Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 97
Filtrar
Más filtros

Bases de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Int J Mol Sci ; 25(3)2024 Jan 30.
Artículo en Inglés | MEDLINE | ID: mdl-38338982

RESUMEN

We had previously investigated the expression and functional role of C-X-C Motif Chemokine Ligand 12 (CXCL12) during the hair cycle progression. CXCL12 was highly expressed in stromal cells such as dermal fibroblasts (DFs) and inhibition of CXCL12 increased hair growth. Therefore, we further investigated whether a CXCL12 neutralizing antibody (αCXCL12) is effective for androgenic alopecia (AGA) and alopecia areata (AA) and studied the underlying molecular mechanism for treating these diseases. In the AGA model, CXCL12 is highly expressed in DFs. Subcutaneous (s.c.) injection of αCXCL12 significantly induced hair growth in AGA mice, and treatment with αCXCL12 attenuated the androgen-induced hair damage in hair organ culture. Androgens increased the secretion of CXCL12 from DFs through the androgen receptor (AR). Secreted CXCL12 from DFs increased the expression of the AR and C-X-C Motif Chemokine Receptor 4 (CXCR4) in dermal papilla cells (DPCs), which induced hair loss in AGA. Likewise, CXCL12 expression is increased in AA mice, while s.c. injection of αCXCL12 significantly inhibited hair loss in AA mice and reduced the number of CD8+, MHC-I+, and MHC-II+ cells in the skin. In addition, injection of αCXCL12 also prevented the onset of AA and reduced the number of CD8+ cells. Interferon-γ (IFNγ) treatment increased the secretion of CXCL12 from DFs through the signal transducer and activator of transcription 3 (STAT3) pathway, and αCXCL12 treatment protected the hair follicle from IFNγ in hair organ culture. Collectively, these results indicate that CXCL12 is involved in the progression of AGA and AA and antibody therapy for CXCL12 is promising for hair loss treatment.


Asunto(s)
Alopecia Areata , Anticuerpos Neutralizantes , Animales , Ratones , Alopecia/metabolismo , Alopecia Areata/tratamiento farmacológico , Alopecia Areata/metabolismo , Andrógenos/metabolismo , Anticuerpos Neutralizantes/farmacología , Anticuerpos Neutralizantes/metabolismo , Cabello , Folículo Piloso/metabolismo , Piel/metabolismo , Quimiocina CXCL12/inmunología
2.
Mol Cell Biochem ; 478(4): 861-873, 2023 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-36107283

RESUMEN

Given that mast cells are pivotal contributors to allergic diseases, various allergy treatments have been developed to inhibit them. Omalizumab, an anti-immunoglobulin E antibody, is a representative therapy that can alleviate allergy symptoms by inhibiting mast cell degranulation. However, omalizumab cannot reduce the proliferation and accumulation of mast cells, which is a fundamental cause of allergic diseases. c-Kit is essential for the proliferation, survival, and differentiation of mast cells. Excessive c-Kit activation triggers various mast cell diseases, such as asthma, chronic spontaneous urticaria, and mastocytosis. Herein, we generated 2G4, an anti-c-Kit antibody, to develop a therapeutic agent for mast cell diseases. The therapeutic efficacy of 2G4 antibody was evaluated in LAD2, a human mast cell line. 2G4 antibody completely inhibited c-Kit signaling by blocking the binding of stem cell factor, known as the c-Kit ligand. Inhibition of c-Kit signaling led to the suppression of proliferation, migration, and degranulation in LAD2 cells. Moreover, 2G4 antibody suppressed the secretion of pro-inflammatory cytokines, including granulocyte-macrophage colony-stimulating factor, vascular endothelial growth factor, C-C motif chemokine ligand 2, brain-derived neurotrophic factor, and complement component C5/C5a, which can exacerbate allergy symptoms. Taken together, these results suggest that 2G4 antibody has potential as a novel therapeutic agent for mast cell diseases.


Asunto(s)
Hipersensibilidad , Trastornos de la Activación de los Mastocitos , Humanos , Mastocitos/metabolismo , Omalizumab/metabolismo , Factor A de Crecimiento Endotelial Vascular/metabolismo , Proteínas Tirosina Quinasas Receptoras/metabolismo , Proteínas Proto-Oncogénicas c-kit/metabolismo , Factor de Células Madre/metabolismo , Hipersensibilidad/metabolismo , Proliferación Celular , Degranulación de la Célula
3.
Int J Mol Sci ; 23(4)2022 Feb 18.
Artículo en Inglés | MEDLINE | ID: mdl-35216379

RESUMEN

Lung cancer is the leading cause of cancer-related deaths. Small cell lung cancer (SCLC) accounts for 15-25% of all lung cancers. It exhibits a rapid doubling time and a high degree of invasiveness. Additionally, overexpression of c-Kit occurs in 70% of SCLC patients. In this study, we evaluated an antibody-drug conjugate (ADC) that targets c-Kit, which is a potential therapeutic agent for SCLC. First, we generated and characterized 4C9, a fully human antibody that targets c-Kit and specifically binds to SCLC cells expressing c-Kit with a binding affinity of KD = 5.5 × 10-9 M. Then, we developed an ADC using DM1, a microtubule inhibitor, as a payload. 4C9-DM1 efficiently induced apoptosis in SCLC with an IC50 ranging from 158 pM to 4 nM. An in vivo assay using a xenograft mouse model revealed a tumor growth inhibition (TGI) rate of 45% (3 mg/kg) and 59% (5 mg/kg) for 4C9-DM1 alone. Combination treatment with 4C9-DM1 plus carboplatin/etoposide or lurbinectedin resulted in a TGI rate greater than 90% compared with the vehicle control. Taken together, these results indicate that 4C9-DM1 is a potential therapeutic agent for SCLC treatment.


Asunto(s)
Anticuerpos Monoclonales Humanizados/farmacología , Inmunoconjugados/farmacología , Neoplasias Pulmonares/tratamiento farmacológico , Carcinoma Pulmonar de Células Pequeñas/tratamiento farmacológico , Animales , Carboplatino/farmacología , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Etopósido/farmacología , Femenino , Humanos , Neoplasias Pulmonares/metabolismo , Maitansina/farmacología , Ratones , Proteínas Proto-Oncogénicas c-kit/metabolismo , Proteínas Tirosina Quinasas Receptoras/metabolismo , Receptor ErbB-2/metabolismo , Carcinoma Pulmonar de Células Pequeñas/metabolismo , Trastuzumab/farmacología , Moduladores de Tubulina/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto/métodos
4.
Int J Mol Sci ; 23(20)2022 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-36293219

RESUMEN

Ovarian cancer is the fifth leading cause of cancer, followed by front line is mostly platinum agents and PARP inhibitors, and very limited option in later lines. Therefore, there is a need for alternative therapeutic options. Nectin-2, which is overexpressed in ovarian cancer, is a known immune checkpoint that deregulates immune cell function. In this study, we generated a novel anti-nectin-2 antibody (chimeric 12G1, c12G1), and further characterized it using epitope mapping, enzyme-linked immunosorbent assay, surface plasmon resonance, fluorescence-activated cell sorting, and internalization assays. The c12G1 antibody specifically bound to the C2 domain of human nectin-2 with high affinity (KD = 2.90 × 10-10 M), but not to mouse nectin-2. We then generated an antibody-drug conjugate comprising the c12G1 antibody conjugated to DM1 and investigated its cytotoxic effects against cancer cells in vitro and in vivo. c12G1-DM1 induced cell cycle arrest at the mitotic phase in nectin-2-positive ovarian cancer cells, but not in nectin-2-negative cancer cells. c12G1-DM1 induced ~100-fold cytotoxicity in ovarian cancer cells, with an IC50 in the range of 0.1 nM~7.4 nM, compared to normal IgG-DM1. In addition, c12G1-DM1 showed ~91% tumor growth inhibition in mouse xenograft models transplanted with OV-90 cells. These results suggest that c12G1-DM1 could be used as a potential therapeutic agent against nectin-2-positive ovarian cancers.


Asunto(s)
Inmunoconjugados , Maitansina , Neoplasias Ováricas , Humanos , Ratones , Animales , Femenino , Inmunoconjugados/farmacología , Inmunoconjugados/uso terapéutico , Xenoinjertos , Inhibidores de Poli(ADP-Ribosa) Polimerasas/farmacología , Platino (Metal)/farmacología , Ensayos Antitumor por Modelo de Xenoinjerto , Línea Celular Tumoral , Proliferación Celular , Carcinoma Epitelial de Ovario/tratamiento farmacológico , Neoplasias Ováricas/patología , Inmunoglobulina G/farmacología , Inmunoglobulina G/uso terapéutico , Maitansina/uso terapéutico
5.
Arterioscler Thromb Vasc Biol ; 39(10): 2120-2131, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31434494

RESUMEN

OBJECTIVE: Aberrant neovascularization is a leading cause of blindness in several eye diseases, including age-related macular degeneration and proliferative diabetic retinopathy. The identification of key regulators of pathological ocular neovascularization has been a subject of extensive research and great therapeutic interest. Here, we explored the previously unrecognized role of cKIT and its ligand, SCF (stem cell factor), in the pathological ocular neovascularization process. Approach and Results: Compared with normoxia, hypoxia, a crucial driver of neovascularization, caused cKIT to be highly upregulated in endothelial cells, which significantly enhanced the angiogenic response of endothelial cells to SCF. In murine models of pathological ocular neovascularization, such as oxygen-induced retinopathy and laser-induced choroidal neovascularization models, cKIT and SCF expression was significantly increased in ocular tissues, and blockade of cKIT and SCF using cKit mutant mice and anti-SCF neutralizing IgG substantially suppressed pathological ocular neovascularization. Mechanistically, SCF/cKIT signaling induced neovascularization through phosphorylation of glycogen synthase kinase-3ß and enhancement of the nuclear translocation of ß-catenin and the transcription of ß-catenin target genes related to angiogenesis. Inhibition of ß-catenin-mediated transcription using chemical inhibitors blocked SCF-induced in vitro angiogenesis in hypoxia, and injection of a ß-catenin agonist into cKit mutant mice with oxygen-induced retinopathy significantly enhanced pathological neovascularization in the retina. Conclusions; Our data reveal that SCF and cKIT are promising novel therapeutic targets for treating vision-threatening ocular neovascular diseases.


Asunto(s)
Regulación de la Expresión Génica , Enfermedades de la Retina/genética , Enfermedades de la Retina/metabolismo , Neovascularización Retiniana/genética , Factor de Células Madre/genética , Factor A de Crecimiento Endotelial Vascular/metabolismo , Análisis de Varianza , Inhibidores de la Angiogénesis/farmacología , Animales , Células Cultivadas , Modelos Animales de Enfermedad , Células Endoteliales/metabolismo , Ensayo de Inmunoadsorción Enzimática , Femenino , Humanos , Hipoxia/complicaciones , Inmunohistoquímica , Masculino , Ratones , Ratones Endogámicos C57BL , Proteínas Proto-Oncogénicas c-kit/genética , Enfermedades de la Retina/patología , Enfermedades de la Retina/fisiopatología , Transducción de Señal/genética
6.
J Nanobiotechnology ; 17(1): 19, 2019 Jan 29.
Artículo en Inglés | MEDLINE | ID: mdl-30696428

RESUMEN

BACKGROUND: Protein-based Cas9 in vivo gene editing therapeutics have practical limitations owing to their instability and low efficacy. To overcome these obstacles and improve stability, we designed a nanocarrier primarily consisting of lecithin that can efficiently target liver disease and encapsulate complexes of Cas9 with a single-stranded guide RNA (sgRNA) ribonucleoprotein (Cas9-RNP) through polymer fusion self-assembly. RESULTS: In this study, we optimized an sgRNA sequence specifically for dipeptidyl peptidase-4 gene (DPP-4) to modulate the function of glucagon-like peptide 1. We then injected our nanocarrier Cas9-RNP complexes directly into type 2 diabetes mellitus (T2DM) db/db mice, which disrupted the expression of DPP-4 gene in T2DM mice with remarkable efficacy. The decline in DPP-4 enzyme activity was also accompanied by normalized blood glucose levels, insulin response, and reduced liver and kidney damage. These outcomes were found to be similar to those of sitagliptin, the current chemical DPP-4 inhibition therapy drug which requires recurrent doses. CONCLUSIONS: Our results demonstrate that a nano-liposomal carrier system with therapeutic Cas9-RNP has great potential as a platform to improve genomic editing therapies for human liver diseases.


Asunto(s)
Sistemas CRISPR-Cas , Diabetes Mellitus Tipo 2/terapia , Dipeptidil Peptidasa 4/genética , Sistemas de Liberación de Medicamentos , Terapia Genética/métodos , Lecitinas , Liposomas , Animales , Glucemia/efectos de los fármacos , Línea Celular , Dipeptidil Peptidasa 4/metabolismo , Edición Génica , Marcación de Gen , Péptido 1 Similar al Glucagón/sangre , Humanos , Lecitinas/administración & dosificación , Lecitinas/química , Liposomas/administración & dosificación , Liposomas/química , Ratones , Ratones Noqueados , ARN Guía de Kinetoplastida/administración & dosificación , ARN Guía de Kinetoplastida/química , ARN Guía de Kinetoplastida/genética
7.
Int J Mol Sci ; 20(7)2019 Apr 09.
Artículo en Inglés | MEDLINE | ID: mdl-30970566

RESUMEN

The mammalian mediator complex subunit 28 (MED28) is overexpressed in a variety of cancers and it regulates cell migration/invasion and epithelial-mesenchymal transition. However, transcription factors that increase MED28 expression have not yet been identified. In this study, we performed a luciferase reporter assay to identify and characterize the prospective transcription factors, namely E2F transcription factor 1, nuclear respiratory factor 1, E-26 transforming sequence 1, and CCAAT/enhancer-binding protein ß, which increased MED28 expression. In addition, the release from the arrest at the G1-S or G2-M phase transition after cell cycle synchronization using thymidine or nocodazole, respectively, showed enhanced MED28 expression at the G1-S transition and mitosis. Furthermore, the overexpression of MED28 significantly decreased the duration of interphase and mitosis. Conversely, a knockdown of MED28 using si-RNA increased the duration of interphase and mitosis. Of note, the overexpression of MED28 significantly increased micronucleus and nuclear budding in HeLa cells. In addition, flow cytometry and fluorescence microscopy analyses showed that the overexpression of MED28 significantly increased aneuploid cells. Taken together, these results suggest that MED28 expression is increased by oncogenic transcription factors and its overexpression disturbs the cell cycle, which results in genomic instability and aneuploidy.


Asunto(s)
Inestabilidad Genómica , Complejo Mediador/genética , Complejo Mediador/metabolismo , Factores de Transcripción/metabolismo , Aneuploidia , Ciclo Celular/efectos de los fármacos , Inestabilidad Genómica/efectos de los fármacos , Células HEK293 , Células HeLa , Humanos , Células MCF-7 , Nocodazol/farmacología , Regiones Promotoras Genéticas , Timidina/farmacología , Regulación hacia Arriba
8.
Clin Exp Rheumatol ; 36(4): 533-539, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29352840

RESUMEN

OBJECTIVES: Secreted aminoacyl-tRNA synthetase-interacting multifunctional protein-1 (AIMP1) has been reported to have pro-inflammatory properties. The aim of this study was to evaluate the clinical significance of serum AIMP1 in patients with systemic lupus erythematosus (SLE). METHODS: Serum levels of AIMP1 were measured in 160 patients with SLE using a human AIMP1 ELISA kit. Eighty patients were classified as active SLE (SLEDAI-2K ≥ 5), and 80 patients were classified as stable SLE. Correlation between serum AIMP1, SLE disease activity index-2000 (SLEDAI-2K), and laboratory variables related to disease activity or inflammatory burdens were assessed using Pearson's correlation analysis. The optimal cut-off value for serum AIMP1 to predict active SLE was estimated by using a receiver operator characteristic curve, and logistic regression analysis was used to compare the odds ratios (ORs) of laboratory variables in predicting active SLE. RESULTS: The median serum AIMP1 was higher in patients with active SLE than those with stable SLE (8.0 vs. 6.5 ng/ml, p<0.001). Serum AIMP1 demonstrated correlation with SLEDAI-2K and laboratory variables related to disease activity or inflammatory burdens. The optimal cut-off AIMP1 to predict active SLE was 10.09. Multivariate logistic regression analysis including conventional laboratory variables demonstrated that serum AIMP1 ≥10.09 ng/ml (OR 3.919, 95% confidence interval 1.223-12.564, p=0.022) was useful in predicting active SLE. CONCLUSIONS: Serum levels of AIMP1 were associated with disease activity of SLE and could predict active SLE based on SLEDAI-2K.


Asunto(s)
Citocinas/sangre , Lupus Eritematoso Sistémico/sangre , Proteínas de Neoplasias/sangre , Proteínas de Unión al ARN/sangre , Adulto , Biomarcadores , Femenino , Humanos , Modelos Logísticos , Lupus Eritematoso Sistémico/diagnóstico , Masculino , Persona de Mediana Edad , Índice de Severidad de la Enfermedad
9.
Arterioscler Thromb Vasc Biol ; 36(9): 1928-36, 2016 09.
Artículo en Inglés | MEDLINE | ID: mdl-27470512

RESUMEN

OBJECTIVE: Vascular smooth muscle cells (VSMCs) modulate their phenotype between synthetic and contractile states in response to environmental changes; this modulation plays a crucial role in the pathogenesis of restenosis and atherosclerosis. Here, we identified fibroblast growth factor 12 (FGF12) as a novel key regulator of the VSMC phenotype switch. APPROACH AND RESULTS: Using murine models and human specimens, we found that FGF12 was highly expressed in contractile VSMCs of normal vessel walls but was downregulated in synthetic VSMCs from injured and atherosclerotic vessels. In human VSMCs, FGF12 expression was inhibited at the transcriptional level by platelet-derived growth factor-BB. Gain- and loss-of-function experiments showed that FGF12 was both necessary and sufficient for inducing and maintaining the quiescent and contractile phenotypes of VSMCs. FGF12 inhibited cell proliferation through the p53 pathway and upregulated the key factors involved in VSMC lineage differentiation, such as myocardin and serum response factor. Such FGF12-induced phenotypic change was mediated by the p38 MAPK (mitogen-activated protein kinase) pathway. Moreover, FGF12 promoted the differentiation of mouse embryonic stem cells and the transdifferentiation of human dermal fibroblasts into SMC-like cells. Furthermore, adenoviral infection of FGF12 substantially decreased neointima hyperplasia in a rat carotid artery injury model. CONCLUSIONS: In general, FGF family members induce a synthetic VSMC phenotype. Interestingly, the present study showed the unanticipated finding that FGF12 belonging to FGF family, strongly induced the quiescent and contractile VSMC phenotypes and directly promoted VSMC lineage differentiation. These novel findings suggested that FGF12 could be a new therapeutic target for treating restenosis and atherosclerosis.


Asunto(s)
Enfermedades de las Arterias Carótidas/metabolismo , Traumatismos de las Arterias Carótidas/metabolismo , Diferenciación Celular , Plasticidad de la Célula , Factores de Crecimiento de Fibroblastos/metabolismo , Músculo Liso Vascular/metabolismo , Miocitos del Músculo Liso/metabolismo , Regiones no Traducidas 5' , Animales , Apolipoproteínas E/deficiencia , Apolipoproteínas E/genética , Becaplermina , Sitios de Unión , Enfermedades de las Arterias Carótidas/patología , Traumatismos de las Arterias Carótidas/genética , Traumatismos de las Arterias Carótidas/patología , Arteria Carótida Común/metabolismo , Arteria Carótida Común/patología , Diferenciación Celular/efectos de los fármacos , Linaje de la Célula , Plasticidad de la Célula/efectos de los fármacos , Proliferación Celular , Células Cultivadas , Modelos Animales de Enfermedad , Células Madre Embrionarias/metabolismo , Factores de Crecimiento de Fibroblastos/genética , Genotipo , Humanos , Hiperplasia , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Músculo Liso Vascular/efectos de los fármacos , Músculo Liso Vascular/patología , Miocitos del Músculo Liso/efectos de los fármacos , Miocitos del Músculo Liso/patología , Neointima , Fenotipo , Fosfatidilinositol 3-Quinasa/metabolismo , Unión Proteica , Proteínas Proto-Oncogénicas c-sis/farmacología , Interferencia de ARN , Ratas Sprague-Dawley , Transducción de Señal , Transcripción Genética , Transfección , Vasoconstricción , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
10.
Int J Mol Sci ; 18(8)2017 Aug 16.
Artículo en Inglés | MEDLINE | ID: mdl-28813021

RESUMEN

The multipotency and anti-inflammatory effects of mesenchymal stem cells (MSCs) make them attractive for cell therapy in regenerative medicine. A large number of MSCs is required for efficient therapy owing to the low homing efficiency of MSCs to target sites. Furthermore, owing to limitations in obtaining sufficient amounts of MSCs, in vitro expansion of MSCs that preserves their differentiation and proliferative potential is essential. The animal factor included in culture media also limits clinical application. In this study, adipose-derived MSCs showed a significantly higher proliferation rate in STK2, a chemically-defined medium, than in DMEM/FBS. The expression of MSC surface markers was increased in the culture using STK2 compared to that using DMEM/FBS. Tri-lineage differentiation analyses showed that MSCs cultured in STK2 were superior to those cultured in DMEM/FBS. In addition, MSCs cultured in STK2 showed a reduced senescence rate, small and homogenous cell size, and were more genetically stable compared to those cultured in DMEM/FBS. Furthermore, secretome analysis showed that the expression of factors related to proliferation/migration, anti-inflammation, and differentiation were increased in STK2 culture medium compared to DMEM/FBS. Taken together, these results suggest that culture using STK2 medium offers many advantages through which it is possible to obtain safer, superior, and larger numbers of MSCs.


Asunto(s)
Adipocitos/citología , Adipocitos/metabolismo , Técnicas de Cultivo de Célula/métodos , Medios de Cultivo/química , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/metabolismo , Humanos
11.
Biochem Biophys Res Commun ; 478(4): 1682-7, 2016 09 30.
Artículo en Inglés | MEDLINE | ID: mdl-27596970

RESUMEN

Tauroursodeoxycholic acid (TUDCA) is known to prevent apoptosis through the Bax pathway and to promote neovascularization by enhancing the mobilization of stem cells, their differentiation. This study was performed to investigate the effect of TUDCA on erythropoiesis in hematopoietic stem cells (HSCs). Since erythropoiesis of CD34(+) HSCs is divided into four phases, the total cell number, viable cell number, cell viability, cell morphology, and expressed erythroid markers in each phase were examined. The number of viable control cells and their viability did not differ from those of the TUDCA-treated cells in phase I and II. However, TUDCA increased cell viability compared to the control in phases III and IV. Cell distribution differed that the immature erythroid cell number was higher for the TUDCA-treated cells than for the control cells until phase III, but all developed into RBCs in the last. The final RBC number and viability was significantly higher in TUDCA-treated cells compared to the control cells. Taken together, we suggest that TUDCA addition to cell cultures for artificial RBC production could be used as a new protocol for improving the viability of RBCs.


Asunto(s)
Diferenciación Celular/efectos de los fármacos , Eritrocitos/efectos de los fármacos , Células Eritroides/efectos de los fármacos , Células Madre Hematopoyéticas/efectos de los fármacos , Ácido Tauroquenodesoxicólico/farmacología , Adulto , Anciano , Antígenos CD34/metabolismo , Biomarcadores/metabolismo , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Colagogos y Coleréticos/farmacología , Recuento de Eritrocitos , Eritrocitos/citología , Eritrocitos/metabolismo , Células Eritroides/citología , Células Eritroides/metabolismo , Eritropoyesis/efectos de los fármacos , Femenino , Citometría de Flujo , Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/metabolismo , Humanos , Inmunofenotipificación , Masculino , Persona de Mediana Edad , Factores de Tiempo
12.
Stem Cells ; 33(3): 792-805, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25407160

RESUMEN

Although serum bile acid concentrations are approximately 10 µM in healthy subjects, the crosstalk between the biliary system and vascular repair has never been investigated. In this study, tauroursodeoxycholic acid (TUDCA) induced dissociation of CD34(+) hematopoietic stem cells (HSCs) from stromal cells by reducing adhesion molecule expression. TUDCA increased CD34(+) /Sca1(+) progenitors in mice peripheral blood (PB), and CD34(+) , CD31(+) , and c-kit(+) progenitors in human PB. In addition, TUDCA increased differentiation of CD34(+) HSCs into EPC lineage cells via Akt activation. EPC invasion was increased by TUDCA, which was mediated by fibroblast activating protein via Akt activation. Interestingly, TUDCA induced integration of EPCs into human aortic endothelial cells (HAECs) by increasing adhesion molecule expression. In the mouse hind limb ischemia model, TUDCA promoted blood perfusion by enhancing angiogenesis through recruitment of Flk-1(+) /CD34(+) and Sca-1(+) /c-kit(+) progenitors into damaged tissue. In GFP(+) bone marrow-transplanted hind limb ischemia, TUDCA induced recruitment of GFP(+) /c-kit(+) progenitors to the ischemic area, resulting in an increased blood perfusion ratio. Histological analysis suggested that GFP(+) progenitors mobilized from bone marrow, integrated into blood vessels, and differentiated into VEGFR(+) cells. In addition, TUDCA decreased cellular senescence by reducing levels of p53, p21, and reactive oxygen species and increased nitric oxide. Transplantation of TUDCA-primed senescent EPCs in hind limb ischemia significantly improved blood vessel regeneration, as compared with senescent EPCs. Our results suggested that TUDCA promoted neovascularization by enhancing the mobilization of stem/progenitor cells from bone marrow, their differentiation into EPCs, and their integration with preexisting endothelial cells.


Asunto(s)
Vasos Sanguíneos/citología , Vasos Sanguíneos/efectos de los fármacos , Células Madre Hematopoyéticas/citología , Células Madre/citología , Células Madre/efectos de los fármacos , Ácido Tauroquenodesoxicólico/farmacología , Adulto , Animales , Diferenciación Celular/fisiología , Células Endoteliales/citología , Células Endoteliales/metabolismo , Femenino , Células Madre Hematopoyéticas/metabolismo , Humanos , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Desnudos , Neovascularización Patológica/patología , Células Madre/metabolismo
13.
Int J Mol Sci ; 17(9)2016 Aug 24.
Artículo en Inglés | MEDLINE | ID: mdl-27563882

RESUMEN

Hypoxia has diverse stimulatory effects on human adipose-derived stem cells (ASCs). In the present study, we investigated whether hypoxic culture conditions (2% O2) suppress spontaneous mineralization and osteogenic differentiation of ASCs. We also investigated signaling pathways and molecular mechanisms involved in this process. We found that hypoxia suppressed spontaneous mineralization and osteogenic differentiation of ASCs, and up-regulated mRNA and protein expression of Insulin-like growth factor binding proteins (IGFBPs) in ASCs. Although treatment with recombinant IGFBPs did not affect osteogenic differentiation of ASCs, siRNA-mediated inhibition of IGFBP3 attenuated hypoxia-suppressed osteogenic differentiation of ASCs. In contrast, overexpression of IGFBP3 via lentiviral vectors inhibited ASC osteogenic differentiation. These results indicate that hypoxia suppresses spontaneous mineralization and osteogenic differentiation of ASCs via intracellular IGFBP3 up-regulation. We determined that reactive oxygen species (ROS) generation followed by activation of the MAPK and PI3K/Akt pathways play pivotal roles in IGFBP3 expression under hypoxia. For example, ROS scavengers and inhibitors for MAPK and PI3K/Akt pathways attenuated the hypoxia-induced IGFBP3 expression. Inhibition of Elk1 and NF-κB through siRNA transfection also led to down-regulation of IGFBP3 mRNA expression. We next addressed the proliferative potential of ASCs with overexpressed IGFBP3, but IGFBP3 overexpression reduced the proliferation of ASCs. In addition, hypoxia reduced the osteogenic differentiation of bone marrow-derived clonal mesenchymal stem cells. Collectively, our results indicate that hypoxia suppresses the osteogenic differentiation of mesenchymal stem cells via IGFBP3 up-regulation.


Asunto(s)
Diferenciación Celular/fisiología , Hipoxia de la Célula/fisiología , Proteína 3 de Unión a Factor de Crecimiento Similar a la Insulina/metabolismo , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/metabolismo , Osteogénesis/fisiología , Animales , Diferenciación Celular/genética , Hipoxia de la Célula/genética , Proliferación Celular/genética , Proliferación Celular/fisiología , Células Cultivadas , Humanos , Inmunoprecipitación , Proteína 3 de Unión a Factor de Crecimiento Similar a la Insulina/genética , Ratones , Osteogénesis/genética , Especies Reactivas de Oxígeno/metabolismo , Reacción en Cadena en Tiempo Real de la Polimerasa
14.
Pharmacol Res ; 102: 176-83, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26453959

RESUMEN

Liver fibrosis and cirrhosis, the late stage of fibrosis, are threatening diseases that lead to liver failure and patient death. Although aberrantly activated hepatic stellate cells (HSCs) are the main cause of disease initiation, the symptoms are primarily related to damaged hepatocytes. Thus, damaged hepatocytes, as well as HSCs, need to be simultaneously considered as therapeutic targets to develop more efficient treatments. Here, we suggest cromolyn sodium as an anti-fibrotic agent to commonly modulate hepatocytes and hepatic stellate cells. The differentially expressed genes from 6 normal and 40 cirrhotic liver tissues which were collected from GEO data were assessed by pharmacokinetic analysis using a connectivity map to identify agents that commonly revert abnormal hepatocytes and HSCs to normal conditions. Based on a series of analyses, a few candidates were selected. Candidates were tested in vitro to determine their anti-fibrotic efficacy on HSCs and hepatocytes. Cromolyn, which was originally developed as a mast cell stabilizer, showed the potential to ameliorate activated HSCs in vitro. The activation and collagen accumulation for HSC cell lines LX2 and HSC-T6 were reduced by 50% after cromolyn treatment at a low concentration without apoptosis. Furthermore, cromolyn treatment compromised the TGF-ß-induced epithelial mesenchyme transition and replicative senescence rate of hepatocytes, which are generally associated with fibrogenesis. Taken together, cromolyn may be the basis for an effective cure for fibrosis and cirrhosis because it targets both HSCs and hepatocytes.


Asunto(s)
Cromolin Sódico/farmacología , Células Estrelladas Hepáticas/efectos de los fármacos , Hepatocitos/efectos de los fármacos , Cirrosis Hepática/tratamiento farmacológico , Apoptosis/efectos de los fármacos , Células Cultivadas , Senescencia Celular/efectos de los fármacos , Colágeno/metabolismo , Transición Epitelial-Mesenquimal/efectos de los fármacos , Células Estrelladas Hepáticas/metabolismo , Hepatocitos/metabolismo , Humanos , Hígado/efectos de los fármacos , Hígado/metabolismo , Cirrosis Hepática/metabolismo , Factor de Crecimiento Transformador beta/metabolismo
15.
J Biol Chem ; 288(46): 32886-96, 2013 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-24072712

RESUMEN

The peroxisome proliferator-activated receptor γ (PPARγ) is a central regulator of adipogenesis and modulates glucose and lipid metabolism. In this study, herpesvirus-associated ubiquitin-specific protease (HAUSP) was isolated as a binding partner of PPARγ. Both endogenous and exogenous PPARγ associated with HAUSP in co-immunoprecipitation analysis. HAUSP, but not the catalytically inactive HAUSP C223S mutant, increased the stability of both endogenous and exogenous PPARγ through its deubiquitinating activity. Site-directed mutagenesis experiments showed that the Lys(462) residue of PPARγ is critical for ubiquitination. HBX 41,108, a specific inhibitor of HAUSP, abolished the increase in PPARγ stability induced by HAUSP. In addition, knockdown of endogenous HAUSP using siRNA decreased PPARγ protein levels. HAUSP enhanced the transcriptional activity of both exogenous and endogenous PPARγ in luciferase activity assays. Quantitative RT-PCR analysis showed that HAUSP increased the transcript levels of PPARγ target genes in HepG2 cells, resulting in the enhanced uptake of glucose and fatty acids, and vice versa, upon siRNA knockdown of HAUSP. In vivo analysis using adenoviruses confirmed that HAUSP, but not the HAUSP C223S mutant, decreased blood glucose and triglyceride levels, which are associated with the increased expression of endogenous PPARγ and lipid accumulation in the liver. Our results demonstrate that the stability and activity of PPARγ are modulated by the deubiquitinating activity of HAUSP, which may be a target for the development of anti-diabetic drugs.


Asunto(s)
PPAR gamma/metabolismo , Transcripción Genética/fisiología , Ubiquitina Tiolesterasa/metabolismo , Proteasas Ubiquitina-Específicas/metabolismo , Ubiquitinación/fisiología , Adenoviridae , Sustitución de Aminoácidos , Animales , Transporte Biológico Activo/efectos de los fármacos , Transporte Biológico Activo/fisiología , Glucemia/genética , Glucemia/metabolismo , Células COS , Chlorocebus aethiops , Ácidos Grasos/sangre , Ácidos Grasos/genética , Técnicas de Silenciamiento del Gen , Células HeLa , Células Hep G2 , Humanos , Indenos/farmacología , Masculino , Ratones , Mutagénesis Sitio-Dirigida , Mutación Missense , PPAR gamma/genética , Estabilidad Proteica , Pirazinas/farmacología , Transcripción Genética/efectos de los fármacos , Transducción Genética , Ubiquitina Tiolesterasa/antagonistas & inhibidores , Ubiquitina Tiolesterasa/genética , Peptidasa Específica de Ubiquitina 7 , Proteasas Ubiquitina-Específicas/genética , Ubiquitinación/efectos de los fármacos
16.
Cardiovasc Res ; 120(7): 745-755, 2024 May 29.
Artículo en Inglés | MEDLINE | ID: mdl-38507654

RESUMEN

AIMS: In hypoxia, endothelial cells (ECs) proliferate, migrate, and form new vasculature in a process called angiogenesis. Recent studies have suggested that ECs rely on glycolysis to meet metabolic needs for angiogenesis in ischaemic tissues, and several studies have investigated the molecular mechanisms integrating angiogenesis and endothelial metabolism. Here, we investigated the role of stem cell factor (SCF) and its receptor, cKIT, in regulating endothelial glycolysis during hypoxia-driven angiogenesis. METHODS AND RESULTS: SCF and cKIT signalling increased the glucose uptake, lactate production, and glycolysis in human ECs under hypoxia. Mechanistically, SCF and cKIT signalling enhanced the expression of genes encoding glucose transporter 1 (GLUT1) and glycolytic enzymes via Akt- and ERK1/2-dependent increased translation of hypoxia inducible factor 1A (HIF1A). In hypoxic conditions, reduction of glycolysis and HIF-1α expression using chemical inhibitors significantly reduced the SCF-induced in vitro angiogenesis in human ECs. Compared with normal mice, mice with oxygen-induced retinopathy (OIR), characterized by ischaemia-driven pathological retinal neovascularization, displayed increased levels of SCF, cKIT, HIF-1α, GLUT1, and glycolytic enzymes in the retina. Moreover, cKIT-positive neovessels in the retina of mice with OIR showed elevated expression of GLUT1 and glycolytic enzymes. Further, blocking SCF and cKIT signalling using anti-SCF neutralizing IgG and cKIT mutant mice significantly reduced the expression of HIF-1α, GLUT1, and glycolytic enzymes and decreased the pathological neovascularization in the retina of mice with OIR. CONCLUSION: We demonstrated that SCF and cKIT signalling regulate angiogenesis by controlling endothelial glycolysis in hypoxia and elucidated the SCF/cKIT/HIF-1α axis as a novel metabolic regulation pathway during hypoxia-driven pathological angiogenesis.


Asunto(s)
Hipoxia de la Célula , Transportador de Glucosa de Tipo 1 , Glucólisis , Subunidad alfa del Factor 1 Inducible por Hipoxia , Proteínas Proto-Oncogénicas c-kit , Transducción de Señal , Factor de Células Madre , Animales , Humanos , Factor de Células Madre/metabolismo , Factor de Células Madre/genética , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Transportador de Glucosa de Tipo 1/metabolismo , Transportador de Glucosa de Tipo 1/genética , Proteínas Proto-Oncogénicas c-kit/metabolismo , Proteínas Proto-Oncogénicas c-kit/genética , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Ratones Endogámicos C57BL , Células Endoteliales/metabolismo , Células Endoteliales/patología , Neovascularización Retiniana/metabolismo , Neovascularización Retiniana/patología , Neovascularización Retiniana/genética , Ratones , Neovascularización Fisiológica , Células Cultivadas , Modelos Animales de Enfermedad , Glucosa/metabolismo
17.
Biochem Biophys Res Commun ; 432(2): 281-6, 2013 Mar 08.
Artículo en Inglés | MEDLINE | ID: mdl-23399565

RESUMEN

Aminoacyl-tRNA synthetases (ARSs) recognize a specific sequence or structural characteristics of their cognate tRNAs. To contribute to the understanding how these recognition sites were selected, we generated two different RNA libraries containing either 42mer or 70mer random sequence and used them to select RNA aptamers that specifically bound to methionyl-tRNA synthetase (MRS) of Mycobacterium tuberculosis. The aptamer pools selected from the two RNA libraries showed strong binding affinity and selectivity to M. tuberculosis MRS compared to that of the homologous Escherichia coli MRS. The RNA aptamers selected from the two completely unrelated RNA pools shared the octamer sequence including CAU and the anticodon sequence of tRNA(Met). The secondary structure prediction suggested that the octamer motif in the selected aptamers would form a loop similar to the anticodon loop of tRNA(Met). The results suggest that the RNA loop containing CAU triplet could selected as a major recognition site for MRS during evolution more or less regarding, and also showed that species-specific ARS inhibitors can be obtained by in vitro evolution.


Asunto(s)
Evolución Molecular , Metionina-ARNt Ligasa/metabolismo , ARN de Transferencia de Metionina/genética , ARN de Transferencia de Metionina/metabolismo , Aptámeros de Nucleótidos/química , Aptámeros de Nucleótidos/genética , Secuencia de Bases , Evolución Molecular Dirigida , Escherichia coli/enzimología , Datos de Secuencia Molecular , Mycobacterium tuberculosis/enzimología , Conformación de Ácido Nucleico , ARN de Transferencia de Metionina/química
18.
Stem Cells ; 30(8): 1703-13, 2012 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-22696466

RESUMEN

Recent studies suggest that epigenetic modifications, such as DNA methylation and histone modification, can alter the differentiation potential of stem cells or progenitor cells. Specifically, coactivator-associated arginine methyltransferase 1 (CARM1) is known to act as a coactivator for various transcription factors and to regulate gene expression by chromatin remodeling through histone methylation. Here, for the first time, we have used direct protein delivery of CARM1 using cell-penetrating peptide (CPP) to regulate the differentiation potential of human mesenchymal stem cells (hMSCs). Immunofluorescence showed that the CPP-CARM1 protein is successfully delivered into the nuclei of hMSCs. Further experiments using immunofluorescence and Western blotting showed that the delivered CARM1 protein can effectively methylate the arginine 17 residue of histone H3 in both bone marrow (BM)- and adipose-derived (AD)-hMSCs, thus suggesting that the CARM1 protein delivered by the CPP system is biologically active in hMSCs. Chromatin immunoprecipitation (ChIP) assay and genome-wide gene expression profiling supported the result that delivered CARM1 protein can cause chromatin remodeling through histone methylation. Finally, the CPP-CARM1 protein efficiently elevated the differentiation efficiency of BM-hMSCs and AD-hMSCs into adipogenic, osteogenic, and myogenic cell lineages in vitro. Altered expression of critical genes after hMSC differentiation was reconfirmed by real-time reverse transcription polymerase chain reaction (qRT-PCR). Collectively, our results suggest that CPP-CARM1 can elevate the differentiation potential of hMSCs into various cell types, and that this system using CPP is a useful tool for exogenous protein delivery in clinical applications of cell-based therapy.


Asunto(s)
Péptidos de Penetración Celular/metabolismo , Células Madre Mesenquimatosas/citología , Proteína-Arginina N-Metiltransferasas/metabolismo , Diferenciación Celular/fisiología , Péptidos de Penetración Celular/genética , Expresión Génica , Humanos , Células Madre Mesenquimatosas/enzimología , Células Madre Mesenquimatosas/metabolismo , Análisis por Micromatrices , Regiones Promotoras Genéticas , Proteína-Arginina N-Metiltransferasas/genética , Transcripción Genética
19.
Anticancer Res ; 42(7): 3453-3461, 2022 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-35790260

RESUMEN

BACKGROUND/AIM: Cytotoxic payload conjugation to antibodies efficiently suppresses tumors and contributes to the improvement of cancer survival. In our previous study, c-Kit targeting antibody-drug conjugate (2G4-DM1) with DM1, a microtubule inhibitor, efficiently suppressed tumor growth. However, slow-growing c-Kit-positive tumors, such as GIST-48, did not efficiently respond to DM1. In this study, we aimed to treat tumors using 2G4 immunotoxin with Pseudomonas exotoxin A (PE) as a payload. MATERIALS AND METHODS: Modified FcBP-PE24 containing p-benzoyl-L-phenylalanine, unnatural amino acid, was expressed in E. coli and purified. Then, photoconjugation of 2G4 antibody and FcBP-PE24 at 365 nm was carried out and 2G4 immunotoxin was purified using anion exchange chromatography. In vitro cytotoxicity of 2G4 immunotoxins was assessed in HMC-1.2, GIST-48, and MDA-MB-453 cells. Then, in vivo efficacy analysis was performed using C.B-17 SCID mice. RESULTS: 2G4 immunotoxin efficiently induced cytotoxicity in 2G4-DM1-resistant HMC-1.2 and GIST-48 cells by inhibiting protein synthesis but not in c-Kit-negative MDA-MB-453 cells. The results showed ~200-fold or more increase in cytotoxicity against c-Kit-positive cells compared to IC50 of 2G4-DM1. In addition, 2G4 immunotoxin suppressed tumor growth in the in vivo xenograft mouse model. CONCLUSION: 2G4 immunotoxins could be an alternative therapeutic strategy for microtubule inhibitor- resistant cancer cells.


Asunto(s)
Tumores del Estroma Gastrointestinal , Inmunoconjugados , Inmunotoxinas , Animales , Escherichia coli , Humanos , Inmunoconjugados/farmacología , Inmunotoxinas/farmacología , Ratones , Ratones SCID
20.
Mol Oncol ; 16(6): 1290-1308, 2022 03.
Artículo en Inglés | MEDLINE | ID: mdl-34407310

RESUMEN

c-Kit overexpression and activating mutations, which are reported in various cancers, including gastrointestinal stromal tumor (GIST), small-cell lung cancer (SCLC), acute myeloid leukemia, acral melanoma, and systemic mastocytosis (SM), confer resistance to tyrosine kinase inhibitors (TKIs). To overcome TKI resistance, an anti-c-Kit antibody-drug conjugate was developed in this study to treat wild-type and mutant c-Kit-positive cancers. NN2101, a fully human IgG1, was conjugated to DM1, a microtubule inhibitor, through N-succinimidyl-4-(N-maleimidomethyl) cyclohexane-1-carboxylate (SMCC) (to give NN2101-DM1). The antitumor activity of NN2101-DM1 was evaluated in vitro and in vivo using various cancer cell lines. NN2101-DM1 exhibited potent growth-inhibitory activities against c-Kit-positive cancer cell lines. In a mouse xenograft model, NN2101-DM1 exhibited potent growth-inhibitory activities against imatinib-resistant GIST and SM cells. In addition, NN2101-DM1 exhibited a significantly higher anti-cancer effect than carboplatin/etoposide against SCLC cells where c-Kit does not mediate cancer pathogenesis. Furthermore, the combination of NN2101-DM1 with imatinib in imatinib-sensitive GIST cells induced complete remission compared with treatment with NN2101-DM1 or imatinib alone in mouse xenograft models. These results suggest that NN2101-DM1 is a potential therapeutic agent for wild-type and mutant c-Kit-positive cancers.


Asunto(s)
Tumores del Estroma Gastrointestinal , Inmunoconjugados , Neoplasias Pulmonares , Carcinoma Pulmonar de Células Pequeñas , Animales , Línea Celular Tumoral , Proliferación Celular , Resistencia a Antineoplásicos/genética , Tumores del Estroma Gastrointestinal/tratamiento farmacológico , Humanos , Mesilato de Imatinib/farmacología , Mesilato de Imatinib/uso terapéutico , Inmunoconjugados/farmacología , Inmunoconjugados/uso terapéutico , Ratones , Mutación/genética , Inhibidores de Proteínas Quinasas/farmacología , Inhibidores de Proteínas Quinasas/uso terapéutico , Proteínas Proto-Oncogénicas c-kit/genética , Proteínas Proto-Oncogénicas c-kit/metabolismo , Proteínas Tirosina Quinasas Receptoras/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA