Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Junqing Ma is active.

Publication


Featured researches published by Junqing Ma.


Proceedings of the National Academy of Sciences of the United States of America | 2014

Cbfβ deletion in mice recapitulates cleidocranial dysplasia and reveals multiple functions of Cbfβ required for skeletal development

Wei Chen; Junqing Ma; Guochun Zhu; Joel Jules; Mengrui Wu; Matthew McConnell; Fei Tian; Christie Paulson; Xuedong Zhou; Lin Wang; Yi-Ping Li

Significance Cleidocranial dysplasia (CCD) is a hereditary human skeletal disease. Mutations in Runt-related transcription factor 2, which functions as a heterodimer with core binding factor β (Cbfβ), are found in most individuals with CCD. It has been suspected that Cbfβ may be responsible for other CCD cases. The pathogenesis of CCD and the role of Cbfβ in postnatal skeletogenesis remain unclear. There has been no animal model to study this disease. We demonstrate that ablation of Cbfβ in various skeletal cells results in severe craniofacial and skeletal dysplasia with the phenotype recapitulating clinical features of CCD. The findings from this study of Cbfβ in the skeleton provide insight into the role of Cbfβ in postnatal skeletogenesis and pathogenesis of CCD, which may assist in developing new therapies for CCD and osteoporosis. The pathogenesis of cleidocranial dysplasia (CCD) as well as the specific role of core binding factor β (Cbfβ) and the Runt-related transcription factor (RUNX)/Cbfβ complex in postnatal skeletogenesis remain unclear. We demonstrate that Cbfβ ablation in osteoblast precursors, differentiating chondrocytes, osteoblasts, and odontoblasts via Osterix-Cre, results in severe craniofacial dysplasia, skeletal dysplasia, abnormal teeth, and a phenotype recapitulating the clinical features of CCD. Cbfβf/fOsterix-Cre mice have fewer proliferative and hypertrophic chondrocytes, fewer osteoblasts, and almost absent trabecular bone, indicating that Cbfβ may maintain trabecular bone formation through its function in hypertrophic chondrocytes and osteoblasts. Cbfβf/fCollagen, type 1, alpha 1 (Col1α1)–Cre mice show decreased bone mineralization and skeletal deformities, but no radical deformities in teeth, mandibles, or cartilage, indicating that osteoblast lineage-specific ablation of Cbfβ results in milder bone defects and less resemblance to CCD. Activating transcription factor 4 (Atf4) and Osterix protein levels in both mutant mice are dramatically reduced. ChIP assays show that Cbfβ directly associates with the promoter regions of Atf4 and Osterix. Our data further demonstrate that Cbfβ highly up-regulates the expression of Atf4 at the transcriptional regulation level. Overall, our genetic dissection approach revealed that Cbfβ plays an indispensable role in postnatal skeletal development and homeostasis in various skeletal cell types, at least partially by up-regulating the expression of Atf4 and Osterix. It also revealed that CCD may result from functional defects of the Runx2/Cbfβ heterodimeric complex in various skeletal cells. These insights into the role of Cbfβ in postnatal skeletogenesis and CCD pathogenesis may assist in the development of new therapies for CCD and osteoporosis.


Journal of Bone and Mineral Research | 2014

Core Binding Factor Beta (Cbfβ) Controls the Balance of Chondrocyte Proliferation and Differentiation by Upregulating Indian hedgehog (Ihh) Expression and Inhibiting Parathyroid Hormone-Related Protein Receptor (PPR) Expression in Postnatal Cartilage and Bone Formation

Fei Tian; Mengrui Wu; Lianfu Deng; Guochun Zhu; Junqing Ma; Bo Gao; Lin Wang; Yi-Ping Li; Wei Chen

Core binding factor beta (Cbfβ) is essential for embryonic bone morphogenesis. Yet the mechanisms by which Cbfβ regulates chondrocyte proliferation and differentiation as well as postnatal cartilage and bone formation remain unclear. Hence, using paired‐related homeobox transcription factor 1‐Cre (Prx1‐Cre) mice, mesenchymal stem cell–specific Cbfβ‐deficient (Cbfβf/fPrx1‐Cre) mice were generated to study the role of Cbfβ in postnatal cartilage and bone development. These mutant mice survived to adulthood but exhibited severe sternum and limb malformations. Sternum ossification was largely delayed in the Cbfβf/fPrx1‐Cre mice and the xiphoid process was noncalcified and enlarged. In newborn and 7‐day‐old Cbfβf/fPrx1‐Cre mice, the resting zone was dramatically elongated, the proliferation zone and hypertrophic zone of the growth plates were drastically shortened and disorganized, and trabecular bone formation was reduced. Moreover, in 1‐month‐old Cbfβf/fPrx1‐Cre mice, the growth plates were severely deformed and trabecular bone was almost absent. In addition, Cbfβ deficiency impaired intramembranous bone formation both in vivo and in vitro. Interestingly, although the expression of Indian hedgehog (Ihh) was largely reduced, the expression of parathyroid hormone‐related protein (PTHrP) receptor (PPR) was dramatically increased in the Cbfβf/fPrx1‐Cre growth plate, indicating that that Cbfβ deficiency disrupted the Ihh‐PTHrP negative regulatory loop. Chromatin immunoprecipitation (ChIP) analysis and promoter luciferase assay demonstrated that the Runx/Cbfβ complex binds putative Runx‐binding sites of the Ihh promoter regions, and also the Runx/Cbfβ complex directly upregulates Ihh expression at the transcriptional level. Consistently, the expressions of Ihh target genes, including CyclinD1, Ptc, and Pthlh, were downregulated in Cbfβ‐deficient chondrocytes. Taken together, our study reveals not only that Cbfβ is essential for chondrocyte proliferation and differentiation for the growth and maintenance of the skeleton in postnatal mice, but also that it functions in upregulating Ihh expression to promoter chondrocyte proliferation and osteoblast differentiation, and inhibiting PPR expression to enhance chondrocyte differentiation.


Infection and Immunity | 2013

RNA Interference-Mediated Silencing of Atp6i Prevents Both Periapical Bone Erosion and Inflammation in the Mouse Model of Endodontic Disease

Junqing Ma; Wei Chen; Lijie Zhang; Byron Tucker; Guochun Zhu; Hajime Sasaki; Liang Hao; Lin Wang; Hongliang Ci; Hongbing Jiang; Philip Stashenko; Yi-Ping Li

ABSTRACT Dental caries is one of the most prevalent infectious diseases in the United States, affecting approximately 80% of children and the majority of adults. Dental caries may lead to endodontic disease, where the bacterial infection progresses to the root canal system of the tooth, leading to periapical inflammation, bone erosion, severe pain, and tooth loss. Periapical inflammation may also exacerbate inflammation in other parts of the body. Although conventional clinical therapies for this disease are successful in approximately 80% of cases, there is still an urgent need for increased efficacy of treatment. In this study, we applied a novel gene-therapeutic approach using recombinant adeno-associated virus (AAV)-mediated Atp6i RNA interference (RNAi) knockdown of Atp6i/TIRC7 gene expression to simultaneously target periapical bone resorption and periapical inflammation. We found that Atp6i inhibition impaired osteoclast function in vitro and in vivo and decreased the number of T cells in the periapical lesion. Notably, AAV-mediated Atp6i/TIRC7 knockdown gene therapy reduced bacterial infection-stimulated bone resorption by 80% in the mouse model of endodontic disease. Importantly, Atp6i +/− mice with haploinsufficiency of Atp6i exhibited protection similar to that in mice with bacterial infection-stimulated bone erosion and periapical inflammation, which confirms the potential therapeutic effect of AAV-small hairpin RNA (shRNA)-Atp6i/TIRC7. Our results demonstrate that AAV-mediated Atp6i/TIRC7 knockdown in periapical tissues can inhibit endodontic disease development, bone resorption, and inflammation, indicating for the first time that this potential gene therapy may significantly improve the health of those who suffer from endodontic disease.


PLOS ONE | 2013

RNAi-Mediated Silencing of Atp6i and Atp6i Haploinsufficiency Prevents Both Bone Loss and Inflammation in a Mouse Model of Periodontal Disease

Hongbing Jiang; Wei Chen; Guochun Zhu; Lijie Zhang; Byron Tucker; Liang Hao; Shengmei Feng; Hongliang Ci; Junqing Ma; Lin Wang; Philip Stashenko; Yi-Ping Li

Periodontal disease affects about 80% of adults in America, and is characterized by oral bacterial infection-induced gingival inflammation, oral bone resorption, and tooth loss. Periodontitis is also associated with other diseases such as rheumatoid arthritis, diabetes, and heart disease. Although many efforts have been made to develop effective therapies for this disease, none have been very effective and there is still an urgent need for better treatments and preventative strategies. Herein we explored for the first time the possibility that adeno-associated virus (AAV)-mediated RNAi knockdown could be used to treat periodontal disease with improved efficacy. For this purpose, we used AAV-mediated RNAi knockdown of Atp6i/TIRC7 gene expression to target bone resorption and gingival inflammation simultaneously. Mice were infected with the oral pathogen Porphyromonas gingivalis W50 (P. gingivalis) in the maxillary periodontium to induce periodontitis. We found that Atp6i depletion impaired extracellular acidification and osteoclast-mediated bone resorption. Furthermore, local injection of AAV-shRNA-Atp6i/TIRC7 into the periodontal tissues in vivo protected mice from P. gingivalis infection-stimulated bone resorption by >85% and decreased the T-cell number in periodontal tissues. Notably, AAV-mediated Atp6i/TIRC7 knockdown also reduced the expression of osteoclast marker genes and inflammation-induced cytokine genes. Atp6i+/− mice with haploinsufficiency were similarly protected from P. gingivalis infection-stimulated bone loss and gingival inflammation. This suggests that AAV-shRNA-Atp6i/TIRC7 therapeutic treatment may significantly improve the health of millions who suffer from P. gingivalis-mediated periodontal disease.


Journal of Molecular Histology | 2017

GATA4 regulates osteoblastic differentiation and bone remodeling via p38-mediated signaling

Tingting Zhou; Shuyu Guo; Yuxin Zhang; Yajuan Weng; Lin Wang; Junqing Ma

Osteoblasts play a major role in bone remodeling and are regulated by transcription factors. GATA4, a zinc finger transcription factor from the GATA family, has an unclear role in osteoblast differentiation. In this study, the role of GATA4 in osteoblast differentiation was studied both in vitro and in vivo by GATA4 knockdown. GATA4 expression increased during osteoblast differentiation. GATA4 knockdown in osteoblast precursor cells reduced alkaline phosphatase activity and decreased the formation of calcified nodule in an osteogenic-induced cell culture system. In vivo, micro-CT showed that local injection of lentivirus-delivered GATA4 shRNA caused reduced new bone formation during tooth movement. Histological analyses such as total collagen and Goldner’s trichrome staining confirmed these results. In vivo immunohistochemical analysis showed reduced expression of osterix (OSX), osteopontin (OPN), and osteocalcin (OCN) in the shGATA4 group (Pu2009<u20090.05). Consistently, both western blotting and quantitative reverse-transcription PCR proved that expression of osteogenesis-related genes, including OSX, OPN, and OCN, was significantly repressed in the shGATA4 group in vitro (Pu2009<u20090.01). For further analysis of the pathways involved in this process, we examined the MAPK signaling pathway, and found knockdown of GATA4, downregulated p38 signaling pathways (Pu2009<u20090.01). Collectively, these results imply GATA4 is a regulator of osteoblastic differentiation via the p38 signaling pathways.


Journal of Molecular Histology | 2017

Blockade of LGR4 inhibits proliferation and odonto/osteogenic differentiation of stem cells from apical papillae

Meng Zhou; Shuyu Guo; Lichan Yuan; Yuxin Zhang; Mengnan Zhang; Huimin Chen; Mengting Lu; Jianrong Yang; Junqing Ma

During tooth root development, stem cells from apical papillae (SCAPs) are indispensable, and their abilities of proliferation, migration and odontoblast differentiation are linked to root formation. Leucine-rich repeat-containing GPCR 4 (LGR4) modulates the biological processes of proliferation and differentiation in multiple stem cells. In this study, we showed that LGR4 is expressed in all odontoblast cell lineage cells and Hertwig’s epithelial root sheath (HERS) during the mouse root formation in vivo. In vitro we determined that LGR4 is involved in the Wnt/β-catenin signaling pathway regulating proliferation and odonto/osteogenic differentiation of SCAPs. Quantitative reverse-transcription PCR (qRT-PCR) confirmed that LGR4 is expressed during odontogenic differentiation of SCAPs. CCK8 assays and in vitro scratch tests, together with cell cycle flow cytometric analysis, demonstrated that downregulation of LGR4 inhibited SCAPs proliferation, delayed migration and arrested cell cycle progression at the S and G2/M phases. ALP staining revealed that blockade of LGR4 decreased ALP activity. QRT-PCR and Western blot analysis demonstrated that LGR4 silencing reduced the expression of odonto/osteogenic markers (RUNX2, OSX, OPN, OCN and DSPP). Further Western blot and immunofluorescence studies clarified that inhibition of LGR4 disrupted β-catenin stabilization. Taken together, downregulation of LGR4 gene expression inhibited SCAPs proliferation, migration and odonto/osteogenic differentiation by blocking the Wnt/β-catenin signaling pathway. These results indicate that LGR4 might play a vital role in SCAPs proliferationxa0and odontoblastic differentiation.


Artificial Cells Nanomedicine and Biotechnology | 2018

Injectable calcium phosphate scaffold with iron oxide nanoparticles to enhance osteogenesis via dental pulp stem cells

Yang Xia; Huimin Chen; Feimin Zhang; Lin Wang; Bo Chen; Mark A. Reynolds; Junqing Ma; Abraham Schneider; Ning Gu; Hockin H.K. Xu

Abstract Literature search revealed no systematic report on iron oxide nanoparticle-incorporating calcium phosphate cement scaffolds (IONP-CPC). The objectives of this study were to: (1) use γFe2O3 nanoparticles (γIONPs) and αFe2O3 nanoparticles (αIONPs) to develop novel IONP-CPC scaffolds, and (2) investigate human dental pulp stem cells (hDPSCs) seeding on IONP-CPC for bone tissue engineering for the first time. IONP-CPC scaffolds were fabricated. Physiochemical properties of IONP-CPC scaffolds were characterized. hDPSC seeding on scaffolds, cell proliferation, osteogenic differentiation and bone matrix mineral synthesis by cells were measured. Our data demonstrated that the osteogenic differentiation of hDPSCs was markedly enhanced via IONP incorporation into CPC. Substantial increases (about three folds) in ALP activity and osteogenic gene expressions were achieved over those without IONPs. Bone matrix mineral synthesis by the cells was increased by two- to three folds over that without IONPs. The enhanced cellular osteogenesis was attributed to: (1) the surface nanotopography of IONP-CPC scaffold, and (2) the cell internalization of IONPs released from IONP-CPC scaffold. Our results demonstrate that the novel CPC functionalized with IONPs is promising to promote osteoinduction and bone regeneration. In conclusion, it is highly promising to incorporate γIONPs and αIONPs into CPC scaffold for bone tissue engineering, yielding substantially better stem cell attachment, spreading and osteogenic differentiation, and much greater bone mineral synthesis by the seeded cells. Therefore, novel CPC scaffolds containing γIONPs and αIONPs are promising for dental, craniofacial and orthopaedic applications to substantially enhance bone regeneration.


International Journal of Biological Sciences | 2016

Class A Scavenger Receptor Exacerbates Osteoclastogenesis by an Interleukin-6-Mediated Mechanism through ERK and JNK Signaling Pathways

Shuyu Guo; Yuanyuan Ni; Jingjing Ben; Yang Xia; Tingting Zhou; Dongyue Wang; Jieli Ni; Hui Bai; Lin Wang; Junqing Ma; Qi Chen

Osteoclasts originate from bone marrow monocyte/macrophage lineage cells, which are important for bone health. Class A scavenger receptor (SR-A) is a multifunctional molecule that functions during differentiation of monocyte into macrophages and osteoclasts. To further characterize the role of SR-A in osteoclasts, we used the murine tooth movement model (TM) and the murine anterior cruciate ligament transection model of osteoarthritis (ACLT OA). In these two models the bones involved are of different origin and have different properties. Bone resorption was decreased in SR-A-/- mice compared to SR-A+/+ mice. Further evaluation showed that the number of multinucleated osteoclasts in SR-A-/- mice, compared to SR-A+/+ mice, was significantly decreased both in vivo and in vitro. The levels of interleukin-6 (IL-6) produced by osteoclasts were reduced in SR-A-/- mice compared to SR-A+/+ mice. In the in vitro marrow-derived osteoclast formation assay and in both mouse models, osteoclastogenesis was restored to normal in SR-A-/- mice by administration of recombinant murine IL-6. Moreover, neutralization of IL-6 reduced the number of osteoclasts formed in SR-A+/+ mice of TM model. Both extracellular signal-regulated kinase (ERK) and c-Jun N-terminal protein kinase (JNK), but not p38, signaling pathways were downregulated in receptor activator of nuclear factor-κB ligand (RANKL)-stimulated SR-A-/- osteoclasts. Importantly, when treated with either ERK or JNK inhibitor, the numbers of osteoclasts generated from RANKL-induced bone marrow derived-macrophages of SR-A+/+ mice, and their IL-6 production, were significantly decreased. This suggests that SR-A activates the ERK and JNK signaling pathways, and promotes production of IL-6 by osteoclasts to further stimulate osteoclast formation.


Angle Orthodontist | 2016

Effects of nanostructured, diamondlike, carbon coating and nitrocarburizing on the frictional properties and biocompatibility of orthodontic stainless steel wires

Hao Zhang; Shuyu Guo; Dongyue Wang; Tingting Zhou; Lin Wang; Junqing Ma

OBJECTIVEnTo evaluate and compare the effects of nanostructured, diamondlike, carbon (DLC) coating and nitrocarburizing on the frictional properties and biocompatibility of orthodontic stainless steel archwires.nnnMATERIALS AND METHODSnPlasma-enhanced chemical vapor deposition technology was applied to coat DLC films onto the surface of austenitic stainless steel wires, and salt-bath nitrocarburizing technology was employed to achieve surface hardening of other wires. Surface and cross-sectional characteristics, microhardness, modulus of elasticity, friction resistance, corrosion resistance, and cell toxicity of the modified and control wires were analyzed.nnnRESULTSnThe surfaces of the DLC-coated and nitrocarburized wires were both smooth and even. Compared with the control, the DLC-coated wires were increased in surface hardness 1.46 times, decreased in elastic modulus, reduced in kinetic friction coefficient by 40.71%, and decreased in corrosion current density by two orders of magnitude. The nitrocarburized wire was increased in surface hardness 2.39 times, exhibited an unchanged elastic modulus, demonstrated a decrease in maximum static friction force of 22.2%, and rose in corrosion current density two orders of magnitude. Cytotoxicity tests revealed no significant toxicity associated with the modified wires.nnnCONCLUSIONSnDLC coating and nitrocarburizing significantly improved the surface hardness of the wires, reduced friction, and exhibited good biocompatibility. The nanostructured DLC coating provided excellent corrosion resistance and good elasticity, and while the nitrocarburizing technique substantially improved frictional properties, it reduced the corrosion resistance of the stainless steel wires to a lesser extent.


Scientific Reports | 2017

Role of GATA binding protein 4 (GATA4) in the regulation of tooth development via GNAI3

Shuyu Guo; Yuxin Zhang; Tingting Zhou; Dongyue Wang; Yajuan Weng; Lin Wang; Junqing Ma

Transcription factor GATA4 regulates cardiac and osteoblast differentiation. However, its role in tooth development is not clear. Therefore, we generated Wnt1-Cre;GATA4fl/fl mice, with conditional inactivation of the GATA4 gene in the dental papilla mesenchymal cells. Phenotypic analysis showed short root deformity along with reduced expressions of odonto/osteogenic markers. Proliferation (but not apoptosis) of cells around the apical area of the root was attenuated. In vitro, we knocked down GATA4 expression in stem cells of dental apical papilla (SCAPs). Proliferation, migration and odonto/osteogenic differentiation of SCAPs were affected in the shGATA4 group. Overexpression of GATA4 in SCAPs increased mineralization. Based on our previous iTRAQ results, guanine nucleotide binding proteins 3 (GNAI3) is one of the distinct proteins after GATA4 deletion. G protein signaling is involved in bone development, remodeling, and disease. In this study, both GATA4 deletion in the mouse root and knock-down in human SCAPs decreased the expression of GNAI3. Dual-luciferase and ChIP assay confirmed the direct binding of GATA4 to the GNAI3 promoter, both in vitro and in vivo. GNAI3 knock-down significantly decreased the odonto/osteogenic differentiation ability of SCAPs. We thus establish the role of GATA4 as a novel regulator of root development and elucidate its downstream molecular events.

Collaboration


Dive into the Junqing Ma's collaboration.

Top Co-Authors

Avatar

Lin Wang

Nanjing Medical University

View shared research outputs
Top Co-Authors

Avatar

Shuyu Guo

Nanjing Medical University

View shared research outputs
Top Co-Authors

Avatar

Tingting Zhou

Nanjing Medical University

View shared research outputs
Top Co-Authors

Avatar

Dongyue Wang

Nanjing Medical University

View shared research outputs
Top Co-Authors

Avatar

Yuxin Zhang

Nanjing Medical University

View shared research outputs
Top Co-Authors

Avatar

Yi-Ping Li

University of Alabama at Birmingham

View shared research outputs
Top Co-Authors

Avatar

Yajuan Weng

Nanjing Medical University

View shared research outputs
Top Co-Authors

Avatar

Guochun Zhu

University of Alabama at Birmingham

View shared research outputs
Top Co-Authors

Avatar

Wei Chen

University of Alabama at Birmingham

View shared research outputs
Top Co-Authors

Avatar

Yang Xia

University of Maryland

View shared research outputs
Researchain Logo
Decentralizing Knowledge