Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 62
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Mol Cell ; 75(1): 154-171.e5, 2019 07 11.
Artículo en Inglés | MEDLINE | ID: mdl-31056445

RESUMEN

The epigenetic information present in mammalian gametes and whether it is transmitted to the progeny are relatively unknown. We find that many promoters in mouse sperm are occupied by RNA polymerase II (Pol II) and Mediator. The same promoters are accessible in GV and MII oocytes and preimplantation embryos. Sperm distal ATAC-seq sites containing motifs for various transcription factors are conserved in monkeys and humans. ChIP-seq analyses confirm that Foxa1, ERα, and AR occupy distal enhancers in sperm. Accessible sperm enhancers containing H3.3 and H2A.Z are also accessible in oocytes and preimplantation embryos. Furthermore, their interactions with promoters in the gametes persist during early development. Sperm- or oocyte-specific interactions mediated by CTCF and cohesin are only present in the paternal or maternal chromosomes, respectively, in the zygote and 2-cell stages. These interactions converge in both chromosomes by the 8-cell stage. Thus, mammalian gametes contain complex patterns of 3D interactions that can be transmitted to the zygote after fertilization.


Asunto(s)
Factor de Unión a CCCTC/genética , Factor Nuclear 3-beta del Hepatocito/genética , Oocitos/metabolismo , Espermatozoides/metabolismo , Cigoto/metabolismo , Animales , Secuencia de Bases , Factor de Unión a CCCTC/metabolismo , Cromatina/química , Cromatina/metabolismo , Secuencia Conservada , Embrión de Mamíferos , Desarrollo Embrionario/genética , Elementos de Facilitación Genéticos , Receptor alfa de Estrógeno/genética , Receptor alfa de Estrógeno/metabolismo , Receptor beta de Estrógeno/genética , Receptor beta de Estrógeno/metabolismo , Femenino , Regulación del Desarrollo de la Expresión Génica , Factor Nuclear 3-beta del Hepatocito/metabolismo , Humanos , Macaca mulatta , Masculino , Ratones , Oocitos/citología , Oocitos/crecimiento & desarrollo , Receptores Androgénicos/genética , Receptores Androgénicos/metabolismo , Homología de Secuencia de Ácido Nucleico , Espermatozoides/citología , Espermatozoides/crecimiento & desarrollo , Dedos de Zinc/genética , Cigoto/citología , Cigoto/crecimiento & desarrollo
2.
J Assist Reprod Genet ; 39(10): 2413-2430, 2022 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-36066723

RESUMEN

Trinucleotide repeats (TNRs) are dispersed throughout the human genome. About 20 loci are related to human diseases, such as Huntington's disease (HD). A larger TNR instability is predominantly observed in the paternal germ cells in some TNR disorders. Suppressing the expansion during spermatogenesis can provide a unique opportunity to end the vicious cycle of genetic anticipation. Here, using an in vitro differentiation method to derive advanced spermatogenic cells, we investigated the efficacy of two therapeutic agents, araC (cytarabine) and aspirin, on stabilizing TNRs in spermatogenic cells. Two WT patient-derived induced pluripotent stem cell (iPSC) lines and two HD hiPSC lines, with 44 Q and 180 Q, were differentiated into spermatogonial stem cell-like cells (SSCLCs). Both HD cell lines showed CAG tract expansion in SSCLC. When treated with araC and aspirin, HD1 showed moderate but not statistically significant stabilization of TNR. In HD2, 10 nM of aspirin and araC showed significant stabilization of TNR. All cell lines showed increased DNA damage response (DDR) gene expression in SSCLCs while more genes were significantly induced in HD SSCLC. In HD1, araC and aspirin treatment showed general suppression of DNA damage response genes. In HD2, only FAN1, OGG1, and PCNA showed significant suppression. When the methylation profile of HD cells was analyzed, FAN1 and OGG1 showed significant hypermethylation after the aspirin and araC treatment in SSCLC compared to the control. This study underscores the utility of our in vitro spermatogenesis model to study and develop therapies for TNR disorders such as HD.


Asunto(s)
Enfermedad de Huntington , Expansión de Repetición de Trinucleótido , Masculino , Humanos , Expansión de Repetición de Trinucleótido/genética , Enfermedad de Huntington/tratamiento farmacológico , Enfermedad de Huntington/genética , Antígeno Nuclear de Célula en Proliferación/genética , Repeticiones de Trinucleótidos/genética , Células Germinativas , Citarabina , Aspirina
3.
J Med Primatol ; 50(1): 75-78, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-33277721

RESUMEN

Amitriptyline is a tricyclic antidepressant commonly prescribed in humans for pain and sleep disorders and in non-human primates for self-injurious behaviors. Here, we report a clinical case on the teratogenic effect of maternal-fetal amitriptyline exposure.


Asunto(s)
Amitriptilina/efectos adversos , Antidepresivos Tricíclicos/efectos adversos , Macaca mulatta/anomalías , Teratogénesis , Teratógenos , Animales , Femenino , Exposición Materna
4.
J Assist Reprod Genet ; 38(5): 1215-1229, 2021 May.
Artículo en Inglés | MEDLINE | ID: mdl-33611676

RESUMEN

PURPOSE: The expansion of CAG (glutamine; Q) trinucleotide repeats (TNRs) predominantly occurs through male lineage in Huntington's disease (HD). As a result, offspring will have larger CAG repeats compared to their fathers, which causes an earlier onset of the disease called genetic anticipation. This study aims to develop a novel in vitro model to replicate CAG repeat instability in early spermatogenesis and demonstrate the biological process of genetic anticipation by using the HD stem cell model for the first time. METHODS: HD rhesus monkey embryonic stem cells (rESCs) were cultured in vitro for an extended period. Male rESCs were used to derive spermatogenic cells in vitro with a 10-day differentiation. The assessment of CAG repeat instability was performed by GeneScan and curve fit analysis. RESULTS: Spermatogenic cells derived from rESCs exhibit progressive expansion of CAG repeats with high daily expansion rates compared to the extended culture of rESCs. The expansion of CAG repeats is cell type-specific and size-dependent. CONCLUSIONS: Here, we report a novel stem cell model that replicates genome instability and CAG repeat expansion in in vitro derived HD monkey spermatogenic cells. The in vitro spermatogenic cell model opens a new opportunity for studying TNR instability and the underlying mechanism of genetic anticipation, not only in HD but also in other TNR diseases.


Asunto(s)
Células Madre Germinales Adultas/patología , Animales Modificados Genéticamente/genética , Células Madre Embrionarias/patología , Enfermedad de Huntington/genética , Animales , Diferenciación Celular/genética , Modelos Animales de Enfermedad , Inestabilidad Genómica/genética , Humanos , Enfermedad de Huntington/patología , Macaca mulatta/genética , Masculino , Inestabilidad de Microsatélites , Repeticiones de Trinucleótidos/genética
5.
Neurobiol Dis ; 146: 105085, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-32950644

RESUMEN

Fused in sarcoma (FUS) is a RNA/DNA protein involved in multiple nuclear and cytoplasmic functions including transcription, splicing, mRNA trafficking, and stress granule formation. To accomplish these many functions, FUS must shuttle between cellular compartments in a highly regulated manner. When shuttling is disrupted, FUS abnormally accumulates into cytoplasmic inclusions that can be toxic. Disrupted shuttling of FUS into the nucleus is a hallmark of ~10% of frontotemporal lobar degeneration (FTLD) cases, the neuropathology that underlies frontotemporal dementia (FTD). Multiple pathways are known to disrupt nuclear/cytoplasmic shuttling of FUS. In earlier work, we discovered that double-strand DNA breaks (DSBs) trigger DNA-dependent protein kinase (DNA-PK) to phosphorylate FUS (p-FUS) at N-terminal residues leading to the cytoplasmic accumulation of FUS. Therefore, DNA damage may contribute to the development of FTLD pathology with FUS inclusions. In the present study, we examined how DSBs effect FUS phosphorylation in various primate and mouse cellular models. All cell lines derived from human and non-human primates exhibit N-terminal FUS phosphorylation following calicheamicin γ1 (CLM) induced DSBs. In contrast, we were unable to detect FUS phosphorylation in mouse-derived primary neurons or immortalized cell lines regardless of CLM treatment, duration, or concentration. Despite DNA damage induced by CLM treatment, we find that mouse cells do not phosphorylate FUS, likely due to reduced levels and activity of DNA-PK compared to human cells. Taken together, our work reveals that mouse-derived cellular models regulate FUS in an anomalous manner compared to primate cells. This raises the possibility that mouse models may not fully recapitulate the pathogenic cascades that lead to FTLD with FUS pathology.


Asunto(s)
Encéfalo/metabolismo , Daño del ADN/fisiología , ADN/metabolismo , Degeneración Lobar Frontotemporal/metabolismo , Proteína FUS de Unión a ARN/genética , Animales , Degeneración Lobar Frontotemporal/genética , Humanos , Cuerpos de Inclusión/genética , Cuerpos de Inclusión/metabolismo , Ratones , Mutación/genética , Neuronas/metabolismo , Fosforilación , Factores Asociados con la Proteína de Unión a TATA/genética
6.
J Neural Transm (Vienna) ; 125(3): 401-417, 2018 03.
Artículo en Inglés | MEDLINE | ID: mdl-29127484

RESUMEN

Huntington's disease (HD) is a complex neurodegenerative disorder that has no cure. Although treatments can often be given to relieve symptoms, the neuropathology associated with HD cannot be stopped or reversed. HD is characterized by degeneration of the striatum and associated pathways that leads to impairment in motor and cognitive functions as well as psychiatric disturbances. Although cell and rodent models for HD exist, longitudinal study in a transgenic HD nonhuman primate (i.e., rhesus macaque; HD monkeys) shows high similarity in its progression with human patients. Progressive brain atrophy and changes in white matter integrity examined by magnetic resonance imaging are coherent with the decline in cognitive behaviors related to corticostriatal functions and neuropathology. HD monkeys also express higher anxiety and irritability/aggression similar to human HD patients that other model systems have not yet replicated. While a comparative model approach is critical for advancing our understanding of HD pathogenesis, HD monkeys could provide a unique platform for preclinical studies and long-term assessment of translatable outcome measures. This review summarizes the progress in the development of the transgenic HD monkey model and the opportunities for advancing HD preclinical research.


Asunto(s)
Encéfalo/patología , Proteína Huntingtina/genética , Enfermedad de Huntington/genética , Animales , Animales Modificados Genéticamente , Encéfalo/metabolismo , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Enfermedad de Huntington/patología , Macaca mulatta
7.
Am J Hum Genet ; 93(2): 306-12, 2013 Aug 08.
Artículo en Inglés | MEDLINE | ID: mdl-23810380

RESUMEN

Huntington disease (HD) is a dominantly inherited neurodegenerative disorder characterized by dysregulation of various genes. Recently, microRNAs (miRNAs) have been reported to be involved in this dysregulation, suggesting that manipulation of appropriate miRNA regulation may have a therapeutic benefit. Here, we report the beneficial effects of miR-196a (miR196a) on HD in cell, transgenic mouse models, and human induced pluripotent stem cells derived from one individual with HD (HD-iPSCs). In the in vitro results, a reduction of mutant HTT and pathological aggregates, accompanying the overexpression of miR-196a, was observed in HD models of human embryonic kidney cells and mouse neuroblastoma cells. In the in vivo model, HD transgenic mice overexpressing miR-196a revealed the suppression of mutant HTT in the brain and also showed improvements in neuropathological progression, such as decreases of nuclear, intranuclear, and neuropil aggregates and late-stage behavioral phenotypes. Most importantly, miR-196a also decreased HTT expression and pathological aggregates when HD-iPSCs were differentiated into the neuronal stage. Mechanisms of miR-196a in HD might be through the alteration of ubiquitin-proteasome systems, gliosis, cAMP response element-binding protein pathway, and several neuronal regulatory pathways in vivo. Taken together, these results show that manipulating miR-196a provides beneficial effects in HD, suggesting the potential therapeutical role of miR-196a in HD.


Asunto(s)
Enfermedad de Huntington/genética , MicroARNs/genética , Neuronas/metabolismo , Células Madre Pluripotentes/metabolismo , Proteínas de Transporte de Serotonina en la Membrana Plasmática/genética , Animales , Encéfalo/metabolismo , Encéfalo/patología , Diferenciación Celular , Línea Celular Tumoral , Modelos Animales de Enfermedad , Embrión de Mamíferos , Regulación de la Expresión Génica , Humanos , Enfermedad de Huntington/metabolismo , Enfermedad de Huntington/patología , Riñón/metabolismo , Riñón/patología , Ratones , Ratones Transgénicos , MicroARNs/metabolismo , Neuronas/citología , Fenotipo , Células Madre Pluripotentes/citología , Proteínas de Transporte de Serotonina en la Membrana Plasmática/metabolismo , Transfección
8.
Brain Behav Immun ; 58: 181-190, 2016 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-27395434

RESUMEN

Although the most notable clinical symptoms of Huntington's disease (HD) are motor disturbances and brain atrophy, other symptoms include cognitive dysfunction, emotional and hormonal dysregulation. Emotional dysregulation (irritability, anger/aggression, and anxiety) and increased inflammation are early emerging symptoms which can be detected decades before the onset of motor symptoms in HD patients. Despite the advances in understanding the genetic causes of HD there is still no cure or preventative treatment. Thus, to better understand the pathogenesis of HD and develop effective treatments, a holistic understanding of HD is needed, as well as animal models that replicate the full spectrum of HD symptoms. The current study examined the emotional, hormonal, and gene expression responses to an acute stressor of adult male transgenic HD rhesus monkeys (n=2) as compared to wild-type controls (n=2). Results revealed that HD monkeys expressed increased anxiety and irritability/aggression as compared to controls. Reactive cortisol response to the stressor was similar between groups. However, HD monkeys exhibited increased pro-inflammatory cytokines and higher induction of immune pathway genes as compared to controls. Overall, results reveal that HD monkeys exhibit these early emerging symptoms of HD and may be an effective animal model to facilitate the development of new therapeutics for HD patients.


Asunto(s)
Ansiedad , Conducta Animal , Enfermedad de Huntington/inmunología , Enfermedad de Huntington/psicología , Agresión , Animales , Animales Modificados Genéticamente , Proteína C-Reactiva/metabolismo , Modelos Animales de Enfermedad , Expresión Génica , Enfermedad de Huntington/genética , Hidrocortisona/sangre , Interleucina-6/sangre , Leucocitos Mononucleares/metabolismo , Macaca mulatta , Masculino , Estrés Fisiológico , Factor de Necrosis Tumoral alfa/sangre
9.
Nature ; 453(7197): 921-4, 2008 Jun 12.
Artículo en Inglés | MEDLINE | ID: mdl-18488016

RESUMEN

Non-human primates are valuable for modelling human disorders and for developing therapeutic strategies; however, little work has been reported in establishing transgenic non-human primate models of human diseases. Huntington's disease (HD) is an autosomal dominant neurodegenerative disorder characterized by motor impairment, cognitive deterioration and psychiatric disturbances followed by death within 10-15 years of the onset of the symptoms. HD is caused by the expansion of cytosine-adenine-guanine (CAG, translated into glutamine) trinucleotide repeats in the first exon of the human huntingtin (HTT) gene. Mutant HTT with expanded polyglutamine (polyQ) is widely expressed in the brain and peripheral tissues, but causes selective neurodegeneration that is most prominent in the striatum and cortex of the brain. Although rodent models of HD have been developed, these models do not satisfactorily parallel the brain changes and behavioural features observed in HD patients. Because of the close physiological, neurological and genetic similarities between humans and higher primates, monkeys can serve as very useful models for understanding human physiology and diseases. Here we report our progress in developing a transgenic model of HD in a rhesus macaque that expresses polyglutamine-expanded HTT. Hallmark features of HD, including nuclear inclusions and neuropil aggregates, were observed in the brains of the HD transgenic monkeys. Additionally, the transgenic monkeys showed important clinical features of HD, including dystonia and chorea. A transgenic HD monkey model may open the way to understanding the underlying biology of HD better, and to the development of potential therapies. Moreover, our data suggest that it will be feasible to generate valuable non-human primate models of HD and possibly other human genetic diseases.


Asunto(s)
Modelos Animales de Enfermedad , Enfermedad de Huntington/genética , Enfermedad de Huntington/fisiopatología , Macaca mulatta/genética , Proteínas del Tejido Nervioso/genética , Proteínas Nucleares/genética , Expansión de Repetición de Trinucleótido/genética , Animales , Animales Modificados Genéticamente , Animales Recién Nacidos , Encéfalo/metabolismo , Encéfalo/patología , Corea/genética , Corea/fisiopatología , Distonía/genética , Distonía/fisiopatología , Exones/genética , Femenino , Humanos , Proteína Huntingtina , Enfermedad de Huntington/metabolismo , Enfermedad de Huntington/patología , Masculino , Proteínas del Tejido Nervioso/metabolismo , Proteínas Nucleares/metabolismo , Péptidos/genética , Péptidos/metabolismo , Embarazo , Análisis de Supervivencia
10.
BMC Neurosci ; 14: 88, 2013 Aug 17.
Artículo en Inglés | MEDLINE | ID: mdl-23957861

RESUMEN

BACKGROUND: Huntington's Disease (HD) is a progressive neurodegenerative disorder caused by an expansion in the polyglutamine (polyQ) region of the Huntingtin (HTT) gene. The clinical features of HD are characterized by cognitive, psychological, and motor deficits. Molecular instability, a core component in neurological disease progression, can be comprehensively evaluated through longitudinal transcriptomic profiling. Development of animal models amenable to longitudinal examination enables distinct disease-associated mechanisms to be identified. RESULTS: Here we report the first longitudinal study of transgenic monkeys with genomic integration of various lengths of the human HTT gene and a range of polyQ repeats. With this unique group of transgenic HD nonhuman primates (HD monkeys), we profiled over 47,000 transcripts from peripheral blood collected over a 2 year timespan from HD monkeys and age-matched wild-type control monkeys. CONCLUSIONS: Messenger RNAs with expression patterns which diverged with disease progression in the HD monkeys considerably facilitated our search for transcripts with diagnostic or therapeutic potential in the blood of human HD patients, opening up a new avenue for clinical investigation.


Asunto(s)
Enfermedad de Huntington/genética , Proteínas del Tejido Nervioso/genética , Transcriptoma , Animales , Animales Modificados Genéticamente , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Femenino , Perfilación de la Expresión Génica , Humanos , Proteína Huntingtina , Enfermedad de Huntington/sangre , Estudios Longitudinales , Macaca mulatta , Masculino , ARN Mensajero/metabolismo
11.
J Assist Reprod Genet ; 30(1): 81-9, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-23224695

RESUMEN

PURPOSE: To investigate whether in vitro fertilization (IVF) and intracytoplasmic sperm injection (ICSI), influence the embryo's development and its quality using the mouse as a model. METHODS: Assisted fertilization was performed using ICSI and IVF. Fluorescent beads were adhered to the fertilization cone or place of previous sperm injection in the natural mated (NM), IVF and ICSI embryos, respectively. Embryo examination was carried out at the two-cell and blastocyst stage to determine the position of fluorescent bead. Protein expression was detected by fluorescence immunocytochemical staining and confocal microscopic imaging of blastocysts. RESULTS: IVF and ICSI embryos developed at rates comparable to NM group. Embryos show similar expression patterns of two transcription factors, Oct4 and Cdx2. The most preferred place for spermatozoa attachment was the equatorial site of the egg, whether fertilization occurred in vitro or under natural conditions. We also link the sperm entry position (SEP) to embryo morphology and the number of cells at the blastocyst stage, with no influence of the method of fertilization. CONCLUSIONS: IVF and ICSI, do not compromise in vitro pre-implantation development. Additional data, related to sperm entry, could offer further criteria to predict embryos that will implant successfully. Based on embryo morphology, developmental rate and protein expression level of key transcription factors, our results support the view that ART techniques, such as IVF and ICSI, do not perturb embryonic development or quality.


Asunto(s)
Blastocisto/fisiología , Desarrollo Embrionario , Fertilización In Vitro/métodos , Inyecciones de Esperma Intracitoplasmáticas/métodos , Espermatozoides/fisiología , Animales , Factor de Transcripción CDX2 , Técnicas de Cultivo de Embriones/métodos , Femenino , Fertilización , Colorantes Fluorescentes , Regulación del Desarrollo de la Expresión Génica , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Inmunohistoquímica , Técnicas de Maduración In Vitro de los Oocitos , Masculino , Ratones , Modelos Animales , Factor 3 de Transcripción de Unión a Octámeros/genética , Factor 3 de Transcripción de Unión a Octámeros/metabolismo , Recuperación del Oocito , Oocitos/citología , Oocitos/fisiología , Imagen Óptica , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
12.
Biomedicines ; 10(8)2022 Aug 02.
Artículo en Inglés | MEDLINE | ID: mdl-36009409

RESUMEN

Huntington's Disease (HD) is an autosomal dominant disease that results in severe neurodegeneration with no cure. HD is caused by the expanded CAG trinucleotide repeat (TNR) on the Huntingtin gene (HTT). Although the somatic and germline expansion of the CAG repeats has been well-documented, the underlying mechanisms had not been fully delineated. Increased CAG repeat length is associated with a more severe phenotype, greater TNR instability, and earlier age of onset. The direct relationship between CAG repeat length and molecular pathogenesis makes TNR instability a useful measure of symptom severity and tissue susceptibility. Thus, we examined the tissue-specific TNR instability of transgenic nonhuman primate models of Huntington's disease. Our data show a similar profile of CAG repeat expansion in both rHD1 and rHD7, where high instability was observed in testis, liver, caudate, and putamen. CAG repeat expansion was observed in all tissue samples, and tissue- and CAG repeat size-dependent expansion was observed. Correlation analysis of CAG repeat expansion and the gene expression profile of four genes in different tissues, clusterin (CLU), transferrin (TF), ribosomal protein lateral stalk subunit P1 (RPLP1), and ribosomal protein L13a (RPL13A), showed a strong correlation with CAG repeat instability. Overall, our data, along with previously published studies, can be used for studying the biology of CAG repeat instability and identifying new therapeutic targets.

13.
Aging Brain ; 22022.
Artículo en Inglés | MEDLINE | ID: mdl-36589695

RESUMEN

With the ultimate goal of developing a more representative animal model of Alzheimer's disease (AD), two female amyloid-ß-(Aß) precursor protein-transgenic (APPtg) rhesus monkeys were generated by lentiviral transduction of the APP gene into rhesus oocytes, followed by in vitro fertilization and embryo transfer. The APP-transgene included the AD-associated Swedish K670N/M671L and Indiana V717F mutations (APPSWE/IND) regulated by the human polyubiquitin-C promoter. Overexpression of APP was confirmed in lymphocytes and brain tissue. Upon sacrifice at 10 years of age, one of the monkeys had developed Aß plaques and cerebral Aß-amyloid angiopathy in the occipital, parietal, and caudal temporal neocortices. The induction of Aß deposition more than a decade prior to its usual emergence in the rhesus monkey supports the feasibility of creating a transgenic nonhuman primate model for mechanistic analyses and preclinical testing of treatments for Alzheimer's disease and cerebrovascular amyloidosis.

14.
BMC Cell Biol ; 12: 39, 2011 Sep 12.
Artículo en Inglés | MEDLINE | ID: mdl-21910887

RESUMEN

BACKGROUND: Dental pulp stem/stromal cells (DPSCs) are categorized as adult stem cells (ASCs) that retain multipotent differentiation capabilities. DPSCs can be isolated from individuals at any age and are considered to be true personal stem cells, making DPSCs one of the potential options for stem cell therapy. However, the properties of DPSCs from individuals with an inherited genetic disorder, such as Huntington's disease (HD), have not been fully investigated. RESULTS: To examine if mutant huntingtin (htt) protein impacts DPSC properties, we have established DPSCs from tooth germ of transgenic monkeys that expressed both mutant htt and green fluorescent protein (GFP) genes (rHD/G-DPSCs), and from a monkey that expressed only the GFP gene (rG-DPSCs), which served as a control. Although mutant htt and oligomeric htt aggregates were overtly present in rHD/G-DPSCs, all rHD/G-DPSCs and rG-DPSCs shared similar characteristics, including self-renewal, multipotent differentiation capabilities, expression of stemness and differentiation markers, and cell surface antigen profile. CONCLUSIONS: Our results suggest that DPSCs from Huntington monkeys retain ASC properties. Thus DPSCs derived from individuals with genetic disorders such as HD could be a potential source of personal stem cells for therapeutic purposes.


Asunto(s)
Células Madre Adultas/metabolismo , Enfermedad de Huntington/terapia , Proteínas de Transporte de Serotonina en la Membrana Plasmática/metabolismo , Trasplante de Células Madre , Células del Estroma/metabolismo , Células Madre Adultas/patología , Animales , Animales Modificados Genéticamente , Supervivencia Celular/genética , Células Cultivadas , Pulpa Dental/patología , Modelos Animales de Enfermedad , Haplorrinos , Humanos , Enfermedad de Huntington/genética , Enfermedad de Huntington/patología , Mutación/genética , Proteínas de Transporte de Serotonina en la Membrana Plasmática/genética , Células del Estroma/patología , Transgenes/genética
15.
BMC Cell Biol ; 11: 12, 2010 Feb 05.
Artículo en Inglés | MEDLINE | ID: mdl-20132560

RESUMEN

BACKGROUND: Pluripotent stem cells that are capable of differentiating into different cell types and develop robust hallmark cellular features are useful tools for clarifying the impact of developmental events on neurodegenerative diseases such as Huntington's disease. Additionally, a Huntington's cell model that develops robust pathological features of Huntington's disease would be valuable for drug discovery research. RESULTS: To test this hypothesis, a pluripotent Huntington's disease monkey hybrid cell line (TrES1) was established from a tetraploid Huntington's disease monkey blastocyst generated by the fusion of transgenic Huntington's monkey skin fibroblast and a wild-type non-transgenic monkey oocyte. The TrES1 developed key Huntington's disease cellular pathological features that paralleled neural development. It expressed mutant huntingtin and stem cell markers, was capable of differentiating to neural cells, and developed teratoma in severely compromised immune deficient (SCID) mice. Interestingly, the expression of mutant htt, the accumulation of oligomeric mutant htt and the formation of intranuclear inclusions paralleled neural development in vitro , and even mutant htt was ubiquitously expressed. This suggests the development of Huntington's disease cellular features is influenced by neural developmental events. CONCLUSIONS: Huntington's disease cellular features is influenced by neural developmental events. These results are the first to demonstrate that a pluripotent stem cell line is able to mimic Huntington's disease progression that parallels neural development, which could be a useful cell model for investigating the developmental impact on Huntington's disease pathogenesis.


Asunto(s)
Enfermedad de Huntington/patología , Células Madre/citología , Animales , Diferenciación Celular , Embrión de Mamíferos , Genotipo , Haplorrinos , Enfermedad de Huntington/metabolismo , Células Híbridas , Cariotipificación , Ratones , Proteínas Asociadas a Microtúbulos/metabolismo , Neuronas/citología
16.
Hum Mol Genet ; 17(17): 2738-51, 2008 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-18558632

RESUMEN

A number of mouse models expressing mutant huntingtin (htt) with an expanded polyglutamine (polyQ) domain are useful for studying the pathogenesis of Huntington's disease (HD) and identifying appropriate therapies. However, these models exhibit neurological phenotypes that differ in their severity and nature. Understanding how transgenic htt leads to variable neuropathology in animal models would shed light on the pathogenesis of HD and help us to choose HD models for investigation. By comparing the expression of mutant htt at the transcriptional and protein levels in transgenic mice expressing N-terminal or full-length mutant htt, we found that the accumulation and aggregation of mutant htt in the brain is determined by htt context. HD mouse models demonstrating more severe phenotypes show earlier accumulation of N-terminal mutant htt fragments, which leads to the formation of htt aggregates that are primarily present in neuronal nuclei and processes, as well as glial cells. Similarly, transgenic monkeys expressing exon-1 htt with a 147-glutamine repeat (147Q) died early and showed abundant neuropil aggregates in swelling neuronal processes. Fractionation of HD150Q knock-in mice brains revealed an age-dependent accumulation of N-terminal mutant htt fragments in the nucleus and synaptosomes, and this accumulation was most pronounced in the striatum due to decreased proteasomal activity. Our findings suggest that the neuropathological phenotypes of HD stem largely from the accumulation of N-terminal mutant htt fragments and that this accumulation is determined by htt context and cell-type-dependent clearance of mutant htt.


Asunto(s)
Enfermedad de Huntington/fisiopatología , Proteínas de Transporte de Serotonina en la Membrana Plasmática/genética , Proteínas de Transporte de Serotonina en la Membrana Plasmática/metabolismo , Animales , Animales Modificados Genéticamente , Encéfalo/fisiopatología , Haplorrinos , Humanos , Ratones , Ratones Transgénicos , Neuronas/metabolismo , Transcripción Genética
17.
Methods ; 49(1): 78-84, 2009 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-19467335

RESUMEN

Modeling human diseases using nonhuman primates including chimpanzee, rhesus, cynomolgus, marmoset and squirrel monkeys has been reported in the past decades. Due to the high similarity between nonhuman primates and humans, including genome constitution, cognitive behavioral functions, anatomical structure, metabolic, reproductive, and brain functions; nonhuman primates have played an important role in understanding physiological functions of the human body, clarifying the underlying mechanism of human diseases, and the development of novel treatments for human diseases. However, nonhuman primate research has been restricted to cognitive, behavioral, biochemical and pharmacological approaches of human diseases due to the limitation of gene transfer technology in nonhuman primates. The recent advancement in transgenic technology that has led to the generation of the first transgenic monkey in 2001 and a transgenic monkey model of Huntington's disease (HD) in 2008 has changed that focus. The creation of transgenic HD monkeys that replicate key pathological features of human HD patients further suggests the crucial role of nonhuman primates in the future development of biomedicine. These successes have opened the door to genetic manipulation in nonhuman primates and a new era in modeling human inherited genetic disorders. We focused on the procedures in creating transgenic Huntington's disease monkeys, but our work can be applied to transgenesis in other nonhuman primate species.


Asunto(s)
Animales Modificados Genéticamente , Modelos Animales de Enfermedad , Haplorrinos/genética , Enfermedad de Huntington/genética , Animales , Ingeniería Genética/métodos , Humanos
18.
Genes Brain Behav ; 19(4): e12638, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-31943801

RESUMEN

Salient sensory environments experienced by a parental generation can exert intergenerational influences on offspring. While these data provide an exciting new perspective on biological inheritance, questions remain about causes and consequences of intergenerational influences of salient sensory experience. We previously showed that exposing male mice to a salient olfactory experience, like olfactory fear conditioning, resulted in offspring demonstrating a sensitivity to the odor used to condition the paternal generation and possessing enhanced neuroanatomical representation for that odor. In this study, we first injected RNA extracted from sperm of male mice that underwent olfactory fear conditioning into naïve single-cell zygotes and found that adults that developed from these embryos had increased sensitivity and enhanced neuroanatomical representation for the odor (Odor A) with which the paternal male had been conditioned. Next, we found that female, but not male offspring sired by males conditioned with Odor A show enhanced consolidation of a weak single-trial Odor A + shock fear conditioning protocol. Our data provide evidence that RNA found in the paternal germline after exposure to salient sensory experiences can contribute to intergenerational influences of such experiences, and that such intergenerational influences confer an element of adaptation to the offspring. In so doing, our study of intergenerational influences of parental sensory experience adds to existing literature on intergenerational influences of parental exposures to stress and dietary manipulations and suggests that some causes (sperm RNA) and consequences (behavioral flexibility) of intergenerational influences of parental experiences may be conserved across a variety of parental experiences.


Asunto(s)
Percepción Olfatoria/genética , Animales , Femenino , Masculino , Ratones , Ratones Endogámicos C57BL , Linaje , ARN/genética , ARN/metabolismo , Espermatozoides/metabolismo , Cigoto/metabolismo
19.
Biol Reprod ; 81(6): 1189-95, 2009 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-19641177

RESUMEN

The study of alternative model organisms has yielded tremendous insights into the regulation of behavioral and physiological traits not displayed by more widely used animal models, such as laboratory rats and mice. In particular, comparative approaches often exploit species ideally suited for investigating specific phenomenon. For instance, comparative studies of socially monogamous prairie voles and polygamous meadow voles have been instrumental toward gaining an understanding of the genetic and neurobiological basis of social bonding. However, laboratory studies of less commonly used organisms, such as prairie voles, have been limited by a lack of genetic tools, including the ability to manipulate the genome. Here, we show that lentiviral vector-mediated transgenesis is a rapid and efficient approach for creating germline transgenics in alternative laboratory rodents. Injection of a green fluorescent protein (GFP)-expressing lentiviral vector into the perivitelline space of 23 single-cell embryos yielded three live offspring (13 %), one of which (33%) contained germline integration of a GFP transgene driven by the human ubiquitin-C promoter. In comparison, transfer of 23 uninjected embryos yielded six live offspring (26%). Green fluorescent protein is present in all tissues examined and is expressed widely in the brain. The GFP transgene is heritable and stably expressed until at least the F(2) generation. This technology has the potential to allow investigation of specific gene candidates in prairie voles and provides a general protocol to pursue germline transgenic manipulation in many different rodent species.


Asunto(s)
Arvicolinae/genética , Vectores Genéticos/genética , Transducción Genética/métodos , Transgenes/genética , Animales , Animales Modificados Genéticamente , Southern Blotting , Western Blotting , Proteínas Fluorescentes Verdes/genética , Inmunohistoquímica , Lentivirus , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
20.
J Vasc Res ; 46(5): 447-58, 2009.
Artículo en Inglés | MEDLINE | ID: mdl-19155635

RESUMEN

The G protein-coupled P2Y(2) nucleotide receptor (P2Y(2)R) is upregulated in response to stress and tissue injury and has been postulated to play a role in chronic inflammation seen in atherosclerosis, Alzheimer's disease and Sjogren's syndrome. The role of P2Y(2)R upregulation in vivo is poorly understood, in part due to the lack of a P2Y(2)R overexpressing animal model. The P2Y(2)R overexpressing transgenic rat was generated using a lentiviral vector. Rats overexpressing P2Y(2)R showed a significant increase in P2Y(2)R mRNA levels in all tissues screened as compared to nontransgenic rats. Fura 2 imaging of smooth muscle cells (SMCs) isolated from aorta indicated that the percentage of cells exhibiting increases in the intracellular free calcium concentration in response to P2Y(2)R agonists was significantly greater in freshly isolated SMCs from transgenic rats than wild-type controls. Histopathological examination of tissues revealed that P2Y(2)R overexpressing rats develop lymphocytic infiltration in lacrimal glands and kidneys as early as at 3 months of age. These rats show similarities to patients with Sjogren's syndrome who display lymphocyte-mediated tissue damage. This transgenic rat model of P2Y(2)R overexpression may prove useful for linking P2Y(2)R upregulation with chronic inflammatory diseases, neurodegenerative diseases and Sjogren's syndrome.


Asunto(s)
Modelos Animales de Enfermedad , Lentivirus/genética , Receptores Purinérgicos P2/genética , Síndrome de Sjögren/genética , Animales , Animales no Consanguíneos , Aorta/patología , Aorta/fisiología , Femenino , Expresión Génica , Vectores Genéticos , Homocigoto , Músculo Liso Vascular/patología , Músculo Liso Vascular/fisiología , Embarazo , Ratas , Ratas Sprague-Dawley , Ratas Transgénicas , Receptores Purinérgicos P2Y2 , Síndrome de Sjögren/patología , Síndrome de Sjögren/fisiopatología , Transgenes/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA