1
|
Cha J, Tong X, Coate KC, Guo M, Liu JH, Reynolds G, Walker EM, Stein RA, Mchaourab H, Stein R. Defining unique structural features in the MAFA and MAFB transcription factors that control Insulin gene activity. J Biol Chem 2024; 300:107938. [PMID: 39476962 PMCID: PMC11626809 DOI: 10.1016/j.jbc.2024.107938] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2024] [Revised: 10/08/2024] [Accepted: 10/22/2024] [Indexed: 11/06/2024] Open
Abstract
MAFA and MAFB are related basic-leucine-zipper domain-containing transcription factors which have important overlapping and distinct regulatory roles in a variety of cellular contexts, including hormone production in pancreatic islet cells. Here, we first examined how mutating conserved MAF protein-DNA contact sites obtained from X-ray crystal structure analysis impacted their DNA-binding and Insulin enhancer-driven activity. While most of these interactions were essential and their disruption severely compromised activity, we identified that regions outside of these contact sites also contributed to transcriptional activity. AlphaFold 2 (https://alphafold.ebi.ac.uk), an artificial intelligence-based structural prediction program, was used to determine if there were also differences in the three-dimensional organization of the non-DNA binding/dimerization sequences of MAFA and MAFB. This analysis was conducted on the WT proteins as well as the pathogenic MAFASer64Phe and MAFBSer70Ala transactivation domain mutants, with differences revealed between MAFAWT and MAFBWT as well as between MAFASer64Phe and MAFAWT, but not between MAFBSer70Ala and MAFBWT. Moreover, dissimilarities between these proteins were also observed in their ability to cooperatively stimulate Insulin enhancer-driven activity in the presence of other islet-enriched transcription factors. Analysis of MAFA and MAFB chimeras disclosed that these properties were influenced by their unique C-terminal region structural differences predicted by AlphaFold 2. Our findings have revealed key structural features of these closely related proteins that impact their ability to regulate gene expression.
Collapse
Affiliation(s)
- Jeeyeon Cha
- Department of Molecular Physiology & Biophysics, Vanderbilt University, Nashville, Tennessee, USA; Division of Diabetes, Endocrinology, and Metabolism, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Xin Tong
- Department of Molecular Physiology & Biophysics, Vanderbilt University, Nashville, Tennessee, USA
| | - Katie C Coate
- Department of Molecular Physiology & Biophysics, Vanderbilt University, Nashville, Tennessee, USA; Division of Diabetes, Endocrinology, and Metabolism, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA; VA Tennessee Valley Healthcare System, Nashville, Tennessee, USA
| | - Min Guo
- Department of Molecular Physiology & Biophysics, Vanderbilt University, Nashville, Tennessee, USA
| | - Jin-Hua Liu
- Department of Molecular Physiology & Biophysics, Vanderbilt University, Nashville, Tennessee, USA
| | - Garrett Reynolds
- Department of Molecular Physiology & Biophysics, Vanderbilt University, Nashville, Tennessee, USA
| | - Emily M Walker
- Division of Metabolism, Endocrinology & Diabetes, Departments of Molecular & Integrative Physiology and Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Richard A Stein
- Department of Molecular Physiology & Biophysics, Vanderbilt University, Nashville, Tennessee, USA; Center for Applied Artificial Intelligence in Protein Dynamics, Vanderbilt University, Nashville, Tennessee, USA
| | - Hassane Mchaourab
- Department of Molecular Physiology & Biophysics, Vanderbilt University, Nashville, Tennessee, USA; Center for Applied Artificial Intelligence in Protein Dynamics, Vanderbilt University, Nashville, Tennessee, USA
| | - Roland Stein
- Department of Molecular Physiology & Biophysics, Vanderbilt University, Nashville, Tennessee, USA.
| |
Collapse
|
2
|
Cha J, Tong X, Coate KC, Guo M, Liu JH, Reynolds G, Walker EM, Stein RA, Mchaourab H, Stein R. Defining unique structural features in the MAFA and MAFB transcription factors that control Insulin gene activity. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.08.23.554429. [PMID: 37662349 PMCID: PMC10473715 DOI: 10.1101/2023.08.23.554429] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/05/2023]
Abstract
MAFA and MAFB are related basic-leucine-zipper domain containing transcription factors which have important overlapping and distinct regulatory roles in a variety of cellular contexts, including hormone production in pancreatic islet α and β cells. Here we first examined how mutating conserved MAF protein-DNA contacts obtained from X-ray crystal structure analysis impacted their DNA-binding and Insulin enhancer-driven activity. While most of these interactions were essential and their disruption severely compromised activity, we identified that regions outside of the contact areas also contributed to activity. AlphaFold 2, an artificial intelligence-based structural prediction program, was next used to determine if there were also differences in the three-dimensional organization of the non-DNA binding/dimerization sequences of MAFA and MAFB. This analysis was conducted on the wildtype (WT) proteins as well as the pathogenic MAFA Ser64Phe and MAFB Ser70Ala trans -activation domain mutants, with differences revealed between MAFA WT and MAFB WT as well as between MAFA Ser64Phe and MAFA WT , but not between MAFB Ser70Ala and MAFB WT . Moreover, dissimilarities between these proteins were also observed in their ability to cooperatively stimulate Insulin enhancer-driven activity in the presence of other islet-enriched transcription factors. Analysis of MAFA and MAFB chimeras disclosed that these properties were greatly influenced by unique C-terminal region structural differences predicted by AlphaFold 2. Importantly, these results have revealed features of these closely related proteins that are functionally significant in islet biology.
Collapse
|
3
|
Bonello JP, Tse MY, Robinson TJG, Bardana DD, Waldman SD, Pang SC. Expression of Chondrogenic Potential Markers in Cultured Chondrocytes from the Human Knee Joint. Cartilage 2024:19476035241241930. [PMID: 38616342 PMCID: PMC11569588 DOI: 10.1177/19476035241241930] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/29/2023] [Revised: 02/18/2024] [Accepted: 03/07/2024] [Indexed: 04/16/2024] Open
Abstract
OBJECTIVES While substantial progress has been made in engineering cartilaginous constructs for animal models, further research is needed to translate these methodologies for human applications. Evidence suggests that cultured autologous chondrocytes undergo changes in phenotype and gene expression, thereby affecting their proliferation and differentiation capacity. This study was designed to evaluate the expression of chondrogenic markers in cultured human articular chondrocytes from passages 3 (P3) and 7 (P7), beyond the current clinical recommendation of P3. METHODS Cultured autologous chondrocytes were passaged from P3 up to P7, and quantitative polymerase chain reaction (qPCR) was used to assess mRNA expression of chondrogenic markers, including collagen type I (COLI), collagen type II (COLII), aggrecan (AGG), bone morphogenetic protein 4 (BMP4), transcription factor SOX-9 (SOX9), proteoglycan 4 (PGR4), and transformation-related protein 53 (p53), between P3 and P7. RESULTS Except for AGG, no significant differences were found in the expression of markers between passages, suggesting the maintenance of chondrogenic potential in cultured chondrocytes. Differential expression identified between SOX9 and PGR4, as well as between COLI and SOX9, indicates that differences in chondrogenic markers are present between age groups and sexes, respectively. CONCLUSIONS Overall, expression profiles of younger and male chondrocytes exhibit conversion of mature cartilage characteristics compared to their counterparts, with signs of dedifferentiation and loss of phenotype within-group passaging. These results may have implications in guiding the use of higher passaged chondrocytes for engineering constructs and provide a foundation for clinical recommendations surrounding the repair and treatment of articular cartilage pathology in both sexes.
Collapse
Affiliation(s)
- John-Peter Bonello
- Department of Biomedical and Molecular Sciences, Queen’s University, Kingston, ON, Canada
| | - M. Yat Tse
- Department of Biomedical and Molecular Sciences, Queen’s University, Kingston, ON, Canada
| | - Trevor J. G. Robinson
- Department of Biomedical and Molecular Sciences, Queen’s University, Kingston, ON, Canada
| | - Davide D. Bardana
- Division of Surgery, Kingston General Hospital, Kingston, ON, Canada
| | - Stephen D. Waldman
- Department of Chemical Engineering, Toronto Metropolitan University, Toronto, ON, Canada
| | - Stephen C. Pang
- Department of Biomedical and Molecular Sciences, Queen’s University, Kingston, ON, Canada
| |
Collapse
|
4
|
Khoswanto C. Role of matrix metalloproteinases in bone regeneration: Narrative review. J Oral Biol Craniofac Res 2023; 13:539-543. [PMID: 37351418 PMCID: PMC10282173 DOI: 10.1016/j.jobcr.2023.06.002] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Revised: 05/14/2023] [Accepted: 06/07/2023] [Indexed: 06/24/2023] Open
Abstract
Background Matrix metalloproteinases (MMPs) not only work as enzymes but also as degrading enzymes that have been shown to play an important function in extracellular matrix (ECM) regeneration, including bone regeneration. To generate new bone tissue, bone regeneration or repair relies on a series of regulated processes in which MMPs play an important role. Bone cells express the MMPs in an active state, and these MMPs are assumed to have a crucial role, not only for the viability and functionality of osteoclasts, osteoblasts, and osteocytes but also for the formation and development of chondrocytes. Objective This study aimed to review and present the roles of matrix metalloproteinases in bone regeneration. Methods An analysis of the scientific literature on the topics of matrix metalloproteinases in bone regeneration was done on PubMed and Google Scholar. Search results were screened for articles that described or investigated the impacts matrix metalloproteinases have on bones in relation to dentistry. The journals' cited papers were also assessed for relevance and included if they complied with the criteria for inclusion. Accessibility to the full document was one of the prerequisites for admission. Result Bone regeneration are intricate ongoing processes involving numerous MMPs, especially MMP 2, 9 and 13. MMP-2 appears to alter bone growth through influencing osteoclast and osteoblast activity and proliferation, MMP-9-deficient animals have abnormal bone formation exclusively during endochondral ossification, MMP 13 is responsible for osteoclast receptor activation, has been linked to the breakdown bone resorption. Conclusions MMP 2, 9, and 13 play a major protective role in osteogenesis and bone regeneration.
Collapse
Affiliation(s)
- Christian Khoswanto
- Department of Oral Biology Faculty of Dentistry, Airlangga University. Jln. Mayjend. Prof. Dr. Moestopo No. 47, Surabaya, 60132, Indonesia
| |
Collapse
|
5
|
Fujino M, Ojima M, Ishibashi S, Mizuno S, Takahashi S. Generation and mutational analysis of a transgenic murine model of the human MAF mutation. Am J Med Genet A 2023. [PMID: 37186330 DOI: 10.1002/ajmg.a.63220] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Revised: 03/29/2023] [Accepted: 04/14/2023] [Indexed: 05/17/2023]
Abstract
Aymé-Gripp syndrome is an autosomal dominant multisystem disorder. The major clinical features of this syndrome include congenital cataracts, sensorineural hearing loss, intellectual disability, and a distinctive flat facial appearance. MAF has been identified as a causative gene of the syndrome, and heterozygous variants owing to impairment in glycogen synthase kinase 3 (GSK3)-mediated MAF phosphorylation shows related disorders. However, the underlying mechanisms of these types of disorders in affected individuals remain poorly understood. To explore the underlying mechanisms and discover new phenotypes, a murine model with a Maf mutation on a GSK3 phosphorylation motif, p.Thr58Ile, was generated using CRISPR-Cas9 gene editing. This is a homologous mutation to that in human patients. Our murine model exhibited similar phenotypes to those in humans, such as lens abnormalities, short stature, growth retardation, and abnormal skull morphology. The murine model showed decreased brain volume and malocclusion. Considering the sequencing and genotyping data, our models were successfully generated for the first time (to the best of our knowledge). Therefore, this study offers new and unique functional insights into human and murine MAF and novel clinical values of MAF pathogenic variants associated with changes in the functions of several organs based on a viable murine model.
Collapse
Affiliation(s)
- Mitsunori Fujino
- Department of Anatomy and Embryology, University of Tsukuba, Ibaraki, Japan
- Human Biology, School of Integrative and Global Majors, University of Tsukuba, Ibaraki, Japan
| | - Masami Ojima
- Department of Anatomy and Embryology, University of Tsukuba, Ibaraki, Japan
| | - Shun Ishibashi
- Department of Anatomy and Embryology, University of Tsukuba, Ibaraki, Japan
- Medical Sciences, Graduate School of Comprehensive Human Sciences, University of Tsukuba, Ibaraki, Japan
| | - Seiya Mizuno
- Laboratory Animal Resource Center, University of Tsukuba, Ibaraki, Japan
| | - Satoru Takahashi
- Department of Anatomy and Embryology, University of Tsukuba, Ibaraki, Japan
- Laboratory Animal Resource Center, University of Tsukuba, Ibaraki, Japan
- Life Science Center, Tsukuba Advanced Research Alliance (TARA), University of Tsukuba, Ibaraki, Japan
- International Institute for Integrative Sleep Medicine (WPI-IIIS), University of Tsukuba, Ibaraki, Japan
- Transborder Medical Research Center, University of Tsukuba, Ibaraki, Japan
| |
Collapse
|
6
|
Fujino M, Morito N, Hayashi T, Ojima M, Ishibashi S, Kuno A, Koshiba S, Yamagata K, Takahashi S. Transcription factor c-Maf deletion improves streptozotocin-induced diabetic nephropathy by directly regulating Sglt2 and Glut2. JCI Insight 2023; 8:163306. [PMID: 36787192 PMCID: PMC10070115 DOI: 10.1172/jci.insight.163306] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Accepted: 02/01/2023] [Indexed: 02/15/2023] Open
Abstract
The transcription factor c-Maf has been widely studied and has been reported to play a critical role in embryonic kidney development; however, the postnatal functions of c-Maf in adult kidneys remain unknown as c-Maf-null C57BL/6J mice exhibit embryonic lethality. In this study, we investigated the role of c-Maf in adult mouse kidneys by comparing the phenotypes of tamoxifen-inducible (TAM-inducible) c-Maf-knockout mice (c-Maffl/fl; CAG-Cre-ERTM mice named "c-MafΔTAM") with those of c-Maffl/fl control mice, 10 days after TAM injection [TAM(10d)]. In addition, we examined the effects of c-Maf deletion on diabetic conditions by injecting the mice with streptozotocin, 4 weeks before TAM injection. c-MafΔTAM mice displayed primary glycosuria caused by sodium-glucose cotransporter 2 (Sglt2) and glucose transporter 2 (Glut2) downregulation in the kidneys without diabetes, as well as morphological changes and life-threatening injuries in the kidneys on TAM(10d). Under diabetic conditions, c-Maf deletion promoted recovery from hyperglycemia and suppressed albuminuria and diabetic nephropathy by causing similar effects as did Sglt2 knockout and SGLT2 inhibitors. In addition to demonstrating the potentially unique gene regulation of c-Maf, these findings highlight the renoprotective effects of c-Maf deficiency under diabetic conditions and suggest that c-Maf could be a novel therapeutic target gene for treating diabetic nephropathy.
Collapse
Affiliation(s)
- Mitsunori Fujino
- Department of Anatomy and Embryology, Faculty of Medicine
- PhD Program in Human Biology, School of Integrative and Global Majors
| | - Naoki Morito
- Department of Nephrology, Faculty of Medicine; and
| | - Takuto Hayashi
- Department of Anatomy and Embryology, Faculty of Medicine
- Doctoral Program in Biomedical Sciences, Graduate School of Comprehensive Human Sciences, University of Tsukuba, Ibaraki, Japan
| | - Masami Ojima
- Department of Anatomy and Embryology, Faculty of Medicine
| | - Shun Ishibashi
- Department of Anatomy and Embryology, Faculty of Medicine
- Doctoral Program in Biomedical Sciences, Graduate School of Comprehensive Human Sciences, University of Tsukuba, Ibaraki, Japan
| | - Akihiro Kuno
- Department of Anatomy and Embryology, Faculty of Medicine
| | - Seizo Koshiba
- Tohoku Medical Megabank Organization and
- Advanced Research Center for Innovations in Next-Generation Medicine (INGEM), Tohoku University, Sendai, Japan
| | | | - Satoru Takahashi
- Department of Anatomy and Embryology, Faculty of Medicine
- Laboratory Animal Resource Center
- Life Science Center for Survival Dynamics, Tsukuba Advanced Research Alliance (TARA)
- International Institute for Integrative Sleep Medicine (WPI-IIIS), and
- Transborder Medical Research Center, Faculty of Medicine, University of Tsukuba, Ibaraki, Japan
| |
Collapse
|
7
|
Deng Y, Lu L, Zhang H, Fu Y, Liu T, Chen Y. The role and regulation of Maf proteins in cancer. Biomark Res 2023; 11:17. [PMID: 36750911 PMCID: PMC9903618 DOI: 10.1186/s40364-023-00457-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Accepted: 01/22/2023] [Indexed: 02/09/2023] Open
Abstract
The Maf proteins (Mafs) belong to basic leucine zipper transcription factors and are members of the activator protein-1 (AP-1) superfamily. There are two subgroups of Mafs: large Mafs and small Mafs, which are involved in a wide range of biological processes, such as the cell cycle, proliferation, oxidative stress, and inflammation. Therefore, dysregulation of Mafs can affect cell fate and is closely associated with diverse diseases. Accumulating evidence has established both large and small Mafs as mediators of tumor development. In this review, we first briefly describe the structure and physiological functions of Mafs. Then we summarize the upstream regulatory mechanisms that control the expression and activity of Mafs. Furthermore, we discuss recent studies on the critical role of Mafs in cancer progression, including cancer proliferation, apoptosis, metastasis, tumor/stroma interaction and angiogenesis. We also review the clinical implications of Mafs, namely their potential possibilities and limitations as biomarkers and therapeutic targets in cancer.
Collapse
Affiliation(s)
- Yalan Deng
- grid.452223.00000 0004 1757 7615Department of Oncology, NHC Key Laboratory of Cancer Proteomics, Laboratory of Structural Biology, Xiangya Hospital, Central South University, Changsha, 410008 Hunan China
| | - Liqing Lu
- grid.452223.00000 0004 1757 7615Department of Oncology, NHC Key Laboratory of Cancer Proteomics, Laboratory of Structural Biology, Xiangya Hospital, Central South University, Changsha, 410008 Hunan China ,grid.452223.00000 0004 1757 7615Department of Thoracic Surgery, Xiangya Hospital, Central South University, Changsha, 410008 Hunan China
| | - Huajun Zhang
- grid.452223.00000 0004 1757 7615Department of Oncology, NHC Key Laboratory of Cancer Proteomics, Laboratory of Structural Biology, Xiangya Hospital, Central South University, Changsha, 410008 Hunan China ,grid.452223.00000 0004 1757 7615Department of Ultrasonic Imaging, Xiangya Hospital, Central South University, Changsha, 410008 Hunan China
| | - Ying Fu
- grid.452223.00000 0004 1757 7615Department of Oncology, NHC Key Laboratory of Cancer Proteomics, Laboratory of Structural Biology, Xiangya Hospital, Central South University, Changsha, 410008 Hunan China
| | - Ting Liu
- Department of Gastroenterology, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China.
| | - Yongheng Chen
- Department of Oncology, NHC Key Laboratory of Cancer Proteomics, Laboratory of Structural Biology, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China. .,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China.
| |
Collapse
|
8
|
Wang Q, Qin T, Tan H, Ding X, Lin X, Li J, Lin Z, Sun L, Lin H, Chen W. Broadening the genotypic and phenotypic spectrum of MAF in three Chinese Han congenital cataracts families. Am J Med Genet A 2022; 188:2888-2898. [PMID: 36097645 DOI: 10.1002/ajmg.a.62947] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2022] [Revised: 06/06/2022] [Accepted: 06/19/2022] [Indexed: 01/31/2023]
Abstract
Pathogenic variants in the v-maf avian musculoaponeurotic fibrosarcoma oncogene homologue (MAF) encoding a transcription factor (from a unique subclass of basic leucine zipper transcription factors) are associated with isolated congenital cataracts (CCs) and Aymé-Gripp syndrome (AYGRPS). We collected detailed disease histories from, and performed comprehensive ophthalmic and systemic examinations in 269 patients with CCs; we then performed whole-exome sequencing. Pathogenicity assessments were evaluated using multiple predictive tools. The clinical validities of the reported gene-disease relationships for MAF genes (MAF-CCs and MAF-AYGRPS) were assessed using the ClinGen gene curation framework. We identified two novel (c.173C>A, p.Thr58Asn and c.947T>C, p. Leu316Pro) variants and one known (c.173C>T, p.Thr58Ile) MAF missense variant in three patients. We described novel phenotypes including cleft palate, macular hypoplasia, and retinal neovascularization in the peripheral avascular area and analyzed the genotype-phenotype correlations. We demonstrated associations of variants in the MAF C-terminal DNA-binding domain with CCs and associations of variants in the N-terminal transactivation domain of MAF with AYGRPS. We thus expand the genotypic and phenotypic spectrum of the MAF gene. The ClinGen gene curation framework results suggested that variants in different domains of MAF are associated with different diseases.
Collapse
Affiliation(s)
- Qiwei Wang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Centre, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangdong Provincial Clinical Research Centre for Ocular Diseases, Guangzhou, China
| | - Tingfeng Qin
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Centre, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangdong Provincial Clinical Research Centre for Ocular Diseases, Guangzhou, China
| | | | - Xiaoyan Ding
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Centre, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangdong Provincial Clinical Research Centre for Ocular Diseases, Guangzhou, China
| | - Xiaoshan Lin
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Centre, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangdong Provincial Clinical Research Centre for Ocular Diseases, Guangzhou, China
| | - Jing Li
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Centre, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangdong Provincial Clinical Research Centre for Ocular Diseases, Guangzhou, China
| | - Zhuolin Lin
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Centre, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangdong Provincial Clinical Research Centre for Ocular Diseases, Guangzhou, China
| | - Limei Sun
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Centre, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangdong Provincial Clinical Research Centre for Ocular Diseases, Guangzhou, China
| | - Haotian Lin
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Centre, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangdong Provincial Clinical Research Centre for Ocular Diseases, Guangzhou, China
| | - Weirong Chen
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Centre, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangdong Provincial Clinical Research Centre for Ocular Diseases, Guangzhou, China
| |
Collapse
|
9
|
Yang Z, Yi P, Liu Z, Zhang W, Mei L, Feng C, Tu C, Li Z. Stem Cell-Laden Hydrogel-Based 3D Bioprinting for Bone and Cartilage Tissue Engineering. Front Bioeng Biotechnol 2022; 10:865770. [PMID: 35656197 PMCID: PMC9152119 DOI: 10.3389/fbioe.2022.865770] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2022] [Accepted: 04/18/2022] [Indexed: 12/30/2022] Open
Abstract
Tremendous advances in tissue engineering and regenerative medicine have revealed the potential of fabricating biomaterials to solve the dilemma of bone and articular defects by promoting osteochondral and cartilage regeneration. Three-dimensional (3D) bioprinting is an innovative fabrication technology to precisely distribute the cell-laden bioink for the construction of artificial tissues, demonstrating great prospect in bone and joint construction areas. With well controllable printability, biocompatibility, biodegradability, and mechanical properties, hydrogels have been emerging as an attractive 3D bioprinting material, which provides a favorable biomimetic microenvironment for cell adhesion, orientation, migration, proliferation, and differentiation. Stem cell-based therapy has been known as a promising approach in regenerative medicine; however, limitations arise from the uncontrollable proliferation, migration, and differentiation of the stem cells and fortunately could be improved after stem cells were encapsulated in the hydrogel. In this review, our focus was centered on the characterization and application of stem cell-laden hydrogel-based 3D bioprinting for bone and cartilage tissue engineering. We not only highlighted the effect of various kinds of hydrogels, stem cells, inorganic particles, and growth factors on chondrogenesis and osteogenesis but also outlined the relationship between biophysical properties like biocompatibility, biodegradability, osteoinductivity, and the regeneration of bone and cartilage. This study was invented to discuss the challenge we have been encountering, the recent progress we have achieved, and the future perspective we have proposed for in this field.
Collapse
Affiliation(s)
- Zhimin Yang
- Department of Orthopedics, The Second Xiangya Hospital, Central South University, Changsha, China
- Hunan Key Laboratory of Tumor Models and Individualized Medicine, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Ping Yi
- Department of Dermatology, The Second Xiangya Hospital, Central South University, Hunan Key Laboratory of Medical Epigenomics, Changsha, China
| | - Zhongyue Liu
- Department of Orthopedics, The Second Xiangya Hospital, Central South University, Changsha, China
- Hunan Key Laboratory of Tumor Models and Individualized Medicine, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Wenchao Zhang
- Department of Orthopedics, The Second Xiangya Hospital, Central South University, Changsha, China
- Hunan Key Laboratory of Tumor Models and Individualized Medicine, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Lin Mei
- Department of Orthopedics, The Second Xiangya Hospital, Central South University, Changsha, China
- Hunan Key Laboratory of Tumor Models and Individualized Medicine, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Chengyao Feng
- Department of Orthopedics, The Second Xiangya Hospital, Central South University, Changsha, China
- Hunan Key Laboratory of Tumor Models and Individualized Medicine, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Chao Tu
- Department of Orthopedics, The Second Xiangya Hospital, Central South University, Changsha, China
- Hunan Key Laboratory of Tumor Models and Individualized Medicine, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Zhihong Li
- Department of Orthopedics, The Second Xiangya Hospital, Central South University, Changsha, China
- Hunan Key Laboratory of Tumor Models and Individualized Medicine, The Second Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
10
|
Kinoshita A, Ohyama K, Tanimura S, Matsuda K, Kishino T, Negishi Y, Asahina N, Shiraishi H, Hosoki K, Tomiwa K, Ishihara N, Mishima H, Mori R, Nakashima M, Saitoh S, Yoshiura KI. Itpr1 regulates the formation of anterior eye segment tissues derived from neural crest cells. Development 2021; 148:271160. [PMID: 34338282 DOI: 10.1242/dev.188755] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2020] [Accepted: 07/19/2021] [Indexed: 01/23/2023]
Abstract
Mutations in ITPR1 cause ataxia and aniridia in individuals with Gillespie syndrome (GLSP). However, the pathogenic mechanisms underlying aniridia remain unclear. We identified a de novo GLSP mutation hotspot in the 3'-region of ITPR1 in five individuals with GLSP. Furthermore, RNA-sequencing and immunoblotting revealed an eye-specific transcript of Itpr1, encoding a 218amino acid isoform. This isoform is localized not only in the endoplasmic reticulum, but also in the nuclear and cytoplasmic membranes. Ocular-specific transcription was repressed by SOX9 and induced by MAF in the anterior eye segment (AES) tissues. Mice lacking seven base pairs of the last Itpr1 exon exhibited ataxia and aniridia, in which the iris lymphatic vessels, sphincter and dilator muscles, corneal endothelium and stroma were disrupted, but the neural crest cells persisted after completion of AES formation. Our analyses revealed that the 218-amino acid isoform regulated the directionality of actin fibers and the intensity of focal adhesion. The isoform might control the nuclear entry of transcriptional regulators, such as YAP. It is also possible that ITPR1 regulates both AES differentiation and muscle contraction in the iris.
Collapse
Affiliation(s)
- Akira Kinoshita
- Department of Human Genetics, Atomic Bomb Disease Institute, Nagasaki University, Nagasaki 852-8523, Japan
| | - Kaname Ohyama
- Department of Pharmacy Practice, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki 852-3131, Japan
| | - Susumu Tanimura
- Department of Cell Regulation, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki 852-3131, Japan
| | - Katsuya Matsuda
- Department of Tumor and Diagnostic Pathology, Atomic Bomb Disease Institute, Nagasaki University, Nagasaki 852-8523, Japan
| | - Tatsuya Kishino
- Gene Research Center, Center for Frontier Life Sciences, Nagasaki University, Nagasaki 852-8523, Japan
| | - Yutaka Negishi
- Department of Pediatrics and Neonatology, Nagoya City University Graduate School of Medical Sciences, Nagoya 467-8602, Japan
| | - Naoko Asahina
- Department of Pediatrics, Hokkaido University Graduate School of Medicine, Sapporo 060-8638, Japan
| | - Hideaki Shiraishi
- Department of Pediatrics, Hokkaido University Graduate School of Medicine, Sapporo 060-8638, Japan
| | - Kana Hosoki
- Department of Medical Genetics, Osaka Women's and Children's Hospital, Osaka 594-1101, Japan
| | - Kiyotaka Tomiwa
- Department of Pediatrics, Todaiji Ryoiku Hospital for Children, Nara 630-8211, Japan
| | - Naoko Ishihara
- Department of Pediatrics, Fujita Health University School of Medicine, Toyoake 470-1192, Japan
| | - Hiroyuki Mishima
- Department of Human Genetics, Atomic Bomb Disease Institute, Nagasaki University, Nagasaki 852-8523, Japan
| | - Ryoichi Mori
- Department of Pathology, Nagasaki University School of Medicine and Graduate School of Biomedical Sciences, Nagasaki 852-8523, Japan
| | - Masahiro Nakashima
- Department of Tumor and Diagnostic Pathology, Atomic Bomb Disease Institute, Nagasaki University, Nagasaki 852-8523, Japan
| | - Shinji Saitoh
- Department of Pediatrics and Neonatology, Nagoya City University Graduate School of Medical Sciences, Nagoya 467-8602, Japan
| | - Koh-Ichiro Yoshiura
- Department of Human Genetics, Atomic Bomb Disease Institute, Nagasaki University, Nagasaki 852-8523, Japan
| |
Collapse
|
11
|
Jiang Z, Byers S, Casal ML, Smith LJ. Failures of Endochondral Ossification in the Mucopolysaccharidoses. Curr Osteoporos Rep 2020; 18:759-773. [PMID: 33064251 PMCID: PMC7736118 DOI: 10.1007/s11914-020-00626-y] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 09/29/2020] [Indexed: 12/12/2022]
Abstract
PURPOSE OF REVIEW The mucopolysaccharidoses (MPS) are a group of inherited lysosomal storage disorders characterized by abnormal accumulation of glycosaminoglycans (GAGs) in cells and tissues. MPS patients frequently exhibit failures of endochondral ossification during postnatal growth leading to skeletal deformity and short stature. In this review, we outline the current understanding of the cellular and molecular mechanisms underlying failures of endochondral ossification in MPS and discuss associated treatment challenges and opportunities. RECENT FINDINGS Studies in MPS patients and animal models have demonstrated that skeletal cells and tissues exhibit significantly elevated GAG storage from early in postnatal life and that this is associated with impaired cartilage-to-bone conversion in primary and secondary ossification centers, and growth plate dysfunction. Recent studies have begun to elucidate the underlying cellular and molecular mechanisms, including impaired chondrocyte proliferation and hypertrophy, diminished growth factor signaling, disrupted cell cycle progression, impaired autophagy, and increased cell stress and apoptosis. Current treatments such as hematopoietic stem cell transplantation and enzyme replacement therapy fail to normalize endochondral ossification in MPS. Emerging treatments including gene therapy and small molecule-based approaches hold significant promise in this regard. Failures of endochondral ossification contribute to skeletal deformity and short stature in MPS patients, increasing mortality and reducing quality of life. Early intervention is crucial for effective treatment, and there is a critical need for new approaches that normalize endochondral ossification by directly targeting affected cells and signaling pathways.
Collapse
Affiliation(s)
- Zhirui Jiang
- Department of Orthopedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Neurosurgery, Perelman School of Medicine, University of Pennsylvania, 371 Stemmler Hall, 3450 Hamilton Walk, Philadelphia, PA, 19104, USA
| | - Sharon Byers
- Genetics and Molecular Pathology, SA Pathology, Adelaide, SA, Australia
- Paediatrics, The University of Adelaide, Adelaide, SA, Australia
- Genetics and Evolution, The University of Adelaide, Adelaide, SA, Australia
| | - Margret L Casal
- Department of Clinical Sciences and Advanced Medicine, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Lachlan J Smith
- Department of Orthopedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
- Department of Neurosurgery, Perelman School of Medicine, University of Pennsylvania, 371 Stemmler Hall, 3450 Hamilton Walk, Philadelphia, PA, 19104, USA.
| |
Collapse
|
12
|
Yilmaz EN, Zeugolis DI. Electrospun Polymers in Cartilage Engineering-State of Play. Front Bioeng Biotechnol 2020; 8:77. [PMID: 32133352 PMCID: PMC7039817 DOI: 10.3389/fbioe.2020.00077] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2019] [Accepted: 01/29/2020] [Indexed: 12/17/2022] Open
Abstract
Articular cartilage defects remain a clinical challenge. Articular cartilage defects progress to osteoarthritis, which negatively (e.g., remarkable pain, decreased mobility, distress) affects millions of people worldwide and is associated with excessive healthcare costs. Surgical procedures and cell-based therapies have failed to deliver a functional therapy. To this end, tissue engineering therapies provide a promise to deliver a functional cartilage substitute. Among the various scaffold fabrication technologies available, electrospinning is continuously gaining pace, as it can produce nano- to micro- fibrous scaffolds that imitate architectural features of native extracellular matrix supramolecular assemblies and can deliver variable cell populations and bioactive molecules. Herein, we comprehensively review advancements and shortfalls of various electrospun scaffolds in cartilage engineering.
Collapse
Affiliation(s)
- Elif Nur Yilmaz
- Regenerative, Modular & Developmental Engineering Laboratory, National University of Ireland Galway, Galway, Ireland.,Science Foundation Ireland, Centre for Research in Medical Devices, National University of Ireland Galway, Galway, Ireland
| | - Dimitrios I Zeugolis
- Regenerative, Modular & Developmental Engineering Laboratory, National University of Ireland Galway, Galway, Ireland.,Science Foundation Ireland, Centre for Research in Medical Devices, National University of Ireland Galway, Galway, Ireland
| |
Collapse
|
13
|
Imbratta C, Hussein H, Andris F, Verdeil G. c-MAF, a Swiss Army Knife for Tolerance in Lymphocytes. Front Immunol 2020; 11:206. [PMID: 32117317 PMCID: PMC7033575 DOI: 10.3389/fimmu.2020.00206] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2019] [Accepted: 01/27/2020] [Indexed: 12/11/2022] Open
Abstract
Beyond its well-admitted role in development and organogenesis, it is now clear that the transcription factor c-Maf has owned its place in the realm of immune-related transcription factors. Formerly introduced solely as a Th2 transcription factor, the role attributed to c-Maf has gradually broadened over the years and has extended to most, if not all, known immune cell types. The influence of c-Maf is particularly prominent among T cell subsets, where c-Maf regulates the differentiation as well as the function of multiple subsets of CD4 and CD8 T cells, lending it a crucial position in adaptive immunity and anti-tumoral responsiveness. Recent research has also revealed the role of c-Maf in controlling Th17 responses in the intestine, positioning it as an essential factor in intestinal homeostasis. This review aims to present and discuss the recent advances highlighting the particular role played by c-Maf in T lymphocyte differentiation, function, and homeostasis.
Collapse
Affiliation(s)
- Claire Imbratta
- Department of Oncology, University of Lausanne, Lausanne, Switzerland
| | - Hind Hussein
- Laboratoire d'Immunobiologie, Université Libre de Bruxelles, Brussels, Belgium
| | - Fabienne Andris
- Laboratoire d'Immunobiologie, Université Libre de Bruxelles, Brussels, Belgium
| | - Grégory Verdeil
- Department of Oncology, University of Lausanne, Lausanne, Switzerland
| |
Collapse
|
14
|
Fujino M, Tagami A, Ojima M, Mizuno S, Abdellatif AM, Kuno A, Takahashi S. c-MAF deletion in adult C57BL/6J mice induces cataract formation and abnormal differentiation of lens fiber cells. Exp Anim 2020; 69:242-249. [PMID: 31969519 PMCID: PMC7220708 DOI: 10.1538/expanim.19-0137] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022] Open
Abstract
The transcription factor c-MAF is a member of the large MAF family, members of which possess transactivation and bZIP domains. c-MAF plays an important role in lens formation, T-lymphocyte differentiation, hypertrophic chondrocyte differentiation, and kidney development in mouse embryos. However, because homozygous deletion of c-Maf in C57BL/6J mice causes embryonic lethality, the functions of c-MAF in adult mice remain largely uninvestigated. To address this issue, we generated c-Maf floxed (c-Maffl/fl) C57BL/6J mice and established tamoxifen-inducible c-Maf knockout mice (c-Maffl/fl; CAG-Cre-ERTM mice, c-MafΔTAM). After tamoxifen injection, adult c-MafΔTAM mice showed successful deletion of c-Maf protein and developed severe cataracts; cataracts are also seen in human patients who have mutations in the c-MAF DNA binding domain. Furthermore, adult c-MafΔTAM mice exhibited abnormal lens structure and impaired differentiation of lens fiber cells. In summary, we established c-Maffl/fl and c-MafΔTAM C57BL/6J mice, which can be useful animal models for the investigation of c-MAF function in various developmental stages and can also be used as a disease model for cataracts.
Collapse
Affiliation(s)
- Mitsunori Fujino
- Department of Anatomy and Embryology, Faculty of Medicine, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki 305-8575, Japan.,Ph.D. Program in Human Biology, School of Integrative and Global Majors, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki 305-8575, Japan
| | - Asuka Tagami
- Department of Anatomy and Embryology, Faculty of Medicine, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki 305-8575, Japan
| | - Masami Ojima
- Laboratory Animal Resource Center (LARC), University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki 305-8575, Japan
| | - Seiya Mizuno
- Laboratory Animal Resource Center (LARC), University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki 305-8575, Japan
| | - Ahmed M Abdellatif
- Department of Anatomy and Embryology, Faculty of Veterinary Medicine, Mansoura University, Mansoura 35516, Egypt
| | - Akihiro Kuno
- Department of Anatomy and Embryology, Faculty of Medicine, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki 305-8575, Japan
| | - Satoru Takahashi
- Department of Anatomy and Embryology, Faculty of Medicine, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki 305-8575, Japan.,Laboratory Animal Resource Center (LARC), University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki 305-8575, Japan.,Life Science Center, Tsukuba Advanced Research Alliance (TARA), University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki 305-8575, Japan.,International Institute for Integrative Sleep Medicine (WPI-IIIS), University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki 305-8575, Japan.,Transborder Medical Research Center, Faculty of Medicine, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki 305-8575, Japan
| |
Collapse
|
15
|
Niceta M, Barbuti D, Gupta N, Ruggiero C, Tizzano EF, Graul‐Neumann L, Barresi S, Nishimura G, Valenzuela I, López‐Grondona F, Fernandez‐Alvarez P, Leoni C, Zweier C, Tzschach A, Stellacci E, Del Fattore A, Dallapiccola B, Zampino G, Tartaglia M. Skeletal abnormalities are common features in Aymé‐Gripp syndrome. Clin Genet 2019; 97:362-369. [DOI: 10.1111/cge.13651] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2019] [Revised: 09/09/2019] [Accepted: 09/27/2019] [Indexed: 12/31/2022]
Affiliation(s)
- Marcello Niceta
- Genetics and Rare Diseases Research DivisionOspedale Pediatrico Bambino Gesù Rome Italy
| | - Domenico Barbuti
- Radiologia e BioimagingOspedale Pediatrico Bambino Gesù Rome Italy
| | - Neerja Gupta
- Division of Genetics, Department of PediatricsAll Institute of Medical Sciences New Delhi India
| | | | - Eduardo F. Tizzano
- Department of Clinical and Molecular Genetics and Rare Diseases UnitUniversity Hospital Valld'Hebron, Medicine Genetics Group, VHIR Barcelona Spain
| | - Luitgard Graul‐Neumann
- Ambulantes Gesundheitszentrum HumangenetikCharité Universitäts medizin Berlin Berlin Germany
| | - Sabina Barresi
- Genetics and Rare Diseases Research DivisionOspedale Pediatrico Bambino Gesù Rome Italy
| | - Gen Nishimura
- Center for Intractable DiseasesSaitama Medical University Hospital Iruma Japan
| | - Irene Valenzuela
- Department of Clinical and Molecular Genetics and Rare Diseases UnitUniversity Hospital Valld'Hebron, Medicine Genetics Group, VHIR Barcelona Spain
| | - Fermina López‐Grondona
- Department of Clinical and Molecular Genetics and Rare Diseases UnitUniversity Hospital Valld'Hebron, Medicine Genetics Group, VHIR Barcelona Spain
| | - Paula Fernandez‐Alvarez
- Department of Clinical and Molecular Genetics and Rare Diseases UnitUniversity Hospital Valld'Hebron, Medicine Genetics Group, VHIR Barcelona Spain
| | - Chiara Leoni
- Center for Rare Diseases and Birth Defects, Department of Woman and Child Health and Pubblic HealthFondazione‐Policlinico‐Universitario‐A. Gemelli‐IRCCS Rome Italy
| | - Christiane Zweier
- Institute of Human GeneticsFriedrich‐Alexander‐Universität (FAU) Erlangen‐Nürnberg Erlangen Germany
| | - Andreas Tzschach
- Institute of Human Genetics FreiburgUniversity Hospital Freiburg Freiburg Germany
| | - Emilia Stellacci
- Dipartimento di Oncologia e Medicina MolecolareIstituto Superiore di Sanità Rome Italy
| | - Andrea Del Fattore
- Multifactorial Disease and Complex Phenotype Research AreaOspedale Pediatrico Bambino Gesù Rome Italy
| | - Bruno Dallapiccola
- Genetics and Rare Diseases Research DivisionOspedale Pediatrico Bambino Gesù Rome Italy
| | - Giuseppe Zampino
- Center for Rare Diseases and Birth Defects, Department of Woman and Child Health and Pubblic HealthFondazione‐Policlinico‐Universitario‐A. Gemelli‐IRCCS Rome Italy
- Istituto di PediatriaUniversità Cattolica del Sacro Cuore Rome Italy
| | - Marco Tartaglia
- Genetics and Rare Diseases Research DivisionOspedale Pediatrico Bambino Gesù Rome Italy
| |
Collapse
|
16
|
Antagonism of BMP signaling is insufficient to induce fibrous differentiation in primary sclerotome. Exp Cell Res 2019; 378:11-20. [PMID: 30817928 PMCID: PMC6501840 DOI: 10.1016/j.yexcr.2019.01.026] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2018] [Revised: 01/04/2019] [Accepted: 01/18/2019] [Indexed: 01/29/2023]
Abstract
Sclerotome is the embryonic progenitor of the axial skeleton. It was previously shown that Tgfbr2 is required in sclerotome for differentiation of fibrous skeletal tissues including the annulus fibrosus of the intervertebral disc. Alternatively, BMP signaling is required to form the vertebral body through chondrogenesis. In addition, TGFβ added to sclerotome cultures induces expression of markers for fibrous tissue differentiation but not cartilage or bone. The mechanism of how TGFβ signaling regulates this lineage decision in sclerotome is not known and could be due to the production of instructive or inhibitory signals or a combination of the two. Here we show that TGFβ antagonizes BMP/ Smad1/5 signaling in primary sclerotome likely through regulation of Noggin, an extracellular BMP antagonist, to prevent chondrogenesis. We then tested whether inhibition of BMP signaling, and inhibition of chondrogenesis, is sufficient to push cells toward the fibrous cell fate. While Noggin inhibited BMP/ Smad1/5 signaling and the formation of chondrogenic nodules in sclerotome cultures; Noggin and inhibition of BMP signaling through Gremlin or DMH2 were insufficient to induce fibrous tissue differentiation. The results suggest inhibition of BMP signaling is not sufficient to stimulate fibrous tissue differentiation and additional signals are likely required. We propose that TGFβ has a dual role in regulating sclerotome fate. First, it inhibits BMP signaling potentially through Noggin to prevent chondrogenesis and, second, it provides an unknown instructive signal to promote fibrous tissue differentiation in sclerotome. The results have implications for the design of stem cell-based therapies for skeletal diseases.
Collapse
|
17
|
Anand D, Agrawal SA, Slavotinek A, Lachke SA. Mutation update of transcription factor genes FOXE3, HSF4, MAF, and PITX3 causing cataracts and other developmental ocular defects. Hum Mutat 2018; 39:471-494. [PMID: 29314435 PMCID: PMC5839989 DOI: 10.1002/humu.23395] [Citation(s) in RCA: 53] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2017] [Revised: 12/19/2017] [Accepted: 12/22/2017] [Indexed: 02/06/2023]
Abstract
Mutations in the transcription factor genes FOXE3, HSF4, MAF, and PITX3 cause congenital lens defects including cataracts that may be accompanied by defects in other components of the eye or in nonocular tissues. We comprehensively describe here all the variants in FOXE3, HSF4, MAF, and PITX3 genes linked to human developmental defects. A total of 52 variants for FOXE3, 18 variants for HSF4, 20 variants for MAF, and 19 variants for PITX3 identified so far in isolated cases or within families are documented. This effort reveals FOXE3, HSF4, MAF, and PITX3 to have 33, 16, 18, and 7 unique causal mutations, respectively. Loss-of-function mutant animals for these genes have served to model the pathobiology of the associated human defects, and we discuss the currently known molecular function of these genes, particularly with emphasis on their role in ocular development. Finally, we make the detailed FOXE3, HSF4, MAF, and PITX3 variant information available in the Leiden Online Variation Database (LOVD) platform at https://www.LOVD.nl/FOXE3, https://www.LOVD.nl/HSF4, https://www.LOVD.nl/MAF, and https://www.LOVD.nl/PITX3. Thus, this article informs on key variants in transcription factor genes linked to cataract, aphakia, corneal opacity, glaucoma, microcornea, microphthalmia, anterior segment mesenchymal dysgenesis, and Ayme-Gripp syndrome, and facilitates their access through Web-based databases.
Collapse
Affiliation(s)
- Deepti Anand
- Department of Biological Sciences, University of Delaware, Newark, DE 19716 USA
| | - Smriti A. Agrawal
- Department of Biological Sciences, University of Delaware, Newark, DE 19716 USA
| | - Anne Slavotinek
- Department of Pediatrics, Division of Genetics, University of California, UCSF Benioff Children’s Hospital, San Francisco, CA 19716 USA
| | - Salil A. Lachke
- Department of Biological Sciences, University of Delaware, Newark, DE 19716 USA
- Center for Bioinformatics and Computational Biology, University of Delaware, Newark, DE 19711 USA
| |
Collapse
|
18
|
Liu CF, Samsa WE, Zhou G, Lefebvre V. Transcriptional control of chondrocyte specification and differentiation. Semin Cell Dev Biol 2016; 62:34-49. [PMID: 27771362 DOI: 10.1016/j.semcdb.2016.10.004] [Citation(s) in RCA: 103] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2016] [Accepted: 10/18/2016] [Indexed: 12/20/2022]
Abstract
A milestone in the evolutionary emergence of vertebrates was the invention of cartilage, a tissue that has key roles in modeling, protecting and complementing the bony skeleton. Cartilage is elaborated and maintained by chondrocytes. These cells derive from multipotent skeletal progenitors and they perform highly specialized functions as they proceed through sequential lineage commitment and differentiation steps. They form cartilage primordia, the primary skeleton of the embryo. They then transform these primordia either into cartilage growth plates, temporary drivers of skeletal elongation and endochondral ossification, or into permanent tissues, namely articular cartilage. Chondrocyte fate decisions and differentiated activities are controlled by numerous extrinsic and intrinsic cues, and they are implemented at the gene expression level by transcription factors. The latter are the focus of this review. Meritorious efforts from many research groups have led over the last two decades to the identification of dozens of key chondrogenic transcription factors. These regulators belong to all types of transcription factor families. Some have master roles at one or several differentiation steps. They include SOX9 and RUNX2/3. Others decisively assist or antagonize the activities of these masters. They include TWIST1, SOX5/6, and MEF2C/D. Many more have tissue-patterning roles and regulate cell survival, proliferation and the pace of cell differentiation. They include, but are not limited to, homeodomain-containing proteins and growth factor signaling mediators. We here review current knowledge of all these factors, one superclass, class, and family at a time. We then compile all knowledge into transcriptional networks. We also identify remaining gaps in knowledge and directions for future research to fill these gaps and thereby provide novel insights into cartilage disease mechanisms and treatment options.
Collapse
Affiliation(s)
- Chia-Feng Liu
- Department of Cellular and Molecular Medicine, Cleveland Clinic Lerner Research Institute, Cleveland, OH, 44195, USA.
| | - William E Samsa
- Department of Orthopaedics, Case Western Reserve University, Cleveland, OH, USA
| | - Guang Zhou
- Department of Orthopaedics, Case Western Reserve University, Cleveland, OH, USA; Department of Genetics and Genome Sciences, Case Western Reserve University, Cleveland, OH, USA; Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, OH, USA
| | - Véronique Lefebvre
- Department of Cellular and Molecular Medicine, Cleveland Clinic Lerner Research Institute, Cleveland, OH, 44195, USA.
| |
Collapse
|
19
|
Hogrebe NJ, Gooch KJ. Direct influence of culture dimensionality on human mesenchymal stem cell differentiation at various matrix stiffnesses using a fibrous self‐assembling peptide hydrogel. J Biomed Mater Res A 2016; 104:2356-68. [DOI: 10.1002/jbm.a.35755] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2016] [Accepted: 04/19/2016] [Indexed: 11/12/2022]
Affiliation(s)
| | - Keith J. Gooch
- Department of Biomedical EngineeringThe Ohio State UniversityColumbus Ohio
- The Ohio State University, Davis Heart Lung Research InstituteColumbus Ohio
| |
Collapse
|
20
|
Davudian S, Mansoori B, Shajari N, Mohammadi A, Baradaran B. BACH1, the master regulator gene: A novel candidate target for cancer therapy. Gene 2016; 588:30-7. [PMID: 27108804 DOI: 10.1016/j.gene.2016.04.040] [Citation(s) in RCA: 66] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2016] [Revised: 03/28/2016] [Accepted: 04/20/2016] [Indexed: 01/17/2023]
Abstract
BACH1 (BTB and CNC homology 1, basic leucine zipper transcription factor 1) is a transcriptional factor and a member of cap 'n' collar (CNC) and basic region leucine zipper factor family. In contrast to other bZIP family members, BACH1 appeared as a comparatively specific transcription factor. It acts as transcription regulator and is recognized as a recently hypoxia regulator and functions as an inducible repressor for the HO-1 gene in many human cell types in response to stress oxidative. In regard to studies lately, although, BACH1 has been related to the regulation of oxidative stress and heme oxidation, it has never been linked to invasion and metastasis. Recent studies have showed that BACH1 is involved in bone metastasis of breast cancer by up-regulating vital metastatic genes like CXCR4 and MMP1. This newly discovered aspect of BACH1 gene provides new insight into cancer progression study and stands on its master regulator role in metastasis process, raising the possibility of considering it as a potential target for cancer therapy.
Collapse
Affiliation(s)
- Sadaf Davudian
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Behzad Mansoori
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Neda Shajari
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Ali Mohammadi
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Behzad Baradaran
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
21
|
Li H, Hao Z, Zhao L, Liu W, Han Y, Bai Y, Wang J. Comparison of molecular mechanisms of rheumatoid arthritis and osteoarthritis using gene microarrays. Mol Med Rep 2016; 13:4599-605. [PMID: 27082252 PMCID: PMC4878540 DOI: 10.3892/mmr.2016.5144] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2015] [Accepted: 12/11/2015] [Indexed: 01/12/2023] Open
Abstract
The present study aimed to compare the molecular mechanisms of rheumatoid arthritis (RA) and osteoarthritis (OA). The microarray dataset no. GSE29746 was downloaded from Gene Expression Omnibus. After data pre‑processing, differential expression analysis between the RA group and the control, as well as between the OA group and the control was performed using the LIMMA package in R and differentially expressed transcripts (DETs) with |log2fold change (FC)|>1 and P<0.01 were identified. DETs screened from each disease group were then subjected to functional annotation using DAVID. Next, DETs from each group were used to construct individual interaction networks using the BIND database, followed by sub‑network mining using clusterONE. Significant functions of nodes in each sub‑network were also investigated. In total, 19 and 281 DETs were screened from the RA and OA groups, respectively, with only six common DETs. DETs from the RA and OA groups were enriched in 8 and 130 gene ontology (GO) terms, respectively, with four common GO terms, of which to were associated with phospholipase C (PLC) activity. In addition, DETs screened from the OA group were enriched in immune response‑associated GO terms, and those screened from the RA group were largely associated with biological processes linked with the cell cycle and chromosomes. Genes involved in PLC activity and its regulation were indicated to be altered in RA as well as in OA. Alterations in the expression of cell cycle‑associated genes were indicated to be linked with the occurrence of OA, while genes participating in the immune response were involved in the occurrence of RA.
Collapse
Affiliation(s)
- Hongqiang Li
- Department of Orthopedics, The Harbin Fifth Hospital, Harbin, Heilongjiang 150001, P.R. China
| | - Zhenyong Hao
- Department of Orthopedics, The Harbin Fifth Hospital, Harbin, Heilongjiang 150001, P.R. China
| | - Liqiang Zhao
- Department of Orthopedics, The Harbin Fifth Hospital, Harbin, Heilongjiang 150001, P.R. China
| | - Wei Liu
- Department of Orthopedics, The Harbin Fifth Hospital, Harbin, Heilongjiang 150001, P.R. China
| | - Yanlong Han
- Department of Orthopedics, The Harbin Fifth Hospital, Harbin, Heilongjiang 150001, P.R. China
| | - Yunxing Bai
- Department of Orthopedics, The Harbin Fifth Hospital, Harbin, Heilongjiang 150001, P.R. China
| | - Jian Wang
- Department of Orthopedics, The Harbin Fifth Hospital, Harbin, Heilongjiang 150001, P.R. China
| |
Collapse
|
22
|
Yang Y, Cvekl A. Large Maf Transcription Factors: Cousins of AP-1 Proteins and Important Regulators of Cellular Differentiation. ACTA ACUST UNITED AC 2016; 23:2-11. [PMID: 18159220 DOI: 10.23861/ejbm20072347] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
A large number of mammalian transcription factors possess the evolutionary conserved basic and leucine zipper domain (bZIP). The basic domain interacts with DNA while the leucine zipper facilitates homo- and hetero-dimerization. These factors can be grouped into at least seven families: AP-1, ATF/CREB, CNC, C/EBP, Maf, PAR, and virus-encoded bZIPs. Here, we focus on a group of four large Maf proteins: MafA, MafB, c-Maf, and NRL. They act as key regulators of terminal differentiation in many tissues such as bone, brain, kidney, lens, pancreas, and retina, as well as in blood. The DNA-binding mechanism of large Mafs involves cooperation between the basic domain and an adjacent ancillary DNA-binding domain. Many genes regulated by Mafs during cellular differentiation use functional interactions between the Pax/Maf, Sox/Maf, and Ets/Maf promoter and enhancer modules. The prime examples are crystallin genes in lens and glucagon and insulin in pancreas. Novel roles for large Mafs emerged from studying generations of MafA and MafB knockouts and analysis of combined phenotypes in double or triple null mice. In addition, studies of this group of factors in invertebrates revealed the evolutionarily conserved function of these genes in the development of multicellular organisms.
Collapse
Affiliation(s)
- Ying Yang
- Departments of Ophthalmology and Visual Sciences and Molecular Genetics, Albert Einstein College of Medicine, Bronx, New York 10461
| | | |
Collapse
|
23
|
Zhang Y, Sun H, Liu Y, Chen S, Cai S, Zhu Y, Guo P. The Limbal Epithelial Progenitors in the Limbal Niche Environment. Int J Med Sci 2016; 13:835-840. [PMID: 27877075 PMCID: PMC5118754 DOI: 10.7150/ijms.16563] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/21/2016] [Accepted: 09/01/2016] [Indexed: 02/06/2023] Open
Abstract
Limbal epithelial progenitors are stem cells located in limbal palisades of vogt. In this review, we present the audience with recent evidence that limbal epithelial progenitors may be a powerful stem cell resource for the cure of human corneal stem cell deficiency. Further understanding of their mechanism may shed lights to the future successful application of stem cell therapy not only to the eye tissue, but also to the other tissues in the human body.
Collapse
Affiliation(s)
- Yuan Zhang
- Research and Development Department, TissueTech, Inc., 7000 SW 97th Avenue, Suite 212, Miami, FL 33173, USA
| | - Hong Sun
- Department of Ophthalmology, the First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
| | - Yongsong Liu
- Department of Ophthalmology, Yan' An Hospital of Kunming City, Kunming, 650051, China
| | - Shuangling Chen
- Research and Development Department, TissueTech, Inc., 7000 SW 97th Avenue, Suite 212, Miami, FL 33173, USA
| | - Subo Cai
- Department of Ophthalmology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Yingting Zhu
- Research and Development Department, TissueTech, Inc., 7000 SW 97th Avenue, Suite 212, Miami, FL 33173, USA
| | - Ping Guo
- Shenzhen Eye Hospital, School of Optometry & Ophthalmology of Shenzhen University, Shenzhen Key Laboratory of Department of Ophthalmology, Shenzhen, 518000, China
| |
Collapse
|
24
|
Xie Q, McGreal R, Harris R, Gao CY, Liu W, Reneker LW, Musil LS, Cvekl A. Regulation of c-Maf and αA-Crystallin in Ocular Lens by Fibroblast Growth Factor Signaling. J Biol Chem 2015; 291:3947-58. [PMID: 26719333 DOI: 10.1074/jbc.m115.705103] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2015] [Indexed: 12/20/2022] Open
Abstract
Fibroblast growth factor (FGF) signaling regulates a multitude of cellular processes, including cell proliferation, survival, migration, and differentiation. In the vertebrate lens, FGF signaling regulates fiber cell differentiation characterized by high expression of crystallin proteins. However, a direct link between FGF signaling and crystallin gene transcriptional machinery remains to be established. Previously, we have shown that the bZIP proto-oncogene c-Maf regulates expression of αA-crystallin (Cryaa) through binding to its promoter and distal enhancer, DCR1, both activated by FGF2 in cell culture. Herein, we identified and characterized a novel FGF2-responsive region in the c-Maf promoter (-272/-70, FRE). Both c-Maf and Cryaa regulatory regions contain arrays of AP-1 and Ets-binding sites. Chromatin immunoprecipitation (ChIP) assays established binding of c-Jun (an AP-1 factor) and Etv5/ERM (an Ets factor) to these regions in lens chromatin. Analysis of temporal and spatial expression of c-Jun, phospho-c-Jun, and Etv5/ERM in wild type and ERK1/2 deficient lenses supports their roles as nuclear effectors of FGF signaling in mouse embryonic lens. Collectively, these studies show that FGF signaling up-regulates expression of αA-crystallin both directly and indirectly via up-regulation of c-Maf. These molecular mechanisms are applicable for other crystallins and genes highly expressed in terminally differentiated lens fibers.
Collapse
Affiliation(s)
- Qing Xie
- From the Departments of Ophthalmology and Visual Sciences and Genetics, Albert Einstein College of Medicine, Bronx, New York 10461
| | - Rebecca McGreal
- From the Departments of Ophthalmology and Visual Sciences and
| | - Raven Harris
- Genetics, Albert Einstein College of Medicine, Bronx, New York 10461
| | - Chun Y Gao
- Laboratory of Molecular and Developmental Biology, National Eye Institute, Bethesda, Maryland 20892
| | - Wei Liu
- From the Departments of Ophthalmology and Visual Sciences and Genetics, Albert Einstein College of Medicine, Bronx, New York 10461
| | - Lixing W Reneker
- Department of Ophthalmology, Mason Eye Institute, University of Missouri, Columbia, Missouri 65212, and
| | - Linda S Musil
- Department of Biochemistry and Molecular Biology, Oregon Health Science University, Portland, Oregon 97239
| | - Ales Cvekl
- From the Departments of Ophthalmology and Visual Sciences and Genetics, Albert Einstein College of Medicine, Bronx, New York 10461,
| |
Collapse
|
25
|
Pavlovic M, Arnal-Estapé A, Rojo F, Bellmunt A, Tarragona M, Guiu M, Planet E, Garcia-Albéniz X, Morales M, Urosevic J, Gawrzak S, Rovira A, Prat A, Nonell L, Lluch A, Jean-Mairet J, Coleman R, Albanell J, Gomis RR. Enhanced MAF Oncogene Expression and Breast Cancer Bone Metastasis. J Natl Cancer Inst 2015; 107:djv256. [PMID: 26376684 PMCID: PMC4681582 DOI: 10.1093/jnci/djv256] [Citation(s) in RCA: 74] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2014] [Accepted: 08/18/2015] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND There are currently no biomarkers for early breast cancer patient populations at risk of bone metastasis. Identification of mediators of bone metastasis could be of clinical interest. METHODS A de novo unbiased screening approach based on selection of highly bone metastatic breast cancer cells in vivo was used to determine copy number aberrations (CNAs) associated with bone metastasis. The CNAs associated with bone metastasis were examined in independent primary breast cancer datasets with annotated clinical follow-up. The MAF gene encoded within the CNA associated with bone metastasis was subjected to gain and loss of function validation in breast cancer cells (MCF7, T47D, ZR-75, and 4T1), its downstream mechanism validated, and tested in clinical samples. A multivariable Cox cause-specific hazard model with competing events (death) was used to test the association between 16q23 or MAF and bone metastasis. All statistical tests were two-sided. RESULTS 16q23 gain CNA encoding the transcription factor MAF mediates breast cancer bone metastasis through the control of PTHrP. 16q23 gain (hazard ratio (HR) for bone metastasis = 14.5, 95% confidence interval (CI) = 6.4 to 32.9, P < .001) as well as MAF overexpression (HR for bone metastasis = 2.5, 95% CI = 1.7 to 3.8, P < .001) in primary breast tumors were specifically associated with risk of metastasis to bone but not to other organs. CONCLUSIONS These results suggest that MAF is a mediator of breast cancer bone metastasis. 16q23 gain or MAF protein overexpression in tumors may help to select patients at risk of bone relapse.
Collapse
Affiliation(s)
- Milica Pavlovic
- Oncology Program (MP, AAE, AB, MT, MG, XGA, MM, JU, SG, RRG) and Biostatistics and Bioinformatics Unit (EP), Institute for Research in Biomedicine (IRB Barcelona), Barcelona, Spain; Cancer Research Program (FR, AR, JA) and Microarray Analysis Service (LN), IMIM (Hospital del Mar Medical Research Institute), Barcelona, Spain; Pathology Department, IIS-Fundación Jimenez Diaz, Madrid, Spain (FR); Medical Oncology Service, Hospital del Mar, Barcelona, Spain (AR, JA); Department of Oncology and Hematology, Hospital Clínico Universitario, Valencia, Spain (AL); Valencia Central University, Spain (AL); Inbiomotion, Barcelona, Spain (JJM); Sheffield Cancer Research Centre, Sheffield, UK (RC); Universitat Pompeu Fabra, Barcelona, Spain (JA); Translational Genomics, Vall d'Hebron Insitute of Oncology, Barcelona, Spain (AP); Department of Epidemiology, Harvard School of Public Health, Boston, MA (XGA); Institució Catalana de Recerca i Estudis Avançats (ICREA), Barcelona, Spain (RRG)
| | - Anna Arnal-Estapé
- Oncology Program (MP, AAE, AB, MT, MG, XGA, MM, JU, SG, RRG) and Biostatistics and Bioinformatics Unit (EP), Institute for Research in Biomedicine (IRB Barcelona), Barcelona, Spain; Cancer Research Program (FR, AR, JA) and Microarray Analysis Service (LN), IMIM (Hospital del Mar Medical Research Institute), Barcelona, Spain; Pathology Department, IIS-Fundación Jimenez Diaz, Madrid, Spain (FR); Medical Oncology Service, Hospital del Mar, Barcelona, Spain (AR, JA); Department of Oncology and Hematology, Hospital Clínico Universitario, Valencia, Spain (AL); Valencia Central University, Spain (AL); Inbiomotion, Barcelona, Spain (JJM); Sheffield Cancer Research Centre, Sheffield, UK (RC); Universitat Pompeu Fabra, Barcelona, Spain (JA); Translational Genomics, Vall d'Hebron Insitute of Oncology, Barcelona, Spain (AP); Department of Epidemiology, Harvard School of Public Health, Boston, MA (XGA); Institució Catalana de Recerca i Estudis Avançats (ICREA), Barcelona, Spain (RRG)
| | - Federico Rojo
- Oncology Program (MP, AAE, AB, MT, MG, XGA, MM, JU, SG, RRG) and Biostatistics and Bioinformatics Unit (EP), Institute for Research in Biomedicine (IRB Barcelona), Barcelona, Spain; Cancer Research Program (FR, AR, JA) and Microarray Analysis Service (LN), IMIM (Hospital del Mar Medical Research Institute), Barcelona, Spain; Pathology Department, IIS-Fundación Jimenez Diaz, Madrid, Spain (FR); Medical Oncology Service, Hospital del Mar, Barcelona, Spain (AR, JA); Department of Oncology and Hematology, Hospital Clínico Universitario, Valencia, Spain (AL); Valencia Central University, Spain (AL); Inbiomotion, Barcelona, Spain (JJM); Sheffield Cancer Research Centre, Sheffield, UK (RC); Universitat Pompeu Fabra, Barcelona, Spain (JA); Translational Genomics, Vall d'Hebron Insitute of Oncology, Barcelona, Spain (AP); Department of Epidemiology, Harvard School of Public Health, Boston, MA (XGA); Institució Catalana de Recerca i Estudis Avançats (ICREA), Barcelona, Spain (RRG)
| | - Anna Bellmunt
- Oncology Program (MP, AAE, AB, MT, MG, XGA, MM, JU, SG, RRG) and Biostatistics and Bioinformatics Unit (EP), Institute for Research in Biomedicine (IRB Barcelona), Barcelona, Spain; Cancer Research Program (FR, AR, JA) and Microarray Analysis Service (LN), IMIM (Hospital del Mar Medical Research Institute), Barcelona, Spain; Pathology Department, IIS-Fundación Jimenez Diaz, Madrid, Spain (FR); Medical Oncology Service, Hospital del Mar, Barcelona, Spain (AR, JA); Department of Oncology and Hematology, Hospital Clínico Universitario, Valencia, Spain (AL); Valencia Central University, Spain (AL); Inbiomotion, Barcelona, Spain (JJM); Sheffield Cancer Research Centre, Sheffield, UK (RC); Universitat Pompeu Fabra, Barcelona, Spain (JA); Translational Genomics, Vall d'Hebron Insitute of Oncology, Barcelona, Spain (AP); Department of Epidemiology, Harvard School of Public Health, Boston, MA (XGA); Institució Catalana de Recerca i Estudis Avançats (ICREA), Barcelona, Spain (RRG)
| | - Maria Tarragona
- Oncology Program (MP, AAE, AB, MT, MG, XGA, MM, JU, SG, RRG) and Biostatistics and Bioinformatics Unit (EP), Institute for Research in Biomedicine (IRB Barcelona), Barcelona, Spain; Cancer Research Program (FR, AR, JA) and Microarray Analysis Service (LN), IMIM (Hospital del Mar Medical Research Institute), Barcelona, Spain; Pathology Department, IIS-Fundación Jimenez Diaz, Madrid, Spain (FR); Medical Oncology Service, Hospital del Mar, Barcelona, Spain (AR, JA); Department of Oncology and Hematology, Hospital Clínico Universitario, Valencia, Spain (AL); Valencia Central University, Spain (AL); Inbiomotion, Barcelona, Spain (JJM); Sheffield Cancer Research Centre, Sheffield, UK (RC); Universitat Pompeu Fabra, Barcelona, Spain (JA); Translational Genomics, Vall d'Hebron Insitute of Oncology, Barcelona, Spain (AP); Department of Epidemiology, Harvard School of Public Health, Boston, MA (XGA); Institució Catalana de Recerca i Estudis Avançats (ICREA), Barcelona, Spain (RRG)
| | - Marc Guiu
- Oncology Program (MP, AAE, AB, MT, MG, XGA, MM, JU, SG, RRG) and Biostatistics and Bioinformatics Unit (EP), Institute for Research in Biomedicine (IRB Barcelona), Barcelona, Spain; Cancer Research Program (FR, AR, JA) and Microarray Analysis Service (LN), IMIM (Hospital del Mar Medical Research Institute), Barcelona, Spain; Pathology Department, IIS-Fundación Jimenez Diaz, Madrid, Spain (FR); Medical Oncology Service, Hospital del Mar, Barcelona, Spain (AR, JA); Department of Oncology and Hematology, Hospital Clínico Universitario, Valencia, Spain (AL); Valencia Central University, Spain (AL); Inbiomotion, Barcelona, Spain (JJM); Sheffield Cancer Research Centre, Sheffield, UK (RC); Universitat Pompeu Fabra, Barcelona, Spain (JA); Translational Genomics, Vall d'Hebron Insitute of Oncology, Barcelona, Spain (AP); Department of Epidemiology, Harvard School of Public Health, Boston, MA (XGA); Institució Catalana de Recerca i Estudis Avançats (ICREA), Barcelona, Spain (RRG)
| | - Evarist Planet
- Oncology Program (MP, AAE, AB, MT, MG, XGA, MM, JU, SG, RRG) and Biostatistics and Bioinformatics Unit (EP), Institute for Research in Biomedicine (IRB Barcelona), Barcelona, Spain; Cancer Research Program (FR, AR, JA) and Microarray Analysis Service (LN), IMIM (Hospital del Mar Medical Research Institute), Barcelona, Spain; Pathology Department, IIS-Fundación Jimenez Diaz, Madrid, Spain (FR); Medical Oncology Service, Hospital del Mar, Barcelona, Spain (AR, JA); Department of Oncology and Hematology, Hospital Clínico Universitario, Valencia, Spain (AL); Valencia Central University, Spain (AL); Inbiomotion, Barcelona, Spain (JJM); Sheffield Cancer Research Centre, Sheffield, UK (RC); Universitat Pompeu Fabra, Barcelona, Spain (JA); Translational Genomics, Vall d'Hebron Insitute of Oncology, Barcelona, Spain (AP); Department of Epidemiology, Harvard School of Public Health, Boston, MA (XGA); Institució Catalana de Recerca i Estudis Avançats (ICREA), Barcelona, Spain (RRG)
| | - Xabier Garcia-Albéniz
- Oncology Program (MP, AAE, AB, MT, MG, XGA, MM, JU, SG, RRG) and Biostatistics and Bioinformatics Unit (EP), Institute for Research in Biomedicine (IRB Barcelona), Barcelona, Spain; Cancer Research Program (FR, AR, JA) and Microarray Analysis Service (LN), IMIM (Hospital del Mar Medical Research Institute), Barcelona, Spain; Pathology Department, IIS-Fundación Jimenez Diaz, Madrid, Spain (FR); Medical Oncology Service, Hospital del Mar, Barcelona, Spain (AR, JA); Department of Oncology and Hematology, Hospital Clínico Universitario, Valencia, Spain (AL); Valencia Central University, Spain (AL); Inbiomotion, Barcelona, Spain (JJM); Sheffield Cancer Research Centre, Sheffield, UK (RC); Universitat Pompeu Fabra, Barcelona, Spain (JA); Translational Genomics, Vall d'Hebron Insitute of Oncology, Barcelona, Spain (AP); Department of Epidemiology, Harvard School of Public Health, Boston, MA (XGA); Institució Catalana de Recerca i Estudis Avançats (ICREA), Barcelona, Spain (RRG)
| | - Mónica Morales
- Oncology Program (MP, AAE, AB, MT, MG, XGA, MM, JU, SG, RRG) and Biostatistics and Bioinformatics Unit (EP), Institute for Research in Biomedicine (IRB Barcelona), Barcelona, Spain; Cancer Research Program (FR, AR, JA) and Microarray Analysis Service (LN), IMIM (Hospital del Mar Medical Research Institute), Barcelona, Spain; Pathology Department, IIS-Fundación Jimenez Diaz, Madrid, Spain (FR); Medical Oncology Service, Hospital del Mar, Barcelona, Spain (AR, JA); Department of Oncology and Hematology, Hospital Clínico Universitario, Valencia, Spain (AL); Valencia Central University, Spain (AL); Inbiomotion, Barcelona, Spain (JJM); Sheffield Cancer Research Centre, Sheffield, UK (RC); Universitat Pompeu Fabra, Barcelona, Spain (JA); Translational Genomics, Vall d'Hebron Insitute of Oncology, Barcelona, Spain (AP); Department of Epidemiology, Harvard School of Public Health, Boston, MA (XGA); Institució Catalana de Recerca i Estudis Avançats (ICREA), Barcelona, Spain (RRG)
| | - Jelena Urosevic
- Oncology Program (MP, AAE, AB, MT, MG, XGA, MM, JU, SG, RRG) and Biostatistics and Bioinformatics Unit (EP), Institute for Research in Biomedicine (IRB Barcelona), Barcelona, Spain; Cancer Research Program (FR, AR, JA) and Microarray Analysis Service (LN), IMIM (Hospital del Mar Medical Research Institute), Barcelona, Spain; Pathology Department, IIS-Fundación Jimenez Diaz, Madrid, Spain (FR); Medical Oncology Service, Hospital del Mar, Barcelona, Spain (AR, JA); Department of Oncology and Hematology, Hospital Clínico Universitario, Valencia, Spain (AL); Valencia Central University, Spain (AL); Inbiomotion, Barcelona, Spain (JJM); Sheffield Cancer Research Centre, Sheffield, UK (RC); Universitat Pompeu Fabra, Barcelona, Spain (JA); Translational Genomics, Vall d'Hebron Insitute of Oncology, Barcelona, Spain (AP); Department of Epidemiology, Harvard School of Public Health, Boston, MA (XGA); Institució Catalana de Recerca i Estudis Avançats (ICREA), Barcelona, Spain (RRG)
| | - Sylwia Gawrzak
- Oncology Program (MP, AAE, AB, MT, MG, XGA, MM, JU, SG, RRG) and Biostatistics and Bioinformatics Unit (EP), Institute for Research in Biomedicine (IRB Barcelona), Barcelona, Spain; Cancer Research Program (FR, AR, JA) and Microarray Analysis Service (LN), IMIM (Hospital del Mar Medical Research Institute), Barcelona, Spain; Pathology Department, IIS-Fundación Jimenez Diaz, Madrid, Spain (FR); Medical Oncology Service, Hospital del Mar, Barcelona, Spain (AR, JA); Department of Oncology and Hematology, Hospital Clínico Universitario, Valencia, Spain (AL); Valencia Central University, Spain (AL); Inbiomotion, Barcelona, Spain (JJM); Sheffield Cancer Research Centre, Sheffield, UK (RC); Universitat Pompeu Fabra, Barcelona, Spain (JA); Translational Genomics, Vall d'Hebron Insitute of Oncology, Barcelona, Spain (AP); Department of Epidemiology, Harvard School of Public Health, Boston, MA (XGA); Institució Catalana de Recerca i Estudis Avançats (ICREA), Barcelona, Spain (RRG)
| | - Ana Rovira
- Oncology Program (MP, AAE, AB, MT, MG, XGA, MM, JU, SG, RRG) and Biostatistics and Bioinformatics Unit (EP), Institute for Research in Biomedicine (IRB Barcelona), Barcelona, Spain; Cancer Research Program (FR, AR, JA) and Microarray Analysis Service (LN), IMIM (Hospital del Mar Medical Research Institute), Barcelona, Spain; Pathology Department, IIS-Fundación Jimenez Diaz, Madrid, Spain (FR); Medical Oncology Service, Hospital del Mar, Barcelona, Spain (AR, JA); Department of Oncology and Hematology, Hospital Clínico Universitario, Valencia, Spain (AL); Valencia Central University, Spain (AL); Inbiomotion, Barcelona, Spain (JJM); Sheffield Cancer Research Centre, Sheffield, UK (RC); Universitat Pompeu Fabra, Barcelona, Spain (JA); Translational Genomics, Vall d'Hebron Insitute of Oncology, Barcelona, Spain (AP); Department of Epidemiology, Harvard School of Public Health, Boston, MA (XGA); Institució Catalana de Recerca i Estudis Avançats (ICREA), Barcelona, Spain (RRG)
| | - Aleix Prat
- Oncology Program (MP, AAE, AB, MT, MG, XGA, MM, JU, SG, RRG) and Biostatistics and Bioinformatics Unit (EP), Institute for Research in Biomedicine (IRB Barcelona), Barcelona, Spain; Cancer Research Program (FR, AR, JA) and Microarray Analysis Service (LN), IMIM (Hospital del Mar Medical Research Institute), Barcelona, Spain; Pathology Department, IIS-Fundación Jimenez Diaz, Madrid, Spain (FR); Medical Oncology Service, Hospital del Mar, Barcelona, Spain (AR, JA); Department of Oncology and Hematology, Hospital Clínico Universitario, Valencia, Spain (AL); Valencia Central University, Spain (AL); Inbiomotion, Barcelona, Spain (JJM); Sheffield Cancer Research Centre, Sheffield, UK (RC); Universitat Pompeu Fabra, Barcelona, Spain (JA); Translational Genomics, Vall d'Hebron Insitute of Oncology, Barcelona, Spain (AP); Department of Epidemiology, Harvard School of Public Health, Boston, MA (XGA); Institució Catalana de Recerca i Estudis Avançats (ICREA), Barcelona, Spain (RRG)
| | - Lara Nonell
- Oncology Program (MP, AAE, AB, MT, MG, XGA, MM, JU, SG, RRG) and Biostatistics and Bioinformatics Unit (EP), Institute for Research in Biomedicine (IRB Barcelona), Barcelona, Spain; Cancer Research Program (FR, AR, JA) and Microarray Analysis Service (LN), IMIM (Hospital del Mar Medical Research Institute), Barcelona, Spain; Pathology Department, IIS-Fundación Jimenez Diaz, Madrid, Spain (FR); Medical Oncology Service, Hospital del Mar, Barcelona, Spain (AR, JA); Department of Oncology and Hematology, Hospital Clínico Universitario, Valencia, Spain (AL); Valencia Central University, Spain (AL); Inbiomotion, Barcelona, Spain (JJM); Sheffield Cancer Research Centre, Sheffield, UK (RC); Universitat Pompeu Fabra, Barcelona, Spain (JA); Translational Genomics, Vall d'Hebron Insitute of Oncology, Barcelona, Spain (AP); Department of Epidemiology, Harvard School of Public Health, Boston, MA (XGA); Institució Catalana de Recerca i Estudis Avançats (ICREA), Barcelona, Spain (RRG)
| | - Ana Lluch
- Oncology Program (MP, AAE, AB, MT, MG, XGA, MM, JU, SG, RRG) and Biostatistics and Bioinformatics Unit (EP), Institute for Research in Biomedicine (IRB Barcelona), Barcelona, Spain; Cancer Research Program (FR, AR, JA) and Microarray Analysis Service (LN), IMIM (Hospital del Mar Medical Research Institute), Barcelona, Spain; Pathology Department, IIS-Fundación Jimenez Diaz, Madrid, Spain (FR); Medical Oncology Service, Hospital del Mar, Barcelona, Spain (AR, JA); Department of Oncology and Hematology, Hospital Clínico Universitario, Valencia, Spain (AL); Valencia Central University, Spain (AL); Inbiomotion, Barcelona, Spain (JJM); Sheffield Cancer Research Centre, Sheffield, UK (RC); Universitat Pompeu Fabra, Barcelona, Spain (JA); Translational Genomics, Vall d'Hebron Insitute of Oncology, Barcelona, Spain (AP); Department of Epidemiology, Harvard School of Public Health, Boston, MA (XGA); Institució Catalana de Recerca i Estudis Avançats (ICREA), Barcelona, Spain (RRG)
| | - Joël Jean-Mairet
- Oncology Program (MP, AAE, AB, MT, MG, XGA, MM, JU, SG, RRG) and Biostatistics and Bioinformatics Unit (EP), Institute for Research in Biomedicine (IRB Barcelona), Barcelona, Spain; Cancer Research Program (FR, AR, JA) and Microarray Analysis Service (LN), IMIM (Hospital del Mar Medical Research Institute), Barcelona, Spain; Pathology Department, IIS-Fundación Jimenez Diaz, Madrid, Spain (FR); Medical Oncology Service, Hospital del Mar, Barcelona, Spain (AR, JA); Department of Oncology and Hematology, Hospital Clínico Universitario, Valencia, Spain (AL); Valencia Central University, Spain (AL); Inbiomotion, Barcelona, Spain (JJM); Sheffield Cancer Research Centre, Sheffield, UK (RC); Universitat Pompeu Fabra, Barcelona, Spain (JA); Translational Genomics, Vall d'Hebron Insitute of Oncology, Barcelona, Spain (AP); Department of Epidemiology, Harvard School of Public Health, Boston, MA (XGA); Institució Catalana de Recerca i Estudis Avançats (ICREA), Barcelona, Spain (RRG)
| | - Robert Coleman
- Oncology Program (MP, AAE, AB, MT, MG, XGA, MM, JU, SG, RRG) and Biostatistics and Bioinformatics Unit (EP), Institute for Research in Biomedicine (IRB Barcelona), Barcelona, Spain; Cancer Research Program (FR, AR, JA) and Microarray Analysis Service (LN), IMIM (Hospital del Mar Medical Research Institute), Barcelona, Spain; Pathology Department, IIS-Fundación Jimenez Diaz, Madrid, Spain (FR); Medical Oncology Service, Hospital del Mar, Barcelona, Spain (AR, JA); Department of Oncology and Hematology, Hospital Clínico Universitario, Valencia, Spain (AL); Valencia Central University, Spain (AL); Inbiomotion, Barcelona, Spain (JJM); Sheffield Cancer Research Centre, Sheffield, UK (RC); Universitat Pompeu Fabra, Barcelona, Spain (JA); Translational Genomics, Vall d'Hebron Insitute of Oncology, Barcelona, Spain (AP); Department of Epidemiology, Harvard School of Public Health, Boston, MA (XGA); Institució Catalana de Recerca i Estudis Avançats (ICREA), Barcelona, Spain (RRG)
| | - Joan Albanell
- Oncology Program (MP, AAE, AB, MT, MG, XGA, MM, JU, SG, RRG) and Biostatistics and Bioinformatics Unit (EP), Institute for Research in Biomedicine (IRB Barcelona), Barcelona, Spain; Cancer Research Program (FR, AR, JA) and Microarray Analysis Service (LN), IMIM (Hospital del Mar Medical Research Institute), Barcelona, Spain; Pathology Department, IIS-Fundación Jimenez Diaz, Madrid, Spain (FR); Medical Oncology Service, Hospital del Mar, Barcelona, Spain (AR, JA); Department of Oncology and Hematology, Hospital Clínico Universitario, Valencia, Spain (AL); Valencia Central University, Spain (AL); Inbiomotion, Barcelona, Spain (JJM); Sheffield Cancer Research Centre, Sheffield, UK (RC); Universitat Pompeu Fabra, Barcelona, Spain (JA); Translational Genomics, Vall d'Hebron Insitute of Oncology, Barcelona, Spain (AP); Department of Epidemiology, Harvard School of Public Health, Boston, MA (XGA); Institució Catalana de Recerca i Estudis Avançats (ICREA), Barcelona, Spain (RRG)
| | - Roger R Gomis
- Oncology Program (MP, AAE, AB, MT, MG, XGA, MM, JU, SG, RRG) and Biostatistics and Bioinformatics Unit (EP), Institute for Research in Biomedicine (IRB Barcelona), Barcelona, Spain; Cancer Research Program (FR, AR, JA) and Microarray Analysis Service (LN), IMIM (Hospital del Mar Medical Research Institute), Barcelona, Spain; Pathology Department, IIS-Fundación Jimenez Diaz, Madrid, Spain (FR); Medical Oncology Service, Hospital del Mar, Barcelona, Spain (AR, JA); Department of Oncology and Hematology, Hospital Clínico Universitario, Valencia, Spain (AL); Valencia Central University, Spain (AL); Inbiomotion, Barcelona, Spain (JJM); Sheffield Cancer Research Centre, Sheffield, UK (RC); Universitat Pompeu Fabra, Barcelona, Spain (JA); Translational Genomics, Vall d'Hebron Insitute of Oncology, Barcelona, Spain (AP); Department of Epidemiology, Harvard School of Public Health, Boston, MA (XGA); Institució Catalana de Recerca i Estudis Avançats (ICREA), Barcelona, Spain (RRG).
| |
Collapse
|
26
|
Wang W, Rigueur D, Lyons KM. TGFβ signaling in cartilage development and maintenance. ACTA ACUST UNITED AC 2015; 102:37-51. [PMID: 24677722 DOI: 10.1002/bdrc.21058] [Citation(s) in RCA: 193] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2013] [Accepted: 01/16/2014] [Indexed: 12/18/2022]
Abstract
Members of the transforming growth factor beta (TGFβ) superfamily of secreted factors play essential roles in nearly every aspect of cartilage formation and maintenance. However, the mechanisms by which TGFβs transduce their effects in cartilage in vivo remain poorly understood. Mutations in several TGFβ family members, their receptors, extracellular modulators, and intracellular transducers have been described, and these usually impact the development of the cartilaginous skeleton. Furthermore, genome-wide association studies have linked components of the (TGFβ) superfamily to susceptibility to osteoarthritis. This review focuses on recent discoveries from genetic studies in the mouse regarding the regulation of TGFβ signaling in developing growth plate and articular cartilage, as well as the different modes of crosstalk between canonical and noncanonical TGFβ signaling. These new insights into TGFβ signaling in cartilage may open new prospects for therapies that maintain healthy articular cartilage.
Collapse
Affiliation(s)
- Weiguang Wang
- Department of Orthopaedic Surgery and Orthopaedic Institute for Children, David Geffen School of Medicine, University of California, Los Angeles, California, 90095
| | | | | |
Collapse
|
27
|
Niceta M, Stellacci E, Gripp K, Zampino G, Kousi M, Anselmi M, Traversa A, Ciolfi A, Stabley D, Bruselles A, Caputo V, Cecchetti S, Prudente S, Fiorenza M, Boitani C, Philip N, Niyazov D, Leoni C, Nakane T, Keppler-Noreuil K, Braddock S, Gillessen-Kaesbach G, Palleschi A, Campeau P, Lee B, Pouponnot C, Stella L, Bocchinfuso G, Katsanis N, Sol-Church K, Tartaglia M. Mutations Impairing GSK3-Mediated MAF Phosphorylation Cause Cataract, Deafness, Intellectual Disability, Seizures, and a Down Syndrome-like Facies. Am J Hum Genet 2015; 96:816-25. [PMID: 25865493 PMCID: PMC4570552 DOI: 10.1016/j.ajhg.2015.03.001] [Citation(s) in RCA: 93] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2015] [Accepted: 03/02/2015] [Indexed: 11/26/2022] Open
Abstract
Transcription factors operate in developmental processes to mediate inductive events and cell competence, and perturbation of their function or regulation can dramatically affect morphogenesis, organogenesis, and growth. We report that a narrow spectrum of amino-acid substitutions within the transactivation domain of the v-maf avian musculoaponeurotic fibrosarcoma oncogene homolog (MAF), a leucine zipper-containing transcription factor of the AP1 superfamily, profoundly affect development. Seven different de novo missense mutations involving conserved residues of the four GSK3 phosphorylation motifs were identified in eight unrelated individuals. The distinctive clinical phenotype, for which we propose the eponym Aymé-Gripp syndrome, is not limited to lens and eye defects as previously reported for MAF/Maf loss of function but includes sensorineural deafness, intellectual disability, seizures, brachycephaly, distinctive flat facial appearance, skeletal anomalies, mammary gland hypoplasia, and reduced growth. Disease-causing mutations were demonstrated to impair proper MAF phosphorylation, ubiquitination and proteasomal degradation, perturbed gene expression in primary skin fibroblasts, and induced neurodevelopmental defects in an in vivo model. Our findings nosologically and clinically delineate a previously poorly understood recognizable multisystem disorder, provide evidence for MAF governing a wider range of developmental programs than previously appreciated, and describe a novel instance of protein dosage effect severely perturbing development.
Collapse
|
28
|
Caldwell KL, Wang J. Cell-based articular cartilage repair: the link between development and regeneration. Osteoarthritis Cartilage 2015; 23:351-62. [PMID: 25450846 PMCID: PMC4339504 DOI: 10.1016/j.joca.2014.11.004] [Citation(s) in RCA: 113] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/11/2014] [Revised: 10/02/2014] [Accepted: 11/01/2014] [Indexed: 02/02/2023]
Abstract
Clinical efforts to repair damaged articular cartilage (AC) currently face major obstacles due to limited intrinsic repair capacity of the tissue and unsuccessful biological interventions. This highlights a need for better therapeutic strategies. This review summarizes the recent advances in the field of cell-based AC repair. In both animals and humans, AC defects that penetrate into the subchondral bone marrow are mainly filled with fibrocartilaginous tissue through the differentiation of bone marrow mesenchymal stem cells (MSCs), followed by degeneration of repaired cartilage and osteoarthritis (OA). Cell therapy and tissue engineering techniques using culture-expanded chondrocytes, bone marrow MSCs, or pluripotent stem cells with chondroinductive growth factors may generate cartilaginous tissue in AC defects but do not form hyaline cartilage-based articular surface because repair cells often lose chondrogenic activity or result in chondrocyte hypertrophy. The new evidence that AC and synovium develop from the same pool of precursors with similar gene profiles and that synovium-derived chondrocytes have stable chondrogenic activity has promoted use of synovium as a new cell source for AC repair. The recent finding that NFAT1 and NFAT2 transcription factors (TFs) inhibit chondrocyte hypertrophy and maintain metabolic balance in AC is a significant advance in the field of AC repair. The use of synovial MSCs and discovery of upstream transcriptional regulators that help maintain the AC phenotype have opened new avenues to improve the outcome of AC regeneration.
Collapse
Affiliation(s)
| | - Jinxi Wang
- Corresponding Author: Jinxi Wang, Address: University of Kansas Medical Center, Department of Orthopedic Surgery, 3901 Rainbow Blvd., Mail Stop 3017, Kansas City, KS 66160, USA, Phone: +1 913-588-0870, Fax: +1 913-945-7773,
| |
Collapse
|
29
|
Tsuchiya M, Misaka R, Nitta K, Tsuchiya K. Transcriptional factors, Mafs and their biological roles. World J Diabetes 2015; 6:175-183. [PMID: 25685288 PMCID: PMC4317310 DOI: 10.4239/wjd.v6.i1.175] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/18/2014] [Revised: 09/17/2014] [Accepted: 12/10/2014] [Indexed: 02/05/2023] Open
Abstract
The Maf family of transcription factors is characterized by a typical bZip structure; these transcription factors act as important regulators of the development and differentiation of many organs and tissues, including the kidney. The Maf family consists of two subgroups that are characterized according to their structure: large Maf transcription factors and small Maf transcription factors. The large Maf subgroup consists of four proteins, designated as MAFA, MAFB, c-MAF and neural retina-specific leucine zipper. In particular, MAFA is a distinct molecule that has been attracting the attention of researchers because it acts as a strong transactivator of insulin, suggesting that Maf transcription factors are likely to be involved in systemic energy homeostasis. In this review, we focused on the regulation of glucose/energy balance by Maf transcription factors in various organs.
Collapse
|
30
|
Paiva KBS, Granjeiro JM. Bone tissue remodeling and development: Focus on matrix metalloproteinase functions. Arch Biochem Biophys 2014; 561:74-87. [PMID: 25157440 DOI: 10.1016/j.abb.2014.07.034] [Citation(s) in RCA: 119] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2014] [Revised: 07/17/2014] [Accepted: 07/21/2014] [Indexed: 12/25/2022]
|
31
|
Ubiquitination of the transcription factor c-MAF is mediated by multiple lysine residues. Int J Biochem Cell Biol 2014; 57:157-66. [PMID: 25448412 DOI: 10.1016/j.biocel.2014.10.024] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2014] [Revised: 10/20/2014] [Accepted: 10/22/2014] [Indexed: 11/22/2022]
Abstract
The transcription factor c-MAF could be polyubiquitinated and subsequently degraded in the proteasomes. Theoretically, any lysine residues in c-MAF could be ubiquitinated. In the present study, we tried to find out the specific lysine residue(s) mediating c-MAF ubiquitination. Through a series of mutational screens from lysine (K) to arginine (R), we found that any single lysine mutation (K to R) failed to prevent c-MAF ubiquitination, and any single lysine residue alone could not mediate c-MAF ubiquitination, which indicated that multiple lysine residues were required for c-MAF ubiquitination. Bioinformatics and computing analyses revealed that K85 and K350 could mediate c-MAF ubiquitination, which was confirmed by the cell-based assays. However, this duo was not the only pair because the K85R/K350R mutant could also be ubiquitinated. Functionally, both M12 (K85/K350) and W12 (K85R/K350R) mutants increased cyclin D2 promoter-driven luciferase activity, but they were less potent than the lysine-free counterpart (M14). In addition, M14 induced a higher level of expression of cyclin D2 at both mRNA and protein levels. Therefore, we demonstrated that c-MAF ubiquitination is mediated by multiple lysine residues, of which K85 and K350 were sufficient but not the only residues in mediating c-MAF ubiquitination. Moreover, c-MAF was found to be degraded by lysosomes. This study added a novel insight for c-MAF ubiquitination and degradation, suggesting that c-MAF stability is strictly regulated.
Collapse
|
32
|
Mutations in COL27A1 cause Steel syndrome and suggest a founder mutation effect in the Puerto Rican population. Eur J Hum Genet 2014; 23:342-6. [PMID: 24986830 DOI: 10.1038/ejhg.2014.107] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2014] [Revised: 04/09/2014] [Accepted: 04/24/2014] [Indexed: 11/09/2022] Open
Abstract
Osteochondrodysplasias represent a large group of developmental structural disorders that can be caused by mutations in a variety of genes responsible for chondrocyte development, differentiation, mineralization and early ossification. The application of whole-exome sequencing to disorders apparently segregating as Mendelian traits has proven to be an effective approach to disease gene identification for conditions with unknown molecular etiology. We identified a homozygous missense variant p.(Gly697Arg) in COL27A1, in a family with Steel syndrome and no consanguinity. Interestingly, the identified variant seems to have arisen as a founder mutation in the Puerto Rican population.
Collapse
|
33
|
Lu Y, Qiao L, Lei G, Mira RR, Gu J, Zheng Q. Col10a1 gene expression and chondrocyte hypertrophy during skeletal development and disease. ACTA ACUST UNITED AC 2014. [DOI: 10.1007/s11515-014-1310-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
34
|
ADAMTS-12: a multifaced metalloproteinase in arthritis and inflammation. Mediators Inflamm 2014; 2014:649718. [PMID: 24876675 PMCID: PMC4020202 DOI: 10.1155/2014/649718] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2014] [Revised: 03/28/2014] [Accepted: 04/07/2014] [Indexed: 12/12/2022] Open
Abstract
ADAMTS-12 is a member of a disintegrin and metalloproteinase with thrombospondin motifs (ADAMTS) family of proteases, which were known to play important roles in various biological and pathological processes, such as development, angiogenesis, inflammation, cancer, arthritis, and atherosclerosis. In this review, we briefly summarize the structural organization of ADAMTS-12; concentrate on the emerging role of ADAMTS-12 in several pathophysiological conditions, including intervertebral disc degeneration, tumorigenesis and angioinhibitory effects, pediatric stroke, gonad differentiation, trophoblast invasion, and genetic linkage to schizophrenia and asthma, with special focus on its role in arthritis and inflammation; and end with the perspective research of ADAMTS-12 and its potential as a promising diagnostic and therapeutic target in various kinds of diseases and conditions.
Collapse
|
35
|
Hong E, Yik J, Amanatullah DF, Di Cesare PE, Haudenschild DR. c-Maf Transcription Factor Regulates ADAMTS-12 Expression in Human Chondrogenic Cells. Cartilage 2013; 4:177-86. [PMID: 26069660 PMCID: PMC4297105 DOI: 10.1177/1947603512472697] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
OBJECTIVE ADAMTS (a disintegrin and metalloproteinase with thrombospondin type-1 motif) zinc metalloproteinases are important during the synthesis and breakdown of cartilage extracellular matrix. ADAMTS-12 is up-regulated during in vitro chondrogenesis and embryonic limb development; however, the regulation of ADAMTS-12 expression in cartilage remains unknown. The transcription factor c-Maf is a member of Maf family of basic ZIP (bZIP) transcription factors. Expression of c-Maf is highest in hypertrophic chondrocytes during embryonic development and postnatal growth. We hypothesize that c-Maf and ADAMTS-12 are co-expressed during chondrocyte differentiation and that c-Maf regulates ADAMTS-12 expression during chondrogenesis. DESIGN Promoter analysis and species alignments identified potential c-Maf binding sites in the ADAMTS-12 promoter. c-Maf and ADAMTS-12 co-expression was monitored during chondrogenesis of stem cell pellet cultures. Luciferase expression driven by ADAMTS-12 promoter segments was measured in the presence and absence of c-Maf, and synthetic oligonucleotides were used to confirm specific binding of c-Maf to ADAMTS-12 promoter sequences. RESULTS In vitro chondrogenesis from human mesenchymal stem cells revealed co-expression of ADAMTS-12 and c-Maf during differentiation. Truncation and point mutations of the ADAMTS-12 promoter evaluated in reporter assays localized the response to the proximal 315 bp of the ADAMTS-12 promoter, which contained a predicted c-Maf recognition element (MARE) at position -61. Electorphoretic mobility shift assay confirmed that c-Maf directly interacted with the MARE at position -61. CONCLUSIONS These data suggest that c-Maf is involved in chondrocyte differentiation and hypertrophy, at least in part, through the regulation of ADAMTS-12 expression at a newly identified MARE in its proximal promoter.
Collapse
Affiliation(s)
- Eunmee Hong
- Lawrence J. Ellison Musculoskeletal Research Center, Department of Orthopaedic Surgery, University of California Davis Medical Center, Sacramento, CA, USA
| | - Jasper Yik
- Lawrence J. Ellison Musculoskeletal Research Center, Department of Orthopaedic Surgery, University of California Davis Medical Center, Sacramento, CA, USA
| | - Derek F. Amanatullah
- Lawrence J. Ellison Musculoskeletal Research Center, Department of Orthopaedic Surgery, University of California Davis Medical Center, Sacramento, CA, USA
| | - Paul E. Di Cesare
- Lawrence J. Ellison Musculoskeletal Research Center, Department of Orthopaedic Surgery, University of California Davis Medical Center, Sacramento, CA, USA
| | - Dominik R. Haudenschild
- Lawrence J. Ellison Musculoskeletal Research Center, Department of Orthopaedic Surgery, University of California Davis Medical Center, Sacramento, CA, USA
| |
Collapse
|
36
|
Nishikawa K. [Understanding the mechanism of senile osteoporosis]. Nihon Ronen Igakkai Zasshi 2012; 49:314-7. [PMID: 23268972 DOI: 10.3143/geriatrics.49.314] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
|
37
|
Xie Q, Cvekl A. The orchestration of mammalian tissue morphogenesis through a series of coherent feed-forward loops. J Biol Chem 2011; 286:43259-71. [PMID: 21998302 DOI: 10.1074/jbc.m111.264580] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Tissue morphogenesis requires intricate temporal and spatial control of gene expression that is executed through specific gene regulatory networks (GRNs). GRNs are comprised from individual subcircuits of different levels of complexity. An important question is to elucidate the mutual relationship between those genes encoding DNA-binding factors that trigger the subcircuit with those that play major "later" roles during terminal differentiation via expression of specific genes that constitute the phenotype of individual tissues. The ocular lens is a classical model system to study tissue morphogenesis. Pax6 is essential for both lens placode formation and subsequent stages of lens morphogenesis, whereas c-Maf controls terminal differentiation of lens fibers, including regulation of crystallins, key lens structural proteins required for its transparency and refraction. Here, we show that Pax6 directly regulates c-Maf expression during lens development. A 1.3-kb c-Maf promoter with a 1.6-kb upstream enhancer (CR1) recapitulated the endogenous c-Maf expression pattern in lens and retinal pigmented epithelium. ChIP assays revealed binding of Pax6 and c-Maf to multiple regions of the c-Maf locus in lens chromatin. To predict functional Pax6-binding sites, nine novel variants of Pax6 DNA-binding motifs were identified and characterized. Two of these motifs predicted a pair of Pax6-binding sites in the CR1. Mutagenesis of these Pax6-binding sites inactivated transgenic expression in the lens but not in retinal pigmented epithelium. These data establish a novel regulatory role for Pax6 during lens development, link together the Pax6/c-Maf/crystallin regulatory network, and suggest a novel type of GRN subcircuit that controls a major part of embryonic lens development.
Collapse
Affiliation(s)
- Qing Xie
- Department of Genetics, Albert Einstein College of Medicine, Bronx, New York 10461, USA
| | | |
Collapse
|
38
|
Goldring MB, Otero M, Plumb DA, Dragomir C, Favero M, El Hachem K, Hashimoto K, Roach HI, Olivotto E, Borzì RM, Marcu KB, Marcu KB. Roles of inflammatory and anabolic cytokines in cartilage metabolism: signals and multiple effectors converge upon MMP-13 regulation in osteoarthritis. Eur Cell Mater 2011; 21:202-20. [PMID: 21351054 PMCID: PMC3937960 DOI: 10.22203/ecm.v021a16] [Citation(s) in RCA: 352] [Impact Index Per Article: 25.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023] Open
Abstract
Human cartilage is a complex tissue of matrix proteins that vary in amount and orientation from superficial to deep layers and from loaded to unloaded zones. A major challenge to efforts to repair cartilage by stem cell-based and other tissue engineering strategies is the inability of the resident chondrocytes to lay down new matrix with the same structural and resilient properties that it had upon its original formation. This is particularly true of the collagen network, which is susceptible to cleavage once proteoglycans are depleted. Thus, a thorough understanding of the similarities and particularly the marked differences in mechanisms of cartilage remodeling during development, osteoarthritis, and aging may lead to more effective strategies for preventing cartilage damage and promoting repair. To identify and characterize effectors or regulators of cartilage remodeling in these processes, we are using culture models of primary human and mouse chondrocytes and cell lines and mouse genetic models to manipulate gene expression programs leading to matrix remodeling and subsequent chondrocyte hypertrophic differentiation, pivotal processes which both go astray in OA disease. Matrix metalloproteinases (MMP)-13, the major type II collagen-degrading collagenase, is regulated by stress-, inflammation-, and differentiation-induced signals that not only contribute to irreversible joint damage (progression) in OA, but importantly, also to the initiation/onset phase, wherein chondrocytes in articular cartilage leave their natural growth- and differentiation-arrested state. Our work points to common mediators of these processes in human OA cartilage and in early through late stages of OA in surgical and genetic mouse models.
Collapse
Affiliation(s)
- Mary B. Goldring
- Tissue Engineering, Regeneration, and Repair Program, Research Division, The Hospital for Special Surgery, Weill Cornell Medical College, New York, NY 10021, USA,Address for correspondence: Mary B. Goldring, 535 East 70th Street, Caspary Research Building, 5th Floor, New York, NY 10021. USA,
| | - Miguel Otero
- Tissue Engineering, Regeneration, and Repair Program, Research Division, The Hospital for Special Surgery, Weill Cornell Medical College, New York, NY 10021, USA
| | - Darren A. Plumb
- Tissue Engineering, Regeneration, and Repair Program, Research Division, The Hospital for Special Surgery, Weill Cornell Medical College, New York, NY 10021, USA
| | - Cecilia Dragomir
- Tissue Engineering, Regeneration, and Repair Program, Research Division, The Hospital for Special Surgery, Weill Cornell Medical College, New York, NY 10021, USA
| | - Marta Favero
- Tissue Engineering, Regeneration, and Repair Program, Research Division, The Hospital for Special Surgery, Weill Cornell Medical College, New York, NY 10021, USA
| | - Karim El Hachem
- Tissue Engineering, Regeneration, and Repair Program, Research Division, The Hospital for Special Surgery, Weill Cornell Medical College, New York, NY 10021, USA
| | - Ko Hashimoto
- Tissue Engineering, Regeneration, and Repair Program, Research Division, The Hospital for Special Surgery, Weill Cornell Medical College, New York, NY 10021, USA
| | | | - Eleonora Olivotto
- Laboratorio di Immunoreumatologia e Rigenerazione Tissutale, Istituti Ortopedia Rizzoli, 40136 Bologna, Italy
| | - Rosa Maria Borzì
- Laboratorio di Immunoreumatologia e Rigenerazione Tissutale, Istituti Ortopedia Rizzoli, 40136 Bologna, Italy
| | - Kenneth B. Marcu
- Laboratorio di Immunoreumatologia e Rigenerazione Tissutale, Istituti Ortopedia Rizzoli, 40136 Bologna, Italy,Biochemistry and Cell Biology Dept., Stony Brook University, Stony Brook, NY, 11794-5215, USA
| | | |
Collapse
|
39
|
Abstract
Chondrocyte differentiation in the growth plate is an important process for the longitudinal growth of endochondral bones. Sox9 and Runx2 are the most often-studied transcriptional regulators of the chondrocyte differentiation process, but the importance of additional factors is also becoming apparent. Mafs are a subfamily of the basic ZIP (bZIP) transcription factor superfamily, which act as key regulators of tissue-specific gene expression and terminal differentiation in many tissues. There is increasing evidence that c-Maf and its splicing variant Lc-Maf play a role in chondrocyte differentiation in a temporal-spatial manner. This review summarizes the functions of c-Maf in chondrocyte differentiation and discusses the possible role of c-Maf in osteoarthritis progression.
Collapse
Affiliation(s)
| | | | - Dominik R. Haudenschild
- Dominik R. Haudenschild, Department of Orthopaedic Surgery, Division of Orthopaedic Research, University of California Davis Medical Center, 4635 Second Street, Sacramento, CA 95817, USA
| |
Collapse
|
40
|
Nishikawa K, Nakashima T, Takeda S, Isogai M, Hamada M, Kimura A, Kodama T, Yamaguchi A, Owen MJ, Takahashi S, Takayanagi H. Maf promotes osteoblast differentiation in mice by mediating the age-related switch in mesenchymal cell differentiation. J Clin Invest 2010; 120:3455-65. [PMID: 20877012 DOI: 10.1172/jci42528] [Citation(s) in RCA: 140] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2010] [Accepted: 07/14/2010] [Indexed: 12/13/2022] Open
Abstract
Aging leads to the disruption of the homeostatic balance of multiple biological systems. In bone marrow multipotent mesenchymal cells undergo differentiation into various anchorage-dependent cell types, including osteoblasts and adipocytes. With age as well as with treatment of antidiabetic drugs such as thiazolidinediones, mesenchymal cells favor differentiation into adipocytes, resulting in an increased number of adipocytes and a decreased number of osteoblasts, causing osteoporosis. The mechanism behind this differentiation switch is unknown. Here we show an age-related decrease in the expression of Maf in mouse mesenchymal cells, which regulated mesenchymal cell bifurcation into osteoblasts and adipocytes by cooperating with the osteogenic transcription factor Runx2 and inhibiting the expression of the adipogenic transcription factor Pparg. The crucial role of Maf in both osteogenesis and adipogenesis was underscored by in vivo observations of delayed bone formation in perinatal Maf(-/-) mice and an accelerated formation of fatty marrow associated with bone loss in aged Maf(+/-) mice. This study identifies a transcriptional mechanism for an age-related switch in cell fate determination and may provide a molecular basis for novel therapeutic strategies against age-related bone diseases.
Collapse
Affiliation(s)
- Keizo Nishikawa
- Department of Cell Signaling, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Bunkyo-ku, Tokyo, Japan
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Kim HN, Lee EJ, Jung SC, Lee JY, Chung HW, Kim HL. Genetic variants that affect length/height in infancy/early childhood in Vietnamese-Korean families. J Hum Genet 2010; 55:681-90. [DOI: 10.1038/jhg.2010.88] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
|
42
|
Huang BL, Brugger SM, Lyons KM. Stage-specific control of connective tissue growth factor (CTGF/CCN2) expression in chondrocytes by Sox9 and beta-catenin. J Biol Chem 2010; 285:27702-12. [PMID: 20571031 DOI: 10.1074/jbc.m110.108498] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
CCN2/connective tissue growth factor is highly expressed in hypertrophic chondrocytes and is required for chondrogenesis. However, the transcriptional mechanisms controlling its expression in cartilage are largely unknown. The activity of the Ccn2 promoter was, therefore, investigated in osteochondro-progenitor cells and hypertrophic chondrocytes to ascertain these mechanisms. Sox9 and T-cell factor (TCF) x lymphoid enhancer factor (LEF) factors contain HMG domains and bind to related consensus sites. TCF x LEF factors are normally repressive but when bound to DNA in a complex with beta-catenin become activators of gene expression. In silico analysis of the Ccn2 proximal promoter identified multiple consensus TCF x LEF elements, one of which was also a consensus binding site for Sox9. Using luciferase reporter constructs, the TCF x LEF x Sox9 site was found to be involved in stage-specific expression of Ccn2. Luciferase, electrophoretic mobility shift assay (EMSA), and ChIP analysis revealed that Sox9 represses Ccn2 expression by binding to the consensus TCF x LEF x Sox9 site. On the other hand, the same assays showed that in hypertrophic chondrocytes, TCF x LEF x beta-catenin complexes occupy the consensus TCF x LEF x Sox9 site and activate Ccn2 expression. Furthermore, transgenic mice in which lacZ expression is driven under the control of the proximal Ccn2 promoter revealed that the proximal Ccn2 promoter responded to Wnt signaling in cartilage. Hence, we propose that differential occupancy of the TCF x LEF x Sox9 site by Sox9 versus beta-catenin restricts high levels of Ccn2 expression to hypertrophic chondrocytes.
Collapse
Affiliation(s)
- Bau-Lin Huang
- Department of Oral Biology, UCLA School of Dentistry, Los Angeles, California 90095, USA
| | | | | |
Collapse
|
43
|
Guo S, Vanderford NL, Stein R. Phosphorylation within the MafA N terminus regulates C-terminal dimerization and DNA binding. J Biol Chem 2010; 285:12655-61. [PMID: 20208071 DOI: 10.1074/jbc.m110.105759] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Phosphorylation regulates transcription factor activity by influencing dimerization, cellular localization, activation potential, and/or DNA binding. Nevertheless, precisely how this post-translation modification mediates these processes is poorly understood. Here, we examined the role of phosphorylation on the DNA-binding properties of MafA and MafB, closely related transcriptional activators of the basic-leucine zipper (b-Zip) family associated with cell differentiation and oncogenesis. Many common phosphorylation sites were identified by mass spectrometry. However, dephosphorylation only precluded the detection of MafA dimers and consequently dramatically reduced DNA-binding ability. Analysis of MafA/B chimeras revealed that sensitivity to the phosphorylation status of MafA was imparted by sequences spanning the C-terminal dimerization region (amino acids (aa) 279-359), whereas the homologous MafB region (aa 257-323) conveyed phosphorylation-independent DNA binding. Mutational analysis showed that formation of MafA dimers capable of DNA binding required phosphorylation within the distinct N-terminal transactivation domain (aa 1-72) and not the C-terminal b-Zip region. These results demonstrate a novel relationship between the phosphoamino acid-rich transactivation and b-Zip domains in controlling MafA DNA-binding activity.
Collapse
Affiliation(s)
- Shuangli Guo
- From the Department of Molecular Physiology and Biophysics, Vanderbilt University Medical Center, Nashville, Tennessee 37232
| | | | | |
Collapse
|
44
|
Hansen A, Henderson S, Lagos D, Nikitenko L, Coulter E, Roberts S, Gratrix F, Plaisance K, Renne R, Bower M, Kellam P, Boshoff C. KSHV-encoded miRNAs target MAF to induce endothelial cell reprogramming. Genes Dev 2010; 24:195-205. [PMID: 20080955 DOI: 10.1101/gad.553410] [Citation(s) in RCA: 124] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Kaposi sarcoma herpesvirus (KSHV) induces transcriptional reprogramming of endothelial cells. In particular, KSHV-infected lymphatic endothelial cells (LECs) show an up-regulation of genes associated with blood vessel endothelial cells (BECs). Consequently, KSHV-infected tumor cells in Kaposi sarcoma are poorly differentiated endothelial cells, expressing markers of both LECs and BECs. MicroRNAs (miRNAs) are short noncoding RNA molecules that act post-transcriptionally to negatively regulate gene expression. Here we validate expression of the KSHV-encoded miRNAs in Kaposi sarcoma lesions and demonstrate that these miRNAs contribute to viral-induced reprogramming by silencing the cellular transcription factor MAF (musculoaponeurotic fibrosarcoma oncogene homolog). MAF is expressed in LECs but not in BECs. We identify a novel role for MAF as a transcriptional repressor, preventing expression of BEC-specific genes, thereby maintaining the differentiation status of LECs. These findings demonstrate that viral miRNAs could influence the differentiation status of infected cells, and thereby contribute to KSHV-induced oncogenesis.
Collapse
Affiliation(s)
- Amy Hansen
- Cancer Research UK Viral Oncology Group, University College London Cancer Institute, University College London, London WC1E 6BT, United Kingdom
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Li T, Xiao J, Wu Z, Qiu G, Ding Y. Transcriptional activation of human MMP-13 gene expression by c-Maf in osteoarthritic chondrocyte. Connect Tissue Res 2010; 51:48-54. [PMID: 20067416 DOI: 10.3109/03008200902989104] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Matrix metalloproteinase (MMP)-13 has pivotal roles in the pathogenesis of Osteoarthritis (OA) and it is necessary to understand the regulatory mechanisms of MMP-13 expression. MMP-13 gene expression is regulated primarily at the transcriptional level. In this study, we investigated the role of c-maf in regulating MMP-13 transcription. Using transient transfection system with an c-maf construct, and MMP-13 promoter-luciferase constructs with specific mutations in transcription factor binding sites, we found that c-maf can significantly enhance MMP-13 promoter activity via the AP-1 site, By gene suppression with RNAi technology, we could show that c-maf downregulation leads to a reduced expression of MMP13. Chromatin immunoprecipitation assays reveal that c-maf binds to the MMP-13 gene promoter to a region of the MMP-13 promoter containing the AP-1 site. Taken together, these studies demonstrate a new level of transcriptional regulation of MMP-13 expression by the c-maf.
Collapse
Affiliation(s)
- Tao Li
- Department of Orthopaedics, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, & Peking Union Medical College, Beijing, China
| | | | | | | | | |
Collapse
|
46
|
Todoerti K, Lisignoli G, Storti P, Agnelli L, Novara F, Manferdini C, Codeluppi K, Colla S, Crugnola M, Abeltino M, Bolzoni M, Sgobba V, Facchini A, Lambertenghi-Deliliers G, Zuffardi O, Rizzoli V, Neri A, Giuliani N. Distinct transcriptional profiles characterize bone microenvironment mesenchymal cells rather than osteoblasts in relationship with multiple myeloma bone disease. Exp Hematol 2009; 38:141-53. [PMID: 19963035 DOI: 10.1016/j.exphem.2009.11.009] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2009] [Revised: 11/05/2009] [Accepted: 11/24/2009] [Indexed: 01/10/2023]
Abstract
OBJECTIVE Multiple myeloma (MM) is characterized by a high incidence of osteolytic bone lesions, which have been previously correlated with the gene expression profiles of MM cells. The aim of this study was to investigate the transcriptional patterns of cells in the bone microenvironment and their relationships with the presence of osteolysis in MM patients. MATERIALS AND METHODS Both mesenchymal (MSC) and osteoblastic (OB) cells were isolated directly from bone biopsies of MM patients and controls to perform gene expression profiling by microarrays and real-time polymerase chain reaction on selected bone-related genes. RESULTS We identified a series of upregulated and downregulated genes that were differentially expressed in the MSC cells of osteolytic and nonosteolytic patients. Comparison of the osteolytic and nonosteolytic samples also showed that the MSC cells and OB had distinct transcriptional patterns. No significantly modulated genes were found in the OBs of the osteolytic and nonosteolytic patients. CONCLUSIONS Our data suggest that the gene expression profiles of cells of the bone microenvironment are different in MM patients and controls, and that MSC cells, but not OBs, have a distinct transcriptional pattern associated with the occurrence of bone lesions in MM patients. These data support the idea that alterations in MSC cells may be involved in MM bone disease.
Collapse
Affiliation(s)
- Katia Todoerti
- Dipartimento di Scienze Mediche, Università di Milano e U.O. Ematologia 1, Fondazione IRCCS Policlinico, Milan, Italy
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Hartmann C. Transcriptional networks controlling skeletal development. Curr Opin Genet Dev 2009; 19:437-43. [DOI: 10.1016/j.gde.2009.09.001] [Citation(s) in RCA: 56] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2009] [Revised: 09/04/2009] [Accepted: 09/10/2009] [Indexed: 12/18/2022]
|
48
|
Ho L, Stojanovski A, Whetstone H, Wei QX, Mau E, Wunder JS, Alman B. Gli2 and p53 cooperate to regulate IGFBP-3- mediated chondrocyte apoptosis in the progression from benign to malignant cartilage tumors. Cancer Cell 2009; 16:126-36. [PMID: 19647223 DOI: 10.1016/j.ccr.2009.05.013] [Citation(s) in RCA: 66] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/17/2008] [Revised: 03/07/2009] [Accepted: 05/21/2009] [Indexed: 11/17/2022]
Abstract
Clinical evidence suggests that benign cartilage lesions can progress to malignant chondrosarcoma, but the molecular events in this progression are unknown. Mice that develop benign cartilage lesions due to overexpression of Gli2 in chondrocytes developed lesions similar to chondrosarcomas when they were also deficient in p53. Gli2 overexpression and p53 deficiency had opposing effects on chondrocyte differentiation, but had additive effects negatively regulating apoptosis. Regulation of Igfbp3 expression and insulin-like growth factor (IGF) signaling by Gli and p53 integrated their effect on apoptosis. Treatment of human chondrosarcomas or fetal mouse limb explants with IGFBP3 or by blocking IGF increased the apoptosis rate, and mice expressing Gli2 developed substantially fewer tumors when they were also deficient for Igf2. IGF signaling-meditated apoptosis regulates the progression to malignant chondrosarcoma.
Collapse
Affiliation(s)
- Louisa Ho
- Program in Developmental and Stem Cell Biology, Hospital for Sick Children, University of Toronto, ON, Canada
| | | | | | | | | | | | | |
Collapse
|
49
|
Localization of the cis-enhancer element for mouse type X collagen expression in hypertrophic chondrocytes in vivo. J Bone Miner Res 2009; 24:1022-32. [PMID: 19113928 PMCID: PMC2683646 DOI: 10.1359/jbmr.081249] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
The type X collagen gene (Col10a1) is a specific molecular marker of hypertrophic chondrocytes during endochondral bone formation. Mutations in human COL10A1 and altered chondrocyte hypertrophy have been associated with multiple skeletal disorders. However, until recently, the cis-enhancer element that specifies Col10a1 expression in hypertrophic chondrocytes in vivo has remained unidentified. Previously, we and others have shown that the Col10a1 distal promoter (-4.4 to -3.8 kb) may harbor a critical enhancer that mediates its tissue specificity in transgenic mice studies. Here, we report further localization of the cis-enhancer element within this Col10a1 distal promoter by using a similar transgenic mouse approach. We identify a 150-bp Col10a1 promoter element (-4296 to -4147 bp) that is sufficient to direct its tissue-specific expression in vivo. In silico analysis identified several putative transcription factor binding sites including two potential activator protein-1 (AP-1) sites within its 5'- and 3'-ends (-4276 to -4243 and -4166 to -4152 bp), respectively. Interestingly, transgenic mice using a reporter construct deleted for these two AP-1 elements still showed tissue-specific reporter activity. EMSAs using oligonucleotide probes derived from this region and MCT cell nuclear extracts identified DNA/protein complexes that were enriched from cells stimulated to hypertrophy. Moreover, these elements mediated increased reporter activity on transfection into MCT cells. These data define a 90-bp cis-enhancer required for tissue-specific Col10a1 expression in vivo and putative DNA/protein complexes that contribute to the regulation of chondrocyte hypertrophy. This work will enable us to identify candidate transcription factors essential both for skeletal development and for the pathogenesis of skeletal disorders.
Collapse
|
50
|
The transcription factor Lc-Maf participates in Col27a1 regulation during chondrocyte maturation. Exp Cell Res 2009; 315:2293-300. [PMID: 19414009 DOI: 10.1016/j.yexcr.2009.04.020] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2009] [Revised: 04/16/2009] [Accepted: 04/19/2009] [Indexed: 11/20/2022]
Abstract
The transcription factor Lc-Maf, which is a splice variant of c-Maf, is expressed in cartilage undergoing endochondral ossification and participates in the regulation of type II collagen through a cartilage-specific Col2a1 enhancer element. Type XXVII and type XI collagens are also expressed in cartilage during endochondral ossification, and so enhancer/reporter assays were used to determine whether Lc-Maf could regulate cartilage-specific enhancers from the Col27a1 and Col11a2 genes. The Col27a1 enhancer was upregulated over 4-fold by Lc-Maf, while the Col11a2 enhancer was downregulated slightly. To confirm the results of these reporter assays, rat chondrosarcoma (RCS) cells were transiently transfected with an Lc-Maf expression plasmid, and quantitative RT-PCR was performed to measure the expression of endogenous Col27a1 and Col11a2 genes. Endogenous Col27a1 was upregulated 6-fold by Lc-Maf overexpression, while endogenous Col11a2 was unchanged. Finally, in situ hybridization and immunohistochemistry were performed in the radius and ulna of embryonic day 17 mouse forelimbs undergoing endochondral ossification. Results demonstrated that Lc-Maf and Col27a1 mRNAs are coexpressed in proliferating and prehypertrophic regions, as would be predicted if Lc-Maf regulates Col27a1 expression. Type XXVII collagen protein was also most abundant in prehypertrophic and proliferating chondrocytes. Others have shown that mice that are null for Lc-Maf and c-Maf have expanded hypertrophic regions with reduced ossification and delayed vascularization. Separate studies have indicated that Col27a1 may serve as a scaffold for ossification and vascularization. The work presented here suggests that Lc-Maf may affect the process of endochondral ossification by participating in the regulation of Col27a1 expression.
Collapse
|