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

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Mol Psychiatry ; 26(4): 1376-1398, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-31444474

RESUMEN

Aberrant fear is a cornerstone of several psychiatric disorders. Consequently, there is large interest in elucidation of signaling mechanisms that link extracellular cues to changes in neuronal function and structure in brain pathways that are important in the generation and maintenance of fear memory and its behavioral expression. Members of the Plexin-B family of receptors for class 4 semaphorins play important roles in developmental plasticity of neurons, and their expression persists in some areas of the adult nervous system. Here, we aimed to elucidate the role of Semaphorin 4C (Sema4C) and its cognate receptor, Plexin-B2, in the expression of contextual and cued fear memory, setting a mechanistic focus on structural plasticity and exploration of contributing signaling pathways. We observed that Plexin-B2 and Sema4C are expressed in forebrain areas related to fear memory, such as the anterior cingulate cortex, amygdala and the hippocampus, and their expression is regulated by aversive stimuli that induce fear memory. By generating forebrain-specific Plexin-B2 knockout mice and analyzing fear-related behaviors, we demonstrate that Sema4C-PlexinB2 signaling plays a crucial functional role in the recent and remote recall of fear memory. Detailed neuronal morphological analyses revealed that Sema4C-PlexinB2 signaling largely mediates fear-induced structural plasticity by enhancing dendritic ramifications and modulating synaptic density in the adult hippocampus. Analyses on signaling-related mutant mice showed that these functions are mediated by PlexinB2-dependent RhoA activation. These results deliver important insights into the mechanistic understanding of maladaptive plasticity in fear circuits and have implications for novel therapeutic strategies against fear-related disorders.


Asunto(s)
Miedo , Memoria , Proteínas del Tejido Nervioso , Semaforinas , Animales , Moléculas de Adhesión Celular , Ratones , Proteínas del Tejido Nervioso/genética , Neuronas , Semaforinas/genética
2.
Cell Mol Neurobiol ; 38(7): 1369-1382, 2018 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-29992390

RESUMEN

Cerebral cavernous malformations (CCM) are vascular malformations associated with abnormally dilated blood vessels and leaky capillaries that often result in hemorrhages. Despite recent advances, precise understanding of the cellular and molecular mechanism leading to the pathogenesis of CCM remains elusive. Emerging evidence indicates that small nucleolar RNAs (snoRNAs), belonging to the class of non-coding RNAs, may play a significant role as diagnostic markers in human diseases. However, there is no report till date that studied the role of snoRNAs in CCM biology. The objective of the current study was to identify snoRNAs associated with CCM pathogenesis. Using genome-wide small RNA sequencing, we identified a total of 271 snoRNAs reliably expressed in CCM. By applying additional statistical stringency, three snoRNAs (SNORD115-32, SNORD114-22, and SNORD113-3) were found to be significantly downregulated in CCM patient tissue samples (n = 3) as compared to healthy brains (n = 3). Deregulation of the selected snoRNAs was further validated by qRT-PCR. Further, cellular localization via in situ hybridization also confirmed robust reduction in the expression of SNORD115-32 and SNORD114-22 in CCM tissues as compared to the healthy controls. By applying high-throughput sequencing and cellular localization analyses, we report here for the first time the genome-wide expression profile of snoRNAs in CCM tissues and a robust downregulation of candidate snoRNAs in CCM conditions. Future studies should warrant the screening in large CCM patient cohorts and will be helpful in the development of potential biomarkers and improved clinical diagnosis.


Asunto(s)
Regulación hacia Abajo/genética , Genoma Humano , Hemangioma Cavernoso del Sistema Nervioso Central/genética , ARN Nucleolar Pequeño/genética , Análisis de Secuencia de ADN , Adulto , Animales , Análisis por Conglomerados , Femenino , Perfilación de la Expresión Génica , Hemangioma Cavernoso del Sistema Nervioso Central/patología , Humanos , Imagen por Resonancia Magnética , Masculino , Persona de Mediana Edad , ARN Nucleolar Pequeño/metabolismo , Reproducibilidad de los Resultados
3.
Mol Pain ; 122016.
Artículo en Inglés | MEDLINE | ID: mdl-27030713

RESUMEN

BACKGROUND: Stroke patients often suffer from a central neuropathic pain syndrome called central post-stroke pain. This syndrome is characterized by evoked pain hypersensitivity as well as spontaneous, on-going pain in the body area affected by the stroke. Clinical evidence strongly suggests a dysfunction in central pain pathways as an important pathophysiological factor in the development of central post-stroke pain, but the exact underlying mechanisms remain poorly understood. To elucidate the underlying pathophysiology of central post-stroke pain, we generated a mouse model that is based on a unilateral stereotactic lesion of the thalamic ventral posterolateral nucleus, which typically causes central post-stroke pain in humans. RESULTS: Behavioral analysis showed that the sensory changes in our model are comparable to the sensory abnormalities observed in patients suffering from central post-stroke pain. Surprisingly, pharmacological inhibition of spinal and peripheral key components of the pain system had no effect on the induction or maintenance of the evoked hypersensitivity observed in our model. In contrast, microinjection of lidocaine into the thalamic lesion completely reversed injury-induced hypersensitivity. CONCLUSIONS: These results suggest that the evoked hypersensitivity observed in central post-stroke pain is causally linked to on-going neuronal activity in the lateral thalamus.


Asunto(s)
Dolor/etiología , Dolor/fisiopatología , Accidente Cerebrovascular/complicaciones , Accidente Cerebrovascular/fisiopatología , Animales , Colagenasas/administración & dosificación , Modelos Animales de Enfermedad , Hiperalgesia/complicaciones , Hiperalgesia/patología , Hiperalgesia/fisiopatología , Ácido Kaínico/administración & dosificación , Lidocaína/administración & dosificación , Ratones Endogámicos C57BL , Microinyecciones , Fibras Nerviosas Amielínicas/patología , Sensación , Médula Espinal/patología , Médula Espinal/fisiopatología , Canales Catiónicos TRPV/metabolismo , Tálamo/patología , Tálamo/fisiopatología , Núcleos Talámicos Ventrales/patología , Núcleos Talámicos Ventrales/fisiopatología
4.
PLoS Biol ; 10(3): e1001283, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22427743

RESUMEN

Synaptic long-term potentiation (LTP) at spinal neurons directly communicating pain-specific inputs from the periphery to the brain has been proposed to serve as a trigger for pain hypersensitivity in pathological states. Previous studies have functionally implicated the NMDA receptor-NO pathway and the downstream second messenger, cGMP, in these processes. Because cGMP can broadly influence diverse ion-channels, kinases, and phosphodiesterases, pre- as well as post-synaptically, the precise identity of cGMP targets mediating spinal LTP, their mechanisms of action, and their locus in the spinal circuitry are still unclear. Here, we found that Protein Kinase G1 (PKG-I) localized presynaptically in nociceptor terminals plays an essential role in the expression of spinal LTP. Using the Cre-lox P system, we generated nociceptor-specific knockout mice lacking PKG-I specifically in presynaptic terminals of nociceptors in the spinal cord, but not in post-synaptic neurons or elsewhere (SNS-PKG-I(-/-) mice). Patch clamp recordings showed that activity-induced LTP at identified synapses between nociceptors and spinal neurons projecting to the periaqueductal grey (PAG) was completely abolished in SNS-PKG-I(-/-) mice, although basal synaptic transmission was not affected. Analyses of synaptic failure rates and paired-pulse ratios indicated a role for presynaptic PKG-I in regulating the probability of neurotransmitter release. Inositol 1,4,5-triphosphate receptor 1 and myosin light chain kinase were recruited as key phosphorylation targets of presynaptic PKG-I in nociceptive neurons. Finally, behavioural analyses in vivo showed marked defects in SNS-PKG-I(-/-) mice in several models of activity-induced nociceptive hypersensitivity, and pharmacological studies identified a clear contribution of PKG-I expressed in spinal terminals of nociceptors. Our results thus indicate that presynaptic mechanisms involving an increase in release probability from nociceptors are operational in the expression of synaptic LTP on spinal-PAG projection neurons and that PKG-I localized in presynaptic nociceptor terminals plays an essential role in this process to regulate pain sensitivity.


Asunto(s)
Proteínas Quinasas Dependientes de GMP Cíclico/metabolismo , Potenciación a Largo Plazo , Nociceptores/metabolismo , Dolor/patología , Aminoquinolinas/farmacología , Animales , Conducta Animal , Moléculas de Adhesión Celular/genética , Moléculas de Adhesión Celular/metabolismo , Proteína Quinasa Dependiente de GMP Cíclico Tipo I , Proteínas Quinasas Dependientes de GMP Cíclico/genética , Activación Enzimática , Ganglios Espinales/metabolismo , Ganglios Espinales/patología , Eliminación de Gen , Guanilato Ciclasa/antagonistas & inhibidores , Guanilato Ciclasa/metabolismo , Receptores de Inositol 1,4,5-Trifosfato/genética , Receptores de Inositol 1,4,5-Trifosfato/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas de Microfilamentos/genética , Proteínas de Microfilamentos/metabolismo , Fibras Nerviosas/metabolismo , Fibras Nerviosas/patología , Nociceptores/efectos de los fármacos , Nociceptores/patología , Dolor/metabolismo , Técnicas de Placa-Clamp , Fosfoproteínas/genética , Fosfoproteínas/metabolismo , Fosforilación , Transducción de Señal , Especificidad por Sustrato , Transmisión Sináptica
5.
Mol Pain ; 10: 22, 2014 Mar 19.
Artículo en Inglés | MEDLINE | ID: mdl-24642266

RESUMEN

BACKGROUND: We carried out a genome-wide study, using microRNA sequencing (miRNA-seq), aimed at identifying miRNAs in primary sensory neurons that are associated with neuropathic pain. Such scans usually yield long lists of transcripts regulated by nerve injury, but not necessarily related to pain. To overcome this we tried a novel search strategy: identification of transcripts regulated differentially by nerve injury in rat lines very similar except for a contrasting pain phenotype. Dorsal root ganglia (DRGs) L4 and 5 in the two lines were excised 3 days after spinal nerve ligation surgery (SNL) and small RNAs were extracted and sequenced. RESULTS: We identified 284 mature miRNA species expressed in rat DRGs, including several not previously reported, and 3340 unique small RNA sequences. Baseline expression of miRNA was nearly identical in the two rat lines, consistent with their shared genetic background. In both lines many miRNAs were nominally up- or down-regulated following SNL, but the change was similar across lines. Only 3 miRNAs that were expressed abundantly (rno-miR-30d-5p, rno-miR-125b-5p) or at moderate levels (rno-miR-379-5p) were differentially regulated. This makes them prime candidates as novel PNS determinants of neuropathic pain. The first two are known miRNA regulators of the expression of Tnf, Bdnf and Stat3, gene products intimately associated with neuropathic pain phenotype. A few non-miRNA, small noncoding RNAs (sncRNAs) were also differentially regulated. CONCLUSIONS: Despite its genome-wide coverage, our search strategy yielded a remarkably short list of neuropathic pain-related miRNAs. As 2 of the 3 are validated regulators of important pro-nociceptive compounds, it is likely that they contribute to the orchestration of gene expression changes that determine individual variability in pain phenotype. Further research is required to determine whether some of the other known or predicted gene targets of these miRNAs, or of the differentially regulated non-miRNA sncRNAs, also contribute.


Asunto(s)
Regulación de la Expresión Génica/genética , Predisposición Genética a la Enfermedad/genética , MicroARNs/genética , Neuralgia/genética , Animales , Secuencia de Bases , Mapeo Cromosómico , Biología Computacional , Modelos Animales de Enfermedad , Femenino , Genoma/genética , Masculino , MicroARNs/metabolismo , Neuralgia/metabolismo , Ratas
6.
Mol Pain ; 9: 48, 2013 Sep 25.
Artículo en Inglés | MEDLINE | ID: mdl-24067145

RESUMEN

BACKGROUND: Cancer-associated pain is a major cause of poor quality of life in cancer patients and is frequently resistant to conventional therapy. Recent studies indicate that some hematopoietic growth factors, namely granulocyte macrophage colony stimulating factor (GMCSF) and granulocyte colony stimulating factor (GCSF), are abundantly released in the tumor microenvironment and play a key role in regulating tumor-nerve interactions and tumor-associated pain by activating receptors on dorsal root ganglion (DRG) neurons. Moreover, these hematopoietic factors have been highly implicated in postsurgical pain, inflammatory pain and osteoarthritic pain. However, the molecular mechanisms via which G-/GMCSF bring about nociceptive sensitization and elicit pain are not known. RESULTS: In order to elucidate G-/GMCSF mediated transcriptional changes in the sensory neurons, we performed a comprehensive, genome-wide analysis of changes in the transcriptome of DRG neurons brought about by exposure to GMCSF or GCSF. We present complete information on regulated genes and validated profiling analyses and report novel regulatory networks and interaction maps revealed by detailed bioinformatics analyses. Amongst these, we validate calpain 2, matrix metalloproteinase 9 (MMP9) and a RhoGTPase Rac1 as well as Tumor necrosis factor alpha (TNFα) as transcriptional targets of G-/GMCSF and demonstrate the importance of MMP9 and Rac1 in GMCSF-induced nociceptor sensitization. CONCLUSION: With integrative approach of bioinformatics, in vivo pharmacology and behavioral analyses, our results not only indicate that transcriptional control by G-/GMCSF signaling regulates a variety of established pain modulators, but also uncover a large number of novel targets, paving the way for translational analyses in the context of pain disorders.


Asunto(s)
Ganglios Espinales/efectos de los fármacos , Factor Estimulante de Colonias de Granulocitos y Macrófagos/farmacología , Factor Estimulante de Colonias de Macrófagos/farmacología , Células Receptoras Sensoriales/efectos de los fármacos , Animales , Ratones , Ratones Endogámicos C57BL , Transducción de Señal/efectos de los fármacos
7.
Pain ; 162(5): 1334-1351, 2021 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-33492037

RESUMEN

ABSTRACT: Diabetes is a leading cause of peripheral neuropathy (diabetic peripheral neuropathy, DPN), and uncontrolled long-lasting hyperglycemia leads to severe complications. A major proportion of diabetics develop excruciating pain with a variable course. Mechanisms leading to painful DPN are not completely understood and treatment options limited. We hypothesized that epigenetic modulation at the level of microRNA (miRNA) expression triggered by metabolic imbalance and nerve damage regulates the course of pain development. We used clinically relevant preclinical models, genome-wide screening, in silico analyses, cellular assays, miRNA fluorescent in situ hybridization, in vivo molecular manipulations, and behavioral analyses in the current study. We identified miRNAs and their targets that critically impact on nociceptive hypersensitivity in painful DPN. Our analyses identify miR-33 and miR-380 expressed in nociceptive neurons as critical denominators of diabetic pain and miR-124-1 as a mediator of physiological nociception. Our comprehensive analyses on the putative mRNA targets for miR-33 or miR-124-1 identified a set of mRNAs that are regulated after miR-33 or miR-124-1 overexpression in dorsal root ganglia in vivo. Our results shed light on the regulation of DPN pathophysiology and implicate specific miRNAs as novel therapeutic targets for treating painful DPN.


Asunto(s)
Diabetes Mellitus , Neuropatías Diabéticas , MicroARNs , Neuropatías Diabéticas/genética , Ganglios Espinales , Humanos , Hibridación Fluorescente in Situ , MicroARNs/genética , Células Receptoras Sensoriales
8.
Sci Rep ; 9(1): 18203, 2019 12 03.
Artículo en Inglés | MEDLINE | ID: mdl-31796831

RESUMEN

Cerebral cavernous malformations (CCMs) are low-flow vascular malformations in the brain associated with recurrent hemorrhage and seizures. The current treatment of CCMs relies solely on surgical intervention. Henceforth, alternative non-invasive therapies are urgently needed to help prevent subsequent hemorrhagic episodes. Long non-coding RNAs (lncRNAs) belong to the class of non-coding RNAs and are known to regulate gene transcription and involved in chromatin remodeling via various mechanism. Despite accumulating evidence demonstrating the role of lncRNAs in cerebrovascular disorders, their identification in CCMs pathology remains unknown. The objective of the current study was to identify lncRNAs associated with CCMs pathogenesis using patient cohorts having 10 CCM patients and 4 controls from brain. Executing next generation sequencing, we performed whole transcriptome sequencing (RNA-seq) analysis and identified 1,967 lncRNAs and 4,928 protein coding genes (PCGs) to be differentially expressed in CCMs patients. Among these, we selected top 6 differentially expressed lncRNAs each having significant correlative expression with more than 100 differentially expressed PCGs. The differential expression status of the top lncRNAs, SMIM25 and LBX2-AS1 in CCMs was further confirmed by qRT-PCR analysis. Additionally, gene set enrichment analysis of correlated PCGs revealed critical pathways related to vascular signaling and important biological processes relevant to CCMs pathophysiology. Here, by transcriptome-wide approach we demonstrate that lncRNAs are prevalent in CCMs disease and are likely to play critical roles in regulating important signaling pathways involved in the disease progression. We believe, that detailed future investigations on this set of identified lncRNAs can provide useful insights into the biology and, ultimately, contribute in preventing this debilitating disease.


Asunto(s)
Neoplasias del Sistema Nervioso Central/genética , Regulación Neoplásica de la Expresión Génica , Hemangioma Cavernoso del Sistema Nervioso Central/genética , ARN Largo no Codificante/metabolismo , Adulto , Niño , Femenino , Humanos , Masculino , Persona de Mediana Edad , RNA-Seq , Transcriptoma , Adulto Joven
9.
Pain ; 158(11): 2243-2258, 2017 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-28837503

RESUMEN

Neuropathic pain is an integral component of several chronic pain conditions and poses a major health problem worldwide. Despite emerging understanding of mechanisms behind neuropathic pain, the available treatment options are still limited in efficacy or associated with side effects, therefore making it necessary to find viable alternatives. In a genetic screen, we recently identified SerpinA3N, a serine protease inhibitor secreted in response to nerve damage by the dorsal root ganglion neurons and we showed that SerpinA3N acts against induction of neuropathic pain by inhibiting the T-cell- and neutrophil-derived protease, leucocyte elastase (LE). In the current study, via detailed in vivo pharmacology combined with analyses of evoked- and spontaneous pain-related behaviors in mice, we report that on systemic delivery, a single dose of 3 independent LE inhibitors can block established nociceptive hypersensitivity in early and late phases in the spared nerve injury model of traumatic neuropathic pain in mice. We further report the strong efficacy of systemic LE inhibitors in reversing ongoing pain in 2 other clinically relevant mouse models-painful diabetic neuropathy and cancer pain. Detailed immunohistochemical analyses on the peripheral tissue samples revealed that both T-Lymphocytes and neutrophils are the sources of LE on peripheral nerve injury, whereas neutrophils are the primary source of LE in diabetic neuropathic conditions. In summary, our results provide compelling evidence for a strong therapeutic potential of generic LE inhibitors for the treatment of neuropathic pain and other chronic pain conditions harboring a neuropathic pain component.


Asunto(s)
Analgésicos/uso terapéutico , Elastasa de Leucocito/uso terapéutico , Neuralgia/tratamiento farmacológico , Animales , Dolor en Cáncer/tratamiento farmacológico , Dolor Crónico/tratamiento farmacológico , Óxidos S-Cíclicos/uso terapéutico , Modelos Animales de Enfermedad , Femenino , Glicina/análogos & derivados , Glicina/uso terapéutico , Hiperalgesia/tratamiento farmacológico , Hiperalgesia/etiología , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Actividad Motora/efectos de los fármacos , Neuralgia/etiología , Dimensión del Dolor , Umbral del Dolor/efectos de los fármacos , Pregabalina/uso terapéutico , Inhibidores de Serina Proteinasa/uso terapéutico , Sulfonamidas/uso terapéutico , Tiazoles/uso terapéutico
10.
Pain ; 158(9): 1765-1779, 2017 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-28614186

RESUMEN

Pathophysiological mechanisms underlying pain associated with cancer are poorly understood. microRNAs (miRNAs) are a class of noncoding RNAs with emerging functional importance in chronic pain. In a genome-wide screen for miRNAs regulated in dorsal root ganglia (DRG) neurons in a mouse model of bone metastatic pain, we identified miR-34c-5p as a functionally important pronociceptive miRNA. Despite these functional insights and therapeutic potential for miR-34c-5p, its molecular mechanism of action in peripheral sensory neurons remains unknown. Here, we report the identification and validation of key target transcripts of miRNA-34c-5p. In-depth bioinformatics analyses revealed Cav2.3, P2rx6, Oprd1, and Oprm1 as high confidence putative targets for miRNA-34c-5p. Of these, canonical and reciprocal regulation of miR-34c-5p and Cav2.3 was observed in cultured sensory neurons as well as in DRG in vivo in mice with cancer pain. Coexpression of miR-34c-5p and Cav2.3 was observed in peptidergic and nonpeptidergic nociceptors, and luciferase reporter assays confirmed functional binding of miR-34c-5p to the 3' UTR of Cav2.3 transcripts. Importantly, knocking down the expression of Cav2.3 specifically in DRG neurons led to hypersensitivity in mice. In summary, these results show that Cav2.3 is a novel mechanistic target for a key pronociceptive miRNA, miR-34c-5p, in the context of cancer pain and indicate an antinociceptive role for Cav2.3 in peripheral sensory neurons. The current study facilitates a deeper understanding of molecular mechanisms underlying cancer pain and suggests a potential for novel therapeutic strategies targeting miR-34c-5p and Cav2.3 in cancer pain.


Asunto(s)
Canales de Calcio Tipo R/metabolismo , Dolor en Cáncer/metabolismo , Proteínas de Transporte de Catión/metabolismo , Ganglios Espinales/patología , MicroARNs/metabolismo , Células Receptoras Sensoriales/metabolismo , Animales , Neoplasias Óseas/complicaciones , Canales de Calcio Tipo R/genética , Dolor en Cáncer/etiología , Dolor en Cáncer/patología , Proteínas de Transporte de Catión/antagonistas & inhibidores , Proteínas de Transporte de Catión/genética , Dependovirus/genética , Modelos Animales de Enfermedad , Regulación Neoplásica de la Expresión Génica/genética , Ontología de Genes , Células HEK293 , Humanos , Luciferasas/genética , Luciferasas/metabolismo , Ratones , Ratones Endogámicos C57BL , MicroARNs/genética , ARN Mensajero/metabolismo , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , Transducción Genética , Transfección
11.
J Mol Neurosci ; 61(2): 178-188, 2017 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-28181149

RESUMEN

Cerebral cavernous malformations (CCM) are vascular lesions associated with loss-of-function mutations in one of the three genes encoding KRIT1 (CCM1), CCM2, and PDCD10. Recent understanding of the molecular mechanisms that lead to CCM development is limited. The role of microRNAs (miRNAs) has been demonstrated in vascular pathologies resulting in loss of tight junction proteins, increased vascular permeability and endothelial cell dysfunction. Since the relevance of miRNAs in CCM pathophysiology has not been elucidated, the primary aim of the study was to identify the miRNA-mRNA expression network associated with CCM. Using small RNA sequencing, we identified a total of 764 matured miRNAs expressed in CCM patients compared to the healthy brains. The expression of the selected miRNAs was validated by qRT-PCR, and the results were found to be consistent with the sequencing data. Upon application of additional statistical stringency, five miRNAs (let-7b-5p, miR-361-5p, miR-370-3p, miR-181a-2-3p, and miR-95-3p) were prioritized to be top CCM-relevant miRNAs. Further in silico analyses revealed that the prioritized miRNAs have a direct functional relation with mRNAs, such as MIB1, HIF1A, PDCD10, TJP1, OCLN, HES1, MAPK1, VEGFA, EGFL7, NF1, and ENG, which are previously characterized as key regulators of CCM pathology. To date, this is the first study to investigate the role of miRNAs in CCM pathology. By employing cutting edge molecular and in silico analyses on clinical samples, the current study reports global miRNA expression changes in CCM patients and provides a rich source of data set to understand detailed molecular machinery involved in CCM pathophysiology.


Asunto(s)
Regulación hacia Abajo , Hemangioma Cavernoso del Sistema Nervioso Central/genética , MicroARNs/genética , Adulto , Estudios de Casos y Controles , Femenino , Redes Reguladoras de Genes , Humanos , Masculino , Persona de Mediana Edad , Transcriptoma
12.
Nat Commun ; 8(1): 176, 2017 08 02.
Artículo en Inglés | MEDLINE | ID: mdl-28765520

RESUMEN

Semaphorins and their transmembrane receptors, Plexins, are key regulators of axon guidance and development of neuronal connectivity. B-type Plexins respond to Class IV semaphorins and mediate a variety of developmental functions. Here we report that the expression of Plexin-B2 and its high-affinity ligand, Sema4C, persists in peripheral sensory neurons in adult life and is markedly increased in states of persistent pain in mice. Genetic deletion of Sema4C as well as adult-onset loss of Plexin-B2 leads to impairment of the development and duration of inflammatory hypersensitivity. Remarkably, unlike the neurodevelopmental functions of Plexin-B2 that solely rely on Ras signaling, we obtained genetic and pharmacological evidence for a requirement of RhoA-ROCK-dependent mechanisms as well as TRPA1 sensitization in pronociceptive functions of Sema4C-Plexin-B2 signaling in adult life. These results suggest important roles for Plexin-B2 signaling in sensory function that may be of therapeutic relevance in pathological pain.Semaphorins and their receptors are involved in neurodevelopment, but their functions in the adult nervous system are not fully understood. This study finds that semaphorin 4C and its receptor Plexin B are expressed in sensory neurons and are pronociceptive in a mouse model of inflammatory pain.


Asunto(s)
Inflamación , Proteínas del Tejido Nervioso/genética , Nocicepción , Dolor/genética , Semaforinas/genética , Células Receptoras Sensoriales/metabolismo , Animales , Modelos Animales de Enfermedad , Ratones , Proteínas del Tejido Nervioso/metabolismo , Dolor/metabolismo , Receptores de Superficie Celular/genética , Receptores de Superficie Celular/metabolismo , Semaforinas/metabolismo , Transducción de Señal , Canal Catiónico TRPA1/metabolismo , Proteínas de Unión al GTP rho/metabolismo , Quinasas Asociadas a rho/metabolismo , Proteína de Unión al GTP rhoA
13.
Sci Rep ; 6: 25923, 2016 05 17.
Artículo en Inglés | MEDLINE | ID: mdl-27184818

RESUMEN

Proprioceptors are responsible for the conscious sensation of limb position and movement, muscle tension or force, and balance. Recent evidence suggests that Piezo2 is a low threshold mechanosensory receptor in the peripheral nervous system, acting as a transducer for touch sensation and proprioception. Thus, we characterized proprioceptive neurons in the mesencephalic trigeminal nucleus that are involved in processing proprioceptive information from the face and oral cavity. This is a specific population of neurons that produce rapidly adapting mechanically-activated currents that are fully dependent on Piezo2. As such, we analyzed the deficits in balance and coordination caused by the selective deletion of the channel in proprioceptors (conditional knockout). The data clearly shows that Piezo2 fulfills a critical role in a defined homogeneous population of proprioceptor neurons that innervate the head muscles, demonstrating that this ion channel is essential for mammalian proprioceptive mechanotransduction.


Asunto(s)
Canales Iónicos/metabolismo , Propiocepción/fisiología , Células Receptoras Sensoriales/metabolismo , Animales , Músculo Masetero/fisiología , Mecanotransducción Celular , Ratones , Tegmento Mesencefálico/fisiología
14.
Nat Commun ; 6: 6820, 2015 Apr 13.
Artículo en Inglés | MEDLINE | ID: mdl-25865668

RESUMEN

Synaptic plasticity is the cornerstone of processes underlying persistent nociceptive activity-induced changes in normal nociceptive sensitivity. Kalirin-7 is a multifunctional guanine-nucleotide-exchange factor (GEF) for Rho GTPases that is characterized by its localization at excitatory synapses, interactions with glutamate receptors and its ability to dynamically modulate the neuronal cytoskeleton. Here we show that spinally expressed Kalirin-7 is required for persistent nociceptive activity-dependent synaptic long-term potentiation as well as activity-dependent remodelling of synaptic spines in the spinal dorsal horn, thereby orchestrating functional and structural plasticity during the course of inflammatory pain.


Asunto(s)
Factores de Intercambio de Guanina Nucleótido/genética , Potenciación a Largo Plazo/fisiología , Nocicepción/fisiología , Dolor/genética , Asta Dorsal de la Médula Espinal/metabolismo , Sinapsis/metabolismo , Animales , Dependovirus/genética , Regulación de la Expresión Génica , Vectores Genéticos , Factores de Intercambio de Guanina Nucleótido/deficiencia , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Neuropéptidos/genética , Neuropéptidos/metabolismo , Dolor/metabolismo , Dolor/fisiopatología , Receptores de N-Metil-D-Aspartato/genética , Receptores de N-Metil-D-Aspartato/metabolismo , Transducción de Señal , Asta Dorsal de la Médula Espinal/fisiopatología , Proteína de Unión al GTP rac1/genética , Proteína de Unión al GTP rac1/metabolismo
15.
Nat Med ; 21(5): 518-23, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25915831

RESUMEN

Neuropathic pain is a major, intractable clinical problem and its pathophysiology is not well understood. Although recent gene expression profiling studies have enabled the identification of novel targets for pain therapy, classical study designs provide unclear results owing to the differential expression of hundreds of genes across sham and nerve-injured groups, which can be difficult to validate, particularly with respect to the specificity of pain modulation. To circumvent this, we used two outbred lines of rats, which are genetically similar except for being genetically segregated as a result of selective breeding for differences in neuropathic pain hypersensitivity. SerpinA3N, a serine protease inhibitor, was upregulated in the dorsal root ganglia (DRG) after nerve injury, which was further validated for its mouse homolog. Mice lacking SerpinA3N developed more neuropathic mechanical allodynia than wild-type (WT) mice, and exogenous delivery of SerpinA3N attenuated mechanical allodynia in WT mice. T lymphocytes infiltrate the DRG after nerve injury and release leukocyte elastase (LE), which was inhibited by SerpinA3N derived from DRG neurons. Genetic loss of LE or exogenous application of a LE inhibitor (Sivelastat) in WT mice attenuated neuropathic mechanical allodynia. Overall, we reveal a novel and clinically relevant role for a member of the serpin superfamily and a leukocyte elastase and crosstalk between neurons and T cells in the modulation of neuropathic pain.


Asunto(s)
Proteínas de Fase Aguda/metabolismo , Inhibidores Enzimáticos/farmacología , Elastasa de Leucocito/antagonistas & inhibidores , Serpinas/metabolismo , Linfocitos T/citología , Animales , Separación Celular , Dependovirus/genética , Femenino , Ganglios Espinales/metabolismo , Hiperalgesia/fisiopatología , Hibridación in Situ , Masculino , Ratones , Ratones Transgénicos , Neuralgia , Neuronas/metabolismo , Análisis de Secuencia por Matrices de Oligonucleótidos , Dolor/fisiopatología , Fenotipo , Reacción en Cadena de la Polimerasa , Ratas , Regulación hacia Arriba
16.
Trends Mol Med ; 20(8): 437-48, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24986063

RESUMEN

Although noncoding RNAs (ncRNAs) were initially considered to be transcriptional byproducts, recent technological advances have led to a steady increase in our understanding of their importance in gene regulation and disease pathogenesis. In keeping with these developments, pain research is also experiencing rapid growth in the investigation of links between ncRNAs and pathological pain. Although the initial focus was on analyzing expression and dysregulation of candidate miRNAs, elucidation of other ncRNAs and ncRNA-mediated functional mechanisms in pain modulation has just commenced. Here we review the major ncRNA literature available to date with respect to pain modulation and discuss tools and opportunities available for testing the impact of other types of ncRNA on pain.


Asunto(s)
Dolor/genética , ARN no Traducido/genética , Animales , Encéfalo/metabolismo , Encéfalo/patología , Regulación de la Expresión Génica , Humanos , Dolor/patología , Manejo del Dolor/métodos , Médula Espinal/metabolismo , Médula Espinal/patología
17.
Neuron ; 83(1): 104-21, 2014 Jul 02.
Artículo en Inglés | MEDLINE | ID: mdl-24991956

RESUMEN

Wnt signaling represents a highly versatile signaling system, which plays diverse and critical roles in various aspects of neural development. Sensory neurons of the dorsal root ganglia require Wnt signaling for initial cell-fate determination as well as patterning and synapse formation. Here we report that Wnt signaling pathways persist in adult sensory neurons and play a functional role in their sensitization in a pathophysiological context. We observed that Wnt3a recruits the Wnt-calcium signaling pathway and the Wnt planar cell polarity pathway in peripheral nerves to alter pain sensitivity in a modality-specific manner and we elucidated underlying mechanisms. In contrast, biochemical, pharmacological, and genetic studies revealed lack of functional relevance for the classical canonical ß-catenin pathway in peripheral sensory neurons in acute modulation of nociception. Finally, this study provides proof-of-concept for a translational potential for Wnt3a-Frizzled3 signaling in alleviating disease-related pain hypersensitivity in cancer-associated pain in vivo.


Asunto(s)
Receptores Frizzled/fisiología , Ganglios Espinales/metabolismo , Hiperalgesia/metabolismo , Células Receptoras Sensoriales/metabolismo , Vía de Señalización Wnt/fisiología , Proteína Wnt3A/fisiología , Animales , Células Cultivadas , Ganglios Espinales/patología , Células HEK293 , Humanos , Hiperalgesia/patología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Nervios Periféricos/metabolismo , Nervios Periféricos/patología , Células Receptoras Sensoriales/patología
18.
EMBO Mol Med ; 5(11): 1740-58, 2013 11.
Artículo en Inglés | MEDLINE | ID: mdl-24039159

RESUMEN

Cancer pain remains a major challenge and there is an urgent demand for the development of specific mechanism-based therapies. Various diseases are associated with unique signatures of expression of microRNAs (miRNAs), which reveal deep insights into disease pathology. Using a comprehensive approach combining genome-wide miRNA screening, molecular and in silico analyses with behavioural approaches in a clinically relevant model of metastatic bone-cancer pain in mice, we now show that tumour-induced conditions are associated with a marked dysregulation of 57 miRNAs in sensory neurons corresponding to tumour-affected areas. By establishing protocols for interference with disease-induced miRNA dysregulation in peripheral sensory neurons in vivo, we functionally validate six dysregulated miRNAs as significant modulators of tumour-associated hypersensitivity. In silico analyses revealed that their predicted targets include key pain-related genes and we identified Clcn3, a gene encoding a chloride channel, as a key miRNA target in sensory neurons, which is functionally important in tumour-induced nociceptive hypersensitivity in vivo. Our results provide new insights into endogenous gene regulatory mechanisms in cancer pain and open up attractive and viable therapeutic options.


Asunto(s)
Genoma , MicroARNs/genética , Neoplasias/genética , Dolor/genética , Animales , Perfilación de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Humanos , Ratones , Ratones Endogámicos C3H , MicroARNs/metabolismo , Neoplasias/metabolismo , Dolor/metabolismo , Células Receptoras Sensoriales/metabolismo
19.
J Clin Invest ; 121(4): 1608-23, 2011 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-21383497

RESUMEN

α-Amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid-type (AMPA-type) glutamate receptors (AMPARs) play an important role in plasticity at central synapses. Although there is anatomical evidence for AMPAR expression in the peripheral nervous system, the functional role of such receptors in vivo is not clear. To address this issue, we generated mice specifically lacking either of the key AMPAR subunits, GluA1 or GluA2, in peripheral, pain-sensing neurons (nociceptors), while preserving expression of these subunits in the central nervous system. Nociceptor-specific deletion of GluA1 led to disruption of calcium permeability and reduced capsaicin-evoked activation of nociceptors. Deletion of GluA1, but not GluA2, led to reduced mechanical hypersensitivity and sensitization in models of chronic inflammatory pain and arthritis. Further analysis revealed that GluA1-containing AMPARs regulated the responses of nociceptors to painful stimuli in inflamed tissues and controlled the excitatory drive from the periphery into the spinal cord. Consequently, peripherally applied AMPAR antagonists alleviated inflammatory pain by specifically blocking calcium-permeable AMPARs, without affecting physiological pain or eliciting central side effects. These findings indicate an important pathophysiological role for calcium-permeable AMPARs in nociceptors and may have therapeutic implications for the treatment chronic inflammatory pain states.


Asunto(s)
Inflamación/fisiopatología , Nociceptores/fisiología , Dolor/fisiopatología , Receptores AMPA/fisiología , Vías Aferentes/crecimiento & desarrollo , Vías Aferentes/fisiopatología , Animales , Artritis Experimental/fisiopatología , Benzodiazepinas/farmacología , Calcio/metabolismo , Ganglios Espinales/crecimiento & desarrollo , Ganglios Espinales/fisiopatología , Inflamación/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Dolor/tratamiento farmacológico , Permeabilidad , Receptores AMPA/antagonistas & inhibidores , Receptores AMPA/deficiencia , Receptores AMPA/genética , Transmisión Sináptica
20.
Nat Med ; 15(7): 802-7, 2009 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-19525966

RESUMEN

Pain is one of the most severe and debilitating symptoms associated with several forms of cancer. Various types of carcinomas and sarcomas metastasize to skeletal bones and cause spontaneous bone pain and hyperalgesia, which is accompanied by bone degradation and remodeling of peripheral nerves. Despite recent advances, the molecular mechanisms underlying the development and maintenance of cancer-evoked pain are not well understood. Several types of non-hematopoietic tumors secrete hematopoietic colony-stimulating factors that act on myeloid cells and tumor cells. Here we report that receptors and signaling mediators of granulocyte- and granulocyte-macrophage colony-stimulating factors (G-CSF and GM-CSF) are also functionally expressed on sensory nerves. GM-CSF sensitized nerves to mechanical stimuli in vitro and in vivo, potentiated CGRP release and caused sprouting of sensory nerve endings in the skin. Interruption of G-CSF and GM-CSF signaling in vivo led to reduced tumor growth and nerve remodeling, and abrogated bone cancer pain. The key significance of GM-CSF signaling in sensory neurons was revealed by an attenuation of tumor-evoked pain following a sensory nerve-specific knockdown of GM-CSF receptors. These results show that G-CSF and GM-CSF are important in tumor-nerve interactions and suggest that their receptors on primary afferent nerve fibers constitute potential therapeutic targets in cancer pain.


Asunto(s)
Neoplasias Óseas/fisiopatología , Factor Estimulante de Colonias de Granulocitos/fisiología , Factor Estimulante de Colonias de Granulocitos y Macrófagos/fisiología , Dolor Intratable/etiología , Células Receptoras Sensoriales/fisiología , Péptido Relacionado con Gen de Calcitonina/metabolismo , Receptores de Factor Estimulante de Colonias de Granulocito/fisiología , Receptores de Factor Estimulante de Colonias de Granulocitos y Macrófagos/fisiología , Transducción de Señal
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA