Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Sci Rep ; 12(1): 18636, 2022 11 03.
Artículo en Inglés | MEDLINE | ID: mdl-36329090

RESUMEN

Periodontitis is a chronic inflammatory disease characterized by the release of matrix metalloproteinases (MMPs) from resident connective tissue cells in tooth-supporting tissues (periodontium). Platelet activation, and the attendant release of pro-inflammatory chemokines such as platelet factor 4 (CXCL4/PF4), are associated with periodontitis although the associated biochemical pathways remain undefined. Here we report that recombinant PF4 is internalized by cultured human gingival fibroblasts (hGFs), resulting in significant (p < 0.05) upregulation in both the production and release of MMP-2 (gelatinase A). This finding was corroborated by elevated circulating levels of MMP-2 (p < 0.05) in PF4-overexpressing transgenic mice, relative to controls. We also determined that PF4 induces the phosphorylation of NF-κB; notably, the suppression of NF-κB signaling by the inhibitor BAY 11-7082 abrogated PF4-induced MMP-2 upregulation. Moreover, the inhibition of surface glycosaminoglycans (GAGs) blocked both PF4 binding and NF-κB phosphorylation. Partial blockade of PF4 binding to the cells was achieved by treatment with either chondroitinase ABC or heparinase III, suggesting that both chondroitin sulfate and heparan sulfate mediate PF4 signaling. These results identify a novel pathway in which PF4 upregulates MMP-2 release from fibroblasts in an NF-κB- and GAG-dependent manner, and further our comprehension of the role of platelet signaling in periodontal tissue homeostasis.


Asunto(s)
Metaloproteinasa 2 de la Matriz , Periodontitis , Ratones , Animales , Humanos , Metaloproteinasa 2 de la Matriz/genética , Metaloproteinasa 2 de la Matriz/metabolismo , Factor Plaquetario 4/metabolismo , FN-kappa B/metabolismo , Encía , Fibroblastos/metabolismo , Periodontitis/metabolismo , Inhibidores de la Angiogénesis/metabolismo , Metaloproteinasa 3 de la Matriz/metabolismo
2.
Cells ; 11(17)2022 08 31.
Artículo en Inglés | MEDLINE | ID: mdl-36078119

RESUMEN

The effects of electric fields (EFs) on various cell types have been thoroughly studied, and exhibit a well-known regulatory effect on cell processes, implicating their usage in several medical applications. While the specific effect exerted on cells is highly parameter-dependent, the majority of past research has focused primarily on low-frequency alternating fields (<1 kHz) and high-frequency fields (in the order of MHz). However, in recent years, low-intensity (1-3 V/cm) alternating EFs with intermediate frequencies (100-500 kHz) have been of topical interest as clinical treatments for cancerous tumours through their disruption of cell division and the mitotic spindle, which can lead to cell death. These aptly named tumour-treating fields (TTFields) have been approved by the FDA as a treatment modality for several cancers, such as malignant pleural mesothelioma and glioblastoma multiforme, demonstrating remarkable efficacy and a high safety profile. In this work, we report the results of in vitro experiments with HeLa and MCF-10A cells exposed to TTFields for 18 h, imaged in real time using live-cell imaging. Both studied cell lines were exposed to 100 kHz TTFields with a 1-1 duty cycle, which resulted in significant mitotic and cytokinetic arrest. In the experiments with HeLa cells, the effects of the TTFields' frequency (100 kHz vs. 200 kHz) and duty cycle (1-1 vs. 1-0) were also investigated. Notably, the anti-mitotic effect was stronger in the HeLa cells treated with 100 kHz TTFields. Additionally, it was found that single and two-directional TTFields (oriented orthogonally) exhibit a similar inhibitory effect on HeLa cell division. These results provide real-time evidence of the profound ability of TTFields to hinder the process of cell division by significantly delaying both the mitosis and cytokinesis phases of the cell cycle.


Asunto(s)
Glioblastoma , Mesotelioma Maligno , Glioblastoma/terapia , Células HeLa , Humanos , Mitosis , Huso Acromático
3.
Front Med Technol ; 4: 871196, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35600165

RESUMEN

We report the results of experimental investigations involving photobiomodulation (PBM) of living cells, tubulin, and microtubules in buffer solutions exposed to near-infrared (NIR) light emitted from an 810 nm LED with a power density of 25 mW/cm2 pulsed at a frequency of 10 Hz. In the first group of experiments, we measured changes in the alternating current (AC) ionic conductivity in the 50-100 kHz range of HeLa and U251 cancer cell lines as living cells exposed to PBM for 60 min, and an increased resistance compared to the control cells was observed. In the second group of experiments, we investigated the stability and polymerization of microtubules under exposure to PBM. The protein buffer solution used was a mixture of Britton-Robinson buffer (BRB aka PEM) and microtubule cushion buffer. Exposure of Taxol-stabilized microtubules (~2 µM tubulin) to the LED for 120 min resulted in gradual disassembly of microtubules observed in fluorescence microscopy images. These results were compared to controls where microtubules remained stable. In the third group of experiments, we performed turbidity measurements throughout the tubulin polymerization process to quantify the rate and amount of polymerization for PBM-exposed tubulin vs. unexposed tubulin samples, using tubulin resuspended to final concentrations of ~ 22.7 µM and ~ 45.5 µM in the same buffer solution as before. Compared to the unexposed control samples, absorbance measurement results demonstrated a slower rate and reduced overall amount of polymerization in the less concentrated tubulin samples exposed to PBM for 30 min with the parameters mentioned above. Paradoxically, the opposite effect was observed in the 45.5 µM tubulin samples, demonstrating a remarkable increase in the polymerization rates and total polymer mass achieved after exposure to PBM. These results on the effects of PBM on living cells, tubulin, and microtubules are novel, further validating the modulating effects of PBM and contributing to designing more effective PBM parameters. Finally, potential consequences for the use of PBM in the context of neurodegenerative diseases are discussed.

4.
Development ; 149(1)2022 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-35020896

RESUMEN

In early placental development, progenitor cytotrophoblasts (CTB) differentiate along one of two cellular trajectories: the villous or extravillous pathways. CTB committed to the villous pathway fuse with neighboring CTB to form the outer multinucleated syncytiotrophoblast (SCT), whereas CTB committed to the extravillous pathway differentiate into invasive extravillous trophoblasts (EVT). Unfortunately, little is known about the processes controlling human CTB progenitor maintenance and differentiation. To address this, we established a single cell RNA sequencing (scRNA-seq) dataset from first trimester placentas to identify cell states important in trophoblast progenitor establishment, renewal and differentiation. Multiple distinct trophoblast states were identified, representing progenitor CTB, column CTB, SCT precursors and EVT. Lineage trajectory analysis identified a progenitor origin that was reproduced in human trophoblast stem cell organoids. Heightened expression of basal cell adhesion molecule (BCAM) defined this primitive state, where BCAM enrichment or gene silencing resulted in enhanced or diminished organoid growth, respectively. Together, this work describes at high-resolution trophoblast heterogeneity within the first trimester, resolves gene networks within human CTB progenitors and identifies BCAM as a primitive progenitor marker and possible regulator.


Asunto(s)
Moléculas de Adhesión Celular/metabolismo , Linaje de la Célula , Sistema del Grupo Sanguíneo Lutheran/metabolismo , Trofoblastos/metabolismo , Adulto , Moléculas de Adhesión Celular/genética , Diferenciación Celular , Células Cultivadas , Femenino , Regulación del Desarrollo de la Expresión Génica , Redes Reguladoras de Genes , Células Madre Embrionarias Humanas/citología , Células Madre Embrionarias Humanas/metabolismo , Humanos , Sistema del Grupo Sanguíneo Lutheran/genética , Organoides/citología , Organoides/metabolismo , Trofoblastos/citología
5.
Development ; 147(2)2020 01 23.
Artículo en Inglés | MEDLINE | ID: mdl-31871275

RESUMEN

Early placental development and the establishment of the invasive trophoblast lineage take place within a low oxygen environment. However, conflicting and inconsistent findings have obscured the role of oxygen in regulating invasive trophoblast differentiation. In this study, the effect of hypoxic, normoxic and atmospheric oxygen on invasive extravillous pathway progression was examined using a human placental explant model. Here, we show that exposure to low oxygen enhances extravillous column outgrowth and promotes the expression of genes that align with extravillous trophoblast (EVT) lineage commitment. By contrast, supra-physiological atmospheric levels of oxygen promote trophoblast proliferation while simultaneously stalling EVT progression. Low oxygen-induced EVT differentiation coincided with elevated transcriptomic levels of lysyl oxidase (LOX) in trophoblast anchoring columns, in which functional experiments established a role for LOX activity in promoting EVT column outgrowth. The findings of this work support a role for low oxygen in potentiating the differentiation of trophoblasts along the extravillous pathway. In addition, these findings generate insight into new molecular processes controlled by oxygen during early placental development.


Asunto(s)
Diferenciación Celular/efectos de los fármacos , Linaje de la Célula/efectos de los fármacos , Oxígeno/farmacología , Proteína-Lisina 6-Oxidasa/metabolismo , Trofoblastos/citología , Adulto , Hipoxia de la Célula/efectos de los fármacos , Femenino , Regulación del Desarrollo de la Expresión Génica/efectos de los fármacos , Humanos , Modelos Biológicos , Transcriptoma/genética , Adulto Joven
6.
Respir Care ; 62(9): 1131-1136, 2017 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-28559463

RESUMEN

BACKGROUND: Neonatal respiratory distress results in > 1 million annual deaths worldwide. Bubble CPAP is a simple, effective, and widely used therapy for infants in respiratory distress. In low-resource settings, more advanced respiratory support is limited by cost, technical expertise, and sporadic electricity. We sought to develop a safe, inexpensive, and simple solution to provide further respiratory support for these infants. METHODS: A standard bubble CPAP system was modified to provide 2 levels of positive airway pressure (bi-level positive airway pressure) by attaching a novel device. To demonstrate reliability, the system was run with continuous pressure monitoring on full-term and preterm neonatal mannikins with pressure targets of 8/5 cm H2O and 15/5 cm H2O to simulate 2 different modes of noninvasive ventilation (NIV). RESULTS: At a ventilation rate set between 30 and 45 cycles/min, by adjusting the leak rate of the device, the following mean pressures ± SD were demonstrated: term mannikin low-pressure NIV, 7.9 ± 0.2/5.3 ± 0.2 cm H2O; term mannikin high-pressure NIV, 15.1 ± 0.1/6.1 ± 0.1 cm H2O; preterm mannikin low-pressure NIV, 7.9 ± 0.2/5.3 ± 0.2 cm H2O; preterm mannikin high-pressure NIV, 16.5 ± 0.4/5.1 ± 0.1 cm H2O. CONCLUSIONS: The modified bubble CPAP system reliably provided alternating pressures similar to bi-level positive airway pressure modes of respiratory support in neonatal mannikins. The dual-pressure technology is a simple, single connection add-on that can readily be applied to existing bubble CPAP systems.


Asunto(s)
Presión de las Vías Aéreas Positiva Contínua/instrumentación , Ventilación no Invasiva/instrumentación , Síndrome de Dificultad Respiratoria del Recién Nacido/terapia , Ventiladores Mecánicos , Presión de las Vías Aéreas Positiva Contínua/métodos , Humanos , Recién Nacido , Recien Nacido Prematuro , Maniquíes , Ventilación no Invasiva/métodos , Presión , Reproducibilidad de los Resultados
7.
J Biol Chem ; 290(15): 9886-95, 2015 Apr 10.
Artículo en Inglés | MEDLINE | ID: mdl-25694432

RESUMEN

Protein tyrosine phosphatase α (PTPα) promotes integrin-stimulated cell migration in part through the role of Src-phosphorylated PTPα-Tyr(P)-789 in recruiting and localizing p130Cas to focal adhesions. The growth factor IGF-1 also stimulates PTPα-Tyr-789 phosphorylation to positively regulate cell movement. This is in contrast to integrin-induced PTPα phosphorylation, that induced by IGF-1 can occur in cells lacking Src family kinases (SFKs), indicating that an unknown kinase distinct from SFKs can target PTPα. We show that this IGF-1-stimulated tyrosine kinase is Abl. We found that PTPα binds to the scaffold protein RACK1 and that RACK1 coordinates the IGF-1 receptor, PTPα, and Abl in a complex to enable IGF-1-stimulated and Abl-dependent PTPα-Tyr-789 phosphorylation. In cells expressing SFKs, IGF-1-stimulated phosphorylation of PTPα is mediated by RACK1 but is Abl-independent. Furthermore, expressing the SFKs Src and Fyn in SFK-deficient cells switches IGF-1-induced PTPα phosphorylation to occur in an Abl-independent manner, suggesting that SFK activity dominantly regulates IGF-1/IGF-1 receptor signaling to PTPα. RACK1 is a molecular scaffold that integrates growth factor and integrin signaling, and our identification of PTPα as a RACK1 binding protein suggests that RACK1 may coordinate PTPα-Tyr-789 phosphorylation in these signaling networks to promote cell migration.


Asunto(s)
Proteínas de Unión al GTP/metabolismo , Factor I del Crecimiento Similar a la Insulina/farmacología , Proteínas de Neoplasias/metabolismo , Proteínas Proto-Oncogénicas c-abl/metabolismo , Proteínas Tirosina Fosfatasas Clase 4 Similares a Receptores/metabolismo , Receptores de Superficie Celular/metabolismo , Animales , Línea Celular , Línea Celular Tumoral , Células Cultivadas , Fibroblastos/efectos de los fármacos , Fibroblastos/metabolismo , Proteínas de Unión al GTP/genética , Humanos , Immunoblotting , Células MCF-7 , Ratones , Proteínas de Neoplasias/genética , Fosforilación/efectos de los fármacos , Unión Proteica , Proteínas Proto-Oncogénicas c-abl/genética , Pirimidinas/farmacología , Interferencia de ARN , Receptor IGF Tipo 1/genética , Receptor IGF Tipo 1/metabolismo , Proteínas Tirosina Fosfatasas Clase 4 Similares a Receptores/genética , Receptores de Cinasa C Activada , Receptores de Superficie Celular/genética , Tirosina/metabolismo , Familia-src Quinasas/genética , Familia-src Quinasas/metabolismo
8.
J Biol Chem ; 289(3): 1345-54, 2014 Jan 17.
Artículo en Inglés | MEDLINE | ID: mdl-24302722

RESUMEN

Oxidative stress induced by reactive oxygen species (ROS) is associated with various neurological disorders including aging, neurodegenerative diseases, as well as traumatic and ischemic insults. Astrocytes have an important role in the anti-oxidative defense in the brain. The gap junction protein connexin43 (Cx43) forms intercellular channels as well as hemichannels in astrocytes. In the present study, we investigated the contribution of Cx43 to astrocytic death induced by the ROS hydrogen peroxide (H2O2) and the mechanism by which Cx43 exerts its effects. Lack of Cx43 expression or blockage of Cx43 channels resulted in increased ROS-induced astrocytic death, supporting a cell protective effect of functional Cx43 channels. H2O2 transiently increased hemichannel activity, but reduced gap junction intercellular communication (GJIC). GJIC in wild-type astrocytes recovered after 7 h, but was absent in Cx43 knock-out astrocytes. Blockage of Cx43 hemichannels incompletely inhibited H2O2-induced hemichannel activity, indicating the presence of other hemichannel proteins. Panx1, which is predicted to be a major hemichannel contributor in astrocytes, did not appear to have any cell protective effect from H2O2 insults. Our data suggest that GJIC is important for Cx43-mediated ROS resistance. In contrast to hypoxia/reoxygenation, H2O2 treatment decreased the ratio of the hypophosphorylated isoform to total Cx43 level. Cx43 has been reported to promote astrocytic death induced by hypoxia/reoxygenation. We therefore speculate the increase in Cx43 dephosphorylation may account for the facilitation of astrocytic death. Our findings suggest that the role of Cx43 in response to cellular stress is dependent on the activation of signaling pathways leading to alteration of Cx43 phosphorylation states.


Asunto(s)
Astrocitos/metabolismo , Comunicación Celular/fisiología , Conexina 43/metabolismo , Uniones Comunicantes/metabolismo , Estrés Oxidativo/fisiología , Transducción de Señal/fisiología , Animales , Astrocitos/citología , Comunicación Celular/efectos de los fármacos , Hipoxia de la Célula/efectos de los fármacos , Hipoxia de la Célula/fisiología , Conexina 43/genética , Conexinas/genética , Conexinas/metabolismo , Uniones Comunicantes/genética , Peróxido de Hidrógeno/farmacología , Ratones , Ratones Noqueados , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Oxidantes/farmacología , Estrés Oxidativo/efectos de los fármacos , Fosforilación/efectos de los fármacos , Fosforilación/fisiología , Transducción de Señal/efectos de los fármacos
9.
J Biol Chem ; 282(29): 20925-32, 2007 Jul 20.
Artículo en Inglés | MEDLINE | ID: mdl-17507376

RESUMEN

CD45 is a major membrane protein tyrosine phosphatase (PTP) expressed in T cells where it regulates the activity of Lck, a Src family kinase important for T cell receptor-mediated activation. PTPalpha is a more widely expressed transmembrane PTP that has been shown to regulate the Src family kinases, Src and Fyn, and is also present in T cells. Here, PTPalpha was phosphorylated at Tyr-789 in CD45(-) T cells but not in CD45(+) T cells suggesting that CD45 could regulate the phosphorylation of PTPalpha at this site. Furthermore, CD45 could directly dephosphorylate PTPalpha in vitro. Expression of PTPalpha and PTPalpha-Y789F in T cells revealed that the mutant had a reduced ability to decrease Fyn and Cbp phosphorylation, to regulate the kinase activity of Fyn, and to restore T cell receptor-induced signaling events when compared with PTPalpha. Conversely, this mutant had an increased ability to prevent Pyk2 phosphorylation and CD44-mediated cell spreading when compared with PTPalpha. These data demonstrate distinct activities of PTPalpha and PTPalpha-Y789F in T cells and identify CD45 as a regulator of PTPalpha phosphorylation at tyrosine 789 in T cells.


Asunto(s)
Regulación de la Expresión Génica , Mutación , Proteínas Tirosina Fosfatasas/genética , Proteínas Tirosina Fosfatasas/fisiología , Receptores de Superficie Celular/genética , Receptores de Superficie Celular/fisiología , Linfocitos T/metabolismo , Tirosina/química , Animales , Línea Celular Tumoral , Antígenos Comunes de Leucocito/biosíntesis , Linfoma de Células T/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Modelos Biológicos , Fosforilación , Proteínas Tirosina Fosfatasas Clase 4 Similares a Receptores , Transducción de Señal
10.
J Neurochem ; 98(6): 1798-809, 2006 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-16899073

RESUMEN

Mice lacking protein tyrosine phosphatase alpha (PTPalpha) exhibited defects in NMDA receptor (NMDAR)-associated processes such as learning and memory, hippocampal neuron migration, and CA1 hippocampal long-term potentiation (LTP). In vivo molecular effectors linking PTPalpha and the NMDAR have not been reported. Thus the involvement of PTPalpha as an upstream regulator of NMDAR tyrosine phosphorylation was investigated in synaptosomes of wild-type and PTPalpha-null mice. Tyrosine phosphorylation of the NMDAR NR2A and NR2B subunits was reduced upon PTPalpha ablation, indicating a positive effect of this phosphatase on NMDAR phosphorylation via intermediate molecules. The NMDAR is a substrate of src family tyrosine kinases, and reduced activity of src, fyn, yes and lck, but not lyn, was apparent in the absence of PTPalpha. In addition, autophosphorylation of proline-rich tyrosine kinase 2 (Pyk2), a tyrosine kinase linked to NMDAR signaling, was also reduced in PTPalpha-deficient synaptosomes. Altered protein tyrosine phosphorylation was not accompanied by altered expression of the NMDAR or the above tyrosine kinases at any stage of PTPalpha-null mouse development examined. In a human embryonic kidney (HEK) 293 cell expression system, PTPalpha enhanced fyn-mediated NR2A and NR2B tyrosine phosphorylation by several-fold. Together, these findings provide evidence that aberrant NMDAR-associated functions in PTPalpha-null mice are due to impaired NMDAR tyrosine phosphorylation resulting from the reduced activity of probably more than one of the src family kinases src, fyn, yes and lck. Defective NMDAR activity in these mice may also be linked to the loss of PTPalpha as an upstream regulator of Pyk2.


Asunto(s)
Quinasa 2 de Adhesión Focal/antagonistas & inhibidores , Proteínas Tirosina Fosfatasas/deficiencia , Proteínas Tirosina Fosfatasas/fisiología , Receptores de Superficie Celular/deficiencia , Receptores de Superficie Celular/fisiología , Receptores de N-Metil-D-Aspartato/metabolismo , Sinaptosomas/metabolismo , Tirosina/metabolismo , Familia-src Quinasas/antagonistas & inhibidores , Animales , Línea Celular , Humanos , Ratones , Ratones Noqueados , Fosforilación , Proteínas Proto-Oncogénicas c-fyn/metabolismo , Proteínas Tirosina Fosfatasas Clase 4 Similares a Receptores , Sinaptosomas/enzimología , Distribución Tisular
11.
J Immunol ; 175(12): 7947-56, 2005 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-16339530

RESUMEN

A role for the receptor protein tyrosine phosphatase alpha (PTPalpha) in immune cell function and regulation of Src family kinases was investigated using thymocytes from PTPalpha-deficient mice. PTPalpha-null thymocytes develop normally, but unstimulated PTPalpha-/- cells exhibit increased tyrosine phosphorylation of specific proteins, increased Fyn activity, and hyperphosphorylation of Cbp/PAG that promotes its association with C-terminal Src kinase. Elevated Fyn activity in the absence of PTPalpha is due to enhanced phosphorylation of Fyn tyrosines 528 and 417. Some PTPalpha is localized in lipid rafts of thymocytes, and raft-associated Fyn is specifically activated in PTPalpha-/- cells. PTPalpha is not a Cbp/PAG phosphatase, because it is not required for Cbp/PAG dephosphorylation in unstimulated or anti-CD3-stimulated thymocytes. Together, our results indicate that PTPalpha, likely located in lipid rafts, regulates the activity of raft Fyn. In the absence of PTPalpha this population of Fyn is activated and phosphorylates Cbp/PAG to enhance association with C-terminal Src kinase. Although TCR-mediated tyrosine phosphorylation was apparently unaffected by the absence of PTPalpha, the long-term proliferative response of PTPalpha-/- thymocytes was reduced. These findings indicate that PTPalpha is a component of the complex Src family tyrosine kinase regulatory network in thymocytes and is required to suppress Fyn activity in unstimulated cells in a manner that is not compensated for by the major T cell PTP and SFK regulator, CD45.


Asunto(s)
Microdominios de Membrana/metabolismo , Proteínas de la Membrana/metabolismo , Fosfoproteínas/metabolismo , Proteínas Tirosina Fosfatasas/fisiología , Proteínas Proto-Oncogénicas c-fyn/metabolismo , Timo/citología , Animales , Proteína Tirosina Quinasa CSK , Proliferación Celular , Activación Enzimática , Péptidos y Proteínas de Señalización Intercelular , Antígenos Comunes de Leucocito , Microdominios de Membrana/enzimología , Ratones , Ratones Noqueados , Fosforilación , Proteínas Tirosina Fosfatasas/deficiencia , Proteínas Tirosina Quinasas/metabolismo , Proteínas Tirosina Fosfatasas Clase 4 Similares a Receptores , Receptores de Superficie Celular/fisiología , Timo/ultraestructura , Familia-src Quinasas
12.
Biochem Biophys Res Commun ; 314(2): 321-9, 2004 Feb 06.
Artículo en Inglés | MEDLINE | ID: mdl-14733908

RESUMEN

Studies in cultured cells have implicated protein tyrosine phosphatase alpha (PTPalpha) as a potential regulator of insulin signaling. The physiological role of PTPalpha in insulin action was investigated using gene-targeted mice deficient in PTPalpha. PTPalpha-null animals had normal body weights and circulating levels of glucose and insulin in random fed and fasted states. In glucose and insulin tolerance tests, their efficiency of blood glucose clearance was comparable to wild-type mice. Kinetics and extents of insulin-stimulated insulin receptor and IRS-1 tyrosine phosphorylation were similar in wild-type and PTPalpha(-/-) liver, muscle, and adipose tissue. However, the association of IRS-1 and PI 3-K was altered in PTPalpha(-/-) liver, with increased insulin-independent and reduced insulin-stimulated association compared to wild-type samples. This did not affect activation of the downstream signaling effector Akt. Our data indicate that PTPalpha is not a negative regulator of insulin signaling and does not perform an essential role in mediating the physiological action of insulin.


Asunto(s)
Insulina/metabolismo , Proteínas Tirosina Fosfatasas/fisiología , Transducción de Señal , Animales , Glucemia/metabolismo , Peso Corporal , Electroforesis en Gel de Poliacrilamida , Glucosa/metabolismo , Homeostasis , Humanos , Immunoblotting , Insulina/sangre , Proteínas Sustrato del Receptor de Insulina , Cinética , Hígado/metabolismo , Ratones , Fosfatidilinositol 3-Quinasas/metabolismo , Fosfoproteínas/metabolismo , Fosforilación , Pruebas de Precipitina , Proteínas Tirosina Fosfatasas/genética , Proteínas Tirosina Fosfatasas Clase 4 Similares a Receptores , Factores de Tiempo , Distribución Tisular , Tirosina/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...