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

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
J Immunol ; 199(6): 1989-1997, 2017 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-28779023

RESUMEN

Dendritic cells (DCs) are important for adaptive immune responses through the activation of T cells. The molecular interplay between DCs and T cells determines the magnitude of T cell responses or outcomes of functional differentiation of T cells. In this study, we demonstrated that DCs in mice that are Rbpj deficient in CD11c+ cells (Rbpj-/- mice) promoted the differentiation of IL-17A-producing Th17 cells. Rbpj-deficient DCs expressed little Aldh1a2 protein that is required for generating retinoic acid. Those DCs exhibited a reduced ability for differentiating regulatory T cells induced by TGF-ß. Rbpj protein directly regulated Aldh1a2 transcription by binding to its promoter region. The overexpression of Aldh1a2 in Rbpj-deficient DCs negated their Th17-promoting ability. Transfer of naive CD4+ T cells into Rag1-deficient Rbpj-/- mice enhanced colitis with increased Th17 and reduced induced regulatory T cells (iTreg) compared with control Rag1-deficient mice. The cotransfer of iTreg and naive CD4+ T cells into Rag1-deficient Rbpj-/- mice improved colitis compared with transfer of naive CD4+ T cell alone. Furthermore, cotransfer of DCs from Rbpj-/- mice that overexpressed Aldh1a2 or Notch-stimulated DCs together with naive CD4+ T cells into Rbpj-/-Rag1-deficient mice led to reduced colitis with increased iTreg numbers. Therefore, our studies identify Notch signaling in DCs as a crucial balancer of Th17/iTreg, which depends on the direct regulation of Aldh1a2 transcription in DCs.


Asunto(s)
Colitis/inmunología , Células Dendríticas/inmunología , Retinal-Deshidrogenasa/metabolismo , Linfocitos T Reguladores/inmunología , Células Th17/inmunología , Familia de Aldehído Deshidrogenasa 1 , Animales , Antígeno CD11c/metabolismo , Diferenciación Celular , Células Cultivadas , Regulación de la Expresión Génica , Genes RAG-1 , Humanos , Proteína de Unión a la Señal Recombinante J de las Inmunoglobulinas/genética , Interleucina-17/metabolismo , Activación de Linfocitos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Receptores Notch/metabolismo , Retinal-Deshidrogenasa/genética , Linfocitos T Reguladores/trasplante , Tretinoina/metabolismo
2.
Biochem Biophys Res Commun ; 444(4): 628-33, 2014 Feb 21.
Artículo en Inglés | MEDLINE | ID: mdl-24491544

RESUMEN

CD4(+) T cell activation is controlled by signaling through the T cell receptor in addition to various co-receptors, and is also affected by their interactions with effector and regulatory T cells in the microenvironment. Inflammatory bowel diseases (IBD) are caused by the persistent activation and expansion of auto-aggressive CD4(+) T cells that attack intestinal epithelial cells. However, the molecular basis for the persistent activation of CD4(+) T cells in IBD remains unclear. In this study, we investigated how the CD98 heavy chain (CD98hc, Slc3a2) affected the development of colitis in an experimental animal model. Transferring CD98hc-deficient CD4(+)CD25(-) T cells into Rag2(-/-) mice did not cause colitis accompanied by increasing Foxp3(+) inducible regulatory T cells. By comparison, CD98hc-deficient naturally occurring regulatory T cells (nTregs) had a decreased capability to suppress colitis induced by CD4(+)CD25(-) T cells, although CD98hc-deficient mice did not have a defect in the development of nTregs. Blocking CD98hc with an anti-CD98 blocking antibody prevented the development of colitis. Our results indicate that CD98hc regulates the expansion of autoimmune CD4(+) T cells in addition to controlling nTregs functions, which suggests the CD98hc as an important target molecule for establishing strategies for treating colitis.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Colitis/inmunología , Colitis/patología , Colon/patología , Cadena Pesada de la Proteína-1 Reguladora de Fusión/inmunología , Animales , Enfermedades Autoinmunes/inmunología , Linfocitos T CD4-Positivos/metabolismo , Linfocitos T CD4-Positivos/patología , Colitis/genética , Colon/inmunología , Colon/metabolismo , Proteínas de Unión al ADN/genética , Cadena Pesada de la Proteína-1 Reguladora de Fusión/genética , Eliminación de Gen , Subunidad alfa del Receptor de Interleucina-2/genética , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL
3.
J Immunol ; 188(5): 2227-34, 2012 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-22291182

RESUMEN

The interplay of CD4(+) and CD8(+) T cells targeting autoantigens is responsible for the progression of a number of autoimmune diseases, including type 1 diabetes mellitus (T1D). Understanding the molecular mechanisms that regulate T cell activation is crucial for designing effective therapies for autoimmune diseases. We probed a panel of Abs with T cell-modulating activity and identified a mAb specific for the H chain of CD98 (CD98hc) that was able to suppress T cell proliferation. The anti-CD98hc mAb also inhibited Ag-specific proliferation and the acquisition of effector function by CD4(+) and CD8(+) T cells in vitro and in vivo. Injection of the anti-CD98hc mAb completely prevented the onset of cyclophosphamide-induced diabetes in NOD mice. Treatment of diabetic NOD mice with anti-CD98hc reversed the diabetic state to normal levels, coincident with decreased proliferation of CD4(+) T cells. Furthermore, treatment of diabetic NOD mice with CD98hc small interfering RNA resolved T1D. These data indicate that strategies targeting CD98hc might have clinical application for treating T1D and other T cell-mediated autoimmune diseases.


Asunto(s)
Diabetes Mellitus Tipo 1/inmunología , Diabetes Mellitus Tipo 1/prevención & control , Cadena Pesada de la Proteína-1 Reguladora de Fusión/fisiología , Animales , Anticuerpos Monoclonales/administración & dosificación , Anticuerpos Monoclonales/uso terapéutico , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/metabolismo , Linfocitos T CD4-Positivos/patología , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/metabolismo , Linfocitos T CD8-positivos/patología , Ciclofosfamida/administración & dosificación , Diabetes Mellitus Tipo 1/inducido químicamente , Femenino , Cadena Pesada de la Proteína-1 Reguladora de Fusión/genética , Cadena Pesada de la Proteína-1 Reguladora de Fusión/inmunología , Inhibidores de Crecimiento/genética , Inhibidores de Crecimiento/metabolismo , Inhibidores de Crecimiento/fisiología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Endogámicos NOD , Ratones SCID , Ratones Transgénicos , Ratas , Ratas Wistar
4.
Front Immunol ; 14: 1065790, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-36776885

RESUMEN

FXR is a key molecule that modulates anti-inflammatory activity in the intestinal-liver axis. Although FXR has pleiotropic functions including regulation of liver inflammation and activation of macrophages, it remains unclear whether it is involved in macrophage polarization. In this paper we demonstrated that stimulation of macrophages derived from the bone marrow using an FXR agonist activated polarization toward M2 but not M1 macrophages. The treatment of mice with chitin skewed macrophage polarization towards M2 macrophages, while co-treatment with an FXR agonist further promoted the polarization toward M2 macrophages in vivo. This skewed polarization towards M2 macrophages by an FXR agonist was accompanied by increased expression of signaling molecules related to the retinoic acid receptor. Inhibition of the retinoic acid receptor suppressed FXR agonist-mediated M2 macrophage polarization, indicating that this polarization was, at least, partly dependent on the retinoic acid receptor pathway. These data demonstrate that FXR has a role in polarization toward M2 macrophages and suggest a possible therapeutic potential of FXR agonists in M2 macrophage-related conditions.


Asunto(s)
Macrófagos , Receptores Citoplasmáticos y Nucleares , Transducción de Señal , Animales , Ratones , Antiinflamatorios/metabolismo , Macrófagos/metabolismo , Receptores de Ácido Retinoico/metabolismo , Receptores Citoplasmáticos y Nucleares/agonistas
5.
Biochem Biophys Res Commun ; 418(4): 701-7, 2012 Feb 24.
Artículo en Inglés | MEDLINE | ID: mdl-22293205

RESUMEN

B cells are classified into several subsets depending on their functions, marker expression pattern and localization. Marginal zone B (MZB) cells are a distinct lineage from follicular B cells, and regulate host defenses against blood-borne pathogens. Notch2/RBP-J signaling regulates the development of MZB cells by interacting with delta-like 1 ligand, although the target genes for Notch2 signaling remain unclear. We identified Fos as an upregulated gene in LPS-stimulated B cells that received Notch2 signaling. Fos is expressed in CD21(high)CD23(low) MZB cells at a higher level compared to CD21(Int)CD23(high) follicular B cells. Deleting the Notch2 gene in CD19(+) B cells decreased Fos expression in B cells. Overexpression of Fos in Notch2-deficient B cells or bone marrow cells partially restored MZB development. Fos promoter activity was upregulated by Notch2 signaling, indicating that Notch2 directly controls Fos transcription associated with MZB development. These data identify Fos as one of the target genes for Notch2 signaling that is crucial for MZB development.


Asunto(s)
Linfocitos B/fisiología , Regulación del Desarrollo de la Expresión Génica , Proteínas Oncogénicas v-fos/genética , Receptor Notch2/metabolismo , Animales , Linfocitos B/metabolismo , Genes Reporteros , Luciferasas/genética , Ratones , Ratones Endogámicos C57BL , Regiones Promotoras Genéticas , Receptor Notch2/genética , Bazo/citología , Transcripción Genética
6.
Dig Dis Sci ; 57(11): 2955-64, 2012 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-22623042

RESUMEN

BACKGROUND: Clinical observations and experimental colitis models have indicated the importance of intestinal bacteria in the etiology of ulcerative colitis (UC), but a causative bacterial agent has not been identified. AIM: To determine how intestinal bacteria are associated with UC, fecal microbiota and other components were compared for UC patients and healthy adults. METHODS: Fresh feces were collected from 48 UC patients. Fecal microbiota were analyzed by use of terminal-restriction fragment length polymorphism (T-RFLP), real-time PCR, and culture. The concentrations of organic acids, indole, and ammonia, and pH and moisture, which are indicators of the intestinal environment, were measured and compared with healthy control data. RESULTS: T-RFLP data divided the UC patients into four clusters; one cluster was obtained for healthy subjects. The diversity of fecal microbiota was significantly lower in UC patients. There were significantly fewer Bacteroides and Clostridium subcluster XIVab, and the amount of Enterococcus was higher in UC patients than in healthy subjects. The fecal concentration of organic acids was significantly lower in UC patients who were in remission. CONCLUSION: UC patients have imbalances in the intestinal environment-less diversity of fecal microbiota, lower levels of major anaerobic bacteria (Bacteroides and Clostridium subcluster XIVab), and a lower concentration of organic acids.


Asunto(s)
Bacterias/aislamiento & purificación , Colitis Ulcerosa/microbiología , Heces/microbiología , Adulto , Anciano , Estudios de Casos y Controles , ADN Bacteriano/análisis , Femenino , Humanos , Mucosa Intestinal/microbiología , Masculino , Persona de Mediana Edad , Polimorfismo de Longitud del Fragmento de Restricción , Reacción en Cadena en Tiempo Real de la Polimerasa , Estadísticas no Paramétricas
7.
Phytother Res ; 26(2): 191-6, 2012 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-21604310

RESUMEN

Brown seaweed Undaria pinnatifida (Harvey) Suringar is popular as a foodstuff, and used for medical care in East Asian countries. The major components of this seaweed are shown to benefit hypertension and hyperlipidemia, and considered to reduce the risks of infarction and ischemic diseases. Furthermore, the intake of dietary fiber of seaweeds is considered to prevent the production and proliferation of cancer in the gastrointestinal tract. The direct effect of an ethanol extract prepared from Undaria pinnatifida sporophyll (mekabu) on HCT116 human colorectal cancer cells was examined, and the mekabu extract was shown to induce the non-oxidative apoptotic damage to the cells, thus resulting in the reduction of their viabilities in a concentration-dependent manner. Moreover, the cytotoxic effects of carcinostatic drugs, such as 5-fluorouracil (5-FU) and irinotecan (CPT-11), were observed only in the medium containing sera, while the mekabu extract could effectively reduce the cell viabilities even in the serum-free medium. These findings suggest that the mekabu extract may contain a potential active substance inducing the non-oxidative apoptotic cell death probably through a mechanism different from those of 5-FU and CPT-11, and hence mekabu is possibly useful as an auxiliary drug to the chemotherapy of colorectal cancer.


Asunto(s)
Antineoplásicos Fitogénicos/farmacología , Neoplasias Colorrectales/patología , Extractos Vegetales/farmacología , Algas Marinas/química , Undaria/química , Apoptosis/efectos de los fármacos , Camptotecina/análogos & derivados , Camptotecina/farmacología , Supervivencia Celular/efectos de los fármacos , Medios de Cultivo/química , Etanol , Fluorouracilo/farmacología , Células HCT116/efectos de los fármacos , Humanos , Irinotecán , Xantófilas/química , Xantófilas/farmacología
8.
JCI Insight ; 7(7)2022 04 08.
Artículo en Inglés | MEDLINE | ID: mdl-35393946

RESUMEN

Immunoproteasomes regulate the degradation of ubiquitin-coupled proteins and generate peptides that are preferentially presented by MHC class I. Mutations in immunoproteasome subunits lead to immunoproteasome dysfunction, which causes proteasome-associated autoinflammatory syndromes (PRAAS) characterized by nodular erythema and partial lipodystrophy. It remains unclear, however, how immunoproteasome dysfunction leads to inflammatory symptoms. Here, we established mice harboring a mutation in Psmb8 (Psmb8-KI mice) and addressed this question. Psmb8-KI mice showed higher susceptibility to imiquimod-induced skin inflammation (IMS). Blockade of IL-6 or TNF-α partially suppressed IMS in both control and Psmb8-KI mice, but there was still more residual inflammation in the Psmb8-KI mice than in the control mice. DNA microarray analysis showed that treatment of J774 cells with proteasome inhibitors increased the expression of the Cxcl9 and Cxcl10 genes. Deficiency in Cxcr3, the gene encoding the receptor of CXCL9 and CXCL10, in control mice did not change IMS susceptibility, while deficiency in Cxcr3 in Psmb8-KI mice ameliorated IMS. Taken together, these findings demonstrate that this mutation in Psmb8 leads to hyperactivation of the CXCR3 pathway, which is responsible for the increased susceptibility of Psmb8-KI mice to IMS. These data suggest the CXCR3/CXCL10 axis as a new molecular target for treating PRAAS.


Asunto(s)
Lipodistrofia , Inhibidores de Proteasoma , Animales , Quimiocina CXCL10/antagonistas & inhibidores , Inflamación/complicaciones , Inflamación/genética , Lipodistrofia/genética , Ratones , Mutación , Complejo de la Endopetidasa Proteasomal/metabolismo , Inhibidores de Proteasoma/farmacología , Receptores CXCR3/antagonistas & inhibidores
9.
Cell Death Dis ; 12(6): 601, 2021 06 10.
Artículo en Inglés | MEDLINE | ID: mdl-34112763

RESUMEN

The sensing of various extrinsic stimuli triggers the receptor-interacting protein kinase-3 (RIPK3)-mediated signaling pathway, which leads to mixed-lineage kinase-like (MLKL) phosphorylation followed by necroptosis. Although necroptosis is a form of cell death and is involved in inflammatory conditions, the roles of necroptosis in acute pancreatitis (AP) remain unclear. In the current study, we administered caerulein to Ripk3- or Mlkl-deficient mice (Ripk3-/- or Mlkl-/- mice, respectively) and assessed the roles of necroptosis in AP. We found that Ripk3-/- mice had significantly more severe pancreatic edema and inflammation associated with macrophage and neutrophil infiltration than control mice. Consistently, Mlkl-/- mice were more susceptible to caerulein-induced AP, which occurred in a time- and dose-dependent manner, than control mice. Mlkl-/- mice exhibit weight loss, edematous pancreatitis, necrotizing pancreatitis, and acinar cell dedifferentiation in response to tissue damage. Genetic deletion of Mlkl resulted in downregulation of the antiapoptotic genes Bclxl and Cflar in association with increases in the numbers of apoptotic cells, as detected by TUNEL assay. These findings suggest that RIPK3 and MLKL-mediated necroptosis exerts protective effects in AP and caution against the use of necroptosis inhibitors for AP treatment.


Asunto(s)
Necroptosis/fisiología , Pancreatitis/patología , Proteínas Quinasas/fisiología , Proteína Serina-Treonina Quinasas de Interacción con Receptores/fisiología , Animales , Desdiferenciación Celular/genética , Citoprotección/genética , Progresión de la Enfermedad , Femenino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Necroptosis/genética , Activación Neutrófila/genética , Páncreas/metabolismo , Páncreas/patología , Pancreatitis/genética , Proteínas Quinasas/genética , Proteína Serina-Treonina Quinasas de Interacción con Receptores/genética
10.
Sci Rep ; 11(1): 5070, 2021 03 03.
Artículo en Inglés | MEDLINE | ID: mdl-33658595

RESUMEN

Inactivation technology for severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is certainly a critical measure to mitigate the spread of coronavirus disease 2019 (COVID-19). A deep ultraviolet light-emitting diode (DUV-LED) would be a promising candidate to inactivate SARS-CoV-2, based on the well-known antiviral effects of DUV on microorganisms and viruses. However, due to variations in the inactivation effects across different viruses, quantitative evaluations of the inactivation profile of SARS-CoV-2 by DUV-LED irradiation need to be performed. In the present study, we quantify the irradiation dose of DUV-LED necessary to inactivate SARS-CoV-2. For this purpose, we determined the culture media suitable for the irradiation of SARS-CoV-2 and optimized the irradiation apparatus using commercially available DUV-LEDs that operate at a center wavelength of 265, 280, or 300 nm. Under these conditions, we successfully analyzed the relationship between SARS-CoV-2 infectivity and the irradiation dose of the DUV-LEDs at each wavelength without irrelevant biological effects. In conclusion, total doses of 1.8 mJ/cm2 for 265 nm, 3.0 mJ/cm2 for 280 nm, and 23 mJ/cm2 for 300 nm are required to inactivate 99.9% of SARS-CoV-2. Our results provide quantitative antiviral effects of DUV irradiation on SARS-CoV-2, serving as basic knowledge of inactivation technologies against SARS-CoV-2.


Asunto(s)
SARS-CoV-2/efectos de la radiación , Terapia Ultravioleta/métodos , Inactivación de Virus/efectos de la radiación , COVID-19/epidemiología , COVID-19/transmisión , Humanos , SARS-CoV-2/metabolismo , Rayos Ultravioleta , Virosis/prevención & control
11.
Front Immunol ; 11: 591713, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33178225

RESUMEN

The NLRC4 inflammasome assembles in response to detection of bacterial invasion, and NLRC4 activation leads to the production of IL-1ß and IL-18 together with pyroptosis-mediated cell death. Missense activating mutations in NLRC4 cause autoinflammatory disorders whose symptoms are distinctly dependent on the site of mutation and other aspects of the genetic background. To determine the involvement of IL-1ß and IL-18 in the inflammation induced by NLRC4 mutation, we depleted IL-1ß, IL-18, or both cytokines in Nlrc4-transgenic mice in which mutant Nlrc4 is expressed under the MHC class II promoter (Nlrc4-H443P-Tg mice). The deletion of the Il1b or Il18 gene in Nlrc4-H443P-Tg mice reduced the neutrophil numbers in the spleen, and mice with deletion of both genes had an equivalent number of neutrophils compared to wild-type mice. Deletion of Il1b ameliorated but did not eliminate bone marrow hyperplasia, while mice deficient in Il18 showed no bone marrow hyperplasia. In contrast, tail bone deformity remained in the presence of Il18 deficiency, but Il1b deficiency completely abolished bone deformity. The decreased bone density in Nlrc4-H443P-Tg mice was counteracted by Il1b but not Il18 deficiency. Our results demonstrate the distinct effects of IL-1ß and IL-18 on NLRC4-induced inflammation among tissues, which suggests that blockers for each cytokine should be utilized depending on the site of inflammation.


Asunto(s)
Proteínas Reguladoras de la Apoptosis/metabolismo , Proteínas de Unión al Calcio/metabolismo , Susceptibilidad a Enfermedades , Inflamación/etiología , Inflamación/metabolismo , Interleucina-18/metabolismo , Interleucina-1beta/metabolismo , Animales , Proteínas Reguladoras de la Apoptosis/genética , Biomarcadores , Biopsia , Proteínas de Unión al Calcio/genética , Modelos Animales de Enfermedad , Genotipo , Inmunohistoquímica , Inflamasomas/metabolismo , Inflamación/diagnóstico , Ratones , Ratones Transgénicos , Mutación , Microtomografía por Rayos X
12.
J Neurosci Res ; 87(11): 2608-14, 2009 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-19360904

RESUMEN

Progesterone treatment has previously been reported to promote the differentiation of glial cells probably through the production of 5alpha-reduced neurosteroids, resulting in the enhancement of serotonin-stimulated brain-derived neurotrophic factor (BDNF) gene expression, which is considered to contribute to the survival, regeneration, and plasticity of neuronal cells in the brain and hence has been suggested to improve mood disorders and other symptoms in depressive patients. Based on these previous observations, the effects on glial cells of histone deacetylase (HDAC) inhibitors, which are known as agents promoting cell differentiation, were examined using rat C6 glioma cells as a model for in vitro studies. Consequently, trichostatin A (TSA), sodium butyrate (NaB), and valproic acid (VPA) stimulated glial fibrillary acidic protein (GFAP) gene expression, and their stimulatory effects on GFAP gene expression were inhibited by treatment of these cells with finasteride, an inhibitor of the enzyme producing 5alpha-reduced neurosteroids. In addition, HDAC inhibitors enhanced serotonin-stimulated BDNF gene expression, the enhancement of which could be abolished by the inhibition of 5alpha-reduced neurosteroid production in the glioma cells. These results suggest that HDAC inhibitors may be able to promote the differentiation of rat C6 glioma cells through the production of 5alpha-reduced neurosteroids, resulting in the enhancement of serotonin-stimulated BDNF gene expression as a consequence of promoting their differentiation, indicating the possibility that differentiated glial cells may be implicated in preserving the integrity of neural networks as well as improving the function of neuronal cells in the brain.


Asunto(s)
Factor Neurotrófico Derivado del Encéfalo/genética , Diferenciación Celular/efectos de los fármacos , Expresión Génica/efectos de los fármacos , Proteína Ácida Fibrilar de la Glía/genética , Inhibidores de Histona Desacetilasas , Neuroglía/efectos de los fármacos , Animales , Factor Neurotrófico Derivado del Encéfalo/metabolismo , Butiratos/farmacología , Diferenciación Celular/fisiología , Línea Celular Tumoral , Relación Dosis-Respuesta a Droga , Inhibidores Enzimáticos/farmacología , Finasterida/farmacología , Proteína Ácida Fibrilar de la Glía/metabolismo , Ácidos Hidroxámicos/farmacología , Neuroglía/metabolismo , ARN Mensajero/metabolismo , Ratas , Serotonina/metabolismo , Compuestos de Sodio/farmacología , Esteroides/metabolismo , Factores de Tiempo , Ácido Valproico/farmacología
13.
Pharmacology ; 81(2): 164-72, 2008.
Artículo en Inglés | MEDLINE | ID: mdl-18025841

RESUMEN

Cytotoxic effects of desipramine on human colon carcinoma HT29 and HCT116 cells were examined. Desipramine reduced the viability of HT29 cells in a concentration-dependent manner, but failed to cause any significant change in the viability of HCT116 cells by the concentration up to 50 mumol/l, at which an approximately 60% reduction of the viability of HT29 cells was observed. Despite their different sensitivities, desipramine caused the nonoxidative apoptotic damage to both of them. In contrast to HT29 cells, desipramine might cause the apoptotic death of HCT116 cells through the disturbance of mitochondrial function. These results suggest that desipramine may cause the nonoxidative apoptotic damage to different types of human colon carcinoma cells through either a nonmitochondrial or a mitochondrial pathway, which may confer the different sensitivities to this drug on these tumor cells.


Asunto(s)
Adenocarcinoma/metabolismo , Apoptosis/fisiología , Neoplasias del Colon/metabolismo , Desipramina/farmacología , Mitocondrias/metabolismo , Adenocarcinoma/tratamiento farmacológico , Adenocarcinoma/patología , Apoptosis/efectos de los fármacos , Muerte Celular/efectos de los fármacos , Línea Celular Tumoral , Neoplasias del Colon/tratamiento farmacológico , Neoplasias del Colon/patología , Desipramina/uso terapéutico , Relación Dosis-Respuesta a Droga , Células HCT116 , Células HT29 , Humanos , Mitocondrias/efectos de los fármacos , Mitocondrias/patología , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología
14.
Cancer Lett ; 241(2): 228-34, 2006 Sep 28.
Artículo en Inglés | MEDLINE | ID: mdl-16300879

RESUMEN

Clostridium perfringens has been regarded as one of the intestinal bacteria increasing colon cancer risk. In previous studies, we have shown that the oral administration of C. perfringens culture medium can inhibit the mutagen-induced formation of pre-neoplastic lesions in rat colon, thus proposing the existence of factor(s) preventing colon tumorigenesis in this culture medium. However, the properties of effective factor(s) and the mechanism of this inhibitory action still remain to be investigated. Then, the effect of C. perfringens culture medium on human colon adenocarcinoma HT29 cells was examined. The exposure of HT29 cells to C. perfringens culture medium was found to suppress the proliferation of these cells probably through the reduction of immediate early gene egr-1 expression. These observations suggest that C. perfringens culture medium has a cytostatic action on colon tumor cells, which may be responsible for the prevention of pre-neoplastic formation in rat colon.


Asunto(s)
Adenocarcinoma/patología , Clostridium perfringens/fisiología , Neoplasias del Colon/patología , Medios de Cultivo/farmacología , Adenocarcinoma/metabolismo , Proliferación Celular/efectos de los fármacos , Infecciones por Clostridium , Neoplasias del Colon/metabolismo , Proteína 1 de la Respuesta de Crecimiento Precoz/metabolismo , Células HT29 , Calor , Humanos , Immunoblotting , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Linfocitos T/inmunología
15.
Brain Res ; 1085(1): 49-56, 2006 Apr 26.
Artículo en Inglés | MEDLINE | ID: mdl-16581042

RESUMEN

Influence of adrenergic and serotonergic stimulation on glial fibrillary acidic protein (GFAP) gene expression in rat C6 glioma cells was first examined as an in vitro model experiment for investigating the neuronal regulation of glial cell differentiation. Stimulation of these cells with isoproterenol and serotonin elevated GFAP mRNA levels followed by an increase in its protein contents, thus suggesting that both adrenergic and serotonergic stimulation might induce the differentiation of the glioma cells. In addition, progesterone and its 5alpha-reduced metabolite dihydroprogesterone also elevated GFAP mRNA levels in rat C6 glioma cells, consistent with their stimulatory actions on GFAP gene expression observed in rat astrocytes. Further studies showed that the elevation of GFAP mRNA levels induced by isoproterenol and serotonin as well as progesterone was abolished by pretreatment of the glioma cells with finasteride, an inhibitor of 5alpha-reduced steroid production. Moreover, the stimulatory actions of isoproterenol and serotonin on GFAP gene expression were inhibited by pretreatment with a GABA(A) receptor antagonist bicuculline and a progesterone receptor antagonist RU486. These findings suggest that both adrenergic and serotonergic stimulation may indirectly activate GFAP gene expression probably through the production of 5alpha-reduced steroid metabolites in rat C6 glioma cells, proposing the possibility that 5alpha-reduced neurosteroids may play a potential role in the neuronal regulation of glial cell differentiation.


Asunto(s)
3-Oxo-5-alfa-Esteroide 4-Deshidrogenasa/fisiología , Agonistas Adrenérgicos beta/farmacología , Expresión Génica/efectos de los fármacos , Proteína Ácida Fibrilar de la Glía/metabolismo , Isoproterenol/farmacología , Serotonina/farmacología , 20-alfa-Dihidroprogesterona/farmacología , Análisis de Varianza , Animales , Western Blotting/métodos , Línea Celular Tumoral , Interacciones Farmacológicas , Inhibidores Enzimáticos/farmacología , Finasterida/farmacología , Expresión Génica/fisiología , Proteína Ácida Fibrilar de la Glía/genética , Glioma , Progesterona/farmacología , ARN Mensajero/metabolismo , Ratas , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa/métodos , Serotonina/fisiología , Factores de Tiempo
16.
Eur J Pharmacol ; 541(1-2): 17-23, 2006 Jul 10.
Artículo en Inglés | MEDLINE | ID: mdl-16753142

RESUMEN

Preclinical studies have suggested that the long-term use of antidepressants may result in the initiation and/or promotion of tumor in the gastrointestinal tract. However, a possible relationship between the use of antidepressants and the production of colon cancer has not yet been confirmed, and hence requires to be further investigated. To address this issue, the effects of antidepressants on the proliferation of colorectal tumor cells were examined using human HT29 colon carcinoma cells, and tricyclic antidepressant, such as imipramine, desipramine and amitriptyline, were shown to reduce the cell viability in a manner dependent on the time exposing to these drugs. In addition to these drugs, a selective serotonin reuptake inhibitor fluoxetine, but not a monoamine oxidase inhibitor tranylcypromine, caused the reduction of cell viability, similar in extent to that caused by imipramine. Further studies showed that desipramine caused the apoptotic cell death, which could be prevented by neither catalase, reduced-form glutathione (GSH), nor N-acetylcysteine (NAC), without accompanying the disruption of mitochondrial membrane potential within the cells and the release of cytochrome c into the cell cytoplasm. Moreover, desipramine caused the arrest of cell-cycle progression at either G0/G1-phase or G2/M-phase, which might be depending upon the drug concentration. Thus, these results suggest that tricyclic antidepressants may be cytotoxic, and induce the non-oxidative apoptotic death of human HT29 colon carcinoma cells probably through a non-mitochondrial pathway associated with the cell-cycle progression.


Asunto(s)
Antidepresivos Tricíclicos/farmacología , Proliferación Celular/efectos de los fármacos , Amitriptilina/farmacología , Apoptosis/efectos de los fármacos , Ciclo Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Neoplasias del Colon/patología , Neoplasias del Colon/fisiopatología , Fragmentación del ADN/efectos de los fármacos , Desipramina/farmacología , Células HT29 , Humanos , Imipramina/farmacología , Potenciales de la Membrana/efectos de los fármacos , Membranas Mitocondriales/efectos de los fármacos , Membranas Mitocondriales/fisiología , Factores de Tiempo
17.
Sci Rep ; 6: 26791, 2016 05 26.
Artículo en Inglés | MEDLINE | ID: mdl-27225296

RESUMEN

The differentiation of adipocytes is tightly regulated by a variety of intrinsic molecules and also by extrinsic molecules produced by adjacent cells. Dysfunction of adipocyte differentiation causes lipodystrophy, which impairs glucose and lipid homeostasis. Although dysfunction of immunoproteasomes causes partial lipodystrophy, the detailed molecular mechanisms remain to be determined. Here, we demonstrate that Psmb8, a catalytic subunit for immunoproteasomes, directly regulates the differentiation of preadipocytes and additionally the differentiation of preadipocytes to mature adipocytes. Psmb8(-/-) mice exhibited slower weight gain than wild-type mice, and this was accompanied by reduced adipose tissue volume and smaller size of mature adipocytes compared with controls. Blockade of Psmb8 activity in 3T3-L1 cells disturbed the differentiation to mature adipocytes. Psmb8(-/-) mice had fewer preadipocyte precursors, fewer preadipocytes and a reduced ability to differentiate preadipocytes toward mature adipocytes. Our data demonstrate that Psmb8-mediated immunoproteasome activity is a direct regulator of the differentiation of preadipocytes and their ultimate maturation.


Asunto(s)
Adipocitos/citología , Adipogénesis/fisiología , Complejo de la Endopetidasa Proteasomal/fisiología , Células 3T3-L1 , Tejido Adiposo/patología , Animales , Células Cultivadas , Inhibidores de Cisteína Proteinasa/farmacología , Dieta Alta en Grasa , Insulina/farmacología , Leptina/sangre , Leupeptinas/farmacología , Lípidos/sangre , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Oligopéptidos/farmacología , Tamaño de los Órganos , Complejo de la Endopetidasa Proteasomal/deficiencia , Complejo de la Endopetidasa Proteasomal/metabolismo , Inhibidores de Proteasoma/farmacología , Aumento de Peso
18.
Inflamm Regen ; 36: 13, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-29259686

RESUMEN

Recent progress in DNA sequencing technology has made it possible to identify specific genetic mutations in familial disorders. For example, autoinflammatory syndromes are caused by mutations in gene coding for immunoproteasomes. These diseases include Japanese autoinflammatory syndrome with lipodystrophy, Nakajo-Nishimura syndrome, joint contractures, muscular atrophy, microcytic anemia, panniculitis-associated lipodystrophy syndrome, and chronic atypical neutrophilic dermatosis with lipodystrophy and elevated temperature syndrome. Causal mutations of these syndromes are present in gene coding for subunits of the immunoproteasome. Importantly, a genetically modified mouse that lacks the catalytic subunit of immunoproteasomes does not always develop an autoinflammatory syndrome. Analysis of causal gene mutations, assessment of patients' phenotypic changes, and appropriate animal models will be indispensable for clarifying the underlying mechanisms responsible for the development of autoinflammatory syndromes and establishing curative approaches.

20.
J Mol Neurosci ; 27(3): 293-301, 2005.
Artículo en Inglés | MEDLINE | ID: mdl-16280599

RESUMEN

As one of the in vitro model experiments for investigating a possible effect of extracellular environmental stresses on glial cell proliferation, the influence of high salt culture conditions on the growth of rat C6 glioma cells was examined. Exposure to the culture medium containing high concentrations of NaCl reduced the number of viable cells in a concentration-dependent manner without any significant change in their viability. In contrast, proliferation of these cells was not substantially altered by culturing them in hyperosmotic medium containing either sucrose or glycerol, both of which were osmotically almost equivalent to high salt culture medium. On the other hand, expression of the egr-1 gene, an immediate early gene related to the proliferation and differentiation of various cells, was enhanced by culturing glioma cells in high salt medium while the reduction of glial fibrillary acidic protein content, an index of glial cell differentiation, was observed under the same culture conditions. Further studies on the relationship between egr-1 gene expression and the cell cycle showed that cell-cycle progression was arrested at S-phase by culturing glioma cells in high salt medium. Moreover, both resveratrol and CPT-11, which are known to arrest cell-cycle progression, elevated egr-1 mRNA levels in glioma cells. Thus, these observations suggest that high salt culture conditions might suppress the proliferation of rat C6 glioma cells as a consequence of arresting cell-cycle progression at S-phase, resulting secondarily in the compensatory enhancement of egr-1 gene expression.


Asunto(s)
Proliferación Celular , Medios de Cultivo/química , Fase S/fisiología , Cloruro de Sodio/química , Animales , Camptotecina/análogos & derivados , Camptotecina/metabolismo , Línea Celular Tumoral , Supervivencia Celular , Proteína 1 de la Respuesta de Crecimiento Precoz/genética , Proteína 1 de la Respuesta de Crecimiento Precoz/metabolismo , Inhibidores Enzimáticos/metabolismo , Proteína Ácida Fibrilar de la Glía/genética , Proteína Ácida Fibrilar de la Glía/metabolismo , Glioma , Soluciones Hipertónicas , Irinotecán , Concentración Osmolar , Ratas , Resveratrol , Estilbenos/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA