Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
1.
Int Endod J ; 57(1): 50-63, 2024 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-37837219

RESUMO

AIM: This study aimed to investigate the upstream regulators and specific mechanisms of metastasis-associated lung adenocarcinoma transcript 1 (MALAT1) in the odontoblastic differentiation of human dental pulp stem cells (hDPSCs). METHODOLOGY: Human dental pulp stem cells were isolated and cultured, followed by conducting loss- or gain-of-function experiments on ATF4 and loss experiments on MALAT1 to elucidate their respective biological functions in odontoblastic differentiation. Chromatin immunoprecipitation assays and RNA immunoprecipitation were performed to uncover the interaction between ATF4-MALAT1 and MALAT1-JMJD3, respectively. The odontoblastic differentiation was estimated by the mRNA and protein of DSPP and DMP1, as well as alkaline phosphatase staining. RESULTS: Expression of MALAT1 was upregulated in the hDPSCs cultured in an odontoblastic medium, and MALAT1 downregulation suppressed the odontoblastic differentiation of the hDPSCs. Subsequent experiments confirmed that ATF4 promoted odontoblastic differentiation and induced MALAT1 expression by binding to the MALAT1 promoter region. Further experiments revealed that nuclear MALAT1 interacted with JMJD3. MALAT1 knockdown decreased the JMJD3 protein level and demethylase activity, and it enhanced H3K27me3 occupancy of the promoter region of DSPP and DMP1, resulting in the inhibition of DSPP and DMP1 transcription. Importantly, JMJD3 overexpression significantly attenuated the inhibition of odontoblastic differentiation induced by MALAT1 knockdown. CONCLUSIONS: ATF4-regulated MALAT1 plays a positive regulatory role in odontoblastic differentiation of hDPSCs through JMJD3-mediated H3K27me3 modifications of the DSPP and DMP1 promoters.


Assuntos
Diferenciação Celular , Histona Desmetilases com o Domínio Jumonji , Odontoblastos , RNA Longo não Codificante , Humanos , Fator 4 Ativador da Transcrição/metabolismo , Células Cultivadas , Polpa Dentária , Proteínas da Matriz Extracelular/genética , Proteínas da Matriz Extracelular/metabolismo , Histona Desmetilases/metabolismo , Histonas/metabolismo , RNA Longo não Codificante/genética , RNA Longo não Codificante/metabolismo , Células-Tronco , Histona Desmetilases com o Domínio Jumonji/genética , Histona Desmetilases com o Domínio Jumonji/metabolismo
2.
Stem Cells ; 40(9): 818-830, 2022 09 26.
Artigo em Inglês | MEDLINE | ID: mdl-35728620

RESUMO

Nerve growth factor (NGF) is the best-characterized neurotrophin and is primarily recognized for its key role in the embryonic development of the nervous system and neuronal cell survival/differentiation. Recently, unexpected actions of NGF in bone regeneration have emerged as NGF is able to enhance the osteogenic differentiation of mesenchymal stem cells. However, little is known regarding how NGF signaling regulates osteogenic differentiation through epigenetic mechanisms. In this study, using human dental mesenchymal stem cells (DMSCs), we demonstrated that NGF mediates osteogenic differentiation through p75NTR, a low-affinity NGF receptor. P75NTR-mediated NGF signaling activates the JNK cascade and the expression of KDM4B, an activating histone demethylase, by removing repressive H3K9me3 epigenetic marks. Mechanistically, NGF-activated c-Jun binds to the KDM4B promoter region and directly upregulates KDM4B expression. Subsequently, KDM4B directly and epigenetically activates DLX5, a master osteogenic gene, by demethylating H3K9me3 marks. Furthermore, we revealed that KDM4B and c-Jun from the JNK signaling pathway work in concert to regulate NGF-mediated osteogenic differentiation through simultaneous recruitment to the promoter region of DLX5. We identified KDM4B as a key epigenetic regulator during the NGF-mediated osteogenesis both in vitro and in vivo using the calvarial defect regeneration mouse model. In conclusion, our study thoroughly elucidated the molecular and epigenetic mechanisms during NGF-mediated osteogenesis.


Assuntos
Células-Tronco Mesenquimais , Osteogênese , Animais , Diferenciação Celular/genética , Epigênese Genética , Histona Desmetilases/metabolismo , Humanos , Histona Desmetilases com o Domínio Jumonji/genética , Histona Desmetilases com o Domínio Jumonji/metabolismo , Células-Tronco Mesenquimais/metabolismo , Camundongos , Fator de Crescimento Neural/genética , Fator de Crescimento Neural/metabolismo , Osteogênese/genética , Receptor de Fator de Crescimento Neural/genética , Receptor de Fator de Crescimento Neural/metabolismo
3.
Oral Dis ; 29(3): 1137-1148, 2023 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-34739163

RESUMO

OBJECTIVE: Periodontitis is a chronic inflammation of periodontal tissues. This study is expected to assess the effect of LSD1 on the osteogenic differentiation of hPDLSCs in periodontitis. METHODS: hPDLSCs were separated, cultivated, and identified, and then treated by LPS to induce inflammatory microenvironment and subjected to osteogenic differentiation. Subsequently, LSD1 expression was determined, and then silenced to assess its effect on hPDLSCs. Next, the binding relation between LSD1 and miR-590-3p was analyzed. miR-590-3p expression was detected and then overexpressed to evaluate its role in hPDLSCs in periodontitis. Afterward, the relation between LSD1 and OSX was analyzed. H3K4me2 level and OSX transcription were measured, and the role of H3K4me2 was determined. Additionally, the role of OSX in hPDLSCs was verified. RESULTS: LSD1 was poorly expressed after osteogenic differentiation of hPDLSCs while it was rescued upon LPS induction. The osteogenic differentiation of hPDLSC in periodontitis was strengthened upon LSD1 downregulation. Besides, miR-590-3p targeted LSD1 transcription, and LSD1 inhibited OSX transcription via H3K4me2 demethylation. miR-590-3p overexpression improved osteogenic differentiation of hPDLSCs in periodontitis. But this improvement was annulled by OSX inhibition. CONCLUSION: miR-590-3p targeted LSD1 transcription and upregulated H3K4me2 methylation to promote OSX transcription, thereby encouraging osteogenic differentiation of hPDLSCs in periodontitis.


Assuntos
MicroRNAs , Periodontite , Humanos , Diferenciação Celular , Células Cultivadas , Histona Desmetilases/genética , Histona Desmetilases/metabolismo , Lipopolissacarídeos/farmacologia , MicroRNAs/genética , MicroRNAs/metabolismo , Osteogênese/genética , Ligamento Periodontal , Periodontite/genética , Periodontite/metabolismo , Células-Tronco
4.
J Cell Physiol ; 235(11): 8432-8445, 2020 11.
Artigo em Inglês | MEDLINE | ID: mdl-32246725

RESUMO

Enhancing the functions of mesenchymal stem cells (MSCs) is considered a potential approach for promoting tissue regeneration. In the present study, we investigate the role of HOXC8 in regulating differentiation and migration by using stem cells of the apical papilla (SCAPs). Our results showed that overexpression of HOXC8 suppressed the osteo-/dentinogenic differentiation, as detected by measuring alkaline phosphatase activity, in vitro mineralization, and the expressions of dentin sialophosphoprotein, dentin matrix acidic phosphoprotein 1, bone sialoprotein, runt-related transcription factor 2, and osterix in SCAPs, and inhibited in vivo osteo-/dentinogenesis of SCAPs. In addition, knockdown of HOXC8 promoted the osteo-/dentinogenic differentiation potentials of SCAPs. Mechanically, HOXC8 enhanced KDM1A transcription by directly binding to its promoter. HOXC8 and KDM1A also inhibited the migration and chemotaxis abilities of SCAPs. To sum up, HOXC8 negatively regulated the osteo-/dentinogenic differentiation and migration abilities of SCAPs by directly enhancing KDM1A transcription and indicated that HOXC8 and KDM1A could serve as potential targets for enhancing dental MSC mediated tissue regeneration.


Assuntos
Diferenciação Celular/fisiologia , Histona Desmetilases/metabolismo , Proteínas de Homeodomínio/metabolismo , Células-Tronco/metabolismo , Diferenciação Celular/genética , Proliferação de Células/fisiologia , Células Cultivadas , Papila Dentária/metabolismo , Genes Homeobox/fisiologia , Humanos , Células-Tronco Mesenquimais/metabolismo , Osteogênese/fisiologia
5.
Virus Genes ; 53(3): 340-351, 2017 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-28364140

RESUMO

The Jumonji C-domain containing protein 6 (JMJD6) has had a convoluted history, and recent reports indicating a multifactorial role in foot-and-mouth disease virus (FMDV) infection have further complicated the functionality of this protein. It was first identified as the phosphatidylserine receptor on the cell surface responsible for recognizing phosphatidylserine on the surface of apoptotic cells resulting in their engulfment by phagocytic cells. Subsequent study revealed a nuclear subcellular localization, where JMJD6 participated in lysine hydroxylation and arginine demethylation of histone proteins and other non-histone proteins. Interestingly, to date, JMDJ6 remains the only known arginine demethylase with a growing list of known substrate molecules. These conflicting associations rendered the subcellular localization of JMJD6 to be quite nebulous. Further muddying this area, two different groups illustrated that JMJD6 could be induced to redistribute from the cell surface to the nucleus of a cell. More recently, JMJD6 was demonstrated to be a host factor contributing to the FMDV life cycle, where it was not only exploited for its arginine demethylase activity, but also served as an alternative virus receptor. This review attempts to coalesce these divergent roles for a single protein into one cohesive account. Given the diverse functionalities already characterized for JMJD6, it is likely to continue to be a confounding protein resulting in much contention going into the near future.


Assuntos
Vírus da Febre Aftosa/metabolismo , Febre Aftosa/virologia , Histona Desmetilases com o Domínio Jumonji/fisiologia , Replicação Viral/fisiologia , Animais , Arginina/metabolismo , Membrana Celular/metabolismo , Núcleo Celular/enzimologia , Núcleo Celular/genética , Febre Aftosa/enzimologia , Febre Aftosa/genética , Vírus da Febre Aftosa/genética , Vírus da Febre Aftosa/crescimento & desenvolvimento , Histona Desmetilases/metabolismo , Histonas , Interações Hospedeiro-Patógeno , Humanos , Lisina/metabolismo , Fagócitos/virologia , Fosfatidilserinas/metabolismo , Pró-Colágeno-Lisina 2-Oxoglutarato 5-Dioxigenase/metabolismo , Ligação Proteica , Receptores de Superfície Celular/metabolismo
6.
Oral Dis ; 23(6): 709-720, 2017 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-27514027

RESUMO

Histone N-terminal tails of nucleosomes are the sites of complex regulation of gene expression through post-translational modifications. Among these modifications, histone methylation had long been associated with permanent gene inactivation until the discovery of Lys-specific demethylase (LSD1), which is responsible for dynamic gene regulation. There are more than 30 members of the Lys demethylase (KDM) family, and with exception of LSD1 and LSD2, all other KDMs possess the Jumonji C (JmjC) domain exhibiting demethylase activity and require unique cofactors, for example, Fe(II) and α-ketoglutarate. These cofactors have been targeted when devising KDM inhibitors, which may yield therapeutic benefit. KDMs and their counterpart Lys methyltransferases (KMTs) regulate multiple biological processes, including oncogenesis and inflammation. KDMs' functional interactions with retinoblastoma (Rb) and E2 factor (E2F) target promoters illustrate their regulatory role in cell cycle progression and oncogenesis. Recent findings also demonstrate the control of inflammation and immune functions by KDMs, such as KDM6B that regulates the pro-inflammatory gene expression and CD4+ T helper (Th) cell lineage determination. This review will highlight the mechanisms by which KDMs and KMTs regulate the target gene expression and how epigenetic mechanisms may be applied to our understanding of oral inflammation.


Assuntos
Carcinogênese/genética , Ciclo Celular/genética , Periodontite Crônica/genética , Epigênese Genética , Histona Desmetilases/genética , Histona Desmetilases/metabolismo , Processo Alveolar/crescimento & desenvolvimento , Metilação de DNA , Humanos , Dente/crescimento & desenvolvimento
7.
Nature ; 466(7305): 503-7, 2010 Jul 22.
Artigo em Inglês | MEDLINE | ID: mdl-20622853

RESUMO

X-linked mental retardation (XLMR) is a complex human disease that causes intellectual disability. Causal mutations have been found in approximately 90 X-linked genes; however, molecular and biological functions of many of these genetically defined XLMR genes remain unknown. PHF8 (PHD (plant homeo domain) finger protein 8) is a JmjC domain-containing protein and its mutations have been found in patients with XLMR and craniofacial deformities. Here we provide multiple lines of evidence establishing PHF8 as the first mono-methyl histone H4 lysine 20 (H4K20me1) demethylase, with additional activities towards histone H3K9me1 and me2. PHF8 is located around the transcription start sites (TSS) of approximately 7,000 RefSeq genes and in gene bodies and intergenic regions (non-TSS). PHF8 depletion resulted in upregulation of H4K20me1 and H3K9me1 at the TSS and H3K9me2 in the non-TSS sites, respectively, demonstrating differential substrate specificities at different target locations. PHF8 positively regulates gene expression, which is dependent on its H3K4me3-binding PHD and catalytic domains. Importantly, patient mutations significantly compromised PHF8 catalytic function. PHF8 regulates cell survival in the zebrafish brain and jaw development, thus providing a potentially relevant biological context for understanding the clinical symptoms associated with PHF8 patients. Lastly, genetic and molecular evidence supports a model whereby PHF8 regulates zebrafish neuronal cell survival and jaw development in part by directly regulating the expression of the homeodomain transcription factor MSX1/MSXB, which functions downstream of multiple signalling and developmental pathways. Our findings indicate that an imbalance of histone methylation dynamics has a critical role in XLMR.


Assuntos
Encéfalo/embriologia , Encéfalo/enzimologia , Cabeça/embriologia , Histona Desmetilases/metabolismo , Histonas/metabolismo , Fatores de Transcrição/metabolismo , Proteínas de Peixe-Zebra/metabolismo , Peixe-Zebra/embriologia , Animais , Biocatálise , Encéfalo/citologia , Domínio Catalítico , Ciclo Celular , Linhagem Celular Tumoral , Sobrevivência Celular , DNA Intergênico/genética , Regulação da Expressão Gênica , Histona Desmetilases/genética , Histonas/química , Proteínas de Homeodomínio/genética , Humanos , Arcada Osseodentária/citologia , Arcada Osseodentária/embriologia , Lisina/metabolismo , Deficiência Intelectual Ligada ao Cromossomo X/enzimologia , Deficiência Intelectual Ligada ao Cromossomo X/genética , Metilação , Neurônios/citologia , Neurônios/enzimologia , Regiões Promotoras Genéticas , Fatores de Transcrição/deficiência , Fatores de Transcrição/genética , Sítio de Iniciação de Transcrição , Peixe-Zebra/metabolismo , Proteínas de Peixe-Zebra/genética
8.
Acta Crystallogr D Biol Crystallogr ; 71(Pt 9): 1955-64, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-26327385

RESUMO

The JmjC domain-containing proteins belong to a large family of oxygenases possessing distinct substrate specificities which are involved in the regulation of different biological processes, such as gene transcription, RNA processing and translation. Nucleolar protein 66 (NO66) is a JmjC domain-containing protein which has been reported to be a histone demethylase and a ribosome protein 8 (Rpl8) hydroxylase. The present biochemical study confirmed the hydroxylase activity of NO66 and showed that oligomerization is required for NO66 to efficiently catalyze the hydroxylation of Rpl8. The structures of NO66(176-C) complexed with Rpl8(204-224) in a tetrameric form and of the mutant protein M2 in a dimeric form were solved. Based on the results of structural and biochemical analyses, the consensus sequence motif NHXH recognized by NO66 was confirmed. Several potential substrates of NO66 were found by a BLAST search according to the consensus sequence motif. When binding to substrate, the relative positions of each subunit in the NO66 tetramer shift. Oligomerization may facilitate the motion of each subunit in the NO66 tetramer and affect the catalytic activity.


Assuntos
Proteínas Cromossômicas não Histona/química , Histona Desmetilases/química , Proteínas Ribossômicas/química , Sequência de Aminoácidos , Animais , Biopolímeros/química , Catálise , Cromatografia em Gel , Proteínas Cromossômicas não Histona/metabolismo , Cristalografia por Raios X , Dioxigenases , Histona Desmetilases/metabolismo , Humanos , Hidroxilação , Modelos Moleculares , Dados de Sequência Molecular , Conformação Proteica , Homologia de Sequência de Aminoácidos , Especificidade por Substrato
9.
Int Immunopharmacol ; 116: 109796, 2023 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-36731157

RESUMO

Macrophages are highly implicated in the progression of periodontitis, while circadian rhythm disruption (CRD) promotes the inflammatory response of macrophages in many diseases. However, the effects of CRD on periodontitis and the role of macrophages in this process remain unclear. Histone lysinedemethylase6a (Kdm6a), a histone demethylase, has recently been identified as a key regulator of macrophage-induced inflammation. Here, we established an experimental periodontitis model by injecting lipopolysaccharide (LPS) derived from Porphyromonas gingivalis with or without periodontal ligation in mice exposed to an 8-h time shift jet-lag schedule every 3 days. By histomorphometry, tartrate acid phosphatase (TRAP) staining, RT-qPCR, ELISA, immunohistochemistry and immunofluorescence analysis, we found that CRD promoted the inflammatory response, alveolar bone resorption, macrophage infiltration and Kdm6a expression in macrophages. Macrophage-specific Kdm6a knockout mice were further used to elucidate the effects of Kdm6a deficiency on periodontitis. Kdm6a deletion in macrophages rescued periodontal tissue inflammation, osteoclastogenesis, and alveolar bone loss in a mouse model of periodontitis. These findings suggest that CRD may intensify periodontitis by increasing the infiltration and activation of macrophages. Kdm6a promotes the inflammatory response in macrophages, which may participate in aggravated periodontitis via CRD.


Assuntos
Perda do Osso Alveolar , Periodontite , Camundongos , Animais , Perda do Osso Alveolar/metabolismo , Macrófagos , Periodontite/metabolismo , Inflamação/metabolismo , Histona Desmetilases/metabolismo , Porphyromonas gingivalis
10.
PLoS One ; 15(12): e0243844, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33301470

RESUMO

AIMS: Obstructive sleep apnea (OSA) is a widespread disease with high global socio-economic impact. However, detailed pathomechanisms are still unclear, partly because current animal models of OSA do not simulate spontaneous airway obstruction. We tested whether polytetrafluoroethylene (PTFE) injection into the tongue induces spontaneous obstructive apneas. METHODS AND RESULTS: PTFE (100 µl) was injected into the tongue of 31 male C57BL/6 mice and 28 mice were used as control. Spontaneous apneas and inspiratory flow limitations were recorded by whole-body plethysmography and mRNA expression of the hypoxia marker KDM6A was quantified by qPCR. Left ventricular function was assessed by echocardiography and ventricular CaMKII expression was measured by Western blotting. After PTFE injection, mice showed features of OSA such as significantly increased tongue diameters that were associated with significantly and sustained increased frequencies of inspiratory flow limitations and apneas. Decreased KDM6A mRNA levels indicated chronic hypoxemia. 8 weeks after surgery, PTFE-treated mice showed a significantly reduced left ventricular ejection fraction. Moreover, the severity of diastolic dysfunction (measured as E/e') correlated significantly with the frequency of apneas. Accordingly, CaMKII expression was significantly increased in PTFE mice and correlated significantly with the frequency of apneas. CONCLUSIONS: We describe here the first mouse model of spontaneous inspiratory flow limitations, obstructive apneas, and hypoxia by tongue enlargement due to PTFE injection. These mice develop systolic and diastolic dysfunction and increased CaMKII expression. This mouse model offers great opportunities to investigate the effects of obstructive apneas.


Assuntos
Contração Miocárdica , Apneia Obstrutiva do Sono/complicações , Apneia Obstrutiva do Sono/fisiopatologia , Língua/patologia , Disfunção Ventricular Esquerda/etiologia , Disfunção Ventricular Esquerda/fisiopatologia , Animais , Proteína Quinase Tipo 2 Dependente de Cálcio-Calmodulina/metabolismo , Diástole , Modelos Animais de Doenças , Eletrocardiografia , Histona Desmetilases/genética , Histona Desmetilases/metabolismo , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Inalação , Injeções , Pulmão/patologia , Masculino , Camundongos Endogâmicos C57BL , Miocárdio/patologia , Tamanho do Órgão , Politetrafluoretileno/administração & dosagem , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Apneia Obstrutiva do Sono/diagnóstico por imagem , Sístole , Disfunção Ventricular Esquerda/diagnóstico por imagem
11.
Cell Prolif ; 51(4): e12459, 2018 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-29656462

RESUMO

OBJECTIVES: Dental tissue-derived mesenchymal stem cells (MSCs)-mediated pulp-dentin regeneration is considered a potential approach for the regeneration of damaged teeth. Enhancing MSC-mediated pulp-dentin regeneration is based on an understanding of the molecular mechanisms underlying directed cell differentiation process. Histone demethylation enzyme, lysine demethylase 1A (KDM1A) can regulate the differentiation of some MSCs, but its role in dental tissue-derived MSCs is unclear. MATERIAL AND METHODS: We obtained SCAPs from immature teeth. Alkaline phosphatase (ALP) activity assay, Alizarin red staining, quantitative calcium analysis, osteogenesis-related genes expression and in vivo transplantation experiment were used to explore the osteo/dentinogenic differentiation. Co-immunoprecipitation (Co-IP) assay was used to investigate the binding protein. RESULTS: Knock-down of KDM1A reduced ALP activity and mineralization, promoted the expressions of osteo/dentinogenic differentiation markers DSPP, DMP1, BSP and key transcript factors, RUNX2, OSX, DLX2 in SCAPs, and also enhanced the osteo/dentinogenesis in vivo. In addition, KDM1A could associate with PLOD2 to form protein complex. And knock-down of PLOD2 inhibited ALP activity and mineralization, and promoted the expressions of DSPP, DMP1, BSP, RUNX2, OSX and DLX2 in SCAPs. CONCLUSIONS: KDM1A might have different role in different stages of osteo/dentinogenic differentiation process by binding partner with PLOD2, and finally resulted in the inhibited function for the osteo/dentinogenesis in SCAPs. Our studies provided a further understanding of the regulatory mechanisms of dynamic osteo/dentinogenic differentiation process in dental tissue MSCs.


Assuntos
Dentinogênese/fisiologia , Histona Desmetilases/metabolismo , Osteogênese/fisiologia , Pró-Colágeno-Lisina 2-Oxoglutarato 5-Dioxigenase/metabolismo , Animais , Diferenciação Celular , Células Cultivadas , Subunidade alfa 1 de Fator de Ligação ao Core/metabolismo , Papila Dentária/citologia , Feminino , Histona Desmetilases/antagonistas & inibidores , Histona Desmetilases/genética , Proteínas de Homeodomínio/metabolismo , Humanos , Camundongos , Camundongos Nus , Pró-Colágeno-Lisina 2-Oxoglutarato 5-Dioxigenase/antagonistas & inibidores , Pró-Colágeno-Lisina 2-Oxoglutarato 5-Dioxigenase/genética , Ligação Proteica , Interferência de RNA , RNA Interferente Pequeno/metabolismo , Fator de Transcrição Sp7/metabolismo , Transplante de Células-Tronco , Células-Tronco/citologia , Células-Tronco/metabolismo , Fatores de Transcrição/metabolismo
12.
Epigenetics ; 13(5): 557-572, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29927684

RESUMO

Periodontal disease (PD) afflicts 46% of Americans with no effective adjunctive therapies available. While most pharmacotherapy for PD targets bacteria, the host immune response is responsible for driving tissue damage and bone loss in severe disease. Herein, we establish that the histone demethylase KDM4B is a potential drug target for the treatment of PD. Immunohistochemical staining of diseased periodontal epithelium revealed an increased abundance of KDM4B that correlates with inflammation. In murine calvarial sections exposed to Aggregatibacter actinomycetemcomitans lipopolysaccharide (Aa-LPS), immunohistochemical staining revealed a significant increase in KDM4B protein expression. The 8-hydroxyquinoline ML324 is known to inhibit the related demethylase KDM4E in vitro, but has not been evaluated against any other targets. Our studies indicate that ML324 also inhibits KDM4B (IC50: 4.9 µM), and decreases the pro-inflammatory cytokine response to an Aa-LPS challenge in vitro. Our results suggest that KDM4B inhibition-induced immunosuppression works indirectly, requiring new protein synthesis. In addition, fluorescence-stained macrophages exhibited a significant decrease in global monomethyl histone 3 lysine 4 (H3K4me) levels following an Aa-LPS challenge that was prevented by KDM4B inhibition, suggesting this effect is produced through KDM1A-mediated demethylation of H3K4. Finally, ML324 inhibition of KDM4B in osteoclast progenitors produced a significant reduction in Aa-LPS-induced osteoclastogenesis. These data link histone methylation with host immune response to bacterial pathogens in PD, and suggest a previously unreported, alternative mechanism for epigenetic control of the host inflammatory environment. As such, KDM4B represents a new therapeutic target for treating hyper-inflammatory diseases that result in bone destruction.


Assuntos
Citocinas/metabolismo , Histona Desmetilases/metabolismo , Histona Desmetilases com o Domínio Jumonji/antagonistas & inibidores , Osteogênese , Periodontite/metabolismo , Animais , Linhagem Celular , Células Cultivadas , Inibidores Enzimáticos/farmacologia , Humanos , Histona Desmetilases com o Domínio Jumonji/metabolismo , Lipopolissacarídeos/toxicidade , Macrófagos/efeitos dos fármacos , Macrófagos/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Osteoclastos/citologia , Osteoclastos/efeitos dos fármacos , Osteoclastos/metabolismo , Periodontite/etiologia
13.
Int J Biol Sci ; 14(4): 381-389, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29725259

RESUMO

Kabuki syndrome is a rare genetic disorder characterized by distinct dysmorphic facial features, intellectual disability, and multiple developmental abnormalities. Despite more than 350 documented cases, the oro-dental spectrum associated with kabuki syndrome and expression of KMT2D (histone-lysine N-methyltransferase 2D) or KDM6A (lysine-specific demethylase 6A) genes in tooth development have not been well defined. Here, we report seven unrelated Thai patients with Kabuki syndrome having congenital absence of teeth, malocclusion, high-arched palate, micrognathia, and deviated tooth shape and size. Exome sequencing successfully identified that six patients were heterozygous for mutations in KMT2D, and one in KDM6A. Six were novel mutations, of which five were in KMT2D and one in KDM6A. They were truncating mutations including four frameshift deletions and two nonsense mutations. The predicted non-functional KMT2D and KDM6A proteins are expected to cause disease by haploinsufficiency. Our study expands oro-dental, medical, and mutational spectra associated with Kabuki syndrome. We also demonstrate for the first time that KMT2D and KDM6A are expressed in the dental epithelium of human tooth germs.


Assuntos
Anormalidades Múltiplas/genética , Proteínas de Ligação a DNA/genética , Face/anormalidades , Doenças Hematológicas/genética , Histona Desmetilases/genética , Proteínas de Neoplasias/genética , Proteínas Nucleares/genética , Anormalidades Dentárias/patologia , Germe de Dente/metabolismo , Doenças Vestibulares/genética , Anormalidades Múltiplas/metabolismo , Anormalidades Múltiplas/patologia , Proteínas de Ligação a DNA/metabolismo , Face/patologia , Mutação da Fase de Leitura , Doenças Hematológicas/metabolismo , Doenças Hematológicas/patologia , Histona Desmetilases/metabolismo , Humanos , Proteínas de Neoplasias/metabolismo , Proteínas Nucleares/metabolismo , Anormalidades Dentárias/genética , Anormalidades Dentárias/metabolismo , Doenças Vestibulares/metabolismo , Doenças Vestibulares/patologia
14.
Plant Physiol Biochem ; 132: 183-188, 2018 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-30212759

RESUMO

JmjC-domain-containing (JmjC) protein, an important kind of histone demethylase in plants, plays key roles in multiple growth and development processes and in adversity resistance. In this study, we found that OsJMJ703, a known histone demethylase, is expressed in various tissues. Furthermore, over-expression of OsJMJ703 influenced the type of rice panicle, and knock-down of the expression of OsJMJ703 showed an earlier flowering time in rice. In addition, OsJMJ703 is involved in abiotic stress. Transgenic rice of over-expressing OsJMJ703 is sensitive to drought stress, whereas knocking down OsJMJ703 enhances the tolerance to drought stress. This study provides a theoretical basis of the biological function of JmjC protein and further promotes the study of drought resistance.


Assuntos
Secas , Genes de Plantas , Histona Desmetilases/genética , Oryza/enzimologia , Oryza/crescimento & desenvolvimento , Desenvolvimento Vegetal/genética , Estresse Fisiológico/genética , Flores/fisiologia , Regulação da Expressão Gênica de Plantas/efeitos dos fármacos , Técnicas de Silenciamento de Genes , Histona Desmetilases/metabolismo , Especificidade de Órgãos/genética , Oryza/efeitos dos fármacos , Oryza/genética , Proteínas de Plantas/genética , Proteínas de Plantas/metabolismo , Polietilenoglicóis/toxicidade , Estresse Fisiológico/efeitos dos fármacos
15.
Mater Sci Eng C Mater Biol Appl ; 54: 142-9, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-26046277

RESUMO

Over the past decade, theranostic nanoparticles with microsize and multifunctional ability have emerged as a new platform in biomedical field, such as cancer therapy, optical imaging and gene therapy. Gene therapy has been recently shown as a promising tool for tissue engineering as safe and effective nanotechnology-based delivery methods are developed. Controlling adhesion and differentiation of stem cells is critical for tissue regeneration. In this study, we have developed poly-sodium 4-styrenesulfonate (PSS) and poly-allylamine hydrochloride (PAH) coated AuNR-based nanocarriers, which are capable of delivering small interfering RNA (siRNA) against LSD1 to induce the differentiation of human mesenchymal stem cells. To further study the mechanism, we tested the stemness and differentiation genes and found that they have been changed with LSD1 down-regulation. In addition, with the hepatocyte growth factor (HGF), LSD1 siRNA delivery by AuNRs could promote the differentiation of the human mesenchymal stem cells (human MSCs) into a hepatocyte lineage in vitro. Our results suggest for the first time use of AuNRs as nanocarriers of delivery LSD1 siRNA to induce the differentiation of human MSCs into a hepatocyte lineage, and envision the potential application of nanotechnology in tissue remodeling (such as liver and bone) in vivo, eventually translating to clinical applications.


Assuntos
Diferenciação Celular , Ouro/química , Histona Desmetilases/genética , Células-Tronco Mesenquimais/metabolismo , Nanotubos/química , RNA Interferente Pequeno/genética , Adesão Celular , Células Cultivadas , Materiais Revestidos Biocompatíveis/química , Regulação para Baixo , Sistemas de Liberação de Medicamentos , Técnicas de Transferência de Genes , Marcadores Genéticos , Fator de Crescimento de Hepatócito/genética , Fator de Crescimento de Hepatócito/metabolismo , Fator 1-alfa Nuclear de Hepatócito/genética , Fator 1-alfa Nuclear de Hepatócito/metabolismo , Hepatócitos/citologia , Hepatócitos/metabolismo , Histona Desmetilases/metabolismo , Humanos , Células-Tronco Mesenquimais/citologia , Poliaminas/química , Polímeros/química , Regiões Promotoras Genéticas , RNA Interferente Pequeno/metabolismo , Ácidos Sulfônicos/química , Engenharia Tecidual/métodos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA