Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 65
Filtrar
1.
Stem Cell Res ; 79: 103493, 2024 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-39032428

RESUMO

Myelin basic protein (MBP) is a major component of the myelin sheaths of oligodendrocytes in the central nervous system and Schwann cells of the peripheral nervous system. Here we generated heterozygous fluorescent reporter of MBP gene in human induced pluripotent stem cells (hiPSCs). CRISPR/Cas9 genome editing technology was employed to knock in fused tdTomato fluorescent protein and EF1 alpha promoter-driven Bleomycin (Zeocin) resistance gene to the translational MBP C-terminal region. The resulting line, MBP-TEZ, showed tdTomato fluorescence upon oligodendrocyte differentiation. This reporter hiPSC line provides a precedential opportunity for monitoring human myelin formation and degeneration and purifying MBP-expressing cell lineages.


Assuntos
Células-Tronco Pluripotentes Induzidas , Humanos , Células-Tronco Pluripotentes Induzidas/metabolismo , Células-Tronco Pluripotentes Induzidas/citologia , Proteína Básica da Mielina/metabolismo , Proteína Básica da Mielina/genética , Bainha de Mielina/metabolismo , Linhagem Celular , Diferenciação Celular , Sistemas CRISPR-Cas , Genes Reporter , Proteínas Luminescentes/metabolismo , Proteínas Luminescentes/genética , Edição de Genes , Proteína Vermelha Fluorescente
2.
Front Cell Dev Biol ; 12: 1370723, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38989059

RESUMO

Juvenile nephronophthisis is an inherited renal ciliopathy with cystic kidney disease, renal fibrosis, and end-stage renal failure in children and young adults. Mutations in the NPHP1 gene encoding nephrocystin-1 protein have been identified as the most frequently responsible gene and cause the formation of cysts in the renal medulla. The molecular pathogenesis of juvenile nephronophthisis remains elusive, and no effective medicines to prevent end-stage renal failure exist even today. No human cellular models have been available yet. Here, we report a first disease model of juvenile nephronophthisis using patient-derived and gene-edited human induced pluripotent stem cells (hiPSCs) and kidney organoids derived from these hiPSCs. We established NPHP1-overexpressing hiPSCs from patient-derived hiPSCs and NPHP1-deficient hiPSCs from healthy donor hiPSCs. Comparing these series of hiPSCs, we found abnormalities in primary cilia associated with NPHP1 deficiency in hiPSCs. Kidney organoids generated from the hiPSCs lacking NPHP1 formed renal cysts frequently in suspension culture with constant rotation. This cyst formation in patient-derived kidney organoids was rescued by overexpression of NPHP1. Transcriptome analysis on these kidney organoids revealed that loss of NPHP1 caused lower expression of genes related to primary cilia in epithelial cells and higher expression of genes related to the cell cycle. These findings suggested the relationship between abnormality in primary cilia induced by NPHP1 loss and abnormal proliferative characteristics in the formation of renal cysts. These findings demonstrated that hiPSC-based systematic disease modeling of juvenile nephronophthisis contributed to elucidating the molecular pathogenesis and developing new therapies.

3.
Regen Ther ; 26: 71-79, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38828011

RESUMO

Background: Human dental pulp-derived stem cells (hDPSCs) have emerged as a promising source for adult stem cell-based regenerative medicine. Stage-specific embryonic antigen 3 (SSEA3) is a cell surface marker associated with Multilineage-differentiating stress-enduring (Muse) cells, a subpopulation of human bone marrow-derived stem cells (hBMSCs), known for their potent regenerative potential and safety profile. In this study, we investigated the influence of the prolonged culture period and the number of culture passages on the regenerative capacity of hDPSCs and explored the association between SSEA3 expression and their regenerative abilities. Methods: hDPSCs were isolated and cultured for up to 20 passages. Cell proliferation, migration, and osteogenic, adipogenic and neurogenic differentiation potential were assessed at passages 5, 10, and 20. Flow cytometry and immunofluorescence were employed to analyze SSEA3 expression. RNA sequencing (RNA-seq) was performed on SSEA3-positive and SSEA3-negative hDPSCs to identify differentially expressed genes and associated pathways. Results: Our findings demonstrated a progressive decline in hDPSCs proliferation and migration capacity with increasing passage number. Conversely, cell size exhibited a positive correlation with passage number. Early passage hDPSCs displayed superior osteogenic and adipogenic differentiation potential. Notably, SSEA3 expression exhibited a significant negative correlation with passage numbers, reflecting the observed decline in differentiation capacity. RNA-seq analysis revealed distinct transcriptional profiles between SSEA3-positive and SSEA3-negative hDPSCs. SSEA3-positive cells displayed upregulation of genes associated with ectodermal differentiation and downregulation of genes involved in cell adhesion. Conclusions: This study elucidates the impact of passaging on hDPSC behavior and suggests SSEA3 as a valuable biomarker for evaluating stemness and regenerative potential. SSEA3-positive hDPSCs, functionally analogous to Muse cells, represent a promising cell population for developing targeted regenerative therapies with potentially improved clinical outcomes.

4.
Biochem Biophys Res Commun ; 719: 150063, 2024 Jul 30.
Artigo em Inglês | MEDLINE | ID: mdl-38749090

RESUMO

Osteoclasts are multinucleated cells with bone resorption activity. Excessive osteoclast activity has been implicated in osteoporosis, rheumatoid arthritis, and bone destruction due to bone metastases from cancer, making osteoclasts essential target cells in bone and joint diseases. C-terminal domain nuclear envelope phosphatase 1 (Ctdnep1, formerly Dullard) is a negative regulator of transforming growth factor (TGF)-ß superfamily signaling and regulates endochondral ossification in mesenchymal cells during skeletal development. In this study, we investigated the role of Ctdnep1 in the Receptor activator of nuclear factor-kappa B ligand (RANKL)-induced RAW264.7 osteoclast differentiation. Expression of Ctdnep1 did not change during osteoclast differentiation; Ctdnep1 protein localized to the cytoplasm before and after osteoclast differentiation. Small interfering RNA-mediated knockdown of Ctdnep1 increased tartrate-resistant acid phosphatase-positive multinucleated osteoclasts and the expression of osteoclast marker genes, including Acp5, Ctsk, and Nfatc1. Interestingly, the knockdown of Ctdnep1 increased the protein level of Nfatc1 in cells unstimulated with RANKL. Knockdown of Ctdnep1 also enhanced calcium-resorbing activity. Mechanistically, the knockdown of Ctdnep1 increased the phosphorylation of RANKL signaling components. These results suggest that Ctdnep1 negatively regulates osteoclast differentiation by suppressing the RANKL signaling pathway.


Assuntos
Diferenciação Celular , Osteoclastos , Ligante RANK , Animais , Camundongos , Técnicas de Silenciamento de Genes , Fatores de Transcrição NFATC/metabolismo , Fatores de Transcrição NFATC/genética , Osteoclastos/metabolismo , Osteoclastos/citologia , Ligante RANK/metabolismo , Células RAW 264.7 , Fosfoproteínas Fosfatases/genética , Fosfoproteínas Fosfatases/metabolismo
5.
J Cell Physiol ; 239(8): e31297, 2024 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-38769895

RESUMO

Teriparatide is a peptide derived from a parathyroid hormone (PTH) and an osteoporosis therapeutic drug with potent bone formation-promoting activity. To identify novel druggable genes that act downstream of PTH signaling and are potentially involved in bone formation, we screened PTH target genes in mouse osteoblast-like MC3T3-E1 cells. Here we show that Gprc5a, encoding an orphan G protein-coupled receptor, is a novel PTH-inducible gene and negatively regulates osteoblast proliferation and differentiation. PTH treatment induced Gprc5a expression in MC3T3-E1 cells, rat osteosarcoma ROS17/2.8 cells, and mouse femurs. Induction of Gprc5a expression by PTH occurred in the absence of protein synthesis and was mediated primarily via the cAMP pathway, suggesting that Gprc5a is a direct target of PTH signaling. Interestingly, Gprc5a expression was induced additively by co-treatment with PTH and 1α, 25-dihydroxyvitamin D3 (calcitriol), or retinoic acid in MC3T3-E1 cells. Reporter analysis of a 1 kb fragment of human GPRC5A promoter revealed that the promoter fragment showed responsiveness to PTH via the cAMP response element, suggesting that GPRC5A is also a PTH-inducible gene in humans. Gprc5a knockdown promoted cell viability and proliferation, as demonstrated by MTT and BrdU assays. Gprc5a knockdown also promoted osteoblast differentiation, as indicated by gene expression analysis and mineralization assay. Mechanistic studies showed that Gprc5a interacted with BMPR1A and suppressed BMP signaling induced by BMP-2 and constitutively active BMP receptors, ALK2 (ACVR1) Q207D and ALK3 (BMPR1A) Q233D. Thus, our results suggest that Gprc5a is a novel gene induced by PTH that acts in an inhibitory manner on both cell proliferation and osteoblast differentiation and is a candidate for drug targets for osteoporosis.


Assuntos
Diferenciação Celular , Proliferação de Células , Osteoblastos , Hormônio Paratireóideo , Receptores Acoplados a Proteínas G , Receptores Acoplados a Proteínas G/genética , Receptores Acoplados a Proteínas G/metabolismo , Osteoblastos/efeitos dos fármacos , Osteoblastos/metabolismo , Animais , Diferenciação Celular/efeitos dos fármacos , Diferenciação Celular/genética , Proliferação de Células/efeitos dos fármacos , Proliferação de Células/genética , Hormônio Paratireóideo/farmacologia , Camundongos , Ratos , Humanos , Transdução de Sinais/efeitos dos fármacos , Osteogênese/efeitos dos fármacos , Osteogênese/genética , Regiões Promotoras Genéticas/genética , Proteína Morfogenética Óssea 2/genética , Proteína Morfogenética Óssea 2/metabolismo , AMP Cíclico/metabolismo , Tretinoína/farmacologia , Calcitriol/farmacologia
6.
J Cell Physiol ; 239(4): e31197, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38284484

RESUMO

Cytoplasmic polyadenylation element-binding protein 4 (Cpeb4) is an RNA-binding protein that regulates posttranscriptional regulation, such as regulation of messenger RNA stability and translation. In the previous study, we reported that Cpeb4 localizes to nuclear bodies upon induction of osteoclast differentiation by RANKL. However, the mechanisms of the localization of Cpeb4 and osteoclastogenesis by Cpeb4 remain unknown. Here, we show that Cpeb4 localizes to the nuclear bodies by its RNA-binding ability and partially regulates normal splicing during osteoclast differentiation. Sodium dodecyl sulfate-polyacrylamide gel electrophoresis analysis with Phos-tag® revealed that the phosphorylation levels of Cpeb4 were already high in the RAW264.7 cells and were not altered by RANKL treatment. Immunofluorescence showed that exogenous Cpeb4 in HEK293T cells without RANKL stimulation localized to the same foci as shown in RANKL-stimulated RAW264.7 cells. Furthermore, when nuclear export was inhibited by leptomycin B treatment, Cpeb4 accumulated throughout the nucleus. Importantly, RNA recognition motif (RRM) 7 of Cpeb4 was essential for the localization. In contrast, the intrinsically disordered region, RRM1, and zinc finger domain CEBP_ZZ were not necessary for the localization. The mechanistic study showed that Cpeb4 co-localized and interacted with the splicing factors serine/arginine-rich splicing factor 5 (SRSF5) and SRSF6, suggesting that Cpeb4 may be involved in the splicing reaction. RNA-sequencing analysis revealed that the expression of genes related to cell proliferation processes, such as mitotic cell cycle and regulation of cell cycle processes, was elevated in osteoclasts depleted of Cpeb4. Interestingly, the splicing pattern of the inhibitor of DNA binding 2 (Id2) gene, which suppresses osteoclast differentiation, was altered by the depletion of Cpeb4. These results provide new insight into the role of Cpeb4 as a player of normal splicing of Id2 in osteoclast differentiation.


Assuntos
Proteína 2 Inibidora de Diferenciação , Osteoclastos , Splicing de RNA , Proteínas de Ligação a RNA , Humanos , Diferenciação Celular/genética , Células HEK293 , Osteoclastos/citologia , Fosfoproteínas/metabolismo , Fosforilação , Ligante RANK/genética , Ligante RANK/farmacologia , Ligante RANK/metabolismo , Proteínas de Ligação a RNA/genética , Proteínas de Ligação a RNA/metabolismo , Fatores de Processamento de Serina-Arginina/metabolismo , Proteína 2 Inibidora de Diferenciação/genética , Animais , Camundongos
7.
JBMR Plus ; 7(9): e10784, 2023 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-37701150

RESUMO

Gorham-Stout disease (GSD), also called vanishing bone disease, is a rare osteolytic disease, frequently associated with lymphangiomatous tissue proliferation. The causative genetic background has not been noted except for a case with a somatic mutation in KRAS. However, in the present study, we encountered a case of GSD from a consanguineous family member. Whole-exome sequencing (WES) analysis focusing on rare recessive variants with zero homozygotes in population databases identified a homozygous missense variant (c.823G > C, p.Asp275His) in gasdermin D (GSDMD) in the patient and heterozygous in his unaffected brother. Because this variant affects the Asp275 residue that is involved in proteolytic cleavage by caspase-11 (as well as -4 and -5) to generate an activating p30 fragment required for pyroptotic cell death and proinflammation, we confirmed the absence of this cleavage product in peripheral monocytic fractions from the patient. A recent study indicated that a shorter p20 fragment, generated by further cleavage at Asp88, has a cell-autonomous function to suppress the maturation of osteoclasts to resorb bone matrix. Thus, the present study suggests for the first time the existence of hereditary GSD cases or novel GSD-like diseases caused by GSDMD deficiency. © 2023 The Authors. JBMR Plus published by Wiley Periodicals LLC on behalf of American Society for Bone and Mineral Research.

8.
Stem Cell Res ; 61: 102744, 2022 05.
Artigo em Inglês | MEDLINE | ID: mdl-35292424

RESUMO

DiGeorge syndrome (22q11.2 deletion syndrome, or CATCH22 syndrome), caused by hemizygous deletion of chromosome 22q11.2, results in the poor development of multiple organs. Here we have generated DiGeorge syndrome-specific human induced pluripotsnt stem cells (hiPSCs) derived from four patients. These established hiPSC lines showed self-renewal and pluripotency and carried a hemizygous deletion in 22q11.2. Since the molecular pathogenesis of DiGeorge syndrome caused by the 22q11.2 deletion is largely unknown, these cell resources will be useful for recapitulating disease phenotypes and for developing new therapies for DiGeorge syndrome.


Assuntos
Síndrome de DiGeorge , Células-Tronco Pluripotentes Induzidas , Cromossomos Humanos Par 2 , Síndrome de DiGeorge/genética , Humanos , Fenótipo
9.
Biochem Biophys Res Commun ; 528(4): 621-627, 2020 08 06.
Artigo em Inglês | MEDLINE | ID: mdl-32517870

RESUMO

Cytoplasmic polyadenylation element binding (CPEB) proteins are RNA-binding proteins involved in translational regulation of the specific target mRNAs and control function of various organs including brain, liver and hematopoietic system. However, the role of CPEB proteins during osteoclast differentiation remains unclear. Here we show that Cpeb4 is required for RANKL-induced osteoclast differentiation in mouse macrophage-derived RAW264.7 cell line. Cpeb4 mRNA and protein levels are upregulated at the late stage of osteoclast differentiation. Immunofluorescence analysis revealed that Cpeb4 is translocated from cytoplasm to nuclear bodies in response to RANKL stimulation. Inhibition of PI3K-Akt signaling or calcium-NFAT pathways using chemical inhibitors suppressed nuclear localization of Cpeb4. Loss-of-function analysis showed that shRNA-mediated Cpeb4 depletion strongly impaired TRAP-positive osteoclast formation and expression of key differentiation markers including Acp5, Ctsk, Nfatc1 and Dcstamp. These results suggest that Cpeb4 is a positive regulator in osteoclastogenesis downstream of RANKL signaling.


Assuntos
Osteoclastos/citologia , Osteogênese , Proteínas de Ligação a RNA/metabolismo , Animais , Camundongos , Osteoclastos/metabolismo , Ligante RANK/metabolismo , Células RAW 264.7 , RNA Mensageiro/genética , Proteínas de Ligação a RNA/genética , Transdução de Sinais , Regulação para Cima
10.
Stem Cell Res ; 45: 101815, 2020 05.
Artigo em Inglês | MEDLINE | ID: mdl-32361464

RESUMO

Juvenile nephronophthisis is an inherited renal ciliopathy, causing cystic kidney disease, renal fibrosis, and end-stage renal failure. Human induced pluripotent stem cell (hiPSC) lines, derived from two Juvenile nephronophthisis patients, were generated from peripheral blood mononuclear cells by episomal plasmid vectors. Generated hiPSC lines showed self-renewal and pluripotency and carried a large deletion in NPHP1 (Nephrocystin 1) gene. Since the molecular pathogenesis caused by NPHP1 dysfunction remains unclear, these cell resources provide useful tools to establish disease models and to develop new therapies for juvenile nephronophthisis.


Assuntos
Células-Tronco Pluripotentes Induzidas , Proteínas Adaptadoras de Transdução de Sinal , Proteínas do Citoesqueleto , Fibrose , Humanos , Doenças Renais Císticas/congênito , Leucócitos Mononucleares , Proteínas de Membrana/genética
11.
JBMR Plus ; 3(6): e10130, 2019 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-31346562

RESUMO

Profilin 1 (Pfn1), a regulator of actin polymerization, controls cell movement in a context-dependent manner. Pfn1 supports the locomotion of most adherent cells by assisting actin-filament elongation, as has been shown in skeletal progenitor cells in our previous study. However, because Pfn1 has also been known to inhibit migration of certain cells, including T cells, by suppressing branched-end elongation of actin filaments, we hypothesized that its roles in osteoclasts may be different from that of osteoblasts. By investigating the osteoclasts in culture, we first verified that Pfn1-knockdown (KD) enhances bone resorption in preosteoclastic RAW264.7 cells, despite having a comparable number and size of osteoclasts. Pfn1-KD in bone marrow cells showed similar results. Mechanistically, Pfn1-KD osteoclasts appeared more mobile than in controls. In vivo, the osteoclast-specific conditional Pfn1-deficient mice (Pfn1-cKO) by CathepsinK-Cre driver demonstrated postnatal skeletal phenotype, including dwarfism, craniofacial deformities, and long-bone metaphyseal osteolytic expansion, by 8 weeks of age. Metaphyseal and diaphyseal femurs were drastically expanded with suppressed trabecular bone mass as indicated by µCT analysis. Histologically, TRAP-positive osteoclasts were increased at endosteal metaphysis to diaphysis of Pfn1-cKO mice. The enhanced movement of Pfn1-cKO osteoclasts in culture was associated with a slight increase in cell size and podosome belt length, as well as an increase in bone-resorbing activity. Our study, for the first time, demonstrated that Pfn1 has critical roles in inhibiting osteoclast motility and bone resorption, thereby contributing to essential roles in postnatal skeletal homeostasis. Our study also provides novel insight into understanding skeletal deformities in human disorders.

12.
Biochem Biophys Res Commun ; 498(4): 967-974, 2018 04 15.
Artigo em Inglês | MEDLINE | ID: mdl-29548825

RESUMO

Bone mass is determined by coordinated acts of osteoblasts and osteoclasts, which control bone formation and resorption, respectively. Osteoclasts are multinucleated, macrophage/monocyte lineage cells from bone marrow. The Dok-family adaptors Dok-1, Dok-2 and Dok-3 are expressed in the macrophage/monocyte lineage and negatively regulate many signaling pathways, implying roles in osteoclastogenesis. Indeed, mice lacking Dok-1 and Dok-2, the closest homologues with redundant functions, develop osteopenia with increased osteoclast counts compared to the wild-type controls. Here, we demonstrate that Dok-3 knockout (KO) mice also develop osteopenia. However, Dok-3 KO, but not Dok-1/-2 double-KO (DKO), mice develop larger osteoclasts within the normal cell-count range, suggesting a distinctive role for Dok-3. Indeed, Dok-3 KO, but not Dok-1/-2 DKO, bone marrow-derived cells (BMDCs) generated larger osteoclasts with more nuclei due to augmented cell-to-cell fusion in vitro. In addition, while Dok-1/-2 DKO BMDCs generated more osteoclasts, Dok-1/-2/-3 triple-KO (TKO) BMDCs generated osteoclasts increased in both number and size. Furthermore, Dok-1/-2/-3 TKO mice showed the combined effects of Dok-3 and Dok-1/-2 deficiency: severe osteopenia with more and larger osteoclasts. Together, our findings demonstrate that Dok-3 and Dok-1/-2 play distinctive but cooperative roles in osteoclastogenesis and protect mice from osteopenia, providing physiological and pathophysiological insight into bone homeostasis.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/fisiologia , Doenças Ósseas Metabólicas/prevenção & controle , Proteínas de Ligação a DNA/fisiologia , Osteoclastos/citologia , Osteogênese , Fosfoproteínas/fisiologia , Proteínas de Ligação a RNA/fisiologia , Proteínas Adaptadoras de Transdução de Sinal/genética , Animais , Células da Medula Óssea/citologia , Contagem de Células , Técnicas de Cultura de Células , Fusão Celular , Proliferação de Células , Tamanho Celular , Proteínas de Ligação a DNA/genética , Camundongos , Camundongos Knockout , Fosfoproteínas/genética , Proteínas de Ligação a RNA/genética
13.
Genes Cells ; 23(5): 345-356, 2018 May.
Artigo em Inglês | MEDLINE | ID: mdl-29521016

RESUMO

In mammals, the ovarian follicles are regulated at least in part by bone morphogenetic protein (BMP) family members. Dullard (also known as Ctdnep1) gene encodes a phosphatase that suppresses BMP signaling by inactivating or degrading BMP receptors. Here we report that the Col1a1-Cre-induced Dullard mutant mice displayed hemorrhagic ovarian cysts, with red blood cells accumulated in the follicles, resulting in infertility. Cells expressing Cre driven by Col1a1 2.3-kb promoter and their descendants were found in granulosa cells in the ovary and in Sertoli cells in the testis. DullardmRNA was localized to granulosa cells in the ovary. Genes involved in steroid hormone genesis including Cyp11a1, Hsd3b1 and Star were reduced, whereas expression of Smad6 and Smad7, BMP-inducible inhibitory Smads, was up-regulated in the Dullard mutant ovaries. Tamoxifen-inducible Dullard deletion in the whole body using Rosa26-CreER mice also resulted in hemorrhagic ovarian cysts in 2 weeks, which was rescued by administration of LDN-193189, a chemical inhibitor of BMP receptor kinase, suggesting that the hemorrhage in the Dullard-deficient ovarian follicles might be caused by increased BMP signaling. Thus, we conclude that Dullard is essential for ovarian homeostasis at least in part via suppression of BMP signaling.


Assuntos
Colágeno Tipo I/metabolismo , Hemorragia/patologia , Infertilidade Feminina/patologia , Cistos Ovarianos/patologia , Folículo Ovariano/patologia , Fosfoproteínas Fosfatases/deficiência , Animais , Proteínas Morfogenéticas Ósseas/antagonistas & inibidores , Osso e Ossos/metabolismo , Osso e Ossos/patologia , Colágeno Tipo I/genética , Cadeia alfa 1 do Colágeno Tipo I , Feminino , Regulação da Expressão Gênica/efeitos dos fármacos , Hemorragia/metabolismo , Infertilidade Feminina/metabolismo , Masculino , Camundongos , Camundongos Knockout , Cistos Ovarianos/metabolismo , Folículo Ovariano/metabolismo , Fosfoproteínas Fosfatases/fisiologia , Fosforilação , Pirazóis/farmacologia , Pirimidinas/farmacologia , Transdução de Sinais , Proteínas Smad/metabolismo , Testículo/metabolismo , Testículo/patologia
14.
Bone ; 101: 179-190, 2017 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-27170093

RESUMO

Bardet-Biedl Syndrome (BBS) is an autosomal recessive disorder and is classified as one of the ciliopathy. The patients manifest a characteristic craniofacial dysmorphology but the effects of Bbs3 deficiency in the developmental process during the craniofacial pathogenesis are still incompletely understood. Here, we analyzed a cranial development of a BBS model Bbs3-/- mouse. It was previously reported that these mutant mice exhibit a dome-shape cranium. We show that Bbs3-/- mouse embryos present mid-facial hypoplasia and solitary central upper incisor. Morphologically, these mutant mice show synchondrosis of the cranial base midline due to the failure to fuse in association with loss of intrasphenoidal synchondrosis. The cranial base was laterally expanded and longitudinally shortened. In the developing cartilaginous primordium of cranial base, cells present in the midline were less in Bbs3-/- embryos. Expression of BBS3 was observed specifically in a cell population lying between condensed ectomesenchyme in the midline and the ventral midbrain at this stage. Finally, siRNA-based knockdown of Bbs3 in ATDC5 cells impaired migration in culture. Our data suggest that BBS3 is required for the development of cranial base via regulation of cell migration toward the midline where they promote the condensation of ectomesenchyme and form the future cartilaginous templates of cranial base.


Assuntos
Fatores de Ribosilação do ADP/metabolismo , Síndrome de Bardet-Biedl/metabolismo , Base do Crânio/crescimento & desenvolvimento , Base do Crânio/metabolismo , Proteínas de Peixe-Zebra/metabolismo , Fatores de Ribosilação do ADP/genética , Animais , Síndrome de Bardet-Biedl/genética , Feminino , Imunofluorescência , Imuno-Histoquímica , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Mutação/genética , Fenótipo , Microtomografia por Raio-X , Peixe-Zebra , Proteínas de Peixe-Zebra/genética
15.
J Cell Biochem ; 117(4): 970-7, 2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-26378628

RESUMO

CIZ/NMP4 (Cas interacting zinc finger protein, Nmp4, Zfp384) is a transcription factor that is known to regulate matrix related-proteins. To explore the possible pathophysiological role of CIZ/NMP4 in arthritis, we examined CIZ/NMP4 expression in articular cartilage in arthritis model. CIZ/NMP4 was expressed in the articular chondrocytes of mice at low levels while its expression was enhanced when arthritis was induced. Arthritis induction increased clinical score in wild type mice. In contrast, CIZ/NMP4 deficiency suppressed such rise in the levels of arthritis score and swelling of soft tissue. CIZ/NMP4 deficiency also reduced invasion of inflammatory cells in joint tissue. Quantitative PCR analyses of mRNA from joints revealed that arthritis-induced increase in expressions of IL-1ß was suppressed by CIZ/NMP4 deficiency. CIZ/NMP4 bound to IL-1ß promoter and activated its transcription. The increase in CIZ/NMP4 in arthritis was also associated with enhancement in bone resorption and cartilage matrix degradation. In fact, RANKL, a signaling molecule prerequisite for osteoclastogenesis and, MMP-3, a clinical marker for arthritis were increased in joints upon arthritis induction. In contrast, CIZ/NMP4 deficiency suppressed the arthritis-induced increase in bone resorption, expression of RANKL and MMP-3 mRNA. Thus, CIZ/NMP4 plays a role in the development of arthritis at least in part through regulation of key molecules related to the arthritis.


Assuntos
Artrite Experimental/genética , Cartilagem Articular/imunologia , Metaloproteinase 3 da Matriz/imunologia , Proteínas Associadas à Matriz Nuclear/imunologia , Ligante RANK/imunologia , Fatores de Transcrição/imunologia , Animais , Artrite Experimental/induzido quimicamente , Artrite Experimental/imunologia , Artrite Experimental/patologia , Autoanticorpos/biossíntese , Reabsorção Óssea , Cartilagem Articular/patologia , Condrócitos/imunologia , Condrócitos/patologia , Feminino , Regulação da Expressão Gênica , Glucose-6-Fosfato Isomerase/antagonistas & inibidores , Glucose-6-Fosfato Isomerase/genética , Glucose-6-Fosfato Isomerase/imunologia , Soros Imunes/administração & dosagem , Interleucina-1beta/genética , Interleucina-1beta/imunologia , Articulações/imunologia , Articulações/patologia , Masculino , Metaloproteinase 3 da Matriz/genética , Camundongos , Camundongos Knockout , Proteínas Associadas à Matriz Nuclear/deficiência , Proteínas Associadas à Matriz Nuclear/genética , Regiões Promotoras Genéticas , Ligante RANK/genética , Índice de Gravidade de Doença , Transdução de Sinais , Fatores de Transcrição/deficiência , Fatores de Transcrição/genética , Transcrição Gênica
16.
Proc Natl Acad Sci U S A ; 112(50): 15432-7, 2015 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-26621720

RESUMO

Migration of the cells in osteoblastic lineage, including preosteoblasts and osteoblasts, has been postulated to influence bone formation. However, the molecular bases that link preosteoblastic/osteoblastic cell migration and bone formation are incompletely understood. Nck (noncatalytic region of tyrosine kinase; collectively referred to Nck1 and Nck2) is a member of the signaling adaptors that regulate cell migration and cytoskeletal structures, but its function in cells in the osteoblastic lineage is not known. Therefore, we examined the role of Nck in migration of these cells. Nck is expressed in preosteoblasts/osteoblasts, and its knockdown suppresses migration as well as cell spreading and attachment to substrates. In contrast, Nck1 overexpression enhances spreading and increases migration and attachment. As for signaling, Nck double knockdown suppresses migration toward IGF1 (insulin-like growth factor 1). In these cells, Nck1 binds to IRS-1 (insulin receptor substrate 1) based on immunoprecipitation experiments using anti-Nck and anti-IRS-1 antibodies. In vivo, Nck knockdown suppresses enlargement of the pellet of DiI-labeled preosteoblasts/osteoblasts placed in the calvarial defects. Genetic experiments indicate that conditional double deletion of both Nck1 and Nck2 specifically in osteoblasts causes osteopenia. In these mice, Nck double deficiency suppresses the levels of bone-formation parameters such as bone formation rate in vivo. Interestingly, bone-resorption parameters are not affected. Finally, Nck deficiency suppresses repair of bone injury after bone marrow ablation. These results reveal that Nck regulates preosteoblastic/osteoblastic migration and bone mass.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Osso e Ossos/citologia , Movimento Celular , Proteínas Oncogênicas/metabolismo , Osteoblastos/citologia , Osteoblastos/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/deficiência , Animais , Reabsorção Óssea/metabolismo , Reabsorção Óssea/patologia , Osso e Ossos/diagnóstico por imagem , Osso e Ossos/efeitos dos fármacos , Movimento Celular/efeitos dos fármacos , Forma Celular/efeitos dos fármacos , Células Cultivadas , Técnicas de Silenciamento de Genes , Proteínas Substratos do Receptor de Insulina/metabolismo , Fator de Crescimento Insulin-Like I/farmacologia , Camundongos Knockout , Proteínas Oncogênicas/deficiência , Tamanho do Órgão , Osteoblastos/efeitos dos fármacos , Osteogênese/efeitos dos fármacos , Ligação Proteica/efeitos dos fármacos , Radiografia , Crânio/efeitos dos fármacos , Crânio/metabolismo , Cicatrização/efeitos dos fármacos
18.
J Cell Physiol ; 230(11): 2788-95, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-25828538

RESUMO

Ift88 is an intraflagella transport protein, critical for the cilium, and has been shown to be required for the maintenance of chondrocytes and cartilage. However, how Ift88 is controlled by cytokines that play a role in osteoarthritis is not well understood. Therefore, we examined the effects of TGF-ß on the expression of Ift88. We used ATDC5 cells as chondrocytes and analyzed the effects of TGF-ß on gene expression. TGF-ß treatment suppresses the levels of Ift88 mRNA in a dose-dependent manner starting from as low as 0.5 ng/mL and reaching the nadir at around 2 ng/mL. TGF-ß treatment also suppresses the protein levels of Ift88. TGF-ß suppression of Ift88 is still observed when the cells are cultured in the presence of a transcriptional inhibitor while the TGF-ß suppression is weakened in the presence of a protein synthesis inhibitor, cycloheximide. TGF-ß treatment suppresses the levels of Ift88 mRNA stability suggesting the presence of posttranscriptional regulation. TGF-ß treatment reduces the number of cilia positive cells and suppresses average length of cilia. Knockdown of Ift88 by siRNA enhances TGF-ß-induced increase in type II collagen mRNA expression in ATDC5 cells revealing the suppressive role of Ift88 on TGF-ß-induced regulation of extracellular matrix protein expression. TGF-ß also suppresses Ift88 mRNA expression in primary culture of rib chondrocytes. These data indicate that TGF-ß regulates Ift88 gene expression at least in part via posttrascriptional manner.


Assuntos
Cílios/metabolismo , Osteoartrite/genética , Fator de Crescimento Transformador beta/metabolismo , Proteínas Supressoras de Tumor/biossíntese , Animais , Cartilagem Articular/metabolismo , Condrócitos/metabolismo , Cílios/patologia , Regulação da Expressão Gênica/efeitos dos fármacos , Camundongos , Osteoartrite/metabolismo , Osteoartrite/patologia , Fosforilação , Transdução de Sinais , Fator de Crescimento Transformador beta/administração & dosagem , Proteínas Supressoras de Tumor/genética , Proteínas Supressoras de Tumor/metabolismo
19.
Nihon Rinsho ; 73 Suppl 5: 40-6, 2015 Jun.
Artigo em Japonês | MEDLINE | ID: mdl-30457310
20.
J Cell Biochem ; 116(6): 1144-52, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-25536656

RESUMO

ß adrenergic stimulation suppresses bone formation in vivo while its actions in osteoblastic differentiation are still incompletely understood. We therefore examined the effects of ß2 adrenergic stimulation on osteoblast-like MC3T3-E1 cells focusing on BMP-induced alkaline phosphatase expression. Morphologically, isoproterenol treatment suppresses BMP-induced increase in the numbers of alkaline phosphatase-positive small foci in the cultures of MC3T3-E1 cells. Biochemically, isoproterenol treatment suppresses BMP-induced enzymatic activity of alkaline phosphatase in a dose-dependent manner. Isoproterenol suppression of alkaline phosphatase activity is observed even when the cells are treated with high concentrations of BMP. With respect to cell density, isoproterenol treatment tends to suppress BMP-induced increase in alkaline phosphatase expression more in osteoblasts cultured at higher cell density. In terms of treatment protocol, continuous isoproterenol treatment is compared to cyclic treatment. Continuous isoproterenol treatment is more suppressive against BMP-induced increase in alkaline phosphatase expression than cyclic regimen. At molecular level, isoproterenol treatment suppresses BMP-induced enhancement of alkaline phosphatase mRNA expression. Regarding the mode of isoproterenol action, isoproterenol suppresses BMP-induced BRE-luciferase activity. These data indicate that isoproterenol regulates BMP-induced alkaline phosphatase expression in osteoblast-like MC3T3E1 cells.


Assuntos
Fosfatase Alcalina/metabolismo , Proteínas Morfogenéticas Ósseas/farmacologia , Osteoblastos/metabolismo , Receptores Adrenérgicos/metabolismo , Animais , Diferenciação Celular/efeitos dos fármacos , Diferenciação Celular/fisiologia , Linhagem Celular , Camundongos , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA