Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
1.
J Cereb Blood Flow Metab ; : 271678X241248907, 2024 Apr 25.
Artículo en Inglés | MEDLINE | ID: mdl-38661094

RESUMEN

Blood-brain barrier (BBB) disruption is increasingly recognized as an early contributor to the pathophysiology of cerebral ischemia/reperfusion (I/R) injury, and is also a key event in triggering secondary damage to the central nervous system. Recently, long non-coding RNA (lncRNA) have been found to be associated with ischemic stroke. However, the roles of lncRNA in BBB homeostasis remain largely unknown. Here, we report that long intergenic non-coding RNA-p21 (lincRNA-p21) was the most significantly down-regulated lncRNA in human brain microvascular endothelial cells (HBMECs) after oxygen and glucose deprivation/reoxygenation (OGD/R) treatment among candidate lncRNA, which were both sensitive to hypoxia and involved in atherosclerosis. Exogenous brain-endothelium-specific overexpression of lincRNA-p21 could alleviate BBB disruption, diminish infarction volume and attenuate motor function deficits in middle cerebral artery occlusion/reperfusion (MCAO/R) mice. Further results showed that lincRNA-p21 was critical to maintain BBB integrity by inhibiting the degradation of junction proteins under MCAO/R and OGD/R conditions. Specifically, lincRNA-p21 could inhibit autophagy-dependent degradation of occludin by activating PI3K/AKT/mTOR signaling pathway. Besides, lincRNA-p21 could inhibit VE-cadherin degradation by binding with miR-101-3p. Together, we identify that lincRNA-p21 is critical for BBB integrity maintenance, and endothelial lincRNA-p21 overexpression could alleviate cerebral I/R injury in mice, pointing to a potential strategy to treat cerebral I/R injury.

2.
J Cell Biol ; 222(5)2023 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-36995368

RESUMEN

Microvascular basement membrane (BM) plays a pivotal role in the interactions of astrocyte with endothelium to maintain the blood-brain barrier (BBB) homeostasis; however, the significance and precise regulation of the endothelial cell-derived BM component in the BBB remain incompletely understood. Here, we report that conditional knockout of Atg7 in endothelial cells (Atg7-ECKO) leads to astrocyte-microvascular disassociation in the brain. Our results reveal astrocytic endfeet detachment from microvessels and BBB leakage in Atg7-ECKO mice. Furthermore, we find that the absence of endothelial Atg7 downregulates the expression of fibronectin, a major BM component of the BBB, causing significantly reduced coverage of astrocytes along cerebral microvessels. We reveal Atg7 triggers the expression of endothelial fibronectin via regulating PKA activity to affect the phosphorylation of cAMP-responsive element-binding protein. These results suggest that Atg7-regulated endothelial fibronectin production is required for astrocytes adhesion to microvascular wall for maintaining the BBB homeostasis. Thus, endothelial Atg7 plays an essential role in astrocyte-endothelium interactions to maintain the BBB integrity.


Asunto(s)
Astrocitos , Proteína 7 Relacionada con la Autofagia , Barrera Hematoencefálica , Animales , Ratones , Astrocitos/metabolismo , Proteína 7 Relacionada con la Autofagia/genética , Barrera Hematoencefálica/metabolismo , Células Endoteliales/metabolismo , Endotelio/metabolismo , Fibronectinas/metabolismo , Membrana Basal/metabolismo , Adhesión Celular
3.
Cell Rep ; 39(2): 110656, 2022 04 12.
Artículo en Inglés | MEDLINE | ID: mdl-35417709

RESUMEN

Tight junctions (TJs) of brain microvascular endothelial cells (BMECs) play a pivotal role in maintaining the blood-brain barrier (BBB) integrity; however, precise regulation of TJs stability in response to physiological and pathological stimuli remains elusive. Here, using RNA immunoprecipitation with next-generation sequencing (RIP-seq) and functional characterization, we identify SNHG12, a long non-coding RNA (lncRNA), as being critical for maintaining the BBB integrity by directly interacting with TJ protein occludin. The interaction between SNHG12 and occludin is oxygen adaptive and could block Itch (an E3 ubiquitin ligase)-mediated ubiquitination and degradation of occludin in human BMECs. Genetic ablation of endothelial Snhg12 in mice results in occludin reduction and BBB leakage and significantly aggravates hypoxia-induced BBB disruption. The detrimental effects of hypoxia on BBB could be alleviated by exogenous SNHG12 overexpression in brain endothelium. Together, we identify a direct TJ modulator lncRNA SNHG12 that is critical for the BBB integrity maintenance and oxygen adaption.


Asunto(s)
Barrera Hematoencefálica , ARN Largo no Codificante , Animales , Barrera Hematoencefálica/metabolismo , Células Endoteliales/metabolismo , Hipoxia/metabolismo , Ratones , Ocludina/metabolismo , Ocludina/farmacología , Oxígeno/metabolismo , ARN Largo no Codificante/genética , ARN Largo no Codificante/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo
4.
Sci Rep ; 12(1): 292, 2022 01 07.
Artículo en Inglés | MEDLINE | ID: mdl-34997175

RESUMEN

Neurogenesis in the Drosophila central brain progresses dynamically in order to generate appropriate numbers of neurons during different stages of development. Thus, a central challenge in neurobiology is to reveal the molecular and genetic mechanisms of neurogenesis timing. Here, we found that neurogenesis is significantly impaired when a novel mutation, Nuwa, is induced at early but not late larval stages. Intriguingly, when the Nuwa mutation is induced in neuroblasts of olfactory projection neurons (PNs) at the embryonic stage, embryonic-born PNs are generated, but larval-born PNs of the same origin fail to be produced. Through molecular characterization and transgenic rescue experiments, we determined that Nuwa is a loss-of-function mutation in Drosophila septin interacting protein 1 (sip1). Furthermore, we found that SIP1 expression is enriched in neuroblasts, and RNAi knockdown of sip1 using a neuroblast driver results in formation of small and aberrant brains. Finally, full-length SIP1 protein and truncated SIP1 proteins lacking either the N- or C-terminus display different subcellular localization patterns, and only full-length SIP1 can rescue the Nuwa-associated neurogenesis defect. Taken together, these results suggest that SIP1 acts as a crucial factor for specific neurogenesis programs in the early developing larval brain.


Asunto(s)
Encéfalo/metabolismo , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/metabolismo , Neurogénesis , Neuronas/metabolismo , Animales , Animales Modificados Genéticamente , Encéfalo/embriología , Proteínas de Drosophila/genética , Drosophila melanogaster/embriología , Drosophila melanogaster/genética , Regulación del Desarrollo de la Expresión Génica , Larva/genética , Larva/metabolismo , Mutación con Pérdida de Función , Transducción de Señal
5.
Front Mol Neurosci ; 13: 23, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32210761

RESUMEN

The expression of contactin-associated protein 1 (Caspr1) in brain microvascular endothelial cells (BMECs), one of the major cellular components of the neurovascular unit (NVU), has been revealed recently. However, the physiological role of Caspr1 in BMECs remains unclear. We previously reported the nonamyloidogenic processing of amyloid protein precursor (APP) pathway in the human BMECs (HBMECs). In this study, we found Caspr1 depletion reduced the levels of soluble amyloid protein precursor α (sAPPα) in the supernatant of HBMECs, which could be rescued by expression of full-length Caspr1. Our further results showed that ADAM9, the α-secretase essential for processing of APP to generate sAPPα, was decreased in Caspr1-depleted HBMECs. The reduced sAPPα secretion in Caspr1-depleted HBMECs was recovered by expression of exogenous ADAM9. Then, we identified that Caspr1 specifically regulates the expression of ADAM9, but not ADAM10 and ADAM17, at transcriptional level by nuclear factor-κB (NF-κB) signaling pathway. Caspr1 knockout attenuated the activation of NF-κB and prevented the nuclear translocation of p65 in brain endothelial cells, which was reversed by expression of full-length Caspr1. The reduced sAPPα production and ADAM9 expression upon Caspr1 depletion were effectively recovered by NF-κB agonist. The results of luciferase assays indicated that the NF-κB binding sites are located at -859 bp to -571 bp of ADAM9 promoter. Taken together, our results demonstrated that Caspr1 facilitates sAPPα production by transcriptional regulation of α-secretase ADAM9 in brain endothelial cells.

6.
Anat Rec (Hoboken) ; 302(12): 2255-2260, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31265765

RESUMEN

Cerebral angiogenesis is a key event during brain development and recovery from brain injury. We previously demonstrated that Atg7 knockout impaired angiogenesis in the mouse brain. However, the role of Atg7 in angiogenesis is not completely understood. In this study, we used human brain microvascular endothelial cells (HBMECs) to investigate the mechanism of Atg7-regulated cerebral angiogenesis. We found that Atg7 depletion specifically diminished the expression of the ß3 and γ2 chains of laminin-5, a major component of the extracellular matrix. In contrast, autophagy inhibitors did not affect laminin-5 expression, suggesting that Atg7-regulated laminin-5 expression is autophagy-independent. We also found that Atg7-regulated laminin-5 expression occurred at the transcriptional level through NF-κB signaling. Exogenous laminin-5 or the NF-κB agonist betulinic acid effectively rescued tube formation by Atg7-deficient HBMECs. Taken together, our study identified a novel mechanism by which Atg7 regulates laminin-5 expression via NF-κB to modulate tube formation by brain endothelial cells during cerebral angiogenesis. Anat Rec, 302:2255-2260, 2019. © 2019 American Association for Anatomy.


Asunto(s)
Proteína 7 Relacionada con la Autofagia/antagonistas & inhibidores , Autofagia , Encéfalo/irrigación sanguínea , Moléculas de Adhesión Celular/antagonistas & inhibidores , Endotelio Vascular/citología , Neovascularización Fisiológica , ARN Interferente Pequeño/genética , Proteína 7 Relacionada con la Autofagia/genética , Encéfalo/citología , Encéfalo/metabolismo , Moléculas de Adhesión Celular/genética , Endotelio Vascular/metabolismo , Humanos , Morfogénesis , Transducción de Señal , Kalinina
7.
J Neurogenet ; 33(1): 33-40, 2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-30686090

RESUMEN

Diverse types of neurons must be specified in the developing brain to form the functional neural circuits that are necessary for the execution of daily tasks. Here, we describe the participation of Forkhead box class O (FOXO) in cell fate specification of a small subset of Drosophila ventral olfactory projection neurons (vPNs). Using the two-color labeling system, twin-spot MARCM, we determined the temporal birth order of each vPN type, and this characterization served as a foundation to investigate regulators of cell fate specification. Flies deficient for chinmo, a known temporal cell fate regulator, exhibited a partial loss of vPNs, suggesting that the gene plays a complex role in specifying vPN cell fate and is not the only regulator of this process. Interestingly, loss of foxo function resulted in the precocious appearance of late-born vPNs in place of early-born vPNs, whereas overexpression of constitutively active FOXO caused late-born vPNs to take on a morphology reminiscent of earlier born vPNs. Taken together, these data suggest that FOXO temporally regulates vPN cell fate specification. The comprehensive identification of molecules that regulate neuronal fate specification promises to provide a better understanding of the mechanisms governing the formation of functional brain tissue.


Asunto(s)
Diferenciación Celular/fisiología , Proteínas de Drosophila/metabolismo , Factores de Transcripción Forkhead/metabolismo , Neuronas/citología , Vías Olfatorias/citología , Animales , Animales Modificados Genéticamente , Linaje de la Célula/fisiología , Neurogénesis/fisiología , Neuronas/metabolismo , Vías Olfatorias/metabolismo
8.
Med Microbiol Immunol ; 208(1): 59-68, 2019 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-30171337

RESUMEN

Escherichia coli K1 is the most common Gram-negative bacteria causing neonatal meningitis. Polymorphonuclear leukocyte (PMN) transmigration across the blood-brain barrier (BBB) is the hallmark of bacterial meningitis. Reportedly, the deletion of virulence factor cglD (E44:ΔcglD) from E44 is responsible for a less efficient PMN transendothelial migration ability. In the present study, we found that complementation of the cglD gene into E44:ΔcglD mutant strain might restore the PMN count and myeloperoxidase level in a neonatal mouse meningitis. Using human brain microvascular endothelial cells (HBMECs), the main model of the BBB in vitro, we found that E44:ΔcglD mutant strain induced a less efficient PMN adhesion to HBMECs and down-regulated chemokines CXCL1, CXCL6 and CXCL8 and adhesion molecule E-selectin, compared with the E44 strain. Complementation of cglD restored the PMN adhesion to HBMECs and the level of these proteins. E44:ΔcglD mutant strain also induced a less efficient NF-κB pathway activation in HBMECs and reduced the soluble p65 (sp65) level in the cerebral spinal fluid of newborn mice, compared with the E44 strain. Complementation of cglD restored the NF-κB pathway activation and increased the sp65 levels. This suggests that cglD in E44 contributes to NF-κB pathway activation in the brain endothelium to promote PMN adhesion to HBMECs and transendothelial migration. Our identified novel requirement of cglD for immune activation and subsequent PMN entry into the central nervous system suggests that therapies directed at neutralising this molecule will be beneficial in preventing bacterial meningitis progression.


Asunto(s)
Proteínas Bacterianas/metabolismo , Células Endoteliales/efectos de los fármacos , Endotelio/efectos de los fármacos , Escherichia coli/patogenicidad , Meningitis Bacterianas/patología , Neutrófilos/inmunología , Migración Transendotelial y Transepitelial , Factores de Virulencia/metabolismo , Animales , Animales Recién Nacidos , Antígenos Bacterianos/análisis , Adhesión Celular , Células Cultivadas , Líquido Cefalorraquídeo/química , Modelos Animales de Enfermedad , Escherichia coli/clasificación , Escherichia coli/aislamiento & purificación , Femenino , Eliminación de Gen , Prueba de Complementación Genética , Humanos , Recién Nacido , Masculino , Ratones , Polisacáridos Bacterianos/análisis , Factor de Transcripción ReIA/análisis
9.
Front Neurol ; 9: 998, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30555402

RESUMEN

Ischemic strokes often result in cerebral injury due to ischemia/reperfusion (I/R). Although the local inflammatory responses are known to play a primary role in the brain I/R injury, the underlying mechanism remains unclear. In the current study, we investigated the effect of brain endothelial Atg7 (autophagy related 7) depletion in the acute brain injury induced by ischemia and reperfusion. Endothelial knockout of Atg7 in mice (Atg7 eKO) was found to significantly attenuate both the infarct volume and the neurological defects induced by I/R when compared to the controls. In fact, brain inflammatory responses induced by I/R were alleviated by the Atg7 eKO. Furthermore, an increased expression of pro-inflammatory cytokines, including IL-1ß, IL-6, IL-8, and TNF-α, was observed in brain endothelial cells in response to oxygen/glucose depletion/reoxygenation, which was decreased by the shRNA-mediated Atg7 knockdown. Interestingly, Atg7 knockdown reduced IKKß phosphorylation, leading to NF-κB deactivation and downregulation of the pro-inflammatory cytokines mRNA levels. Further, Atg7 transcriptional regulation function is independent of its role in autophagy. Taken together, our results demonstrated that brain endothelial Atg7 contributes to brain damage during I/R by modulating the expression of pro-inflammatory cytokines. Depletion of Atg7 in brain endothelium has a neuroprotective effect against the ischemia/reperfusion-induced acute cerebral injury during stroke.

10.
Nat Commun ; 9(1): 2296, 2018 06 12.
Artículo en Inglés | MEDLINE | ID: mdl-29895952

RESUMEN

Escherichia coli is the leading cause of neonatal Gram-negative bacterial meningitis, but the pathogenesis of E. coli meningitis remains elusive. E. coli penetration of the blood-brain barrier (BBB) is the critical step for development of meningitis. Here, we identify Caspr1, a single-pass transmembrane protein, as a host receptor for E. coli virulence factor IbeA to facilitate BBB penetration. Genetic ablation of endothelial Caspr1 and blocking IbeA-Caspr1 interaction effectively prevent E. coli penetration into the brain during meningitis in rodents. IbeA interacts with extracellular domain of Caspr1 to activate focal adhesion kinase signaling causing E. coli internalization into the brain endothelial cells of BBB. E. coli can invade hippocampal neurons causing apoptosis dependent on IbeA-Caspr1 interaction. Our results indicate that E. coli exploits Caspr1 as a host receptor for penetration of BBB resulting in meningitis, and that Caspr1 might be a useful target for prevention or therapy of E. coli meningitis.


Asunto(s)
Moléculas de Adhesión Celular Neuronal/metabolismo , Escherichia coli/patogenicidad , Meningitis por Escherichia coli/metabolismo , Animales , Apoptosis , Barrera Hematoencefálica , Encéfalo/metabolismo , Membrana Celular/metabolismo , Supervivencia Celular , Células Endoteliales/metabolismo , Proteínas de Escherichia coli/metabolismo , Femenino , Quinasa 1 de Adhesión Focal/metabolismo , Células HEK293 , Hipocampo/metabolismo , Humanos , Masculino , Proteínas de la Membrana/metabolismo , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Microcirculación , Neuronas/metabolismo , Ratas , Ratas Sprague-Dawley
11.
Int J Mol Sci ; 18(5)2017 May 03.
Artículo en Inglés | MEDLINE | ID: mdl-28467355

RESUMEN

The formation of brain vasculature is an essential step during central nervous system development. The molecular mechanism underlying brain angiogenesis remains incompletely understood. The role of Atg7, an autophagy-related protein, in brain angiogenesis was investigated in this study. We found that the microvessel density in mice brains with endothelial-specific knockout of Atg7 (Atg7 EKO) was significantly decreased compared to wild-type control. Consistently, in vitro angiogenesis assays showed that Atg7 knockdown impaired angiogenesis in brain microvascular endothelial cells. Further results indicated that knockdown of Atg7 reduced interleukin-6 (IL-6) expression in brain microvascular endothelial cells, which is mediated by NF-κB-dependent transcriptional control. Interestingly, exogenous IL-6 restored the impaired angiogenesis and reduced cell motility caused by Atg7 knockdown. These results demonstrated that Atg7 has proangiogenic activity in brain angiogenesis which is mediated by IL-6 production in a NF-κB-dependent manner.


Asunto(s)
Proteína 7 Relacionada con la Autofagia/metabolismo , Encéfalo/irrigación sanguínea , Interleucina-6/metabolismo , FN-kappa B/metabolismo , Neovascularización Fisiológica/fisiología , Análisis de Varianza , Animales , Proteína 7 Relacionada con la Autofagia/genética , Movimiento Celular/fisiología , Células Cultivadas , Modelos Animales de Enfermedad , Células Endoteliales , Humanos , Ratones , Ratones Noqueados , Microvasos/crecimiento & desarrollo , Microvasos/metabolismo , Neovascularización Fisiológica/genética
12.
Sci Rep ; 6: 39141, 2016 12 23.
Artículo en Inglés | MEDLINE | ID: mdl-28008974

RESUMEN

MicroRNA-34 (miR-34) is crucial for preventing chronic large-scale neurite degeneration in the aged brain of Drosophila melanogaster. Here we investigated the role of miR-34 in two other types of large-scale axon degeneration in Drosophila: axotomy-induced axon degeneration in olfactory sensory neurons (OSNs) and developmentally related axon pruning in mushroom body (MB) neurons. Ectopically overexpressed miR-34 did not inhibit axon degeneration in OSNs following axotomy, whereas ectopically overexpressed miR-34 in differentiated MB neurons impaired γ axon pruning. Intriguingly, the miR-34-induced γ axon pruning defect resulted from downregulating the expression of ecdysone receptor B1 (EcR-B1) in differentiated MB γ neurons. Notably, the separate overexpression of EcR-B1 or a transforming growth factor- ß receptor Baboon, whose activation can upregulate the EcR-B1 expression, in MB neurons rescued the miR-34-induced γ axon pruning phenotype. Future investigations of miR-34 targets that regulate the expression of EcR-B1 in MB γ neurons are warranted to elucidate pathways that regulate axon pruning, and to provide insight into mechanisms that control large-scale axon degeneration in the nervous system.


Asunto(s)
Regulación hacia Abajo , Drosophila melanogaster/crecimiento & desarrollo , MicroARNs/genética , Cuerpos Pedunculados/citología , Receptores de Esteroides/metabolismo , Animales , Axotomía , Diferenciación Celular , Drosophila melanogaster/genética , Regulación del Desarrollo de la Expresión Génica , Cuerpos Pedunculados/crecimiento & desarrollo , Plasticidad Neuronal , Neuronas Receptoras Olfatorias/citología
13.
PLoS One ; 11(5): e0155384, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27163287

RESUMEN

In the Drosophila olfactory system, odorant information is sensed by olfactory sensory neurons and relayed from the primary olfactory center, the antennal lobe (AL), to higher olfactory centers via olfactory projection neurons (PNs). A major portion of the AL is constituted with dendrites of four groups of PNs, anterodorsal PNs (adPNs), lateral PNs (lPNs), lateroventral PNs (lvPNs) and ventral PNs (vPNs). Previous studies have been focused on the development and function of adPNs and lPNs, while the investigation on those of lvPNs and vPNs received less attention. Here, we study the molecular and cellular mechanisms underlying the morphogenesis of a putative male-pheromone responding vPN, the DA1 vPN. Using an intersection strategy to remove background neurons labeled within a DA1 vPN-containing GAL4 line, we depicted morphological changes of the DA1 vPN that occurs at the pupal stage. We then conducted a pilot screen using RNA interference knock-down approach to identify cell surface molecules, including Down syndrome cell adhesion molecule 1 and Semaphorin-1a, that might play essential roles for the DA1 vPN morphogenesis. Taken together, by revealing molecular and cellular basis of the DA1 vPN morphogenesis, we should provide insights into future comprehension of how vPNs are assembled into the olfactory neural circuitry.


Asunto(s)
Drosophila melanogaster/genética , Regulación del Desarrollo de la Expresión Génica , Morfogénesis/genética , Vías Olfatorias/metabolismo , Neuronas Receptoras Olfatorias/metabolismo , Atractivos Sexuales/metabolismo , Animales , Animales Modificados Genéticamente , Moléculas de Adhesión Celular , Dendritas/metabolismo , Dendritas/ultraestructura , Proteínas de Drosophila/antagonistas & inhibidores , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/anatomía & histología , Drosophila melanogaster/crecimiento & desarrollo , Drosophila melanogaster/metabolismo , Masculino , Moléculas de Adhesión de Célula Nerviosa/antagonistas & inhibidores , Moléculas de Adhesión de Célula Nerviosa/genética , Moléculas de Adhesión de Célula Nerviosa/metabolismo , Vías Olfatorias/ultraestructura , Neuronas Receptoras Olfatorias/ultraestructura , Pupa/anatomía & histología , Pupa/genética , Pupa/crecimiento & desarrollo , Pupa/metabolismo , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , Semaforinas/antagonistas & inhibidores , Semaforinas/genética , Semaforinas/metabolismo , Atractivos Sexuales/genética , Transducción de Señal , Olfato/fisiología , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
14.
J Pineal Res ; 58(4): 429-38, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25752481

RESUMEN

Melatonin induces apoptosis in many different cancer cell lines, including colorectal cancer. However, the precise mechanisms involved remain largely unresolved. In this study, we provide evidence to reveal a new mechanism by which melatonin induces apoptosis of colorectal cancer LoVo cells. Melatonin at pharmacological concentrations significantly suppressed cell proliferation and induced apoptosis in a dose-dependent manner. The observed apoptosis was accompanied by the melatonin-induced dephosphorylation and nuclear import of histone deacetylase 4 (HDAC4). Pretreatment with a HDAC4-specific siRNA effectively attenuated the melatonin-induced apoptosis, indicating that nuclear localization of HDAC4 is required for melatonin-induced apoptosis. Moreover, constitutively active Ca(2+) /calmodulin-dependent protein kinase II alpha (CaMKIIα) abrogated the melatonin-induced HDAC4 nuclear import and apoptosis of LoVo cells. Furthermore, melatonin decreased H3 acetylation on bcl-2 promoter, leading to a reduction of bcl-2 expression, whereas constitutively active CaMKIIα(T286D) or HDAC4-specific siRNA abrogated the effect of melatonin. In conclusion, the present study provides evidence that melatonin-induced apoptosis in colorectal cancer LoVo cells largely depends on the nuclear import of HDAC4 and subsequent H3 deacetylation via the inactivation of CaMKIIα.


Asunto(s)
Apoptosis/efectos de los fármacos , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/metabolismo , Neoplasias Colorrectales/metabolismo , Histona Desacetilasas/metabolismo , Melatonina/farmacología , Proteínas Represoras/metabolismo , Acetilación/efectos de los fármacos , Línea Celular Tumoral , Inmunoprecipitación de Cromatina , Humanos
15.
Brain Res ; 1604: 15-24, 2015 Apr 16.
Artículo en Inglés | MEDLINE | ID: mdl-25661252

RESUMEN

Histone deacetylase 5 (HDAC5) undergoes signal-dependent shuttling between the nucleus and cytoplasm, which is regulated in part by calcium/calmodulin-dependent kinase (CaMK)-mediated phosphorylation. Here, we report that HDAC5 regulates the survival of cortical neurons in pathological conditions. HDAC5 was evenly localized to the nucleus and cytoplasm in cultured cortical neurons. However, in response to 50µM NMDA conditions that induced neuronal cell apoptosis, nuclear-distributed HDAC5 was rapidly phosphorylated and translocated into cytoplasm of the cultured cortical neurons. Treatment with a CaMKII inhibitor KN93 suppressed HDAC5 phosphorylation and nuclear translocation induced by NMDA, whereas constitutively active CaMKIIα (T286D) stimulated HDAC5 nuclear export. Importantly, we showed that ectopic expression of nuclear-localized HDAC5 in cortical neurons suppressed NMDA-induced apoptosis. Finally, inactivation of HDAC5 by treatment with the class II-specific HDAC inhibitor trichostatin A (TSA) promoted NMDA-induced neuronal cell apoptosis. Altogether, these data identify HDAC5 and its intracellular translocation as key effectors of multiple pathways that regulate neuronal cell apoptosis.


Asunto(s)
Apoptosis/fisiología , Histona Desacetilasas/metabolismo , Neuronas/citología , Neuronas/enzimología , Acetilación , Animales , Apoptosis/efectos de los fármacos , Bencilaminas/farmacología , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/metabolismo , Núcleo Celular/metabolismo , Células Cultivadas , Corteza Cerebral/citología , Corteza Cerebral/enzimología , Activación Enzimática , Inhibidores de Histona Desacetilasas/farmacología , N-Metilaspartato/farmacología , Neuronas/efectos de los fármacos , Fosforilación , Transporte de Proteínas , Ratas , Ratas Sprague-Dawley , Sulfonamidas/farmacología
16.
Biomaterials ; 35(25): 6998-7007, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24857291

RESUMEN

The development of diagnostic/therapeutic strategies against metastasis-related molecular targets is critical for improving the survival rate of cancer patients. Subtractive Cell-SELEX was performed using highly metastatic colorectal cancer (CRC) LoVo cells and non-metastatic HCT-8 cells as the target and negative cells, respectively, for the selection of metastatic-specific aptamers. This process generated seven aptamers that displayed highly specific binding to the target cells with Kds in the nanomolar range. Based on the distinct chemical/biological properties of their individual cell surface targets, the aptamers were separately functionalized: the receptor-targeting aptamer W14 was used as a carrier for doxorubicin, resulting in the specific delivery of the drug to the target cells and a significant reduction of its cytotoxicity to non-target cells, and the non-receptor-binding aptamer W3 was used as a molecular probe conjugated to quantum dots for the targeted imaging of metastatic cancer cell lines, spontaneous lung metastasis murine tissue, and metastatic CRC patient tissues. In addition, these aptamers can be used in combination due to their lack of detectable mutual-binding interference. The study demonstrates that a panel of aptamers that recognize distinct features of target molecules can be obtained through single Cell-SELEX selection, and the selected aptamers may be individually functionalized for specific applications and/or utilized in combination.


Asunto(s)
Aptámeros de Nucleótidos/química , Neoplasias Colorrectales/metabolismo , Sistemas de Liberación de Medicamentos/métodos , Técnica SELEX de Producción de Aptámeros/métodos , Animales , Células CHO , Línea Celular Tumoral , Cricetulus , Diagnóstico por Imagen/métodos , Doxorrubicina/farmacología , Femenino , Humanos , Ratones , Ratones Desnudos , Sondas Moleculares , Células 3T3 NIH , Puntos Cuánticos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA