Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
Cell Rep ; 42(4): 112295, 2023 04 25.
Artículo en Inglés | MEDLINE | ID: mdl-36947543

RESUMEN

Corticospinal tract (CST) neurons innervate the deep spinal dorsal horn to sustain chronic neuropathic pain. The majority of neurons targeted by the CST are interneurons expressing the transcription factor c-Maf. Here, we used intersectional genetics to decipher the function of these neurons in dorsal horn sensory circuits. We find that excitatory c-Maf (c-MafEX) neurons receive sensory input mainly from myelinated fibers and target deep dorsal horn parabrachial projection neurons and superficial dorsal horn neurons, thereby connecting non-nociceptive input to nociceptive output structures. Silencing c-MafEX neurons has little effect in healthy mice but alleviates mechanical hypersensitivity in neuropathic mice. c-MafEX neurons also receive input from inhibitory c-Maf and parvalbumin neurons, and compromising inhibition by these neurons caused mechanical hypersensitivity and spontaneous aversive behaviors reminiscent of c-MafEX neuron activation. Our study identifies c-MafEX neurons as normally silent second-order nociceptors that become engaged in pathological pain signaling upon loss of inhibitory control.


Asunto(s)
Neuralgia , Asta Dorsal de la Médula Espinal , Animales , Ratones , Asta Dorsal de la Médula Espinal/patología , Médula Espinal , Células del Asta Posterior/fisiología , Transmisión Sináptica , Interneuronas/fisiología , Proteínas Proto-Oncogénicas c-maf
2.
Dis Model Mech ; 16(1)2023 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-36695500

RESUMEN

Parkinson's disease (PD), an age-dependent neurodegenerative disease, is characterised by the selective loss of dopaminergic neurons in the substantia nigra (SN). Mitochondrial dysfunction is a hallmark of PD, and mutations in PINK1, a gene necessary for mitochondrial fitness, cause PD. Drosophila melanogaster flies with pink1 mutations exhibit mitochondrial defects and dopaminergic cell loss and are used as a PD model. To gain an integrated view of the cellular changes caused by defects in the PINK1 pathway of mitochondrial quality control, we combined metabolomics and transcriptomics analysis in pink1-mutant flies with human induced pluripotent stem cell (iPSC)-derived neural precursor cells (NPCs) with a PINK1 mutation. We observed alterations in cysteine metabolism in both the fly and human PD models. Mitochondrial dysfunction in the NPCs resulted in changes in several metabolites that are linked to cysteine synthesis and increased glutathione levels. We conclude that alterations in cysteine metabolism may compensate for increased oxidative stress in PD, revealing a unifying mechanism of early-stage PD pathology that may be targeted for drug development. This article has an associated First Person interview with the first author of the paper.


Asunto(s)
Proteínas de Drosophila , Células Madre Pluripotentes Inducidas , Células-Madre Neurales , Enfermedades Neurodegenerativas , Enfermedad de Parkinson , Animales , Humanos , Drosophila melanogaster/metabolismo , Cisteína , Enfermedad de Parkinson/metabolismo , Células-Madre Neurales/metabolismo , Células Madre Pluripotentes Inducidas/metabolismo , Proteínas Quinasas/metabolismo , Proteínas de Drosophila/metabolismo , Proteínas Serina-Treonina Quinasas/genética
3.
J Vis Exp ; (135)2018 05 12.
Artículo en Inglés | MEDLINE | ID: mdl-29806830

RESUMEN

Selective manipulation of spinal neuronal subpopulations has mainly been achieved by two different methods: 1) Intersectional genetics, whereby double or triple transgenic mice are generated in order to achieve selective expression of a reporter or effector gene (e.g., from the Rosa26 locus) in the desired spinal population. 2) Intraspinal injection of Cre-dependent recombinant adeno-associated virus (rAAV); here Cre-dependent AAV vectors coding for the reporter or effector gene of choice are injected into the spinal cord of mice expressing Cre recombinase in the desired neuronal subpopulation. This protocol describes how to generate Cre-dependent rAAV vectors and how to transduce neurons in the dorsal horn of the lumbar spinal cord segments L3-L5 with rAAVs. As the lumbar spinal segments L3-L5 are innervated by those peripheral sensory neurons that transmit sensory information from the hindlimbs, spontaneous behavior and responses to sensory tests applied to the hindlimb ipsilateral to the injection side can be analyzed in order to interrogate the function of the manipulated neurons in sensory processing. We provide examples of how this technique can be used to analyze genetically defined subsets of spinal neurons. The main advantages of virus-mediated transgene expression in Cre transgenic mice compared to classical reporter mouse-induced transgene expression are the following: 1) Different Cre-dependent rAAVs encoding various reporter or effector proteins can be injected into a single Cre transgenic line, thus overcoming the need to create several multiple transgenic mouse lines. 2) Intraspinal injection limits manipulation of Cre-expressing cells to the injection site and to the time after injection. The main disadvantages are: 1) Reporter gene expression from rAAVs is more variable. 2) Surgery is required to transduce the spinal neurons of interest. Which of the two methods is more appropriate depends on the neuron population and research question to be addressed.


Asunto(s)
Adenoviridae/genética , Terapia Genética/métodos , Médula Espinal/fisiología , Médula Espinal/virología , Transgenes , Animales , Ratones , Ratones Transgénicos , Médula Espinal/citología , Médula Espinal/metabolismo
4.
J Neurosci ; 37(43): 10358-10371, 2017 10 25.
Artículo en Inglés | MEDLINE | ID: mdl-28951448

RESUMEN

Glycoprotein-deleted rabies virus-mediated monosynaptic tracing has become a standard method for neuronal circuit mapping, and is applied to virtually all parts of the rodent nervous system, including the spinal cord and primary sensory neurons. Here we identified two classes of unmyelinated sensory neurons (nonpeptidergic and C-fiber low-threshold mechanoreceptor neurons) resistant to direct and trans-synaptic infection from the spinal cord with rabies viruses that carry glycoproteins in their envelopes and that are routinely used for infection of CNS neurons (SAD-G and N2C-G). However, the same neurons were susceptible to infection with EnvA-pseudotyped rabies virus in tumor virus A receptor transgenic mice, indicating that resistance to retrograde infection was due to impaired virus adsorption rather than to deficits in subsequent steps of infection. These results demonstrate an important limitation of rabies virus-based retrograde tracing of sensory neurons in adult mice, and may help to better understand the molecular machinery required for rabies virus spread in the nervous system. In this study, mice of both sexes were used.SIGNIFICANCE STATEMENT To understand the neuronal bases of behavior, it is important to identify the underlying neural circuitry. Rabies virus-based monosynaptic tracing has been used to identify neuronal circuits in various parts of the nervous system. This has included connections between peripheral sensory neurons and their spinal targets. These connections form the first synapse in the somatosensory pathway. Here we demonstrate that two classes of unmyelinated sensory neurons, which account for >40% of dorsal root ganglia neurons, display resistance to rabies infection. Our results are therefore critical for interpreting monosynaptic rabies-based tracing in the sensory system. In addition, identification of rabies-resistant neurons might provide a means for future studies addressing rabies pathobiology.


Asunto(s)
Ganglios Espinales/química , Red Nerviosa/química , Técnicas de Trazados de Vías Neuroanatómicas/métodos , Virus de la Rabia , Células Receptoras Sensoriales/química , Animales , Femenino , Ganglios Espinales/citología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Red Nerviosa/citología , Células del Asta Posterior/química
5.
J Neurochem ; 142(5): 721-733, 2017 09.
Artículo en Inglés | MEDLINE | ID: mdl-28700081

RESUMEN

Recombinant adeno-associated virus (rAAV) vector-mediated gene transfer into genetically defined neuron subtypes has become a powerful tool to study the neuroanatomy of neuronal circuits in the brain and to unravel their functions. More recently, this methodology has also become popular for the analysis of spinal cord circuits. To date, a variety of naturally occurring AAV serotypes and genetically modified capsid variants are available but transduction efficiency in spinal neurons, target selectivity, and the ability for retrograde tracing are only incompletely characterized. Here, we have compared the transduction efficiency of seven commonly used AAV serotypes after intraspinal injection. We specifically analyzed local transduction of different types of dorsal horn neurons, and retrograde transduction of dorsal root ganglia (DRG) neurons and of neurons in the rostral ventromedial medulla (RVM) and the somatosensory cortex (S1). Our results show that most of the tested rAAV vectors have similar transduction efficiency in spinal neurons. All serotypes analyzed were also able to transduce DRG neurons and descending RVM and S1 neurons via their spinal axon terminals. When comparing the commonly used rAAV serotypes to the recently developed serotype 2 capsid variant rAAV2retro, a > 20-fold increase in transduction efficiency of descending supraspinal neurons was observed. Conversely, transgene expression in retrogradely transduced neurons was strongly reduced when the human synapsin 1 (hSyn1) promoter was used instead of the strong ubiquitous hybrid cytomegalovirus enhancer/chicken ß-actin promoter (CAG) or cytomegalovirus (CMV) promoter fragments. We conclude that the use of AAV2retro greatly increases transduction of neurons connected to the spinal cord via their axon terminals, while the hSyn1 promoter can be used to minimize transgene expression in retrogradely connected neurons of the DRG or brainstem. Cover Image for this issue: doi. 10.1111/jnc.13813.


Asunto(s)
Adenoviridae , Vectores Genéticos/farmacología , Red Nerviosa/fisiología , Regiones Promotoras Genéticas/fisiología , Serogrupo , Médula Espinal/fisiología , Animales , Pollos , Femenino , Masculino , Ratones , Ratones Endogámicos C57BL , Red Nerviosa/efectos de los fármacos , Neuronas/efectos de los fármacos , Neuronas/fisiología , Regiones Promotoras Genéticas/efectos de los fármacos , Proteínas Recombinantes/farmacología , Médula Espinal/efectos de los fármacos
6.
Neuron ; 85(6): 1289-304, 2015 Mar 18.
Artículo en Inglés | MEDLINE | ID: mdl-25789756

RESUMEN

The gate control theory of pain proposes that inhibitory neurons of the spinal dorsal horn exert critical control over the relay of nociceptive signals to higher brain areas. Here we investigated how the glycinergic subpopulation of these neurons contributes to modality-specific pain and itch processing. We generated a GlyT2::Cre transgenic mouse line suitable for virus-mediated retrograde tracing studies and for spatially precise ablation, silencing, and activation of glycinergic neurons. We found that these neurons receive sensory input mainly from myelinated primary sensory neurons and that their local toxin-mediated ablation or silencing induces localized mechanical, heat, and cold hyperalgesia; spontaneous flinching behavior; and excessive licking and biting directed toward the corresponding skin territory. Conversely, local pharmacogenetic activation of the same neurons alleviated neuropathic hyperalgesia and chloroquine- and histamine-induced itch. These results establish glycinergic neurons of the spinal dorsal horn as key elements of an inhibitory pain and itch control circuit.


Asunto(s)
Red Nerviosa/fisiopatología , Neuronas/citología , Dolor/fisiopatología , Prurito/fisiopatología , Asta Dorsal de la Médula Espinal/citología , Animales , Modelos Animales de Enfermedad , Glicina/metabolismo , Hiperalgesia/patología , Ratones , Ratones Transgénicos , Red Nerviosa/metabolismo , Red Nerviosa/patología , Enfermedades del Sistema Nervioso Periférico/genética , Enfermedades del Sistema Nervioso Periférico/patología , Enfermedades del Sistema Nervioso Periférico/fisiopatología , Células del Asta Posterior/fisiología
7.
Hum Gene Ther Methods ; 25(1): 14-32, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24090197

RESUMEN

RNA silencing is an established method for investigating gene function and has attracted particular interest because of the potential for generating RNA-based therapeutics. Using lentiviral vectors as an efficient delivery system that offers stable, long-term expression in postmitotic cells further enhances the applicability of an RNA-based gene therapy for the CNS. In this review we provide an overview of both lentiviral vectors and RNA silencing along with design considerations for generating lentiviral vectors capable of RNA silencing. We go on to describe the current preclinical data regarding lentiviral vector-mediated RNA silencing for CNS disorders and discuss the concerns of side effects associated with lentiviral vectors and small interfering RNAs and how these might be mitigated.


Asunto(s)
Enfermedades del Sistema Nervioso Central/genética , Enfermedades del Sistema Nervioso Central/metabolismo , Vectores Genéticos/metabolismo , Lentivirus/genética , Interferencia de ARN , Enfermedades del Sistema Nervioso Central/patología , Vectores Genéticos/química , Vectores Genéticos/genética , Humanos , ARN Interferente Pequeño/metabolismo
8.
Mol Ther ; 21(1): 49-56, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-22910296

RESUMEN

Neuropathic pain is a chronic condition that is often refractory to treatment with available therapies and thus an unmet medical need. We have previously shown that the voltage-gated sodium channel Na(v)1.3 is upregulated in peripheral and central nervous system (CNS) of rats following nerve injury, and that it contributes to nociceptive neuron hyperexcitability in neuropathic conditions. To evaluate the therapeutic potential of peripheral Na(v)1.3 knockdown at a specific segmental level, we constructed adeno-associated viral (AAV) vector expressing small hairpin RNA against rat Na(v)1.3 and injected it into lumbar dorsal root ganglion (DRG) of rats with spared nerve injury (SNI). Our data show that direct DRG injection provides a model that can be used for proof-of-principle studies in chronic pain with respect to peripheral delivery route of gene transfer constructs, high transduction efficiency, flexibility in terms of segmental localization, and limited behavioral effects of the surgical procedure. We show that knockdown of Na(v)1.3 in lumbar 4 (L4) DRG results in an attenuation of nerve injury-induced mechanical allodynia in the SNI model. Taken together, our studies support the contribution of peripheral Na(v)1.3 to pain in adult rats with neuropathic pain, validate Na(v)1.3 as a target, and provide validation for this approach of AAV-mediated peripheral gene therapy.


Asunto(s)
Dependovirus/genética , Ganglios Espinales/metabolismo , Técnicas de Silenciamiento del Gen , Vectores Genéticos , Canal de Sodio Activado por Voltaje NAV1.3/fisiología , Enfermedades del Sistema Nervioso Periférico/prevención & control , ARN Interferente Pequeño/genética , Animales , Secuencia de Bases , Línea Celular , Cartilla de ADN , Regulación hacia Abajo , Masculino , Canal de Sodio Activado por Voltaje NAV1.3/genética , Técnicas de Placa-Clamp , Ratas , Ratas Sprague-Dawley , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
9.
J Gene Med ; 14(5): 299-315, 2012 May.
Artículo en Inglés | MEDLINE | ID: mdl-22499506

RESUMEN

BACKGROUND: Knocking down neuronal LINGO-1 using short hairpin RNAs (shRNAs) might enhance axon regeneration in the central nervous system (CNS). Integration-deficient lentiviral vectors have great potential as a therapeutic delivery system for CNS injuries. However, recent studies have revealed that shRNAs can induce an interferon response resulting in off-target effects and cytotoxicity. METHODS: CNS neurones were transduced with integration-deficient lentiviral vectors in vitro. The transcriptional effect of shRNA expression was analysed using quantitative real time-polymerase chain reaction and northern blots were used to assess shRNA production. RESULTS: Integration-deficient lentiviral vectors efficiently transduced CNS neurones and knocked down LINGO-1 mRNA in vitro. However, an increase in cell death was observed when lentiviral vectors encoding an shRNA were applied or when high vector concentrations were used. We demonstrate that high doses of vector or the use of vectors encoding shRNAs can induce an up-regulation of interferon-stimulated genes (2',5'-oligoadenylate synthase 1 and protein kinase R although not myxovirus resistance 1) and a down-regulation of off-target genes (including p75(NTR) and Nogo receptor 1). Furthermore, the northern blot demonstrated that these negative consequences occur even when lentiviral vectors express low levels of shRNAs. Taken together, these results may explain why neurite outgrowth was not enhanced on an inhibitory substrate following transduction with lentiviral vectors encoding an shRNA targeting LINGO-1. CONCLUSIONS: These findings highlight the importance of including appropriate controls to verify silencing specificity and the requirement to check for an interferon response when conducting RNA interference experiments. However, the potential benefits that RNA interference and viral vectors offer to gene-based therapies to CNS injuries cannot be overlooked and demand further investigation.


Asunto(s)
Sistema Nervioso Central , Proteínas de la Membrana/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Neuritas/metabolismo , ARN Interferente Pequeño/genética , Regeneración/genética , Animales , Sistema Nervioso Central/crecimiento & desarrollo , Sistema Nervioso Central/metabolismo , Regulación del Desarrollo de la Expresión Génica , Técnicas de Transferencia de Gen/efectos adversos , Vectores Genéticos , Células HEK293 , Células HeLa , Humanos , Interferones/metabolismo , Lentivirus , Proteínas de la Membrana/genética , Proteínas del Tejido Nervioso/genética , Interferencia de ARN , ARN Mensajero/genética , ARN Mensajero/metabolismo , ARN Interferente Pequeño/efectos adversos , Ratas
10.
Brain ; 133(Pt 6): 1823-35, 2010 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-20478850

RESUMEN

The Na(v)1.7 sodium channel is preferentially expressed in nocioceptive dorsal root ganglion and sympathetic ganglion neurons. Gain-of-function mutations in Na(v)1.7 produce the nocioceptor hyperexcitability underlying inherited erythromelalgia, characterized in most kindreds by early-age onset of severe pain. Here we describe a mutation (Na(v)1.7-G616R) in a pedigree with adult-onset of pain in some family members. The mutation shifts the voltage-dependence of channel fast-inactivation in a depolarizing direction in the adult-long, but not in the neonatal-short splicing isoform of Na(v)1.7 in dorsal root ganglion neurons. Altered inactivation does not depend on the age of the dorsal root ganglion neurons in which the mutant is expressed. Expression of the mutant adult-long, but not the mutant neonatal-short, isoform of Na(v)1.7 renders dorsal root ganglion neurons hyperexcitable, reducing the current threshold for generation of action potentials, increasing spontaneous activity and increasing the frequency of firing in response to graded suprathreshold stimuli. This study shows that a change in relative expression of splice isoforms can contribute to time-dependent manifestation of the functional phenotype of a sodium channelopathy.


Asunto(s)
Empalme Alternativo , Eritromelalgia/genética , Canales de Sodio/genética , Adolescente , Edad de Inicio , Anciano de 80 o más Años , Animales , Animales Recién Nacidos , Niño , Eritromelalgia/fisiopatología , Femenino , Ganglios Espinales/fisiopatología , Humanos , Técnicas In Vitro , Masculino , Persona de Mediana Edad , Canal de Sodio Activado por Voltaje NAV1.7 , Dolor/genética , Dolor/fisiopatología , Fenotipo , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Ratas , Ratas Sprague-Dawley , Canales de Sodio/metabolismo , Adulto Joven
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA