Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Biochim Biophys Acta Rev Cancer ; 1878(5): 188950, 2023 09.
Artículo en Inglés | MEDLINE | ID: mdl-37419192

RESUMEN

Ovarian cancer (OVCA) is the second most common gynecological cancer and one of the leading causes of cancer related mortality among women. Recent studies suggest that among ovarian cancer patients at least 70% of the cases experience the involvement of lymph nodes and metastases through lymphatic vascular network. However, the impact of lymphatic system in the growth, spread and the evolution of ovarian cancer, its contribution towards the landscape of ovarian tissue resident immune cells and their metabolic responses is still a major knowledge gap. In this review first we present the epidemiological aspect of the OVCA, the lymphatic architecture of the ovary, we discuss the role of lymphatic circulation in regulation of ovarian tumor microenvironment, metabolic basis of the upregulation of lymphangiogenesis which is often observed during progression of ovarian metastasis and ascites development. Further we describe the implication of several mediators which influence both lymphatic vasculature as well as ovarian tumor microenvironment and conclude with several therapeutic strategies for targeting lymphatic vasculature in ovarian cancer progression in present day.


Asunto(s)
Vasos Linfáticos , Neoplasias Ováricas , Humanos , Femenino , Vasos Linfáticos/metabolismo , Vasos Linfáticos/patología , Neoplasias Ováricas/patología , Linfangiogénesis/fisiología , Ganglios Linfáticos/patología , Microambiente Tumoral
2.
Front Behav Neurosci ; 16: 1008556, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36338879

RESUMEN

Early-life adversity (ELA) can induce persistent neurological changes and increase the risk for developing affective or substance use disorders. Disruptions to the reward circuitry of the brain and pathways serving motivation and emotion have been implicated in the link between ELA and altered adult behavior. The molecular mechanisms that mediate the long-term effects of ELA, however, are not fully understood. We examined whether ELA in the form of neonatal maternal separation (MatSep) modifies behavior and synaptic protein expression in adults. We hypothesized that MatSep would affect dopaminergic and glutamatergic signaling and enhance sensitivity to methamphetamine (Meth) reward or increase anxiety. Male Wistar rats were subjected to MatSep for 180 min/d on postnatal days (PND) 2-14 and allowed to grow to adulthood (PND 60) with no further manipulation. The hippocampus (Hipp), medial prefrontal cortex (mPFC), nucleus accumbens (NAc), and caudate putamen (CPu) were isolated from one subgroup of animals and subjected to Western blot and protein quantitation for tyrosine hydroxylase (TH), α-synuclein (ALPHA), NMDA receptor (NMDAR), dopamine receptor-1 (D1) and -2 (D2), dopamine transporter (DAT), and postsynaptic density 95 (PSD95). Separate group of animals were tested for anxiety-like behavior and conditioned place preference (CPP) to Meth at 0.0, 0.1, and 1.0 mg/kg doses. MatSep rats displayed an increase in basal levels of anxiety-like behavior compared to control animals. MatSep rats also demonstrated CPP to Meth, but their responses did not differ significantly from controls at any drug dose. Increased NMDAR, D2, and ALPHA expression was observed in the NAc and CPu following MatSep; D2 and ALPHA levels were also elevated in the mPFC, along with DAT. MatSep rats had reduced D1 expression in the mPFC and Hipp, with the Hipp also showing a reduction in D2. Only the CPu showed elevated TH and decreased DAT expression levels. No significant changes were found in PSD95 expression in MatSep rats. In conclusion, ELA is associated with long-lasting and region-specific changes in synaptic protein expression that diminish dopamine neurotransmission and increase anxiety-like behavior in adults. These findings illustrate potential mechanisms through which ELA may increase vulnerability to stress-related illness.

3.
Drug Chem Toxicol ; 45(4): 1698-1706, 2022 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-33297789

RESUMEN

The venom of Crotalus ornatus (vCo) poses a threat to human health, as it contains a mixture of toxins that can cause cytotoxic, necrotic, and hemolytic effects. The present study assessed methanolic and acetone extracts from leaves and flowers of Larrea tridentata, as well as the bark of Quercus virginiana as potential suppressors of the toxic effects of vCo in vitro. The content of total phenols, flavonoids, and tannins of the plant extracts were quantified for the suppression of vCo cytotoxicity in two cell culture models, human lymphocytes and porcine aortic endothelial (PAE) cells. Extracts from Q. virginiana displayed a greater concentration of total phenols, flavonoids, and tannins. Co-incubation of lymphocytes and PAE cells with fixed concentrations of vCo and plant extracts resulted in decreased vCo-induced cytotoxicity. A 24-hour co-incubation of lymphocytes with vCo (2.36 ± 0.17 µg/mL) and 0.5 µg/mL of methanolic leaf extract from L. tridentata (LLM) significantly suppressed the venom-induced cytotoxicity by 37.33 ± 8.33%. Similarly, the LLM extract (4 µg/mL) caused a significant decrease in vCo cytotoxicity after 24 hours in PAE cells. In contrast, while the acetone extract of Q. virginiana bark (QA) suppressed cytotoxicity by 29.20 ± 3.51% (p < 0.001) in lymphocytes, it failed to protect PAE cells against vCo after 24 hours. In PAE cells, a shorter 4-hour co-incubation showed significant suppression of cytotoxicity with both extracts. Our results collectively suggest that LLM and QA possess cytoprotective properties against the in vitro toxic effects of vCo, and thus establish extracts from these plants as potential therapeutic interventions against Crotalus envenomation.


Asunto(s)
Larrea , Quercus , Acetona , Animales , Crotalus , Flavonoides , Metanol , Fenoles , Extractos Vegetales/toxicidad , Porcinos , Taninos , Ponzoñas
4.
Front Mol Neurosci ; 14: 661993, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34447296

RESUMEN

Exposure to early-life stress (ELS) can persistently modify neuronal circuits and functions, and contribute to the expression of misfolded and aggregated proteins that are hallmarks of several neurodegenerative diseases. The healthy brain is able to clear dysfunctional proteins through the ubiquitin-proteasome system (UPS) and the autophagy-lysosomal pathway (ALP). Accumulating evidence indicates that impairment of these pathways contributes to enhanced protein aggregation and neurodegeneration. While stress is a known precipitant of neurological decline, few specific mechanistic links underlying this relationship have been identified. We hypothesized that neonatal maternal separation (MatSep), a well-established model of ELS, has the ability to alter the levels of UPS and ALP components in the brain, and thus has the potential to disrupt proteostasis. The expression of proteostasis-associated protein markers was evaluated by immunoblotting in the hippocampus and cortex of adult Wistar rats that were previously subjected to MatSep. We observed multiple sex- and MatSep-specific changes in the expression of proteins in the ALP, mitophagy, and UPS pathways, particularly in the hippocampus of adult animals. In contrast, MatSep had limited influence on proteostasis marker expression in the cortex of adult animals. Our results indicate that MatSep can selectively modify the intracellular protein degradation machinery in ways that may impact the development and progression of neurodegenerative disease.

5.
J Alzheimers Dis ; 83(4): 1691-1702, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34420960

RESUMEN

BACKGROUND: Fluoxetine (FLX) represents the antidepressant of choice for the management of pediatric mood-related illnesses. Accumulating preclinical evidence suggests that ontogenic FLX exposure leads to deregulated affect-related phenotypes in adulthood. Mood-related symptomatology constitutes a risk-factor for various neurological disorders, including Alzheimer's disease (AD), making it possible for juvenile FLX history to exacerbate the development of neurodegenerative diseases. OBJECTIVE: Because AD is characterized by the pathological accumulation of hyperphosphorylated tau, which can result from impaired function of protein degradation pathways, such as autophagy and the ubiquitin-proteasome system (UPS), we evaluated the long-term effects of adolescent FLX exposure on these pathways, using mice as a model system. METHODS: We subjected C57BL/6 adolescent male mice to FLX (20 mg/kg/day) from postnatal day (PD) 35 to PD49. Twenty-one days after the last FLX injection (i.e., adulthood; PD70), mice were euthanized and, using immunoblotting analysis, we evaluated protein markers of autophagy (Beclin-1, LC3-II, p62) and the UPS (K48-pUb), as well as AD-associated forms of phosphorylated tau, within the hippocampus and prefrontal cortex. RESULTS: Juvenile FLX pre-exposure mediated long-term changes in the expression of protein markers (increased LC3-II and decreased p62) that is consistent with autophagy activation, particularly in the prefrontal cortex. Furthermore, FLX history induced persistent accumulation of AD-associated variants of tau in both the hippocampus and prefrontal cortexConclusion: Adolescent FLX treatment may have enduring effects in the neuronal protein degradation machinery, which could adversely influence clearance of abnormal proteins, potentially predisposing individuals to developing AD in later life.


Asunto(s)
Enfermedad de Alzheimer/patología , Autofagia/efectos de los fármacos , Fluoxetina , Hipocampo/patología , Corteza Prefrontal/patología , Proteínas tau , Adolescente , Animales , Antidepresivos de Segunda Generación/administración & dosificación , Antidepresivos de Segunda Generación/farmacología , Encéfalo/efectos de los fármacos , Encéfalo/patología , Fluoxetina/administración & dosificación , Fluoxetina/farmacología , Humanos , Immunoblotting , Masculino , Ratones , Ratones Endogámicos C57BL , Fosforilación
6.
Sci Rep ; 11(1): 7758, 2021 04 08.
Artículo en Inglés | MEDLINE | ID: mdl-33833356

RESUMEN

The objective of this study was to evaluate whether juvenile fluoxetine (FLX) exposure induces long-term changes in baseline responses to anxiety-inducing environments, and if so, whether its re-exposure in adulthood would ameliorate this anxiety-like phenotype. An additional goal was to assess the impact of adolescent FLX pretreatment, and its re-exposure in adulthood, on serotonin transporters (5-HTT) and brain-derived-neurotrophic-factor (BDNF)-related signaling markers (TrkB-ERK1/2-CREB-proBDNF-mBDNF) within the hippocampus and prefrontal cortex. To do this, female C57BL/6 mice were exposed to FLX in drinking water during postnatal-days (PD) 35-49. After a 21-day washout-period (PD70), mice were either euthanized (tissue collection) or evaluated on anxiety-related tests (open field, light/dark box, elevated plus-maze). Juvenile FLX history resulted in a persistent avoidance-like profile, along with decreases in BDNF-signaling markers, but not 5-HTTs or TrkB receptors, within both brain regions. Interestingly, FLX re-exposure in adulthood reversed the enduring FLX-induced anxiety-related responses across all behavioral tasks, while restoring ERK2-CREB-proBDNF markers to control levels and increasing mBDNF within the prefrontal cortex, but not the hippocampus. Collectively, these results indicate that adolescent FLX history mediates neurobehavioral adaptations that endure into adulthood, which are indicative of a generalized anxiety-like phenotype, and that this persistent effect is ameliorated by later-life FLX re-exposure, in a prefrontal cortex-specific manner.


Asunto(s)
Ansiedad/tratamiento farmacológico , Fluoxetina/administración & dosificación , Inhibidores Selectivos de la Recaptación de Serotonina/administración & dosificación , Animales , Conducta Animal/efectos de los fármacos , Femenino , Ratones , Ratones Endogámicos C57BL
7.
Biochem Biophys Res Commun ; 549: 98-104, 2021 04 16.
Artículo en Inglés | MEDLINE | ID: mdl-33667715

RESUMEN

The ßγ subunit of heterotrimeric G proteins, a key molecule in the G protein-coupled receptors (GPCRs) signaling pathway, has been shown to be an important factor in the modulation of the microtubule cytoskeleton. Gßγ has been shown to bind to tubulin, stimulate microtubule assembly, and promote neurite outgrowth of PC12 cells. In this study, we demonstrate that in addition to microtubules, Gßγ also interacts with actin filaments, and this interaction increases during NGF-induced neuronal differentiation of PC12 cells. We further demonstrate that the Gßγ-actin interaction occurs independently of microtubules as nocodazole, a well-known microtubule depolymerizing agent did not inhibit Gßγ-actin complex formation in PC12 cells. A confocal microscopic analysis of NGF-treated PC12 cells revealed that Gßγ co-localizes with both actin and microtubule cytoskeleton along neurites, with specific co-localization of Gßγ with actin at the distal end of these neuronal processes. Furthermore, we show that Gßγ interacts with the actin cytoskeleton in primary hippocampal and cerebellar rat neurons. Our results indicate that Gßγ serves as an important modulator of the neuronal cytoskeleton by interacting with both microtubules and actin filaments, and is likely to participate in various aspects of neuronal differentiation including axon and growth cone formation.


Asunto(s)
Citoesqueleto de Actina/metabolismo , Diferenciación Celular , Subunidades beta de la Proteína de Unión al GTP/metabolismo , Subunidades gamma de la Proteína de Unión al GTP/metabolismo , Neuronas/citología , Neuronas/metabolismo , Citoesqueleto de Actina/efectos de los fármacos , Actinas/metabolismo , Animales , Axones/efectos de los fármacos , Axones/metabolismo , Diferenciación Celular/efectos de los fármacos , Citoesqueleto/efectos de los fármacos , Citoesqueleto/metabolismo , Hipocampo/citología , Modelos Biológicos , Factor de Crecimiento Nervioso/farmacología , Neuronas/efectos de los fármacos , Células PC12 , Polimerizacion/efectos de los fármacos , Unión Proteica/efectos de los fármacos , Ratas , Ratas Sprague-Dawley
8.
Brain Res ; 1749: 147117, 2020 12 15.
Artículo en Inglés | MEDLINE | ID: mdl-32971085

RESUMEN

Early life stress alters the function and feedback regulation of the hypothalamic-pituitaryadrenal (HPA) axis, and can contribute to neuroinflammation and neurodegeneration by modifying peripheral blood mononuclear cell (PBMC) activity. The retina, as part of the nervous system, is sensitive to immune changes induced by stress. However, the consequences of stress experienced at an early age on retinal development have not yet been elucidated. Here we aimed to evaluate the impact of maternal separation (MatSep) across three stages of the lifespan (adolescent, adult, and aged) on the retina, as well as on progression through the cell cycle and mitochondrial activity in PBMCs from female Wistar rats. Newborn pups were separated from their mother from postnatal day (PND) 2 until PND 14 for 3 h/day. Retinal analysis from the MatSep groups showed architectural alterations such as a diminished thickness of retinal layers, as well as increased expression of proinflammatory markers DJ-1, Iba-1, and CD45 and the gliotic marker GFAP. Additionally, MatSep disrupted the cell cycle and caused long-term increases in mitochondrial activity in PBMCs from adolescent and adult rats. Changes in the cell cycle profile of the PBMCs from aged MatSep rats were undetected. However, these PBMCs exhibited increased sensitivity to H2O2-induced oxidative stress in vitro. Therefore, these results suggest that early life stress can have long-term effects on retinal structure and function, possibly elicited by neonatal immune preconditioning.


Asunto(s)
Leucocitos Mononucleares/metabolismo , Privación Materna , Retina/metabolismo , Estrés Psicológico/metabolismo , Animales , Ciclo Celular/fisiología , Femenino , Mitocondrias/metabolismo , Estrés Oxidativo/fisiología , Ratas , Ratas Wistar
9.
Artículo en Inglés | MEDLINE | ID: mdl-32095734

RESUMEN

BACKGROUND: Anxiety disorders are the most common neuropathologies worldwide, but the precise neuronal mechanisms that underlie these disorders remain unknown. The hippocampus plays a role in mediating anxiety-related responses, which can be modeled in rodents using behavioral assays, such as the elevated plus maze. Yet, the molecular markers that underlie affect-related behavior on the elevated plus maze are not well understood. METHODS: We used herpes simplex virus vector delivery to overexpress extracellular signal-regulated kinase-2, a signaling molecule known to be involved in depression and anxiety, within the dorsal hippocampus of adult Sprague-Dawley male rats. Three days post virus delivery, we assessed anxiety-like responses on the elevated plus maze or general locomotor activity on the open field test. RESULTS: When compared to controls, rats overexpressing extracellular signal-regulated kinase-2 in the dorsal hippocampus displayed an anxiolytic-like phenotype, per increases in time spent in the open arms, and less time in the closed arms, of the elevated plus maze. Furthermore, no changes in locomotor activity as a function of virus infusion were observed on the open field test between the experimental groups. CONCLUSION: This investigation demonstrates that virus-mediated increases of extracellular signal-regulated kinase-2 signaling, within the hippocampus, plays a critical role in decreasing anxiogenic responses on the rat elevated plus maze. As such, our data provide construct validity, at least in part, to the molecular mechanisms that mediate anxiolytic-like behavior in rodent models for the study of anxiety.

10.
Neurobiol Stress ; 9: 105-112, 2018 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-30450376

RESUMEN

Neurodegenerative diseases are characterized by an irreversible and progressive loss of neuronal structure and function. While many alterations to normal cellular processes occur during neurodegeneration, a pathological accumulation of aggregated proteins constitutes a hallmark of several neurodegenerative disorders. Alzheimer's disease, specifically, is pathologically defined by the formation of amyloid plaques and tangles of hyperphosphorylated tau protein. Stress has emerged as an important factor in the development and progression of neurodegenerative diseases, including Alzheimer's. Very little is known, however, regarding the effects of stress on the mechanisms controlling abnormal protein aggregation and clearance. Chronic stress activates the hypothalamic-pituitary-adrenal (HPA) axis, causing an excessive secretion of glucocorticoids that are capable of impacting diverse physiological and cellular processes. The present review focuses on the influence of stress on a key feature of Alzheimer's disease pathology, emphasizing the relationship between tau phosphorylation and accumulation and its connection to HPA axis dysfunction.

11.
Biochem Biophys Res Commun ; 503(1): 102-108, 2018 09 03.
Artículo en Inglés | MEDLINE | ID: mdl-29852176

RESUMEN

Microtubules (MTs) constitute a crucial part of the cytoskeleton and are essential for cell division and differentiation, cell motility, intracellular transport, and cell morphology. Precise regulation of MT assembly and dynamics is essential for the performance of these functions. Although much progress has been made in identifying and characterizing the cellular factors that regulate MT assembly and dynamics, signaling events in this process is not well understood. Gßγ, an important component of the G protein-coupled receptor (GPCR) signaling pathway, has been shown to promote MT assembly in vitro and in cultured NIH3T3 and PC12 cells. Using the MT depolymerizing agent nocodazole, it has been demonstrated that the association of Gßγ with polymerized tubulin is critical for MT assembly. More recently, Gßγ has been shown to play a key role in NGF-induced neuronal differentiation of PC12 cells through its interaction with tubulin/MTs and modulation of MT assembly. Although NGF is known to exert its effect through tyrosine kinase receptor TrkA, the result suggests a possible involvement of GPCRs in this process. The present study was undertaken to determine whether agonist activation of GPCR utilizes Gßγ to promote MT assembly. We used isoproterenol and UK 14,304, agonists for two different GPCRs (ß- and α2-adrenergic receptors, respectively) known to activate Gs and Gi respectively, with an opposing effect on production of cAMP. The results demonstrate that the agonist activation of ß- and α2-adrenergic receptors promotes the association of Gßγ with MTs and stimulates MT assembly in NIH3T3 cells. Interestingly, the effects of these two agonists were more prominent when the cellular level of MT assembly was low (30% or less). In contrast to MT assembly, actin polymerization was not affected by isoproterenol or UK 14, 304 indicating that the effects of these agonists are limited to MTs. Thus, it appears that, upon cellular demand, GPCRs may utilize Gßγ to promote MT assembly. Stimulation of MT assembly appears to be a novel function of G protein-mediated signaling.


Asunto(s)
Subunidades beta de la Proteína de Unión al GTP/metabolismo , Subunidades gamma de la Proteína de Unión al GTP/metabolismo , Microtúbulos/metabolismo , Receptores Adrenérgicos alfa 2/metabolismo , Receptores Adrenérgicos beta/metabolismo , Tubulina (Proteína)/química , Tubulina (Proteína)/metabolismo , Agonistas de Receptores Adrenérgicos alfa 2/farmacología , Agonistas Adrenérgicos beta/farmacología , Animales , Diferenciación Celular , Ratones , Células 3T3 NIH , Neuronas/citología , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Células PC12 , Polimerizacion , Multimerización de Proteína , Ratas , Transducción de Señal
12.
J Bioenerg Biomembr ; 50(1): 59-69, 2018 02.
Artículo en Inglés | MEDLINE | ID: mdl-29313294

RESUMEN

The plasma membrane Ca2+-ATPase (PMCA) removes Ca2+ from the cytosol into the extracellular space. Its catalytic activity can be stimulated by calmodulin (CaM) or by limited proteolysis. We evaluated the effect of chlorpromazine (CPZ) and dimethyl sulfoxide (DMSO) over the hydrolytic activity of PMCA. Activity was monitored in three different forms: native, CaM-activated and proteolyzed by trypsin. CPZ appears to inhibit PMCA without directly interfering with the C-terminal site, since it is affected by CaM and proteolysis. Although the treatment of PMCA with trypsin and CaM produces an activation, it also produces an enzymatic form that is more sensitive to inhibition by CPZ. The same case was observed in the DMSO inhibition experiments. In the absence of CPZ, DMSO produces a progressive loss of activity, but in the presence of CPZ the profile of activity against DMSO changes and produces a recovery of activity, indicating a possible partition of CPZ by the solvent. Increasing Ca2+ concentrations indicated that CPZ interacts with PMCA rather than with CaM. This observation is supported by docking analysis that suggests that the CPZ-PMCA interaction is non-competitive. We propose that CPZ interacts with the state of lower affinity for Ca2 +.


Asunto(s)
Clorpromazina/farmacología , Dimetilsulfóxido/farmacología , Membrana Eritrocítica/enzimología , ATPasas Transportadoras de Calcio de la Membrana Plasmática/metabolismo , Biocatálisis/efectos de los fármacos , Calmodulina/farmacología , Antagonistas de Dopamina/farmacología , Interacciones Farmacológicas , Humanos , Simulación del Acoplamiento Molecular , Tripsina/farmacología
13.
BMC Cancer ; 17(1): 621, 2017 Sep 05.
Artículo en Inglés | MEDLINE | ID: mdl-28874141

RESUMEN

BACKGROUND: Prostate cancer is a major contributor to mortality worldwide, and significant efforts are being undertaken to decipher specific cellular and molecular pathways underlying the disease. Chronic stress is known to suppress reproductive function and promote tumor progression in several cancer models, but our understanding of the mechanisms through which stress contributes to cancer development and progression is incomplete. We therefore examined the relationship between stress, modulation of the gonadotropin-releasing hormone (GnRH) system, and changes in the expression of cancer-related genes in the rat prostate. METHODS: Adult male rats were acutely or repeatedly exposed to restraint stress, and compared to unstressed controls and groups that were allowed 14 days of recovery from the stress. Prostate tissue was collected and frozen for gene expression analyses by PCR array before the rats were transcardially perfused; and brain tissues harvested and immunohistochemically stained for Fos to determine neuronal activation. RESULTS: Acute stress elevated Fos expression in the paraventricular nucleus of the hypothalamus (PVH), an effect that habituated with repeated stress exposure. Data from the PCR arrays showed that repeated stress significantly increases the transcript levels of several genes associated with cellular proliferation, including proto-oncogenes. Data from another array platform showed that both acute and repeated stress can induce significant changes in metastatic gene expression. The functional diversity of genes with altered expression, which includes transcription factors, growth factor receptors, apoptotic genes, and extracellular matrix components, suggests that stress is able to induce aberrant changes in pathways that are deregulated in prostate cancer. CONCLUSIONS: Our findings further support the notion that stress can affect cancer outcomes, perhaps by interfering with neuroendocrine mechanisms involved in the control of reproduction.


Asunto(s)
Expresión Génica , Oncogenes , Próstata/metabolismo , Estrés Fisiológico , Estrés Psicológico , Animales , Biomarcadores , Transformación Celular Neoplásica , Sistema Endocrino/metabolismo , Hipotálamo/metabolismo , Masculino , Metástasis de la Neoplasia , Neoplasias de la Próstata/etiología , Neoplasias de la Próstata/metabolismo , Neoplasias de la Próstata/patología , Proteínas Proto-Oncogénicas c-fos/genética , Proteínas Proto-Oncogénicas c-fos/metabolismo , Ratas , Transducción de Señal
14.
BMC Biochem ; 17: 5, 2016 Mar 11.
Artículo en Inglés | MEDLINE | ID: mdl-26968444

RESUMEN

BACKGROUND: 1,2-Dichlorobenzene (1,2-DCB) is a benzene-derived molecule with two Cl atoms that is commonly utilized in the synthesis of pesticides. 1,2-DCB can be absorbed by living creatures and its effects on naturally-occurring enzymatic systems, including the effects on Ca(2+)-ATPases, have been poorly studied. Therefore, we aimed to study the effect of 1,2-DCB on the Ca(2+)-ATPase from sarcoplasmic reticulum (SERCA), a critical regulator of intracellular Ca(2+) concentration. RESULTS: Concentrations of 0.05-0.2 mM of 1,2-DCB were able to stimulate the hydrolytic activity of SERCA in a medium-containing Ca(2+)-ionophore. At higher concentrations (0.25-0.75 mM), 1,2-DCB inhibited the ATP hydrolysis to ~80 %. Moreover, ATP hydrolysis and Ca(2+) uptake in a medium supported by K-oxalate showed that starting at 0.05 mM,1,2-DCB was able to uncouple the ratio of hydrolysis/Ca(2+) transported. The effect of this compound on the integrity of the SR membrane loaded with Ca(2+) remained unaffected. Finally, the analysis of phosphorylation of SERCA by [γ-(32)P]ATP, starting under different conditions at 0° or 25 °C showed a reduction in the phosphoenzyme levels by 1,2-DCB, mostly at 0 °C. CONCLUSIONS: The temperature-dependent decreased levels of phosphoenzyme by 1,2-DCB could be due to the acceleration of the dephosphorylation mechanism - E2P · Ca2 state to E2 and Pi, which explains the uncoupling of the ATP hydrolysis from the Ca(2+) transport.


Asunto(s)
Clorobencenos/metabolismo , Plaguicidas/metabolismo , ATPasas Transportadoras de Calcio del Retículo Sarcoplásmico/metabolismo , Retículo Sarcoplasmático/enzimología , Adenosina Trifosfato/metabolismo , Animales , Biocatálisis , Calcio/metabolismo , Clorobencenos/química , Hidrólisis , Músculo Esquelético/metabolismo , Ácido Oxálico/química , Ácido Oxálico/metabolismo , Plaguicidas/química , Fosforilación , Conejos , ATPasas Transportadoras de Calcio del Retículo Sarcoplásmico/antagonistas & inhibidores , Temperatura
15.
BMC Neurosci ; 15: 132, 2014 Dec 31.
Artículo en Inglés | MEDLINE | ID: mdl-25552352

RESUMEN

BACKGROUND: Assembly and disassembly of microtubules (MTs) is critical for neurite outgrowth and differentiation. Evidence suggests that nerve growth factor (NGF) induces neurite outgrowth from PC12 cells by activating the receptor tyrosine kinase, TrkA. G protein-coupled receptors (GPCRs) as well as heterotrimeric G proteins are also involved in regulating neurite outgrowth. However, the possible connection between these pathways and how they might ultimately converge to regulate the assembly and organization of MTs during neurite outgrowth is not well understood. RESULTS: Here, we report that Gßγ, an important component of the GPCR pathway, is critical for NGF-induced neuronal differentiation of PC12 cells. We have found that NGF promoted the interaction of Gßγ with MTs and stimulated MT assembly. While Gßγ-sequestering peptide GRK2i inhibited neurite formation, disrupted MTs, and induced neurite damage, the Gßγ activator mSIRK stimulated neurite outgrowth, which indicates the involvement of Gßγ in this process. Because we have shown earlier that prenylation and subsequent methylation/demethylation of γ subunits are required for the Gßγ-MTs interaction in vitro, small-molecule inhibitors (L-28 and L-23) targeting prenylated methylated protein methyl esterase (PMPMEase) were tested in the current study. We found that these inhibitors disrupted Gßγ and ΜΤ organization and affected cellular morphology and neurite outgrowth. In further support of a role of Gßγ-MT interaction in neuronal differentiation, it was observed that overexpression of Gßγ in PC12 cells induced neurite outgrowth in the absence of added NGF. Moreover, overexpressed Gßγ exhibited a pattern of association with MTs similar to that observed in NGF-differentiated cells. CONCLUSIONS: Altogether, our results demonstrate that ßγ subunit of heterotrimeric G proteins play a critical role in neurite outgrowth and differentiation by interacting with MTs and modulating MT rearrangement.


Asunto(s)
Subunidades beta de la Proteína de Unión al GTP/metabolismo , Subunidades gamma de la Proteína de Unión al GTP/metabolismo , Microtúbulos/metabolismo , Factor de Crecimiento Nervioso/metabolismo , Neuritas/fisiología , Animales , Hidrolasas de Éster Carboxílico/antagonistas & inhibidores , Hidrolasas de Éster Carboxílico/metabolismo , Aumento de la Célula , Células Cultivadas , Cerebelo/citología , Cerebelo/fisiología , Hipocampo/citología , Hipocampo/fisiología , Neurogénesis/fisiología , Neuronas/citología , Neuronas/fisiología , Células PC12 , Ratas , Ratas Sprague-Dawley , Tubulina (Proteína)/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...