1
|
Nagel S, Meyer C, Pommerenke C. IRX-related homeobox gene MKX is a novel oncogene in acute myeloid leukemia. PLoS One 2024; 19:e0315196. [PMID: 39689089 DOI: 10.1371/journal.pone.0315196] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2024] [Accepted: 11/21/2024] [Indexed: 12/19/2024] Open
Abstract
Homeobox genes encode transcription factors which organize differentiation processes in all tissue types including the hematopoietic compartment. Recently, we have reported physiological expression of TALE-class homeobox gene IRX1 in early myelopoiesis restricted to the megakaryocyte-erythroid-progenitor stage and in early B-cell development to the pro-B-cell stage. In contrast, sister homeobox genes IRX2, IRX3 and IRX5 are aberrantly activated in the corresponding malignancies acute myeloid leukemia (AML) and B-cell progenitor acute lymphoid leukemia. Here, we examined the role of IRX-related homeobox gene MKX (also termed IRXL1 or mohawk) in normal and malignant hematopoiesis. Screening of public datasets revealed silent MKX in normal myelopoiesis and B-cell differentiation, and aberrant expression in subsets of AML and multiple myeloma (MM) cell lines and patients. To investigate its dysregulation and oncogenic function we used AML cell line OCI-AML3 as model which strongly expressed MKX at both RNA and protein levels. We found that IRX5, JUNB and NFkB activated MKX in this cell line, while downregulated GATA2 and STAT5 inhibited its expression. MKX downstream analysis was conducted by siRNA-mediated knockdown and RNA-sequencing in OCI-AML3, and by comparative expression profiling analysis of a public dataset from MM patients. Analysis of these data revealed activation of CCL2 which in turn promoted proliferation. Furthermore, MKX upregulated SESN3 and downregulated BCL2L11, which may together underlie decreased etoposide-induced apoptosis. Finally, myeloid differentiation genes CEBPD and GATA2 were respectively up- and downregulated by MKX. Taken together, our study identified MKX as novel aberrantly expressed homeobox gene in AML and MM, highlighting the function of IRX1 in normal myelopoiesis and B-cell development, and of IRX-related genes in corresponding malignancies. Our data merit further investigation of MKX and its deregulated target genes to serve as novel markers and/or potential therapeutic targets in AML patient subsets.
Collapse
Affiliation(s)
- Stefan Nagel
- Dept. of Human and Animal Cell Lines, Leibniz-Institute DSMZ, Braunschweig, Germany
| | - Corinna Meyer
- Dept. of Human and Animal Cell Lines, Leibniz-Institute DSMZ, Braunschweig, Germany
| | - Claudia Pommerenke
- Dept. of Human and Animal Cell Lines, Leibniz-Institute DSMZ, Braunschweig, Germany
| |
Collapse
|
2
|
Marr N, Zamboulis DE, Werling D, Felder AA, Dudhia J, Pitsillides AA, Thorpe CT. The tendon interfascicular basement membrane provides a vascular niche for CD146+ cell subpopulations. Front Cell Dev Biol 2023; 10:1094124. [PMID: 36699014 PMCID: PMC9869387 DOI: 10.3389/fcell.2022.1094124] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2022] [Accepted: 12/23/2022] [Indexed: 01/11/2023] Open
Abstract
Introduction: The interfascicular matrix (IFM; also known as the endotenon) is critical to the mechanical adaptations and response to load in energy-storing tendons, such as the human Achilles and equine superficial digital flexor tendon (SDFT). We hypothesized that the IFM is a tendon progenitor cell niche housing an exclusive cell subpopulation. Methods: Immunolabelling of equine superficial digital flexor tendon was used to identify the interfascicular matrix niche, localising expression patterns of CD31 (endothelial cells), Desmin (smooth muscle cells and pericytes), CD146 (interfascicular matrix cells) and LAMA4 (interfascicular matrix basement membrane marker). Magnetic-activated cell sorting was employed to isolate and compare in vitro properties of CD146+ and CD146- subpopulations. Results: Labelling for CD146 using standard histological and 3D imaging of large intact 3D segments revealed an exclusive interfascicular cell subpopulation that resides in proximity to a basal lamina which forms extensive, interconnected vascular networks. Isolated CD146+ cells exhibited limited mineralisation (osteogenesis) and lipid production (adipogenesis). Discussion: This study demonstrates that the interfascicular matrix is a unique tendon cell niche, containing a vascular-rich network of basement membrane, CD31+ endothelial cells, Desmin+ mural cells, and CD146+ cell populations that are likely essential to tendon structure and/or function. Contrary to our hypothesis, interfascicular CD146+ subpopulations did not exhibit stem cell-like phenotypes. Instead, our results indicate CD146 as a pan-vascular marker within the tendon interfascicular matrix. Together with previous work demonstrating that endogenous tendon CD146+ cells migrate to sites of injury, our data suggest that their mobilisation to promote intrinsic repair involves changes in their relationships with local interfascicular matrix vascular and basement membrane constituents.
Collapse
Affiliation(s)
- Neil Marr
- Comparative Biomedical Sciences, Royal Veterinary College, London, United Kingdom
| | - Danae E. Zamboulis
- Comparative Biomedical Sciences, Royal Veterinary College, London, United Kingdom
| | - Dirk Werling
- Pathobiology and Population Sciences, Centre for Vaccinology and Regenerative Medicine, Royal Veterinary College, Hatfield, United Kingdom
| | - Alessandro A. Felder
- Research Software Development Group, Advanced Research Computing, University College London, London, United Kingdom
| | - Jayesh Dudhia
- Clinical Sciences and Services, Royal Veterinary College, Hatfield, United Kingdom
| | | | - Chavaunne T. Thorpe
- Comparative Biomedical Sciences, Royal Veterinary College, London, United Kingdom
| |
Collapse
|
3
|
Peserico A, Barboni B, Russo V, Bernabò N, El Khatib M, Prencipe G, Cerveró-Varona A, Haidar-Montes AA, Faydaver M, Citeroni MR, Berardinelli P, Mauro A. Mammal comparative tendon biology: advances in regulatory mechanisms through a computational modeling. Front Vet Sci 2023; 10:1175346. [PMID: 37180059 PMCID: PMC10174257 DOI: 10.3389/fvets.2023.1175346] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Accepted: 04/03/2023] [Indexed: 05/15/2023] Open
Abstract
There is high clinical demand for the resolution of tendinopathies, which affect mainly adult individuals and animals. Tendon damage resolution during the adult lifetime is not as effective as in earlier stages where complete restoration of tendon structure and property occurs. However, the molecular mechanisms underlying tendon regeneration remain unknown, limiting the development of targeted therapies. The research aim was to draw a comparative map of molecules that control tenogenesis and to exploit systems biology to model their signaling cascades and physiological paths. Using current literature data on molecular interactions in early tendon development, species-specific data collections were created. Then, computational analysis was used to construct Tendon NETworks in which information flow and molecular links were traced, prioritized, and enriched. Species-specific Tendon NETworks generated a data-driven computational framework based on three operative levels and a stage-dependent set of molecules and interactions (embryo-fetal or prepubertal) responsible, respectively, for signaling differentiation and morphogenesis, shaping tendon transcriptional program and downstream modeling of its fibrillogenesis toward a mature tissue. The computational network enrichment unveiled a more complex hierarchical organization of molecule interactions assigning a central role to neuro and endocrine axes which are novel and only partially explored systems for tenogenesis. Overall, this study emphasizes the value of system biology in linking the currently available disjointed molecular data, by establishing the direction and priority of signaling flows. Simultaneously, computational enrichment was critical in revealing new nodes and pathways to watch out for in promoting biomedical advances in tendon healing and developing targeted therapeutic strategies to improve current clinical interventions.
Collapse
|
4
|
Ding L, Zhou B, Hou Y, Xu L. Stem cells in tendon regeneration and factors governing tenogenesis. Curr Stem Cell Res Ther 2022; 17:503-512. [PMID: 35086458 DOI: 10.2174/1574888x17666220127111135] [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: 08/23/2021] [Revised: 11/16/2021] [Accepted: 12/08/2021] [Indexed: 11/22/2022]
Abstract
Tendons are connective tissue structures of paramount importance to the human ability of locomotion. Tendinopathy and tendon rupture can be resistant to treatment and often recurs, thus resulting in a significant health problem with a relevant social impact worldwide. Unfortunately, existing treatment approaches are suboptimal. A better understanding of the basic biology of tendons may provide a better way to solve these problems and promote tendon regeneration. Stem cells, either obtained from tendons or non-tendon sources, such as bone marrow (BMSCs), adipose tissue (AMSCs), as well as embryonic stem cells (ESCs) and induced pluripotent stem cells (iPSCs), have received increasing attention toward enhancing tendon healing. There are many studies showing that stem cells can contribute to improving tendon healing. Hence, in this review, the current knowledge of BMSCs, AMSCs, TSPCs, ESCs and iPSCs for tendon regeneration, as well as the advantages and limitations among them, has been highlighted. Moreover, the transcriptional and bioactive factors governing tendon healing processes have been discussed.
Collapse
Affiliation(s)
- Lingli Ding
- Lingnan Medical Research Center, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - BingYu Zhou
- Lingnan Medical Research Center, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yonghui Hou
- Key Laboratory of Orthopaedics & Traumatology, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, PR China
| | - Liangliang Xu
- Lingnan Medical Research Center, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, China
| |
Collapse
|
5
|
He P, Ruan D, Huang Z, Wang C, Xu Y, Cai H, Liu H, Fei Y, Heng BC, Chen W, Shen W. Comparison of Tendon Development Versus Tendon Healing and Regeneration. Front Cell Dev Biol 2022; 10:821667. [PMID: 35141224 PMCID: PMC8819183 DOI: 10.3389/fcell.2022.821667] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Accepted: 01/07/2022] [Indexed: 12/27/2022] Open
Abstract
Tendon is a vital connective tissue in human skeletal muscle system, and tendon injury is very common and intractable in clinic. Tendon development and repair are two closely related but still not fully understood processes. Tendon development involves multiple germ layer, as well as the regulation of diversity transcription factors (Scx et al.), proteins (Tnmd et al.) and signaling pathways (TGFβ et al.). The nature process of tendon repair is roughly divided in three stages, which are dominated by various cells and cell factors. This review will describe the whole process of tendon development and compare it with the process of tendon repair, focusing on the understanding and recent advances in the regulation of tendon development and repair. The study and comparison of tendon development and repair process can thus provide references and guidelines for treatment of tendon injuries.
Collapse
Affiliation(s)
- Peiwen He
- Department of Orthopedic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Orthopedics Research Institute of Zhejiang University, Hangzhou, China
- Institute of Sports Medicine, Zhejiang University, Hangzhou, China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou, China
| | - Dengfeng Ruan
- Department of Orthopedic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Orthopedics Research Institute of Zhejiang University, Hangzhou, China
- Institute of Sports Medicine, Zhejiang University, Hangzhou, China
| | - Zizhan Huang
- Department of Orthopedic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Orthopedics Research Institute of Zhejiang University, Hangzhou, China
- Institute of Sports Medicine, Zhejiang University, Hangzhou, China
| | - Canlong Wang
- Department of Orthopedic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Orthopedics Research Institute of Zhejiang University, Hangzhou, China
- Institute of Sports Medicine, Zhejiang University, Hangzhou, China
| | - Yiwen Xu
- Department of Orthopedic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Orthopedics Research Institute of Zhejiang University, Hangzhou, China
- Institute of Sports Medicine, Zhejiang University, Hangzhou, China
| | - Honglu Cai
- Department of Orthopedic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Orthopedics Research Institute of Zhejiang University, Hangzhou, China
- Institute of Sports Medicine, Zhejiang University, Hangzhou, China
| | - Hengzhi Liu
- Department of Orthopedic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Orthopedics Research Institute of Zhejiang University, Hangzhou, China
- Institute of Sports Medicine, Zhejiang University, Hangzhou, China
| | - Yang Fei
- Department of Orthopedic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Orthopedics Research Institute of Zhejiang University, Hangzhou, China
- Institute of Sports Medicine, Zhejiang University, Hangzhou, China
| | - Boon Chin Heng
- Central Laboratory, Peking University School of Stomatology, Bejing, China
| | - Weishan Chen
- Department of Orthopedic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Orthopedics Research Institute of Zhejiang University, Hangzhou, China
- Institute of Sports Medicine, Zhejiang University, Hangzhou, China
- *Correspondence: Weishan Chen, ; Weiliang Shen,
| | - Weiliang Shen
- Department of Orthopedic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Orthopedics Research Institute of Zhejiang University, Hangzhou, China
- Institute of Sports Medicine, Zhejiang University, Hangzhou, China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou, China
- Dr. Li Dak Sum and Yip Yio Chin Center for Stem Cell and Regenerative Medicine, Zhejiang University, Hangzhou, China
- China Orthopaedic Regenerative Medicine (CORMed), Hangzhou, China
- *Correspondence: Weishan Chen, ; Weiliang Shen,
| |
Collapse
|
6
|
Zhang Y, Zhang J, Sun C, Wu F. Identification of the occurrence and potential mechanisms of heterotopic ossification associated with 17-beta-estradiol targeting MKX by bioinformatics analysis and cellular experiments. PeerJ 2022; 9:e12696. [PMID: 35036166 PMCID: PMC8734462 DOI: 10.7717/peerj.12696] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Accepted: 12/06/2021] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Tendon heterotopic ossification (HO) is a common condition occurring secondary to tendon injury or surgical trauma that significantly affects the patient's quality of life. The treatment of tendon HO remains challenging due to a lack of clarity regarding the pathological mechanism. Mohawk (MKX) is a key factor in preventing tendon HO; however, its upstream regulatory mechanism remains to be understood. This study aimed to identify potential compounds that target and regulate MKX and explore their functional mechanisms. METHODS Bioinformatics analysis of MKX-related compounds and proteins was performed based on data from the STITCH and OncoBinder databases. Subsequently, the SymMap database was used to study MKX-related traditional Chinese medicine drugs and symptoms. Next, the OncoBinder genomic and proteomic discovery model was applied to identify potential regulators of MKX. The analytical tool Expert Protein Analysis System for proteomics was used to predict the three-dimensional structure of MKX, and the AutoDockTools software was used to identify pockets of activity at potential sites for molecular docking. Furthermore, we evaluated the effect of different doses of 17-beta-estradiol on bone marrow-derived mesenchymal stem cells (BM-MSCs). RESULTS By predicting the three-dimensional structure of MKX and simulating molecular docking, Pro-Tyr and 17-beta-Estradiol were found to target and bind to MKX. Analysis of the STITCH and OncoBinder databases showed that MKX had a significant regulatory correlation with suppressor interacting 3 A/histone deacetylase 1 (SIN3A/HDAC1). The GO and KEGG pathway enrichment analysis revealed that the functions of MKX and its associated proteins were mainly enriched in osteogenic-related pathways. Assessment of the proliferation of BM-MSCs revealed that 17-beta-estradiol possibly upregulated the mRNA expression of the HDAC1-SIN3A/BMP pathway-related RUNX2, thereby promoting the proliferation of BM-MSCs. CONCLUSIONS The compounds Pro-Tyr and 17-beta-Estradiol may bind to MKX and thus affect the interaction of MKX with SIN3A/HDAC1.
Collapse
Affiliation(s)
- Yunpeng Zhang
- Department of surgery, Shanghai Fengxian District Central Hospital, Shanghai, China
| | - Jingwei Zhang
- Department of Orthopedics, Shanghai Fengxian District Central Hospital, Shanghai, China.,Shanghai University of Medicine & Health Sciences Affiliated Sixth People's Hospital South Campus, Shanghai, China
| | - Chenyu Sun
- Department of Internal Medicine, AMITA Health Saint Joseph Hospital Chicago, Chicago, Illinois, United States of America
| | - Fan Wu
- Department of surgery, Shanghai Fengxian District Central Hospital, Shanghai, China
| |
Collapse
|
7
|
Lin J, Yang Y, Zhou W, Dai C, Chen X, Xie Y, Han S, Liu H, Hu Y, Tang C, Bunpetch V, Zhang D, Chen Y, Zou X, Chen D, Liu W, Ouyang H. Single cell analysis reveals inhibition of angiogenesis attenuates the progression of heterotopic ossification in Mkx -/- mice. Bone Res 2022; 10:4. [PMID: 34996891 PMCID: PMC8741758 DOI: 10.1038/s41413-021-00175-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2020] [Accepted: 09/01/2021] [Indexed: 11/09/2022] Open
Abstract
Tendon heterotopic ossification (HO) is characterized by bone formation inside tendon tissue, which severely debilitates people in their daily life. Current therapies fail to promote functional tissue repair largely due to our limited understanding of HO pathogenesis. Here, we investigate the pathological mechanism and propose a potential treatment method for HO. Immunofluorescence assays showed that the Mohawk (MKX) expression level was decreased in human tendon HO tissue, coinciding with spontaneous HO and the upregulated expression of osteochondrogenic and angiogenic genes in the tendons of Mkx-/- mice. Single-cell RNA sequencing analyses of wild-type and Mkx-/- tendons identified three cell types and revealed the excessive activation of osteochondrogenic genes during the tenogenesis of Mkx-/- tendon cells. Single-cell analysis revealed that the gene expression program of angiogenesis, which is strongly associated with bone formation, was activated in all cell types during HO. Moreover, inhibition of angiogenesis by the small-molecule inhibitor BIBF1120 attenuated bone formation and angiogenesis in the Achilles tendons of both Mkx mutant mice and a rat traumatic model of HO. These findings provide new insights into the cellular mechanisms of tendon HO and highlight the inhibition of angiogenesis with BIBF1120 as a potential treatment strategy for HO.
Collapse
Affiliation(s)
- Junxin Lin
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cells and Regenerative Medicine, and Department of Orthopedic Surgery of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.,Department of Sports Medicine, Zhejiang University School of Medicine, Hangzhou, China.,Zhejiang University-University of Edinburgh Institute, Zhejiang University School of Medicine, and Key Laboratory of Tissue Engineering and Regenerative Medicine of Zhejiang Province, Hangzhou, China
| | - Yuwei Yang
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cells and Regenerative Medicine, and Department of Orthopedic Surgery of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.,Zhejiang University-University of Edinburgh Institute, Zhejiang University School of Medicine, and Key Laboratory of Tissue Engineering and Regenerative Medicine of Zhejiang Province, Hangzhou, China
| | - Wenyan Zhou
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cells and Regenerative Medicine, and Department of Orthopedic Surgery of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.,Zhejiang University-University of Edinburgh Institute, Zhejiang University School of Medicine, and Key Laboratory of Tissue Engineering and Regenerative Medicine of Zhejiang Province, Hangzhou, China
| | - Chao Dai
- Zhejiang University-University of Edinburgh Institute, Zhejiang University School of Medicine, and Key Laboratory of Tissue Engineering and Regenerative Medicine of Zhejiang Province, Hangzhou, China
| | - Xiao Chen
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cells and Regenerative Medicine, and Department of Orthopedic Surgery of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.,Department of Sports Medicine, Zhejiang University School of Medicine, Hangzhou, China.,Zhejiang University-University of Edinburgh Institute, Zhejiang University School of Medicine, and Key Laboratory of Tissue Engineering and Regenerative Medicine of Zhejiang Province, Hangzhou, China.,China Orthopedic Regenerative Medicine Group (CORMed), Hangzhou, China
| | - Yuanhao Xie
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cells and Regenerative Medicine, and Department of Orthopedic Surgery of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.,Department of Sports Medicine, Zhejiang University School of Medicine, Hangzhou, China
| | - Shan Han
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cells and Regenerative Medicine, and Department of Orthopedic Surgery of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.,Zhejiang University-University of Edinburgh Institute, Zhejiang University School of Medicine, and Key Laboratory of Tissue Engineering and Regenerative Medicine of Zhejiang Province, Hangzhou, China
| | - Huanhuan Liu
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cells and Regenerative Medicine, and Department of Orthopedic Surgery of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.,Zhejiang University-University of Edinburgh Institute, Zhejiang University School of Medicine, and Key Laboratory of Tissue Engineering and Regenerative Medicine of Zhejiang Province, Hangzhou, China
| | - Yejun Hu
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cells and Regenerative Medicine, and Department of Orthopedic Surgery of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.,Department of Sports Medicine, Zhejiang University School of Medicine, Hangzhou, China
| | - Chenqi Tang
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cells and Regenerative Medicine, and Department of Orthopedic Surgery of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.,Department of Sports Medicine, Zhejiang University School of Medicine, Hangzhou, China
| | - Varitsara Bunpetch
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cells and Regenerative Medicine, and Department of Orthopedic Surgery of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.,Zhejiang University-University of Edinburgh Institute, Zhejiang University School of Medicine, and Key Laboratory of Tissue Engineering and Regenerative Medicine of Zhejiang Province, Hangzhou, China
| | - Dandan Zhang
- Department of Pathology, Zhejiang University School of Medicine, Hangzhou, China
| | - Yishan Chen
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cells and Regenerative Medicine, and Department of Orthopedic Surgery of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.,Zhejiang University-University of Edinburgh Institute, Zhejiang University School of Medicine, and Key Laboratory of Tissue Engineering and Regenerative Medicine of Zhejiang Province, Hangzhou, China
| | - Xiaohui Zou
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cells and Regenerative Medicine, and Department of Orthopedic Surgery of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.,Zhejiang University-University of Edinburgh Institute, Zhejiang University School of Medicine, and Key Laboratory of Tissue Engineering and Regenerative Medicine of Zhejiang Province, Hangzhou, China.,Clinical Research Center, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Di Chen
- Zhejiang University-University of Edinburgh Institute, Zhejiang University School of Medicine, and Key Laboratory of Tissue Engineering and Regenerative Medicine of Zhejiang Province, Hangzhou, China.,Center for Reproductive Medicine, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Wanlu Liu
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cells and Regenerative Medicine, and Department of Orthopedic Surgery of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China. .,Zhejiang University-University of Edinburgh Institute, Zhejiang University School of Medicine, and Key Laboratory of Tissue Engineering and Regenerative Medicine of Zhejiang Province, Hangzhou, China.
| | - Hongwei Ouyang
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cells and Regenerative Medicine, and Department of Orthopedic Surgery of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China. .,Department of Sports Medicine, Zhejiang University School of Medicine, Hangzhou, China. .,Zhejiang University-University of Edinburgh Institute, Zhejiang University School of Medicine, and Key Laboratory of Tissue Engineering and Regenerative Medicine of Zhejiang Province, Hangzhou, China. .,China Orthopedic Regenerative Medicine Group (CORMed), Hangzhou, China.
| |
Collapse
|
8
|
Citeroni MR, Ciardulli MC, Russo V, Della Porta G, Mauro A, El Khatib M, Di Mattia M, Galesso D, Barbera C, Forsyth NR, Maffulli N, Barboni B. In Vitro Innovation of Tendon Tissue Engineering Strategies. Int J Mol Sci 2020; 21:E6726. [PMID: 32937830 PMCID: PMC7555358 DOI: 10.3390/ijms21186726] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2020] [Revised: 09/06/2020] [Accepted: 09/07/2020] [Indexed: 12/12/2022] Open
Abstract
Tendinopathy is the term used to refer to tendon disorders. Spontaneous adult tendon healing results in scar tissue formation and fibrosis with suboptimal biomechanical properties, often resulting in poor and painful mobility. The biomechanical properties of the tissue are negatively affected. Adult tendons have a limited natural healing capacity, and often respond poorly to current treatments that frequently are focused on exercise, drug delivery, and surgical procedures. Therefore, it is of great importance to identify key molecular and cellular processes involved in the progression of tendinopathies to develop effective therapeutic strategies and drive the tissue toward regeneration. To treat tendon diseases and support tendon regeneration, cell-based therapy as well as tissue engineering approaches are considered options, though none can yet be considered conclusive in their reproduction of a safe and successful long-term solution for full microarchitecture and biomechanical tissue recovery. In vitro differentiation techniques are not yet fully validated. This review aims to compare different available tendon in vitro differentiation strategies to clarify the state of art regarding the differentiation process.
Collapse
Affiliation(s)
- Maria Rita Citeroni
- Unit of Basic and Applied Biosciences, Faculty of Bioscience and Agro-Food and Environmental Technology, University of Teramo, 64100 Teramo, Italy; (V.R.); (A.M.); (M.E.K.); (M.D.M.); (B.B.)
| | - Maria Camilla Ciardulli
- Department of Medicine, Surgery and Dentistry, University of Salerno, Via S. Allende, 84081 Baronissi (SA), Italy; (M.C.C.); (G.D.P.); (N.M.)
| | - Valentina Russo
- Unit of Basic and Applied Biosciences, Faculty of Bioscience and Agro-Food and Environmental Technology, University of Teramo, 64100 Teramo, Italy; (V.R.); (A.M.); (M.E.K.); (M.D.M.); (B.B.)
| | - Giovanna Della Porta
- Department of Medicine, Surgery and Dentistry, University of Salerno, Via S. Allende, 84081 Baronissi (SA), Italy; (M.C.C.); (G.D.P.); (N.M.)
- Interdepartment Centre BIONAM, Università di Salerno, via Giovanni Paolo I, 84084 Fisciano (SA), Italy
| | - Annunziata Mauro
- Unit of Basic and Applied Biosciences, Faculty of Bioscience and Agro-Food and Environmental Technology, University of Teramo, 64100 Teramo, Italy; (V.R.); (A.M.); (M.E.K.); (M.D.M.); (B.B.)
| | - Mohammad El Khatib
- Unit of Basic and Applied Biosciences, Faculty of Bioscience and Agro-Food and Environmental Technology, University of Teramo, 64100 Teramo, Italy; (V.R.); (A.M.); (M.E.K.); (M.D.M.); (B.B.)
| | - Miriam Di Mattia
- Unit of Basic and Applied Biosciences, Faculty of Bioscience and Agro-Food and Environmental Technology, University of Teramo, 64100 Teramo, Italy; (V.R.); (A.M.); (M.E.K.); (M.D.M.); (B.B.)
| | - Devis Galesso
- Fidia Farmaceutici S.p.A., via Ponte della Fabbrica 3/A, 35031 Abano Terme (PD), Italy; (D.G.); (C.B.)
| | - Carlo Barbera
- Fidia Farmaceutici S.p.A., via Ponte della Fabbrica 3/A, 35031 Abano Terme (PD), Italy; (D.G.); (C.B.)
| | - Nicholas R. Forsyth
- Guy Hilton Research Centre, School of Pharmacy and Bioengineering, Keele University, Thornburrow Drive, Stoke on Trent ST4 7QB, UK;
| | - Nicola Maffulli
- Department of Medicine, Surgery and Dentistry, University of Salerno, Via S. Allende, 84081 Baronissi (SA), Italy; (M.C.C.); (G.D.P.); (N.M.)
- Department of Musculoskeletal Disorders, Faculty of Medicine and Surgery, University of Salerno, Via San Leonardo 1, 84131 Salerno, Italy
- Centre for Sports and Exercise Medicine, Barts and The London School of Medicine and Dentistry, Mile End Hospital, Queen Mary University of London, 275 Bancroft Road, London E1 4DG, UK
- School of Pharmacy and Bioengineering, Keele University School of Medicine, Thornburrow Drive, Stoke on Trent ST5 5BG, UK
| | - Barbara Barboni
- Unit of Basic and Applied Biosciences, Faculty of Bioscience and Agro-Food and Environmental Technology, University of Teramo, 64100 Teramo, Italy; (V.R.); (A.M.); (M.E.K.); (M.D.M.); (B.B.)
| |
Collapse
|
9
|
Nguyen PK, Pan XS, Li J, Kuo CK. Roadmap of molecular, compositional, and functional markers during embryonic tendon development. Connect Tissue Res 2018; 59:495-508. [PMID: 30231651 PMCID: PMC6669275 DOI: 10.1080/03008207.2018.1511710] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Tendon is a specialized connective tissue that connects muscle to bone, thereby enabling musculoskeletal movement. Tendon injury leads to formation of tissue with aberrant functional properties. Current approaches to treat tendon injuries, including surgical repair and tissue engineering, have not achieved normal tendon. A roadmap of markers could help with identifying when mis-steps occur during aberrant tendon formation and providing instructions for normal tendon formation. We propose this roadmap should be based on the embryo-the perfect model of tissue formation. Our prior studies have shown that adult mesenchymal stem cells mimic tendon progenitor cell behavior when treated with tendon developmental cues. Although transcription factors and extracellular matrix molecules are commonly used to assess tendon development, we have shown that these markers do not reliably reflect functional property elaboration. Thus, evaluating tendon formation on the basis of a combination of these molecular, compositional, and functional markers is important. In this review, we highlight various tendon markers with focus on their temporal profiles and roles in tendon development to outline a roadmap that may be useful for informing tendon healing and tissue engineering strategies.
Collapse
Affiliation(s)
- Phong K. Nguyen
- Department of Biomedical Engineering, University of Rochester, Rochester NY, USA.,Center for Musculoskeletal Research, University of Rochester School of Medicine, Rochester NY, USA
| | - Xuan Sabrina Pan
- Department of Biomedical Engineering, University of Rochester, Rochester NY, USA.,Center for Musculoskeletal Research, University of Rochester School of Medicine, Rochester NY, USA
| | - Jiewen Li
- Department of Biomedical Engineering, University of Rochester, Rochester NY, USA.,Center for Musculoskeletal Research, University of Rochester School of Medicine, Rochester NY, USA
| | - Catherine K. Kuo
- Department of Biomedical Engineering, University of Rochester, Rochester NY, USA.,Center for Musculoskeletal Research, University of Rochester School of Medicine, Rochester NY, USA.,Department of Orthopaedics, University of Rochester School of Medicine, Rochester NY, USA.,Genetics, Development, and Stem Cells Program, University of Rochester School of Medicine, Rochester NY, USA.,Materials Science Graduate Program, University of Rochester, Rochester NY, USA
| |
Collapse
|
10
|
Lim J, Munivez E, Jiang MM, Song IW, Gannon F, Keene DR, Schweitzer R, Lee BH, Joeng KS. mTORC1 Signaling is a Critical Regulator of Postnatal Tendon Development. Sci Rep 2017; 7:17175. [PMID: 29215029 PMCID: PMC5719403 DOI: 10.1038/s41598-017-17384-0] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2017] [Accepted: 11/24/2017] [Indexed: 12/11/2022] Open
Abstract
Tendons transmit contractile forces between musculoskeletal tissues. Whereas the biomechanical properties of tendons have been studied extensively, the molecular mechanisms regulating postnatal tendon development are not well understood. Here we examine the role of mTORC1 signaling in postnatal tendon development using mouse genetic approaches. Loss of mTORC1 signaling by removal of Raptor in tendons caused severe tendon defects postnatally, including decreased tendon thickness, indicating that mTORC1 is necessary for postnatal tendon development. By contrast, activation of mTORC1 signaling in tendons increased tendon cell numbers and proliferation. In addition, Tsc1 conditional knockout mice presented severely disorganized collagen fibers and neovascularization in the tendon midsubstance. Interestingly, collagen fibril diameter was significantly reduced in both Raptor and Tsc1 conditional knockout mice, albeit with variations in severity. We performed RNA-seq analysis using Achilles tendons to investigate the molecular changes underlying these tendon phenotypes. Raptor conditional knockout mice showed decreased extracellular matrix (ECM) structure-related gene expression, whereas Tsc1 conditional knockout mice exhibited changes in genes regulating TGF-β/BMP/FGF signaling, as well as in genes controlling ECM structure and disassembly. Collectively, our studies suggest that maintaining physiological levels of mTORC1 signaling is essential for postnatal tendon development and maturation.
Collapse
Affiliation(s)
- Joohyun Lim
- Department of Molecular and Human Genetics, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030, USA
| | - Elda Munivez
- Department of Molecular and Human Genetics, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030, USA
| | - Ming-Ming Jiang
- Department of Molecular and Human Genetics, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030, USA
| | - I-Wen Song
- Department of Molecular and Human Genetics, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030, USA
| | - Francis Gannon
- Department of Pathology, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030, USA
| | - Douglas R Keene
- Research Center, Shriners Hospital for Children, Portland, OR, 97239, USA
| | - Ronen Schweitzer
- Research Center, Shriners Hospital for Children, Portland, OR, 97239, USA
| | - Brendan H Lee
- Department of Molecular and Human Genetics, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030, USA.
| | - Kyu Sang Joeng
- Department of Molecular and Human Genetics, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030, USA
| |
Collapse
|
11
|
Havis E, Bonnin MA, Esteves de Lima J, Charvet B, Milet C, Duprez D. TGFβ and FGF promote tendon progenitor fate and act downstream of muscle contraction to regulate tendon differentiation during chick limb development. Development 2016; 143:3839-3851. [PMID: 27624906 DOI: 10.1242/dev.136242] [Citation(s) in RCA: 76] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2016] [Accepted: 08/25/2016] [Indexed: 01/02/2023]
Abstract
The molecular programme underlying tendon development has not been fully identified. Interactions with components of the musculoskeletal system are important for limb tendon formation. Limb tendons initiate their development independently of muscles; however, muscles are required for further tendon differentiation. We show that both FGF/ERK MAPK and TGFβ/SMAD2/3 signalling pathways are required and sufficient for SCX expression in chick undifferentiated limb cells, whereas the FGF/ERK MAPK pathway inhibits Scx expression in mouse undifferentiated limb mesodermal cells. During differentiation, muscle contraction is required to maintain SCX, TNMD and THBS2 expression in chick limbs. The activities of FGF/ERK MAPK and TGFβ/SMAD2/3 signalling pathways are decreased in tendons under immobilisation conditions. Application of FGF4 or TGFβ2 ligands prevents SCX downregulation in immobilised limbs. TGFβ2 but not FGF4 prevent TNMD and THBS2 downregulation under immobilisation conditions. We did not identify any intracellular crosstalk between both signalling pathways in their positive effect on SCX expression. Independently of each other, both FGF and TGFβ promote tendon commitment of limb mesodermal cells and act downstream of mechanical forces to regulate tendon differentiation during chick limb development.
Collapse
Affiliation(s)
- Emmanuelle Havis
- Sorbonne Universités, UPMC Univ Paris 06, CNRS UMR 7622, Inserm U1156, IBPS-Developmental Biology Laboratory, Paris F-75005, France
| | - Marie-Ange Bonnin
- Sorbonne Universités, UPMC Univ Paris 06, CNRS UMR 7622, Inserm U1156, IBPS-Developmental Biology Laboratory, Paris F-75005, France
| | - Joana Esteves de Lima
- Sorbonne Universités, UPMC Univ Paris 06, CNRS UMR 7622, Inserm U1156, IBPS-Developmental Biology Laboratory, Paris F-75005, France
| | - Benjamin Charvet
- Sorbonne Universités, UPMC Univ Paris 06, CNRS UMR 7622, Inserm U1156, IBPS-Developmental Biology Laboratory, Paris F-75005, France
| | - Cécile Milet
- Sorbonne Universités, UPMC Univ Paris 06, CNRS UMR 7622, Inserm U1156, IBPS-Developmental Biology Laboratory, Paris F-75005, France
| | - Delphine Duprez
- Sorbonne Universités, UPMC Univ Paris 06, CNRS UMR 7622, Inserm U1156, IBPS-Developmental Biology Laboratory, Paris F-75005, France
| |
Collapse
|
12
|
Kawasaki T, Takahashi M, Yajima H, Mori Y, Kawakami K. Six1 is required for mouse dental follicle cell and human periodontal ligament-derived cell proliferation. Dev Growth Differ 2016; 58:530-45. [DOI: 10.1111/dgd.12291] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2016] [Revised: 03/28/2016] [Accepted: 04/12/2016] [Indexed: 12/23/2022]
Affiliation(s)
- Tatsuki Kawasaki
- Department of Oral and Maxillofacial Surgery; Jichi Medical University; 3311-1, Yakushiji Shimotsuke Tochigi 329-0498 Japan
- Division of Biology; Center for Molecular Medicine; Jichi Medical University; 3311-1, Yakushiji Shimotsuke Tochigi 329-0498 Japan
| | - Masanori Takahashi
- Division of Biology; Center for Molecular Medicine; Jichi Medical University; 3311-1, Yakushiji Shimotsuke Tochigi 329-0498 Japan
| | - Hiroshi Yajima
- Division of Biology; Center for Molecular Medicine; Jichi Medical University; 3311-1, Yakushiji Shimotsuke Tochigi 329-0498 Japan
| | - Yoshiyuki Mori
- Department of Oral and Maxillofacial Surgery; Jichi Medical University; 3311-1, Yakushiji Shimotsuke Tochigi 329-0498 Japan
| | - Kiyoshi Kawakami
- Division of Biology; Center for Molecular Medicine; Jichi Medical University; 3311-1, Yakushiji Shimotsuke Tochigi 329-0498 Japan
| |
Collapse
|
13
|
Subramanian A, Schilling TF. Tendon development and musculoskeletal assembly: emerging roles for the extracellular matrix. Development 2016; 142:4191-204. [PMID: 26672092 DOI: 10.1242/dev.114777] [Citation(s) in RCA: 105] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Tendons and ligaments are extracellular matrix (ECM)-rich structures that interconnect muscles and bones. Recent work has shown how tendon fibroblasts (tenocytes) interact with muscles via the ECM to establish connectivity and strengthen attachments under tension. Similarly, ECM-dependent interactions between tenocytes and cartilage/bone ensure that tendon-bone attachments form with the appropriate strength for the force required. Recent studies have also established a close lineal relationship between tenocytes and skeletal progenitors, highlighting the fact that defects in signals modulated by the ECM can alter the balance between these fates, as occurs in calcifying tendinopathies associated with aging. The dynamic fine-tuning of tendon ECM composition and assembly thus gives rise to the remarkable characteristics of this unique tissue type. Here, we provide an overview of the functions of the ECM in tendon formation and maturation that attempts to integrate findings from developmental genetics with those of matrix biology.
Collapse
Affiliation(s)
- Arul Subramanian
- Department of Developmental and Cell Biology, University of California, Irvine, Irvine, CA 92697-2300, USA
| | - Thomas F Schilling
- Department of Developmental and Cell Biology, University of California, Irvine, Irvine, CA 92697-2300, USA
| |
Collapse
|
14
|
Gtf2ird1-Dependent Mohawk Expression Regulates Mechanosensing Properties of the Tendon. Mol Cell Biol 2016; 36:1297-309. [PMID: 26884464 PMCID: PMC4836271 DOI: 10.1128/mcb.00950-15] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2015] [Accepted: 02/08/2016] [Indexed: 11/20/2022] Open
Abstract
Mechanoforces experienced by an organ are translated into biological information for cellular sensing and response. In mammals, the tendon connective tissue experiences and resists physical forces, with tendon-specific mesenchymal cells called tenocytes orchestrating extracellular matrix (ECM) turnover. We show that Mohawk (Mkx), a tendon-specific transcription factor, is essential in mechanoresponsive tenogenesis through regulation of its downstream ECM genes such as type I collagens and proteoglycans such as fibromodulin both in vivo and in vitro Wild-type (WT) mice demonstrated an increase in collagen fiber diameter and density in response to physical treadmill exercise, whereas in Mkx(-/-) mice, tendons failed to respond to the same mechanical stimulation. Furthermore, functional screening of the Mkx promoter region identified several upstream transcription factors that regulate Mkx In particular, general transcription factor II-I repeat domain-containing protein 1 (Gtf2ird1) that is expressed in the cytoplasm of unstressed tenocytes translocated into the nucleus upon mechanical stretching to activate the Mkx promoter through chromatin regulation. Here, we demonstrate that Gtf2ird1 is essential for Mkx transcription, while also linking mechanical forces to Mkx-mediated tendon homeostasis and regeneration.
Collapse
|
15
|
Liu H, Zhang C, Zhu S, Lu P, Zhu T, Gong X, Zhang Z, Hu J, Yin Z, Heng BC, Chen X, Ouyang HW. Mohawk promotes the tenogenesis of mesenchymal stem cells through activation of the TGFβ signaling pathway. Stem Cells 2015; 33:443-55. [PMID: 25332192 DOI: 10.1002/stem.1866] [Citation(s) in RCA: 123] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2014] [Revised: 09/09/2014] [Accepted: 09/18/2014] [Indexed: 12/14/2022]
Abstract
The transcription factor Mohawk (Mkx) is expressed in developing tendons and is an important regulator of tenogenic differentiation. However, the exact roles of Mkx in tendinopathy and tendon repair remain unclear. Using gene expression Omnibus datasets and immunofluorescence assays, we found that Mkx expression level was dramatically lower in human tendinopathy tissue and it is activated at specific stages of tendon development. In mesenchymal stem cells (MSCs), ectopic Mkx expression strikingly promoted tenogenesis more efficiently than Scleraxis (Scx), a well-known master transcription factor of tendon. Significantly higher levels of tenogenic gene expression and collagen fibril growth were observed with Mkx versus Scx. Interestingly, it was observed that Mkx dramatically upregulated Scx through binding to the Tgfb2 promoter. Additionally, the transplantation of Mkx-expressing-MSC sheets promoted tendon repair in a mouse model of Achilles-tendon defect. Taken together, these data shed light on previously unrecognized roles of Mkx in tendinopathy, tenogenesis, and tendon repair as well as in regulating the TGFβ pathway.
Collapse
Affiliation(s)
- Huanhuan Liu
- Center for Stem Cell and Tissue Engineering, School of Medicine, Zhejiang University, Hangzhou, People's Republic of China; Zhejiang Provincial Key Laboratory of Tissue Engineering and Regenerative Medicine, Hangzhou, People's Republic of China
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
16
|
Gaut L, Duprez D. Tendon development and diseases. WILEY INTERDISCIPLINARY REVIEWS-DEVELOPMENTAL BIOLOGY 2015; 5:5-23. [PMID: 26256998 DOI: 10.1002/wdev.201] [Citation(s) in RCA: 67] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/20/2015] [Revised: 06/15/2015] [Accepted: 06/20/2015] [Indexed: 12/22/2022]
Abstract
Tendon is a uniaxial connective tissue component of the musculoskeletal system. Tendon is involved in force transmission between muscle and bone. Tendon injury is very common and debilitating but tendon repair remains a clinical challenge for orthopedic medicine. In vertebrates, tendon is mainly composed of type I collagen fibrils, displaying a parallel organization along the tendon axis. The tendon-specific spatial organization of type I collagen provides the mechanical properties for tendon function. In contrast to other components of the musculoskeletal system, tendon biology is poorly understood. An important goal in tendon biology is to understand the mechanisms involved in the production and assembly of type I collagen fibrils during development, postnatal formation, and healing processes in order to design new therapies for tendon repair. In this review we highlight the current understanding of the molecular and mechanical signals known to be involved in tenogenesis during development, and how development provides insights into tendon healing processes. WIREs Dev Biol 2016, 5:5-23. doi: 10.1002/wdev.201 For further resources related to this article, please visit the WIREs website.
Collapse
Affiliation(s)
- Ludovic Gaut
- CNRS UMR 7622, IBPS-Developmental Biology Laboratory, Paris, France.,Sorbonne Universités, UPMC Univ Paris 06, IBPS-Developmental Biology Laboratory, Paris, France.,Inserm U1156, Paris, France
| | - Delphine Duprez
- CNRS UMR 7622, IBPS-Developmental Biology Laboratory, Paris, France.,Sorbonne Universités, UPMC Univ Paris 06, IBPS-Developmental Biology Laboratory, Paris, France.,Inserm U1156, Paris, France
| |
Collapse
|
17
|
Milet C, Duprez D. The Mkx homeoprotein promotes tenogenesis in stem cells and improves tendon repair. ANNALS OF TRANSLATIONAL MEDICINE 2015; 3:S33. [PMID: 26046080 DOI: 10.3978/j.issn.2305-5839.2015.03.64] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Subscribe] [Scholar Register] [Received: 02/18/2015] [Accepted: 02/20/2015] [Indexed: 12/27/2022]
Affiliation(s)
- Cécile Milet
- 1 CNRS UMR 7622, IBPS-Developmental Biology Laboratory, F-75005, Paris, France ; 2 Sorbonne Universités, UPMC Univ Paris 06, IBPS-Developmental Biology Laboratory, F-75005, Paris, France ; 3 Inserm U1156, F-75005, Paris, France
| | - Delphine Duprez
- 1 CNRS UMR 7622, IBPS-Developmental Biology Laboratory, F-75005, Paris, France ; 2 Sorbonne Universités, UPMC Univ Paris 06, IBPS-Developmental Biology Laboratory, F-75005, Paris, France ; 3 Inserm U1156, F-75005, Paris, France
| |
Collapse
|
18
|
Huang AH, Lu HH, Schweitzer R. Molecular regulation of tendon cell fate during development. J Orthop Res 2015; 33:800-12. [PMID: 25664867 DOI: 10.1002/jor.22834] [Citation(s) in RCA: 86] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/10/2014] [Accepted: 01/16/2015] [Indexed: 02/04/2023]
Abstract
Although there have been several advances identifying novel mediators of tendon induction, differentiation, and patterning, much of the basic landscape of tendon biology from developmental stages onward remain almost completely undefined. During the New Frontiers in Tendon Research meeting, a group of developmental biologists with expertise across musculoskeletal disciplines identified key challenges for the tendon development field. The tools generated and the molecular regulators identified in developmental research have enhanced mechanistic studies in tendon injury and repair, both by defining benchmarks for success, as well as informing regenerative strategies. To address the needs of the orthopedic research community, this review will therefore focus on three key areas in tendon development that may have critical implications for the fields of tendon repair/regeneration and tendon tissue engineering, including functional markers of tendon cell identity, signaling regulators of tendon induction and differentiation, and in vitro culture models for tendon cell differentiation. Our goal is to provide a useful list of the currently known molecular players and their function in tendon differentiation within the context of development.
Collapse
Affiliation(s)
- Alice H Huang
- Department of Orthopaedics, Icahn School of Medicine at Mount Sinai, New York, NY
| | | | | |
Collapse
|
19
|
Chuang HN, Hsiao KM, Chang HY, Wu CC, Pan H. The homeobox transcription factor Irxl1 negatively regulates MyoD expression and myoblast differentiation. FEBS J 2014; 281:2990-3003. [PMID: 24814716 DOI: 10.1111/febs.12837] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2014] [Revised: 04/07/2014] [Accepted: 04/30/2014] [Indexed: 11/29/2022]
Abstract
Irxl1/Mkx (Iroquois homeobox-like 1/Mohawk) encodes a member of the TALE subfamily of homeodomain proteins. It is expressed in multiple mesoderm-derived tissues and has recently been shown to regulate tendon differentiation during mouse embryonic development. Previously we showed that knockdown of Irxl1 in zebrafish caused a deficit in neural crest cells which consequently resulted in deformation of craniofacial muscles and arch cartilages. Here, we further demonstrate that loss of Irxl1 function results in deformed somites with disordered muscle fibers and myotendinous junctions. Because expression of myoD is increased in the somites of Irxl1 knockdown morphants, we test whether Irxl1 negatively regulates myoD expression. When stable C2C12 myoblasts overexpressing Irxl1/Mkx were induced to differentiate, myotube formation was inhibited and protein levels of myoD and myosin heavy chain were decreased accordingly. A series of deletion constructs of myoD promoter fragments were tested by luciferase reporter assays, which identified a promoter fragment that is necessary and sufficient for Irxl1-mediated repression. Direct interaction of Irxl1 and myoD promoter was subsequently elucidated by yeast one-hybrid assays, electrophoretic mobility shift assays and chromatin immunoprecipitation analysis. Furthermore, mouse Mkx also binds to and represses myoD promoter. These results indicate that Irxl1/Mkx can repress myoD expression through direct binding to its promoter and may thus play a negative regulatory role in muscle differentiation.
Collapse
Affiliation(s)
- Han-Ni Chuang
- Department of Life Science and Institute of Molecular Biology, National Chung Cheng University, Chia-Yi, Taiwan; Department of Biomedical Sciences, Chung Shan Medical University, Taichung, Taiwan
| | | | | | | | | |
Collapse
|
20
|
Liu H, Zhu S, Zhang C, Lu P, Hu J, Yin Z, Ma Y, Chen X, OuYang H. Crucial transcription factors in tendon development and differentiation: their potential for tendon regeneration. Cell Tissue Res 2014; 356:287-98. [PMID: 24705622 DOI: 10.1007/s00441-014-1834-8] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2013] [Accepted: 01/30/2014] [Indexed: 12/22/2022]
Abstract
Tendons that connect muscles to bone are often the targets of sports injuries. The currently unsatisfactory state of tendon repair is largely attributable to the limited understanding of basic tendon biology. A number of tendon lineage-related transcription factors have recently been uncovered and provide clues for the better understanding of tendon development. Scleraxis and Mohawk have been identified as critical transcription factors in tendon development and differentiation. Other transcription factors, such as Sox9 and Egr1/2, have also been recently reported to be involved in tendon development. However, the molecular mechanisms and application of these transcription factors remain largely unclear and this prohibits their use in tendon therapy. Here, we systematically review and analyze recent findings and our own data concerning tendon transcription factors and tendon regeneration. Based on these findings, we provide interaction and temporal programming maps of transcription factors, as a basis for future tendon therapy. Finally, we discuss future directions for tendon regeneration with differentiation and trans-differentiation approaches based on transcription factors.
Collapse
Affiliation(s)
- Huanhuan Liu
- Center for Stem Cell and Tissue Engineering, School of Medicine, Zhejiang University, 866 Yu Hang Tang Road, Hangzhou, 310058, China
| | | | | | | | | | | | | | | | | |
Collapse
|
21
|
The Mohawk homeobox transcription factor regulates the differentiation of tendons and volar plates. J Orthop Sci 2014; 19:172-80. [PMID: 24166359 PMCID: PMC3943675 DOI: 10.1007/s00776-013-0485-z] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/18/2013] [Accepted: 10/08/2013] [Indexed: 12/27/2022]
Abstract
BACKGROUND Mohawk (Mkx) is a homeodomain-containing transcription factor that is expressed in various mesoderm-derived tissues, particularly in developing tendons. In this study, we investigate the exact expression pattern and functions of Mkx in forelimbs. METHODS We analyzed the forelimbs of Mkx knockout mice [from embryonic day (E) 18.5 to postnatal day (P) 28 weeks] by using knocked-in Venus signals, Masson trichrome staining, and hematoxylin and eosin (H&E) staining. RESULTS We detected Venus signals in forelimb tendons, pulleys, and volar plates (VPs) in P21 mice. In-depth histological analysis showed that compared to the wild-type mice, the Mkx knockout mice showed significant hypoplasia in the flexor digitorum profundus tendons from E18.5. The VPs and pulleys appeared normal until P0; however, by P14, they became increasingly thicker in Mkx-null mice compared to wild-type mice. The fiber alignment was particularly disrupted in VPs of Mkx-null mice. CONCLUSIONS These results suggest that Mkx is an important regulator of the differentiation of VPs and pulleys, as well as of tendon differentiation.
Collapse
|
22
|
Nakahara H, Hasegawa A, Otabe K, Ayabe F, Matsukawa T, Onizuka N, Ito Y, Ozaki T, Lotz MK, Asahara H. Transcription factor Mohawk and the pathogenesis of human anterior cruciate ligament degradation. ACTA ACUST UNITED AC 2013; 65:2081-9. [PMID: 23686683 DOI: 10.1002/art.38020] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2012] [Accepted: 05/09/2013] [Indexed: 01/04/2023]
Abstract
OBJECTIVE To investigate the expression and function of Mohawk (MKX) in human adult anterior cruciate ligament (ACL) tissue and ligament cells from normal and osteoarthritis (OA)-affected knees. METHODS Knee joints were obtained at autopsy (within 24-48 hours postmortem) from 13 donors with normal knees (mean ± SD age 36.9 ± 11.0 years), 16 donors with knee OA (age 79.7 ± 11.4 years), and 8 aging donors without knee OA (age 76.9 ± 12.9 years). All cartilage surfaces were graded macroscopically. MKX expression was analyzed by immunohistochemistry and quantitative polymerase chain reaction. ACL-derived cells were used to study regulation of MKX expression by interleukin-1β (IL-1β). MKX was knocked down with small interfering RNA (siRNA) to analyze the function of MKX in extracellular matrix (ECM) production and differentiation in ACL-derived cells. RESULTS The expression of MKX was significantly decreased in ACL-derived cells from OA knees compared with normal knees. Consistent with this finding, immunohistochemistry analysis showed that MKX-positive cells were significantly reduced in ACL tissue from OA donors, in particular in cells located in disorientated fibers. In ACL-derived cells, IL-1β strongly suppressed MKX expression and reduced expression of the ligament ECM genes COL1A1 and TNXB. In contrast, SOX9, a chondrocyte master transcription factor, was up-regulated by IL-1β treatment. Importantly, knockdown of MKX expression with siRNA up-regulated SOX9 expression in ACL-derived cells, whereas the expression of COL1A1 and TNXB was reduced. CONCLUSION Reduced expression of MKX is a feature of degenerated ACL in OA-affected joints, and this may be mediated in part by IL-1β. MKX appears necessary to maintain the tissue-specific cellular differentiation status and ECM production in adult human tendons and ligaments.
Collapse
|
23
|
Dyment NA, Liu CF, Kazemi N, Aschbacher-Smith LE, Kenter K, Breidenbach AP, Shearn JT, Wylie C, Rowe DW, Butler DL. The paratenon contributes to scleraxis-expressing cells during patellar tendon healing. PLoS One 2013; 8:e59944. [PMID: 23555841 PMCID: PMC3608582 DOI: 10.1371/journal.pone.0059944] [Citation(s) in RCA: 100] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2012] [Accepted: 02/19/2013] [Indexed: 01/27/2023] Open
Abstract
The origin of cells that contribute to tendon healing, specifically extrinsic epitenon/paratenon cells vs. internal tendon fibroblasts, is still debated. The purpose of this study is to determine the location and phenotype of cells that contribute to healing of a central patellar tendon defect injury in the mouse. Normal adult patellar tendon consists of scleraxis-expressing (Scx) tendon fibroblasts situated among aligned collagen fibrils. The tendon body is surrounded by paratenon, which consists of a thin layer of cells that do not express Scx and collagen fibers oriented circumferentially around the tendon. At 3 days following injury, the paratenon thickens as cells within the paratenon proliferate and begin producing tenascin-C and fibromodulin. These cells migrate toward the defect site and express scleraxis and smooth muscle actin alpha by day 7. The thickened paratenon tissue eventually bridges the tendon defect by day 14. Similarly, cells within the periphery of the adjacent tendon struts express these markers and become disorganized. Cells within the defect region show increased expression of fibrillar collagens (Col1a1 and Col3a1) but decreased expression of tenogenic transcription factors (scleraxis and mohawk homeobox) and collagen assembly genes (fibromodulin and decorin). By contrast, early growth response 1 and 2 are upregulated in these tissues along with tenascin-C. These results suggest that paratenon cells, which normally do not express Scx, respond to injury by turning on Scx and assembling matrix to bridge the defect. Future studies are needed to determine the signaling pathways that drive these cells and whether they are capable of producing a functional tendon matrix. Understanding this process may guide tissue engineering strategies in the future by stimulating these cells to improve tendon repair.
Collapse
Affiliation(s)
- Nathaniel A Dyment
- Department of Reconstructive Sciences, College of Dental Medicine, University of Connecticut Health Center, Farmington, Connecticut, United States of America.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Noorai RE, Freese NH, Wright LM, Chapman SC, Clark LA. Genome-wide association mapping and identification of candidate genes for the rumpless and ear-tufted traits of the Araucana chicken. PLoS One 2012; 7:e40974. [PMID: 22844420 PMCID: PMC3402462 DOI: 10.1371/journal.pone.0040974] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2012] [Accepted: 06/17/2012] [Indexed: 11/18/2022] Open
Abstract
Araucana chickens are known for their rounded, tailless rumps and tufted ears. Inheritance studies have shown that the rumpless (Rp) and ear-tufted (Et) loci each act in an autosomal dominant fashion, segregate independently, and are associated with an increased rate of embryonic mortality. To find genomic regions associated with Rp and Et, we generated genome-wide SNP profiles for a diverse population of 60 Araucana chickens using the 60 K chicken SNP BeadChip. Genome-wide association studies using 40 rumpless and 11 tailed birds showed a strong association with rumpless on Gga 2 (Praw = 2.45×10−10, Pgenome = 0.00575), and analysis of genotypes revealed a 2.14 Mb haplotype shared by all rumpless birds. Within this haplotype, a 0.74 Mb critical interval containing two Iroquois homeobox genes, Irx1 and Irx2, was unique to rumpless Araucana chickens. Irx1 and Irx2 are central for developmental prepatterning, but neither gene is known to have a role in mechanisms leading to caudal development. A second genome-wide association analysis using 30 ear-tufted and 28 non-tufted birds revealed an association with tufted on Gga 15 (Praw = 6.61×10−7, Pgenome = 0.0981). We identified a 0.58 Mb haplotype common to tufted birds and harboring 7 genes. Because homozygosity for Et is nearly 100% lethal, we employed a heterozygosity mapping approach to prioritize candidate gene selection. A 60 kb region heterozygous in all Araucana chickens contains the complete coding sequence for TBX1 and partial sequence for GNB1L. TBX1 is an important transcriptional regulator of embryonic development and a key genetic determinant of human DiGeorge syndrome. Herein, we describe localization of Rp and Et and identification of positional candidate genes.
Collapse
Affiliation(s)
- Rooksana E. Noorai
- Department of Genetics and Biochemistry, Clemson University, Clemson, South Carolina, United States of America
| | - Nowlan H. Freese
- Department of Biological Sciences, Clemson University, Clemson, South Carolina, United States of America
| | - Lindsay M. Wright
- Department of Genetics and Biochemistry, Clemson University, Clemson, South Carolina, United States of America
| | - Susan C. Chapman
- Department of Biological Sciences, Clemson University, Clemson, South Carolina, United States of America
- * E-mail: (SCC); (LAC)
| | - Leigh Anne Clark
- Department of Genetics and Biochemistry, Clemson University, Clemson, South Carolina, United States of America
- * E-mail: (SCC); (LAC)
| |
Collapse
|