1
|
Chung PH, Lin FH, Liu IH. Enhancing intrinsic TGF-β signaling via heparan sulfate glycosaminoglycan regulation to promote mesenchymal stem cell capabilities and chondrogenesis for cartilage repair. Int J Biol Macromol 2024; 282:137242. [PMID: 39505166 DOI: 10.1016/j.ijbiomac.2024.137242] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2024] [Revised: 10/21/2024] [Accepted: 11/02/2024] [Indexed: 11/08/2024]
Abstract
Osteoarthritis burdens patients due to the limited regenerative capacity of chondrocytes. Traditional cartilage repair often falls short, necessitating innovative approaches. Mesenchymal stem cells (MSCs) show promise for regeneration. Heparan sulfate glycosaminoglycans (HS-GAGs) regulate cellular functions, making them a target for cartilage repair. This study highlights how Heparinase III (HepIII) cleaves intact HS-GAGs in bone marrow-derived MSCs (BM-MSCs), enhancing their capabilities and specifically promoting chondrogenesis. HepIII-treated BM-MSCs cultured in a hanging drop device for three days, significantly increased cell number and aggregation into a cell sphere with early chondrogenesis. HepIII promoted BM-MSCs toward chondrogenesis, increasing type II collagen, intact HS-GAGs, and sulfated GAG content, while upregulating chondrogenic and heparan sulfate proteoglycan genes. Treatment with the TGF-β inhibitor (SB-431542) in HepIII-treated BM-MSCs demonstrated enhanced intrinsic transforming growth factor-β (TGF-β) signaling and fibronectin expression. This approach also boosted BM-MSC self-renewal, immunosuppressive potential, and modified acetylated histone signatures, offering a cost-effective strategy for cartilage repair by addressing inflammation, metabolic changes, and the high costs of traditional TGF-β methods. From the results, HepIII-treated BM-MSCs show potential for use in combination with other biopolymers as injectable gels to improve cartilage repair in osteoarthritis patients in the near future.
Collapse
Affiliation(s)
- Pei-Hsuan Chung
- Department of Animal Science and Technology, National Taiwan University, Taipei 106, Taiwan.
| | - Feng-Huei Lin
- Department of Biomedical Engineering, College of Medicine and College of Engineering, National Taiwan University, Taipei 106, Taiwan; Institute of Biomedical Engineering and Nanomedicine, National Health Research Institutes, Miaoli county 350, Taiwan.
| | - I-Hsuan Liu
- Department of Animal Science and Technology, National Taiwan University, Taipei 106, Taiwan; Research Center for Developmental Biology and Regenerative Medicine, National Taiwan University, Taipei 106, Taiwan.
| |
Collapse
|
2
|
Koosha E, Brenna CTA, Ashique AM, Jain N, Ovens K, Koike T, Kitagawa H, Eames BF. Proteoglycan inhibition of canonical BMP-dependent cartilage maturation delays endochondral ossification. Development 2024; 151:dev201716. [PMID: 38117077 PMCID: PMC10820745 DOI: 10.1242/dev.201716] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2023] [Accepted: 11/28/2023] [Indexed: 12/21/2023]
Abstract
During endochondral ossification, chondrocytes secrete a proteoglycan (PG)-rich extracellular matrix that can inhibit the process of cartilage maturation, including expression of Ihh and Col10a1. Because bone morphogenetic proteins (BMPs) can promote cartilage maturation, we hypothesized that cartilage PGs normally inhibit BMP signalling. Accordingly, BMP signalling was evaluated in chondrocytes of wild-type and PG mutant (fam20b-/-) zebrafish and inhibited with temporal control using the drug DMH1 or an inducible dominant-negative BMP receptor transgene (dnBMPR). Compared with wild type, phospho-Smad1/5/9, but not phospho-p38, was increased in fam20b-/- chondrocytes, but only after they secreted PGs. Phospho-Smad1/5/9 was decreased in DMH1-treated or dnBMPR-activated wild-type chondrocytes, and DMH1 also decreased phospho-p38 levels. ihha and col10a1a were decreased in DMH1-treated or dnBMPR-activated chondrocytes, and less perichondral bone formed. Finally, early ihha and col10a1a expression and early perichondral bone formation of fam20b mutants were rescued with DMH1 treatment or dnBMPR activation. Therefore, PG inhibition of canonical BMP-dependent cartilage maturation delays endochondral ossification, and these results offer hope for the development of growth factor therapies for skeletal defects of PG diseases.
Collapse
Affiliation(s)
- Elham Koosha
- Department of Anatomy, Physiology, and Pharmacology, University of Saskatchewan, Saskatoon, SK S7N 5E5, Canada
| | - Connor T. A. Brenna
- Department of Anatomy, Physiology, and Pharmacology, University of Saskatchewan, Saskatoon, SK S7N 5E5, Canada
| | - Amir M. Ashique
- Department of Anatomy, Physiology, and Pharmacology, University of Saskatchewan, Saskatoon, SK S7N 5E5, Canada
| | - Niteesh Jain
- Department of Anatomy, Physiology, and Pharmacology, University of Saskatchewan, Saskatoon, SK S7N 5E5, Canada
| | - Katie Ovens
- Department of Computer Science, University of Calgary, Calgary, AB T2N 1N4, Canada
| | - Toshiyasu Koike
- Department of Biochemistry, Kobe Pharmaceutical University, Higashinada-ku, Kobe 658-0003, Japan
| | - Hiroshi Kitagawa
- Department of Biochemistry, Kobe Pharmaceutical University, Higashinada-ku, Kobe 658-0003, Japan
| | - B. Frank Eames
- Department of Anatomy, Physiology, and Pharmacology, University of Saskatchewan, Saskatoon, SK S7N 5E5, Canada
| |
Collapse
|
3
|
Lovett BM, Hill KE, Randolph EM, Wang L, Schwarzbauer JE. Nucleation of fibronectin fibril assembly requires binding between heparin and the 13th type III module of fibronectin. J Biol Chem 2023; 299:104622. [PMID: 36933809 PMCID: PMC10124947 DOI: 10.1016/j.jbc.2023.104622] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Revised: 02/28/2023] [Accepted: 03/06/2023] [Indexed: 03/18/2023] Open
Abstract
Fibronectin (FN), a critical component of the extracellular matrix, is assembled into fibrils through a cell-mediated process. Heparan sulfate (HS) binds to the III13 module of FN, and fibroblasts lacking this glycosaminoglycan exhibit reduced FN fibril assembly. To determine if HS depends on III13 to control FN assembly, we deleted both III13 alleles in NIH 3T3 cells using the CRISPR-Cas9 system. ΔIII13 cells assembled fewer FN matrix fibrils and less DOC-insoluble FN matrix than wildtype cells. Little if any mutant FN matrix was assembled when purified ΔIII13 FN was provided to Chinese hamster ovary (CHO) cells, showing that lack of III13 caused the deficiency in assembly by ΔIII13 cells. Addition of heparin promoted the assembly of wildtype FN by CHO cells, but it had no effect on the assembly of ΔIII13 FN. Furthermore, heparin binding stabilized the folded conformation of III13 and prevented it from self-associating with increasing temperature suggesting that stabilization by HS/heparin binding might regulate interactions between III13 and other FN modules. This effect would be particularly important at matrix assembly sites where our data show that ΔIII13 cells require both exogenous wildtype FN and heparin in the culture medium to maximize assembly site formation. Our results show that heparin-promoted growth of fibril nucleation sites is dependent on III13. We conclude that HS/heparin binds to III13 to promote and control the nucleation and development of FN fibrils.
Collapse
Affiliation(s)
- Benjamin M Lovett
- Department of Molecular Biology, Princeton University, Princeton, New Jersey, USA
| | - Katherine E Hill
- Department of Molecular Biology, Princeton University, Princeton, New Jersey, USA
| | - Ellie M Randolph
- Department of Molecular Biology, Princeton University, Princeton, New Jersey, USA
| | - Luqiong Wang
- Department of Molecular Biology, Princeton University, Princeton, New Jersey, USA
| | - Jean E Schwarzbauer
- Department of Molecular Biology, Princeton University, Princeton, New Jersey, USA.
| |
Collapse
|
4
|
Pathogenic Roles of Heparan Sulfate and Its Use as a Biomarker in Mucopolysaccharidoses. Int J Mol Sci 2022; 23:ijms231911724. [PMID: 36233030 PMCID: PMC9570396 DOI: 10.3390/ijms231911724] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2022] [Revised: 09/29/2022] [Accepted: 09/30/2022] [Indexed: 11/17/2022] Open
Abstract
Heparan sulfate (HS) is an essential glycosaminoglycan (GAG) as a component of proteoglycans, which are present on the cell surface and in the extracellular matrix. HS-containing proteoglycans not only function as structural constituents of the basal lamina but also play versatile roles in various physiological processes, including cell signaling and organ development. Thus, inherited mutations of genes associated with the biosynthesis or degradation of HS can cause various diseases, particularly those involving the bones and central nervous system (CNS). Mucopolysaccharidoses (MPSs) are a group of lysosomal storage disorders involving GAG accumulation throughout the body caused by a deficiency of GAG-degrading enzymes. GAGs are stored differently in different types of MPSs. Particularly, HS deposition is observed in patients with MPS types I, II, III, and VII, all which involve progressive neuropathy with multiple CNS system symptoms. While therapies are available for certain symptoms in some types of MPSs, significant unmet medical needs remain, such as neurocognitive impairment. This review presents recent knowledge on the pathophysiological roles of HS focusing on the pathogenesis of MPSs. We also discuss the possible use and significance of HS as a biomarker for disease severity and therapeutic response in MPSs.
Collapse
|
5
|
Two Modulators of Skeletal Development: BMPs and Proteoglycans. J Dev Biol 2022; 10:jdb10020015. [PMID: 35466193 PMCID: PMC9036252 DOI: 10.3390/jdb10020015] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2022] [Revised: 03/21/2022] [Accepted: 03/23/2022] [Indexed: 12/27/2022] Open
Abstract
During embryogenesis, skeletal development is tightly regulated by locally secreted growth factors that interact with proteoglycans (PGs) in the extracellular matrix (ECM). Bone morphogenetic proteins (BMPs) are multifunctional growth factors that play critical roles in cartilage maturation and bone formation. BMP signals are transduced from plasma membrane receptors to the nucleus through both canonical Smad and noncanonical p38 mitogen-activated protein kinase (MAPK) pathways. BMP signalling is modulated by a variety of endogenous and exogenous molecular mechanisms at different spatiotemporal levels and in both positive and negative manners. As an endogenous example, BMPs undergo extracellular regulation by PGs, which generally regulate the efficiency of ligand-receptor binding. BMP signalling can also be exogenously perturbed by a group of small molecule antagonists, such as dorsomorphin and its derivatives, that selectively bind to and inhibit the intracellular kinase domain of BMP type I receptors. In this review, we present a current understanding of BMPs and PGs functions in cartilage maturation and osteoblast differentiation, highlighting BMP–PG interactions. We also discuss the identification of highly selective small-molecule BMP receptor type I inhibitors. This review aims to shed light on the importance of BMP signalling and PGs in cartilage maturation and bone formation.
Collapse
|
6
|
Mashima R, Okuyama T, Ohira M. Physiology and Pathophysiology of Heparan Sulfate in Animal Models: Its Biosynthesis and Degradation. Int J Mol Sci 2022; 23:1963. [PMID: 35216081 PMCID: PMC8876164 DOI: 10.3390/ijms23041963] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2022] [Revised: 02/05/2022] [Accepted: 02/08/2022] [Indexed: 12/17/2022] Open
Abstract
Heparan sulfate (HS) is a type of glycosaminoglycan that plays a key role in a variety of biological functions in neurology, skeletal development, immunology, and tumor metastasis. Biosynthesis of HS is initiated by a link of xylose to Ser residue of HS proteoglycans, followed by the formation of a linker tetrasaccharide. Then, an extension reaction of HS disaccharide occurs through polymerization of many repetitive units consisting of iduronic acid and N-acetylglucosamine. Subsequently, several modification reactions take place to complete the maturation of HS. The sulfation positions of N-, 2-O-, 6-O-, and 3-O- are all mediated by specific enzymes that may have multiple isozymes. C5-epimerization is facilitated by the epimerase enzyme that converts glucuronic acid to iduronic acid. Once these enzymatic reactions have been completed, the desulfation reaction further modifies HS. Apart from HS biosynthesis, the degradation of HS is largely mediated by the lysosome, an intracellular organelle with acidic pH. Mucopolysaccharidosis is a genetic disorder characterized by an accumulation of glycosaminoglycans in the body associated with neuronal, skeletal, and visceral disorders. Genetically modified animal models have significantly contributed to the understanding of the in vivo role of these enzymes. Their role and potential link to diseases are also discussed.
Collapse
Affiliation(s)
- Ryuichi Mashima
- Department of Clinical Laboratory Medicine, National Center for Child Health and Development, 2-10-1 Okura, Setagaya-ku, Tokyo 157-8535, Japan; (T.O.); (M.O.)
| | | | | |
Collapse
|
7
|
Hill KE, Lovett BM, Schwarzbauer JE. Heparan sulfate is necessary for the early formation of nascent fibronectin and collagen I fibrils at matrix assembly sites. J Biol Chem 2022; 298:101479. [PMID: 34890641 PMCID: PMC8801470 DOI: 10.1016/j.jbc.2021.101479] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Revised: 12/02/2021] [Accepted: 12/03/2021] [Indexed: 11/30/2022] Open
Abstract
Fibronectin (FN), an essential component of the extracellular matrix (ECM), is assembled via a cell-mediated process in which integrin receptors bind secreted FN and mediate its polymerization into fibrils that extend between cells, ultimately forming an insoluble matrix. Our previous work using mutant Chinese hamster ovary (CHO) cells identified the glycosaminoglycan heparan sulfate (HS) and its binding to FN as essential for the formation of insoluble FN fibrils. In this study, we investigated the contributions of HS at an early stage of the assembly process using knockdown of exostosin-1 (EXT1), one of the glycosyltransferases required for HS chain synthesis. NIH 3T3 fibroblasts with decreased EXT1 expression exhibited a significant reduction in both FN and type I collagen in the insoluble matrix. We show that FN fibril formation is initiated at matrix assembly sites, and while these sites were formed by cells with EXT1 knockdown, their growth was stunted compared with wild-type cells. The most severe defect observed was in the polymerization of nascent FN fibrils, which was reduced 2.5-fold upon EXT1 knockdown. This defect was rescued by the addition of exogenous soluble heparin chains long enough to simultaneously bind multiple FN molecules. The activity of soluble heparin in this process indicates that nascent fibril formation depends on HS more so than on the protein component of a specific HS proteoglycan. Together, our results suggest that heparin or HS is necessary for concentrating and localizing FN molecules at sites of early fibril assembly.
Collapse
Affiliation(s)
- Katherine E Hill
- Department of Molecular Biology, Princeton University, Princeton, New Jersey, USA
| | - Benjamin M Lovett
- Department of Molecular Biology, Princeton University, Princeton, New Jersey, USA
| | - Jean E Schwarzbauer
- Department of Molecular Biology, Princeton University, Princeton, New Jersey, USA.
| |
Collapse
|
8
|
Chen J, Sun T, You Y, Wu B, Wang X, Wu J. Proteoglycans and Glycosaminoglycans in Stem Cell Homeostasis and Bone Tissue Regeneration. Front Cell Dev Biol 2021; 9:760532. [PMID: 34917612 PMCID: PMC8669051 DOI: 10.3389/fcell.2021.760532] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2021] [Accepted: 10/25/2021] [Indexed: 12/20/2022] Open
Abstract
Stem cells maintain a subtle balance between self-renewal and differentiation under the regulatory network supported by both intracellular and extracellular components. Proteoglycans are large glycoproteins present abundantly on the cell surface and in the extracellular matrix where they play pivotal roles in facilitating signaling transduction and maintaining stem cell homeostasis. In this review, we outline distinct proteoglycans profiles and their functions in the regulation of stem cell homeostasis, as well as recent progress and prospects of utilizing proteoglycans/glycosaminoglycans as a novel glycomics carrier or bio-active molecules in bone regeneration.
Collapse
Affiliation(s)
- Jiawen Chen
- School of Stomatology, Southern Medical University, Guangzhou, China
| | - Tianyu Sun
- Department of Periodontology, Stomatological Hospital, Southern Medical University, Guangzhou, China
| | - Yan You
- School of Stomatology, Southern Medical University, Guangzhou, China
| | - Buling Wu
- School of Stomatology, Southern Medical University, Guangzhou, China.,Department of Endodontics, Shenzhen Stomatology Hospital, Southern Medical University, Shenzhen, China
| | - Xiaofang Wang
- Department of Biomedical Sciences, Texas A&M University College of Dentistry, Dallas, TX, United states
| | - Jingyi Wu
- Center of Oral Implantology, Stomatological Hospital, Southern Medical University, Guangzhou, China
| |
Collapse
|
9
|
Takahata Y, Hagino H, Kimura A, Urushizaki M, Kobayashi S, Wakamori K, Fujiwara C, Nakamura E, Yu K, Kiyonari H, Bando K, Murakami T, Komori T, Hata K, Nishimura R. Smoc1 and Smoc2 regulate bone formation as downstream molecules of Runx2. Commun Biol 2021; 4:1199. [PMID: 34667264 PMCID: PMC8526618 DOI: 10.1038/s42003-021-02717-7] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2019] [Accepted: 09/21/2021] [Indexed: 11/21/2022] Open
Abstract
Runx2 is an essential transcription factor for bone formation. Although osteocalcin, osteopontin, and bone sialoprotein are well-known Runx2-regulated bone-specific genes, the skeletal phenotypes of knockout (KO) mice for these genes are marginal compared with those of Runx2 KO mice. These inconsistencies suggest that unknown Runx2-regulated genes play important roles in bone formation. To address this, we attempted to identify the Runx2 targets by performing RNA-sequencing and found Smoc1 and Smoc2 upregulation by Runx2. Smoc1 or Smoc2 knockdown inhibited osteoblastogenesis. Smoc1 KO mice displayed no fibula formation, while Smoc2 KO mice had mild craniofacial phenotypes. Surprisingly, Smoc1 and Smoc2 double KO (DKO) mice manifested no skull, shortened tibiae, and no fibulae. Endochondral bone formation was also impaired at the late stage in the DKO mice. Collectively, these results suggest that Smoc1 and Smoc2 function as novel targets for Runx2, and play important roles in intramembranous and endochondral bone formation. Takahata et al. investigate the functional role of SMOC1/2 proteins in skeletal development. They reveal a genetic pathway that includes Bmp2 and Runx2 inducing expression of the paralogous Smoc genes, which may offer novel and effective therapeutic strategies associated with various bone and cartilage diseases.
Collapse
Affiliation(s)
- Yoshifumi Takahata
- Department of Molecular and Cellular Biochemistry, Osaka University Graduate School of Dentistry, Osaka, Japan.
| | - Hiromasa Hagino
- Department of Molecular and Cellular Biochemistry, Osaka University Graduate School of Dentistry, Osaka, Japan
| | - Ayaka Kimura
- Department of Molecular and Cellular Biochemistry, Osaka University Graduate School of Dentistry, Osaka, Japan
| | - Mitsuki Urushizaki
- Department of Molecular and Cellular Biochemistry, Osaka University Graduate School of Dentistry, Osaka, Japan
| | - Sachi Kobayashi
- Department of Molecular and Cellular Biochemistry, Osaka University Graduate School of Dentistry, Osaka, Japan
| | - Kanta Wakamori
- Department of Molecular and Cellular Biochemistry, Osaka University Graduate School of Dentistry, Osaka, Japan
| | - Chika Fujiwara
- Department of Molecular and Cellular Biochemistry, Osaka University Graduate School of Dentistry, Osaka, Japan
| | - Eriko Nakamura
- Department of Molecular and Cellular Biochemistry, Osaka University Graduate School of Dentistry, Osaka, Japan
| | - Kayon Yu
- Department of Molecular and Cellular Biochemistry, Osaka University Graduate School of Dentistry, Osaka, Japan
| | - Hiroshi Kiyonari
- Laboratory for Animal Resources and Genetic Engineering, RIKEN Center for Biosystems Dynamics Research, Hyogo, Japan
| | - Kana Bando
- Laboratory for Animal Resources and Genetic Engineering, RIKEN Center for Biosystems Dynamics Research, Hyogo, Japan
| | - Tomohiko Murakami
- Department of Molecular and Cellular Biochemistry, Osaka University Graduate School of Dentistry, Osaka, Japan
| | - Toshihisa Komori
- Basic and Translational Research Center for Hard Tissue Disease, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Kenji Hata
- Department of Molecular and Cellular Biochemistry, Osaka University Graduate School of Dentistry, Osaka, Japan
| | - Riko Nishimura
- Department of Molecular and Cellular Biochemistry, Osaka University Graduate School of Dentistry, Osaka, Japan
| |
Collapse
|
10
|
Matsuzawa T, Morita M, Shimane A, Otsuka R, Mei Y, Irie F, Yamaguchi Y, Yanai K, Yoshikawa T. Heparan sulfate promotes differentiation of white adipocytes to maintain insulin sensitivity and glucose homeostasis. J Biol Chem 2021; 297:101006. [PMID: 34310946 PMCID: PMC8379462 DOI: 10.1016/j.jbc.2021.101006] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2021] [Revised: 07/16/2021] [Accepted: 07/22/2021] [Indexed: 11/29/2022] Open
Abstract
Heparan sulfate (HS), a highly sulfated linear polysaccharide, is involved in diverse biological functions in various tissues. Although previous studies have suggested a possible contribution of HS to the differentiation of white adipocytes, there has been no direct evidence supporting this. Here, we inhibited the synthesis of HS chains in 3T3-L1 cells using CRISPR–Cas9 technology, resulting in impaired differentiation of adipocytes with attenuated bone morphogenetic protein 4 (BMP4)–fibroblast growth factor 1 (FGF1) signaling pathways. HS reduction resulted in reduced glucose uptake and decreased insulin-dependent intracellular signaling. We then made heterozygous mutant mice for the Ext1 gene, which encodes an enzyme essential for the HS biosynthesis, specifically in the visceral white adipose tissue (Fabp4-Cre+::Ext1flox/WT mice, hereafter called Ext1Δ/WT) to confirm the importance of HS in vivo. The expression levels of transcription factors that control adipocyte differentiation, such as peroxisome proliferator–activated receptor gamma, were reduced in Ext1Δ/WT adipocytes, which contained smaller, unilocular lipid droplets, reduced levels of enzymes involved in lipid synthesis, and altered expression of BMP4–FGF1 signaling molecules. Furthermore, we examined the impact of HS reduction in visceral white adipose tissue on systemic glucose homeostasis. We observed that Ext1Δ/WT mice showed glucose intolerance because of insulin resistance. Our results demonstrate that HS plays a crucial role in the differentiation of white adipocytes through BMP4–FGF1 signaling pathways, thereby contributing to insulin sensitivity and glucose homeostasis.
Collapse
Affiliation(s)
- Takuro Matsuzawa
- Department of Pharmacology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Masanobu Morita
- Department of Environmental Medicine and Molecular Toxicology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Ai Shimane
- Department of Pharmacology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Rina Otsuka
- Department of Pharmacology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Yu Mei
- Department of Pharmacology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Fumitoshi Irie
- Human Genetics Program, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, California, USA
| | - Yu Yamaguchi
- Human Genetics Program, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, California, USA
| | - Kazuhiko Yanai
- Department of Pharmacology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Takeo Yoshikawa
- Department of Pharmacology, Tohoku University Graduate School of Medicine, Sendai, Japan.
| |
Collapse
|
11
|
Dubail J, Cormier-Daire V. Chondrodysplasias With Multiple Dislocations Caused by Defects in Glycosaminoglycan Synthesis. Front Genet 2021; 12:642097. [PMID: 34220933 PMCID: PMC8242584 DOI: 10.3389/fgene.2021.642097] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Accepted: 05/04/2021] [Indexed: 11/13/2022] Open
Abstract
Chondrodysplasias with multiple dislocations form a group of severe disorders characterized by joint laxity and multiple dislocations, severe short stature of pre- and post-natal onset, hand anomalies, and/or vertebral anomalies. The majority of chondrodysplasias with multiple dislocations have been associated with mutations in genes encoding glycosyltransferases, sulfotransferases, and transporters implicated in the synthesis or sulfation of glycosaminoglycans, long and unbranched polysaccharides composed of repeated disaccharide bond to protein core of proteoglycan. Glycosaminoglycan biosynthesis is a tightly regulated process that occurs mainly in the Golgi and that requires the coordinated action of numerous enzymes and transporters as well as an adequate Golgi environment. Any disturbances of this chain of reactions will lead to the incapacity of a cell to construct correct glycanic chains. This review focuses on genetic and glycobiological studies of chondrodysplasias with multiple dislocations associated with glycosaminoglycan biosynthesis defects and related animal models. Strong comprehension of the molecular mechanisms leading to those disorders, mostly through extensive phenotypic analyses of in vitro and/or in vivo models, is essential for the development of novel biomarkers for clinical screenings and innovative therapeutics for these diseases.
Collapse
Affiliation(s)
- Johanne Dubail
- Université de Paris, INSERM UMR 1163, Institut Imagine, Paris, France
| | - Valérie Cormier-Daire
- Université de Paris, INSERM UMR 1163, Institut Imagine, Paris, France.,Service de Génétique Clinique, Centre de Référence Pour Les Maladies Osseuses Constitutionnelles, AP-HP, Hôpital Necker-Enfants Malades, Paris, France
| |
Collapse
|
12
|
Heparan Sulfate Deficiency in Cartilage: Enhanced BMP-Sensitivity, Proteoglycan Production and an Anti-Apoptotic Expression Signature after Loading. Int J Mol Sci 2021; 22:ijms22073726. [PMID: 33918436 PMCID: PMC8038223 DOI: 10.3390/ijms22073726] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Revised: 03/29/2021] [Accepted: 03/31/2021] [Indexed: 12/15/2022] Open
Abstract
Osteoarthritis (OA) represents one major cause of disability worldwide still evading efficient pharmacological or cellular therapies. Severe degeneration of extracellular cartilage matrix precedes the loss of mobility and disabling pain perception in affected joints. Recent studies showed that a reduced heparan sulfate (HS) content protects cartilage from degradation in OA-animal models of joint destabilization but the underlying mechanisms remained unclear. We aimed to clarify whether low HS-content alters the mechano-response of chondrocytes and to uncover pathways relevant for HS-related chondro-protection in response to loading. Tissue-engineered cartilage with HS-deficiency was generated from rib chondrocytes of mice carrying a hypomorphic allele of Exostosin 1 (Ext1), one of the main HS-synthesizing enzymes, and wildtype (WT) littermate controls. Engineered cartilage matured for 2 weeks was exposed to cyclic unconfined compression in a bioreactor. The molecular loading response was determined by transcriptome profiling, bioinformatic data processing, and qPCR. HS-deficient chondrocytes expressed 3-6% of WT Ext1-mRNA levels. Both groups similarly raised Sox9, Col2a1 and Acan levels during maturation. However, HS-deficient chondrocytes synthesized and deposited 50% more GAG/DNA. TGFβ and FGF2-sensitivity of Ext1gt/gt chondrocytes was similar to WT cells but their response to BMP-stimulation was enhanced. Loading induced similar activation of mechano-sensitive ERK and P38-signaling in WT and HS-reduced chondrocytes. Transcriptome analysis reflected regulation of cell migration as major load-induced biological process with similar stimulation of common (Fosl1, Itgα5, Timp1, and Ngf) as well as novel mechano-regulated genes (Inhba and Dhrs9). Remarkably, only Ext1-hypomorphic cartilage responded to loading by an expression signature of negative regulation of apoptosis with pro-apoptotic Bnip3 being selectively down-regulated. HS-deficiency enhanced BMP-sensitivity, GAG-production and fostered an anti-apoptotic expression signature after loading, all of which may protect cartilage from load-induced erosion.
Collapse
|
13
|
Al-Zayed Z, Al-Rijjal RA, Al-Ghofaili L, BinEssa HA, Pant R, Alrabiah A, Al-Hussainan T, Zou M, Meyer BF, Shi Y. Mutation spectrum of EXT1 and EXT2 in the Saudi patients with hereditary multiple exostoses. Orphanet J Rare Dis 2021; 16:100. [PMID: 33632255 PMCID: PMC7905910 DOI: 10.1186/s13023-021-01738-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Accepted: 02/11/2021] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Hereditary Multiple Exostoses (HME), also known as Multiple Osteochondromas (MO) is a rare genetic disorder characterized by multiple benign cartilaginous bone tumors, which are caused by mutations in the genes for exostosin glycosyltransferase 1 (EXT1) and exostosin glycosyltransferase 2 (EXT2). The genetic defects have not been studied in the Saudi patients. AIM OF STUDY We investigated mutation spectrum of EXT1 and EXT2 in 22 patients from 17 unrelated families. METHODS Genomic DNA was extracted from peripheral leucocytes. The coding regions and intron-exon boundaries of both EXT1 and EXT2 genes were screened for mutations by PCR-sequencing analysis. Gross deletions were analyzed by MLPA analysis. RESULTS EXT1 mutations were detected in 6 families (35%) and 3 were novel mutations: c.739G > T (p. E247*), c.1319delG (p.R440Lfs*4), and c.1786delA (p.S596Afs*25). EXT2 mutations were detected in 7 families (41%) and 3 were novel mutations: c.541delG (p.D181Ifs*89), c.583delG (p.G195Vfs*75), and a gross deletion of approximately 10 kb including promoter and exon 1. Five patients from different families had no family history and carried de novo mutations (29%, 5/17). No EXT1 and EXT2 mutations were found in the remaining four families. In total, EXT1 and EXT2 mutations were found in 77% (13/17) of Saudi HME patients. CONCLUSION EXT1 and EXT2 mutations contribute significantly to the pathogenesis of HME in the Saudi population. In contrast to high mutation rate in EXT 1 (65%) and low mutation rate in EXT2 (25%) in other populations, the frequency of EXT2 mutations are much higher (41%) and comparable to that of EXT1 among Saudi patients. De novo mutations are also common and the six novel EXT1/EXT2 mutations further expands the mutation spectrum of HME.
Collapse
Affiliation(s)
- Zayed Al-Zayed
- Department of Orthopedics, King Faisal Specialist Hospital and Research Center, Riyadh, Saudi Arabia.,College of Medicine, Alfaisal University, Riyadh, Saudi Arabia
| | - Roua A Al-Rijjal
- Department of Genetics, MBC 3, Centre for Genomic Medicine, King Faisal Specialist Hospital and Research Center, P.O. Box 3354, Riyadh, 11211, Saudi Arabia
| | | | - Huda A BinEssa
- Department of Genetics, MBC 3, Centre for Genomic Medicine, King Faisal Specialist Hospital and Research Center, P.O. Box 3354, Riyadh, 11211, Saudi Arabia
| | - Rajeev Pant
- Department of Orthopedics, King Faisal Specialist Hospital and Research Center, Riyadh, Saudi Arabia
| | - Anwar Alrabiah
- Department of Orthopedics, King Faisal Specialist Hospital and Research Center, Riyadh, Saudi Arabia.,College of Medicine, Alfaisal University, Riyadh, Saudi Arabia
| | - Thamer Al-Hussainan
- Department of Orthopedics, King Faisal Specialist Hospital and Research Center, Riyadh, Saudi Arabia.,College of Medicine, Alfaisal University, Riyadh, Saudi Arabia
| | - Minjing Zou
- Department of Genetics, MBC 3, Centre for Genomic Medicine, King Faisal Specialist Hospital and Research Center, P.O. Box 3354, Riyadh, 11211, Saudi Arabia
| | - Brian F Meyer
- Department of Genetics, MBC 3, Centre for Genomic Medicine, King Faisal Specialist Hospital and Research Center, P.O. Box 3354, Riyadh, 11211, Saudi Arabia
| | - Yufei Shi
- Department of Genetics, MBC 3, Centre for Genomic Medicine, King Faisal Specialist Hospital and Research Center, P.O. Box 3354, Riyadh, 11211, Saudi Arabia.
| |
Collapse
|
14
|
Kawashima K, Ogawa H, Komura S, Ishihara T, Yamaguchi Y, Akiyama H, Matsumoto K. Heparan sulfate deficiency leads to hypertrophic chondrocytes by increasing bone morphogenetic protein signaling. Osteoarthritis Cartilage 2020; 28:1459-1470. [PMID: 32818603 PMCID: PMC7606622 DOI: 10.1016/j.joca.2020.08.003] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/08/2020] [Revised: 08/03/2020] [Accepted: 08/11/2020] [Indexed: 02/02/2023]
Abstract
OBJECTIVE Exostosin-1 (EXT1) and EXT2 are the major genetic etiologies of multiple hereditary exostoses and are essential for heparan sulfate (HS) biosynthesis. Previous studies investigating HS in several mouse models of multiple hereditary exostoses have reported that aberrant bone morphogenetic protein (BMP) signaling promotes osteochondroma formation in Ext1-deficient mice. This study examined the mechanism underlying the effects of HS deficiency on BMP/Smad signaling in articular cartilage in a cartilage-specific Ext-/- mouse model. METHOD We generated mice with a conditional Ext1 knockout in cartilage tissue (Ext1-cKO mice) using Prg4-Cre transgenic mice. Structural cartilage alterations were histologically evaluated and phospho-Smad1/5/9 (pSmad1/5/9) expression in mouse chondrocytes was analyzed. The effect of pharmacological intervention of BMP signaling using a specific inhibitor was assessed in the articular cartilage of Ext1-cKO mice. RESULTS Hypertrophic chondrocytes were significantly more abundant (P = 0.021) and cartilage thickness was greater in Ext1-cKO mice at 3 months postnatal than in control littermates (P = 0.036 for femur; and P < 0.001 for tibia). However, osteoarthritis did not spontaneously occur before the 1-year follow-up. matrix metalloproteinase (MMP)-13 and adamalysin-like metalloproteinases with thrombospondin motifs(ADAMTS)-5 were upregulated in hypertrophic chondrocytes of transgenic mice. Immunostaining and western blotting revealed that pSmad1/5/9-positive chondrocytes were more abundant in the articular cartilage of Ext1-cKO mice than in control littermates. Furthermore, the BMP inhibitor significantly decreased the number of hypertrophic chondrocytes in Ext1-cKO mice (P = 0.007). CONCLUSIONS HS deficiency in articular chondrocytes causes chondrocyte hypertrophy, wherein upregulated BMP/Smad signaling partially contributes to this phenotype. HS might play an important role in maintaining the cartilaginous matrix by regulating BMP signaling.
Collapse
Affiliation(s)
- K. Kawashima
- Department of Orthopaedic Surgery, Gifu University Graduate School of Medicine, 1-1 Yanagido, Gifu, Japan
| | - H. Ogawa
- Department of Orthopaedic Surgery, Gifu University Graduate School of Medicine, 1-1 Yanagido, Gifu, Japan
| | - S. Komura
- Department of Orthopaedic Surgery, Gifu University Graduate School of Medicine, 1-1 Yanagido, Gifu, Japan
| | - T. Ishihara
- Innovative and Clinical Research Promotion Center, Gifu University Hospital, 1-1 Yanagido, Gifu, Japan
| | - Y. Yamaguchi
- Human Genetics Program, Sanford Burnham Prebys Medical Discovery Institute, 10901 North Torrey Pines Road, La Jolla, CA, 92037, USA
| | - H. Akiyama
- Department of Orthopaedic Surgery, Gifu University Graduate School of Medicine, 1-1 Yanagido, Gifu, Japan
| | - K. Matsumoto
- Department of Orthopaedic Surgery, Gifu University Graduate School of Medicine, 1-1 Yanagido, Gifu, Japan,Address correspondence and reprint requests to: K. Matsumoto, Department of Orthopedic Surgery, Gifu University Graduate School of Medicine, 1-1 Yanagido, Gifu, 501-1194, Japan. Tel.: 81-58-230-6333; Fax: 81-58-230-6334. (K. Matsumoto)
| |
Collapse
|
15
|
Severmann AC, Jochmann K, Feller K, Bachvarova V, Piombo V, Stange R, Holzer T, Brachvogel B, Esko J, Pap T, Hoffmann D, Vortkamp A. An altered heparan sulfate structure in the articular cartilage protects against osteoarthritis. Osteoarthritis Cartilage 2020; 28:977-987. [PMID: 32315715 PMCID: PMC8422443 DOI: 10.1016/j.joca.2020.04.002] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/23/2020] [Revised: 04/05/2020] [Accepted: 04/09/2020] [Indexed: 02/02/2023]
Abstract
OBJECTIVE Osteoarthritis (OA) is a progressive degenerative disease of the articular cartilage caused by an unbalanced activity of proteases, cytokines and other secreted proteins. Since heparan sulfate (HS) determines the activity of many extracellular factors, we investigated its role in OA progression. METHODS To analyze the role of the HS level, OA was induced by anterior cruciate ligament transection (ACLT) in transgenic mice carrying a loss-of-function allele of Ext1 in clones of chondrocytes (Col2-rtTA-Cre;Ext1e2fl/e2fl). To study the impact of the HS sulfation pattern, OA was surgically induced in mice with a heterozygous (Ndst1+/-) or chondrocyte-specific (Col2-Cre;Ndst1fl/fl) loss-of-function allele of the sulfotransferase Ndst1. OA progression was evaluated using the OARSI scoring system. To investigate expression and activity of cartilage degrading proteases, femoral head explants of Ndst1+/- mutants were analyzed by qRT-PCR, Western Blot and gelatin zymography. RESULTS All investigated mouse strains showed reduced OA scores (Col2-rtTA-Cre;Ext1e2fl/e2fl: 0.83; 95% HDI 0.72-0.96; Ndst1+/-: 0.83, 95% HDI 0.74-0.9; Col2-Cre;Ndst1fl/fl: 0.87, 95% HDI 0.76-1). Using cartilage explant cultures of Ndst1 animals, we detected higher amounts of aggrecan degradation products in wildtype samples (NITEGE 4.24-fold, 95% HDI 1.05-18.55; VDIPEN 1.54-fold, 95% HDI 1.54-2.34). Accordingly, gelatin zymography revealed lower Mmp2 activity in mutant samples upon RA-treatment (0.77-fold, 95% HDI: 0.60-0.96). As expression of major proteases and their inhibitors was not altered, HS seems to regulate cartilage degeneration by affecting protease activity. CONCLUSION A decreased HS content or a reduced sulfation level protect against OA progression by regulating protease activity rather than expression.
Collapse
Affiliation(s)
- A-C Severmann
- Department of Developmental Biology, Center for Medical Biotechnology, Faculty Biology, University Duisburg-Essen, Germany.
| | - K Jochmann
- Department of Developmental Biology, Center for Medical Biotechnology, Faculty Biology, University Duisburg-Essen, Germany.
| | - K Feller
- Department of Developmental Biology, Center for Medical Biotechnology, Faculty Biology, University Duisburg-Essen, Germany.
| | - V Bachvarova
- Department of Developmental Biology, Center for Medical Biotechnology, Faculty Biology, University Duisburg-Essen, Germany.
| | - V Piombo
- Department of Developmental Biology, Center for Medical Biotechnology, Faculty Biology, University Duisburg-Essen, Germany.
| | - R Stange
- Zentrum für Muskuloskelettale Medizin, Westfälische Wilhelms-Universität Münster, Germany.
| | - T Holzer
- Center for Biochemistry, Department of Pediatrics and Adolescent Medicine, Experimental Neonatology, Medical Faculty, University of Cologne, Germany.
| | - B Brachvogel
- Center for Biochemistry, Department of Pediatrics and Adolescent Medicine, Experimental Neonatology, Medical Faculty, University of Cologne, Germany.
| | - J Esko
- Department of Cellular and Molecular Medicine, Glycobiology Research & Training Center, University of California, San Diego, La Jolla, CA, 92093-0687, USA.
| | - T Pap
- Zentrum für Muskuloskelettale Medizin, Westfälische Wilhelms-Universität Münster, Germany.
| | - D Hoffmann
- Department Bioinformatics and Computational Biophysics, Center for Medical Biotechnology, Faculty Biology, University Duisburg-Essen, Germany.
| | - A Vortkamp
- Department of Developmental Biology, Center for Medical Biotechnology, Faculty Biology, University Duisburg-Essen, Germany.
| |
Collapse
|
16
|
Kodama K, Takahashi H, Oiji N, Nakano K, Okamura T, Niimi K, Takahashi E, Guo L, Ikegawa S, Furuichi T. CANT1 deficiency in a mouse model of Desbuquois dysplasia impairs glycosaminoglycan synthesis and chondrocyte differentiation in growth plate cartilage. FEBS Open Bio 2020; 10:1096-1103. [PMID: 32277574 PMCID: PMC7262921 DOI: 10.1002/2211-5463.12859] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2019] [Revised: 03/24/2020] [Accepted: 04/03/2020] [Indexed: 12/19/2022] Open
Abstract
Desbuquois dysplasia (DD) type 1 is a rare skeletal dysplasia characterized by a short stature, round face, progressive scoliosis, and joint laxity. The causative gene has been identified as calcium‐activated nucleotidase 1 (CANT1), which encodes a nucleotidase that preferentially hydrolyzes UDP to UMP and phosphate. In this study, we generated Cant1 KO mice using CRISPR/Cas9‐mediated genome editing. All F0 mice possessing frameshift deletions at both Cant1 alleles exhibited a dwarf phenotype. Germline transmission of the edited allele was confirmed in an F0 heterozygous mouse, and KO mice were generated by crossing of the heterozygous breeding pairs. Cant1 KO mice exhibited skeletal defects, including short stature, thoracic kyphosis, and delta phalanx, all of which are observed in DD type 1 patients. The glycosaminoglycan (GAG) content and extracellular matrix space were reduced in the growth plate cartilage of mutants, and proliferating chondrocytes lost their typical flat shape and became round. Chondrocyte differentiation, especially terminal differentiation to hypertrophic chondrocytes, was impaired in Cant1 KO mice. These findings indicate that CANT1 is involved in the synthesis of GAG and regulation of chondrocyte differentiation in the cartilage and contribute to a better understanding of the pathogenesis of DD type 1.
Collapse
Affiliation(s)
- Kazuki Kodama
- Laboratory of Laboratory Animal Science and Medicine, Co-Department of Veterinary Medicine, Faculty of Agriculture, Iwate University, Morioka, Japan
| | - Hiroaki Takahashi
- Laboratory of Laboratory Animal Science and Medicine, Co-Department of Veterinary Medicine, Faculty of Agriculture, Iwate University, Morioka, Japan
| | - Nobuyasu Oiji
- Laboratory of Laboratory Animal Science and Medicine, Co-Department of Veterinary Medicine, Faculty of Agriculture, Iwate University, Morioka, Japan
| | - Kenta Nakano
- Department of Laboratory Animal Medicine, Research Institute, National Center for Global Health and Medicine (NCGM), Tokyo, Japan.,Laboratory of Laboratory Animal Science and Medicine, School of Veterinary Medicine, Kitasato University, Towada, Japan
| | - Tadashi Okamura
- Department of Laboratory Animal Medicine, Research Institute, National Center for Global Health and Medicine (NCGM), Tokyo, Japan.,Section of Animal Models, Department of Infectious Diseases, Research Institute, National Center for Global Health and Medicine (NCGM), Tokyo, Japan
| | - Kimie Niimi
- Support Unit for Animal Resources Development, Research Resources Division, RIKEN Center for Brain Science, Saitama, Japan
| | - Eiki Takahashi
- Support Unit for Animal Resources Development, Research Resources Division, RIKEN Center for Brain Science, Saitama, Japan
| | - Long Guo
- Laboratory for Bone and Joint Diseases, RIKEN Center for Integrative Medical Sciences, Tokyo, Japan
| | - Shiro Ikegawa
- Laboratory for Bone and Joint Diseases, RIKEN Center for Integrative Medical Sciences, Tokyo, Japan
| | - Tatsuya Furuichi
- Laboratory of Laboratory Animal Science and Medicine, Co-Department of Veterinary Medicine, Faculty of Agriculture, Iwate University, Morioka, Japan.,Department of Basic Veterinary Science, United Graduate School of Veterinary Science, Gifu University, Japan
| |
Collapse
|
17
|
Bachvarova V, Dierker T, Esko J, Hoffmann D, Kjellen L, Vortkamp A. Chondrocytes respond to an altered heparan sulfate composition with distinct changes of heparan sulfate structure and increased levels of chondroitin sulfate. Matrix Biol 2020; 93:43-59. [PMID: 32201365 DOI: 10.1016/j.matbio.2020.03.006] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2019] [Revised: 03/11/2020] [Accepted: 03/12/2020] [Indexed: 01/27/2023]
Abstract
Heparan sulfate (HS) regulates the activity of many signaling molecules critical for the development of endochondral bones. Even so, mice with a genetically altered HS metabolism display a relatively mild skeletal phenotype compared to the defects observed in other tissues and organs pointing to a reduced HS dependency of growth-factor signaling in chondrocytes. To understand this difference, we have investigated the glycosaminoglycan (GAG) composition in two mouse lines that produce either reduced levels of HS (Ext1gt/gt mice) or HS lacking 2-O-sulfation (Hs2st1-/- mice). Analysis by RPIP-HPLC revealed an increased level of sulfated disaccarides not affected by the mutation in both mouse lines indicating that chondrocytes attempt to restore a critical level of sulfation. In addition, in both mutant lines we also detected significantly elevated levels of CS. Size exclusion chromatography further demonstrated that Ext1gt/gt mutants produce more but shorter CS chains, while the CS chains produced by (Hs2st1-/- mice) mutants are of similar length to that of wild type littermates indicating that chondrocytes produce more rather than longer CS chains. Expression analysis revealed an upregulation of aggrecan, which likely carries most of the additionally produced CS. Together the results of this study demonstrate for the first time that not only a reduced HS synthesis but also an altered HS structure leads to increased levels of CS in mammalian tissues. Furthermore, as chondrocytes produce 100-fold more CS than HS the increased CS levels point to an active, precursor-independent mechanism that senses the quality of HS in a vast excess of CS. Interestingly, reducing the level of cell surface CS by chondroitinase treatment leads to reduced Bmp2 induced Smad1/5/9 phosphorylation. In addition, Erk phosphorylation is increased independent of Fgf18 treatment indicating that both, HS and CS, affect growth factor signaling in chondrocytes in distinct manners.
Collapse
Affiliation(s)
- Velina Bachvarova
- Department of Developmental Biology, Faculty of Biology and Centre for Medical Biotechnology, University of Duisburg-Essen, Universitätsstr 1-5,45117 Essen, Germany.
| | - Tabea Dierker
- Department of Medical Biochemistry and Microbiology, and Science for Life Laboratory, Uppsala University, Box 582, Uppsala, Sweden.
| | - Jeffrey Esko
- Department of Cellular and Molecular Medicine, UCSD, United States.
| | - Daniel Hoffmann
- Department of Bioinformatics and Computational Biophysics, Faculty of Biology and Centre for Medical Biotechnology, University of Duisburg-Essen, Germany.
| | - Lena Kjellen
- Department of Medical Biochemistry and Microbiology, and Science for Life Laboratory, Uppsala University, Box 582, Uppsala, Sweden.
| | - Andrea Vortkamp
- Department of Developmental Biology, Faculty of Biology and Centre for Medical Biotechnology, University of Duisburg-Essen, Universitätsstr 1-5,45117 Essen, Germany.
| |
Collapse
|
18
|
Wang X, Cornelis FMF, Lories RJ, Monteagudo S. Exostosin-1 enhances canonical Wnt signaling activity during chondrogenic differentiation. Osteoarthritis Cartilage 2019; 27:1702-1710. [PMID: 31330188 DOI: 10.1016/j.joca.2019.07.007] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/11/2019] [Revised: 07/07/2019] [Accepted: 07/09/2019] [Indexed: 02/02/2023]
Abstract
OBJECTIVE Exostosin-1 (Ext1) encodes a glycosyltransferase required for heparan sulfate (HS) chain elongation in HS-proteoglycan biosynthesis. HS chains serve as binding partners for signaling proteins, affecting their distribution and activity. The Wnt/β-catenin pathway emerged as critical regulator of chondrogenesis. Yet, how EXT1 and HS affect Wnt/β-catenin signaling during chondrogenesis remains unexplored. METHOD Ext1 was stably knocked-down or overexpressed in ATDC5 chondrogenic cells cultured as micromasses. HS content was determined using ELISA. Chondrogenic markers Sox9, Col2a1, Aggrecan, and Wnt direct target gene Axin2 were measured by RT-qPCR. Proteoglycan content was evaluated by Alcian blue and DMMB assay, canonical Wnt signaling activation by β-catenin Western blot and TOP/FOP assay. ATDC5 cells and human articular chondrocytes were treated with Wnt activators CHIR99021 and recombinant WNT3A. RESULTS Ext1 knock-down reduced HS, and increased chondrogenic markers and proteoglycan accumulation. Ext1 knock-down reduced active Wnt/β-catenin signaling. Conversely, Ext1 overexpressing cells, with higher HS content, showed decreased chondrogenic differentiation and enhanced Wnt/β-catenin signaling. Wnt/β-catenin signaling activation led to a down-regulation of Ext1 expression in ATDC5 cells and in human articular chondrocytes. CONCLUSIONS EXT1 affects chondrogenic differentiation of precursor cells, in part via changes in the activity of Wnt/β-catenin signaling. Wnt/β-catenin signaling controls Ext1 expression, suggesting a regulatory loop between EXT1 and Wnt/β-catenin signaling during chondrogenesis.
Collapse
Affiliation(s)
- X Wang
- Laboratory of Tissue Homeostasis and Disease, Skeletal Biology and Engineering Research Center, KU Leuven, Leuven, Belgium.
| | - F M F Cornelis
- Laboratory of Tissue Homeostasis and Disease, Skeletal Biology and Engineering Research Center, KU Leuven, Leuven, Belgium.
| | - R J Lories
- Laboratory of Tissue Homeostasis and Disease, Skeletal Biology and Engineering Research Center, KU Leuven, Leuven, Belgium; Division of Rheumatology, University Hospitals Leuven, Leuven, Belgium.
| | - S Monteagudo
- Laboratory of Tissue Homeostasis and Disease, Skeletal Biology and Engineering Research Center, KU Leuven, Leuven, Belgium.
| |
Collapse
|
19
|
Yang C, Zhang R, Lin H, Wang H. Insights into the molecular regulatory network of pathomechanisms in osteochondroma. J Cell Biochem 2019; 120:16362-16369. [PMID: 31211456 DOI: 10.1002/jcb.29155] [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: 03/25/2019] [Revised: 05/17/2019] [Accepted: 05/22/2019] [Indexed: 12/21/2022]
Abstract
Osteochondroma is a benign autosomal dominant hereditary disease characterized by abnormal proliferation of cartilage in the long bone. It is divided into solitary osteochondroma and hereditary multiple exostoses (HMEs). The exostosin-1 (EXT-1) and exostosin-2 (EXT-2) gene mutations are well-defined molecular mechanisms in the pathogenesis of HME. EXT-1 and EXT-2 encode glycosyltransferases that are necessary for the synthesis of heparin sulfate. Accumulating evidence suggests that mutations in the EXT family induce changes in isolated hypogonadotropic hypogonadism-parathyroid hormone-related protein, bone morphogenetic protein, and fibroblast growth factor signaling pathways. Studies have also found that a large number of microRNAs (miRNAs) are abnormally expressed in osteochondroma tissues, and some of them also participate in several major signaling pathways. The regulation of miRNA expression could be another breakthrough in the treatment of osteochondroma. Although the pathogenesis of osteochondroma is very complicated, significant progress has been made in recent years. It is hoped that the pathogenesis of osteochondroma will be clearly understood and the most effective methods for the prevention and treatment of osteochondroma will be determined. This review provides an update on the recent progress in the interpretation of the underlying molecular mechanisms of osteochondroma.
Collapse
Affiliation(s)
- Congyi Yang
- School of Basic Medical Sciences, Nanchang University, Nanchang, Jiangxi, China.,Queen Mary School, Nanchang University, Nanchang, Jiangxi, China
| | - Ruiqian Zhang
- School of Basic Medical Sciences, Nanchang University, Nanchang, Jiangxi, China.,Queen Mary School, Nanchang University, Nanchang, Jiangxi, China
| | - Hui Lin
- School of Basic Medical Sciences, Nanchang University, Nanchang, Jiangxi, China
| | - Hongmei Wang
- School of Basic Medical Sciences, Nanchang University, Nanchang, Jiangxi, China
| |
Collapse
|
20
|
Levings DC, Nakato H. Loss of heparan sulfate in the niche leads to tumor-like germ cell growth in the Drosophila testis. Glycobiology 2018; 28:32-41. [PMID: 29069438 PMCID: PMC5993100 DOI: 10.1093/glycob/cwx090] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2017] [Revised: 10/10/2017] [Accepted: 10/17/2017] [Indexed: 12/15/2022] Open
Abstract
The stem cell niche normally prevents aberrant stem cell behaviors that lead to cancer formation. Recent studies suggest that some cancers are derived from endogenous populations of adult stem cells that have somehow escaped from normal control by the niche. However, the molecular mechanisms by which the niche retains stem cells locally and tightly controls their divisions are poorly understood. Here, we demonstrate that the presence of heparan sulfate (HS), a class glygosaminoglycan chains, in the Drosophila germline stem cell niche prevents tumor formation in the testis. Loss of HS in the niche, called the hub, led to gross changes in the morphology of testes as well as the formation of both somatic and germline tumors. This loss of hub HS resulted in ectopic signaling events in the Jak/Stat pathway outside the niche. This ectopic Jak/Stat signaling disrupted normal somatic cell differentiation, leading to the formation of tumors. Our finding indicates a novel non-autonomous role for niche HS in ensuring the integrity of the niche and preventing tumor formation.
Collapse
Affiliation(s)
- Daniel C Levings
- Department of Genetics, Cell Biology and Development, The University of Minnesota, 6-160 Jackson Hall, 321 Church St SE, Minneapolis, MN 55455, USA
| | - Hiroshi Nakato
- Department of Genetics, Cell Biology and Development, The University of Minnesota, 6-160 Jackson Hall, 321 Church St SE, Minneapolis, MN 55455, USA
| |
Collapse
|
21
|
Monteiro AF, Saraiva Gil F, Rato M, Osório T, Tavares E. An extremely rare association of multiple familial trichoepitheliomas and hereditary multiple osteochondromas. Int J Dermatol 2018; 57:e157-e159. [PMID: 30246361 DOI: 10.1111/ijd.14251] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/23/2018] [Revised: 07/11/2018] [Accepted: 08/31/2018] [Indexed: 11/30/2022]
Affiliation(s)
- Ana F Monteiro
- Dermatovenereology Department, Hospital de Santarém EPE, Santarém, Portugal
| | | | - Margarida Rato
- Dermatovenereology Department, Hospital de Santarém EPE, Santarém, Portugal
| | - Tomás Osório
- Department of Orthopedic Surgery, Unidade Local de Saúde do Baixo Alentejo, EPE - Hospital José Joaquim Fernandes, Beja, Portugal
| | - Ermelindo Tavares
- Dermatovenereology Department, Hospital de Vila Franca de Xira, Vila Franca de Xira, Portugal
| |
Collapse
|
22
|
Piombo V, Jochmann K, Hoffmann D, Wuelling M, Vortkamp A. Signaling systems affecting the severity of multiple osteochondromas. Bone 2018; 111:71-81. [PMID: 29545125 DOI: 10.1016/j.bone.2018.03.010] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/14/2017] [Revised: 02/12/2018] [Accepted: 03/09/2018] [Indexed: 01/01/2023]
Abstract
Multiple osteochondromas (MO) syndrome is a dominant autosomal bone disorder characterized by the formation of cartilage-capped bony outgrowths that develop at the juxtaposition of the growth plate of endochondral bones. MO has been linked to mutations in either EXT1 or EXT2, two glycosyltransferases required for the synthesis of heparan sulfate (HS). The establishment of mouse mutants demonstrated that a clonal, homozygous loss of Ext1 in a wild type background leads to the development of osteochondromas. Here we investigate mechanisms that might contribute to the variation in the severity of the disease observed in human patients. Our results show that residual amounts of HS are sufficient to prevent the development of osteochondromas strongly supporting that loss of heterozygosity is required for osteochondroma formation. Furthermore, we demonstrate that different signaling pathways affect size and frequency of the osteochondromas thereby modulating the severity of the disease. Reduced Fgfr3 signaling, which regulates proliferation and differentiation of chondrocytes, increases osteochondroma number, while activated Fgfr3 signaling reduces osteochondroma size. Both, activation and reduction of Wnt/β-catenin signaling decrease osteochondroma size and frequency by interfering with the chondrogenic fate of the mutant cells. Reduced Ihh signaling does not change the development of the osteochondromas, while elevated Ihh signaling increases the cellularity and inhibits chondrocyte differentiation in a subset of osteochondromas and might thus predispose osteochondromas to the transformation into chondrosarcomas.
Collapse
Affiliation(s)
- Virginia Piombo
- Department of Developmental Biology, Centre of Medical Biotechnology, Faculty of Biology, University of Duisburg-Essen, Essen, Germany
| | - Katja Jochmann
- Department of Developmental Biology, Centre of Medical Biotechnology, Faculty of Biology, University of Duisburg-Essen, Essen, Germany
| | - Daniel Hoffmann
- Research Group Bioinformatics, Centre of Medical Biotechnology, Faculty of Biology, University of Duisburg-Essen, Essen, Germany
| | - Manuela Wuelling
- Department of Developmental Biology, Centre of Medical Biotechnology, Faculty of Biology, University of Duisburg-Essen, Essen, Germany
| | - Andrea Vortkamp
- Department of Developmental Biology, Centre of Medical Biotechnology, Faculty of Biology, University of Duisburg-Essen, Essen, Germany.
| |
Collapse
|
23
|
Inubushi T, Lemire I, Irie F, Yamaguchi Y. Palovarotene Inhibits Osteochondroma Formation in a Mouse Model of Multiple Hereditary Exostoses. J Bone Miner Res 2018; 33:658-666. [PMID: 29120519 PMCID: PMC5895492 DOI: 10.1002/jbmr.3341] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/08/2017] [Revised: 11/06/2017] [Accepted: 11/08/2017] [Indexed: 12/12/2022]
Abstract
Multiple hereditary exostoses (MHE), also known as multiple osteochondromas (MO), is an autosomal dominant disorder characterized by the development of multiple cartilage-capped bone tumors (osteochondromas). The large majority of patients with MHE carry loss-of-function mutations in the EXT1 or EXT2 gene, which encodes a glycosyltransferase essential for heparan sulfate (HS) biosynthesis. Increasing evidence suggests that enhanced bone morphogenetic protein (BMP) signaling resulting from loss of HS expression plays a role in osteochondroma formation in MHE. Palovarotene (PVO) is a retinoic acid receptor γ selective agonist, which is being investigated as a potential drug for fibrodysplasia ossificans progressiva (FOP), another genetic bone disorder with features that overlap with those of MHE. Here we show that PVO inhibits osteochondroma formation in the Fsp1Cre ;Ext1flox/flox model of MHE. Four-week daily treatment with PVO starting at postnatal day (P) 14 reduced the number of osteochondromas that develop in these mice by up to 91% in a dose-dependent manner. An inhibition of long bone growth observed in animals treated from P14 was almost entirely abrogated by delaying the initiation of treatment to P21. We also found that PVO attenuates BMP signaling in Fsp1Cre ;Ext1flox/flox mice and that aberrant chondrogenic fate determination of Ext1-deficient perichondrial progenitor cells in these mice is restored by PVO. Together, the present data support further preclinical and clinical investigations of PVO as a potential therapeutic agent for MHE. © 2017 American Society for Bone and Mineral Research.
Collapse
Affiliation(s)
- Toshihiro Inubushi
- Human Genetics Program, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA
| | | | - Fumitoshi Irie
- Human Genetics Program, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA
| | - Yu Yamaguchi
- Human Genetics Program, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA
| |
Collapse
|
24
|
Nozawa S, Inubushi T, Irie F, Takigami I, Matsumoto K, Shimizu K, Akiyama H, Yamaguchi Y. Osteoblastic heparan sulfate regulates osteoprotegerin function and bone mass. JCI Insight 2018; 3:89624. [PMID: 29415886 DOI: 10.1172/jci.insight.89624] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2016] [Accepted: 01/10/2018] [Indexed: 02/05/2023] Open
Abstract
Bone remodeling is a highly coordinated process involving bone formation and resorption, and imbalance of this process results in osteoporosis. It has long been recognized that long-term heparin therapy often causes osteoporosis, suggesting that heparan sulfate (HS), the physiological counterpart of heparin, is somehow involved in bone mass regulation. The role of endogenous HS in adult bone, however, remains unclear. To determine the role of HS in bone homeostasis, we conditionally ablated Ext1, which encodes an essential glycosyltransferase for HS biosynthesis, in osteoblasts. Resultant conditional mutant mice developed severe osteopenia. Surprisingly, this phenotype is not due to impairment in bone formation but to enhancement of bone resorption. We show that osteoprotegerin (OPG), which is known as a soluble decoy receptor for RANKL, needs to be associated with the osteoblast surface in order to efficiently inhibit RANKL/RANK signaling and that HS serves as a cell surface binding partner for OPG in this context. We also show that bone mineral density is reduced in patients with multiple hereditary exostoses, a genetic bone disorder caused by heterozygous mutations of Ext1, suggesting that the mechanism revealed in this study may be relevant to low bone mass conditions in humans.
Collapse
Affiliation(s)
- Satoshi Nozawa
- Human Genetics Program, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, California, USA.,Department of Orthopedic Surgery, Gifu University, Gifu, Japan
| | - Toshihiro Inubushi
- Human Genetics Program, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, California, USA
| | - Fumitoshi Irie
- Human Genetics Program, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, California, USA
| | - Iori Takigami
- Department of Orthopedic Surgery, Gifu University, Gifu, Japan
| | - Kazu Matsumoto
- Department of Orthopedic Surgery, Gifu University, Gifu, Japan
| | - Katsuji Shimizu
- Department of Orthopedic Surgery, Gifu University, Gifu, Japan
| | | | - Yu Yamaguchi
- Human Genetics Program, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, California, USA
| |
Collapse
|
25
|
Shimbo M, Suzuki R, Fuseya S, Sato T, Kiyohara K, Hagiwara K, Okada R, Wakui H, Tsunakawa Y, Watanabe H, Kimata K, Narimatsu H, Kudo T, Takahashi S. Postnatal lethality and chondrodysplasia in mice lacking both chondroitin sulfate N-acetylgalactosaminyltransferase-1 and -2. PLoS One 2017; 12:e0190333. [PMID: 29287114 PMCID: PMC5747463 DOI: 10.1371/journal.pone.0190333] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2017] [Accepted: 12/12/2017] [Indexed: 02/04/2023] Open
Abstract
Chondroitin sulfate (CS) is a sulfated glycosaminoglycan (GAG) chain. In cartilage, CS plays important roles as the main component of the extracellular matrix (ECM), existing as side chains of the major cartilage proteoglycan, aggrecan. Six glycosyltransferases are known to coordinately synthesize the backbone structure of CS; however, their in vivo synthetic mechanism remains unknown. Previous studies have suggested that two glycosyltransferases, Csgalnact1 (t1) and Csgalnact2 (t2), are critical for initiation of CS synthesis in vitro. Indeed, t1 single knockout mice (t1 KO) exhibit slight dwarfism and a reduction in CS content in cartilage compared with wild-type (WT) mice. To reveal the synergetic roles of t1 and t2 in CS synthesis in vivo, we generated systemic single and double knockout (DKO) mice and cartilage-specific t1 and t2 double knockout (Col2-DKO) mice. DKO mice exhibited postnatal lethality, whereas t2 KO mice showed normal size and skeletal development. Col2-DKO mice survived to adulthood and showed severe dwarfism compared with t1 KO mice. Histological analysis of epiphyseal cartilage from Col2-DKO mice revealed disrupted endochondral ossification, characterized by drastic GAG reduction in the ECM. Moreover, DKO cartilage had reduced chondrocyte proliferation and an increased number of apoptotic chondrocytes compared with WT cartilage. Conversely, primary chondrocyte cultures from Col2-DKO knee cartilage had the same proliferation rate as WT chondrocytes and low GAG expression levels, indicating that the chondrocytes themselves had an intact proliferative ability. Quantitative RT-PCR analysis of E18.5 cartilage showed that the expression levels of Col2a1 and Ptch1 transcripts tended to decrease in DKO compared with those in WT mice. The CS content in DKO cartilage was decreased compared with that in t1 KO cartilage but was not completely absent. These results suggest that aberrant ECM caused by CS reduction disrupted endochondral ossification. Overall, we propose that both t1 and t2 are necessary for CS synthesis and normal chondrocyte differentiation but are not sufficient for all CS synthesis in cartilage.
Collapse
Affiliation(s)
- Miki Shimbo
- Department of Anatomy and Embryology, Faculty of Medicine, University of Tsukuba, Tsukuba, Ibaraki, Japan
- Doctoral Program in Biomedical Sciences, Graduate School of Comprehensive Human Sciences, University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Riku Suzuki
- Department of Anatomy and Embryology, Faculty of Medicine, University of Tsukuba, Tsukuba, Ibaraki, Japan
- Ph.D. Program in Human Biology, School of Integrative and Global Majors, University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Sayaka Fuseya
- Department of Anatomy and Embryology, Faculty of Medicine, University of Tsukuba, Tsukuba, Ibaraki, Japan
- Master’s Program in Medical Sciences, Graduate School of Comprehensive Human Sciences, University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Takashi Sato
- Glycoscience and Glycotechnology Research Group, Biotechnology Research Institute for Drug Discovery, National Institute of Advanced Industrial Science and Technology (AIST), Tsukuba, Ibaraki, Japan
| | - Katsue Kiyohara
- Glycoscience and Glycotechnology Research Group, Biotechnology Research Institute for Drug Discovery, National Institute of Advanced Industrial Science and Technology (AIST), Tsukuba, Ibaraki, Japan
| | - Kozue Hagiwara
- Glycoscience and Glycotechnology Research Group, Biotechnology Research Institute for Drug Discovery, National Institute of Advanced Industrial Science and Technology (AIST), Tsukuba, Ibaraki, Japan
| | - Risa Okada
- Department of Anatomy and Embryology, Faculty of Medicine, University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Hiromasa Wakui
- Department of Anatomy and Embryology, Faculty of Medicine, University of Tsukuba, Tsukuba, Ibaraki, Japan
- Master’s Program in Medical Sciences, Graduate School of Comprehensive Human Sciences, University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Yuki Tsunakawa
- Department of Anatomy and Embryology, Faculty of Medicine, University of Tsukuba, Tsukuba, Ibaraki, Japan
- Ph.D. Program in Human Biology, School of Integrative and Global Majors, University of Tsukuba, Tsukuba, Ibaraki, Japan
| | | | - Koji Kimata
- Multidisciplinary Pain Center, Aichi Medical University, Aichi, Japan
| | - Hisashi Narimatsu
- Glycoscience and Glycotechnology Research Group, Biotechnology Research Institute for Drug Discovery, National Institute of Advanced Industrial Science and Technology (AIST), Tsukuba, Ibaraki, Japan
| | - Takashi Kudo
- Department of Anatomy and Embryology, Faculty of Medicine, University of Tsukuba, Tsukuba, Ibaraki, Japan
- Laboratory Animal Resource Center (LARC), University of Tsukuba, Tsukuba, Ibaraki, Japan
- * E-mail: (TK); (ST)
| | - Satoru Takahashi
- Department of Anatomy and Embryology, Faculty of Medicine, University of Tsukuba, Tsukuba, Ibaraki, Japan
- Laboratory Animal Resource Center (LARC), University of Tsukuba, Tsukuba, Ibaraki, Japan
- Transborder Medical Research Center, Faculty of Medicine, University of Tsukuba, Tsukuba, Ibaraki, Japan
- * E-mail: (TK); (ST)
| |
Collapse
|
26
|
Heparin-fibronectin interactions in the development of extracellular matrix insolubility. Matrix Biol 2017; 67:107-122. [PMID: 29223498 DOI: 10.1016/j.matbio.2017.11.012] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2017] [Revised: 11/29/2017] [Accepted: 11/30/2017] [Indexed: 12/15/2022]
Abstract
During extracellular matrix (ECM) assembly, fibronectin (FN) fibrils are irreversibly converted into a detergent-insoluble form which, through FN's multi-domain structure, can interact with collagens, matricellular proteins, and growth factors to build a definitive matrix. FN also has heparin/heparan sulfate (HS) binding sites. Using HS-deficient CHO cells, we show that the addition of soluble heparin significantly increased the amount of FN matrix that these cells assemble. Sulfated HS glycosaminoglycan (GAG) mimetics similarly increased FN assembly and demonstrated a dependence on GAG sulfation. The length of the heparin chains also plays a role in assembly. Chains of sufficient length to bind to two FN molecules gave maximal stimulation of assembly whereas shorter heparin had less of an effect. Using a decellularized fibroblast matrix for proteolysis, detergent fractionation, and mass spectrometry, we found that the predominant domain within insoluble fibril fragments is FN's major heparin-binding domain HepII (modules III12-14). Multiple HepII domains bind simultaneously to a single heparin chain in size exclusion chromatography analyses. We propose a model in which heparin/HS binding to the HepII domain connects multiple FNs together to facilitate the formation of protein interactions for insoluble fibril assembly.
Collapse
|
27
|
Hu Z, Wang H, Wang Y, Zhou H, Shi F, Zhao J, Zhang S, Cao X. Genome‑wide analysis and prediction of functional long noncoding RNAs in osteoblast differentiation under simulated microgravity. Mol Med Rep 2017; 16:8180-8188. [PMID: 28990099 PMCID: PMC5779904 DOI: 10.3892/mmr.2017.7671] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2017] [Accepted: 08/17/2017] [Indexed: 01/12/2023] Open
Abstract
Long noncoding RNAs (lncRNAs) have been regarded as important regulators in numerous biological processes during cell development. However, the holistic lncRNA expression pattern and potential functions during osteoblast differentiation under simulated microgravity remain unknown. In the present study, a high throughput microarray assay was performed to detect lncRNA and mRNA expression profiles during MC3TC-E1 pre-osteoblast cell osteo-differentiation under simulated microgravity. The expression of 857 lncRNAs and 2,264 mRNAs was significantly altered when MC3T3-E1 cells were exposed to simulated microgravity. A relatively consistent distribution pattern on the chromosome and a co-expression network were observed between the differentially-expressed lncRNAs and mRNAs. Genomic context analysis further identified 132 differentially-expressed lncRNAs and nearby coding gene pairs. Subsequently, 3 lncRNAs were screened out for their possible function in osteoblast differentiation, based on their co-expression association and potential cis-acting regulatory pattern with the deregulated mRNAs. The present study aimed to provide a comprehensive understanding of and a foundation for future studies into lncRNA function in mechanical signal-mediated osteoblast differentiation.
Collapse
Affiliation(s)
- Zebing Hu
- The Key Laboratory of Aerospace Medicine, Ministry of Education, The Fourth Military Medical University, Xi'an, Shaanxi 710032, P.R. China
| | - Han Wang
- The Key Laboratory of Aerospace Medicine, Ministry of Education, The Fourth Military Medical University, Xi'an, Shaanxi 710032, P.R. China
| | - Yixuan Wang
- The Key Laboratory of Aerospace Medicine, Ministry of Education, The Fourth Military Medical University, Xi'an, Shaanxi 710032, P.R. China
| | - Hua Zhou
- The Key Laboratory of Aerospace Medicine, Ministry of Education, The Fourth Military Medical University, Xi'an, Shaanxi 710032, P.R. China
| | - Fei Shi
- The Key Laboratory of Aerospace Medicine, Ministry of Education, The Fourth Military Medical University, Xi'an, Shaanxi 710032, P.R. China
| | - Jiangdong Zhao
- The Key Laboratory of Aerospace Medicine, Ministry of Education, The Fourth Military Medical University, Xi'an, Shaanxi 710032, P.R. China
| | - Shu Zhang
- The Key Laboratory of Aerospace Medicine, Ministry of Education, The Fourth Military Medical University, Xi'an, Shaanxi 710032, P.R. China
| | - Xinsheng Cao
- The Key Laboratory of Aerospace Medicine, Ministry of Education, The Fourth Military Medical University, Xi'an, Shaanxi 710032, P.R. China
| |
Collapse
|
28
|
Inubushi T, Nozawa S, Matsumoto K, Irie F, Yamaguchi Y. Aberrant perichondrial BMP signaling mediates multiple osteochondromagenesis in mice. JCI Insight 2017; 2:90049. [PMID: 28768899 DOI: 10.1172/jci.insight.90049] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2016] [Accepted: 06/23/2017] [Indexed: 01/20/2023] Open
Abstract
Multiple hereditary exostoses (MHE) is characterized by the development of numerous benign bony tumors (osteochondromas). Although it has been well established that MHE is caused by mutations in EXT1 and EXT2, which encode glycosyltransferase essential for heparan sulfate (HS) biosynthesis, the cellular origin and molecular mechanisms of MHE remain elusive. Here, we show that in Ext1 mutant mice, osteochondromas develop from mesenchymal stem cell-like progenitor cells residing in the perichondrium, and we show that enhanced BMP signaling in these cells is the primary signaling defect that leads to osteochondromagenesis. We demonstrate that progenitor cells in the perichondrium, including those in the groove of Ranvier, highly express HS and that Ext1 ablation targeted to the perichondrium results in the development of osteochondromas. Ext1-deficient perichondrial progenitor cells show enhanced BMP signaling and increased chondrogenic differentiation both in vitro and in vivo. Consistent with the functional role for enhanced BMP signaling in osteochondromagenesis, administration of the small molecule BMP inhibitor LDN-193189 suppresses osteochondroma formation in two MHE mouse models. Together, our results demonstrate a role for enhanced perichondrial BMP signaling in osteochondromagenesis in mice, and they suggest the possibility of pharmacological treatment of MHE with BMP inhibitors.
Collapse
|
29
|
You WK, Stallcup WB. Localization of VEGF to Vascular ECM Is an Important Aspect of Tumor Angiogenesis. Cancers (Basel) 2017; 9:cancers9080097. [PMID: 28788063 PMCID: PMC5575600 DOI: 10.3390/cancers9080097] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2017] [Revised: 07/22/2017] [Accepted: 07/26/2017] [Indexed: 12/02/2022] Open
Abstract
Our research has identified several examples in which reduced VEGF-A binding to deficient vascular extracellular matrix leads to deficits in tumor vascularization and tumor growth: (1) germline ablation of collagen VI in the stroma of intracranial B16F10 melanomas; (2) knockdown of the Tks5 scaffolding protein in MDA-MB-231 mammary tumor cells; (3) germline ablation of NG2 proteoglycan in the stroma of MMTV-PyMT mammary tumors; and (4) myeloid-specific ablation of NG2 in the stroma of intracranial B16F10 melanomas. Tumor hypoxia is increased in each of the four types of experimental mice, accompanied by increases in total VEGF-A. However, while VEGF-A is highly associated with tumor blood vessels in control mice, it is much more diffusely distributed in tumors in all four sets of experimental mice, likely due to reduced extent of the vascular extracellular matrix. In parallel to lost VEGF-A localization, tumor vessels in each case have smaller diameters and are leakier than tumor vessels in control mice. Tumor growth is decreased as a result of this poor vascular function. The fact that the observed vascular changes occur in the absence of alterations in vascular density suggests that examination of vessel structure and function is more useful than vascular density for understanding the importance of angiogenesis in tumor progression.
Collapse
Affiliation(s)
| | - William B Stallcup
- Tumor Microenvironment and Cancer Immunology Program, Cancer Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA 92037, USA.
| |
Collapse
|
30
|
Ayerst BI, Merry CLR, Day AJ. The Good the Bad and the Ugly of Glycosaminoglycans in Tissue Engineering Applications. Pharmaceuticals (Basel) 2017; 10:E54. [PMID: 28608822 PMCID: PMC5490411 DOI: 10.3390/ph10020054] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2017] [Revised: 06/05/2017] [Accepted: 06/05/2017] [Indexed: 12/14/2022] Open
Abstract
High sulfation, low cost, and the status of heparin as an already FDA- and EMA- approved product, mean that its inclusion in tissue engineering (TE) strategies is becoming increasingly popular. However, the use of heparin may represent a naïve approach. This is because tissue formation is a highly orchestrated process, involving the temporal expression of numerous growth factors and complex signaling networks. While heparin may enhance the retention and activity of certain growth factors under particular conditions, its binding 'promiscuity' means that it may also inhibit other factors that, for example, play an important role in tissue maintenance and repair. Within this review we focus on articular cartilage, highlighting the complexities and highly regulated processes that are involved in its formation, and the challenges that exist in trying to effectively engineer this tissue. Here we discuss the opportunities that glycosaminoglycans (GAGs) may provide in advancing this important area of regenerative medicine, placing emphasis on the need to move away from the common use of heparin, and instead focus research towards the utility of specific GAG preparations that are able to modulate the activity of growth factors in a more controlled and defined manner, with less off-target effects.
Collapse
Affiliation(s)
- Bethanie I Ayerst
- Wellcome Trust Centre for Cell-Matrix Research, Division of Cell-Matrix Biology & Regenerative Medicine, School of Biology, Faculty of Biology, Medicine & Health, The University of Manchester, Manchester Academic Health Science Centre, Manchester M13 9PL, UK.
| | - Catherine L R Merry
- Stem Cell Glycobiology Group, Wolfson Centre for Stem Cells, Tissue Engineering & Modelling (STEM), Centre for Biomolecular Sciences, University of Nottingham, University Park, Nottingham NG7 2RD, UK.
| | - Anthony J Day
- Wellcome Trust Centre for Cell-Matrix Research, Division of Cell-Matrix Biology & Regenerative Medicine, School of Biology, Faculty of Biology, Medicine & Health, The University of Manchester, Manchester Academic Health Science Centre, Manchester M13 9PL, UK.
| |
Collapse
|
31
|
Pacifici M. Hereditary Multiple Exostoses: New Insights into Pathogenesis, Clinical Complications, and Potential Treatments. Curr Osteoporos Rep 2017; 15:142-152. [PMID: 28466453 PMCID: PMC5510481 DOI: 10.1007/s11914-017-0355-2] [Citation(s) in RCA: 71] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
PURPOSE OF REVIEW Hereditary multiple exostoses (HME) is a complex musculoskeletal pediatric disorder characterized by osteochondromas that form next to the growth plates of many skeletal elements, including long bones, ribs, and vertebrae. Due to its intricacies and unresolved issues, HME continues to pose major challenges to both clinicians and biomedical researchers. The purpose of this review is to describe and analyze recent advances in this field and point to possible targets and strategies for future biologically based therapeutic intervention. RECENT FINDINGS Most HME cases are linked to loss-of-function mutations in EXT1 or EXT2 that encode glycosyltransferases responsible for heparan sulfate (HS) synthesis, leading to HS deficiency. Recent genomic inquiries have extended those findings but have yet to provide a definitive genotype-phenotype correlation. Clinical studies emphasize that in addition to the well-known skeletal problems caused by osteochondromas, HME patients can experience, and suffer from, other symptoms and health complications such as chronic pain and nerve impingement. Laboratory work has produced novel insights into alterations in cellular and molecular mechanisms instigated by HS deficiency and subtending onset and growth of osteochondroma and how such changes could be targeted toward therapeutic ends. HME is a rare and orphan disease and, as such, is being studied only by a handful of clinical and basic investigators. Despite this limitation, significant advances have been made in the last few years, and the future bodes well for deciphering more thoroughly its pathogenesis and, in turn, identifying the most effective treatment for osteochondroma prevention.
Collapse
Affiliation(s)
- Maurizio Pacifici
- Translational Research Program in Pediatric Orthopaedics, Abramson Research Center, 902D, Division of Orthopaedic Surgery, Department of Surgery, The Children's Hospital of Philadelphia, Philadelphia, PA, 19104, USA.
| |
Collapse
|
32
|
Sinha S, Mundy C, Bechtold T, Sgariglia F, Ibrahim MM, Billings PC, Carroll K, Koyama E, Jones KB, Pacifici M. Unsuspected osteochondroma-like outgrowths in the cranial base of Hereditary Multiple Exostoses patients and modeling and treatment with a BMP antagonist in mice. PLoS Genet 2017; 13:e1006742. [PMID: 28445472 PMCID: PMC5425227 DOI: 10.1371/journal.pgen.1006742] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2016] [Revised: 05/10/2017] [Accepted: 04/05/2017] [Indexed: 11/18/2022] Open
Abstract
Hereditary Multiple Exostoses (HME) is a rare pediatric disorder caused by loss-of-function mutations in the genes encoding the heparan sulfate (HS)-synthesizing enzymes EXT1 or EXT2. HME is characterized by formation of cartilaginous outgrowths-called osteochondromas- next to the growth plates of many axial and appendicular skeletal elements. Surprisingly, it is not known whether such tumors also form in endochondral elements of the craniofacial skeleton. Here, we carried out a retrospective analysis of cervical spine MRI and CT scans from 50 consecutive HME patients that included cranial skeletal images. Interestingly, nearly half of the patients displayed moderate defects or osteochondroma-like outgrowths in the cranial base and specifically in the clivus. In good correlation, osteochondromas developed in the cranial base of mutant Ext1f/f;Col2-CreER or Ext1f/f;Aggrecan-CreER mouse models of HME along the synchondrosis growth plates. Osteochondroma formation was preceded by phenotypic alteration of cells at the chondro-perichondrial boundary and was accompanied by ectopic expression of major cartilage matrix genes -collagen 2 and collagen X- within the growing ectopic masses. Because chondrogenesis requires bone morphogenetic protein (BMP) signaling, we asked whether osteochondroma formation could be blocked by a BMP signaling antagonist. Systemic administration with LDN-193189 effectively inhibited osteochondroma growth in conditional Ext1-mutant mice. In vitro studies with mouse embryo chondrogenic cells clarified the mechanisms of LDN-193189 action that turned out to include decreases in canonical BMP signaling pSMAD1/5/8 effectors but interestingly, concurrent increases in such anti-chondrogenic mechanisms as pERK1/2 and Chordin, Fgf9 and Fgf18 expression. Our study is the first to reveal that the cranial base can be affected in patients with HME and that osteochondroma formation is amenable to therapeutic drug intervention.
Collapse
Affiliation(s)
- Sayantani Sinha
- Translational Research Program in Pediatric Orthopaedics, Division of Orthopaedic Surgery, The Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania, United States of America
| | - Christina Mundy
- Translational Research Program in Pediatric Orthopaedics, Division of Orthopaedic Surgery, The Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania, United States of America
| | - Till Bechtold
- Translational Research Program in Pediatric Orthopaedics, Division of Orthopaedic Surgery, The Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania, United States of America
| | - Federica Sgariglia
- Translational Research Program in Pediatric Orthopaedics, Division of Orthopaedic Surgery, The Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania, United States of America
| | - Mazen M. Ibrahim
- Translational Research Program in Pediatric Orthopaedics, Division of Orthopaedic Surgery, The Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania, United States of America
| | - Paul C. Billings
- Translational Research Program in Pediatric Orthopaedics, Division of Orthopaedic Surgery, The Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania, United States of America
| | - Kristen Carroll
- Shriner’s Hospital for Children, Salt Lake City, Utah, United States of America
- Department of Orthopaedics, University of Utah School of Medicine, Salt Lake City, Utah, United States of America
| | - Eiki Koyama
- Translational Research Program in Pediatric Orthopaedics, Division of Orthopaedic Surgery, The Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania, United States of America
| | - Kevin B. Jones
- Department of Orthopaedics, University of Utah School of Medicine, Salt Lake City, Utah, United States of America
- Department of Oncological Sciences and Huntsman Cancer Institute, University of Utah School of Medicine, Salt Lake City, Utah, United States of America
- * E-mail: (MP); (KBJ)
| | - Maurizio Pacifici
- Translational Research Program in Pediatric Orthopaedics, Division of Orthopaedic Surgery, The Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania, United States of America
- * E-mail: (MP); (KBJ)
| |
Collapse
|
33
|
Migliorini E, Horn P, Haraszti T, Wegner SV, Hiepen C, Knaus P, Richter RP, Cavalcanti-Adam EA. Enhanced Biological Activity of BMP-2 Bound to Surface-Grafted Heparan Sulfate. ACTA ACUST UNITED AC 2017; 1:e1600041. [DOI: 10.1002/adbi.201600041] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2016] [Indexed: 01/08/2023]
Affiliation(s)
- Elisa Migliorini
- Department of Biophysical Chemistry; Institute of Physical Chemistry; Heidelberg University; Im Neuenheimer Feld 253 69120 Heidelberg Germany
- Department of Cellular Biophysics; Max Planck Institute for Medical Research; Heisenbergstr. 3 D-70569 Stuttgart Germany
| | - Patrick Horn
- Department of Medicine V; Heidelberg University; INF 410 69120 Heidelberg Germany
| | - Tamás Haraszti
- DWI - Leibniz Institute for Interactive Materials; Forkenbeckstr. 50 52056 Aachen Germany
| | - Seraphine V. Wegner
- Department of Biophysical Chemistry; Institute of Physical Chemistry; Heidelberg University; Im Neuenheimer Feld 253 69120 Heidelberg Germany
- Department of Cellular Biophysics; Max Planck Institute for Medical Research; Heisenbergstr. 3 D-70569 Stuttgart Germany
- Max Planck Institute for Polymer Research; Ackermannweg 10 55128 Mainz Germany
| | - Christian Hiepen
- Institute of Biochemistry; Freie Universität Berlin; Thielallee 63 14195 Berlin Germany
| | - Petra Knaus
- Institute of Biochemistry; Freie Universität Berlin; Thielallee 63 14195 Berlin Germany
| | - Ralf P. Richter
- School of Biomedical Sciences and School of Physics and Astronomy; University of Leeds; Leeds LS2 9JT UK
- Biosurfaces Lab; CIC biomaGUNE; Paseo Miramon 182 20009 San Sebastian Spain
- Laboratory of Interdisciplinary Physics; University Grenoble-Alpes and CNRS; 140 Rue de la Physique 38402 St. Martin d'Hères France
| | - Elisabetta Ada Cavalcanti-Adam
- Department of Biophysical Chemistry; Institute of Physical Chemistry; Heidelberg University; Im Neuenheimer Feld 253 69120 Heidelberg Germany
- Department of Cellular Biophysics; Max Planck Institute for Medical Research; Heisenbergstr. 3 D-70569 Stuttgart Germany
| |
Collapse
|
34
|
Thomas JT, Eric Dollins D, Andrykovich KR, Chu T, Stultz BG, Hursh DA, Moos M. SMOC can act as both an antagonist and an expander of BMP signaling. eLife 2017; 6:e17935. [PMID: 28323621 PMCID: PMC5360445 DOI: 10.7554/elife.17935] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2016] [Accepted: 02/14/2017] [Indexed: 01/19/2023] Open
Abstract
The matricellular protein SMOC (Secreted Modular Calcium binding protein) is conserved phylogenetically from vertebrates to arthropods. We showed previously that SMOC inhibits bone morphogenetic protein (BMP) signaling downstream of its receptor via activation of mitogen-activated protein kinase (MAPK) signaling. In contrast, the most prominent effect of the Drosophila orthologue, pentagone (pent), is expanding the range of BMP signaling during wing patterning. Using SMOC deletion constructs we found that SMOC-∆EC, lacking the extracellular calcium binding (EC) domain, inhibited BMP2 signaling, whereas SMOC-EC (EC domain only) enhanced BMP2 signaling. The SMOC-EC domain bound HSPGs with a similar affinity to BMP2 and could expand the range of BMP signaling in an in vitro assay by competition for HSPG-binding. Together with data from studies in vivo we propose a model to explain how these two activities contribute to the function of Pent in Drosophila wing development and SMOC in mammalian joint formation.
Collapse
Affiliation(s)
- J Terrig Thomas
- Division of Cellular and Gene Therapies, Office of Tissues and Advanced Therapies, U.S. Food and Drug Administration, Silver Spring, United States
| | - D Eric Dollins
- Division of Cellular and Gene Therapies, Office of Tissues and Advanced Therapies, U.S. Food and Drug Administration, Silver Spring, United States
| | - Kristin R Andrykovich
- Division of Cellular and Gene Therapies, Office of Tissues and Advanced Therapies, U.S. Food and Drug Administration, Silver Spring, United States
| | - Tehyen Chu
- Division of Cellular and Gene Therapies, Office of Tissues and Advanced Therapies, U.S. Food and Drug Administration, Silver Spring, United States
| | - Brian G Stultz
- Division of Cellular and Gene Therapies, Office of Tissues and Advanced Therapies, U.S. Food and Drug Administration, Silver Spring, United States
| | - Deborah A Hursh
- Division of Cellular and Gene Therapies, Office of Tissues and Advanced Therapies, U.S. Food and Drug Administration, Silver Spring, United States
| | - Malcolm Moos
- Division of Cellular and Gene Therapies, Office of Tissues and Advanced Therapies, U.S. Food and Drug Administration, Silver Spring, United States
| |
Collapse
|
35
|
Volpi S, Yamazaki Y, Brauer PM, van Rooijen E, Hayashida A, Slavotinek A, Sun Kuehn H, Di Rocco M, Rivolta C, Bortolomai I, Du L, Felgentreff K, Ott de Bruin L, Hayashida K, Freedman G, Marcovecchio GE, Capuder K, Rath P, Luche N, Hagedorn EJ, Buoncompagni A, Royer-Bertrand B, Giliani S, Poliani PL, Imberti L, Dobbs K, Poulain FE, Martini A, Manis J, Linhardt RJ, Bosticardo M, Rosenzweig SD, Lee H, Puck JM, Zúñiga-Pflücker JC, Zon L, Park PW, Superti-Furga A, Notarangelo LD. EXTL3 mutations cause skeletal dysplasia, immune deficiency, and developmental delay. J Exp Med 2017; 214:623-637. [PMID: 28148688 PMCID: PMC5339678 DOI: 10.1084/jem.20161525] [Citation(s) in RCA: 68] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2016] [Revised: 12/10/2016] [Accepted: 01/10/2017] [Indexed: 12/05/2022] Open
Abstract
Volpi et al. demonstrate that hypomorphic EXTL3 mutations cause abnormalities of heparan sulfate composition, affect signaling in response to growth factors and cytokines, and perturb thymopoiesis, resulting in a novel genetic disease associating skeletal dysplasia, T cell immunodeficiency, and neurodevelopmental delay. We studied three patients with severe skeletal dysplasia, T cell immunodeficiency, and developmental delay. Whole-exome sequencing revealed homozygous missense mutations affecting exostosin-like 3 (EXTL3), a glycosyltransferase involved in heparan sulfate (HS) biosynthesis. Patient-derived fibroblasts showed abnormal HS composition and altered fibroblast growth factor 2 signaling, which was rescued by overexpression of wild-type EXTL3 cDNA. Interleukin-2–mediated STAT5 phosphorylation in patients’ lymphocytes was markedly reduced. Interbreeding of the extl3-mutant zebrafish (box) with Tg(rag2:green fluorescent protein) transgenic zebrafish revealed defective thymopoiesis, which was rescued by injection of wild-type human EXTL3 RNA. Targeted differentiation of patient-derived induced pluripotent stem cells showed a reduced expansion of lymphohematopoietic progenitor cells and defects of thymic epithelial progenitor cell differentiation. These data identify EXTL3 mutations as a novel cause of severe immune deficiency with skeletal dysplasia and developmental delay and underline a crucial role of HS in thymopoiesis and skeletal and brain development.
Collapse
Affiliation(s)
- Stefano Volpi
- Unita' Operativa Pediatria 2, Istituto Giannina Gaslini, 16148 Genoa, Italy
| | - Yasuhiro Yamazaki
- Laboratory of Host Defenses, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, 20892
| | - Patrick M Brauer
- Department of Immunology, Sunnybrook Research Institute, University of Toronto, Toronto, Ontario M5S, Canada
| | - Ellen van Rooijen
- Stem Cell Program, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115
| | - Atsuko Hayashida
- Division of Respiratory Diseases, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115
| | - Anne Slavotinek
- Department of Pediatrics, Division of Genetics, University of California, San Francisco, San Francisco, CA 94143
| | - Hye Sun Kuehn
- Department of Laboratory Medicine, National Institutes of Health Clinical Center, National Institutes of Health, Bethesda, MD, 20892
| | - Maja Di Rocco
- Unit of Rare Diseases, Department of Pediatrics, Istituto Giannina Gaslini, 16148 Genoa, Italy
| | - Carlo Rivolta
- Department of Computational Biology, Unit of Medical Genetics, University of Lausanne, 1015 Lausanne, Switzerland
| | - Ileana Bortolomai
- San Raffaele Telethon Institute for Gene Therapy, Istituto di Ricerca e Cura a Carattere Scientifico San Raffaele Scientific Institute, 20132 Milan, Italy.,Consiglio Nazionale delle Ricerche-Istituto di Ricerca Genetica e Biomedica, Milan Unit, 20138 Milan, Italy
| | - Likun Du
- Division of Immunology, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115
| | - Kerstin Felgentreff
- Division of Immunology, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115
| | - Lisa Ott de Bruin
- Division of Immunology, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115
| | - Kazutaka Hayashida
- Division of Respiratory Diseases, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115
| | - George Freedman
- Department of Pediatrics, University of California, San Francisco, San Francisco, CA 94143
| | - Genni Enza Marcovecchio
- San Raffaele Telethon Institute for Gene Therapy, Istituto di Ricerca e Cura a Carattere Scientifico San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Kelly Capuder
- Division of Immunology, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115
| | - Prisni Rath
- Tata Consultancy Services Innovation Labs, Telangana 500081, India
| | - Nicole Luche
- Division of Immunology, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115
| | - Elliott J Hagedorn
- Stem Cell Program, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115
| | | | - Beryl Royer-Bertrand
- Department of Computational Biology, Unit of Medical Genetics, University of Lausanne, 1015 Lausanne, Switzerland.,Division of Genetic Medicine, Lausanne University Hospital, University of Lausanne, 1015 Lausanne, Switzerland
| | - Silvia Giliani
- A. Nocivelli Institute for Molecular Medicine, University of Brescia, 25123 Brescia, Italy
| | - Pietro Luigi Poliani
- Department of Molecular and Translational Medicine, University of Brescia, 25123 Brescia, Italy
| | - Luisa Imberti
- Centro di ricerca emato-oncologica AIL, Spedali Civili, 25123 Brescia, Italy
| | - Kerry Dobbs
- Laboratory of Host Defenses, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, 20892
| | - Fabienne E Poulain
- Department of Biological Sciences, University of South Carolina, Columbia, SC 29208
| | - Alberto Martini
- Unita' Operativa Pediatria 2, Istituto Giannina Gaslini, 16148 Genoa, Italy
| | - John Manis
- Department of Laboratory Medicine, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115
| | - Robert J Linhardt
- Department of Chemistry and Chemical Biology, Rensselaer Polytechnic Institute, Troy, NY 12180
| | - Marita Bosticardo
- San Raffaele Telethon Institute for Gene Therapy, Istituto di Ricerca e Cura a Carattere Scientifico San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Sergio Damian Rosenzweig
- Department of Laboratory Medicine, National Institutes of Health Clinical Center, National Institutes of Health, Bethesda, MD, 20892
| | - Hane Lee
- Department of Pathology and Laboratory Medicine, University of California at Los Angeles, Los Angeles, CA 90095
| | - Jennifer M Puck
- Department of Pediatrics, University of California, San Francisco, San Francisco, CA 94143
| | - Juan Carlos Zúñiga-Pflücker
- Department of Immunology, Sunnybrook Research Institute, University of Toronto, Toronto, Ontario M5S, Canada
| | - Leonard Zon
- Stem Cell Program, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115
| | - Pyong Woo Park
- Division of Respiratory Diseases, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115
| | - Andrea Superti-Furga
- Division of Genetic Medicine, Lausanne University Hospital, University of Lausanne, 1015 Lausanne, Switzerland
| | - Luigi D Notarangelo
- Laboratory of Host Defenses, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, 20892
| |
Collapse
|
36
|
Matsuo I, Kimura-Yoshida C. Extracellular distribution of diffusible growth factors controlled by heparan sulfate proteoglycans during mammalian embryogenesis. Philos Trans R Soc Lond B Biol Sci 2015; 369:rstb.2013.0545. [PMID: 25349453 DOI: 10.1098/rstb.2013.0545] [Citation(s) in RCA: 74] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
During mouse embryogenesis, diffusible growth factors, i.e. fibroblast growth factors, Wnt, bone morphogenetic protein and Hedgehog family members, emanating from localized areas can travel through the extracellular space and reach their target cells to specify the cell fate and form tissue architectures in coordination. However, the mechanisms by which these growth factors travel great distances to their target cells and control the signalling activity as morphogens remain an enigma. Recent studies in mice and other model animals have revealed that heparan sulfate proteoglycans (HSPGs) located on the cell surface (e.g. syndecans and glypicans) and in the extracellular matrix (ECM; e.g. perlecan and agrin) play crucial roles in the extracellular distribution of growth factors. Principally, the function of HSPGs depends primarily on the fine features and localization of their heparan sulfate glycosaminoglycan chains. Cell-surface-tethered HSPGs retain growth factors as co-receptors and/or endocytosis mediators, and enzymatic release of HSPGs from the cell membrane allows HSPGs to transport or move multiple growth factors. By contrast, ECM-associated HSPGs function as a reservoir or barrier in a context-dependent manner. This review is focused on our current understanding of the extracellular distribution of multiple growth factors controlled by HSPGs in mammalian development.
Collapse
Affiliation(s)
- Isao Matsuo
- Department of Molecular Embryology, Osaka Medical Center and Research Institute for Maternal and Child Health, Osaka Prefectural Hospital Organization, 840 Murodo-cho, Izumi, Osaka 594-1101, Japan
| | - Chiharu Kimura-Yoshida
- Department of Molecular Embryology, Osaka Medical Center and Research Institute for Maternal and Child Health, Osaka Prefectural Hospital Organization, 840 Murodo-cho, Izumi, Osaka 594-1101, Japan
| |
Collapse
|
37
|
Matsumoto Y, Matsumoto K, Harimaya K, Okada S, Doi T, Iwamoto Y. Scoliosis in patients with multiple hereditary exostoses. EUROPEAN SPINE JOURNAL : OFFICIAL PUBLICATION OF THE EUROPEAN SPINE SOCIETY, THE EUROPEAN SPINAL DEFORMITY SOCIETY, AND THE EUROPEAN SECTION OF THE CERVICAL SPINE RESEARCH SOCIETY 2015; 24:1568-73. [PMID: 25794701 DOI: 10.1007/s00586-015-3883-4] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/07/2014] [Revised: 03/12/2015] [Accepted: 03/12/2015] [Indexed: 10/23/2022]
Abstract
PURPOSE To investigate the prevalence of and to identify independent predictors associated with scoliosis in patients with multiple hereditary exostoses (MHE). METHODS Fifty patients with MHE were clinically examined, and the diagnosis of scoliosis was made based on radiographs. To classify disease severity, three classes based on the presence of deformities and functional limitations were defined. Significant independent predictors of scoliosis in MHE were statistically analyzed. RESULTS Scoliosis was present in 36 patients (MHE-scoliosis) (72 %). In the MHE-scoliosis group, the mean primary curve was 15.3° ± 5.7° (range 10°-34°) and the mean minor curve was 10.6° ± 7° (range 6°-32°). Left curve was predominant (72 %), and the apex was located in the thoracolumbar or lumbar spine in 64 % of patients. Univariable and multivariable analyses confirmed that MHE severity was a significant predictor of moderate scoliosis (≥20°). CONCLUSIONS Our study confirmed that scoliosis is a common feature of MHE and disease severity is a predictor of moderate scoliosis (≥20°).
Collapse
Affiliation(s)
- Yoshihiro Matsumoto
- Department of Orthopaedic Surgery, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan,
| | | | | | | | | | | |
Collapse
|
38
|
Nakamura R, Nakamura F, Fukunaga S. Disruption of endogenous perlecan function improves differentiation of rat articular chondrocytesin vitro. Anim Sci J 2014; 86:449-58. [DOI: 10.1111/asj.12309] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2014] [Accepted: 07/09/2014] [Indexed: 01/11/2023]
Affiliation(s)
- Ryosuke Nakamura
- Laboratory of Animal By-Product Science; Research Faculty of Agriculture; Hokkaido University; Sapporo Japan
| | - Fumio Nakamura
- Laboratory of Animal By-Product Science; Research Faculty of Agriculture; Hokkaido University; Sapporo Japan
| | - Shigeharu Fukunaga
- Laboratory of Animal By-Product Science; Research Faculty of Agriculture; Hokkaido University; Sapporo Japan
| |
Collapse
|
39
|
Mizumoto S, Yamada S, Sugahara K. Human genetic disorders and knockout mice deficient in glycosaminoglycan. BIOMED RESEARCH INTERNATIONAL 2014; 2014:495764. [PMID: 25126564 PMCID: PMC4122003 DOI: 10.1155/2014/495764] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/09/2014] [Accepted: 06/08/2014] [Indexed: 12/20/2022]
Abstract
Glycosaminoglycans (GAGs) are constructed through the stepwise addition of respective monosaccharides by various glycosyltransferases and maturated by epimerases and sulfotransferases. The structural diversity of GAG polysaccharides, including their sulfation patterns and sequential arrangements, is essential for a wide range of biological activities such as cell signaling, cell proliferation, tissue morphogenesis, and interactions with various growth factors. Studies using knockout mice of enzymes responsible for the biosynthesis of the GAG side chains of proteoglycans have revealed their physiological functions. Furthermore, mutations in the human genes encoding glycosyltransferases, sulfotransferases, and related enzymes responsible for the biosynthesis of GAGs cause a number of genetic disorders including chondrodysplasia, spondyloepiphyseal dysplasia, and Ehlers-Danlos syndromes. This review focused on the increasing number of glycobiological studies on knockout mice and genetic diseases caused by disturbances in the biosynthetic enzymes for GAGs.
Collapse
Affiliation(s)
- Shuji Mizumoto
- Department of Pathobiochemistry, Faculty of Pharmacy, Meijo University, 150 Yagotoyama, Tempaku-ku, Nagoya 468-8503, Japan
| | - Shuhei Yamada
- Department of Pathobiochemistry, Faculty of Pharmacy, Meijo University, 150 Yagotoyama, Tempaku-ku, Nagoya 468-8503, Japan
| | - Kazuyuki Sugahara
- Laboratory of Proteoglycan Signaling and Therapeutics, Frontier Research Center for Post-Genomic Science and Technology, Graduate School of Life Science, Hokkaido University, West-11, North-21, Kita-ku, Sapporo, Hokkaido 001-0021, Japan
| |
Collapse
|
40
|
Cuellar A, Inui A, James MA, Borys D, Reddi AH. Immunohistochemical Localization of Bone Morphogenetic Proteins (BMPs) and their Receptors in Solitary and Multiple Human Osteochondromas. J Histochem Cytochem 2014; 62:488-98. [PMID: 24789804 DOI: 10.1369/0022155414535781] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2013] [Accepted: 04/11/2014] [Indexed: 12/14/2022] Open
Abstract
The expression of bone morphogenetic proteins (BMPs) and their cognate receptors (BMPRs) in osteochondromas has not been investigated. We determined the immunohistochemical localization and distribution of BMP-2/4, -6 and -7; BMP receptors BMPR-1A, BMPR-1B and BMPR-2; signal transducing proteins phosphorylated Smad1/5/8; and BMP antagonist noggin in the cartilaginous cap of solitary (SO) and multiple (MO) human osteochondromas and compared these with bovine growth plate and articular cartilage. The distribution and localization patterns for BMP-6, BMP-7, BMPR-1A and BMPR-2 were similar between the cartilaginous cap and the growth plate. BMP-2/4 and BMPR-1B were present throughout the growth plate. However, BMP-2/4 and phosphorylated Smad1/5/8 were mainly detected in proliferating chondrocytes of the cartilaginous cap. Also, BMPR-1B was found in hypertrophic chondrocytes of SO and proliferating chondrocytes of MO. Noggin was observed in resting chondrocytes and, to a lesser extent, in clustered proliferating chondrocytes in SO. On the other hand, noggin in MO was observed in proliferating chondrocytes. Since BMPs can stimulate proliferation and hypertrophic differentiation of chondrocytes, these findings suggest that there is an imbalance of BMP-2/4 and noggin interactions that may lead to abnormal regulation of chondrocyte proliferation and differentiation in the cartilaginous cap of human osteochondromas.
Collapse
Affiliation(s)
- Araceli Cuellar
- Lawrence Ellison Center for Tissue Regeneration and Repair (AC, AI, AHR), University of California Davis, Sacramento, CaliforniaDepartment of Orthopaedic Surgery (AC, AI, AHR), University of California Davis, Sacramento, CaliforniaDepartment of Pathology (DB), University of California Davis, Sacramento, CaliforniaShriners Hospital for Children Northern California, Sacramento, California (MAJ)
| | - Atsuyuki Inui
- Lawrence Ellison Center for Tissue Regeneration and Repair (AC, AI, AHR), University of California Davis, Sacramento, CaliforniaDepartment of Orthopaedic Surgery (AC, AI, AHR), University of California Davis, Sacramento, CaliforniaDepartment of Pathology (DB), University of California Davis, Sacramento, CaliforniaShriners Hospital for Children Northern California, Sacramento, California (MAJ)
| | - Michelle A James
- Lawrence Ellison Center for Tissue Regeneration and Repair (AC, AI, AHR), University of California Davis, Sacramento, CaliforniaDepartment of Orthopaedic Surgery (AC, AI, AHR), University of California Davis, Sacramento, CaliforniaDepartment of Pathology (DB), University of California Davis, Sacramento, CaliforniaShriners Hospital for Children Northern California, Sacramento, California (MAJ)
| | - Dariusz Borys
- Lawrence Ellison Center for Tissue Regeneration and Repair (AC, AI, AHR), University of California Davis, Sacramento, CaliforniaDepartment of Orthopaedic Surgery (AC, AI, AHR), University of California Davis, Sacramento, CaliforniaDepartment of Pathology (DB), University of California Davis, Sacramento, CaliforniaShriners Hospital for Children Northern California, Sacramento, California (MAJ)
| | - A Hari Reddi
- Lawrence Ellison Center for Tissue Regeneration and Repair (AC, AI, AHR), University of California Davis, Sacramento, CaliforniaDepartment of Orthopaedic Surgery (AC, AI, AHR), University of California Davis, Sacramento, CaliforniaDepartment of Pathology (DB), University of California Davis, Sacramento, CaliforniaShriners Hospital for Children Northern California, Sacramento, California (MAJ)
| |
Collapse
|
41
|
Jochmann K, Bachvarova V, Vortkamp A. Reprint of: Heparan sulfate as a regulator of endochondral ossification and osteochondroma development. Matrix Biol 2014; 35:239-47. [PMID: 24726293 DOI: 10.1016/j.matbio.2014.04.001] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2013] [Revised: 11/17/2013] [Accepted: 11/17/2013] [Indexed: 12/19/2022]
Abstract
Most elements of the vertebrate skeleton are formed by endochondral ossification. This process is initiated with mesenchymal cells that condense and differentiate into chondrocytes. These undergo several steps of differentiation from proliferating into hypertrophic chondrocytes, which are subsequently replaced by bone. Chondrocyte proliferation and differentiation are tightly controlled by a complex network of signaling molecules. During recent years, it has become increasingly clear that heparan sulfate (HS) carrying proteoglycans play a critical role in controlling the distribution and activity of these secreted factors. In this review we summarize the current understanding of the role of HS in regulating bone formation. In human, mutations in the HS synthetizing enzymes Ext1 and Ext2 induce the Multiple Osteochondroma syndrome, a skeletal disorder characterized by short stature and the formation of benign cartilage-capped tumors. We review the current insight into the origin of the disease and discuss its possible molecular basis. In addition, we summarize the existing insight into the role of HS as a regulator of signal propagation and signaling strength in the developing skeleton.
Collapse
Affiliation(s)
- Katja Jochmann
- Department of Developmental Biology, Faculty of Biology and Centre for Medical Biotechnology, University of Duisburg-Essen, Essen, Germany.
| | - Velina Bachvarova
- Department of Developmental Biology, Faculty of Biology and Centre for Medical Biotechnology, University of Duisburg-Essen, Essen, Germany.
| | - Andrea Vortkamp
- Department of Developmental Biology, Faculty of Biology and Centre for Medical Biotechnology, University of Duisburg-Essen, Essen, Germany.
| |
Collapse
|
42
|
Jones KB, Pacifici M, Hilton MJ. Multiple hereditary exostoses (MHE): elucidating the pathogenesis of a rare skeletal disorder through interdisciplinary research. Connect Tissue Res 2014; 55:80-8. [PMID: 24409815 DOI: 10.3109/03008207.2013.867957] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Abstract An interdisciplinary and international group of clinicians and scientists gathered in Philadelphia, PA, to attend the fourth International Research Conference on Multiple Hereditary Exostoses (MHE), a rare and severe skeletal disorder. MHE is largely caused by autosomal dominant mutations in EXT1 or EXT2, genes encoding Golgi-associated glycosyltransferases responsible for heparan sulfate (HS) synthesis. HS chains are key constituents of cell surface- and extracellular matrix-associated proteoglycans, which are known regulators of skeletal development. MHE affected individuals are HS-deficient, can display skeletal growth retardation and deformities, and consistently develop benign, cartilage-capped bony outgrowths (termed exostoses or osteochondromas) near the growth plates of many skeletal elements. Nearly 2% of patients will have their exostoses progress to malignancy, becoming peripheral chondrosarcomas. Current treatments are limited to the surgical removal of symptomatic exostoses. No definitive treatments have been established to inhibit further formation and growth of exostoses, prevent transition to malignancy, or address other medical problems experienced by MHE patients, including chronic pain. Thus, the goals of the Conference were to assess our current understanding of MHE pathogenesis, identify key gaps in information, envision future therapeutic strategies and discuss ways to test and implement them. This report provides an assessment of the exciting and promising findings in MHE and related fields presented at the Conference and a discussion of the future MHE research directions. The Conference underlined the critical usefulness of gathering experts in several research fields to forge new alliances and identify cross-fertilization areas to benefit both basic and translational biomedical research on the skeleton.
Collapse
Affiliation(s)
- Kevin B Jones
- Department of Orthopaedics and Center for Children's Cancer Research, Huntsman Cancer Institute, University of Utah School of Medicine , Salt Lake City, UT , USA
| | | | | |
Collapse
|
43
|
Jochmann K, Bachvarova V, Vortkamp A. Heparan sulfate as a regulator of endochondral ossification and osteochondroma development. Matrix Biol 2013; 34:55-63. [PMID: 24370655 DOI: 10.1016/j.matbio.2013.11.003] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2013] [Revised: 11/17/2013] [Accepted: 11/17/2013] [Indexed: 12/18/2022]
Abstract
Most elements of the vertebrate skeleton are formed by endochondral ossification. This process is initiated with mesenchymal cells that condense and differentiate into chondrocytes. These undergo several steps of differentiation from proliferating into hypertrophic chondrocytes, which are subsequently replaced by bone. Chondrocyte proliferation and differentiation are tightly controlled by a complex network of signaling molecules. During recent years, it has become increasingly clear that heparan sulfate (HS) carrying proteoglycans play a critical role in controlling the distribution and activity of these secreted factors. In this review we summarize the current understanding of the role of HS in regulating bone formation. In human, mutations in the HS synthetizing enzymes Ext1 and Ext2 induce the Multiple Osteochondroma syndrome, a skeletal disorder characterized by short stature and the formation of benign cartilage-capped tumors. We review the current insight into the origin of the disease and discuss its possible molecular basis. In addition, we summarize the existing insight into the role of HS as a regulator of signal propagation and signaling strength in the developing skeleton.
Collapse
Affiliation(s)
- Katja Jochmann
- Department of Developmental Biology, Faculty of Biology and Centre for Medical Biotechnology, University Duisburg-Essen, Essen, Germany.
| | - Velina Bachvarova
- Department of Developmental Biology, Faculty of Biology and Centre for Medical Biotechnology, University Duisburg-Essen, Essen, Germany.
| | - Andrea Vortkamp
- Department of Developmental Biology, Faculty of Biology and Centre for Medical Biotechnology, University Duisburg-Essen, Essen, Germany.
| |
Collapse
|
44
|
Sgariglia F, Candela ME, Huegel J, Jacenko O, Koyama E, Yamaguchi Y, Pacifici M, Enomoto-Iwamoto M. Epiphyseal abnormalities, trabecular bone loss and articular chondrocyte hypertrophy develop in the long bones of postnatal Ext1-deficient mice. Bone 2013; 57:220-31. [PMID: 23958822 PMCID: PMC4107462 DOI: 10.1016/j.bone.2013.08.012] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/13/2013] [Revised: 08/05/2013] [Accepted: 08/13/2013] [Indexed: 11/25/2022]
Abstract
Long bones are integral components of the limb skeleton. Recent studies have indicated that embryonic long bone development is altered by mutations in Ext genes and consequent heparan sulfate (HS) deficiency, possibly due to changes in activity and distribution of HS-binding/growth plate-associated signaling proteins. Here we asked whether Ext function is continuously required after birth to sustain growth plate function and long bone growth and organization. Compound transgenic Ext1(f/f);Col2CreERT mice were injected with tamoxifen at postnatal day 5 (P5) to ablate Ext1 in cartilage and monitored over time. The Ext1-deficient mice exhibited growth retardation already by 2weeks post-injection, as did their long bones. Mutant growth plates displayed a severe disorganization of chondrocyte columnar organization, a shortened hypertrophic zone with low expression of collagen X and MMP-13, and reduced primary spongiosa accompanied, however, by increased numbers of TRAP-positive osteoclasts at the chondro-osseous border. The mutant epiphyses were abnormal as well. Formation of a secondary ossification center was significantly delayed but interestingly, hypertrophic-like chondrocytes emerged within articular cartilage, similar to those often seen in osteoarthritic joints. Indeed, the cells displayed a large size and round shape, expressed collagen X and MMP-13 and were surrounded by an abundant Perlecan-rich pericellular matrix not seen in control articular chondrocytes. In addition, ectopic cartilaginous outgrowths developed on the lateral side of mutant growth plates over time that resembled exostotic characteristic of children with Hereditary Multiple Exostoses, a syndrome caused by Ext mutations and HS deficiency. In sum, the data do show that Ext1 is continuously required for postnatal growth and organization of long bones as well as their adjacent joints. Ext1 deficiency elicits defects that can occur in human skeletal conditions including trabecular bone loss, osteoarthritis and HME.
Collapse
Affiliation(s)
- Federica Sgariglia
- Translational Research Program in Pediatric Orthopaedics, Division of Orthopaedic Surgery, Department of Surgery, The Children's Hospital of Philadelphia, Philadelphia, PA 19104
| | - Maria Elena Candela
- Translational Research Program in Pediatric Orthopaedics, Division of Orthopaedic Surgery, Department of Surgery, The Children's Hospital of Philadelphia, Philadelphia, PA 19104
| | - Julianne Huegel
- Translational Research Program in Pediatric Orthopaedics, Division of Orthopaedic Surgery, Department of Surgery, The Children's Hospital of Philadelphia, Philadelphia, PA 19104
| | - Olena Jacenko
- Department of Animal Biology, School of Veterinary Medicine, University of Philadelphia, Philadelphia, PA 19104
| | - Eiki Koyama
- Translational Research Program in Pediatric Orthopaedics, Division of Orthopaedic Surgery, Department of Surgery, The Children's Hospital of Philadelphia, Philadelphia, PA 19104
- Department of Orthopaedic Surgery, Perelman School of Medicine, University of Philadelphia, Philadelphia, PA 19104
| | - Yu Yamaguchi
- Sanford Children's Health Research Center, Sanford-Burnham Medical Research Institute, La Jolla, CA, 92037
| | - Maurizio Pacifici
- Translational Research Program in Pediatric Orthopaedics, Division of Orthopaedic Surgery, Department of Surgery, The Children's Hospital of Philadelphia, Philadelphia, PA 19104
- Department of Orthopaedic Surgery, Perelman School of Medicine, University of Philadelphia, Philadelphia, PA 19104
| | - Motomi Enomoto-Iwamoto
- Translational Research Program in Pediatric Orthopaedics, Division of Orthopaedic Surgery, Department of Surgery, The Children's Hospital of Philadelphia, Philadelphia, PA 19104
- Department of Orthopaedic Surgery, Perelman School of Medicine, University of Philadelphia, Philadelphia, PA 19104
| |
Collapse
|
45
|
Huegel J, Sgariglia F, Enomoto-Iwamoto M, Koyama E, Dormans JP, Pacifici M. Heparan sulfate in skeletal development, growth, and pathology: the case of hereditary multiple exostoses. Dev Dyn 2013; 242:1021-32. [PMID: 23821404 DOI: 10.1002/dvdy.24010] [Citation(s) in RCA: 63] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2013] [Revised: 06/21/2013] [Accepted: 06/22/2013] [Indexed: 12/14/2022] Open
Abstract
Heparan sulfate (HS) is an essential component of cell surface and matrix-associated proteoglycans. Due to their sulfation patterns, the HS chains interact with numerous signaling proteins and regulate their distribution and activity on target cells. Many of these proteins, including bone morphogenetic protein family members, are expressed in the growth plate of developing skeletal elements, and several skeletal phenotypes are caused by mutations in those proteins as well as in HS-synthesizing and modifying enzymes. The disease we discuss here is hereditary multiple exostoses (HME), a disorder caused by mutations in HS synthesizing enzymes EXT1 and EXT2, leading to HS deficiency. The exostoses are benign cartilaginous-bony outgrowths, form next to growth plates, can cause growth retardation and deformities, chronic pain and impaired motion, and progress to malignancy in 2-5% of patients. We describe recent advancements on HME pathogenesis and exostosis formation deriving from studies that have determined distribution, activities and roles of signaling proteins in wild-type and HS-deficient cells and tissues. Aberrant distribution of signaling factors combined with aberrant responsiveness of target cells to those same factors appear to be a major culprit in exostosis formation. Insights from these studies suggest plausible and cogent ideas about how HME could be treated in the future.
Collapse
Affiliation(s)
- Julianne Huegel
- Translational Research Program in Pediatric Orthopaedics, Division of Orthopaedic Surgery, The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA.
| | | | | | | | | | | |
Collapse
|
46
|
Cell biology of osteochondromas: bone morphogenic protein signalling and heparan sulphates. INTERNATIONAL ORTHOPAEDICS 2013; 37:1591-6. [PMID: 23771188 DOI: 10.1007/s00264-013-1906-5] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/07/2013] [Accepted: 04/18/2013] [Indexed: 01/01/2023]
Abstract
Frequent benign outgrowths from bone known as osteochondromas, exhibiting typical endochondral ossification, are reported from single to multiple lesions. Characterised by a high incidence of osteochondromas and skeletal deformities, multiple hereditary exostoses (MHE) is the most common inherited musculoskeletal condition. While factors for severity remain unknown, mutations in exostosin 1 and exostosin 2 genes, encoding glycosyltransferases involved in the biosynthesis of ubiquitously expressed heparan sulphate (HS) chains, are associated with MHE. HS-binding bone morphogenetic proteins (BMPs) are multifunctional proteins involved in the morphogenesis of bone and cartilage. HS and HS proteoglycans are involved in BMP-mediated morphogenesis by regulating their gradient formation and activity. Mutations in exostosin genes disturb HS biosynthesis, subsequently affecting its functional role in the regulation of signalling pathways. As BMPs are the primordial morphogens for bone development, we propose the hypothesis that BMP signalling may be critical in osteochondromas. For this reason, the outcomes of exostosin mutations on HS biosynthesis and interactions within osteochondromas and MHE are reviewed. Since BMPs are HS binding proteins, the interactions of HS with the BMP signalling pathway are discussed. The impact of mouse models in the quest to better understand the cell biology of osteochondromas is discussed. Several challenges and questions still remain and further investigations are needed to explore new approaches for better understanding of the pathogenesis of osteochondromas.
Collapse
|
47
|
Huegel J, Mundy C, Sgariglia F, Nygren P, Billings PC, Yamaguchi Y, Koyama E, Pacifici M. Perichondrium phenotype and border function are regulated by Ext1 and heparan sulfate in developing long bones: a mechanism likely deranged in Hereditary Multiple Exostoses. Dev Biol 2013; 377:100-12. [PMID: 23458899 PMCID: PMC3733121 DOI: 10.1016/j.ydbio.2013.02.008] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2012] [Revised: 02/07/2013] [Accepted: 02/13/2013] [Indexed: 10/27/2022]
Abstract
During limb skeletogenesis the cartilaginous long bone anlagen and their growth plates become delimited by perichondrium with which they interact functionally. Yet, little is known about how, despite being so intimately associated with cartilage, perichondrium acquires and maintains its distinct phenotype and exerts its border function. Because perichondrium becomes deranged and interrupted by cartilaginous outgrowths in Hereditary Multiple Exostoses (HME), a pediatric disorder caused by EXT mutations and consequent heparan sulfate (HS) deficiency, we asked whether EXT genes and HS normally have roles in establishing its phenotype and function. Indeed, conditional Ext1 ablation in perichondrium and lateral chondrocytes flanking the epiphyseal region of mouse embryo long bone anlagen - a region encompassing the groove of Ranvier - caused ectopic cartilage formation. A similar response was observed when HS function was disrupted in long bone anlagen explants by genetic, pharmacological or enzymatic means, a response preceded by ectopic BMP signaling within perichondrium. These treatments also triggered excess chondrogenesis and cartilage nodule formation and overexpression of chondrogenic and matrix genes in limb bud mesenchymal cells in micromass culture. Interestingly, the treatments disrupted the peripheral definition and border of the cartilage nodules in such a way that many nodules overgrew and fused with each other into large amorphous cartilaginous masses. Interference with HS function reduced the physical association and interactions of BMP2 with HS and increased the cell responsiveness to endogenous and exogenous BMP proteins. In sum, Ext genes and HS are needed to establish and maintain perichondrium's phenotype and border function, restrain pro-chondrogenic signaling proteins including BMPs, and restrict chondrogenesis. Alterations in these mechanisms may contribute to exostosis formation in HME, particularly at the expense of regions rich in progenitor cells including the groove of Ranvier.
Collapse
Affiliation(s)
- Julianne Huegel
- Division of Orthopaedic Surgery, The Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA.
| | | | | | | | | | | | | | | |
Collapse
|
48
|
Matsuo I, Kimura-Yoshida C. Extracellular modulation of Fibroblast Growth Factor signaling through heparan sulfate proteoglycans in mammalian development. Curr Opin Genet Dev 2013; 23:399-407. [PMID: 23465883 DOI: 10.1016/j.gde.2013.02.004] [Citation(s) in RCA: 76] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2013] [Revised: 02/04/2013] [Accepted: 02/07/2013] [Indexed: 12/27/2022]
Abstract
Fibroblast Growth Factor (FGF) signaling plays crucial roles in multiple cellular processes including cell proliferation, differentiation, survival, and migration during mammalian embryogenesis. In the extracellular matrix, as well as at the cell surface, the movement of FGF ligands to target cells and the subsequent complex formations with their receptors are positively and negatively controlled extracellularly by heparan sulfate proteoglycans (HSPGs) such as syndecans, glypicans, and perlecan. Additionally, spreading of HSPGs by cleavage with sheddases such as proteinases and heparanases, and the overall length and sulfation level of specific heparan sulfate structures further generate a great diversity of FGF signaling outcomes. This review presents our current understanding of the regulatory mechanisms of FGF signaling in extracellular spaces through HSPGs in mammalian development.
Collapse
Affiliation(s)
- Isao Matsuo
- Department of Molecular Embryology, Osaka Medical Center and Research Institute for Maternal and Child Health, Osaka Prefectural Hospital Organization, 840 Murodo-cho, Izumi, Osaka 594-1101, Japan.
| | | |
Collapse
|
49
|
Kraushaar DC, Rai S, Condac E, Nairn A, Zhang S, Yamaguchi Y, Moremen K, Dalton S, Wang L. Heparan sulfate facilitates FGF and BMP signaling to drive mesoderm differentiation of mouse embryonic stem cells. J Biol Chem 2012; 287:22691-700. [PMID: 22556407 DOI: 10.1074/jbc.m112.368241] [Citation(s) in RCA: 71] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Heparan sulfate (HS) has been implicated in regulating cell fate decisions during differentiation of embryonic stem cells (ESCs) into advanced cell types. However, the necessity and the underlying molecular mechanisms of HS in early cell lineage differentiation are still largely unknown. In this study, we examined the potential of EXT1(-/-) mouse ESCs (mESCs), that are deficient in HS, to differentiate into primary germ layer cells. We observed that EXT1(-/-) mESCs lost their differentiation competence and failed to differentiate into Pax6(+)-neural precursor cells and mesodermal cells. More detailed analyses highlighted the importance of HS for the induction of Brachyury(+) pan-mesoderm as well as normal gene expression associated with the dorso-ventral patterning of mesoderm. Examination of developmental cell signaling revealed that EXT1 ablation diminished FGF and BMP but not Wnt signaling. Furthermore, restoration of FGF and BMP signaling each partially rescued mesoderm differentiation defects. We further show that BMP4 is more prone to degradation in EXT1(-/-) mESCs culture medium compared with that of wild type cells. Therefore, our data reveal that HS stabilizes BMP ligand and thereby maintains the BMP signaling output required for normal mesoderm differentiation. In summary, our study demonstrates that HS is required for ESC pluripotency, in particular lineage specification into mesoderm through facilitation of FGF and BMP signaling.
Collapse
Affiliation(s)
- Daniel C Kraushaar
- Complex Carbohydrate Research Center and Department of Biochemistry and Molecular Biology, University of Georgia, Athens, Georgia 30602, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
50
|
McCarthy KJ, Wassenhove-McCarthy DJ. The glomerular basement membrane as a model system to study the bioactivity of heparan sulfate glycosaminoglycans. MICROSCOPY AND MICROANALYSIS : THE OFFICIAL JOURNAL OF MICROSCOPY SOCIETY OF AMERICA, MICROBEAM ANALYSIS SOCIETY, MICROSCOPICAL SOCIETY OF CANADA 2012; 18:3-21. [PMID: 22258721 PMCID: PMC3351113 DOI: 10.1017/s1431927611012682] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/31/2023]
Abstract
The glomerular basement membrane and its associated cells are critical elements in the renal ultrafiltration process. Traditionally the anionic charge associated with several carbohydrate moieties in the glomerular basement membrane are thought to form a charge selective barrier that restricts the transmembrane flux of anionic proteins across the glomerular basement membrane into the urinary space. The charge selective function, along with the size selective component of the basement membrane, serves to limit the efflux of plasma proteins from the capillary lumen. Heparan sulfate glycosaminoglycans are anionically charged carbohydrate structures attached to proteoglycan core proteins and have a role in establishing the charge selective function of the glomerular basement membrane. Although there are a large number of studies in the literature that support this concept, the results of several recent studies using molecular genetic approaches to minimize the anionic charge of the glomerular basement membrane would suggest that the role of heparan sulfate glycosaminoglycans in the glomerular capillary wall are still not yet entirely resolved, suggesting that this research area still requires new and novel exploration.
Collapse
Affiliation(s)
- Kevin J McCarthy
- Department of Pathology, LSU Health Sciences Center-Shreveport, 1501 Kings Highway, Shreveport, LA 71130-3932, USA.
| | | |
Collapse
|