Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 65
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
Mol Psychiatry ; 28(3): 1351-1364, 2023 03.
Artigo em Inglês | MEDLINE | ID: mdl-36434054

RESUMO

Spatial learning and memory flexibility are known to require long-term potentiation (LTP) and long-term depression (LTD), respectively, on a cellular basis. We previously showed that cyclin Y (CCNY), a synapse-remodeling cyclin, is a novel actin-binding protein and an inhibitory regulator of functional and structural LTP in vitro. In this study, we report that Ccny knockout (KO) mice exhibit enhanced LTP and weak LTD at Schaffer collateral-CA1 synapses in the hippocampus. In accordance with enhanced LTP, Ccny KO mice showed improved spatial learning and memory. However, although previous studies reported that normal LTD is necessary for memory flexibility, Ccny KO mice intriguingly showed improved memory flexibility, suggesting that weak LTD could exert memory flexibility when combined with enhanced LTP. At the molecular level, CCNY modulated spatial learning and memory flexibility by distinctively affecting the cofilin-actin signaling pathway in the hippocampus. Specifically, CCNY inhibited cofilin activation by original learning, but reversed such inhibition by reversal learning. Furthermore, viral-mediated overexpression of a phosphomimetic cofilin-S3E in hippocampal CA1 regions enhanced LTP, weakened LTD, and improved spatial learning and memory flexibility, thus mirroring the phenotype of Ccny KO mice. In contrast, the overexpression of a non-phosphorylatable cofilin-S3A in hippocampal CA1 regions of Ccny KO mice reversed the synaptic plasticity, spatial learning, and memory flexibility phenotypes observed in Ccny KO mice. Altogether, our findings demonstrate that LTP and LTD cooperatively regulate memory flexibility. Moreover, CCNY suppresses LTP while facilitating LTD in the hippocampus and negatively regulates spatial learning and memory flexibility through the control of cofilin-actin signaling, proposing CCNY as a learning regulator modulating both memorizing and forgetting processes.


Assuntos
Actinas , Aprendizagem Espacial , Camundongos , Animais , Hipocampo/metabolismo , Potenciação de Longa Duração/fisiologia , Plasticidade Neuronal/fisiologia , Sinapses/metabolismo , Camundongos Knockout , Ciclinas/genética , Ciclinas/metabolismo , Fatores de Despolimerização de Actina/metabolismo
2.
J Korean Med Sci ; 36(28): e204, 2021 Jul 19.
Artigo em Inglês | MEDLINE | ID: mdl-34282607

RESUMO

BACKGROUND: The coronavirus disease 2019 (COVID-19) pandemic began in December 2019. While it has not yet ended, COVID-19 has already created transitions in health care, one of which is a decrease in medical use for health-related issues other than COVID-19 infection. Korean soldiers are relatively homogeneous in terms of age and physical condition. They show a similar disease distribution pattern every year and are directly affected by changes in government attempts to control COVID-19 with nonpharmaceutical interventions. This study aimed to identify the changes in patterns of outpatient visits and admissions to military hospitals for a range of disease types during a pandemic. METHODS: Outpatient attendance and admission data from all military hospitals in South Korea from January 2016 to December 2020 were analyzed. Only active enlisted soldiers aged 18-32 years were included. Outpatient visits where there was a diagnosis of pneumonia, acute upper respiratory tract infection, infectious conjunctivitis, infectious enteritis, asthma, allergic rhinitis, allergic conjunctivitis, atopic dermatitis, urticaria, and fractures were analyzed. Admissions for pneumonia, acute enteritis, and fractures were also analyzed. All outpatient visits and admissions in 2020 for each disease were counted on a weekly basis and compared with the average number of visits over the same period of each year from 2016 to 2019. The corrected value was calculated by dividing the ratio of total weekly number of outpatient visits or admissions to the corresponding medical department in 2020 to the average in 2016-2019. RESULTS: A total of 5,813,304 cases of outpatient care and 143,022 cases of admission were analyzed. For pneumonia, the observed and corrected numbers of outpatient visits and admissions in 2020 decreased significantly compared with the average of other years (P < 0.001). The results were similar for outpatient visits for acute upper respiratory tract infection and infectious conjunctivitis (P < 0.001), while the corrected number of outpatient visits for infectious enteritis showed a significant increase in 2020 (P = 0.005). The corrected number of outpatient visits for asthma in 2020 did not differ from the average of the previous 4 years but the number of visits for the other allergic diseases increased significantly (P < 0.001). For fractures, the observed and corrected numbers of outpatient visits and admissions in 2020 decreased significantly compared with the average of other years (P < 0.001). CONCLUSION: During the COVID-19 pandemic, outpatient visits to military hospitals for respiratory and conjunctival infections and fractures decreased, whereas visits for allergic diseases did not change or increased only slightly. Admissions for pneumonia decreased significantly in 2020, while those for acute enteritis and fractures also decreased, but showed an increased proportion compared with previous years. These results are important because they illustrate the changing patterns in lifestyle as a result of public encouragement to adopt nonpharmaceutical interventions during the pandemic and their effect on medical needs for both infectious and noninfectious diseases in a select group.


Assuntos
COVID-19/epidemiologia , Hospitais Militares/estatística & dados numéricos , SARS-CoV-2 , Adulto , Assistência Ambulatorial/estatística & dados numéricos , Feminino , Humanos , Hipersensibilidade/epidemiologia , Masculino , República da Coreia/epidemiologia , Infecções Respiratórias/epidemiologia
3.
Int J Mol Sci ; 22(15)2021 Jul 26.
Artigo em Inglês | MEDLINE | ID: mdl-34360715

RESUMO

Protein arginine methyltransferase 1 (PRMT1) is a major enzyme responsible for the formation of methylarginine in mammalian cells; however, its function in vivo is not well understood due to its early embryonic lethality in null mice exhibiting spontaneous DNA damage, cell cycle delays, and defects in check point activation. Here, we generated germ cell-specific Prmt1 knock-out (KO) mice to evaluate the function of PRMT1 in spermatogenesis. Our findings demonstrate that PRMT1 is vital for male fertility in mice. Spermatogenesis in Prmt1 KO mice was arrested at the zygotene-like stage of the first meiotic division due to an elevated number of DNA double-strand breaks (DSBs). There was a loss of methylation in meiotic recombination 11 (MRE11), the key endonuclease in MRE11/RAD50/NBS 1 (MRN) complex, resulting in the accumulation of SPO11 protein in DSBs. The ATM-mediated negative feedback control over SPO11 was lost and, consequently, the repair pathway of DSBs was highly affected in PRMT1 deficient male germ cells. Our findings provide a novel insight into the role of PRMT1-mediated asymmetric demethylation in mouse spermatogenesis.


Assuntos
Células Germinativas/enzimologia , Meiose , Proteína-Arginina N-Metiltransferases/metabolismo , Espermatogênese , Hidrolases Anidrido Ácido/genética , Hidrolases Anidrido Ácido/metabolismo , Animais , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Quebras de DNA de Cadeia Dupla , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Endodesoxirribonucleases/genética , Endodesoxirribonucleases/metabolismo , Feminino , Proteína Homóloga a MRE11/genética , Proteína Homóloga a MRE11/metabolismo , Masculino , Camundongos , Camundongos Knockout , Proteína-Arginina N-Metiltransferases/genética
4.
Carcinogenesis ; 41(10): 1341-1352, 2020 10 15.
Artigo em Inglês | MEDLINE | ID: mdl-32706861

RESUMO

Pathological changes in the epigenetic landscape of chromatin are hallmarks of cancer. The caudal-type homeobox gene CDX2 is not expressed in normal gastric epithelia but rather in adult intestinal epithelia, and it is overexpressed in intestinal metaplasia (IM). However, it remains unclear how CDX2 transcription is suppressed in normal gastric epithelial cells and overexpressed in IM. Here, we demonstrate that methylation of the CDX2 promoter increases with age in Helicobacter pylori-positive, noncancerous gastric tissue, whereas the promoter is demethylated in paired gastric tumors in which CDX2 is upregulated. Moreover, we also found that the CDX2 promoter is demethylated in IM as well as gastric tumor. Immunohistochemistry revealed that CDX2 is present in foci of parts of the gastric mucosae but highly expressed in IM as well as in gastric tumors, suggesting that the elevated level of CDX2 in IM and gastric tumors may be attributable to promoter demethylation. Our data suggest that CDX2 repression may be associated with promoter methylation in noncancerous H. pylori-positive mucosa but its upregulation might be attributable to increased promoter activity mediated by chromatin remodeling during gastric carcinogenesis.


Assuntos
Fator de Transcrição CDX2/genética , Desmetilação do DNA , Metilação de DNA , Mucosa Gástrica/microbiologia , Regulação Neoplásica da Expressão Gênica , Helicobacter pylori , Regiões Promotoras Genéticas , Neoplasias Gástricas/genética , Neoplasias Gástricas/microbiologia , Adulto , Fatores Etários , Idoso , Linhagem Celular Tumoral , Feminino , Inativação Gênica , Humanos , Masculino , Pessoa de Meia-Idade , Regulação para Cima
5.
Int J Mol Sci ; 21(22)2020 Nov 16.
Artigo em Inglês | MEDLINE | ID: mdl-33207561

RESUMO

Non-alcoholic fatty liver disease (NAFLD) is a complex disease that is affected by genetic predisposition and epigenetic modification. Deregulation of epigenetic pathways is now recognized as a frequent event in NAFLD, and understanding the mechanistic roles of these epigenetic factors may lead to new strategies for NAFLD treatment. Enhancer of zeste homolog 2 (EZH2) catalyzes methylation on Lys 27 of histone H3, which leads to chromatin compaction and gene silencing. EZH2 regulates embryonic development and cell lineage determination and is related to many human diseases. Recent studies show that EZH2 has critical roles in liver development, homeostasis, and regeneration. Moreover, aberrant activation of EZH2 promotes NAFLD progression. Several EZH2 inhibitors have been developed and studied both in vitro and in clinical trials. In this review, we summarize our current understanding of the role of EZH2 in NAFLD and highlight its potential as a novel therapeutic target for NAFLD treatment.


Assuntos
Sistemas de Liberação de Medicamentos , Proteína Potenciadora do Homólogo 2 de Zeste , Fígado/metabolismo , Hepatopatia Gordurosa não Alcoólica , Proteína Potenciadora do Homólogo 2 de Zeste/antagonistas & inibidores , Proteína Potenciadora do Homólogo 2 de Zeste/metabolismo , Humanos , Hepatopatia Gordurosa não Alcoólica/tratamento farmacológico , Hepatopatia Gordurosa não Alcoólica/metabolismo
6.
Stem Cells ; 36(10): 1578-1588, 2018 10.
Artigo em Inglês | MEDLINE | ID: mdl-30005139

RESUMO

Previous studies have reported that vitamin C (VC) promotes neural stem/precursor cell (NSC) differentiation toward dopamine (DA) neurons via DNA hydroxymethylation-induced transcriptional activation of DA neuron-specific genes. To further understand the VC effects on NSC differentiation, we profiled the transcriptome and DNA methylome/hydroxymethylome using high-throughput sequencing. Interestingly, RNA sequencing analyses have shown that, in addition to DA neuronal genes, astrocytic genes Gfap, Slc1a3, and S100a16 were also upregulated in NSC cultures differentiated with VC treatment. Consistently, enhanced GFAP+ astrocytic yields were manifested in the differentiated cultures with VC treatment, collectively indicating that VC promotes astrocytic differentiation. In genome-wide hydroxymethylome analyses, VC treatment induces enrichment of DNA hydroxymethylation (5-hydroxymethyl cytosine; 5hmC) near the consensus binding motifs of nuclear factor I (NFI). Furthermore, we showed that VC significantly enhanced recruitment of NFI and STAT3, key transcription factors for astrogenesis, in the 5hmC-enriched regions of the astrocyte-specific genes. These findings suggest that VC play important roles in astrocytogenesis during brain development. Stem Cells 2018;36:1578-1588.


Assuntos
Ácido Ascórbico/farmacologia , Astrócitos/metabolismo , Metilação de DNA , Células-Tronco Neurais/metabolismo , Animais , Astrócitos/efeitos dos fármacos , Diferenciação Celular/efeitos dos fármacos , Diferenciação Celular/fisiologia , Humanos , Células-Tronco Neurais/efeitos dos fármacos , Ratos
7.
Mol Cancer ; 17(1): 175, 2018 12 18.
Artigo em Inglês | MEDLINE | ID: mdl-30563517

RESUMO

Even when targets responsible for chemoresistance are identified, drug development is often hampered due to the poor druggability of these proteins. We systematically analyzed therapy-resistance with a large-scale cancer cell transcriptome and drug-response datasets and predicted the candidate drugs based on the gene expression profile. Our results implicated the epithelial-mesenchymal transition as a common mechanism underlying resistance to chemotherapeutic drugs. Notably, we identified ITGB3, whose expression was abundant in both drug resistance and mesenchymal status, as a promising target to overcome chemoresistance. We also confirmed that depletion of ITGB3 sensitized cancer cells to conventional chemotherapeutic drugs by modulating the NF-κB signaling pathway. Considering the poor druggability of ITGB3 and the lack of feasible drugs to directly inhibit this protein, we took an in silico screening for drugs mimicking the transcriptome-level changes caused by knockdown of ITGB3. This approach successfully identified atorvastatin as a novel candidate for drug repurposing, paving an alternative path to drug screening that is applicable to undruggable targets.


Assuntos
Resistencia a Medicamentos Antineoplásicos/genética , Transição Epitelial-Mesenquimal/genética , Integrina beta3/genética , Neoplasias Pulmonares/genética , Células A549 , Linhagem Celular Tumoral , Descoberta de Drogas/métodos , Humanos , NF-kappa B/genética , Farmacogenética/métodos , Transdução de Sinais/genética
8.
Br J Cancer ; 119(11): 1347-1357, 2018 11.
Artigo em Inglês | MEDLINE | ID: mdl-30420612

RESUMO

BACKGROUND: The IDF-11774, a novel clinical candidate for cancer therapy, targets HSP70 and inhibits mitochondrial respiration, resulting in the activation of AMPK and reduction in HIF-1α accumulation. METHODS: To identify genes that have synthetic lethality to IDF-11774, RNA interference screening was conducted, using pooled lentiviruses expressing a short hairpin RNA library. RESULTS: We identified ATP6V0C, encoding the V0 subunit C of lysosomal V-ATPase, knockdown of which induced a synergistic growth-inhibitory effect in HCT116 cells in the presence of IDF-11774. The synthetic lethality of IDF-11774 with ATP6V0C possibly correlates with IDF-11774-mediated autolysosome formation. Notably, the synergistic effect of IDF-11774 and the ATP6V0C inhibitor, bafilomycin A1, depended on the PIK3CA genetic status and Bcl-2 expression, which regulates autolysosome formation and apoptosis. Similarly, in an experiment using conditionally reprogramed cells derived from colorectal cancer patients, synergistic growth inhibition was observed in cells with low Bcl-2 expression. CONCLUSIONS: Bcl-2 is a biomarker for the synthetic lethal interaction of IDF-11774 with ATP6V0C, which is clinically applicable for the treatment of cancer patients with IDF-11774 or autophagy-inducing anti-cancer drugs.


Assuntos
Adamantano/análogos & derivados , Neoplasias Colorretais/enzimologia , Piperazinas/farmacologia , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , ATPases Vacuolares Próton-Translocadoras/antagonistas & inibidores , Adamantano/farmacologia , Animais , Apoptose/efeitos dos fármacos , Autofagia/efeitos dos fármacos , Linhagem Celular Tumoral , Classe I de Fosfatidilinositol 3-Quinases/genética , Neoplasias Colorretais/patologia , Feminino , Humanos , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Macrolídeos/farmacologia , Camundongos , Ensaios Antitumorais Modelo de Xenoenxerto
9.
Virus Genes ; 53(6): 918-921, 2017 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-28795266

RESUMO

Zika virus (ZIKV) (genus Flavivirus, family Flaviviridae) is an emerging pathogen associated with microcephaly and Guillain-Barré syndrome. The rapid spread of ZIKV disease in over 60 countries and the large numbers of travel-associated cases have caused worldwide concern. Thus, intensified surveillance of cases among immigrants and tourists from ZIKV-endemic areas is important for disease control and prevention. In this study, using Next Generation Sequencing, we reported the first whole-genome sequence of ZIKV strain AFMC-U, amplified from the urine of a traveler returning to Korea from the Philippines. Phylogenetic analysis showed geographic-specific clustering. Our results underscore the importance of examining urine in the diagnosis of ZIKV infection.


Assuntos
Infecção por Zika virus/virologia , Humanos , Filipinas , Filogenia , República da Coreia , Viagem , Sequenciamento Completo do Genoma/métodos , Zika virus/genética
10.
Hum Mol Genet ; 23(3): 657-67, 2014 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-24087792

RESUMO

DNA methylation and hydroxymethylation have been implicated in normal development and differentiation, but our knowledge is limited about the genome-wide distribution of 5-methylcytosine (5 mC) and 5-hydroxymethylcytosine (5 hmC) during cellular differentiation. Using an in vitro model system of gradual differentiation of human embryonic stem (hES) cells into ventral midbrain-type neural precursor cells and terminally into dopamine neurons, we observed dramatic genome-wide changes in 5 mC and 5 hmC patterns during lineage commitment. The 5 hmC pattern was dynamic in promoters, exons and enhancers. DNA hydroxymethylation within the gene body was associated with gene activation. The neurogenesis-related genes NOTCH1, RGMA and AKT1 acquired 5 hmC in the gene body and were up-regulated during differentiation. DNA methylation in the promoter was associated with gene repression. The pluripotency-related genes POU5F1, ZFP42 and HMGA1 acquired 5 mC in their promoters and were down-regulated during differentiation. Promoter methylation also acted as a locking mechanism to maintain gene silencing. The mesoderm development-related genes NKX2-8, TNFSF11 and NFATC1 acquired promoter methylation during neural differentiation even though they were already silenced in hES cells. Our findings will help elucidate the molecular mechanisms underlying lineage-specific differentiation of pluripotent stem cells during human embryonic development.


Assuntos
Diferenciação Celular/fisiologia , Metilação de DNA , Células-Tronco Embrionárias/fisiologia , Neurônios/fisiologia , 5-Metilcitosina/metabolismo , Linhagem da Célula/genética , Células Cultivadas , Citosina/análogos & derivados , Citosina/metabolismo , Células-Tronco Embrionárias/citologia , Proteínas Ligadas por GPI/genética , Regulação da Expressão Gênica , Inativação Gênica , Proteínas de Homeodomínio/genética , Humanos , Mesoderma/fisiologia , Proteínas do Tecido Nervoso/genética , Neurogênese/genética , Neurônios/citologia , Regiões Promotoras Genéticas , Proteínas Proto-Oncogênicas c-akt/genética , Ligante RANK/genética , Receptor Notch1/genética , Fatores de Transcrição/genética
11.
Stem Cells ; 33(4): 1320-32, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25535150

RESUMO

Intracellular Vitamin C (VC) is maintained at high levels in the developing brain by the activity of sodium-dependent VC transporter 2 (Svct2), suggesting specific VC functions in brain development. A role of VC as a cofactor for Fe(II)-2-oxoglutarate-dependent dioxygenases has recently been suggested. We show that VC supplementation in neural stem cell cultures derived from embryonic midbrains greatly enhanced differentiation toward midbrain-type dopamine (mDA) neurons, the neuronal subtype associated with Parkinson's disease. VC induced gain of 5-hydroxymethylcytosine (5hmC) and loss of H3K27m3 in DA phenotype gene promoters, which are catalyzed by Tet1 and Jmjd3, respectively. Consequently, VC enhanced DA phenotype gene transcriptions in the progenitors by Nurr1, a transcription factor critical for mDA neuron development, to be more accessible to the gene promoters. Further mechanism studies including Tet1 and Jmjd3 knockdown/inhibition experiments revealed that both the 5hmC and H3K27m3 changes, specifically in the progenitor cells, are indispensible for the VC-mediated mDA neuron differentiation. We finally show that in Svct2 knockout mouse embryos, mDA neuron formation in the developing midbrain decreased along with the 5hmC/H3k27m3 changes. These findings together indicate an epigenetic role of VC in midbrain DA neuron development.


Assuntos
Ácido Ascórbico/farmacologia , Diferenciação Celular/fisiologia , Dioxigenases/metabolismo , Neurônios Dopaminérgicos/metabolismo , Epigênese Genética/fisiologia , Histona Desmetilases com o Domínio Jumonji/metabolismo , Animais , Diferenciação Celular/efeitos dos fármacos , Células Cultivadas , Neurônios Dopaminérgicos/efeitos dos fármacos , Epigênese Genética/efeitos dos fármacos , Mesencéfalo/citologia , Mesencéfalo/efeitos dos fármacos , Mesencéfalo/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Neurogênese/efeitos dos fármacos , Neurogênese/fisiologia , Ratos , Ratos Sprague-Dawley
12.
Genome Res ; 22(6): 1026-35, 2012 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-22421545

RESUMO

H2B monoubiquitylation (H2Bub1), which is required for multiple methylations of both H3K4 and H3K79, has been implicated in gene expression in numerous organisms ranging from yeast to human. However, the molecular crosstalk between H2Bub1 and other modifications, especially the methylations of H3K4 and H3K79, remains unclear in vertebrates. To better understand the functional role of H2Bub1, we measured genome-wide histone modification patterns in human cells. Our results suggest that H2Bub1 has dual roles, one that is H3 methylation dependent, and another that is H3 methylation independent. First, H2Bub1 is a 5'-enriched active transcription mark and co-occupies with H3K79 methylations in actively transcribed regions. Second, this study shows for the first time that H2Bub1 plays a histone H3 methylations-independent role in chromatin architecture. Furthermore, the results of this work indicate that H2Bub1 is largely positioned at the exon-intron boundaries of highly expressed exons, and it demonstrates increased occupancy in skipped exons compared with flanking exons in the human and mouse genomes. Our findings collectively suggest that a potentiating mechanism links H2Bub1 to both H3K79 methylations in actively transcribed regions and the exon-intron structure of highly expressed exons via the regulation of nucleosome dynamics during transcription elongation.


Assuntos
Cromatina/genética , Éxons , Histonas/metabolismo , Íntrons , Transcrição Gênica , Animais , Linhagem Celular Tumoral , Cromatina/metabolismo , Imunoprecipitação da Cromatina , Drosophila/genética , Regulação da Expressão Gênica , Genoma Humano , Histonas/genética , Humanos , Metilação , Camundongos , Neoplasias Embrionárias de Células Germinativas/genética , Ubiquitinação
13.
Clin Mol Hepatol ; 2024 Jul 25.
Artigo em Inglês | MEDLINE | ID: mdl-39048522

RESUMO

Background/Aims: Blocking the complement system is a promising strategy to impede the progression of metabolic dysfunction-associated steatotic liver disease (MASLD). However, the interplay between complement and MASLD remains to be elucidated. This comprehensive approach aimed to investigate the potential association between complement dysregulation and the histological severity of MASLD. Methods: Liver biopsy specimens were procured from a cohort comprising 106 Korean individuals, which included 31 controls, 17 with isolated steatosis, and 58 with metabolic dysfunction-associated steatohepatitis (MASH). Utilizing the Infinium Methylation EPIC array, thorough analysis of methylation alterations in 61 complement genes was conducted. The expression and methylation of nine complement genes in a murine MASH model were examined using quantitative RT-PCR and pyrosequencing. Results: Methylome and transcriptome analyses of liver biopsies revealed significant (P <0.05) hypermethylation and downregulation of C1R, C1S, C3, C6, C4BPA, and SERPING1, as well as hypomethylation (P <0.0005) and upregulation (P <0.05) of C5AR1, C7, and CD59, in association with the histological severity of MASLD. Furthermore, DNA methylation and the relative expression of nine complement genes in a MASH diet mouse model aligned with human data. Conclusions: Our research provides compelling evidence that epigenetic alterations in complement genes correlate with MASLD severity, offering valuable insights into the mechanisms driving MASLD progression, and suggests that inhibiting the function of certain complement proteins may be a promising strategy for managing MASLD.

14.
J Biol Chem ; 287(47): 39698-709, 2012 Nov 16.
Artigo em Inglês | MEDLINE | ID: mdl-23012353

RESUMO

Histone-modifying enzymes play a pivotal role in gene expression and repression. In human, DOT1L (Dot1-like) is the only known histone H3 lysine 79 methyltransferase. hDOT1L is associated with transcriptional activation, but the general mechanism connecting hDOT1L to active transcription remains largely unknown. Here, we report that hDOT1L interacts with the phosphorylated C-terminal domain of actively transcribing RNA polymerase II (RNAPII) through a region conserved uniquely in multicellular DOT1 proteins. Genome-wide profiling analyses indicate that the occupancy of hDOT1L largely overlaps with that of RNAPII at actively transcribed genes, especially surrounding transcriptional start sites, in embryonic carcinoma NCCIT cells. We also find that C-terminal domain binding or H3K79 methylations by hDOT1L is important for the expression of target genes such as NANOG and OCT4 and a marker for pluripotency in NCCIT cells. Our results indicate that a functional interaction between hDOT1L and RNAPII targets hDOT1L and subsequent H3K79 methylations to actively transcribed genes.


Assuntos
Regulação da Expressão Gênica/fisiologia , Histona-Lisina N-Metiltransferase/metabolismo , Histonas/metabolismo , Metiltransferases/metabolismo , RNA Polimerase II/metabolismo , Transcrição Gênica/fisiologia , Estudo de Associação Genômica Ampla , Células HEK293 , Histona Metiltransferases , Histona-Lisina N-Metiltransferase/genética , Histonas/genética , Humanos , Metilação , Metiltransferases/genética , Ligação Proteica/fisiologia , RNA Polimerase II/genética
15.
Hum Mol Genet ; 20(14): 2722-33, 2011 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-21505074

RESUMO

Hepatocytes that have differentiated from human embryonic stem cells (hESCs) have great potential for the treatment of liver disease as well as for drug testing. Moreover, in vitro hepatogenesis is a powerful model system for studying the molecular mechanisms underlying liver development. DNA methylation is an important epigenetic mechanism that influences differential gene expression during embryonic development. We profiled gene expression and DNA methylation of three cell states of in vitro hepatogenesis-hESC, definitive endoderm and hepatocyte-using microarray analysis. Among 525 state-specific expressed genes, 67 showed significant negative correlation between gene expression and DNA methylation. State-specific expression and methylation of target genes were validated by quantitative reverse transcription-polymerase chain reaction and pyrosequencing, respectively. To elucidate genome-scale methylation changes beyond the promoter, we also performed high-throughput sequencing of methylated DNA captured by the MBD2 protein. We found dynamic methylation changes in intergenic regions of the human genome during differentiation. This study provides valuable methylation markers for the lineage commitment of in vitro hepatogenesis and should help elucidate the molecular mechanisms underlying stem cell differentiation and liver development.


Assuntos
Diferenciação Celular/fisiologia , Metilação de DNA/fisiologia , Células-Tronco Embrionárias/metabolismo , Epigênese Genética/fisiologia , Hepatócitos/metabolismo , Regiões Promotoras Genéticas/fisiologia , Linhagem Celular , Células-Tronco Embrionárias/citologia , Estudo de Associação Genômica Ampla , Hepatócitos/citologia , Humanos
16.
Mol Cells ; 46(5): 298-308, 2023 May 31.
Artigo em Inglês | MEDLINE | ID: mdl-36896596

RESUMO

Gastric cancer (GC) is a complex disease influenced by multiple genetic and epigenetic factors. Chronic inflammation caused by Helicobacter pylori infection and dietary risk factors can result in the accumulation of aberrant DNA methylation in gastric mucosa, which promotes GC development. Tensin 4 (TNS4), a member of the Tensin family of proteins, is localized to focal adhesion sites, which connect the extracellular matrix and cytoskeletal network. We identified upregulation of TNS4 in GC using quantitative reverse transcription PCR with 174 paired samples of GC tumors and adjacent normal tissues. Transcriptional activation of TNS4 occurred even during the early stage of tumor development. TNS4 depletion in GC cell lines that expressed high to moderate levels of TNS4, i.e., SNU-601, KATO III, and MKN74, reduced cell proliferation and migration, whereas ectopic expression of TNS4 in those lines that expressed lower levels of TNS4, i.e., SNU-638, MKN1, and MKN45 increased colony formation and cell migration. The promoter region of TNS4 was hypomethylated in GC cell lines that showed upregulation of TNS4. We also found a significant negative correlation between TNS4 expression and CpG methylation in 250 GC tumors based on The Cancer Genome Atlas (TCGA) data. This study elucidates the epigenetic mechanism of TNS4 activation and functional roles of TNS4 in GC development and progression and suggests a possible approach for future GC treatments.


Assuntos
Infecções por Helicobacter , Helicobacter pylori , Neoplasias Gástricas , Humanos , Linhagem Celular Tumoral , Metilação de DNA , Epigênese Genética , Regulação Neoplásica da Expressão Gênica , Infecções por Helicobacter/genética , Helicobacter pylori/metabolismo , Neoplasias Gástricas/genética , Neoplasias Gástricas/patologia , Tensinas/genética , Tensinas/metabolismo
17.
Leukemia ; 37(4): 807-819, 2023 04.
Artigo em Inglês | MEDLINE | ID: mdl-36932165

RESUMO

Clinical effect of donor-derived natural killer cell infusion (DNKI) after HLA-haploidentical hematopoietic cell transplantation (HCT) was evaluated in high-risk myeloid malignancy in phase 2, randomized trial. Seventy-six evaluable patients (aged 21-70 years) were randomized to receive DNKI (N = 40) or not (N = 36) after haploidentical HCT. For the HCT conditioning, busulfan, fludarabine, and anti-thymocyte globulin were administered. DNKI was given twice 13 and 20 days after HCT. Four patients in the DNKI group failed to receive DNKI. In the remaining 36 patients, median DNKI doses were 1.0 × 108/kg and 1.4 × 108/kg on days 13 and 20, respectively. Intention-to-treat analysis showed a lower disease progression for the DNKI group (30-month cumulative incidence, 35% vs 61%, P = 0.040; subdistribution hazard ratio, 0.50). Furthermore, at 3 months after HCT, the DNKI patients showed a 1.8- and 2.6-fold higher median absolute blood count of NK and T cells, respectively. scRNA-sequencing analysis in seven study patients showed that there was a marked increase in memory-like NK cells in DNKI patients which, in turn, expanded the CD8+ effector-memory T cells. In high-risk myeloid malignancy, DNKI after haploidentical HCT reduced disease progression. This enhanced graft-vs-leukemia effect may be related to the DNKI-induced, post-HCT expansion of NK and T cells. Clinical trial number: NCT02477787.


Assuntos
Doença Enxerto-Hospedeiro , Transplante de Células-Tronco Hematopoéticas , Leucemia Mieloide Aguda , Humanos , Interleucina-15 , Doença Enxerto-Hospedeiro/patologia , Células Matadoras Naturais/patologia , Progressão da Doença , Leucemia Mieloide Aguda/terapia , Leucemia Mieloide Aguda/patologia , Condicionamento Pré-Transplante
18.
Carcinogenesis ; 33(8): 1494-501, 2012 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-22610074

RESUMO

In this study, the promoter of the gene coiled-coil domain-containing 67 (CCDC67) was found to be frequently methylated in gastric cancer cell lines and in primary gastric tumors, as examined by restriction landmark genomic scanning. In addition, CCDC67 expression was down-regulated in 72.7% of gastric cancer cell lines tested. In most cases, gene down-regulation was associated with CpG hypermethylation in the CCDC67 promoter. Treatment with 5-aza-2'-deoxycytidine and/or trichostatin A restored CCDC67 expression in down-regulated cell lines. Pyrosequencing analysis of 150 paired primary gastric cancer samples revealed that promoter CpG methylation was increased in 74% of tested tumors compared with paired adjacent normal tissues, and this hypermethylation correlated significantly with down-regulation of CCDC67. CCDC67 protein was localized to the cell membrane by immunocytochemistry. Stable transfection of a CCDC67 gene in one gastric cancer cell line inhibited adhesion-dependent and -independent colony formation, and CCDC67 expression suppressed tumorigenesis in nude mice. We suggest that CCDC67 is a putative tumor suppressor gene that is silenced in gastric cancers by promoter CpG methylation and that it may play an important role in cell signaling and migration related to tumorigenesis.


Assuntos
Epigênese Genética , Genes Supressores de Tumor , Neoplasias Gástricas/genética , Proteínas Supressoras de Tumor/genética , Animais , Proliferação de Células , Humanos , Imuno-Histoquímica , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Proteínas Associadas aos Microtúbulos , Regiões Promotoras Genéticas , RNA Mensageiro/genética , Reação em Cadeia da Polimerase em Tempo Real , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Neoplasias Gástricas/patologia
19.
Biochem Biophys Res Commun ; 423(1): 38-44, 2012 Jun 22.
Artigo em Inglês | MEDLINE | ID: mdl-22627140

RESUMO

PKM2 is an isoenzyme of the glycolytic enzyme pyruvate kinase that promotes aerobic glycolysis. Here, we describe an important role for PKM2 in regulating the survival of gastric cancer (GC) cells. We showed that PKM2 was overexpressed in gastric tumor tissues compared to normal tissues and its expression level was associated with poor survival of gastric cancer patients. We also showed that PKM2 affected cell survival by regulating Bcl-xL at the transcriptional level. PKM2 knockdown partially affected the stability of NF-kB subunit p65, suggesting that post-translational regulation of p65 by PKM2 is one of plausible mechanisms for the increased cell growth. Therefore, PKM2 may function as an upstream molecule that regulates p65 function and thus enhances the growth of tumor cells.


Assuntos
Biomarcadores Tumorais/metabolismo , Regulação Neoplásica da Expressão Gênica , Piruvato Quinase/metabolismo , Neoplasias Gástricas/genética , Neoplasias Gástricas/patologia , Proteína bcl-X/genética , Biomarcadores Tumorais/genética , Linhagem Celular Tumoral , Proliferação de Células , Técnicas de Silenciamento de Genes , Humanos , Prognóstico , Piruvato Quinase/genética , Fator de Transcrição RelA/metabolismo , Transcrição Gênica
20.
Cancers (Basel) ; 14(24)2022 Dec 14.
Artigo em Inglês | MEDLINE | ID: mdl-36551669

RESUMO

The loss-of-function variants are thought to be associated with inflammation in the stomach. We here aimed to evaluate the extent and role of methylation at the SSTR2 promoter in inflammation and gastric tumor formation. A whole-genome bisulfite sequencing analysis revealed that the SSTR2 promoter was significantly hypermethylated in gastric tumors, dysplasia, and intestinal metaplasia compared to non-tumor tissues from patients with gastric cancer. Using public data, we confirmed SSTR2 promoter methylation in primary gastric tumors and intestinal metaplasia, and even aged gastric mucosae infected with Helicobacter pylori, suggesting that aberrant methylation is initiated in normal gastric mucosa. The loss-of-function of SSTR2 in SNU638 cell-induced cell proliferation in vitro, while stable transfection of SSTR2 in AGS and MKN74 cells inhibited cell proliferation and tumorigenesis in vitro and in vivo. As revealed by a comparison of target genes differentially expressed in these cells with hallmark molecular signatures, inflammation-related pathways were distinctly induced in SSTR2-KO SNU638 cell. By contrast, inflammation-related pathways were inhibited in AGS and MKN74 cells ectopically expressing SSTR2. Collectively, we propose that SSTR2 silencing upon promoter methylation is initiated in aged gastric mucosae infected with H. pylori and promotes the establishment of an inflammatory microenvironment via the intrinsic pathway. These findings provide novel insights into the initiation of gastric carcinogenesis.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA