHIELD limits post-infarction adverse ventricular remodeling by increasing angiogenesis and reducing fibrosis. Encapsulation of HGFdf in SHIELD improves clinical translatability by enabling minimally-invasive delivery and subsequent retention and sustained administration of this novel, potent angiogenic protein analog. Biotechnol. Bioeng. 2017;114: 2379-2389. © 2017 Wiley Periodicals, Inc.


Assuntos
Preparações de Ação Retardada/administração & dosagem , Fator de Crescimento de Hepatócito/administração & dosagem , Hidrogéis/química , Infarto do Miocárdio/tratamento farmacológico , Engenharia de Proteínas/métodos , Proteínas Recombinantes/administração & dosagem , Disfunção Ventricular Esquerda/prevenção & controle , Proteínas Angiogênicas/administração & dosagem , Proteínas Angiogênicas/química , Proteínas Angiogênicas/genética , Animais , Preparações de Ação Retardada/química , Difusão , Fator de Crescimento de Hepatócito/análogos & derivados , Fator de Crescimento de Hepatócito/genética , Injeções , Masculino , Infarto do Miocárdio/diagnóstico por imagem , Infarto do Miocárdio/patologia , Ratos , Ratos Sprague-Dawley , Ratos Wistar , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Resistência ao Cisalhamento , Resultado do Tratamento , Disfunção Ventricular Esquerda/diagnóstico por imagem , Disfunção Ventricular Esquerda/patologia , Viscosidade
14.
Surg Endosc ; 31(7): 2789-2797, 2017 07.
Artigo em Inglês | MEDLINE | ID: mdl-27752816

RESUMO

BACKGROUND: As hepatic surgery has become safer and more commonly performed, the extent of hepatic resections has increased. When there is not enough expected hepatic reserve to facilitate primary resection of hepatic tumors, a clinical adjunct to facilitating primary resection is portal vein embolization (PVE). PVE allows the hepatic remnant to increase to an appropriate size prior to resection via hepatocyte regeneration; however, PVE is not always successful in facilitating adequate regeneration. One of the strongest trophic factors for hepatocyte regeneration is hepatocyte growth factor (HGF). The purpose of this study was to improve hepatic regeneration with perioperative HGF infusions in an animal model that mimics PVE. METHODS: Portal branch ligation (PBL) in rodents is equivalent to PVE in humans. We performed left-sided PBL in Sprague-Dawley rodents with the experimental group receiving perioperative HGF infusions. Baseline and postoperative liver volumetrics were obtained with CT scanning methods as performed in clinical practice. Baseline and postoperative liver functions were assessed via indocyanine green (ICG) elimination testing. RESULTS: HGF infused rodents had statistically significant increase in all postoperative liver volumetrics. Most clinically relevant were increased right liver volumes (RLV), 14.10 versus 7.85 cm3 (p value 0.0001), and increased degree of hypertrophy (DH %), 159.23 versus 47.11 % (p value 0.0079). HGF infused rodents also had a quick return to baseline liver function, 2.38 days compared to 6.13 days (p value 0.0001). CONCLUSION: Perioperative HGF infusions significantly increase hepatic regeneration following PBL in rodents. Perioperative HGF infusions following PVE are a possible adjunct to increase the amount of patients able to successfully undergo primary resection for hepatic tumors. Further basic science is warranted in examining the use of HGF infusions to increase hepatic regeneration and translating that basic science work to clinical practice.


Assuntos
Fator de Crescimento de Hepatócito/farmacologia , Regeneração Hepática/efeitos dos fármacos , Fígado/efeitos dos fármacos , Veia Porta/cirurgia , Cuidados Pré-Operatórios/métodos , Animais , Embolização Terapêutica , Hepatectomia , Fator de Crescimento de Hepatócito/administração & dosagem , Infusões Parenterais , Ligadura , Fígado/diagnóstico por imagem , Fígado/crescimento & desenvolvimento , Fígado/cirurgia , Tamanho do Órgão , Ratos , Ratos Sprague-Dawley , Tomografia Computadorizada por Raios X
15.
Int J Nanomedicine ; 11: 4875-4890, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27729786

RESUMO

Inflammatory response is a major cause of grafts dysfunction in islet transplantation. Hepatocyte growth factor (HGF) had shown anti-inflammatory activity in multiple diseases. In this study, we aim to deliver HGF by self-assembling peptide/heparin (SAP/Hep) hybrid gel to protect ß-cell from inflammatory injury. The morphological and slow release properties of SAPs were analyzed. Rat INS-1 ß-cell line was treated with tumor necrosis factor α in vitro and transplanted into rat kidney capsule in vivo, and the viability, apoptosis, function, and inflammation of ß-cells were evaluated. Cationic KLD1R and KLD2R self-assembled to nanofiber hydrogel, which showed higher binding affinity for Hep and HGF because of electrostatic interaction. Slow release of HGF from cationic SAP/Hep gel is a two-step mechanism involving binding affinity with Hep and molecular diffusion. In vitro and in vivo results showed that HGF-loaded KLD2R/Hep gel promoted ß-cell survival and insulin secretion, and inhibited cell apoptosis, cytokine release, T-cell infiltration, and activation of NFκB/p38 MAPK pathways in ß-cells. This study suggested that SAP/Hep gel is a promising carrier for local delivery of bioactive proteins in islet transplantation.


Assuntos
Linfócitos B/citologia , Sobrevivência Celular/efeitos dos fármacos , Fator de Crescimento de Hepatócito/administração & dosagem , Hidrogéis/química , Inflamação/tratamento farmacológico , Células Secretoras de Insulina/citologia , Animais , Apoptose/efeitos dos fármacos , Cátions/química , Linhagem Celular , Dicroísmo Circular , Citocinas/metabolismo , Preparações de Ação Retardada , Difusão , Glucose/química , Heparina/química , Humanos , Insulina/química , Células Secretoras de Insulina/efeitos dos fármacos , Transplante das Ilhotas Pancreáticas , Luz , Masculino , Microscopia Eletrônica de Varredura , Microscopia Eletrônica de Transmissão , Nanofibras/química , Ratos , Espectroscopia de Infravermelho com Transformada de Fourier , Eletricidade Estática
16.
Ann Vasc Surg ; 36: 273-282, 2016 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-27423724

RESUMO

BACKGROUND: Current commercially available drug-eluting stents (DESs) are criticized for the problem of stent thrombosis by induced impaired re-endothelialization (RE). The solving of this challenge could be boosted by endothelial progenitor cells (EPCs). The purpose of this study was to examine the effects of hepatocyte growth factor (HGF) on this process. METHODS: The abundance and functional capacity of circulating EPC was analyzed by a fluorescence-activated cell sorter and western blot. The in vivo effect of HGF on DES patency, RE, and neointimal formation was investigated in a hypercholesterolemic rabbit model. RESULTS: After 7 days of HGF administration, the number of CD34+/CD133+ progenitor cells had increased significantly. HGF also significantly inhibited the onset of senescence of EPC due to a decrease in protein expression of p53 and p21. In the in vivo study, HGF-treated DES had a higher patency rate than the control group (11/12 vs. 6/12, P = 0.032). Moreover, the HGF-treated group exhibited better RE (control group: 69.5 ± 12.9%, HGF group: 88.8 ± 8.4%, P = 0.006), but significantly smaller areas of neointima (control group: 0.68 ± 0.15 mm2, HGF group: 0.45 ± 0.18 mm2, P = 0.02). CONCLUSION: HGF efficiently ameliorates the vascular response to stent implantation, and has an important redeeming influence on the deleterious endothelial effects of DES.


Assuntos
Angioplastia com Balão/instrumentação , Fármacos Cardiovasculares/administração & dosagem , Artérias Carótidas/efeitos dos fármacos , Doenças das Artérias Carótidas/terapia , Stents Farmacológicos , Células Progenitoras Endoteliais/efeitos dos fármacos , Fator de Crescimento de Hepatócito/administração & dosagem , Paclitaxel/administração & dosagem , Reepitelização/efeitos dos fármacos , Angioplastia com Balão/efeitos adversos , Animais , Apoptose/efeitos dos fármacos , Artérias Carótidas/diagnóstico por imagem , Artérias Carótidas/metabolismo , Artérias Carótidas/patologia , Doenças das Artérias Carótidas/diagnóstico por imagem , Doenças das Artérias Carótidas/etiologia , Doenças das Artérias Carótidas/metabolismo , Movimento Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Esquema de Medicação , Células Progenitoras Endoteliais/metabolismo , Células Progenitoras Endoteliais/patologia , Hipercolesterolemia/complicações , Injeções Subcutâneas , Masculino , Neointima , Coelhos , Fatores de Tempo
17.
Expert Opin Ther Targets ; 20(9): 1087-98, 2016 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-26941128

RESUMO

INTRODUCTION: Bone marrow-derived mesenchymal stem cells (MSCs) can differentiate into multiple cell types, including osteoblasts, chondrocytes, and adipocytes. These pluripotent cells secrete hepatocyte growth factor (HGF), which regulates cell growth, survival, motility, migration, mitogenesis and is important for tissue development/regeneration. HGF has four splice variants, NK1, NK2, NK3, and NK4 which have varying functions and affinities for the HGF receptor, cMET. HGF promotes osteoblastic differentiation of MSCs into bone forming cells, playing a role in bone development, health and repair. AREAS COVERED: This review will focus on the effects of HGF in osteogenesis, bone repair and bone health, including structural and functional insights into the role of HGF in the body. EXPERT OPINION: Approximately 6.2 million Americans experience a fracture annually, with 5-10% being mal- or non-union fractures. HGF is important in priming MSCs for osteogenic differentiation in vitro and is currently being studied to assess its role during bone repair in vivo. Due to the high turnover rate of systemic HGF, non-classic modes of HGF-treatment, including naked-plasmid HGF delivery and the use of HGF splice variants (NK1 & NK2) are being studied to find safe and efficacious treatments for bone disorders, such as mal- or non-union fractures.


Assuntos
Osso e Ossos/metabolismo , Fator de Crescimento de Hepatócito/metabolismo , Osteogênese/genética , Adipócitos/citologia , Processamento Alternativo/genética , Animais , Diferenciação Celular , Condrócitos/citologia , Regulação da Expressão Gênica , Fator de Crescimento de Hepatócito/administração & dosagem , Fator de Crescimento de Hepatócito/genética , Humanos , Células-Tronco Mesenquimais/citologia , Osteoblastos/citologia
18.
Am J Ther ; 23(6): e1602-e1611, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-26164021

RESUMO

Our study assessed the effect of bone marrow mesenchymal stem cells (BMSCs) expressing inducible hepatocyte growth factor (HGF) on the recovery of femoral head necrosis (FHN). BMSCs were isolated by density gradient centrifugation. A recombinant AdTRE-HGF was constructed as the response plasmid and Adeno-X Tet-on as the regulator vector. The regulator and the response vectors were coinfected into BMSCs and induced at 0, 200, 500, 1000, and 1200 ng/mL doxycycline (Dox). After 3 days, the concentration of HGF was determined using enzyme-linked immunosorbent assay. Forty rabbits were selected to establish the FHN model and divided into 4 experimental groups. After the rabbits were killed by ketamine overdose, the restoration of FHN was assessed. The distribution of HGF-positive cells was observed by immunohistochemical method. Enzyme-linked immunosorbent assay results showed that 1000 ng/mL Dox induced the highest HGF expression level, even higher than the 1200 ng/mL Dox induction. The highest osteonecrosis incidence and empty lacunae percentage were found in group A compared with all the other groups (all P < 0.05). Furthermore, dramatically lower osteonecrosis incidence and empty lacunae percentage were found in group C compared with those of groups B and D (all P < 0.05). A significantly higher level of HGF protein was detected in group C compared with the other groups (all P < 0.05). Our study successfully developed the AdTRE-HGF, a recombinant adenovirus carrying HGF gene, for high expression of HGF in BMSCs. Importantly, introduction of BMSCs expressing HGF successfully produced the desired therapeutic effect in reversing FHN, in a Dox-dependent manner.


Assuntos
Doxiciclina/farmacologia , Necrose da Cabeça do Fêmur/terapia , Fator de Crescimento de Hepatócito/biossíntese , Fator de Crescimento de Hepatócito/uso terapêutico , Células-Tronco Mesenquimais/metabolismo , Animais , Relação Dose-Resposta a Droga , Doxiciclina/administração & dosagem , Ensaio de Imunoadsorção Enzimática , Vetores Genéticos , Fator de Crescimento de Hepatócito/administração & dosagem , Masculino , Coelhos
19.
Anticancer Res ; 35(4): 1985-95, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25862851

RESUMO

Vitamin K plays a role in controlling cell growth. Anti-angiogenic effects of sorafenib lead to impairment of vitamin K uptake and induction of des-γ-carboxyprothrombin release by hepatocellular carcinoma (HCC) cells. We examined sorafenib and vitamin K individually and in combination regarding their ability to suppress migration and metastatic potential of HCC cells. HepG2 cells (HCC cell line) were treated with hepatocyte growth factor (HGF). E-Cadherin expression, phospho-MET (p-MET), and phospho-extracellular signal-regulated kinase (p-ERK) levels and cell migration were evaluated. HGF-stimulated HepG2 cells, which were treated with a combination of sorafenib and vitamin K, showed significantly increased expression of E-cadherin and impairment of migration ability compared to when treated with either agent alone. This combination therapy also induced marked inhibition of epithelial-mesenchymal transition phenotype; inhibition of HGF-stimulated cell proliferation, invasion and migration; and inhibition of HGF/c-MET signaling pathway. Levels of p-MET and p-ERK were also significantly reduced by this combination. Our experimental study demonstrated that sorafenib and vitamin K can function synergistically to inhibit the migration and proliferation of HCC cells. Combination therapy with sorafenib and vitamin K appears to be worthy of clinical trial with expectation of synergistic therapeutic effects.


Assuntos
Carcinoma Hepatocelular/tratamento farmacológico , Neoplasias Hepáticas/tratamento farmacológico , Niacinamida/análogos & derivados , Compostos de Fenilureia/administração & dosagem , Vitamina K/administração & dosagem , Carcinoma Hepatocelular/patologia , Movimento Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Sinergismo Farmacológico , Células Hep G2 , Fator de Crescimento de Hepatócito/administração & dosagem , Humanos , Neoplasias Hepáticas/patologia , Niacinamida/administração & dosagem , Sorafenibe
20.
J Biomed Mater Res A ; 103(9): 3012-25, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-25727843

RESUMO

One of the main cause of ineffective cell therapy in repairing the damaged heart is the poor yield of grafted cells. To overcome this drawback, rats with 4-week-old myocardial infarction (MI) were injected in the border zone with human adipose-derived stem cells (ADSCs) conveyed by poly(lactic-co-glycolic acid) microcarriers (PAMs) releasing hepatocyte growth factor (HGF) and insulin-like growth factor-1 (IGF-1) (GFsPAMs). According to treatments, animals were subdivided into different groups: MI_ADSC, MI_ADSC/PAM, MI_GFsPAM, MI_ADSC/GFsPAM, and untreated MI_V. Two weeks after injection, a 31% increase in ADSC engraftment was observed in MI_ADSC/PAM compared with MI_ADSC (p < 0.05). A further ADSC retention was obtained in MI_ADSC/GFsPAM with respect to MI_ADSC (106%, p < 0.05) and MI_ADSC/PAM (57%, p < 0.05). A 130% higher density of blood vessels of medium size was present in MI_ADSC/GFsPAM compared with MI_ADSC (p < 0.01). MI_ADSC/GFsPAM also improved, albeit slightly, left ventricular remodeling and hemodynamics with respect to the other groups. Notably, ADSCs and/or PAMs, with or without HGF/IGF-1, trended to induce arrhythmias in electrically driven, Langendorff-perfused, hearts of all groups. Thus, PAMs releasing HGF/IGF-1 markedly increase ADSC engraftment 2 weeks after injection and stimulate healing in chronically infarcted myocardium, but attention should be paid to potentially negative electrophysiological consequences.


Assuntos
Fator de Crescimento de Hepatócito/administração & dosagem , Fator de Crescimento Insulin-Like I/administração & dosagem , Infarto do Miocárdio/tratamento farmacológico , Infarto do Miocárdio/terapia , Transplante de Células-Tronco/métodos , Tecido Adiposo/citologia , Animais , Arritmias Cardíacas/etiologia , Materiais Biomiméticos/química , Modelos Animais de Doenças , Portadores de Fármacos/administração & dosagem , Humanos , Ácido Láctico , Masculino , Teste de Materiais , Microesferas , Infarto do Miocárdio/patologia , Neovascularização Fisiológica/efeitos dos fármacos , Ácido Poliglicólico , Copolímero de Ácido Poliláctico e Ácido Poliglicólico , Ratos , Ratos Wistar , Transplante de Células-Tronco/efeitos adversos , Remodelação Ventricular , Cicatrização/efeitos dos fármacos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA