Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 6 de 6
Filtrar
Más filtros













Base de datos
Intervalo de año de publicación
1.
Front Mol Neurosci ; 17: 1398839, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38783904

RESUMEN

Chronic pain is common and inadequately treated, making the development of safe and effective analgesics a high priority. Our previous data indicate that carbonic anhydrase-8 (CA8) expression in dorsal root ganglia (DRG) mediates analgesia via inhibition of neuronal ER inositol trisphosphate receptor-1 (ITPR1) via subsequent decrease in ER calcium release and reduction of cytoplasmic free calcium, essential to the regulation of neuronal excitability. This study tested the hypothesis that novel JDNI8 replication-defective herpes simplex-1 viral vectors (rdHSV) carrying a CA8 transgene (vHCA8) reduce primary afferent neuronal excitability. Whole-cell current clamp recordings in small DRG neurons showed that vHCA8 transduction caused prolongation of their afterhyperpolarization (AHP), an essential regulator of neuronal excitability. This AHP prolongation was completely reversed by the specific Kv7 channel inhibitor XE-991. Voltage clamp recordings indicate an effect via Kv7 channels in vHCA8-infected small DRG neurons. These data demonstrate for the first time that vHCA8 produces Kv7 channel activation, which decreases neuronal excitability in nociceptors. This suppression of excitability may translate in vivo as non-opioid dependent behavioral- or clinical analgesia, if proven behaviorally and clinically.

2.
Mamm Genome ; 31(9-12): 287-294, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-33247772

RESUMEN

Intracellular calcium is critical in orchestrating neuronal excitability and analgesia. Carbonic anhydrase-8 (CA8) regulates intracellular calcium signaling through allosteric inhibition of neuronal inositol trisphosphate receptor 1 (ITPR1) to produce profound analgesia. Recently, we reported the "G" allele at rs6471859 represents cis-eQTL regulating alternative splicing of a 1697 bp transcript (CA8-204G) with a retained intron, alternative polyadenylation site and a new stop codon producing a functional 26 kDa peptide with an extended exon 3. In this study we show the reversion mutation (G to C) at rs6471859 within the CA8-204G expression vector also produced a stable 1697 bp transcript (CA8-204C) coding for a smaller peptide (~ 22 kDa) containing only the first three CA8 exons. Surprisingly, this peptide inhibited ITPR1 (pITPR1) activation, ITPR1-mediated calcium release in vitro; and produced profound analgesia in vivo. This is the first report showing CA8-204C codes for a functional peptide sufficient to regulate calcium signaling and produce profound analgesia.


Asunto(s)
Analgesia , Biomarcadores de Tumor/genética , Calcio/metabolismo , ADN Complementario , Mutación , Péptidos/genética , Adenosina Trifosfato/metabolismo , Animales , Biomarcadores de Tumor/química , Dependovirus/genética , Técnicas de Transferencia de Gen , Vectores Genéticos/genética , Humanos , Ratones , Dolor/etiología , Dolor/metabolismo , Transducción Genética
3.
PLoS Genet ; 15(6): e1008226, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-31199789

RESUMEN

Carbonic anhydrase-8 (CA8) is an intracellular protein that functions as an allosteric inhibitor of inositol trisphosphate receptor-1 (ITPR1) critical to intracellular Ca++ release, synaptic functions and neuronal excitability. We showed previously that murine nociception and analgesic responses are regulated by the expression of this gene in dorsal root ganglion (DRG) associated with a cis-eQTL. In this report, we identify an exon-level cis-eQTL (rs6471859) that regulates human DRG CA8 alternative splicing, producing a truncated 1,697bp transcript (e.g., CA8-204). Our functional genomic studies show the "G" allele at rs6471859 produces a cryptic 3'UTR splice site regulating expression of CA8-204. We developed constructs to study the expression and function of the naturally occurring CA8-204G transcript (G allele at rs6471859), CA8-204C (C allele at rs6471859 reversion mutation) and CA8-201 (full length transcript). CA8-204G transcript expression occurred predominantly in non-neuronal cells (HEK293), while CA8-204C expression was restricted to neuronal derived cells (NBL) in vitro. CA8-204G produced a stable truncated transcript in HEK293 cells that was barely detectable in NBL cells. We also show CA8-204 produces a stable peptide that inhibits pITPR1 and Ca++ release in HEK293 cells. These results imply homozygous G/G individuals at rs6471859, which are common in the general population, produce exclusively CA8-204G that is barely detectable in neuronal cells. CA8 null mutations that greatly impact neuronal functions are associated with severe forms of spinal cerebellar ataxia, and our data suggest G/G homozygotes should display a similar phenotype. To address this question, we show in vivo using AAV8-FLAG-CA8-204G and AAV8-V5-CA8-201 gene transfer delivered via intra-neural sciatic nerve injection (SN), that these viral constructs are able to transduce DRG cells and produce similar analgesic and anti-hyperalgesic responses to inflammatory pain. Immunohistochemistry (IHC) examinations of DRG tissues further show CA8-204G peptide is expressed in advillin expressing neuronal cells, but to a lesser extent compared to glial cells. These findings explain why G/G homozygotes that exclusively produce this truncated functional peptide in DRG evade a severe phenotype. These genomic studies significantly advance the literature regarding structure-function studies on CA8-ITPR1 critical to calcium signaling pathways, synaptic functioning, neuronal excitability and analgesic responses.


Asunto(s)
Biomarcadores de Tumor/genética , Señalización del Calcio/genética , Receptores de Inositol 1,4,5-Trifosfato/genética , Neuronas/metabolismo , Dolor/genética , Empalme Alternativo/genética , Animales , Biomarcadores de Tumor/farmacología , Cerebelo/efectos de los fármacos , Cerebelo/metabolismo , Ganglios Espinales/metabolismo , Ganglios Espinales/patología , Técnicas de Transferencia de Gen , Células HEK293 , Humanos , Ratones , Mutación/genética , Neuronas/patología , Especificidad de Órganos , Dolor/patología , Péptidos/genética , Péptidos/farmacología , Sitios de Carácter Cuantitativo/genética , Sitios de Empalme de ARN/genética , Nervio Ciático/efectos de los fármacos , Nervio Ciático/metabolismo
4.
Gene Ther ; 25(4): 297-311, 2018 07.
Artículo en Inglés | MEDLINE | ID: mdl-29789638

RESUMEN

Carbonic anhydrase-8 (Car8; murine gene symbol) is an allosteric inhibitor of inositol trisphosphate receptor-1 (ITPR1), which regulates neuronal intracellular calcium release. We previously reported that wild-type Car8 overexpression corrects the baseline allodynia and hyperalgesia associated with calcium dysregulation in the waddle (wdl) mouse due to a 19 bp deletion in exon 8 of the Car8 gene. In this report, we provide preliminary evidence that overexpression of the human wild-type ortholog of Car8 (CA8WT), but not the reported CA8 S100P loss-of-function mutation (CA8MT), inhibits nerve growth factor (NGF)-induced phosphorylation of ITPR1, TrkA (NGF high-affinity receptor), and ITPR1-mediated cytoplasmic free calcium release in vitro. In addition, we show that gene transfer using AAV8-V5-CA8WT viral particles via sciatic nerve injection demonstrates retrograde transport to dorsal root ganglia (DRG) producing prolonged V5-CA8WT expression, pITPR1 and pTrkA inhibition, and profound analgesia and anti-hyperalgesia in male C57BL/6J mice. AAV8-V5-CA8WT-mediated overexpression prevented and treated allodynia and hyperalgesia associated with chronic neuropathic pain produced by the spinal nerve ligation (SNL) model. These AAV8-V5-CA8 data provide a proof-of-concept for precision medicine through targeted gene therapy of NGF-responsive somatosensory neurons as a long-acting local analgesic able to prevent and treat chronic neuropathic pain through regulating TrkA signaling, ITPR1 activation, and intracellular free calcium release by ITPR1.


Asunto(s)
Biomarcadores de Tumor/genética , Terapia Genética/métodos , Hiperalgesia/terapia , Receptores de Inositol 1,4,5-Trifosfato/efectos de los fármacos , Factor de Crecimiento Nervioso/antagonistas & inhibidores , Analgesia/métodos , Animales , Biomarcadores de Tumor/biosíntesis , Dependovirus/genética , Modelos Animales de Enfermedad , Humanos , Hiperalgesia/genética , Receptores de Inositol 1,4,5-Trifosfato/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Factor de Crecimiento Nervioso/genética , Factor de Crecimiento Nervioso/metabolismo , Neuralgia/genética , Neuralgia/terapia , Neuronas/metabolismo , Manejo del Dolor/métodos , Fosforilación , Transducción de Señal
5.
Neuroreport ; 28(18): 1215-1220, 2017 Dec 13.
Artículo en Inglés | MEDLINE | ID: mdl-28902707

RESUMEN

Recently, we showed that murine dorsal root ganglion (DRG) Car8 expression is a cis-regulated eQTL that determines analgesic responses. In this report, we show that transduction through sciatic nerve injection of DRG with human wild-type carbonic anhydrase-8 using adeno-associated virus viral particles (AAV8-V5-CA8WT) produces analgesia in naive male C57BL/6J mice and antihyperalgesia after carrageenan treatment. A peak mean increase of about 4 s in thermal hindpaw withdrawal latency equaled increases in thermal withdrawal latency produced by 10 mg/kg intraperitoneal morphine in these mice. Allometric conversion of this intraperitoneal morphine dose in mice equals an oral morphine dose of about 146 mg in a 60-kg adult. Our work quantifies for the first time analgesia and antihyperalgesia in an inflammatory pain model after DRG transduction by CA8 gene therapy.


Asunto(s)
Analgésicos Opioides/uso terapéutico , Biomarcadores de Tumor/uso terapéutico , Hiperalgesia/terapia , Morfina/uso terapéutico , Manejo del Dolor/métodos , Umbral del Dolor/fisiología , Dolor/fisiopatología , Adenoviridae/genética , Animales , Biomarcadores de Tumor/genética , Carragenina/toxicidad , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Ganglios Espinales/efectos de los fármacos , Ganglios Espinales/metabolismo , Humanos , Inflamación/inducido químicamente , Inflamación/complicaciones , Masculino , Ratones , Ratones Endogámicos C57BL , Dolor/tratamiento farmacológico , Dolor/etiología , Dimensión del Dolor/efectos de los fármacos , Umbral del Dolor/efectos de los fármacos , Tiempo de Reacción/efectos de los fármacos , Transducción Genética
6.
PLoS One ; 10(3): e0118273, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25734498

RESUMEN

Calcium dysregulation is causally linked with various forms of neuropathology including seizure disorders, multiple sclerosis, Huntington's disease, Alzheimer's, spinal cerebellar ataxia (SCA) and chronic pain. Carbonic anhydrase-8 (Car8) is an allosteric inhibitor of inositol trisphosphate receptor-1 (ITPR1), which regulates intracellular calcium release fundamental to critical cellular functions including neuronal excitability, neurite outgrowth, neurotransmitter release, mitochondrial energy production and cell fate. In this report we test the hypothesis that Car8 regulation of ITPR1 and cytoplasmic free calcium release is critical to nociception and pain behaviors. We show Car8 null mutant mice (MT) exhibit mechanical allodynia and thermal hyperalgesia. Dorsal root ganglia (DRG) from MT also demonstrate increased steady-state ITPR1 phosphorylation (pITPR1) and cytoplasmic free calcium release. Overexpression of Car8 wildtype protein in MT nociceptors complements Car8 deficiency, down regulates pITPR1 and abolishes thermal and mechanical hypersensitivity. We also show that Car8 nociceptor overexpression alleviates chronic inflammatory pain. Finally, inflammation results in downregulation of DRG Car8 that is associated with increased pITPR1 expression relative to ITPR1, suggesting a possible mechanism of acute hypersensitivity. Our findings indicate Car8 regulates the ITPR1-cytosolic free calcium pathway that is critical to nociception, inflammatory pain and possibly other neuropathological states. Car8 and ITPR1 represent new therapeutic targets for chronic pain.


Asunto(s)
Biomarcadores de Tumor/genética , Calcio/metabolismo , Dolor Crónico/genética , Ganglios Espinales/metabolismo , Hiperalgesia/genética , Receptores de Inositol 1,4,5-Trifosfato/genética , Proteínas del Tejido Nervioso/genética , Animales , Biomarcadores de Tumor/deficiencia , Señalización del Calcio , Dolor Crónico/metabolismo , Dolor Crónico/fisiopatología , Citosol/metabolismo , Modelos Animales de Enfermedad , Femenino , Ganglios Espinales/fisiopatología , Regulación de la Expresión Génica , Prueba de Complementación Genética , Hiperalgesia/metabolismo , Hiperalgesia/fisiopatología , Inflamación , Receptores de Inositol 1,4,5-Trifosfato/metabolismo , Potenciación a Largo Plazo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas del Tejido Nervioso/deficiencia , Neuronas/metabolismo , Neuronas/patología , Nocicepción/fisiología , Fosforilación
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA