Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
J Bone Miner Res ; 39(5): 611-626, 2024 May 24.
Artigo em Inglês | MEDLINE | ID: mdl-38477792

RESUMO

The impaired bone healing in tooth extraction sockets due to periodontitis presents a major obstacle to restoring oral health. The mechanisms regulating the osteogenic capacity of jawbone-derived stromal cells in the periodontitis microenvironment remain elusive. Leptin receptor (LepR) expressing stromal cells, which largely overlap with Cxcl12-abundant reticular (CAR) cells in bone tissue, rapidly proliferate and differentiate into bone-forming cells during extraction socket healing to support alveolar bone repair. In this study, we identify that CCRL2 is significantly expressed and inhibits osteogenesis in LepR+/CAR cells of alveolar bones with periodontitis. The Ccrl2-KO mice exhibit significant improvements in bone healing in extraction sockets with periodontitis. Specifically, the binding of CCRL2 to SFRP1 on the surface of LepR+/CAR cells can amplify the suppressive effect of SFRP1 on Wnt signaling under inflammation, thus hindering the osteogenic differentiation of LepR+/CAR cells and resulting in poor bone healing in extraction sockets with periodontitis. Together, we clarify that the CCRL2 receptor of LepR+/CAR cells can respond to periodontitis and crosstalk with Wnt signaling to deteriorate extraction socket healing.


The impaired bone healing in tooth extraction sockets due to periodontitis presents a major obstacle to restoring oral health. Alterations in the cellular activity of LepR+/CAR cells, an essential stromal cell population for extraction socket healing, in the periodontitis microenvironment have yet to be determined. In this study, we identify that CCRL2, as a potent agent of inflammation-bone crosstalk, is significantly expressed and inhibits osteogenesis in LepR+/CAR cells of alveolar bones with periodontitis. Specifically, the binding of CCRL2 to SFRP1 on the surface of LepR+/CAR cells can amplify the suppressive effect of SFRP1 on the Wnt/ß-catenin signaling under inflammation, thus hindering the osteogenic differentiation of LepR+/CAR cells and resulting in poor bone healing in tooth extraction sockets with periodontitis.


Assuntos
Osteogênese , Periodontite , Receptores para Leptina , Via de Sinalização Wnt , Animais , Periodontite/metabolismo , Periodontite/patologia , Receptores para Leptina/metabolismo , Receptores para Leptina/deficiência , Receptores para Leptina/genética , Camundongos , Camundongos Knockout , Células Estromais/metabolismo , Células Estromais/patologia , Masculino , Humanos , Processo Alveolar/patologia , Processo Alveolar/metabolismo , Cicatrização , Proteínas de Membrana/metabolismo
2.
Artigo em Inglês | MEDLINE | ID: mdl-38498783

RESUMO

PURPOSE: To explore the contribution of paired-related homeobox 1-positive cells to the implant-induced osseointegration process in adult alveolar bone and the potential underlying mechanisms. MATERIALS AND METHODS: Cre recombinase-induced lineage tracing and cell ablation were conducted in a murine dental implant model. Scratch and transwell assays were used to assess MC3T3-E1 cell migration after paired-related homeobox 1 overexpression. Single-cell RNA sequencing were applied to identify potential genes involved in pairedrelated homeobox 1-positive cells-driven osteogenesis. RESULTS: Paired-related homeobox 1- positive cells were observed to accumulate in the peri-implant area in a time-dependent manner. The number of these cells were found to reach its maximum on day 14. Osseointegration in mice were noticeably impaired after ablation of paired-related homeobox 1-positive cells. Further, it was discovered that paired-related homeobox 1 promotes MC3T3- E1 cell migration, a process which is indispensable for sound healing of peri-implant tissue. Finally, Semaphorin 3C was detected exclusively and abundantly expressed by paired-related homeobox 1-positive cells. Knockdown of semaphorin 3C in paired-related homeobox 1- positive cells significantly weakened their osteogenic potential. CONCLUSION: Our data suggest that paired-related homeobox 1-positive cells contribute to the osseointegration process under stress stimulation and semaphorin 3C may play a critical role in paired-related homeobox 1- positive cell-driven osteogenesis. Paired-related homeobox 1 could significantly promote MC3T3-E1 cell migration.

3.
Am J Pathol ; 193(2): 213-232, 2023 02.
Artigo em Inglês | MEDLINE | ID: mdl-36410421

RESUMO

This study explored the role of transient receptor potential channel melastatin 2 (TRPM2)-mediated activation of NOD-, LRR-, and pyrin domain-containing protein 3 (NLRP3) inflammasome in osteogenesis during healing of tooth extraction sockets. Tooth extraction socket tissue samples were collected from patients with or without periodontitis. In a TRPM2 knockout mouse model of socket healing, mice with or without periodontitis and their wild-type littermates were used for comparing the socket healing phenotypes. Micro-computed tomography imaging, three-dimensional reconstruction of the sockets, and hematoxylin and eosin staining for histopathologic analysis were performed. Immunofluorescence, immunohistochemistry, and Western blot analysis were used for evaluation of protein expression; the mRNA levels were evaluated by quantitative RT-PCR. Osteogenic, chondrogenic, and adipogenic differentiation potential of human bone marrow mesenchymal stem cells (BMMSCs) was evaluated. Calcium deposition was evaluated using Alizarin Red S staining. NLRP3 and CASP1 were up-regulated in tooth sockets of periodontitis patients. NLRP3 knockdown promoted the osteogenic differentiation of maxillary BMMSCs under inflammatory conditions. TRPM2 was up-regulated in the tooth extraction socket tissue of periodontitis. Inhibiting TRPM2 expression mitigated the NLRP3 inflammasome and its deleterious effect on osteogenesis. Activation of the TRPM2 ion channel regulated osteogenesis of BMMSCs under inflammatory conditions via Ca2+ influx, the mitochondrial dynamics, and pyroptosis. Targeting the TRPM2/Ca2+/NLRP3 axis could be beneficial in the healing process of the tooth extraction sockets of patients with periodontitis.


Assuntos
Periodontite , Canais de Cátion TRPM , Canais de Potencial de Receptor Transitório , Humanos , Camundongos , Animais , Inflamassomos/metabolismo , Osteogênese/fisiologia , Alvéolo Dental/metabolismo , Proteína 3 que Contém Domínio de Pirina da Família NLR/genética , Proteína 3 que Contém Domínio de Pirina da Família NLR/metabolismo , Canais de Cátion TRPM/genética , Canais de Cátion TRPM/metabolismo , Microtomografia por Raio-X , Camundongos Endogâmicos NOD , Extração Dentária
4.
Cell Res ; 32(9): 814-830, 2022 09.
Artigo em Inglês | MEDLINE | ID: mdl-35821090

RESUMO

Bone regeneration originates from proliferation and differentiation of osteoprogenitors via either endochondral or intramembranous ossification; and the regeneration capacities decline with age and estrogen loss. Maxillary sinus floor lifting (MSFL) is a commonly used surgical procedure for guiding bone regeneration in maxilla. Radiographic analysis of 1210 clinical cases of maxilla bone regeneration after MSFL revealed that the intrasinus osteogenic efficacy was independent of age and gender, however; and this might be related to the Schneiderian membrane that lines the sinus cavity. In view of the particularity of this biological process, our present study aimed to elucidate the underlying mechanism of MSFL-induced bone regeneration. We first established a murine model to simulate the clinical MSFL. By single-cell RNA-sequencing and flow cytometry-based bulk RNA-sequencing, we identified a novel Krt14+Ctsk+ subset of cells that display both epithelial and mesenchymal properties and the transcriptomic feature of osteoprogenitors. Dual recombinases-mediated lineage tracing and loss-of-function analyses showed that these Krt14+Ctsk+ progenitors contribute to both MSFL-induced osteogenesis and physiological bone homeostasis by differentiating into Krt14-Ctsk+ descendants which show robust osteogenic capacity. In addition, we detected a similar population of Krt14+Ctsk+ cells in human samples of Schneiderian membrane, which show a highly similar osteogenic potential and transcriptomic feature to the corresponding cells in mice. The identification of this Krt14+Ctsk+ population, featured by osteoprogenitor characteristics and dual epithelial-mesenchymal properties, provides new insight into the understanding of bone regeneration and may open more possibilities for clinical applications.


Assuntos
Seio Maxilar , Levantamento do Assoalho do Seio Maxilar , Animais , Regeneração Óssea , Diferenciação Celular , Homeostase , Humanos , Camundongos , Osteogênese/fisiologia , RNA , Levantamento do Assoalho do Seio Maxilar/métodos
5.
Redox Biol ; 40: 101849, 2021 04.
Artigo em Inglês | MEDLINE | ID: mdl-33486152

RESUMO

Periodontitis is the sixth most prevalent diseases around the globe, which is closely related to many systemic diseases and affects general health. As the leading cause of tooth loss, periodontitis is characterized by irreversible alveolar bone loss and activated osteoclastogenic process, which might be closely related to the activated intracellular reactive oxygen species (ROS) in osteoclasts. Here, we demonstrated triggering receptor expressed on myeloid cells 2 (Trem2) as a key regulator of osteoclastogenesis with the regulation of intracellular ROS signals in periodontitis. In the present study, the expression of Trem2 was significantly upregulated in human alveolar bones diagnosed with chronic periodontitis, as assessed by RNA-seq. In the mice model of periodontitis, the alveolar bone resorption was impeded in the presence of the conditional knockout of Trem2 in osteoclasts. Furthermore, we identified Trem2/DAP12/Syk-dependent cascade as a vital intracellular signaling for the amplification of reactive oxygen species (ROS) signals in osteoclastogenesis, while the accumulation of soluble Aß42 oligomers (Aßo) in periodontitis microenvironment further strengthened the signals and enhanced osteoclastogenesis through direct interactions with Trem2. Collectively, Trem2 mediated ROS signal amplification cascade was crucial in the process of osteoclastogenesis in periodontitis, suggesting the potential of Trem2 as a target for the prevention and treatment of bone destruction in periodontitis.


Assuntos
Osteogênese , Periodontite , Humanos , Glicoproteínas de Membrana/genética , Osteoclastos , Periodontite/genética , Ligante RANK , Espécies Reativas de Oxigênio , Receptores Imunológicos/genética , Quinase Syk/genética
6.
Nat Commun ; 11(1): 4593, 2020 09 14.
Artigo em Inglês | MEDLINE | ID: mdl-32929070

RESUMO

Gene-targeted animal models that are generated by injecting Cas9 and sgRNAs into zygotes are often accompanied by undesired double-strand break (DSB)-induced byproducts and random biallelic targeting due to uncontrollable Cas9 targeting activity. Here, we establish a parental allele-specific gene-targeting (Past-CRISPR) method, based on the detailed observation that pronuclear transfer-mediated cytoplasmic dilution can effectively terminate Cas9 activity. We apply this method in embryos to efficiently target the given parental alleles of a gene of interest and observed little genomic mosaicism because of the spatiotemporal control of Cas9 activity. This method allows us to rapidly explore the function of individual parent-of-origin effects and to construct animal models with a single genomic change. More importantly, Past-CRISPR could also be used for therapeutic applications or disease model construction.


Assuntos
Alelos , Sistemas CRISPR-Cas/genética , Núcleo Celular/genética , Edição de Genes , Terapia de Substituição Mitocondrial , Animais , Sequência de Bases , Modelos Animais de Doenças , Nanismo/genética , Perda do Embrião/genética , Feminino , Marcação de Genes , Genes Dominantes , Impressão Genômica , Heterozigoto , Camundongos Endogâmicos C57BL , Camundongos Knockout , Mutação , Reprodutibilidade dos Testes , Fatores de Tempo
7.
Aging (Albany NY) ; 12(6): 5362-5383, 2020 03 25.
Artigo em Inglês | MEDLINE | ID: mdl-32209725

RESUMO

Reactive oxygen species (ROS) play a pivotal role in the development of pathological cardiac hypertrophy. Delphinidin, a natural flavonoid, was reported to exert marked antioxidative effects. Therefore, we investigated whether delphinidin ameliorates pathological cardiac hypertrophy via inhibiting oxidative stress. In this study, male C57BL/6 mice were treated with DMSO or delphinidin after surgery. Neonatal rat cardiomyocytes (NRCMs) were treated with angiotensin II (Ang II) and delphinidin in vitro. Eighteen-month-old mice were administered delphinidin to investigate the effect of delphinidin on aging-related cardiac hypertrophy. Through analyses of hypertrophic cardiomyocyte growth, fibrosis and cardiac function, delphinidin was demonstrated to confer resistance to aging- and transverse aortic constriction (TAC)-induced cardiac hypertrophy in vivo and attenuate Ang II-induced cardiomyocyte hypertrophy in vitro by significantly suppressing hypertrophic growth and the deposition of fibrosis. Mechanistically, delphinidin reduced ROS accumulation upon Ang II stimulation through the direct activation of AMP-activated protein kinase (AMPK) and subsequent inhibition of the activity of Rac1 and expression of p47phox. In addition, excessive levels of ERK1/2, P38 and JNK1/2 phosphorylation induced by oxidative stress were abrogated by delphinidin. Delphinidin was conclusively shown to repress pathological cardiac hypertrophy by modulating oxidative stress through the AMPK/NADPH oxidase (NOX)/mitogen-activated protein kinase (MAPK) signaling pathway.


Assuntos
Proteínas Quinases Ativadas por AMP/metabolismo , Antocianinas/farmacologia , Cardiomegalia/metabolismo , Angiotensina II/farmacologia , Animais , Fibrose/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Miócitos Cardíacos/metabolismo , NADPH Oxidases/metabolismo , Estresse Oxidativo , Fosforilação , Ratos , Espécies Reativas de Oxigênio/metabolismo , Transdução de Sinais , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
8.
Front Med ; 13(5): 575-589, 2019 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-31065929

RESUMO

Fractures are frequently occurring diseases that endanger human health. Crucial to fracture healing is cartilage formation, which provides a bone-regeneration environment. Cartilage consists of both chondrocytes and extracellular matrix (ECM). The ECM of cartilage includes collagens and various types of proteoglycans (PGs), which play important roles in maintaining primary stability in fracture healing. The PG form of dentin matrix protein 1 (DMP1-PG) is involved in maintaining the health of articular cartilage and bone. Our previous data have shown that DMP1-PG is richly expressed in the cartilaginous calluses of fracture sites. However, the possible significant role of DMP1-PG in chondrogenesis and fracture healing is unknown. To further detect the potential role of DMP1-PG in fracture repair, we established a mouse fracture model by using a glycosylation site mutant DMP1 mouse (S89G-DMP1 mouse). Upon inspection, fewer cartilaginous calluses and down-regulated expression levels of chondrogenesis genes were observed in the fracture sites of S89G-DMP1 mice. Given the deficiency of DMP1-PG, the impaired IL-6/JAK/STAT signaling pathway was observed to affect the chondrogenesis of fracture healing. Overall, these results suggest that DMP1-PG is an indispensable proteoglycan in chondrogenesis during fracture healing.


Assuntos
Condrogênese , Proteínas da Matriz Extracelular/genética , Consolidação da Fratura , Glicosilação , Animais , Cartilagem/metabolismo , Condrócitos/metabolismo , Modelos Animais de Doenças , Proteínas da Matriz Extracelular/química , Regulação da Expressão Gênica no Desenvolvimento , Masculino , Camundongos , Microtomografia por Raio-X
9.
Protein Cell ; 10(8): 620-621, 2019 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-30159643

RESUMO

In the original publication, the label of Fig. 2C should be read as "GFAP/lectin/DAPI" not "DMP1/GFAP/lectin/DAPI".

10.
Protein Cell ; 9(3): 298-309, 2018 03.
Artigo em Inglês | MEDLINE | ID: mdl-28822114

RESUMO

The blood-brain barrier (BBB) is a tight boundary formed between endothelial cells and astrocytes, which separates and protects brain from most pathogens as well as neural toxins in circulation. However, detailed molecular players involved in formation of BBB are not completely known. Dentin matrix protein 1 (DMP1)-proteoglycan (PG), which is known to be involved in mineralization of bones and dentin, is also expressed in soft tissues including brain with unknown functions. In the present study, we reported that DMP1-PG was expressed in brain astrocytes and enriched in BBB units. The only glycosylation site of DMP1 is serine89 (S89) in the N-terminal domain of the protein in mouse. Mutant mice with DMP1 point mutations changing S89 to glycine (S89G), which completely eradicated glycosylation of the protein, demonstrated severe BBB disruption. Another breed of DMP1 mutant mice, which lacked the C-terminal domain of DMP1, manifested normal BBB function. The polarity of S89G-DMP1 astrocytes was disrupted and cell-cell adhesion was loosened. Through a battery of analyses, we found that DMP1 glycosylation was critically required for astrocyte maturation both in vitro and in vivo. S89G-DMP1 mutant astrocytes failed to express aquaporin 4 and had reduced laminin and ZO1 expression, which resulted in disruption of BBB. Interestingly, overexpression of wild-type DMP1-PG in mouse brain driven by the nestin promoter elevated laminin and ZO1 expression beyond wild type levels and could effectively resisted intravenous mannitol-induced BBB reversible opening. Taken together, our study not only revealed a novel element, i.e., DMP1-PG, that regulated BBB formation, but also assigned a new function to DMP1-PG.


Assuntos
Astrócitos/citologia , Astrócitos/metabolismo , Barreira Hematoencefálica/citologia , Barreira Hematoencefálica/metabolismo , Proteínas da Matriz Extracelular/metabolismo , Animais , Células Cultivadas , Proteínas da Matriz Extracelular/genética , Feminino , Glicosilação , Masculino , Camundongos , Proteoglicanas/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa
11.
Biochem Biophys Res Commun ; 490(2): 356-363, 2017 08 19.
Artigo em Inglês | MEDLINE | ID: mdl-28647374

RESUMO

The well-known matrix protein Dentin matrix protein 1 (DMP1) is expressed by osteoblasts and osteocytes in bone, and it controls bone mineralization. Recently, it has been found that DMP1 is also expressed in other cell types, such as chondrocytes. Nestin+ cells are one important type of progenitor cell in bone marrow and are associated with bone remodeling. In our preliminary experiment, DMP1 could also be detected in Nestin+ cells in bone marrow. This study was designed to explore the effect on bone of DMP1 in Nestin+ cells. A transgenic mouse model with DMP1 expression driven by the Nestin promoter was generated. In vivo and in vitro experiments revealed that overexpression of DMP1 in Nestin+ cells could limit the proliferation and osteogenic differentiation of BMMSCs, subsequently leading to decreased bone mass. Lower expression of bone matrix protein and a lower bone deposition rate were also observed. Meanwhile, overexpression of DMP1 in Nestin+ cells had no influence on osteoclast activity. These data indicate that DMP1 plays negative roles in differentiation of Nestin+ cells and bone formation.


Assuntos
Osso e Ossos/fisiopatologia , Proteínas da Matriz Extracelular/genética , Nestina/genética , Osteogênese , Animais , Densidade Óssea , Remodelação Óssea , Osso e Ossos/metabolismo , Osso e Ossos/patologia , Proliferação de Células , Proteínas da Matriz Extracelular/metabolismo , Regulação da Expressão Gênica , Masculino , Células-Tronco Mesenquimais/citologia , Células-Tronco Mesenquimais/metabolismo , Células-Tronco Mesenquimais/patologia , Camundongos , Camundongos Transgênicos , Nestina/metabolismo , Regiões Promotoras Genéticas , Regulação para Cima
12.
J Mol Histol ; 47(5): 475-83, 2016 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-27541035

RESUMO

Osteoarthritis (OA) was a degenerative joint disease characterized by articular cartilage degradation and extensive remodeling of the subchondral bone. Multiple lines of evidence indicated that Osteoprotegerin (OPG), a member of TNF receptor superfamily that was expressed in the chondrocytes of articular cartilage and adjacent locations in the physiological setting, was involved in maintaining integrity of articular cartilage. OPG could prevent subchondral bone from resorption, and also protect cartilage from degradation. In this study, we used Osteoprotegerin-knockout mice (Opg-KO mice) to find out the role of OPG in articular cartilage. We examined articular cartilage in the femoral head of Opg-KO mice began in early adulthood using modern molecular and imaging methods. We found cartilage changes starting from adulthood and progressively with age, reminiscent of pathological changes in OA. Deficiency of OPG caused thinned articular cartilage and extensive remodeling of the subchondral bone in femoral head in comparison with wild-type mice (WT mice). Also, the articular cartilage of femoral head expressed significantly less of Aggrecan, Col-II and Col-X, but more Col-I and Matrix Metalloproteinases-13 (Mmp-13) than WT mice both at gene and protein level. Moreover, increased chondrocyte apoptosis and decreased chondrocyte proliferation were observed in femoral head of Opg-KO mice compared to WT mice. These data suggested that OPG played an important role in maintaining the homeostasis of articular cartilage of femoral head.


Assuntos
Cartilagem Articular/metabolismo , Cartilagem Articular/patologia , Cabeça do Fêmur/metabolismo , Cabeça do Fêmur/patologia , Osteogênese/genética , Osteoprotegerina/deficiência , Animais , Contagem de Células , Sobrevivência Celular/genética , Condrócitos/metabolismo , Modelos Animais de Doenças , Matriz Extracelular/metabolismo , Cabeça do Fêmur/diagnóstico por imagem , Camundongos , Camundongos Knockout , Osteoartrite/diagnóstico por imagem , Osteoartrite/genética , Osteoartrite/metabolismo , Osteoartrite/patologia , Osteoclastos/metabolismo , Microtomografia por Raio-X
13.
Sci Rep ; 5: 17518, 2015 Dec 04.
Artigo em Inglês | MEDLINE | ID: mdl-26634432

RESUMO

Proteoglycans play important roles in regulating osteogenesis. Dentin matrix protein 1 (DMP1) is a highly expressed bone extracellular matrix protein that regulates both bone development and phosphate metabolism. After glycosylation, an N-terminal fragment of DMP1 protein was identified as a new proteoglycan (DMP1-PG) in bone matrix. In vitro investigations showed that Ser(89) is the key glycosylation site in mouse DMP1. However, the specific role of DMP1 glycosylation is still not understood. In this study, a mutant DMP1 mouse model was developed in which the glycosylation site S(89) was substituted with G(89) (S89G-DMP1). The glycosylation level of DMP1 was down-regulated in the bone matrix of S89G-DMP1 mice. Compared with wild type mice, the long bones of S89G-DMP1 mice showed developmental changes, including the speed of bone remodeling and mineralization, the morphology and activities of osteocytes, and activities of both osteoblasts and osteoclasts. These findings indicate that glycosylation of DMP1 is a key posttranslational modification process during development and that DMP1-PG functions as an indispensable proteoglycan in osteogenesis.


Assuntos
Proteínas da Matriz Extracelular/genética , Glicosilação , Osteogênese/genética , Animais , Matriz Óssea/metabolismo , Proteínas da Matriz Extracelular/biossíntese , Regulação da Expressão Gênica no Desenvolvimento , Camundongos , Osteoblastos/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...