Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 33
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
PLoS Genet ; 12(5): e1006083, 2016 05.
Artículo en Inglés | MEDLINE | ID: mdl-27203582

RESUMEN

The Huntington's disease (HD) protein, huntingtin (HTT), is a large protein consisting of 3144 amino acids and has conserved N-terminal sequences that are followed by a polyglutamine (polyQ) repeat. Loss of Htt is known to cause embryonic lethality in mice, whereas polyQ expansion leads to adult neuronal degeneration. Whether N-terminal HTT is essential for neuronal development or contributes only to late-onset neurodegeneration remains unknown. We established HTT knock-in mice (N160Q-KI) expressing the first 208 amino acids of HTT with 160Q, and they show age-dependent HTT aggregates in the brain and neurological phenotypes. Importantly, the N-terminal mutant HTT also preferentially accumulates in the striatum, the brain region most affected in HD, indicating the importance of N-terminal HTT in selective neuropathology. That said, homozygous N160Q-KI mice are also embryonic lethal, suggesting that N-terminal HTT alone is unable to support embryonic development. Using Htt knockout neurons, we found that loss of Htt selectively affects the survival of developing neuronal cells, but not astrocytes, in culture. This neuronal degeneration could be rescued by a truncated HTT lacking the first 237 amino acids, but not by N-terminal HTT (1-208 amino acids). Also, the rescue effect depends on the region in HTT known to be involved in intracellular trafficking. Thus, the N-terminal HTT region may not be essential for the survival of developing neurons, but when carrying a large polyQ repeat, can cause selective neuropathology. These findings imply a possible therapeutic benefit of removing the N-terminal region of HTT containing the polyQ repeat to treat the neurodegeneration in HD.


Asunto(s)
Proteína Huntingtina/genética , Enfermedad de Huntington/genética , Degeneración Nerviosa/genética , Proteínas del Tejido Nervioso/genética , Péptidos/genética , Animales , Cuerpo Estriado/metabolismo , Cuerpo Estriado/patología , Técnicas de Sustitución del Gen , Humanos , Proteína Huntingtina/metabolismo , Proteína Huntingtina/uso terapéutico , Enfermedad de Huntington/patología , Enfermedad de Huntington/terapia , Ratones , Mutación , Degeneración Nerviosa/patología , Degeneración Nerviosa/terapia , Proteínas del Tejido Nervioso/metabolismo , Proteínas del Tejido Nervioso/uso terapéutico , Neuronas/metabolismo , Neuronas/patología , Péptidos/metabolismo , Péptidos/uso terapéutico , Fenotipo
2.
Hum Mol Genet ; 24(8): 2308-17, 2015 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-25552648

RESUMEN

Parkinson's disease (PD) is an age-dependent neurodegenerative disease that can be caused by genetic mutations in α-synuclein (α-syn) or duplication of wild-type α-syn; PD is characterized by the deposition of α-syn aggregates, indicating a gain of toxicity from accumulation of α-syn. Although the major neuropathologic feature of PD is the degeneration of dopaminergic (DA) neurons in the substantia nigra, non-motor symptoms including anxiety, cognitive defect and sleep disorder precede the onset of motor impairment, and many clinical symptoms of PD are not caused by degeneration of DA neurons. Non-human primate models of PD are important for revealing the early pathology in PD and identifying effective treatments. We established transgenic PD rhesus monkeys that express mutant α-syn (A53T). Six transgenic A53T monkeys were produced via lentiviral vector expressing A53T in fertilized monkey eggs and subsequent embryo transfer to surrogates. Transgenic A53T is expressed in the monkey brain and causes age-dependent non-motor symptoms, including cognitive defects and anxiety phenotype, without detectable sleeping disorders. The transgenic α-syn monkeys demonstrate the specific early symptoms caused by mutant α-syn and provide insight into treatment of early PD.


Asunto(s)
Modelos Animales de Enfermedad , Macaca mulatta , Enfermedad de Parkinson/genética , alfa-Sinucleína/genética , Animales , Animales Modificados Genéticamente/genética , Animales Modificados Genéticamente/metabolismo , Encéfalo/metabolismo , Encéfalo/patología , Neuronas Dopaminérgicas/metabolismo , Femenino , Humanos , Macaca mulatta/genética , Macaca mulatta/metabolismo , Masculino , Enfermedad de Parkinson/metabolismo , Enfermedad de Parkinson/patología , alfa-Sinucleína/metabolismo , alfa-Sinucleína/toxicidad
3.
Proc Natl Acad Sci U S A ; 111(15): 5706-11, 2014 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-24706802

RESUMEN

Ubiquitination of misfolded proteins, a common feature of many neurodegenerative diseases, is mediated by different lysine (K) residues in ubiquitin and alters the levels of toxic proteins. In Huntington disease, polyglutamine expansion causes N-terminal huntingtin (Htt) to misfold, inducing neurodegeneration. Here we report that shorter N-terminal Htt fragments are more stable than longer fragments and find differential ubiquitination via K63 of ubiquitin. Aging decreases proteasome-mediated Htt degradation, at the same time increasing K63-mediated ubiquitination and subsequent Htt aggregation in HD knock-in mice. The association of Htt with the K48-specific E3 ligase, Ube3a, is decreased in aged mouse brain. Overexpression of Ube3a in HD mouse brain reduces K63-mediated ubiquitination and Htt aggregation, enhancing its degradation via the K48 ubiquitin-proteasome system. Our findings suggest that aging-dependent Ube3a levels result in differential ubiquitination and degradation of Htt fragments, thereby contributing to the age-related neurotoxicity of mutant Htt.


Asunto(s)
Proteínas del Tejido Nervioso/metabolismo , Proteínas Nucleares/metabolismo , Proteolisis , Ubiquitina-Proteína Ligasas/metabolismo , Ubiquitinación/fisiología , Factores de Edad , Animales , Cicloheximida , Técnica del Anticuerpo Fluorescente , Técnicas de Sustitución del Gen , Células HEK293 , Humanos , Proteína Huntingtina , Inmunoprecipitación , Ratones
4.
J Neurosci ; 35(21): 8345-58, 2015 May 27.
Artículo en Inglés | MEDLINE | ID: mdl-26019347

RESUMEN

Parkinson's disease (PD) is an age-dependent neurodegenerative disease that often occurs in those over age 60. Although rodents and small animals have been used widely to model PD and investigate its pathology, their short life span makes it difficult to assess the aging-related pathology that is likely to occur in PD patient brains. Here, we used brain tissues from rhesus monkeys at 2-3, 7-8, and >15 years of age to examine the expression of Parkin, PINK1, and α-synuclein, which are known to cause PD via loss- or gain-of-function mechanisms. We found that α-synuclein is increased in the older monkey brains, whereas Parkin and PINK1 are decreased or remain unchanged. Because of the gain of toxicity of α-synuclein, we performed stereotaxic injection of lentiviral vectors expressing mutant α-synuclein (A53T) into the substantia nigra of monkeys and found that aging also increases the accumulation of A53T in neurites and its associated neuropathology. A53T also causes more extensive reactive astrocytes and axonal degeneration in monkey brain than in mouse brain. Using monkey brain tissues, we found that A53T interacts with neurofascin, an adhesion molecule involved in axon subcellular targeting and neurite outgrowth. Aged monkey brain tissues show an increased interaction of neurofascin with A53T. Overexpression of A53T causes neuritic toxicity in cultured neuronal cells, which can be attenuated by transfected neurofascin. These findings from nonhuman primate brains reveal age-dependent pathological and molecular changes that could contribute to the age-dependent neuropathology in PD.


Asunto(s)
Envejecimiento/genética , Envejecimiento/patología , Encéfalo/patología , Degeneración Nerviosa/genética , Degeneración Nerviosa/patología , alfa-Sinucleína/genética , Envejecimiento/metabolismo , Animales , Encéfalo/metabolismo , Haplorrinos , Macaca mulatta , Masculino , Ratones , Ratones Endogámicos C57BL , Mutación/genética , Degeneración Nerviosa/metabolismo , alfa-Sinucleína/biosíntesis
5.
Hum Mol Genet ; 23(10): 2678-93, 2014 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-24381309

RESUMEN

Mutations in TAR DNA-binding protein 43 (TDP-43) are associated with familial forms of amyotrophic lateral sclerosis and frontotemporal lobar degeneration. Although recent studies have revealed that mutant TDP-43 in neuronal and glial cells is toxic, how mutant TDP-43 causes primarily neuronal degeneration in an age-dependent manner remains unclear. Using adeno-associated virus (AAV) that expresses mutant TDP-43 (M337V) ubiquitously, we found that mutant TDP-43 accumulates preferentially in neuronal cells in the postnatal mouse brain. We then ubiquitously or selectively expressed mutant TDP-43 in neuronal and glial cells in the striatum of adult mouse brains via stereotaxic injection of AAV vectors and found that it also preferentially accumulates in neuronal cells. Expression of mutant TDP-43 in neurons in the striatum causes more severe degeneration, earlier death and more robust symptoms in mice than expression of mutant TDP-43 in glial cells; however, aging increases the expression of mutant TDP-43 in glial cells, and expression of mutant TDP-43 in older mice caused earlier onset of phenotypes and more severe neuropathology than that in younger mice. Although expression of mutant TDP-43 in glial cells via stereotaxic injection does not lead to robust neurological phenotypes, systemic inhibition of the proteasome activity via MG132 in postnatal mice could exacerbate glial TDP-43-mediated toxicity and cause mice to die earlier. Consistently, this inhibition increases the expression of mutant TDP-43 in glial cells in mouse brains. Thus, the differential accumulation of mutant TDP-43 in neuronal versus glial cells contributes to the preferential toxicity of mutant TDP-43 in neuronal cells and age-dependent pathology.


Asunto(s)
Encéfalo/patología , Proteínas de Unión al ADN/genética , Neuroglía/metabolismo , Neuronas/metabolismo , Esclerosis Amiotrófica Lateral/genética , Esclerosis Amiotrófica Lateral/metabolismo , Esclerosis Amiotrófica Lateral/patología , Animales , Encéfalo/metabolismo , Proteínas de Unión al ADN/metabolismo , Expresión Génica , Células HEK293 , Humanos , Leupeptinas/farmacología , Ratones Endogámicos C57BL , Ratones Transgénicos , Mutación Missense , Células PC12 , Inhibidores de Proteasoma/farmacología , Proteolisis , Ratas
6.
J Neurosci ; 34(25): 8411-22, 2014 Jun 18.
Artículo en Inglés | MEDLINE | ID: mdl-24948797

RESUMEN

Huntington's disease (HD) belongs to a family of neurodegenerative diseases caused by misfolded proteins and shares the pathological hallmark of selective accumulation of misfolded proteins in neuronal cells. Polyglutamine expansion in the HD protein, huntingtin (Htt), causes selective neurodegeneration that is more severe in the striatum and cortex than in other brain regions, but the mechanism behind this selectivity is unknown. Here we report that in HD knock-in mice, the expression levels of mutant Htt (mHtt) are higher in brain tissues than in peripheral tissues. However, the expression of N-terminal mHtt via stereotaxic injection of viral vectors in mice also results in greater accumulation of mHtt in the striatum than in muscle. We developed an in vitro assay that revealed that extracts from the striatum and cortex promote the formation of high-molecular weight (HMW) mHtt compared with the relatively unaffected cerebellar and peripheral tissue extracts. Inhibition of ubiquitin-activating enzyme E1 (Ube1) increased the levels of HMW mHtt in the relatively unaffected tissues. Importantly, the expression levels of Ube1 are lower in brain tissues than peripheral tissues and decline in the nuclear fraction with age, which is correlated with the increased accumulation of mHtt in the brain and neuronal nuclei during aging. Our findings suggest that decreased targeting of misfolded Htt to the proteasome for degradation via Ube1 may underlie the preferential accumulation of toxic forms of mHtt in the brain and its selective neurodegeneration.


Asunto(s)
Química Encefálica/genética , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Enzimas Activadoras de Ubiquitina/fisiología , Animales , Activación Enzimática/genética , Femenino , Técnicas de Sustitución del Gen , Células HEK293 , Humanos , Proteína Huntingtina , Masculino , Ratones , Mutación , Proteínas del Tejido Nervioso/química , Proteínas Nucleares/química , Distribución Tisular/genética , Enzimas Activadoras de Ubiquitina/química , Enzimas Activadoras de Ubiquitina/genética
7.
J Neurosci ; 33(19): 8172-84, 2013 May 08.
Artículo en Inglés | MEDLINE | ID: mdl-23658157

RESUMEN

Mutations in the Abelson helper integration site-1 (AHI1) gene result in N-terminal Ahi1 fragments and cause Joubert syndrome, an autosomal recessive brain malformation disorder associated with delayed development. How AHI1 mutations lead to delayed development remains unclear. Here we report that full-length, but not N-terminal, Ahi1 binds Hap1, a huntingtin-associated protein that is essential for the postnatal survival of mice and that this binding is regulated during neuronal differentiation by nerve growth factor. Nerve growth factor induces dephosphorylation of Hap1A and decreases its association with Ahi1, correlating with increased Hap1A distribution in neurite tips. Consistently, Ahi1 associates with phosphorylated Hap1A in cytosolic, but not in synaptosomal, fractions isolated from mouse brain, suggesting that Ahi1 functions mainly in the soma of neurons. Mass spectrometry analysis of cytosolic Ahi1 immunoprecipitates reveals that Ahi1 also binds Cend1 (cell cycle exit and neuronal differentiation protein 1)/BM88, a neuronal protein that mediates neuronal differentiation and is highly expressed in postnatal mouse brain. Loss of Ahi1 reduces the levels of Cend1 in the hypothalamus of Ahi1 KO mice, which show retarded growth during postnatal days. Overexpressed Ahi1 can stabilize Cend1 in cultured cells. Furthermore, overexpression of Cend1 can rescue the neurite extension defects of hypothalamic neurons from Ahi1 KO mice. Our findings suggest that Cend1 is involved in Ahi1-associated hypothalamic neuronal differentiation in early development, giving us fresh insight into the mechanism behind the delayed development in Joubert syndrome.


Asunto(s)
Diferenciación Celular/fisiología , Regulación de la Expresión Génica/genética , Proteínas de la Membrana/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Neuronas/fisiología , Proteínas Proto-Oncogénicas/deficiencia , Proteínas Adaptadoras del Transporte Vesicular , Factores de Edad , Animales , Animales Recién Nacidos , Diferenciación Celular/efectos de los fármacos , Células Cultivadas , Regulación de la Expresión Génica/efectos de los fármacos , Suspensión Trasera/fisiología , Humanos , Hipotálamo/citología , Hipotálamo/crecimiento & desarrollo , Inmunoprecipitación , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Actividad Motora/efectos de los fármacos , Actividad Motora/genética , Mutación/genética , Factor de Crecimiento Nervioso/farmacología , Neuritas/efectos de los fármacos , Neuritas/fisiología , Neuronas/ultraestructura , Fosforilación/efectos de los fármacos , Ratas , Natación , Transfección
8.
Nat Genet ; 37(2): 198-204, 2005 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-15654337

RESUMEN

Proteins with polyglutamine (polyQ) expansions accumulate in the nucleus and affect gene expression. The mechanism by which mutant huntingtin (htt) accumulates intranuclearly is not known; wild-type htt, a 350-kDa protein of unknown function, is normally found in the cytoplasm. N-terminal fragments of mutant htt, which contain a polyQ expansion (>37 glutamines), have no conserved nuclear localization sequences or nuclear export sequences but can accumulate in the nucleus and cause neurological problems in transgenic mice. Here we report that N-terminal htt shuttles between the cytoplasm and nucleus in a Ran GTPase-independent manner. Small N-terminal htt fragments interact with the nuclear pore protein translocated promoter region (Tpr), which is involved in nuclear export. PolyQ expansion and aggregation decrease this interaction and increase the nuclear accumulation of htt. Reducing the expression of Tpr by RNA interference or deletion of ten amino acids of N-terminal htt, which are essential for the interaction of htt with Tpr, increased the nuclear accumulation of htt. These results suggest that Tpr has a role in the nuclear export of N-terminal htt and that polyQ expansion reduces this nuclear export to cause the nuclear accumulation of htt.


Asunto(s)
Enfermedad de Huntington/genética , Péptidos/metabolismo , Proteínas Proto-Oncogénicas/fisiología , Secuencia de Aminoácidos , Núcleo Celular/metabolismo , Células Cultivadas , Humanos , Datos de Secuencia Molecular , Mutación , Proteínas de Complejo Poro Nuclear/química , Regiones Promotoras Genéticas , Proteínas Proto-Oncogénicas/genética , Transfección
9.
Front Endocrinol (Lausanne) ; 15: 1328748, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38572474

RESUMEN

Background: In observational studies, the relationship between coffee intake and bone mineral density (BMD) is contradictory. However, residual confounding tends to bias the results of these studies. Therefore, we used a two-sample Mendelian randomization (MR) approach to further investigate the potential causal relationship between the two. Methods: Genetic instrumental variables (IVs) associated with coffee intake were derived from genome-wide association studies (GWAS) of the Food Frequency Questionnaire (FFQ) in 428,860 British individuals and matched using phenotypes in PhenoScanner. Summarized data on BMD were obtained from 537,750 participants, including total body BMD (TB-BMD), TB-BMD in five age brackets ≥60, 45-60, 30-45, 15-30, and 0-15 years, and BMD in four body sites: the lumbar spine, the femoral neck, the heel, and the ultradistal forearm. We used inverse variance weighting (IVW) methods as the primary analytical method for causal inference. In addition, several sensitivity analyses (MR-Egger, Weighted median, MR-PRESSO, Cochran's Q test, and Leave-one-out test) were used to test the robustness of the results. Results: After Bonferroni correction, Coffee intake has a potential positive correlation with total body BMD (effect estimate [Beta]: 0.198, 95% confidence interval [Cl]: 0.05-0.35, P=0.008). In subgroup analyses, coffee intake was potentially positively associated with TB-BMD (45-60, 30-45 years) (Beta: 0.408, 95% Cl: 0.12-0.69, P=0.005; Beta: 0.486, 95% Cl: 0.12-0.85, P=0.010). In addition, a significant positive correlation with heel BMD was also observed (Beta: 0.173, 95% Cl: 0.08-0.27, P=0.002). The results of the sensitivity analysis were generally consistent. Conclusion: The results of the present study provide genetic evidence for the idea that coffee intake is beneficial for bone density. Further studies are needed to reveal the biological mechanisms and offer solid support for clinical guidelines on osteoporosis prevention.


Asunto(s)
Densidad Ósea , Café , Humanos , Densidad Ósea/genética , Estudio de Asociación del Genoma Completo , Análisis de la Aleatorización Mendeliana , Cuello Femoral
10.
Hum Mol Genet ; 20(7): 1424-37, 2011 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-21245084

RESUMEN

An expanded polyglutamine tract (>37 glutamines) in the N-terminal region of huntingtin (htt) causes htt to accumulate in the nucleus, leading to transcriptional dysregulation in Huntington disease (HD). In HD knock-in mice that express full-length mutant htt at the endogenous level, mutant htt preferentially accumulates in the nuclei of striatal neurons, which are affected most profoundly in HD. The mechanism underlying this preferential nuclear accumulation of mutant htt in striatal neurons remains unknown. Here, we report that serine 16 (S16) in htt is important for the generation of small N-terminal fragments that are able to accumulate in the nucleus and form aggregates. Phosphorylation of N-terminal S16 in htt promotes the nuclear accumulation of small N-terminal fragments and reduces the interaction of N-terminal htt with the nuclear pore complex protein Tpr. Mouse brain striatal tissues show increased S16 phosphorylation and a decreased association between mutant N-terminal htt and Tpr. These findings provide mechanistic insight into the nuclear accumulation of mutant htt and the selective neuropathology of HD, revealing potential therapeutic targets for treating this disease.


Asunto(s)
Cuerpo Estriado/metabolismo , Mutación , Proteínas del Tejido Nervioso/metabolismo , Neuronas/metabolismo , Poro Nuclear/metabolismo , Proteínas Nucleares/metabolismo , Animales , Cuerpo Estriado/patología , Células HEK293 , Humanos , Proteína Huntingtina , Enfermedad de Huntington/genética , Enfermedad de Huntington/metabolismo , Enfermedad de Huntington/terapia , Ratones , Ratones Mutantes , Proteínas del Tejido Nervioso/genética , Neuronas/fisiología , Poro Nuclear/genética , Poro Nuclear/patología , Proteínas Nucleares/genética , Fosforilación , Ácido Poliglutámico/genética , Ácido Poliglutámico/metabolismo , Estructura Terciaria de Proteína
11.
Cell Mol Life Sci ; 69(8): 1305-17, 2012 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-21544547

RESUMEN

Hap1 was originally identified as a neuronal protein that interacts with huntingtin, the Huntington's disease (HD) protein. Later studies revealed that Hap1 participates in intracellular trafficking in neuronal cells and that this trafficking function can be adversely affected by mutant huntingtin. Hap1 is also present in pancreatic ß-cells and other endocrine cells; however, the role of Hap1 in these endocrine cells remains unknown. Using the Cre-loxP system, we generated conditional Hap1 knockout mice to selectively deplete the expression of Hap1 in mouse pancreatic ß-cells. Mutant mice with Hap1 deficiency in pancreatic ß-cells had impaired glucose tolerance and decreased insulin release in response to intraperitoneally injected glucose. Using cultured pancreatic ß-cell lines and isolated mouse pancreatic islets, we confirmed that decreasing Hap1 could reduce glucose-mediated insulin release. Electron microscopy suggested that there was a reduced number of insulin-containing vesicles docked at the plasma membrane of pancreatic islets in Hap1 mutant mice following intraperitoneal glucose injection. Glucose treatment decreased the phosphorylation of Hap1A in cultured ß-cells and in mouse pancreatic tissues. Moreover, this glucose treatment increased Hap1's association with kinesin light chain and dynactin p150, both of which are involved in microtubule-dependent trafficking. These studies suggest that Hap1 is important for insulin release from ß-cells via dephosphorylation that can regulate its intracellular trafficking function.


Asunto(s)
Células Secretoras de Insulina/metabolismo , Insulina/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Animales , Línea Celular , Glucosa/metabolismo , Enfermedad de Huntington/genética , Enfermedad de Huntington/metabolismo , Células Secretoras de Insulina/citología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Mutación , Proteínas del Tejido Nervioso/genética , Fosforilación
12.
Front Immunol ; 14: 1291206, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-38173728

RESUMEN

Background: Observational studies have shown that changes in circulating cytokine/growth factor levels occur throughout the initiation and progression of ankylosing spondylitis (AS), yet whether they are etiologic or downstream effects remains unclear. In this study, we performed a summarized-level bidirectional Mendelian randomization (MR) analysis to shed light on the causal relationship between the two. Methods: Genetic instrumental-variables (IVs) associated with circulating cytokine/growth factor levels were derived from a genome-wide association study (GWAS) of 8,293 European individuals, whereas summary data for the AS were obtained from a FinnGen GWAS of 166,144 participants. We used the inverse-variance-weighted (IVW) method as the main analysis for causal inference. Furthermore, several sensitivity analyses (MR-Egger, weighted median, MR-PRESSO and Cochran's Q test) were utilized to examine the robustness of the results. Finally, reverse MR analysis was performed to assess reverse causality between AS and circulating cytokine/growth factor levels. Results: After Bonferroni correction, circulating levels of Cutaneous T-cell attracting (CTACK) and Monocyte specific chemokine 3 (MCP-3) were positively associated with a higher risk of AS (odds ratio [OR]: 1.224, 95% confidence interval [95% Cl]: 1.022 ~ 1.468, P = 0.028; OR: 1.250, 95% Cl: 1.016 ~ 1.539, P = 0.035). In addition, elevated circulating levels of Basic fibroblast growth factor (FGF-basic), Granulocyte colony-stimulating factor (G-CSF) and MCP-3 was considered a consequence of AS disease (ß = 0.023, P = 0.017; ß = 0.017, P = 0.025; ß = 0.053, P = 0.025). The results of the sensitivity analysis were generally consistent. Conclusion: The present study supplies genetic evidence for the relationship between circulating cytokine levels and AS. Targeted interventions of specific cytokines may help to reduce the risk of AS initiation and progression.


Asunto(s)
Citocinas , Espondilitis Anquilosante , Humanos , Estudio de Asociación del Genoma Completo , Análisis de la Aleatorización Mendeliana , Espondilitis Anquilosante/genética , Factor Estimulante de Colonias de Granulocitos
13.
Hum Mol Genet ; 19(12): 2445-55, 2010 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-20354076

RESUMEN

An expanded polyglutamine (polyQ) domain in the N-terminal region of huntingtin (htt) causes misfolding and accumulation of htt in neuronal cells and the subsequent neurodegeneration of Huntington's disease (HD). Clearing the misfolded htt is critical for preventing neuropathology, and this process is mediated primarily by both the ubiquitin-proteasome system (UPS) and autophagy. Although overexpression of mutant htt can inhibit UPS activity in cultured cells, mutant htt does not inhibit global UPS activity in the brains of HD transgenic mice. These findings underscore the importance of investigating the function of the UPS and autophagy in the brain when mutant proteins are not overexpressed. When cultured PC12 cells were treated with either UPS or autophagy inhibitors, more N-terminal mutant htt fragments accumulated via inhibition of the UPS. Furthermore, in HD CAG repeat knock-in mouse brain, inhibiting the UPS also resulted in a greater accumulation of N-terminal, but not full-length, mutant htt than inhibiting autophagy did. Our findings suggest that impairment of the UPS may be more important for the accumulation of N-terminal mutant htt and might therefore make an attractive therapeutic target.


Asunto(s)
Enfermedad de Huntington/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Proteínas Nucleares/metabolismo , Péptidos/metabolismo , Inhibidores de Proteasoma , Ubiquitina/antagonistas & inhibidores , Animales , Autofagia/efectos de los fármacos , Encéfalo/metabolismo , Línea Celular , Técnicas de Sustitución del Gen , Humanos , Proteína Huntingtina , Ratones , Mutación , Proteínas del Tejido Nervioso/genética , Proteínas Nucleares/genética , Células PC12 , Péptidos/genética , Complejo de la Endopetidasa Proteasomal/metabolismo , Ratas , Ubiquitina/metabolismo
14.
Hum Mol Genet ; 19(20): 3983-94, 2010 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-20660116

RESUMEN

Neurodegeneration is a hallmark of many neurological diseases, including Alzheimer's, Parkinson's and the polyglutamine diseases, which are all caused by misfolded proteins that accumulate in neuronal cells of the brain. Although apoptosis is believed to contribute to neurodegeneration in these cases, genetic mouse models of these diseases often fail to replicate apoptosis and overt neurodegeneration in the brain. Using nuclear transfer, we generated transgenic Huntington's disease (HD) pigs that express N-terminal (208 amino acids) mutant huntingtin with an expanded polyglutamine tract (105Q). Postnatal death, dyskinesia and chorea-like movement were observed in some transgenic pigs that express mutant huntingtin. Importantly, the transgenic HD pigs, unlike mice expressing the same transgene, displayed typical apoptotic neurons with DNA fragmentation in their brains. Also, expression of mutant huntingtin resulted in more neurons with activated caspase-3 in transgenic pig brains than that in transgenic mouse brains. Our findings suggest that species differences determine neuropathology and underscore the importance of large mammalian animals for modeling neurological disorders.


Asunto(s)
Encéfalo/patología , Modelos Animales de Enfermedad , Enfermedad de Huntington , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Neuronas/metabolismo , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Porcinos Enanos , Animales , Animales Modificados Genéticamente/metabolismo , Apoptosis , Encéfalo/metabolismo , Caspasa 3/biosíntesis , Fragmentación del ADN , Expresión Génica , Proteína Huntingtina , Enfermedad de Huntington/genética , Enfermedad de Huntington/metabolismo , Enfermedad de Huntington/patología , Ratones , Ratones Transgénicos , Proteínas Mutantes/genética , Proteínas Mutantes/metabolismo , Degeneración Nerviosa/genética , Degeneración Nerviosa/metabolismo , Degeneración Nerviosa/patología , Enfermedades Neurodegenerativas , Neuronas/patología , Deficiencias en la Proteostasis , Especificidad de la Especie , Porcinos/genética , Porcinos/metabolismo , Porcinos Enanos/genética , Porcinos Enanos/metabolismo
15.
Proc Natl Acad Sci U S A ; 106(52): 22480-5, 2009 Dec 29.
Artículo en Inglés | MEDLINE | ID: mdl-20018729

RESUMEN

Huntington disease (HD) is an inherited neurological disorder caused by a polyglutamine expansion in the protein huntingtin and is characterized by selective neurodegeneration that preferentially occurs in striatal medium spiny neurons. Because the medium spiny neurons are innervated abundantly by glutamatergic axons from cortical neurons, the preferential degeneration in the striatal neurons supports the glutamate excitotoxicity theory for HD pathogenesis. Thus, glutamate uptake by glia may be particularly important for preventing glutamate excitotoxicity in HD. Although mutant huntingtin is expressed ubiquitously in various types of cells, it accumulates and forms aggregates in fewer glial cells than in neuronal cells. It remains largely unknown whether and how mutant huntingtin in glia can contribute to the neurological symptoms of HD. We generated transgenic mice that express N-terminal mutant huntingtin in astrocytes, a major type of glial cell that remove extracellular glutamate in the brain. Although transgenic mutant huntingtin in astrocytes is expressed below the endogenous level, it can cause age-dependent neurological phenotypes in transgenic mice. Mice expressing mutant huntingtin show body weight loss, have motor function deficits, and die earlier than wild-type or control transgenic mice. We also found that mutant huntingtin in astrocytes decreases the expression of glutamate transporter by increasing its binding to Sp1 and reducing the association of Sp1 with the promoter of glutamate transporter. These results imply an important role for glial mutant huntingtin in HD pathology and suggest possibilities for treatment.


Asunto(s)
Astrocitos/fisiología , Encéfalo/fisiopatología , Enfermedad de Huntington/genética , Enfermedad de Huntington/fisiopatología , Mutación , Proteínas del Tejido Nervioso/genética , Proteínas Nucleares/genética , Envejecimiento/genética , Envejecimiento/patología , Envejecimiento/fisiología , Animales , Secuencia de Bases , Encéfalo/patología , Cartilla de ADN/genética , Transportador 2 de Aminoácidos Excitadores/genética , Transportador 2 de Aminoácidos Excitadores/metabolismo , Expresión Génica , Proteína Ácida Fibrilar de la Glía/genética , Gliosis/genética , Gliosis/patología , Ácido Glutámico/metabolismo , Humanos , Proteína Huntingtina , Enfermedad de Huntington/patología , Ratones , Ratones Transgénicos , Proteínas del Tejido Nervioso/fisiología , Proteínas Nucleares/fisiología , Fenotipo , Regiones Promotoras Genéticas , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Factor de Transcripción Sp1/metabolismo
16.
J Biol Chem ; 285(14): 10653-61, 2010 Apr 02.
Artículo en Inglés | MEDLINE | ID: mdl-20145253

RESUMEN

Huntington disease (HD) is caused by an expansion of the polyglutamine (polyQ) repeat (>37Q) in huntingtin (htt), and age of onset is inversely correlated with the length of the polyQ repeat. Mutant htt with expanded polyQ is ubiquitously expressed in various types of cells, including glia, but causes selective neurodegeneration. Our recent study demonstrated that expression of the N-terminal mutant htt with a large polyQ repeat (160Q) in astrocytes is sufficient to induce neurological symptoms in mice (Bradford, J., Shin, J. Y., Roberts, M., Wang, C. E., Li, X.-J., and Li, S. H. (2009) Proc. Natl. Acad. Sci. U.S.A. 106, 22480-22485). Because glia-neuron interactions are critical for maintaining the normal function and survival of neurons in the brain and because mutant htt is more abundant in neurons than in glial cells, it is important to investigate whether glial htt can still contribute to HD pathology when mutant htt is abundantly expressed in neuronal cells. We generated transgenic mice that express mutant htt with 98Q in astrocytes. Unlike our recently generated htt-160Q transgenic mice, htt-98Q mice do not show obvious neurological phenotypes, suggesting that the length of the polyQ repeat determines the severity of glial dysfunction. However, htt-98Q mice show increased susceptibility to glutamate-induced seizure. Mice expressing mutant htt in astrocytes were mated with N171-82Q mice that express mutant htt primarily in neuronal cells. Double transgenic mice expressing mutant htt in both neuronal and glial cells display more severe neurological symptoms and earlier death than N171-82Q mice. These findings indicate a role of glial mutant htt in exacerbating HD neuropathology and underscore the importance of improving glial function in treating HD.


Asunto(s)
Modelos Animales de Enfermedad , Enfermedad de Huntington/patología , Mutación/genética , Proteínas del Tejido Nervioso/metabolismo , Neuroglía/metabolismo , Proteínas Nucleares/metabolismo , Convulsiones/patología , Animales , Conducta Animal , Western Blotting , Encéfalo/citología , Encéfalo/metabolismo , Células Cultivadas , Femenino , Ácido Glutámico/toxicidad , Humanos , Proteína Huntingtina , Enfermedad de Huntington/genética , Técnicas para Inmunoenzimas , Masculino , Ratones , Ratones Transgénicos , Proteínas del Tejido Nervioso/genética , Neuroglía/citología , Neuronas/citología , Neuronas/metabolismo , Proteínas Nucleares/genética , Fenotipo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Convulsiones/inducido químicamente , Convulsiones/metabolismo , Expansión de Repetición de Trinucleótido/fisiología
17.
J Clin Invest ; 118(8): 2785-95, 2008 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-18636121

RESUMEN

Joubert syndrome is an autosomal recessive disorder characterized by congenital malformation of the cerebellum and brainstem, with abnormal decussation in the brain. Mutations in the Abelson helper integration site 1 gene, which encodes the protein AHI1, have been shown to cause Joubert syndrome. In this study, we found that mouse Ahi1 formed a stable complex with huntingtin-associated protein 1 (Hap1), which is critical for neonatal development and involved in intracellular trafficking. Hap1-knockout mice showed significantly reduced Ahi1 levels, defective cerebellar development, and abnormal axonal decussation. Suppression of Ahi1 also decreased the level of Hap1; and truncated Ahi1, which corresponds to the mutations in Joubert syndrome, inhibited neurite outgrowth in neuronal culture. Reducing Hap1 expression suppressed the level and internalization of TrkB, a neurotrophic factor receptor that mediates neurogenesis and neuronal differentiation, which led to decreased TrkB signaling. These findings provide insight into the pathogenesis of Joubert syndrome and demonstrate the critical role of the Ahi1-Hap1 complex in early brain development.


Asunto(s)
Tronco Encefálico/anomalías , Cerebelo/anomalías , Proteínas del Tejido Nervioso/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Proteínas Adaptadoras del Transporte Vesicular , Animales , Línea Celular , Células Cultivadas , Humanos , Inmunohistoquímica , Riñón/citología , Ratones , Ratones Noqueados , Neuronas/citología , Neuronas/metabolismo , Transfección
18.
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
19.
J Cell Biol ; 171(6): 1001-12, 2005 Dec 19.
Artículo en Inglés | MEDLINE | ID: mdl-16365166

RESUMEN

Huntington disease (HD) is characterized by the preferential loss of striatal medium-sized spiny neurons (MSNs) in the brain. Because MSNs receive abundant glutamatergic input, their vulnerability to excitotoxicity may be largely influenced by the capacity of glial cells to remove extracellular glutamate. However, little is known about the role of glia in HD neuropathology. Here, we report that mutant huntingtin accumulates in glial nuclei in HD brains and decreases the expression of glutamate transporters. As a result, mutant huntingtin (htt) reduces glutamate uptake in cultured astrocytes and HD mouse brains. In a neuron-glia coculture system, wild-type glial cells protected neurons against mutant htt-mediated neurotoxicity, whereas glial cells expressing mutant htt increased neuronal vulnerability. Mutant htt in cultured astrocytes decreased their protection of neurons against glutamate excitotoxicity. These findings suggest that decreased glutamate uptake caused by glial mutant htt may critically contribute to neuronal excitotoxicity in HD.


Asunto(s)
Neuroglía/metabolismo , Proteínas de Transporte de Serotonina en la Membrana Plasmática/genética , Proteínas de Transporte de Serotonina en la Membrana Plasmática/metabolismo , Adulto , Anciano , Animales , Astrocitos/metabolismo , Encéfalo/metabolismo , Encéfalo/patología , Células Cultivadas , Relación Dosis-Respuesta a Droga , Transportador 2 de Aminoácidos Excitadores/metabolismo , Regulación de la Expresión Génica , Ácido Glutámico/metabolismo , Humanos , Enfermedad de Huntington/metabolismo , Ratones , Ratones Transgénicos , Persona de Mediana Edad , Ratas , Proteínas de Transporte de Serotonina en la Membrana Plasmática/toxicidad
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA