Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 5 de 5
Filtrar
Más filtros











Intervalo de año de publicación
1.
Proc Natl Acad Sci U S A ; 115(41): 10434-10439, 2018 10 09.
Artículo en Inglés | MEDLINE | ID: mdl-30249655

RESUMEN

HERV-K HML-2 (HK2) has been proliferating in the germ line of humans at least as recently as 250,000 years ago, with some integrations that remain polymorphic in the modern human population. One of the solitary HK2 LTR polymorphic integrations lies between exons 17 and 18 of RASGRF2, a gene that affects dopaminergic activity and is thus related to addiction. Here we show that this antisense HK2 integration (namely RASGRF2-int) is found more frequently in persons who inject drugs compared with the general population. In a Greek HIV-1-positive population (n = 202), we found RASGRF2-int 2.5 times (14 versus 6%) more frequently in patients infected through i.v. drug use compared with other transmission route controls (P = 0.03). Independently, in a United Kingdom-based hepatitis C virus-positive population (n = 184), we found RASGRF2-int 3.6 times (34 versus 9.5%) more frequently in patients infected during chronic drug abuse compared with controls (P < 0.001). We then tested whether RASGRF2-int could be mechanistically responsible for this association by modulating transcription of RASGRF2 We show that the CRISPR/Cas9-mediated insertion of HK2 in HEK293 cells in the exact RASGRF2 intronic position found in the population resulted in significant transcriptional and phenotypic changes. We also explored mechanistic features of other intronic HK2 integrations and show that HK2 LTRs can be responsible for generation of cis-natural antisense transcripts, which could interfere with the transcription of nearby genes. Our findings suggest that RASGRF2-int is a strong candidate for dopaminergic manipulation, and emphasize the importance of accurate mapping of neglected HERV polymorphisms in human genomic studies.


Asunto(s)
Células Madre de Carcinoma Embrionario/metabolismo , Retrovirus Endógenos/genética , Abuso de Sustancias por Vía Intravenosa/genética , Transcripción Genética , Integración Viral/genética , Factores de Intercambio de Guanina Nucleótido ras/genética , Estudios de Casos y Controles , Niño , Estudios de Cohortes , Células Madre de Carcinoma Embrionario/patología , Femenino , Genoma Humano , Humanos , Masculino , Células Tumorales Cultivadas
2.
J Biol Chem ; 286(46): 39836-47, 2011 Nov 18.
Artículo en Inglés | MEDLINE | ID: mdl-21965668

RESUMEN

Painful peripheral neuropathy is a significant clinical problem; however, its pathological mechanism and effective treatments remain elusive. Increased peripheral expression of tetrodotoxin-resistant voltage-gated sodium channel 1.8 (NaV1.8) has been shown to associate with chronic pain symptoms in humans and experimental animals. Sciatic nerve entrapment (SNE) injury was used to develop neuropathic pain symptoms in rats, resulting in increased NaV1.8 mRNA in the injured nerve but not in dorsal root ganglia (DRG). To study the role of NaV1.8 mRNA in the pathogenesis of SNE-induced painful neuropathy, NaV1.8 shRNA vector was delivered by subcutaneous injection of cationized gelatin/plasmid DNA polyplex into the rat hindpaw and its subsequent retrograde transport via sciatic nerve to DRG. This in vivo NaV1.8 shRNA treatment reversibly and repeatedly attenuated the SNE-induced pain symptoms, an effect that became apparent following a distinct lag period of 3-4 days and lasted for 4-6 days before returning to pretreatment levels. Surprisingly, apparent knockdown of NaV1.8 mRNA occurred only in the injured nerve, not in the DRG, during the pain alleviation period. Levels of heteronuclear NaV1.8 RNA were unaffected by SNE or shRNA treatments, suggesting that transcription of the Scn10a gene encoding NaV1.8 was unchanged. Based on these data, we postulate that increased axonal mRNA transport results in accumulation of functional NaV1.8 protein in the injured nerve and the development of painful neuropathy symptoms. Thus, targeted delivery of agents that interfere with axonal NaV1.8 mRNA may represent effective neuropathic pain treatments.


Asunto(s)
Axones/metabolismo , Dolor Crónico/metabolismo , Ganglios Espinales/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Enfermedades del Sistema Nervioso Periférico/metabolismo , ARN Mensajero/biosíntesis , Nervio Ciático/lesiones , Canales de Sodio/metabolismo , Animales , Axones/patología , Dolor Crónico/genética , Dolor Crónico/patología , Ganglios Espinales/patología , Técnicas de Silenciamiento del Gen , Terapia Genética/métodos , Vectores Genéticos/farmacología , Canal de Sodio Activado por Voltaje NAV1.8 , Proteínas del Tejido Nervioso/genética , Enfermedades del Sistema Nervioso Periférico/genética , Enfermedades del Sistema Nervioso Periférico/patología , Enfermedades del Sistema Nervioso Periférico/terapia , ARN Mensajero/genética , Ratas , Ratas Sprague-Dawley , Canales de Sodio/genética
3.
PLoS One ; 6(1): e16204, 2011 Jan 19.
Artículo en Inglés | MEDLINE | ID: mdl-21283824

RESUMEN

BACKGROUND: After dental extraction, the external surface of alveolar bone undergoes resorption at various rates, and a group of patients develop excessive jawbone atrophy. Oral mucosa overlying the atrophied jawbone is unusually thin; therefore, we have hypothesized that excessive jawbone atrophy may be associated with abnormal oral mucosa contraction. FGFR1OP2/wit3.0, a cytoskeleton molecule initially identified in oral wound fibroblasts, has been shown to induce oral mucosa contraction after dental extraction. This study examined the genetic association between single nucleotide polymorphisms (SNPs) of FGFR1OP2/wit3.0 and excessive atrophy of edentulous mandible. METHODS AND FINDINGS: First, the expression of FGFR1OP2/wit3.0 was determined in gingival tissues of 8 subjects before and after dental extraction. In situ hybridization revealed that all subject increased FGFR1OP2/wit3.0 expression in the post-operative oral mucosa tissues; however, significantly high levels of FGFR1OP2/wit3.0 were observed in 3 out of 8 subjects. In a separate study, 20 long-term edentulous subjects (66.4 ± 9.4 years) were recruited. Tag-SNPs in the FGFR1OP2/wit3.0 allele were determined by Taqman-based polymerase chain reaction. The mandibular bone height was determined following the American College of Prosthodontists (ACP) protocol. Subjects with minor allele of rs840869 or rs859024 were found in the highly atrophied group by the ACP classification (Chi square test, p = 0.0384 and p = 0.0565, respectively; Fisher's Exact, p= 0.0515 and p = 0.2604, respectively). The linear regression analysis indicated a suggestive association between rs859024 and the decreased bone heights (Mann-Whitney, p = 0.06). The average bone height of the subjects with rs840869 or rs859024 minor alleles (10.6 ± 3.2 mm and 9.6 ± 3.2 mm, respectively) was significantly smaller than that of those subjects with the major alleles (14.2 ± 4.5 mm, p<0.05). CONCLUSIONS: The patients with the minor allele of rs840869 or rs859024 were associated with excessive atrophy of edentulous mandible. This study may provide the basis for a genetic marker identifying susceptible individuals to develop jawbone atrophy after dental extraction.


Asunto(s)
Estudios de Asociación Genética , Arcada Edéntula/genética , Maxilares/patología , Mandíbula , Polimorfismo de Nucleótido Simple , Proteínas Proto-Oncogénicas/genética , Anciano , Alelos , Atrofia/genética , Marcadores Genéticos , Humanos , Persona de Mediana Edad
4.
Cell Adh Migr ; 4(3): 396-9, 2010.
Artículo en Inglés | MEDLINE | ID: mdl-20448469

RESUMEN

Wound closure and infection control are the primary goal of wound management. A variety of disinfectants and antimicrobial agents are widely available today and routinely achieve infection control. On the contrary, wound closure still remains a challenging goal. Cell adhesion, migration and contraction play significant roles in creating contractile force of patent wound margins and in contributing to wound closure. Modulations of these cellular behaviors have been investigated in the context of wound contraction; however, therapeutic strategy to achieve wound closure has not been established. Recently, we have reported that a previously unknown cytoskeleton molecule, wound inducible transcript-3.0 (wit3.0) also known as fibroblast growth factor receptor 1 oncogene partner 2 (FGFR1OP2), can significantly modulate fibroblast-driven wound closure in vitro and in vivo. The dynamic role of cytoskeleton in different experimental models may provide a novel platform for designing the therapeutic target of wound management.


Asunto(s)
Proteínas del Citoesqueleto/metabolismo , Cicatrización de Heridas/fisiología , Actinas/metabolismo , Animales , Drosophila melanogaster/embriología , Embrión no Mamífero/metabolismo , Embrión no Mamífero/patología , Feto/metabolismo , Feto/patología , Humanos , Mucosa Bucal/metabolismo , Mucosa Bucal/patología , Ratas
5.
Am J Pathol ; 176(1): 108-21, 2010 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-19959814

RESUMEN

Wounds created in the oral cavity heal rapidly and leave minimal scarring. We have examined a role of a previously isolated cDNA from oral wounds encoding wound inducible transcript-3.0 (wit3.0), also known as fibroblast growth factor receptor 1 oncogene partner 2 (FGFR1OP2). FGFR1OP2/wit3.0 was highly expressed in oral wound fibroblasts without noticeable up-regulation of alpha-smooth muscle actin. In silico analyses, denaturing and nondenaturing gel Western blot, and immunocytology together demonstrated that FGFR1OP2/wit3.0 were able to dimerize and oligomerize through coiled-coil structures and appeared to associate with cytoskeleton networks in oral wound fibroblasts. Overexpression of FGFR1OP2/wit3.0 increased the floating collagen gel contraction of naïve oral fibroblasts to the level of oral wound fibroblasts, which was in turn attenuated by small-interfering RNA knockdown. The FGFR1OP2/wit3.0 synthesis did not affect the expression of collagen I as well as procontractile peptides such as alpha-smooth muscle actin, and transforming growth factor-beta1 had no effect on FGFR1OP2/wit3.0 expression. Fibroblastic cells derived from embryonic stem cells carrying FGFR1OP2/wit3.0 (+/-) mutation showed significant retardation in cell migration. Thus, we postulate that FGFR1OP2/wit3.0 may regulate cell motility and stimulate wound closure. FGFR1OP2/wit3.0 was not up-regulated during skin wound healing; however, when treated with FGFR1OP2/wit3.0 -expression vector, the skin wound closure was significantly accelerated, resulting in the limited granulation tissue formation. Our data suggest that FGFR1OP2/wit3.0 may possess a therapeutic potential for wound management.


Asunto(s)
Proteínas del Citoesqueleto/metabolismo , Citoesqueleto/metabolismo , Fibroblastos/metabolismo , Fibroblastos/patología , Cadenas Pesadas de Miosina/metabolismo , Proteínas/metabolismo , Cicatrización de Heridas , Animales , Secuencia de Bases , Movimiento Celular/efectos de los fármacos , Colágeno/metabolismo , Proteínas del Citoesqueleto/genética , Citoesqueleto/efectos de los fármacos , Fibroblastos/efectos de los fármacos , Geles , Técnicas de Silenciamiento del Gen , Humanos , Ratones , Datos de Secuencia Molecular , Mucosa Bucal/efectos de los fármacos , Mucosa Bucal/metabolismo , Mucosa Bucal/patología , Mutación/genética , Cadenas Pesadas de Miosina/genética , Polimorfismo de Nucleótido Simple/genética , Transporte de Proteínas/efectos de los fármacos , Proteínas/genética , ARN Interferente Pequeño/metabolismo , Ratas , Factor de Crecimiento Transformador beta1/farmacología , Cicatrización de Heridas/efectos de los fármacos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA