Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Cell Metab ; 32(6): 1041-1051.e6, 2020 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-33207244

RESUMEN

Diabetes is associated with increased mortality from severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2). Given literature suggesting a potential association between SARS-CoV-2 infection and diabetes induction, we examined pancreatic expression of angiotensin-converting enzyme 2 (ACE2), the key entry factor for SARS-CoV-2 infection. Specifically, we analyzed five public scRNA-seq pancreas datasets and performed fluorescence in situ hybridization, western blotting, and immunolocalization for ACE2 with extensive reagent validation on normal human pancreatic tissues across the lifespan, as well as those from coronavirus disease 2019 (COVID-19) cases. These in silico and ex vivo analyses demonstrated prominent expression of ACE2 in pancreatic ductal epithelium and microvasculature, but we found rare endocrine cell expression at the mRNA level. Pancreata from individuals with COVID-19 demonstrated multiple thrombotic lesions with SARS-CoV-2 nucleocapsid protein expression that was primarily limited to ducts. These results suggest SARS-CoV-2 infection of pancreatic endocrine cells, via ACE2, is an unlikely central pathogenic feature of COVID-19-related diabetes.


Asunto(s)
Enzima Convertidora de Angiotensina 2/genética , COVID-19/genética , Páncreas/metabolismo , SARS-CoV-2/fisiología , Internalización del Virus , Enzima Convertidora de Angiotensina 2/análisis , Enzima Convertidora de Angiotensina 2/metabolismo , COVID-19/complicaciones , COVID-19/metabolismo , Diabetes Mellitus Tipo 2/complicaciones , Diabetes Mellitus Tipo 2/genética , Diabetes Mellitus Tipo 2/metabolismo , Expresión Génica , Humanos , Páncreas/irrigación sanguínea , Serina Endopeptidasas/análisis , Serina Endopeptidasas/genética , Serina Endopeptidasas/metabolismo , Donantes de Tejidos
2.
Nat Commun ; 11(1): 3265, 2020 06 29.
Artículo en Inglés | MEDLINE | ID: mdl-32601271

RESUMEN

The culture of live pancreatic tissue slices is a powerful tool for the interrogation of physiology and pathology in an in vitro setting that retains near-intact cytoarchitecture. However, current culture conditions for human pancreatic slices (HPSs) have only been tested for short-term applications, which are not permissive for the long-term, longitudinal study of pancreatic endocrine regeneration. Using a culture system designed to mimic the physiological oxygenation of the pancreas, we demonstrate high viability and preserved endocrine and exocrine function in HPS for at least 10 days after sectioning. This extended lifespan allowed us to dynamically lineage trace and quantify the formation of insulin-producing cells in HPS from both non-diabetic and type 2 diabetic donors. This technology is expected to be of great impact for the conduct of real-time regeneration/developmental studies in the human pancreas.


Asunto(s)
Islotes Pancreáticos/citología , Páncreas/citología , Técnicas de Cultivo de Tejidos/métodos , Animales , Humanos , Estudios Longitudinales , Ratones , Modelos Biológicos , Regeneración , Células Madre/citología
4.
JCI Insight ; 5(8)2020 04 23.
Artículo en Inglés | MEDLINE | ID: mdl-32324170

RESUMEN

In type 1 diabetes (T1D), autoimmune destruction of pancreatic ß cells leads to insulin deficiency and loss of glycemic control. However, knowledge about human pancreas pathophysiology in T1D remains incomplete. To address this limitation, we established a pancreas tissue slice platform of donor organs with and without diabetes, facilitating the first live cell studies of human pancreas in T1D pathogenesis to our knowledge. We show that pancreas tissue slices from organ donors allow thorough assessment of processes critical for disease development, including insulin secretion, ß cell physiology, endocrine cell morphology, and immune infiltration within the same donor organ. Using this approach, we compared detailed pathophysiological profiles for 4 pancreata from donors with T1D with 19 nondiabetic control donors. We demonstrate that ß cell loss, ß cell dysfunction, alterations of ß cell physiology, and islet infiltration contributed differently to individual cases of T1D, allowing insight into pathophysiology and heterogeneity of T1D pathogenesis. Thus, our study demonstrates that organ donor pancreas tissue slices represent a promising and potentially novel approach in the search for successful prevention and reversal strategies of T1D.


Asunto(s)
Diabetes Mellitus Tipo 1/fisiopatología , Células Secretoras de Insulina/metabolismo , Células Secretoras de Insulina/patología , Páncreas/fisiopatología , Técnicas de Cultivo de Tejidos , Adolescente , Adulto , Niño , Preescolar , Femenino , Humanos , Masculino , Donantes de Tejidos , Adulto Joven
5.
IUBMB Life ; 67(8): 634-44, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-26099053

RESUMEN

A gradual decline in insulin response is known to precede the onset of type 1 diabetes (T1D). To track age-related changes in the ß-cell function of nonobese diabetic (NOD) mice, the most commonly used animal model for T1D, and to establish differences between those who do and do not become hyperglycemic, we performed a long-term longitudinal oral glucose tolerance test (OGTT) study (10-42 weeks) in combination with immunofluorescence imaging of islet morphology and cell proliferation. We observed a clear biphasic decline in insulin secretion (AUC0-30 min ) even in euglycemic animals. A first phase (10-28 weeks) consisted of a relatively rapid decline and paralleled diabetes development in the same cohort of animals. This was followed by a second phase (29-42 weeks) during which insulin secretion declined much slower while no additional animals became diabetic. Blood glucose profiles showed a corresponding, but less pronounced change: the area under the concentration curve (AUC0-150 min ) increased with age, and fit with a bilinear model indicated a rate-change in the trendline around 28 weeks. In control NOD scids, no such changes were observed. Islet morphology also changed with age as islets become surrounded by mononuclear infiltrates, and, in all mice, islets with immune cell infiltration around them showed increased ß-cell proliferation. In conclusion, insulin secretion declines in a biphasic manner in all NOD mice. This trend, as well as increased ß-cell proliferation, is present even in the NODs that never become diabetic, whereas, it is absent in control NOD scid mice.


Asunto(s)
Envejecimiento/metabolismo , Diabetes Mellitus Experimental/metabolismo , Diabetes Mellitus Tipo 1/metabolismo , Células Secretoras de Insulina/metabolismo , Insulina/metabolismo , Envejecimiento/patología , Animales , Glucemia , Proliferación Celular , Diabetes Mellitus Experimental/patología , Diabetes Mellitus Tipo 1/patología , Modelos Animales de Enfermedad , Prueba de Tolerancia a la Glucosa , Humanos , Secreción de Insulina , Células Secretoras de Insulina/patología , Ratones , Ratones Endogámicos NOD
6.
Biomed Eng Online ; 14: 28, 2015 Mar 28.
Artículo en Inglés | MEDLINE | ID: mdl-25889474

RESUMEN

BACKGROUND: In type 1 diabetic patients, who have lost their ability to produce insulin, transplantation of pancreatic islet cells can normalize metabolic control in a manner that is not achievable with exogenous insulin. To be successful, this procedure has to address the problems caused by the immune and autoimmune responses to the graft. Islet encapsulation using various techniques and materials has been and is being extensively explored as a possible approach. Within this framework, it is of considerable interest to characterize the effect encapsulation has on the insulin response of pancreatic islets. METHODS: To improve our ability to quantitatively describe the glucose-stimulated insulin release (GSIR) of pancreatic islets in general and of micro-encapsulated islets in particular, we performed dynamic perifusion experiments with frequent sampling. We used unencapsulated and microencapsulated murine islets in parallel and fitted the results with a complex local concentration-based finite element method (FEM) computational model. RESULTS: The high-resolution dynamic perifusion experiments allowed good characterization of the first-phase and second-phase insulin secretion, and we observed a slightly delayed and blunted first-phase insulin response for microencapsulated islets when compared to free islets. Insulin secretion profiles of both free and encapsulated islets could be fitted well by a COMSOL Multiphysics model that couples hormone secretion and nutrient consumption kinetics with diffusive and convective transport. This model, which was further validated and calibrated here, can be used for arbitrary geometries and glucose stimulation sequences and is well suited for the quantitative characterization of the insulin response of cultured, perifused, transplanted, or encapsulated islets. CONCLUSIONS: The present high-resolution GSIR experiments allowed for direct characterization of the effect microencapsulation has on the time-profile of insulin secretion. The multiphysics model, further validated here with the help of these experimental results, can be used to increase our understanding of the challenges that have to be faced in the design of bioartificial pancreas-type devices and to advance their further optimization.


Asunto(s)
Simulación por Computador , Glucosa/farmacología , Insulina/metabolismo , Islotes Pancreáticos/efectos de los fármacos , Islotes Pancreáticos/metabolismo , Animales , Cápsulas , Análisis de Elementos Finitos , Secreción de Insulina , Cinética , Ratones , Modelos Biológicos , Perfusión
7.
Steroids ; 85: 36-43, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24747770

RESUMEN

Glucocorticoids are an important class of anti-inflammatory/immunosuppressive drugs. A crucial part of their anti-inflammatory action results from their ability to repress proinflammatory transcription factors such as nuclear factor-κB (NF-κB) and activator protein-1 (AP-1) upon binding to the glucocorticoid receptor (GR). Accordingly, sensor cells quantifying their effect on inflammatory signal-induced NF-κB activation can provide useful information regarding their potencies as well as their transrepression abilities. Here, we report results obtained on their effect in suppressing both the TNFα- and the CD40L-induced activation of NF-κB in sensor cells that contain an NF-κB-inducible SEAP construct. In these cells, we confirmed concentration-dependent NF-κB activation for both TNFα and CD40L at low nanomolar concentrations (EC50). Glucocorticoids tested included hydrocortisone, prednisolone, dexamethasone, loteprednol etabonate, triamcinolone acetonide, beclomethasone dipropionate, and clobetasol propionate. They all caused significant, but only partial inhibition of these activations in concentration-dependent manners that could be well described by sigmoid response-functions. Despite the limitations of only partial maximum inhibitions, this cell-based assay could be used to quantitate the suppressing ability of glucocorticoids (transrepression potency) on the expression of proinflammatory transcription factors caused by two different cytokines in parallel both in a detailed, full dose-response format as well as in a simpler single-dose format. Whereas inhibitory potencies obtained in the TNF assay correlated well with consensus glucocorticoid potencies (receptor-binding affinities, Kd, RBA, at the GR) for all compounds, the non-halogenated steroids (hydrocortisone, prednisolone, and loteprednol etabonate) were about an order of magnitude more potent than expected in the CD40 assay in this system.


Asunto(s)
Ligando de CD40/biosíntesis , Inflamación/tratamiento farmacológico , FN-kappa B/biosíntesis , Factor de Transcripción AP-1/biosíntesis , Factor de Necrosis Tumoral alfa/biosíntesis , Antiinflamatorios/farmacología , Regulación de la Expresión Génica/efectos de los fármacos , Glucocorticoides/administración & dosificación , Glucocorticoides/química , Humanos , Inflamación/genética , Inflamación/patología , Transducción de Señal/efectos de los fármacos , Activación Transcripcional/efectos de los fármacos , Triamcinolona/administración & dosificación
8.
Stem Cells Transl Med ; 3(3): 277-89, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24375542

RESUMEN

The possibility of using human embryonic stem (hES) cell-derived ß cells as an alternative to cadaveric islets for the treatment of type 1 diabetes is now widely acknowledged. However, current differentiation methods consistently fail to generate meaningful numbers of mature, functional ß cells. In order to address this issue, we set out to explore the role of oxygen modulation in the maturation of pancreatic progenitor (PP) cells differentiated from hES cells. We have previously determined that oxygenation is a powerful driver of murine PP differentiation along the endocrine lineage of the pancreas. We hypothesized that targeting physiological oxygen partial pressure (pO2) levels seen in mature islets would help the differentiation of PP cells along the ß-cell lineage. This hypothesis was tested both in vivo (by exposing PP-transplanted immunodeficient mice to a daily hyperbaric oxygen regimen) and in vitro (by allowing PP cells to mature in a perfluorocarbon-based culture device designed to carefully adjust pO2 to a desired range). Our results show that oxygen modulation does indeed contribute to enhanced maturation of PP cells, as evidenced by improved engraftment, segregation of α and ß cells, body weight maintenance, and rate of diabetes reversal in vivo, and by elevated expression of pancreatic endocrine makers, ß-cell differentiation yield, and insulin production in vitro. Our studies confirm the importance of oxygen modulation as a key variable to consider in the design of ß-cell differentiation protocols and open the door to future strategies for the transplantation of fully mature ß cells.


Asunto(s)
Diabetes Mellitus Experimental/inmunología , Diabetes Mellitus Experimental/terapia , Células Madre Embrionarias/efectos de los fármacos , Huésped Inmunocomprometido , Células Secretoras de Insulina/efectos de los fármacos , Oxígeno/farmacología , Animales , Técnicas de Cultivo de Célula , Diferenciación Celular , Diabetes Mellitus Experimental/patología , Células Madre Embrionarias/citología , Células Madre Embrionarias/inmunología , Fluorocarburos/farmacología , Células Secretoras de Glucagón/citología , Células Secretoras de Glucagón/efectos de los fármacos , Células Secretoras de Glucagón/inmunología , Glucosa/metabolismo , Glucosa/farmacología , Supervivencia de Injerto , Humanos , Insulina/biosíntesis , Células Secretoras de Insulina/citología , Células Secretoras de Insulina/inmunología , Células Secretoras de Insulina/trasplante , Ratones , Ratones Desnudos , Consumo de Oxígeno/fisiología
9.
Cell Transplant ; 22(9): 1723-33, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23068091

RESUMEN

Conventional culture vessels are not designed for physiological oxygen (O2) delivery. Both hyperoxia and hypoxia-commonly observed when culturing cells in regular plasticware-have been linked to reduced cellular function and death. Pancreatic islets, used for the clinical treatment of diabetes, are especially sensitive to sub- and supraphysiological O2 concentrations. A result of current culture standards is that a high percentage of islet preparations are never transplanted because of cell death and loss of function in the 24-48 h postisolation. Here, we describe a new culture system designed to provide quasiphysiological oxygenation to islets in culture. The use of dishes where islets rest atop a perfluorocarbon (PFC)-based membrane, coupled with a careful adjustment of environmental O2 concentration to target the islet physiological pO2 range, resulted in dramatic gains in viability and function. These observations underline the importance of approximating culture conditions as closely as possible to those of the native microenvironment, and fill a widely acknowledged gap in our ability to preserve islet functionality in vitro. As stem cell-derived insulin-producing cells are likely to suffer from the same limitations as those observed in real islets, our findings are especially timely in the context of current efforts to define renewable sources for transplantation.


Asunto(s)
Técnicas de Cultivo de Célula/instrumentación , Técnicas de Cultivo de Célula/métodos , Fluorocarburos , Células Secretoras de Insulina/efectos de los fármacos , Células Secretoras de Insulina/fisiología , Oxígeno/administración & dosificación , Oxígeno/metabolismo , Animales , Muerte Celular/fisiología , Supervivencia Celular/fisiología , Femenino , Humanos , Células Secretoras de Insulina/citología , Células Secretoras de Insulina/metabolismo , Islotes Pancreáticos/citología , Ratones , Ratones Desnudos
10.
PLoS One ; 6(8): e22364, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21857924

RESUMEN

Alongside Pdx1 and Beta2/NeuroD, the transcription factor MafA has been shown to be instrumental in the maintenance of the beta cell phenotype. Indeed, a combination of MafA, Pdx1 and Ngn3 (an upstream regulator of Beta2/NeuroD) was recently reported to lead to the effective reprogramming of acinar cells into insulin-producing beta cells. These experiments set the stage for the development of new strategies to address the impairment of glycemic control in diabetic patients. However, the clinical applicability of reprogramming in this context is deemed to be poor due to the need to use viral vehicles for the delivery of the above factors. Here we describe a recombinant transducible version of the MafA protein (TAT-MafA) that penetrates across cell membranes with an efficiency of 100% and binds to the insulin promoter in vitro. When injected in utero into living mouse embryos, TAT-MafA significantly up-regulates target genes and induces enhanced insulin production as well as cytoarchitectural changes consistent with faster islet maturation. As the latest addition to our armamentarium of transducible proteins (which already includes Pdx1 and Ngn3), the purification and characterization of a functional TAT-MafA protein opens the door to prospective therapeutic uses that circumvent the use of viral delivery. To our knowledge, this is also the first report on the use of protein transduction in utero.


Asunto(s)
Insulina/metabolismo , Islotes Pancreáticos/metabolismo , Factores de Transcripción Maf de Gran Tamaño/metabolismo , Páncreas/metabolismo , Útero/metabolismo , Animales , Animales Recién Nacidos , Western Blotting , Línea Celular Tumoral , Células Cultivadas , Femenino , Expresión Génica , Productos del Gen tat/genética , Productos del Gen tat/metabolismo , Insulina/genética , Islotes Pancreáticos/citología , Factores de Transcripción Maf de Gran Tamaño/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Páncreas/embriología , Embarazo , Regiones Promotoras Genéticas/genética , Unión Proteica , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transfección
11.
Cell Transplant ; 17(7): 793-802, 2008.
Artículo en Inglés | MEDLINE | ID: mdl-19044206

RESUMEN

Pancreatic islet transplantation can provide insulin independence to diabetic patients. However, apoptosis of islets often leads to early graft failure. Genetic engineering with protective gene(s) can improve the viability of these cells. Here we show successful transduction of human islets with a feline immunodeficiency virus (FIV) vector expressing both a cytoprotective (cFLIP) gene and the green fluorescent protein (GFP). Despite using low virus titers to maximize safety, transduced islets expressed both genes, resulting in improved beta-cell metabolic activity and viability. Although only approximately 10% of total islet cells were transduced, the significant viability advantages suggest a "barrier" effect in which protecting the periphery of the islet shields the core. These results provide the first demonstration that a lentiviral vector can express two genes in islets. Furthermore, the engineered islets are resistant to a variety of apoptotic stimuli, suggesting the potential of this approach in enhancing the viability of transplanted cells.


Asunto(s)
Proteína Reguladora de Apoptosis Similar a CASP8 y FADD/genética , Técnicas de Transferencia de Gen , Vectores Genéticos/genética , Rechazo de Injerto/prevención & control , Proteínas Fluorescentes Verdes/genética , Virus de la Inmunodeficiencia Felina/genética , Islotes Pancreáticos/fisiología , Antibióticos Antineoplásicos/metabolismo , Apoptosis/fisiología , Proteína Reguladora de Apoptosis Similar a CASP8 y FADD/metabolismo , Citocinas/metabolismo , Doxorrubicina/metabolismo , Vectores Genéticos/metabolismo , Glucosa/metabolismo , Proteínas Fluorescentes Verdes/metabolismo , Humanos , Virus de la Inmunodeficiencia Felina/metabolismo , Insulina/metabolismo , Receptor fas/metabolismo
12.
Neurochem Res ; 30(5): 603-11, 2005 May.
Artículo en Inglés | MEDLINE | ID: mdl-16176063

RESUMEN

We studied pathways involved in the proliferation of rat C6 glioma cells induced by lysophosphatidic acid (LPA), a phospholipid with diverse biological functions. LPA induced a dose-responsive proliferation of C6 cells after 48 h. Proliferation was blocked by inhibitors of the sodium/proton exchanger type 1 (NHE1), Rho-associated kinase, the phosphatidylinositol 3-kinase/Akt pathway (PI3K/Akt), protein kinase C (PKC) and extracellular signal regulated kinase kinase (MEK). Phospho-specific antibodies were used to investigate the pathways involved. LPA induced transient (10 min) phosphorylations of ERK 1/2, Akt and the transcription factor CREB. The LPA-induced phosphorylation of ERK 1/2 and CREB was blocked by inhibition of PI3K, PKC and MEK, but that of Akt was only inhibited by wortmannin, the PI3K inhibitor. Inhibition of Rho kinase or NHE1 did not reduce the LPA-induced phosphorylation of ERK, Akt or CREB. The results were compared with the effects of LPA on transduction pathways in other cell types.


Asunto(s)
Proliferación Celular/efectos de los fármacos , Glioma/patología , Lisofosfolípidos/farmacología , Transducción de Señal , Proteínas de Transporte de Catión/metabolismo , Línea Celular Tumoral , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/metabolismo , Glioma/enzimología , Glioma/metabolismo , Humanos , Proteínas de la Membrana/metabolismo , Fosforilación , Proteínas Quinasas/metabolismo , Intercambiador 1 de Sodio-Hidrógeno , Intercambiadores de Sodio-Hidrógeno/metabolismo
13.
Chem Res Toxicol ; 16(11): 1433-9, 2003 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-14615969

RESUMEN

In this study, a new series of trihalomethyl-substituted pyrimidines and dihydropyrimidines were synthesized and tested as potential NTPDase inhibitors. For this purpose, synaptosomes from rat cerebral cortex were used as the enzyme source and ATP and ADP were used as the substrate. Among the new compounds, 4-methyl-2-methylsulfanyl-6-trichloromethylpyrimidine (2b) was found to be the most effective noncompetitive inhibitor, with an estimated K(i) value of 0.18 and 0.55 mM for ATP and ADP, respectively. Other pyrimidines inhibited NTPDase activity with the following rank order of inhibitory potency: 3,6-dimethyl-2-methylsulfanyl-4-trifluoromethyl-3,4-dihydro-pyrimidin-4-ol (3a) > 5-methyl-2-(4-methyl-6-trifluoromethyl-pyrimidin-2-yl)-3-trifluoromethyl-3,4-dihydro-2H-3-pyrazol-3-ol (6a) > 5-bromo-4,6-dimethoxy-4-trichloromethyl-1,2,3,4-tetrahydro-2-pyrimidin-2-one (9) for ATP and 6a > 9 > 3a for ADP. Our results demonstrate that a novel group of pyrimidines compounds can act as inhibitors of ATP and ADP hydrolysis in synaptosomes from rat cerebral cortex. These results can contribute for the understanding of the NTPDase activity and for further studies involving new compounds that can enlist as it inhibitors.


Asunto(s)
Adenosina Difosfato/antagonistas & inhibidores , Adenosina Trifosfato/antagonistas & inhibidores , Corteza Cerebral/efectos de los fármacos , Pirimidinas/farmacocinética , Sinaptosomas/efectos de los fármacos , Adenosina Difosfato/química , Adenosina Difosfato/metabolismo , Adenosina Trifosfato/química , Adenosina Trifosfato/metabolismo , Animales , Brasil , Corteza Cerebral/citología , Corteza Cerebral/enzimología , Inhibidores Enzimáticos/síntesis química , Inhibidores Enzimáticos/química , Inhibidores Enzimáticos/farmacocinética , Hidrólisis/efectos de los fármacos , Pirazoles/síntesis química , Pirazoles/farmacocinética , Pirimidinas/síntesis química , Pirimidinas/química , Pirimidinonas/síntesis química , Pirimidinonas/farmacocinética , Ratas , Sinaptosomas/enzimología
14.
Brain Res ; 946(1): 12-23, 2002 Aug 09.
Artículo en Inglés | MEDLINE | ID: mdl-12133590

RESUMEN

Primary cultures of astrocytes exhibit a polygonal morphology, but on treatment with agents that increase cAMP they change to stellate cells. We found that astrocyte stellation also occurred on replacing the culture medium with saline buffered with HEPES. However, stellation did not occur when the medium was replaced with saline buffered with bicarbonate/CO(2) provided Ca(2+) was present. Since exposure of astrocytes to media lacking bicarbonate results in a decrease in intracellular pH (pH(i)) we sought evidence for an association between pH(i) and morphology. Astrocytic pH(i) was monitored for 60 min after transferring the cells to HEPES or bicarbonate-buffered saline. HEPES-induced stellation was associated with transient acidification which coincided with the morphological changes. Acidification was not observed in cells transferred to bicarbonate-saline. However when cytoplasmic acidification of cells in bicarbonate-saline was induced pharmacologically, rapid stellation occurred. Stellation induced by cAMP is reversed by activation of the RhoA pathway with lysophosphatidic acid (LPA). Here we found that LPA inhibited HEPES-induced stellation, but only with Ca(2+) present. Inhibition of stellation by LPA+Ca(2+) was associated with transient acidification followed by modest alkanization. A close association of tyrosine phosphorylation with stellation and pH(i) was observed. Thus incubation of astrocytes in HEPES-saline with orthovanadate to inhibit dephosphorylation abolished stellation and acidification; conversely incubation of cells in bicarbonate-saline with genistein to inhibit tyrosine kinases caused stellation and major acidification. Acidification may be one of several factors resulting in stellation, but it is not a necessary factor since stellation without acidification was observed in bicarbonate-saline lacking Ca(2+).


Asunto(s)
Astrocitos/citología , Astrocitos/efectos de los fármacos , Cloruro de Sodio/farmacología , Cloruro de Amonio/farmacología , Animales , Bicarbonatos/farmacología , Cationes/análisis , Tamaño de la Célula/efectos de los fármacos , Células Cultivadas , Medios de Cultivo/química , Medios de Cultivo/farmacología , Combinación de Medicamentos , HEPES/farmacología , Concentración de Iones de Hidrógeno , Membranas Intracelulares/metabolismo , Litio/farmacología , Lisofosfolípidos/farmacología , Fosforilación , Potasio/farmacología , Ratas , Ratas Wistar , Intercambiadores de Sodio-Hidrógeno/farmacología , Tirosina/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...