Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 82
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Immunol Res ; 70(2): 208-215, 2022 04.
Artículo en Inglés | MEDLINE | ID: mdl-35014010

RESUMEN

Gliostatin/thymidine phosphorylase (GLS/TP) is known to have angiogenic and arthritogenic activities in the pathogenesis of rheumatoid arthritis (RA). The novel oral Janus kinase (JAK) inhibitor baricitinib has demonstrated high efficacy in RA. However, the effect of baricitinib on fibroblast-like synoviocytes (FLSs), a key component of invasive synovitis, has not been still elucidated. This study investigated whether GLS/TP production could be regulated by JAK/signal transducers and activators of transcription (STAT) signaling in FLSs derived from patients with RA. FLSs were cultured and stimulated by interferon (IFN)γ in the presence of baricitinib. Expression levels of GLS/TP were determined using reverse transcription-polymerase chain reaction (RT-PCR), enzyme-linked immunosorbent assay (ELISA), and immunocytochemistry. Phosphorylation of STAT proteins was investigated by Western blot. In cultured FLSs, GLS/TP mRNA and protein levels were significantly induced by treatment with IFNγ and these inductions were suppressed by baricitinib treatment. Baricitinib inhibited IFNγ-induced STAT1 phosphorylation, while JAK/STAT activation played a pivotal role in IFNγ-mediated GLS/TP upregulation in RA. These results suggested that baricitinib suppressed IFNγ-induced GLS/TP expression by inhibiting JAK/STAT signaling, resulting in the attenuation of neovascularization, synovial inflammation, and cartilage destruction.


Asunto(s)
Artritis Reumatoide , Inhibidores de las Cinasas Janus , Sinoviocitos , Artritis Reumatoide/metabolismo , Azetidinas , Células Cultivadas , Fibroblastos , Humanos , Inhibidores de las Cinasas Janus/metabolismo , Inhibidores de las Cinasas Janus/farmacología , Purinas , Pirazoles , Sulfonamidas , Timidina Fosforilasa/genética , Timidina Fosforilasa/metabolismo
2.
Exp Cell Res ; 404(1): 112630, 2021 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-33971195

RESUMEN

The proximal tubules, which are part of the kidney, maintain blood homeostasis by absorbing amino acids, glucose, water, and ions such as sodium (Na), potassium, and bicarbonate. Proximal tubule dysfunction is associated with the pathogenesis of many kidney diseases. Renal proximal tubular epithelial cells (RPTECs) are responsible for the main functions of the proximal tubules. Therefore, in vitro experiments using RPTECs would greatly enhance our understanding of nephron physiology and pathobiology. It is preferable to use immortalized cell lines, such as human kidney-2 (HK-2) cells, because they are derived from humans and maintain growth indefinitely. However, tissue-specific RPTEC phenotypes, including apical-basal polarization, are frequently lost in conventional two-dimensional culture methods in part due to microenvironmental deficiencies. To overcome this limitation, we developed a three-dimensional (3D) spheroid culture method for HK-2 cells using an extracellular matrix. HK-2 spheroids in 3D culture formed a tubule-like architecture with cellular polarity and showed markedly restored Na transport function. 3D culture of HK-2 cells also increased expression of kidney development-related genes, including WNT9B. Models of human renal tubules using HK-2 spheroids will greatly improve our understanding of the physiology and pathobiology of the kidney.


Asunto(s)
Polaridad Celular/fisiología , Células Epiteliales/citología , Túbulos Renales Proximales/citología , Túbulos Renales/metabolismo , Transporte Biológico , Línea Celular , Matriz Extracelular/metabolismo , Humanos , Riñón/metabolismo , Sodio/metabolismo
3.
J Pharmacol Sci ; 146(1): 65-69, 2021 May.
Artículo en Inglés | MEDLINE | ID: mdl-33858657

RESUMEN

The blood-brain barrier (BBB) is mainly formed by brain capillary endothelial cells (BCECs) and is exposed to hypoxic environments under pathological conditions. The effects of hypoxia on the expression and activity of Ca2+-activated Cl- (ClCa) channels, TMEM16A, were examined in bovine brain endothelial t-BBEC117 cells and mouse BCECs. The expression of TMEM16A was upregulated by hypoxia. Whole-cell ClCa currents increased under hypoxia. Hypoxia also increased cell proliferation and trans-endothelial permeability, which were attenuated by ClCa channel blockers or TMEM16A siRNA. These findings are useful for elucidating the pathological role of TMEM16A ClCa channels in the BBB during cerebral ischemia.


Asunto(s)
Anoctamina-1/genética , Anoctamina-1/metabolismo , Barrera Hematoencefálica/citología , Barrera Hematoencefálica/patología , Encéfalo/citología , Proliferación Celular , Células Endoteliales/metabolismo , Células Endoteliales/patología , Expresión Génica/genética , Hipoxia-Isquemia Encefálica/genética , Hipoxia-Isquemia Encefálica/patología , Regulación hacia Arriba/genética , Animales , Bovinos , Línea Celular , Ratones , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo
4.
Mol Pharmacol ; 98(1): 61-71, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-32358165

RESUMEN

The blood-brain barrier (BBB) is essential for the maintenance of homeostasis in the brain. Brain capillary endothelial cells (BCECs) comprise the BBB, and thus a delicate balance between their proliferation and death is required. Although the activity of ion channels in BCECs is involved in BBB functions, the underlying molecular mechanisms remain unclear. In the present study, the molecular components of Ca2+-activated Cl- (ClCa) channels and their physiological roles were examined using mouse BCECs (mBCECs) and a cell line derived from bovine BCECs, t-BBEC117. Expression analyses revealed that TMEM16A was strongly expressed in mBCECs and t-BBEC117 cells. In t-BBEC117 cells, whole-cell Cl- currents were sensitive to the ClCa channel blockers, 100 µM niflumic acid and 10 µM T16Ainh-A01, and were also reduced markedly by small-interfering RNA (siRNA) knockdown of TMEM16A. Importantly, block of ClCa currents with ClCa channel blockers or TMEM16A siRNA induced membrane hyperpolarization. Moreover, treatment with TMEM16A siRNA caused an increase in resting cytosolic Ca2+ concentration ([Ca2+]cyt). T16Ainh-A01 reduced cell viability in a concentration-dependent manner. Either ClCa channel blockers or TMEM16A siRNA also curtailed cell proliferation and migration. Furthermore, ClCa channel blockers attenuated the trans-endothelial permeability. In combination, these results strongly suggest that TMEM16A contributes to ClCa channel conductance and can regulate both the resting membrane potential and [Ca2+]cyt in BCECs. Our data also reveal how these BCECs may be involved in the maintenance of BBB functions, as both the proliferation and migration are altered following changes in channel activity. SIGNIFICANCE STATEMENT: In brain capillary endothelial cells (BCECs) of the blood-brain barrier (BBB), TMEM16A is responsible for Ca2+-activated Cl- channels and can regulate both the resting membrane potential and cytosolic Ca2+ concentration, contributing to the proliferation and migration of BCECs. The present study provides novel information on the molecular mechanisms underlying the physiological functions of BCECs in the BBB and a novel target for therapeutic drugs for disorders associated with dysfunctions in the BBB.


Asunto(s)
Anoctamina-1/metabolismo , Barrera Hematoencefálica/metabolismo , Encéfalo/citología , Calcio/metabolismo , Canales de Cloruro/metabolismo , Animales , Anoctamina-1/antagonistas & inhibidores , Barrera Hematoencefálica/citología , Barrera Hematoencefálica/diagnóstico por imagen , Encéfalo/efectos de los fármacos , Encéfalo/metabolismo , Bovinos , Línea Celular , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Células Endoteliales/citología , Células Endoteliales/efectos de los fármacos , Células Endoteliales/metabolismo , Células HEK293 , Humanos , Masculino , Potenciales de la Membrana/efectos de los fármacos , Ratones , Ácido Niflúmico/farmacología , Pirimidinas/farmacología , Tiazoles/farmacología
5.
Biochem Biophys Res Commun ; 523(1): 153-158, 2020 02 26.
Artículo en Inglés | MEDLINE | ID: mdl-31839216

RESUMEN

Brain capillary endothelial cells (BCECs) form the blood-brain barrier (BBB) and play an essential role in the regulation of its functions. Oxidative stress accumulates excessive reactive oxygen species (ROS) and facilitates the death of BCECs, leading to a dysfunctional BBB. However, the mechanisms underlying the death of BCECs under oxidative stress remain unclear. In the present study, the effects of oxidative stress on cell viability, ROS production, intracellular Ca2+ concentration, and protein expression were examined using a cell line derived from bovine BCECs, t-BBEC117. When t-BBEC117 cells were exposed to oxidative stress induced by hydrogen peroxide (H2O2, 10-100 µM), cell growth was inhibited in a dose-dependent manner. Oxidative stress by 30 µM H2O2 increased the production of ROS and its effects were blocked by the ROS scavenger, 10 mM N-acetyl-l-cysteine (NAC). In addition, oxidative stress reduced store-operated Ca2+ entry (SOCE) and this decrease was recovered by NAC or the Orai channel activator, 5 µM 2-aminoethyl diphenylborinate (2-APB). The siRNA knockdown of Orai1 revealed that Orai1 was mainly responsible for SOCE channels and its activity was decreased by oxidative stress. However, the protein expression of Orai1 and STIM1 was not affected by oxidative stress. Oxidative stress-induced cell death was rescued by 2-APB, NAC, or the STIM-Orai activating region. In conclusion, oxidative stress reduces Orai1-mediated SOCE and, thus, facilitates the death of BCECs.


Asunto(s)
Acetilcisteína/farmacología , Calcio/metabolismo , Células Endoteliales/efectos de los fármacos , Proteína ORAI1/antagonistas & inhibidores , Estrés Oxidativo , Animales , Barrera Hematoencefálica/efectos de los fármacos , Barrera Hematoencefálica/metabolismo , Bovinos , Muerte Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Células Endoteliales/metabolismo , Peróxido de Hidrógeno/antagonistas & inhibidores , Peróxido de Hidrógeno/farmacología , Proteína ORAI1/metabolismo , Estrés Oxidativo/efectos de los fármacos
6.
Neurobiol Dis ; 120: 51-62, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-30176352

RESUMEN

Krabbe disease (KD), or globoid cell leukodystrophy, is an inherited lysosomal storage disease with leukodystrophy caused by a mutation in the galactosylceramidase (GALC) gene. The majority of patients show the early onset form of KD dominated by cerebral demyelination with apoptotic oligodendrocyte (OL) death. However, the initial pathophysiological changes in developing OLs remain poorly understood. Here, we show that OLs of twitcher mice, an authentic mouse model of KD, exhibited developmental defects and impaired myelin formation in vivo and in vitro. In twitcher mouse brain, abnormal myelination and reduced expression of myelin genes during the period of most active OL differentiation and myelination preceded subsequent progressive OL death and demyelination. Importantly, twitcher mouse OL precursor cells proliferated normally, but their differentiation and survival were intrinsically defective. These defects were associated with aberrant accumulation of endogenous psychosine (galactosylsphingosine) and reduced activation of the Erk1/2 and Akt/mTOR pathways before apoptotic cell death. Collectively, our results demonstrate that GALC deficiency in developing KD OLs profoundly affects their differentiation and maturation, indicating the critical contribution of OL dysfunction to KD pathogenesis.


Asunto(s)
Modelos Animales de Enfermedad , Leucodistrofia de Células Globoides/metabolismo , Oligodendroglía/metabolismo , Psicosina/metabolismo , Animales , Proliferación Celular/fisiología , Células Cultivadas , Leucodistrofia de Células Globoides/genética , Leucodistrofia de Células Globoides/patología , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Oligodendroglía/patología , Psicosina/genética
7.
Nihon Ronen Igakkai Zasshi ; 55(3): 358-366, 2018.
Artículo en Japonés | MEDLINE | ID: mdl-30122702

RESUMEN

AIM: Awareness reform aims to enable survival in an aging society, and ultimately, improve healthcare. An ideal way to achieve this is by implementing Advance directive (Ad) and Advance Care Planning (ACP), which do not usually include postmortem events. This study aims to create opportunities for Ad and ACP to include the postmortem period as a trigger for this awareness reform. METHODS: We conducted an Ad/ACP enlightenment lecture, and a questionnaire survey pre- and post-lecture for the elderly in old New Town, which is known for its aging society. The questionnaire comprised 38 multiple-choice questions covering 6 themes assuming an advanced state of dementia. RESULTS: There were 35 participants (7 men and 22 women) aged 40-89 years. Several people left during the lecture, making it difficult to capture the precise transformation effect with regard to changing of mind. However, the effect of enlightenment was identified as a result of the consciousness survey. A statistically significant change in consciousness occurred in response to social contribution after death. Furthermore, notably more people wanted emergency transportation compared to those wanting resuscitation and extension of life. CONCLUSIONS: The medical treatment desired might vary over time. Even the desire for life extension may differ significantly among individuals. This survey indicated a divergent view between the general public and medical staff, regarding a series of medical actions. We must persistently promote opportunities for enlightenment in cooperation with the general public (i.e., the communities and families we serve).


Asunto(s)
Planificación Anticipada de Atención , Educación del Paciente como Asunto , Adulto , Directivas Anticipadas , Anciano , Anciano de 80 o más Años , Donación Directa de Tejido , Femenino , Registros de Salud Personal , Humanos , Masculino , Persona de Mediana Edad , Encuestas y Cuestionarios
8.
Am J Physiol Cell Physiol ; 315(2): C202-C213, 2018 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-29768048

RESUMEN

Brain capillary endothelial cells (BCECs) play a central role in maintenance of blood-brain barrier (BBB) function and, therefore, are essential for central nervous system homeostasis and integrity. Although brain ischemia damages BCECs and causes disruption of BBB, the related influence of hypoxia on BCECs is not well understood. Hypoxic stress can upregulate functional expression of specific K+ currents in endothelial cells, e.g., Kir2.1 channels without any alterations in the mRNA level, in t-BBEC117, a cell line derived from bovine BCECs. The hyperpolarization of membrane potential due to Kir2.1 channel upregulation significantly facilitates cell proliferation. In the present study, the mechanisms underlying the hypoxia-induced Kir2.1 upregulation was examined. We emphasize the involvement of dynamin2, a protein known to be involved in a number of surface expression pathways. Hypoxic culture upregulated dynamin2 expression in t-BBEC117 cells. The inhibition of dynamin2 by Dynasore canceled hypoxia-induced upregulation of Kir2.1 currents by reducing surface expression. On the contrary, Kir2.1 currents and proteins in t-BBEC117 cultured under normoxia were increased by overexpression of dynamin2, but not by dominant-negative dynamin2. Molecular imaging based on bimolecular fluorescence complementation, double-immunostaining, and coimmunoprecipitation assays revealed that dynamin2 can directly bind to the Kir2.1 channel. Moreover, hypoxic culture downregulated hypoxic-inducible factor-1α (HIF-1α) expression. Knockdown of HIF-1α increased dynamin2 expression in t-BBEC117 cells, in both normoxic and hypoxic culture conditions. In summary, our results demonstrated that hypoxia downregulates HIF-1α, increases dynamin2 expression, and facilitates Kir2.1 surface expression, resulting in hyperpolarization of membrane potential and subsequent increase in Ca2+ influx in BCECs.


Asunto(s)
Encéfalo/metabolismo , Hipoxia de la Célula/fisiología , Células Endoteliales/metabolismo , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Canales de Potasio de Rectificación Interna/metabolismo , Regulación hacia Arriba/fisiología , Animales , Calcio/metabolismo , Bovinos , Línea Celular , Movimiento Celular/fisiología , Proliferación Celular/fisiología , Sistema Nervioso Central/metabolismo , Regulación hacia Abajo/fisiología , Dinaminas/metabolismo , Humanos
9.
Clin Exp Rheumatol ; 36(4): 559-567, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29352846

RESUMEN

OBJECTIVES: Gliostatin (GLS) is known to have angiogenic and arthritogenic activity, and GLS expression levels in serum from patients with rheumatoid arthritis (RA) are significantly correlated with the disease activity. Tofacitinib is a novel oral Janus kinase (JAK) inhibitor and is effective in treating RA. However, the mechanism of action of tofacitinib in fibroblast-like synoviocytes (FLSs) has not been elucidated. The purpose of this study was to investigate the modulatory effects of tofacitinib on serum GLS levels in patients with RA and GLS production in FLSs derived from patients with RA. METHODS: Six patients with RA who had failed therapy with at least one TNF inhibitor and were receiving tofacitinib therapy were included in the study. Serum samples were collected to measure CRP, MMP-3 and GLS expression. FLSs derived from patients with RA were cultured and stimulated by TNFα with or without tofacitinib. GLS expression levels were determined using reverse transcription-polymerase chain reaction (RT-PCR), EIA and immunocytochemistry, and signal transducer and activator of transcription (STAT) protein phosphorylation levels were determined by western blotting. RESULTS: Treatment with tofacitinib decreased serum GLS levels in all patients. GLS mRNA and protein expression levels were significantly increased by treatment with TNF-α alone, and these increases were suppressed by treatment with tofacitinib, which also inhibited TNF-α-induced STAT1 phosphorylation. CONCLUSIONS: JAK/STAT activation plays a pivotal role in TNF-α-mediated GLS up-regulation in RA. Suppression of GLS expression in FLSs has been suggested to be one of the mechanisms through which tofacitinib exerts its anti-inflammatory effects.


Asunto(s)
Artritis Reumatoide/tratamiento farmacológico , Inhibidores de las Cinasas Janus/farmacología , Piperidinas/farmacología , Pirimidinas/farmacología , Pirroles/farmacología , Sinoviocitos/metabolismo , Timidina Fosforilasa/genética , Factor de Necrosis Tumoral alfa/antagonistas & inhibidores , Anciano , Anciano de 80 o más Años , Femenino , Fibroblastos/metabolismo , Humanos , Masculino , Persona de Mediana Edad , Piperidinas/uso terapéutico , Pirimidinas/uso terapéutico , Pirroles/uso terapéutico , Factor de Transcripción STAT1/metabolismo
10.
Mod Rheumatol ; 28(3): 495-505, 2018 May.
Artículo en Inglés | MEDLINE | ID: mdl-28741989

RESUMEN

OBJECTIVES: Gliostatin (GLS) has angiogenic and arthritogenic activities and enzymatic activity as thymidine phosphorylase. Aberrant GLS production has been observed in the synovial membranes of patients with rheumatoid arthritis (RA). Matrix metalloproteinases (MMPs) are involved in joint destruction. Promoters of GLS and some MMP genes contain Sp1 binding sites. We examined the inhibitory effect of the Sp1 inhibitor mithramycin on GLS-induced GLS and MMP expression in cultured fibroblast-like synoviocytes (FLSs). METHODS: Synovial tissue samples were obtained from patients with RA. FLSs pretreated with mithramycin were cultured with GLS. The mRNA expression levels of GLS and MMP-1, MMP-2, MMP-3, MMP-9, and MMP-13 were determined using reverse transcription polymerase chain reactions. Protein levels were measured using enzyme immunoassay and gelatin zymography. RESULTS: GLS upregulated the expression of GLS itself and of MMP-1, MMP-3, MMP-9, and MMP-13, an effect significantly reduced by treatment with mithramycin. GLS and mithramycin had no effect on MMP-2 expression. CONCLUSIONS: Mithramycin downregulated the increased expression of GLS and MMP-1, MMP-3, MMP-9, and MMP-13 in FLSs treated with GLS. Because GLS plays a pathological role in RA, blocking GLS stimulation using an agent such as mithramycin may be a novel approach to antirheumatic therapy.


Asunto(s)
Artritis Reumatoide/metabolismo , Metaloproteinasas de la Matriz/metabolismo , Plicamicina/farmacología , Sinoviocitos/efectos de los fármacos , Timidina Fosforilasa/metabolismo , Anciano , Anciano de 80 o más Años , Antirreumáticos/farmacología , Artritis Reumatoide/patología , Células Cultivadas , Femenino , Humanos , Masculino , Metaloproteinasas de la Matriz/genética , Persona de Mediana Edad , Sinoviocitos/metabolismo , Timidina Fosforilasa/genética
11.
Mol Med Rep ; 16(3): 3095-3102, 2017 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-28713972

RESUMEN

AT motif binding factor 1 (ATBF1) is a transcriptional regulator that functions as a tumour suppressor to negatively affect cancer cell growth. In the present study four specific polyclonal antibodies against ATBF1 were generated, and the expression and intracellular localization of ATBF1 in colonic mucosae, polyps, adenoma and adenocarcinoma tissue samples were investigated. The four polyclonal antibodies produced were as follows: MB34 and MB49, which recognize the N­ and C­terminal fragments of ATBF1, respectively; and D1­120 and MB44, which recognize the middle fragments of ATBF1 that contain three nuclear localization signals (NLS). In total, 191 colon samples were examined by immunohistochemical analysis. In addition, colon cancer cells were transfected with four ATBF1 expression vectors, and the subcellular localization of each fragment was examined. Normal colon mucosal cells were not observed to express ATBF1. However, a small number of hyperplastic polyps, serrated adenomas and tubular adenomas expressed ATBF1. Colon cancer cells were observed to express D1­120­ and MB44­reactive middle fragments of ATBF1 in their cell nuclei. However, the N­ and C­terminal fragments of ATBF1 did not translocate to the nucleus. Transfection of ATBF1 fragments revealed cleavage of the ATBF1 protein and nuclear translocation of the cleaved middle portion containing the NLS. A positive correlation between the cytoplasmic localization of the N­ and C­termini of ATBF1, nuclear localization of the middle portion of ATBF1 and malignant cancer cell invasion was observed. In conclusion, the results of the present study suggest that alterations in the expression and subcellular localization of ATBF1, as a result of post­transcriptional modifications, are associated with malignant features of colon tumours.


Asunto(s)
Neoplasias del Colon/metabolismo , Neoplasias del Colon/patología , Proteínas de Homeodominio/metabolismo , Diferenciación Celular , Núcleo Celular/metabolismo , Humanos , Mucosa Intestinal/metabolismo , Mucosa Intestinal/patología , Transporte de Proteínas , Coloración y Etiquetado , Fracciones Subcelulares/metabolismo
12.
Brain Res ; 1662: 65-74, 2017 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-28257780

RESUMEN

Erythropoietin (EPO), a hematopoietic hormonal cytokine induced in response to hypoxia, has neuroprotective effects. EPO receptor (EPOR) is expressed in microglia, resident immune cells in the brain. However, the effect of EPO on microglial activation is not clear. In the present study, we demonstrated that the EPOR is highly expressed in microglia, rather than in neurons or astrocytes, in in vitro experiments. Therefore, we investigated whether EPO could attenuate lipopolysaccharide (LPS)-mediated activation of microglia in vitro. The BV-2 microglial cell line was treated with LPS in the absence or presence of EPO. In the presence of EPO, microglial expression of LPS-induced inflammatory cytokine genes was significantly decreased. In addition, EPO suppressed the LPS-induced phagocytic activity of BV-2 cells towards fluorescent beads, as well as induction of inducible nitric oxide synthase. In in vivo experiments, EPO significantly decreased the LPS-induced expression of inflammatory cytokine genes in mouse brains. Furthermore, morphological analysis of cortical microglia in the brains of mice stimulated with LPS revealed that combined treatment with EPO alleviated LPS-induced morphological changes in the microglia. These data indicate that EPO attenuates microglial activation, including morphological changes in vivo, phagocytosis in vitro, and the production of inflammatory cytokines in vivo and in vitro. Further investigation of EPO modulation of LPS-induced microglial activation may contribute to the development of novel neuroprotective therapies.


Asunto(s)
Eritropoyetina/metabolismo , Microglía/fisiología , Receptores de Eritropoyetina/metabolismo , Animales , Astrocitos/fisiología , Encéfalo/metabolismo , Técnicas de Cultivo de Célula , Citocinas , Eritropoyetina/uso terapéutico , Inflamación/metabolismo , Lipopolisacáridos/farmacología , Activación de Macrófagos , Ratones , Microglía/efectos de los fármacos , Neuronas/fisiología , Fármacos Neuroprotectores/metabolismo , Óxido Nítrico/metabolismo , Óxido Nítrico Sintasa de Tipo II/metabolismo , Fagocitosis/efectos de los fármacos , Ratas , Receptores de Eritropoyetina/genética
13.
Clin Orthop Relat Res ; 475(6): 1693-1701, 2017 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-28155209

RESUMEN

BACKGROUND: The p53 protein in mesenchymal stem cells (MSCs) regulates differentiation to osteogenic or adipogenic lineage. Because p53 function is depressed in most malignancies, if MSCs in malignancy also have p53 hypofunction, differentiation therapy to osteogenic or adipogenic lineage may be an effective treatment. We therefore wished to begin to explore this idea by evaluating atypical lipomatous tumor/well-differentiated liposarcoma (ALT/WDL) cells, because murine double minute 2 (MDM2) gene amplification, which leads to p53 hypofunction, is found in almost all ALT/WDLs. QUESTIONS/PURPOSES: We compared osteogenic and adipogenic differentiation potency between MSCs isolated and cultured from normal adipose tissues and ALT/WDLs from the same patients. METHODS: During tumor resections in six patients with ALT/WDL, we analyzed 3 mL of tumor, and for comparison, we harvested a similar amount of normal-appearing subcutaneous adipose tissue from an area remote from the tumor for comparison. Adipogenic differentiation potency was quantitatively assessed using spectrometry after oil red O staining. Osteogenic differentiation potency was semiquantitatively assessed by measuring a specific colored area after alkaline phosphatase (ALP) and alizarin red S staining. ALP is related to preosseous cellular metabolism, and alizarin red is related to calcium deposits in cell culture. There were three observers for each assessment, and each assessment (including induced-differentiation and histologic analysis) was performed in duplicate. We then analyzed the mechanism of the difference of osteogenic differentiation potency using the MDM2-specific inhibitor Nutlin-3 at various concentrations. RESULTS: In terms of adipogenic differentiation potency, contrary to our expectations, more fatty acid droplets were observed in MSCs derived from normal fat than in MSCs derived from ALT/WDL, although we found no significant difference between MSCs derived from ALT/WDL and MSCs derived from normal fat; the mean differentiation potency values (normal adipose tissue versus ALT/WDL) (± SD) were 0.34 (SD, ± 0.13; 95% CI, 0.24-0.44) versus 0.25 (SD, ± 0.10; 95% CI, 0.18-0.33; p = 0.22). By contrast, we found greater osteogenic differentiation potency in MSCs derived from ALT/WDL than in MSCs derived from normal fat. The mean differentiation potency values (normal adipose tissue versus ALT/WDL) (±SD) based on ALP staining was 1.0 versus 17 (SD, ± 36; 95% CI, -2.8 to 38; p = 0.04). However, we found no differences based on alizarin red S staining; mean differentiation potency value (normal adipose tissue versus ALT/WDL) (± SD) was 1.0 versus 4.2 (SD, ± 4.8; 95% CI, 1.3-7.2; p = 0.58). The gap of osteogenic differentiation potency between MSCs from normal adipose tissue and ALT/WDL was decreased as MDM2-inhibitor Nutlin-3 concentration increased. CONCLUSIONS: MSCs derived from ALT/WDL had higher osteogenic differentiation potency based on ALP staining, which disappeared as Nutlin-3 concentration increased, suggesting that could be caused by amplified MDM2 in ALT/WDL. Future laboratory studies might mechanistically confirm the gene and protein expression, and based on the mechanism of the gap of differentiation potency, if p53 contrast between MSCs in tumor and normal tissue could be stimulated, less-toxic and more-effective differentiation therapy to MSCs in malignancies might be developed.


Asunto(s)
Tejido Adiposo/citología , Diferenciación Celular/fisiología , Lipoma/patología , Liposarcoma/patología , Células Madre Mesenquimatosas/fisiología , Femenino , Humanos , Masculino , Persona de Mediana Edad , Osteogénesis/fisiología
14.
BMC Cancer ; 16(1): 805, 2016 10 18.
Artículo en Inglés | MEDLINE | ID: mdl-27756245

RESUMEN

BACKGROUND: Pathological stage and grade have limited ability to predict the outcomes of superficial urothelial bladder carcinoma at initial transurethral resection (TUR). AT-motif binding factor 1 (ATBF1) is a tumor suppressive transcription factor that is normally localized to the nucleus but has been detected in the cytoplasm in several cancers. Here, we examined the diagnostic value of the intracellular localization of ATBF1 as a marker for the identification of high risk urothelial bladder carcinoma. METHODS: Seven anti-ATBF1 antibodies were generated to cover the entire ATBF1 sequence. Four human influenza hemagglutinin-derived amino acid sequence-tagged expression vectors with truncated ATBF1 cDNA were constructed to map the functional domains of nuclear localization signals (NLSs) with the consensus sequence KR[X10-12]K. A total of 117 samples from initial TUR of human bladder carcinomas were analyzed. None of the patients had received chemotherapy or radiotherapy before pathological evaluation. RESULTS: ATBF1 nuclear localization was regulated synergistically by three NLSs on ATBF1. The cytoplasmic fragments of ATBF1 lacked NLSs. Patients were divided into two groups according to positive nuclear staining of ATBF1, and significant differences in overall survival (P = 0.021) and intravesical recurrence-free survival (P = 0.013) were detected between ATBF1+ (n = 110) and ATBF1- (n = 7) cases. Multivariate analysis revealed that ATBF1 staining was an independent prognostic factor for intravesical recurrence-free survival after adjusting for cellular grading and pathological staging (P = 0.008). CONCLUSIONS: Cleavage of ATBF1 leads to the cytoplasmic localization of ATBF1 fragments and downregulates nuclear ATBF1. Alterations in the subcellular localization of ATBF1 due to fragmentation of the protein are related to the malignant character of urothelial carcinoma. Pathological evaluation using anti-ATBF1 antibodies enabled the identification of highly malignant cases that had been overlooked at initial TUR. Nuclear localization of ATBF1 indicates better prognosis of urothelial carcinoma.


Asunto(s)
Biomarcadores de Tumor/metabolismo , Carcinoma de Células Transicionales/metabolismo , Proteínas de Homeodominio/metabolismo , Neoplasias de la Vejiga Urinaria/metabolismo , Adulto , Anciano , Anciano de 80 o más Años , Animales , Western Blotting , Células COS , Carcinoma de Células Transicionales/patología , Línea Celular Tumoral , Núcleo Celular/metabolismo , Chlorocebus aethiops , Citoplasma/metabolismo , Progresión de la Enfermedad , Femenino , Células HEK293 , Humanos , Inmunohistoquímica , Estimación de Kaplan-Meier , Masculino , Persona de Mediana Edad , Análisis Multivariante , Neoplasias de la Vejiga Urinaria/patología
15.
Stem Cells ; 34(11): 2733-2743, 2016 11.
Artículo en Inglés | MEDLINE | ID: mdl-27339271

RESUMEN

Bone homeostasis comprises the balance between bone-forming osteoblasts and bone-resorbing osteoclasts (OCs), with an acceleration of osteoclastic bone resorption leading to osteoporosis. OCs can be generated from bone marrow cells (BMCs) under the tightly regulated local bone environment. However, it remained difficult to identify the critical cells responsible for providing an osteoclastogenesis niche. In this study, we used a fluorescence-activated cell sorting technique to determine the cell populations important for forming an appropriate microenvironment for osteoclastogenesis and to verify the associated interactions between osteoclast precursor cells and non-OCs. We isolated and removed a small cell population specific for osteoclastogenesis (CXCR4+ CD45- ) from mouse BMCs and cultured the remaining cells with receptor activator of nuclear factor-kappa B ligand (RANKL) and macrophage-colony stimulating factor. The resulting cultures showed significantly less large osteoclast formation. Quantitative RT-PCR analysis revealed that these CXCR4+ CD45- cells expressed low levels of RANK and RANKL, but high levels of critical chemokines including stromal cell derived factor 1 (SDF-1), chemokine (C-X-C motif) ligand 7 (CXCL7), and chemokine (C-X3-C motif) ligand 1 (CX3CL1). Furthermore, an SDF-1-specific antibody strongly suppressed OC formation in RAW264.7 cells and antibodies against SDF-1, CXCL7, and CX3CL1 suppressed OC formation in BMCs. These results suggest that isolated CXCR4+ CD45- cells support an appropriate microenvironment for osteoclastogenesis with a direct effect on the cells expressing SDF-1, CXCL7, and CX3CL1 receptors. The regulation of CXCR4+ CD45- cell function might therefore inform therapeutic strategies for diseases involving loss of bone homeostasis. Stem Cells 2016;34:2733-2743.


Asunto(s)
Quimiocina CX3CL1/genética , Quimiocina CXCL12/genética , Quimiocinas CXC/genética , Antígenos Comunes de Leucocito/genética , Osteoblastos/metabolismo , Osteoclastos/metabolismo , Receptores CXCR4/genética , Animales , Anticuerpos/farmacología , Células de la Médula Ósea/citología , Células de la Médula Ósea/efectos de los fármacos , Células de la Médula Ósea/metabolismo , Diferenciación Celular , Quimiocina CX3CL1/metabolismo , Quimiocina CXCL12/antagonistas & inhibidores , Quimiocina CXCL12/metabolismo , Quimiocinas CXC/metabolismo , Citometría de Flujo , Regulación de la Expresión Génica , Antígenos Comunes de Leucocito/deficiencia , Factor Estimulante de Colonias de Macrófagos/farmacología , Masculino , Ratones , Osteoblastos/citología , Osteoblastos/efectos de los fármacos , Osteoclastos/citología , Osteoclastos/efectos de los fármacos , Osteogénesis/genética , Cultivo Primario de Células , Ligando RANK/genética , Ligando RANK/metabolismo , Ligando RANK/farmacología , Receptor Activador del Factor Nuclear kappa-B/genética , Receptor Activador del Factor Nuclear kappa-B/metabolismo , Receptores CXCR4/metabolismo , Transducción de Señal , Tibia/citología , Tibia/efectos de los fármacos , Tibia/metabolismo
16.
Biochem Biophys Res Commun ; 476(4): 386-392, 2016 08 05.
Artículo en Inglés | MEDLINE | ID: mdl-27235552

RESUMEN

The blood-brain barrier (BBB) is mainly composed of brain capillary endothelial cells (BCECs), astrocytes and pericytes. Brain ischemia causes hypoxic encephalopathy and damages BBB. However, it remains still unclear how hypoxia affects BCECs. In the present study, t-BBEC117 cells, an immortalized bovine brain endothelial cell line, were cultured under hypoxic conditions at 4-5% oxygen for 72 h. This hypoxic stress caused hyperpolarization of resting membrane potential. Patch-clamp recordings revealed a marked increase in Ba(2+)-sensitive inward rectifier K(+) current in t-BBEC117 cells after hypoxic culture. Western blot and real-time PCR analyses showed that Kir2.1 expression was significantly up-regulated at protein level but not at mRNA level after the hypoxic culture. Ca(2+) imaging study revealed that the hypoxic stress enhanced store-operated Ca(2+) (SOC) entry, which was significantly reduced in the presence of 100 µM Ba(2+). On the other hand, the expression of SOC channels such as Orai1, Orai2, and transient receptor potential channels was not affected by hypoxic stress. MTT assay showed that the hypoxic stress significantly enhanced t-BBEC117 cell proliferation, which was inhibited by approximately 60% in the presence of 100 µM Ba(2+). We first show here that moderate cellular stress by cultivation under hypoxic conditions hyperpolarizes membrane potential via the up-regulation of functional Kir2.1 expression and presumably enhances Ca(2+) entry, resulting in the facilitation of BCEC proliferation. These findings suggest potential roles of Kir2.1 expression in functional changes of BCECs in BBB following ischemia.


Asunto(s)
Encéfalo/citología , Células Endoteliales/metabolismo , Canales de Potasio de Rectificación Interna/metabolismo , Animales , Calcio/metabolismo , Bovinos , Hipoxia de la Célula/fisiología , Proliferación Celular , Células Cultivadas , Potenciales de la Membrana , Canales de Potasio de Rectificación Interna/genética , Estrés Fisiológico , Regulación hacia Arriba
17.
J Pharmacol Sci ; 130(4): 219-25, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26976493

RESUMEN

We aimed to evaluate mineralocorticoid receptor (MR) expression in rat bladder and the physiological role of the MR-epithelial sodium channel (ENaC) pathway in controlling bladder function in 10-12-week-old, male Sprague-Dawley rats. First, we examined the mRNA expression of MR and localization of MR and ENaC-α proteins in the urinary bladder. MR mRNA expression was observed in untreated-rat urinary bladders, and MR and ENaC-α proteins were localized in the epithelium. Next, rats were treated with vehicle (controls) or fludrocortisone (an MR agonist) for 3 days, and ENaC-α protein expression levels and bladder function were evaluated on day 4. ENaC-α protein expression was significantly higher in fludrocortisone-treated rats than in controls. In addition, cystometry was performed during intravesical infusion of saline and amiloride (an ENaC inhibitor). While intercontraction intervals (ICIs) during saline infusion were significantly shorter in the fludrocortisone group than in the controls, infusion of amiloride normalized the ICIs in the fludrocortisone group. However, no intra- or inter-group differences in maximum intravesical pressure were observed. Taken together, MR protein is localized in the rat urinary bladder epithelium, and may regulate ENaC expression and bladder afferent input. The MR-ENaC pathway may be a therapeutic target for ameliorating storage symptoms.


Asunto(s)
Receptores de Mineralocorticoides/metabolismo , Receptores de Mineralocorticoides/fisiología , Canales de Sodio/fisiología , Vejiga Urinaria/metabolismo , Vejiga Urinaria/fisiología , Administración Intravesical , Amilorida/administración & dosificación , Amilorida/farmacología , Animales , Epitelio/metabolismo , Epitelio/fisiología , Fludrocortisona/farmacología , Expresión Génica , Masculino , ARN Mensajero/genética , ARN Mensajero/metabolismo , Ratas Sprague-Dawley , Receptores de Mineralocorticoides/genética , Transducción de Señal/genética , Transducción de Señal/fisiología , Canales de Sodio/metabolismo
18.
Microbiol Immunol ; 60(1): 47-55, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26616436

RESUMEN

Performance status (PS) frequently improves occurs in cancer patients who have been infused with their own lymphokine-activated killer T cells (LAK-T). In the present study, a culture supernatant of LAK-T (LAK-T sup) administered to 8-week-old rats caused neurogenesis as evidenced by increased 5-ethynyl-2'-deoxyuridine staining of brain tissues. Intravenous injection of granulocyte-macrophage colony stimulating factor (GM-CSF), a major cytokine in LAK-T sup, had a similar effect. Furthermore, LAK-T sup induced Ca(++) increase in rat hippocampal brain slices that was detected in neuronal cells by emission of Fluo-8 NW at 520 nm. The same effect was observed with an rGM-CSF solution. GM-CSF may activate neuronal cells by stimulating the glial cells that surround and attach to them. If so, GM-CSF and LAK-T sup may improve the motor neurons of patients with amyotrophic lateral sclerosis. The neurogenerative effect of GM-CSF in LAK-T sup may also help improve brain function in aged adults including those with dementia such as Alzheimer's disease.


Asunto(s)
Anticuerpos Monoclonales/farmacología , Interleucina-2/farmacología , Células Asesinas Activadas por Linfocinas/inmunología , Neuronas/inmunología , Linfocitos T Citotóxicos/efectos de los fármacos , Adulto , Enfermedad de Alzheimer/terapia , Esclerosis Amiotrófica Lateral/terapia , Animales , Encéfalo/efectos de los fármacos , Encéfalo/inmunología , Encéfalo/patología , Proliferación Celular/fisiología , Citocinas/inmunología , Citometría de Flujo , Factor Estimulante de Colonias de Granulocitos y Macrófagos/farmacología , Humanos , Inmunoterapia Adoptiva/métodos , Interleucina-2/inmunología , Células Asesinas Activadas por Linfocinas/citología , Células Asesinas Activadas por Linfocinas/efectos de los fármacos , Células Asesinas Activadas por Linfocinas/trasplante , Masculino , Neurogénesis/inmunología , Neuronas/citología , Neuronas/efectos de los fármacos , Ratas , Ratas Sprague-Dawley , Ratas Wistar , Linfocitos T Citotóxicos/inmunología , Regulación hacia Arriba
19.
Biochem Biophys Res Commun ; 459(3): 457-62, 2015 Apr 10.
Artículo en Inglés | MEDLINE | ID: mdl-25748572

RESUMEN

Store-operated Ca(2+) entry (SOCE) via Orai1 and STIM1 complex is supposed to have obligatory roles in the regulation of cellular functions of vascular endothelial cells, while little is known about the contribution of Orai2. Quantitative PCR and Western blot analyses indicated the expression of Orai2 and STIM2, in addition to Orai1 and STIM1 in bovine brain capillary endothelial cell line, t-BBEC117. During the exponential growth of t-BBEC117, the knockdown of Orai1 and STIM1 significantly reduced the SOCE activity, whereas Orai2 and STIM2 siRNAs had no effect. To examine whether endogenous SOCE activity contributes to the regulation of cell cycle progression, t-BBEC117 were synchronized using double thymidine blockage. At the G2/M phase, Ca(2+) influx via SOCE was decreased and Orai2 expression was increased compared to the G0/G1 phase. When Orai2 was knocked down at the G2/M phase, the decrease in SOCE was removed, and cell proliferation was partly attenuated. Taken together, Orai1 significantly contributes to cell proliferation via the functional expression, which is presumably independent of the cell cycle phases. In construct, Orai2 is specifically up-regulated during the G2/M phase, negatively modulates the SOCE activity, and may contribute to the regulation of cell cycle progression in brain capillary endothelial cells.


Asunto(s)
Encéfalo/irrigación sanguínea , Canales de Calcio/metabolismo , Señalización del Calcio , Animales , Barrera Hematoencefálica/citología , Barrera Hematoencefálica/metabolismo , Encéfalo/citología , Encéfalo/metabolismo , Canales de Calcio/genética , Bovinos , Puntos de Control del Ciclo Celular/fisiología , Línea Celular , Proliferación Celular , Células Endoteliales/citología , Células Endoteliales/metabolismo , Técnicas de Silenciamiento del Gen , Glicoproteínas de Membrana/antagonistas & inhibidores , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/metabolismo , ARN Interferente Pequeño/genética , Regulación hacia Arriba
20.
Cancer Lett ; 356(2 Pt B): 846-54, 2015 Jan 28.
Artículo en Inglés | MEDLINE | ID: mdl-25444894

RESUMEN

The human epidermal growth factor receptor 2 (HER2)-targeting agent, trastuzumab, is effective for HER2-overexpressing gastric cancer therapy. As oncolytic reovirus is currently undergoing clinical trials internationally, we wanted to explore whether combination therapy using trastuzumab and reovirus might provide a novel, more effective therapeutic option for gastric cancer. Cell proliferation and cell apoptosis were examined in vitro, while molecular analysis of pathways responsible for cell damage was examined using polymerase chain reaction array. Activation of the proteins related to apoptosis, cell growth and survival was detected by Western blotting. Mouse tumor xenograft models were used to examine antitumor activity in vivo. Reovirus sensitized HER2-overexpressing gastric cancer cells to undergo apoptosis. Both in vitro and in vivo studies provided evidence that the combination therapy is a more powerful modality against HER2-overexpressing gastric cancer cells than treatment using a single agent. Molecular analysis indicated that combination therapy induced significantly higher levels of tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) in cancer cells. Antibody against TRAIL strongly inhibited cell toxicity caused by the combined treatment. These data suggest that reovirus may augment trastuzumab-induced cytotoxicity in gastric cancer cells.


Asunto(s)
Anticuerpos Monoclonales Humanizados/farmacología , Viroterapia Oncolítica , Receptor ErbB-2/metabolismo , Reoviridae/genética , Neoplasias Gástricas/terapia , Ligando Inductor de Apoptosis Relacionado con TNF/metabolismo , Animales , Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Western Blotting , Proliferación Celular/efectos de los fármacos , Terapia Combinada , Humanos , Técnicas para Inmunoenzimas , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , ARN Mensajero/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Receptor ErbB-2/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Neoplasias Gástricas/genética , Neoplasias Gástricas/patología , Ligando Inductor de Apoptosis Relacionado con TNF/genética , Trastuzumab , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...