1
|
Qi L, Xiao L, Fu R, Nie Q, Zhang X, Luo W. Genetic characteristics and selection signatures between Southern Chinese local and commercial chickens. Poult Sci 2024; 103:103863. [PMID: 38810566 PMCID: PMC11166977 DOI: 10.1016/j.psj.2024.103863] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2024] [Revised: 04/25/2024] [Accepted: 05/13/2024] [Indexed: 05/31/2024] Open
Abstract
The introduction of exotic breeds and the cultivation of new lines by breeding companies have posed challenges to native chickens in South China, including loss of breed characteristics, decreased genetic diversity, and declining purity. Understanding the population genetic structure and genetic diversity of native chickens in South China is crucial for further advancements in breeding efforts. In this study, we analyzed the population genetic structure and genetic diversity of 321 individuals from 10 different breeds in South China. By comparing commercial chickens with native ones, we identified selection signatures occurring between local chickens and commercial breeds. The analysis of population genetic structure revealed that the native chicken populations in South China exhibited a considerable level of genetic diversity. Moreover, the commercial lines of Xiaobai chicken and Huangma chicken displayed even higher levels of genetic diversity, which distinguished them from other native varieties at the clustering level. However, certain individuals within these commercial varieties showed a discernible genetic relationship with the native populations. Notably, both commercial varieties also retained a significant degree of genetic similarity to their respective native counterparts. In order to investigate the genomic changes occurring during the commercialization of native chickens, we employed 4 methods (Fst, ROD, XPCLR, and XPEHH) to identify potential candidate regions displaying selective signatures in Southern Chinese native chicken population. A total of 168 (identified by Fst and ROD) and 86 (identified by XPCLR and XPEHH) overlapping genes were discovered. Functional annotation analysis revealed that these genes may be associated with reproduction and growth (SAMSN1, HYLS1, ROBO3, FGF14, PRSS23), musculoskeletal development (DNER, MYBPC1, DGKB, ORC1, KLF10), disease resistance and environmental adaptability (PUS3, CRB2, CALD1, USP15, SGCD, LTBP1), as well as egg production (ADGRB3, ACSF3). Overall, native chickens in South China harbor numerous selective sweep regions compared to commercial chickens, enriching valuable genomic resources for future genetic research and breeding conservation.
Collapse
Affiliation(s)
- Lin Qi
- State Key Laboratory of Livestock and Poultry Breeding, & Lingnan Guangdong Laboratory of Agriculture, South China Agricultural University, Guangzhou 510642, China; Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding, and Key Lab of Chicken Genetics, Breeding and Reproduction, Ministry of Agriculture and Rural Affair, South China Agricultural University, 510642, China; Department of Animal Genetics, Breeding and Reproduction, College of Animal Science, South China Agricultural University, Guangzhou Guangzhou 510642, China
| | - Liangchao Xiao
- State Key Laboratory of Livestock and Poultry Breeding, & Lingnan Guangdong Laboratory of Agriculture, South China Agricultural University, Guangzhou 510642, China; Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding, and Key Lab of Chicken Genetics, Breeding and Reproduction, Ministry of Agriculture and Rural Affair, South China Agricultural University, 510642, China; Department of Animal Genetics, Breeding and Reproduction, College of Animal Science, South China Agricultural University, Guangzhou Guangzhou 510642, China
| | - Rong Fu
- State Key Laboratory of Livestock and Poultry Breeding, & Lingnan Guangdong Laboratory of Agriculture, South China Agricultural University, Guangzhou 510642, China; Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding, and Key Lab of Chicken Genetics, Breeding and Reproduction, Ministry of Agriculture and Rural Affair, South China Agricultural University, 510642, China; Department of Animal Genetics, Breeding and Reproduction, College of Animal Science, South China Agricultural University, Guangzhou Guangzhou 510642, China
| | - Qinghua Nie
- State Key Laboratory of Livestock and Poultry Breeding, & Lingnan Guangdong Laboratory of Agriculture, South China Agricultural University, Guangzhou 510642, China; Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding, and Key Lab of Chicken Genetics, Breeding and Reproduction, Ministry of Agriculture and Rural Affair, South China Agricultural University, 510642, China; Department of Animal Genetics, Breeding and Reproduction, College of Animal Science, South China Agricultural University, Guangzhou Guangzhou 510642, China
| | - Xiquan Zhang
- State Key Laboratory of Livestock and Poultry Breeding, & Lingnan Guangdong Laboratory of Agriculture, South China Agricultural University, Guangzhou 510642, China; Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding, and Key Lab of Chicken Genetics, Breeding and Reproduction, Ministry of Agriculture and Rural Affair, South China Agricultural University, 510642, China; Department of Animal Genetics, Breeding and Reproduction, College of Animal Science, South China Agricultural University, Guangzhou Guangzhou 510642, China
| | - Wen Luo
- State Key Laboratory of Livestock and Poultry Breeding, & Lingnan Guangdong Laboratory of Agriculture, South China Agricultural University, Guangzhou 510642, China; Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding, and Key Lab of Chicken Genetics, Breeding and Reproduction, Ministry of Agriculture and Rural Affair, South China Agricultural University, 510642, China; Department of Animal Genetics, Breeding and Reproduction, College of Animal Science, South China Agricultural University, Guangzhou Guangzhou 510642, China.
| |
Collapse
|
2
|
Kandarpa M, Robinson D, Wu YM, Qin T, Pettit K, Li Q, Luker G, Sartor M, Chinnaiyan A, Talpaz M. Broad Next-Generation Integrated Sequencing of Myelofibrosis Identifies Disease-Specific and Age-Related Genomic Alterations. Clin Cancer Res 2024; 30:1972-1983. [PMID: 38386293 PMCID: PMC11061602 DOI: 10.1158/1078-0432.ccr-23-0372] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Revised: 05/18/2023] [Accepted: 02/20/2024] [Indexed: 02/23/2024]
Abstract
PURPOSE Myeloproliferative neoplasms (MPN) are characterized by the overproduction of differentiated myeloid cells. Mutations in JAK2, CALR, and MPL are considered drivers of Bcr-Abl-ve MPN, including essential thrombocythemia (ET), polycythemia vera (PV), prefibrotic primary myelofibrosis (prePMF), and overt myelofibrosis (MF). However, how these driver mutations lead to phenotypically distinct and/or overlapping diseases is unclear. EXPERIMENTAL DESIGN To compare the genetic landscape of MF to ET/PV/PrePMF, we sequenced 1,711 genes for mutations along with whole transcriptome RNA sequencing of 137 patients with MPN. RESULTS In addition to driver mutations, 234 and 74 genes were found to be mutated in overt MF (N = 106) and ET/PV/PrePMF (N = 31), respectively. Overt MF had more mutations compared with ET/PV/prePMF (5 vs. 4 per subject, P = 0.006). Genes frequently mutated in MF included high-risk genes (ASXL1, SRSF2, EZH2, IDH1/2, and U2AF1) and Ras pathway genes. Mutations in NRAS, KRAS, SRSF2, EZH2, IDH2, and NF1 were exclusive to MF. Advancing age, higher DIPSS, and poor overall survival (OS) correlated with increased variants in MF. Ras mutations were associated with higher leukocytes and platelets and poor OS. The comparison of gene expression showed upregulation of proliferation and inflammatory pathways in MF. Notably, ADGRL4, DNASE1L3, PLEKHGB4, HSPG2, MAMDC2, and DPYSL3 were differentially expressed in hematopoietic stem and differentiated cells. CONCLUSIONS Our results illustrate that evolution of MF from ET/PV/PrePMF likely advances with age, accumulation of mutations, and activation of proliferative pathways. The genes and pathways identified by integrated genomics approach provide insight into disease transformation and progression and potential targets for therapeutic intervention.
Collapse
Affiliation(s)
- Malathi Kandarpa
- Division of Hematology/Oncology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan
| | - Dan Robinson
- Michigan Center for Translational Pathology, University of Michigan Medical School, Ann Arbor, Michigan
- Department of Pathology, University of Michigan Medical School, Ann Arbor, Michigan
| | - Yi-Mi Wu
- Michigan Center for Translational Pathology, University of Michigan Medical School, Ann Arbor, Michigan
- Department of Pathology, University of Michigan Medical School, Ann Arbor, Michigan
| | - Tingting Qin
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, Michigan
| | - Kristen Pettit
- Division of Hematology/Oncology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan
| | - Qing Li
- Division of Hematology/Oncology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan
- Department of Cell and Developmental Biology, University of Michigan, Ann Arbor, Michigan
| | - Gary Luker
- Department of Radiology, University of Michigan, Ann Arbor, Michigan
| | - Maureen Sartor
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, Michigan
| | - Arul Chinnaiyan
- Michigan Center for Translational Pathology, University of Michigan Medical School, Ann Arbor, Michigan
- Department of Pathology, University of Michigan Medical School, Ann Arbor, Michigan
- Department of Urology, University of Michigan Medical School, Ann Arbor, Michigan
| | - Moshe Talpaz
- Division of Hematology/Oncology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan
| |
Collapse
|
3
|
Peng R, Shang J, Jiang N, Chi-Jen H, Gu Y, Xing B, Hu R, Wu B, Wang D, Xu X, Lu H. Klf10 is involved in extracellular matrix calcification of chondrocytes alleviating chondrocyte senescence. J Transl Med 2024; 22:52. [PMID: 38217021 PMCID: PMC10790269 DOI: 10.1186/s12967-023-04666-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2023] [Accepted: 10/27/2023] [Indexed: 01/14/2024] Open
Abstract
Osteoarthritis (OA) is a chronic degenerative disease resulting joint disability and pain. Accumulating evidences suggest that chondrocyte extracellular matrix calcification plays an important role in the development of OA. Here, we showed that Krüppel-like factor 10 (Klf10) was involved in the regulation of chondrocyte extracellular matrix calcification by regulating the expression of Frizzled9. Knockdown of Klf10 attenuated TBHP induced calcification and reduced calcium content in chondrocytes. Restoring extracellular matrix calcification of chondrocytes could aggravate chondrocyte senescence. Destabilization of a medial meniscus (DMM) mouse model of OA, in vivo experiments revealed that knockdown Klf10 improved the calcification of articular cartilage and ameliorated articular cartilage degeneration. These findings suggested that knockdown Klf10 inhibited extracellular matrix calcification-related changes in chondrocytes and alleviated chondrocyte senescence.
Collapse
Affiliation(s)
- Rong Peng
- Department of Orthopedics, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, 519000, Guangdong, China
- Guangdong Provincial Key Laboratory of Biomedical Imaging, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, 519000, Guangdong, China
| | - Jie Shang
- Department of Orthopedics, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, 519000, Guangdong, China
- Guangdong Provincial Key Laboratory of Biomedical Imaging, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, 519000, Guangdong, China
| | - Ning Jiang
- Department of Orthopedics, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, 519000, Guangdong, China
- Department of Orthopedics, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, 26400, Shandong, China
| | - Hsu Chi-Jen
- Department of Orthopedics, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, 519000, Guangdong, China
- Guangdong Provincial Key Laboratory of Biomedical Imaging, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, 519000, Guangdong, China
| | - Yu Gu
- Department of Orthopedics, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, 519000, Guangdong, China
- Guangdong Provincial Key Laboratory of Biomedical Imaging, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, 519000, Guangdong, China
| | - Baizhou Xing
- Department of Orthopedics, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, 519000, Guangdong, China
- Guangdong Provincial Key Laboratory of Biomedical Imaging, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, 519000, Guangdong, China
| | - Renan Hu
- Department of Orthopedics, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, 519000, Guangdong, China
- Guangdong Provincial Key Laboratory of Biomedical Imaging, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, 519000, Guangdong, China
| | - Biao Wu
- Department of Orthopedics, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, 519000, Guangdong, China
- Guangdong Provincial Key Laboratory of Biomedical Imaging, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, 519000, Guangdong, China
| | - Dawei Wang
- Department of Orthopedics, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, 519000, Guangdong, China.
- Guangdong Provincial Key Laboratory of Biomedical Imaging, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, 519000, Guangdong, China.
| | - Xianghe Xu
- Department of Orthopedics, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, 519000, Guangdong, China.
- Guangdong Provincial Key Laboratory of Biomedical Imaging, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, 519000, Guangdong, China.
| | - Huading Lu
- Department of Orthopedics, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, 519000, Guangdong, China.
- Guangdong Provincial Key Laboratory of Biomedical Imaging, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, 519000, Guangdong, China.
| |
Collapse
|
4
|
Huang L, Liang L, Ji Z, Chen S, Liu M, Huang Q, Huang Z, Sun S, Ding J, Chen J, Huang X, Zheng S, Deng W, Huang Y, Li T. Proteomics profiling of CD4 + T-cell-derived exosomes from patients with rheumatoid arthritis. Int Immunopharmacol 2023; 122:110560. [PMID: 37423153 DOI: 10.1016/j.intimp.2023.110560] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2023] [Revised: 06/17/2023] [Accepted: 06/22/2023] [Indexed: 07/11/2023]
Abstract
OBJECTIVES Our study profiled the CD4 + T-cell-derived exosomes from patients with rheumatoid arthritis (RA) using proteomics. METHODS Proteomic analysis of CD4 + T-cell-derived exosomes was performed by tandem mass tags (TMT) combined with LC-MS/MS. We validated the most significantly upregulated and downregulated proteins using ELISA and WB. RESULTS The proteomic results showed that there were 3 upregulated differentially expressed proteins and 31 downregulated differentially expressed proteins in the RA group. The results indicated that dihydropyrimidinase-related protein 3 (DPYSL3) was significantly upregulated in CD4 + T-cell-derived exosomes, whereas proteasome activator complex subunit 1 (PSME1) was significantly downregulated in the RA group. Bioinformatics analysis showed that proteins were enriched in "positive regulation of gene expression", "antigen processing and presentation", "acute-phase response" and "PI3K-AKT signaling" pathways. ELISA verified that compared to the control group, the RA group showed significant upregulation of DPYSL3, and downregulation of PSME1 in CD4 + T-cell-derived exosomes. CONCLUSIONS The proteomic analysis results of CD4 + T-cell-derived exosomes from patients with RA suggest that these differentially expressed proteins may be involved in RA pathogenesis. DPYSL3 and PSME1 may become useful biomarkers for RA.
Collapse
Affiliation(s)
- Lixin Huang
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou, China; Department of Rheumatology and Immunology, Guangdong Second Provincial General Hospital, Guangzhou, China
| | - Ling Liang
- The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Zhuyi Ji
- The Affiliated Guangdong Second Provincial General Hospital of Jinan University, Guangzhou, China
| | - Shuyang Chen
- Department of Rheumatology and Immunology, Guangdong Second Provincial General Hospital, Guangzhou, China
| | - Meng Liu
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou, China; Department of Rheumatology and Immunology, Guangdong Second Provincial General Hospital, Guangzhou, China
| | - Qidang Huang
- Department of Rheumatology and Immunology, Guangdong Second Provincial General Hospital, Guangzhou, China
| | - Zhixiang Huang
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou, China; Department of Rheumatology and Immunology, Guangdong Second Provincial General Hospital, Guangzhou, China
| | - Shanmiao Sun
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou, China; Department of Rheumatology and Immunology, Guangdong Second Provincial General Hospital, Guangzhou, China
| | - Jiali Ding
- Department of Rheumatology and Immunology, Guangdong Second Provincial General Hospital, Guangzhou, China
| | - Jiajun Chen
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou, China; Department of Rheumatology and Immunology, Guangdong Second Provincial General Hospital, Guangzhou, China
| | - Xuechan Huang
- Department of Rheumatology and Immunology, Guangdong Second Provincial General Hospital, Guangzhou, China
| | - Shaoling Zheng
- Department of Rheumatology and Immunology, Guangdong Second Provincial General Hospital, Guangzhou, China
| | - Weiming Deng
- Department of Rheumatology and Immunology, Guangdong Second Provincial General Hospital, Guangzhou, China.
| | - Yukai Huang
- Department of Rheumatology and Immunology, Guangdong Second Provincial General Hospital, Guangzhou, China.
| | - Tianwang Li
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou, China; Department of Rheumatology and Immunology, Guangdong Second Provincial General Hospital, Guangzhou, China; Department of Rheumatology and Immunology, Zhaoqing Central People's Hospital, Zhaoqing, China; The Affiliated Guangdong Second Provincial General Hospital of Jinan University, Guangzhou, China.
| |
Collapse
|
5
|
Kim NH, Jung SK, Lee J, Chang PS, Kang SH. Modulation of osteogenic differentiation by Escherichia coli-derived recombinant bone morphogenetic protein-2. AMB Express 2022; 12:106. [PMID: 35947236 PMCID: PMC9365917 DOI: 10.1186/s13568-022-01443-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2022] [Accepted: 07/28/2022] [Indexed: 11/10/2022] Open
Abstract
Recombinant human bone morphogenetic protein-2 (rhBMP-2), a key regulator of osteogenesis, induces the differentiation of mesenchymal cells into cartilage or bone tissues. Early orthopedic and dental studies often used mammalian cell-derived rhBMP-2, especially Chinese hamster ovary (CHO) cells. However, CHO cell-derived rhBMP-2 (C-rhBMP-2) presents disadvantages such as high cost and low production yield. To overcome these problems, Escherichia coli-derived BMP-2 (E-rhBMP-2) was developed; however, the E-rhBMP-2-induced signaling pathways and gene expression profiles during osteogenesis remain unclear. Here, we investigated the E-rhBMP-2-induced osteogenic differentiation pattern in C2C12 cells and elucidated the difference in biological characteristics between E-rhBMP-2 and C-rhBMP-2 via surface plasmon resonance, western blotting, qRT-PCR, RNA-seq, and alkaline phosphatase assays. The binding affinities of E-rhBMP-2 and C-rhBMP-2 towards BMP receptors were similar, both being confirmed at the nanomolecular level. However, the phosphorylation of Smad1/5/9 at 3 h after treatment with E-rhBMP-2 was significantly lower than that on treatment with C-rhBMP-2. The expression profiles of osteogenic marker genes were similar in both the E-rhBMP-2 and C-rhBMP-2 groups, but the gene expression level in the E-rhBMP-2 group was lower than that in the C-rhBMP-2 group at each time point. Taken together, our results suggest that the osteogenic signaling pathways induced by E-rhBMP-2 and C-rhBMP-2 both follow the general Smad-signaling pathway, but the difference in intracellular phosphorylation intensity results in distinguishable transcription profiles on osteogenic marker genes and biological activities of each rhBMP-2. These findings provide an extensive understanding of the biological properties of E-rhBMP-2 and the signaling pathways during osteogenic differentiation.
Collapse
Affiliation(s)
- Nam-Hyun Kim
- Life Science Institute, Daewoong Pharmaceutical, Yongin, Gyeonggido, Republic of Korea.,Department of Agricultural Biotechnology, Seoul National University, Seoul, Republic of Korea
| | - Seon-Kyong Jung
- Life Science Institute, Daewoong Pharmaceutical, Yongin, Gyeonggido, Republic of Korea
| | - Juno Lee
- Department of Agricultural Biotechnology, Seoul National University, Seoul, Republic of Korea
| | - Pahn-Shick Chang
- Department of Agricultural Biotechnology, Seoul National University, Seoul, Republic of Korea. .,Research Institute of Agriculture and Life Sciences, Seoul National University, Seoul, Republic of Korea. .,Center for Food and Bioconvergence, Seoul National University, Seoul, Republic of Korea. .,Center for Agricultural Microorganism and Enzyme, Seoul National University, Seoul, Republic of Korea.
| | - Seung-Hoon Kang
- Life Science Institute, Daewoong Pharmaceutical, Yongin, Gyeonggido, Republic of Korea.
| |
Collapse
|
6
|
Yadav PS, Feng S, Cong Q, Kim H, Liu Y, Yang Y. Stat3 loss in mesenchymal progenitors causes Job syndrome-like skeletal defects by reducing Wnt/β-catenin signaling. Proc Natl Acad Sci U S A 2021; 118:e2020100118. [PMID: 34172578 PMCID: PMC8256036 DOI: 10.1073/pnas.2020100118] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Job syndrome is a rare genetic disorder caused by STAT3 mutations and primarily characterized by immune dysfunction along with comorbid skeleton developmental abnormalities including osteopenia, recurrent fracture of long bones, and scoliosis. So far, there is no definitive cure for the skeletal defects in Job syndrome, and treatments are limited to management of clinical symptoms only. Here, we have investigated the molecular mechanism whereby Stat3 regulates skeletal development and osteoblast differentiation. We showed that removing Stat3 function in the developing limb mesenchyme or osteoprogenitor cells in mice resulted in shortened and bow limbs with multiple fractures in long bones that resembled the skeleton symptoms in the Job Syndrome. However, Stat3 loss did not alter chondrocyte differentiation and hypertrophy in embryonic development, while osteoblast differentiation was severely reduced. Genome-wide transcriptome analyses as well as biochemical and histological studies showed that Stat3 loss resulted in down-regulation of Wnt/β-catenin signaling. Restoration of Wnt/β-catenin signaling by injecting BIO, a small molecule inhibitor of GSK3, or crossing with a Lrp5 gain of function (GOF) allele, rescued the bone reduction phenotypes due to Stat3 loss to a great extent. These studies uncover the essential functions of Stat3 in maintaining Wnt/β-catenin signaling in early mesenchymal or osteoprogenitor cells and provide evidence that bone defects in the Job Syndrome are likely caused by Wnt/β-catenin signaling reduction due to reduced STAT3 activities in bone development. Enhancing Wnt/β-catenin signaling could be a therapeutic approach to reduce bone symptoms of Job syndrome patients.
Collapse
Affiliation(s)
- Prem Swaroop Yadav
- Department of Developmental Biology, Harvard School of Dental Medicine, Boston, MA 02115
| | - Shuhao Feng
- Department of Developmental Biology, Harvard School of Dental Medicine, Boston, MA 02115
| | - Qian Cong
- Department of Developmental Biology, Harvard School of Dental Medicine, Boston, MA 02115
| | - Hanjun Kim
- Department of Developmental Biology, Harvard School of Dental Medicine, Boston, MA 02115
| | - Yuchen Liu
- Department of Developmental Biology, Harvard School of Dental Medicine, Boston, MA 02115
| | - Yingzi Yang
- Department of Developmental Biology, Harvard School of Dental Medicine, Boston, MA 02115;
- Harvard Stem Cell Institute, Cambridge, MA 02138
| |
Collapse
|
7
|
Kammoun M, Piquereau J, Nadal‐Desbarats L, Même S, Beuvin M, Bonne G, Veksler V, Le Fur Y, Pouletaut P, Même W, Szeremeta F, Constans J, Bruinsma ES, Nelson Holte MH, Najafova Z, Johnsen SA, Subramaniam M, Hawse JR, Bensamoun SF. Novel role of Tieg1 in muscle metabolism and mitochondrial oxidative capacities. Acta Physiol (Oxf) 2020; 228:e13394. [PMID: 31560161 DOI: 10.1111/apha.13394] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2018] [Revised: 09/20/2019] [Accepted: 09/23/2019] [Indexed: 12/26/2022]
Abstract
AIM Tieg1 is involved in multiple signalling pathways, human diseases, and is highly expressed in muscle where its functions are poorly understood. METHODS We have utilized Tieg1 knockout (KO) mice to identify novel and important roles for this transcription factor in regulating muscle ultrastructure, metabolism and mitochondrial functions in the soleus and extensor digitorum longus (EDL) muscles. RNA sequencing, immunoblotting, transmission electron microscopy, MRI, NMR, histochemical and mitochondrial function assays were performed. RESULTS Loss of Tieg1 expression resulted in altered sarcomere organization and a significant decrease in mitochondrial number. Histochemical analyses demonstrated an absence of succinate dehydrogenase staining and a decrease in cytochrome c oxidase (COX) enzyme activity in KO soleus with similar, but diminished, effects in the EDL. Decreased complex I, COX and citrate synthase (CS) activities were detected in the soleus muscle of KO mice indicating altered mitochondrial function. Complex I activity was also diminished in KO EDL. Significant decreases in CS and respiratory chain complex activities were identified in KO soleus. 1 H-NMR spectra revealed no significant metabolic difference between wild-type and KO muscles. However, 31 P spectra revealed a significant decrease in phosphocreatine and ATPγ. Altered expression of 279 genes, many of which play roles in mitochondrial and muscle function, were identified in KO soleus muscle. Ultimately, all of these changes resulted in an exercise intolerance phenotype in Tieg1 KO mice. CONCLUSION Our findings have implicated novel roles for Tieg1 in muscle including regulation of gene expression, metabolic activity and organization of tissue ultrastructure. This muscle phenotype resembles diseases associated with exercise intolerance and myopathies of unknown consequence.
Collapse
Affiliation(s)
- Malek Kammoun
- Biomechanics and Bioengineering Laboratory Alliance Sorbonne Universités Université de Technologie de Compiègne UMR CNRS 7338 Compiègne France
| | - Jerome Piquereau
- Signalling and Cardiovascular Pathophysiology ‐ UMR‐S 1180 Université Paris‐Sud INSERM Université Paris‐Saclay Châtenay‐Malabry France
| | | | - Sandra Même
- CNRS UPR4301 Centre de Biophysique Moléculaire Orléans France
| | - Maud Beuvin
- Inserm U974 Centre de Recherche en Myologie Sorbonne Université Paris France
| | - Gisèle Bonne
- Inserm U974 Centre de Recherche en Myologie Sorbonne Université Paris France
| | - Vladimir Veksler
- Signalling and Cardiovascular Pathophysiology ‐ UMR‐S 1180 Université Paris‐Sud INSERM Université Paris‐Saclay Châtenay‐Malabry France
| | - Yann Le Fur
- Aix‐Marseille University CNRS CRMBM Marseille France
| | - Philippe Pouletaut
- Biomechanics and Bioengineering Laboratory Alliance Sorbonne Universités Université de Technologie de Compiègne UMR CNRS 7338 Compiègne France
| | - William Même
- CNRS UPR4301 Centre de Biophysique Moléculaire Orléans France
| | | | - Jean‐Marc Constans
- Institut Faire Faces EA Chimère Imagerie et Radiologie Médicale CHU Amiens Amiens France
| | | | | | - Zeynab Najafova
- Department of General, Visceral and Pediatric Surgery University Medical Center Göttingen Göttingen Germany
| | - Steven A. Johnsen
- Department of General, Visceral and Pediatric Surgery University Medical Center Göttingen Göttingen Germany
| | | | - John R. Hawse
- Department of Biochemistry and Molecular Biology Mayo Clinic Rochester MN USA
| | - Sabine F. Bensamoun
- Biomechanics and Bioengineering Laboratory Alliance Sorbonne Universités Université de Technologie de Compiègne UMR CNRS 7338 Compiègne France
| |
Collapse
|
8
|
Xu R, Khan SK, Zhou T, Gao B, Zhou Y, Zhou X, Yang Y. Gα s signaling controls intramembranous ossification during cranial bone development by regulating both Hedgehog and Wnt/β-catenin signaling. Bone Res 2018; 6:33. [PMID: 30479847 PMCID: PMC6242855 DOI: 10.1038/s41413-018-0034-7] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2018] [Revised: 09/11/2018] [Accepted: 09/27/2018] [Indexed: 02/05/2023] Open
Abstract
How osteoblast cells are induced is a central question for understanding skeletal formation. Abnormal osteoblast differentiation leads to a broad range of devastating craniofacial diseases. Here we have investigated intramembranous ossification during cranial bone development in mouse models of skeletal genetic diseases that exhibit craniofacial bone defects. The GNAS gene encodes Gαs that transduces GPCR signaling. GNAS activation or loss-of-function mutations in humans cause fibrous dysplasia (FD) or progressive osseous heteroplasia (POH) that shows craniofacial hyperostosis or craniosynostosis, respectively. We find here that, while Hh ligand-dependent Hh signaling is essential for endochondral ossification, it is dispensable for intramembranous ossification, where Gαs regulates Hh signaling in a ligand-independent manner. We further show that Gαs controls intramembranous ossification by regulating both Hh and Wnt/β-catenin signaling. In addition, Gαs activation in the developing cranial bone leads to reduced ossification but increased cartilage presence due to reduced cartilage dissolution, not cell fate switch. Small molecule inhibitors of Hh and Wnt signaling can effectively ameliorate cranial bone phenotypes in mice caused by loss or gain of Gnas function mutations, respectively. Our work shows that studies of genetic diseases provide invaluable insights in both pathological bone defects and normal bone development, understanding both leads to better diagnosis and therapeutic treatment of bone diseases.
Collapse
Affiliation(s)
- Ruoshi Xu
- 1Department of Developmental Biology, Harvard School of Dental Medicine, 188 Longwood Avenue, Boston, MA USA.,2State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Cariology and Endodontology, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Sanjoy Kumar Khan
- 1Department of Developmental Biology, Harvard School of Dental Medicine, 188 Longwood Avenue, Boston, MA USA
| | - Taifeng Zhou
- 1Department of Developmental Biology, Harvard School of Dental Medicine, 188 Longwood Avenue, Boston, MA USA.,3Department of Orthopaedic Surgery, Guangdong Provincial Key Laboratory of Orthopedics and Traumatology, First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Bo Gao
- 1Department of Developmental Biology, Harvard School of Dental Medicine, 188 Longwood Avenue, Boston, MA USA.,4Department of Spine Surgery, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Yaxing Zhou
- 1Department of Developmental Biology, Harvard School of Dental Medicine, 188 Longwood Avenue, Boston, MA USA
| | - Xuedong Zhou
- 2State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Cariology and Endodontology, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Yingzi Yang
- 1Department of Developmental Biology, Harvard School of Dental Medicine, 188 Longwood Avenue, Boston, MA USA
| |
Collapse
|
9
|
Chawla S, Sharma A, Bandyopadhyay A, Ghosh S. Developmental Biology-Inspired Strategies To Engineer 3D Bioprinted Bone Construct. ACS Biomater Sci Eng 2018; 4:3545-3560. [DOI: 10.1021/acsbiomaterials.8b00757] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Affiliation(s)
- Shikha Chawla
- Regenerative Engineering Laboratory, Department of Textile Technology, Indian Institute of Technology Delhi, New Delhi 110016, India
| | - Aarushi Sharma
- Regenerative Engineering Laboratory, Department of Textile Technology, Indian Institute of Technology Delhi, New Delhi 110016, India
| | - Amitabha Bandyopadhyay
- Department of Biological Sciences & Bioengineering (BSBE), Indian Institute of Technology Kanpur, Kanpur 208016, India
| | - Sourabh Ghosh
- Regenerative Engineering Laboratory, Department of Textile Technology, Indian Institute of Technology Delhi, New Delhi 110016, India
| |
Collapse
|
10
|
Cleves PA, Hart JC, Agoglia RM, Jimenez MT, Erickson PA, Gai L, Miller CT. An intronic enhancer of Bmp6 underlies evolved tooth gain in sticklebacks. PLoS Genet 2018; 14:e1007449. [PMID: 29902209 PMCID: PMC6019817 DOI: 10.1371/journal.pgen.1007449] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2017] [Revised: 06/26/2018] [Accepted: 05/25/2018] [Indexed: 12/30/2022] Open
Abstract
Threespine stickleback fish offer a powerful system to dissect the genetic basis of morphological evolution in nature. Marine sticklebacks have repeatedly invaded and adapted to numerous freshwater environments throughout the Northern hemisphere. In response to new diets in freshwater habitats, changes in craniofacial morphology, including heritable increases in tooth number, have evolved in derived freshwater populations. Using a combination of quantitative genetics and genome resequencing, here we fine-mapped a quantitative trait locus (QTL) regulating evolved tooth gain to a cluster of ten QTL-associated single nucleotide variants, all within intron four of Bone Morphogenetic Protein 6 (Bmp6). Transgenic reporter assays revealed this intronic region contains a tooth enhancer. We induced mutations in Bmp6, revealing required roles for survival, growth, and tooth patterning. Transcriptional profiling of Bmp6 mutant dental tissues identified significant downregulation of a set of genes whose orthologs were previously shown to be expressed in quiescent mouse hair stem cells. Collectively these data support a model where mutations within a Bmp6 intronic tooth enhancer contribute to evolved tooth gain, and suggest that ancient shared genetic circuitry regulates the regeneration of diverse vertebrate epithelial appendages including mammalian hair and fish teeth.
Collapse
Affiliation(s)
- Phillip A. Cleves
- Department of Molecular and Cell Biology, University of California-Berkeley, Berkeley CA, United States of America
| | - James C. Hart
- Department of Molecular and Cell Biology, University of California-Berkeley, Berkeley CA, United States of America
| | - Rachel M. Agoglia
- Department of Molecular and Cell Biology, University of California-Berkeley, Berkeley CA, United States of America
| | - Monica T. Jimenez
- Department of Molecular and Cell Biology, University of California-Berkeley, Berkeley CA, United States of America
| | - Priscilla A. Erickson
- Department of Molecular and Cell Biology, University of California-Berkeley, Berkeley CA, United States of America
| | - Linda Gai
- Department of Molecular and Cell Biology, University of California-Berkeley, Berkeley CA, United States of America
| | - Craig T. Miller
- Department of Molecular and Cell Biology, University of California-Berkeley, Berkeley CA, United States of America
| |
Collapse
|
11
|
Memon A, Lee WK. KLF10 as a Tumor Suppressor Gene and Its TGF-β Signaling. Cancers (Basel) 2018; 10:E161. [PMID: 29799499 PMCID: PMC6025274 DOI: 10.3390/cancers10060161] [Citation(s) in RCA: 52] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2018] [Revised: 05/15/2018] [Accepted: 05/23/2018] [Indexed: 12/17/2022] Open
Abstract
Krüppel-like factor 10 (KLF10), originally named TGF-β (Transforming growth factor beta) inducible early gene 1 (TIEG1), is a DNA-binding transcriptional regulator containing a triple C2H2 zinc finger domain. By binding to Sp1 (specificity protein 1) sites on the DNA and interactions with other regulatory transcription factors, KLF10 encourages and suppresses the expression of multiple genes in many cell types. Many studies have investigated its signaling cascade, but other than the TGF-β/Smad signaling pathway, these are still not clear. KLF10 plays a role in proliferation, differentiation as well as apoptosis, just like other members of the SP (specificity proteins)/KLF (Krüppel-like Factors). Recently, several studies reported that KLF10 KO (Knock out) is associated with defects in cell and organs such as osteopenia, abnormal tendon or cardiac hypertrophy. Since KLF10 was first discovered, several studies have defined its role in cancer as a tumor suppressor. KLF10 demonstrate anti-proliferative effects and induce apoptosis in various carcinoma cells including pancreatic cancer, leukemia, and osteoporosis. Collectively, these data indicate that KLF10 plays a significant role in various biological processes and diseases, but its role in cancer is still unclear. Therefore, this review was conducted to describe and discuss the role and function of KLF10 in diseases, including cancer, with a special emphasis on its signaling with TGF-β.
Collapse
Affiliation(s)
- Azra Memon
- Laboratory of Developmental Genetics, Department of Biomedical Sciences, School of Medicine, Inha University, Incheon 22212, Korea.
| | - Woon Kyu Lee
- Laboratory of Developmental Genetics, Department of Biomedical Sciences, School of Medicine, Inha University, Incheon 22212, Korea.
| |
Collapse
|
12
|
Kumar Y, Biswas T, Thacker G, Kanaujiya JK, Kumar S, Shukla A, Khan K, Sanyal S, Chattopadhyay N, Bandyopadhyay A, Trivedi AK. BMP signaling-driven osteogenesis is critically dependent on Prdx-1 expression-mediated maintenance of chondrocyte prehypetrophy. Free Radic Biol Med 2018; 118:1-12. [PMID: 29452246 DOI: 10.1016/j.freeradbiomed.2018.02.016] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/03/2017] [Revised: 01/29/2018] [Accepted: 02/10/2018] [Indexed: 10/18/2022]
Abstract
During endochondral ossification, cartilage template is eventually replaced by bone. This process involves several well characterized, stereotypic, molecular and cellular changes in the cartilage primordia. These steps involve transition from resting to proliferative and then pre-hypertrophic to finally hypertrophic cartilage. BMP signaling is necessary and sufficient for osteogenesis. However, the specific step(s) of endochondral ossification in which BMP signaling plays an essential role is not yet known. In this study we have identified Prdx1, a known scavenger of ROS, to be expressed in pre-hypertrophic chondrocytes in a BMP signaling-dependent manner. We demonstrate that BMP signaling inhibition increases ROS levels in osteogenic cells. Further, Prdx1 regulates osteogenesis in vivo by helping maintenance of Ihh expressing pre-hypertrophic cells, in turn regulating these cells' transition into hypertrophy. Therefore, our data suggests that one of the key roles of BMP signaling in endochondral ossification is to maintain pre-hypertrophic state.
Collapse
Affiliation(s)
- Yogesh Kumar
- Biochemistry Division, CSIR-Central Drug Research Institute (CSIR-CDRI), Sector-10, Jankipuram Extension, Lucknow 226031, UP, India
| | - Tathagata Biswas
- Department of Biological Sciences and Bioengineering, Indian Institute of Technology Kanpur, Kanpur 208016, India
| | - Gatha Thacker
- Biochemistry Division, CSIR-Central Drug Research Institute (CSIR-CDRI), Sector-10, Jankipuram Extension, Lucknow 226031, UP, India
| | - Jitendra Kumar Kanaujiya
- Biochemistry Division, CSIR-Central Drug Research Institute (CSIR-CDRI), Sector-10, Jankipuram Extension, Lucknow 226031, UP, India
| | - Sandeep Kumar
- Department of Biological Sciences and Bioengineering, Indian Institute of Technology Kanpur, Kanpur 208016, India
| | - Anukampa Shukla
- Biochemistry Division, CSIR-Central Drug Research Institute (CSIR-CDRI), Sector-10, Jankipuram Extension, Lucknow 226031, UP, India
| | - Kainat Khan
- Division of Endocrinology and Center for Research in Anabolic Skeletal Targets in Health and Illness (ASTHI), CSIR-Central Drug Research Institute (CSIR-CDRI), Sector-10, Jankipuram Extension, Lucknow 226031, UP, India
| | - Sabyasachi Sanyal
- Biochemistry Division, CSIR-Central Drug Research Institute (CSIR-CDRI), Sector-10, Jankipuram Extension, Lucknow 226031, UP, India
| | - Naibedya Chattopadhyay
- Division of Endocrinology and Center for Research in Anabolic Skeletal Targets in Health and Illness (ASTHI), CSIR-Central Drug Research Institute (CSIR-CDRI), Sector-10, Jankipuram Extension, Lucknow 226031, UP, India
| | - Amitabha Bandyopadhyay
- Department of Biological Sciences and Bioengineering, Indian Institute of Technology Kanpur, Kanpur 208016, India.
| | - Arun Kumar Trivedi
- Biochemistry Division, CSIR-Central Drug Research Institute (CSIR-CDRI), Sector-10, Jankipuram Extension, Lucknow 226031, UP, India.
| |
Collapse
|
13
|
Yang Y, Jiang Y, Xie D, Liu M, Song N, Zhu J, Fan J, Zhu C. Inhibition of cell-adhesion protein DPYSL3 promotes metastasis of lung cancer. Respir Res 2018; 19:41. [PMID: 29514686 PMCID: PMC5842641 DOI: 10.1186/s12931-018-0740-0] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2017] [Accepted: 02/15/2018] [Indexed: 01/05/2023] Open
Abstract
BACKGROUND Our previous screening study suggested that the cell-adhesions protein Dihydropyrimidinase-like 3 (DPYSL3) was a candidate metastatic lung cancer related molecule. This study aimed to analyze the correlation between DPYSL3 and metastatic lung cancer. METHODS Stable DPYSL3 knockdown Lewis lung carcinoma (LLC) cells were constructed with a retroviral system. Cell migration and invasion assays were performed to determine the role of DPYSL3 in LLC cells' migration and invasion changes. A metastatic lung tumor model in which the stable DPYSL3 knockdown LLC cells were injected through tail vein was used to analyze the role of DPYSL3 in tumor metastasis in vivo. The correlation between DPYSL3 expression and the survival time of lung cancer patients were analyzed in KMPLOT database. RESULTS Knockdown of DPYSL3 promoted the migratory and invasive of LLC cells compared to the control group. Meanwhile, the motility of LLC cells was also increased with the inhibition of DPYSL3. The TGFβ-induced EMT increased when DPYSL3 was inhibited. The expression of EMT markers, TWIST1 and N-cadherin, significantly increased to almost two times with the knockdown of DPYSL3. Furthermore, inhibition of DPYSL3 promoted the progression of metastatic xenograft in C57BL/6 mice. The expression level of DPYSL3 decreased in lung cancer patients with distant metastasis. CONCLUSIONS Knockdown of DPYSL3 promoted the metastatic ability of LLC cells in vitro and in vivo.
Collapse
Affiliation(s)
- Yang Yang
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital affiliated Tongji University, 507 Zhengmin road, Shanghai, 200433 China
| | - Yan Jiang
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital affiliated Tongji University, 507 Zhengmin road, Shanghai, 200433 China
| | - Dong Xie
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital affiliated Tongji University, 507 Zhengmin road, Shanghai, 200433 China
| | - Ming Liu
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital affiliated Tongji University, 507 Zhengmin road, Shanghai, 200433 China
| | - Nan Song
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital affiliated Tongji University, 507 Zhengmin road, Shanghai, 200433 China
| | - Junjie Zhu
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital affiliated Tongji University, 507 Zhengmin road, Shanghai, 200433 China
| | - Jiang Fan
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital affiliated Tongji University, 507 Zhengmin road, Shanghai, 200433 China
| | - Chenfang Zhu
- Department of General Surgery, Shanghai Ninth People’s Hospital, Shanghai JiaoTong University School of Medicine, Discipline Construction Research Center of China Hospital Development Institute, Shanghai Jiao Tong University, 639 Zhizaoju road, Shanghai, 200011 China
| |
Collapse
|
14
|
Saxena M, Agnihotri N, Sen J. Perturbation of canonical and non-canonical BMP signaling affects migration, polarity and dendritogenesis of mouse cortical neurons. Development 2018; 145:dev.147157. [PMID: 29180570 DOI: 10.1242/dev.147157] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2016] [Accepted: 11/16/2017] [Indexed: 01/07/2023]
Abstract
Bone morphogenetic protein (BMP) signaling has been implicated in the regulation of patterning of the forebrain and as a regulator of neurogenesis and gliogenesis in the mammalian cortex. However, its role in other aspects of cortical development in vivo remains unexplored. We hypothesized that BMP signaling might regulate additional processes during the development of cortical neurons after observing active BMP signaling in a spatiotemporally dynamic pattern in the mouse cortex. Our investigation revealed that BMP signaling specifically regulates the migration, polarity and the dendritic morphology of upper layer cortical neurons born at E15.5. On further dissection of the role of canonical and non-canonical BMP signaling in each of these processes, we found that migration of these neurons is regulated by both pathways. Their polarity, however, appears to be affected more strongly by canonical BMP signaling, whereas dendritic branch formation appears to be somewhat more strongly affected by LIMK-mediated non-canonical BMP signaling.
Collapse
Affiliation(s)
- Monika Saxena
- Department of Biological Sciences and Bioengineering, Indian Institute of Technology Kanpur, Kanpur-208016, Uttar Pradesh, India
| | - Nitin Agnihotri
- Department of Biological Sciences and Bioengineering, Indian Institute of Technology Kanpur, Kanpur-208016, Uttar Pradesh, India
| | - Jonaki Sen
- Department of Biological Sciences and Bioengineering, Indian Institute of Technology Kanpur, Kanpur-208016, Uttar Pradesh, India
| |
Collapse
|
15
|
Induced GnasR201H expression from the endogenous Gnas locus causes fibrous dysplasia by up-regulating Wnt/β-catenin signaling. Proc Natl Acad Sci U S A 2017; 115:E418-E427. [PMID: 29158412 DOI: 10.1073/pnas.1714313114] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
Fibrous dysplasia (FD; Online Mendelian Inheritance in Man no. 174800) is a crippling skeletal disease caused by activating mutations of the GNAS gene, which encodes the stimulatory G protein Gαs FD can lead to severe adverse conditions such as bone deformity, fracture, and severe pain, leading to functional impairment and wheelchair confinement. So far there is no cure, as the underlying molecular and cellular mechanisms remain largely unknown and the lack of appropriate animal models has severely hampered FD research. Here we have investigated the cellular and molecular mechanisms underlying FD and tested its potential treatment by establishing a mouse model in which the human FD mutation (R201H) has been conditionally knocked into the corresponding mouse Gnas locus. We found that the germ-line FD mutant was embryonic lethal, and Cre-induced Gnas FD mutant expression in early osteochondral progenitors, osteoblast cells, or bone marrow stromal cells (BMSCs) recapitulated FD features. In addition, mosaic expression of FD mutant Gαs in BMSCs induced bone marrow fibrosis both cell autonomously and non-cell autonomously. Furthermore, Wnt/β-catenin signaling was up-regulated in FD mutant mouse bone and BMSCs undergoing osteogenic differentiation, as we have found in FD human tissue previously. Reduction of Wnt/β-catenin signaling by removing one Lrp6 copy in an FD mutant line significantly rescued the phenotypes. We demonstrate that induced expression of the FD Gαs mutant from the mouse endogenous Gnas locus exhibits human FD phenotypes in vivo, and that inhibitors of Wnt/β-catenin signaling may be repurposed for treating FD and other bone diseases caused by Gαs activation.
Collapse
|
16
|
Baas R, van Teeffelen HAAM, Tjalsma SJD, Timmers HTM. The mixed lineage leukemia 4 (MLL4) methyltransferase complex is involved in transforming growth factor beta (TGF-β)-activated gene transcription. Transcription 2017; 9:67-74. [PMID: 28976802 PMCID: PMC5834223 DOI: 10.1080/21541264.2017.1373890] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
Sma and Mad related (SMAD)-mediated Transforming Growth Factor β (TGF-β) and Bone Morphogenetic Protein (BMP) signaling is required for various cellular processes. The activated heterotrimeric SMAD protein complexes associate with nuclear proteins such as the histone acetyltransferases p300, PCAF and the Mixed Lineage Leukemia 4 (MLL4) subunit Pax Transactivation domain-Interacting Protein (PTIP) to regulate gene transcription. We investigated the functional role of PTIP and PTIP Interacting protein 1 (PA1) in relation to TGF-β-activated SMAD signaling. We immunoprecipitated PTIP and PA1 with all SMAD family members to identify the TGF-β and not BMP-specific SMADs as interacting proteins. Gene silencing experiments of MLL4 and the subunits PA1 and PTIP confirm TGF-β-specific genes to be regulated by the MLL4 complex, which links TGF-β signaling to transcription regulation by the MLL4 methyltransferase complex.
Collapse
Affiliation(s)
- Roy Baas
- a Molecular Cancer Research and Stem Cells, Regenerative Medicine Center, Center for Molecular Medicine , University Medical Center Utrecht , Utrecht , The Netherlands
| | - Hetty A A M van Teeffelen
- a Molecular Cancer Research and Stem Cells, Regenerative Medicine Center, Center for Molecular Medicine , University Medical Center Utrecht , Utrecht , The Netherlands
| | - Sjoerd J D Tjalsma
- a Molecular Cancer Research and Stem Cells, Regenerative Medicine Center, Center for Molecular Medicine , University Medical Center Utrecht , Utrecht , The Netherlands
| | - H Th Marc Timmers
- a Molecular Cancer Research and Stem Cells, Regenerative Medicine Center, Center for Molecular Medicine , University Medical Center Utrecht , Utrecht , The Netherlands
| |
Collapse
|
17
|
Abdallah BM, Figeac F, Larsen KH, Ditzel N, Keshari P, Isa A, Jafari A, Andersen TL, Delaisse JM, Goshima Y, Ohshima T, Kassem M. CRMP4 Inhibits Bone Formation by Negatively Regulating BMP and RhoA Signaling. J Bone Miner Res 2017; 32:913-926. [PMID: 28019696 DOI: 10.1002/jbmr.3069] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/24/2016] [Revised: 12/18/2016] [Accepted: 12/21/2016] [Indexed: 11/10/2022]
Abstract
We identified the neuroprotein collapsing response mediator protein-4 (CRMP4) as a noncanonical osteogenic factor that regulates the differentiation of mouse bone marrow skeletal stem cells (bone marrow stromal stem cells [mBMSCs]) into osteoblastic cells. CRMP4 is the only member of the CRMP1-CRMP5 family to be expressed by mBMSCs and in osteoprogenitors of both adult mouse and human bones. In vitro gain-of-function and loss-of-function of CRMP4 in murine stromal cells revealed its inhibitory effect on osteoblast differentiation. In addition, Crmp4-deficient mice (Crmp4-/- ) displayed a 40% increase in bone mass, increased mineral apposition rate, and bone formation rate, compared to wild-type controls. Increased bone mass in Crmp4-/- mice was associated with enhanced BMP2 signaling and BMP2-induced osteoblast differentiation in Crmp4-/- osteoblasts (OBs). Furthermore, Crmp4-/- OBs exhibited enhanced activation of RhoA/focal adhesion kinase (FAK) signaling that led to cytoskeletal changes with increased cell spreading. In addition, Crmp4-/- OBs exhibited increased cell proliferation that was mediated via inhibiting cyclin-dependent kinase inhibitor 1B, p27Kip1 and upregulating cyclin D1 expression which are targets of RhoA signaling pathway. Our findings identify CRMP4 as a novel negative regulator of osteoblast differentiation. © 2016 American Society for Bone and Mineral Research.
Collapse
Affiliation(s)
- Basem M Abdallah
- Molecular Endocrinology Laboratory (KMEB), Department of Endocrinology, Odense University Hospital & University of Southern Denmark, Odense, Denmark.,Department of Biological Sciences, College of Science, King Faisal University, Hofuf, Saudi Arabia.,Faculty of Science, Helwan University, Cairo, Egypt
| | - Florence Figeac
- Molecular Endocrinology Laboratory (KMEB), Department of Endocrinology, Odense University Hospital & University of Southern Denmark, Odense, Denmark
| | - Kenneth H Larsen
- Molecular Endocrinology Laboratory (KMEB), Department of Endocrinology, Odense University Hospital & University of Southern Denmark, Odense, Denmark
| | - Nicholas Ditzel
- Molecular Endocrinology Laboratory (KMEB), Department of Endocrinology, Odense University Hospital & University of Southern Denmark, Odense, Denmark
| | - Pankaj Keshari
- Department of Neurology, Oslo University Hospital and Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Adiba Isa
- Molecular Endocrinology Laboratory (KMEB), Department of Endocrinology, Odense University Hospital & University of Southern Denmark, Odense, Denmark
| | - Abbas Jafari
- Department of Cellular and Molecular Medicine, DanStem (Danish Stem Cell Center), Panum Institute, University of Copenhagen, Copenhagen, Denmark
| | - Thomas L Andersen
- Department of Clinical Cell Biology, Vejle/Lillebaelt Hospital, Institute of Regional Health Research, University of Southern Denmark, Vejle, Denmark
| | - Jean-Marie Delaisse
- Department of Clinical Cell Biology, Vejle/Lillebaelt Hospital, Institute of Regional Health Research, University of Southern Denmark, Vejle, Denmark
| | - Yoshio Goshima
- Department of Molecular Pharmacology and Neurobiology, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Toshio Ohshima
- Department of Life Science and Medical Bioscience, Waseda University, Tokyo, Japan
| | - Moustapha Kassem
- Molecular Endocrinology Laboratory (KMEB), Department of Endocrinology, Odense University Hospital & University of Southern Denmark, Odense, Denmark.,Department of Cellular and Molecular Medicine, DanStem (Danish Stem Cell Center), Panum Institute, University of Copenhagen, Copenhagen, Denmark.,Stem Cell Unit, Department of Anatomy, Faculty of Medicine, King Saud University, Riyadh, Saudi Arabia
| |
Collapse
|
18
|
Khan MP, Khan K, Yadav PS, Singh AK, Nag A, Prasahar P, Mittal M, China SP, Tewari MC, Nagar GK, Tewari D, Trivedi AK, Sanyal S, Bandyopadhyay A, Chattopadhyay N. BMP signaling is required for adult skeletal homeostasis and mediates bone anabolic action of parathyroid hormone. Bone 2016; 92:132-144. [PMID: 27567726 DOI: 10.1016/j.bone.2016.08.018] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/02/2016] [Revised: 08/15/2016] [Accepted: 08/22/2016] [Indexed: 10/21/2022]
Abstract
Bmp2 and Bmp4 genes were ablated in adult mice (KO) using a conditional gene knockout technology. Bones were evaluated by microcomputed tomography (μCT), bone strength tester, histomorphometry and serum biochemical markers of bone turnover. Drill-hole was made at femur metaphysis and bone regeneration in the hole site was measured by calcein binding and μCT. Mice were either sham operated (ovary intact) or ovariectomized (OVX), and treated with human parathyroid hormone (PTH), 17β-estradiol (E2) or vehicle. KO mice displayed trabecular bone loss, diminished osteoid formation and reduced biomechanical strength compared with control (expressing Bmp2 and Bmp4). Both osteoblast and osteoclast functions were impaired in KO mice. Bone histomorphomtery and serum parameters established a low turnover bone loss in KO mice. Bone regeneration at the drill-hole site in KO mice was lower than control. However, deletion of Bmp2 gene alone had no effect on skeleton, an outcome similar to that reported previously for deletion of Bmp4 gene. Both PTH and E2 resulted in skeletal preservation in control-OVX, whereas in KO-OVX, E2 but not PTH was effective which suggested that the skeletal action of PTH required Bmp ligands but E2 did not. To determine cellular effects of Bmp2 and Bmp4, we used bone marrow stromal cells in which PTH but not E2 stimulated both Bmp2 and Bmp4 synthesis leading to increased Smad1/5 phosphorylation. Taken together, we conclude that Bmp2 and Bmp4 are essential for maintaining adult skeletal homeostasis and mediating the anabolic action of PTH.
Collapse
Affiliation(s)
- Mohd Parvez Khan
- Division of Endocrinology and Center for Research in Anabolic Skeletal Targets in Health and Illness (ASTHI), CSIR-Central Drug Research Institute, Sector 10 Jankipuram Extension, Sitapur Road, Lucknow 226031, India
| | - Kainat Khan
- Division of Endocrinology and Center for Research in Anabolic Skeletal Targets in Health and Illness (ASTHI), CSIR-Central Drug Research Institute, Sector 10 Jankipuram Extension, Sitapur Road, Lucknow 226031, India
| | - Prem Swaroop Yadav
- Department of Biological Sciences and Bioengineering, Indian Institute of Technology Kanpur 208016, India
| | - Abhishek Kumar Singh
- Division of Biochemistry, CSIR-Central Drug Research Institute, Sector 10 Jankipuram Extension, Sitapur Road, Lucknow 226031, India
| | - Aditi Nag
- Department of Biological Sciences and Bioengineering, Indian Institute of Technology Kanpur 208016, India
| | - Paritosh Prasahar
- Department of Biological Sciences and Bioengineering, Indian Institute of Technology Kanpur 208016, India
| | - Monika Mittal
- Division of Endocrinology and Center for Research in Anabolic Skeletal Targets in Health and Illness (ASTHI), CSIR-Central Drug Research Institute, Sector 10 Jankipuram Extension, Sitapur Road, Lucknow 226031, India; AcSIR, CSIR-Central Drug Research Institute, Lucknow 226031, India
| | - Shyamsundar Pal China
- Division of Endocrinology and Center for Research in Anabolic Skeletal Targets in Health and Illness (ASTHI), CSIR-Central Drug Research Institute, Sector 10 Jankipuram Extension, Sitapur Road, Lucknow 226031, India; AcSIR, CSIR-Central Drug Research Institute, Lucknow 226031, India
| | - Mahesh Chandra Tewari
- Division of Endocrinology and Center for Research in Anabolic Skeletal Targets in Health and Illness (ASTHI), CSIR-Central Drug Research Institute, Sector 10 Jankipuram Extension, Sitapur Road, Lucknow 226031, India
| | - Geet Kumar Nagar
- Division of Endocrinology and Center for Research in Anabolic Skeletal Targets in Health and Illness (ASTHI), CSIR-Central Drug Research Institute, Sector 10 Jankipuram Extension, Sitapur Road, Lucknow 226031, India
| | - Deepshikha Tewari
- Division of Endocrinology and Center for Research in Anabolic Skeletal Targets in Health and Illness (ASTHI), CSIR-Central Drug Research Institute, Sector 10 Jankipuram Extension, Sitapur Road, Lucknow 226031, India
| | - Arun Kumar Trivedi
- Division of Biochemistry, CSIR-Central Drug Research Institute, Sector 10 Jankipuram Extension, Sitapur Road, Lucknow 226031, India
| | - Sabyasachi Sanyal
- Division of Biochemistry, CSIR-Central Drug Research Institute, Sector 10 Jankipuram Extension, Sitapur Road, Lucknow 226031, India
| | - Amitabha Bandyopadhyay
- Department of Biological Sciences and Bioengineering, Indian Institute of Technology Kanpur 208016, India.
| | - Naibedya Chattopadhyay
- Division of Endocrinology and Center for Research in Anabolic Skeletal Targets in Health and Illness (ASTHI), CSIR-Central Drug Research Institute, Sector 10 Jankipuram Extension, Sitapur Road, Lucknow 226031, India; AcSIR, CSIR-Central Drug Research Institute, Lucknow 226031, India.
| |
Collapse
|
19
|
Yadav PS, Khan MP, Prashar P, Duggal S, Rath SK, Chattopadhyay N, Bandyopadhyay A. Characterization of BMP signaling dependent osteogenesis using a BMP depletable avianized bone marrow stromal cell line (TVA-BMSC). Bone 2016; 91:39-52. [PMID: 27424936 DOI: 10.1016/j.bone.2016.07.010] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/13/2015] [Revised: 06/22/2016] [Accepted: 07/14/2016] [Indexed: 01/08/2023]
Abstract
Adipogenesis, chondrogenesis and osteogenesis are BMP signaling dependent differentiation processes. However, the molecular networks operating downstream of BMP signaling to bring about these distinct fates are yet to be fully elucidated. We have developed a novel Bone Marrow Stromal Cell (BMSC) derived mouse cell line as a powerful in vitro platform to conduct such experiments. This cell line is a derivative of BMSCs isolated from a tamoxifen inducible Bmp2 and Bmp4 double conditional knock-out mouse strain. These BMSCs are immortalized and stably transfected with avian retroviral receptor TVA (TVA-BMSCs), enabling an easy method for stable transduction of multiple genes in these cells. In TVA-BMSCs multiple components of BMP signaling pathway can be manipulated simultaneously. Using this cell line we have demonstrated that for osteogenesis, BMP signaling is required only for the first three days. We have further demonstrated that Klf10, an osteogenic transcription factor which is transcribed in developing bones in a BMP signaling dependent manner, can largely compensate for the loss of BMP signaling during osteogenesis of BMSCs. TVA-BMSCs can undergo chondrogenesis and adipogenesis, and hence may be used for dissection of the molecular networks downstream of BMP signaling in these differentiation processes as well.
Collapse
Affiliation(s)
- Prem Swaroop Yadav
- Department of Biological Sciences and Bioengineering, Indian Institute of Technology, Kanpur, Uttar Pradesh 208016, India
| | - Mohd Parvez Khan
- Department of Biological Sciences and Bioengineering, Indian Institute of Technology, Kanpur, Uttar Pradesh 208016, India
| | - Paritosh Prashar
- Department of Biological Sciences and Bioengineering, Indian Institute of Technology, Kanpur, Uttar Pradesh 208016, India
| | - Shivali Duggal
- Department of Biological Sciences and Bioengineering, Indian Institute of Technology, Kanpur, Uttar Pradesh 208016, India
| | - Srikanta Kumar Rath
- Department of Biological Sciences and Bioengineering, Indian Institute of Technology, Kanpur, Uttar Pradesh 208016, India
| | - Naibedya Chattopadhyay
- Department of Biological Sciences and Bioengineering, Indian Institute of Technology, Kanpur, Uttar Pradesh 208016, India
| | - Amitabha Bandyopadhyay
- Department of Biological Sciences and Bioengineering, Indian Institute of Technology, Kanpur, Uttar Pradesh 208016, India.
| |
Collapse
|
20
|
Saxena M, Prashar P, Yadav PS, Sen J. Mouse bone marrow stromal cells differentiate to neuron-like cells upon inhibition of BMP signaling. Differentiation 2016; 92:1-9. [DOI: 10.1016/j.diff.2016.03.003] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2015] [Revised: 03/06/2016] [Accepted: 03/17/2016] [Indexed: 10/22/2022]
|
21
|
Nagai J, Baba R, Ohshima T. CRMPs Function in Neurons and Glial Cells: Potential Therapeutic Targets for Neurodegenerative Diseases and CNS Injury. Mol Neurobiol 2016; 54:4243-4256. [PMID: 27339876 DOI: 10.1007/s12035-016-0005-1] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2016] [Accepted: 06/14/2016] [Indexed: 12/19/2022]
Abstract
Neurodegeneration in the adult mammalian central nervous system (CNS) is fundamentally accelerated by its intrinsic neuronal mechanisms, including its poor regenerative capacity and potent extrinsic inhibitory factors. Thus, the treatment of neurodegenerative diseases faces many obstacles. The degenerative processes, consisting of axonal/dendritic structural disruption, abnormal axonal transport, release of extracellular factors, and inflammation, are often controlled by the cytoskeleton. From this perspective, regulators of the cytoskeleton could potentially be a therapeutic target for neurodegenerative diseases and CNS injury. Collapsin response mediator proteins (CRMPs) are known to regulate the assembly of cytoskeletal proteins in neurons, as well as control axonal growth and neural circuit formation. Recent studies have provided some novel insights into the roles of CRMPs in several inhibitory signaling pathways of neurodegeneration, in addition to its functions in neurological disorders and CNS repair. Here, we summarize the roles of CRMPs in axon regeneration and its emerging functions in non-neuronal cells, especially in inflammatory responses. We also discuss the direct and indirect targeting of CRMPs as a novel therapeutic strategy for neurological diseases.
Collapse
Affiliation(s)
- Jun Nagai
- Department of Life Science and Medical Bioscience, Graduate School of Advanced Science and Engineering, Waseda University, TWIns, 2-2 Wakamatsu-cho Shinjuku-ku, Tokyo, 162-8480, Japan.,Research Fellow of Japan Society for the Promotion of Science, Tokyo, Japan
| | - Rina Baba
- Department of Life Science and Medical Bioscience, Graduate School of Advanced Science and Engineering, Waseda University, TWIns, 2-2 Wakamatsu-cho Shinjuku-ku, Tokyo, 162-8480, Japan
| | - Toshio Ohshima
- Department of Life Science and Medical Bioscience, Graduate School of Advanced Science and Engineering, Waseda University, TWIns, 2-2 Wakamatsu-cho Shinjuku-ku, Tokyo, 162-8480, Japan.
| |
Collapse
|
22
|
Subramaniam M, Pitel KS, Withers SG, Drissi H, Hawse JR. TIEG1 enhances Osterix expression and mediates its induction by TGFβ and BMP2 in osteoblasts. Biochem Biophys Res Commun 2016; 470:528-533. [PMID: 26801561 DOI: 10.1016/j.bbrc.2016.01.112] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2016] [Accepted: 01/18/2016] [Indexed: 01/03/2023]
Abstract
Deletion of TIEG1/KLF10 in mice results in an osteopenic skeletal phenotype with significant decreases in both bone mineral density and content throughout the skeleton. Calvarial osteoblasts isolated from TIEG1 knockout (KO) mice display numerous changes in gene expression and exhibit significant delays in their mineralization rates relative to wild-type (WT) controls. Here, we demonstrate that loss of TIEG1 expression in osteoblasts results in decreased levels of Osterix mRNA. Suppression of TIEG1 expression in WT osteoblasts leads to decreased Osterix expression while restoration of TIEG1 expression in TIEG1 KO osteoblasts results in increased levels of Osterix. Transient transfection and chromatin immunoprecipitation assays reveal that TIEG1 directly binds to and activates the Osterix promoter and demonstrate that the zinc finger-containing DNA binding domain of TIEG1 is necessary for this regulation. Furthermore, we reveal that TIEG1 expression is essential for the induction of Osterix expression by important bone-related cytokines such as TGFβ and BMP2 in osteoblast cells. Taken together, these data implicate an important role for TIEG1 in regulating the expression of Osterix, a master regulator of osteoblast differentiation and bone formation, and suggest that decreased expression of Osterix, as well as impaired TGFβ and BMP2 signaling, contribute to the observed osteopenic bone phenotype of TIEG1 KO mice.
Collapse
Affiliation(s)
| | - Kevin S Pitel
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN 55905, USA
| | - Sarah G Withers
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN 55905, USA
| | - Hicham Drissi
- Department of Orthopaedic Surgery, UConn Musculoskeletal Institute, UConn Health, Farmington, CT 06030, USA
| | - John R Hawse
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN 55905, USA.
| |
Collapse
|
23
|
A comprehensive mRNA expression analysis of developing chicken articular cartilage. Gene Expr Patterns 2016; 20:22-31. [DOI: 10.1016/j.gep.2015.11.001] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2015] [Revised: 10/25/2015] [Accepted: 11/02/2015] [Indexed: 01/01/2023]
|