Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 65
Filtrar
1.
PLoS Biol ; 21(8): e3002281, 2023 08.
Artigo em Inglês | MEDLINE | ID: mdl-37643163

RESUMO

The central circadian clock of the suprachiasmatic nucleus (SCN) is a network consisting of various types of neurons and glial cells. Individual cells have the autonomous molecular machinery of a cellular clock, but their intrinsic periods vary considerably. Here, we show that arginine vasopressin (AVP) neurons set the ensemble period of the SCN network in vivo to control the circadian behavior rhythm. Artificial lengthening of cellular periods by deleting casein kinase 1 delta (CK1δ) in the whole SCN lengthened the free-running period of behavior rhythm to an extent similar to CK1δ deletion specific to AVP neurons. However, in SCN slices, PER2::LUC reporter rhythms of these mice only partially and transiently recapitulated the period lengthening, showing a dissociation between the SCN shell and core with a period instability in the shell. In contrast, in vivo calcium rhythms of both AVP and vasoactive intestinal peptide (VIP) neurons in the SCN of freely moving mice demonstrated stably lengthened periods similar to the behavioral rhythm upon AVP neuron-specific CK1δ deletion, without changing the phase relationships between each other. Furthermore, optogenetic activation of AVP neurons acutely induced calcium increase in VIP neurons in vivo. These results indicate that AVP neurons regulate other SCN neurons, such as VIP neurons, in vivo and thus act as a primary determinant of the SCN ensemble period.


Assuntos
Arginina Vasopressina , Cálcio , Animais , Camundongos , Neurônios , Núcleo Supraquiasmático , Neuroglia , Peptídeo Intestinal Vasoativo
2.
Endocr J ; 71(3): 245-252, 2024 Mar 28.
Artigo em Inglês | MEDLINE | ID: mdl-38143086

RESUMO

11Beta-hydroxysteroid dehydrogenase 1 (11ß-HSD1) is a key enzyme involved in metabolic syndrome. Transcript-specific epigenetic regulation of the gene encoding 11ß-HSD1 (HSD11B1) has been reported. We examined the mRNA level and methylation status of the HSD11B1 promoter region in the adipose tissue of patients with primary aldosteronism (PA). We compared 10 tissue specimens from patients with PA caused by aldosterone-producing adenoma (APA) with 8 adipose tissue specimens from patients with subclinical Cushing's syndrome (SCS) caused by cortisol-producing adenomas, 4 tissue specimens from patients with Cushing's adenoma (Cu), or 7 tissue specimens from patients with non-functioning adrenal adenoma (NFA). PA, SCS, and Cu were diagnosed according to the guideline of the Japan Endocrine Society. The mRNA level of HSD11B1 was quantified using real-time PCR. Isolated DNA was treated with bisulfite and amplified using primers specific to the human HSD11B1 promoter region. The glycohemoglobin level was significantly higher in patients with APA, SCS, or Cu than in those with NFA (p < 0.05). Blood pressure was significantly higher in patients with APA than in those with SCS, Cu, or NFA (p < 0.01). The HSD11B1 mRNA level was significantly increased in the adipose tissues of APA or SCS patients compared with Cu or NFA patients (p < 0.05). The methylation ratio was significantly lower in SCS patients than in APA, Cu, or NFA patients (p < 0.05). HSD11B1 expression is partly controlled by an epigenetic mechanism in human tissues. The pathophysiological role of epigenetic regulation of HSD11B1 expression in adipose tissue requires further study.


Assuntos
Adenoma , Adenoma Adrenocortical , Hiperaldosteronismo , Humanos , 11-beta-Hidroxiesteroide Desidrogenase Tipo 1/genética , 11-beta-Hidroxiesteroide Desidrogenase Tipo 1/metabolismo , Epigênese Genética , Tecido Adiposo/metabolismo , Adenoma Adrenocortical/metabolismo , Hiperaldosteronismo/genética , Hiperaldosteronismo/metabolismo , Adenoma/metabolismo , RNA Mensageiro/metabolismo
3.
Cancer Sci ; 114(10): 3946-3956, 2023 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-37545017

RESUMO

Multitargeted receptor tyrosine kinase inhibitors, including vascular endothelial growth factor (VEGF) inhibitors, such as sunitinib, have been used as the primary targeted agents for patients with recurrent or distant metastasis of advanced renal cell carcinoma (RCC). However, endogenous or acquired sunitinib resistance has become a significant therapeutic problem. Therefore, we focused on mechanisms of sunitinib resistance in RCC. First, we undertook RNA sequencing analysis using previously established sunitinib-resistant RCC (SUR-Caki1, SUR-ACHN, and SUR-A498) cells. The results showed increased expression of secretogranin II (SCG2, chromogranin C) in SUR-RCC cells compared to parental cells. The Cancer Genome Atlas database showed that SCG2 expression was increased in RCC compared to normal renal cells. In addition, the survival rate of the SCG2 high-expression group was significantly lower than that of the RCC low-expression group. Thus, we investigated the involvement of SCG2 in sunitinib-resistant RCC. In vitro analysis showed that migratory and invasive abilities were suppressed by SCG2 knockdown SUR cells. As SCG2 was previously reported to be associated with angiogenesis, we undertook a tube formation assay. The results showed that suppression of SCG2 inhibited angiogenesis. Furthermore, coimmunoprecipitation assays revealed a direct interaction between SCG2 and hypoxia-inducible factor 1α (HIF1α). Expression levels of VEGF-A and VEGF-C downstream of HIF1α were found to be decreased in SCG2 knockdown SUR cells. In conclusion, SCG2 could be associated with sunitinib resistance through VEGF regulation in RCC cells. These findings could lead to a better understanding of the VHL/HIF/VEGF pathway and the development of new therapeutic strategies for sunitinib-resistant RCC.

4.
Biochem Biophys Res Commun ; 669: 19-29, 2023 08 20.
Artigo em Inglês | MEDLINE | ID: mdl-37262949

RESUMO

ASH2L (Absent-Small-Homeotic-2-Like protein) is a core subunit of the COMPASS (COMplex of Proteins ASsociated with Set1) complex, the most notable writer of the methylation of histone H3 lysine 4 (H3K4). The COMPASS complex regulates active promoters or enhancers for gene expression, and its dysfunction is associated with aberrant development and disease. Here, we demonstrated that ASH2L mediated the cell invasion and migration activity of triple-negative breast cancer cells through the interaction with the COMPASS components and the target genomic regions. Transcriptome analysis indicated a potential correlation between ASH2L and the genes involved in inflammatory/immune responses. Among them, we found that the intrinsic expression of IL1B (interleukin 1 beta), an essential proinflammatory gene, was directly regulated by ASH2L. These results revealed a novel role of ASH2L on the maintenance of breast cancer malignancy possibly through H3K4 methylation of the target inflammatory/immune responsive genes.


Assuntos
Histonas , Neoplasias de Mama Triplo Negativas , Humanos , Histonas/metabolismo , Metilação , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Lisina/metabolismo , Neoplasias de Mama Triplo Negativas/genética , Histona-Lisina N-Metiltransferase/genética , Histona-Lisina N-Metiltransferase/metabolismo , Epigênese Genética , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo
5.
Cancer Sci ; 113(10): 3376-3389, 2022 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-35838233

RESUMO

Although the human papillomavirus (HPV) vaccine is effective for preventing cervical cancers, this vaccine does not eliminate pre-existing infections, and alternative strategies have been warranted. Here, we report that FOXP4 is a new target molecule for differentiation therapy of cervical intraepithelial neoplasia (CIN). An immunohistochemical study showed that FOXP4 was expressed in columnar epithelial, reserve, and immature squamous cells, but not in mature squamous cells of the normal uterine cervix. In contrast with normal mature squamous cells, FOXP4 was expressed in atypical squamous cells in CIN and squamous cell carcinoma lesions. The FOXP4-positive areas significantly increased according to the CIN stages from CIN1 to CIN3. In monolayer cultures, downregulation of FOXP4 attenuated proliferation and induced squamous differentiation in CIN1-derived HPV 16-positive W12 cells via an ELF3-dependent pathway. In organotypic raft cultures, FOXP4-downregulated W12 cells showed mature squamous phenotypes of CIN lesions. In human keratinocyte-derived HaCaT cells, FOXP4 downregulation also induced squamous differentiation via an ELF3-dependent pathway. These findings suggest that downregulation of FOXP4 inhibits cell proliferation and promotes the differentiation of atypical cells in CIN lesions. Based on these results, we propose that FOXP4 is a novel target molecule for nonsurgical CIN treatment that inhibits CIN progression by inducing squamous differentiation.


Assuntos
Carcinoma de Células Escamosas , Infecções por Papillomavirus , Displasia do Colo do Útero , Neoplasias do Colo do Útero , Carcinoma de Células Escamosas/patologia , Proteínas de Ligação a DNA , Feminino , Fatores de Transcrição Forkhead , Humanos , Papillomaviridae , Infecções por Papillomavirus/patologia , Proteínas Proto-Oncogênicas c-ets , Sulfonamidas , Fatores de Transcrição , Neoplasias do Colo do Útero/patologia
6.
Hepatology ; 74(4): 1971-1993, 2021 10.
Artigo em Inglês | MEDLINE | ID: mdl-33931882

RESUMO

BACKGROUND AND AIMS: Synthetic cyclin-dependent kinase (CDK) 4/6 inhibitors exert antitumor effects by forcing RB1 in unphosphorylated status, causing not only cell cycle arrest but also cellular senescence, apoptosis, and increased immunogenicity. These agents currently have an indication in advanced breast cancers and are in clinical trials for many other solid tumors. HCC is one of promising targets of CDK4/6 inhibitors. RB family dysfunction is often associated with the initiation of HCC; however, this is revivable, as RB family members are not frequently mutated or deleted in this malignancy. APPROACH AND RESULTS: Loss of all Rb family members in transformation related protein 53 (Trp53)-/- mouse liver resulted in liver tumor reminiscent of human HCC, and re-expression of RB1 sensitized these tumors to a CDK4/6 inhibitor, palbociclib. Introduction of an unphosphorylatable form of RB1 (RB7LP) into multiple liver tumor cell lines induced effects similar to palbociclib. By screening for compounds that enhance the efficacy of RB7LP, we identified an I kappa B kinase (IKK)ß inhibitor Bay 11-7082. Consistently, RB7LP expression and treatment with palbociclib enhanced IKKα/ß phosphorylation and NF-κB activation. Combination therapy using palbociclib with Bay 11-7082 was significantly more effective in hepatoblastoma and HCC treatment than single administration. Moreover, blockade of IKK-NF-κB or AKT pathway enhanced effects of palbociclib on RB1-intact KRAS Kirsten rat sarcoma viral oncogene homolog mutated lung and colon cancers. CONCLUSIONS: In conclusion, CDK4/6 inhibitors have a potential to treat a wide variety of RB1-intact cancers including HCC when combined with an appropriate kinase inhibitor.


Assuntos
Carcinoma Hepatocelular/tratamento farmacológico , Proliferação de Células/efeitos dos fármacos , Neoplasias Hepáticas/tratamento farmacológico , Piperazinas/farmacologia , Inibidores de Proteínas Quinases/farmacologia , Piridinas/farmacologia , Aminopiridinas/farmacologia , Aminopiridinas/uso terapêutico , Animais , Benzimidazóis/farmacologia , Benzimidazóis/uso terapêutico , Carcinoma Hepatocelular/genética , Proliferação de Células/genética , Sobrevivência Celular/efeitos dos fármacos , Quinase 4 Dependente de Ciclina/antagonistas & inibidores , Quinase 6 Dependente de Ciclina/antagonistas & inibidores , Células Hep G2 , Humanos , Técnicas In Vitro , Neoplasias Hepáticas/genética , Neoplasias Hepáticas Experimentais/tratamento farmacológico , Neoplasias Hepáticas Experimentais/genética , Camundongos , Transplante de Neoplasias , Piperazinas/uso terapêutico , Inibidores de Proteínas Quinases/uso terapêutico , Purinas/farmacologia , Purinas/uso terapêutico , Piridinas/uso terapêutico , Proteína do Retinoblastoma , Proteína Supressora de Tumor p53/genética , Proteínas de Xenopus
7.
Int J Mol Sci ; 23(14)2022 Jul 11.
Artigo em Inglês | MEDLINE | ID: mdl-35886985

RESUMO

Recently, it was demonstrated that the expression of BMAL1 was decreased in the endometrium of women suffering from recurrent spontaneous abortion. To investigate the pathological roles of uterine clock genes during pregnancy, we produced conditional deletion of uterine Bmal1 (cKO) mice and found that cKO mice could receive embryo implantation but not sustain pregnancy. Gene ontology analysis of microarray suggested that uterine NK (uNK) cell function was suppressed in cKO mice. Histological examination revealed the poor formation of maternal vascular spaces in the placenta. In contrast to WT mice, uNK cells in the spongiotrophoblast layer, where maternal uNK cells are directly in contact with fetal trophoblast, hardly expressed an immunosuppressive NK marker, CD161, in cKO mice. By progesterone supplementation, pregnancy could be sustained until the end of pregnancy in some cKO mice. Although this treatment did not improve the structural abnormalities of the placenta, it recruited CD161-positive NK cells into the spongiotrophoblast layer in cKO mice. These findings indicate that the uterine clock system may be critical for pregnancy maintenance after embryo implantation.


Assuntos
Fatores de Transcrição ARNTL , Morte Fetal , Neovascularização Patológica , Placenta , Fatores de Transcrição ARNTL/genética , Fatores de Transcrição ARNTL/imunologia , Animais , Implantação do Embrião/genética , Feminino , Morte Fetal/etiologia , Células Matadoras Naturais/imunologia , Glicoproteínas de Membrana/imunologia , Camundongos , Neovascularização Patológica/genética , Neovascularização Patológica/imunologia , Placenta/irrigação sanguínea , Placenta/imunologia , Gravidez/genética , Gravidez/imunologia , Complicações na Gravidez/genética , Complicações na Gravidez/imunologia , Natimorto/genética , Útero/imunologia
8.
Cancer Sci ; 112(9): 3810-3821, 2021 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-34145929

RESUMO

Epidermal growth factor receptor tyrosine kinase inhibitors (EGFR-TKIs) are effective in patients with non-small-cell lung cancer (NSCLC) harboring EGFR mutations. However, due to acquired resistance to EGFR-TKIs, even patients on third-generation osimertinib have a poor prognosis. Resistance mechanisms are still not fully understood. Here, we demonstrate that the increased expression of MUSASHI-2 (MSI2), an RNA-binding protein, is a novel mechanism for resistance to EGFR-TKIs. We found that after a long-term exposure to gefitinib, the first-generation EGFR-TKI lung cancer cells harboring the EGFR-TKI-sensitive mutations became resistant to both gefitinib and osimertinib. Although other mutations in EGFR were not found, expression levels of Nanog, a stemness core protein, and activities of aldehyde dehydrogenase (ALDH) were increased, suggesting that cancer stem-like properties were increased. Transcriptome analysis revealed that MSI2 was among the stemness-related genes highly upregulated in EGFR-TKI-resistant cells. Knockdown of MSI2 reduced cancer stem-like properties, including the expression levels of Nanog, a core stemness factor. We demonstrated that knockdown of MSI2 restored sensitivity to osimertinib or gefitinib in EGFR-TKI-resistant cells to levels similar to those of parental cells in vitro. An RNA immunoprecipitation (RIP) assay revealed that antibodies against MSI2 were bound to Nanog mRNA, suggesting that MSI2 increases Nanog expression by binding to Nanog mRNA. Moreover, overexpression of MSI2 or Nanog conferred resistance to osimertinib or gefitinib in parental cells. Finally, MSI2 knockdown greatly increased the sensitivity to osimertinib in vivo. Collectively, our findings provide proof of principle that targeting the MSI2-Nanog axis in combination with EGFR-TKIs would effectively prevent the emergence of acquired resistance.


Assuntos
Acrilamidas/farmacologia , Adenocarcinoma de Pulmão/metabolismo , Compostos de Anilina/farmacologia , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Gefitinibe/farmacologia , Neoplasias Pulmonares/metabolismo , Inibidores de Proteínas Quinases/farmacologia , Proteínas de Ligação a RNA/metabolismo , Regulação para Cima , Células A549 , Acrilamidas/uso terapêutico , Adenocarcinoma de Pulmão/patologia , Compostos de Anilina/uso terapêutico , Carcinoma Pulmonar de Células não Pequenas/tratamento farmacológico , Carcinoma Pulmonar de Células não Pequenas/genética , Carcinoma Pulmonar de Células não Pequenas/metabolismo , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/genética , Resistencia a Medicamentos Antineoplásicos/genética , Receptores ErbB/antagonistas & inibidores , Receptores ErbB/genética , Gefitinibe/uso terapêutico , Perfilação da Expressão Gênica , Técnicas de Silenciamento de Genes , Humanos , Neoplasias Pulmonares/patologia , Mutação , Proteína Homeobox Nanog/metabolismo , Inibidores de Proteínas Quinases/uso terapêutico , Proteínas de Ligação a RNA/genética , Transcriptoma , Transfecção
9.
Cancer Sci ; 111(7): 2336-2348, 2020 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-32437590

RESUMO

Dietary fat consumption during accelerated stages of mammary gland development, such as peripubertal maturation or pregnancy, is known to increase the risk for breast cancer. However, the underlying molecular mechanisms are not fully understood. Here we examined the gene expression profile of mouse mammary epithelial cells (MMECs) on exposure to a high-fat diet (HFD) or control diet (CD). Trp53-/- female mice were fed with the experimental diets for 5 weeks during the peripubertal period (3-8 weeks of age). The treatment showed no significant difference in body weight between the HFD-fed mice and CD-fed mice. However, gene set enrichment analysis predicted a significant enrichment of c-Myc target genes in animals fed HFD. Furthermore, we detected enhanced activity and stabilization of c-Myc protein in MMECs exposed to a HFD. This was accompanied by augmented c-Myc phosphorylation at S62 with a concomitant increase in ERK phosphorylation. Moreover, MMECs derived from HFD-fed Trp53-/- mouse showed increased colony- and sphere-forming potential that was dependent on c-Myc. Further, oleic acid, a major fatty acid constituent of the HFD, and TAK-875, an agonist to G protein-coupled receptor 40 (a receptor for oleic acid), enhanced c-Myc stabilization and MMEC proliferation. Overall, our data indicate that HFD influences MMECs by stabilizing an oncoprotein, pointing to a novel mechanism underlying dietary fat-mediated mammary carcinogenesis.


Assuntos
Dieta Hiperlipídica , Epitélio/metabolismo , Glândulas Mamárias Animais/metabolismo , Glândulas Mamárias Animais/patologia , Proteínas Proto-Oncogênicas c-myc/metabolismo , Maturidade Sexual , Animais , Linhagem Celular Tumoral , Feminino , Genes p53 , Humanos , Camundongos , Camundongos Knockout , Obesidade/etiologia , Obesidade/metabolismo , Proteínas Proto-Oncogênicas c-myc/genética , Puberdade , Células Tumorais Cultivadas
10.
Cell Tissue Res ; 381(2): 229-237, 2020 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-32418130

RESUMO

The submandibular gland (SMG) of newborn mice has no mature acini but has the rudiments of acini called terminal tubules (TT). The TT are composed of TT cells with dark secretory granules and proacinar cells with lighter secretory granules, the latter being considered the immediate precursor of mature acinar cells. TT cells contain a specific secretory protein, submandibular gland protein C (SMGC) and they decrease in number postnatally at a higher rate in males than in females. In the present study, in order to clarify the biological roles of TT cells and their secretory product SMGC, we generated a knockout (KO) mouse strain deficient in SMGC. The KO mice of both sexes grew normally, had normal reproductive capacity and had normal acinar and duct systems in the SMG in adult ages. However, through the neonatal and early postnatal stages, the KO mice were deficient not only in the production of SMGC but also in TT cells. With electron microscopy of the SMG of newborn KO mice, TT cells with characteristic granules were absent and replaced by undifferentiated ductal cells, whereas proacinar cells were normal. These results suggested that the absence of SMGC inhibits the development of TT cells and that the absence of SMGC and TT cells has no notable influence on the postnatal development of the acinar and duct systems in the SMG.


Assuntos
Células Acinares , Diferenciação Celular , Mucinas/fisiologia , Glândula Submandibular , Células Acinares/citologia , Células Acinares/metabolismo , Animais , Animais Recém-Nascidos , Feminino , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Glândula Submandibular/citologia , Glândula Submandibular/metabolismo
11.
EMBO Rep ; 19(1): 73-88, 2018 01.
Artigo em Inglês | MEDLINE | ID: mdl-29217659

RESUMO

p63, more specifically its ΔNp63α isoform, plays essential roles in squamous cell carcinomas (SCCs), yet the mechanisms controlling its nuclear transport remain unknown. Nucleoporins (NUPs) are a family of proteins building nuclear pore complexes (NPC) and mediating nuclear transport across the nuclear envelope. Recent evidence suggests a cell type-specific function for certain NUPs; however, the significance of NUPs in SCC biology remains unknown. In this study, we show that nucleoporin 62 (NUP62) is highly expressed in stratified squamous epithelia and is further elevated in SCCs. Depletion of NUP62 inhibits proliferation and augments differentiation of SCC cells. The impaired ability to maintain the undifferentiated status is associated with defects in ΔNp63α nuclear transport. We further find that differentiation-inducible Rho kinase reduces the interaction between NUP62 and ΔNp63α by phosphorylation of phenylalanine-glycine regions of NUP62, attenuating ΔNp63α nuclear import. Our results characterize NUP62 as a gatekeeper for ΔNp63α and uncover its role in the control of cell fate through regulation of ΔNp63α nuclear transport in SCC.


Assuntos
Carcinoma de Células Escamosas/genética , Regulação Neoplásica da Expressão Gênica , Neoplasias de Cabeça e Pescoço/genética , Glicoproteínas de Membrana/genética , Proteínas de Membrana/genética , Complexo de Proteínas Formadoras de Poros Nucleares/genética , Neoplasias Cutâneas/genética , Neoplasias do Colo do Útero/genética , Quinases Associadas a rho/genética , Transporte Ativo do Núcleo Celular/genética , Sequência de Aminoácidos , Atlas como Assunto , Carcinoma de Células Escamosas/metabolismo , Carcinoma de Células Escamosas/patologia , Diferenciação Celular , Linhagem Celular Tumoral , Núcleo Celular/metabolismo , Proliferação de Células , Biologia Computacional , Citosol/metabolismo , Feminino , Células HEK293 , Neoplasias de Cabeça e Pescoço/metabolismo , Neoplasias de Cabeça e Pescoço/patologia , Humanos , Glicoproteínas de Membrana/metabolismo , Proteínas de Membrana/metabolismo , Complexo de Proteínas Formadoras de Poros Nucleares/metabolismo , Fosforilação , Fatores de Troca de Nucleotídeo Guanina Rho/genética , Fatores de Troca de Nucleotídeo Guanina Rho/metabolismo , Transdução de Sinais , Neoplasias Cutâneas/metabolismo , Neoplasias Cutâneas/patologia , Neoplasias do Colo do Útero/metabolismo , Neoplasias do Colo do Útero/patologia , Quinases Associadas a rho/metabolismo
12.
Brain ; 142(3): 560-573, 2019 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-30715177

RESUMO

The leukodystrophies cause severe neurodevelopmental defects from birth and follow an incurable and progressive course that often leads to premature death. It has recently been reported that abnormalities in aminoacyl t-RNA synthetase (ARS) genes are linked to various unique leukodystrophies and leukoencephalopathies. Aminoacyl t-RNA synthetase proteins are fundamentally known as the first enzymes of translation, catalysing the conjugation of amino acids to cognate tRNAs for protein synthesis. It is known that certain aminoacyl t-RNA synthetase have multiple non-canonical roles in both transcription and translation, and their disruption results in varied and complicated phenotypes. We clinically and genetically studied seven patients (six male and one female; aged 2 to 12 years) from five unrelated families who all showed the same phenotypes of severe developmental delay or arrest (7/7), hypotonia (6/7), deafness (7/7) and inability to speak (6/7). The subjects further developed intractable epilepsy (7/7) and nystagmus (6/6) with increasing age. They demonstrated characteristic laboratory data, including increased lactate and/or pyruvate levels (7/7), and imaging findings (7/7), including calcification and abnormal signals in the white matter and pathological involvement (2/2) of the corticospinal tracts. Through whole-exome sequencing, we discovered genetic abnormalities in lysyl-tRNA synthetase (KARS). All patients harboured the variant [c.1786C>T, p.Leu596Phe] KARS isoform 1 ([c.1702C>T, p.Leu568Phe] of KARS isoform 2) either in the homozygous state or compound heterozygous state with the following KARS variants, [c.879+1G>A; c.1786C>T, p.Glu252_Glu293del; p.Leu596Phe] ([c.795+1G>A; c.1702C>T, p.Glu224_Glu255del; p.Leu568Phe]) and [c.650G>A; c.1786C>T, p.Gly217Asp; p.Leu596Phe] ([c.566G>A; c.1702C>T, p.Gly189Asp; p.Leu568Phe]). Moreover, similarly disrupted lysyl-tRNA synthetase (LysRS) proteins showed reduced enzymatic activities and abnormal CNSs in Xenopus embryos. Additionally, LysRS acts as a non-canonical inducer of the immune response and has transcriptional activity. We speculated that the complex functions of the abnormal LysRS proteins led to the severe phenotypes in our patients. These KARS pathological variants are novel, including the variant [c.1786C>T; p.Leu596Phe] (c.1702C>T; p.Leu568Phe) shared by all patients in the homozygous or compound-heterozygous state. This common position may play an important role in the development of severe progressive leukodystrophy. Further research is warranted to further elucidate this relationship and to investigate how specific mutated LysRS proteins function to understand the broad spectrum of KARS-related diseases.


Assuntos
Leucodistrofia de Células Globoides/genética , Leucodistrofia de Células Globoides/fisiopatologia , Lisina-tRNA Ligase/genética , Aminoacil-tRNA Sintetases/genética , Aminoacil-tRNA Sintetases/fisiologia , Animais , Criança , Pré-Escolar , Modelos Animais de Doenças , Feminino , Homozigoto , Humanos , Leucoencefalopatias/genética , Lisina-tRNA Ligase/fisiologia , Masculino , Mutação , Linhagem , Fenótipo , Sequenciamento do Exoma , Xenopus laevis
13.
PLoS Genet ; 12(1): e1005802, 2016 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-26820155

RESUMO

Previously, we identified an adolescent idiopathic scoliosis susceptibility locus near human ladybird homeobox 1 (LBX1) and FLJ41350 by a genome-wide association study. Here, we characterized the associated non-coding variant and investigated the function of these genes. A chromosome conformation capture assay revealed that the genome region with the most significantly associated single nucleotide polymorphism (rs11190870) physically interacted with the promoter region of LBX1-FLJ41350. The promoter in the direction of LBX1, combined with a 590-bp region including rs11190870, had higher transcriptional activity with the risk allele than that with the non-risk allele in HEK 293T cells. The ubiquitous overexpression of human LBX1 or either of the zebrafish lbx genes (lbx1a, lbx1b, and lbx2), but not FLJ41350, in zebrafish embryos caused body curvature followed by death prior to vertebral column formation. Such body axis deformation was not observed in transcription activator-like effector nucleases mediated knockout zebrafish of lbx1b or lbx2. Mosaic expression of lbx1b driven by the GATA2 minimal promoter and the lbx1b enhancer in zebrafish significantly alleviated the embryonic lethal phenotype to allow observation of the later onset of the spinal curvature with or without vertebral malformation. Deformation of the embryonic body axis by lbx1b overexpression was associated with defects in convergent extension, which is a component of the main axis-elongation machinery in gastrulating embryos. In embryos overexpressing lbx1b, wnt5b, a ligand of the non-canonical Wnt/planar cell polarity (PCP) pathway, was significantly downregulated. Injection of mRNA for wnt5b or RhoA, a key downstream effector of Wnt/PCP signaling, rescued the defective convergent extension phenotype and attenuated the lbx1b-induced curvature of the body axis. Thus, our study presents a novel pathological feature of LBX1 and its zebrafish homologs in body axis deformation at various stages of embryonic and subsequent growth in zebrafish.


Assuntos
Proteínas de Homeodomínio/genética , Morfogênese/genética , Escoliose/genética , Fatores de Transcrição/genética , Proteínas Wnt/genética , Proteínas de Peixe-Zebra/genética , Adolescente , Animais , Polaridade Celular/genética , Regulação da Expressão Gênica no Desenvolvimento , Células HEK293 , Proteínas de Homeodomínio/biossíntese , Humanos , Polimorfismo de Nucleotídeo Único , Escoliose/patologia , Fatores de Transcrição/biossíntese , Proteínas Wnt/biossíntese , Via de Sinalização Wnt/genética , Proteína Wnt-5a , Peixe-Zebra , Proteínas de Peixe-Zebra/biossíntese
14.
Cancer Sci ; 109(12): 4045-4055, 2018 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-30281889

RESUMO

At the invasive front of adenocarcinomas, single cells and multicellular structures exist; the latter include glands and cell clusters, such as tumor buddings and poorly differentiated clusters. Recent reports suggest the importance of collective cell migration in metastasis; however, it is technically difficult to observe the movement of multicellular structures in vivo. We utilized MDCK cells as a model for epithelial cells and established a method to quantify their motility in 3D structures in vitro. A single MDCK cell grows as a cell cluster in the gel and later proliferates and forms a cyst. Active K-RAS expression induced rotation of both the cell clusters and the cysts. The rotation speed of cell clusters was 4 times higher than that of cysts. The screening of inhibitors for their effects on cell clusters and cysts revealed that cyclin B1 and ß-catenin were the key molecules for their rotation, respectively. Regulators for cyst rotation, such as vorinostat and ß-catenin, were not effective for inducing cell cluster rotation. These results indicate that the signaling pathways of cell dynamics are different between cell clusters and cysts. As cell clusters are related to lymph node involvement and the prognosis of various carcinomas, our in vitro quantitative system may be useful for the screening of drugs to prevent lymphatic invasion.


Assuntos
Ciclina B1/metabolismo , Células Epiteliais/citologia , Proteínas Proto-Oncogênicas p21(ras)/metabolismo , beta Catenina/metabolismo , Animais , Movimento Celular , Proliferação de Células , Cães , Células Epiteliais/metabolismo , Células Madin Darby de Rim Canino , Modelos Biológicos , Transdução de Sinais , Análise de Célula Única
15.
Am J Pathol ; 187(5): 1177-1185, 2017 May.
Artigo em Inglês | MEDLINE | ID: mdl-28427592

RESUMO

The activation of phosphatidylinositol 3-kinase-AKTs-mammalian target of rapamycin cell signaling pathway leads to cell overgrowth and abnormal migration and results in various types of cortical malformations, such as hemimegalencephaly (HME), focal cortical dysplasia, and tuberous sclerosis complex. However, the pathomechanism underlying abnormal cell migration remains unknown. With the use of fetal mouse brain, we performed causative gene analysis of the resected brain tissues from a patient with HME and investigated the pathogenesis. We obtained a novel somatic mutation of the MTOR gene, having approximately 11% and 7% mutation frequency in the resected brain tissues. Moreover, we revealed that the MTOR mutation resulted in hyperphosphorylation of its downstream molecules, S6 and 4E-binding protein 1, and delayed cell migration on the radial glial fiber and did not affect other cells. We suspect cell-autonomous migration arrest on the radial glial foot by the active MTOR mutation and offer potential explanations for why this may lead to cortical malformations such as HME.


Assuntos
Epilepsia Resistente a Medicamentos/genética , Hemimegalencefalia/genética , Malformações do Desenvolvimento Cortical do Grupo II/genética , Serina-Treonina Quinases TOR/genética , Animais , Células Cultivadas , Epilepsia Resistente a Medicamentos/cirurgia , Eletroencefalografia , Feminino , Hemimegalencefalia/cirurgia , Humanos , Lactente , Malformações do Desenvolvimento Cortical do Grupo II/cirurgia , Camundongos , Tomografia por Emissão de Pósitrons combinada à Tomografia Computadorizada , Serina-Treonina Quinases TOR/metabolismo , Transfecção , Regulação para Cima
17.
Nat Genet ; 37(1): 31-40, 2005 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-15608638

RESUMO

Mutations in MECP2 are associated with Rett syndrome, an X-linked neurodevelopmental disorder. To identify genes targeted by Mecp2, we sequenced 100 in vivo Mecp2-binding sites in mouse brain. Several sequences mapped to an imprinted gene cluster on chromosome 6, including Dlx5 and Dlx6, whose transcription was roughly two times greater in brains of Mecp2-null mice compared with those of wild-type mice. The maternally expressed gene DLX5 showed a loss of imprinting in lymphoblastoid cells from individuals with Rett syndrome. Because Dlx5 regulates production of enzymes that synthesize gamma-aminobutyric acid (GABA), loss of imprinting of Dlx5 may alter GABAergic neuron activity in individuals with Rett syndrome. In mouse brain, Dlx5 imprinting was relaxed, yet Mecp2-mediated silent-chromatin structure existed at the Dlx5-Dlx6 locus in brains of wild-type, but not Mecp2-null, mice. Mecp2 targeted histone deacetylase 1 to a sharply defined, approximately 1-kb region at the Dlx5-Dlx6 locus and promoted repressive histone methylation at Lys9 at this site. Chromatin immunoprecipitation-combined loop assays showed that Mecp2 mediated the silent chromatin-derived 11-kb chromatin loop at the Dlx5-Dlx6 locus. This loop was absent in chromatin of brains of Mecp2-null mice, and Dlx5-Dlx6 interacted with far distant sequences, forming distinct active chromatin-associated loops. These results show that formation of a silent-chromatin loop is a new mechanism underlying gene regulation by Mecp2.


Assuntos
Cromatina/metabolismo , Proteínas Cromossômicas não Histona/genética , Proteínas de Ligação a DNA/genética , Impressão Genômica , Proteínas de Homeodomínio/genética , Proteínas Repressoras/genética , Síndrome de Rett/genética , Animais , Cromatina/genética , Ilhas de CpG/fisiologia , Metilação de DNA , Perfilação da Expressão Gênica , Proteínas de Homeodomínio/metabolismo , Humanos , Proteína 2 de Ligação a Metil-CpG , Camundongos , Família Multigênica , Neurônios/metabolismo , Testes de Precipitina , Fatores de Transcrição , Ácido gama-Aminobutírico/metabolismo
18.
Sci Rep ; 14(1): 330, 2024 01 03.
Artigo em Inglês | MEDLINE | ID: mdl-38172165

RESUMO

Adverse early life experiences are well-established risk factors for neurological disorders later in life. However, the molecular mechanisms underlying the impact of adverse experiences on neurophysiological systems throughout life remain incompletely understood. Previous studies suggest that social attachment to parents in early development are indispensable for infants to grow into healthy adults. In situations where multiple offspring are born in a single birth in common marmosets, human hand-rearing is employed to ensure the survival of the offspring in captivity. However, hand-reared marmosets often exhibit behavioral abnormalities, including abnormal vocalizations, excessive attachment to the caretaker, and aggressive behavior. In this study, comprehensive transcriptome analyses were conducted on hippocampus tissues, a neuroanatomical region sensitive to social attachment, obtained from human hand-reared (N = 6) and parent-reared male marmosets (N = 5) at distinct developmental stages. Our analyses revealed consistent alterations in a subset of genes, including those related to neurodevelopmental diseases, across different developmental stages, indicating their continuous susceptibility to the effects of early parental deprivation. These findings highlight the dynamic nature of gene expression in response to early life experiences and suggest that the impact of early parental deprivation on gene expression may vary across different stages of development.


Assuntos
Callithrix , Pais , Animais , Adulto , Humanos , Masculino , Callithrix/fisiologia , Relações Familiares , Encéfalo , Expressão Gênica
19.
J Pharm Sci ; 113(7): 1996-2000, 2024 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-38641061

RESUMO

Sodium-phosphate transporter NPT4 (SLC17A3) is a membrane transporter for organic anionic compounds localized on the apical membranes of kidney proximal tubular epithelial cells and plays a role in the urinary excretion of organic anionic compounds. However, its physiological role has not been sufficiently elucidated because its substrate specificity is yet to be determined. The present study aimed to comprehensively explore the physiological substrates of NPT4 in newly developed Slc17a3-/- mice using a metabolomic approach. Metabolomic analysis showed that the plasma concentrations of 11 biological substances, including 3-indoxyl sulfate, were more than two-fold higher in Slc17a3-/- mice than in wild-type mice. Moreover, urinary excretion of 3-indoxyl sulfate was reduced in Slc17a3-/- mice compared to that in wild-type mice. The uptake of 3-indoxyl sulfate by NPT4-expressing Xenopus oocytes was significantly higher than that by water-injected oocytes. The calculated Km and Vmax values for NPT4-mediated 3-indoxyl sulfate uptake were 4.52 ± 1.18 mM and 1.45 ± 0.14 nmol/oocyte/90 min, respectively. In conclusion, the present study revealed that 3-indoxyl sulfate is a novel substrate of NPT4 based on the metabolomic analysis of Slc17a3-/- mice, suggesting that NPT4 regulates systemic exposure to 3-indoxyl sulfate by regulating its urinary excretion.


Assuntos
Indicã , Camundongos Knockout , Oócitos , Toxinas Urêmicas , Animais , Masculino , Camundongos , Indicã/metabolismo , Rim/metabolismo , Metabolômica/métodos , Camundongos Endogâmicos C57BL , Oócitos/metabolismo , Transportadores de Ânions Orgânicos/metabolismo , Transportadores de Ânions Orgânicos/genética , Toxinas Urêmicas/metabolismo , Xenopus laevis
20.
Commun Biol ; 7(1): 740, 2024 Jun 18.
Artigo em Inglês | MEDLINE | ID: mdl-38890503

RESUMO

Although low estrogen is considered to suppress uterine endometrial carcinoma, the most cases occur in the postmenopausal stage. After menopause, the production of androgen level also declines. Therefore, to resolve the above enigma, we hypothesize that the postmenopausal decline of androgen is a trigger of its progression. In the present study, to validate this hypothesis, we examine the pathological roles of androgen/AR by analyzing clinical data, culturing endometrioid cancer cell lines, and using murine models. Clinical data show that androgen receptor (AR) expression and serum dihydrotestosterone (DHT) are associated with lower disease-free survival (DFS). DHT suppresses malignant behaviors in AR-transfected human endometrial cancer cells (ECC). In ovariectomized Ptenff/PRcre/+ mice, DHT decreases the proliferation of spontaneously developed murine ECC. In AR-transfected human ECC and Ptenff/PRcre/+ mice, DHT suppresses FOXP4 expression. FOXP4-overexpressed human ECC increases, while FOXP4-knocked-down ECC shows decreased malignant behaviors. DHT/AR-mediated ECC suppression is restored by FOXP4 overexpression. The high FOXP4 expression is significantly correlated with low postoperative DFS. These findings indicate that the androgen/AR system suppresses the malignant activity of endometrial carcinoma and that downstream FOXP4 is another target molecule. These findings will also impact developments in clinical approaches to elderly health.


Assuntos
Androgênios , Neoplasias do Endométrio , Fatores de Transcrição Forkhead , Receptores Androgênicos , Feminino , Neoplasias do Endométrio/metabolismo , Neoplasias do Endométrio/genética , Humanos , Animais , Camundongos , Fatores de Transcrição Forkhead/metabolismo , Fatores de Transcrição Forkhead/genética , Receptores Androgênicos/metabolismo , Receptores Androgênicos/genética , Androgênios/metabolismo , Linhagem Celular Tumoral , Di-Hidrotestosterona/metabolismo , Di-Hidrotestosterona/farmacologia , Regulação Neoplásica da Expressão Gênica , Pessoa de Meia-Idade , Proliferação de Células
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA