Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
1.
J Neurosci ; 43(6): 885-901, 2023 02 08.
Artículo en Inglés | MEDLINE | ID: mdl-36535770

RESUMEN

Memory formation and maintenance is a dynamic process involving the modulation of the actin cytoskeleton at synapses. Understanding the signaling pathways that contribute to actin modulation is important for our understanding of synapse formation and function, as well as learning and memory. Here, we focused on the importance of the actin regulator, noncatalytic region of tyrosine kinase adaptor protein 1 (NCK1), in hippocampal dependent behaviors and development. We report that male mice lacking NCK1 have impairments in both short-term and working memory, as well as spatial learning. Additionally, we report sex differences in memory impairment showing that female mice deficient in NCK1 fail at reversal learning in a spatial learning task. We find that NCK1 is expressed in postmitotic neurons but is dispensable for neuronal proliferation and migration in the developing hippocampus. Morphologically, NCK1 is not necessary for overall neuronal dendrite development. However, neurons lacking NCK1 have lower dendritic spine and synapse densities in vitro and in vivo EM analysis reveal increased postsynaptic density (PSD) thickness in the hippocampal CA1 region of NCK1-deficient mice. Mechanistically, we find the turnover of actin-filaments in dendritic spines is accelerated in neurons that lack NCK1. Together, these findings suggest that NCK1 contributes to hippocampal-dependent memory by stabilizing actin dynamics and dendritic spine formation.SIGNIFICANCE STATEMENT Understanding the molecular signaling pathways that contribute to memory formation, maintenance, and elimination will lead to a better understanding of the genetic influences on cognition and cognitive disorders and will direct future therapeutics. Here, we report that the noncatalytic region of tyrosine kinase adaptor protein 1 (NCK1) adaptor protein modulates actin-filament turnover in hippocampal dendritic spines. Mice lacking NCK1 show sex-dependent deficits in hippocampal memory formation tasks, have altered postsynaptic densities, and reduced synaptic density. Together, our work implicates NCK1 in the regulation of actin cytoskeleton dynamics and normal synapse development which is essential for memory formation.


Asunto(s)
Actinas , Espinas Dendríticas , Animales , Femenino , Masculino , Ratones , Actinas/metabolismo , Espinas Dendríticas/metabolismo , Hipocampo/metabolismo , Neuronas/fisiología , Proteínas Tirosina Quinasas/metabolismo , Sinapsis/fisiología , Memoria
2.
Basic Res Cardiol ; 117(1): 20, 2022 04 07.
Artículo en Inglés | MEDLINE | ID: mdl-35389129

RESUMEN

Accumulating evidence suggests that autophagy dysfunction plays a critical role in myocardial ischemia/reperfusion (I/R) injury. However, the underling mechanism of malfunctional autophagy in the cardiomyocytes subjected to I/R has not been well defined. As a result, there is no effective therapeutic option by targeting autophagy to prevent myocardial I/R injury. Here, we used both an in vitro and an in vivo I/R model to monitor autophagic flux in the cardiomyocytes, by exposing neonatal rat ventricular myocytes to hypoxia/reoxygenation and by subjecting mice to I/R, respectively. We observed that the autophagic flux in the cardiomyocytes subjected to I/R was blocked in both in vitro and in vivo models. Down-regulating a lysosomal cationic channel, TRPML1, markedly restored the blocked myocardial autophagic flux induced by I/R, demonstrating that TRPML1 directly contributes to the blocked autophagic flux in the cardiomyocytes subjected to I/R. Mechanistically, TRPML1 is activated secondary to ROS elevation following ischemia/reperfusion, which in turn induces the release of lysosomal zinc into the cytosol and ultimately blocks the autophagic flux in cardiomyocytes, presumably by disrupting the fusion between autophagosomes and lysosomes. As a result, the inhibited myocardial autophagic flux induced by TRPML1 disrupted mitochondria turnover and resulted in mass accumulation of damaged mitochondria and further ROS release, which directly led to cardiomyocyte death. More importantly, pharmacological and genetic inhibition of TRPML1 channels greatly reduced infarct size and rescued heart function in mice subjected to I/R in vivo by restoring impaired myocardial autophagy. In summary, our study demonstrates that secondary to ROS elevation, activation of TRPML1 results in autophagy inhibition in the cardiomyocytes subjected to I/R, which directly leads to cardiomyocyte death by disrupting mitochondria turnover. Therefore, targeting TRPML1 represents a novel therapeutic strategy to protect against myocardial I/R injury.


Asunto(s)
Daño por Reperfusión Miocárdica , Animales , Apoptosis , Autofagia , Ratones , Daño por Reperfusión Miocárdica/tratamiento farmacológico , Miocardio , Miocitos Cardíacos , Ratas , Especies Reactivas de Oxígeno
3.
J Neurochem ; 134(6): 1008-14, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-25913325

RESUMEN

As our understanding of motor circuit function increases, our need to understand how circuits form to ensure proper function becomes increasingly important. Recently, deleted in colorectal cancer (DCC) has been shown to be important in the development of spinal circuits necessary for gait. Importantly, humans with mutation in DCC show mirror movement disorders pointing to the significance of DCC in the development of spinal circuits for coordinated movement. Although DCC binds a number of ligands, the intracellular signaling cascade leading to the aberrant spinal circuits remains unknown. Here, we show that the non-catalytic region of tyrosine kinase adaptor (NCK) proteins 1 and 2 are distributed in the developing spinal cord. Using dissociated dorsal spinal neuron cultures we show that NCK proteins are necessary for the outgrowth and growth cone architecture of DCC(+ve) dorsal spinal neurons. Consistent with a role for NCK in DCC signaling, we show that loss of NCK proteins leads to a reduction in the thickness of TAG1(+ve) commissural bundles in the floor plate and loss of DCC mRNA in vivo. We suggest that DCC signaling functions through NCK1 and NCK2 and that both proteins are necessary for the establishment of normal spinal circuits necessary for gait. Reduction in NCK proteins in the developing CNS leads to a reduction in TAG1(+ve) commissural tract thickness, a reduction in growth cone complexity of DCC(+ve) spinal interneurons, and a reduction in DCC mRNA. These are consistent with an in vivo role for NCK in the development of critical DCC spinal circuits, and may be important for the normal development of spinal circuits critical for walking.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Vías Nerviosas/embriología , Neurogénesis/fisiología , Proteínas Oncogénicas/metabolismo , Receptores de Superficie Celular/metabolismo , Médula Espinal/embriología , Proteínas Supresoras de Tumor/metabolismo , Animales , Receptor DCC , Conos de Crecimiento/metabolismo , Células HEK293 , Humanos , Inmunoprecipitación , Ratones , Ratones Noqueados , Vías Nerviosas/metabolismo , Neuronas/metabolismo , Médula Espinal/metabolismo , Transfección
4.
Autophagy ; : 1-11, 2024 Mar 24.
Artículo en Inglés | MEDLINE | ID: mdl-38522082

RESUMEN

MCOLN1/TRPML1 is a nonselective cationic channel specifically localized to the late endosome and lysosome. With its property of mediating the release of several divalent cations such as Ca2+, Zn2+ and Fe2+ from the lysosome to the cytosol, MCOLN1 plays a pivotal role in regulating a variety of cellular events including endocytosis, exocytosis, lysosomal biogenesis, lysosome reformation, and especially in Macroautophagy/autophagy. Autophagy is a highly conserved catabolic process that maintains cytoplasmic integrity by removing superfluous proteins and damaged organelles. Acting as the terminal compartments, lysosomes are crucial for the completion of the autophagy process. This review delves into the emerging role of MCOLN1 in controlling the autophagic process by regulating lysosomal ionic homeostasis, thereby governing the fundamental functions of lysosomes. Furthermore, this review summarizes the physiological relevance as well as molecular mechanisms through which MCOLN1 orchestrates autophagy, consequently influencing mitochondria turnover, cell apoptosis and migration. In addition, we have illustrated the implications of MCOLN1-regulated autophagy in the pathological process of cancer and myocardial ischemia-reperfusion (I/R) injury. In summary, given the involvement of MCOLN1-mediated autophagy in the pathogenesis of cancer and myocardial I/R injury, targeting MCOLN1 May provide clues for developing new therapeutic strategies for the treatment of these diseases. Exploring the regulation of MCOLN1-mediated autophagy in diverse diseases contexts will surely broaden our understanding of this pathway and offer its potential as a promising drug target.Abbreviation: CCCP:carbonyl cyanide3-chlorophenylhydrazone; CQ:chloroquine; HCQ: hydroxychloroquine;I/R: ischemia-reperfusion; MAP1LC3/LC3:microtubule associated protein 1 light chain 3; MCOLN1/TRPML1:mucolipin TRP cation channel 1; MLIV: mucolipidosis type IV; MTORC1:MTOR complex 1; ROS: reactive oxygenspecies; SQSTM1/p62: sequestosome 1.

5.
Autophagy ; 18(12): 3053-3055, 2022 12.
Artículo en Inglés | MEDLINE | ID: mdl-35491864

RESUMEN

Accumulating evidence suggests that macroautophagy/autophagy dysfunction plays a critical role in myocardial ischemia-reperfusion (I/R) injury. However, the underlying mechanisms responsible for malfunctional autophagy in cardiomyocytes subjected to I/R are poorly understood. As a result, there are no effective therapeutic options that target autophagy to prevent myocardial I/R injury. We recently revealed that MCOLN1/TRPML1, a lysosomal cationic channel, directly contributes to the inhibition of autophagic flux in cardiomyocytes post I/R. We found that MCOLN1 is activated secondary to reactive oxygen species (ROS) elevation following I/R, which in turn induces the release of lysosomal zinc into the cytosol. This ultimately blocks autophagic flux in cardiomyocytes by disrupting the fusion between autophagosomes containing engulfed mitochondria and lysosomes. Furthermore, we discovered that the MCOLN1-mediated inhibition of autophagy induced by I/R impairs mitochondrial function, which results in further detrimental ROS release that directly contributes to cardiomyocyte death. More importantly, restoration of blocked autophagic flux in cardiomyocytes subjected to I/R achieved by blocking MCOLN1 channels significantly rescues cardiomyocyte death in vitro and greatly improves cardiac function of mice subjected to I/R in vivo. Therefore, targeting MCOLN1 represents a novel therapeutic strategy to protect against myocardial I/R injury.Abbreviations: I/R: ischemia-reperfusion; MAP1LC3/LC3: microtubule associated protein 1 light chain 3; MCOLN1/TRPML1: mucolipin TRP cation channel 1; ROS: reactive oxygen species; SQSTM1/p62: sequestosome 1.


Asunto(s)
Daño por Reperfusión Miocárdica , Canales de Potencial de Receptor Transitorio , Ratones , Animales , Miocitos Cardíacos/metabolismo , Autofagia , Especies Reactivas de Oxígeno/metabolismo , Daño por Reperfusión Miocárdica/prevención & control , Autofagosomas/metabolismo , Canales de Potencial de Receptor Transitorio/metabolismo
6.
Cancer Lett ; 541: 215752, 2022 08 10.
Artículo en Inglés | MEDLINE | ID: mdl-35644286

RESUMEN

Previous studies have demonstrated that autophagy tightly regulates apoptosis. However, the underlying mechanism whereby autophagy regulates apoptosis remains unclear. Here, we discover a "autophagy inhibition-mitochondrial turnover disruption-ROS elevation-DNA damage-p53 transactivation-apoptosis" axis that explicates the process of autophagy modulating apoptosis. We found that autophagy inhibition induced by TRPML1, a cationic channel localized in the lysosome, results in accumulation of damaged mitochondria via blocking the mitophagic flux to lysosomes in human melanoma and glioblastoma cells. The disrupted mitochondria turnover leads to ROS elevation, which in turn causes severe damage to DNA in these cancer cells. Damage to DNA resulted from TRPML1-mediated autophagy inhibition subsequently activates p53, which ultimately triggers mitochondrial mediated apoptosis by modulating pro- and anti-apoptosis proteins in these cancer cells. As a result, by triggering apoptosis, TRPML1-induced autophagy inhibition greatly suppresses growth of human melanoma and glioma both in vitro and in vivo. In summary, our findings define the mechanism underling the regulation of autophagy inhibition in apoptosis and represent TRPML1 as a novel target for potentially treating melanoma and glioblastoma in the clinical setting.


Asunto(s)
Glioblastoma , Melanoma , Canales de Potencial de Receptor Transitorio/metabolismo , Apoptosis , Autofagia , Glioblastoma/tratamiento farmacológico , Glioblastoma/genética , Glioblastoma/metabolismo , Humanos , Lisosomas/metabolismo , Melanoma/metabolismo , Mitocondrias/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Proteína p53 Supresora de Tumor/genética , Proteína p53 Supresora de Tumor/metabolismo
7.
Cancer Lett ; 525: 179-197, 2022 01 28.
Artículo en Inglés | MEDLINE | ID: mdl-34752845

RESUMEN

The transient receptor potential melastatin-subfamily member 7 (TRPM7) is a ubiquitous cation channel possessing kinase activity. TRPM7 mediates a variety of physiological responses by conducting flow of cations such as Ca2+, Mg2+, and Zn2+. Here, we show that the activation of TRPM7 channel stimulated by chemical agonists of TRPM7, Clozapine or Naltriben, inhibited autophagy via mediating Zn2+ release to the cytosol, presumably from the intracellular Zn2+-accumulating vesicles where TRPM7 localizes. Zn2+ release following the activation of TRPM7 disrupted the fusion between autophagosomes and lysosomes by disturbing the interaction between Sxt17 and VAMP8 which determines fusion status of autophagosomes and lysosomes. Ultimately, the disrupted fusion resulting from stimulation of TRPM7 channels arrested autophagy. Functionally, we demonstrate that the autophagy inhibition mediated by TRPM7 triggered cell death and suppressed metastasis of cancer cells in vitro, more importantly, restricted tumor growth and metastasis in vivo, by evoking apoptosis, cell cycle arrest, and reactive oxygen species (ROS) elevation. These findings represent a strategy for stimulating TRPM7 to combat cancer.


Asunto(s)
Neoplasias/tratamiento farmacológico , Células Madre Neoplásicas/efectos de los fármacos , Proteínas Serina-Treonina Quinasas/genética , Proteínas R-SNARE/genética , Canales Catiónicos TRPM/genética , Apoptosis/efectos de los fármacos , Autofagosomas/efectos de los fármacos , Autofagia/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Clozapina/farmacología , Humanos , Lisosomas/efectos de los fármacos , Naltrexona/análogos & derivados , Naltrexona/farmacología , Metástasis de la Neoplasia , Neoplasias/genética , Transducción de Señal/efectos de los fármacos , Canales Catiónicos TRPM/agonistas , Zinc/farmacología
8.
Autophagy ; 18(8): 1932-1954, 2022 08.
Artículo en Inglés | MEDLINE | ID: mdl-34878954

RESUMEN

Compelling evidence has demonstrated that macroautophagy/autophagy plays an important role in regulating multiple steps of metastatic cascades; however, the precise role of autophagy in metastasis remains unclear. This study demonstrates that autophagy inhibition induced by MCOLN1/TRPML1 suppresses cancer metastasis by evoking the ROS-mediated TP53/p53 pathway. First, we found that MCOLN1-mediated autophagy inhibition not only profoundly inhibits both migration and invasion in malignant melanoma and glioma cell lines in vitro, but also suppresses melanoma metastasis in vivo. Second, our study reveals that autophagy inhibition induced by MCOLN1 leads to damaged mitochondria accumulation followed by large quantities of ROS release. Third, we demonstrate that the elevated ROS resulting from autophagy inhibition subsequently triggers TP53 activity, which in turn modulates expression of its downstream targets that are involved in a broad spectrum of the metastatic cascade to suppress metastasis including MMP members and TWIST. In summary, our findings have established a mechanism by which autophagy inhibition suppresses metastasis via the ROS-TP53 signaling pathway. More importantly, our study demonstrates that autophagy inhibition through stimulation of MCOLN1 could evidently be one of the therapeutic potentials for combating cancer metastasis.Abbreviations: 3-MA: 3-methyladenine; AA: amino acid; ATG5: autophagy related 5; ATG12: autophagy-related 12; Baf-A1: bafilomycin A1; CCCP: carbonyl cyanide m-chlorophenylhydrazone; CQ: chloroquine; DMEM: Dulbecco's Modified Eagle Medium; EMT: epithelial-mesenchymal transition; FBS: fetal bovine serum; GAPDH: glyceraldehyde-3-phosphate dehydrogenase; HEK: human embryonic kidney; LAMP1: lysosomal-associated membrane protein 1; MAP1LC3/LC3: microtubule-associated protein 1 light chain 3; MCOLN1/TRPML1: mucolipin TRP cation channel 1; MMP: matrix metallopeptidase; NC: negative control; NRK: normal rat kidney; PBS: phosphate-buffered saline; shRNA: short hairpin RNA; siRNA: short interfering RNA; SQSTM1/p62: sequestosome 1; ULK1: unc-51 like autophagy-activating kinase 1.


Asunto(s)
Neoplasias , Canales de Potencial de Receptor Transitorio , Autofagia/fisiología , Humanos , Mitocondrias/metabolismo , Metástasis de la Neoplasia , Neoplasias/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Canales de Potencial de Receptor Transitorio/metabolismo , Proteína p53 Supresora de Tumor/metabolismo
9.
Pulm Pharmacol Ther ; 24(1): 42-8, 2011 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-20970516

RESUMEN

Prostaglandin (PG)E2 mediates its effects via activation of four distinct PGE2 receptors, termed EP1₋4, all of which are present on the model human airway epithelial cell line, Calu-3. We previously reported that acute activation of the EP4 subtype of the PGE2 receptor is associated with increased anion efflux from these cells, via the CFTR chloride channel. In the present study we examine the effects of longer term activation of the EP4 receptor in Calu-3 cells in an attempt to determine whether this would prove beneficial or detrimental to the airway epithelial cell environment. Using PGE1-OH, an EP4 receptor selective agonist, we determined that EP4 receptor activation was associated with increased phosphorylation of extracellular signal-related kinases (ERKs) and induction of the transcription factor early growth response factor-1 (Egr-1). Additionally, using specific enzyme-linked immunosorbent assays and quantitative PCR, we detected increased production of PGE2, IL-6, IL-8 and the chemokine monocyte chemotactic protein-1 (MCP-1) at both the protein and gene level in response to EP4 receptor activation. Intriguingly, the enhanced production of PGE2 in response to EP4 receptor activation raises the possibility of a positive feedback situation. Generally, within the airways, PGE2 is considered to have pro-inflammatory effects, whilst the enhanced production of IL-6, IL-8 and MCP-1 would be associated with the recruitment and activation of inflammatory cells to the airways. Thus, we conclude that chronic activation of the EP4 receptor is associated with increased production of mediators likely to increase the pro-inflammatory milieu of airway epithelial cells.


Asunto(s)
Citocinas/biosíntesis , Dinoprostona/biosíntesis , Células Epiteliales/metabolismo , Subtipo EP4 de Receptores de Prostaglandina E/fisiología , Tráquea/metabolismo , Células Cultivadas , Quimiocina CCL2/biosíntesis , Proteína 1 de la Respuesta de Crecimiento Precoz/análisis , Activación Enzimática , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Humanos , Interleucina-6/biosíntesis , Interleucina-8/biosíntesis
10.
Autophagy ; 17(12): 4401-4422, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-33890549

RESUMEN

Macroautophagy/autophagy is elevated to ensure the high demand for nutrients for the growth of cancer cells. Here we demonstrated that MCOLN1/TRPML1 is a pharmaceutical target of oncogenic autophagy in cancers such as pancreatic cancer, breast cancer, gastric cancer, malignant melanoma, and glioma. First, we showed that activating MCOLN1, by increasing expression of the channel or using the MCOLN1 agonists, ML-SA5 or MK6-83, arrests autophagic flux by perturbing fusion between autophagosomes and lysosomes. Second, we demonstrated that MCOLN1 regulates autophagy by mediating the release of zinc from the lysosome to the cytosol. Third, we uncovered that zinc influx through MCOLN1 blocks the interaction between STX17 (syntaxin 17) in the autophagosome and VAMP8 in the lysosome and thereby disrupting the fusion process that is determined by the two SNARE proteins. Furthermore, we demonstrated that zinc influx originating from the extracellular fluid arrests autophagy by the same mechanism as lysosomal zinc, confirming the fundamental function of zinc as a participant in membrane trafficking. Last, we revealed that activating MCOLN1 with the agonists, ML-SA5 or MK6-83, triggers cell death of a number of cancer cells by evoking autophagic arrest and subsequent apoptotic response and cell cycle arrest, with little or no effect observed on normal cells. Consistent with the in vitro results, administration of ML-SA5 in Patu 8988 t xenograft mice profoundly suppresses tumor growth and improves survival. These results establish that a lysosomal cation channel, MCOLN1, finely controls oncogenic autophagy in cancer by mediating zinc influx into the cytosol.Abbreviation: Abbreviations: 3-MA: 3-methyladenine; AA: amino acid; ATG12: autophagy related 12; Baf-A1: bafilomycin A1; BAPTA-am: 1,2-bis(2-aminophenoxy)ethane-N, N,N',N'-tetraacetic acid tetrakis-acetoxymethyl ester; co-IP: coimmunoprecipitaion; CQ: chloroquine; DMEM: Dulbecco's Modified Eagle Medium; FBS: fetal bovine serum; GAPDH: glyceraldehyde- 3-phosphate dehydrogenase; HCQ: hydroxychloroquine; HEK: human embryonic kidney; LAMP1: lysosomal associated membrane protein 1; MAP1LC3/LC3: microtubule associated protein 1 light chain 3; MCOLN1/TRPML1: mucolipin TRP cation channel 1; MTORC1: mechanistic target of rapamycin kinase complex 1; NC: negative control; NRK: normal rat kidney epithelial cells; PBS: phosphate-buffered saline; PtdIns3K: phosphatidylinositol 3-kinase; RPS6KB/S6K: ribosomal protein S6 kinase B; shRNA: short hairpin RNA; siRNA: short interfering RNA; SNARE: soluble N-ethylmaleimide-sensitive factor (NSF) attachment protein receptor; SQSTM1/p62: sequestosome 1; STX17: syntaxin 17; TPEN: N,N,N',N'-tetrakis(2-pyridylmethyl)ethylenediamine; TTM: tetrathiomolybdate; ULK1: unc-51 like autophagy activating kinase 1; VAMP8: vesicle associated membrane protein 8; Zn2+: zinc.


Asunto(s)
Neoplasias , Canales de Potencial de Receptor Transitorio , Animales , Autofagosomas/metabolismo , Autofagia/fisiología , Humanos , Lisosomas/metabolismo , Ratones , Neoplasias/metabolismo , Oncogenes , Preparaciones Farmacéuticas/metabolismo , Ratas , Canales de Potencial de Receptor Transitorio/metabolismo , Zinc/metabolismo , Zinc/farmacología
11.
Clin Exp Pharmacol Physiol ; 37(3): 337-42, 2010 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-19769605

RESUMEN

1. The aim of the present study was to examine the effects of inhibition of the Na(+)/H(+) exchanger (NHE-1) on cardiopulmonary performance, oxygen carrying capacity and tissue inflammation in a pig model of traumatic haemorrhage-resuscitation. 2. In 12 instrumented anaesthetized pigs, traumatic haemorrhage was modelled by producing tibia fractures, followed by haemorrhage of 25 mL/kg for 20 min, and then a 4 mm hepatic arterial tear with surgical repair after 20 min. Animals then underwent low-volume fluid resuscitation with either Hextend (vehicle; n = 6; Hospira, Lake Forest, IL, USA) or 3 mg/kg BIIB513 (an NHE-1 inhibitor) + Hextend (n = 6). The experiment was terminated 6 h after the beginning of resuscitation. 3. Compared with vehicle-treated controls, the addition of NHE-1 inhibition with BIIB513 significantly improved the left ventricle stroke work index and attenuated increases in pulmonary arterial pressure and pulmonary vascular resistance. Furthermore, BIIB513 treatment significantly increased the oxygenated haemoglobin ratio, blood oxygen content and mixed venous blood oxygen saturation and improved blood oxygen delivery. In addition, BIIB513 treatment reduced lung tissue levels of interleukin-6 by 80%, tumour necrosis factor-alpha by 37% and myeloperoxidase activity by 38%. Nuclear factor-kappaB DNA binding activity in the lung was also slightly and significantly attenuated following BIIB513 treatment. 4. In conclusion, the present study shows that NHE-1 inhibition facilitates the response to fluid resuscitation after traumatic haemorrhage by improving cardiac function, pulmonary vascular function and oxygen carrying capacity, which results in reduced tissue inflammatory injury.


Asunto(s)
Reanimación Cardiopulmonar/métodos , Mediadores de Inflamación/fisiología , Oxígeno/metabolismo , Choque Hemorrágico/metabolismo , Intercambiadores de Sodio-Hidrógeno/antagonistas & inhibidores , Intercambiadores de Sodio-Hidrógeno/metabolismo , Animales , Mediadores de Inflamación/farmacología , Pulmón/efectos de los fármacos , Pulmón/metabolismo , Pulmón/patología , Masculino , Mesilatos/farmacología , Mesilatos/uso terapéutico , Miocardio/metabolismo , Miocardio/patología , Resucitación/métodos , Choque Hemorrágico/tratamiento farmacológico , Choque Hemorrágico/patología , Porcinos
12.
Neuroscience ; 448: 107-125, 2020 11 10.
Artículo en Inglés | MEDLINE | ID: mdl-32946951

RESUMEN

Anxiety disorder (AD) is characterized by the development of maladaptive neuronal circuits and changes to the excitatory/inhibitory (E/I) balance of the central nervous system. Although AD is considered to be heritable, specific genetic markers remain elusive. Recent genome-wide association studies (GWAS) studies have identified non-catalytic region of tyrosine kinase adaptor protein 1 (NCK1), a gene that codes for an intracellular adaptor protein involved in actin dynamics, as an important gene in the regulation of mood. Using a murine model in which NCK1 is inactivated, we show that male, but not female, mice display increased levels of context-dependent anxiety-like behaviors along with an increase in circulating serum corticosterone relative to control. Treatment of male NCK1 mutant mice with a positive allosteric modulator of the GABAA receptor rescued the anxiety-like behaviors implicating NCK1 in regulating neuronal excitability. These defects are not attributable to apparent defects in gross brain structure or in axon guidance. However, when challenged in an approach-avoidance conflict paradigm, male NCK1-deficient mice have decreased neuronal activation in the prefrontal cortex (PFC), as well as decreased activation of inhibitory interneurons in the basolateral amygdala (BLA). Finally, NCK1 deficiency results in loss of dendritic spine density in principal neurons of the BLA. Taken together, these data implicate NCK1 in the control of E/I balance in BLA. Our work identifies a novel role for NCK1 in the regulation of sex-specific neuronal circuitry necessary for controlling anxiety-like behaviors. Further, our work points to this animal model as a useful preclinical tool for the study of novel anxiolytics and its significance towards understanding sex differences in anxiolytic function.


Asunto(s)
Complejo Nuclear Basolateral , Estudio de Asociación del Genoma Completo , Amígdala del Cerebelo , Animales , Ansiedad , Trastornos de Ansiedad , Femenino , Masculino , Ratones
13.
Pflugers Arch ; 458(3): 471-80, 2009 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-19139916

RESUMEN

The objective of the study was to investigate the role of tyrosine phosphorylation in the regulation of KCNQ1/KCNE1 channels. Large whole-cell time- and voltage-dependent K(+) currents were present in human embryonic kidney 293 cells cotransfected with human KCNQ1 and KCNE1 but not in control nontransfected cells. The time- and voltage-dependent current had biophysical properties typical of cardiac KCNQ1/KCNE1 current and was almost completely abolished by KCNQ1 blocker chromanol 293B (50 microM). Both KCNQ1/KCNE1 and KCNQ1 current were inhibited in a voltage-independent manner by tyrosine kinase (PTK) inhibitor tyrphostin A25 (100 microM), but not by PTK-inactive tyrphostin A1 (100 microM), suggesting involvement of tyrosine phosphorylation in maintaining channel activity. This view was strengthened by the finding that phosphotyrosyl phosphatase inhibitor monoperoxo(picolinato)-oxo-vanadate(V) (200 microM) reversed the inhibition of current by tyrphostin A25. However, the channel-pertinent tyrosine phosphorylation modulated by these compounds does not appear to be on the channel itself because inhibition of current by tyrphostin A25 was unaffected by single and multiple mutations of KCNQ1 cytoplasmically accessible tyrosine residues. Inhibition by tyrphostin A25 was unaffected by intracellularly applied diC8 phosphatidylinositol-4,5-bisphosphate (diC8 PIP(2); 25 microM), and based on the results obtained from cell surface biotinylation experiments, it was not due to loss of channels from the membrane. We conclude that tyrphostin A25 inhibits KCNQ1/KCNE1 current by lowering tyrosine phosphorylation on unidentified nonchannel protein(s) that directly or indirectly regulate the open probability of the KCNQ1 pore in a PIP(2)-independent manner.


Asunto(s)
Activación del Canal Iónico/fisiología , Canal de Potasio KCNQ1/fisiología , Riñón/fisiología , Potenciales de la Membrana/fisiología , Canales de Potasio con Entrada de Voltaje/fisiología , Potasio/metabolismo , Proteínas Tirosina Quinasas/metabolismo , Línea Celular , Retroalimentación/fisiología , Humanos , Mutación
14.
J Cell Biol ; 217(2): 715-730, 2018 02 05.
Artículo en Inglés | MEDLINE | ID: mdl-29317530

RESUMEN

The actin cytoskeleton is essential for the structural changes in dendritic spines that lead to the formation of new synapses. Although the molecular mechanisms underlying spine formation are well characterized, the events that drive spine maturation during development are largely unknown. In this study, we demonstrate that Angiomotin (AMOT-130) is necessary for spine stabilization. AMOT-130 is enriched in mature dendritic spines and functions to stabilize the actin cytoskeleton by coupling F-actin to postsynaptic protein scaffolds. These functions of AMOT are transiently restricted during postnatal development by phosphorylation imposed by the kinase Lats1. Our study proposes that AMOT-130 is essential for normal spine morphogenesis and identifies Lats1 as an upstream regulator in this process. Moreover, our findings may link AMOT-130 loss and the related spine defects to neurological disorders.


Asunto(s)
Espinas Dendríticas/metabolismo , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Proteínas de la Membrana/metabolismo , Angiomotinas , Animales , Células Cultivadas , Ratas , Ratas Sprague-Dawley
15.
Mol Cell Biol ; 35(13): 2265-77, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-25918243

RESUMEN

Deregulation of cellular polarity proteins and their associated complexes leads to changes in cell migration and proliferation. The nitric oxide synthase 1 adaptor protein (NOS1AP) associates with the tumor suppressor protein Scribble to control cell migration and oncogenic transformation. However, how NOS1AP is linked to the cell signaling events that curb oncogenic progression has remained elusive. Here we identify several novel NOS1AP isoforms, NOS1APd, NOS1APe, and NOS1APf, with distinct cellular localizations. We show that isoforms with a membrane-interacting phosphotyrosine binding (PTB) domain can associate with Scribble and recognize acidic phospholipids. In a screen to identify novel binding proteins, we have discovered a complex consisting of NOS1AP and the transcriptional coactivator YAP linking NOS1AP to the Hippo signaling pathway. Silencing of NOS1AP reduces the phosphorylation of YAP and of the upstream kinase Lats1. Conversely, expression of NOS1AP promotes YAP and Lats1 phosphorylation, which correlates with reduced TEAD activity and restricted cell proliferation. Together, these data implicate a role for NOS1AP in the regulation of core Hippo signaling and are consistent with the idea that NOS1AP functions as a tumor suppressor.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Proteínas Reguladoras de la Apoptosis/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Transducción de Señal , Proteínas Adaptadoras Transductoras de Señales/análisis , Secuencia de Aminoácidos , Animales , Proteínas Reguladoras de la Apoptosis/análisis , Células HEK293 , Hipocampo/metabolismo , Humanos , Modelos Moleculares , Datos de Secuencia Molecular , Isoformas de Proteínas/análisis , Isoformas de Proteínas/metabolismo , Ratas , Ratas Wistar , Proteínas Señalizadoras YAP
16.
Br J Pharmacol ; 140(4): 750-8, 2003 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-14534156

RESUMEN

The effect of preconditioning with U50488 H (UP), a selective kappa-opioid receptor (kappa-OR) agonist, on infarct size and intracellular Ca2+ ([Ca2+]i) in the heart subjected to ischaemic insults were studied and evaluated. U50488 H administered intravenously reduced the infarct size 18-48 h after administration in isolated hearts subjected to regional ischaemia/reperfusion (I/R). The effect was dose dependent. A peak effect was reached at 10 mg x kg-1 U50488 H and at 24 h after administration. The effect of 10 mg x kg-1 U50488 H at 24 h after administration was abolished by nor-binaltorphimine (nor-BNI), a selective kappa-OR antagonist, indicating the effect was kappa-OR mediated. The infarct reducing effect of U50488 H was attenuated when a selective blocker of mitochondrial (5-hydroxydecanoic acid, 5-HD) or sarcolemmal (HRM-1098) ATP-sensitive potassium channel (KATP) was coadministered with U50488 H 24 h before ischaemia or when 5-HD was administered just before ischaemia. U50488 H also attenuated the elevation in [Ca2+]i and reduction in electrically induced [Ca2+]i transient in cardiomyocytes subjected to ischaemic insults. The effects were reversed by blockade of KATP channel, which abolished the protective effect of preconditioning with U50488 H. The results indicated that mitochondrial KATP channel serves as both a trigger and a mediator, while sarcolemmal KATP channel as a trigger only, of delayed cardioprotection of kappa-OR stimulation. The effects of these channels may result from prevention/attenuation of [Ca2+]i overload induced by ischaemic insults.


Asunto(s)
3,4-Dicloro-N-metil-N-(2-(1-pirrolidinil)-ciclohexil)-bencenacetamida, (trans)-Isómero/farmacología , Calcio/metabolismo , Corazón/fisiología , Naltrexona/análogos & derivados , Canales de Potasio/fisiología , Receptores Opioides kappa/efectos de los fármacos , 3,4-Dicloro-N-metil-N-(2-(1-pirrolidinil)-ciclohexil)-bencenacetamida, (trans)-Isómero/antagonistas & inhibidores , 3,4-Dicloro-N-metil-N-(2-(1-pirrolidinil)-ciclohexil)-bencenacetamida, (trans)-Isómero/uso terapéutico , Animales , Calcio/antagonistas & inhibidores , Señalización del Calcio/efectos de los fármacos , Señalización del Calcio/fisiología , Circulación Coronaria/efectos de los fármacos , Vasos Coronarios/lesiones , Relación Dosis-Respuesta a Droga , Corazón/efectos de los fármacos , Frecuencia Cardíaca/efectos de los fármacos , Frecuencia Cardíaca/fisiología , Inyecciones Intravenosas , Precondicionamiento Isquémico Miocárdico/métodos , Masculino , Infarto del Miocardio/complicaciones , Infarto del Miocardio/tratamiento farmacológico , Daño por Reperfusión Miocárdica/complicaciones , Daño por Reperfusión Miocárdica/tratamiento farmacológico , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/metabolismo , Naltrexona/farmacología , Bloqueadores de los Canales de Potasio/metabolismo , Bloqueadores de los Canales de Potasio/farmacología , Bloqueadores de los Canales de Potasio/uso terapéutico , Canales de Potasio/clasificación , Canales de Potasio/efectos de los fármacos , Ratas/fisiología , Ratas Sprague-Dawley , Receptores Opioides kappa/antagonistas & inhibidores , Receptores Opioides kappa/metabolismo , Factores de Tiempo
17.
Resuscitation ; 83(6): 774-81, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-22155220

RESUMEN

This study evaluated the effects of sodium-hydrogen exchanger (NHE1) inhibition on enhancing fluid resuscitation outcomes in traumatic hemorrhagic shock, and examined the mechanisms related to NHE1 inhibitor-induced protection and recovery from hemorrhagic shock. Traumatic hemorrhage was modeled in anesthetized pigs by producing tibia fractures followed by hemorrhage of 25 ml/kg for 20 min, and then a 4mm hepatic arterial tear with surgical repair after 20 min. Animals then underwent low volume fluid resuscitation with either hextend (n=6) or 3mg/kg BIIB513 (NHE1 inhibitor)+hextend (n=6). The experiment was terminated 6h after the beginning of resuscitation. In association with traumatic hemorrhagic shock, there was a decrease in cardiac index, stimulation of the inflammatory response, myocardial, liver and kidney injury. The administration of the NHE1 inhibitor at the time of resuscitation attenuated shock-resuscitation-induced myocardial hypercontracture and resulted in a significant increase in stroke volume index, compared to vehicle-treated controls. NHE1 inhibition also reduced the inflammatory response, and lessened myocardial, liver and kidney injury. In addition, NHE1 inhibition reduced NF-κB activation and iNOS expression, and attenuated of ERK1/2 phosphorylation. Results from the present study indicate that NHE1 inhibition prevents multiple organ injury by attenuating shock-resuscitation-induced myocardial hypercontracture and by inhibiting NF-κB activation and neutrophil infiltration, reducing iNOS expression and ERK1/2 phosphorylation, thereby, reducing systemic inflammation and thus multi-organ injury.


Asunto(s)
Mesilatos/uso terapéutico , Resucitación , Choque Hemorrágico/tratamiento farmacológico , Intercambiadores de Sodio-Hidrógeno/antagonistas & inhibidores , Animales , Fluidoterapia , Hemodinámica , Derivados de Hidroxietil Almidón/uso terapéutico , Inflamación/prevención & control , Interleucina-10/sangre , Interleucina-6/sangre , Sistema de Señalización de MAP Quinasas , Masculino , Insuficiencia Multiorgánica/prevención & control , Isquemia Miocárdica/prevención & control , FN-kappa B/metabolismo , Óxido Nítrico Sintasa de Tipo II/metabolismo , Fosforilación , Sustitutos del Plasma/uso terapéutico , Choque Hemorrágico/fisiopatología , Choque Hemorrágico/terapia , Sus scrofa , Troponina I/sangre
18.
J Gen Physiol ; 135(3): 229-45, 2010 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-20142516

RESUMEN

Rapid chloride permeation through the cystic fibrosis transmembrane conductance regulator (CFTR) Cl(-) channel is dependent on the presence of fixed positive charges in the permeation pathway. Here, we use site-directed mutagenesis and patch clamp recording to show that the functional role played by one such positive charge (K95) in the inner vestibule of the pore can be "transplanted" to a residue in a different transmembrane (TM) region (S1141). Thus, the mutant channel K95S/S1141K showed Cl(-) conductance and open-channel blocker interactions similar to those of wild-type CFTR, thereby "rescuing" the effects of the charge-neutralizing K95S mutation. Furthermore, the function of K95C/S1141C, but not K95C or S1141C, was inhibited by the oxidizing agent copper(II)-o-phenanthroline, and this inhibition was reversed by the reducing agent dithiothreitol, suggesting disulfide bond formation between these two introduced cysteine side chains. These results suggest that the amino acid side chains of K95 (in TM1) and S1141 (in TM12) are functionally interchangeable and located closely together in the inner vestibule of the pore. This allowed us to investigate the functional effects of increasing the number of fixed positive charges in this vestibule from one (in wild type) to two (in the S1141K mutant). The S1141K mutant had similar Cl(-) conductance as wild type, but increased susceptibility to channel block by cytoplasmic anions including adenosine triphosphate, pyrophosphate, 5-nitro-2-(3-phenylpropylamino)benzoic acid, and Pt(NO(2))(4)(2-) in inside-out membrane patches. Furthermore, in cell-attached patch recordings, apparent voltage-dependent channel block by cytosolic anions was strengthened by the S1141K mutation. Thus, the Cl(-) channel function of CFTR is maximal with a single fixed positive charge in this part of the inner vestibule of the pore, and increasing the number of such charges to two causes a net decrease in overall Cl(-) transport through a combination of failure to increase Cl(-) conductance and increased susceptibility to channel block by cytosolic substances.


Asunto(s)
Regulador de Conductancia de Transmembrana de Fibrosis Quística/fisiología , Activación del Canal Iónico/fisiología , Potenciales de la Membrana/fisiología , Cationes , Humanos , Mutagénesis Sitio-Dirigida , Técnicas de Placa-Clamp , Conformación Proteica , Relación Estructura-Actividad
19.
Science ; 322(5901): 587-90, 2008 Oct 24.
Artículo en Inglés | MEDLINE | ID: mdl-18948540

RESUMEN

Studies of nitric oxide over the past two decades have highlighted the fundamental importance of gaseous signaling molecules in biology and medicine. The physiological role of other gases such as carbon monoxide and hydrogen sulfide (H2S) is now receiving increasing attention. Here we show that H2S is physiologically generated by cystathionine gamma-lyase (CSE) and that genetic deletion of this enzyme in mice markedly reduces H2S levels in the serum, heart, aorta, and other tissues. Mutant mice lacking CSE display pronounced hypertension and diminished endothelium-dependent vasorelaxation. CSE is physiologically activated by calcium-calmodulin, which is a mechanism for H2S formation in response to vascular activation. These findings provide direct evidence that H2S is a physiologic vasodilator and regulator of blood pressure.


Asunto(s)
Presión Sanguínea , Cistationina gamma-Liasa/metabolismo , Sulfuro de Hidrógeno/metabolismo , Hipertensión/fisiopatología , Vasodilatación , Animales , Aorta/metabolismo , Calcio/metabolismo , Calmodulina/metabolismo , Cistationina gamma-Liasa/deficiencia , Cistationina gamma-Liasa/genética , Cisteína/sangre , Endotelio Vascular/metabolismo , Homocisteína/sangre , Sulfuro de Hidrógeno/sangre , Arterias Mesentéricas/fisiología , Cloruro de Metacolina/farmacología , Ratones , Ratones Noqueados , Miocardio/metabolismo , Oxidación-Reducción , Sulfuros/farmacología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA