Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 28
Filtrar
1.
PLoS Genet ; 16(5): e1008361, 2020 05.
Artigo em Inglês | MEDLINE | ID: mdl-32463812

RESUMO

Osteocalcin (OCN), the most abundant noncollagenous protein in the bone matrix, is reported to be a bone-derived endocrine hormone with wide-ranging effects on many aspects of physiology, including glucose metabolism and male fertility. Many of these observations were made using an OCN-deficient mouse allele (Osc-) in which the 2 OCN-encoding genes in mice, Bglap and Bglap2, were deleted in ES cells by homologous recombination. Here we describe mice with a new Bglap and Bglap2 double-knockout (dko) allele (Bglap/2p.Pro25fs17Ter) that was generated by CRISPR/Cas9-mediated gene editing. Mice homozygous for this new allele do not express full-length Bglap or Bglap2 mRNA and have no immunodetectable OCN in their serum. FTIR imaging of cortical bone in these homozygous knockout animals finds alterations in the collagen maturity and carbonate to phosphate ratio in the cortical bone, compared with wild-type littermates. However, µCT and 3-point bending tests do not find differences from wild-type littermates with respect to bone mass and strength. In contrast to the previously reported OCN-deficient mice with the Osc-allele, serum glucose levels and male fertility in the OCN-deficient mice with the Bglap/2pPro25fs17Ter allele did not have significant differences from wild-type littermates. We cannot explain the absence of endocrine effects in mice with this new knockout allele. Possible explanations include the effects of each mutated allele on the transcription of neighboring genes, or differences in genetic background and environment. So that our findings can be confirmed and extended by other interested investigators, we are donating this new Bglap and Bglap2 double-knockout strain to the Jackson Laboratories for academic distribution.


Assuntos
Sistema Endócrino/fisiologia , Osteocalcina/genética , Animais , Densidade Óssea/genética , Osso e Ossos/metabolismo , Sistema Endócrino/metabolismo , Feminino , Fertilidade/genética , Resistência à Insulina/genética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Osteocalcina/deficiência
2.
Anal Chem ; 93(5): 2811-2819, 2021 02 09.
Artigo em Inglês | MEDLINE | ID: mdl-33475355

RESUMO

Bacterial riboswitch RNAs are attractive targets for novel antibiotics against antibiotic-resistant superbacteria. Their binding to cognate metabolites is essential for the regulation of bacterial gene expression. Despite the importance of RNAs as therapeutic targets, the development of RNA-targeted, small-molecule drugs is limited by current biophysical methods. Here, we monitored the specific interaction between the adenine-sensing riboswitch aptamer domain (ARS) and adenine at the single-molecule level using α-hemolysin (αHL) nanopores. During adenine-induced tertiary folding, adenine-bound ARS intermediates exhibited characteristic nanopore events, including a two-level ionic current blockade and a ∼ 5.6-fold longer dwell time than that of free RNA. In a proof-of-concept experiment, tertiary RNA folding-targeted drug screening was performed using a protein nanopore, which resulted in the discovery of three new ARS-targeting hit compounds from a natural compound library. Taken together, these results reveal that αHL nanopores are a valuable platform for ultrasensitive, label-free, and single-molecule-based drug screening against therapeutic RNA targets.


Assuntos
Nanoporos , Riboswitch , Avaliação Pré-Clínica de Medicamentos , Proteínas Hemolisinas , Dobramento de RNA
3.
J Cell Physiol ; 235(12): 9785-9794, 2020 12.
Artigo em Inglês | MEDLINE | ID: mdl-32529635

RESUMO

Skeletal homeostasis is sensitive to perturbations in Wnt signaling. Beyond its role in the bone, Wnt is a major target for pharmaceutical inhibition in a wide range of diseases, most notably cancers. Numerous clinical trials for Wnt-based candidates are currently underway, and Wnt inhibitors will likely soon be approved for clinical use. Given the bone-suppressive effects accompanying Wnt inhibition, there is a need to expose alternate pathways/molecules that can be targeted to counter the deleterious effects of Wnt inhibition on bone properties. Activation of the Pi3k/Akt pathway via Pten deletion is one possible osteoanabolic pathway to exploit. We investigated whether the osteopenic effects of ß-catenin deletion from bone cells could be rescued by Pten deletion in the same cells. Mice carrying floxed alleles for Pten and ß-catenin were bred to Dmp1-Cre mice to delete Pten alone, ß-catenin alone, or both genes from the late-stage osteoblast/osteocyte population. The mice were assessed for bone mass, density, strength, and formation parameters to evaluate the potential rescue effect of Pten deletion in Wnt-impaired mice. Pten deletion resulted in high bone mass and ß-catenin deletion resulted in low bone mass. Compound mutants had bone properties similar to ß-catenin mutant mice, or surprisingly in some assays, were further compromised beyond ß-catenin mutants. Pten inhibition, or one of its downstream nodes, is unlikely to protect against the bone-wasting effects of Wnt/ßcat inhibition. Other avenues for preserving bone mass in the presence of Wnt inhibition should be explored to alleviate the skeletal side effects of Wnt inhibitor-based therapies.


Assuntos
Proteínas da Matriz Extracelular/genética , Neoplasias/tratamento farmacológico , PTEN Fosfo-Hidrolase/genética , beta Catenina/genética , Animais , Doenças Ósseas Metabólicas/tratamento farmacológico , Doenças Ósseas Metabólicas/genética , Proliferação de Células/genética , Modelos Animais de Doenças , Humanos , Camundongos , Neoplasias/genética , Osteoblastos/efeitos dos fármacos , Osteogênese/efeitos dos fármacos , Osteogênese/genética , Fosfatidilinositol 3-Quinases/genética , Proteínas Proto-Oncogênicas c-akt/genética , Via de Sinalização Wnt/efeitos dos fármacos
4.
Nucleic Acids Res ; 44(17): 8407-16, 2016 09 30.
Artigo em Inglês | MEDLINE | ID: mdl-27288441

RESUMO

Retinoic acid-inducible gene I (RIG-I) recognizes specific molecular patterns of viral RNAs for inducing type I interferon. The C-terminal domain (CTD) of RIG-I binds to double-stranded RNA (dsRNA) with the 5'-triphosphate (5'-PPP), which induces a conformational change in RIG-I to an active form. It has been suggested that RIG-I detects infection of influenza A virus by recognizing the 5'-triphosphorylated panhandle structure of the viral RNA genome. Influenza panhandle RNA has a unique structure with a sharp helical bending. In spite of extensive studies of how viral RNAs activate RIG-I, whether the structural elements of the influenza panhandle RNA confer the ability to activate RIG-I signaling has been poorly explored. Here, we investigated the dynamics of the influenza panhandle RNA in complex with RIG-I CTD using NMR spectroscopy and showed that the bending structure of the panhandle RNA negates the requirement of a 5'-PPP moiety for RIG-I activation.


Assuntos
Proteína DEAD-box 58/metabolismo , Vírus da Influenza A/genética , Conformação de Ácido Nucleico , Polifosfatos/metabolismo , RNA Viral/química , Pareamento de Bases , Calorimetria , Proteína DEAD-box 58/química , Humanos , Hidrogênio/metabolismo , Interferon beta/metabolismo , Espectroscopia de Ressonância Magnética , Modelos Biológicos , Ligação Proteica , Domínios Proteicos , Estabilidade de RNA , RNA Viral/metabolismo , Reação em Cadeia da Polimerase em Tempo Real , Receptores Imunológicos , Termodinâmica
6.
Molecules ; 23(10)2018 Oct 22.
Artigo em Inglês | MEDLINE | ID: mdl-30360415

RESUMO

Eukaryotic transcription initiation is mediated by interactions between transcriptional activators and the mediator coactivator complex. Molecular interaction of p53 transcription factor with mediator complex subunit 25 (MED25) is essential for its target gene transcription. In this study, we characterized the molecular interaction between p53 transactivation domain (p53TAD) and activator interaction domain (ACID) of MED25 using nuclear magnetic resonance (NMR) spectroscopy. The NMR chemical shift perturbation and isothermal titration calorimetry (ITC) data showed that p53TAD interacted with MED25 ACID mainly through the p53TAD2 sequence motif. Taken together with the mutagenesis data, the refined structural model of MED25 ACID/p53TAD2 peptide complex showed that an amphipathic α-helix of p53TAD2 peptide bound an elongated hydrophobic groove of MED25 ACID. Furthermore, our results revealed the highly conserved mechanism of MED25 interaction with intrinsically unfolded acidic TADs from the transcriptional activators p53, ERM (Ets-related molecule), and herpes simplex virus protein 16 (VP16).


Assuntos
Complexo Mediador/química , Complexo Mediador/metabolismo , Domínios Proteicos , Domínios e Motivos de Interação entre Proteínas , Proteína Supressora de Tumor p53/química , Proteína Supressora de Tumor p53/metabolismo , Sítios de Ligação , Sequência Conservada , Humanos , Espectroscopia de Ressonância Magnética , Complexo Mediador/genética , Modelos Moleculares , Mutação , Peptídeos/química , Peptídeos/metabolismo , Ligação Proteica , Conformação Proteica , Relação Estrutura-Atividade , Proteína Supressora de Tumor p53/genética
7.
Calcif Tissue Int ; 100(4): 361-373, 2017 04.
Artigo em Inglês | MEDLINE | ID: mdl-28013361

RESUMO

Recently, we demonstrated that osteoblast-specific overexpression of human WNT16 increased both cortical and trabecular bone mass and structure in mice. To further identify the cell-specific role of Wnt16 in bone homeostasis, we created transgenic (TG) mice overexpressing human WNT16 in osteocytes using Dmp1 promoter (Dmp1-hWNT16 TG) on C57BL/6 (B6) background. We analyzed bone phenotypes and serum bone biomarkers, performed gene expression analysis and measured dynamic bone histomorphometry in Dmp1-hWNT16 TG and wild-type (WT) mice. Compared to WT mice, Dmp1-hWNT16 TG mice exhibited significantly higher whole-body, spine and femoral aBMD, BMC and trabecular (BV/TV, Tb.N, and Tb.Th) and cortical (bone area and thickness) parameters in both male and female at 12 weeks of age. Femur stiffness and ultimate force were also significantly improved in the Dmp1-hWNT16 TG female mice, compared to sex-matched WT littermates. In addition, female Dmp1-hWNT16 TG mice displayed significantly higher MS/BS, MAR and BFR/BS compared to the WT mice. Gene expression analysis demonstrated significantly higher mRNA level of Alp in both male and female Dmp1-hWNT16 TG mice and significantly higher levels of Osteocalcin, Opg and Rankl in the male Dmp1-hWNT16 TG mice in bone tissue compared to sex-matched WT mice. These results indicate that WNT16 plays a critical role for acquisition of both cortical and trabecular bone mass and strength. Strategies designed to use WNT16 as a target for therapeutic interventions will be valuable to treat osteoporosis and other low bone mass conditions.


Assuntos
Densidade Óssea/fisiologia , Osteócitos/metabolismo , Proteínas Wnt/metabolismo , Animais , Densidade Óssea/genética , Osso e Ossos/metabolismo , Feminino , Fêmur/metabolismo , Fêmur/patologia , Humanos , Masculino , Camundongos , Camundongos Transgênicos , Osteoblastos/metabolismo , Osteocalcina/metabolismo , Osteoporose/genética , Osteoporose/metabolismo , Proteínas Wnt/genética
8.
J Cell Physiol ; 231(1): 162-71, 2016 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-26058470

RESUMO

Core binding factor ß (Cbfß) is a partner protein of Runx family transcription factors with minimally characterized function in cartilage. Here we address the role of Cbfß in cartilage by generating chondrocyte-specific Cbfß-deficient mice (Cbfb(Δch/Δch) ) from Cbfb-floxed mice crossed with mice expressing Cre from the Col2a1 promoter. Cbfb(Δch/Δch) mice died soon after birth and exhibited delayed endochondral bone formation, shorter appendicular skeleton length with increased proliferative chondrocytes, and nearly absent hypertrophic chondrocyte zones. Immunohistochemical and quantitative real-time PCR analyses showed that the number and size of proliferative chondrocytes increased and the expression of chondrocyte maturation markers at the growth plates, including Runx2, osterix, and osteopontin, significantly diminished in Cbfb(Δch/Δch) mice compared to wild type mice. With regard to signaling pathways, both PTHrP-Ihh and BMP signaling were compromised in Cbfb(Δch/Δch) mice. Mechanistically, Cbfß deficiency in chondrocytes caused a decrease of protein levels of Runx transcription factors by accelerating polyubiquitination-mediated proteosomal degradation in vitro. Indeed, Runx2 and Runx3, but not Runx1, decreased in Cbfb(Δch/Δch) mice. Collectively, these findings indicate that Cbfß plays a critical role for chondrocyte differentiation through stabilizing Runx2 and Runx3 proteins in cartilage.


Assuntos
Diferenciação Celular/genética , Condrócitos/citologia , Condrogênese/genética , Subunidade beta de Fator de Ligação ao Core/metabolismo , Lâmina de Crescimento/metabolismo , Animais , Cartilagem/fisiologia , Subunidade beta de Fator de Ligação ao Core/genética , Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Osteogênese/fisiologia
9.
Bone ; 143: 115708, 2021 02.
Artigo em Inglês | MEDLINE | ID: mdl-33164872

RESUMO

The cysteine knot protein sclerostin is an osteocyte-derived secreted inhibitor of the Wnt co-receptors LRP5 and LRP6. LRP5 plays a dominant role in bone homeostasis, but we previously reported that Sost/sclerostin suppression significantly increased osteogenesis regardless of Lrp5 presence or absence. Those observations suggested that the bone forming effects of sclerostin inhibition can occur through Lrp6 (when Lrp5 is suppressed), or through other yet undiscovered mechanisms independent of Lrp5/6. To distinguish between these two possibilities, we generated mice with compound deletion of Lrp5 and Lrp6 selectively in bone, and treated them with sclerostin monoclonal antibody (Scl-mAb). All mice were homozygous flox for both Lrp5 and Lrp6 (Lrp5f/f; Lrp6f/f), and varied only in whether or not they carried the Dmp1-Cre transgene. Positive (Cre+) and negative (Cre-) mice were injected with Scl-mAb or vehicle from 4.5 to 14 weeks of age. Vehicle-treated Cre+ mice exhibited significantly reduced skeletal properties compared to vehicle-treated Cre- mice, as assessed by DXA, µCT, pQCT, and histology, indicating that Lrp5/6 deletions were effective and efficient. Scl-mAb treatment improved nearly every bone-related parameter among Cre- mice, but the same treatment in Cre+ mice resulted in little to no improvement in skeletal properties. For the few endpoints where Cre+ mice responded to Scl-mAb, it is likely that antibody-induced promotion of Wnt signaling occurred in cell types earlier in the mesenchymal/osteoblast differentiation pathway than the Dmp1-expressing stage. This latter conclusion was supported by changes in some histomorphometric parameters. In conclusion, unlike with the deletion of Lrp5 alone, the bone-selective late-stage co-deletion of Lrp5 and Lrp6 significantly impairs or completely nullifies the osteogenic action of Scl-mAb, and highlights a major role for both Lrp5 and Lrp6 in the mechanism of action for the bone-building effects of sclerostin antibody.


Assuntos
Glicoproteínas , Peptídeos e Proteínas de Sinalização Intercelular , Proteínas Adaptadoras de Transdução de Sinal , Animais , Osso e Ossos/metabolismo , Glicoproteínas/metabolismo , Proteína-5 Relacionada a Receptor de Lipoproteína de Baixa Densidade/genética , Proteína-5 Relacionada a Receptor de Lipoproteína de Baixa Densidade/metabolismo , Proteína-6 Relacionada a Receptor de Lipoproteína de Baixa Densidade/genética , Proteína-6 Relacionada a Receptor de Lipoproteína de Baixa Densidade/metabolismo , Camundongos , Osteoblastos/metabolismo
10.
J Cell Biochem ; 110(1): 97-103, 2010 May.
Artigo em Inglês | MEDLINE | ID: mdl-20225274

RESUMO

Cleidocranial dysplasia (CCD) is caused by haploinsufficiency in RUNX2 function. We have previously identified a series of RUNX2 mutations in Korean CCD patients, including a novel R131G missense mutation in the Runt-homology domain. Here, we examine the functional consequences of the RUNX2(R131G) mutation, which could potentially affect DNA binding, nuclear localization signal, and/or heterodimerization with core-binding factor-beta (CBF-beta). Immunofluorescence microscopy and western blot analysis with subcellular fractions show that RUNX2(R131G) is localized in the nucleus. Immunoprecipitation analysis reveals that heterodimerization with CBF-beta is retained. However, precipitation assays with biotinylated oligonucleotides and reporter gene assays with RUNX2 responsive promoters together reveal that DNA-binding activity and consequently the transactivation of potential of RUNX2(R131G) is abrogated. We conclude that loss of DNA binding, but not nuclear localization or CBF-beta heterodimerization, causes RUNX2 haploinsufficiency in patients with the RUNX2(R131G) mutation. Retention of specific functions including nuclear localization and binding to CBF-beta of the RUNX2(R131G) mutation may render the mutant protein an effective competitor that interferes with wild-type function.


Assuntos
Substituição de Aminoácidos/genética , Displasia Cleidocraniana/genética , Subunidade alfa 1 de Fator de Ligação ao Core/genética , Subunidade alfa 1 de Fator de Ligação ao Core/metabolismo , Subunidade beta de Fator de Ligação ao Core/metabolismo , DNA/metabolismo , Mutação/efeitos dos fármacos , Motivos de Aminoácidos , Animais , Células CHO , Núcleo Celular/metabolismo , Subunidade alfa 1 de Fator de Ligação ao Core/química , Subunidade alfa 2 de Fator de Ligação ao Core/química , Subunidade alfa 2 de Fator de Ligação ao Core/metabolismo , Subunidade beta de Fator de Ligação ao Core/química , Cricetinae , Cricetulus , Células HeLa , Humanos , Proteínas Mutantes/química , Proteínas Mutantes/metabolismo , Mutação/genética , Ligação Proteica , Multimerização Proteica , Estrutura Terciária de Proteína , Transporte Proteico , Ativação Transcricional/genética
11.
J Cell Biochem ; 109(6): 1148-57, 2010 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-20108254

RESUMO

Here, we show the involvement of signaling pathways to induce the gene expression of bone morphogenetic protein (BMP) in the osteogenic activity of physcion-8-O-beta-D-glucopyranoside (physcion-Glu); it stimulated osteoblast differentiation in mouse osteoblast MC3T3-E1 subclone 4 cells and induced BMP-2 gene expression and activation of Akt and ERK/MAP kinases. Physcion-Glu-induced BMP-2 expression and mineralization were attenuated by LY294002, an inhibitor of PI3K that lies upstream of Akt and MAP kinases, suggesting that physcion-Glu induces osteoblast differentiation via PI3K-Akt/MAP kinase signaling pathways, which play important roles in inducing BMP-2 gene expression. Physcion-Glu also enhanced BMP-2-induced commitment of mouse bi-potential mesenchymal precursor C2C12 cells into osteoblasts while inducing the transcription of several osteogenic BMP isoforms, such as BMP-2, -4, -7, and -9. Osteogenic synergy between BMP-2 and physcion-Glu was supported by the fact that noggin inhibited BMP-2 and physcion-Glu-induced alkaline phosphatase expression and activity. Considering that physcion-Glu induced Runx2 activity and the nuclear translocation of p-Smad, physcion-Glu could act by enhancing the BMP signaling pathway that induces Smad activation and translocation to activate Runx2. In conclusion, physcion-Glu could enhance the commitment of mesenchymal progenitors into osteoblasts and their differentiation by activating signaling pathways to induce BMP gene expression.


Assuntos
Proteína Morfogenética Óssea 2/metabolismo , Emodina/análogos & derivados , Glucosídeos/química , Glucosídeos/farmacologia , Células-Tronco Mesenquimais/citologia , Células-Tronco Mesenquimais/efeitos dos fármacos , Osteoblastos/citologia , Transdução de Sinais/efeitos dos fármacos , Animais , Western Blotting , Proteína Morfogenética Óssea 2/genética , Diferenciação Celular/efeitos dos fármacos , Linhagem Celular , Cromonas/farmacologia , Emodina/química , Camundongos , Morfolinas/farmacologia , Fosfatidilinositol 3-Quinases/metabolismo , Inibidores de Fosfoinositídeo-3 Quinase , Reação em Cadeia da Polimerase , Regiões Promotoras Genéticas/genética
12.
Bone ; 131: 115084, 2020 02.
Artigo em Inglês | MEDLINE | ID: mdl-31648079

RESUMO

Enhancing LRP5 signaling and inhibiting TGFß signaling have each been reported to increase bone mass and improve bone strength in wild-type mice. Monotherapy targeting LRP5 signaling, or TGFß signaling, also improved bone properties in mouse models of Osteogenesis Imperfecta (OI). We investigated whether additive or synergistic increases in bone properties would be attained if enhanced LRP5 signaling was combined with TGFß inhibition. We crossed an Lrp5 high bone mass (HBM) allele (Lrp5A214V) into the Col1a2G610C/+ mouse model of OI. At 6-weeks-of-age we began treating mice with an antibody that inhibits TGFß1, ß2, and ß3 (mAb 1D11), or with an isotype-matched control antibody (mAb 13C4). At 12-weeks-old, we observed that combining enhanced LRP5 signaling with inhibited TGFß signaling produced an additive effect on femoral and vertebral trabecular bone volumes, but not on cortical bone volumes. Although enhanced LRP5 signaling increased femur strength in a 3-point bending assay in Col1a2G610C/+ mice, femur strength did not improve further with TGFß inhibition. Neither enhanced LRP5 signaling nor TGFß inhibition, alone or in combination, improved femur 3-point-bending post-yield displacement in Col1a2G610C/+ mice. These pre-clinical studies indicate combination therapies that target LRP5 and TGFß signaling should increase trabecular bone mass in patients with OI more than targeting either signaling pathway alone. Whether additive increases in trabecular bone mass will occur in, and clinically benefit, patients with OI needs to be determined.


Assuntos
Osteogênese Imperfeita , Animais , Osso Esponjoso/diagnóstico por imagem , Colágeno Tipo I , Osso Cortical , Modelos Animais de Doenças , Humanos , Proteína-5 Relacionada a Receptor de Lipoproteína de Baixa Densidade/genética , Camundongos , Osteogênese Imperfeita/tratamento farmacológico , Osteogênese Imperfeita/genética , Transdução de Sinais , Fator de Crescimento Transformador beta
13.
Bone ; 120: 166-175, 2019 03.
Artigo em Inglês | MEDLINE | ID: mdl-30409757

RESUMO

High-bone-mass (HBM)-causing missense mutations in the low density lipoprotein receptor-related protein-5 (Lrp5) are associated with increased osteoanabolic action and protection from disuse- and ovariectomy-induced osteopenia. These mutations (e.g., A214V and G171V) confer resistance to endogenous secreted Lrp5/6 inhibitors, such as sclerostin (SOST) and Dickkopf homolog-1 (DKK1). Cells in the osteoblast lineage are responsive to canonical Wnt stimulation, but recent work has indicated that osteoclasts exhibit both indirect and direct responsiveness to canonical Wnt. Whether Lrp5-HBM receptors, expressed in osteoclasts, might alter osteoclast differentiation, activity, and consequent net bone balance in the skeleton, is not known. To address this, we bred mice harboring heterozygous Lrp5 HBM-causing conditional knock-in alleles to Ctsk-Cre transgenic mice and studied the phenotype using DXA, µCT, histomorphometry, serum assays, and primary cell culture. Mice with HBM alleles induced in Ctsk-expressing cells (TG) exhibited higher bone mass and architectural properties compared to non-transgenic (NTG) counterparts. In vivo and in vitro measurements of osteoclast activity, population density, and differentiation yielded significant reductions in osteoclast-related parameters in female but not male TG mice. Droplet digital PCR performed on osteocyte enriched cortical bone tubes from TG and NTG mice revealed that ~8-17% of the osteocyte population (depending on sex) underwent recombination of the conditional Lrp5 allele in the presence of Ctsk-Cre. Further, bone formation parameters in the midshaft femur cortex show a small but significant increase in anabolic action on the endocortical but not periosteal surface. These findings suggest that Wnt/Lrp5 signaling in osteoclasts affects osteoclastogenesis and activity in female mice, but also that some of the changes in bone mass in TG mice might be due to Cre expression in the osteocyte population.


Assuntos
Osso e Ossos/metabolismo , Catepsina K/metabolismo , Proteína-5 Relacionada a Receptor de Lipoproteína de Baixa Densidade/genética , Mutação/genética , Absorciometria de Fóton , Alelos , Animais , Biomarcadores/sangue , Células da Medula Óssea/metabolismo , Reabsorção Óssea/sangue , Reabsorção Óssea/patologia , Osso e Ossos/diagnóstico por imagem , Diferenciação Celular , Feminino , Integrases/metabolismo , Masculino , Camundongos Transgênicos , Tamanho do Órgão/genética , Osteoclastos/metabolismo , Osteoclastos/patologia , Osteogênese/genética , Periósteo/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Recombinação Genética/genética , Transgenes , Microtomografia por Raio-X
14.
FEBS Lett ; 592(20): 3367-3379, 2018 10.
Artigo em Inglês | MEDLINE | ID: mdl-30207377

RESUMO

In-cell NMR spectroscopy has emerged as a powerful technique for monitoring biomolecular interactions at an atomic level inside intact cells. However, current methodologies are inadequate at charting intracellular interactions of nonlabeled proteins and require their prior isotopic labeling. Herein, we describe for the first time the monitoring of the quercetin-alanine bioconjugate interaction with the nonlabeled antiapoptotic protein Bcl-2 inside living human cancer cells. STD and Tr-NOESY in-cell NMR methodologies were successfully applied in the investigation of the binding, which was further validated in vitro. In-cell NMR proved a very promising strategy for the real-time probing of the interaction profile of potential drugs with their therapeutic targets in native cellular environments and could, thus, open a new avenue in drug discovery.


Assuntos
Alanina/metabolismo , Espectroscopia de Ressonância Magnética/métodos , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Quercetina/metabolismo , Alanina/química , Humanos , Células Jurkat , Modelos Moleculares , Conformação Molecular , Ligação Proteica , Domínios Proteicos , Proteínas Proto-Oncogênicas c-bcl-2/química , Proteínas Proto-Oncogênicas c-bcl-2/genética , Quercetina/química
15.
JCI Insight ; 3(11)2018 06 07.
Artigo em Inglês | MEDLINE | ID: mdl-29875318

RESUMO

The WNT pathway has become an attractive target for skeletal therapies. High-bone-mass phenotypes in patients with loss-of-function mutations in the LRP5/6 inhibitor Sost (sclerosteosis), or in its downstream enhancer region (van Buchem disease), highlight the utility of targeting Sost/sclerostin to improve bone properties. Sclerostin-neutralizing antibody is highly osteoanabolic in animal models and in human clinical trials, but antibody-based inhibition of another potent LRP5/6 antagonist, Dkk1, is largely inefficacious for building bone in the unperturbed adult skeleton. Here, we show that conditional deletion of Dkk1 from bone also has negligible effects on bone mass. Dkk1 inhibition increases Sost expression, suggesting a potential compensatory mechanism that might explain why Dkk1 suppression lacks anabolic action. To test this concept, we deleted Sost from osteocytes in, or administered sclerostin neutralizing antibody to, mice with a Dkk1-deficient skeleton. A robust anabolic response to Dkk1 deletion was manifest only when Sost/sclerostin was impaired. Whole-body DXA scans, µCT measurements of the femur and spine, histomorphometric measures of femoral bone formation rates, and biomechanical properties of whole bones confirmed the anabolic potential of Dkk1 inhibition in the absence of sclerostin. Further, combined administration of sclerostin and Dkk1 antibody in WT mice produced a synergistic effect on bone gain that greatly exceeded individual or additive effects of the therapies, confirming the therapeutic potential of inhibiting multiple WNT antagonists for skeletal health. In conclusion, the osteoanabolic effects of Dkk1 inhibition can be realized if sclerostin upregulation is prevented. Anabolic therapies for patients with low bone mass might benefit from a strategy that accounts for the compensatory milieu of WNT inhibitors in bone tissue.


Assuntos
Anabolizantes/administração & dosagem , Glicoproteínas/antagonistas & inibidores , Hiperostose/tratamento farmacológico , Osteogênese/efeitos dos fármacos , Sindactilia/tratamento farmacológico , Via de Sinalização Wnt/efeitos dos fármacos , Proteínas Adaptadoras de Transdução de Sinal , Animais , Anticorpos Neutralizantes/administração & dosagem , Proteínas Morfogenéticas Ósseas/genética , Modelos Animais de Doenças , Feminino , Fêmur/citologia , Fêmur/diagnóstico por imagem , Fêmur/patologia , Marcadores Genéticos/genética , Glicoproteínas/genética , Glicoproteínas/metabolismo , Humanos , Hiperostose/diagnóstico por imagem , Hiperostose/genética , Hiperostose/patologia , Peptídeos e Proteínas de Sinalização Intercelular/genética , Mutação com Perda de Função , Masculino , Camundongos , Osteócitos , Coluna Vertebral/citologia , Coluna Vertebral/diagnóstico por imagem , Coluna Vertebral/patologia , Sindactilia/diagnóstico por imagem , Sindactilia/genética , Sindactilia/patologia , Resultado do Tratamento , Regulação para Cima/efeitos dos fármacos , Microtomografia por Raio-X
16.
Bone ; 90: 127-32, 2016 09.
Artigo em Inglês | MEDLINE | ID: mdl-27297606

RESUMO

Osteogenesis Imperfecta (OI) comprises a group of genetic skeletal fragility disorders. The mildest form of OI, Osteogenesis Imperfecta type I, is frequently caused by haploinsufficiency mutations in COL1A1, the gene encoding the α1(I) chain of type 1 collagen. Children with OI type I have a 95-fold higher fracture rate compared to unaffected children. Therapies for OI type I in the pediatric population are limited to anti-catabolic agents. In adults with osteoporosis, anabolic therapies that enhance Wnt signaling in bone improve bone mass, and ongoing clinical trials are determining if these therapies also reduce fracture risk. We performed a proof-of-principle experiment in mice to determine whether enhancing Wnt signaling in bone could benefit children with OI type I. We crossed a mouse model of OI type I (Col1a1(+/Mov13)) with a high bone mass (HBM) mouse (Lrp5(+/p.A214V)) that has increased bone strength from enhanced Wnt signaling. Offspring that inherited the OI and HBM alleles had higher bone mass and strength than mice that inherited the OI allele alone. However, OI+HBM and OI mice still had bones with lower ductility compared to wild-type mice. We conclude that enhancing Wnt signaling does not make OI bone normal, but does improve bone properties that could reduce fracture risk. Therefore, agents that enhance Wnt signaling are likely to benefit children and adults with OI type 1.


Assuntos
Osso e Ossos/patologia , Osso e Ossos/fisiopatologia , Colágeno Tipo I/metabolismo , Osteogênese Imperfeita/patologia , Osteogênese Imperfeita/fisiopatologia , Via de Sinalização Wnt , Alelos , Animais , Fenômenos Biomecânicos , Densidade Óssea , Matriz Óssea/patologia , Osso e Ossos/diagnóstico por imagem , Osso e Ossos/metabolismo , Calcificação Fisiológica , Osso Esponjoso/diagnóstico por imagem , Osso Esponjoso/patologia , Osso Esponjoso/fisiopatologia , Colágeno Tipo I/genética , Cadeia alfa 1 do Colágeno Tipo I , Osso Cortical/diagnóstico por imagem , Osso Cortical/patologia , Osso Cortical/fisiopatologia , Modelos Animais de Doenças , Feminino , Fêmur/diagnóstico por imagem , Fêmur/patologia , Fêmur/fisiopatologia , Proteína-5 Relacionada a Receptor de Lipoproteína de Baixa Densidade/metabolismo , Vértebras Lombares/diagnóstico por imagem , Vértebras Lombares/patologia , Vértebras Lombares/fisiopatologia , Masculino , Camundongos Endogâmicos C57BL , Tamanho do Órgão , Microtomografia por Raio-X
17.
Endocrinology ; 157(8): 3047-57, 2016 08.
Artigo em Inglês | MEDLINE | ID: mdl-27253995

RESUMO

ß-Catenin (ßcat) is a major downstream signaling node in canonical Wingless-related integration site (Wnt) signaling pathway, and its activity is crucial for canonical Wnt signal transduction. Wnt signaling has recently been implicated in the osteo-anabolic response to PTH, a potent calcium-regulating factor. We investigated whether ßcat is essential for the anabolic action of intermittent PTH by generating male mice with adult-onset deletion of ßcat in a subpopulation of bone cells (osteocytes and late-stage osteoblasts), treating them with an anabolic regimen of PTH, and measuring the skeletal responses. Male (10kb)Dmp1-CreERt2 transgenic mice that also harbored floxed loss-of-function ßcat alleles (ßcat(f/f)) were induced for Cre activity using tamoxifen, then injected daily with human PTH 1-34 (30 µg/kg) or vehicle for 5 weeks. Mice in which ßcat was deleted showed either total lack of bone mineral density (BMD) gain, or BMD loss, and did not respond to PTH treatment. However, bone mass measurements in the trabecular compartment of the femur and spine revealed PTH-induced bone gain whether ßcat was deleted or not. PTH-stimulated increases in periosteal and cancellous bone formation rates were not impaired by ßcat deletion, but resorption markers and cortical porosity were significantly increased in induced mice, particularly induced mice treated with PTH. These results suggest that ßcat is required for net-positive BMD effects of PTH therapy but that the anabolic effects per se of PTH treatment might not require osteocytic/osteoblastic ßcat.


Assuntos
Proteínas da Matriz Extracelular/genética , Deleção de Genes , Osteogênese , Hormônio Paratireóideo/farmacologia , beta Catenina/genética , Fatores Etários , Anabolizantes/metabolismo , Anabolizantes/farmacologia , Animais , Densidade Óssea/efeitos dos fármacos , Densidade Óssea/genética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Mutagênese , Osteogênese/efeitos dos fármacos , Osteogênese/genética , Hormônio Paratireóideo/metabolismo
18.
Endocrinology ; 157(2): 722-36, 2016 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-26584014

RESUMO

Previous genome-wide association studies have identified common variants in genes associated with bone mineral density (BMD) and risk of fracture. Recently, we identified single nucleotide polymorphisms (SNPs) in Wingless-type mouse mammary tumor virus integration site (WNT)16 that were associated with peak BMD in premenopausal women. To further identify the role of Wnt16 in bone mass regulation, we created transgenic (TG) mice overexpressing human WNT16 in osteoblasts. We compared bone phenotypes, serum biochemistry, gene expression, and dynamic bone histomorphometry between TG and wild-type (WT) mice. Compared with WT mice, WNT16-TG mice exhibited significantly higher whole-body areal BMD and bone mineral content (BMC) at 6 and 12 weeks of age in both male and female. Microcomputer tomography analysis of trabecular bone at distal femur revealed 3-fold (male) and 14-fold (female) higher bone volume/tissue volume (BV/TV), and significantly higher trabecular number and trabecular thickness but lower trabecular separation in TG mice compared with WT littermates in both sexes. The cortical bone at femur midshaft also displayed significantly greater bone area/total area and cortical thickness in the TG mice in both sexes. Serum biochemistry analysis showed that male TG mice had higher serum alkaline phosphatase, osteocalcin, osteoprotegerin (OPG), OPG to receptor activator of NF-kB ligand (tumor necrosis family ligand superfamily, number 11; RANKL) ratio as compared with WT mice. Also, lower carboxy-terminal collagen cross-link (CTX) to tartrate-resistant acid phosphatase 5, isoform b (TRAPc5b) ratio was observed in TG mice compared with WT littermates in both male and female. Histomorphometry data demonstrated that both male and female TG mice had significantly higher cortical and trabecular mineralizing surface/bone surface and bone formation rate compared with sex-matched WT mice. Gene expression analysis demonstrated higher expression of Alp, OC, Opg, and Opg to Rankl ratio in bone tissue in the TG mice compared with WT littermates. Our data indicate that WNT16 is critical for positive regulation of both cortical and trabecular bone mass and structure and that this molecule might be targeted for therapeutic interventions to treat osteoporosis.


Assuntos
Densidade Óssea/genética , Fêmur/diagnóstico por imagem , Osteoblastos/metabolismo , Osteogênese/genética , RNA Mensageiro/metabolismo , Proteínas Wnt/genética , Fosfatase Ácida/genética , Fosfatase Ácida/metabolismo , Fosfatase Alcalina/genética , Fosfatase Alcalina/metabolismo , Animais , Osso e Ossos/diagnóstico por imagem , Colágeno Tipo I/genética , Colágeno Tipo I/metabolismo , Feminino , Fêmur/metabolismo , Humanos , Isoenzimas/genética , Isoenzimas/metabolismo , Masculino , Camundongos , Camundongos Transgênicos , Osteocalcina/genética , Osteocalcina/metabolismo , Osteoporose , Osteoprotegerina/genética , Osteoprotegerina/metabolismo , Peptídeos/genética , Peptídeos/metabolismo , Ligante RANK/genética , Ligante RANK/metabolismo , Reação em Cadeia da Polimerase em Tempo Real , Fosfatase Ácida Resistente a Tartarato , Via de Sinalização Wnt , Microtomografia por Raio-X
19.
FEBS Lett ; 589(20 Pt B): 3037-43, 2015 Oct 07.
Artigo em Inglês | MEDLINE | ID: mdl-26318859

RESUMO

FAAP20 (Fanconi anemia-associated protein 20) is a subunit of the Fanconi anemia (FA) core complex that repairs interstrand cross-links. To understand the molecular basis for the FA core complex-mediated recruitment of Rev1 to the DNA lesion, we characterized the interactions among FAAP20-UBZ4, Rev1-BRCT, and ubiquitin using NMR. We found that FAAP20-UBZ4 binds not only ubiquitin but also Rev1-BRCT. Mapping the protein-protein interactions showed that FAAP20-UBZ4 has distinct binding surfaces for ubiquitin and Rev1-BRCT. In addition, the chemical exchange patterns indicated that the interaction between FAAP20-UBZ4 and ubiquitin might enhance the binding affinity between FAAP20-UBZ4 and Rev1-BRCT. These results provide new insight into the Rev1 recognition mechanism by FAAP20.


Assuntos
Proteínas de Grupos de Complementação da Anemia de Fanconi/química , Proteínas Nucleares/química , Nucleotidiltransferases/química , Estrutura Terciária de Proteína , Sequência de Aminoácidos , Sítios de Ligação/genética , Fenômenos Biofísicos , Proteínas de Grupos de Complementação da Anemia de Fanconi/genética , Proteínas de Grupos de Complementação da Anemia de Fanconi/metabolismo , Humanos , Espectroscopia de Ressonância Magnética , Modelos Moleculares , Dados de Sequência Molecular , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Nucleotidiltransferases/genética , Nucleotidiltransferases/metabolismo , Ligação Proteica , Mapeamento de Interação de Proteínas , Homologia de Sequência de Aminoácidos , Propriedades de Superfície , Ubiquitina/química , Ubiquitina/metabolismo
20.
Neurosci Lett ; 602: 95-8, 2015 Aug 18.
Artigo em Inglês | MEDLINE | ID: mdl-26151587

RESUMO

Neural progenitor cells generate various types of neurons and glia in a tightly regulated manner. During primary neurogenesis, retinoic acid (RA) acts earlier than Notch signaling and regulates differentiation and proliferation by upregulating proneural and neurogenic genes in the neural plate. However, the relationship between Notch signaling and the retinoid pathway during late neurogenesis remains unclear. We investigated the role of Mindbomb (Mib)-mediated Notch signaling in the differentiation of neural progenitors during late neurogenesis by overexpressing Mib and administering RA to Tg[hsp70-Mib:EGFP]. The majority of cells in the p3 domain differentiated into GABAergic Kolmer-Agduhr (KA) cells in Tg[hsp70-mib:EGFP] embryos heat-shocked during late neurogenesis, whereas these phenotypes were suppressed by exogenous RA. Our observations suggest that Mib-mediated Notch signaling plays a critical role in the temporal differentiation of neural progenitors, and that the generation of late-born KA″ cells is regulated by the interplay between Mib and RA.


Assuntos
Neurônios/citologia , Receptores Notch/fisiologia , Medula Espinal/citologia , Tretinoína/fisiologia , Animais , Animais Geneticamente Modificados , Diferenciação Celular , Embrião não Mamífero , Neurônios GABAérgicos/citologia , Neurônios GABAérgicos/metabolismo , Proteínas de Choque Térmico HSP70/genética , Interneurônios/citologia , Interneurônios/metabolismo , Neurônios Motores/citologia , Neurônios Motores/metabolismo , Células-Tronco Neurais/citologia , Células-Tronco Neurais/metabolismo , Neurogênese , Neurônios/metabolismo , Regiões Promotoras Genéticas , Células Receptoras Sensoriais/citologia , Células Receptoras Sensoriais/metabolismo , Transdução de Sinais , Medula Espinal/embriologia , Medula Espinal/metabolismo , Tretinoína/farmacologia , Ubiquitina-Proteína Ligases/genética , Ubiquitina-Proteína Ligases/fisiologia , Peixe-Zebra , Proteínas de Peixe-Zebra/genética , Proteínas de Peixe-Zebra/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA