Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 37
Filtrar
Mais filtros

Bases de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
Biochim Biophys Acta ; 1850(6): 1169-79, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-25665484

RESUMO

BACKGROUND: Cysteine-rich secretory protein (CRISP) is present in majority of vertebrate including human. The physiological role of this protein is not characterized. We report that a CRISP isolated from Echis carinatus sochureki venom (ES-CRISP) inhibits angiogenesis. METHODS: The anti-angiogenic activity of purified ES-CRISP from snake venom was investigated in vitro using endothelial cells assays such as proliferation, migration and tube formation in Matrigel, as well as in vivo in quail embryonic CAM system. The modulatory effect of ES-CRISP on the expression of major angiogenesis factors and activation of angiogenesis pathways was tested by qRT-PCR and Western blot. RESULTS: The amino acid sequence of ES-CRISP was found highly similar to other members of this snake venom protein family, and shares over 50% identity with human CRISP-3. ES-CRISP supported adhesion to endothelial cells, although it was also internalized into the cytoplasm in a granule-like manner. It blocked EC proliferation, migration and tube formation in Matrigel. In the embryonic quail CAM system, ES-CRISP abolished neovascularization process induced by exogenous growth factors (bFGF, vpVEGF) and by developing gliomas. CRISP modulates the expression of several factors at the mRNA level, which were characterized as regulators of angiogenesis and blocked activation of MAPK Erk1/2 induced by VEGF. CONCLUSIONS: ES-CRISP was characterized as a negative regulator of the angiogenesis, by direct interaction with endothelial cells. GENERAL SIGNIFICANCE: The presented work may lead to the development of novel angiostatic therapy, as well as contribute to the identification of the physiological relevance of this functionally uncharacterized protein.


Assuntos
Inibidores da Angiogênese/farmacologia , Membrana Corioalantoide/irrigação sanguínea , Células Endoteliais/efeitos dos fármacos , Glioma/irrigação sanguínea , Células Endoteliais da Veia Umbilical Humana/efeitos dos fármacos , Neovascularização Patológica , Neovascularização Fisiológica/efeitos dos fármacos , Venenos de Víboras/farmacologia , Sequência de Aminoácidos , Inibidores da Angiogênese/química , Inibidores da Angiogênese/isolamento & purificação , Inibidores da Angiogênese/metabolismo , Proteínas Angiogênicas/genética , Proteínas Angiogênicas/metabolismo , Animais , Linhagem Celular Tumoral , Movimento Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Relação Dose-Resposta a Droga , Células Endoteliais/metabolismo , Glioma/patologia , Células Endoteliais da Veia Umbilical Humana/metabolismo , Humanos , Dados de Sequência Molecular , Conformação Proteica , Codorniz , Transdução de Sinais/efeitos dos fármacos , Venenos de Víboras/química , Venenos de Víboras/isolamento & purificação , Venenos de Víboras/metabolismo
2.
Am J Physiol Lung Cell Mol Physiol ; 309(11): L1273-85, 2015 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-26408553

RESUMO

There is a clear unmet clinical need for novel biotechnology-based therapeutic approaches to lung repair and/or replacement, such as tissue engineering of whole bioengineered lungs. Recent studies have demonstrated the feasibility of decellularizing the whole organ by removal of all its cellular components, thus leaving behind the extracellular matrix as a complex three-dimensional (3D) biomimetic scaffold. Implantation of decellularized lung scaffolds (DLS), which were recellularized with patient-specific lung (progenitor) cells, is deemed the ultimate alternative to lung transplantation. Preclinical studies demonstrated that, upon implantation in rodent models, bioengineered lungs that were recellularized with airway and vascular cells were capable of gas exchange for up to 14 days. However, the long-term applicability of this concept is thwarted in part by the failure of current approaches to reconstruct a physiologically functional, quiescent endothelium lining the entire vascular tree of reseeded lung scaffolds, as inferred from the occurrence of hemorrhage into the airway compartment and thrombosis in the vasculature in vivo. In this review, we explore the idea that successful whole lung bioengineering will critically depend on 1) preserving and/or reestablishing the integrity of the subendothelial basement membrane, especially of the ultrathin respiratory membrane separating airways and capillaries, during and following decellularization and 2) restoring vascular physiological functionality including the barrier function and quiescence of the endothelial lining following reseeding of the vascular compartment. We posit that physiological reconstitution of the pulmonary vascular tree in its entirety will significantly promote the clinical translation of the next generation of bioengineered whole lungs.


Assuntos
Pulmão/irrigação sanguínea , Neovascularização Fisiológica , Engenharia Tecidual/métodos , Alicerces Teciduais/química , Animais , Matriz Extracelular/metabolismo , Humanos , Modelos Biológicos
3.
Br Med Bull ; 115(1): 45-56, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-26063231

RESUMO

INTRODUCTION: Mesenchymal stem cells (MSCs) of different biological sources are in Phase 1 clinical trials and are being considered for Phase 2 therapy of lung disorders, and lung (progenitor) cells derived from pluripotent stem cells (SCs) are under development in preclinical animal models. SOURCES OF DATA: PubMed.gov and ClinicalTrials.gov. AREAS OF AGREEMENT: There is consensus about the therapeutic potential of transplanted SCs, mainly MSCs, primarily involves paracrine 'bystander' effects that confer protection of the epithelial and endothelial linings of the lung caused by inflammation and/or fibrosis and lead to increased survival in animal models. Clinical trials of Phase 1 indicate safety and suggest that the efficacy of SC therapy in patients with various lung diseases will require a higher dosage than previously evaluated. AREAS OF CONTROVERSY: A growing interest in the re-epithelialization and re-endothelialization of damaged lung tissue involves the putative pulmonary differentiation of exogenous MSCs. Currently, it is not clear whether or not the observed regeneration of distal airways/vasculature is derived from lung-resident and/or transplanted SCs. GROWING POINTS: Important topics under investigation include optimization of the cell source with a decrease in cell population heterogeneity characterized by defined markers, route of delivery for effective treatment, potential dose and therapeutic protocol of SC application, development of quantitative assays and biomarkers of lung disease and repair, and the potential use of tissue engineered lung. AREAS TIMELY FOR DEVELOPING RESEARCH: Ability of MSCs to differentiate into epithelial cells of the lung, use of autologous induced pluripotent SCs (iPSCs) derived from the patients, complete biochemical characterization of the secretome of SCs used for therapy, and the incorporation of simultaneous and/or subsequent treatment with drugs which also aid in lung repair and regeneration. CAUTIONARY NOTE: Although safety of MSC-based cell therapy was proved in Phase 1, efficacy, long-term survival and preservation of lung respiratory function need to be further evaluated, cautioning against hastily translating SCs therapy from animal models of lung injury to clinical trials of patients with lung disorders.


Assuntos
Pneumopatias/terapia , Transplante de Células-Tronco Mesenquimais/métodos , Animais , Efeito Espectador , Diferenciação Celular/fisiologia , Modelos Animais de Doenças , Humanos , Pneumopatias/fisiopatologia , Transplante de Células-Tronco Mesenquimais/efeitos adversos , Engenharia Tecidual/métodos
4.
Biomedicines ; 12(7)2024 Jul 17.
Artigo em Inglês | MEDLINE | ID: mdl-39062165

RESUMO

Rasagiline (Azilect®) is a selective monoamine oxidase B (MAO-B) inhibitor that provides symptomatic benefits in Parkinson's disease (PD) treatment and has been found to exert preclinical neuroprotective effects. Here, we investigated the neuroprotective signaling pathways of acute rasagiline treatment for 22 h in PC12 neuronal cultures exposed to oxygen-glucose deprivation (OGD) for 4 h, followed by 18 h of reoxygenation (R), causing 40% aponecrotic cell death. In this study, 3-10 µM rasagiline induced dose-dependent neuroprotection of 20-80%, reduced the production of the neurotoxic reactive oxygen species by 15%, and reduced the nuclear translocation of glyceraldehyde-3-phosphate dehydrogenase (GAPDH) by 75-90%. In addition, 10 µM rasagiline increased protein kinase B (Akt) phosphorylation by 50% and decreased the protein expression of the ischemia-induced α-synuclein protein by 50% in correlation with the neuroprotective effect. Treatment with 1-5 µM rasagiline induced nuclear shuttling of transcription factor Nrf2 by 40-90% and increased the mRNA levels of the antioxidant enzymes heme oxygenase-1, (NAD (P) H- quinone dehydrogenase, and catalase by 1.8-2.0-fold compared to OGD/R insult. These results indicate that rasagiline provides neuroprotection to the ischemic neuronal cultures through the inhibition of α-synuclein and GAPDH-mediated aponecrotic cell death, as well as via mitochondrial protection, by increasing mitochondria-specific antioxidant enzymes through a mechanism involving the Akt/Nrf2 redox-signaling pathway. These findings may be exploited for neuroprotective drug development in PD and stroke therapy.

5.
J Cardiovasc Pharmacol ; 62(3): 270-7, 2013 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-23644989

RESUMO

Nerve growth factor (NGF) has been reported to play an important role in physiological and pathological angiogenesis. Based on these observations, we hypothesized that NGF may induce the formation of functional blood vessels in a hindlimb ischemic rabbit model. Hindlimb ischemia was induced in 34 rabbits bilaterally by endovascular embolization of femoral arteries. On the 7th, 14th, and 20th postembolization days, NGF was injected intramuscularly, in 1 ischemic limb, and vehicle was injected in the contralateral control limb. On the 40th day, newly developed collateral vessels (diameter >500 µm) were quantified by transauricular intraarterial subtraction angiography. Perfusion analysis of an in vivo dynamic computed tomography study was performed to the limbs to investigate the hemodynamic recovery of the distal ischemic tissues. Functional estimation of limb perfusion showed a statistically significant increase of blood flow and blood volume for NGF. However, the increase of the collateral vessels was not detectable angiographically, providing evidence for the existence of a NGF-stimulated capillary angiogenic network but not increase of arteriogenesis. The combination of NGF with either tropomyosin-related kinase type A or vascular endothelial growth factor receptor 2 antagonists abolished the NGF-induced hemodynamic recovery. These findings provide new insights into understanding the involvement of NGF in vascular formation and its applications in therapeutic angiogenesis.


Assuntos
Indutores da Angiogênese/uso terapêutico , Modelos Animais de Doenças , Isquemia/tratamento farmacológico , Músculo Esquelético/efeitos dos fármacos , Fator de Crescimento Neural/uso terapêutico , Receptor trkA/agonistas , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/agonistas , Indutores da Angiogênese/administração & dosagem , Indutores da Angiogênese/antagonistas & inibidores , Indutores da Angiogênese/isolamento & purificação , Animais , Capilares/diagnóstico por imagem , Capilares/efeitos dos fármacos , Capilares/patologia , Hemodinâmica/efeitos dos fármacos , Membro Posterior , Injeções Intramusculares , Isquemia/induzido quimicamente , Isquemia/diagnóstico por imagem , Isquemia/patologia , Masculino , Camundongos , Músculo Esquelético/irrigação sanguínea , Músculo Esquelético/diagnóstico por imagem , Músculo Esquelético/patologia , Neovascularização Fisiológica/efeitos dos fármacos , Fator de Crescimento Neural/administração & dosagem , Fator de Crescimento Neural/antagonistas & inibidores , Fator de Crescimento Neural/isolamento & purificação , Inibidores de Proteínas Quinases/efeitos adversos , Coelhos , Radiografia , Distribuição Aleatória , Receptor trkA/antagonistas & inibidores , Receptor trkA/metabolismo , Fluxo Sanguíneo Regional/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/antagonistas & inibidores , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/metabolismo
6.
Int J Mol Sci ; 14(7): 14669-88, 2013 Jul 12.
Artigo em Inglês | MEDLINE | ID: mdl-23857061

RESUMO

In this study, we present the applicability of imaging epidermal growth factor (EGF) receptor levels in preclinical models of COLO205 carcinoma cells in vitro, mice with orthotopic tumors and ex vivo colorectal tumor biopsies, using EGF-labeled with IRDye800CW (EGF-NIR). The near infrared (NIR) bio-imaging of COLO205 cultures indicated specific and selective binding, reflecting EGF receptors levels. In vivo imaging of tumors in mice showed that the highest signal/background ratio between tumor and adjacent tissue was achieved 48 hours post-injection. Dissected colorectal cancer tissues from different patients demonstrated ex vivo specific imaging using the NIR bio-imaging platform of the heterogeneous distributed EGF receptors. Moreover, in the adjacent gastrointestinal tissue of the same patients, which by Western blotting was demonstrated as EGF receptor negative, no labeling with EGF-NIR probe was detected. Present results support the concept of tumor imaging by measuring EGF receptor levels using EGF-NIR probe. This platform is advantageous for EGF receptor bio-imaging of the NCI-60 recommended panel of tumor cell lines including 6-9 colorectal cell lines, since it avoids radioactive probes and is appropriate for use in the clinical setting using NIR technologies in a real-time manner.


Assuntos
Neoplasias Colorretais/metabolismo , Receptores ErbB/metabolismo , Espectroscopia de Luz Próxima ao Infravermelho , Animais , Linhagem Celular Tumoral , Neoplasias Colorretais/patologia , Fator de Crescimento Epidérmico/química , Fator de Crescimento Epidérmico/metabolismo , Receptores ErbB/antagonistas & inibidores , Receptores ErbB/genética , Humanos , Camundongos , Camundongos Nus , Ligação Proteica , Interferência de RNA , RNA Interferente Pequeno/metabolismo , Transplante Heterólogo , Imagem Corporal Total
7.
Artigo em Inglês | MEDLINE | ID: mdl-23314533

RESUMO

BACKGROUND: Nerve growth factor (NGF) is a neurotrophin that supports the survival and differentiation of sympathetic neurons, and its increased expression after myocardial infarct was correlated with cardiac sympathetic hyperinnervation and arrhythmias. However, it is unclear whether NGF protects the heart during infarct. In this study, we sought to address this issue in rat heart exposed to ischemia/reperfusion injury (IRI). METHODS: NGF was administered intravenously (IV), 15 min before ischemia, at different concentrations in the absence or presence of inhibitors of phosphatidylinositol-3 kinase (PI3K) or nitric oxide synthase (NOS) in different groups of rats (n=6) with left coronary occlusion for 30 min followed by 120-min reperfusion. The area at risk and infarct to risk ratios were determined from sections stained with 1% 2,3,5-triphenylterazolium chloride. RESULTS: NGF treatment at doses of 0.015-15 µg/kg, with an optimal dose of 0.15 µg/kg given IV before ischemia, reduced the infarct size from about 60% at the area of risk to about 25%, indicating cardioprotection by about 60%. The infarct-sparing effects of NGF were partially abolished by the inhibition of PI3K and NOS using wortmannin and N(G)-monomethyl-l-arginine, respectively. CONCLUSIONS: We have demonstrated for the first time that NGF attenuates myocardial infarct damage in an in vivo rat model of myocardial regional IRI. This cardioprotective effect is proposed to be related to the activities of PI3K and NOS. This suggests that NGF has a potential therapeutic role in the treatment of IRI.


Assuntos
Traumatismo por Reperfusão Miocárdica/prevenção & controle , Fator de Crescimento Neural/farmacologia , Óxido Nítrico Sintase/metabolismo , Fosfatidilinositol 3-Quinase/metabolismo , Androstadienos/farmacologia , Animais , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Masculino , Traumatismo por Reperfusão Miocárdica/patologia , Fator de Crescimento Neural/administração & dosagem , Óxido Nítrico Sintase/antagonistas & inibidores , Inibidores de Fosfoinositídeo-3 Quinase , Ratos , Ratos Sprague-Dawley , Wortmanina , ômega-N-Metilarginina/farmacologia
8.
J Cell Physiol ; 226(1): 165-72, 2011 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-20658530

RESUMO

The tuftelin protein isoforms undergo post-translation modifications, and are ubiquitously expressed in various tissues in embryos, adults, and tumors. Developmental and pathological studies suggested an apparent correlation between oxygen deprivation and tuftelin expression. The aim of the study was therefore to investigate the effect of a pathological insult (hypoxia) and a physiological growth factor (NGF), which antagonistically regulate HIF1 expression, on tuftelin expression using the neuronal PC12 cell model. In the present study, we first demonstrated the expression of tuftelin in PC12 cells, providing an experimental system to investigate the pathophysiological role of tuftelin. Furthermore, we demonstrated the induction of tuftelin during hypoxia by oxygen deprivation and during chemical hypoxia by cobalt chloride. Down-regulation of HIF1α mRNA blocked hypoxia-induced HIF1α expression, and reduced by 89% hypoxia-induced tuftelin expression. In mice, intraperitoneal injection of cobalt chloride significantly induced tuftelin mRNA and protein expression in the brain. During NGF-mediated PC12 differentiation, tuftelin expression was significantly induced in correlation with neurite outgrowth. This induction was partially blocked by K252a, a selective antagonist of the NGF receptor TrkA, indicating the involvement of the TrkA-signaling pathways in tuftelin induction by NGF. Revealing the physiological role of tuftelin will clarify mechanisms related to the "hypoxic genome," and NGF-induced neurotrophic and angiogenic effects.


Assuntos
Proteínas do Esmalte Dentário/metabolismo , Regulação da Expressão Gênica/efeitos dos fármacos , Fator de Crescimento Neural/farmacologia , Consumo de Oxigênio/fisiologia , Glândulas Suprarrenais/efeitos dos fármacos , Glândulas Suprarrenais/metabolismo , Animais , Diferenciação Celular , Cobalto/toxicidade , Proteínas do Esmalte Dentário/genética , Subunidade alfa do Fator 1 Induzível por Hipóxia/genética , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Oxigênio/farmacologia , Células PC12 , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , RNA Interferente Pequeno , Ratos , Receptor trkA/genética , Receptor trkA/metabolismo , Transdução de Sinais
9.
Eur J Oral Sci ; 119 Suppl 1: 55-61, 2011 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-22243227

RESUMO

Tuftelin, an acidic protein, thought to play a role in the initial stages of ectodermal enamel mineralization, has since been detected in mesenchymal-derived tissues. During bone/cartilage development and regeneration, mesenchymal stem cells (MSCs) undergo an avascular period in a hypoxic environment, involving induction of hypoxia-inducible factor 1-alpha (HIF-1-alpha), a key component in this process. In the present study we investigated, in a mouse mesenchymal C3H10T1/2 stem cell model, the hypothesis that oxygen stress modulates tuftelin 1 expression in relation to HIF-1-alpha (Hif1a), in a mouse mesenchymal C3H10T1/2 stem cell model. The results of the present study showed a biphasic induction of tuftelin, similar to the pattern of HIF-1-alpha expression, in MSCs subjected to a hypoxic insult of 1% O(2) through a period of 2-24 h. Immunocytochemistry analysis of the cells exposed to hypoxic insult for 2-24 h revealed the same biphasic pattern of tuftelin protein expression. Tuftelin localization appears to be mainly in the cytoplasm, and concentrated at the perinuclear region of the cells by 24 h of hypoxic insult. Based on our previous studies using the neuronal PC12 cell model, in which tuftelin induction was mediated by Hif1a, we propose that tuftelin is a member of oxygen-sensitive genes and implicated in the adaptive mechanisms regulating MSC function.


Assuntos
Hipóxia Celular/fisiologia , Proteínas do Esmalte Dentário/biossíntese , Subunidade alfa do Fator 1 Induzível por Hipóxia/biossíntese , Células-Tronco Mesenquimais/metabolismo , Adaptação Fisiológica , Animais , Morte Celular , Células Cultivadas , Proteínas do Esmalte Dentário/genética , Regulação da Expressão Gênica no Desenvolvimento , Subunidade alfa do Fator 1 Induzível por Hipóxia/genética , L-Lactato Desidrogenase/metabolismo , Camundongos , Camundongos Endogâmicos C3H
10.
Nanomedicine ; 7(4): 480-8, 2011 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-21272665

RESUMO

A new liposome-based near-infrared probe that combines both imaging and targeting abilities was developed for application in medical imaging. The near-infrared fluorescent molecule indocyanine green (ICG), and the cetuximab monoclonal antibody for epidermal growth factor receptor (EGFR) were attached to liposomes by passive adsorption. It was found that ICG molecules adsorbed to the liposomes are more fluorescent than free ICG and have a larger quantum yield. Cetuximab-adsorbed fluorescent liposomes preserved EGFR recognition, as is evident from internalization and selective binding to A431 colon carcinoma cells overexpressing EGFR. The binding of cetuximab-targeted fluorescent liposomes to A431 compared with IEC-6 cells (normal enterocytes expressing physiological EGFR levels) was greater by a factor of 3.5, ensuring imaging abilities with available fluorescent equipment. Due to relatively high quantum yield and specific tumor cell-recognizing ability, this technology deserves further in vivo evaluation for imaging and diagnostic purposes. FROM THE CLINICAL EDITOR: A new liposome-based near-infrared probe combining both imaging and targeting abilities is reported. Due to relatively high quantum yield and EGFR-expressing tumor cell specificity, this technology deserves further in vivo evaluation for imaging and diagnostic purposes.


Assuntos
Anticorpos Monoclonais/química , Diagnóstico por Imagem/métodos , Espectroscopia de Luz Próxima ao Infravermelho/métodos , Anticorpos Monoclonais Humanizados , Linhagem Celular Tumoral , Cetuximab , Neoplasias do Colo/metabolismo , Neoplasias do Colo/patologia , Microscopia Crioeletrônica , Ensaio de Imunoadsorção Enzimática , Receptores ErbB/metabolismo , Humanos , Verde de Indocianina/química , Lipossomos/química , Microscopia Confocal , Microscopia Eletrônica de Transmissão
11.
Arch Ital Biol ; 149(2): 233-45, 2011 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-21701995

RESUMO

Stem cells have an extremely high potential to treat many devastating diseases, including neuronal injuries. Albeit the need for human neuronal stem cells, their quantities are very limited by relying on early human embryos as the main source. Therefore, progenitors of other origins, such as human umbilical cord blood (CB) are being considered. In the last decade, various populations isolated from the CB were reported to differentiate in vitro towards a neural phenotype. The conditions to induce the cell differentiation are not conclusive and may include addition of chemicals, cytokines and growth factors, including the nerve growth factor (NGF). Some CB cells were found to express the TrkANGF receptor, suggesting an endogenous role for this growth factor also in the CB environment. The ability of CB and derived stem cell populations to protect against neurological deficits was shown, both in vitro and in vivo, in models of ischemic brain injuries. In rodent models of stroke, heatstroke, brain trauma and brain damage at birth, CB cells either by intravenous injection or intrastriatal transplantation, were found to reduce the infarct size and the neurological deficits caused by the injury. The restorative effects of CB were suggested to be mediated by mechanisms other than cell replacement. Some of the proposed mechanisms involve reduced inflammation, nerve fiber reorganization by trophic actions, increased cell survival and enhanced angiogenesis. Furthermore, treatment with CB was found to have a therapeutic window of days compared with the present 36 hour window for the treatment of stroke with clinically available tools such as recombinant tissue plasminogen activator. Considering the encouraging results with whole CB and derived cells transplantation in ischemic injury models and since CB is widely available and have been used clinically, they may be an excellent source of cells for treatment of human brain ischemic disorders.


Assuntos
Lesões Encefálicas/prevenção & controle , Isquemia Encefálica/cirurgia , Transplante de Células-Tronco de Sangue do Cordão Umbilical/métodos , Sangue Fetal/citologia , Células-Tronco Hematopoéticas/fisiologia , Animais , Lesões Encefálicas/etiologia , Isquemia Encefálica/complicações , Diferenciação Celular , Humanos , Fator de Crescimento Neural/metabolismo , Fator de Crescimento Neural/uso terapêutico , Fármacos Neuroprotetores/metabolismo , Fármacos Neuroprotetores/uso terapêutico
12.
Mol Cell Biochem ; 339(1-2): 201-13, 2010 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-20148355

RESUMO

Nerve growth factor (NGF) supports the survival and differentiation of sympathetic and sensory neurons and is also mitogenic for a variety of tumors. K252a, an antagonist of NGF receptor TrkA, was previously used as a pharmacological tool to study NGF actions and as a lead compound for developing anti-tumor drugs. Since recently, NGF was characterized as an angiogenic factor, we sought to investigate the angiostatic properties of K252a on endothelial cells (ECs). For this purpose, we used a murine brain microcapillary ECs model in which we found autocrine release of NGF in the culture medium and activation of TrkA receptor-induced downstream signaling molecules Erk1/2, Akt, and PLCgamma. In this model, we demonstrated the angiostatic property of K252a based on its ability to affect several important angiogenic steps. K252a, but not its cell membrane impermeable analogue K252b at 100 nM: (i) inhibited the proliferation of the ECs by 45 +/- 9%; (ii) reduced by 70 +/- 4% the migration of the ECs measured in a wound-closure model; (iii) reduced by 29 +/- 9% the formation of tube-like structures of the ECs cultured on Matrigel; (iv) stimulated by 100 +/- 25% the collagen deposition by the ECs, a process responsible for the increased endothelial barrier functions expressed by 22 +/- 5% reduction of paracellular permeability and by 17 +/- 3% elevation of transendothelial electrical resistance. These data suggest that NGF/TrkA may represent a target for the development of novel, K252a-derived multikinase inhibitors drugs with anti-tumor and angiostatic dual activities.


Assuntos
Encéfalo/efeitos dos fármacos , Encéfalo/metabolismo , Carbazóis/farmacologia , Células Endoteliais/efeitos dos fármacos , Inibidores Enzimáticos/farmacologia , Alcaloides Indólicos/farmacologia , Receptor trkA/antagonistas & inibidores , Receptor trkA/metabolismo , Animais , Western Blotting , Encéfalo/citologia , Diferenciação Celular/efeitos dos fármacos , Permeabilidade da Membrana Celular/efeitos dos fármacos , Movimento Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Colágeno/metabolismo , Combinação de Medicamentos , Células Endoteliais/metabolismo , Laminina/metabolismo , Camundongos , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Fator de Crescimento Neural/genética , Fator de Crescimento Neural/metabolismo , Fosforilação/efeitos dos fármacos , Proteoglicanas/metabolismo , RNA Mensageiro/genética , Receptor trkA/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa
13.
Nephrol Dial Transplant ; 25(2): 373-80, 2010 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-19828461

RESUMO

BACKGROUND: Renal cell carcinoma (RCC) is considered resistant to ionizing radiation. Recently, the extracellular matrix (ECM) has been shown to play a role in both drug resistance and radiation resistance (RR). While fibronectin has been extensively investigated in the context of RR, the role of type I collagen [col(I)], a principal constituent of the ECM in tumour metastases, in RR of RCC is unknown. METHODS: RCC cell adhesion to matrix was studied via pre-coating a variety of ECM glycoproteins onto plates. Cancer cell apoptosis and cell cycle were evaluated with flow cytometry using annexin V and propidium iodide stains, respectively. Activation of cellular survival signalling was analysed with western blots, and specific molecular inhibitors were correspondingly employed to block signalling. Hypoxia (<1%) was induced via N(2)/CO(2) gas flow in a specialized chamber. RESULTS: While adherence to col(I) enhanced RCC cell proliferation in general, col(I) and fibronectin, but not fibrinogen, could confer specific anti-apoptotic RR to RCC cells. The radioprotective effect of col(I) was maintained during both hypoxia/reoxygenation and normoxia conditions. In contrast to intact col(I), micronized col(I), lacking the natural fibrillar structure, was not radioprotective. The effect of col(I) in RCC cells is mediated via attenuation of apoptosis rather than cell cycle redistribution, involving the PI3 kinase/Akt pathway but not the MAP kinase pathway. CONCLUSIONS: Adherence to col(I) appears to be a relevant environmental cue enhancing RR in RCC cells, Akt dependently. Our results support inhibition of the PI3-kinase/Akt pathway as a radiosensitizing approach.


Assuntos
Carcinoma de Células Renais/patologia , Carcinoma de Células Renais/radioterapia , Neoplasias Renais/patologia , Neoplasias Renais/radioterapia , Proteína Oncogênica v-akt/fisiologia , Adesão Celular , Colágeno Tipo I , Humanos , Falha de Tratamento , Células Tumorais Cultivadas/efeitos da radiação
14.
Int J Mol Sci ; 11(9): 3513-28, 2010 Sep 21.
Artigo em Inglês | MEDLINE | ID: mdl-20957109

RESUMO

The use of stem cells for reparative medicine was first proposed more than three decades ago. Hematopoietic stem cells from bone marrow, peripheral blood and human umbilical cord blood (CB) have gained major use for treatment of hematological indications. CB, however, is also a source of cells capable of differentiating into various non-hematopoietic cell types, including neural cells. Several animal model reports have shown that CB cells may be used for treatment of neurological injuries. This review summarizes the information available on the origin of CB-derived neuronal cells and the mechanisms proposed to explain their action. The potential use of stem/progenitor cells for treatment of ischemic brain injuries is discussed. Issues that remain to be resolved at the present stage of preclinical trials are addressed.


Assuntos
Isquemia Encefálica/cirurgia , Transplante de Células-Tronco de Sangue do Cordão Umbilical , Animais , Humanos , Células-Tronco Neurais/citologia , Células-Tronco Neurais/transplante , Neurogênese
15.
J Mol Neurosci ; 37(3): 225-37, 2009 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-18629654

RESUMO

There is a paucity of quantitative methods for evaluating the morphological differentiation of neuronal cells in a three-dimensional (3-D) system to assist in quality control of neural tissue engineering constructs for use in reparative medicine. Neuronal cells tend to aggregate in the 3-D scaffolds, hindering the application of two-dimensional (2-D) morphological methods to quantitate neuronal differentiation. To address this problem, we developed a stable transfectant green fluorescence protein (GFP)-PC12 neuronal cell model, in which the differentiation process in 3-D can be monitored with high sensitivity by fluorescence microscopy. Under 2-D conditions, the green cells showed collagen adherence, round morphology, proliferation properties, expression of the nerve growth factor (NGF) receptors TrkA and p75(NTR), stimulation of extracellular signal-regulated kinase phosphorylation by NGF and were able to differentiate in a dose-dependent manner upon NGF treatment, like wild-type (wt)-PC12 cells. When grown within 3-D collagen gels, upon NGF treatment, the GFP-PC12 cells differentiated, expressing long neurite outgrowths. We describe here a new validated method to measure NGF-induced differentiation in 3-D. Having properties similar to those of wt-PC12 and an ability to grow and differentiate in 3-D structures, these highly visualized GFP-expressing PC12 cells may serve as an ideal model for investigating various aspects of differentiation to serve in neural engineering.


Assuntos
Diferenciação Celular/fisiologia , Colágeno/metabolismo , Géis/metabolismo , Proteínas de Fluorescência Verde/metabolismo , Neurônios/fisiologia , Células PC12 , Animais , Proliferação de Células , Proteínas de Fluorescência Verde/genética , Neurônios/citologia , Células PC12/citologia , Células PC12/fisiologia , Ratos , Receptores de Fator de Crescimento Neural/genética , Receptores de Fator de Crescimento Neural/metabolismo
16.
Peptides ; 29(9): 1620-5, 2008 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-18584915

RESUMO

Pardaxin is a fish toxin belonging to the alpha-helical, pore-forming peptide family, used in toxicological and biophysical research to study toxin-cell and -lipid-artificial membranes interactions. We investigated the membrane interaction of two pardaxin analogues using a colorimetric phospholipid/polydiacetylene biomimetic assay. In this assay, polydiacetylene undergoes visible, concentration dependent, blue-red transformation induced through interactions of pardaxins with the vesicle membrane. Pardaxins P4 and P5, are composed of 33 amino acids, but differ in a single amino acid substitution at the carboxy-terminal (G31 to D31, respectively) known to decrease the pore forming activity. Addition of pardaxins in the colorimetric assay induced dose-dependent color transitions with different kinetics. The colorimetric analysis could distinguish between different pardaxins-membrane interaction profiles, suggesting bilayer surface association for P4 and vesicle membrane penetration for P5. The colorimetric assay could distinguish between pardaxins membrane interaction profiles although circular dichroism spectra of vesicle-interacting pardaxins did not indicate a significant difference in the secondary structure between these two toxin analogues. The colorimetric platform utilized in the present report represents a useful assay with general applications for studying membrane interactions of peptides in general and pore-forming toxins in particular, and may become an important tool for evaluating quantitative toxin structure-activity relationship.


Assuntos
Venenos de Peixe/farmacologia , Lipídeos de Membrana/fisiologia , Membranas Artificiais , Fosfolipídeos/fisiologia , Sequência de Aminoácidos , Animais , Materiais Biomiméticos , Colorimetria/métodos , Relação Dose-Resposta a Droga , Venenos de Peixe/análise , Venenos de Peixe/genética , Dados de Sequência Molecular , Mutação Puntual , Polímero Poliacetilênico , Polímeros , Poli-Inos , Relação Estrutura-Atividade
17.
Stem Cells Dev ; 24(5): 663-76, 2015 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-25226206

RESUMO

We investigated the effects of hypoxia on spontaneous (SP)- and activin A (AA)-induced definitive endoderm (DE) differentiation of mouse embryonic stem cells (mESCs) and their subsequent differentiation into distal pulmonary epithelial cells. SP differentiation for 6 days of mESCs toward endoderm at hypoxia of 1% O2, but not at 3% or 21% (normoxia), increased the expression of Sox17 and Foxa2 by 31- and 63-fold above maintenance culture, respectively. Treatment of mESCs with 20 ng/mL AA for 6 days under hypoxia further increased the expression of DE marker genes Sox17, Foxa2, and Cxcr4 by 501-, 1,483-, and 126-fold above maintenance cultures, respectively. Transient exposure to hypoxia, as short as 24 h, was sufficient to enhance AA-induced endoderm formation. The involvement of hypoxia-inducible factor (HIF)-1α and reactive oxygen species (ROS) in the AA-induced endoderm enrichment was assessed using HIF-1α(-/-) mESCs and the ROS scavenger N-acetylcysteine (NAC). Under SP conditions, HIF-1α(-/-) mESCs failed to increase the expression of endodermal marker genes but rather shifted toward ectoderm. Hypoxia induced only a marginal potentiation of AA-induced endoderm differentiation in HIF-1α(-/-) mESCs. Treatment of mESCs with AA and NAC led to a dose-dependent decrease in Sox17 and Foxa2 expression. In addition, the duration of exposure to hypoxia in the course of a recently reported lung differentiation protocol resulted in differentially enhanced expression of distal lung epithelial cell marker genes aquaporin 5 (Aqp5), surfactant protein C (Sftpc), and secretoglobin 1a1 (Scgb1a1) for alveolar epithelium type I, type II, and club cells, respectively. Our study is the first to show the effects of in vitro hypoxia on efficient formation of DE and lung lineages. We suggest that the extent of hypoxia and careful timing may be important components of in vitro differentiation bioprocesses for the differential generation of distal lung epithelial cells from pluripotent progenitors.


Assuntos
Diferenciação Celular , Endoderma/citologia , Pulmão/citologia , Células-Tronco Embrionárias Murinas/fisiologia , Animais , Hipóxia Celular , Células Cultivadas , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Pulmão/embriologia , Camundongos , Espécies Reativas de Oxigênio/metabolismo
18.
Cell Signal ; 27(6): 1225-36, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-25748048

RESUMO

Direct interaction of α9ß1 integrin with nerve growth factor (NGF) has been previously reported to induce pro-proliferative and pro-survival activities of non-neuronal cells. We investigated participation of p75(NTR) in α9ß1 integrin-dependent cellular response to NGF stimulation. Using selective transfection of glioma cell lines with these receptors, we showed a strong, cation-independent association of α9 integrin subunit with p75(NTR) on the cellular membrane by selective immunoprecipitation experiments. The presence of the α9/p75(NTR) complex increases NGF-dependent cell adhesion, proliferation and migration. Other integrin subunits including ß1 were not found in complex with p75(NTR). FRET analysis indicated that p75(NTR) and α9 integrin subunit are not closely associated through their cytoplasmic domains, most probably because of the molecular interference with other cytoplasmic proteins such as paxillin. Interaction of α9ß1 integrin with another ligand, VCAM-1 was not modulated by the p75(NTR). α9/p75(NTR) complex elevated NGF-dependent activation of MAPK Erk1/2 arty for integrin that may create active complexes with other types of receptors belonging to the TNF superfamily.


Assuntos
Proliferação de Células/efeitos dos fármacos , Integrinas/metabolismo , Fator de Crescimento Neural/farmacologia , Proteínas do Tecido Nervoso/metabolismo , Receptores de Fator de Crescimento Neural/metabolismo , Animais , Adesão Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Transferência Ressonante de Energia de Fluorescência , Células HEK293 , Humanos , Imuno-Histoquímica , Integrinas/química , Integrinas/genética , Camundongos , Microscopia Confocal , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Fator de Crescimento Neural/isolamento & purificação , Proteínas do Tecido Nervoso/química , Proteínas do Tecido Nervoso/genética , Paxilina/metabolismo , Ligação Proteica , Receptor trkA/metabolismo , Receptores de Fator de Crescimento Neural/química , Receptores de Fator de Crescimento Neural/genética , Molécula 1 de Adesão de Célula Vascular/metabolismo
19.
Prog Brain Res ; 146: 387-401, 2004.
Artigo em Inglês | MEDLINE | ID: mdl-14699975

RESUMO

Inflammatory processes in the central nervous system (CNS) are considered neurotoxic, although recent studies suggest that they also can be beneficial and confer neuroprotection (neuroprotective autoimmunity). Cells from the immune system have been detected in CNS injury and found to produce and secrete a variety of neurotrophins such as NGF, BDNF, NT-3 and NT-4/5, and to express (similarly to neuronal cells), members of the tyrosine kinase (Trk) receptor family such as TrkA, TrkB and TrkC. Indeed, autocrine and paracrine interactions are observed at the site of CNS injury, resulting in a variety of homologic-heterologic modulations of immune and neuronal cell function. The end result of the inflammatory process, neurotoxicity and/or neuroprotection, is a function of the fine balance between the two cellular systems, i.e., of the complex signaling relationships between anti-inflammatory neuroprotective factors (neurotrophins and other chemical mediators) and proinflammatory neurotoxic factors (TNF, free radicals, certain cytokines, etc.). Autoimmune neuroprotection is a novel therapeutic approach aimed at shifting the balance between the immune and neuronal cells towards survival pathways in a variety of CNS injuries. This review focuses on data supporting this concept and its future therapeutical implications for optic nerve injury and multiple sclerosis.


Assuntos
Sistema Imunitário/fisiologia , Fatores de Crescimento Neural/uso terapêutico , Sistema Nervoso/imunologia , Fármacos Neuroprotetores/uso terapêutico , Traumatismos do Sistema Nervoso/prevenção & controle , Animais , Humanos , Inflamação/induzido quimicamente , Inflamação/prevenção & controle , Fatores de Crescimento Neural/metabolismo , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Fármacos Neuroprotetores/metabolismo , Receptor trkC/genética , Receptor trkC/metabolismo , Traumatismos do Sistema Nervoso/metabolismo
20.
J Mol Neurosci ; 54(3): 574-85, 2014 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-25078264

RESUMO

Nerve growth factor (NGF) treatment causes a profound down-regulation of epidermal growth factor (EGF) receptors (EGFR) during the neuronal differentiation of PC12 cells. This process was characterized by a progressive decrease in EGFR level, as measured by (125)I-EGF binding and Scatchard analysis, tyrosine phosphorylation, Western blotting, and bio-imaging using EGF-labeled with a near-infrared probe. Differentiation of the cells with NGF for 5-7 days produces a 95 % reduction in the amount of (35)S-methionine-labeled EGFR. This down-regulation does not occur in PC12-nnr5 cells, which lack the TrkA NGF receptor but is reconstituted in these cells upon their stable transfection with TrkA. The process of NGF-induced EGFR down-regulation was inhibited by K252a, a TrkA antagonist and by anti-TrkA antibodies but not by Thx-B, a blocker of the interaction of NGF with p75(NTR) receptors. NGF-induced (heterologous) down-regulation, but not EGF-induced (homologous) down-regulation of EGFR, was blocked in Ras-deficient PC12 cells. NGF treatment for 5-7 days of PC12 cells, grown in suspension or in 3D collagen gels, induces down-regulation of EGFR independent of neurite outgrowth. The messenger RNA (mRNA) for EGFR decreased in a comparable fashion. This process was correlated temporally with a decrease in the transcription of the EGFR gene. Treatment with NGF also increased the cellular content of GCF2, a putative inhibitory transcription factor of the EGFR gene. The temporal increase in GCF2, like the decrease in the EGFR mRNA, was not seen in TrkA deficient PC12 cells nor in cells expressing dominant-negative Ras. The results suggest that NGF-induced down-regulation of the EGFR is under transcriptional control, is TrkA and Ras-dependent, may involve transcriptional repression by GCF2, and independent of mechanisms that lead to NGF-induced neurite outgrowth in PC12cells. This heterologous down-regulation of EGFR would appear to be an efficient mean of desensitizing the neuron to proliferative stimuli, thereby representing a safety latch for initiating and sustaining NGF-induced neuronal differentiation.


Assuntos
Regulação para Baixo , Receptores ErbB/metabolismo , Fator de Crescimento Neural/farmacologia , Transcrição Gênica , Animais , Fator de Crescimento Epidérmico/farmacologia , Receptores ErbB/genética , Neuritos/efeitos dos fármacos , Neuritos/metabolismo , Células PC12 , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Ratos , Receptor trkA/metabolismo , Proteínas ras/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA