1
|
Wang C, Fan M, Heo SC, Adams SM, Li T, Liu Y, Li Q, Loebel C, Burdick JA, Lu XL, Birk DE, Alisafaei F, Mauck RL, Han L. Structure, Mechanics, and Mechanobiology of Fibrocartilage Pericellular Matrix Mediated by Type V Collagen. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025:e14750. [PMID: 40407177 DOI: 10.1002/advs.202414750] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/11/2024] [Revised: 05/01/2025] [Indexed: 05/28/2025]
Abstract
The pericellular matrix (PCM) is the immediate microniche surrounding cells in various tissues, regulating matrix turnover, cell-matrix interactions, and disease. This study elucidates the structure-mechanical properties and mechanobiology of the PCM in fibrocartilage, using the murine meniscus as the model. The fibrocartilage PCM is comprised of thin, randomly oriented collagen fibrils that entrap proteoglycans, contrasting with the densely packed, highly aligned collagen fibers in the bulk extracellular matrix (ECM). Compared to the ECM, the PCM exhibits lower modulus and greater isotropy, but has similar relative viscoelastic properties. In Col5a1+/- menisci, the reduction of collagen V results in thicker, more heterogeneous collagen fibrils, reduced modulus, loss of isotropy and faster viscoelastic relaxation in the PCM. Such altered PCM leads to impaired matrix-to-cell strain transmission, and in turn, disrupts mechanotransduction of meniscal cells, as illustrated by reduced calcium signaling activities and alters expression of matrix genes. In vitro, Col5a1+/- cells produce a weakened PCM with inferior properties and reduced protection of cells against tensile stretch. These findings highlight the PCM as a distinctive microstructure in fibrocartilage mechanobiology, underscoring a pivotal role of collagen V in PCM function. Targeting the PCM or its constituents offers potential for improving meniscus regeneration, osteoarthritis intervention and broader fibrocartilage-related therapies.
Collapse
Affiliation(s)
- Chao Wang
- School of Biomedical Engineering, Science and Health Systems, Drexel University, Philadelphia, PA, 19104, USA
| | - Mingyue Fan
- School of Biomedical Engineering, Science and Health Systems, Drexel University, Philadelphia, PA, 19104, USA
| | - Su Chin Heo
- McKay Orthopaedic Research Laboratory, Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Sheila M Adams
- Department of Molecular Pharmacology and Physiology, Morsani School of Medicine, University of South Florida, Tampa, FL, 33612, USA
| | - Thomas Li
- School of Biomedical Engineering, Science and Health Systems, Drexel University, Philadelphia, PA, 19104, USA
| | - Yuchen Liu
- School of Biomedical Engineering, Science and Health Systems, Drexel University, Philadelphia, PA, 19104, USA
| | - Qing Li
- School of Biomedical Engineering, Science and Health Systems, Drexel University, Philadelphia, PA, 19104, USA
| | - Claudia Loebel
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Jason A Burdick
- BioFrontiers Institute and Department of Chemical and Biological Engineering, University of Colorado, Boulder, CO, 80309, USA
| | - X Lucas Lu
- Department of Mechanical Engineering, University of Delaware, Newark, DE, 19716, USA
| | - David E Birk
- Department of Molecular Pharmacology and Physiology, Morsani School of Medicine, University of South Florida, Tampa, FL, 33612, USA
| | - Farid Alisafaei
- Department of Mechanical and Industrial Engineering, New Jersey Institute of Technology, Newark, NJ, 07102, USA
| | - Robert L Mauck
- McKay Orthopaedic Research Laboratory, Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
- Translational Musculoskeletal Research Center, Corporal Michael J. Crescenz Veterans Administration Medical Center, Philadelphia, PA, 19104, USA
| | - Lin Han
- School of Biomedical Engineering, Science and Health Systems, Drexel University, Philadelphia, PA, 19104, USA
| |
Collapse
|
2
|
Woods JP, Rackley A, Kwon HR, Olson LE. PDGFRα signaling regulates cartilage and fibrous tissue differentiation during synovial joint development. Nat Commun 2025; 16:4041. [PMID: 40301343 PMCID: PMC12041487 DOI: 10.1038/s41467-025-59207-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Accepted: 04/14/2025] [Indexed: 05/01/2025] Open
Abstract
Synovial joints develop from mesenchymal structures called interzones, with progenitor cells differentiating into specialized cartilaginous and fibrous tissues of the joint. Platelet-derived growth factor receptor-α (PDGFRα) is a tyrosine kinase expressed by cells of the limb bud, but its role in limb development is unknown. To investigate PDGFRα function, we generated mice expressing mutant PDGFRα with a point mutation (D842V) that increases receptor signaling. Mutant hindlimbs are immobile with knee joints fused by cartilage and lacking ligaments and menisci. The interzone marker Gdf5 is initially expressed at E12.5 but is downregulated thereafter, suggesting a defect in interzone maintenance. Omics analysis of the joint tissues identifies ectopic cartilage matrix expressing genes for cartilage and fibrotic tissue. Thus, elevated PDGFRα signaling corrupts joint development by downregulating Gdf5 and redirecting interzone progenitors into a fibrocartilage fate. This suggests that tight regulation of tyrosine kinase activity is necessary for the development of the mouse knee joint.
Collapse
Affiliation(s)
- John P Woods
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
- Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Alex Rackley
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
| | - Hae Ryong Kwon
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA.
| | - Lorin E Olson
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA.
- Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA.
| |
Collapse
|
3
|
Katoku-Kikyo N, Kawakami H, Cantor M, Kawakami Y, Kikyo N. METTL14 regulates chondrogenesis through the GDF5-RUNX-extracellular matrix gene axis during limb development. Nat Commun 2025; 16:4072. [PMID: 40307229 PMCID: PMC12043825 DOI: 10.1038/s41467-025-59346-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2024] [Accepted: 04/18/2025] [Indexed: 05/02/2025] Open
Abstract
m6A RNA methylation is essential for many aspects of mammalian development but its roles in chondrogenesis remain largely unknown. Here, we show that m6A is necessary for chondrogenesis and limb morphogenesis using limb progenitor-specific knockout mice of Mettl14, an essential subunit in the m6A methyltransferase complex. The knockout disrupts cartilage anlagen formation in limb buds with 11 downregulated proteins known to dysregulate chondrogenesis and shorten limb skeletons upon mutation in mice and humans. Further studies show a gene regulatory hierarchy among the 11 proteins. m6A stabilizes the transcript and increases the protein level of GDF5, a BMP family member. This activates the chondrogenic transcription factor genes Runx2 and Runx3, whose mRNAs are also stabilized by m6A. They promote the transcription of six collagen genes and two other chondrogenic genes, Ddrgk1 and Pbxip1. Thus, this study uncovers an m6A-based cascade essential for chondrogenesis during limb skeletal development.
Collapse
Affiliation(s)
- Nobuko Katoku-Kikyo
- Stem Cell Institute, University of Minnesota, Minneapolis, MN, USA
- Department of Genetics, Cell Biology, and Development, University of Minnesota, Minneapolis, MN, USA
| | - Hiroko Kawakami
- Stem Cell Institute, University of Minnesota, Minneapolis, MN, USA
- Department of Genetics, Cell Biology, and Development, University of Minnesota, Minneapolis, MN, USA
| | - Max Cantor
- Stem Cell Institute, University of Minnesota, Minneapolis, MN, USA
- Department of Genetics, Cell Biology, and Development, University of Minnesota, Minneapolis, MN, USA
| | - Yasuhiko Kawakami
- Stem Cell Institute, University of Minnesota, Minneapolis, MN, USA.
- Department of Genetics, Cell Biology, and Development, University of Minnesota, Minneapolis, MN, USA.
| | - Nobuaki Kikyo
- Stem Cell Institute, University of Minnesota, Minneapolis, MN, USA.
- Department of Genetics, Cell Biology, and Development, University of Minnesota, Minneapolis, MN, USA.
| |
Collapse
|
4
|
Kaneko Y, Tomiyama K, Yasuda M, Komaki Y, Ogura T, Takahashi R, Yamamoto M. Identification of gene mutations associated with the phenotype of short-limb mice emerging from a foundation colony of severely immunodeficient mice. Exp Anim 2025; 74:122-131. [PMID: 39261059 PMCID: PMC11742472 DOI: 10.1538/expanim.24-0045] [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: 04/30/2024] [Accepted: 09/02/2024] [Indexed: 09/13/2024] Open
Abstract
Here, we report the identification of causative genes for limb-shortening in individuals repeatedly found in a population of severely immunodeficient NOG mice maintained via sibling mating. First, we conducted a pedigree survey to determine whether limb-shortening was a recessive genetic trait and then identified it using a crossing test. Simultaneously, the symptoms were identified in detail using pathological analysis. Accordingly, a mouse strain exhibiting a recessive trait caused by a single gene trait and similar symptoms was identified, suggesting growth differentiation factor 5 (Gdf5) as a causative gene. Genome walking via PCR and sequence analysis of Gdf5 revealed a deletion of approximately 1.1 kb from the latter half of exon 2 of Gdf5. Furthermore, we established NOG-Gdf5bpJic by removing other modified genes and confirmed that the inheritance pattern was reconfirmed semi-dominant. In recent years, regenerative medicine research using immunodeficient mice has been actively conducted, and this murine strain is expected to contribute to niche stem cell analysis and transplantation research.
Collapse
Affiliation(s)
- Yui Kaneko
- Central Institute for Experimental Medicine and Life Science, 3-25-12 Tonomachi, Kawasaki-ku, Kawasaki, Kanagawa 210-0821, Japan
| | - Kayo Tomiyama
- Central Institute for Experimental Medicine and Life Science, 3-25-12 Tonomachi, Kawasaki-ku, Kawasaki, Kanagawa 210-0821, Japan
| | - Masahiko Yasuda
- Central Institute for Experimental Medicine and Life Science, 3-25-12 Tonomachi, Kawasaki-ku, Kawasaki, Kanagawa 210-0821, Japan
| | - Yuji Komaki
- Central Institute for Experimental Medicine and Life Science, 3-25-12 Tonomachi, Kawasaki-ku, Kawasaki, Kanagawa 210-0821, Japan
| | - Tomoyuki Ogura
- Central Institute for Experimental Medicine and Life Science, 3-25-12 Tonomachi, Kawasaki-ku, Kawasaki, Kanagawa 210-0821, Japan
| | - Riichi Takahashi
- Central Institute for Experimental Medicine and Life Science, 3-25-12 Tonomachi, Kawasaki-ku, Kawasaki, Kanagawa 210-0821, Japan
| | - Masafumi Yamamoto
- Central Institute for Experimental Medicine and Life Science, 3-25-12 Tonomachi, Kawasaki-ku, Kawasaki, Kanagawa 210-0821, Japan
| |
Collapse
|
5
|
Alexander KA, Tseng HW, Lao HW, Girard D, Barbier V, Ungerer JPJ, McWhinney BC, Samuel SG, Fleming W, Winkler IG, Salga M, Genêt F, Banzet S, Ruitenberg MJ, Lévesque JP. A glucocorticoid spike derails muscle repair to heterotopic ossification after spinal cord injury. Cell Rep Med 2024; 5:101849. [PMID: 39657663 PMCID: PMC11722129 DOI: 10.1016/j.xcrm.2024.101849] [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: 10/01/2023] [Revised: 08/02/2024] [Accepted: 11/11/2024] [Indexed: 12/12/2024]
Abstract
Why severe injury to the central nervous system (CNS) triggers the development of large neurogenic heterotopic ossifications (NHOs) within periarticular muscles remains unknown. We report that spinal cord injury (SCI) triggers a rapid corticosterone spike in mice, which is causal for NHO development because treatments with corticosterone or the synthetic glucocorticoid (GC) receptor (GR) agonist dexamethasone are sufficient to trigger heterotopic ossification and upregulate the expression of osteoinductive and osteogenic differentiation genes in injured muscles even without SCI. The central role for GR signaling in causing NHO is further demonstrated in mice deleted for the GR gene (Nr3c1), which no longer develop NHO after SCI. Furthermore, administration of clinical GR antagonists inhibits NHO development in mice with SCI. This study identifies endogenous GC as causing pathological NHO after CNS injury and suggests that GR antagonists may be of prophylactic use to prevent NHO development in victims of severe CNS injuries.
Collapse
Affiliation(s)
- Kylie A Alexander
- Mater Research Institute - The University of Queensland, Translational Research Institute, Woolloongabba, QLD 4102, Australia.
| | - Hsu-Wen Tseng
- Mater Research Institute - The University of Queensland, Translational Research Institute, Woolloongabba, QLD 4102, Australia
| | - Hong Wa Lao
- School of Biomedical Sciences, Faculty of Medicine, The University of Queensland, St Lucia, QLD 4067, Australia
| | - Dorothée Girard
- Institut de Recherche Biomédicale des Armées, 92140 Clamart, France; INSERM, UMR-MD U1197 SToRM, 92140 Clamart, France
| | - Valérie Barbier
- Mater Research Institute - The University of Queensland, Translational Research Institute, Woolloongabba, QLD 4102, Australia
| | - Jacobus P J Ungerer
- School of Biomedical Sciences, Faculty of Medicine, The University of Queensland, St Lucia, QLD 4067, Australia; Department of Chemical Pathology, Pathology Queensland, Herston, QLD 4029, Australia
| | - Brett C McWhinney
- Department of Chemical Pathology, Pathology Queensland, Herston, QLD 4029, Australia
| | - Selwin G Samuel
- Mater Research Institute - The University of Queensland, Translational Research Institute, Woolloongabba, QLD 4102, Australia
| | - Whitney Fleming
- Mater Research Institute - The University of Queensland, Translational Research Institute, Woolloongabba, QLD 4102, Australia
| | - Ingrid G Winkler
- Mater Research Institute - The University of Queensland, Translational Research Institute, Woolloongabba, QLD 4102, Australia
| | - Marjorie Salga
- Unité Péri-Opératoire du Handicap, Physical and Rehabilitation Medicine Department, Hôpital Raymond-Poincaré, Assistance Publique Hôpitaux de Paris (APHP), 92380 Garches, France
| | - François Genêt
- Unité Péri-Opératoire du Handicap, Physical and Rehabilitation Medicine Department, Hôpital Raymond-Poincaré, Assistance Publique Hôpitaux de Paris (APHP), 92380 Garches, France; Université Versailles Saint-Quentin-en-Yvelines, UFR Simone Veil - Santé, END:ICAP, INSERM U1179, 78180 Montigny-le-Bretonneux, France
| | - Sébastien Banzet
- Institut de Recherche Biomédicale des Armées, 92140 Clamart, France; INSERM, UMR-MD U1197 SToRM, 92140 Clamart, France
| | - Marc J Ruitenberg
- School of Biomedical Sciences, Faculty of Medicine, The University of Queensland, St Lucia, QLD 4067, Australia
| | - Jean-Pierre Lévesque
- Mater Research Institute - The University of Queensland, Translational Research Institute, Woolloongabba, QLD 4102, Australia.
| |
Collapse
|
6
|
McClure JJ, McIlroy GD, Symons RA, Clark SM, Cunningham I, Han W, Kania K, Colella F, Rochford JJ, De Bari C, Roelofs AJ. Disentangling the detrimental effects of local from systemic adipose tissue dysfunction on articular cartilage in the knee. Osteoarthritis Cartilage 2024; 32:1552-1565. [PMID: 39103079 DOI: 10.1016/j.joca.2024.07.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Revised: 07/11/2024] [Accepted: 07/12/2024] [Indexed: 08/07/2024]
Abstract
OBJECTIVE Obesity increases osteoarthritis (OA) risk due to adipose tissue dysfunction with associated metabolic syndrome and excess weight. Lipodystrophy syndromes exhibit systemic metabolic and inflammatory abnormalities similar to obesity without biomechanical overloading. Here, we used lipodystrophy mouse models to investigate the effects of systemic versus intra-articular adipose tissue dysfunction on the knee. METHODS Intra-articular adipose tissue development was studied using reporter mice. Mice with selective lipodystrophy of intra-articular adipose tissue were generated by conditional knockout (cKO) of Bscl2 in Gdf5-lineage cells, and compared with whole-body Bscl2 knockout (KO) mice with generalised lipodystrophy and associated systemic metabolic dysfunction. OA was induced by surgically destabilising the medial meniscus (DMM) and obesity by high-fat diet (HFD). Gene expression was analysed by quantitative RT-PCR and tissues were analysed histologically. RESULTS The infrapatellar fat pad (IFP), in contrast to overlying subcutaneous adipose tissue, developed from a template established from the Gdf5-expressing joint interzone during late embryogenesis, and was populated shortly after birth by adipocytes stochastically arising from Pdgfrα-expressing Gdf5-lineage progenitors. While female Bscl2 KO mice with generalised lipodystrophy developed spontaneous knee cartilage damage, Bscl2 cKO mice with intra-articular lipodystrophy did not, despite the presence of synovial hyperplasia and inflammation of the residual IFP. Furthermore, male Bscl2 cKO mice showed no worse cartilage damage after DMM. However, female Bscl2 cKO mice showed increased susceptibility to the cartilage-damaging effects of HFD-induced obesity. CONCLUSION Our findings emphasise the prevalent role of systemic metabolic and inflammatory effects in impairing cartilage homeostasis, with a modulatory role for intra-articular adipose tissue.
Collapse
Affiliation(s)
- Jessica J McClure
- Arthritis & Regenerative Medicine Laboratory, Centre for Arthritis and Musculoskeletal Health, University of Aberdeen, Foresterhill, Aberdeen AB25 2ZD, UK
| | - George D McIlroy
- The Rowett Institute and Aberdeen Cardiovascular and Diabetes Centre, University of Aberdeen, Foresterhill, Aberdeen AB25 2ZD, UK
| | - Rebecca A Symons
- Arthritis & Regenerative Medicine Laboratory, Centre for Arthritis and Musculoskeletal Health, University of Aberdeen, Foresterhill, Aberdeen AB25 2ZD, UK
| | - Susan M Clark
- Arthritis & Regenerative Medicine Laboratory, Centre for Arthritis and Musculoskeletal Health, University of Aberdeen, Foresterhill, Aberdeen AB25 2ZD, UK
| | - Iain Cunningham
- Arthritis & Regenerative Medicine Laboratory, Centre for Arthritis and Musculoskeletal Health, University of Aberdeen, Foresterhill, Aberdeen AB25 2ZD, UK
| | - Weiping Han
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research (A⁎STAR), Singapore
| | - Karolina Kania
- Arthritis & Regenerative Medicine Laboratory, Centre for Arthritis and Musculoskeletal Health, University of Aberdeen, Foresterhill, Aberdeen AB25 2ZD, UK
| | - Fabio Colella
- Arthritis & Regenerative Medicine Laboratory, Centre for Arthritis and Musculoskeletal Health, University of Aberdeen, Foresterhill, Aberdeen AB25 2ZD, UK
| | - Justin J Rochford
- The Rowett Institute and Aberdeen Cardiovascular and Diabetes Centre, University of Aberdeen, Foresterhill, Aberdeen AB25 2ZD, UK
| | - Cosimo De Bari
- Arthritis & Regenerative Medicine Laboratory, Centre for Arthritis and Musculoskeletal Health, University of Aberdeen, Foresterhill, Aberdeen AB25 2ZD, UK
| | - Anke J Roelofs
- Arthritis & Regenerative Medicine Laboratory, Centre for Arthritis and Musculoskeletal Health, University of Aberdeen, Foresterhill, Aberdeen AB25 2ZD, UK.
| |
Collapse
|
7
|
Coveney CR, Maridas D, Chen H, Muthuirulan P, Liu Z, Jagoda E, Yarlagadda S, Movahhedi M, Proffen B, Rosen V, Kiapour AM, Capellini TD. Complex regulatory interactions at GDF5 shape joint morphology and osteoarthritis disease risk. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.11.01.621374. [PMID: 39554166 PMCID: PMC11565913 DOI: 10.1101/2024.11.01.621374] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/19/2024]
Abstract
Our ability to pinpoint causal variants using GWAS is dependent on understanding the dynamic epigenomic and epistatic context of each associated locus. Being the best studied skeletal locus, GDF5 associates with many diseases and has a complex cis-regulatory architecture. We interrogate GDF5 regulatory interactions and model disease variants in vitro and in vivo. For all regulatory regions we see that local epigenetic activation/repression impacts patterns of joint-specific expression and disease risk. By modeling the most cited risk variant in mice we found that it had no impact on expression, joint morphology, or disease. Yet, we identified significant epistatic expression interactions between this risk variant and others lying within regulatory regions subject to repression or activation. These findings are important lessons on how regulatory interactions and local epistasis work in the etiology of disease risk, and that assessment of individual variants of high GWAS significance need not alone be considered causal.
Collapse
Affiliation(s)
- Clarissa R Coveney
- Department of Human Evolutionary Biology, Harvard University, Cambridge, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - David Maridas
- Department of Developmental Biology, Harvard School of Dental Medicine, Boston, MA, USA
| | - Hao Chen
- Department of Developmental Biology, Beckman Center B300, Stanford University School of Medicine, Stanford, California 94305, USA
| | | | - Zun Liu
- Department of Human Evolutionary Biology, Harvard University, Cambridge, MA, USA
| | - Evelyn Jagoda
- Department of Human Evolutionary Biology, Harvard University, Cambridge, MA, USA
| | - Siddharth Yarlagadda
- Department of Human Evolutionary Biology, Harvard University, Cambridge, MA, USA
| | - Mohammadreza Movahhedi
- Department of Orthopaedic Surgery and Sports Medicine, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Benedikt Proffen
- Department of Orthopaedic Surgery and Sports Medicine, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Vicki Rosen
- Department of Developmental Biology, Harvard School of Dental Medicine, Boston, MA, USA
| | - Ata M Kiapour
- Department of Orthopaedic Surgery and Sports Medicine, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Terence D Capellini
- Department of Human Evolutionary Biology, Harvard University, Cambridge, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| |
Collapse
|
8
|
Horbaly H, Hubbe M. Systemic versus local patterns of limb joint articular morphology inferred from relative distances from morphological centroid. Anat Rec (Hoboken) 2024; 307:3519-3528. [PMID: 38817037 DOI: 10.1002/ar.25506] [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: 02/26/2024] [Revised: 05/02/2024] [Accepted: 05/04/2024] [Indexed: 06/01/2024]
Abstract
Joint morphogenesis is a complex process known to require the interaction of developmental cascades and mechanical loading, yet many details of this interaction are incompletely understood. While prior work has established populational patterns of joint morphological (co)variance, exploring how these patterns manifest within the individual provides information on the deployment of morphogenic processes as either systemic or local influences on joint shape. To better identify the patterns of variance-generating morphogenic processes, this study investigates the degree to which individual joint shapes deviate from population averages systematically across the body. Using three-dimensional landmark data from 200 adult skeletons, we ranked individuals based on their distances from morphological centroids for eight major joints. Spearman correlations assessed associations between ranks across various articular pairings, testing hypotheses regarding systemic versus localized variance. Results reveal low coordination between deviations observed in conarticular surfaces, functional analogs, and same-bone surfaces; however strong associations exist between antimeres, suggesting the left-right deployment of variance-generating morphogenic patterns is highly consistent. These results support a model of localized rather than systemic processes driving variation in joint shape. While more remains to be elucidated about the specifics of articular surface morphogenesis, these findings advance our understanding of the systems of variance generation at play during development and growth of our definitive joint morphology.
Collapse
Affiliation(s)
- Haley Horbaly
- Department of Health and Human Performance, Congdon School of Health Sciences, High Point University, High Point, North Carolina, USA
- Department of Physician Assistant Studies, Congdon School of Health Sciences, High Point University, High Point, North Carolina, USA
| | - Mark Hubbe
- Department of Anthropology, The Ohio State University, Columbus, Ohio, USA
| |
Collapse
|
9
|
Abdelrazek I, Knaus A, Javanmardi B, Krawitz P, Horn D, Abdalla E, Kumar S. Acromesomelic Dysplasia With Homozygosity for a Likely Pathogenic BMPR1B Variant: Postaxial Polydactyly as a Novel Clinical Finding. Mol Genet Genomic Med 2024; 12:e70023. [PMID: 39441036 PMCID: PMC11497645 DOI: 10.1002/mgg3.70023] [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: 03/18/2024] [Revised: 09/05/2024] [Accepted: 09/25/2024] [Indexed: 10/25/2024] Open
Abstract
BACKGROUND Acromesomelic chondrodysplasias are a rare subgroup of the clinically and genetically heterogeneous osteochondrodysplasias that are characterised by abnormalities in the limb development and short stature. Here, we report a 2-year-old boy, offspring of consanguineous parents, with acromesomelic dysplasia and postaxial polydactyly in which exome sequencing identified a novel homozygous missense variant in BMPR1B. The patient showed skeletal malformation of both hands and feet that included complex brachydactyly with the thumbs most severely affected, postaxial polydactyly of both hands, shortened toes as well as a bilateral hypoplasia of the fibula. METHODS Whole trio exome sequencing was conducted to identify potential genetic variants in the patient. RESULTS The analysis identified the biallelic variant NM_001203.3:c.821A > G;p.(Gln274Arg) in BMPR1B, a gene encoding bone morphogenetic protein receptor 1B. CONCLUSION The skeletal phenotype can be brought in line with the phenotypes of previously reported cases of BMPR1B-associated chondrodysplasias. However, the postaxial polydactyly described here is a novel clinical finding in a BMPR1B-related case; notably, it has previously been reported in other acromesomelic dysplasia cases caused by homozygous pathogenic variants in GDF5-a gene which encodes for growth differentiation factor 5, a high-affinity ligand to BMPR1B.
Collapse
Affiliation(s)
- Ibrahim M. Abdelrazek
- Department of Human GeneticsMedical Research Institute, Alexandria UniversityAlexandriaEgypt
| | - Alexej Knaus
- Institute for Genomic Statistics and Bioinformatics, Medical FacultyUniversity of Bonn, University Hospital BonnBonnGermany
| | - Behnam Javanmardi
- Institute for Genomic Statistics and Bioinformatics, Medical FacultyUniversity of Bonn, University Hospital BonnBonnGermany
| | - Peter M. Krawitz
- Institute for Genomic Statistics and Bioinformatics, Medical FacultyUniversity of Bonn, University Hospital BonnBonnGermany
| | - Denise Horn
- Institute of Medical Genetics and Human GeneticsCharité‐Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt‐Universität Zu BerlinBerlinGermany
| | - Ebtesam M. Abdalla
- Department of Human GeneticsMedical Research Institute, Alexandria UniversityAlexandriaEgypt
| | - Sheetal Kumar
- Institute of Human Genetics, Medical FacultyUniversity of Bonn, University Hospital BonnBonnGermany
| |
Collapse
|
10
|
Li Q, Jiang S, Lei K, Han H, Chen Y, Lin W, Xiong Q, Qi X, Gan X, Sheng R, Wang Y, Zhang Y, Ma J, Li T, Lin S, Zhou C, Chen D, Yuan Q. Metabolic rewiring during bone development underlies tRNA m7G-associated primordial dwarfism. J Clin Invest 2024; 134:e177220. [PMID: 39255038 PMCID: PMC11473147 DOI: 10.1172/jci177220] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2023] [Accepted: 08/26/2024] [Indexed: 09/12/2024] Open
Abstract
Translation of mRNA to protein is tightly regulated by transfer RNAs (tRNAs), which are subject to various chemical modifications that maintain structure, stability, and function. Deficiency of tRNA N7-methylguanosine (m7G) modification in patients causes a type of primordial dwarfism, but the underlying mechanism remains unknown. Here we report that the loss of m7G rewires cellular metabolism, leading to the pathogenesis of primordial dwarfism. Conditional deletion of the catalytic enzyme Mettl1 or missense mutation of the scaffold protein Wdr4 severely impaired endochondral bone formation and bone mass accrual. Mechanistically, Mettl1 knockout decreased abundance of m7G-modified tRNAs and inhibited translation of mRNAs relating to cytoskeleton and Rho GTPase signaling. Meanwhile, Mettl1 knockout enhanced cellular energy metabolism despite incompetent proliferation and osteogenic commitment. Further exploration revealed that impairment of Rho GTPase signaling upregulated the level of branched-chain amino acid transaminase 1 (BCAT1) that rewired cell metabolism and restricted intracellular α-ketoglutarate (αKG). Supplementation of αKG ameliorated the skeletal defect of Mettl1-deficient mice. In addition to the selective translation of metabolism-related mRNAs, we further revealed that Mettl1 knockout globally regulated translation via integrated stress response (ISR) and mammalian target of rapamycin complex 1 (mTORC1) signaling. Restoring translation by targeting either ISR or mTORC1 aggravated bone defects of Mettl1-deficient mice. Overall, our study unveils a critical role of m7G tRNA modification in bone development by regulation of cellular metabolism and indicates suspension of translation initiation as a quality control mechanism in response to tRNA dysregulation.
Collapse
Affiliation(s)
- Qiwen Li
- State Key Laboratory of Oral Diseases and National Center for Stomatology and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Shuang Jiang
- State Key Laboratory of Oral Diseases and National Center for Stomatology and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Kexin Lei
- State Key Laboratory of Oral Diseases and National Center for Stomatology and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Hui Han
- Center for Translational Medicine, Precision Medicine Institute, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Yaqian Chen
- State Key Laboratory of Oral Diseases and National Center for Stomatology and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Weimin Lin
- State Key Laboratory of Oral Diseases and National Center for Stomatology and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Qiuchan Xiong
- State Key Laboratory of Oral Diseases and National Center for Stomatology and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Xingying Qi
- State Key Laboratory of Oral Diseases and National Center for Stomatology and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Xinyan Gan
- State Key Laboratory of Oral Diseases and National Center for Stomatology and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Rui Sheng
- State Key Laboratory of Oral Diseases and National Center for Stomatology and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Yuan Wang
- State Key Laboratory of Oral Diseases and National Center for Stomatology and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Yarong Zhang
- State Key Laboratory of Oral Diseases and National Center for Stomatology and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Jieyi Ma
- Center for Translational Medicine, Precision Medicine Institute, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Tao Li
- West China–Washington Mitochondria and Metabolism Center and Department of Anesthesiology, West China Hospital, Sichuan University, Chengdu, China
| | - Shuibin Lin
- Center for Translational Medicine, Precision Medicine Institute, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Chenchen Zhou
- State Key Laboratory of Oral Diseases and National Center for Stomatology and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Demeng Chen
- Otorhinolaryngology Hospital, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Quan Yuan
- State Key Laboratory of Oral Diseases and National Center for Stomatology and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| |
Collapse
|
11
|
Zhao X, Liu S, Yang Z, Li Y. Molecular mechanisms and genetic factors contributing to the developmental dysplasia of the hip. Front Genet 2024; 15:1413500. [PMID: 39156961 PMCID: PMC11327038 DOI: 10.3389/fgene.2024.1413500] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2024] [Accepted: 07/16/2024] [Indexed: 08/20/2024] Open
Abstract
The most prevalent hip disease in neonates is developmental dysplasia of the hip (DDH). A timely and accurate diagnosis is required to provide the most effective treatment for pediatric patients with DDH. Heredity and gene variation have been the subject of increased attention and research worldwide as one of the factors contributing to the pathogenesis of DDH. Genome-wide association studies (GWAS), genome-wide linkage analyses (GWLA), and exome sequencing (ES) have identified variants in numerous genes and single-nucleotide polymorphisms (SNPs) as being associated with susceptibility to DDH in sporadic and DDH family patients. Furthermore, the DDH phenotype can be observed in animal models that exhibit susceptibility genes or loci, including variants in CX3CR1, KANSL1, and GDF5. The dentification of noncoding RNAs and de novo gene variants in patients with DDH-related syndrome has enhanced our understanding of the genes implicated in DDH. This article reviews the most recent molecular mechanisms and genetic factors that contribute to DDH.
Collapse
Affiliation(s)
- Xiaoming Zhao
- Department of Pediatric Orthopaedics, Shenyang Orthopaedic Hospital, Shenyang, China
| | - Shuai Liu
- College of Police Dog Technology, Criminal Investigation Police University of China, Shenyang, China
| | - Zhonghua Yang
- Department of Pediatric Surgery, Shengjing Hospital of China Medical University, Shenyang, China
- Key Laboratory of Health Ministry for Congenital Malformation, Shengjing Hospital of China Medical University, Shenyang, China
| | - Yong Li
- Department of Pediatric Surgery, Shengjing Hospital of China Medical University, Shenyang, China
| |
Collapse
|
12
|
Wang C, Fan M, Heo SJ, Adams SM, Li T, Liu Y, Li Q, Loebel C, Alisafaei F, Burdick JA, Lu XL, Birk DE, Mauck RL, Han L. Structure-Mechanics Principles and Mechanobiology of Fibrocartilage Pericellular Matrix: A Pivotal Role of Type V Collagen. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.26.600498. [PMID: 38979323 PMCID: PMC11230444 DOI: 10.1101/2024.06.26.600498] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 07/10/2024]
Abstract
The pericellular matrix (PCM) is the immediate microniche surrounding resident cells in various tissue types, regulating matrix turnover, cell-matrix cross-talk and disease initiation. This study elucidated the structure-mechanical properties and mechanobiological functions of the PCM in fibrocartilage, a family of connective tissues that sustain complex tensile and compressive loads in vivo. Studying the murine meniscus as the model tissue, we showed that fibrocartilage PCM contains thinner, random collagen fibrillar networks that entrap proteoglycans, a structure distinct from the densely packed, highly aligned collagen fibers in the bulk extracellular matrix (ECM). In comparison to the ECM, the PCM has a lower modulus and greater isotropy, but similar relative viscoelastic properties. In Col5a1 +/- menisci, the reduction of collagen V, a minor collagen localized in the PCM, resulted in aberrant fibril thickening with increased heterogeneity. Consequently, the PCM exhibited a reduced modulus, loss of isotropy and faster viscoelastic relaxation. This disrupted PCM contributes to perturbed mechanotransduction of resident meniscal cells, as illustrated by reduced intracellular calcium signaling, as well as upregulated biosynthesis of lysyl oxidase and tenascin C. When cultured in vitro, Col5a1 +/- meniscal cells synthesized a weakened nascent PCM, which had inferior properties towards protecting resident cells against applied tensile stretch. These findings underscore the PCM as a distinctive microstructure that governs fibrocartilage mechanobiology, and highlight the pivotal role of collagen V in PCM function. Targeting the PCM or its molecular constituents holds promise for enhancing not only meniscus regeneration and osteoarthritis intervention, but also addressing diseases across various fibrocartilaginous tissues.
Collapse
Affiliation(s)
- Chao Wang
- School of Biomedical Engineering, Science and Health Systems, Drexel University, Philadelphia, PA 19104, United States
| | - Mingyue Fan
- School of Biomedical Engineering, Science and Health Systems, Drexel University, Philadelphia, PA 19104, United States
| | - Su-Jin Heo
- McKay Orthopaedic Research Laboratory, Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, United States
| | - Sheila M. Adams
- Department of Molecular Pharmacology and Physiology, Morsani School of Medicine, University of South Florida, Tampa, FL 33612, United States
| | - Thomas Li
- School of Biomedical Engineering, Science and Health Systems, Drexel University, Philadelphia, PA 19104, United States
| | - Yuchen Liu
- School of Biomedical Engineering, Science and Health Systems, Drexel University, Philadelphia, PA 19104, United States
| | - Qing Li
- School of Biomedical Engineering, Science and Health Systems, Drexel University, Philadelphia, PA 19104, United States
| | - Claudia Loebel
- Department of Materials Science and Engineering, University of Michigan, Ann Arbor, MI 48109, United States
| | - Farid Alisafaei
- Department of Mechanical and Industrial Engineering, New Jersey Institute of Technology, Newark, NJ 07102, United States
| | - Jason A. Burdick
- BioFrontiers Institute and Department of Chemical and Biological Engineering, University of Colorado, Boulder, CO 80309, United States
| | - X. Lucas Lu
- Department of Mechanical Engineering, University of Delaware, Newark, DE 19716, United States
| | - David E. Birk
- Department of Molecular Pharmacology and Physiology, Morsani School of Medicine, University of South Florida, Tampa, FL 33612, United States
| | - Robert L. Mauck
- McKay Orthopaedic Research Laboratory, Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, United States
- Translational Musculoskeletal Research Center, Corporal Michael J. Crescenz Veterans Administration Medical Center, Philadelphia, PA 19104, United States
| | - Lin Han
- School of Biomedical Engineering, Science and Health Systems, Drexel University, Philadelphia, PA 19104, United States
| |
Collapse
|
13
|
Leung AOW, Poon ACH, Wang X, Feng C, Chen P, Zheng Z, To MK, Chan WCW, Cheung M, Chan D. Suppression of apoptosis impairs phalangeal joint formation in the pathogenesis of brachydactyly type A1. Nat Commun 2024; 15:2229. [PMID: 38472182 PMCID: PMC10933404 DOI: 10.1038/s41467-024-45053-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Accepted: 01/12/2024] [Indexed: 03/14/2024] Open
Abstract
Apoptosis occurs during development when a separation of tissues is needed. Synovial joint formation is initiated at the presumptive site (interzone) within a cartilage anlagen, with changes in cellular differentiation leading to cavitation and tissue separation. Apoptosis has been detected in phalangeal joints during development, but its role and regulation have not been defined. Here, we use a mouse model of brachydactyly type A1 (BDA1) with an IhhE95K mutation, to show that a missing middle phalangeal bone is due to the failure of the developing joint to cavitate, associated with reduced apoptosis, and a joint is not formed. We showed an intricate relationship between IHH and interacting partners, CDON and GAS1, in the interzone that regulates apoptosis. We propose a model in which CDON/GAS1 may act as dependence receptors in this context. Normally, the IHH level is low at the center of the interzone, enabling the "ligand-free" CDON/GAS1 to activate cell death for cavitation. In BDA1, a high concentration of IHH suppresses apoptosis. Our findings provided new insights into the role of IHH and CDON in joint formation, with relevance to hedgehog signaling in developmental biology and diseases.
Collapse
Affiliation(s)
- Adrian On Wah Leung
- School of Biomedical Sciences, The University of Hong Kong, Pokfulam, Hong Kong, China
| | - Andrew Chung Hin Poon
- School of Biomedical Sciences, The University of Hong Kong, Pokfulam, Hong Kong, China
| | - Xue Wang
- School of Biomedical Sciences, The University of Hong Kong, Pokfulam, Hong Kong, China
| | - Chen Feng
- School of Biomedical Sciences, The University of Hong Kong, Pokfulam, Hong Kong, China
- Hebei Orthopedic Clinical Research Center, The Third Hospital of Hebei Medical University, 050051, Shijiazhuang, Hebei, China
| | - Peikai Chen
- School of Biomedical Sciences, The University of Hong Kong, Pokfulam, Hong Kong, China
- Department of Orthopaedics Surgery and Traumatology, The University of Hong Kong -Shenzhen Hospital (HKU-SZH), Shenzhen, China
| | - Zhengfan Zheng
- School of Biomedical Sciences, The University of Hong Kong, Pokfulam, Hong Kong, China
| | - Michael KaiTsun To
- Department of Orthopaedics Surgery and Traumatology, The University of Hong Kong -Shenzhen Hospital (HKU-SZH), Shenzhen, China
- Department of Orthopaedics and Traumatology, The University of Hong Kong, Pokfulam, Hong Kong, China
| | - Wilson Cheuk Wing Chan
- School of Biomedical Sciences, The University of Hong Kong, Pokfulam, Hong Kong, China.
- Department of Orthopaedics Surgery and Traumatology, The University of Hong Kong -Shenzhen Hospital (HKU-SZH), Shenzhen, China.
| | - Martin Cheung
- School of Biomedical Sciences, The University of Hong Kong, Pokfulam, Hong Kong, China
| | - Danny Chan
- School of Biomedical Sciences, The University of Hong Kong, Pokfulam, Hong Kong, China.
| |
Collapse
|
14
|
Wei D, Su Y, Leung PCK, Li Y, Chen ZJ. Roles of bone morphogenetic proteins in endometrial remodeling during the human menstrual cycle and pregnancy. Hum Reprod Update 2024; 30:215-237. [PMID: 38037193 DOI: 10.1093/humupd/dmad031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Revised: 10/17/2023] [Indexed: 12/02/2023] Open
Abstract
BACKGROUND During the human menstrual cycle and pregnancy, the endometrium undergoes a series of dynamic remodeling processes to adapt to physiological changes. Insufficient endometrial remodeling, characterized by inadequate endometrial proliferation, decidualization and spiral artery remodeling, is associated with infertility, endometriosis, dysfunctional uterine bleeding, and pregnancy-related complications such as preeclampsia and miscarriage. Bone morphogenetic proteins (BMPs), a subset of the transforming growth factor-β (TGF-β) superfamily, are multifunctional cytokines that regulate diverse cellular activities, such as differentiation, proliferation, apoptosis, and extracellular matrix synthesis, are now understood as integral to multiple reproductive processes in women. Investigations using human biological samples have shown that BMPs are essential for regulating human endometrial remodeling processes, including endometrial proliferation and decidualization. OBJECTIVE AND RATIONALE This review summarizes our current knowledge on the known pathophysiological roles of BMPs and their underlying molecular mechanisms in regulating human endometrial proliferation and decidualization, with the goal of promoting the development of innovative strategies for diagnosing, treating and preventing infertility and adverse pregnancy complications associated with dysregulated human endometrial remodeling. SEARCH METHODS A literature search for original articles published up to June 2023 was conducted in the PubMed, MEDLINE, and Google Scholar databases, identifying studies on the roles of BMPs in endometrial remodeling during the human menstrual cycle and pregnancy. Articles identified were restricted to English language full-text papers. OUTCOMES BMP ligands and receptors and their transduction molecules are expressed in the endometrium and at the maternal-fetal interface. Along with emerging technologies such as tissue microarrays, 3D organoid cultures and advanced single-cell transcriptomics, and given the clinical availability of recombinant human proteins and ongoing pharmaceutical development, it is now clear that BMPs exert multiple roles in regulating human endometrial remodeling and that these biomolecules (and their receptors) can be targeted for diagnostic and therapeutic purposes. Moreover, dysregulation of these ligands, their receptors, or signaling determinants can impact endometrial remodeling, contributing to infertility or pregnancy-related complications (e.g. preeclampsia and miscarriage). WIDER IMPLICATIONS Although further clinical trials are needed, recent advancements in the development of recombinant BMP ligands, synthetic BMP inhibitors, receptor antagonists, BMP ligand sequestration tools, and gene therapies have underscored the BMPs as candidate diagnostic biomarkers and positioned the BMP signaling pathway as a promising therapeutic target for addressing infertility and pregnancy complications related to dysregulated human endometrial remodeling.
Collapse
Affiliation(s)
- Daimin Wei
- Center for Reproductive Medicine, Shandong University, Jinan, Shandong, China
- Medical Integration and Practice Center, Shandong University, Jinan, Shandong, China
- State Key Laboratory of Reproductive Medicine and Offspring Health, Shandong University, Jinan, Shandong, China
| | - Yaxin Su
- Center for Reproductive Medicine, Shandong University, Jinan, Shandong, China
| | - Peter C K Leung
- Department of Obstetrics and Gynaecology, BC Children's Hospital Research Institute, University of British Columbia, Vancouver, BC, Canada
| | - Yan Li
- Center for Reproductive Medicine, Shandong University, Jinan, Shandong, China
- Medical Integration and Practice Center, Shandong University, Jinan, Shandong, China
- State Key Laboratory of Reproductive Medicine and Offspring Health, Shandong University, Jinan, Shandong, China
| | - Zi-Jiang Chen
- Center for Reproductive Medicine, Shandong University, Jinan, Shandong, China
- State Key Laboratory of Reproductive Medicine and Offspring Health, Shandong University, Jinan, Shandong, China
- Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Shanghai, China
- Research Unit of Gametogenesis and Health of ART-Offspring, Chinese Academy of Medical Sciences (No.2021RU001), Jinan, Shandong, China
| |
Collapse
|
15
|
Fox SC, Waskiewicz AJ. Transforming growth factor beta signaling and craniofacial development: modeling human diseases in zebrafish. Front Cell Dev Biol 2024; 12:1338070. [PMID: 38385025 PMCID: PMC10879340 DOI: 10.3389/fcell.2024.1338070] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Accepted: 01/18/2024] [Indexed: 02/23/2024] Open
Abstract
Humans and other jawed vertebrates rely heavily on their craniofacial skeleton for eating, breathing, and communicating. As such, it is vital that the elements of the craniofacial skeleton develop properly during embryogenesis to ensure a high quality of life and evolutionary fitness. Indeed, craniofacial abnormalities, including cleft palate and craniosynostosis, represent some of the most common congenital abnormalities in newborns. Like many other organ systems, the development of the craniofacial skeleton is complex, relying on specification and migration of the neural crest, patterning of the pharyngeal arches, and morphogenesis of each skeletal element into its final form. These processes must be carefully coordinated and integrated. One way this is achieved is through the spatial and temporal deployment of cell signaling pathways. Recent studies conducted using the zebrafish model underscore the importance of the Transforming Growth Factor Beta (TGF-β) and Bone Morphogenetic Protein (BMP) pathways in craniofacial development. Although both pathways contain similar components, each pathway results in unique outcomes on a cellular level. In this review, we will cover studies conducted using zebrafish that show the necessity of these pathways in each stage of craniofacial development, starting with the induction of the neural crest, and ending with the morphogenesis of craniofacial elements. We will also cover human skeletal and craniofacial diseases and malformations caused by mutations in the components of these pathways (e.g., cleft palate, craniosynostosis, etc.) and the potential utility of zebrafish in studying the etiology of these diseases. We will also briefly cover the utility of the zebrafish model in joint development and biology and discuss the role of TGF-β/BMP signaling in these processes and the diseases that result from aberrancies in these pathways, including osteoarthritis and multiple synostoses syndrome. Overall, this review will demonstrate the critical roles of TGF-β/BMP signaling in craniofacial development and show the utility of the zebrafish model in development and disease.
Collapse
|
16
|
Iacobescu GL, Iacobescu L, Popa MIG, Covache-Busuioc RA, Corlatescu AD, Cirstoiu C. Genomic Determinants of Knee Joint Biomechanics: An Exploration into the Molecular Basis of Locomotor Function, a Narrative Review. Curr Issues Mol Biol 2024; 46:1237-1258. [PMID: 38392197 PMCID: PMC10888373 DOI: 10.3390/cimb46020079] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2023] [Revised: 01/20/2024] [Accepted: 01/25/2024] [Indexed: 02/24/2024] Open
Abstract
In recent years, the nexus between genetics and biomechanics has garnered significant attention, elucidating the role of genomic determinants in shaping the biomechanical attributes of human joints, specifically the knee. This review seeks to provide a comprehensive exploration of the molecular basis underlying knee joint locomotor function. Leveraging advancements in genomic sequencing, we identified specific genetic markers and polymorphisms tied to key biomechanical features of the knee, such as ligament elasticity, meniscal resilience, and cartilage health. Particular attention was devoted to collagen genes like COL1A1 and COL5A1 and their influence on ligamentous strength and injury susceptibility. We further investigated the genetic underpinnings of knee osteoarthritis onset and progression, as well as the potential for personalized rehabilitation strategies tailored to an individual's genetic profile. We reviewed the impact of genetic factors on knee biomechanics and highlighted the importance of personalized orthopedic interventions. The results hold significant implications for injury prevention, treatment optimization, and the future of regenerative medicine, targeting not only knee joint health but joint health in general.
Collapse
Affiliation(s)
- Georgian-Longin Iacobescu
- Orthopaedics and Traumatology Department, "Carol Davila" University of Medicine and Pharmacy, 050474 Bucharest, Romania
- University Emergency Hospital, 050098 Bucharest, Romania
| | - Loredana Iacobescu
- Orthopaedics and Traumatology Department, "Carol Davila" University of Medicine and Pharmacy, 050474 Bucharest, Romania
- University Emergency Hospital, 050098 Bucharest, Romania
| | - Mihnea Ioan Gabriel Popa
- Orthopaedics and Traumatology Department, "Carol Davila" University of Medicine and Pharmacy, 050474 Bucharest, Romania
- University Emergency Hospital, 050098 Bucharest, Romania
| | - Razvan-Adrian Covache-Busuioc
- Orthopaedics and Traumatology Department, "Carol Davila" University of Medicine and Pharmacy, 050474 Bucharest, Romania
| | - Antonio-Daniel Corlatescu
- Orthopaedics and Traumatology Department, "Carol Davila" University of Medicine and Pharmacy, 050474 Bucharest, Romania
| | - Catalin Cirstoiu
- Orthopaedics and Traumatology Department, "Carol Davila" University of Medicine and Pharmacy, 050474 Bucharest, Romania
- University Emergency Hospital, 050098 Bucharest, Romania
| |
Collapse
|
17
|
Wu M, Wu S, Chen W, Li YP. The roles and regulatory mechanisms of TGF-β and BMP signaling in bone and cartilage development, homeostasis and disease. Cell Res 2024; 34:101-123. [PMID: 38267638 PMCID: PMC10837209 DOI: 10.1038/s41422-023-00918-9] [Citation(s) in RCA: 122] [Impact Index Per Article: 122.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2023] [Accepted: 12/15/2023] [Indexed: 01/26/2024] Open
Abstract
Transforming growth factor-βs (TGF-βs) and bone morphometric proteins (BMPs) belong to the TGF-β superfamily and perform essential functions during osteoblast and chondrocyte lineage commitment and differentiation, skeletal development, and homeostasis. TGF-βs and BMPs transduce signals through SMAD-dependent and -independent pathways; specifically, they recruit different receptor heterotetramers and R-Smad complexes, resulting in unique biological readouts. BMPs promote osteogenesis, osteoclastogenesis, and chondrogenesis at all differentiation stages, while TGF-βs play different roles in a stage-dependent manner. BMPs and TGF-β have opposite functions in articular cartilage homeostasis. Moreover, TGF-β has a specific role in maintaining the osteocyte network. The precise activation of BMP and TGF-β signaling requires regulatory machinery at multiple levels, including latency control in the matrix, extracellular antagonists, ubiquitination and phosphorylation in the cytoplasm, nucleus-cytoplasm transportation, and transcriptional co-regulation in the nuclei. This review weaves the background information with the latest advances in the signaling facilitated by TGF-βs and BMPs, and the advanced understanding of their diverse physiological functions and regulations. This review also summarizes the human diseases and mouse models associated with disordered TGF-β and BMP signaling. A more precise understanding of the BMP and TGF-β signaling could facilitate the development of bona fide clinical applications in treating bone and cartilage disorders.
Collapse
Affiliation(s)
- Mengrui Wu
- Department of Cell and Developmental Biology, College of Life Sciences, Zhejiang University, Hangzhou, Zhejiang, China.
| | - Shali Wu
- Department of Cell and Developmental Biology, College of Life Sciences, Zhejiang University, Hangzhou, Zhejiang, China
| | - Wei Chen
- Division in Cellular and Molecular Medicine, Department of Pathology and Laboratory Medicine, Tulane University School of Medicine, Tulane University, New Orleans, LA, USA
| | - Yi-Ping Li
- Division in Cellular and Molecular Medicine, Department of Pathology and Laboratory Medicine, Tulane University School of Medicine, Tulane University, New Orleans, LA, USA.
| |
Collapse
|
18
|
Grall E, Feregrino C, Fischer S, De Courten A, Sacher F, Hiscock TW, Tschopp P. Self-organized BMP signaling dynamics underlie the development and evolution of digit segmentation patterns in birds and mammals. Proc Natl Acad Sci U S A 2024; 121:e2304470121. [PMID: 38175868 PMCID: PMC10786279 DOI: 10.1073/pnas.2304470121] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2023] [Accepted: 11/03/2023] [Indexed: 01/06/2024] Open
Abstract
Repeating patterns of synovial joints are a highly conserved feature of articulated digits, with variations in joint number and location resulting in diverse digit morphologies and limb functions across the tetrapod clade. During the development of the amniote limb, joints form iteratively within the growing digit ray, as a population of distal progenitors alternately specifies joint and phalanx cell fates to segment the digit into distinct elements. While numerous molecular pathways have been implicated in this fate choice, it remains unclear how they give rise to a repeating pattern. Here, using single-cell RNA sequencing and spatial gene expression profiling, we investigate the transcriptional dynamics of interphalangeal joint specification in vivo. Combined with mathematical modeling, we predict that interactions within the BMP signaling pathway-between the ligand GDF5, the inhibitor NOGGIN, and the intracellular effector pSMAD-result in a self-organizing Turing system that forms periodic joint patterns. Our model is able to recapitulate the spatiotemporal gene expression dynamics observed in vivo, as well as phenocopy digit malformations caused by BMP pathway perturbations. By contrasting in silico simulations with in vivo morphometrics of two morphologically distinct digits, we show how changes in signaling parameters and growth dynamics can result in variations in the size and number of phalanges. Together, our results reveal a self-organizing mechanism that underpins amniote digit segmentation and its evolvability and, more broadly, illustrate how Turing systems based on a single molecular pathway may generate complex repetitive patterns in a wide variety of organisms.
Collapse
Affiliation(s)
- Emmanuelle Grall
- Zoology, Department of Environmental Sciences, University of Basel, Basel4051, Switzerland
| | - Christian Feregrino
- Zoology, Department of Environmental Sciences, University of Basel, Basel4051, Switzerland
| | - Sabrina Fischer
- Zoology, Department of Environmental Sciences, University of Basel, Basel4051, Switzerland
| | - Aline De Courten
- Zoology, Department of Environmental Sciences, University of Basel, Basel4051, Switzerland
| | - Fabio Sacher
- Zoology, Department of Environmental Sciences, University of Basel, Basel4051, Switzerland
| | - Tom W. Hiscock
- Institute of Medical Sciences, University of Aberdeen, AberdeenAB25 2ZD, Scotland, United Kingdom
| | - Patrick Tschopp
- Zoology, Department of Environmental Sciences, University of Basel, Basel4051, Switzerland
| |
Collapse
|
19
|
Shikatani EA, Wang T, Dingwell LS, White-Dzuro C, Momen A, Husain M. GDF5 deficiency prevents cardiac rupture following acute myocardial infarction in mice. Cardiovasc Pathol 2024; 68:107581. [PMID: 37838075 DOI: 10.1016/j.carpath.2023.107581] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/18/2023] [Revised: 09/19/2023] [Accepted: 10/09/2023] [Indexed: 10/16/2023] Open
Abstract
BACKGROUND We previously showed that growth differentiation factor 5 (GDF5) limits infarct expansion post-myocardial infarction (MI). We now examine the acute post-MI role of GDF5 in cardiac rupture. METHODS AND RESULTS Following permanent ligation of the left anterior descending artery, GDF5 deficiency (i.e., GDF5 knockout mice) reduced the incidence of cardiac rupture (4/24 vs. 17/24; P < .05), and improved survival over 28-d compared to wild-type (WT) mice (79% vs. 25%; P < .0001). Moreover, at 3-d post-MI, GDF5-deficient mice manifest: (a) reduced heart weight/body weight ratio (P < .0001) without differences in infarct size or cardiomyocyte size; (b) increased infarct zone expression of Col1a1 (P < .05) and Col3a1 (P < .01), suggesting increased myocardial fibrosis; and (c) reduced aortic and left ventricular peak systolic pressures (P ≤ .05), suggesting reduced afterload. Despite dysregulated inflammatory markers and reduced circulating monocytes in GDF5-deficient mice at 3-d post-MI, reciprocal bone marrow transplantation (BMT) failed to implicate GDF5 in BM-derived cells, suggesting the involvement of tissue-resident GDF5 expression in cardiac rupture. CONCLUSIONS Loss of GDF5 reduces cardiac rupture post-MI with increased myocardial fibrosis and lower afterload, albeit at the cost of chronic adverse remodeling.
Collapse
Affiliation(s)
- Eric A Shikatani
- Toronto General Hospital Research Institute, University Health Network, Toronto, Ontario, Canada; Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada
| | - Tao Wang
- Toronto General Hospital Research Institute, University Health Network, Toronto, Ontario, Canada; Heart and Stroke Richard Lewar Centre of Excellence, Ted Rogers Centre for Heart Research, and Peter Munk Cardiac Centre, Toronto, Ontario, Canada; Department of Physiology, University of Toronto, Toronto, Ontario, Canada
| | - Luke S Dingwell
- Toronto General Hospital Research Institute, University Health Network, Toronto, Ontario, Canada; Heart and Stroke Richard Lewar Centre of Excellence, Ted Rogers Centre for Heart Research, and Peter Munk Cardiac Centre, Toronto, Ontario, Canada; Institute of Medical Science, University of Toronto, Toronto, Ontario, Canada
| | - Colin White-Dzuro
- Toronto General Hospital Research Institute, University Health Network, Toronto, Ontario, Canada
| | - Abdul Momen
- Toronto General Hospital Research Institute, University Health Network, Toronto, Ontario, Canada
| | - Mansoor Husain
- Toronto General Hospital Research Institute, University Health Network, Toronto, Ontario, Canada; Heart and Stroke Richard Lewar Centre of Excellence, Ted Rogers Centre for Heart Research, and Peter Munk Cardiac Centre, Toronto, Ontario, Canada; Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada; Department of Physiology, University of Toronto, Toronto, Ontario, Canada; Institute of Medical Science, University of Toronto, Toronto, Ontario, Canada; Department of Medicine, University of Toronto, Toronto, Ontario, Canada.
| |
Collapse
|
20
|
Franco RAG, McKenna E, Shajib MS, Guillesser B, Robey PG, Crawford RW, Doran MR, Futrega K. Microtissue Culture Provides Clarity on the Relative Chondrogenic and Hypertrophic Response of Bone-Marrow-Derived Stromal Cells to TGF-β1, BMP-2, and GDF-5. Cells 2023; 13:37. [PMID: 38201241 PMCID: PMC10778331 DOI: 10.3390/cells13010037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Revised: 11/27/2023] [Accepted: 12/04/2023] [Indexed: 01/12/2024] Open
Abstract
Chondrogenic induction of bone-marrow-derived stromal cells (BMSCs) is typically accomplished with medium supplemented with growth factors (GF) from the transforming growth factor-beta (TGF-β)/bone morphogenetic factor (BMP) superfamily. In a previous study, we demonstrated that brief (1-3 days) stimulation with TGF-β1 was sufficient to drive chondrogenesis and hypertrophy using small-diameter microtissues generated from 5000 BMSC each. This biology is obfuscated in typical large-diameter pellet cultures, which suffer radial heterogeneity. Here, we investigated if brief stimulation (2 days) of BMSC microtissues with BMP-2 (100 ng/mL) or growth/differentiation factor (GDF-5, 100 ng/mL) was also sufficient to induce chondrogenic differentiation, in a manner comparable to TGF-β1 (10 ng/mL). Like TGF-β1, BMP-2 and GDF-5 are reported to stimulate chondrogenic differentiation of BMSCs, but the effects of transient or brief use in culture have not been explored. Hypertrophy is an unwanted outcome in BMSC chondrogenic differentiation that renders engineered tissues unsuitable for use in clinical cartilage repair. Using three BMSC donors, we observed that all GFs facilitated chondrogenesis, although the efficiency and the necessary duration of stimulation differed. Microtissues treated with 2 days or 14 days of TGF-β1 were both superior at producing extracellular matrix and expression of chondrogenic gene markers compared to BMP-2 and GDF-5 with the same exposure times. Hypertrophic markers increased proportionally with chondrogenic differentiation, suggesting that these processes are intertwined for all three GFs. The rapid action, or "temporal potency", of these GFs to induce BMSC chondrogenesis was found to be as follows: TGF-β1 > BMP-2 > GDF-5. Whether briefly or continuously supplied in culture, TGF-β1 was the most potent GF for inducing chondrogenesis in BMSCs.
Collapse
Affiliation(s)
- Rose Ann G. Franco
- Centre for Biomedical Technologies (CBT), School of Mechanical, Medical and Process Engineering, Faculty of Engineering, Queensland University of Technology (QUT), Brisbane, QLD 4000, Australia
- Translational Research Institute (TRI), Brisbane, QLD 4102, Australia
| | - Eamonn McKenna
- Centre for Biomedical Technologies (CBT), School of Mechanical, Medical and Process Engineering, Faculty of Engineering, Queensland University of Technology (QUT), Brisbane, QLD 4000, Australia
- Translational Research Institute (TRI), Brisbane, QLD 4102, Australia
| | - Md. Shafiullah Shajib
- Centre for Biomedical Technologies (CBT), School of Mechanical, Medical and Process Engineering, Faculty of Engineering, Queensland University of Technology (QUT), Brisbane, QLD 4000, Australia
- Translational Research Institute (TRI), Brisbane, QLD 4102, Australia
- School of Biomedical Science, Faculty of Health, Queensland University of Technology (QUT), Brisbane, QLD 4000, Australia
| | - Bianca Guillesser
- Centre for Biomedical Technologies (CBT), School of Mechanical, Medical and Process Engineering, Faculty of Engineering, Queensland University of Technology (QUT), Brisbane, QLD 4000, Australia
- Translational Research Institute (TRI), Brisbane, QLD 4102, Australia
- School of Biomedical Science, Faculty of Health, Queensland University of Technology (QUT), Brisbane, QLD 4000, Australia
| | - Pamela G. Robey
- Skeletal Biology Section, National Institute of Dental and Craniofacial Research (NIDCR), National Institutes of Health (NIH), Department of Health and Human Services, Bethesda, MD 20892, USA
| | - Ross W. Crawford
- Centre for Biomedical Technologies (CBT), School of Mechanical, Medical and Process Engineering, Faculty of Engineering, Queensland University of Technology (QUT), Brisbane, QLD 4000, Australia
| | - Michael R. Doran
- Centre for Biomedical Technologies (CBT), School of Mechanical, Medical and Process Engineering, Faculty of Engineering, Queensland University of Technology (QUT), Brisbane, QLD 4000, Australia
- Translational Research Institute (TRI), Brisbane, QLD 4102, Australia
- School of Biomedical Science, Faculty of Health, Queensland University of Technology (QUT), Brisbane, QLD 4000, Australia
- Skeletal Biology Section, National Institute of Dental and Craniofacial Research (NIDCR), National Institutes of Health (NIH), Department of Health and Human Services, Bethesda, MD 20892, USA
- Mater Research Institute—University of Queensland (UQ), Translational Research Institute (TRI), Brisbane, QLD 4102, Australia
| | - Kathryn Futrega
- Centre for Biomedical Technologies (CBT), School of Mechanical, Medical and Process Engineering, Faculty of Engineering, Queensland University of Technology (QUT), Brisbane, QLD 4000, Australia
- Skeletal Biology Section, National Institute of Dental and Craniofacial Research (NIDCR), National Institutes of Health (NIH), Department of Health and Human Services, Bethesda, MD 20892, USA
| |
Collapse
|
21
|
Flore L, Francalacci P, Massidda M, Robledo R, Calò CM. Influence of Different Evolutive Forces on GDF5 Gene Variability. Genes (Basel) 2023; 14:1895. [PMID: 37895244 PMCID: PMC10606091 DOI: 10.3390/genes14101895] [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: 09/11/2023] [Accepted: 09/26/2023] [Indexed: 10/29/2023] Open
Abstract
The GDF5 gene is involved in the development of skeletal elements, synovial joint formation, tendons, ligaments, and cartilage. Several polymorphisms are present within the gene, and two of them, rs143384 and 143383, were reported to be correlated with osteoarticular disease or muscle flexibility. The aim of this research is to verify if the worldwide distribution of the rs143384 polymorphism among human populations was shaped by selective pressure, or if it was the result of random genetic drift events. Ninety-four individuals of both the male and female sexes, 18-28 years old, from Sardinia were analyzed. We observed the following genotype frequencies: 28.72% of AA homozygotes, 13.83% of GG homozygotes, and 57.45% of AG heterozygotes. The allele frequencies were 0.574 for allele A and 0.426 for allele G. The relationships between the populations were verified via Multidimensional Scaling (MDS). Our data show (i) a clear heterogeneity within the African populations; (ii) a strong differentiation between the African populations and the other populations; and that (iii) the Sardinian population is placed within the European cluster. To reveal possible traces of selective pressure, the Population Branch Statistic (PBS) was calculated; both the rs143384 and 143383 SNPs have low PBS values, suggesting that there are no signals of selective pressure in those areas of the gene.
Collapse
Affiliation(s)
- Laura Flore
- Department of Life and Environment Sciences, University of Cagliari, 09042 Cagliari, Italy; (L.F.); (P.F.); (C.M.C.)
| | - Paolo Francalacci
- Department of Life and Environment Sciences, University of Cagliari, 09042 Cagliari, Italy; (L.F.); (P.F.); (C.M.C.)
| | - Myosotis Massidda
- Department of Medical Sciences and Public Health, University of Cagliari, 09042 Cagliari, Italy;
| | - Renato Robledo
- Department of Biomedical Sciences, University of Cagliari, 09042 Cagliari, Italy
| | - Carla Maria Calò
- Department of Life and Environment Sciences, University of Cagliari, 09042 Cagliari, Italy; (L.F.); (P.F.); (C.M.C.)
| |
Collapse
|
22
|
Caxaria S, Kouvatsos N, Eldridge SE, Alvarez‐Fallas M, Thorup A, Cici D, Barawi A, Arshed A, Strachan D, Carletti G, Huang X, Bharde S, Deniz M, Wilson J, Thomas BL, Pitzalis C, Cantatore FP, Sayilekshmy M, Sikandar S, Luyten FP, Pap T, Sherwood JC, Day AJ, Dell'Accio F. Disease modification and symptom relief in osteoarthritis using a mutated GCP-2/CXCL6 chemokine. EMBO Mol Med 2022; 15:e16218. [PMID: 36507558 PMCID: PMC9832835 DOI: 10.15252/emmm.202216218] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Revised: 11/09/2022] [Accepted: 11/11/2022] [Indexed: 12/14/2022] Open
Abstract
We showed that the chemokine receptor C-X-C Motif Chemokine Receptor 2 (CXCR2) is essential for cartilage homeostasis. Here, we reveal that the CXCR2 ligand granulocyte chemotactic protein 2 (GCP-2) was expressed, during embryonic development, within the prospective permanent articular cartilage, but not in the epiphyseal cartilage destined to be replaced by bone. GCP-2 expression was retained in adult articular cartilage. GCP-2 loss-of-function inhibited extracellular matrix production. GCP-2 treatment promoted chondrogenesis in vitro and in human cartilage organoids implanted in nude mice in vivo. To exploit the chondrogenic activity of GCP-2, we disrupted its chemotactic activity, by mutagenizing a glycosaminoglycan binding sequence, which we hypothesized to be required for the formation of a GCP-2 haptotactic gradient on endothelia. This mutated version (GCP-2-T) had reduced capacity to induce transendothelial migration in vitro and in vivo, without affecting downstream receptor signaling through AKT, and chondrogenic activity. Intra-articular adenoviral overexpression of GCP-2-T, but not wild-type GCP-2, reduced pain and cartilage loss in instability-induced osteoarthritis in mice. We suggest that GCP-2-T may be used for disease modification in osteoarthritis.
Collapse
Affiliation(s)
- Sara Caxaria
- William Harvey Research Institute, Barts and the London School of Medicine and DentistryQueen Mary University of LondonLondonUK
| | - Nikolaos Kouvatsos
- Wellcome Centre for Cell‐Matrix Research, Manchester Academic Health Science CentreUniversity of ManchesterManchesterUK
| | - Suzanne E Eldridge
- William Harvey Research Institute, Barts and the London School of Medicine and DentistryQueen Mary University of LondonLondonUK
| | - Mario Alvarez‐Fallas
- William Harvey Research Institute, Barts and the London School of Medicine and DentistryQueen Mary University of LondonLondonUK
| | - Anne‐Sophie Thorup
- William Harvey Research Institute, Barts and the London School of Medicine and DentistryQueen Mary University of LondonLondonUK
| | - Daniela Cici
- Rheumatology Clinic, Department of Medical and Surgical SciencesUniversity of FoggiaFoggiaItaly
| | - Aida Barawi
- William Harvey Research Institute, Barts and the London School of Medicine and DentistryQueen Mary University of LondonLondonUK
| | - Ammaarah Arshed
- William Harvey Research Institute, Barts and the London School of Medicine and DentistryQueen Mary University of LondonLondonUK
| | - Danielle Strachan
- William Harvey Research Institute, Barts and the London School of Medicine and DentistryQueen Mary University of LondonLondonUK
| | - Giulia Carletti
- William Harvey Research Institute, Barts and the London School of Medicine and DentistryQueen Mary University of LondonLondonUK
| | - Xinying Huang
- William Harvey Research Institute, Barts and the London School of Medicine and DentistryQueen Mary University of LondonLondonUK
| | - Sabah Bharde
- William Harvey Research Institute, Barts and the London School of Medicine and DentistryQueen Mary University of LondonLondonUK
| | - Melody Deniz
- William Harvey Research Institute, Barts and the London School of Medicine and DentistryQueen Mary University of LondonLondonUK
| | - Jacob Wilson
- William Harvey Research Institute, Barts and the London School of Medicine and DentistryQueen Mary University of LondonLondonUK
| | - Bethan L Thomas
- William Harvey Research Institute, Barts and the London School of Medicine and DentistryQueen Mary University of LondonLondonUK
| | - Costantino Pitzalis
- William Harvey Research Institute, Barts and the London School of Medicine and DentistryQueen Mary University of LondonLondonUK
| | | | - Manasi Sayilekshmy
- William Harvey Research Institute, Barts and the London School of Medicine and DentistryQueen Mary University of LondonLondonUK
| | - Shafaq Sikandar
- William Harvey Research Institute, Barts and the London School of Medicine and DentistryQueen Mary University of LondonLondonUK
| | - Frank P Luyten
- Department of Development and Regeneration, Skeletal Biology and Engineering Research CenterKU LeuvenLeuvenBelgium
| | - Thomas Pap
- Institute of Musculoskeletal MedicineUniversity Hospital MünsterMünsterGermany
| | - Joanna C Sherwood
- Institute of Musculoskeletal MedicineUniversity Hospital MünsterMünsterGermany
| | - Anthony J Day
- Wellcome Centre for Cell‐Matrix Research, Manchester Academic Health Science CentreUniversity of ManchesterManchesterUK,Lydia Becker Institute of Immunology and Inflammation, Faculty of Biology, Medicine & Health, Manchester Academic Health Science CentreUniversity of ManchesterManchesterUK
| | - Francesco Dell'Accio
- William Harvey Research Institute, Barts and the London School of Medicine and DentistryQueen Mary University of LondonLondonUK
| |
Collapse
|
23
|
Zhang CH, Gao Y, Hung HH, Zhuo Z, Grodzinsky AJ, Lassar AB. Creb5 coordinates synovial joint formation with the genesis of articular cartilage. Nat Commun 2022; 13:7295. [PMID: 36435829 PMCID: PMC9701237 DOI: 10.1038/s41467-022-35010-0] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2020] [Accepted: 11/15/2022] [Indexed: 11/28/2022] Open
Abstract
While prior work has established that articular cartilage arises from Prg4-expressing perichondrial cells, it is not clear how this process is specifically restricted to the perichondrium of synovial joints. We document that the transcription factor Creb5 is necessary to initiate the expression of signaling molecules that both direct the formation of synovial joints and guide perichondrial tissue to form articular cartilage instead of bone. Creb5 promotes the generation of articular chondrocytes from perichondrial precursors in part by inducing expression of signaling molecules that block a Wnt5a autoregulatory loop in the perichondrium. Postnatal deletion of Creb5 in the articular cartilage leads to loss of both flat superficial zone articular chondrocytes coupled with a loss of both Prg4 and Wif1 expression in the articular cartilage; and a non-cell autonomous up-regulation of Ctgf. Our findings indicate that Creb5 promotes joint formation and the subsequent development of articular chondrocytes by driving the expression of signaling molecules that both specify the joint interzone and simultaneously inhibit a Wnt5a positive-feedback loop in the perichondrium.
Collapse
Affiliation(s)
- Cheng-Hai Zhang
- Department of Biological Chemistry and Molecular Pharmacology, Blavatnik Institute at Harvard Medical School, 240 Longwood Ave., Boston, MA, 02115, USA.
| | - Yao Gao
- Department of Biological Chemistry and Molecular Pharmacology, Blavatnik Institute at Harvard Medical School, 240 Longwood Ave., Boston, MA, 02115, USA
| | - Han-Hwa Hung
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
| | - Zhu Zhuo
- Bioinformatics Core, Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA, 02115, USA
| | - Alan J Grodzinsky
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
| | - Andrew B Lassar
- Department of Biological Chemistry and Molecular Pharmacology, Blavatnik Institute at Harvard Medical School, 240 Longwood Ave., Boston, MA, 02115, USA.
| |
Collapse
|
24
|
The molecular genetics of human appendicular skeleton. Mol Genet Genomics 2022; 297:1195-1214. [PMID: 35907958 DOI: 10.1007/s00438-022-01930-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2021] [Accepted: 07/09/2022] [Indexed: 10/16/2022]
Abstract
Disorders that result from de-arrangement of growth, development and/or differentiation of the appendages (limbs and digit) are collectively called as inherited abnormalities of human appendicular skeleton. The bones of appendicular skeleton have central role in locomotion and movement. The different types of appendicular skeletal abnormalities are well described in the report of "Nosology and Classification of Genetic skeletal disorders: 2019 Revision". In the current article, we intend to present the embryology, developmental pathways, disorders and the molecular genetics of the appendicular skeletal malformations. We mainly focused on the polydactyly, syndactyly, brachydactyly, split-hand-foot malformation and clubfoot disorders. To our knowledge, only nine genes of polydactyly, five genes of split-hand-foot malformation, nine genes for syndactyly, eight genes for brachydactyly and only single gene for clubfoot have been identified to be involved in disease pathophysiology. The current molecular genetic data will help life sciences researchers working on the rare skeletal disorders. Moreover, the aim of present systematic review is to gather the published knowledge on molecular genetics of appendicular skeleton, which would help in genetic counseling and molecular diagnosis.
Collapse
|
25
|
Le Dréau G. BuMPing Into Neurogenesis: How the Canonical BMP Pathway Regulates Neural Stem Cell Divisions Throughout Space and Time. Front Neurosci 2022; 15:819990. [PMID: 35153664 PMCID: PMC8829030 DOI: 10.3389/fnins.2021.819990] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Accepted: 12/28/2021] [Indexed: 11/13/2022] Open
Abstract
Bone morphogenetic proteins (BMPs) are secreted factors that contribute to many aspects of the formation of the vertebrate central nervous system (CNS), from the initial shaping of the neural primordium to the maturation of the brain and spinal cord. In particular, the canonical (SMAD1/5/8-dependent) BMP pathway appears to play a key role during neurogenesis, its activity dictating neural stem cell fate decisions and thereby regulating the growth and homeostasis of the CNS. In this mini-review, I summarize accumulating evidence demonstrating how the canonical BMP activity promotes the amplification and/or maintenance of neural stem cells at different times and in diverse regions of the vertebrate CNS, and highlight findings suggesting that this function is evolutionarily conserved.
Collapse
|
26
|
Anantha J, Goulding SR, Tuboly E, O'Mahony AG, Moloney GM, Lomansey G, McCarthy CM, Collins LM, Sullivan AM, O'Keeffe GW. NME1 Protects Against Neurotoxin-, α-Synuclein- and LRRK2-Induced Neurite Degeneration in Cell Models of Parkinson's Disease. Mol Neurobiol 2022; 59:61-76. [PMID: 34623600 PMCID: PMC8786793 DOI: 10.1007/s12035-021-02569-6] [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: 12/11/2020] [Accepted: 09/14/2021] [Indexed: 11/26/2022]
Abstract
Parkinson's disease (PD) is a neurodegenerative disease characterised by the progressive degeneration of midbrain dopaminergic neurons, coupled with the intracellular accumulation of α-synuclein. Axonal degeneration is a central part of the pathology of PD. While the majority of PD cases are sporadic, some are genetic; the G2019S mutation in leucine-rich repeat kinase 2 (LRRK2) is the most common genetic form. The application of neurotrophic factors to protect dopaminergic neurons is a proposed experimental therapy. One such neurotrophic factor is growth differentiation factor (GDF)5. GDF5 is a dopaminergic neurotrophic factor that has been shown to upregulate the expression of a protein called nucleoside diphosphate kinase A (NME1). However, whether NME1 is neuroprotective in cell models of axonal degeneration of relevance to PD is unknown. Here we show that treatment with NME1 can promote neurite growth in SH-SY5Y cells, and in cultured dopaminergic neurons treated with the neurotoxin 6-hydroxydopamine (6-OHDA). Similar effects of NME1 were found in SH-SY5Y cells and dopaminergic neurons overexpressing human wild-type α-synuclein, and in stable SH-SY5Y cell lines carrying the G2019S LRRK2 mutation. We found that the effects of NME1 require the RORα/ROR2 receptors. Furthermore, increased NF-κB-dependent transcription was partially required for the neurite growth-promoting effects of NME1. Finally, a combined bioinformatics and biochemical analysis of the mitochondrial oxygen consumption rate revealed that NME1 enhanced mitochondrial function, which is known to be impaired in PD. These data show that recombinant NME1 is worthy of further study as a potential therapeutic agent for axonal protection in PD.
Collapse
Affiliation(s)
- Jayanth Anantha
- Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland
| | - Susan R Goulding
- Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland
| | - Eszter Tuboly
- Department of Pharmacology and Therapeutics, University College Cork, Cork, Ireland
| | - Adam G O'Mahony
- Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland
| | - Gerard M Moloney
- Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland
- APC Microbiome Ireland, University College Cork, Cork, Ireland
| | - Gareth Lomansey
- Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland
| | - Cathal M McCarthy
- Department of Pharmacology and Therapeutics, University College Cork, Cork, Ireland
| | - Louise M Collins
- Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland
- Department of Physiology, University College Cork, Cork, Ireland
- Parkinson's Disease Research Cluster (PDRC), University College Cork, Cork, Ireland
| | - Aideen M Sullivan
- Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland.
- APC Microbiome Ireland, University College Cork, Cork, Ireland.
- Parkinson's Disease Research Cluster (PDRC), University College Cork, Cork, Ireland.
| | - Gerard W O'Keeffe
- Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland.
- APC Microbiome Ireland, University College Cork, Cork, Ireland.
- Parkinson's Disease Research Cluster (PDRC), University College Cork, Cork, Ireland.
| |
Collapse
|
27
|
Towler OW, Shore EM. BMP signaling and skeletal development in fibrodysplasia ossificans progressiva (FOP). Dev Dyn 2022; 251:164-177. [PMID: 34133058 PMCID: PMC9068236 DOI: 10.1002/dvdy.387] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2021] [Revised: 05/07/2021] [Accepted: 05/20/2021] [Indexed: 01/03/2023] Open
Abstract
Fibrodysplasia ossificans progressiva (FOP) is an ultra-rare genetic disease caused by increased BMP pathway signaling due to mutation of ACVR1, a bone morphogenetic protein (BMP) type 1 receptor. The primary clinical manifestation of FOP is extra-skeletal bone formation (heterotopic ossification) within soft connective tissues. However, the underlying ACVR1 mutation additionally alters skeletal bone development and nearly all people born with FOP have bilateral malformation of the great toes as well as other skeletal malformations at diverse anatomic sites. The specific mechanisms through which ACVR1 mutations and altered BMP pathway signaling in FOP influence skeletal bone formation during development remain to be elucidated; however, recent investigations are providing a clearer understanding of the molecular and developmental processes associated with ACVR1-regulated skeletal formation.
Collapse
Affiliation(s)
- Oscar Will Towler
- The Center for Research in FOP & Related Disorders, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA,Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Eileen M. Shore
- The Center for Research in FOP & Related Disorders, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA,Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA,Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| |
Collapse
|
28
|
Rubin S, Agrawal A, Stegmaier J, Krief S, Felsenthal N, Svorai J, Addadi Y, Villoutreix P, Stern T, Zelzer E. Application of 3D MAPs pipeline identifies the morphological sequence chondrocytes undergo and the regulatory role of GDF5 in this process. Nat Commun 2021; 12:5363. [PMID: 34508093 PMCID: PMC8433335 DOI: 10.1038/s41467-021-25714-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Accepted: 08/19/2021] [Indexed: 02/08/2023] Open
Abstract
The activity of epiphyseal growth plates, which drives long bone elongation, depends on extensive changes in chondrocyte size and shape during differentiation. Here, we develop a pipeline called 3D Morphometric Analysis for Phenotypic significance (3D MAPs), which combines light-sheet microscopy, segmentation algorithms and 3D morphometric analysis to characterize morphogenetic cellular behaviors while maintaining the spatial context of the growth plate. Using 3D MAPs, we create a 3D image database of hundreds of thousands of chondrocytes. Analysis reveals broad repertoire of morphological changes, growth strategies and cell organizations during differentiation. Moreover, identifying a reduction in Smad 1/5/9 activity together with multiple abnormalities in cell growth, shape and organization provides an explanation for the shortening of Gdf5 KO tibias. Overall, our findings provide insight into the morphological sequence that chondrocytes undergo during differentiation and highlight the ability of 3D MAPs to uncover cellular mechanisms that may regulate this process.
Collapse
Affiliation(s)
- Sarah Rubin
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot, Israel
| | - Ankit Agrawal
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot, Israel
| | - Johannes Stegmaier
- Institute of Imaging and Computer Vision, RWTH Aachen University, Aachen, Germany
- Institute for Automation and Applied Informatics, Karlsruhe Institute of Technology, Karlsruhe, Germany
| | - Sharon Krief
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot, Israel
| | - Neta Felsenthal
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot, Israel
| | - Jonathan Svorai
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot, Israel
| | - Yoseph Addadi
- Department of Life Science Core Facilities, Weizmann Institute of Science, Rehovot, Israel
| | - Paul Villoutreix
- LIS (UMR 7020), IBDM (UMR 7288), Turing Center For Living Systems, Aix-Marseille University, Marseille, France.
| | - Tomer Stern
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot, Israel.
| | - Elazar Zelzer
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot, Israel.
| |
Collapse
|
29
|
Wilkinson JM, Zeggini E. The Genetic Epidemiology of Joint Shape and the Development of Osteoarthritis. Calcif Tissue Int 2021; 109:257-276. [PMID: 32393986 PMCID: PMC8403114 DOI: 10.1007/s00223-020-00702-6] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/13/2020] [Accepted: 04/29/2020] [Indexed: 02/06/2023]
Abstract
Congruent, low-friction relative movement between the articulating elements of a synovial joint is an essential pre-requisite for sustained, efficient, function. Where disorders of joint formation or maintenance exist, mechanical overloading and osteoarthritis (OA) follow. The heritable component of OA accounts for ~ 50% of susceptible risk. Although almost 100 genetic risk loci for OA have now been identified, and the epidemiological relationship between joint development, joint shape and osteoarthritis is well established, we still have only a limited understanding of the contribution that genetic variation makes to joint shape and how this modulates OA risk. In this article, a brief overview of synovial joint development and its genetic regulation is followed by a review of current knowledge on the genetic epidemiology of established joint shape disorders and common shape variation. A summary of current genetic epidemiology of OA is also given, together with current evidence on the genetic overlap between shape variation and OA. Finally, the established genetic risk loci for both joint shape and osteoarthritis are discussed.
Collapse
Affiliation(s)
- J Mark Wilkinson
- Department of Oncology and Metabolism, University of Sheffield, Sheffield, UK.
| | - Eleftheria Zeggini
- Institute of Translational Genomics, Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg, Germany
| |
Collapse
|
30
|
Waldmann L, Leyhr J, Zhang H, Allalou A, Öhman-Mägi C, Haitina T. The role of Gdf5 in the development of the zebrafish fin endoskeleton. Dev Dyn 2021; 251:1535-1549. [PMID: 34242444 DOI: 10.1002/dvdy.399] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2021] [Revised: 06/22/2021] [Accepted: 07/08/2021] [Indexed: 11/12/2022] Open
Abstract
BACKGROUND The development of the vertebrate limb skeleton requires a complex interaction of multiple factors to facilitate the correct shaping and positioning of bones and joints. Growth and differentiation factor 5 (Gdf5) is involved in patterning appendicular skeletal elements including joints. Expression of gdf5 in zebrafish has been detected in fin mesenchyme condensations and segmentation zones as well as the jaw joint, however, little is known about the functional role of Gdf5 outside of Amniota. RESULTS We generated CRISPR/Cas9 knockout of gdf5 in zebrafish and analyzed the resulting phenotype at different developmental stages. Homozygous gdf5 mutant zebrafish displayed changes in segmentation of the endoskeletal disc and, as a consequence, loss of posterior radials in the pectoral fins. Mutant fish also displayed disorganization and reduced length of endoskeletal elements in the median fins, while joints and mineralization seemed unaffected. CONCLUSIONS Our study demonstrates the importance of Gdf5 in the development of the zebrafish pectoral and median fin endoskeleton and reveals that the severity of the effect increases from anterior to posterior elements. Our findings are consistent with phenotypes observed in the human and mouse appendicular skeleton in response to Gdf5 knockout, suggesting a broadly conserved role for Gdf5 in Osteichthyes.
Collapse
Affiliation(s)
- Laura Waldmann
- Department of Organismal Biology, Uppsala University, Uppsala, Sweden
| | - Jake Leyhr
- Department of Organismal Biology, Uppsala University, Uppsala, Sweden
| | - Hanqing Zhang
- Division of Visual Information and Interaction, Department of Information Technology, Uppsala University, Uppsala, Sweden.,Science for Life Laboratory BioImage Informatics Facility, Uppsala, Sweden
| | - Amin Allalou
- Division of Visual Information and Interaction, Department of Information Technology, Uppsala University, Uppsala, Sweden.,Science for Life Laboratory BioImage Informatics Facility, Uppsala, Sweden
| | - Caroline Öhman-Mägi
- Department of Materials Science and Engineering, Uppsala University, Uppsala, Sweden
| | - Tatjana Haitina
- Department of Organismal Biology, Uppsala University, Uppsala, Sweden
| |
Collapse
|
31
|
Muthuirulan P, Zhao D, Young M, Richard D, Liu Z, Emami A, Portilla G, Hosseinzadeh S, Cao J, Maridas D, Sedlak M, Menghini D, Cheng L, Li L, Ding X, Ding Y, Rosen V, Kiapour AM, Capellini TD. Joint disease-specificity at the regulatory base-pair level. Nat Commun 2021; 12:4161. [PMID: 34230488 PMCID: PMC8260791 DOI: 10.1038/s41467-021-24345-9] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Accepted: 06/16/2021] [Indexed: 02/06/2023] Open
Abstract
Given the pleiotropic nature of coding sequences and that many loci exhibit multiple disease associations, it is within non-coding sequence that disease-specificity likely exists. Here, we focus on joint disorders, finding among replicated loci, that GDF5 exhibits over twenty distinct associations, and we identify causal variants for two of its strongest associations, hip dysplasia and knee osteoarthritis. By mapping regulatory regions in joint chondrocytes, we pinpoint two variants (rs4911178; rs6060369), on the same risk haplotype, which reside in anatomical site-specific enhancers. We show that both variants have clinical relevance, impacting disease by altering morphology. By modeling each variant in humanized mice, we observe joint-specific response, correlating with GDF5 expression. Thus, we uncouple separate regulatory variants on a common risk haplotype that cause joint-specific disease. By broadening our perspective, we finally find that patterns of modularity at GDF5 are also found at over three-quarters of loci with multiple GWAS disease associations.
Collapse
Affiliation(s)
| | - Dewei Zhao
- Department of Orthopedics, Affiliated Zhongshan Hospital of Dalian University, Dalian, China
| | - Mariel Young
- Department of Human Evolutionary Biology, Harvard University, Cambridge, MA, USA
| | - Daniel Richard
- Department of Human Evolutionary Biology, Harvard University, Cambridge, MA, USA
| | - Zun Liu
- Department of Human Evolutionary Biology, Harvard University, Cambridge, MA, USA
| | - Alireza Emami
- Department of Orthopaedic Surgery, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Gabriela Portilla
- Department of Orthopaedic Surgery, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Shayan Hosseinzadeh
- Department of Orthopaedic Surgery, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Jiaxue Cao
- Department of Human Evolutionary Biology, Harvard University, Cambridge, MA, USA.,Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, China
| | - David Maridas
- Department of Developmental Biology, Harvard School of Dental Medicine, Boston, MA, USA
| | - Mary Sedlak
- Department of Human Evolutionary Biology, Harvard University, Cambridge, MA, USA
| | - Danilo Menghini
- Department of Orthopaedic Surgery, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Liangliang Cheng
- Department of Orthopedics, Affiliated Zhongshan Hospital of Dalian University, Dalian, China
| | - Lu Li
- Department of Orthopedics, Affiliated Zhongshan Hospital of Dalian University, Dalian, China
| | - Xinjia Ding
- Department of Surgery, the Second Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Yan Ding
- Department of Pediatrics, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Vicki Rosen
- Department of Developmental Biology, Harvard School of Dental Medicine, Boston, MA, USA
| | - Ata M Kiapour
- Department of Orthopaedic Surgery, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA.
| | - Terence D Capellini
- Department of Human Evolutionary Biology, Harvard University, Cambridge, MA, USA. .,Broad Institute of MIT and Harvard, Cambridge, MA, USA.
| |
Collapse
|
32
|
Controlling BMP growth factor bioavailability: The extracellular matrix as multi skilled platform. Cell Signal 2021; 85:110071. [PMID: 34217834 DOI: 10.1016/j.cellsig.2021.110071] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Revised: 06/26/2021] [Accepted: 06/29/2021] [Indexed: 01/23/2023]
Abstract
Bone morphogenetic proteins (BMPs) belong to the TGF-β superfamily of signaling ligands which comprise a family of pluripotent cytokines regulating a multitude of cellular events. Although BMPs were originally discovered as potent factors extractable from bone matrix that are capable to induce ectopic bone formation in soft tissues, their mode of action has been mostly studied as soluble ligands in absence of the physiologically relevant cellular microenvironment. This micro milieu is defined by supramolecular networks of extracellular matrix (ECM) proteins that specifically target BMP ligands, present them to their cellular receptors, and allow their controlled release. Here we focus on functional interactions and mechanisms that were described to control BMP bioavailability in a spatio-temporal manner within the respective tissue context. Structural disturbance of the ECM architecture due to mutations in ECM proteins leads to dysregulated BMP signaling as underlying cause for connective tissue disease pathways. We will provide an overview about current mechanistic concepts of how aberrant BMP signaling drives connective tissue destruction in inherited and chronic diseases.
Collapse
|
33
|
Whole-Genome Sequencing Identifies Two Novel Rare Mutations in BMP5 and BMP2 in Monozygotic Twins With Microtia. J Craniofac Surg 2021; 33:e212-e217. [PMID: 34183628 DOI: 10.1097/scs.0000000000007689] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
ABSTRACT Microtia is a rare congenital anomaly of the ear; it is regulated by both genetic and environmental factors. However, the mechanisms underlying its pathogenesis are unknown. In this study, the genomes of 2-year-old twin sisters with right microtia were sequenced using human genome-wide sequencing, an approach useful for identifying mutations in genes responsible for congenital microtia. The phenotypes of the twin sisters included congenital microtia on the right side, abnormal auricle shape in the right external ear, a peanut shape for the residual ear, and complete atresia of the right external auditory canal. In the twin sisters, we identified a previously unknown mutation in BMP5(exon4:c.833- 4C>G), as well as a new mutation (exon2:c.G332T:p.S111I) in BMP2, both of which were confirmed using polymerase chain reaction-based amplification of the corresponding genome regions, followed by first-generation sequencing. The exon4:c.833-4C>G mutation in human BMP5 may be the main cause of microtia in the twin sisters. A pathogenic mutation in human BMP2 (exon2:c.G332T:p.S111I) may be responsible for the facial deformity in the twin sisters. Thus, our study demonstrates the potential of genome-wide sequencing for identifying novel mutations associated with microtia on the whole-genome scale and extends the mutation spectrum of BMP5. Additionally, our data suggest that BMP2 is another pathogenic gene associated with microtia.
Collapse
|
34
|
Sun L, Cao Y, Kong Q, Huang X, Yu Z, Sun D, Ren W, Yang G, Xu S. Over-expression of the bottlenose dolphin Hoxd13 gene in zebrafish provides new insights into the cetacean flipper formation. Genomics 2021; 113:2925-2933. [PMID: 34166750 DOI: 10.1016/j.ygeno.2021.06.028] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Revised: 04/28/2021] [Accepted: 06/19/2021] [Indexed: 11/24/2022]
Abstract
Cetaceans have evolved elongated soft-tissue flipper with digits made of hyperphalangy. Cetaceans were found to have 2-3 more alanine residues in Hoxd13 than other mammals, which were suggested to be related to their flipper. However, how Hoxd13 regulates other genes and induces hyperphalangy in cetaceans remain poorly understood. Here, we overexpressed the bottlenose dolphin Hoxd13 in zebrafish (Danio rerio). Combined with transcriptome data and evolutionary analyses, our results revealed that the Wingless/Integrated (Wnt) and Hedgehog signaling pathways and multiple genes might regulate hyperphalangy development in cetaceans. Meanwhile, the Notch and mitogen-activated protein kinase (Mapk) signaling pathways and Fibroblast growth factor receptor 1 (Fgfr1) are probably correlated with interdigital tissues retained in the cetacean flipper. In conclusion, this is the first study to use a transgenic zebrafish to explore the molecular evolution of Hoxd13 in cetaceans, and it provides new insights into cetacean flipper formation.
Collapse
Affiliation(s)
- Linxia Sun
- Jiangsu Key Laboratory for Biodiversity and Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing 210023, China
| | - Yang Cao
- Jiangsu Key Laboratory for Biodiversity and Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing 210023, China
| | - Qian Kong
- Jiangsu Key Laboratory for Biodiversity and Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing 210023, China
| | - Xin Huang
- Jiangsu Key Laboratory for Biodiversity and Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing 210023, China
| | - Zhenpeng Yu
- Jiangsu Key Laboratory for Biodiversity and Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing 210023, China
| | - Di Sun
- Jiangsu Key Laboratory for Biodiversity and Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing 210023, China
| | - Wenhua Ren
- Jiangsu Key Laboratory for Biodiversity and Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing 210023, China
| | - Guang Yang
- Jiangsu Key Laboratory for Biodiversity and Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing 210023, China.
| | - Shixia Xu
- Jiangsu Key Laboratory for Biodiversity and Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing 210023, China.
| |
Collapse
|
35
|
Guo S, Cui L, Xiao C, Wang C, Zhu B, Liu X, Li Y, Liu X, Wang D, Li S. The Mechanisms and Functions of GDF-5 in Intervertebral Disc Degeneration. Orthop Surg 2021; 13:734-741. [PMID: 33817978 PMCID: PMC8126946 DOI: 10.1111/os.12942] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/21/2020] [Revised: 01/01/2021] [Accepted: 01/01/2021] [Indexed: 01/15/2023] Open
Abstract
Intervertebral disc degeneration (IDD) is widely recognized as the main cause of low back pain, which leads to disability in aging populations and induces great losses both socially and economically worldwide. Unfortunately, current treatments for IDD are aimed at relieving symptoms instead of preserving disc structure and function. Researchers are forged to find new promising biological therapeutics to stop, and even reverse, IVD degeneration. Recently, the injection of growth factors has been shown to be a promising biological therapy for IDD. A number of growth factors have been investigated to modulate the synthesis of the extracellular matrix (ECM) through a variety of pathogenetic biological mechanisms, including suppressing inflammatory process and down-regulating degrading enzymes. However, growth factors, including Transforming Growth Factor-β (TGF-β), Fibroblast Growth Factor (FGF), and Insulin-like Growth Factor-1 (IGF-1), may induce unwanted blood vessel in-growth, which accelerates the process of IDD. On the contrary, studies have demonstrated that injection of GDF-5 into the intervertebral disc of mice can effectively alleviate the degeneration of the intervertebral disc, which elicits their response via BMPRII and will not induce blood vessel in-growth. This finding suggests that GDF-5 is more suitable for use in IDD treatment compared with the three other growth factors. Substantial evidence has suggested that GDF-5 may maintain the structure and function of the intervertebral disc (IVD). GDF-5 plays an important role in IDD and is a very promising therapeutic agent for IDD. This review is focused on the mechanisms and functions of GDF-5 in IDD.
Collapse
Affiliation(s)
- Sheng Guo
- Spinal Surgery DepartmentHospital (T.C.M) Affiliated to Southwest Medical UniversityLuzhouChina
| | - Liqiang Cui
- Department of Spine SurgeryMianyang Orthopaedic HospitalMianyangChina
| | - Changming Xiao
- Spinal Surgery DepartmentHospital (T.C.M) Affiliated to Southwest Medical UniversityLuzhouChina
| | - Chenglong Wang
- Spinal Surgery DepartmentHospital (T.C.M) Affiliated to Southwest Medical UniversityLuzhouChina
| | - Bin Zhu
- Institute of Physical EducationSouthwest Medical UniversityLuzhouChina
| | - Xueli Liu
- Institute of Physical EducationSouthwest Medical UniversityLuzhouChina
| | - Yujie Li
- Institute of Physical EducationSouthwest Medical UniversityLuzhouChina
| | - Xinyue Liu
- Institute of Physical EducationSouthwest Medical UniversityLuzhouChina
| | - Dingxuan Wang
- Institute of Physical EducationSouthwest Medical UniversityLuzhouChina
| | - Sen Li
- Spinal Surgery DepartmentHospital (T.C.M) Affiliated to Southwest Medical UniversityLuzhouChina
| |
Collapse
|
36
|
Iwan A, Moskalewski S, Hyc A. Growth factor profile in calcified cartilage from the metaphysis of a calf costochondral junction, the site of initial bone formation. Biomed Rep 2021; 14:54. [PMID: 33884197 PMCID: PMC8056382 DOI: 10.3892/br.2021.1430] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2020] [Accepted: 03/11/2021] [Indexed: 12/25/2022] Open
Abstract
Endochondral bone formation is orchestrated by growth factors produced by chondrocytes and deposited in the cartilage matrix. Whilst some of these factors have been identified, the complete list and their relationship remains unknown. In the present study, the growth factors were isolated from non-calcified and calcified cartilage of costochondral junctions. Cartilage dissected from the ribs of 6-20-week-old calves was purchased from a local butcher within 24 h of the death of the animal. The isolation involved hyaluronidase digestion, guanidinium hydrochloride (GuHCl) extraction, HCl decalcification and GuHCl extraction of the decalcified matrix. Growth factors were purified by heparin chromatography and their quantities were estimated using ELISA. Decalcified cartilage was also used for protein sequence analysis (data are available via ProteomeXchange; ID, PXD021781). Bone morphogenetic protein-7 (BMP-7), growth/differentiation factor-5 (GDF-5) and NEL-like protein-1 (NELL-1), all known growth factors that stimulate bone formation, quantitatively accounted for the majority of the material obtained in all steps of isolation. Thus, cartilage serves as a store for growth factors. During initial bone formation septoclasts release osteoclastogenesis-stimulating factors deposited in non-calcified cartilage. Osteoclasts dissolve calcified cartilage and transport the released factors required for the stimulation of osteoprogenitor cells to deposit osteoid. High concentrations of BMP-7, GDF-5 and NELL-1 at the site of initial bone formation may suggest that their synergistic action favours osteogenesis.
Collapse
Affiliation(s)
- Anna Iwan
- Department of Histology and Embryology, Medical University of Warsaw, Warsaw PL02004, Poland
| | - Stanisław Moskalewski
- Department of Histology and Embryology, Medical University of Warsaw, Warsaw PL02004, Poland
| | - Anna Hyc
- Department of Histology and Embryology, Medical University of Warsaw, Warsaw PL02004, Poland
| |
Collapse
|
37
|
Sun K, Guo J, Yao X, Guo Z, Guo F. Growth differentiation factor 5 in cartilage and osteoarthritis: A possible therapeutic candidate. Cell Prolif 2021; 54:e12998. [PMID: 33522652 PMCID: PMC7941218 DOI: 10.1111/cpr.12998] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Revised: 01/01/2021] [Accepted: 01/05/2021] [Indexed: 12/11/2022] Open
Abstract
Growth differentiation factor 5 (GDF-5) is essential for cartilage development and homeostasis. The expression and function of GDF-5 are highly associated with the pathogenesis of osteoarthritis (OA). OA, characterized by progressive degeneration of joint, particularly in cartilage, causes severe social burden. However, there is no effective approach to reverse the progression of this disease. Over the past decades, extensive studies have demonstrated the protective effects of GDF-5 against cartilage degeneration and defects. Here, we summarize the current literature describing the role of GDF-5 in development of cartilage and joints, and the association between the GDF-5 gene polymorphisms and OA susceptibility. We also shed light on the protective effects of GDF-5 against OA in terms of direct GDF-5 supplementation and modulation of the GDF-5-related signalling. Finally, we discuss the current limitations in the application of GDF-5 for the clinical treatment of OA. This review provides a comprehensive insight into the role of GDF-5 in cartilage and emphasizes GDF-5 as a potential therapeutic candidate in OA.
Collapse
Affiliation(s)
- Kai Sun
- Department of OrthopedicsTongji Medical CollegeTongji HospitalHuazhong University of Science and TechnologyWuhanChina
| | - Jiachao Guo
- Department of OrthopedicsTongji Medical CollegeTongji HospitalHuazhong University of Science and TechnologyWuhanChina
| | - Xudong Yao
- Department of OrthopedicsTongji Medical CollegeTongji HospitalHuazhong University of Science and TechnologyWuhanChina
| | - Zhou Guo
- Department of OrthopedicsTongji Medical CollegeTongji HospitalHuazhong University of Science and TechnologyWuhanChina
| | - Fengjing Guo
- Department of OrthopedicsTongji Medical CollegeTongji HospitalHuazhong University of Science and TechnologyWuhanChina
| |
Collapse
|
38
|
Profile of David M. Kingsley. Proc Natl Acad Sci U S A 2021; 118:2025633118. [PMID: 33431699 DOI: 10.1073/pnas.2025633118] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
|
39
|
Sampath TK, Vukicevic S. Biology of bone morphogenetic protein in bone repair and regeneration: A role for autologous blood coagulum as carrier. Bone 2020; 141:115602. [PMID: 32841742 DOI: 10.1016/j.bone.2020.115602] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Revised: 08/10/2020] [Accepted: 08/11/2020] [Indexed: 12/12/2022]
Abstract
BMPs were purified from demineralized bone matrix based on their ability to induce new bone in vivo and they represent a large member of the TGF-β superfamily of proteins. BMPs serve as morphogenic signals for mesenchymal stem cell migration, proliferation and subsequently differentiation into cartilage and bone during embryonic development. A BMP when implanted with a collagenous carrier in a rat subcutaneous site is capable of inducing new bone by mimicking the cellular events of embryonic bone formation. Based on this biological principle, BMP2 and BMP7 containing collagenous matrix as carrier have been developed as bone graft substitutes for spine fusion and long bone fractures. Here, we describe a novel autologous bone graft substitute that contains BMP6 delivered within an autologous blood coagulum as carrier and summarize the biology of osteogenic BMPs in the context of bone repair and regeneration specifically the critical role that carrier plays to support osteogenesis.
Collapse
Affiliation(s)
- T Kuber Sampath
- perForm Biologics Inc., Holliston, MA 01746, United States of America.
| | - Slobodan Vukicevic
- Laboratory for Mineralized Tissues, Center for Translational and Clinical Research, University of Zagreb School of Medicine, 10000 Zagreb, Croatia
| |
Collapse
|
40
|
Frohlich J, Vinciguerra M. Candidate rejuvenating factor GDF11 and tissue fibrosis: friend or foe? GeroScience 2020; 42:1475-1498. [PMID: 33025411 PMCID: PMC7732895 DOI: 10.1007/s11357-020-00279-w] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2020] [Accepted: 09/22/2020] [Indexed: 12/13/2022] Open
Abstract
Growth differentiation factor 11 (GDF11 or bone morphogenetic protein 11, BMP11) belongs to the transforming growth factor-β superfamily and is closely related to other family member-myostatin (also known as GDF8). GDF11 was firstly identified in 2004 due to its ability to rejuvenate the function of multiple organs in old mice. However, in the past few years, the heralded rejuvenating effects of GDF11 have been seriously questioned by many studies that do not support the idea that restoring levels of GDF11 in aging improves overall organ structure and function. Moreover, with increasing controversies, several other studies described the involvement of GDF11 in fibrotic processes in various organ setups. This review paper focuses on the GDF11 and its pro- or anti-fibrotic actions in major organs and tissues, with the goal to summarize our knowledge on its emerging role in regulating the progression of fibrosis in different pathological conditions, and to guide upcoming research efforts.
Collapse
Affiliation(s)
- Jan Frohlich
- International Clinical Research Center, St. Anne's University Hospital, Pekarska 53, 656 91, Brno, Czech Republic
| | - Manlio Vinciguerra
- International Clinical Research Center, St. Anne's University Hospital, Pekarska 53, 656 91, Brno, Czech Republic.
- Institute for Liver and Digestive Health, Division of Medicine, University College London (UCL), London, UK.
| |
Collapse
|
41
|
Saito K, Chiba Y, Yamada A, Fukumoto S. Identification and function analysis of ameloblast differentiation-related molecules using mouse incisors. PEDIATRIC DENTAL JOURNAL 2020. [DOI: 10.1016/j.pdj.2020.08.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
|
42
|
Towler OW, Peck SH, Kaplan FS, Shore EM. Dysregulated BMP signaling through ACVR1 impairs digit joint development in fibrodysplasia ossificans progressiva (FOP). Dev Biol 2020; 470:136-146. [PMID: 33217406 DOI: 10.1016/j.ydbio.2020.11.004] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2019] [Revised: 11/01/2020] [Accepted: 11/10/2020] [Indexed: 12/30/2022]
Abstract
The development of joints in the mammalian skeleton depends on the precise regulation of multiple interacting signaling pathways including the bone morphogenetic protein (BMP) pathway, a key regulator of joint development, digit patterning, skeletal growth, and chondrogenesis. Mutations in the BMP receptor ACVR1 cause the rare genetic disease fibrodysplasia ossificans progressiva (FOP) in which extensive and progressive extra-skeletal bone forms in soft connective tissues after birth. These mutations, which enhance BMP-pSmad1/5 pathway activity to induce ectopic bone, also affect skeletal development. FOP can be diagnosed at birth by symmetric, characteristic malformations of the great toes (first digits) that are associated with decreased joint mobility, shortened digit length, and absent, fused, and/or malformed phalanges. To elucidate the role of ACVR1-mediated BMP signaling in digit skeletal development, we used an Acvr1R206H/+;Prrx1-Cre knock-in mouse model that mimics the first digit phenotype of human FOP. We have determined that the effects of increased Acvr1-mediated signaling by the Acvr1R206H mutation are not limited to the first digit but alter BMP signaling, Gdf5+ joint progenitor cell localization, and joint development in a manner that differently affects individual digits during embryogenesis. The Acvr1R206H mutation leads to delayed and disrupted joint specification and cleavage in the digits and alters the development of cartilage and endochondral ossification at sites of joint morphogenesis. These findings demonstrate an important role for ACVR1-mediated BMP signaling in the regulation of joint and skeletal formation, show a direct link between failure to restrict BMP signaling in the digit joint interzone and failure of joint cleavage at the presumptive interzone, and implicate impaired, digit-specific joint development as the proximal cause of digit malformation in FOP.
Collapse
Affiliation(s)
- O Will Towler
- Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, 3450 Hamilton Walk, 309A Stemmler Hall, Philadelphia, PA 19104, United States; Center for Research in FOP & Related Disorders, Perelman School of Medicine, University of Pennsylvania, 3450 Hamilton Walk, 309A Stemmler Hall, Philadelphia, PA 19104, United States
| | - Sun H Peck
- Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, 3450 Hamilton Walk, 309A Stemmler Hall, Philadelphia, PA 19104, United States; Center for Research in FOP & Related Disorders, Perelman School of Medicine, University of Pennsylvania, 3450 Hamilton Walk, 309A Stemmler Hall, Philadelphia, PA 19104, United States
| | - Frederick S Kaplan
- Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, 3450 Hamilton Walk, 309A Stemmler Hall, Philadelphia, PA 19104, United States; Department of Medicine, Perelman School of Medicine, University of Pennsylvania, 3400 Civic Center Boulevard, Perelman Center for Advanced Medicine, Philadelphia, PA 19104, United States; Center for Research in FOP & Related Disorders, Perelman School of Medicine, University of Pennsylvania, 3450 Hamilton Walk, 309A Stemmler Hall, Philadelphia, PA 19104, United States
| | - Eileen M Shore
- Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, 3450 Hamilton Walk, 309A Stemmler Hall, Philadelphia, PA 19104, United States; Department of Genetics, Perelman School of Medicine, University of Pennsylvania, 415 Curie Boulevard, Clinical Research Building, Philadelphia, PA 19104, United States; Center for Research in FOP & Related Disorders, Perelman School of Medicine, University of Pennsylvania, 3450 Hamilton Walk, 309A Stemmler Hall, Philadelphia, PA 19104, United States.
| |
Collapse
|
43
|
Anantha J, Goulding SR, Wyatt SL, Concannon RM, Collins LM, Sullivan AM, O'Keeffe GW. STRAP and NME1 Mediate the Neurite Growth-Promoting Effects of the Neurotrophic Factor GDF5. iScience 2020; 23:101457. [PMID: 32853992 PMCID: PMC7452236 DOI: 10.1016/j.isci.2020.101457] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2020] [Revised: 07/17/2020] [Accepted: 08/10/2020] [Indexed: 12/23/2022] Open
Abstract
Loss of midbrain dopaminergic (mDA) neurons and their axons is central to Parkinson's disease (PD). Growth differentiation factor (GDF)5 is a potential neurotrophic factor for PD therapy. However, the molecular mediators of its neurotrophic action are unknown. Our proteomics analysis shows that GDF5 increases the expression of serine threonine receptor-associated protein kinase (STRAP) and nucleoside diphosphate kinase (NME)1 in the SH-SY5Y neuronal cell line. GDF5 overexpression increased NME1 expression in adult rat brain in vivo. NME and STRAP mRNAs are expressed in developing and adult rodent midbrain. Expression of both STRAP and NME1 is necessary and sufficient for the promotion of neurite growth in SH-SY5Y cells by GDF5. NME1 treatment increased neurite growth in both SH-SY5Y cells and cultured mDA neurons. Expression patterns of NME and STRAP are altered in PD midbrain. NME1 and STRAP are thus key mediators of GDF5's neurotrophic effects, rationalizing their future study as therapeutic targets for PD.
Collapse
Affiliation(s)
- Jayanth Anantha
- Department of Anatomy & Neuroscience, University College Cork (UCC), Cork, Ireland
| | - Susan R. Goulding
- Department of Anatomy & Neuroscience, University College Cork (UCC), Cork, Ireland
- Department of Biological Sciences, Cork Institute of Technology, Cork, Ireland
| | - Sean L. Wyatt
- School of Biosciences, Cardiff University, Museum Avenue, Cardiff, UK
| | - Ruth M. Concannon
- Department of Anatomy & Neuroscience, University College Cork (UCC), Cork, Ireland
| | - Louise M. Collins
- Department of Anatomy & Neuroscience, University College Cork (UCC), Cork, Ireland
- Department of Physiology, UCC, Cork, Ireland
| | - Aideen M. Sullivan
- Department of Anatomy & Neuroscience, University College Cork (UCC), Cork, Ireland
- APC Microbiome Ireland, UCC, Cork, Ireland
- Cork Neuroscience Centre, UCC, Cork, Ireland
| | - Gerard W. O'Keeffe
- Department of Anatomy & Neuroscience, University College Cork (UCC), Cork, Ireland
- APC Microbiome Ireland, UCC, Cork, Ireland
- Cork Neuroscience Centre, UCC, Cork, Ireland
| |
Collapse
|
44
|
Lee H, Hwangbo H, Ji SY, Kim MY, Kim SY, Kim DH, Hong SH, Lee SJ, Assefa F, Kim GY, Park EK, Park JH, Lee BJ, Jeon YJ, Choi YH. Gamma Aminobutyric Acid-Enriched Fermented Oyster ( Crassostrea gigas) Increases the Length of the Growth Plate on the Proximal Tibia Bone in Sprague-Dawley Rats. Molecules 2020; 25:molecules25194375. [PMID: 32977643 PMCID: PMC7582314 DOI: 10.3390/molecules25194375] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2020] [Revised: 09/21/2020] [Accepted: 09/22/2020] [Indexed: 11/16/2022] Open
Abstract
Bone growth during childhood and puberty determines an adult’s final stature. Although several prior studies have reported that fermented oyster (FO) consisting of a high amount of gamma aminobutyric acid can be attributed to bone health, there is no research on the efficacy of FO on growth regulation and the proximal tibial growth plate. Therefore, in this study, we investigated the effect of FO oral administration on hepatic and serum growth regulator levels and the development of the proximal tibial growth plate in young Sprague-Dawley rats. Both oral administration of FO (FO 100, 100 mg/kg FO and FO 200, 200 mg/kg FO) and subcutaneous injection of recombinant human growth hormone (rhGH, 200 μg/kg of rhGH) for two weeks showed no toxicity. Circulating levels of growth hormone (GH) significantly increased in the FO 200 group. The expression and secretion of insulin-like growth factor-1 (IGF-1) and insulin-like growth factor binding protein-3 (IGFBP-3) were enhanced by FO administration. FO administration promoted the expression of bone morphogenic proteins IGF-1 and IGFBP-3 in the proximal tibial growth plate. This positive effect of FO resulted in incremental growth of the entire plate length by expanding the proliferating and hypertrophic zones in the proximal tibial growth plate. Collectively, our results suggested that oral administration of FO is beneficial for bone health, which may ultimately result in increased height.
Collapse
Affiliation(s)
- Hyesook Lee
- Anti-Aging Research Center, Dong-eui University, Busan 47340, Korea; (H.L.); (H.H.); (S.Y.J.); (M.Y.K.); (S.Y.K.); (D.H.K.); (S.H.H.)
- Department of Biochemistry, Dong-eui University College of Korean Medicine, Busan 47227, Korea
| | - Hyun Hwangbo
- Anti-Aging Research Center, Dong-eui University, Busan 47340, Korea; (H.L.); (H.H.); (S.Y.J.); (M.Y.K.); (S.Y.K.); (D.H.K.); (S.H.H.)
- Department of Molecular Biology, Pusan National University, Busan 46241, Korea
| | - Seon Yeong Ji
- Anti-Aging Research Center, Dong-eui University, Busan 47340, Korea; (H.L.); (H.H.); (S.Y.J.); (M.Y.K.); (S.Y.K.); (D.H.K.); (S.H.H.)
- Department of Biochemistry, Dong-eui University College of Korean Medicine, Busan 47227, Korea
| | - Min Yeong Kim
- Anti-Aging Research Center, Dong-eui University, Busan 47340, Korea; (H.L.); (H.H.); (S.Y.J.); (M.Y.K.); (S.Y.K.); (D.H.K.); (S.H.H.)
- Department of Biochemistry, Dong-eui University College of Korean Medicine, Busan 47227, Korea
| | - So Young Kim
- Anti-Aging Research Center, Dong-eui University, Busan 47340, Korea; (H.L.); (H.H.); (S.Y.J.); (M.Y.K.); (S.Y.K.); (D.H.K.); (S.H.H.)
- Department of Molecular Biology, Pusan National University, Busan 46241, Korea
| | - Da Hye Kim
- Anti-Aging Research Center, Dong-eui University, Busan 47340, Korea; (H.L.); (H.H.); (S.Y.J.); (M.Y.K.); (S.Y.K.); (D.H.K.); (S.H.H.)
- Department of Smart Bio-Health, Dong-eui University, Busan 47340, Korea
| | - Su Hyun Hong
- Anti-Aging Research Center, Dong-eui University, Busan 47340, Korea; (H.L.); (H.H.); (S.Y.J.); (M.Y.K.); (S.Y.K.); (D.H.K.); (S.H.H.)
- Department of Biochemistry, Dong-eui University College of Korean Medicine, Busan 47227, Korea
| | - Su Jeong Lee
- Department of Pathology and Regenerative Medicine, School of Dentistry, Kyungpook National University, Daegu 41940, Korea; (S.J.L.); (F.A.); (E.K.P.)
| | - Freshet Assefa
- Department of Pathology and Regenerative Medicine, School of Dentistry, Kyungpook National University, Daegu 41940, Korea; (S.J.L.); (F.A.); (E.K.P.)
| | - Gi-Young Kim
- Department of Marine Life Science, Jeju National University, Jeju 63243, Korea; (G.-Y.K.); (Y.-J.J.)
| | - Eui Kyun Park
- Department of Pathology and Regenerative Medicine, School of Dentistry, Kyungpook National University, Daegu 41940, Korea; (S.J.L.); (F.A.); (E.K.P.)
| | - Joung-Hyun Park
- Ocean Fisheries & Biology Center, Marine Bioprocess Co., Ltd., Busan 46048, Korea; (J.-H.P.); (B.-J.L.)
| | - Bae-Jin Lee
- Ocean Fisheries & Biology Center, Marine Bioprocess Co., Ltd., Busan 46048, Korea; (J.-H.P.); (B.-J.L.)
| | - You-Jin Jeon
- Department of Marine Life Science, Jeju National University, Jeju 63243, Korea; (G.-Y.K.); (Y.-J.J.)
| | - Yung Hyun Choi
- Anti-Aging Research Center, Dong-eui University, Busan 47340, Korea; (H.L.); (H.H.); (S.Y.J.); (M.Y.K.); (S.Y.K.); (D.H.K.); (S.H.H.)
- Department of Biochemistry, Dong-eui University College of Korean Medicine, Busan 47227, Korea
- Correspondence: ; Tel.: +82-51-890-3319
| |
Collapse
|
45
|
Peng L, Jin S, Lu J, Ouyang C, Guo J, Xie Z, Shen H, Wang P. Association between growth differentiation factor 5 rs143383 genetic polymorphism and the risk of knee osteoarthritis among Caucasian but not Asian: a meta-analysis. Arthritis Res Ther 2020; 22:215. [PMID: 32928309 PMCID: PMC7488690 DOI: 10.1186/s13075-020-02306-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Accepted: 08/31/2020] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND A few months ago, the Bioscience Reports journal showed that growth differentiation factor 5 (GDF5) rs143383 genetic polymorphism increases the susceptibility of knee osteoarthritis (KOA), but previous studies' results have debates about available data. Considering the availability of more recent data, we focus on clarifying the relationship of KOA and GDF5 rs143383 genetic polymorphism by a meta-analysis of case-control trial data. METHODS The eligible studies from the time of database established to Oct. 2019 were collected from PubMed, Springer, Cochrane library, Web of Science, China National Knowledge Infrastructure (CNKI), and Wan Fang library. Odds ratios (OR) and 95% confidence intervals (CI) were used to estimate the association between these polymorphisms and KOA risk. The meta-analysis was completed by STATA 18.0 software. RESULTS A total of 196 studies were collected, 16 of them included in final meta-analysis (7997 cases and 12,684 controls). There was significant association between GDF5 rs143383 polymorphism and KOA in all genetic models (for Allele model (C versus T): OR = 0.84 (95% CI = 0.76-0.91); dominate model (CC+CT versus TT): OR = 0.80 (95% CI = 0.72-0.90); recessive model (CC versus CT+TT): OR = 0.79 (95% CI = 0.68-0.92); heterozygote model (CT versus CC+TT): OR = 0.89 (95% CI = 0.80-0.97); homozygous model (CC versus TT): OR = 0.71 (95% CI = 0.60-0.85)). In the subgroup analysis, we obtained the results that there is no significance among Asians. CONCLUSION GDF5 rs143383 genetic polymorphism increases the risk of KOA among Caucasians; CC genotype and C allele are protective factors for the susceptibility of KOA among Caucasians.
Collapse
Affiliation(s)
- Lei Peng
- Department of Orthopedics, The Eighth Affiliated Hospital, Sun Yat-sen University, 3025# Shen Nan Road, Shenzhen, 518033, People's Republic of China.,Department of Orthopedics, The Second Affiliated Hospital, Sun Yat-sen University, 107# Yan Jiang Road West, Guangzhou, 510120, People's Republic of China.,Department of Orthopedics, The Second Affiliated Hospital of Hunan Normal University, The 921 Central Hospital of the People's Liberation Army, Hongshan bridge, Changsha, People's Republic of China
| | - Song Jin
- Department of Orthopedics, The Eighth Affiliated Hospital, Sun Yat-sen University, 3025# Shen Nan Road, Shenzhen, 518033, People's Republic of China
| | - Jiping Lu
- Department of Orthopedics, The Second Affiliated Hospital of Hunan Normal University, The 921 Central Hospital of the People's Liberation Army, Hongshan bridge, Changsha, People's Republic of China
| | - Chao Ouyang
- Department of Orthopedics, The Second Affiliated Hospital of Hunan Normal University, The 921 Central Hospital of the People's Liberation Army, Hongshan bridge, Changsha, People's Republic of China
| | - Jiang Guo
- Department of Orthopedics, The Eighth Affiliated Hospital, Sun Yat-sen University, 3025# Shen Nan Road, Shenzhen, 518033, People's Republic of China.,Department of Orthopedics, The Second Affiliated Hospital, Sun Yat-sen University, 107# Yan Jiang Road West, Guangzhou, 510120, People's Republic of China
| | - Zhongyu Xie
- Department of Orthopedics, The Eighth Affiliated Hospital, Sun Yat-sen University, 3025# Shen Nan Road, Shenzhen, 518033, People's Republic of China.,Department of Orthopedics, The Second Affiliated Hospital, Sun Yat-sen University, 107# Yan Jiang Road West, Guangzhou, 510120, People's Republic of China
| | - Huiyong Shen
- Department of Orthopedics, The Eighth Affiliated Hospital, Sun Yat-sen University, 3025# Shen Nan Road, Shenzhen, 518033, People's Republic of China. .,Department of Orthopedics, The Second Affiliated Hospital, Sun Yat-sen University, 107# Yan Jiang Road West, Guangzhou, 510120, People's Republic of China.
| | - Peng Wang
- Department of Orthopedics, The Eighth Affiliated Hospital, Sun Yat-sen University, 3025# Shen Nan Road, Shenzhen, 518033, People's Republic of China. .,Department of Orthopedics, The Second Affiliated Hospital, Sun Yat-sen University, 107# Yan Jiang Road West, Guangzhou, 510120, People's Republic of China.
| |
Collapse
|
46
|
Eldridge SE, Barawi A, Wang H, Roelofs AJ, Kaneva M, Guan Z, Lydon H, Thomas BL, Thorup AS, Fernandez BF, Caxaria S, Strachan D, Ali A, Shanmuganathan K, Pitzalis C, Whiteford JR, Henson F, McCaskie AW, De Bari C, Dell'Accio F. Agrin induces long-term osteochondral regeneration by supporting repair morphogenesis. Sci Transl Med 2020; 12:12/559/eaax9086. [PMID: 32878982 DOI: 10.1126/scitranslmed.aax9086] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2019] [Revised: 04/03/2020] [Accepted: 08/13/2020] [Indexed: 12/11/2022]
Abstract
Cartilage loss leads to osteoarthritis, the most common cause of disability for which there is no cure. Cartilage regeneration, therefore, is a priority in medicine. We report that agrin is a potent chondrogenic factor and that a single intraarticular administration of agrin induced long-lasting regeneration of critical-size osteochondral defects in mice, with restoration of tissue architecture and bone-cartilage interface. Agrin attracted joint resident progenitor cells to the site of injury and, through simultaneous activation of CREB and suppression of canonical WNT signaling downstream of β-catenin, induced expression of the chondrogenic stem cell marker GDF5 and differentiation into stable articular chondrocytes, forming stable articular cartilage. In sheep, an agrin-containing collagen gel resulted in long-lasting regeneration of bone and cartilage, which promoted increased ambulatory activity. Our findings support the therapeutic use of agrin for joint surface regeneration.
Collapse
Affiliation(s)
- Suzanne E Eldridge
- Centre for Experimental Medicine and Rheumatology, William Harvey Research Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London EC1M 6BQ, UK.
| | - Aida Barawi
- Centre for Experimental Medicine and Rheumatology, William Harvey Research Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London EC1M 6BQ, UK
| | - Hui Wang
- Arthritis and Regenerative Medicine Laboratory, Aberdeen Centre for Arthritis and Musculoskeletal Health, University of Aberdeen, Aberdeen AB25 2ZD, UK
| | - Anke J Roelofs
- Arthritis and Regenerative Medicine Laboratory, Aberdeen Centre for Arthritis and Musculoskeletal Health, University of Aberdeen, Aberdeen AB25 2ZD, UK
| | - Magdalena Kaneva
- Centre for Experimental Medicine and Rheumatology, William Harvey Research Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London EC1M 6BQ, UK
| | - Zeyu Guan
- Centre for Experimental Medicine and Rheumatology, William Harvey Research Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London EC1M 6BQ, UK
| | - Helen Lydon
- Division of Trauma and Orthopaedic Surgery, Department of Surgery, University of Cambridge, Addenbrooke's Hospital, Cambridge CB2 2QQ, UK
| | - Bethan L Thomas
- Centre for Biochemical Pharmacology, William Harvey Research Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London EC1M 6BQ, UK
| | - Anne-Sophie Thorup
- Centre for Experimental Medicine and Rheumatology, William Harvey Research Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London EC1M 6BQ, UK
| | - Beatriz F Fernandez
- Centre for Experimental Medicine and Rheumatology, William Harvey Research Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London EC1M 6BQ, UK
| | - Sara Caxaria
- Centre for Experimental Medicine and Rheumatology, William Harvey Research Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London EC1M 6BQ, UK
| | - Danielle Strachan
- Centre for Experimental Medicine and Rheumatology, William Harvey Research Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London EC1M 6BQ, UK
| | - Ahmed Ali
- Centre for Experimental Medicine and Rheumatology, William Harvey Research Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London EC1M 6BQ, UK
| | - Kanatheepan Shanmuganathan
- Centre for Experimental Medicine and Rheumatology, William Harvey Research Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London EC1M 6BQ, UK
| | - Costantino Pitzalis
- Centre for Experimental Medicine and Rheumatology, William Harvey Research Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London EC1M 6BQ, UK
| | - James R Whiteford
- Comparative Musculoskeletal Biology Group, Department of Veterinary Medicine, University of Cambridge, Madingley Road, Cambridge CB3 0ES, UK
| | - Frances Henson
- Centre for Microvascular Research, William Harvey Research Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London EC1M 6BQ, UK
| | - Andrew W McCaskie
- Division of Trauma and Orthopaedic Surgery, Department of Surgery, University of Cambridge, Addenbrooke's Hospital, Cambridge CB2 2QQ, UK
| | - Cosimo De Bari
- Arthritis and Regenerative Medicine Laboratory, Aberdeen Centre for Arthritis and Musculoskeletal Health, University of Aberdeen, Aberdeen AB25 2ZD, UK
| | - Francesco Dell'Accio
- Centre for Experimental Medicine and Rheumatology, William Harvey Research Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London EC1M 6BQ, UK.
| |
Collapse
|
47
|
Wang M, Zhao J, Wang Y, Mao Y, Zhao X, Huang P, Liu Q, Ma Y, Yao Y, Yang Z, Yuan W, Cui W, Payne TJ, Li MD. Genome-wide DNA methylation analysis reveals significant impact of long-term ambient air pollution exposure on biological functions related to mitochondria and immune response. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2020; 264:114707. [PMID: 32388307 DOI: 10.1016/j.envpol.2020.114707] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/24/2019] [Revised: 04/15/2020] [Accepted: 04/29/2020] [Indexed: 05/28/2023]
Abstract
Exposure to long-term ambient air pollution is believed to have adverse effects on human health. However, the mechanisms underlying these impacts are poorly understood. DNA methylation, a crucial epigenetic modification, is susceptible to environmental factors and likely involved in these processes. We conducted a whole-genome bisulfite sequencing study on 120 participants from a highly polluted region (HPR) and a less polluted region (LPR) in China, where the HPR had much higher concentrations of five air pollutants (PM2.5, PM10, SO2, NO2, and CO) (fold difference 1.6 to 6.6 times; P value 1.80E-07 to 3.19E-23). Genome-wide methylation analysis revealed 371 DMRs in subjects from the two areas and these DMRs were located primarily in gene regulatory elements such as promoters and enhancers. Gene enrichment analysis showed that DMR-related genes were significantly enriched in diseases related to pulmonary disorders and cancers and in biological processes related to mitochondrial assembly and cytokine production. Further, HPR participants showed a higher mtDNA copy number. Of those identified DMRs, 15 were significantly correlated with mtDNA copy number. Finally, cytokine assay indicated that an increased plasma interleukin-5 level was associated with greater air pollution. Taken together, our findings suggest that exposure to long-term ambient air pollution can lead to alterations in DNA methylation whose functions relate to mitochondria and immune responses.
Collapse
Affiliation(s)
- Maiqiu Wang
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Zhejiang University School of Medicine, Hangzhou, China
| | - Junsheng Zhao
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Zhejiang University School of Medicine, Hangzhou, China
| | - Yan Wang
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Zhejiang University School of Medicine, Hangzhou, China
| | - Ying Mao
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Zhejiang University School of Medicine, Hangzhou, China
| | - Xinyi Zhao
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Zhejiang University School of Medicine, Hangzhou, China
| | - Peng Huang
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Zhejiang University School of Medicine, Hangzhou, China
| | - Qiang Liu
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Zhejiang University School of Medicine, Hangzhou, China
| | - Yunlong Ma
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Zhejiang University School of Medicine, Hangzhou, China
| | - Yinghao Yao
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Zhejiang University School of Medicine, Hangzhou, China
| | - Zhongli Yang
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Zhejiang University School of Medicine, Hangzhou, China
| | - Wenji Yuan
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Zhejiang University School of Medicine, Hangzhou, China
| | - Wenyan Cui
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Zhejiang University School of Medicine, Hangzhou, China
| | - Thomas J Payne
- ACT Center for Tobacco Treatment, Education and Research, Department of Otolaryngology and Communicative Sciences, University of Mississippi Medical Center, Jackson, MS, USA
| | - Ming D Li
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Zhejiang University School of Medicine, Hangzhou, China; Research Center for Air Pollution and Health, Zhejiang University, Hangzhou, China.
| |
Collapse
|
48
|
Rahmani Del Bakhshayesh A, Babaie S, Tayefi Nasrabadi H, Asadi N, Akbarzadeh A, Abedelahi A. An overview of various treatment strategies, especially tissue engineering for damaged articular cartilage. ARTIFICIAL CELLS NANOMEDICINE AND BIOTECHNOLOGY 2020; 48:1089-1104. [DOI: 10.1080/21691401.2020.1809439] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Affiliation(s)
- Azizeh Rahmani Del Bakhshayesh
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Department of Tissue Engineering, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Soraya Babaie
- Department of Nanotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Hamid Tayefi Nasrabadi
- Department of Tissue Engineering, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Nahideh Asadi
- Department of Nanotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Abolfazl Akbarzadeh
- Department of Nanotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Ali Abedelahi
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Department of Tissue Engineering, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
49
|
Thorup AS, Dell'Accio F, Eldridge SE. Lessons from joint development for cartilage repair in the clinic. Dev Dyn 2020; 250:360-376. [PMID: 32738003 DOI: 10.1002/dvdy.228] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Revised: 07/23/2020] [Accepted: 07/24/2020] [Indexed: 12/19/2022] Open
Abstract
More than 250 years ago, William Hunter stated that when cartilage is destroyed it never recovers. In the last 20 years, the understanding of the mechanisms that lead to joint formation and the knowledge that some of these mechanisms are reactivated in the homeostatic responses of cartilage to injury has offered an unprecedented therapeutic opportunity to achieve cartilage regeneration. Very large investments in ambitious clinical trials are finally revealing that, although we do not have perfect medicines yet, disease modification is a feasible possibility for human osteoarthritis.
Collapse
Affiliation(s)
- Anne-Sophie Thorup
- Centre for Experimental Medicine and Rheumatology, William Harvey Research Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Francesco Dell'Accio
- Centre for Experimental Medicine and Rheumatology, William Harvey Research Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Suzanne E Eldridge
- Centre for Experimental Medicine and Rheumatology, William Harvey Research Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| |
Collapse
|
50
|
Vallittu PK, Posti JP, Piitulainen JM, Serlo W, Määttä JA, Heino TJ, Pagliari S, Syrjänen SM, Forte G. Biomaterial and implant induced ossification: in vitro and in vivo findings. J Tissue Eng Regen Med 2020; 14:1157-1168. [PMID: 32415757 PMCID: PMC7496445 DOI: 10.1002/term.3056] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2020] [Revised: 04/30/2020] [Accepted: 05/04/2020] [Indexed: 12/12/2022]
Abstract
Material-induced ossification is suggested as a suitable approach to heal large bone defects. Fiber-reinforced composite-bioactive glasses (FRC-BGs) display properties that could enhance the ossification of calvarial defects. Here, we analyzed the healing processes of a FRC-BG implant in vivo from the perspective of material-induced ossification. Histological analysis of the implant, which was removed 5 months after insertion, showed the formation of viable, noninflammatory mesenchymal tissue with newly-formed mineralized woven bone, as well as nonmineralized connective tissue with capillaries and larger blood vessels. The presence of osteocytes was detected within the newly generated bone matrix. To expand our understanding on the osteogenic properties of FRC-BG, we cultured human adipose tissue-derived mesenchymal stromal cells (AD-MSCs) in the presence of two different BGs (45S5 and S53P4) and Al2 O3 control. AD-MSCs grew and proliferated on all the scaffolds tested, as well as secreted abundant extracellular matrix, when osteogenic differentiation was appropriately stimulated. 45S5 and S53P4 induced enhanced expression of COL2A1, COL10A1, COL5A1 collagen subunits, and pro-osteogenic genes BMP2 and BMP4. The concomitant downregulation of BMP3 was also detected. Our findings show that FRC-BG can support the vascularization of the implant and the formation of abundant connective tissue in vivo. Specifically, BG 45S5 and BG S53P4 are suited to evoke the osteogenic potential of host mesenchymal stromal cells. In conclusion, FRC-BG implant demonstrated material-induced ossification both in vitro and in vivo.
Collapse
Affiliation(s)
- Pekka K. Vallittu
- Department of Biomaterials ScienceInstitute of Dentistry, University of Turku and City of Turku, Welfare DivisionTurkuFinland
| | - Jussi P. Posti
- Division of Clinical Neurosciences, Department of Neurosurgery, Turku Brain Injury CentreTurku University Hospital and University of TurkuTurkuFinland
| | - Jaakko M. Piitulainen
- Division of Surgery and Cancer Diseases, Department of Otorhinolaryngology ‐ Head and Neck Surgery, Turku University HospitalTurku Finland and University of TurkuTurkuFinland
| | - Willy Serlo
- PEDEGO Research Unit, University of Oulu, Oulu, Finland and Department of Children and AdolescentsOulu University HospitalOuluFinland
| | | | | | - Stefania Pagliari
- International Clinical Research Center of St. Anne's University Hospital BrnoBrnoCzech Republic
| | - Stina M. Syrjänen
- Department of Oral Pathology and Radiology, Institute of DentistryUniversity of TurkuTurkuFinland
| | - Giancarlo Forte
- International Clinical Research Center of St. Anne's University Hospital BrnoBrnoCzech Republic
| |
Collapse
|