Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 361
Filtrar
1.
Neuromolecular Med ; 26(1): 18, 2024 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-38691185

RESUMEN

Seipin is a key regulator of lipid metabolism, the deficiency of which leads to severe lipodystrophy. Hypothalamus is the pivotal center of brain that modulates appetite and energy homeostasis, where Seipin is abundantly expressed. Whether and how Seipin deficiency leads to systemic metabolic disorders via hypothalamus-involved energy metabolism dysregulation remains to be elucidated. In the present study, we demonstrated that Seipin-deficiency induced hypothalamic inflammation, reduction of anorexigenic pro-opiomelanocortin (POMC), and elevation of orexigenic agonist-related peptide (AgRP). Importantly, administration of rosiglitazone, a thiazolidinedione antidiabetic agent, rescued POMC and AgRP expression, suppressed hypothalamic inflammation, and restored energy homeostasis in Seipin knockout mice. Our findings offer crucial insights into the mechanism of Seipin deficiency-associated energy imbalance and indicates that rosiglitazone could serve as potential intervening agent towards metabolic disorders linked to Seipin.


Asunto(s)
Proteína Relacionada con Agouti , Metabolismo Energético , Subunidades gamma de la Proteína de Unión al GTP , Homeostasis , Hipotálamo , Ratones Noqueados , Proopiomelanocortina , Rosiglitazona , Animales , Ratones , Hipotálamo/metabolismo , Metabolismo Energético/efectos de los fármacos , Proopiomelanocortina/genética , Proopiomelanocortina/biosíntesis , Proteína Relacionada con Agouti/genética , Subunidades gamma de la Proteína de Unión al GTP/genética , Rosiglitazona/farmacología , Masculino , Enfermedades Neuroinflamatorias/etiología , Ratones Endogámicos C57BL , Hipoglucemiantes/farmacología , Hipoglucemiantes/uso terapéutico , Neuropéptidos/genética , Neuropéptidos/deficiencia , Regulación de la Expresión Génica/efectos de los fármacos
2.
J Neurosci ; 41(33): 7148-7159, 2021 08 18.
Artículo en Inglés | MEDLINE | ID: mdl-34210784

RESUMEN

Following stroke, the survival of neurons and their ability to reestablish connections is critical to functional recovery. This is strongly influenced by the balance between neuronal excitation and inhibition. In the acute phase of experimental stroke, lethal hyperexcitability can be attenuated by positive allosteric modulation of GABAA receptors (GABAARs). Conversely, in the late phase, negative allosteric modulation of GABAAR can correct the suboptimal excitability and improves both sensory and motor recovery. Here, we hypothesized that octadecaneuropeptide (ODN), an endogenous allosteric modulator of the GABAAR synthesized by astrocytes, influences the outcome of ischemic brain tissue and subsequent functional recovery. We show that ODN boosts the excitability of cortical neurons, which makes it deleterious in the acute phase of stroke. However, if delivered after day 3, ODN is safe and improves motor recovery over the following month in two different paradigms of experimental stroke in mice. Furthermore, we bring evidence that, during the subacute period after stroke, the repairing cortex can be treated with ODN by means of a single hydrogel deposit into the stroke cavity.SIGNIFICANCE STATEMENT Stroke remains a devastating clinical challenge because there is no efficient therapy to either minimize neuronal death with neuroprotective drugs or to enhance spontaneous recovery with neurorepair drugs. Around the brain damage, the peri-infarct cortex can be viewed as a reservoir of plasticity. However, the potential of wiring new circuits in these areas is restrained by a chronic excess of GABAergic inhibition. Here we show that an astrocyte-derived peptide, can be used as a delayed treatment, to safely correct cortical excitability and facilitate sensorimotor recovery after stroke.


Asunto(s)
Inhibidor de la Unión a Diazepam/uso terapéutico , Agonistas de Receptores de GABA-A/uso terapéutico , Neuronas/efectos de los fármacos , Neuropéptidos/uso terapéutico , Fragmentos de Péptidos/uso terapéutico , Receptores de GABA-A/efectos de los fármacos , Accidente Cerebrovascular/tratamiento farmacológico , Adulto , Animales , Astrocitos/metabolismo , Depresión de Propagación Cortical/fisiología , Inhibidor de la Unión a Diazepam/deficiencia , Inhibidor de la Unión a Diazepam/fisiología , Implantes de Medicamentos , Potenciales Evocados Somatosensoriales , Femenino , Agonistas de Receptores de GABA-A/farmacología , Humanos , Hidrogeles , Infarto de la Arteria Cerebral Media/tratamiento farmacológico , Trombosis Intracraneal/tratamiento farmacológico , Trombosis Intracraneal/etiología , Luz , Ratones , Ratones Endogámicos C57BL , N-Metilaspartato/toxicidad , Neuronas/fisiología , Neuropéptidos/deficiencia , Neuropéptidos/fisiología , Técnicas de Placa-Clamp , Fragmentos de Péptidos/deficiencia , Fragmentos de Péptidos/fisiología , Ratas , Rosa Bengala/efectos de la radiación , Rosa Bengala/toxicidad , Método Simple Ciego , Accidente Cerebrovascular/etiología
3.
Nat Commun ; 12(1): 2335, 2021 04 20.
Artículo en Inglés | MEDLINE | ID: mdl-33879799

RESUMEN

Current therapeutic options for treating colorectal cancer have little clinical efficacy and acquired resistance during treatment is common, even following patient stratification. Understanding the mechanisms that promote therapy resistance may lead to the development of novel therapeutic options that complement existing treatments and improve patient outcome. Here, we identify RAC1B as an important mediator of colorectal tumourigenesis and a potential target for enhancing the efficacy of EGFR inhibitor treatment. We find that high RAC1B expression in human colorectal cancer is associated with aggressive disease and poor prognosis and deletion of Rac1b in a mouse colorectal cancer model reduces tumourigenesis. We demonstrate that RAC1B interacts with, and is required for efficient activation of the EGFR signalling pathway. Moreover, RAC1B inhibition sensitises cetuximab resistant human tumour organoids to the effects of EGFR inhibition, outlining a potential therapeutic target for improving the clinical efficacy of EGFR inhibitors in colorectal cancer.


Asunto(s)
Neoplasias Colorrectales/etiología , Neoplasias Colorrectales/metabolismo , Proteína de Unión al GTP rac1/metabolismo , Animales , Antineoplásicos Inmunológicos/farmacología , Carcinogénesis , Línea Celular Tumoral , Cetuximab/farmacología , Neoplasias Colorrectales/genética , Resistencia a Antineoplásicos , Receptores ErbB/antagonistas & inhibidores , Receptores ErbB/metabolismo , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Neuropéptidos/deficiencia , Neuropéptidos/genética , Neuropéptidos/metabolismo , Transducción de Señal , Regulación hacia Arriba , Vía de Señalización Wnt , Proteína de Unión al GTP rac1/deficiencia , Proteína de Unión al GTP rac1/genética
4.
Sci Rep ; 11(1): 2372, 2021 01 27.
Artículo en Inglés | MEDLINE | ID: mdl-33504822

RESUMEN

Neuronal calcium sensor 1 (NCS1) regulates a wide range of cellular functions throughout the mammalian nervous systems. Altered NCS1 expression is associated with neurodevelopmental and neurodegenerative diseases. Previous studies focused on affective and cognitive behaviors in NCS1 knockout (KO) mice, but little is known about the physiological and pathological states associated with the loss of NCS1 in the peripheral nervous system. We previously reported that NCS1 expression was reduced following paclitaxel-induced peripheral neuropathy. Here, we comprehensively investigated the phenotypes of NCS1-KO mice through a battery of behavioral tests examining both central and peripheral nervous systems. Generally, only mild differences were observed in thermal sensation and memory acquisition between NCS1-WT and -KO male mice, but not in female mice. No differences were observed in motor performance, affective behaviors, and hearing in both sexes. These results suggest that NCS1 plays a modulatory role in sensory perceptions and cognition, particularly in male mice. NCS1 has been proposed as a pharmacological target for various diseases. Therefore, the sex-specific effects of NCS1 loss may be of clinical interest. As we examined a constitutive KO model, future studies focusing on various conditional KO models will further elucidate the precise physiological significance of NCS1.


Asunto(s)
Proteínas Sensoras del Calcio Neuronal/deficiencia , Neuropéptidos/deficiencia , Fenotipo , Desempeño Psicomotor , Animales , Conducta Animal , Masculino , Ratones , Ratones Noqueados , Actividad Motora , Estimulación Física , Sensación , Temperatura
5.
Sci Rep ; 10(1): 16947, 2020 10 12.
Artículo en Inglés | MEDLINE | ID: mdl-33046791

RESUMEN

Protein 4.1N, a member of the protein 4.1 family, is highly expressed in the brain. But its function remains to be fully defined. Using 4.1N-/- mice, we explored the function of 4.1N in vivo. We show that 4.1N-/- mice were born at a significantly reduced Mendelian ratio and exhibited high mortality between 3 to 5 weeks of age. Live 4.1N-/- mice were smaller than 4.1N+/+ mice. Notably, while there were no significant differences in organ/body weight ratio for most of the organs, the testis/body and ovary/body ratio were dramatically decreased in 4.1N-/- mice, demonstrating selective effects of 4.1N deficiency on the development of the reproductive systems. Histopathology of the reproductive organs showed atrophy of both testis and ovary. Specifically, in the testis there is a lack of spermatogenesis, lack of leydig cells and lack of mature sperm. Similarly, in the ovary there is a lack of follicular development and lack of corpora lutea formation, as well as lack of secretory changes in the endometrium. Examination of pituitary glands revealed that the secretory granules were significantly decreased in pituitary glands of 4.1N-/- compared to 4.1N+/+. Moreover, while GnRH was expressed in both neuronal cell body and axons in the hypothalamus of 4.1N+/+ mice, it was only expressed in the cell body but not the axons of 4.1N-/- mice. Our findings uncover a novel role for 4.1N in the axis of hypothalamus-pituitary gland-reproductive system.


Asunto(s)
Proteínas del Citoesqueleto/deficiencia , Proteínas del Citoesqueleto/fisiología , Genitales/metabolismo , Genitales/patología , Proteínas de la Membrana/deficiencia , Proteínas de la Membrana/fisiología , Neuropéptidos/deficiencia , Neuropéptidos/fisiología , Sistemas Neurosecretores/metabolismo , Sistemas Neurosecretores/patología , Animales , Proteínas del Citoesqueleto/genética , Femenino , Hormona Liberadora de Gonadotropina/genética , Hormona Liberadora de Gonadotropina/metabolismo , Humanos , Hipotálamo/metabolismo , Hipotálamo/patología , Masculino , Proteínas de la Membrana/genética , Ratones Noqueados , Neuropéptidos/genética , Tamaño de los Órganos , Ovario/patología , Hipófisis/metabolismo , Hipófisis/patología , Espermatogénesis/genética , Testículo/patología
6.
J Biol Chem ; 295(29): 9948-9958, 2020 07 17.
Artículo en Inglés | MEDLINE | ID: mdl-32471868

RESUMEN

Neurofibromatosis type 1 (NF1) is a common cancer predisposition syndrome caused by mutations in the NF1 tumor suppressor gene. NF1 encodes neurofibromin, a GTPase-activating protein for RAS proto-oncogene GTPase (RAS). Plexiform neurofibromas are a hallmark of NF1 and result from loss of heterozygosity of NF1 in Schwann cells, leading to constitutively activated p21RAS. Given the inability to target p21RAS directly, here we performed an shRNA library screen of all human kinases and Rho-GTPases in a patient-derived NF1-/- Schwann cell line to identify novel therapeutic targets to disrupt PN formation and progression. Rho family members, including Rac family small GTPase 1 (RAC1), were identified as candidates. Corroborating these findings, we observed that shRNA-mediated knockdown of RAC1 reduces cell proliferation and phosphorylation of extracellular signal-regulated kinase (ERK) in NF1-/- Schwann cells. Genetically engineered Nf1flox/flox;PostnCre+ mice, which develop multiple PNs, also exhibited increased RAC1-GTP and phospho-ERK levels compared with Nf1flox/flox;PostnCre- littermates. Notably, mice in which both Nf1 and Rac1 loci were disrupted (Nf1flox/floxRac1flox/flox;PostnCre+) were completely free of tumors and had normal phospho-ERK activity compared with Nf1flox/flox ;PostnCre+ mice. We conclude that the RAC1-GTPase is a key downstream node of RAS and that genetic disruption of the Rac1 allele completely prevents PN tumor formation in vivo in mice.


Asunto(s)
Técnicas de Silenciamiento del Gen , Neoplasias Primarias Secundarias , Neurofibroma Plexiforme , Neurofibromatosis 1 , Neuropéptidos/deficiencia , Proteína de Unión al GTP rac1/deficiencia , Animales , Ratones , Ratones Noqueados , Neoplasias Primarias Secundarias/enzimología , Neoplasias Primarias Secundarias/genética , Neoplasias Primarias Secundarias/patología , Neoplasias Primarias Secundarias/prevención & control , Neurofibroma Plexiforme/enzimología , Neurofibroma Plexiforme/genética , Neurofibroma Plexiforme/prevención & control , Neurofibromatosis 1/enzimología , Neurofibromatosis 1/genética , Neurofibromatosis 1/patología , Neurofibromina 1/deficiencia , Neurofibromina 1/metabolismo , Neuropéptidos/metabolismo , Proto-Oncogenes Mas , Proteína de Unión al GTP rac1/metabolismo
7.
Amino Acids ; 52(4): 543-553, 2020 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-32236698

RESUMEN

The aim of the current study was to investigate whether doublecortin (DCX), insulin-like growth factor receptor 1 (IGF-1R) and metabotropic glutamate receptor 5 (mGluR5) levels are indeed modified in the aging rat hippocampal individual subareas (rather than total hippocampal tissue as in previous reports) at the protein and mRNA level and whether the methylation status contributes to these changes. Since the aging population is not homogeneous in terms of spatial memory performance, we examined whether changes in DCX, IGF-1R and mGluR5 are linked to cognitive aging. Aged (22 months) male Sprague Dawley rats were trained in the hole-board, a spatial memory task, and were subdivided according to performance to aged impaired and aged unimpaired groups. Age- and memory performance-dependent changes in mRNA steady-state levels, protein levels and DNA methylation status of DCX, IGF-1R and mGluR5 were evaluated by RT-PCR, immunoblotting and bisulfite pyrosequencing. Extending previous findings, we detected decreased DCX protein and mRNA levels in dentate gyrus (DG) of aged animals. IGF-1 signaling is a key event and herein we show that mRNA levels for IGF-1R were unchanged although reduced at the protein level. This finding may simply reflect that these protein levels are regulated at the level of protein synthesis as well as protein degradation. We provide evidence that promoter methylation is not involved in regulation of mRNA and protein levels of DCX, IGF-1R and mGluR5 during aging. Moreover, there was no significant difference between aged rats with impaired and aged rats with unimpaired memory at the protein and mRNA level. Findings propose that changes in the abovementioned protein levels may not be relevant for performance in the spatial memory task used in aged rats.


Asunto(s)
Hipocampo/metabolismo , Proteínas Asociadas a Microtúbulos/deficiencia , Neuropéptidos/deficiencia , Receptor IGF Tipo 1/deficiencia , Envejecimiento/metabolismo , Animales , Cognición , Metilación de ADN , Proteínas de Dominio Doblecortina , Proteína Doblecortina , Masculino , Proteínas Asociadas a Microtúbulos/análisis , Proteínas Asociadas a Microtúbulos/genética , Neuropéptidos/análisis , Neuropéptidos/genética , Regiones Promotoras Genéticas , Ratas , Ratas Sprague-Dawley , Receptor IGF Tipo 1/análisis , Receptor IGF Tipo 1/genética , Receptor del Glutamato Metabotropico 5/análisis , Receptor del Glutamato Metabotropico 5/genética , Receptor del Glutamato Metabotropico 5/metabolismo , Memoria Espacial
8.
Artículo en Inglés | MEDLINE | ID: mdl-32114486

RESUMEN

INTRODUCTION: 26RFa (pyroglutamyl RFamide peptide (QRFP)) is a biologically active peptide that has been found to control feeding behavior by stimulating food intake, and to regulate glucose homeostasis by acting as an incretin. The aim of the present study was thus to investigate the impact of 26RFa gene knockout on the regulation of energy and glucose metabolism. RESEARCH DESIGN AND METHODS: 26RFa mutant mice were generated by homologous recombination, in which the entire coding region of prepro26RFa was replaced by the iCre sequence. Energy and glucose metabolism was evaluated through measurement of complementary parameters. Morphological and physiological alterations of the pancreatic islets were also investigated. RESULTS: Our data do not reveal significant alteration of energy metabolism in the 26RFa-deficient mice except the occurrence of an increased basal metabolic rate. By contrast, 26RFa mutant mice exhibited an altered glycemic phenotype with an increased hyperglycemia after a glucose challenge associated with an impaired insulin production, and an elevated hepatic glucose production. Two-dimensional and three-dimensional immunohistochemical experiments indicate that the insulin content of pancreatic ß cells is much lower in the 26RFa-/- mice as compared with the wild-type littermates. CONCLUSION: Disruption of the 26RFa gene induces substantial alteration in the regulation of glucose homeostasis, with in particular a deficit in insulin production by the pancreatic islets. These findings further support the notion that 26RFa is an important regulator of glucose homeostasis.


Asunto(s)
Glucemia/metabolismo , Homeostasis/genética , Neuropéptidos/deficiencia , Neuropéptidos/genética , Animales , Metabolismo Energético/genética , Conducta Alimentaria , Técnicas de Inactivación de Genes , Hiperglucemia/genética , Hiperglucemia/metabolismo , Insulina/biosíntesis , Células Secretoras de Insulina/metabolismo , Locomoción/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Fenotipo
9.
Arterioscler Thromb Vasc Biol ; 40(3): 714-732, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-31996022

RESUMEN

OBJECTIVE: Calcification of atherosclerotic plaque is traditionally associated with increased cardiovascular event risk; however, recent studies have found increased calcium density to be associated with more stable disease. 3-hydroxy-3-methylglutaryl coenzymeA reductase inhibitors or statins reduce cardiovascular events. Invasive clinical studies have found that statins alter both the lipid and calcium composition of plaque but the molecular mechanisms of statin-mediated effects on plaque calcium composition remain unclear. We recently defined a macrophage Rac (Ras-related C3 botulinum toxin substrate)-IL-1ß (interleukin-1 beta) signaling axis to be a key mechanism in promoting atherosclerotic calcification and sought to define the impact of statin therapy on this pathway. Approach and Results: Here, we demonstrate that statin therapy is independently associated with elevated coronary calcification in a high-risk patient population and that statins disrupt the complex between Rac1 and its inhibitor RhoGDI (Rho GDP-dissociation inhibitor), leading to increased active (GTP bound) Rac1 in primary monocytes/macrophages. Rac1 activation is prevented by rescue with the isoprenyl precursor geranylgeranyl diphosphate. Statin-treated macrophages exhibit increased activation of NF-κB (nuclear factor kappa-light-chain-enhancer of activated B cells), increased IL-1ß mRNA, and increased Rac1-dependent IL-1ß protein secretion in response to inflammasome stimulation. Using an animal model of calcific atherosclerosis, inclusion of statin in the atherogenic diet led to a myeloid Rac1-dependent increase in atherosclerotic calcification, which was associated with increased serum IL-1ß expression, increased plaque Rac1 activation, and increased plaque expression of the osteogenic markers, alkaline phosphatase and RUNX2 (Runt-related transcription factor 2). CONCLUSIONS: Statins are capable of increasing atherosclerotic calcification through disinhibition of a macrophage Rac1-IL-1ß signaling axis.


Asunto(s)
Aterosclerosis/tratamiento farmacológico , Atorvastatina/uso terapéutico , Inhibidores de Hidroximetilglutaril-CoA Reductasas/uso terapéutico , Macrófagos/efectos de los fármacos , Neuropéptidos/metabolismo , Placa Aterosclerótica , Calcificación Vascular/enzimología , Proteína de Unión al GTP rac1/metabolismo , Anciano , Animales , Aterosclerosis/enzimología , Aterosclerosis/genética , Aterosclerosis/patología , Células Cultivadas , Modelos Animales de Enfermedad , Humanos , Interleucina-1beta/genética , Interleucina-1beta/metabolismo , Macrófagos/enzimología , Macrófagos/patología , Masculino , Ratones Noqueados para ApoE , Neuropéptidos/deficiencia , Neuropéptidos/genética , Prenilación , Receptores de Factor Estimulante de Colonias de Granulocitos y Macrófagos/genética , Receptores de Factor Estimulante de Colonias de Granulocitos y Macrófagos/metabolismo , Estudios Retrospectivos , Transducción de Señal , Calcificación Vascular/genética , Calcificación Vascular/patología , Proteína de Unión al GTP rac1/deficiencia , Proteína de Unión al GTP rac1/genética , Inhibidor alfa de Disociación del Nucleótido Guanina rho/metabolismo
10.
Biochem Biophys Res Commun ; 521(2): 521-526, 2020 01 08.
Artículo en Inglés | MEDLINE | ID: mdl-31677791

RESUMEN

Neuromedin U (NMU) has a precursor that contains one additional peptide consisting of 33 or 36 amino acid residues. Recently, we identified this second peptide from rat brain and designated it neuromedin U precursor-related peptide (NURP), showing it to stimulate prolactin release from the pituitary when injected via the intracerebroventricular (icv) route. Here, we examined whether NMU, like NURP, also stimulates prolactin release. Unlike NURP, icv injection of NMU significantly decreased the secretion of prolactin from the pituitary. This suppression of prolactin release by NMU was observed in hyper-prolactin states such as lactation, stress, pseudopregnancy, domperidone (dopamine antagonist) administration, and icv injection of NURP. Immunohistochemical analysis revealed that icv injection of NMU induced cFos expression in dopaminergic neurons of the arcuate nucleus, but not the substantia nigra. Mice with double knockout of NMU and neuromedin S (NMS), the latter also binding to NMU receptors, showed a significant increase of the plasma prolactin level after domperidone treatment relative to wild-type mice. These results suggest that NMU and NURP may play important reciprocal roles in physiological prolactin secretion.


Asunto(s)
Núcleo Arqueado del Hipotálamo/citología , Neuronas Dopaminérgicas/metabolismo , Neuropéptidos/fisiología , Prolactina/metabolismo , Animales , Ratones , Neuropéptidos/deficiencia , Neuropéptidos/genética , Hipófisis/metabolismo , Proteínas Proto-Oncogénicas c-fos/metabolismo , Ratas , Receptores de Neurotransmisores
11.
Sci Rep ; 9(1): 19697, 2019 12 23.
Artículo en Inglés | MEDLINE | ID: mdl-31873146

RESUMEN

Excitatory neurons of the mammalian cerebral cortex are organized into six functional layers characterized by unique patterns of connectivity, as well as distinctive physiological and morphological properties. Cortical layers appear after a highly regulated migration process in which cells move from the deeper, proliferative zone toward the superficial layers. Importantly, defects in this radial migration process have been implicated in neurodevelopmental and psychiatric diseases. Here we report that during the final stages of migration, transcription factor Neurogenic Differentiation 2 (Neurod2) contributes to terminal cellular localization within the cortical plate. In mice, in utero knockdown of Neurod2 resulted in reduced numbers of neurons localized to the uppermost region of the developing cortex, also termed the primitive cortical zone. Our ChIP-Seq and RNA-Seq analyses of genes regulated by NEUROD2 in the developing cortex identified a number of key target genes with known roles in Reelin signaling, a critical regulator of neuronal migration. Our focused analysis of regulation of the Reln gene, encoding the extracellular ligand REELIN, uncovered NEUROD2 binding to conserved E-box elements in multiple introns. Furthermore, we demonstrate that knockdown of NEUROD2 in primary cortical neurons resulted in a strong increase in Reln gene expression at the mRNA level, as well as a slight upregulation at the protein level. These data reveal a new role for NEUROD2 during the late stages of neuronal migration, and our analysis of its genomic targets offers new genes with potential roles in cortical lamination.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Moléculas de Adhesión Celular Neuronal/genética , Corteza Cerebral/metabolismo , Proteínas de la Matriz Extracelular/genética , Proteínas del Tejido Nervioso/genética , Neuropéptidos/metabolismo , Serina Endopeptidasas/genética , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/deficiencia , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Sitios de Unión/genética , Diferenciación Celular , Movimiento Celular , Células Cultivadas , Corteza Cerebral/citología , Corteza Cerebral/crecimiento & desarrollo , Dendritas/metabolismo , Femenino , Regulación del Desarrollo de la Expresión Génica , Técnicas de Silenciamiento del Gen , Ratones , Neuronas/citología , Neuronas/metabolismo , Neuropéptidos/deficiencia , Neuropéptidos/genética , Embarazo , ARN Mensajero/genética , ARN Mensajero/metabolismo , ARN Interferente Pequeño/genética , RNA-Seq , Proteína Reelina
12.
Elife ; 82019 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-31613218

RESUMEN

In Drosophila, ~150 neurons expressing molecular clock proteins regulate circadian behavior. Sixteen of these neurons secrete the neuropeptide Pdf and have been called 'master pacemakers' because they are essential for circadian rhythms. A subset of Pdf+ neurons (the morning oscillator) regulates morning activity and communicates with other non-Pdf+ neurons, including a subset called the evening oscillator. It has been assumed that the molecular clock in Pdf+ neurons is required for these functions. To test this, we developed and validated Gal4-UAS based CRISPR tools for cell-specific disruption of key molecular clock components, period and timeless. While loss of the molecular clock in both the morning and evening oscillators eliminates circadian locomotor activity, the molecular clock in either oscillator alone is sufficient to rescue circadian locomotor activity in the absence of the other. This suggests that clock neurons do not act in a hierarchy but as a distributed network to regulate circadian activity.


Asunto(s)
Relojes Circadianos/genética , Ritmo Circadiano/genética , Proteínas de Drosophila/genética , Drosophila melanogaster/genética , Neuronas/metabolismo , Neuropéptidos/genética , Proteínas Circadianas Period/genética , Animales , Encéfalo/citología , Encéfalo/metabolismo , Encéfalo/efectos de la radiación , Sistemas CRISPR-Cas , Comunicación Celular , Linaje de la Célula/genética , Relojes Circadianos/efectos de los fármacos , Ritmo Circadiano/efectos de los fármacos , Oscuridad , Proteínas de Drosophila/deficiencia , Drosophila melanogaster/metabolismo , Drosophila melanogaster/efectos de la radiación , Retroalimentación Fisiológica , Edición Génica , Regulación de la Expresión Génica , Fototransducción/genética , Locomoción/genética , Locomoción/efectos de la radiación , Red Nerviosa/metabolismo , Red Nerviosa/efectos de la radiación , Neuronas/citología , Neuronas/efectos de la radiación , Neuropéptidos/deficiencia , Proteínas Circadianas Period/deficiencia , Factores de Transcripción/deficiencia , Factores de Transcripción/genética
13.
Elife ; 82019 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-31613223

RESUMEN

Animal circadian rhythms persist in constant darkness and are driven by intracellular transcription-translation feedback loops. Although these cellular oscillators communicate, isolated mammalian cellular clocks continue to tick away in darkness without intercellular communication. To investigate these issues in Drosophila, we assayed behavior as well as molecular rhythms within individual brain clock neurons while blocking communication within the ca. 150 neuron clock network. We also generated CRISPR-mediated neuron-specific circadian clock knockouts. The results point to two key clock neuron groups: loss of the clock within both regions but neither one alone has a strong behavioral phenotype in darkness; communication between these regions also contributes to circadian period determination. Under these dark conditions, the clock within one region persists without network communication. The clock within the famous PDF-expressing s-LNv neurons however was strongly dependent on network communication, likely because clock gene expression within these vulnerable sLNvs depends on neuronal firing or light.


Asunto(s)
Encéfalo/metabolismo , Relojes Circadianos/genética , Ritmo Circadiano/genética , Drosophila melanogaster/genética , Regulación de la Expresión Génica , Fototransducción/genética , Neuronas/metabolismo , Animales , Factores de Transcripción con Cremalleras de Leucina de Carácter Básico/deficiencia , Factores de Transcripción con Cremalleras de Leucina de Carácter Básico/genética , Encéfalo/citología , Encéfalo/efectos de la radiación , Sistemas CRISPR-Cas , Comunicación Celular , Linaje de la Célula/genética , Relojes Circadianos/efectos de los fármacos , Ritmo Circadiano/efectos de los fármacos , Oscuridad , Proteínas de Drosophila/deficiencia , Proteínas de Drosophila/genética , Drosophila melanogaster/metabolismo , Drosophila melanogaster/efectos de la radiación , Retroalimentación Fisiológica , Edición Génica , Red Nerviosa/metabolismo , Red Nerviosa/efectos de la radiación , Neuronas/citología , Neuronas/efectos de la radiación , Neuropéptidos/deficiencia , Neuropéptidos/genética , Proteínas Circadianas Period/deficiencia , Proteínas Circadianas Period/genética , Factores de Transcripción/deficiencia , Factores de Transcripción/genética
14.
Elife ; 82019 09 16.
Artículo en Inglés | MEDLINE | ID: mdl-31524598

RESUMEN

Neurexophilins are secreted neuropeptide-like glycoproteins, and neurexophilin1 and neurexophilin3 are ligands for the presynaptic cell adhesion molecule α-neurexin. Neurexophilins are more selectively expressed in the brain than α-neurexins, however, which led us to ask whether neurexophilins modulate the function of α-neurexin in a context-specific manner. We characterized the expression and function of neurexophilin4 in mice and found it to be expressed in subsets of neurons responsible for feeding, emotion, balance, and movement. Deletion of Neurexophilin4 caused corresponding impairments, most notably in motor learning and coordination. We demonstrated that neurexophilin4 interacts with α-neurexin and GABAARs in the cerebellum. Loss of Neurexophilin4 impaired cerebellar Golgi-granule inhibitory neurotransmission and synapse number, providing a partial explanation for the motor learning and coordination deficits observed in the Neurexophilin4 null mice. Our data illustrate how selectively expressed Neurexophilin4, an α-neurexin ligand, regulates specific synapse function and modulates cerebellar motor control.


Asunto(s)
Cerebelo/fisiología , Glicoproteínas/metabolismo , Actividad Motora , Neuronas Motoras/fisiología , Neuropéptidos/metabolismo , Animales , Eliminación de Gen , Regulación de la Expresión Génica , Glicoproteínas/deficiencia , Ratones Endogámicos C57BL , Ratones Noqueados , Neuropéptidos/deficiencia
15.
Peptides ; 121: 170131, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-31408662

RESUMEN

Extensive proliferation of vascular smooth muscle cell (VSMC) contributes to intimal hyperplasia following vascular injury, in which endoplasmic reticulum stress (ERS) plays a critical role. Intermedin (IMD) is a vascular paracrine/autocrine peptide exerting numerous beneficial effects in cardiovascular diseases. IMD overexpression could alleviate intimal hyperplasia. Here, we investigated whether endogenous IMD protects against intimal hyperplasia by inhibiting endoplasmic reticulum stress. The mouse left common carotid-artery ligation-injury model was established to induce intimal hyperplasia using IMD-/-mice and C57BL/6 J wild-type (WT) mice. Platelet-derived growth factor-BB (PDGF-BB) was used to stimulate the proliferation of VSMC. IMD-/- mice displayed exacerbated intimal hyperplasia induced by complete ligation of the left carotid artery at 14 d and 28 d compared to WT mice. However, IMD-deficiency had no effect on blood pressure, plasma triglyceride, and fasting blood glucose levels in mice. Furthermore, VSMCs derived from IMD-/- mice showed increased cell proliferation and dramatically elevated levels of glucose regulated protein 78 (GRP78), activating transcription factor 4 (ATF4), ATF6 mRNA under PDGF-BB treatment compared to WT mice-derived VSMCs. In addition, exogenous administration of IMD significantly attenuated PDGF-BB-induced cell proliferation and GRP78, phosphorylase-inositol requiring enzyme 1α, ATF4, and ATF6 protein levels. Thus, endogenous IMD may counteract ERS to exert protective role in response to vascular injury and IMD is expected to be a therapeutic target for the prevention and treatment of restenosis.


Asunto(s)
Estrés del Retículo Endoplásmico/genética , Hiperplasia/genética , Miocitos del Músculo Liso/metabolismo , Neuropéptidos/genética , Túnica Íntima/metabolismo , Factor de Transcripción Activador 4 , Factor de Transcripción Activador 6/genética , Factor de Transcripción Activador 6/metabolismo , Animales , Becaplermina/farmacología , Arterias Carótidas/cirugía , Proliferación Celular/efectos de los fármacos , Modelos Animales de Enfermedad , Chaperón BiP del Retículo Endoplásmico , Estrés del Retículo Endoplásmico/efectos de los fármacos , Regulación de la Expresión Génica , Proteínas de Choque Térmico , Hiperplasia/metabolismo , Hiperplasia/patología , Hiperplasia/prevención & control , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Músculo Liso Vascular/citología , Músculo Liso Vascular/metabolismo , Miocitos del Músculo Liso/citología , Miocitos del Músculo Liso/efectos de los fármacos , Neuropéptidos/deficiencia , Cultivo Primario de Células , Transducción de Señal , Túnica Íntima/patología
16.
Sci Rep ; 9(1): 7675, 2019 05 22.
Artículo en Inglés | MEDLINE | ID: mdl-31118438

RESUMEN

Membrane ruffling and lamellipodia formation promote the motility of adherent cells in two-dimensional motility assays by mechano-sensing of the microenvironment and initiation of focal adhesions towards their surroundings. Lamellipodium formation is stimulated by small Rho GTPases of the Rac subfamily, since genetic removal of these GTPases abolishes lamellipodium assembly. The relevance of lamellipodial or invadopodial structures for facilitating cellular mechanics and 3D cell motility is still unclear. Here, we hypothesized that Rac1 affects cell mechanics and facilitates 3D invasion. Thus, we explored whether fibroblasts that are genetically deficient for Rac1 (lacking Rac2 and Rac3) harbor altered mechanical properties, such as cellular deformability, intercellular adhesion forces and force exertion, and exhibit alterations in 3D motility. Rac1 knockout and control cells were analyzed for changes in deformability by applying an external force using an optical stretcher. Five Rac1 knockout cell lines were pronouncedly more deformable than Rac1 control cells upon stress application. Using AFM, we found that cell-cell adhesion forces are increased in Rac1 knockout compared to Rac1-expressing fibroblasts. Since mechanical deformability, cell-cell adhesion strength and 3D motility may be functionally connected, we investigated whether increased deformability of Rac1 knockout cells correlates with changes in 3D motility. All five Rac1 knockout clones displayed much lower 3D motility than Rac1-expressing controls. Moreover, force exertion was reduced in Rac1 knockout cells, as assessed by 3D fiber displacement analysis. Interference with cellular stiffness through blocking of actin polymerization by Latrunculin A could not further reduce invasion of Rac1 knockout cells. In contrast, Rac1-expressing controls treated with Latrunculin A were again more deformable and less invasive, suggesting actin polymerization is a major determinant of observed Rac1-dependent effects. Together, we propose that regulation of 3D motility by Rac1 partly involves cellular mechanics such as deformability and exertion of forces.


Asunto(s)
Fibroblastos/enzimología , Neuropéptidos/fisiología , Proteína de Unión al GTP rac1/fisiología , Citoesqueleto de Actina/fisiología , Animales , Biopolímeros , Adhesión Celular , Línea Celular , Movimiento Celular/efectos de los fármacos , Movimiento Celular/fisiología , Colágeno , Elasticidad , Matriz Extracelular , Fibroblastos/fisiología , Fibroblastos/ultraestructura , Técnicas de Inactivación de Genes , Ratones , Microscopía de Fuerza Atómica , Microscopía Confocal , Neuropéptidos/antagonistas & inhibidores , Neuropéptidos/deficiencia , Neuropéptidos/genética , Seudópodos/fisiología , Pironas/farmacología , Quinolinas/farmacología , Reología , Propiedades de Superficie , Proteína de Unión al GTP rac1/antagonistas & inhibidores , Proteína de Unión al GTP rac1/deficiencia , Proteína de Unión al GTP rac1/genética
17.
Neurosci Bull ; 35(4): 673-687, 2019 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-30888607

RESUMEN

Ras-related C3 botulinum toxin substrate 1 (Rac1), a member of the Rho GTPase family which plays important roles in dendritic spine morphology and plasticity, is a key regulator of cytoskeletal reorganization in dendrites and spines. Here, we investigated whether and how Rac1 modulates synaptic transmission in mouse retinal ganglion cells (RGCs) using selective conditional knockout of Rac1 (Rac1-cKO). Rac1-cKO significantly reduced the frequency of AMPA receptor-mediated miniature excitatory postsynaptic currents, while glycine/GABAA receptor-mediated miniature inhibitory postsynaptic currents were not affected. Although the total GluA1 protein level was increased in Rac1-cKO mice, its expression in the membrane component was unchanged. Rac1-cKO did not affect spine-like branch density in single dendrites, but significantly reduced the dendritic complexity, which resulted in a decrease in the total number of dendritic spine-like branches. These results suggest that Rac1 selectively affects excitatory synaptic transmission in RGCs by modulating dendritic complexity.


Asunto(s)
Dendritas/metabolismo , Neuropéptidos/metabolismo , Células Ganglionares de la Retina/metabolismo , Células Ganglionares de la Retina/fisiología , Transmisión Sináptica/genética , Proteína de Unión al GTP rac1/metabolismo , Animales , Dendritas/ultraestructura , Espinas Dendríticas/metabolismo , Potenciales Postsinápticos Excitadores/fisiología , Antagonistas de Receptores de GABA-A , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas del Tejido Nervioso/metabolismo , Neuropéptidos/deficiencia , Receptores AMPA/metabolismo , Receptores de GABA-A/metabolismo , Receptores de Glicina/antagonistas & inhibidores , Receptores de N-Metil-D-Aspartato/metabolismo , Transducción de Señal , Sinapsis/metabolismo , Proteína de Unión al GTP rac1/deficiencia
18.
Brain Res ; 1711: 146-155, 2019 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-30689978

RESUMEN

Subcortical band heterotopia (SBH), also known as doublecortex syndrome, is a malformation of cortical development resulting from mutations in the doublecortin gene (DCX). It is characterized by a lack of migration of cortical neurons that accumulate in the white matter forming a heterotopic band. Patients with SBH may present mild to moderate intellectual disability as well as epilepsy. The SBH condition can be modeled in rats by in utero knockdown (KD) of Dcx. The affected cells form an SBH reminiscent of that observed in human patients and the animals develop a chronic epileptic condition in adulthood. Here, we investigated if the presence of a SBH is sufficient to induce cognitive impairment in juvenile Dcx-KD rats, before the onset of epilepsy. Using a wide range of behavioral tests, we found that the presence of SBH did not appear to affect motor control or somatosensory processing. In addition, cognitive abilities such as learning, short-term and long-term memory, were normal in pre-epileptic Dcx-KD rats. We suggest that the SBH presence is not sufficient to impair these behavioral functions.


Asunto(s)
Conducta Animal , Lisencefalias Clásicas y Heterotopias Subcorticales en Banda/psicología , Cognición , Modelos Animales de Enfermedad , Epilepsia/genética , Discapacidad Intelectual/genética , Animales , Ansiedad/genética , Enfermedades Asintomáticas , Movimiento Celular , Lisencefalias Clásicas y Heterotopias Subcorticales en Banda/complicaciones , Lisencefalias Clásicas y Heterotopias Subcorticales en Banda/embriología , Lisencefalias Clásicas y Heterotopias Subcorticales en Banda/genética , Proteínas de Dominio Doblecortina , Proteína Doblecortina , Electroporación , Conducta Exploratoria , Sustancia Gris/anomalías , Sustancia Gris/embriología , Aprendizaje , Aprendizaje por Laberinto , Memoria , Proteínas Asociadas a Microtúbulos/deficiencia , Proteínas Asociadas a Microtúbulos/genética , Mosaicismo , Neuropéptidos/deficiencia , Neuropéptidos/genética , ARN Interferente Pequeño/administración & dosificación , ARN Interferente Pequeño/toxicidad , Ratas , Prueba de Desempeño de Rotación con Aceleración Constante , Sensación , Sustancia Blanca/anomalías , Sustancia Blanca/embriología
19.
Hippocampus ; 29(7): 569-578, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-30387892

RESUMEN

The hippocampus is generally considered as a brain center for learning and memory. We have recently established an electroporation-mediated gene transfer method to investigate the development of neonatal dentate granule cells in vivo. Using this new technique, we introduced knockdown vectors against Rac1 small GTPase into precursors for dentate granule cells at postnatal day 0. After 21 days, Rac1-deficient cells were frequently mispositioned between the granule cell layer (GCL) and hilus. About 60% of these mislocalized cells expressed a dentate granule cell marker, Prox1. Both the dendritic spine density and the ratio of mature spine were reduced when Rac1 was silenced. Notably, the deficient cells have immature thin processes during migrating in the early neonatal period. Knockdown of another Rac isoform, Rac3, also resulted in mislocalization of neonatally born dentate granule cells. In addition, knockdown of Cdc42, another Rho family protein, also caused mislocalization of the cell, although the effects were moderate compared to Rac1 and 3. Despite the ectopic localization, Rac3- or Cdc42-disrupted mispositioned cells expressed Prox1. These results indicate that Rho signaling pathways differentially regulate the proper localization and differentiation of dentate granule cells.


Asunto(s)
Giro Dentado/enzimología , Giro Dentado/crecimiento & desarrollo , Neuropéptidos/metabolismo , Proteína de Unión al GTP cdc42/metabolismo , Proteínas de Unión al GTP rac/metabolismo , Proteína de Unión al GTP rac1/metabolismo , Animales , Animales Recién Nacidos , Diferenciación Celular , Movimiento Celular , Giro Dentado/citología , Técnicas de Silenciamiento del Gen , Técnicas de Transferencia de Gen , Proteínas de Homeodominio/metabolismo , Ratones , Ratones Endogámicos ICR , Neurogénesis , Neuropéptidos/deficiencia , Neuropéptidos/genética , Interferencia de ARN , Transducción de Señal , Proteínas Supresoras de Tumor/metabolismo , Proteína de Unión al GTP cdc42/deficiencia , Proteína de Unión al GTP cdc42/genética , Proteínas de Unión al GTP rac/deficiencia , Proteínas de Unión al GTP rac/genética , Proteína de Unión al GTP rac1/deficiencia , Proteína de Unión al GTP rac1/genética
20.
J Cell Biol ; 218(1): 350-379, 2019 01 07.
Artículo en Inglés | MEDLINE | ID: mdl-30523041

RESUMEN

Growth cones are complex, motile structures at the tip of an outgrowing neurite. They often exhibit a high density of filopodia (thin actin bundles), which complicates the unbiased quantification of their morphologies by software. Contemporary image processing methods require extensive tuning of segmentation parameters, require significant manual curation, and are often not sufficiently adaptable to capture morphology changes associated with switches in regulatory signals. To overcome these limitations, we developed Growth Cone Analyzer (GCA). GCA is designed to quantify growth cone morphodynamics from time-lapse sequences imaged both in vitro and in vivo, but is sufficiently generic that it may be applied to nonneuronal cellular structures. We demonstrate the adaptability of GCA through the analysis of growth cone morphological variation and its relation to motility in both an unperturbed system and in the context of modified Rho GTPase signaling. We find that perturbations inducing similar changes in neurite length exhibit underappreciated phenotypic nuance at the scale of the growth cone.


Asunto(s)
Conos de Crecimiento/ultraestructura , Procesamiento de Imagen Asistido por Computador/estadística & datos numéricos , Imagen Molecular/normas , Neuronas/ultraestructura , Programas Informáticos , Imagen de Lapso de Tiempo/normas , Proteínas de Unión al GTP rho/genética , Animales , Línea Celular Tumoral , Movimiento Celular , Forma de la Célula/genética , Regulación de la Expresión Génica , Heterogeneidad Genética , Conos de Crecimiento/metabolismo , Factores de Intercambio de Guanina Nucleótido/deficiencia , Factores de Intercambio de Guanina Nucleótido/genética , Ratones , Imagen Molecular/métodos , Neuronas/metabolismo , Neuropéptidos/deficiencia , Neuropéptidos/genética , Fosfoproteínas/deficiencia , Fosfoproteínas/genética , Proteínas Serina-Treonina Quinasas/deficiencia , Proteínas Serina-Treonina Quinasas/genética , Seudópodos/metabolismo , Seudópodos/ultraestructura , Factores de Intercambio de Guanina Nucleótido Rho/deficiencia , Factores de Intercambio de Guanina Nucleótido Rho/genética , Transducción de Señal , Imagen de Lapso de Tiempo/métodos , Proteína de Unión al GTP cdc42/deficiencia , Proteína de Unión al GTP cdc42/genética , Proteína de Unión al GTP rac1/deficiencia , Proteína de Unión al GTP rac1/genética , Proteínas de Unión al GTP rho/deficiencia , Proteína de Unión al GTP rhoA
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA