Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 59
Filtrar
1.
Int J Mol Sci ; 25(8)2024 Apr 09.
Artigo em Inglês | MEDLINE | ID: mdl-38673728

RESUMO

BTB and CNC homology 1 (BACH1) represses the expression of genes involved in the metabolism of iron, heme and reactive oxygen species. While BACH1 is rapidly degraded when it is bound to heme, it remains unclear how BACH1 degradation is regulated under other conditions. We found that FBXO22, a ubiquitin ligase previously reported to promote BACH1 degradation, polyubiquitinated BACH1 only in the presence of heme in a highly purified reconstitution assay. In parallel to this regulatory mechanism, TANK binding kinase 1 (TBK1), a protein kinase that activates innate immune response and regulates iron metabolism via ferritinophagy, was found to promote BACH1 degradation when overexpressed in 293T cells. While TBK1 phosphorylated BACH1 at multiple serine and threonine residues, BACH1 degradation was observed with not only the wild-type TBK1 but also catalytically impaired TBK1. The BACH1 degradation in response to catalytically impaired TBK1 was not dependent on FBXO22 but involved both autophagy-lysosome and ubiquitin-proteasome pathways judging from its suppression by using inhibitors of lysosome and proteasome. Chemical inhibition of TBK1 in hepatoma Hepa1 cells showed that TBK1 was not required for the heme-induced BACH1 degradation. Its inhibition in Namalwa B lymphoma cells increased endogenous BACH1 protein. These results suggest that TBK1 promotes BACH1 degradation in parallel to the FBXO22- and heme-dependent pathway, placing BACH1 as a downstream effector of TBK1 in iron metabolism or innate immune response.


Assuntos
Fatores de Transcrição de Zíper de Leucina Básica , Proteínas F-Box , Heme , Proteínas Serina-Treonina Quinases , Proteólise , Receptores Citoplasmáticos e Nucleares , Humanos , Heme/metabolismo , Fatores de Transcrição de Zíper de Leucina Básica/metabolismo , Fatores de Transcrição de Zíper de Leucina Básica/genética , Fosforilação , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Serina-Treonina Quinases/genética , Proteínas F-Box/metabolismo , Proteínas F-Box/genética , Células HEK293 , Ubiquitinação , Linhagem Celular Tumoral , Lisossomos/metabolismo , Autofagia , Complexo de Endopeptidases do Proteassoma/metabolismo
2.
Front Endocrinol (Lausanne) ; 14: 1172060, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37547309

RESUMO

Organismal energy balance is controlled by inter-tissue communication mediated by the nervous system and hormones, the disruption of which causes metabolic syndrome exemplified by diabetes and obesity. Fat-storing adipose tissue, especially those located in subcutaneous white adipose tissue, secretes leptin in a proportion of fat mass, inhibiting the accumulation of organismal fat by suppressing appetite and promoting energy expenditure. With a prevalence of obesity that exhibits hyperleptinemia, most of the investigation on leptin has been focused on how it works and how it does not, which is expected to be a clue for treating obesity. In contrast, how it is synthesized, transported, and excreted, all of which are relevant to the homeostasis of blood leptin concentration, are not much understood. Of note, acute leptin reduction after hyperleptinemia in the context of obesity exhibited a beneficial effect on obesity and insulin sensitivity, indicating that manipulation of circulating leptin level may provide a therapeutic strategy. Technological advances such as "omics" analysis combined with sophisticated gene-engineered mice studies in the past decade enabled a deeper understanding of leptin's action in more detail. Here, we summarize the updated understanding of the action as well as regulation of leptin and point out the emerging direction of research on leptin.


Assuntos
Diabetes Mellitus , Leptina , Camundongos , Animais , Leptina/metabolismo , Adipocinas/metabolismo , Tecido Adiposo/metabolismo , Obesidade/metabolismo , Diabetes Mellitus/metabolismo
4.
Am J Physiol Cell Physiol ; 323(6): C1633-C1641, 2022 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-36189972

RESUMO

Alzheimer's disease is an intractable disease, and the accumulation of amyloid ß in the brain is thought to be involved in the onset of the disease. Additionally, abnormal protein accumulation due to autophagic deficiency may also be involved in disease progression. Autophagy involves a mechanism called selective autophagy. However, the relationship between selective autophagy and the amyloid precursor protein (APP) remains unclear. In the present study, we analyzed the interaction between p62, an adapter protein, and an APP-related molecule and found that p62 interacted with the COOH-terminal fragment of APP (C60). When C60 and p62 are overexpressed, aggregates are formed and C60 is degraded by autophagy. These aggregates cannot be easily degraded, even with a reducing agent. We also found that autophagosome- and lysosome marker-positive vesicles were formed in the C60- and p62-expressing cells. Superresolution technology also revealed that p62-C60-positive autophagosomes were formed in the cells. Overall, these results suggest that p62 may bind with C60 to form aggregates and induce autophagy in autophagosomes. These results reveal one of the mechanisms underlying the progression of Alzheimer's disease, in which selective autophagy may be involved.


Assuntos
Doença de Alzheimer , Precursor de Proteína beta-Amiloide , Humanos , Precursor de Proteína beta-Amiloide/genética , Precursor de Proteína beta-Amiloide/metabolismo , Doença de Alzheimer/genética , Doença de Alzheimer/metabolismo , Peptídeos beta-Amiloides , Autofagia , Autofagossomos/metabolismo , Proteína Sequestossoma-1/genética , Proteína Sequestossoma-1/metabolismo
5.
Surg Case Rep ; 8(1): 148, 2022 Aug 02.
Artigo em Inglês | MEDLINE | ID: mdl-35915344

RESUMO

BACKGROUND: Splenic artery aneurysms usually rupture into the free peritoneal space and rarely into the gastrointestinal tract. We report the case of a patient with a giant splenic artery aneurysm that ruptured in to the stomach with hemorrhagic shock and was successfully treated with emergency surgery. CASE PRESENTATION: A 59-year-old man presented to the emergency department with chest pain and syncope. Contrast-enhanced computed tomography showed splenic artery aneurysm with active contrast extravasation. He developed upper gastrointestinal (UGI) bleeding and hypovolemic shock. We diagnosed a splenic artery aneurysm ruptured in to the stomach, performed emergency distal splenopancreatectomy including the aneurysm and partial gastric resection, and could prevent patient death. CONCLUSIONS: This report shows that splenic artery aneurysm can cause UGI bleeding. Thus, clinicians should be alert about this condition when managing patients with UGI bleeding and/or splenic artery aneurysm.

6.
J Pharmacol Exp Ther ; 381(2): 68-78, 2022 05.
Artigo em Inglês | MEDLINE | ID: mdl-35241633

RESUMO

Parkinson's disease (PD) is the second most common neurodegenerative disease. Although many monogenic variants have been identified that cause familial PD, most cases are sporadic and the mechanisms of sporadic PD onset remain unclear. We previously identified midnolin (MIDN) as a novel genetic risk factor for PD in a Japanese population. MIDN copy number loss was strongly associated with sporadic PD, which was replicated in a British population. Furthermore, suppression of MIDN expression in rat pheochromocytoma cells inhibits neurite outgrowth and expression of Parkin ubiquitin ligase. However, the detailed molecular mechanisms of MIDN expression are unknown. We, therefore, investigated the molecular mechanism of MIDN expression in human neuroblastoma SH-SY5Y cells. We found that MIDN expression was promoted by insulin via extracellular-signal regulated kinase1/2 and phosphoinositide 3-kinase-dependent pathways. In addition, MIDN promoter activity was enhanced by mutations at transcription factor AP-2 consensus sequences and reduced by mutations at cAMP response element-binding protein and activator protein 1 (AP-1) consensus sequences. The dominant-negative cAMP response element-binding protein mutant did not block MIDN promoter activity, but both the pharmacological inhibitor and decoy oligodeoxynucleotide for AP-1 significantly blocked its activity. Additionally, DNA binding of c-FOS and c-JUN to the AP-1 consensus sequence in the MIDN promoter was enhanced by insulin as determined by chromatin immunoprecipitation, which suggested that AP-1 positively regulated MIDN expression. Taken together, this study reveals molecular mechanisms of MIDN gene expression induced by insulin in neuronal cells, and drugs which promote MIDN expression may have potential to be a novel medicine for PD. SIGNIFICANCE STATEMENT: We demonstrated that insulin promotes midnolin expression via extracellular-signal regulated kinase 1/2 and phosphoinositide 3-kinase pathways. Furthermore, we identified the important region of the MIDN promoter and showed that transcription factors, including activator protein 1, positively regulate MIDN expression, whereas transcription factor AP-2 negatively regulates basal and insulin-induced MIDN expression. We believe that our observations are important and that they contribute to the development of novel drugs to treat Parkinson's disease.


Assuntos
Doenças Neurodegenerativas , Doença de Parkinson , Animais , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/metabolismo , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Expressão Gênica , Humanos , Insulina/farmacologia , Proteínas Nucleares , Doença de Parkinson/genética , Doença de Parkinson/metabolismo , Fosfatidilinositol 3-Quinase/genética , Fosfatidilinositol 3-Quinase/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Ratos , Fatores de Risco , Fator de Transcrição AP-1/genética , Fator de Transcrição AP-1/metabolismo , Fator de Transcrição AP-2/genética , Fator de Transcrição AP-2/metabolismo , Transcrição Gênica
7.
Cell Rep ; 38(12): 110541, 2022 03 22.
Artigo em Inglês | MEDLINE | ID: mdl-35320725

RESUMO

The histone chaperone complex FACT comprises SPT16 and SSRP1 and contributes to DNA replication, transcription, and repair, but how it plays such various roles is unclear. Here, we show that human SPT16 is ubiquitylated at lysine-674 (K674) by the DCAF14-CRL4 ubiquitin ligase. K674 is located in the middle domain of SPT16, and the corresponding residue of the yeast ortholog is critical for binding to histone H3.1-H4. We show that the middle domain of human SPT16 binds to histone H3.1-H4 and that this binding is inhibited by K674 ubiquitylation. Cells with heterozygous knockin of a K674R mutant of SPT16 manifest reduction of both SPT16 ubiquitylation and H3.1 in chromatin, a reduced population in mid S phase, impaired proliferation, and increased susceptibility to S phase stress. Our data thus indicate that SPT16 ubiquitylation by DCAF14-CRL4 regulates FACT binding to histones and may thereby control DNA replication-coupled histone incorporation into chromatin.


Assuntos
Histonas , Proteínas de Saccharomyces cerevisiae , Cromatina , Proteínas de Ligação a DNA , Proteínas de Grupo de Alta Mobilidade , Chaperonas de Histonas , Humanos , Lisina , Receptores de Interleucina-17 , Saccharomyces cerevisiae , Fatores de Elongação da Transcrição , Ubiquitina-Proteína Ligases , Ubiquitinação
8.
Front Genet ; 13: 1022339, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36685966

RESUMO

Genomic analysis has revealed that the genes for various chromatin regulators are mutated in many individuals with neurodevelopmental disorders (NDDs), emphasizing the important role of chromatin regulation in nervous system development and function. Chromatin regulation is mediated by writers, readers, and erasers of histone and DNA modifications, with such proteins being defined by specific domains. One of these domains is the SET domain, which is present in enzymes that catalyze histone methylation. Heterozygous loss-of-function mutations of the SETD5 (SET domain containing 5) gene have been identified in individuals with an NDD designated IDD23 (intellectual developmental disorder, autosomal dominant 23). KBG syndrome (named after the initials of the last names of the first three families identified with the condition) is characterized by features that either overlap with or are distinct from those of IDD23 and was initially thought to be caused only by mutations in the ANKRD11 (ankyrin repeat domain containing 11) gene. However, recent studies have identified SETD5 mutations in some KBG syndrome patients without ANKRD11 mutations. Here we summarize the neurobehavioral characterization of Setd5 +/- mice performed by four independent research groups, compare IDD23 and KBG phenotypes, and address the utility and future development of mouse models for elucidation of the mechanisms underlying NDD pathogenesis, with a focus on SETD5 and its related proteins.

9.
Stem Cell Reports ; 16(6): 1527-1541, 2021 06 08.
Artigo em Inglês | MEDLINE | ID: mdl-34048688

RESUMO

Amyotrophic lateral sclerosis (ALS) is an adult-onset incurable motor neuron (MN) disease. The reasons for selective MN vulnerability in ALS are unknown. Axonal pathology is among the earliest signs of ALS. We searched for novel modulatory genes in human MN axon shortening affected by TARDBP mutations. In transcriptome analysis of RNA present in the axon compartment of human-derived induced pluripotent stem cell (iPSC)-derived MNs, PHOX2B (paired-like homeobox protein 2B) showed lower expression in TARDBP mutant axons, which was consistent with axon qPCR and in situ hybridization. PHOX2B mRNA stability was reduced in TARDBP mutant MNs. Furthermore, PHOX2B knockdown reduced neurite length in human MNs. Finally, phox2b knockdown in zebrafish induced short spinal axons and impaired escape response. PHOX2B is known to be highly express in other types of neurons maintained after ALS progression. Collectively, TARDBP mutations induced loss of axonal resilience, which is an important ALS-related phenotype mediated by PHOX2B downregulation.


Assuntos
Esclerose Lateral Amiotrófica/metabolismo , Axônios/metabolismo , Proteínas de Ligação a DNA/metabolismo , Proteínas de Homeodomínio/metabolismo , Células-Tronco Pluripotentes Induzidas/metabolismo , Neurônios Motores/metabolismo , Fatores de Transcrição/metabolismo , Peixe-Zebra/metabolismo , Esclerose Lateral Amiotrófica/genética , Animais , Proteínas de Ligação a DNA/genética , Regulação da Expressão Gênica , Técnicas de Silenciamento de Genes/métodos , Proteínas de Homeodomínio/genética , Humanos , Mutação , Fenótipo , Fatores de Transcrição/genética , Transcriptoma
10.
iScience ; 23(9): 101491, 2020 Aug 21.
Artigo em Inglês | MEDLINE | ID: mdl-32891887

RESUMO

C21ORF2 and NEK1 have been identified as amyotrophic lateral sclerosis (ALS)-associated genes. Both genes are also mutated in certain ciliopathies, suggesting that they might contribute to the same signaling pathways. Here we show that FBXO3, the substrate receptor of an SCF ubiquitin ligase complex, binds and ubiquitylates C21ORF2, thereby targeting it for proteasomal degradation. C21ORF2 stabilizes the kinase NEK1, with the result that loss of FBXO3 stabilizes not only C21ORF2 but also NEK1. Conversely, NEK1-mediated phosphorylation stabilizes C21ORF2 by attenuating its interaction with FBXO3. We found that the ALS-associated V58L mutant of C21ORF2 is more susceptible to phosphorylation by NEK1, with the result that it is not ubiquitylated by FBXO3 and therefore accumulates together with NEK1. Expression of C21ORF2(V58L) in motor neurons induced from mouse embryonic stem cells impaired neurite outgrowth. We suggest that inhibition of NEK1 activity is a potential therapeutic approach to ALS associated with C21ORF2 mutation.

11.
Sci Adv ; 6(38)2020 09.
Artigo em Inglês | MEDLINE | ID: mdl-32948596

RESUMO

TET2 DNA dioxygenase is frequently mutated in human hematopoietic malignancies and functionally inactivated in many solid tumors through a nonmutational mechanism. We recently found that TET2 mediates the interferon-JAK-STAT pathway to stimulate chemokine expression and tumor infiltration of lymphocytes (TILs). TET2 is monoubiquitylated at K1299, which promotes its activity. Here, we report that USP15 is a TET2 deubiquitinase and inhibitor. USP15 catalyzes the removal of K1299-linked monoubiquitin and negatively regulates TET2 activity. Gene expression profiling demonstrates that TET2 and USP15 oppositely regulate genes involved in multiple inflammatory pathways, and TET2 is a major target of USP15 function. Deletion of Usp15 in melanoma stimulates chemokine expression and TILs in a TET2-dependent manner, leading to increased response to immunotherapy and extended life span of tumor-bearing mice. These results reveal a previously unknown regulator of TET2 activity and suggest USP15 as a potential therapeutic target for immunotherapy of solid tumors.

12.
PLoS One ; 15(6): e0235279, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32598341

RESUMO

Blue laser/light imaging (BLI) is an image-enhanced endoscopy (IEE) technique that can provide an accurate diagnosis by closely observing the surface structure of various colonic lesions. However, complete correspondence between endoscopic images and pathological images has not been demonstrated. The aim of this study was to accurately compare endoscopic images and the pathological images using a three-dimensionally (3D) reconstructed pathological model. Continuous thin layer sections were prepared from colonic tissue specimens and immunohistochemically stained for CD34 and CAM5.2. Three-dimensional reconstructed images were created by superimposing immunohistochemically stained pathological images. The endoscopic image with magnifying BLI was compared with the top view of the 3D reconstructed image to identify any one-to-one correspondence between the endoscopic images and histopathological images using the gland orifices and microvessels as a guide. Using 3D reconstructed pathological images, we were able to identify the location on the endoscope image in cases of colonic adenocarcinoma, adenoma and normal mucosa. As a result, the horizontal plane of the endoscopic image and the vertical plane of the 2D pathological specimen were able to be compared, and we successfully determined the visible blood vessel depth and performed a detailed evaluation on magnifying BLI. Examples are as follows: (1) The median vasculature depth from the mucosal surface that could be recognized as vasculature on magnifying BLI was 29.4 µm. The median depth of unrecognizable vessels on magnifying BLI was 218.8 µm, which was significantly deeper than recognizable vessels. (2) Some brownish structures were suggested to potentially be not only dense vessels, vessel expansions, corrupted vessels but also bleeding or extravasation of erythrocytes. Overall, we demonstrated a new approach to matching endoscopic images and pathological findings using a 3D-reconstructed pathological model immunohistochemically stained for CD34 and CAM5.2. This approach may increase the overall understanding of endoscopic images and positively contribute to making more accurate endoscopic diagnoses.


Assuntos
Adenocarcinoma/patologia , Neoplasias do Colo/patologia , Detecção Precoce de Câncer/métodos , Endoscopia/métodos , Imageamento Tridimensional/métodos , Idoso , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Imagem de Banda Estreita
13.
iScience ; 23(4): 101030, 2020 Apr 24.
Artigo em Inglês | MEDLINE | ID: mdl-32299058

RESUMO

Haploinsufficiency of SETD5 is implicated in syndromic autism spectrum disorder (ASD), but the molecular mechanism underlying the pathological role of this protein has remained unclear. We have now shown that Setd5+/- mice manifest ASD-related behavioral phenotypes and that the expression of ribosomal protein genes and rDNA is disturbed in the brain of these mice. SETD5 recruited the HDAC3 complex to the rDNA promoter, resulting in removal of the histone mark H4K16ac and its reader protein TIP5, a repressor of rDNA expression. Depletion of SETD5 attenuated rDNA expression, translational activity, and neural cell proliferation, whereas ablation of TIP5 in SETD5-deficient cells rescued these effects. Translation of cyclin D1 mRNA was specifically down-regulated in SETD5-insufficient cells. Our results thus suggest that SETD5 positively regulates rDNA expression via an HDAC3-mediated epigenetic mechanism and that such regulation is essential for translation of cyclin D1 mRNA and neural cell proliferation.

14.
Adv Exp Med Biol ; 1217: 147-171, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-31898227

RESUMO

The CRL1 complex, also known as the SCF complex, is a ubiquitin ligase that in mammals consists of an adaptor protein (SKP1), a scaffold protein (CUL1), a RING finger protein (RBX1, also known as ROC1), and one of about 70 F-box proteins. Given that the F-box proteins determine the substrate specificity of the CRL1 complex, the variety of these proteins allows the generation of a large number of ubiquitin ligases that promote the degradation or regulate the function of many substrate proteins and thereby control numerous key cellular processes. The physiological and pathological functions of these many CRL1 ubiquitin ligases have been studied by the generation and characterization of knockout mouse models that lack specific CRL1 components. In this chapter, we provide a comprehensive overview of these mouse models and discuss the role of each CRL1 component in mouse physiology and pathology.


Assuntos
Proteínas Culina/metabolismo , Proteínas Ligases SKP Culina F-Box/metabolismo , Animais , Proteínas Culina/química , Camundongos , Camundongos Knockout , Modelos Animais , Complexos Multiproteicos/química , Complexos Multiproteicos/metabolismo , Proteínas Ligases SKP Culina F-Box/química
15.
Hum Mol Genet ; 28(20): 3486-3497, 2019 10 15.
Artigo em Inglês | MEDLINE | ID: mdl-31577344

RESUMO

Amyotrophic lateral sclerosis (ALS) is an adult-onset motor neuron disease characterized by a progressive decline in motor function. Genetic analyses have identified several genes mutated in ALS patients, and one of them is Cyclin F gene (CCNF), the product of which (Cyclin F) serves as the substrate-binding module of a SKP1-CUL1-F-box protein (SCF) ubiquitin ligase complex. However, the role of Cyclin F in ALS pathogenesis has remained unclear. Here, we show that Cyclin F binds to valosin-containing protein (VCP), which is also reported to be mutated in ALS, and that the two proteins colocalize in the nucleus. VCP was found to bind to the NH2-terminal region of Cyclin F and was not ubiquitylated by SCFCyclin F in transfected cells. Instead, the ATPase activity of VCP was enhanced by Cyclin F in vitro. Furthermore, whereas ALS-associated mutations of CCNF did not affect the stability of Cyclin F or disrupt formation of the SCFCyclin F complex, amino acid substitutions in the VCP binding region increased the binding ability of Cyclin F to VCP and activity of VCP as well as mislocalization of the protein in the cytoplasm. We also provided evidence that the ATPase activity of VCP promotes cytoplasmic aggregation of transactivation responsive region (TAR) DNA-binding protein 43, which is commonly observed in degenerating neurons in ALS patients. Given that mutations of VCP identified in ALS patients also increase its ATPase activity, our results suggest that Cyclin F mutations may contribute to ALS pathogenesis by increasing the ATPase activity of VCP in the cytoplasm, which in turn increases TDP-43 aggregates.


Assuntos
Esclerose Lateral Amiotrófica/genética , Ciclinas/metabolismo , Citoplasma/metabolismo , Mutação/genética , Proteínas Ligases SKP Culina F-Box/metabolismo , Proteína com Valosina/metabolismo , Adenosina Trifosfatases/genética , Adenosina Trifosfatases/metabolismo , Animais , Ciclinas/genética , Masculino , Camundongos , Ligação Proteica , Proteínas Ligases SKP Culina F-Box/genética , Ubiquitinação , Proteína com Valosina/genética
16.
EBioMedicine ; 45: 362-378, 2019 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-31262712

RESUMO

BACKGROUND: The characteristic structure of motor neurons (MNs), particularly of the long axons, becomes damaged in the early stages of amyotrophic lateral sclerosis (ALS). However, the molecular pathophysiology of axonal degeneration remains to be fully elucidated. METHOD: Two sets of isogenic human-induced pluripotent stem cell (hiPSCs)-derived MNs possessing the single amino acid difference (p.H517D) in the fused in sarcoma (FUS) were constructed. By combining MN reporter lentivirus, MN specific phenotype was analyzed. Moreover, RNA profiling of isolated axons were conducted by applying the microfluidic devices that enable axon bundles to be produced for omics analysis. The relationship between the target gene, which was identified as a pathological candidate in ALS with RNA-sequencing, and the MN phenotype was confirmed by intervention with si-RNA or overexpression to hiPSCs-derived MNs and even in vivo. The commonality was further confirmed with other ALS-causative mutant hiPSCs-derived MNs and human pathology. FINDINGS: We identified aberrant increasing of axon branchings in FUS-mutant hiPSCs-derived MN axons compared with isogenic controls as a novel phenotype. We identified increased level of Fos-B mRNA, the binding target of FUS, in FUS-mutant MNs. While Fos-B reduction using si-RNA or an inhibitor ameliorated the observed aberrant axon branching, Fos-B overexpression resulted in aberrant axon branching even in vivo. The commonality of those phenotypes was further confirmed with other ALS causative mutation than FUS. INTERPRETATION: Analyzing the axonal fraction of hiPSC-derived MNs using microfluidic devices revealed that Fos-B is a key regulator of FUS-mutant axon branching. FUND: Japan Agency for Medical Research and development; Japanese Ministry of Education, Culture, Sports, Science and Technology Clinical Research, Innovation and Education Center, Tohoku University Hospital; Japan Intractable Diseases (Nanbyo) Research Foundation; the Kanae Foundation for the Promotion of Medical Science; and "Inochi-no-Iro" ALS research grant.


Assuntos
Esclerose Lateral Amiotrófica/genética , Proteínas Proto-Oncogênicas c-fos/genética , Proteína FUS de Ligação a RNA/genética , Esclerose Lateral Amiotrófica/patologia , Animais , Axônios/metabolismo , Axônios/patologia , Diferenciação Celular/genética , Linhagem Celular , Edição de Genes/métodos , Sequenciamento de Nucleotídeos em Larga Escala , Humanos , Células-Tronco Pluripotentes Induzidas/metabolismo , Lentivirus/genética , Neurônios Motores/metabolismo , Mutação , Neurogênese/genética , Fenótipo , RNA Interferente Pequeno/genética
17.
Dev Biol ; 445(2): 178-188, 2019 01 15.
Artigo em Inglês | MEDLINE | ID: mdl-30391586

RESUMO

ß-TrCP is the substrate recognition subunit of an SCF-type ubiquitin ligase. We recently showed that deletion of the genes for both ß-TrCP1 and ß-TrCP2 paralogs in germ cells of male mice resulted in accumulation of the transcription factor DMRT1 and spermatogenic failure, whereas systemic ß-TrCP1 knockout combined with ß-TrCP2 knockdown had previously been shown to lead to disruption of testicular organization and accumulation of the transcription factor SNAIL. Here we investigated ß-TrCP function in Sertoli cells by generating mice with targeted deletion of the ß-TrCP2 gene in Sertoli cells on a background of whole-body ß-TrCP1 knockout. Loss of ß-TrCP in Sertoli cells caused infertility due to a reduction in the number of mature sperm. Whereas spermatogonia were not affected, male germ cells entered meiosis prematurely and the number of round spermatids was reduced in the mutant mice. Extracts of Sertoli cells and of the testis from the mutant mice manifested accumulation of SNAIL, and expression of the SNAIL target gene for E-cadherin was down-regulated in Sertoli cells from these animals. Our results indicate that ß-TrCP in Sertoli cells regulates Sertoli cell-germ cell interaction through degradation of SNAIL, with such regulation being critical for sperm development.


Assuntos
Células de Sertoli/metabolismo , Espermatogênese/fisiologia , Proteínas Contendo Repetições de beta-Transducina/metabolismo , Animais , Infertilidade Masculina/genética , Infertilidade Masculina/metabolismo , Infertilidade Masculina/patologia , Masculino , Meiose/genética , Meiose/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , RNA/genética , RNA/metabolismo , Células de Sertoli/patologia , Fatores de Transcrição da Família Snail/metabolismo , Espermátides/metabolismo , Espermátides/patologia , Espermatogênese/genética , Espermatogônias/metabolismo , Espermatogônias/patologia , Proteínas Contendo Repetições de beta-Transducina/deficiência , Proteínas Contendo Repetições de beta-Transducina/genética
18.
Mol Cell ; 70(5): 961-970.e5, 2018 06 07.
Artigo em Inglês | MEDLINE | ID: mdl-29883611

RESUMO

HIV-1 expresses several accessory proteins to counteract host anti-viral restriction factors to facilitate viral replication and disease progression. One such protein, Vpr, has been implicated in affecting multiple cellular processes, but its mechanism remains elusive. Here we report that Vpr targets TET2 for polyubiquitylation by the VprBP-DDB1-CUL4-ROC1 E3 ligase and subsequent degradation. Genetic inactivation or Vpr-mediated degradation of TET2 enhances HIV-1 replication and substantially sustains expression of the pro-inflammatory cytokine interleukin-6 (IL-6). This process correlates with reduced recruitment of histone deacetylase 1 and 2 to the IL-6 promoter, thus enhancing its histone H3 acetylation level during resolution phase. Blocking IL-6 signaling reduced the ability of Vpr to enhance HIV-1 replication. We conclude that HIV-1 Vpr degrades TET2 to sustain IL-6 expression to enhance viral replication and disease progression. These results suggest that disrupting the Vpr-TET2-IL6 axis may prove clinically beneficial to reduce both viral replication and inflammation during HIV-1 infection.


Assuntos
Proteínas de Transporte/metabolismo , Proteínas de Ligação a DNA/metabolismo , HIV-1/metabolismo , Mediadores da Inflamação/metabolismo , Interleucina-6/metabolismo , Monócitos/virologia , Proteínas Proto-Oncogênicas/metabolismo , Replicação Viral , Produtos do Gene vpr do Vírus da Imunodeficiência Humana/metabolismo , Sítios de Ligação , Proteínas de Transporte/genética , Proteínas de Ligação a DNA/genética , Dioxigenases , Células HEK293 , HIV-1/genética , HIV-1/crescimento & desenvolvimento , HIV-1/patogenicidade , Histona Desacetilase 1/metabolismo , Histona Desacetilase 2/metabolismo , Interações Hospedeiro-Patógeno , Humanos , Interleucina-6/genética , Células Jurkat , Monócitos/enzimologia , Regiões Promotoras Genéticas , Proteínas Serina-Treonina Quinases , Proteólise , Proteínas Proto-Oncogênicas/genética , Transdução de Sinais , Células THP-1 , Ubiquitina-Proteína Ligases , Ubiquitinação , Produtos do Gene vpr do Vírus da Imunodeficiência Humana/genética
19.
Genet Med ; 20(5): 486-494, 2018 04.
Artigo em Inglês | MEDLINE | ID: mdl-28817111

RESUMO

PurposeNeonatal encephalopathy, which is characterized by a decreased level of consciousness, occurs in 1-7/1,000 live-term births. In more than half of term newborns, there is no identifiable etiological factor. To identify underlying genetic defects, we applied whole-exome sequencing (WES) in term newborns with neonatal encephalopathy as a prospective cohort study.MethodsTerm newborns with neonatal encephalopathy and no history of perinatal asphyxia were included. WES was performed using patient and both parents' DNA.ResultsNineteen patients fulfilling inclusion criteria were enrolled. Five patients were excluded owing to withdrawal of consent, no parental DNA samples, or a genetic diagnosis prior to WES. Fourteen patients underwent WES. We confirmed a genetic diagnosis in five patients (36%): epileptic encephalopathy associated with autosomal dominant de novo variants in SCN2A (p.Met1545Val), KCNQ2 (p.Asp212Tyr), and GNAO1 (p.Gly40Arg); lipoic acid synthetase deficiency due to compound heterozygous variants in LIAS (p.Ala253Pro and p.His236Gln); and encephalopathy associated with an X-linked variant in CUL4B (p.Asn211Ser).ConclusionWES is helpful at arriving genetic diagnoses in neonatal encephalopathy and/or seizures and brain damage. It will increase our understanding and probably enable us to develop targeted neuroprotective treatment strategies.


Assuntos
Encefalopatias/genética , Predisposição Genética para Doença , Estudo de Associação Genômica Ampla , Doenças do Recém-Nascido/genética , Encefalopatias/diagnóstico , Eletroencefalografia , Feminino , Humanos , Recém-Nascido , Doenças do Recém-Nascido/diagnóstico , Imageamento por Ressonância Magnética , Masculino , Gravidez , Análise de Sequência de DNA , Sequenciamento do Exoma
20.
Mol Cell Biol ; 38(5)2018 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-29203640

RESUMO

Recognition of gene promoters by RNA polymerase II is mediated by general transcription factor IID (TFIID), which has been thought to be a static complex and to play a passive role in the regulation of gene expression under the instruction of gene-specific transcription factors. Here we show that transforming growth factor ß (TGF-ß) induced degradation of the TFIID subunit TAF7 in cultured mouse mammary epithelial cells and that this effect was required for proliferative arrest in response to TGF-ß stimulation. TGF-ß stimulated transcription of the gene for the ubiquitin ligase TRIM26, which was shown to ubiquitylate TAF7 and thereby to target it for proteasomal degradation. Sustained exposure of cells to TGF-ß resulted in recovery from proliferative arrest in association with amplification of the Myc proto-oncogene, with MYC inhibiting TRIM26 induction by TGF-ß. Our data thus show that TFIID is not simply a general mediator of transcription but contributes to the regulation of transcription in response to cell stimulation, playing a key role in the cytostatic function of TGF-ß.


Assuntos
Proteínas Proto-Oncogênicas c-myc/metabolismo , Fatores Associados à Proteína de Ligação a TATA/metabolismo , Fator de Transcrição TFIID/metabolismo , Fator de Crescimento Transformador beta/farmacologia , Proteínas com Motivo Tripartido/metabolismo , Ubiquitina-Proteína Ligases/metabolismo , Animais , Divisão Celular/fisiologia , Proliferação de Células/efeitos dos fármacos , Proliferação de Células/fisiologia , Genes myc , Glândulas Mamárias Animais/citologia , Glândulas Mamárias Animais/efeitos dos fármacos , Glândulas Mamárias Animais/metabolismo , Camundongos , Proteínas Proto-Oncogênicas c-myc/genética , Elementos Reguladores de Transcrição , Fatores Associados à Proteína de Ligação a TATA/antagonistas & inibidores , Fatores Associados à Proteína de Ligação a TATA/genética , Fator de Transcrição TFIID/antagonistas & inibidores , Fator de Transcrição TFIID/genética , Proteínas com Motivo Tripartido/genética , Células Tumorais Cultivadas , Ubiquitina-Proteína Ligases/genética , Ubiquitinação
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA