Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 32
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
MicroPubl Biol ; 20222022.
Artigo em Inglês | MEDLINE | ID: mdl-36320616

RESUMO

Opatic atrophy 1 (Opa1) is a mitochondrial GTPase that regulates mitochondrial fusion and maintenance of cristae architecture. Osteoclasts are mitochondrial rich-cells. However, the role of Opa1 in osteoclasts remains unclear. Here, we demonstrate that Opa1- deficient osteoclast precursor cells do not undergo efficient osteoclast differentiation and exhibit abnormal cristae morphology. Thus, Opa1 is a key factor in osteoclast differentiation through regulation of mitochondrial dynamics.

2.
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
3.
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
4.
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
5.
EMBO Rep ; 22(12): e53035, 2021 12 06.
Artigo em Inglês | MEDLINE | ID: mdl-34661337

RESUMO

Oxygen plays an important role in diverse biological processes. However, since quantitation of the partial pressure of cellular oxygen in vivo is challenging, the extent of oxygen perturbation in situ and its cellular response remains underexplored. Using two-photon phosphorescence lifetime imaging microscopy, we determine the physiological range of oxygen tension in osteoclasts of live mice. We find that oxygen tension ranges from 17.4 to 36.4 mmHg, under hypoxic and normoxic conditions, respectively. Physiological normoxia thus corresponds to 5% and hypoxia to 2% oxygen in osteoclasts. Hypoxia in this range severely limits osteoclastogenesis, independent of energy metabolism and hypoxia-inducible factor activity. We observe that hypoxia decreases ten-eleven translocation (TET) activity. Tet2/3 cooperatively induces Prdm1 expression via oxygen-dependent DNA demethylation, which in turn activates NFATc1 required for osteoclastogenesis. Taken together, our results reveal that TET enzymes, acting as functional oxygen sensors, regulate osteoclastogenesis within the physiological range of oxygen tension, thus opening new avenues for research on in vivo response to oxygen perturbation.


Assuntos
Desmetilação do DNA , Osteoclastos , Animais , Diferenciação Celular/genética , Hipóxia Celular , Hipóxia/metabolismo , Camundongos , Osteoclastos/metabolismo , Oxigênio/metabolismo
6.
Nat Commun ; 12(1): 2136, 2021 04 09.
Artigo em Inglês | MEDLINE | ID: mdl-33837198

RESUMO

Osteoclastic bone resorption and osteoblastic bone formation/replenishment are closely coupled in bone metabolism. Anabolic parathyroid hormone (PTH), which is commonly used for treating osteoporosis, shifts the balance from osteoclastic to osteoblastic, although it is unclear how these cells are coordinately regulated by PTH. Here, we identify a serine protease inhibitor, secretory leukocyte protease inhibitor (SLPI), as a critical mediator that is involved in the PTH-mediated shift to the osteoblastic phase. Slpi is highly upregulated in osteoblasts by PTH, while genetic ablation of Slpi severely impairs PTH-induced bone formation. Slpi induction in osteoblasts enhances its differentiation, and increases osteoblast-osteoclast contact, thereby suppressing osteoclastic function. Intravital bone imaging reveals that the PTH-mediated association between osteoblasts and osteoclasts is disrupted in the absence of SLPI. Collectively, these results demonstrate that SLPI regulates the communication between osteoblasts and osteoclasts to promote PTH-induced bone anabolism.


Assuntos
Reabsorção Óssea/tratamento farmacológico , Osteogênese/fisiologia , Hormônio Paratireóideo/administração & dosagem , Inibidor Secretado de Peptidases Leucocitárias/metabolismo , Animais , Reabsorção Óssea/patologia , Diferenciação Celular/efeitos dos fármacos , Diferenciação Celular/fisiologia , Linhagem Celular , Modelos Animais de Doenças , Feminino , Fêmur/citologia , Fêmur/diagnóstico por imagem , Fêmur/efeitos dos fármacos , Fêmur/patologia , Humanos , Masculino , Camundongos , Camundongos Knockout , Osteoblastos/efeitos dos fármacos , Osteoblastos/metabolismo , Osteoclastos/efeitos dos fármacos , Osteoclastos/metabolismo , Osteogênese/efeitos dos fármacos , Cultura Primária de Células , RNA-Seq , Inibidor Secretado de Peptidases Leucocitárias/genética , Regulação para Cima/efeitos dos fármacos , Microtomografia por Raio-X
7.
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
9.
Commun Biol ; 3(1): 496, 2020 09 08.
Artigo em Inglês | MEDLINE | ID: mdl-32901092

RESUMO

Space flight produces an extreme environment with unique stressors, but little is known about how our body responds to these stresses. While there are many intractable limitations for in-flight space research, some can be overcome by utilizing gene knockout-disease model mice. Here, we report how deletion of Nrf2, a master regulator of stress defense pathways, affects the health of mice transported for a stay in the International Space Station (ISS). After 31 days in the ISS, all flight mice returned safely to Earth. Transcriptome and metabolome analyses revealed that the stresses of space travel evoked ageing-like changes of plasma metabolites and activated the Nrf2 signaling pathway. Especially, Nrf2 was found to be important for maintaining homeostasis of white adipose tissues. This study opens approaches for future space research utilizing murine gene knockout-disease models, and provides insights into mitigating space-induced stresses that limit the further exploration of space by humans.


Assuntos
Fator 2 Relacionado a NF-E2/metabolismo , Voo Espacial , Aumento de Peso , Gordura Abdominal/patologia , Tecido Adiposo Branco/patologia , Envelhecimento/sangue , Envelhecimento/metabolismo , Animais , Osso e Ossos/patologia , Regulação da Expressão Gênica , Homeostase , Metaboloma , Camundongos Knockout , Músculos/patologia , Fator 2 Relacionado a NF-E2/deficiência , Fator 2 Relacionado a NF-E2/genética , Análise de Sequência de RNA , Estresse Fisiológico , Aumento de Peso/genética
10.
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
11.
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
12.
Clin Calcium ; 28(2): 201-210, 2018.
Artigo em Japonês | MEDLINE | ID: mdl-29371485

RESUMO

Fluorescence imaging technology using two-photon excitation microscopy has been developed and utilized to observe cell dynamics in various developmental processes and pathological conditions in vivo. This technology is absolutely dependent on the fluorescent labelling technique of specific cells in a living state in vivo using various methods such as genetic engineering, chemiluminescent probes or fluorescent-conjugated antibodies. In this article, we demonstrate the methods of genetic engineering, particularly how to generate a genetically modified mouse(reporter mouse)that expresses fluorescent protein endogenously in the specific cells. In consideration of mouse genetic engineering technologies and the current state of bioresources, we describe the transgenic method, the knock-in method, the Cre/loxP-mediated recombination method and the genomic editing method by CRISPR/Cas9 system that have been used widely for generation of reporter mice. Among these methods, it is important to carefully select the suitable method according to the research purpose. We would like to compare the methods comprehensively.


Assuntos
Osso e Ossos/química , Osso e Ossos/citologia , Animais , Sobrevivência Celular , Proteínas Luminescentes/análise , Camundongos , Camundongos Transgênicos
13.
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
14.
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
15.
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
16.
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
17.
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
18.
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
20.
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
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...