Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 47
Filtrar
1.
J Biol Chem ; 300(2): 105629, 2024 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-38199563

RESUMO

In contrast to stage-specific transcription factors, the role of ubiquitous transcription factors in neuronal development remains a matter of scrutiny. Here, we demonstrated that a ubiquitous factor NF-Y is essential for neural progenitor maintenance during brain morphogenesis. Deletion of the NF-YA subunit in neural progenitors by using nestin-cre transgene in mice resulted in significant abnormalities in brain morphology, including a thinner cerebral cortex and loss of striatum during embryogenesis. Detailed analyses revealed a progressive decline in multiple neural progenitors in the cerebral cortex and ganglionic eminences, accompanied by induced apoptotic cell death and reduced cell proliferation. In neural progenitors, the NF-YA short isoform lacking exon 3 is dominant and co-expressed with cell cycle genes. ChIP-seq analysis from the cortex during early corticogenesis revealed preferential binding of NF-Y to the cell cycle genes, some of which were confirmed to be downregulated following NF-YA deletion. Notably, the NF-YA short isoform disappears and is replaced by its long isoform during neuronal differentiation. Forced expression of the NF-YA long isoform in neural progenitors resulted in a significant decline in neuronal count, possibly due to the suppression of cell proliferation. Collectively, we elucidated a critical role of the NF-YA short isoform in maintaining neural progenitors, possibly by regulating cell proliferation and apoptosis. Moreover, we identified an isoform switch in NF-YA within the neuronal lineage in vivo, which may explain the stage-specific role of NF-Y during neuronal development.


Assuntos
Fator de Ligação a CCAAT , Córtex Cerebral , Animais , Camundongos , Fator de Ligação a CCAAT/genética , Fator de Ligação a CCAAT/metabolismo , Córtex Cerebral/citologia , Córtex Cerebral/crescimento & desenvolvimento , Córtex Cerebral/metabolismo , Regulação da Expressão Gênica , Neurogênese , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Fatores de Transcrição/metabolismo
2.
Biol Reprod ; 110(3): 465-475, 2024 Mar 13.
Artigo em Inglês | MEDLINE | ID: mdl-37995271

RESUMO

The mammalian oviductal lumen is a specialized chamber that provides an environment that strictly regulates fertilization and early embryogenesis, but the regulatory mechanisms to gametes and zygotes are unclear. We evaluated the oviductal regulation of early embryonic development using Ovgp1 (encoding an oviductal humoral factor, OVGP1)-knockout golden hamsters. The experimental results revealed the following: (1) female Ovgp1-knockout hamsters failed to produce litters; (2) in the oviducts of Ovgp1-knockout animals, fertilized eggs were sometimes identified, but their morphology showed abnormal features; (3) the number of implantations in the Ovgp1-knockout females was low; (4) even if implantations occurred, the embryos developed abnormally and eventually died; and (5) Ovgp1-knockout female ovaries transferred to wild-type females resulted in the production of Ovgp1-knockout egg-derived OVGP1-null litters, but the reverse experiment did not. These results suggest that OVGP1-mediated physiological events are crucial for reproductive process in vivo, from fertilization to early embryonic development. This animal model shows that the fate of the zygote is determined not only genetically, but also by the surrounding oviductal microenvironment.


Assuntos
Tubas Uterinas , Oviductos , Humanos , Gravidez , Animais , Cricetinae , Feminino , Mesocricetus , Células Germinativas , Ovário , Mamíferos , Glicoproteínas
3.
J Biol Chem ; 295(29): 9768-9785, 2020 07 17.
Artigo em Inglês | MEDLINE | ID: mdl-32499373

RESUMO

Huntington disease (HD) is a neurodegenerative disorder caused by expanded CAG repeats in the Huntingtin gene. Results from previous studies have suggested that transcriptional dysregulation is one of the key mechanisms underlying striatal medium spiny neuron (MSN) degeneration in HD. However, some of the critical genes involved in HD etiology or pathology could be masked in a common expression profiling assay because of contamination with non-MSN cells. To gain insight into the MSN-specific gene expression changes in presymptomatic R6/2 mice, a common HD mouse model, here we used a transgenic fluorescent protein marker of MSNs for purification via FACS before profiling gene expression with gene microarrays and compared the results of this "FACS-array" with those obtained with homogenized striatal samples (STR-array). We identified hundreds of differentially expressed genes (DEGs) and enhanced detection of MSN-specific DEGs by comparing the results of the FACS-array with those of the STR-array. The gene sets obtained included genes ubiquitously expressed in both MSNs and non-MSN cells of the brain and associated with transcriptional regulation and DNA damage responses. We proposed that the comparative gene expression approach using the FACS-array may be useful for uncovering the gene cascades affected in MSNs during HD pathogenesis.


Assuntos
Corpo Estriado/metabolismo , Citometria de Fluxo , Doença de Huntington/metabolismo , Transcriptoma , Animais , Corpo Estriado/patologia , Modelos Animais de Doenças , Proteína Huntingtina/genética , Proteína Huntingtina/metabolismo , Doença de Huntington/genética , Doença de Huntington/patologia , Masculino , Camundongos , Camundongos Transgênicos
4.
Int J Mol Sci ; 22(14)2021 Jul 12.
Artigo em Inglês | MEDLINE | ID: mdl-34299063

RESUMO

Regulatory T cells (Tregs) suppress immune responses and maintain immunological self-tolerance and homeostasis. We currently investigated relationships between skin barrier condition and Treg behavior using skin barrier-disrupted mice. Skin barrier disruption was induced by repeated topical application of 4% sodium dodecyl sulfate (SDS) on mice. The number of CD4+ forkhead box protein P3 (Foxp3)+ Tregs was higher in 4% SDS-treated skins than in controls. This increasing was correlated with the degree of acanthosis. The numbers of interleukin (IL)-10+ and transforming growth factor (TGF)-ß+ Tregs also increased in 4% SDS-treated skins. Localization of IL-33 in keratinocytes shifted from nucleus to cytoplasm after skin barrier disruption. Notably, IL-33 promoted the migration of Tregs in chemotaxis assay. The skin infiltration of Tregs was cancelled in IL-33 neutralizing antibody-treated mice and IL-33 knockout mice. Thus, keratinocyte-derived IL-33 may induce Treg migration into barrier-disrupted skin to control the phase transition between healthy and inflammatory conditions.


Assuntos
Movimento Celular , Quimiotaxia , Dermatite/patologia , Interleucina-33/fisiologia , Pele/patologia , Linfócitos T Reguladores/imunologia , Animais , Dermatite/imunologia , Dermatite/metabolismo , Fatores de Transcrição Forkhead/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Pele/metabolismo
5.
Mol Cell ; 44(2): 279-89, 2011 Oct 21.
Artigo em Inglês | MEDLINE | ID: mdl-22017874

RESUMO

Selective macroautophagy (autophagy) of ubiquitinated protein is implicated as a compensatory mechanism of the ubiquitin-proteasome system. p62/SQSTM1 is a key molecule managing autophagic clearance of polyubiquitinated proteins. However, little is known about mechanisms controlling autophagic degradation of polyubiquitinated proteins. Here, we show that the specific phosphorylation of p62 at serine 403 (S403) in its ubiquitin-associated (UBA) domain increases the affinity between UBA and polyubiquitin chain, resulting in efficiently targeting polyubiquitinated proteins in "sequestosomes" and stabilizing sequestosome structure as a cargo of ubiquitinated proteins for autophagosome entry. Casein kinase 2 (CK2) phosphorylates S403 of p62 directly. Furthermore, CK2 overexpression or phosphatase inhibition reduces the formation of inclusion bodies of the polyglutamine-expanded huntingtin exon1 fragment in a p62-dependent manner. We propose that phosphorylation of p62 at S403 regulates autophagic clearance of ubiquitinated proteins and protein aggregates that are poorly degraded by proteasomes.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/genética , Autofagia , Serina/genética , Proteínas Ubiquitinadas/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Caseína Quinase II/genética , Caseína Quinase II/metabolismo , Humanos , Proteína Huntingtina , Mutação , Proteínas do Tecido Nervoso/metabolismo , Proteínas Nucleares/metabolismo , Fosforilação , Complexo de Endopeptidases do Proteassoma/metabolismo , Proteína Sequestossoma-1 , Serina/metabolismo , Transfecção
6.
Hum Mol Genet ; 24(3): 740-56, 2015 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-25274774

RESUMO

In some neurological diseases caused by repeat expansions such as myotonic dystrophy, the RNA-binding protein muscleblind-like 1 (MBNL1) accumulates in intranuclear inclusions containing mutant repeat RNA. The interaction between MBNL1 and mutant RNA in the nucleus is a key event leading to loss of MBNL function, yet the details of this effect have been elusive. Here, we investigated the mechanism and significance of MBNL1 nuclear localization. We found that MBNL1 contains two classes of nuclear localization signal (NLS), a classical bipartite NLS and a novel conformational NLS. Alternative splicing of exon 7 acts as a switch between these NLS types and couples MBNL1 activity and intracellular localization. Depending on its nuclear localization, MBNL1 promoted nuclear accumulation of mutant RNA containing a CUG or CAG repeat, some of which produced proteins containing homopolymeric tracts such as polyglutamine. Furthermore, MBNL1 repressed the expression of these homopolymeric proteins including those presumably produced through repeat-associated non-ATG (RAN) translation. These results suggest that nuclear retention of expanded RNA reflects a novel role of MBNL proteins in repressing aberrant protein expression and may provide pathological and therapeutic implications for a wide range of repeat expansion diseases associated with nuclear RNA retention and/or RAN translation.


Assuntos
Núcleo Celular/metabolismo , Proteínas de Ligação a DNA/metabolismo , Sinais de Localização Nuclear/genética , RNA Mensageiro/metabolismo , Proteínas de Ligação a RNA/metabolismo , Expansão das Repetições de Trinucleotídeos , Processamento Alternativo , Animais , Células COS , Linhagem Celular Tumoral , Núcleo Celular/genética , Chlorocebus aethiops , Proteínas de Ligação a DNA/química , Regulação da Expressão Gênica , Humanos , Camundongos , Mutação , Sinais de Localização Nuclear/metabolismo , RNA Mensageiro/genética , Proteínas de Ligação a RNA/química
7.
Hum Mol Genet ; 24(4): 1092-105, 2015 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-25305080

RESUMO

Huntington's disease (HD) is a dominantly inherited genetic disease caused by mutant huntingtin (htt) protein with expanded polyglutamine (polyQ) tracts. A neuropathological hallmark of HD is the presence of neuronal inclusions of mutant htt. p62 is an important regulatory protein in selective autophagy, a process by which aggregated proteins are degraded, and it is associated with several neurodegenerative disorders including HD. Here, we investigated the effect of p62 depletion in three HD model mice: R6/2, HD190QG and HD120QG mice. We found that loss of p62 in these models led to longer life spans and reduced nuclear inclusions, although cytoplasmic inclusions increased with polyQ length. In mouse embryonic fibroblasts (MEFs) with or without p62, mutant htt with a nuclear localization signal (NLS) showed no difference in nuclear inclusion between the two MEF types. In the case of mutant htt without NLS, however, p62 depletion increased cytoplasmic inclusions. Furthermore, to examine the effect of impaired autophagy in HD model mice, we crossed R6/2 mice with Atg5 conditional knockout mice. These mice also showed decreased nuclear inclusions and increased cytoplasmic inclusions, similar to HD mice lacking p62. These data suggest that the genetic ablation of p62 in HD model mice enhances cytoplasmic inclusion formation by interrupting autophagic clearance of polyQ inclusions. This reduces polyQ nuclear influx and paradoxically ameliorates disease phenotypes by decreasing toxic nuclear inclusions.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/genética , Doença de Huntington/genética , Corpos de Inclusão Intranuclear/genética , Fenótipo , Animais , Autofagia , Corpo Estriado/metabolismo , Corpo Estriado/patologia , Modelos Animais de Doenças , Feminino , Hipocampo/metabolismo , Hipocampo/patologia , Doença de Huntington/mortalidade , Doença de Huntington/patologia , Espaço Intracelular/metabolismo , Longevidade/genética , Camundongos , Camundongos Knockout , Peptídeos/genética , Proteólise
8.
Biochim Biophys Acta ; 1842(9): 1472-84, 2014 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-24769000

RESUMO

Polyglutamine diseases are a family of inherited neurodegenerative diseases caused by the expansion of CAG repeats within the coding region of target genes. Still the mechanism(s) by which polyglutamine proteins are ubiquitinated and degraded remains obscure. Here, for the first time, we demonstrate that Mahogunin 21 ring finger 1 E3 ubiquitin protein ligase is depleted in cells that express expanded-polyglutamine proteins. MGRN1 co-immunoprecipitates with expanded-polyglutamine huntingtin and ataxin-3 proteins. Furthermore, we show that MGRN1 is predominantly colocalized and recruits with polyglutamine aggregates in both cellular and transgenic mouse models. Finally, we demonstrate that the partial depletion of MGRN1 increases the rate of aggregate formation and cell death, whereas the overexpression of MGRN1 reduces the frequency of aggregate formation and provides cytoprotection against polyglutamine-induced proteotoxicity. These observations suggest that stimulating the activity of MGRN1 ubiquitin ligase might be a potential therapeutic target to eliminate the cytotoxic threat in polyglutamine diseases.


Assuntos
Apoptose , Proteínas do Tecido Nervoso/metabolismo , Peptídeos/metabolismo , Dobramento de Proteína , Ubiquitina-Proteína Ligases/fisiologia , Ubiquitina/metabolismo , Animais , Western Blotting , Proliferação de Células , Células Cultivadas , Imunofluorescência , Humanos , Proteína Huntingtina , Imunoprecipitação , Masculino , Camundongos , Camundongos Transgênicos , Chaperonas Moleculares/genética , Chaperonas Moleculares/metabolismo , Proteínas do Tecido Nervoso/genética , Complexo de Endopeptidases do Proteassoma/metabolismo , Proteólise , RNA Mensageiro/genética , Reação em Cadeia da Polimerase em Tempo Real , Reação em Cadeia da Polimerase Via Transcriptase Reversa
9.
Biochem Biophys Res Commun ; 463(4): 1196-202, 2015 Aug 07.
Artigo em Inglês | MEDLINE | ID: mdl-26086102

RESUMO

Dominant mutations in Cu,Zn-superoxide dismutase (SOD1) cause a familial form of amyotrophic lateral sclerosis (ALS). A pathological hallmark of the familial ALS is the formation of mutant SOD1 aggregates, leading to the proposal that SOD1 gains toxicities through protein misfolding triggered by mutations. Nevertheless, molecular requirements for mutant SOD1 to acquire pathogenicity still remain obscure. Here, we show that Cys residues in SOD1 are essential to exerting toxicities of SOD1 in a Caenorhabditis elegans model. Exogenous expression of wild-type as well as pathogenic mutant SOD1 fused with a fluorescent protein in C. elegans resulted in the accumulation of disulfide-reduced SOD1 and retarded the worm's motility. In contrast, little effects of exogenously expressed SOD1 on the motility were observed when all four Cys residues in SOD1 were replaced with Ser. Taken together, we propose that deregulation of Cys chemistry in SOD1 proteins is involved in the pathogenesis of SOD1-related ALS.


Assuntos
Esclerose Lateral Amiotrófica/enzimologia , Caenorhabditis elegans/efeitos dos fármacos , Cisteína/metabolismo , Superóxido Dismutase/metabolismo , Animais , Modelos Animais de Doenças , Superóxido Dismutase/química
10.
J Biol Chem ; 287(41): 34764-75, 2012 Oct 05.
Artigo em Inglês | MEDLINE | ID: mdl-22891249

RESUMO

Aggregation of protein molecules is a pathological hallmark of many neurodegenerative diseases. Abnormal modifications have often been observed in the aggregated proteins, supporting the aggregation mechanism regulated by post-translational modifications on proteins. Modifications are in general assumed to occur in soluble proteins before aggregation, but actually it remains quite obscure when proteins are modified in the course of the aggregation. Here we focus upon aggregation of huntingtin (HTT), which causes a neurodegenerative disorder, Huntington disease, and we show that oxidation of a methionine residue in HTT occurs in vitro and also in vivo. Copper ions as well as added hydrogen peroxide are found to oxidize the methionine residue, but notably, this oxidative modification occurs only in the aggregated HTT but not in the soluble state. Furthermore, the methionine oxidation creates additional interactions among HTT aggregates and alters overall morphologies of the aggregates. We thus reveal that protein aggregates can be a target of oxidative modifications and propose that such a "post-aggregation" modification is a relevant factor to regulate properties of protein aggregates.


Assuntos
Cobre/química , Peróxido de Hidrogênio/química , Mutação , Proteínas do Tecido Nervoso/química , Oxidantes/química , Processamento de Proteína Pós-Traducional , Cobre/metabolismo , Humanos , Proteína Huntingtina , Doença de Huntington/genética , Doença de Huntington/metabolismo , Peróxido de Hidrogênio/farmacologia , Metionina/química , Metionina/metabolismo , Proteínas do Tecido Nervoso/genética , Oxidantes/farmacologia , Oxirredução/efeitos dos fármacos
11.
Biochem Biophys Res Commun ; 436(2): 121-7, 2013 Jun 28.
Artigo em Inglês | MEDLINE | ID: mdl-23524259

RESUMO

Repetitive transcranial magnetic stimulation (rTMS) is a new tool that has been used for the treatment of patients with neuropsychiatric disorders. However, the mechanisms underlying the effects of rTMS are still unclear. We analyzed the changes in mRNA expression in mouse brain that occurred after rTMS with an Affymetrix GeneChip. Following 20days of rTMS, many genes were differentially expressed in the mouse brain. Downregulation of Period 2 and 3 mRNA expression levels and a subsequent decrease in food and water intake were observed. HSP70 mRNA expression levels were upregulated after transient and chronic rTMS. In N2A 150Q cells, an upregulation of HSP70 mRNA and protein levels and subsequent cell-protective effects were observed after chronic rTMS. In addition, dopamine receptor 2 mRNA expression levels were downregulated, and a subsequent decrease in the binding of [(3)H]raclopride was observed. These results indicated that the modulation of several genes may be involved in the therapeutic mechanisms of chronic rTMS for patients with neuropsychiatric disorders.


Assuntos
Encéfalo/metabolismo , Perfilação da Expressão Gênica , Transtornos Mentais/genética , Transtornos Mentais/terapia , Estimulação Magnética Transcraniana/métodos , Animais , Ligação Competitiva , Western Blotting , Linhagem Celular Tumoral , Regulação para Baixo , Proteínas de Choque Térmico HSP72/genética , Proteínas de Choque Térmico HSP72/metabolismo , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Proteínas Circadianas Period/genética , Racloprida/metabolismo , Receptores de Dopamina D2/genética , Receptores de Dopamina D2/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Trítio , Regulação para Cima
12.
Nucleic Acids Res ; 39(7): 2781-98, 2011 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-21109527

RESUMO

TLS (translocated in liposarcoma), also known as FUS (fused in sarcoma), is an RNA/DNA-binding protein that plays regulatory roles in transcription, pre-mRNA splicing and mRNA transport. Mutations in TLS are responsible for familial amyotrophic lateral sclerosis (ALS) type 6. Furthermore, TLS-containing intracellular inclusions are found in polyglutamine diseases, sporadic ALS, non-SOD1 familial ALS and a subset of frontotemporal lobar degeneration, indicating a pathological significance of TLS in a wide variety of neurodegenerative diseases. Here, we identified TLS domains that determine intracellular localization of the murine TLS. Among them, PY-NLS located in the C-terminus is a strong determinant of intracellular localization as well as splicing regulation of an E1A-derived minigene. Disruption of PY-NLS promoted the formation of cytoplasmic granules that were partially overlapped with stress granules and P-bodies. Some of the ALS-linked mutations altered both intracellular localization and splicing regulation of TLS, while most mutations alone did not affect splicing regulation. However, phospho-mimetic substitution of Ser505 (or Ser513 in human) could enhance the effects of ALS mutations, highlighting interplay between post-translational modification and ALS-linked mutations. These results demonstrate that ALS-linked mutations can variably cause loss of nuclear functions of TLS depending on the degree of impairment in nuclear localization.


Assuntos
Esclerose Lateral Amiotrófica/genética , Mutação , Splicing de RNA , Proteína FUS de Ligação a RNA/genética , Proteínas E1A de Adenovirus/genética , Proteínas E1A de Adenovirus/metabolismo , Animais , Linhagem Celular , Grânulos Citoplasmáticos/química , Proteínas de Fluorescência Verde/genética , Humanos , Camundongos , Proteína FUS de Ligação a RNA/análise , Proteínas Recombinantes de Fusão/análise
13.
Hum Mol Genet ; 19(11): 2099-112, 2010 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-20185558

RESUMO

In polyglutamine diseases including Huntington's disease (HD), mutant proteins containing expanded polyglutamine stretches form nuclear aggregates in neurons. Although analysis of their disease models suggested a significance of transcriptional dysregulation in these diseases, how it mediates the specific neuronal cell dysfunction remains obscure. Here we performed a comprehensive analysis of altered DNA binding of multiple transcription factors using R6/2 HD model mice brains that express an N-terminal fragment of mutant huntingtin (mutant Nhtt). We found a reduction of DNA binding of Brn-2, a POU domain transcription factor involved in differentiation and function of hypothalamic neurosecretory neurons. We provide evidence supporting that Brn-2 loses its function through two pathways, its sequestration by mutant Nhtt and its reduced transcription, leading to reduced expression of hypothalamic neuropeptides. In contrast to Brn-2, its functionally related protein, Brn-1, was not sequestered by mutant Nhtt but was upregulated in R6/2 brain, except in hypothalamus. Our data indicate that functional suppression of Brn-2 together with a region-specific lack of compensation by Brn-1 mediates hypothalamic cell dysfunction by mutant Nhtt.


Assuntos
DNA/metabolismo , Proteínas de Homeodomínio/metabolismo , Doença de Huntington/metabolismo , Hipotálamo/citologia , Proteínas do Tecido Nervoso/metabolismo , Neurônios/metabolismo , Proteínas Nucleares/metabolismo , Fatores do Domínio POU/metabolismo , Animais , Ensaio de Desvio de Mobilidade Eletroforética , Proteínas de Homeodomínio/genética , Proteína Huntingtina , Doença de Huntington/genética , Doença de Huntington/patologia , Hipotálamo/metabolismo , Imuno-Histoquímica , Hibridização In Situ , Masculino , Camundongos , Microscopia de Fluorescência , Mutação/genética , Proteínas do Tecido Nervoso/genética , Neurônios/patologia , Proteínas Nucleares/genética , Fatores do Domínio POU/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa
14.
EMBO J ; 27(6): 827-39, 2008 Mar 19.
Artigo em Inglês | MEDLINE | ID: mdl-18288205

RESUMO

In Huntington's disease (HD), mutant Huntingtin, which contains expanded polyglutamine stretches, forms nuclear aggregates in neurons. The interactions of several transcriptional factors with mutant Huntingtin, as well as altered expression of many genes in HD models, imply the involvement of transcriptional dysregulation in the HD pathological process. The precise mechanism remains obscure, however. Here, we show that mutant Huntingtin aggregates interact with the components of the NF-Y transcriptional factor in vitro and in HD model mouse brain. An electrophoretic mobility shift assay using HD model mouse brain lysates showed reduction in NF-Y binding to the promoter region of HSP70, one of the NF-Y targets. RT-PCR analysis revealed reduced HSP70 expression in these brains. We further clarified the importance of NF-Y for HSP70 transcription in cultured neurons. These data indicate that mutant Huntingtin sequesters NF-Y, leading to the reduction of HSP70 gene expression in HD model mice brain. Because suppressive roles of HSP70 on the HD pathological process have been shown in several HD models, NF-Y could be an important target of mutant Huntingtin.


Assuntos
Fator de Ligação a CCAAT/metabolismo , Proteínas de Choque Térmico HSP70/antagonistas & inibidores , Proteínas de Choque Térmico HSP70/genética , Mutação , Proteínas do Tecido Nervoso/genética , Proteínas Nucleares/genética , Animais , Fator de Ligação a CCAAT/antagonistas & inibidores , Linhagem Celular Tumoral , Modelos Animais de Doenças , Proteínas de Choque Térmico HSP70/biossíntese , Proteína Huntingtina , Doença de Huntington/genética , Doença de Huntington/metabolismo , Masculino , Camundongos , Camundongos Transgênicos , Proteínas do Tecido Nervoso/fisiologia , Proteínas Nucleares/fisiologia , Ligação Proteica/genética
15.
Proc Natl Acad Sci U S A ; 106(24): 9679-84, 2009 Jun 16.
Artigo em Inglês | MEDLINE | ID: mdl-19487684

RESUMO

A hallmark of polyglutamine diseases, including Huntington disease (HD), is the formation of beta-sheet-rich aggregates, called amyloid, of causative proteins with expanded polyglutamines. However, it has remained unclear whether the polyglutamine amyloid is a direct cause or simply a secondary manifestation of the pathology. Here we show that huntingtin-exon1 (thtt) with expanded polyglutamines remarkably misfolds into distinct amyloid conformations under different temperatures, such as 4 degrees C and 37 degrees C. The 4 degrees C amyloid has loop/turn structures together with mostly beta-sheets, including exposed polyglutamines, whereas the 37 degrees C amyloid has more extended and buried beta-sheets. By developing a method to efficiently introduce amyloid into mammalian cells, we found that the formation of the 4 degrees C amyloid led to substantial toxicity, whereas the toxic effects of the 37 degrees C amyloid were very small. Importantly, thtt amyloids in different brain regions of HD mice also had distinct conformations. The thermolabile thtt amyloid with loop/turn structures in the striatum showed higher toxicity, whereas the rigid thtt amyloid with more extended beta-sheets in the hippocampus and cerebellum had only mild toxic effects. These studies show that the thtt protein with expanded polyglutamines can misfold into distinct amyloid conformations and, depending on the conformations, the amyloids can be either toxic or nontoxic. Thus, the amyloid conformation of thtt may be a critical determinant of cytotoxicity in HD.


Assuntos
Amiloide , Sobrevivência Celular/fisiologia , Éxons , Proteínas do Tecido Nervoso/química , Proteínas Nucleares/química , Animais , Encéfalo/metabolismo , Proteína Huntingtina , Camundongos , Proteínas do Tecido Nervoso/metabolismo , Proteínas do Tecido Nervoso/fisiologia , Proteínas Nucleares/metabolismo , Proteínas Nucleares/fisiologia , Conformação Proteica , Temperatura
16.
Hum Mol Genet ; 18(22): 4239-54, 2009 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-19661183

RESUMO

PQBP1 (polyglutamine tract-binding protein 1) is a causative gene for a relatively frequent X-linked syndromic and non-syndromic mental retardation (MR). To analyze behavioral abnormalities of these patients from molecular basis, we developed a knock-down (KD) mouse model. The KD mice possess a transgene expressing 498 bp double-strand RNA that is endogenously cleaved to siRNA suppressing PQBP1 efficiently. After confirming that PQBP1 is selectively suppressed to nearly 50% of the control mice, we performed behavioral analyses of PQBP1-KD mice. The KD mice possessed normal ability in ordinary memory tests including water-maze test, whereas they showed abnormal anxiety-related behavior in light/dark exploration test and open-field test and showed obvious declines of anxiety-related cognition in the repetitive elevated plus maze or novel object recognition test. Correspondingly, we found c-fos upregulation and histone H3 acetylation after behavior tests were declined in neurons of amygdala, prefrontal cortex and hippocampus. Furthermore, we found that 4-phenylbutyric acid, an HDAC inhibitor, efficiently improved expression of these genes and rescued the abnormal phenotypes in adult PQBP1-KD mice. These results suggested that PQBP1 dysfunction in regulating gene expression might underlie the abnormal behavior and cognition of PQBP1-KD mice and that the recovery of expression of such PQBP1 target genes might improve the symptoms in adult patients.


Assuntos
Ansiedade/genética , Ansiedade/psicologia , Proteínas de Transporte/genética , Cognição , Proteínas Nucleares/genética , Animais , Ansiedade/metabolismo , Proteínas de Transporte/metabolismo , Proteínas de Ligação a DNA , Modelos Animais de Doenças , Feminino , Expressão Gênica , Técnicas de Silenciamento de Genes , Humanos , Masculino , Aprendizagem em Labirinto , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteínas Nucleares/metabolismo
17.
Nat Med ; 10(2): 148-54, 2004 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-14730359

RESUMO

Inhibition of polyglutamine-induced protein aggregation could provide treatment options for polyglutamine diseases such as Huntington disease. Here we showed through in vitro screening studies that various disaccharides can inhibit polyglutamine-mediated protein aggregation. We also found that various disaccharides reduced polyglutamine aggregates and increased survival in a cellular model of Huntington disease. Oral administration of trehalose, the most effective of these disaccharides, decreased polyglutamine aggregates in cerebrum and liver, improved motor dysfunction and extended lifespan in a transgenic mouse model of Huntington disease. We suggest that these beneficial effects are the result of trehalose binding to expanded polyglutamines and stabilizing the partially unfolded polyglutamine-containing protein. Lack of toxicity and high solubility, coupled with efficacy upon oral administration, make trehalose promising as a therapeutic drug or lead compound for the treatment of polyglutamine diseases. The saccharide-polyglutamine interaction identified here thus provides a new therapeutic strategy for polyglutamine diseases.


Assuntos
Doença de Huntington/tratamento farmacológico , Doença de Huntington/patologia , Peptídeos/metabolismo , Trealose/uso terapêutico , Animais , Encéfalo/citologia , Encéfalo/metabolismo , Encéfalo/patologia , Morte Celular/fisiologia , Linhagem Celular , Modelos Animais de Doenças , Glucose/administração & dosagem , Glucose/metabolismo , Humanos , Proteína Huntingtina , Doença de Huntington/metabolismo , Fígado/citologia , Fígado/metabolismo , Fígado/patologia , Camundongos , Camundongos Transgênicos , Atividade Motora/fisiologia , Mioglobina/genética , Mioglobina/metabolismo , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/metabolismo , Neurônios/metabolismo , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo
18.
J Neurosci ; 29(16): 5153-62, 2009 Apr 22.
Artigo em Inglês | MEDLINE | ID: mdl-19386911

RESUMO

A pathological hallmark of the Huntington's disease (HD) is intracellular inclusions containing a huntingtin (Htt) protein with an elongated polyglutamine tract. Aggregation of mutant Htt causes abnormal protein-protein interactions, and the functional dysregulation of aggregate-interacting proteins (AIPs) has been proposed as a pathomechanism of HD. Despite this, a molecular mechanism remains unknown how Htt aggregates sequester AIPs. We note an RNA-binding protein, TIA-1, as a model of AIPs containing a Q/N-rich sequence and suggest that in vitro and in vivo Htt fibrillar aggregates function as a structural template for inducing insoluble fibrillation of TIA-1. It is also plausible that such a cross-seeding activity of Htt aggregates represses the physiological function of TIA-1. We thus propose that Htt aggregates act as an intracellular hub for the cross-seeded fibrillation of Q/N-rich AIPs and that a cross-seeding reaction is a molecular origin to cause diverse pathologies in a polyglutamine disease.


Assuntos
Glutamina/metabolismo , Doença de Huntington/genética , Doença de Huntington/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Proteínas Nucleares/metabolismo , Peptídeos/metabolismo , Animais , Asparagina/metabolismo , Linhagem Celular Tumoral , Células Cultivadas , Modelos Animais de Doenças , Humanos , Proteína Huntingtina , Corpos de Inclusão Intranuclear/metabolismo , Corpos de Inclusão Intranuclear/patologia , Camundongos , Camundongos Transgênicos , Proteínas do Tecido Nervoso/genética , Proteínas Nucleares/genética , Peptídeos/genética , Estrutura Terciária de Proteína/genética , Transporte Proteico/genética , Proteínas de Ligação a RNA/química , Proteínas de Ligação a RNA/metabolismo , Solubilidade
19.
Hum Mol Genet ; 17(20): 3223-35, 2008 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-18658163

RESUMO

Huntington's disease (HD) is a fatal neurodegenerative disorder. Despite a tremendous effort to develop therapeutic tools in several HD models, there is no effective cure at present. Acidosis has been observed previously in cellular and in in vivo models as well as in the brains of HD patients. Here we challenged HD models with amiloride (Ami) derivative benzamil (Ben), a chemical agent used to rescue acid-sensing ion channel (ASIC)-dependent acidotoxicity, to examine whether chronic acidosis is an important part of the HD pathomechanism and whether these drugs could be used as novel therapeutic agents. Ben markedly reduced the huntingtin-polyglutamine (htt-polyQ) aggregation in an inducible cellular system, and the therapeutic value of Ben was successfully recapitulated in the R6/2 animal model of HD. To reveal the mechanism of action, Ben was found to be able to alleviate the inhibition of the ubiquitin-proteasome system (UPS) activity, resulting in enhanced degradation of soluble htt-polyQ specifically in its pathological range. More importantly, we were able to demonstrate that blocking the expression of a specific isoform of ASIC (asic1a), one of the many molecular targets of Ben, led to an enhancement of UPS activity and this blockade also decreased htt-polyQ aggregation in the striatum of R6/2 mice. In conclusion, we believe that chemical compounds that target ASIC1a or pharmacological alleviation of UPS inhibition would be an effective and promising approach to combat HD and other polyQ-related disorders.


Assuntos
Doença de Huntington/tratamento farmacológico , Proteínas do Tecido Nervoso/antagonistas & inibidores , Canais Iônicos Sensíveis a Ácido , Adulto , Amilorida/análogos & derivados , Amilorida/farmacologia , Animais , Células Cultivadas , Modelos Animais de Doenças , Feminino , Humanos , Doença de Huntington/genética , Doença de Huntington/metabolismo , Técnicas In Vitro , Masculino , Camundongos , Camundongos Transgênicos , Pessoa de Meia-Idade , Mutação , Proteínas do Tecido Nervoso/genética , Peptídeos/química , Peptídeos/genética , Interferência de RNA , Ratos , Proteínas Recombinantes de Fusão/química , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Proteínas da Membrana Plasmática de Transporte de Serotonina/química , Proteínas da Membrana Plasmática de Transporte de Serotonina/genética , Proteínas da Membrana Plasmática de Transporte de Serotonina/metabolismo , Bloqueadores dos Canais de Sódio/farmacologia , Canais de Sódio/genética , Solubilidade
20.
J Histochem Cytochem ; 67(11): 813-824, 2019 11.
Artigo em Inglês | MEDLINE | ID: mdl-31424977

RESUMO

The arrangement of immature germ cells changes regularly and periodically along the axis of the seminiferous tubule, and is used to describe the progression of spermatogenesis. This description is based primarily on the changes in the acrosome and the nuclear morphology of haploid spermatids. However, such criteria cannot be applied under pathological conditions with arrested spermatid differentiation. In such settings, the changes associated with the differentiation of premeiotic germ cells must be analyzed. Here, we found that the unique bipolar motor protein, KIF11 (kinesin-5/Eg5), which functions in spindle formation during mitosis and meiosis in oocytes and early embryos, is expressed in premeiotic germ cells (spermatogonia and spermatocytes). Thus, we aimed to investigate whether KIF11 could be used to describe the progression of incomplete spermatogenesis. Interestingly, KIF11 expression was barely observed in haploid spermatids and Sertoli cells. The KIF11 staining allowed us to evaluate the progression of meiotic processes, by providing the time axis of spindle formation in both normal and spermatogenesis-arrested mutant mice. Accordingly, KIF11 has the potential to serve as an excellent marker to describe spermatogenesis, even in the absence of spermatid development.


Assuntos
Cinesinas/análise , Túbulos Seminíferos/citologia , Espermatogênese , Animais , Masculino , Meiose , Camundongos , Camundongos Endogâmicos C57BL , Túbulos Seminíferos/ultraestrutura , Espermátides/citologia , Espermatócitos/citologia , Espermatogônias/citologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA