Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Sci Rep ; 12(1): 3497, 2022 03 10.
Artigo em Inglês | MEDLINE | ID: mdl-35273210

RESUMO

Oxygen is a key regulator of both development and homeostasis. To study the role of oxygen, a variety of in vitro and ex vivo cell and tissue models have been used in biomedical research. However, because of ambiguity surrounding the level of oxygen that cells experience in vivo, the cellular pathway related to oxygenation state and hypoxia have been inadequately studied in many of these models. Here, we devised a method to determine the oxygen tension in bone marrow monocytes using two-photon phosphorescence lifetime imaging microscopy with the cell-penetrating phosphorescent probe, BTPDM1. Phosphorescence lifetime imaging revealed the physiological level of oxygen tension in monocytes to be 5.3% in live mice exposed to normal air. When the mice inhaled hypoxic air, the level of oxygen tension in bone marrow monocytes decreased to 2.4%. By performing in vitro cell culture experiment within the physiological range of oxygen tension, hypoxia changed the molecular phenotype of monocytes, leading to enhanced the expression of CD169 and CD206, which are markers of a unique subset of macrophages in bone marrow, osteal macrophages. This current study enables the determination of the physiological range of oxygen tension in bone marrow with spatial resolution at a cellular level and application of this information on oxygen tension in vivo to in vitro assays. Quantifying oxygen tension in tissues can provide invaluable information on metabolism under physiological and pathophyisological conditions. This method will open new avenues for research on oxygen biology.


Assuntos
Medula Óssea , Microscopia , Animais , Medula Óssea/metabolismo , Hipóxia/metabolismo , Camundongos , Monócitos/metabolismo , Oxigênio/metabolismo , Fótons
2.
Nat Commun ; 13(1): 1066, 2022 02 24.
Artigo em Inglês | MEDLINE | ID: mdl-35210428

RESUMO

Bone metabolism is regulated by the cooperative activity between bone-forming osteoblasts and bone-resorbing osteoclasts. However, the mechanisms mediating the switch between the osteoblastic and osteoclastic phases have not been fully elucidated. Here, we identify a specific subset of mature osteoblast-derived extracellular vesicles that inhibit bone formation and enhance osteoclastogenesis. Intravital imaging reveals that mature osteoblasts secrete and capture extracellular vesicles, referred to as small osteoblast vesicles (SOVs). Co-culture experiments demonstrate that SOVs suppress osteoblast differentiation and enhance the expression of receptor activator of NF-κB ligand, thereby inducing osteoclast differentiation. We also elucidate that the SOV-enriched microRNA miR-143 inhibits Runt-related transcription factor 2, a master regulator of osteoblastogenesis, by targeting the mRNA expression of its dimerization partner, core-binding factor ß. In summary, we identify SOVs as a mode of cell-to-cell communication, controlling the dynamic transition from bone-forming to bone-resorbing phases in vivo.


Assuntos
Reabsorção Óssea , Osteogênese , Reabsorção Óssea/metabolismo , Diferenciação Celular , Humanos , Osteoblastos/metabolismo , Osteoclastos/metabolismo , Osteogênese/genética , Ligante RANK/metabolismo , Transdução de Sinais
3.
Autophagy ; 18(10): 2323-2332, 2022 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-35025696

RESUMO

Maintenance of bone integrity is mediated by the balanced actions of osteoblasts and osteoclasts. Because macroautophagy/autophagy regulates osteoblast mineralization, osteoclast differentiation, and their secretion from osteoclast cells, autophagy deficiency in osteoblasts or osteoclasts can disrupt this balance. However, it remains unclear whether upregulation of autophagy becomes beneficial for suppression of bone-associated diseases. In this study, we found that genetic upregulation of autophagy in osteoblasts facilitated bone formation. We generated mice in which autophagy was specifically upregulated in osteoblasts by deleting the gene encoding RUBCN/Rubicon, a negative regulator of autophagy. The rubcnflox/flox;Sp7/Osterix-Cre mice showed progressive skeletal abnormalities in femur bones. Consistent with this, RUBCN deficiency in osteoblasts resulted in elevated differentiation and mineralization, as well as an increase in the elevated expression of key transcription factors involved in osteoblast function such as Runx2 and Bglap/Osteocalcin. Furthermore, RUBCN deficiency in osteoblasts accelerated autophagic degradation of NOTCH intracellular domain (NICD) and downregulated the NOTCH signaling pathway, which negatively regulates osteoblast differentiation. Notably, osteoblast-specific deletion of RUBCN alleviated the phenotype in a mouse model of osteoporosis. We conclude that RUBCN is a key regulator of bone homeostasis. On the basis of these findings, we propose that medications targeting RUBCN or autophagic degradation of NICD could be used to treat age-related osteoporosis and bone fracture.Abbreviations: ALPL: alkaline phosphatase, liver/bone/kidney; BCIP/NBT: 5-bromo-4-chloro-3'-indolyl phosphate/nitro blue tetrazolium; BMD: bone mineral density; BV/TV: bone volume/total bone volume; MAP1LC3/LC3: microtubule-associated protein 1 light chain 3; MTOR: mechanistic target of rapamycin kinase; NICD: NOTCH intracellular domain; RB1CC1/FIP200: RB1-inducible coiled-coil 1; RUBCN/Rubicon: RUN domain and cysteine-rich domain containing, Beclin 1-interacting protein; SERM: selective estrogen receptor modulator; TNFRSF11B/OCIF: tumor necrosis factor receptor superfamily, member 11b (osteoprotegerin).


Assuntos
Osteogênese , Osteoporose , Fosfatase Alcalina/metabolismo , Animais , Autofagia/fisiologia , Proteína Beclina-1/metabolismo , Diferenciação Celular , Subunidade alfa 1 de Fator de Ligação ao Core/metabolismo , Cisteína/metabolismo , Camundongos , Proteínas Associadas aos Microtúbulos/metabolismo , Osteoblastos/patologia , Osteocalcina/metabolismo , Osteoporose/metabolismo , Osteoporose/patologia , Osteoprotegerina/metabolismo , Fosfatos/metabolismo , Receptores Notch , Moduladores Seletivos de Receptor Estrogênico/metabolismo , Sirolimo , Serina-Treonina Quinases TOR/metabolismo
4.
J Bone Miner Metab ; 39(3): 353-359, 2021 May.
Artigo em Inglês | MEDLINE | ID: mdl-33106978

RESUMO

INTRODUCTION: Overexpression studies have been commonly used to yield significant advances in cell biology. In vitro osteoclast culturing involves the differentiation of bone marrow-derived monocyte macrophage precursors (BMMs) in medium supplemented with macrophage colony-stimulating factor and receptor activator of nuclear factor-kB ligand (RANKL) into mature osteoclasts. Retroviral vectors are the gold standards for efficient gene delivery into BMMs. While this strategy is effective in BMMs that are in the early stages of differentiation, it is ineffective in RANKL-treated BMMs such as mono- and multinucleated osteoclasts. This study attempted to enhance gene delivery into differentiated BMMs using liposome-mediated RNA transfection. MATERIAL AND METHODS: BMMs were transfected with an EYFP overexpression plasmid or EYFP RNA by lipofection, or transduced with a retroviral vector expressing EYFP. EYFP expression was assessed by flow cytometry. RESULTS: We performed overexpression analyses using enhanced yellow fluorescent protein (EYFP). Although EYFP expression was observed 24 h after infection of BMMs with a recombinant retrovirus containing EYFP, expression of EYFP was observed within 3 h of transfection with EYFP RNA. Moreover, the efficiency of EYFP RNA for gene delivery into BMMs was comparable to that of retroviral transduction of EYFP. In contrast, while very few BMMs stimulated by RANKL for two days expressed EYFP after retroviral infection, more than half of the cells expressed EYFP after transfection with EYFP RNA. CONCLUSION: RNA-mediated gene delivery is quick and easy method for performing gain-of-function analyses in primary osteoclast precursors and mature osteoclasts.


Assuntos
Mutação com Ganho de Função , Técnicas de Transferência de Genes , Osteoclastos/metabolismo , Animais , Proteínas de Bactérias/metabolismo , Células da Medula Óssea/metabolismo , Diferenciação Celular , Células Cultivadas , Proteínas Luminescentes/metabolismo , Camundongos Endogâmicos C57BL , Transdução de Sinais , Transfecção
5.
J Bone Miner Res ; 33(10): 1785-1798, 2018 10.
Artigo em Inglês | MEDLINE | ID: mdl-29893999

RESUMO

Osteoclast differentiation is a dynamic differentiation process, which is accompanied by dramatic changes in metabolic status as well as in gene expression. Recent findings have revealed an essential connection between metabolic reprogramming and dynamic gene expression changes during osteoclast differentiation. However, the upstream regulatory mechanisms that drive these metabolic changes in osteoclastogenesis remain to be elucidated. Here, we demonstrate that induced deletion of a tumor suppressor gene, Folliculin (Flcn), in mouse osteoclast precursors causes severe osteoporosis in 3 weeks through excess osteoclastogenesis. Flcn-deficient osteoclast precursors reveal cell autonomous accelerated osteoclastogenesis with increased sensitivity to receptor activator of NF-κB ligand (RANKL). We demonstrate that Flcn regulates oxidative phosphorylation and purine metabolism through suppression of nuclear localization of the transcription factor Tfe3, thereby inhibiting expression of its target gene Pgc1. Metabolome studies revealed that Flcn-deficient osteoclast precursors exhibit significant augmentation of oxidative phosphorylation and nucleotide production, resulting in an enhanced purinergic signaling loop that is composed of controlled ATP release and autocrine/paracrine purinergic receptor stimulation. Inhibition of this purinergic signaling loop efficiently blocks accelerated osteoclastogenesis in Flcn-deficient osteoclast precursors. Here, we demonstrate an essential and novel role of the Flcn-Tfe3-Pgc1 axis in osteoclastogenesis through the metabolic reprogramming of oxidative phosphorylation and purine metabolism. © 2018 The Authors Journal of Bone and Mineral Research published by Wiley Periodicals, Inc. on behalf of American Society for Bone and Mineral Research (ASBMR).


Assuntos
Osteoclastos/metabolismo , Osteogênese , Proteínas Proto-Oncogênicas/metabolismo , Proteínas Supressoras de Tumor/metabolismo , Animais , Fatores de Transcrição de Zíper de Leucina e Hélice-Alça-Hélix Básicos/metabolismo , Medula Óssea/patologia , Camundongos , Camundongos Knockout , Biogênese de Organelas , Osteoclastos/patologia , Osteoporose/metabolismo , Osteoporose/patologia , Fosforilação Oxidativa , Purinas/metabolismo , Células RAW 264.7 , Transdução de Sinais , Fatores de Transcrição/metabolismo , Regulação para Cima
6.
Sci Rep ; 8(1): 7504, 2018 05 14.
Artigo em Inglês | MEDLINE | ID: mdl-29760402

RESUMO

Bidirectional transcription has been proposed to play a role associated with enhancer activity. Transcripts called enhancer RNAs (eRNAs) play important roles in gene regulation; however, their roles in osteoclasts are unknown. To analyse eRNAs in osteoclasts comprehensively, we used cap-analysis of gene expression (CAGE) to detect adjacent transcription start sites (TSSs) that were distant from promoters for protein-coding gene expression. When comparing bidirectional TSSs between osteoclast precursors and osteoclasts, we found that bidirectional TSSs were located in the 5'-flanking regions of the Nrp2 and Dcstamp genes. We also detected bidirectional TSSs in the intron region of the Nfatc1 gene. To investigate the role of bidirectional transcription in osteoclasts, we performed loss of function analyses using the CRISPR/Cas9 system. Targeted deletion of the DNA regions between the bidirectional TSSs led to decreased expression of the bidirectional transcripts, as well as the protein-coding RNAs of Nrp2, Dcstamp, and Nfatc1, suggesting that these transcripts act as eRNAs. Furthermore, osteoclast differentiation was impaired by targeted deletion of bidirectional eRNA regions. The combined results show that eRNAs play important roles in osteoclastogenic gene regulation, and may therefore provide novel insights to elucidate the transcriptional mechanisms that control osteoclast differentiation.


Assuntos
Elementos Facilitadores Genéticos , Perfilação da Expressão Gênica/métodos , Osteoclastos/citologia , Ligante RANK/farmacologia , Capuzes de RNA/metabolismo , Animais , Sistemas CRISPR-Cas , Diferenciação Celular/efeitos dos fármacos , Células Cultivadas , Regulação da Expressão Gênica , Proteínas de Membrana/genética , Camundongos , Fatores de Transcrição NFATC/genética , Neuropilina-2/genética , Osteoclastos/química , Osteoclastos/efeitos dos fármacos , Regiões Promotoras Genéticas , Sítio de Iniciação de Transcrição
7.
Clin Calcium ; 26(5): 713-9, 2016 May.
Artigo em Japonês | MEDLINE | ID: mdl-27117617

RESUMO

Osteoclasts dramatically alter their metabolic activity during cell differentiation. This change in the metabolic status is termed'metabolic reprogramming',but its role in osteoclast is not fully understood. Using metabolomics approach, we found that metabolic reprogramming during osteoclast differentiation increased intracellular S-adenosyle methionine (SAM), a metabolite of the methionine cycle. SAM is the universal methyl donor for methylation reactions, including histone and DNA methylation. Furthermore, SAM-mediated DNA methylation is required for osteoclast differentiation. These findings reveal the novel role of SAM metabolism in regulating osteoclast differentiation.


Assuntos
Diferenciação Celular/genética , Reprogramação Celular/fisiologia , Hematopoese/fisiologia , Metionina/metabolismo , Osteoclastos/citologia , Animais , Diferenciação Celular/imunologia , Reprogramação Celular/imunologia , Metilação de DNA/genética , Humanos , Osteoclastos/imunologia
8.
Mol Cell Biol ; 36(11): 1610-20, 2016 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-27001307

RESUMO

Bone homeostasis is maintained by a balance in activity between bone-resorbing osteoclasts and bone-forming osteoblasts. Shifting the balance toward bone resorption causes osteolytic bone diseases such as rheumatoid arthritis and periodontitis. Osteoclast differentiation is regulated by receptor activator of nuclear factor κB ligand (RANKL), which, under some pathological conditions, is produced by T and B lymphocytes and synoviocytes. However, the mechanism underlying bone destruction in other diseases is little understood. Bone destruction caused by cholesteatoma, an epidermal cyst in the middle ear resulting from hyperproliferation of keratinizing squamous epithelium, can lead to lethal complications. In this study, we succeeded in generating a model for cholesteatoma, epidermal cyst-like tissue, which has the potential for inducing osteoclastogenesis in mice. Furthermore, an in vitro coculture system composed of keratinocytes, fibroblasts, and osteoclast precursors was used to demonstrate that keratinocytes stimulate osteoclast differentiation through the induction of RANKL in fibroblasts. Thus, this study demonstrates that intercellular communication between keratinocytes and fibroblasts is involved in the differentiation and function of osteoclasts, which may provide the molecular basis of a new therapeutic strategy for cholesteatoma-induced bone destruction.


Assuntos
Colesteatoma/patologia , Fibroblastos/citologia , Queratinócitos/citologia , Osteoclastos/citologia , Ligante RANK/metabolismo , Animais , Comunicação Celular , Diferenciação Celular , Células Cultivadas , Colesteatoma/metabolismo , Técnicas de Cocultura , Modelos Animais de Doenças , Fibroblastos/metabolismo , Queratinócitos/metabolismo , Camundongos , Osteoclastos/metabolismo
9.
Nat Med ; 21(3): 281-7, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25706873

RESUMO

Metabolic reprogramming occurs in response to the cellular environment to mediate differentiation, but the fundamental mechanisms linking metabolic processes to differentiation programs remain to be elucidated. During osteoclast differentiation, a shift toward more oxidative metabolic processes occurs. In this study we identified the de novo DNA methyltransferase 3a (Dnmt3a) as a transcription factor that couples these metabolic changes to osteoclast differentiation. We also found that receptor activator of nuclear factor-κB ligand (RANKL), an essential cytokine for osteoclastogenesis, induces this metabolic shift towards oxidative metabolism, which is accompanied by an increase in S-adenosylmethionine (SAM) production. We found that SAM-mediated DNA methylation by Dnmt3a regulates osteoclastogenesis via epigenetic repression of anti-osteoclastogenic genes. The importance of Dnmt3a in bone homeostasis was underscored by the observations that Dnmt3a-deficient osteoclast precursor cells do not differentiate efficiently into osteoclasts and that mice with an osteoclast-specific deficiency in Dnmt3a have elevated bone mass due to a smaller number of osteoclasts. Furthermore, inhibition of DNA methylation by theaflavin-3,3'-digallate abrogated bone loss in models of osteoporosis. Thus, this study reveals the role of epigenetic processes in the regulation of cellular metabolism and differentiation, which may provide the molecular basis for a new therapeutic strategy for a variety of bone disorders.


Assuntos
Reabsorção Óssea/genética , Diferenciação Celular/genética , DNA (Citosina-5-)-Metiltransferases/genética , Osteoclastos/citologia , Osteogênese/genética , Ligante RANK/genética , S-Adenosilmetionina/metabolismo , Animais , Biflavonoides/farmacologia , Catequina/análogos & derivados , Catequina/farmacologia , Diferenciação Celular/efeitos dos fármacos , Diferenciação Celular/fisiologia , DNA (Citosina-5-)-Metiltransferases/antagonistas & inibidores , DNA (Citosina-5-)-Metiltransferases/fisiologia , Metilação de DNA/genética , DNA Metiltransferase 3A , Regulação para Baixo , Expressão Gênica , Camundongos , Osteogênese/efeitos dos fármacos , Osteogênese/fisiologia , Ligante RANK/fisiologia , Fatores de Transcrição
10.
J Biol Chem ; 290(10): 6326-37, 2015 Mar 06.
Artigo em Inglês | MEDLINE | ID: mdl-25605732

RESUMO

Cell cycle-arrested cancer cells are resistant to conventional chemotherapy that acts on the mitotic phases of the cell cycle, although the molecular mechanisms involved in halting cell cycle progression remain unclear. Here, we demonstrated that RFPL4A, an uncharacterized ubiquitin ligase, induced G1 retention and thus conferred decreased sensitivity to chemotherapy in the human colorectal cancer cell line, HCT116. Long term time lapse observations in HCT116 cells bearing a "fluorescence ubiquitin-based cell cycle indicator" identified a characteristic population that is viable but remains in the G1 phase for an extended period of time (up to 56 h). Microarray analyses showed that expression of RFPL4A was significantly up-regulated in these G1-arrested cells, not only in HCT116 cells but also in other cancer cell lines, and overexpression of RFPL4A increased the G1 population and decreased sensitivity to chemotherapy. However, knockdown of RFPL4A expression caused the cells to resume mitosis and induced their susceptibility to anti-cancer drugs in vitro and in vivo. These results indicate that RFPL4A is a novel factor that increases the G1 population and decreases sensitivity to chemotherapy and thus may be a promising therapeutic target for refractory tumor conditions.


Assuntos
Antineoplásicos/administração & dosagem , Neoplasias Colorretais/tratamento farmacológico , Neoplasias Colorretais/genética , Ubiquitina-Proteína Ligases/biossíntese , Apoptose/efeitos dos fármacos , Ciclo Celular/efeitos dos fármacos , Ciclo Celular/genética , Neoplasias Colorretais/patologia , Fase G1/efeitos dos fármacos , Fase G1/genética , Regulação Neoplásica da Expressão Gênica , Células HCT116 , Humanos , Análise em Microsséries
11.
J Bone Miner Metab ; 32(3): 331-6, 2014 May.
Artigo em Inglês | MEDLINE | ID: mdl-24366621

RESUMO

The development of methods for differentiation of embryonic stem cells (ESCs) and induced pluripotent stem cell (iPSCs) into functional cells have helped to analyze the mechanism regulating cellular processes and to explore cell-based assays for drug discovery. Although several reports have demonstrated methods for differentiation of mouse ESCs into osteoclast-like cells, it remains unclear whether these methods are applicable for differentiation of iPSCs to osteoclasts. In this study, we developed a simple method for stepwise differentiation of mouse ESCs and iPSCs into bone-resorbing osteoclasts based upon a monoculture approach consisting of three steps. First, based on conventional hanging-drop methods, embryoid bodies (EBs) were produced from mouse ESCs or iPSCs. Second, EBs were cultured in medium supplemented with macrophage colony-stimulating factor (M-CSF), and differentiated to osteoclast precursors, which expressed CD11b. Finally, ESC- or iPSC-derived osteoclast precursors stimulated with receptor activator of nuclear factor-B ligand (RANKL) and M-CSF formed large multinucleated osteoclast-like cells that expressed tartrate-resistant acid phosphatase and were capable of bone resorption. Molecular analysis showed that the expression of osteoclast marker genes such as Nfatc1, Ctsk, and Acp5 are increased in a RANKL-dependent manner. Thus, our procedure is simple and easy and would be helpful for stem cell-based bone research.


Assuntos
Diferenciação Celular/fisiologia , Células-Tronco Embrionárias/fisiologia , Técnicas In Vitro/métodos , Células-Tronco Pluripotentes Induzidas/fisiologia , Osteoclastos/fisiologia , Fosfatase Ácida/metabolismo , Animais , Reabsorção Óssea/metabolismo , Reabsorção Óssea/fisiopatologia , Linhagem Celular , Células-Tronco Embrionárias/metabolismo , Células-Tronco Pluripotentes Induzidas/metabolismo , Isoenzimas/metabolismo , Fator Estimulador de Colônias de Macrófagos/metabolismo , Camundongos , Osteoclastos/metabolismo , Ligante RANK/metabolismo , Receptor Ativador de Fator Nuclear kappa-B/metabolismo , Fosfatase Ácida Resistente a Tartarato
12.
PLoS One ; 8(12): e83629, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24386239

RESUMO

The mechanism behind the spatiotemporal control of cancer cell dynamics and its possible association with cell proliferation has not been well established. By exploiting the intravital imaging technique, we found that cancer cell motility and invasive properties were closely associated with the cell cycle. In vivo inoculation of human colon cancer cells bearing fluorescence ubiquitination-based cell cycle indicator (Fucci) demonstrated an unexpected phenomenon: S/G2/M cells were more motile and invasive than G1 cells. Microarray analyses showed that Arhgap11a, an uncharacterized Rho GTPase-activating protein (RhoGAP), was expressed in a cell-cycle-dependent fashion. Expression of ARHGAP11A in cancer cells suppressed RhoA-dependent mechanisms, such as stress fiber formation and focal adhesion, which made the cells more prone to migrate. We also demonstrated that RhoA suppression by ARHGAP11A induced augmentation of relative Rac1 activity, leading to an increase in the invasive properties. RNAi-based inhibition of Arhgap11a reduced the invasion and in vivo expansion of cancers. Additionally, analysis of human specimens showed the significant up-regulation of Arhgap11a in colon cancers, which was correlated with clinical invasion status. The present study suggests that ARHGAP11A, a cell cycle-dependent RhoGAP, is a critical regulator of cancer cell mobility and is thus a promising therapeutic target in invasive cancers.


Assuntos
Ciclo Celular , Movimento Celular , Proteínas Ativadoras de GTPase/metabolismo , Neoplasias/metabolismo , Neoplasias/patologia , Ciclo Celular/genética , Linhagem Celular Tumoral , Movimento Celular/genética , Neoplasias Colorretais/genética , Neoplasias Colorretais/metabolismo , Neoplasias Colorretais/patologia , Ativação Enzimática , Proteínas Ativadoras de GTPase/antagonistas & inibidores , Proteínas Ativadoras de GTPase/genética , Expressão Gênica , Técnicas de Silenciamento de Genes , Células HCT116 , Humanos , Invasividade Neoplásica , Neoplasias/genética
14.
J Bone Miner Res ; 26(10): 2463-72, 2011 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-21713993

RESUMO

Serum calcium and phosphate homeostasis is critically regulated by parathyroid hormone (PTH) secreted by the parathyroid glands. Parathyroid glands develop from the bilateral parathyroid-thymus common primordia. In mice, the expression of transcription factor Glial cell missing 2 (Gcm2) begins in the dorsal/anterior part of the primordium on embryonic day 9.5 (E9.5), specifying the parathyroid domain. The parathyroid primordium then separates from the thymus primordium and migrates to its adult location beside the thyroid gland by E15.5. Genetic ablation of gcm2 results in parathyroid agenesis in mice, indicating that Gcm2 is essential for early parathyroid organogenesis. However, the regulation of parathyroid development at later stages is not well understood. Here we show that transcriptional activator v-maf musculoaponeurotic fibrosarcoma oncogene homologue B (MafB) is developmentally expressed in parathyroid cells after E11.5. MafB expression was lost in the parathyroid primordium of gcm2 null mice. The parathyroid glands of mafB(+/-) mice were mislocalized between the thymus and thyroid. In mafB(-/-) mice, the parathyroid did not separate from the thymus. Furthermore, in mafB(-/-) mice, PTH expression and secretion were impaired; expression levels of renal cyp27b1, one of the target genes of PTH, was decreased; and bone mineralization was reduced. We also demonstrate that although Gcm2 alone does not stimulate the PTH gene promoter, it associates with MafB to synergistically activate PTH expression. Taken together, our results suggest that MafB regulates later steps of parathyroid development, that is, separation from the thymus and migration toward the thyroid. MafB also regulates the expression of PTH in cooperation with Gcm2.


Assuntos
Fator de Transcrição MafB/metabolismo , Proteínas Nucleares/metabolismo , Glândulas Paratireoides/embriologia , Hormônio Paratireóideo/metabolismo , Fatores de Transcrição/metabolismo , Animais , Sequência de Bases , Primers do DNA , Ensaio de Desvio de Mobilidade Eletroforética , Imuno-Histoquímica , Camundongos , Camundongos Endogâmicos C57BL , Hormônio Paratireóideo/sangue , Reação em Cadeia da Polimerase em Tempo Real
15.
J Clin Invest ; 120(10): 3455-65, 2010 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-20877012

RESUMO

Aging leads to the disruption of the homeostatic balance of multiple biological systems. In bone marrow multipotent mesenchymal cells undergo differentiation into various anchorage-dependent cell types, including osteoblasts and adipocytes. With age as well as with treatment of antidiabetic drugs such as thiazolidinediones, mesenchymal cells favor differentiation into adipocytes, resulting in an increased number of adipocytes and a decreased number of osteoblasts, causing osteoporosis. The mechanism behind this differentiation switch is unknown. Here we show an age-related decrease in the expression of Maf in mouse mesenchymal cells, which regulated mesenchymal cell bifurcation into osteoblasts and adipocytes by cooperating with the osteogenic transcription factor Runx2 and inhibiting the expression of the adipogenic transcription factor Pparg. The crucial role of Maf in both osteogenesis and adipogenesis was underscored by in vivo observations of delayed bone formation in perinatal Maf(-/-) mice and an accelerated formation of fatty marrow associated with bone loss in aged Maf(+/-) mice. This study identifies a transcriptional mechanism for an age-related switch in cell fate determination and may provide a molecular basis for novel therapeutic strategies against age-related bone diseases.


Assuntos
Envelhecimento , Células-Tronco Mesenquimais/citologia , Osteoblastos/citologia , Proteínas Proto-Oncogênicas c-maf/fisiologia , Adipogenia , Animais , Diferenciação Celular , Linhagem da Célula , Células Cultivadas , Subunidade alfa 1 de Fator de Ligação ao Core/análise , Camundongos , Camundongos Endogâmicos C57BL , Osteogênese , Osteoporose/etiologia , Osteoporose/terapia , PPAR gama/antagonistas & inibidores , PPAR gama/genética , Espécies Reativas de Oxigênio/metabolismo , Proteína Supressora de Tumor p53/fisiologia
16.
Proc Natl Acad Sci U S A ; 107(7): 3117-22, 2010 Feb 16.
Artigo em Inglês | MEDLINE | ID: mdl-20133620

RESUMO

Regulation of irreversible cell lineage commitment depends on a delicate balance between positive and negative regulators, which comprise a sophisticated network of transcription factors. Receptor activator of NF-kappaB ligand (RANKL) stimulates the differentiation of bone-resorbing osteoclasts through the induction of nuclear factor of activated T cells c1 (NFATc1), the essential transcription factor for osteoclastogenesis. Osteoclast-specific robust induction of NFATc1 is achieved through an autoamplification mechanism, in which NFATc1 is constantly activated by calcium signaling while the negative regulators of NFATc1 are suppressed. However, it has been unclear how such negative regulators are repressed during osteoclastogenesis. Here we show that B lymphocyte-induced maturation protein-1 (Blimp1; encoded by Prdm1), which is induced by RANKL through NFATc1 during osteoclastogenesis, functions as a transcriptional repressor of anti-osteoclastogenic genes such as Irf8 and Mafb. Overexpression of Blimp1 leads to an increase in osteoclast formation, and Prdm1-deficient osteoclast precursor cells do not undergo osteoclast differentiation efficiently. The importance of Blimp1 in bone homeostasis is underscored by the observation that mice with an osteoclast-specific deficiency in the Prdm1 gene exhibit a high bone mass phenotype caused by a decreased number of osteoclasts. Thus, NFATc1 choreographs the determination of cell fate in the osteoclast lineage by inducing the repression of negative regulators as well as through its effect on positive regulators.


Assuntos
Diferenciação Celular/fisiologia , Osteoclastos/citologia , Proteínas Repressoras/metabolismo , Fatores de Transcrição/metabolismo , Animais , Células Cultivadas , Imunoprecipitação da Cromatina , Primers do DNA/genética , Citometria de Fluxo , Immunoblotting , Fatores Reguladores de Interferon/metabolismo , Fator de Transcrição MafB/metabolismo , Camundongos , Camundongos Mutantes , Fatores de Transcrição NFATC/metabolismo , Oligonucleotídeos/genética , Osteoclastos/fisiologia , Reação em Cadeia da Polimerase , Fator 1 de Ligação ao Domínio I Regulador Positivo , Ligante RANK/metabolismo , Interferência de RNA , Elementos Reguladores de Transcrição/fisiologia , Organismos Livres de Patógenos Específicos
17.
Dev Growth Differ ; 52(2): 245-50, 2010 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-20100247

RESUMO

Phenotypic rescue experiments have been commonly used in zebrafish since it is convenient to study the causality of mutant phenotypes just by injecting mRNA into embryos. However, this strategy is only effective for phenotypes at early embryonic stages due to mRNA instability. For later developmental stages, DNA constructs are used to express exogenous genes, while it is usually ineffective owing to the problem of mosaicism. This study attempted to solve the problem by using Tol2-mediated transgenesis. As a model case, we used vlad tepes (vlt), a zebrafish gata1 mutant, whose phenotypes have never been able to be rescued at later stages by transient rescue experiments. Blood cell-specific transgenic expression of gata1 was driven by its own promoter/enhancer elements. The co-injection of a Tol2-donor plasmid containing gata1 cDNA and transposase mRNA efficiently rescued the bloodless phenotypes of vlt even in day 12 larvae when definitive erythropoiesis took place with primitive erythropoiesis. This Tol2-mediated rescue is therefore considered to be a quick and easy method for analyzing the mutant phenotypes in zebrafish.


Assuntos
Elementos de DNA Transponíveis/genética , Fator de Transcrição GATA1/genética , Mutagênese Insercional , Mutação/genética , Transcrição Gênica/genética , Proteínas de Peixe-Zebra/genética , Peixe-Zebra/genética , Animais , Regulação da Expressão Gênica no Desenvolvimento/genética , Fenótipo , Peixe-Zebra/embriologia
18.
Arch Biochem Biophys ; 477(1): 139-45, 2008 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-18585365

RESUMO

We previously demonstrated that Nrf2 regulates oxidized LDL-mediated CD36 expression in macrophages. The current study aimed to determine the mechanism of Nrf2-mediated macrophage CD36 induction. Treatment with the Nrf2 activator diethylmaleate, but not PPARgamma specific ligands, caused marked upregulation of CD36 in mouse macrophage RAW264.7 cells. Similarly, Nrf2 activators induced CD36 expression in bone marrow-derived macrophages in a Nrf2-dependent manner. Induced expression of the three alternative first exons of mouse CD36, deemed 1A, 1B, and 1C, occurred upon Nrf2 activation with exon1A mainly contributing to the CD36 expression. Four antioxidant response elements (AREs) lie within close proximity to these three exons, and chromatin immunoprecipitation assays demonstrated that two AREs upstream of exon1A, the distal 1A-ARE1, and the proximal 1A-ARE2, were Nrf2-responsive. Luciferase reporter assays conclusively demonstrated that 1A-ARE2 is the critical regulatory element for the Nrf2-mediated gene expression. Thus Nrf2 directly regulates CD36 gene expression by binding to 1A-ARE2.


Assuntos
Antioxidantes/metabolismo , Antígenos CD36/genética , Éxons , Macrófagos/metabolismo , Fator 2 Relacionado a NF-E2/fisiologia , Elementos de Resposta/fisiologia , Animais , Antioxidantes/farmacologia , Western Blotting , Linhagem Celular , Imunoprecipitação da Cromatina , Regulação da Expressão Gênica , Humanos , Camundongos , Camundongos Knockout , Fator 2 Relacionado a NF-E2/genética , Elementos de Resposta/efeitos dos fármacos , Elementos de Resposta/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa
19.
Science ; 319(5863): 624-7, 2008 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-18239127

RESUMO

Cathepsin K was originally identified as an osteoclast-specific lysosomal protease, the inhibitor of which has been considered might have therapeutic potential. We show that inhibition of cathepsin K could potently suppress autoimmune inflammation of the joints as well as osteoclastic bone resorption in autoimmune arthritis. Furthermore, cathepsin K-/- mice were resistant to experimental autoimmune encephalomyelitis. Pharmacological inhibition or targeted disruption of cathepsin K resulted in defective Toll-like receptor 9 signaling in dendritic cells in response to unmethylated CpG DNA, which in turn led to attenuated induction of T helper 17 cells, without affecting the antigen-presenting ability of dendritic cells. These results suggest that cathepsin K plays an important role in the immune system and may serve as a valid therapeutic target in autoimmune diseases.


Assuntos
Artrite Experimental/imunologia , Artrite Experimental/metabolismo , Doenças Autoimunes/metabolismo , Catepsinas/metabolismo , Transdução de Sinais , Receptor Toll-Like 9/metabolismo , Animais , Antígenos CD/metabolismo , Artrite Experimental/tratamento farmacológico , Doenças Autoimunes/tratamento farmacológico , Doenças Autoimunes/imunologia , Reabsorção Óssea , Catepsina K , Catepsinas/antagonistas & inibidores , Catepsinas/deficiência , Citocinas/metabolismo , DNA/imunologia , DNA/metabolismo , Células Dendríticas/efeitos dos fármacos , Células Dendríticas/imunologia , Fosfatos de Dinucleosídeos/imunologia , Fosfatos de Dinucleosídeos/metabolismo , Encefalomielite Autoimune Experimental/imunologia , Encefalomielite Autoimune Experimental/metabolismo , Endossomos/metabolismo , Adjuvante de Freund/imunologia , Ativação Linfocitária/efeitos dos fármacos , Masculino , Camundongos , Osteoporose/tratamento farmacológico , Inibidores de Proteases/farmacologia , Ratos , Linfócitos T/efeitos dos fármacos , Linfócitos T/enzimologia , Linfócitos T/imunologia
20.
J Biol Chem ; 282(21): 15862-71, 2007 May 25.
Artigo em Inglês | MEDLINE | ID: mdl-17374603

RESUMO

GATA-1 is the key transcription factor for the development of the erythroid, megakaryocytic, eosinophilic, and mast cell lineages. GATA-1 possesses the ability to self-associate, and this characteristic has been suggested to be important for GATA-1 function. To elucidate the roles self-associated GATA-1 plays during hematopoietic cell development in vivo, in this study we prepared GATA-1 mutants in which three lysine residues potentially contributing to the self-association (Lys-245, Lys-246, and Lys-312) are substituted in combination with alanines. Of the mutants, 3KA harboring alanine substitutions in all three lysines showed reduced self-association activity without considerable interference in the modification of GATA-1 by acetylation. We generated transgenic mouse lines that express these GATA-1 mutants utilizing the Gata1 hematopoietic regulatory domain, and crossed the mice to Gata1 knockdown (GATA-1.05) mutant mice. Although NKA (K245A and K246A) and CKA (K312A) mutants almost fully rescued the GATA-1.05 mice from anemia and embryonic lethality, the 3KA mutant only partially rescued the GATA-1.05 mutant mice. Even with the higher than endogenous level expression, GATA-1.05/Y::3KA embryos were prone to die at various stages in mid-to-late gestation. Live birth and an anemic phenotype were restored in some embryos depending on the expression level of the 3KA transgene. The expression of the transferrin receptor and heme biosynthesis enzymes was impaired in the yolk sac and liver of the 3KA-rescued embryos. Immature erythroid cells with insufficient expression of the transferrin receptor accumulated in the livers of 3KA-rescued embryos. These results provide the first convincing line of evidence that the self-association of GATA-1 is important for proper mammalian erythroid development in vivo.


Assuntos
Células Precursoras Eritroides/metabolismo , Eritropoese/fisiologia , Fator de Transcrição GATA1/metabolismo , Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Processamento de Proteína Pós-Traducional/fisiologia , Acetilação , Substituição de Aminoácidos , Anemia/genética , Anemia/metabolismo , Animais , Cruzamentos Genéticos , Perda do Embrião/genética , Perda do Embrião/metabolismo , Eosinófilos/citologia , Eosinófilos/metabolismo , Células Precursoras Eritroides/citologia , Fator de Transcrição GATA1/genética , Heme/biossíntese , Fígado/citologia , Fígado/embriologia , Mastócitos/citologia , Mastócitos/metabolismo , Camundongos , Camundongos Transgênicos , Mutação de Sentido Incorreto , Ligação Proteica/genética , Estrutura Terciária de Proteína/genética , Receptores da Transferrina/biossíntese , Saco Vitelino/citologia , Saco Vitelino/embriologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA