Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 32
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Biochem Biophys Res Commun ; 643: 30-38, 2023 Feb 05.
Artículo en Inglés | MEDLINE | ID: mdl-36586156

RESUMEN

Breast cancer is the most commonly diagnosed cancer and a leading cause of cancer-related death among women worldwide. Somatostatin (SST) and Cannabinoids have an anti-proliferative and pro-apoptotic effect, but the mechanisms of their actions remain elusive. In the present study, we have evaluated the effects of SST, Cannabidiol (CBD) alone or in combination on receptor expression, cell proliferation and apoptosis and related downstream signalling pathways in MDA-MB-231 and MCF-7 breast cancer cells. The results presented here demonstrate the cell type and agonist-dependent changes in receptor expression at the cell membrane, inhibition of cell proliferation and increased apoptosis following treatment with SST and CBD alone and in combination. In comparison to MDA-MB-231 cells, MCF-7 cells treated with SST alone and in combination with CBD exhibited inhibition of phosphorylated Protein Kinase B (pAKT) and phosphorylated-Phosphoinositide 3-Kinase (pPI3K) expression. Importantly, inhibition of PI3K/AKT activation was accompanied by enhanced PTEN expression in MCF-7 cells. These results highlight the possible interaction between SSTR and CBR subtypes with the implication in the modulation of receptor expression, cell viability and signal transduction pathways in a breast cancer cell type-dependent manner.


Asunto(s)
Neoplasias de la Mama , Cannabidiol , Femenino , Humanos , Apoptosis , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/metabolismo , Cannabidiol/farmacología , Línea Celular Tumoral , Proliferación Celular , Células MCF-7 , Fosfatidilinositol 3-Quinasas/metabolismo , Transducción de Señal , Somatostatina/farmacología
2.
Biomedicines ; 10(2)2022 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-35203546

RESUMEN

During brain development, neurite formation plays a critical role in neuronal communication and cognitive function. In the present study, we compared developmental changes in the expression of crucial markers that govern the functional activity of neurons, including somatostatin (SST), choline acetyltransferase (ChAT), tyrosine hydroxylase (TH), brain nitric oxide synthase (bNOS), gamma-aminobutyric acid (GABA), glutamic acid decarboxylase (GAD-65) and synaptic vesicle protein synaptophysin (SYP) in non-differentiated and retinoic acid (RA)-induced differentiated SH-SY5Y cells. We further determined the role of SST in regulating subcellular distribution and expression of neurotransmitters. Our results indicate that SST potentiates RA-induced differentiation of SH-SY5Y cells and involves regulating the subcellular distribution and expression of neurotransmitter markers and synaptophysin translocation to neurites in a time-dependent manner, anticipating the therapeutic implication of SST in neurodegeneration.

3.
Biochim Biophys Acta Mol Basis Dis ; 1868(3): 166325, 2022 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-34921975

RESUMEN

OBJECTIVES: Glaucoma is characterized by progressive damage of the retinal ganglion cells (RGCs), resulting in irreversible vision loss. Cannabinoids (CBs) ameliorate several factors that contribute to the progression of glaucoma, including increased intraocular pressure (IOP), degeneration of RGC and optical nerve (ON) damage. However, a direct correlation of specific CBs with the molecular events pertaining to glaucoma pathology is not well established. Therefore, this study aims to evaluate the role of cannabinol (CBN) on RGC protection, modulation of IOP, and its effects on the level of extracellular matrix (ECM) proteins using both in vitro and in vivo models of glaucoma. METHODS AND RESULTS: When exposed to elevated hydrostatic pressure, CBN, in a dose-dependent manner, protected differentiated mouse 661W retinal ganglion precursor-like cells from pressure-induced toxicity. In human trabecular meshwork cells (hTM), CBN attenuated changes in the ECM proteins, including fibronectin and α-smooth muscle actin (α-SMA), as well as mitogen-activated protein kinases (phospho-ERK1/2) in the presence or absence of transforming growth factor-beta 2 (TGF-ß2) induced stress. Ocular pharmacokinetic parameters were evaluated post-intravitreal (IVT) CBN delivery in vivo. Furthermore, we demonstrated that IVT-administered CBN improved pattern electroretinogram (pERG) amplitudes and reduced IOP in a rat episcleral vein laser photocoagulation model of glaucoma. CONCLUSION: CBN promotes neuroprotection, abrogates changes in ECM protein, and normalizes the IOP levels in the eye. Therefore, our observations in the present study indicate a therapeutic potential for CBN in the treatment of glaucoma.


Asunto(s)
Cannabinol/farmacología , Modelos Animales de Enfermedad , Glaucoma/tratamiento farmacológico , Presión Intraocular/efectos de los fármacos , Fármacos Neuroprotectores/farmacología , Células Ganglionares de la Retina/efectos de los fármacos , Malla Trabecular/efectos de los fármacos , Animales , Glaucoma/metabolismo , Glaucoma/patología , Masculino , Ratones , Ratas , Ratas Wistar , Células Ganglionares de la Retina/metabolismo , Células Ganglionares de la Retina/patología , Malla Trabecular/metabolismo , Malla Trabecular/patología , Factor de Crecimiento Transformador beta2/metabolismo
4.
Biomedicines ; 9(1)2021 Jan 02.
Artículo en Inglés | MEDLINE | ID: mdl-33401710

RESUMEN

Somatostatin is involved in the regulation of multiple signaling pathways and affords neuroprotection in response to neurotoxins. In the present study, we investigated the role of Somatostatin-14 (SST) in cell viability and the regulation of phosphorylation of Collapsin Response Mediator Protein 2 (CRMP2) (Ser522) via the blockade of Ca2+ accumulation, along with the inhibition of cyclin-dependent kinase 5 (CDK5) and Calpain activation in differentiated SH-SY5Y cells. Cell Viability and Caspase 3/7 assays suggest that the presence of SST ameliorates mitochondrial stability and cell survival pathways while augmenting pro-apoptotic pathways activated by Aß. SST inhibits the phosphorylation of CRMP2 at Ser522 site, which is primarily activated by CDK5. Furthermore, SST effectively regulates Ca2+ influx in the presence of Aß, directly affecting the activity of calpain in differentiated SH-SY5Y cells. We also demonstrated that SSTR2 mediates the protective effects of SST. In conclusion, our results highlight the regulatory role of SST in intracellular Ca2+ homeostasis. The neuroprotective role of SST via axonal regeneration and synaptic integrity is corroborated by regulating changes in CRMP2; however, SST-mediated changes in the blockade of Ca2+ influx, calpain expression, and toxicity did not correlate with CDK5 expression and p35/25 accumulation. To summarize, our findings suggest two independent mechanisms by which SST mediates neuroprotection and confirms the therapeutic implications of SST in AD as well as in other neurodegenerative diseases where the effective regulation of calcium homeostasis is required for a better prognosis.

5.
Endocrine ; 70(1): 92-106, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32335798

RESUMEN

PURPOSE: Propylthiouracil (PTU)-induced hypothyroidism is a well-established model for assessing hormonal and morphological changes in thyroid as well as other central and peripheral tissues. Somatostatin (SST) is known to regulate hormonal secretion and synthesis in endocrine tissues; however, nothing is currently known about the distribution of SST and its receptor in hypothyroidism. METHOD: In the present study, the comparative immunohistochemical distribution of SST and somatostatin receptors (SSTRs) were analyzed in PTU-induced hypothyroid rats. Rats were treated with PTU for 15 days followed by a co-administration of levothyroxine (LVT) for 15 days. After PTU and LVT treatments (day 30), rats were further administered LVT alone for 15 more days (day 45). The subcellular distribution of SST and SSTR subtypes was determined by peroxidase immunohistochemistry in the thyroid gland collected from control and treated rats. RESULTS: SST and SSTR subtypes were found to be moderately expressed in control thyroid tissues. SST and SSTR subtypes like immunoreactivity increased significantly in follicular and parafollicular epithelial cells in the thyroid of PTU-treated rats. The PTU-induced changes in the expression of SST and SSTR subtypes were suppressed by the administration of the LVT. In addition to thyroid tissues, SST and SSTRs expression was also changed in non-follicular tissues including blood vessels, smooth muscle cells, and connective tissue following treatments. CONCLUSION: The present study revealed a distinct subcellular distribution of SST and SSTR subtypes in the thyroid and provides a new insight for the role of SST and SSTR subtypes in hypothyroidism in addition to its well-established role in negative regulation of hormonal secretion.


Asunto(s)
Hipotiroidismo , Receptores de Somatostatina , Animales , Hipotiroidismo/inducido químicamente , Propiltiouracilo/toxicidad , Ratas , Somatostatina
6.
J Mol Neurosci ; 68(1): 120-134, 2019 May.
Artículo en Inglés | MEDLINE | ID: mdl-30879180

RESUMEN

Somatostatin (SST) is a growth hormone inhibitory peptide involved in regulation of several physiological responses of cells including neurotransmission, cell migration, maturation, and neurite formation. In the present study, we examined the role of SST in all-trans retinoic acid (RA)-induced progression of neurite outgrowth in SH-SY5Y cells. We also determined the morphological and developmental changes in prominent intracellular markers of neurite growth including microtubule-associated protein 2 (MAP2), neuron-specific III ß-tubulin (TUJ1), and Tau. Here, we present evidence that SST is a molecular determinant in regulating the transition of SH-SY5Y cells from non-neuronal entity to neuronal phenotype in response to RA. The results from present study reveal that SST changes the distributional pattern of MAP2/Tau and TUJ1, and activates extracellular signal-regulated kinase (ERK1/2) signaling pathway through SST receptors (SSTRs). The expression of MAP2 and Tau remains elevated upon treatment with RA and SST alone or in combination. Importantly, we identified that the cells displaying strong co-expression of SST and TUJ1 are more likely to bear elongated neurite formation than cells devoid of such expression. These findings show that the site-specific expression of MAP2 and TUJ1 is an essential determinant of neurite outgrowth in SH-SY5Y cells in RA-mediated differentiation. Taken together, results presented here further substantiates the role of SST in the promotion of neurite formation and elongation in SH-SY5Y cells in combination with RA. Investigating how SST can improve neurite formation in neurodegenerative disease may help to develop new therapeutic approach in improving cognitive function and memory loss.


Asunto(s)
Proteínas Asociadas a Microtúbulos/metabolismo , Proyección Neuronal , Somatostatina/farmacología , Tretinoina/farmacología , Línea Celular Tumoral , Humanos , Sistema de Señalización de MAP Quinasas , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Receptores de Somatostatina/metabolismo , Tubulina (Proteína)/metabolismo
7.
Mol Neurobiol ; 56(1): 292-306, 2019 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-29700775

RESUMEN

In Alzheimer's disease (AD), the impaired clearance of ß-amyloid peptide (Aß) due to disrupted tight junction and transporter proteins is the prominent cause of disease progression. Somatostatin (SST) blocks the aggregation of Aß and inflammation whereas reduction of SST levels in the CSF and brain tissue is associated with impaired cognitive function and memory loss. However, the role of SST in preservation of blood-brain barrier (BBB) integrity and functionality in Aß-induced toxicity is not known. In the present study using human CMEC/D3 cells, we demonstrate that SST prevents Aß-induced BBB permeability by regulating LRP1 and RAGE expression and improving the disrupted tight junction proteins. Furthermore, SST abrogates Aß-induced JNK phosphorylation and expression of MMP2. Taken together, results presented here suggest that SST might serve as a therapeutic intervention in AD via targeting multiple pathways responsible for neurotoxicity, impaired BBB function, and disease progression.


Asunto(s)
Péptidos beta-Amiloides/toxicidad , Barrera Hematoencefálica/patología , Somatostatina/farmacología , Barrera Hematoencefálica/efectos de los fármacos , Células Cultivadas , Citocinas/metabolismo , Humanos , Interleucina-1beta/metabolismo , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Proteína 1 Relacionada con Receptor de Lipoproteína de Baja Densidad/metabolismo , Metaloproteinasa 2 de la Matriz/metabolismo , Fármacos Neuroprotectores/farmacología , Permeabilidad , Fosforilación/efectos de los fármacos , Receptor para Productos Finales de Glicación Avanzada/metabolismo , Proteínas de Uniones Estrechas/metabolismo , Factores de Tiempo
8.
Biochim Biophys Acta Mol Basis Dis ; 1864(2): 359-373, 2018 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-29104117

RESUMEN

The present study describes the status of somatostatin receptors (SSTRs) and their colocalization with insulin (ß), glucagon (α) and somatostatin (δ) producing cells in the pancreatic islets of 11weeks old R6/2 Huntington's Disease transgenic (HD tg) and age-matched wild type (wt) mice. We also determined expression of tyrosine hydroxylase (TH), glutamic acid decarboxylase (GAD) and presynaptic marker synaptophysin (SYP) in addition to signal transduction pathways associated with diabetes. In R6/2 mice, islets are relatively smaller in size, exhibit enhanced expression and nuclear inclusion of mHtt along with the loss of insulin, glucagon and somatostatin expression. In comparison to wt, R6/2 mice display enhanced mRNA for all SSTRs except SSTR2. In the pancreatic lysate, SSTR1, 4 and 5 immunoreactivity decreases whereas SSTR3 immunoreactivity increases with no discernible changes in SSTR2 immunoreactivity. Furthermore, at the cellular level, R6/2 mice exhibit a receptor specific distributional pattern of SSTRs like immunoreactivity and colocalization with ß, α and δ cells. While GAD expression is increased, TH and SYP immunoreactivity was decreased in R6/2 mice, anticipating a cross-talk between the CNS and pancreas in diabetes pathophysiology. We also dissected out the changes in signaling pathway and found decreased activation and expression of PKA, AKT, ERK1/2 and STAT3 in R6/2 mice pancreas. These findings suggest that the impaired organization of SSTRs within islets may lead to perturbed hormonal regulation and signaling. These interconnected complex events might shed new light on the pathogenesis of diabetes in neurodegenerative diseases and the role of SSTRs in potential therapeutic intervention.


Asunto(s)
Enfermedad de Huntington/genética , Páncreas/metabolismo , Receptores de Somatostatina/genética , Receptores de Somatostatina/metabolismo , Transducción de Señal , Animales , Diabetes Mellitus/metabolismo , Modelos Animales de Enfermedad , Regulación de la Expresión Génica , Glucagón/metabolismo , Glutamato Descarboxilasa/metabolismo , Insulina/metabolismo , Islotes Pancreáticos/metabolismo , Masculino , Ratones , Ratones Transgénicos , Microscopía Confocal , ARN Mensajero/metabolismo , Somatostatina/metabolismo , Sinaptofisina/metabolismo , Tirosina 3-Monooxigenasa/metabolismo
9.
Neurosci Lett ; 640: 81-87, 2017 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-28104430

RESUMEN

In the present study, we describe the distribution of GABAC ρ3 receptor immunoreactivity in the cortex, striatum and hippocampus of wild type (wt) and 11 weeks old HD transgenic (tg) R6/2 mouse brain. In the brain of wt mice, GABAC ρ3 immunoreactivity is well expressed in neuronal cells, nerve fibers and axonal processes. In comparison to wt, GABAC ρ3 receptor like immunoreactivity decreases significantly in all three brain regions of R6/2 mice. The altered distributional pattern and significant changes in GABAC ρ3 receptor immunoreactivity as seen in the R6/2 mouse brain might be a plausible molecular mechanism for excitotoxicity in HD pathogenesis due to the loss of inhibitory input.


Asunto(s)
Encéfalo/metabolismo , Enfermedad de Huntington/metabolismo , Neuronas/metabolismo , Receptores de GABA/metabolismo , Animales , Axones/metabolismo , Corteza Cerebral/metabolismo , Cuerpo Estriado/metabolismo , Hipocampo/metabolismo , Ratones , Ratones Transgénicos , Especificidad de Órganos
10.
Neuroscience ; 340: 218-231, 2017 01 06.
Artículo en Inglés | MEDLINE | ID: mdl-27984180

RESUMEN

Endocannabinoids and somatostatin (SST) play critical roles in several pathophysiological conditions via binding to different receptor subtypes. Cannabinoid receptor 1 (CB1R) and somatostatin receptors (SSTRs) are expressed in several brain regions and share overlapping functions. Whether these two prominent members of G-protein-coupled receptor (GPCR) family interact with each other and constitute a functional receptor complex is not known. In the present study, we investigated the colocalization of CB1R and SSTR5 in rat brain, and studied receptor internalization, interaction and signal transduction pathways in HEK-293 cells cotransfected with human cannabinoid receptor 1 (hCB1R) and hSSTR5. Our results showed that CB1R and SSTR5 colocalized in rat brain cortex, striatum, and hippocampus. CB1R was expressed in SSTR5 immunoprecipitate prepared from the brain tissue lysate, indicating their association in a system where these receptors are endogenously expressed. In cotransfected HEK-293 cells, SSTR5 and CB1R existed in a constitutive heteromeric complex under basal condition, which was disrupted upon agonist treatments. Furthermore, concurrent receptor activation led to preferential formation of SSTR5 homodimer and dissociation of CB1R homodimer. We also discovered that second messenger cyclic adenosine monophosphate and downstream signaling pathways were modulated in a SSTR5-dominant and concentration-dependent manner in the presence of receptor-specific agonist. In conclusion, with predominant role of SSTR5, the functional consequences of crosstalk between SSTR5 and CB1R resulting in the regulation of receptor trafficking and signal transduction pathways open new therapeutic avenue in cancer biology and excitotoxicity.


Asunto(s)
Receptor Cannabinoide CB1/metabolismo , Receptores de Somatostatina/metabolismo , Animales , Encéfalo/metabolismo , Membrana Celular/efectos de los fármacos , Membrana Celular/metabolismo , AMP Cíclico/metabolismo , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Células HEK293 , Humanos , Masculino , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Neurotransmisores/farmacología , Fosfatidilinositol 3-Quinasas/metabolismo , Multimerización de Proteína , Ratas Sprague-Dawley , Receptor Cannabinoide CB1/agonistas , Receptor Cannabinoide CB1/genética , Receptores de Somatostatina/agonistas , Receptores de Somatostatina/genética , Transducción de Señal/fisiología
11.
J Mol Neurosci ; 55(2): 480-91, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25001005

RESUMEN

Despite several overlapping functions of cannabinoid receptor 1 (CB1 receptor), somatostatin (SST), and neuronal nitric oxide synthase (nNOS) in the hypothalamus, nothing is currently known whether CB1 receptor-positive neurons coexpress SST and nNOS. In the present study, we describe the colocalization of CB1 receptor with SST and nNOS in the rat brain hypothalamus. In the hypothalamus, the distributional patterns and colocalization of CB1 receptor with SST and nNOS were selective and region specific. CB1 receptor and SST exhibited comparable colocalization (<60%) in paraventricular nucleus (PVN) and periventricular nucleus (PeVN), followed by 20% colocalization in ventromedial hypothalamic nucleus (VMH). Neurons showing colocalization between CB1 receptor and nNOS in PeVN constituted >80%, followed by 60 and 30% in PVN and VMH, respectively. In contrast, SST- and nNOS-positive neurons displayed comparable colocalization (>55%) in PeVN and VMH, followed by PVN (~20%). In the median eminence, CB1 receptor-, SST-, and nNOS-like immunoreactivity was mostly confined to the nerve fibers. The morphological colocalization of CB1 receptor with SST and nNOS shed new light on the understanding of their roles in regulation of physiological and pharmacological response to certain stimuli in hypothalamic nuclei specifically in food intake and energy balance.


Asunto(s)
Hipotálamo/metabolismo , Óxido Nítrico Sintasa de Tipo I/metabolismo , Receptor Cannabinoide CB1/metabolismo , Somatostatina/metabolismo , Animales , Hipotálamo/citología , Masculino , Neuronas/metabolismo , Ratas , Ratas Sprague-Dawley
12.
Biochim Biophys Acta ; 1843(4): 735-45, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24412308

RESUMEN

In the present study, we report that somatostatin receptor 2 (SSTR2) plays a crucial role in modulation of ß1AR and ß2AR mediated signaling pathways that are associated with increased intracellular Ca(2+) and cardiac complications. In H9c2 cells, SSTR2 colocalizes with ß1AR or ß2AR in receptor specific manner. SSTR2 selective agonist inhibits isoproterenol and formoterol stimulated cAMP formation and PKA phosphorylation in concentration dependent manner. In the presence of SSTR2 agonist, the expression of PKCα and PKCß was comparable to the basal condition, however SSTR2 agonist inhibits isoproterenol or formoterol induced PKCα and PKCß expression, respectively. Furthermore, the activation of SSTR2 not only inhibits calcineurin expression and its activity, but also blocks NFAT dephosphorylation and its nuclear translocation. SSTR2 selective agonist abrogates isoproterenol mediated increase in cell size and protein content (an index of hypertrophy). Taken together, the results described here provide direct evidence in support of cardiac protective role of SSTR2 via modulation of Ca(2+) associated signaling pathways attributed to cardiac hypertrophy.


Asunto(s)
Calcio/metabolismo , AMP Cíclico/metabolismo , Fosforilación/efectos de los fármacos , Receptores Adrenérgicos beta 1/metabolismo , Receptores de Somatostatina/metabolismo , Animales , Línea Celular , Etanolaminas/farmacología , Fumarato de Formoterol , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Isoproterenol/farmacología , Miocitos Cardíacos/citología , Miocitos Cardíacos/metabolismo , Proteína Quinasa C beta/biosíntesis , Proteína Quinasa C beta/metabolismo , Proteína Quinasa C-alfa/biosíntesis , Proteína Quinasa C-alfa/metabolismo , Ratas , Receptores Adrenérgicos beta 2/metabolismo , Receptores de Somatostatina/agonistas , Transducción de Señal/efectos de los fármacos , Somatostatina/agonistas , Somatostatina/metabolismo
13.
PLoS One ; 9(1): e85193, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24416361

RESUMEN

Pain relief is the principal action of opioids. Somatostatin (SST), a growth hormone inhibitory peptide is also known to alleviate pain even in cases when opioids fail. Recent studies have shown that mice are prone to sustained pain and devoid of analgesic effect in the absence of somatostatin receptor 4 (SSTR4). In the present study, using brain slices, cultured neurons and HEK-293 cells, we showed that SSTR4 and δ-Opioid receptor (δOR) exist in a heteromeric complex and function in synergistic manner. SSTR4 and δOR co-expressed in cortical/striatal brain regions and spinal cord. Using cultured neuronal cells, we describe the heterogeneous complex formation of SSTR4 and δOR at neuronal cell body and processes. Cotransfected cells display inhibition of cAMP/PKA and co-activation of SSTR4 and δOR oppose receptor trafficking induced by individual receptor activation. Furthermore, downstream signaling pathways either associated with withdrawal or pain relief are modulated synergistically with a predominant role of SSTR4. Inhibition of cAMP/PKA and activation of ERK1/2 are the possible cellular adaptations to prevent withdrawal induced by chronic morphine use. Our results reveal direct intra-membrane interaction between SSTR4 and δOR and provide insights for the molecular mechanism for the anti-nociceptive property of SST in combination with opioids as a potential therapeutic approach to avoid undesirable withdrawal symptoms.


Asunto(s)
Dolor/metabolismo , Receptores Opioides delta/metabolismo , Receptores de Somatostatina/metabolismo , Transducción de Señal , Somatostatina/metabolismo , Analgésicos Opioides/farmacología , Animales , Corteza Cerebral/efectos de los fármacos , Corteza Cerebral/metabolismo , Corteza Cerebral/fisiopatología , Cuerpo Estriado/efectos de los fármacos , Cuerpo Estriado/metabolismo , Cuerpo Estriado/fisiopatología , AMP Cíclico/metabolismo , Proteínas Quinasas Dependientes de AMP Cíclico/genética , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Regulación de la Expresión Génica , Células HEK293 , Humanos , Ratones , Proteína Quinasa 1 Activada por Mitógenos/genética , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/genética , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Morfina/farmacología , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Dolor/genética , Dolor/fisiopatología , Dolor/prevención & control , Cultivo Primario de Células , Multimerización de Proteína , Receptores Opioides delta/genética , Receptores de Somatostatina/genética , Somatostatina/farmacología , Médula Espinal/efectos de los fármacos , Médula Espinal/metabolismo , Médula Espinal/fisiopatología
14.
Biochem Pharmacol ; 86(4): 497-507, 2013 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-23770458

RESUMEN

Multiple sclerosis (MS) is an inflammatory neurological disorder associated with demyelination, impaired blood brain barrier (BBB), axonal damage and neuronal loss. In the present study, we measured somatostatin (SST) and tumor necrosis factor-α (TNF-α) like immunoreactivity in CSF samples from MS and non-MS patients. We also examined the role of SST in cytokines and lipopolysaccharide (LPS)-induced damage to the BBB using human brain endothelial cells in culture. Most of the cerebrospinal fluid (CSF) samples studied from definite MS patients exhibited lower somatostatin (SST)-like immunoreactivity and higher expression of TNF-α in comparison to non-MS patients. Treatment of cells with cytokines and LPS blocked SST secretion and decreased SST expression. Human brain endothelial cells expressed all five somatostatin receptors (SSTRs) with increased expression of SSTR2 and 4 upon treatment with cytokines and LPS. Cytokines and LPS-induced disruption of the tight junction proteins Zonula occludens (ZO-1) organization was restored in presence of SST, SSTR2 or SSTR4 selective agonists. Furthermore, inflammation induced changes in extracellular signal-regulated kinases (ERK1/2 and ERK5) signaling and altered expression of endothelial and inducible nitric oxide synthase are modulated in presence of SST. These data indicate that decreased levels of SST contribute to failure of the BBB in MS.


Asunto(s)
Barrera Hematoencefálica/metabolismo , Citocinas/metabolismo , Esclerosis Múltiple/metabolismo , Somatostatina/metabolismo , Barrera Hematoencefálica/efectos de los fármacos , Encéfalo/irrigación sanguínea , Citocinas/farmacología , Células Endoteliales/efectos de los fármacos , Células Endoteliales/metabolismo , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Femenino , Humanos , Interferón gamma/farmacología , Lipopolisacáridos/farmacología , Masculino , Microvasos/citología , Microvasos/metabolismo , Óxido Nítrico Sintasa de Tipo I/metabolismo , Óxido Nítrico Sintasa de Tipo III/metabolismo , Fosforilación , Receptores de Somatostatina/agonistas , Receptores de Somatostatina/metabolismo , Transducción de Señal , Somatostatina/líquido cefalorraquídeo , Somatostatina/farmacología , Factor de Necrosis Tumoral alfa/líquido cefalorraquídeo , Factor de Necrosis Tumoral alfa/farmacología , Proteína de la Zonula Occludens-1/metabolismo
15.
Int J Cardiol ; 167(3): 1012-22, 2013 Aug 10.
Artículo en Inglés | MEDLINE | ID: mdl-22465343

RESUMEN

BACKGROUND: Somatostatin (SST), a growth hormone inhibitory peptide plays key role in regulation of cell proliferation via modulation of mitogen activated protein kinases (MAPKs) and cell survival pathway. In cardiac physiology, ß-Adrenergic receptors (ß-ARs) play crucial role in regulation of downstream signaling pathways in receptor specific manner. The aim of the current study was to delineate the mechanistic insight for the role of SST on ß-AR mediated signaling which promotes hypertrophy and apoptosis in rat fetal cardiomyocytes (H9c2 cells). Accordingly, SST dependent changes in signaling molecules including second messenger cAMP, PKA/CREB as well as MAPKs including ERK and p38 which are key mediators of hypertrophy and apoptosis were analyzed. METHODS AND RESULTS: In the present study, we determined receptor specific effects on intracellular cAMP levels, signaling by western blot analysis and apoptosis by using JC-1 and Hoechst-33258 staining. Here, we present the data which indicates that SST inhibits isoproterenol induced hypertrophy and apoptosis in H9c2 cells. Importantly, SST inhibits ß-ARs agonist induced cAMP activation and SST mediated inhibition of cAMP was enhanced in presence of ß-ARs antagonist. SST enhances ß2AR agonist formoterol mediated effects on PKA, CREB and ERK1/2 phosphorylations whereas it inhibits isoproterenol mediated ERK1/2 and p38 signaling in concentration dependent manner. CONCLUSIONS: Taken together, these results presented here provide a novel insight for the potential role of SST in regulation of ß-AR mediated effects on hypertrophy and modulation of hypertrophy promoting signaling in H9c2 cells.


Asunto(s)
Isoproterenol/toxicidad , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/patología , Transducción de Señal/efectos de los fármacos , Somatostatina/farmacología , Animales , Cardiomegalia/sangre , Cardiomegalia/inducido químicamente , Cardiomegalia/patología , Línea Celular , Isoproterenol/sangre , Isoproterenol/farmacología , Ratas , Transducción de Señal/fisiología , Somatostatina/sangre
16.
Proc Natl Acad Sci U S A ; 109(25): 9917-22, 2012 Jun 19.
Artículo en Inglés | MEDLINE | ID: mdl-22665789

RESUMEN

A chemically diverse family of small-molecule signals, the ascarosides, control developmental diapause (dauer), olfactory learning, and social behaviors of the nematode model organism, Caenorhabditis elegans. The ascarosides act upstream of conserved signaling pathways, including the insulin, TGF-ß, serotonin, and guanylyl cyclase pathways; however, the sensory processes underlying ascaroside function are poorly understood. Because ascarosides often are multifunctional and show strongly synergistic effects, characterization of their receptors will be essential for understanding ascaroside biology and may provide insight into molecular mechanisms that produce synergistic outcomes in small-molecule sensing. Based on DAF-8 immunoprecipitation, we here identify two G-protein-coupled receptors, DAF-37 and DAF-38, which cooperatively mediate ascaroside perception. daf-37 mutants are defective in all responses to ascr#2, one of the most potent dauer-inducing ascarosides, although this mutant responds normally to other ascarosides. In contrast, daf-38 mutants are partially defective in responses to several different ascarosides. Through cell-specific overexpression, we show that DAF-37 regulates dauer when expressed in ASI neurons and adult behavior when expressed in ASK neurons. Using a photoaffinity-labeled ascr#2 probe and amplified luminescence assays (AlphaScreen), we demonstrate that ascr#2 binds to DAF-37. Photobleaching fluorescent energy transfer assays revealed that DAF-37 and DAF-38 form heterodimers, and we show that heterodimerization strongly increases cAMP inhibition in response to ascr#2. These results suggest that that the ascarosides' intricate signaling properties result in part from the interaction of highly structure-specific G-protein-coupled receptors such as DAF-37 with more promiscuous G-protein-coupled receptors such as DAF-38.


Asunto(s)
Caenorhabditis elegans/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Transducción de Señal , Animales , Caenorhabditis elegans/genética , AMP Cíclico/metabolismo , Dimerización , Inmunoprecipitación , Neuronas/metabolismo , Etiquetas de Fotoafinidad , Conformación Proteica , Receptores Acoplados a Proteínas G/química
17.
Pharmaceuticals (Basel) ; 5(5): 417-46, 2012 Apr 27.
Artículo en Inglés | MEDLINE | ID: mdl-24281555

RESUMEN

G-protein coupled receptors (GPCRs) are cell surface proteins responsible for translating >80% of extracellular reception to intracellular signals. The extracellular information in the form of neurotransmitters, peptides, ions, odorants etc is converted to intracellular signals via a wide variety of effector molecules activating distinct downstream signaling pathways. All GPCRs share common structural features including an extracellular N-terminal, seven-transmembrane domains (TMs) linked by extracellular/intracellular loops and the C-terminal tail. Recent studies have shown that most GPCRs function as dimers (homo- and/or heterodimers) or even higher order of oligomers. Protein-protein interaction among GPCRs and other receptor proteins play a critical role in the modulation of receptor pharmacology and functions. Although ~50% of the current drugs available in the market target GPCRs, still many GPCRs remain unexplored as potential therapeutic targets, opening immense possibility to discover the role of GPCRs in pathophysiological conditions. This review explores the existing information and future possibilities of GPCRs as tools in clinical pharmacology and is specifically focused for the role of somatostatin receptors (SSTRs) in pathophysiology of diseases and as the potential candidate for drug discovery.

18.
J Mol Signal ; 6: 9, 2011 Aug 12.
Artículo en Inglés | MEDLINE | ID: mdl-21838893

RESUMEN

BACKGROUND: In the present study, we describe heterodimerization between human-Somatostatin Receptor 5 (hSSTR5) and ß2-Adrenergic Receptor (ß2AR) and its impact on the receptor trafficking, coupling to adenylyl cyclase and signaling including mitogen activated protein kinases and calcineurin-NFAT pathways. METHODS: We used co-immunoprecipitation, photobleaching- fluorescence resonance energy transfer and Fluorescence assisted cell sorting analysis to characterize heterodimerization between SSTR5 and ß2AR. RESULTS: Our results indicate that hSSTR5/ß2AR exist as preformed heterodimers in the basal condition which is enhanced upon co-activation of both receptors. In contrast, the activation of individual receptors leads to the dissociation of heterodimers. Receptor coupling to adenylyl cyclase displayed predominant effect of ß2AR, however, somatostatin mediated inhibition of cAMP was enhanced upon blocking ß2AR. Our results indicate hSSTR5 mediated significant activation of ERK1/2 and inhibition of phospho-p38. The phospho-NFAT level was enhanced in cotransfected cells indicating the blockade of calcineurin mediated dephosphorylation of NFAT upon receptor heterodimerization. CONCLUSION: These data for the first time unveil a novel insight for the role of hSSTR5/ß2AR in the modulation of signaling pathways which has not been addressed earlier.

19.
Biochim Biophys Acta ; 1813(6): 1172-89, 2011 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-21419811

RESUMEN

Epidermal growth factor (EGF) regulates normal and tumor cell proliferation via epidermal growth factor receptor (EGFR) phosphorylation, homo- or heterodimerization and activation of mitogen-activated protein kinases (MAPKs) and PI3K/AKT cell survival pathways. In contrast, SST via activation of five different receptor subtypes inhibits cell proliferation and has been potential target in tumor treatment. To gain further insight for the effect of SSTRs on EGFR activated signaling, we determine the role of SSTR1 and SSTR1/5 in human embryonic kidney (HEK) 293 cells. We here demonstrate that cells transfected with SSTR1 or SSTR1/5 negatively regulates EGF mediated effects attributed to the inhibition of EGFR phosphorylation, MAPKs as well as the cell survival signaling. Furthermore, SSTR effects were significantly enhanced in cells when EGFR was knock down using siRNA or treated with selective antagonist (AG1478). Most importantly, the presence of SSTR in addition to modulating signaling pathways leads to the dissociation of the constitutive and EGF induced heteromeric complex of EGFR/ErbB2. Furthermore, cells cotransfected with SSTR1/5 display pronounced effect of SST on the signaling and dissociation of the EGFR/ErbB2 heteromeric complex than the cells expressing SSTR1 alone. Taken together this study provides the first evidence that the presence of SSTR controls EGF mediated cell survival pathway via dissociation of ErbB heteromeric complex. We propose that the activation of SSTR and blockade of EGFR might serve novel therapeutic approach in inhibition of tumor proliferation.


Asunto(s)
Receptores ErbB/metabolismo , Receptores de Somatostatina/metabolismo , Transducción de Señal/fisiología , Western Blotting , Inhibidor p27 de las Quinasas Dependientes de la Ciclina/genética , Inhibidor p27 de las Quinasas Dependientes de la Ciclina/metabolismo , Factor de Crecimiento Epidérmico/farmacología , Receptores ErbB/antagonistas & inhibidores , Receptores ErbB/genética , Células HEK293 , Humanos , Inmunoprecipitación , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Fosforilación/efectos de los fármacos , Unión Proteica , Multimerización de Proteína , Proteína Tirosina Fosfatasa no Receptora Tipo 6/genética , Proteína Tirosina Fosfatasa no Receptora Tipo 6/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Quinazolinas , Interferencia de ARN , Receptor ErbB-2/genética , Receptor ErbB-2/metabolismo , Receptores de Somatostatina/genética , Transducción de Señal/efectos de los fármacos , Somatostatina/farmacología , Tirfostinos/farmacología , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
20.
Biochim Biophys Acta ; 1813(3): 390-402, 2011 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-21194548

RESUMEN

In the present study, we describe the role of cytoplasmic terminal (C-tail) domain in regulating coupling to adenylyl cyclase, signaling, and apoptosis in human embryonic kidney (HEK-293) cells transfected with wild type (wt)-hSSTR3 and C-tail deleted mutants. Cells transfected with wt-hSSTR3 and C-tail mutants show comparable membrane expression; however, display decreased expression in presence of agonist. wt-hSSTR3 exists as preformed homodimer at cell surface in basal conditions and decreases in response to agonist. Cells expressing C-tail mutants also show evidence of homodimerization with the same intensity as wt-hSSTR3. The agonist-dependent inhibition of cyclic adenosine monophosphate (cAMP) was lost in cells expressing C-tail mutants. Agonist treatment in cells expressing wt-hSSTR3 resulted in inhibition of cell proliferation, increased expression of PARP-1, and TUNEL positivity in proliferating cell nuclear antigen (PCNA)-positive cells. The agonist mediated increase in membrane expression of protein tyrosine phosphatase (PTP) seen with wt-hSSTR3 was diminished in C-tail mutants, which was accompanied with the loss of receptor's ability to induce apoptosis. Taken together, our data provide new insights into C-tail-dependent regulation of cell signaling and apoptosis by hSSTR3.


Asunto(s)
Adenilil Ciclasas/metabolismo , Apoptosis , Receptores de Somatostatina/química , Receptores de Somatostatina/metabolismo , Transducción de Señal , Secuencia de Aminoácidos , Línea Celular , Proliferación Celular , Humanos , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Datos de Secuencia Molecular , Mutación , Poli(ADP-Ribosa) Polimerasa-1 , Poli(ADP-Ribosa) Polimerasas/genética , Multimerización de Proteína , Estructura Terciaria de Proteína , Transporte de Proteínas , Proteína Tirosina Fosfatasa no Receptora Tipo 6/metabolismo , Receptores de Somatostatina/agonistas , Receptores de Somatostatina/genética , Regulación hacia Arriba
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...