RESUMO
Neuroblastoma (NB) is the most common extracranial solid tumor that originates from multipotent neural crest cells. NB cell populations that express embryonic stem cell-associated genes have been identified and shown to retain a multipotent phenotype. However, whether somatic reprogramming of NB cells can produce similar stem-cell like populations is unknown. Here, we sought to reprogram NB cell lines using an integration-free Sendai virus vector system. Of four NB cell lines examined, only SH-IN cells formed induced pluripotent stem cell-like colonies (SH-IN 4F colonies) at approximately 6 weeks following transduction. These SH-IN 4F colonies were alkaline phosphatase-positive. Array comparative genomic hybridization analysis indicated identical genomic aberrations in the SH-IN 4F cells as in the parental cells. SH-IN 4F cells had the ability to differentiate into the three embryonic germ layers in vitro, but rather formed NBs in vivo. Furthermore, SH-IN 4F cells exhibited resistance to cisplatin treatment and differentiated into endothelial-like cells expressing CD31 in the presence of vascular endothelial growth factor. These results suggest that SH-IN 4F cells are partially reprogrammed NB cells, and could be a suitable model for investigating the plasticity of aggressive tumors.
Assuntos
Plasticidade Celular/genética , Reprogramação Celular/genética , Células-Tronco Pluripotentes Induzidas/citologia , Neuroblastoma/genética , Neuroblastoma/patologia , Diferenciação Celular , Linhagem Celular Tumoral , Cisplatino/farmacologia , Hibridização Genômica Comparativa , Resistencia a Medicamentos Antineoplásicos , Células Endoteliais/citologia , Vetores Genéticos/genética , Proteínas de Homeodomínio/biossíntese , Proteínas de Homeodomínio/genética , Humanos , Células-Tronco Pluripotentes Induzidas/virologia , Fator 4 Semelhante a Kruppel , Fatores de Transcrição Kruppel-Like/biossíntese , Fatores de Transcrição Kruppel-Like/genética , Proteína Homeobox Nanog , Fator 3 de Transcrição de Octâmero/biossíntese , Fator 3 de Transcrição de Octâmero/genética , Regiões Promotoras Genéticas/genética , Proteínas Proto-Oncogênicas c-myc/biossíntese , Proteínas Proto-Oncogênicas c-myc/genética , Fatores de Transcrição SOXB1/biossíntese , Fatores de Transcrição SOXB1/genética , Vírus SendaiRESUMO
7-Methoxy-6-[4-(4-methyl-1,3-thiazol-2-yl)-1H-imidazol-5-yl]-1,3-benzothiazole 11 (TASP0382088) was synthesized and evaluated as transforming growth factor-ß (TGF-ß) type I receptor (also known as activin receptor-like kinase 5 or ALK5) inhibitor. Compound 11, a potent and selective ALK5 inhibitor, exhibited good enzyme inhibitory activity (IC50=4.8 nM) as well as inhibitory activity against TGF-ß-induced Smad2/3 phosphorylation at a cellular level (IC50=17 nM). The introduction of a methoxy group to the benzothiazole ring in 1 and the break up of the planarity between the imidazole ring and the thiazole ring improved the solubility in the lotion base of 11. Furthermore, the topical application of 3% 11 lotion significantly inhibited Smad2 phosphorylation in mouse skin at 8 h after application (71% inhibition, compared with vehicle-treated animals).
Assuntos
Alopecia/tratamento farmacológico , Benzotiazóis/farmacologia , Inibidores Enzimáticos/química , Inibidores Enzimáticos/farmacologia , Proteínas Serina-Treonina Quinases/antagonistas & inibidores , Receptores de Fatores de Crescimento Transformadores beta/antagonistas & inibidores , Animais , Benzotiazóis/química , Feminino , Imidazóis/química , Imidazóis/farmacologia , Camundongos , Camundongos Endogâmicos C57BL , Fosforilação/efeitos dos fármacos , Proteínas Serina-Treonina Quinases/metabolismo , Receptor do Fator de Crescimento Transformador beta Tipo I , Receptores de Fatores de Crescimento Transformadores beta/metabolismo , Proteína Smad2/antagonistas & inibidores , Proteína Smad2/metabolismo , Proteína Smad3/antagonistas & inibidores , Proteína Smad3/metabolismo , Fator de Crescimento Transformador beta/antagonistas & inibidores , Fator de Crescimento Transformador beta/metabolismoRESUMO
Sphingosine 1-phosphate receptor type 1 (S1P(1)) was shown to be essential for vascular maturation during embryonic development and it has been demonstrated that substantial crosstalk exists between S1P(1) and other pro-angiogenic growth factors, such as vascular endothelial growth factor (VEGF) and basic fibroblast growth factor. We developed a novel S1P(1)-selective antagonist, TASP0277308, which is structurally unrelated to S1P as well as previously described S1P(1) antagonists. TASP0277308 inhibited S1P- as well as VEGF-induced cellular responses, including migration and proliferation of human umbilical vein endothelial cells. Furthermore, TASP0277308 effectively blocked a VEGF-induced tube formation in vitro and significantly suppressed tumor cell-induced angiogenesis in vivo. These findings revealed that S1P(1) is a critical component of VEGF-related angiogenic responses and also provide evidence for the efficacy of TASP0277308 for anti-cancer therapies.
Assuntos
Neovascularização Fisiológica/efeitos dos fármacos , Receptores de Lisoesfingolipídeo/antagonistas & inibidores , Sulfonas/farmacologia , Triazóis/farmacologia , Linhagem Celular Tumoral , Células Endoteliais da Veia Umbilical Humana/efeitos dos fármacos , Células Endoteliais da Veia Umbilical Humana/fisiologia , Humanos , Lisofosfolipídeos/metabolismo , Lisofosfolipídeos/farmacologia , Neoplasias/irrigação sanguínea , Neovascularização Patológica/metabolismo , Receptores de Lisoesfingolipídeo/metabolismo , Esfingosina/análogos & derivados , Esfingosina/metabolismo , Esfingosina/farmacologia , Receptores de Esfingosina-1-Fosfato , Fator A de Crescimento do Endotélio Vascular/farmacologiaRESUMO
We recently demonstrated efficient antitumor immunity against murine tumors using dendritic cells (DCs) activated by recombinant Sendai viruses (rSeVs), and proposed a new concept, "immunostimulatory virotherapy," for cancer immunotherapy. However, there has been little information on the efficacy of this method in preventing metastatic diseases. In this study, we investigated the efficacy of vaccinating DCs activated by fusion gene-deleted nontransmissible rSeV (rSeV/dF) using a murine model of lung metastasis. Bolus and i.v. administration of DCs harboring rSeV/dF-expressing GFP without pulsation of tumor Ag (DC-rSeV/dF-GFP) 2 days before tumor inoculation showed efficient prevention against lung metastasis of c1300 neuroblastoma, but not of RM-9 prostatic cancer. We found that the timing of DC therapy was critical for the inhibition of pulmonary metastasis of RM-9, and that the optimal effect of DCs was seen 28 days before tumor inoculation. Interestingly, the antimetastatic effect was sustained for over 3 mo, even when administered DCs were already cleared from the lung and organs related to the immune system. Although NK cell activity had already declined to baseline at the time of tumor inoculation, Ab-mediated depletion studies revealed that CD4+ cells as well as the presence of, but not the activation of, NK cells were crucial to the prevention of lung metastasis. These results are the first demonstration of efficient inhibition of lung metastasis via bolus administration of virally activated DCs that was sustained and NK/CD4+ cell-dependent, and may suggest a potentially new mechanism of DC-based immunotherapy for advanced malignancies.
Assuntos
Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD4-Positivos/patologia , Células Dendríticas/imunologia , Células Matadoras Naturais/imunologia , Células Matadoras Naturais/patologia , Neoplasias Pulmonares/prevenção & controle , Neoplasias Pulmonares/secundário , Vírus Sendai/imunologia , Animais , Proliferação de Células , Citotoxicidade Imunológica/genética , Células Dendríticas/virologia , Neoplasias Pulmonares/patologia , Linfonodos/imunologia , Linfonodos/patologia , Masculino , Camundongos , Camundongos Endogâmicos A , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Nus , Camundongos Transgênicos , Neuroblastoma/imunologia , Terapia Viral Oncolítica , Neoplasias da Próstata/imunologia , Neoplasias da Próstata/virologia , Vírus Sendai/genética , Fatores de Tempo , Vacinas Sintéticas/genética , Vacinas Sintéticas/imunologiaRESUMO
Glioblastoma multiforme (GM), the most frequent primary malignant brain tumor, is highly invasive due to the expression of proteases, including urokinase-type plasminogen activator (uPA). Here, we show the potential of our new and powerful recombinant Sendai virus (rSeV) showing uPA-specific cell-to-cell fusion activity [rSeV/dMFct14 (uPA2), named "BioKnife"] for GM treatment, an effect that was synergistically enhanced by arming BioKnife with the interferon-ß (IFN-ß) gene. BioKnife killed human GM cell lines efficiently in a uPA-dependent fashion, and this killing was prevented by PA inhibitor-1. Rat gliosarcoma 9L cells expressing both uPA and its functional receptor uPAR (9L-L/R) exhibited high uPA activity on the cellular surface and were highly susceptible to BioKnife. Although parent 9L cells (9L-P) were resistant to BioKnife and to BioKnife expressing IFN-ß (BioKnife-IFNß), cell-cell fusion of 9L-L/R strongly facilitated the expression of IFN-ß, and in turn, IFN-ß significantly accelerated the fusion activity of BioKnife. A similar synergy was seen in a rat orthotopic brain GM model with 9L-L/R in vivo; therefore, these results suggest that BioKnife-IFNß may have significant potential to improve the survival of GM patients in a clinical setting.
Assuntos
Glioblastoma/terapia , Interferon beta/metabolismo , Vírus Oncolíticos/fisiologia , Vírus Sendai/fisiologia , Ativador de Plasminogênio Tipo Uroquinase/metabolismo , Animais , Western Blotting , Linhagem Celular , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/genética , Ensaio de Imunoadsorção Enzimática , Feminino , Glioblastoma/genética , Glioblastoma/metabolismo , Humanos , Interferon beta/genética , Imageamento por Ressonância Magnética , Camundongos , Camundongos Nus , Vírus Oncolíticos/genética , Inibidor 1 de Ativador de Plasminogênio/farmacologia , Ratos , Ratos Endogâmicos F344 , Receptores de Ativador de Plasminogênio Tipo Uroquinase/genética , Receptores de Ativador de Plasminogênio Tipo Uroquinase/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Vírus Sendai/genética , Ativador de Plasminogênio Tipo Uroquinase/genéticaRESUMO
Gene therapy for cystic fibrosis (CF) is making encouraging progress into clinical trials. However, further improvements in transduction efficiency are desired. To develop a novel gene transfer vector that is improved and truly effective for CF gene therapy, a simian immunodeficiency virus (SIV) was pseudotyped with envelope proteins from Sendai virus (SeV), which is known to efficiently transduce unconditioned airway epithelial cells from the apical side. This novel vector was evaluated in mice in vivo and in vitro directed toward CF gene therapy. Here, we show that (i) we can produce relevant titers of an SIV vector pseudotyped with SeV envelope proteins for in vivo use, (ii) this vector can transduce the respiratory epithelium of the murine nose in vivo at levels that may be relevant for clinical benefit in CF, (iii) this can be achieved in a single formulation, and without the need for preconditioning, (iv) expression can last for 15 months, (v) readministration is feasible, (vi) the vector can transduce human air-liquid interface (ALI) cultures, and (vii) functional CF transmembrane conductance regulator (CFTR) chloride channels can be generated in vitro. Our data suggest that this lentiviral vector may provide a step change in airway transduction efficiency relevant to a clinical programme of gene therapy for CF.
Assuntos
Fibrose Cística/terapia , Terapia Genética , Vetores Genéticos , Lentivirus/genética , Vírus Sendai/genética , Proteínas do Envelope Viral/genética , Animais , Diferenciação Celular , Linhagem Celular , Feminino , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Transdução GenéticaRESUMO
Dendritic cells (DCs) have a crucial role to play in fighting nonself organisms and cells, including tumors. Clinically, numerous DC vaccinations have been attempted for cancer immunotherapy since the first trial, published in 1995, but with limited success. We found that Sendai virus (SeV) vector infection induces maturation of DCs and produces more powerful antitumor immunity against DCs in mouse models. We used a SeV vector as an immune booster for tumors and believe that this novel therapy, designated as "immunostimulatory virotherapy," will offer potent treatment for tumors.
Assuntos
Imunoterapia , Neoplasias/imunologia , Neoplasias/terapia , Vírus Sendai/genética , Animais , Linhagem Celular Tumoral , Separação Celular , Citometria de Fluxo , Vetores Genéticos/genética , Camundongos , Transplante de Neoplasias , Neoplasias/genética , Neoplasias/patologiaRESUMO
BACKGROUND: This study was designed to assess the therapeutic effect of urokinase-targeted recombinant oncolytic Sendai virus, termed "BioKnife," on anaplastic thyroid carcinoma (ATC). METHODS: Urokinase activity was investigated in human ATC cell lines, and BioKnife cytotoxicity against the cell lines was evaluated in vitro. Orthotopic mouse models of ATC were treated with three intratumoral injections of BioKnife, control virus, or phosphate-buffered saline (PBS) and were observed daily until >20% weight loss occurred. RESULTS: All three ATC cell lines showed a high level of urokinase activity. BioKnife induced urokinase-dependent cell fusion and cytotoxicity in all cell lines. Orthotopic models treated with BioKnife showed significantly prolonged survival compared with models treated with control virus or PBS (BioKnife 41.6 ± 15.0, control virus 17.0 ± 2.9, PBS 17.7 ± 6.3 days). CONCLUSIONS: BioKnife exerted therapeutic effects in orthotopic ATC mouse models. Thus, BioKnife represents a possible treatment option for ATC.
Assuntos
Terapia Viral Oncolítica , Vírus Sendai/fisiologia , Carcinoma Anaplásico da Tireoide/terapia , Neoplasias da Glândula Tireoide/terapia , Ativador de Plasminogênio Tipo Uroquinase/metabolismo , Animais , Apoptose , Fusão Celular , Linhagem Celular Tumoral , Modelos Animais de Doenças , Células Gigantes/metabolismo , Camundongos Endogâmicos BALB C , Carcinoma Anaplásico da Tireoide/metabolismo , Neoplasias da Glândula Tireoide/metabolismo , Ensaios Antitumorais Modelo de XenoenxertoRESUMO
In clinical N0 (cN0) cases with head and neck squamous cell carcinoma (HNSCC), a treatment selection is still controversial: elective neck dissection or watchful waiting. We focused on sentinel lymph node (SLN)-targeted therapy using the urokinase-type plasminogen activator (uPA)-dependent oncolytic Sendai virus "BioKnife." The objectives of this study were to investigate BioKnife migration into SLNs and elucidate its antitumor effect on lymph node metastases (LNM). We established an orthotopic nude mouse model of HNSCC, with LNM being frequently induced. We inoculated HSC-3-M3, human highly metastatic tongue squamous cell carcinoma cells, in the tongue of the nude mice, and after 2 weeks, we injected BioKnife into the primary tumor. We tracked BioKnife migration into the SLNs by immunostaining, RT-PCR, and an in vivo imaging system. We also examined its antitumor effects and mechanisms through serial section analysis of lymph nodes. GFP reporter expression was clearly visible in the lymph nodes of virus groups, which corresponded to SLNs. Relative GFP mRNA was significantly increased in both the tongues and lymph nodes in the virus groups compared with that in the control group (P < 0.05). Serial section analysis showed that BioKnife infected cancer cells and exhibited significant antitumor effect against LNM compared with the control groups (P < 0.05). We detected apoptosis in LNM infected by BioKnife. BioKnife migrated into SLNs after its injection into the primary tumor and effectively suppressed LNM, suggesting that SLN-targeted therapy using BioKnife has great potential to provide a novel and promising alternative to elective neck dissection in cN0 patients with HNSCC.
Assuntos
Terapia Viral Oncolítica , Vírus Oncolíticos , Vírus Sendai , Linfonodo Sentinela/patologia , Carcinoma de Células Escamosas de Cabeça e Pescoço/patologia , Carcinoma de Células Escamosas de Cabeça e Pescoço/terapia , Animais , Apoptose/efeitos dos fármacos , Linhagem Celular Tumoral , Modelos Animais de Doenças , Humanos , Metástase Linfática , Camundongos , Micrometástase de Neoplasia , Vírus Oncolíticos/genética , Vírus Sendai/genética , Linfonodo Sentinela/metabolismo , Carcinoma de Células Escamosas de Cabeça e Pescoço/diagnóstico por imagem , Ensaios Antitumorais Modelo de XenoenxertoRESUMO
BACKGROUND: The objectives of this study were to demonstrate anti-metastatic effect of BioKnife, uPA activity-dependent oncolytic Sendai virus, after BioKnife treatment for primary tumor, and analyze its mechanisms in a simulated metastasis mouse model of head and neck squamous cell carcinoma (HNSCC). METHODS: We established a simulated metastasis mouse model using a murine HNSCC cell line "SCCVII." We assessed a tumor size and an induction of tumor-specific immunoresponses using cytotoxic T-lymphocyte (CTL) assay, flow cytometry (FCM) in spleen and immunohistochemistry (IHC) in secondary tumor. RESULTS: Secondary tumors were significantly smaller in BioKnife-treated group. CTL activities were significantly improved in BioKnife group. FCM revealed that induction of dendritic cells and CD4+ /CD8+ lymphocytes was significantly higher in BioKnife group. IHC showed that CD8+ lymphocytes invaded secondary tumor. CONCLUSION: Tumor-specific immunoresponses induced by BioKnife has great potential to be a novel, safe, and less invasive option for control and prevention of metastasis.
Assuntos
Carcinoma de Células Escamosas/imunologia , Carcinoma de Células Escamosas/terapia , Neoplasias de Cabeça e Pescoço/imunologia , Neoplasias de Cabeça e Pescoço/terapia , Terapia Viral Oncolítica , Vírus Oncolíticos/fisiologia , Vírus Sendai/fisiologia , Animais , Linfócitos T CD4-Positivos/metabolismo , Linfócitos T CD8-Positivos/metabolismo , Linhagem Celular Tumoral , Células Dendríticas/metabolismo , Modelos Animais de Doenças , Imunocompetência , Camundongos Endogâmicos C3H , Ativador de Plasminogênio Tipo Uroquinase , Ensaios Antitumorais Modelo de XenoenxertoRESUMO
BACKGROUND: We previously demonstrated that a new lentiviral vector derived from nonpathogenic simian immunodeficiency virus (SIVagm) was efficient and safe for long-lasting retinal gene transfer, and that it provided the significant therapeutic effect of expressing human pigment epithelium-derived factor (hPEDF) in Royal College of Surgeons (RCS) rats. In the present study, to obtain a more pronounced outcome, we assessed the potential synergistic effect of the simultaneous gene transfer of hPEDF and human fibroblast growth factor-2 (hFGF-2) by improved third-generation SIV on RCS rats and retinal degeneration slow (rds) mice, because the former targets the primary neurons, including photoreceptor cells (PCs), whereas the latter is effective for targeting secondary neural cells, including Muller cells. METHODS: Vector solution (SIV-hPEDF, SIV-hFGF-2, a 1 : 1 mixture of SIV-hPEDF and SIV-hFGF-2, or SIV-enhanced green fluorescent protein) was injected into the peripheral subretinal space of 3-week-old RCS rats or rds mice. Histopathological and electroretinographic assessments were made at several points after gene transfer. RESULTS: Administration of SIV-hPEDF or SIV-hFGF-2 significantly delayed the histological PC degeneration and electrical deficit in RCS rats, and these delays were synergistically and significantly pronounced by SIV-hPEDF + SIV-hFGF-2 (1 : 1 mixture). In rds mice, functional therapeutic effects were observed even by SIV-PEDF, or SIV-FGF-2 alone and, moreover, both SIV-PEDF and SIV-FGF-2 showed higher therapeutic effects. CONCLUSIONS: These synergistic rescues of retinitis pigmentosa (RP) model animals are the 'proof concept' that the 'dual' expression of hPEDF and hFGF-2 dramatically improved therapeutic efficacy by keeping lower titers. This strategy may contribute to safer and more effective gene therapy for RP.
Assuntos
Proteínas do Olho/genética , Fator 2 de Crescimento de Fibroblastos/genética , Fatores de Crescimento Neural/genética , Retinose Pigmentar/terapia , Serpinas/genética , Vírus da Imunodeficiência Símia/genética , Animais , Modelos Animais de Doenças , Eletrorretinografia , Proteínas do Olho/uso terapêutico , Fator 2 de Crescimento de Fibroblastos/uso terapêutico , Vetores Genéticos , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Humanos , Camundongos , Fatores de Crescimento Neural/uso terapêutico , Ratos , Serpinas/uso terapêutico , TransgenesRESUMO
The utility of recombinant Sendai virus (rSeV) has been considerably examined over the last decade as a potent gene transfer candidate in a cytoplasmic gene expression system. Such risks as excessive immune responses associated with this virus administration in vivo however have limited its applicability in clinical settings as is the case with other viral vectors including adenoviruses. In consequence of extensive assessment on the mechanisms of immune responses against SeV, we found that ex vivo infection of immature dendritic cells (DCs) with SeV demonstrates their spontaneous maturation and activation. We applied this result to create a unique, representative, and powerful agent to activate DCs, namely rSeV-modified DCs (rSeV/DCs), for use in cancer immunotherapy. Use of this system in vivo resulted in the induction of efficient antitumor immunity against vascularized rodent tumors, including melanoma, hepatocellular carcinoma, neuroblastoma, squamous cell carcinoma, and prostatic cancer, and it even frequently associated with elimination of those tumors. These results indicate that rSeV could be a powerful immune booster for DC-based cancer immunotherapy that is worth investigating further. We propose a conceptual term "immunostimulatory virotherapy" to describe this new method of cancer therapy using the rSeV/DCs system.
Assuntos
Terapia Genética/instrumentação , Terapia Genética/métodos , Imunoterapia/métodos , Neoplasias/metabolismo , Neoplasias/terapia , Vírus Sendai/genética , Animais , Células Dendríticas/metabolismo , Técnicas de Transferência de Genes , Vetores Genéticos , Humanos , Modelos Genéticos , Mutação , Tretinoína/metabolismoRESUMO
The utility of recombinant Sendai virus (rSeV) has been considerably examined over the last decade as a potent gene transfer candidate in a cytoplasmic gene expression system. Such risks as excessive immune responses associated with this virus administration in vivo however have limited its applicability in clinical settings as is the case with other viral vectors including adenoviruses. In consequence of extensive assessment on the mechanisms of immune responses against SeV, we found that ex vivo infection of immature dendritic cells (DCs) with SeV demonstrates their spontaneous maturation and activation. We applied this result to create a unique, representative, and powerful agent to activate DCs, namely rSeV-modified DCs (rSeV/DCs), for use in cancer immunotherapy. Use of this system in vivo resulted in the induction of efficient antitumor immunity against vascularized rodent tumors, including melanoma, hepatocellular carcinoma, neuroblastoma, squamous cell carcinoma, and prostatic cancer, and it even frequently associated with elimination of those tumors. These results indicate that rSeV could be a powerful immune booster for DC-based cancer immunotherapy that is worth investigating further. We propose a conceptual term "immunostimulatory virotherapy" to describe this new method of cancer therapy using the rSeV/DCs system.
Assuntos
Terapia Genética/métodos , Imunoterapia/métodos , Neoplasias/imunologia , Vírus Sendai/imunologia , Células Dendríticas/imunologia , Humanos , Imunização/métodos , Vírus Sendai/genéticaRESUMO
BACKGROUND: Androgenic alopecia (AGA) occurs as a result of the contraction of the anagen phase because of the action of androgens on hair follicles. TGF-ß production from dermal papillae is enhanced by androgens, and growth inhibition of hair-follicle cells is induced by TGF-ß, and the hair cycle progresses from the anagen phase to the catagen phase. We investigated both the in vitro and in vivo potency of the newly identified ALK5 inhibitor TP0427736 {6-[4-(4-methyl-1,3-thiazol-2-yl)-1H-imidazol-5-yl]-1,3-benzothiazole}. METHODS: For in vitro study, kinase inhibitory activity was evaluated with ELISA, and inhibitory activity against TGF-ß-induced Smad2/3 phosphorylation in A549 cells and TGF-ß-induced growth inhibition of human outer root sheath cells were assayed using ELISA. For in vivo study, we used a mouse model that had been synchronized through dorsal hair depilation. RESULTS: TP0427736 inhibited ALK5 kinase activity with an IC50 of 2.72nM; this effect was 300-fold higher than the inhibitory effect on ALK3. In cell-based assays, TP0427736 inhibited Smad2/3 phosphorylation in A549 cells and decreased the growth inhibition of human outer root sheath cells. The topical application of TP0427736 significantly decreased Smad2 phosphorylation in mouse skin, and its repeated application suppressed the shortening of average hair follicle length during the transition from the late anagen phase to the catagen phase. CONCLUSIONS: TP0427736, a potent ALK5 inhibitor with appropriate in vitro and in vivo profiles, may serve as a potential new therapy for AGA.ã.
Assuntos
Alopecia/tratamento farmacológico , Benzotiazóis/farmacologia , Folículo Piloso/efeitos dos fármacos , Imidazóis/farmacologia , Proteínas Serina-Treonina Quinases/antagonistas & inibidores , Receptores de Fatores de Crescimento Transformadores beta/antagonistas & inibidores , Administração Tópica , Alopecia/patologia , Animais , Benzotiazóis/administração & dosagem , Receptores de Proteínas Morfogenéticas Ósseas Tipo I/antagonistas & inibidores , Ensaio de Imunoadsorção Enzimática , Humanos , Imidazóis/administração & dosagem , Concentração Inibidora 50 , Camundongos , Fosforilação , Inibidores de Proteínas Quinases/administração & dosagem , Inibidores de Proteínas Quinases/farmacologia , Receptor do Fator de Crescimento Transformador beta Tipo I , Proteína Smad2/metabolismo , Proteína Smad3/metabolismo , Fator de Crescimento Transformador beta/metabolismoRESUMO
OBJECTIVE: To assess the expression and distribution of a neurotrophic/antiangiogenic factor, pigment epithelium-derived factor (PEDF), related to angiogenesis that is a possibly key event during atherogenesis in human atherosclerotic plaques. METHODS AND RESULTS: Twenty fresh aortic samples were used for reverse-transcription polymerase chain reaction (RT-PCR), Western blot, and immunohistochemistry (IHC). In addition, 80 stocked paraffin blocks of coronary arteries from 40 autopsy cases were also used. IHC revealed divergent staining patterns for PEDF in both the aortas and the coronary arteries tested, ie, "cytoplasmic staining" or "extracellular deposition," were observed, respectively. In the areas showing cytoplasmic staining, double PEDF was expressed in a majority of the foamy macrophages and in some smooth muscle cells, and the PEDF-positive cell frequency was positively correlated with that of microvessels in a cell-rich area in the coronary arteries (P<0.0001). Inversely, extracellular deposition of PEDF was seen in acellular areas and was negatively correlated with the number of microvessels (P=0.0003). CONCLUSIONS: These results suggest that PEDF may function as an antiangiogenic factor when it is deposited onto the extracellular matrix. Thus, PEDF may play a significant role in determining the balance of angiogenesis/ antiangiogenesis during atherogenesis.
Assuntos
Doença da Artéria Coronariana/metabolismo , Doença da Artéria Coronariana/patologia , Proteínas do Olho/genética , Proteínas do Olho/metabolismo , Fatores de Crescimento Neural/genética , Fatores de Crescimento Neural/metabolismo , Serpinas/genética , Serpinas/metabolismo , Adulto , Idoso , Idoso de 80 Anos ou mais , Aorta/metabolismo , Aorta/patologia , Doença da Artéria Coronariana/imunologia , Vasos Coronários/metabolismo , Vasos Coronários/patologia , Citoplasma/metabolismo , Espaço Extracelular/metabolismo , Feminino , Células Espumosas/patologia , Humanos , Recém-Nascido , Macrófagos/patologia , Masculino , Pessoa de Meia-Idade , Neovascularização Patológica/imunologia , Neovascularização Patológica/metabolismo , Neovascularização Patológica/patologia , RNA Mensageiro/análise , Retina/metabolismo , Túnica Íntima/imunologia , Túnica Íntima/metabolismo , Túnica Íntima/patologiaRESUMO
Genes and proteins of human origin are often administered to monkeys for research purposes, however, it can be difficult to obtain sufficient levels of the products in vivo due to immunological clearance. In this study, we showed that human erythropoietin (hEPO) induces generation of anti-hEPO antibody in cynomolgus macaques (n=2), although 92% of amino acid residues are common between the human and macaque EPO. The administered hEPO was thus eliminated from the animals. On the other hand, when an immunosuppressant, cyclosporin A (CyA), was administered (6 mg/kg) intramuscularly every other day in combination with hEPO (n=2), no anti-hEPO antibody was generated and high serum levels of hEPO were obtained during administration of hEPO, resulting in an increase in serum hemoglobin levels. No adverse effects associated with CyA were observed. Thus, CyA treatment is useful for prevention of immune responses associated with the administration of human proteins in monkeys.
Assuntos
Anticorpos/sangue , Ciclosporina/farmacologia , Eritropoetina/imunologia , Imunossupressores/farmacologia , Macaca fascicularis/imunologia , Animais , Formação de Anticorpos/efeitos dos fármacos , Feminino , Hemoglobinas/análise , Humanos , Hipersensibilidade Imediata/prevenção & controle , Hipersensibilidade Imediata/veterinária , Macaca fascicularis/sangue , MasculinoRESUMO
Direct reprogramming is a promising, simple and low-cost approach to generate target cells from somatic cells without using induced pluripotent stem cells. Recently, peripheral blood mononuclear cells (PBMCs) have attracted considerable attention as a somatic cell source for reprogramming. As a cell source, PBMCs have an advantage over dermal fibroblasts with respect to the ease of collecting tissues. Based on our studies involving generation of photosensitive photoreceptor cells from human iris cells and human dermal fibroblasts by transduction of photoreceptor-related transcription factors via retrovirus vectors, we transduced these transcription factors into PBMCs via Sendai virus vectors. We found that retinal disease-related genes were efficiently detected in CRX-transduced cells, most of which are crucial to photoreceptor functions. In functional studies, a light-induced inward current was detected in some CRX-transduced cells. Moreover, by modification of the culture conditions including additional transduction of RAX1 and NEUROD1, we found a greater variety of retinal disease-related genes than that observed in CRX-transduced PBMCs. These data suggest that CRX acts as a master control gene for reprogramming PBMCs into photoreceptor-like cells and that our induced photoreceptor-like cells might contribute to individualized drug screening and disease modeling of inherited retinal degeneration.
RESUMO
Vascular endothelial growth factor (VEGF) plays key roles in tumor angiogenesis. Therefore, VEGF and its receptors are considered to be primary targets for antiangiogenic strategy during cancer chemotherapy. Our previous study reported that VGA1155, a low-molecular-weight inhibitor of the binding of VEGF, inhibited VEGF binding to KDR/Flk-1 receptor-overexpressing cells. In the present study, the antitumor effects and antimetastatic effect of VGA1155 were examined in vivo. VGA1155 suppressed the growth of human lung, breast, colon and epidermoid cancers (LC-6, HT29, MX-1, Col-1 and A431) in the nude mouse xenograft model, and pulmonary metastasis of melanoma in the spontaneous metastasis model. These results suggest that VGA1155 has antitumor effects in vivo through the inhibition of VEGF binding to its receptors.
Assuntos
Benzoatos/farmacologia , Receptores de Fatores de Crescimento do Endotélio Vascular/antagonistas & inibidores , Fator A de Crescimento do Endotélio Vascular/antagonistas & inibidores , Animais , Antineoplásicos/farmacologia , Neoplasias da Mama/irrigação sanguínea , Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/patologia , Carcinoma Pulmonar de Células não Pequenas/irrigação sanguínea , Carcinoma Pulmonar de Células não Pequenas/tratamento farmacológico , Carcinoma Pulmonar de Células não Pequenas/patologia , Carcinoma de Células Escamosas/irrigação sanguínea , Carcinoma de Células Escamosas/tratamento farmacológico , Carcinoma de Células Escamosas/patologia , Processos de Crescimento Celular/efeitos dos fármacos , Neoplasias do Colo/irrigação sanguínea , Neoplasias do Colo/tratamento farmacológico , Neoplasias do Colo/patologia , Humanos , Neoplasias Pulmonares/irrigação sanguínea , Neoplasias Pulmonares/tratamento farmacológico , Neoplasias Pulmonares/patologia , Melanoma Experimental/irrigação sanguínea , Melanoma Experimental/tratamento farmacológico , Melanoma Experimental/patologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Neovascularização Patológica/tratamento farmacológico , Receptores de Fatores de Crescimento do Endotélio Vascular/metabolismo , Fator A de Crescimento do Endotélio Vascular/metabolismo , Ensaios Antitumorais Modelo de XenoenxertoRESUMO
During the past several years, many articles have described how human embryonic stem (ES) cells and adult hematopoietic stem cells (HSCs) can differentiate into cardiac muscle, blood vessels, and various other types of cells. The articles raised the expectation that these stem cells may become useful for the treatment of a variety of diseases, including cardiovascular diseases. Genetic manipulation of ES cells and HSCs would be important for such future applications of the cells. Until now, retroviral vectors have been used primarily for stable expression of transgenes in murine ES cells and HSCs. Because murine models may not predict reliably the biology of ES cells and HSCs in humans, we have utilized primate ES cells and HSCs as targets of gene transfer. We have shown that primate ES cells and HSCs can be transduced efficiently with lentiviral vectors derived from the simian immunodeficiency virus, and that the high transgene expression persists without transcriptional silencing. This highly efficient gene transfer method allows for safe and faithful gene delivery to primate ES cells and HSCs to test potential research and therapeutic applications.
Assuntos
Embrião de Mamíferos/citologia , Terapia Genética , Vetores Genéticos/genética , Células-Tronco Hematopoéticas/citologia , Vírus da Imunodeficiência Símia/genética , Animais , Doenças Cardiovasculares/genética , Doenças Cardiovasculares/terapia , Diferenciação Celular/genética , Técnicas de Transferência de Genes , HumanosRESUMO
Vascular endothelial growth factor (VEGF) plays a pivotal role in the processes of angiogenesis, which is essential for the growth of solid tumors and their metastasis. Because VEGF is a critical factor in tumor survival, inhibiting VEGF would provide significant benefits in tumor therapy. To identify a compound that inhibits the binding of VEGF to its receptor, we used a high throughput screening method, finding that small molecular compounds inhibited VEGF binding. Among active compounds, 5-[N-methyl-N-(4-octadecyloxyphenyl)acetyl]amino-2-methylthiobenzoic acid (VGA1155) was selected for its potent inhibition of binding. VGA1155 inhibited [(125)I] VEGF binding to two cell lines, NIH3T3-fms-like tyrosine kinase-1 (VEGF receptor 1 transfected) cells and NIH3T3-kinase insert domain containing receptor/fetal liver kinase-1 (KDR/Flk-1; VEGF receptor 2 transfected), in a concentration-dependent manner. VGA1155 did not inhibit the binding of several other growth factors or cytokines to their receptors. Based on the results of surface plasmon resonance analysis using Biacore S51 system, it appears that this binding inhibitory property may be based on the association of VGA1155 with VEGF receptor 2 (KDR/Flk-1). Further, the interference in VEGF binding by VGA1155 in turn induces the inhibition of VEGF-induced KDR/Flk-1 autophosphorylation. VGA1155 also reduced intradermal VEGF-induced vascular permeability in guinea pigs. These findings indicate that VGA1155 inhibits not only VEGF binding to its receptors through association with KDR/Flk-1 but also VEGF function in vivo. These VGA1155 activities may provide a useful basis for the development of antiangiogenic and antitumor agents.