Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
1.
Hum Mol Genet ; 28(20): 3355-3368, 2019 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-31332443

RESUMEN

MicroRNA-204 (miR-204) is expressed in pulmonary, renal, mammary and eye tissue, and its reduction can result in multiple diseases including cancer. We first generated miR-204-/- mice to study the impact of miR-204 loss on retinal and retinal pigment epithelium (RPE) structure and function. The RPE is fundamentally important for maintaining the health and integrity of the retinal photoreceptors. miR-204-/- eyes evidenced areas of hyper-autofluorescence and defective photoreceptor digestion, along with increased microglia migration to the RPE. Migratory Iba1+ microglial cells were localized to the RPE apical surface where they participated in the phagocytosis of photoreceptor outer segments (POSs) and contributed to a persistent build-up of rhodopsin. These structural, molecular and cellular outcomes were accompanied by decreased light-evoked electrical responses from the retina and RPE. In parallel experiments, we suppressed miR-204 expression in primary cultures of human RPE using anti-miR-204. In vitro suppression of miR-204 in human RPE similarly showed abnormal POS clearance and altered expression of autophagy-related proteins and Rab22a, a regulator of endosome maturation. Together, these in vitro and in vivo experiments suggest that the normally high levels of miR-204 in RPE can mitigate disease onset by preventing generation of oxidative stress and inflammation originating from intracellular accumulation of undigested photoreactive POS lipids. More generally, these results implicate RPE miR-204-mediated regulation of autophagy and endolysosomal interaction as a critical determinant of normal RPE/retina structure and function.


Asunto(s)
MicroARNs/metabolismo , Fagocitosis/fisiología , Fagosomas/metabolismo , Retina/metabolismo , Epitelio Pigmentado de la Retina/metabolismo , Animales , Células Cultivadas , Quimiocinas/metabolismo , Citocinas/metabolismo , Electrofisiología , Femenino , Citometría de Flujo , Humanos , Proteína 2 de la Membrana Asociada a los Lisosomas/metabolismo , Masculino , Ratones , Ratones Noqueados , MicroARNs/genética , Fagocitosis/genética , Fagosomas/fisiología , Retina/fisiología , Epitelio Pigmentado de la Retina/fisiología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
2.
J Neurosci Res ; 98(8): 1646-1661, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-32537945

RESUMEN

Inhibitory pathways from Golgi tendon organs project widely between muscles crossing different joints and axes of rotation. Evidence suggests that the strength and distribution of this intermuscular inhibition is dependent on motor task and corresponding signals from the brainstem. The purpose of the present study was to investigate whether this sensory network is altered after spinal cord hemisection as a potential explanation for motor deficits observed after spinal cord injury (SCI). Force feedback was assessed between the long toe flexor and ankle plantarflexor (flexor hallucis longus), and the three major ankle extensors, (combined gastrocnemius, soleus, and plantaris muscles) in the hind limbs of unanesthetized, decerebrate, female cats. Data were collected from animals with intact spinal cords (control) and lateral spinal hemisections (LSHs) including chronic LSH (4-20 weeks), subchronic LSH (2 weeks), and acute LSH. Muscles were stretched individually and in pairwise combinations to measure intermuscular feedback between the toe flexor and each of the ankle extensors. In control animals, three patterns were observed (balanced inhibition between toe flexor and ankle extensors, stronger inhibition from toe flexor to ankle extensor, and vice versa). Following spinal hemisection, only strong inhibition from toe flexors onto ankle extensors was observed independent of survival time. The results suggest immediate and permanent reorganization of force feedback in the injured spinal cord. The altered strength and distribution of force feedback after SCI may be an important future target for rehabilitation.


Asunto(s)
Tobillo , Músculo Esquelético/fisiología , Traumatismos de la Médula Espinal , Dedos del Pie , Animales , Gatos , Femenino , Miembro Posterior , Reflejo/fisiología , Médula Espinal
3.
Adv Exp Med Biol ; 1074: 633-640, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29721997

RESUMEN

Previous work suggests that replacing diseased Retinal Pigment Epithelium (RPE) with a healthy autologous RPE sheet can provide vision rescue for AMD patients. We differentiated iPSCs into RPE using a directed differentiation protocol. RPE cells at the immature RPE stage were purified and seeded onto either electrospun poly(lactic-co-glycolic acid) (PLGA) scaffolds or non-biodegradable polyester cell culture inserts and compared the two tissues. In vitro, PLGA and polyester substrates produced functionally similar results. Following in vitro evaluation, we tested RPE tissue in animal models for safety and function. Safety studies were conducted in RNU rats using an injection composed of intact cells and homogenized scaffolds. To assess function and develop surgical procedures, the tissues were implanted into an acute RPE injury model pig eye and evaluated using optical coherence tomography (OCT), multifocal ERG (mfERG), and histology. Subretinal injection studies in rats demonstrated safety of the implant. Biodegradability and biocompatibility data from a pig model demonstrated that PLGA scaffold is safe, with the added benefit of being resorbed by the body over time, leaving no foreign material in the eye. We confirmed that biodegradable substrates provide suitable support for RPE maturation and transplantation.


Asunto(s)
Células Epiteliales/trasplante , Células Madre Pluripotentes Inducidas/trasplante , Degeneración Macular/terapia , Epitelio Pigmentado de la Retina/citología , Animales , Células Madre Pluripotentes Inducidas/citología , Células Madre Pluripotentes Inducidas/patología , Modelos Animales , Ratas , Ratas Desnudas , Reproducibilidad de los Resultados , Trasplante de Células Madre/efectos adversos , Porcinos , Teratoma/etiología
4.
Hum Mol Genet ; 20(14): 2770-82, 2011 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-21515588

RESUMEN

Recent studies have implicated an N-terminal caspase-6 cleavage product of mutant huntingtin (htt) as an important mediator of toxicity in Huntington's disease (HD). To directly assess the consequences of such fragments on neurologic function, we produced transgenic mice that express a caspase-6 length N-terminal fragment of mutant htt (N586) with both normal (23Q) and disease (82Q) length glutamine repeats. In contrast to mice expressing N586-23Q, mice expressing N586-82Q accumulate large cytoplasmic inclusion bodies that can be visualized with antibodies to epitopes throughout the N586 protein. However, biochemical analyses of aggregated mutant huntingtin in these mice demonstrated that the inclusion bodies are composed largely of a much smaller htt fragment (terminating before residue 115), with lesser amounts of full-length N586-82Q fragments. Mice expressing the N586-82Q fragment show symptoms typical of previously generated mice expressing mutant huntingtin fragments, including failure to maintain weight, small brain weight and reductions in specific mRNAs in the striatum. Uniquely, these N586-82Q mice develop a progressive movement disorder that includes dramatic deficits in motor performance on the rotarod and ataxia. Our findings suggest that caspase-6-derived fragments of mutant htt are capable of inducing novel HD-related phenotypes, but these fragments are not terminal cleavage products as they are subject to further proteolysis. In this scenario, mutant htt fragments derived from caspase 6, or possibly other proteases, could mediate HD pathogenesis via a 'hit and run' type of mechanism in which caspase-6, or other larger N-terminal fragments, mediate a neurotoxic process before being cleaved to a smaller fragment that accumulates pathologically.


Asunto(s)
Cuerpo Estriado/metabolismo , Expresión Génica , Enfermedad de Huntington/metabolismo , Cuerpos de Inclusión/metabolismo , Mutación Missense , Proteínas del Tejido Nervioso/biosíntesis , Proteínas Nucleares/biosíntesis , Sustitución de Aminoácidos , Animales , Ataxia/genética , Ataxia/metabolismo , Ataxia/patología , Caspasa 6 , Cuerpo Estriado/patología , Humanos , Proteína Huntingtina , Enfermedad de Huntington/genética , Enfermedad de Huntington/patología , Cuerpos de Inclusión/patología , Ratones , Ratones Transgénicos , Proteínas del Tejido Nervioso/genética , Proteínas Nucleares/genética , ARN Mensajero/biosíntesis , ARN Mensajero/genética , Prueba de Desempeño de Rotación con Aceleración Constante
5.
Nat Commun ; 12(1): 7293, 2021 12 15.
Artículo en Inglés | MEDLINE | ID: mdl-34911940

RESUMEN

Age-related Macular Degeneration (AMD), a blinding eye disease, is characterized by pathological protein- and lipid-rich drusen deposits underneath the retinal pigment epithelium (RPE) and atrophy of the RPE monolayer in advanced disease stages - leading to photoreceptor cell death and vision loss. Currently, there are no drugs that stop drusen formation or RPE atrophy in AMD. Here we provide an iPSC-RPE AMD model that recapitulates drusen and RPE atrophy. Drusen deposition is dependent on AMD-risk-allele CFH(H/H) and anaphylatoxin triggered alternate complement signaling via the activation of NF-κB and downregulation of autophagy pathways. Through high-throughput screening we identify two drugs, L-745,870, a dopamine receptor antagonist, and aminocaproic acid, a protease inhibitor that reduce drusen deposits and restore RPE epithelial phenotype in anaphylatoxin challenged iPSC-RPE with or without the CFH(H/H) genotype. This comprehensive iPSC-RPE model replicates key AMD phenotypes, provides molecular insight into the role of CFH(H/H) risk-allele in AMD, and discovers two candidate drugs to treat AMD.


Asunto(s)
Ácido Aminocaproico/farmacología , Células Madre Pluripotentes Inducidas/efectos de los fármacos , Degeneración Macular/tratamiento farmacológico , Piridinas/farmacología , Pirroles/farmacología , Epitelio Pigmentado de la Retina/efectos de los fármacos , Alelos , Factor H de Complemento/genética , Factor H de Complemento/metabolismo , Evaluación Preclínica de Medicamentos , Humanos , Células Madre Pluripotentes Inducidas/metabolismo , Degeneración Macular/genética , Degeneración Macular/metabolismo , Modelos Biológicos , Fenotipo , Epitelio Pigmentado de la Retina/metabolismo
6.
Mol Ther ; 17(11): 1857-67, 2009 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-19707186

RESUMEN

Glial cell line-derived neurotrophic factor (GDNF) gene transfer is being developed as a treatment for Parkinson's disease (PD). Due to the potential for side effects, external transgene regulation should enhance this strategy's safety profile. Here, we demonstrate dynamic control during long-term expression of GDNF using a recombinant adeno-associated virus (rAAV)-based bicistronic tetracycline (tet)-off construct. Nigrostriatal GDNF overexpression induces body weight alterations in rodents, enabling longitudinal in vivo tracking of GDNF expression after nigral vector delivery. Regulated GDNF expression was highly sensitive to dietary doxycycline (DOX), displaying undetectable striatal GDNF levels at serum DOX levels below those required for antimicrobial activity. However, in the absence of DOX, striatal GDNF levels exceeded levels required for efficacy in PD models. We also demonstrate the absence of a series of known GDNF-associated side effects when using direct intrastriatal vector delivery. Therefore, this single rAAV vector system meets most of the requirements for an experimental reagent for treatment of PD.


Asunto(s)
Dependovirus/genética , Doxiciclina/farmacología , Factor Neurotrófico Derivado de la Línea Celular Glial/metabolismo , Animales , Peso Corporal/efectos de los fármacos , Peso Corporal/genética , Cromatografía Líquida de Alta Presión , Densitometría , Doxiciclina/sangre , Ensayo de Inmunoadsorción Enzimática , Regulación de la Expresión Génica/efectos de los fármacos , Vectores Genéticos/genética , Factor Neurotrófico Derivado de la Línea Celular Glial/genética , Inmunohistoquímica , Masculino , Ratas , Ratas Sprague-Dawley , Tetraciclina/farmacología
7.
Mol Ther ; 17(6): 980-91, 2009 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-19277011

RESUMEN

Intraventricular administration of glial cell line-derived neurotrophic factor (GDNF) in primate and humans to study Parkinson's disease (PD) has revealed the potential for GDNF to induce weight loss. Our previous data indicate that bilateral continuous hypothalamic GDNF overexpression via recombinant adeno-associated virus (rAAV) results in significant failure to gain weight in young rats and weight loss in aged rats. Based on these previous results, we hypothesized that because the nigrostriatal tract passes through the lateral hypothalamus, motor hyperactivity mediated by nigrostriatal dopamine (DA) may have been responsible for the previously observed effect on body weight. In this study, we compared bilateral injections of rAAV2/5-GDNF in hypothalamus versus substantia nigra (SN) in aged Brown-Norway X Fisher 344 rats. Nigrostriatal GDNF overexpression resulted in significantly greater weight loss than rats treated in hypothalamus. The nigral or hypothalamic GDNF-induced weight loss was unrelated to motor activity levels of the rats, though some of the weight loss could be attributed to a transient reduction in food intake. Forebrain DA levels did not account for the observed effects on body weight, although GDNF-induced increases in nucleus accumbens DA may have partially contributed to this effect in the hypothalamic GDNF-treated group. However, only nigrostriatal GDNF overexpression induced activation of phosphorylated extracellular signal-regulated kinase (p-ERK) in a small population of corticotrophin-releasing factor [corticotrophin-releasing hormone (CRH)] neurons located specifically in the medial parvocellullar division (MPD) of the paraventricular nucleus of the hypothalamus. Activation of these hypothalamic CRH neurons likely accounted for the observed metabolic effects leading to weight loss in obese rats.


Asunto(s)
Envejecimiento/fisiología , Factor Neurotrófico Derivado de la Línea Celular Glial/fisiología , Obesidad/genética , Pérdida de Peso/genética , Adiposidad/genética , Animales , Western Blotting , Peso Corporal/genética , Catecolaminas/metabolismo , Cromatografía Líquida de Alta Presión , Dependovirus/genética , Dopamina/metabolismo , Ingestión de Alimentos/genética , Ensayo de Inmunoadsorción Enzimática , Factor Neurotrófico Derivado de la Línea Celular Glial/genética , Factor Neurotrófico Derivado de la Línea Celular Glial/metabolismo , Hipotálamo/metabolismo , Inmunohistoquímica , Masculino , Neuropéptido Y/metabolismo , Obesidad/metabolismo , Ratas , Ratas Endogámicas F344 , Ratas Sprague-Dawley , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Sustancia Negra/metabolismo
8.
Sci Transl Med ; 11(475)2019 01 16.
Artículo en Inglés | MEDLINE | ID: mdl-30651323

RESUMEN

Considerable progress has been made in testing stem cell-derived retinal pigment epithelium (RPE) as a potential therapy for age-related macular degeneration (AMD). However, the recent reports of oncogenic mutations in induced pluripotent stem cells (iPSCs) underlie the need for robust manufacturing and functional validation of clinical-grade iPSC-derived RPE before transplantation. Here, we developed oncogenic mutation-free clinical-grade iPSCs from three AMD patients and differentiated them into clinical-grade iPSC-RPE patches on biodegradable scaffolds. Functional validation of clinical-grade iPSC-RPE patches revealed specific features that distinguished transplantable from nontransplantable patches. Compared to RPE cells in suspension, our biodegradable scaffold approach improved integration and functionality of RPE patches in rats and in a porcine laser-induced RPE injury model that mimics AMD-like eye conditions. Our results suggest that the in vitro and in vivo preclinical functional validation of iPSC-RPE patches developed here might ultimately be useful for evaluation and optimization of autologous iPSC-based therapies.


Asunto(s)
Degeneración Retiniana/terapia , Epitelio Pigmentado de la Retina/citología , Células Madre/citología , Animales , Modelos Animales de Enfermedad , Degeneración Macular/patología , Degeneración Macular/terapia , Ratas , Degeneración Retiniana/patología , Porcinos
10.
Exp Neurol ; 228(2): 173-82, 2011 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-21192926

RESUMEN

The discovery of the gene mutation responsible for Huntington's disease (HD), huntingtin, in 1993 allowed for a better understanding of the pathology of and enabled the development of animal models. HD is caused by the expansion of a polyglutamine repeat region in the N-terminal of the huntingtin protein. Here we examine the behavioral, transcriptional, histopathological and anatomical characteristics of a knock-in HD mouse model with a 140 polyglutamine expansion in the huntingtin protein. This CAG 140 model contains a portion of the human exon 1 with 140 CAG repeats knocked into the mouse huntingtin gene. We have longitudinally examined the rearing behavior, accelerating rotarod, constant speed rotarod and gait for age-matched heterozygote, homozygote and non-transgenic mice and have found a significant difference in the afflicted mice. However, while there were significant differences between the non-transgenic and the knock-in mice, these behaviors were not progressive. As in HD, we show that the CAG 140 mice also have a significant decrease in striatally enriched mRNA transcripts. In addition, striatal neuronal intranuclear inclusion density increases with age. Lastly these CAG 140 mice show slight cortical thinning compared to non-transgenic mice, similarly to the cortical thinning recently reported in HD.


Asunto(s)
Conducta Animal/fisiología , Técnicas de Sustitución del Gen/métodos , Enfermedad de Huntington/genética , Enfermedad de Huntington/patología , Proteínas del Tejido Nervioso/genética , Proteínas Nucleares/genética , Expansión de Repetición de Trinucleótido/genética , Animales , Estudios Transversales , Modelos Animales de Enfermedad , Femenino , Marcha/genética , Tamización de Portadores Genéticos/métodos , Homocigoto , Humanos , Proteína Huntingtina , Enfermedad de Huntington/psicología , Estudios Longitudinales , Masculino , Ratones , Ratones Endogámicos C57BL , Actividad Motora/genética , Prueba de Desempeño de Rotación con Aceleración Constante
11.
Mol Ther ; 13(3): 463-83, 2006 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-16412695

RESUMEN

Recombinant adeno-associated virus (rAAV) is derived from a small human parvovirus with an excellent safety profile. In addition, this viral vector efficiently transduces and supports long-term transgene expression in the nervous system. These properties make rAAV a reasonable candidate vector for treating neurological disorders. Indeed, rAAV is currently being used in five early stage clinical trials for various neurodegenerative disorders. Therefore, we will review the currently available preclinical data using rAAV in animal models of central nervous system (CNS) disorders. Moreover, potential caveats for rAAV-based gene therapy in the CNS are also presented.


Asunto(s)
ADN Recombinante/uso terapéutico , Dependovirus/genética , Terapia Genética/métodos , Vectores Genéticos/uso terapéutico , Enfermedades Neurodegenerativas/terapia , Animales , Terapia Genética/tendencias , Humanos , Enfermedades Neurodegenerativas/genética , Enfermedades Neurodegenerativas/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA