1
|
Chermside-Scabbo CJ, Shuster JT, Erdmann-Gilmore P, Tycksen E, Zhang Q, Townsend RR, Silva MJ. A proteomics approach to study mouse long bones: examining baseline differences and mechanical loading-induced bone formation in young-adult and old mice. Aging (Albany NY) 2024; 16:12726-12768. [PMID: 39400554 PMCID: PMC11501390 DOI: 10.18632/aging.206131] [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: 06/20/2023] [Accepted: 09/23/2024] [Indexed: 10/15/2024]
Abstract
With aging, bone mass declines and the anabolic effects of skeletal loading diminish. While much research has focused on gene transcription, how bone ages and loses its mechanoresponsiveness at the protein level remains unclear. We developed a novel proteomics approach and performed a paired mass spectrometry and RNA-seq analysis on tibias from young-adult (5-month) and old (22-month) mice. We report the first correlation estimate between the bone proteome and transcriptome (Spearman ρ = 0.40), which is in line with other tissues but indicates that a relatively low amount of variation in protein levels is explained by the variation in transcript levels. Of 71 shared targets that differed with age, eight were associated with bone mineral density in previous GWAS, including understudied targets Asrgl1 and Timp2. We used complementary RNA in situ hybridization to confirm that Asrgl1 and Timp2 had reduced expression in osteoblasts/osteocytes in old bones. We also found evidence for reduced TGF-beta signaling with aging, in particular Tgfb2. Next, we defined proteomic changes following mechanical loading. At the protein level, bone differed more with age than with loading, and aged bone had fewer loading-induced changes. Overall, our findings underscore the need for complementary protein-level assays in skeletal biology research.
Collapse
Affiliation(s)
- Christopher J. Chermside-Scabbo
- Department of Orthopaedic Surgery, Washington University School of Medicine, St. Louis, MO 63110, USA
- Medical Scientist Training Program, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - John T. Shuster
- Department of Orthopaedic Surgery, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Petra Erdmann-Gilmore
- Department of Medicine, Proteomics Core, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Eric Tycksen
- Department of Genetics, McDonnell Genome Institute, Washington University School of Medicine, St. Louis, MO 63108, USA
| | - Qiang Zhang
- Department of Medicine, Proteomics Core, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - R. Reid Townsend
- Department of Medicine, Proteomics Core, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Matthew J. Silva
- Department of Orthopaedic Surgery, Washington University School of Medicine, St. Louis, MO 63110, USA
- Department of Biomedical Engineering, Washington University, St. Louis, MO 63105, USA
| |
Collapse
|
2
|
Meshcheryakova A, Bohdan S, Zimmermann P, Jaritz M, Pietschmann P, Mechtcheriakova D. RNA-Binding Proteins as Novel Effectors in Osteoblasts and Osteoclasts: A Systems Biology Approach to Dissect the Transcriptional Landscape. Int J Mol Sci 2024; 25:10417. [PMID: 39408753 PMCID: PMC11476634 DOI: 10.3390/ijms251910417] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2024] [Revised: 09/17/2024] [Accepted: 09/24/2024] [Indexed: 10/20/2024] Open
Abstract
Bone health is ensured by the coordinated action of two types of cells-the osteoblasts that build up bone structure and the osteoclasts that resorb the bone. The loss of balance in their action results in pathological conditions such as osteoporosis. Central to this study is a class of RNA-binding proteins (RBPs) that regulates the biogenesis of miRNAs. In turn, miRNAs represent a critical level of regulation of gene expression and thus control multiple cellular and biological processes. The impact of miRNAs on the pathobiology of various multifactorial diseases, including osteoporosis, has been demonstrated. However, the role of RBPs in bone remodeling is yet to be elucidated. The aim of this study is to dissect the transcriptional landscape of genes encoding the compendium of 180 RBPs in bone cells. We developed and applied a multi-modular integrative analysis algorithm. The core methodology is gene expression analysis using the GENEVESTIGATOR platform, which is a database and analysis tool for manually curated and publicly available transcriptomic data sets, and gene network reconstruction using the Ingenuity Pathway Analysis platform. In this work, comparative insights into gene expression patterns of RBPs in osteoblasts and osteoclasts were obtained, resulting in the identification of 24 differentially expressed genes. Furthermore, the regulation patterns upon different treatment conditions revealed 20 genes as being significantly up- or down-regulated. Next, novel gene-gene associations were dissected and gene networks were reconstructed. Additively, a set of osteoblast- and osteoclast-specific gene signatures were identified. The consolidation of data and information gained from each individual analytical module allowed nominating novel promising candidate genes encoding RBPs in osteoblasts and osteoclasts and will significantly enhance the understanding of potential regulatory mechanisms directing intracellular processes in the course of (patho)physiological bone turnover.
Collapse
Affiliation(s)
- Anastasia Meshcheryakova
- Department of Pathophysiology and Allergy Research, Center of Pathophysiology, Infectiology and Immunology, Medical University of Vienna, 1090 Vienna, Austria
| | - Serhii Bohdan
- Department of Pathophysiology and Allergy Research, Center of Pathophysiology, Infectiology and Immunology, Medical University of Vienna, 1090 Vienna, Austria
| | | | - Markus Jaritz
- Research Institute of Molecular Pathology, Vienna Biocenter, 1030 Vienna, Austria
| | - Peter Pietschmann
- Department of Pathophysiology and Allergy Research, Center of Pathophysiology, Infectiology and Immunology, Medical University of Vienna, 1090 Vienna, Austria
| | - Diana Mechtcheriakova
- Department of Pathophysiology and Allergy Research, Center of Pathophysiology, Infectiology and Immunology, Medical University of Vienna, 1090 Vienna, Austria
| |
Collapse
|
3
|
Jacobsen KK, Børte S, Laborie LB, Kristiansen H, Schäfer A, Gundersen T, Zayats T, Slagsvold Winsvold BK, Rosendahl K. COL11A1 is associated with developmental dysplasia of the hip and secondary osteoarthritis in the HUNT study. OSTEOARTHRITIS AND CARTILAGE OPEN 2024; 6:100424. [PMID: 38283578 PMCID: PMC10820335 DOI: 10.1016/j.ocarto.2023.100424] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Revised: 10/19/2023] [Accepted: 12/01/2023] [Indexed: 01/30/2024] Open
Abstract
Objective Developmental dysplasia of the hip (DDH) is a congenital condition affecting 2-3% of all infants. DDH increases the risk of osteoarthritis, is the cause of 30 % of all total hip arthroplasties (THAs) in adults <40 years of age and can result in loss of life quality. Our aim was to explore the genetic background of DDH in order to improve diagnosis, management and longterm outcome. Design We used the large, ongoing, longitudinal Trøndelag Health Study (HUNT) database. Case definition was based on ICD-9/-10 diagnoses of DDH, or osteoarthritis secondary to DDH. Analyses were performed using SAIGE software, with covariates including sex, batch, birth year and principal components. We included only single nucleotide polymorphisms (SNPs) with minor allele frequency (MAF) ≥ 0.01, R2≥ 0.8 and Hardy-Weinberg equilibrium (HWE) P-value ≥ 0.0001. Significance level was set at p < 5 × 10-8. Meta-analysis using data from DDH and primary osteoarthritis genome-wide association studies (GWASs) was done using METAL software. The study was approved by the regional ethical committee. Results Analysis included 69,500 individuals, of which 408 cases, and 8,531,386 SNPs. Two SNPs near COL11A1 were significantly associated with DDH; rs713162 (β = -0.43, SE = 0.07, p = 8.4 × 10-9) and rs6577334 (β = -0.43, SE = 0.08, p = 8.9 × 10-9). COL11A1 has previously been associated with acetabular dysplasia and osteoarthritis. Meta-analysis supported previous GWAS findings of both DDH and primary osteoarthritis. Conclusions This large, genome-wide case-control study indicates an association between COL11A1 and DDH and is an important contribution to investigating the etiology of DDH, with further research needed.
Collapse
Affiliation(s)
- Kaya Kvarme Jacobsen
- Department of Orthopedic Surgery, District General Hospital of Førde, Førde, Norway
| | - Sigrid Børte
- Research and Communication Unit for Musculoskeletal Health, Division of Clinical Neuroscience, Oslo University Hospital, Ullevaal, Oslo, Norway
- Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
- K. G. Jebsen Center for Genetic Epidemiology, Department of Public Health and Nursing, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology, Trondheim, Norway
| | - Lene Bjerke Laborie
- Department of Clinical Medicine, University of Bergen, Bergen, Norway
- Section for Pediatric Radiology, Department of Radiology, Haukeland University Hospital, Bergen, Norway
| | - Hege Kristiansen
- Department of Paediatrics, District General Hospital of Førde, Førde, Norway
- Department of Clinical Science, University of Bergen, Bergen, Norway
| | - Annette Schäfer
- Department of Paediatrics, District General Hospital of Førde, Førde, Norway
| | - Trude Gundersen
- Department of Clinical Medicine, University of Bergen, Bergen, Norway
- Department of Orthopaedic Surgery, Haukeland University Hospital, Bergen, Norway
| | - Tetyana Zayats
- Analytic and Translational Genetics Unit, Massachusetts General Hospital, Boston, MA, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- PROMENTA, Department of Psychology, University of Oslo, Oslo, Norway
| | - Bendik Kristoffer Slagsvold Winsvold
- K. G. Jebsen Center for Genetic Epidemiology, Department of Public Health and Nursing, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology, Trondheim, Norway
- Department of Research and Innovation, Division of Clinical Neuroscience, Oslo University Hospital, Oslo, Norway
- Department of Neurology, Oslo University Hospital, Oslo, Norway
| | - Karen Rosendahl
- Department of Radiology, University Hospital of North-Norway, Tromsø, Norway
- Department of Clinical Medicine, UiT, The Arctic University of Norway, Tromsø, Norway
| |
Collapse
|
4
|
Sun Y, You Y, Wu Q, Hu R, Dai K. Genetically inspired organoids prevent joint degeneration and alleviate chondrocyte senescence via Col11a1-HIF1α-mediated glycolysis-OXPHOS metabolism shift. Clin Transl Med 2024; 14:e1574. [PMID: 38314968 PMCID: PMC10840017 DOI: 10.1002/ctm2.1574] [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: 09/09/2023] [Revised: 01/16/2024] [Accepted: 01/22/2024] [Indexed: 02/07/2024] Open
Abstract
INTRODUCTION Developmental dysplasia of hip (DDH) is a hip joint disorder leading to subsequent osteoarthritis. Previous studies suggested collagen XI alpha 1 (COL11A1) as a potential gene in hip dysplasia and chondrocyte degeneration. However, no genetic association has reported COL11A1-related cellular therapy as treatment of DDH and joint degeneration. METHODS AND RESULTS We report identified genetic association between COL11A1 locus and DDH with genome-wide association study (GWAS). Further exome sequencing for familial DDH patients was conducted in different populations to identify potential pathogenic Col11A1 variants for familiar DDH. Further studies demonstrated involvement of COL11A1 expression was down-regulated in femoral head cartilage of DDH patients and Col11a1-KO mice with induced DDH. Col11a1-KO mice demonstrated aggravated joint degeneration and severe OA phenotype. To explore the underlying mechanism of Col11a1 in cartilage and DDH development, we generated scRNA-seq profiles for DDH and Col11a1-KO cartilage, demonstrating disrupted chondrocyte homeostasis and cellular senescence caused by Col11a1-HIF1α-mediated glycolysis-OXPHOS shift in chondrocytes. Genetically and biologically inspired, we further fabricated an intra-articular injection therapy to preventing cartilage degeneration by generating a Col11a1-over-expressed (OE) SMSC mini-organoids. Col11a1-OE organoids demonstrated superior chondrogenesis and ameliorated cartilage degeneration in DDH mice via regulating cellular senescence by up-regulated Col11a1/HIF1α-mediated glycolysis in chondrocytes. CONCLUSION We reported association between COL11A1 loci and DDH with GWAS and exome sequencing. Further studies demonstrated involvement of COL11A1 in DDH patients and Col11a1-KO mice. ScRNA-seq for DDH and Col11a1-KO cartilage demonstrated disrupted chondrocyte homeostasis and cellular senescence caused by Col11a1-HIF1α-mediated glycolysis-OXPHOS shift in chondrocytes. Genetically and biologically inspired, an intra-articular injection therapy was fabricated to prevent cartilage degeneration with Col11a1-OE SMSC organoids. Col11a1-OE organoids ameliorated cartilage degeneration in DDH mice via regulating cellular senescence by up-regulated Col11a1/HIF1α-mediated glycolysis in chondrocytes.
Collapse
Affiliation(s)
- Ye Sun
- Department of OrthopaedicsThe First Affiliated Hospital of Nanjing Medical UniversityJiangsuChina
- Department of Orthopaedic SurgeryShanghai Key Laboratory of Orthopaedic ImplantsShanghai Ninth People's Hospital, Shanghai Jiao Tong University School of MedicineShanghaiChina
| | - Yongqing You
- Department of Renal DiseasesAffiliated Hospital of Nanjing University of Chinese MedicineNanjingChina
| | - Qiang Wu
- Department of Orthopaedic SurgeryShanghai Key Laboratory of Orthopaedic ImplantsShanghai Ninth People's Hospital, Shanghai Jiao Tong University School of MedicineShanghaiChina
| | - Rui Hu
- Department of OrthopaedicsThe First Affiliated Hospital of Nanjing Medical UniversityJiangsuChina
| | - Kerong Dai
- Department of Orthopaedic SurgeryShanghai Key Laboratory of Orthopaedic ImplantsShanghai Ninth People's Hospital, Shanghai Jiao Tong University School of MedicineShanghaiChina
| |
Collapse
|
5
|
Yu H, Khanshour AM, Ushiki A, Otomo N, Koike Y, Einarsdottir E, Fan Y, Antunes L, Kidane YH, Cornelia R, Sheng RR, Zhang Y, Pei J, Grishin NV, Evers BM, Cheung JPY, Herring JA, Terao C, Song YQ, Gurnett CA, Gerdhem P, Ikegawa S, Rios JJ, Ahituv N, Wise CA. Association of genetic variation in COL11A1 with adolescent idiopathic scoliosis. eLife 2024; 12:RP89762. [PMID: 38277211 PMCID: PMC10945706 DOI: 10.7554/elife.89762] [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] [Indexed: 01/27/2024] Open
Abstract
Adolescent idiopathic scoliosis (AIS) is a common and progressive spinal deformity in children that exhibits striking sexual dimorphism, with girls at more than fivefold greater risk of severe disease compared to boys. Despite its medical impact, the molecular mechanisms that drive AIS are largely unknown. We previously defined a female-specific AIS genetic risk locus in an enhancer near the PAX1 gene. Here, we sought to define the roles of PAX1 and newly identified AIS-associated genes in the developmental mechanism of AIS. In a genetic study of 10,519 individuals with AIS and 93,238 unaffected controls, significant association was identified with a variant in COL11A1 encoding collagen (α1) XI (rs3753841; NM_080629.2_c.4004C>T; p.(Pro1335Leu); p=7.07E-11, OR = 1.118). Using CRISPR mutagenesis we generated Pax1 knockout mice (Pax1-/-). In postnatal spines we found that PAX1 and collagen (α1) XI protein both localize within the intervertebral disc-vertebral junction region encompassing the growth plate, with less collagen (α1) XI detected in Pax1-/- spines compared to wild-type. By genetic targeting we found that wild-type Col11a1 expression in costal chondrocytes suppresses expression of Pax1 and of Mmp3, encoding the matrix metalloproteinase 3 enzyme implicated in matrix remodeling. However, the latter suppression was abrogated in the presence of the AIS-associated COL11A1P1335L mutant. Further, we found that either knockdown of the estrogen receptor gene Esr2 or tamoxifen treatment significantly altered Col11a1 and Mmp3 expression in chondrocytes. We propose a new molecular model of AIS pathogenesis wherein genetic variation and estrogen signaling increase disease susceptibility by altering a PAX1-COL11a1-MMP3 signaling axis in spinal chondrocytes.
Collapse
Affiliation(s)
- Hao Yu
- Center for Translational Research, Scottish Rite for ChildrenDallasUnited States
| | - Anas M Khanshour
- Center for Translational Research, Scottish Rite for ChildrenDallasUnited States
| | - Aki Ushiki
- Department of Bioengineering and Therapeutic Sciences, University of California, San FranciscoSan FranciscoUnited States
- Institute for Human Genetics, University of California, San FranciscoSan FranciscoUnited States
| | - Nao Otomo
- Laboratory of Bone and Joint Diseases, RIKEN Center for Integrative Medical SciencesTokyoJapan
| | - Yoshinao Koike
- Laboratory of Bone and Joint Diseases, RIKEN Center for Integrative Medical SciencesTokyoJapan
- Laboratory for Statistical and Translational Genetics, RIKEN Center for Integrative Medical SciencesYokohamaJapan
| | - Elisabet Einarsdottir
- Science for Life Laboratory, Department of Gene Technology, KTH-Royal Institute of TechnologySolnaSweden
| | - Yanhui Fan
- School of Biomedical Sciences, The University of Hong KongHong Kong SARChina
| | - Lilian Antunes
- Department of Neurology, Washington University in St. LouisSt. LouisUnited States
| | - Yared H Kidane
- Center for Translational Research, Scottish Rite for ChildrenDallasUnited States
| | - Reuel Cornelia
- Center for Translational Research, Scottish Rite for ChildrenDallasUnited States
| | - Rory R Sheng
- Department of Bioengineering and Therapeutic Sciences, University of California, San FranciscoSan FranciscoUnited States
- Institute for Human Genetics, University of California, San FranciscoSan FranciscoUnited States
| | - Yichi Zhang
- Department of Bioengineering and Therapeutic Sciences, University of California, San FranciscoSan FranciscoUnited States
- Institute for Human Genetics, University of California, San FranciscoSan FranciscoUnited States
- School of Pharmaceutical Sciences, Tsinghua UniversityBeijingChina
| | - Jimin Pei
- Department of Biophysics, University of Texas Southwestern Medical CenterDallasUnited States
| | - Nick V Grishin
- Department of Biophysics, University of Texas Southwestern Medical CenterDallasUnited States
| | - Bret M Evers
- Department of Pathology, University of Texas Southwestern Medical CenterDallasUnited States
- Department of Ophthalmology, University of Texas Southwestern Medical CenterDallasUnited States
| | - Jason Pui Yin Cheung
- Department of Orthopaedics and Traumatology LKS Faculty of Medicine, The University of Hong KongHong Kong SARChina
| | - John A Herring
- Department of Orthopedic Surgery, Scottish Rite for ChildrenDallasUnited States
- Department of Orthopaedic Surgery, University of Texas Southwestern Medical CenterDallasUnited States
| | - Chikashi Terao
- Laboratory for Statistical and Translational Genetics, RIKEN Center for Integrative Medical SciencesYokohamaJapan
| | - You-qiang Song
- School of Biomedical Sciences, The University of Hong KongHong Kong SARChina
| | - Christina A Gurnett
- Department of Neurology, Washington University in St. LouisSt. LouisUnited States
| | - Paul Gerdhem
- Department of Surgical Sciences, Uppsala UniversityUppsalaSweden
- Department of Orthopaedics and Hand Surgery, Uppsala University HospitalUppsalaSweden
- Department of Clinical Science, Intervention & Technology (CLINTEC), Karolinska Institutet, Stockholm, Uppsala UniversityUppsalaSweden
| | - Shiro Ikegawa
- Laboratory of Bone and Joint Diseases, RIKEN Center for Integrative Medical SciencesTokyoJapan
| | - Jonathan J Rios
- Center for Translational Research, Scottish Rite for ChildrenDallasUnited States
- Department of Orthopaedic Surgery, University of Texas Southwestern Medical CenterDallasUnited States
- Eugene McDermott Center for Human Growth and Development, University of Texas Southwestern Medical CenterDallasUnited States
- Department of Pediatrics, University of Texas Southwestern Medical CenterDallasUnited States
| | - Nadav Ahituv
- Department of Bioengineering and Therapeutic Sciences, University of California, San FranciscoSan FranciscoUnited States
- Institute for Human Genetics, University of California, San FranciscoSan FranciscoUnited States
| | - Carol A Wise
- Center for Translational Research, Scottish Rite for ChildrenDallasUnited States
- Department of Orthopaedic Surgery, University of Texas Southwestern Medical CenterDallasUnited States
- Eugene McDermott Center for Human Growth and Development, University of Texas Southwestern Medical CenterDallasUnited States
- Department of Pediatrics, University of Texas Southwestern Medical CenterDallasUnited States
| |
Collapse
|
6
|
Yu H, Khanshour AM, Ushiki A, Otomo N, Koike Y, Einarsdottir E, Fan Y, Antunes L, Kidane YH, Cornelia R, Sheng R, Zhang Y, Pei J, Grishin NV, Evers BM, Cheung JPY, Herring JA, Terao C, Song YQ, Gurnett CA, Gerdhem P, Ikegawa S, Rios JJ, Ahituv N, Wise CA. Association of genetic variation in COL11A1 with adolescent idiopathic scoliosis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.05.26.542293. [PMID: 37292598 PMCID: PMC10245954 DOI: 10.1101/2023.05.26.542293] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Adolescent idiopathic scoliosis (AIS) is a common and progressive spinal deformity in children that exhibits striking sexual dimorphism, with girls at more than five-fold greater risk of severe disease compared to boys. Despite its medical impact, the molecular mechanisms that drive AIS are largely unknown. We previously defined a female-specific AIS genetic risk locus in an enhancer near the PAX1 gene. Here we sought to define the roles of PAX1 and newly-identified AIS-associated genes in the developmental mechanism of AIS. In a genetic study of 10,519 individuals with AIS and 93,238 unaffected controls, significant association was identified with a variant in COL11A1 encoding collagen (α1) XI (rs3753841; NM_080629.2_c.4004C>T; p.(Pro1335Leu); P=7.07e-11, OR=1.118). Using CRISPR mutagenesis we generated Pax1 knockout mice (Pax1-/-). In postnatal spines we found that PAX1 and collagen (α1) XI protein both localize within the intervertebral disc (IVD)-vertebral junction region encompassing the growth plate, with less collagen (α1) XI detected in Pax1-/- spines compared to wildtype. By genetic targeting we found that wildtype Col11a1 expression in costal chondrocytes suppresses expression of Pax1 and of Mmp3, encoding the matrix metalloproteinase 3 enzyme implicated in matrix remodeling. However, this suppression was abrogated in the presence of the AIS-associated COL11A1P1335L mutant. Further, we found that either knockdown of the estrogen receptor gene Esr2, or tamoxifen treatment, significantly altered Col11a1 and Mmp3 expression in chondrocytes. We propose a new molecular model of AIS pathogenesis wherein genetic variation and estrogen signaling increase disease susceptibility by altering a Pax1-Col11a1-Mmp3 signaling axis in spinal chondrocytes.
Collapse
Affiliation(s)
- Hao Yu
- Center for Pediatric Bone Biology and Translational Research, Scottish Rite for Children, Dallas, TX, USA
| | - Anas M Khanshour
- Center for Pediatric Bone Biology and Translational Research, Scottish Rite for Children, Dallas, TX, USA
| | - Aki Ushiki
- Department of Bioengineering and Therapeutic Sciences, University of California San Francisco, San Francisco, CA, USA
- Institute for Human Genetics, University of California San Francisco, San Francisco, CA, USA
| | - Nao Otomo
- Laboratory of Bone and Joint Diseases, RIKEN Center for Integrative Medical Sciences, Tokyo, JP
| | - Yoshinao Koike
- Laboratory of Bone and Joint Diseases, RIKEN Center for Integrative Medical Sciences, Tokyo, JP
- Laboratory for Statistical and Translational Genetics, RIKEN Center for Integrative Medical Sciences, Yokohama, JP
| | - Elisabet Einarsdottir
- Science for Life Laboratory, Department of Gene Technology, KTH-Royal Institute of Technology, Solna, SE
| | - Yanhui Fan
- School of Biomedical Sciences, The University of Hong Kong, Hong Kong SAR, CN
| | - Lilian Antunes
- Department of Neurology, Washington University in St. Louis, St. Louis, MO, USA
| | - Yared H Kidane
- Center for Pediatric Bone Biology and Translational Research, Scottish Rite for Children, Dallas, TX, USA
| | - Reuel Cornelia
- Center for Pediatric Bone Biology and Translational Research, Scottish Rite for Children, Dallas, TX, USA
| | - Rory Sheng
- Department of Bioengineering and Therapeutic Sciences, University of California San Francisco, San Francisco, CA, USA
- Institute for Human Genetics, University of California San Francisco, San Francisco, CA, USA
| | - Yichi Zhang
- Department of Bioengineering and Therapeutic Sciences, University of California San Francisco, San Francisco, CA, USA
- Institute for Human Genetics, University of California San Francisco, San Francisco, CA, USA
- School of Pharmaceutical Sciences, Tsinghua University, Beijing, CN
| | - Jimin Pei
- Department of Biophysics, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Nick V Grishin
- Department of Biophysics, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Bret M Evers
- Department of Pathology, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Department of Ophthalmology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Jason Pui Yin Cheung
- Department of Orthopaedics and Traumatology LKS Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, CN
| | - John A Herring
- Department of Orthopedic Surgery, Scottish Rite for Children, Dallas, TX, USA
- Department of Orthopaedic Surgery, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Chikashi Terao
- Laboratory for Statistical and Translational Genetics, RIKEN Center for Integrative Medical Sciences, Yokohama, JP
| | - You-Qiang Song
- School of Biomedical Sciences, The University of Hong Kong, Hong Kong SAR, CN
| | - Christina A Gurnett
- Department of Neurology, Washington University in St. Louis, St. Louis, MO, USA
| | - Paul Gerdhem
- Department of Clinical Science, Intervention & Technology (CLINTEC), Karolinska Institutet, Stockholm, Uppsala University, Uppsala, SE
- Department of Surgical Sciences, Uppsala University and
- Department of Orthopaedics and Hand Surgery, Uppsala University Hospital, Uppsala, SE
| | - Shiro Ikegawa
- Laboratory of Bone and Joint Diseases, RIKEN Center for Integrative Medical Sciences, Tokyo, JP
| | - Jonathan J Rios
- Center for Pediatric Bone Biology and Translational Research, Scottish Rite for Children, Dallas, TX, USA
- Department of Orthopaedic Surgery, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Eugene McDermott Center for Human Growth and Development, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Nadav Ahituv
- Department of Bioengineering and Therapeutic Sciences, University of California San Francisco, San Francisco, CA, USA
- Institute for Human Genetics, University of California San Francisco, San Francisco, CA, USA
| | - Carol A Wise
- Center for Pediatric Bone Biology and Translational Research, Scottish Rite for Children, Dallas, TX, USA
- Department of Orthopaedic Surgery, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Eugene McDermott Center for Human Growth and Development, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX, USA
| |
Collapse
|
7
|
Guo S, Yan M, Li X, Zhang S, Liu Z, Li K, Liu P, Liu Y, Sun G, Fu Q. Single-cell RNA-seq analysis reveals that immune cells induce human nucleus pulposus ossification and degeneration. Front Immunol 2023; 14:1224627. [PMID: 37638033 PMCID: PMC10449260 DOI: 10.3389/fimmu.2023.1224627] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2023] [Accepted: 07/24/2023] [Indexed: 08/29/2023] Open
Abstract
Background and aims Determining the transcriptomes and molecular mechanism underlying human degenerative nucleus pulposus (NP) is of critical importance for treating intervertebral disc degeneration (IDD). Here, we aimed to elucidate the detailed molecular mechanism of NP ossification and IDD using single-cell RNA sequencing. Methods Single-cell RNA-seq and bioinformatic analysis were performed to identify NP cell populations with gene signatures, biological processes and pathways, and subpopulation analysis, RNA velocity analysis, and cell-to-cell communication analysis were performed in four IDD patients. We also verified the effects of immune cells on NP ossification using cultured NP cells and a well-established rat IDD model. Results We identified five cell populations with gene expression profiles in degenerative NP at single-cell resolution. GO database analysis showed that degenerative NP-associated genes were mainly enriched in extracellular matrix organization, immune response, and ossification. Gene set enrichment analysis showed that rheumatoid arthritis signaling, antigen processing and presentation signaling were activated in the blood cell cluster. We revealed that stromal cells, which are progenitor cells, differentiated toward an ossification phenotype and delineated interactions between immune cells (macrophages and T cells) and stromal cells. Immune factors such as TNF-α, CD74 and CCL-3 promoted the differentiation of stromal cells toward an ossification phenotype in vitro. Blocking TNF-α with a specific inhibitor successfully reversed NP ossification and modified NP morphology in vivo. Conclusion Our study revealed an increase in macrophages and T cells in degenerative NP, which induced stromal cell differentiation toward an ossification phenotype, and contributed to the identification of a novel therapeutic target to delay IDD.
Collapse
Affiliation(s)
- Song Guo
- Department of Spine Surgery, Shanghai Jiaotong University First People’s Hospital, Shanghai, China
| | - Meijun Yan
- Department of Spine Surgery, Shanghai Jiaotong University First People’s Hospital, Shanghai, China
| | - Xinhua Li
- Department of Spine Surgery, Shanghai Jiaotong University First People’s Hospital, Shanghai, China
| | - Shuya Zhang
- Department of Neurologic Surgery, Mayo Clinic, Rochester, MN, United States
| | - Zhong Liu
- Department of Spine Surgery, Shanghai Jiaotong University First People’s Hospital, Shanghai, China
| | - Kewei Li
- Department of Spine Surgery, Shanghai Jiaotong University First People’s Hospital, Shanghai, China
| | - Pengcheng Liu
- Department of Spine Surgery, Shanghai Jiaotong University First People’s Hospital, Shanghai, China
| | - Yanbin Liu
- Department of Spine Surgery, Shanghai Jiaotong University First People’s Hospital, Shanghai, China
| | - Guixin Sun
- Department of Traumatology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Qiang Fu
- Department of Spine Surgery, Shanghai Jiaotong University First People’s Hospital, Shanghai, China
| |
Collapse
|
8
|
Yue M, Liu Y, Zhang P, Li Z, Zhou Y. Integrative Analysis Reveals the Diverse Effects of 3D Stiffness upon Stem Cell Fate. Int J Mol Sci 2023; 24:9311. [PMID: 37298263 PMCID: PMC10253631 DOI: 10.3390/ijms24119311] [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: 04/24/2023] [Revised: 05/09/2023] [Accepted: 05/23/2023] [Indexed: 06/12/2023] Open
Abstract
The origin of life and native tissue development are dependent on the heterogeneity of pluripotent stem cells. Bone marrow mesenchymal stem cells (BMMSCs) are located in a complicated niche with variable matrix stiffnesses, resulting in divergent stem cell fates. However, how stiffness drives stem cell fate remains unknown. For this study, we performed whole-gene transcriptomics and precise untargeted metabolomics sequencing to elucidate the complex interaction network of stem cell transcriptional and metabolic signals in extracellular matrices (ECMs) with different stiffnesses, and we propose a potential mechanism involved in stem cell fate decision. In a stiff (39~45 kPa) ECM, biosynthesis of aminoacyl-tRNA was up-regulated, and increased osteogenesis was also observed. In a soft (7~10 kPa) ECM, biosynthesis of unsaturated fatty acids and deposition of glycosaminoglycans were increased, accompanied by enhanced adipogenic/chondrogenic differentiation of BMMSCs. In addition, a panel of genes responding to the stiffness of the ECM were validated in vitro, mapping out the key signaling network that regulates stem cells' fate decisions. This finding of "stiffness-dependent manipulation of stem cell fate" provides a novel molecular biological basis for development of potential therapeutic targets within tissue engineering, from both a cellular metabolic and a biomechanical perspective.
Collapse
Affiliation(s)
- Muxin Yue
- Department of Prosthodontics, Peking University School and Hospital of Stomatology, Beijing 100081, China; (M.Y.); (Y.L.); (P.Z.)
- National Center for Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices & Beijing Key Laboratory of Digital Stomatology, Beijing 100081, China
| | - Yunsong Liu
- Department of Prosthodontics, Peking University School and Hospital of Stomatology, Beijing 100081, China; (M.Y.); (Y.L.); (P.Z.)
- National Center for Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices & Beijing Key Laboratory of Digital Stomatology, Beijing 100081, China
| | - Ping Zhang
- Department of Prosthodontics, Peking University School and Hospital of Stomatology, Beijing 100081, China; (M.Y.); (Y.L.); (P.Z.)
- National Center for Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices & Beijing Key Laboratory of Digital Stomatology, Beijing 100081, China
| | - Zheng Li
- Department of Prosthodontics, Peking University School and Hospital of Stomatology, Beijing 100081, China; (M.Y.); (Y.L.); (P.Z.)
- National Center for Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices & Beijing Key Laboratory of Digital Stomatology, Beijing 100081, China
| | - Yongsheng Zhou
- Department of Prosthodontics, Peking University School and Hospital of Stomatology, Beijing 100081, China; (M.Y.); (Y.L.); (P.Z.)
- National Center for Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices & Beijing Key Laboratory of Digital Stomatology, Beijing 100081, China
| |
Collapse
|
9
|
Luo J, Zhang T, Wang W, Zhang D. Genome-wide association study of handgrip strength in the Northern Chinese adult twins. Connect Tissue Res 2023; 64:117-125. [PMID: 35876483 DOI: 10.1080/03008207.2022.2104160] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
PURPOSE Currently, new loci related to handgrip strength have been identified in genome-wide association studies. However, this topic is an understudied area in the Chinese population. MATERIALS AND METHODS A total of 135 dizygotic twin pairs recruited from the Qingdao Twin Registry system were included in the present study. Using GEMMA, VEAGSE2, and PASCAL software for SNP-based analysis, gene-based analysis, and pathway-based analysis, respectively. The resulting SNPs were subjected to eQTL analysis. RESULTS Although none of the loci reach the statistically significant level (p < 5 × 10-8), we found 19 SNPs exceeding the suggestive significant level (p < 1 × 10-5). After imputation, 162 SNPs reached suggestive evidence level for handgrip strength. A total of 1,118 genes reached the nominal significance level (p < 0.05) in gene-based analysis. A total of 626 potential biological pathways were associated with handgrip strength (p < 0.05). The results of eQTL analysis were mainly enriched in tissues such as the muscle-skeletal, brain, visceral fat, and brain-cortical. CONCLUSIONS Genetic variants may involve in regulatory domains, functional genes, and biological pathways that mediate handgrip strength.
Collapse
Affiliation(s)
- Jia Luo
- Department of Epidemiology and Health Statistics, Public Health College, Qingdao University, Qingdao, Shandong Province, China
| | - Tianhao Zhang
- Department of Epidemiology and Health Statistics, Public Health College, Qingdao University, Qingdao, Shandong Province, China
| | - Weijing Wang
- Department of Epidemiology and Health Statistics, Public Health College, Qingdao University, Qingdao, Shandong Province, China
| | - Dongfeng Zhang
- Department of Epidemiology and Health Statistics, Public Health College, Qingdao University, Qingdao, Shandong Province, China
| |
Collapse
|
10
|
Jiang TM. Unveiling the Time Course Mechanism of Bone Fracture Healing by Transcriptional Profiles. Comb Chem High Throughput Screen 2023; 26:149-162. [PMID: 35418283 DOI: 10.2174/1386207325666220412134311] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2021] [Revised: 02/03/2022] [Accepted: 02/14/2022] [Indexed: 11/22/2022]
Abstract
BACKGROUND Bone fracture healing is a time-consuming and high-priority orthopedic problem worldwide. OBJECTIVE Discovering the potential mechanism of bone healing at a time course and transcriptional level may better help manage bone fracture. METHODS In this study, we analyze a time-course bone fracture healing transcriptional dataset in a rat model (GSE592, GSE594, and GSE1371) of Gene Expression Omnibus (GEO). RNA was obtained from female Sprague-Dawley rats with a femoral fracture at the initial time (day 3) as well as early (week 1), middle (week 2), and late (week 4) time periods, with nonfracture rats used as control. Gene Ontology (GO) functional analysis and pathway examinations were performed for further measurements of GSEA and hub genes. RESULTS Results indicated that the four stages of bone fracture healing at the initial, early, middle, and late time periods represent the phases of hematoma formation, callus formation, callus molding, and mature lamellar bone formation, respectively. Extracellular organization was positively employed throughout the four stages. At the hematoma formation phase, the muscle contraction process was downregulated. Antibacterial peptide pathway was downregulated at all phases. The upregulation of Fn1 (initial, early, middle, and late time periods), Col3a1 (initial, early, and middle time periods), Col11a1 (initial and early time periods), Mmp9 (middle and late time periods), Mmp13 (early, middle, and late time periods) and the downregulation of RatNP-3b (initial, early, middle, and late time periods) were possible symbols for bone fracture healing and may be used as therapeutic targets. CONCLUSION These findings suggest some new potential pathways and genes in the process of bone fracture healing and further provide insights that can be used in targeted molecular therapy for bone fracture healing.
Collapse
Affiliation(s)
- Tong-Meng Jiang
- School of Materials Science and Engineering, Zhejiang University, Hangzhou, 310027, China
| |
Collapse
|
11
|
Wang X, Li G, Jiang Y, Tang J, Fan Y, Ren J. Genomic insights into the conservation and population genetics of two Chinese native goat breeds. J Anim Sci 2022; 100:skac274. [PMID: 35998083 PMCID: PMC9585554 DOI: 10.1093/jas/skac274] [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: 10/14/2021] [Accepted: 08/20/2022] [Indexed: 11/14/2022] Open
Abstract
Chinese goats are an important group of goats worldwide. However, there are few studies on the conservation priority, genetic relationship, and potential gene flow between Chinese and global goat breeds. Here, we genotyped 239 goats from conservation populations of the Chinese Guangfeng and Ganxi breeds using the GoatSNP50 BeadChip. The conservation priority, population structure, selection signatures and introgression of these goats were analyzed in the context of 36 global goat breeds. First, we showed that Guangfeng and Ganxi goats had the largest effective population sizes across the global breeds 13 generations ago. Nevertheless, Ganxi goats have recently experienced a high degree of inbreeding, resulting in their conservation priority based on total gene and allelic diversities being lower than that of most other Chinese breeds (including Guangfeng goats). Population structure and admixture analyses showed that an average of 18% of Guangfeng genomic components were introgressed from Boer goats approximately 18-yr ago. Next, we reconstructed the subfamily structure of the core populations of Guangfeng and Ganxi goats, and proposed reasonable conservation strategies for inbreeding management. Moreover, a list of candidate genes under selection for fertility, immunity, growth, and meat quality were detected in Guangfeng and Ganxi goats. Finally, we identified some genes related to body development and reproduction, which were introgressed from Boer goats and may be beneficial for improving performance and productivity of Guangfeng goats. In conclusion, this study not only provides new insights into the conservation and utilization of Guangfeng and Ganxi goats but also enriches our understanding of artificial introgression from exotic goats into Chinese local goats.
Collapse
Affiliation(s)
- Xiaopeng Wang
- Guangdong Laboratory for Lingnan Modern Agriculture, College of Animal Science, South China Agricultural University, Guangzhou 510642, China
| | - Guixin Li
- Guangdong Laboratory for Lingnan Modern Agriculture, College of Animal Science, South China Agricultural University, Guangzhou 510642, China
| | - Yongchuang Jiang
- Guangdong Laboratory for Lingnan Modern Agriculture, College of Animal Science, South China Agricultural University, Guangzhou 510642, China
| | - Jianhong Tang
- Guangdong Laboratory for Lingnan Modern Agriculture, College of Animal Science, South China Agricultural University, Guangzhou 510642, China
- Laboratory Animal Engineering Research Center of Ganzhou, Gannan Medical University, Ganzhou 341000, China
| | - Yin Fan
- Department of Animal Science, Jiangxi Biotech Vocational College, Nanchang 330200, China
| | - Jun Ren
- Guangdong Laboratory for Lingnan Modern Agriculture, College of Animal Science, South China Agricultural University, Guangzhou 510642, China
| |
Collapse
|
12
|
Reeck JC, Oxford JT. The Shape of the Jaw-Zebrafish Col11a1a Regulates Meckel's Cartilage Morphogenesis and Mineralization. J Dev Biol 2022; 10:jdb10040040. [PMID: 36278545 PMCID: PMC9590009 DOI: 10.3390/jdb10040040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Revised: 08/19/2022] [Accepted: 09/14/2022] [Indexed: 11/25/2022] Open
Abstract
The expression of the col11a1a gene is essential for normal skeletal development, affecting both cartilage and bone. Loss of function mutations have been shown to cause abnormalities in the growth plate of long bones, as well as in craniofacial development. However, the specific effects on Meckel's cartilage have not been well studied. To further understand the effect of col11a1a gene function, we analyzed the developing jaw in zebrafish using gene knockdown by the injection of an antisense morpholino oligonucleotide using transgenic Tg(sp7:EGFP) and Tg(Fli1a:EGFP) EGFP reporter fish, as well as wildtype AB zebrafish. Our results demonstrate that zebrafish col11a1a knockdown impairs the cellular organization of Meckel's cartilage in the developing jaw and alters the bone formation that occurs adjacent to the Meckel's cartilage. These results suggest roles for Col11a1a protein in cartilage intermediates of bone development, the subsequent mineralization of the bony collar of long bones, and that which occurs adjacent to Meckel's cartilage in the developing jaw.
Collapse
|
13
|
Reeck JC, Hardy MJ, Pu X, Keller-Peck C, Oxford JT. Authentication of a novel antibody to zebrafish collagen type XI alpha 1 chain (Col11a1a). BMC Res Notes 2021; 14:359. [PMID: 34526111 PMCID: PMC8444443 DOI: 10.1186/s13104-021-05770-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2021] [Accepted: 09/01/2021] [Indexed: 11/10/2022] Open
Abstract
OBJECTIVE Extracellular matrix proteins play important roles in embryonic development and antibodies that specifically detect these proteins are essential to understanding their function. The zebrafish embryo is a popular model for vertebrate development but suffers from a dearth of authenticated antibody reagents for research. Here, we describe a novel antibody designed to detect the minor fibrillar collagen chain Col11a1a in zebrafish (AB strain). RESULTS The Col11a1a antibody was raised in rabbit against a peptide comprising a unique sequence within the zebrafish Col11a1a gene product. The antibody was affinity-purified and characterized by ELISA. The antibody is effective for immunoblot and immunohistochemistry applications. Protein bands identified by immunoblot were confirmed by mass spectrometry and sensitivity to collagenase. Col11a1a knockout zebrafish were used to confirm specificity of the antibody. The Col11a1a antibody labeled cartilaginous structures within the developing jaw, consistent with previously characterized Col11a1 antibodies in other species. Col11a1a within formalin-fixed paraffin-embedded zebrafish were recognized by the antibody. The antibodies and the approaches described here will help to address the lack of well-defined antibody reagents in zebrafish research.
Collapse
Affiliation(s)
- Jonathon C. Reeck
- Department of Biological Sciences, Biomolecular Sciences Graduate Program, and Biomolecular Research Center, Boise State University, Boise, ID 83725 USA
| | - Makenna J. Hardy
- Biomolecular Sciences Graduate Program, Biomolecular Research Center, Boise State University, Boise, ID 83725 USA
| | - Xinzhu Pu
- Biomolecular Research Center, Boise State University, Boise, ID 83725 USA
| | | | - Julia Thom Oxford
- Department of Biological Sciences, Biomolecular Sciences Graduate Program, and Biomolecular Research Center, Boise State University, Boise, ID 83725 USA
| |
Collapse
|
14
|
Gorski JP, Franz NT, Pernoud D, Keightley A, Eyre DR, Oxford JT. A repeated triple lysine motif anchors complexes containing bone sialoprotein and the type XI collagen A1 chain involved in bone mineralization. J Biol Chem 2021; 296:100436. [PMID: 33610546 PMCID: PMC8008188 DOI: 10.1016/j.jbc.2021.100436] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Revised: 02/05/2021] [Accepted: 02/16/2021] [Indexed: 01/16/2023] Open
Abstract
While details remain unclear, initiation of woven bone mineralization is believed to be mediated by collagen and potentially nucleated by bone sialoprotein (BSP). Interestingly, our recent publication showed that BSP and type XI collagen form complexes in mineralizing osteoblastic cultures. To learn more, we examined the protein composition of extracellular sites of de novo hydroxyapatite deposition which were enriched in BSP and Col11a1 containing an alternatively spliced "6b" exonal sequence. An alternate splice variant "6a" sequence was not similarly co-localized. BSP and Col11a1 co-purify upon ion-exchange chromatography or immunoprecipitation. Binding of the Col11a1 "6b" exonal sequence to bone sialoprotein was demonstrated with overlapping peptides. Peptide 3, containing three unique lysine-triplet sequences, displayed the greatest binding to osteoblastic cultures; peptides containing fewer lysine triplet motifs or derived from the "6a" exon yielded dramatically lower binding. Similar results were obtained with 6-carboxyfluorescein (FAM)-conjugated peptides and western blots containing extracts from osteoblastic cultures. Mass spectroscopic mapping demonstrated that FAM-peptide 3 bound to 90 kDa BSP and its 18 to 60 kDa fragments, as well as to 110 kDa nucleolin. In osteoblastic cultures, FAM-peptide 3 localized to biomineralization foci (site of BSP) and to nucleoli (site of nucleolin). In bone sections, biotin-labeled peptide 3 bound to sites of new bone formation which were co-labeled with anti-BSP antibodies. These results establish the fluorescent peptide 3 conjugate as the first nonantibody-based method to identify BSP on western blots and in/on cells. Further examination of the "6b" splice variant interactions will likely reveal new insights into bone mineralization during development.
Collapse
Affiliation(s)
- Jeff P Gorski
- Center of Excellence in Mineralized Tissue Research, School of Dentistry, University of Missouri-Kansas City, Kansas City, Missouri, USA; Department of Oral and Craniofacial Sciences, School of Dentistry, University of Missouri-Kansas City, Kansas City, Missouri, USA.
| | - Nichole T Franz
- Center of Excellence in Mineralized Tissue Research, School of Dentistry, University of Missouri-Kansas City, Kansas City, Missouri, USA; Department of Oral and Craniofacial Sciences, School of Dentistry, University of Missouri-Kansas City, Kansas City, Missouri, USA
| | - Daniel Pernoud
- Department of Oral and Craniofacial Sciences, School of Dentistry, University of Missouri-Kansas City, Kansas City, Missouri, USA
| | - Andrew Keightley
- Department of Ophthalmology and Proteomics Core Facility, University of Missouri-Kansas City School of Medicine, Kansas City, Missouri, USA
| | - David R Eyre
- Department of Orthopaedics and Sports Medicine, University of Washington, Seattle, Washington, USA
| | - Julia Thom Oxford
- Department of Biological Sciences, Center of Biomedical Research Excellence in Matrix Biology, Boise State University, Boise, Idaho, USA
| |
Collapse
|
15
|
Bielajew BJ, Hu JC, Athanasiou KA. Collagen: quantification, biomechanics, and role of minor subtypes in cartilage. NATURE REVIEWS. MATERIALS 2020; 5:730-747. [PMID: 33996147 PMCID: PMC8114887 DOI: 10.1038/s41578-020-0213-1] [Citation(s) in RCA: 111] [Impact Index Per Article: 27.8] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 05/28/2020] [Indexed: 05/02/2023]
Abstract
Collagen is a ubiquitous biomaterial in vertebrate animals. Although each of its 28 subtypes contributes to the functions of many different tissues in the body, most studies on collagen or collagenous tissues have focussed on only one or two subtypes. With recent developments in analytical chemistry, especially mass spectrometry, significant advances have been made toward quantifying the different collagen subtypes in various tissues; however, high-throughput and low-cost methods for collagen subtype quantification do not yet exist. In this Review, we introduce the roles of collagen subtypes and crosslinks, and describe modern assays that enable a deep understanding of tissue physiology and disease states. Using cartilage as a model tissue, we describe the roles of major and minor collagen subtypes in detail; discuss known and unknown structure-function relationships; and show how tissue engineers may harness the functional characteristics of collagen to engineer robust neotissues.
Collapse
Affiliation(s)
- Benjamin J. Bielajew
- Department of Biomedical Engineering, University of California, Irvine, Irvine, CA 92617, USA
| | - Jerry C. Hu
- Department of Biomedical Engineering, University of California, Irvine, Irvine, CA 92617, USA
| | - Kyriacos A. Athanasiou
- Department of Biomedical Engineering, University of California, Irvine, Irvine, CA 92617, USA
| |
Collapse
|
16
|
Yang H, Zhou J, Pan H, Wei W, Gao R, Zhou Q. Mesenchymal stem cells derived-exosomes as a new therapeutic strategy for acute soft tissue injury. Cell Biochem Funct 2020; 39:107-115. [PMID: 32638406 DOI: 10.1002/cbf.3570] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2020] [Revised: 05/26/2020] [Accepted: 05/31/2020] [Indexed: 12/15/2022]
Abstract
The study aimed to investigate the role of exosomes derived from rat bone marrow mesenchymal stem cells (rBMSCs) in acute soft tissue injury and its related mechanisms. Exosomes were isolated from rBMSCs and characterized by Nanosight NS300 particle size analyser (NTA), transmission electron microscopy (TEM), and western blot. Twenty four rats were randomly divided into four groups (n = 6): control group, strike group, rBMSCs group, and rBMSCs-exo group. Haematoxylin-eosin (HE) staining was used to observe the morphology. Real-time quantification PCR (RT-qPCR) and western blot were used to analyse the expression of IL-1A, IL-12A, COL11A1, COL4A4, and Wnt4. NTA, TEM and western blot results showed that exosomes isolated from rBMSCs were cup-shaped morphology with a size of about 100 nm. HE staining showed that there was severe soft tissue inflammation in strike group, and the symptoms were alleviated after rBMSCs and rBMSCs-exo treatment. RT-qPCR and western blot indicated that in the strike group, the expression levels of IL-1A and IL-12A were significantly increased, and their expressions were decreased markedly by exosomes treatment. In addition, after treatment, the expression levels of COL11A1 and Wnt4 were up-regulated, while the expression of COL4A4 was down-regulated. Exosomes isolated from rBMSCs could improve acute soft tissue injury, and may be used as a new therapeutic strategy acute soft tissue injury. SIGNIFICANCE OF THE STUDY: Acute soft tissue injury is a common clinical exercise injury, which has a significant impact on people's health and work ability. Exosomes have been attracting increasing attention as a media of cell-to-cell communication. This study showed that exosomes isolated from rBMSCs could improve acute soft tissue injury by inhibiting inflammatory response, regulating the levels of COL11A1 and COL4A4, and up-regulating the expression of Wnt4. These will provide a new therapy strategy of acute soft tissue injury, and improve our understanding of the occurrence and development in acute soft tissue injury.
Collapse
Affiliation(s)
- Hongchang Yang
- Physical Education Department, Hohai University, Nanjing, Jiangsu, China
| | - Jing Zhou
- Department of Clinical Medicine, Jiangsu Health Vocational College, Nanjing, Jiangsu, China
| | - Hongyao Pan
- Physical Education Department, Hohai University, Nanjing, Jiangsu, China
| | - Wei Wei
- Physical Education Department, Hohai University, Nanjing, Jiangsu, China
| | - Run Gao
- The Department of Rehabilitation, Brain Hospital Affiliated to Nanjing Medical University, Nanjing, Jiangsu, China
| | - Qiang Zhou
- Physical Education Department, Hohai University, Nanjing, Jiangsu, China
| |
Collapse
|
17
|
Chen Y, Wu B, Lin J, Yu D, Du X, Sheng Z, Yu Y, An C, Zhang X, Li Q, Zhu S, Sun H, Zhang X, Zhang S, Zhou J, Bunpetch V, El-Hashash A, Ji J, Ouyang H. High-Resolution Dissection of Chemical Reprogramming from Mouse Embryonic Fibroblasts into Fibrocartilaginous Cells. Stem Cell Reports 2020; 14:478-492. [PMID: 32084387 PMCID: PMC7066361 DOI: 10.1016/j.stemcr.2020.01.013] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2019] [Revised: 01/23/2020] [Accepted: 01/23/2020] [Indexed: 01/20/2023] Open
Abstract
Articular cartilage injury and degeneration causing pain and loss of quality-of-life has become a serious problem for increasingly aged populations. Given the poor self-renewal of adult human chondrocytes, alternative functional cell sources are needed. Direct reprogramming by small molecules potentially offers an oncogene-free and cost-effective approach to generate chondrocytes, but has yet to be investigated. Here, we directly reprogrammed mouse embryonic fibroblasts into PRG4+ chondrocytes using a 3D system with a chemical cocktail, VCRTc (valproic acid, CHIR98014, Repsox, TTNPB, and celecoxib). Using single-cell transcriptomics, we revealed the inhibition of fibroblast features and activation of chondrogenesis pathways in early reprograming, and the intermediate cellular process resembling cartilage development. The in vivo implantation of chemical-induced chondrocytes at defective articular surfaces promoted defect healing and rescued 63.4% of mechanical function loss. Our approach directly converts fibroblasts into functional cartilaginous cells, and also provides insights into potential pharmacological strategies for future cartilage regeneration. A chemical method to derive functional murine articular chondrocytes from fibroblasts Chemical-induced chondrocytes promote in vivo regeneration of articular defects In single-cell analysis, intermediate reprogramming events resemble cartilage development
Collapse
Affiliation(s)
- Yishan Chen
- Department of Orthopaedic Surgery, Second Affiliated Hospital and Zhejiang University-University of Edinburgh Institute and School of Basic Medicine, Zhejiang University School of Medicine, Hangzhou 310058, China; Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cells and Regenerative Medicine, Zhejiang University School of Medicine, Hangzhou 310058, China; Key Laboratory of Tissue Engineering and Regenerative Medicine of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Bingbing Wu
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cells and Regenerative Medicine, Zhejiang University School of Medicine, Hangzhou 310058, China; Key Laboratory of Tissue Engineering and Regenerative Medicine of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Junxin Lin
- Department of Orthopaedic Surgery, Second Affiliated Hospital and Zhejiang University-University of Edinburgh Institute and School of Basic Medicine, Zhejiang University School of Medicine, Hangzhou 310058, China; Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cells and Regenerative Medicine, Zhejiang University School of Medicine, Hangzhou 310058, China; Key Laboratory of Tissue Engineering and Regenerative Medicine of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Dongsheng Yu
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cells and Regenerative Medicine, Zhejiang University School of Medicine, Hangzhou 310058, China; Key Laboratory of Tissue Engineering and Regenerative Medicine of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Xiaotian Du
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cells and Regenerative Medicine, Zhejiang University School of Medicine, Hangzhou 310058, China; Key Laboratory of Tissue Engineering and Regenerative Medicine of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Zixuan Sheng
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cells and Regenerative Medicine, Zhejiang University School of Medicine, Hangzhou 310058, China; Key Laboratory of Tissue Engineering and Regenerative Medicine of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Yeke Yu
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cells and Regenerative Medicine, Zhejiang University School of Medicine, Hangzhou 310058, China; Key Laboratory of Tissue Engineering and Regenerative Medicine of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Chengrui An
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cells and Regenerative Medicine, Zhejiang University School of Medicine, Hangzhou 310058, China; Key Laboratory of Tissue Engineering and Regenerative Medicine of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Xiaoan Zhang
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cells and Regenerative Medicine, Zhejiang University School of Medicine, Hangzhou 310058, China; Key Laboratory of Tissue Engineering and Regenerative Medicine of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Qikai Li
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cells and Regenerative Medicine, Zhejiang University School of Medicine, Hangzhou 310058, China; Key Laboratory of Tissue Engineering and Regenerative Medicine of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Shouan Zhu
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cells and Regenerative Medicine, Zhejiang University School of Medicine, Hangzhou 310058, China; Key Laboratory of Tissue Engineering and Regenerative Medicine of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Heng Sun
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cells and Regenerative Medicine, Zhejiang University School of Medicine, Hangzhou 310058, China; Key Laboratory of Tissue Engineering and Regenerative Medicine of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Xianzhu Zhang
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cells and Regenerative Medicine, Zhejiang University School of Medicine, Hangzhou 310058, China; Key Laboratory of Tissue Engineering and Regenerative Medicine of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Shufang Zhang
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cells and Regenerative Medicine, Zhejiang University School of Medicine, Hangzhou 310058, China; Key Laboratory of Tissue Engineering and Regenerative Medicine of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou 310058, China; China Orthopedic Regenerative Medicine Group (CORMed), Hangzhou 310058, China
| | - Jing Zhou
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cells and Regenerative Medicine, Zhejiang University School of Medicine, Hangzhou 310058, China; Key Laboratory of Tissue Engineering and Regenerative Medicine of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Varitsara Bunpetch
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cells and Regenerative Medicine, Zhejiang University School of Medicine, Hangzhou 310058, China; Key Laboratory of Tissue Engineering and Regenerative Medicine of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Ahmed El-Hashash
- Department of Orthopaedic Surgery, Second Affiliated Hospital and Zhejiang University-University of Edinburgh Institute and School of Basic Medicine, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Junfeng Ji
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cells and Regenerative Medicine, Zhejiang University School of Medicine, Hangzhou 310058, China; Key Laboratory of Tissue Engineering and Regenerative Medicine of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Hongwei Ouyang
- Department of Orthopaedic Surgery, Second Affiliated Hospital and Zhejiang University-University of Edinburgh Institute and School of Basic Medicine, Zhejiang University School of Medicine, Hangzhou 310058, China; Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cells and Regenerative Medicine, Zhejiang University School of Medicine, Hangzhou 310058, China; Key Laboratory of Tissue Engineering and Regenerative Medicine of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou 310058, China; Department of Sports Medicine, Zhejiang University School of Medicine, Hangzhou 310058, China; China Orthopedic Regenerative Medicine Group (CORMed), Hangzhou 310058, China.
| |
Collapse
|
18
|
Johanson Z, Martin K, Fraser G, James K. The Synarcual of the Little Skate, Leucoraja erinacea: Novel Development Among the Vertebrates. Front Ecol Evol 2019. [DOI: 10.3389/fevo.2019.00012] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
|
19
|
Seegmiller RE, Foster C, Burnham JL. Understanding chondrodysplasia (cho): A comprehensive review of cho as an animal model of birth defects, disorders, and molecular mechanisms. Birth Defects Res 2019; 111:237-247. [PMID: 30719872 DOI: 10.1002/bdr2.1473] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2019] [Accepted: 01/18/2019] [Indexed: 11/11/2022]
Abstract
BACKGROUND The mutant chondrodysplasia (cho) is a cartilage-targeting disorder in C57BL mice that results in dwarfing and other malformations stemming from this collagenopathy. Clarke Fraser made the discovery of the mutation accidentally in the early 1960s during the thalidomide tragedy. METHODS For this review we identified key research on cho as since its discovery. Relevant data were compiled to make a comprehensive review that details discoveries associated with the cho mutation, that describes the associated phenotypes and molecular mechanisms, and that provides a discussion surrounding its current clinical relevance. RESULTS Mechanistically, cho acts by hindering chondrogenesis and endochondral bone formation. The phenotype results from a 1-nt deletion in the gene encoding the alpha 1 chain of type XI collagen. For more than half a century, researchers have studied the pathogenesis of the cho mutation in relation to a variety of mouse models of human birth defects and disease. These studies have resulted in several discoveries linking cho with such human disorders as dwarfism, tracheal stenosis, cleft palate, pulmonary hypoplasia, and osteoarthritis (OA). CONCLUSION The study of cho has led to numerous advances in understanding human birth defects, congenital disorders, and adult human disease. The most recent studies have suggested a role for the TGF-Beta, HtrA1, Ddr2, and Mmp-13 pathway in the degradation of articular cartilage and the development of OA in cho/+ mice. We have shown that the anti-hypertension drug Losartan is a TGF-Beta blocker that could be used to treat OA in Stickler syndrome, and thereby rescue the WT phenotype.
Collapse
Affiliation(s)
- Robert E Seegmiller
- Department of Physiology and Developmental Biology, Brigham Young University, Provo, Utah
| | - Cameron Foster
- Department of Physiology and Developmental Biology, Brigham Young University, Provo, Utah
| | - Jared L Burnham
- Department of Physiology and Developmental Biology, Brigham Young University, Provo, Utah
| |
Collapse
|
20
|
Seegmiller RE, Cook N, Goodwin K, Leishman T, Graf M. Assessment of Gross Fetal Malformations: The Modernized Wilson Technique and Skeletal Staining. Methods Mol Biol 2019; 1965:421-434. [PMID: 31069690 DOI: 10.1007/978-1-4939-9182-2_27] [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] [Indexed: 12/13/2022]
Abstract
Teratology is the study of anatomical and physiological abnormalities, commonly known as birth defects. If an embryo is exposed to a harmful substance, or teratogen, during the critical period of development, an ensuing malformation may occur. These malformations and their associated mechanisms are studied and analyzed in laboratory animals in order to prevent them from occurring in humans. Rodents such as rats and mice have commonly been used in such studies because of their similarity to humans. In 1959, James G. Wilson designed, developed, and tested a protocol on how to observe and analyze structural malformations in rodent fetuses, which included: external examination, skeletal evaluation, soft tissue analysis, and data collection/analysis. For standardization purposes, i.e., to normalize findings from one lab to another, it is important that this protocol be followed with precision. Although many years have passed since Wilson initially created this protocol, it is still widely used to this day, and only minor changes have been made to his instructions such as the chemical reagents used in the experiments and methods of analysis of the experimental data. Such testing has resulted in major advances in the dissemination of teratology information, including the identification of an increasing number of teratogens and the understanding of the pathogenesis of birth defects. While mechanistically birth defect prevention will include the understanding of individual genomes and pharmacogenomics, overall, morphological assessment will still be required as an integral part of birth defects research. As the interaction between teratogenic and genetic factors is better understood, it is anticipated that the incidence of most types of defects will substantially be reduced.
Collapse
Affiliation(s)
- Robert E Seegmiller
- Department of Physiology and Developmental Biology, College of Life Sciences, Brigham Young University, Provo, UT, USA.
| | - Nathan Cook
- Department of Physiology and Developmental Biology, College of Life Sciences, Brigham Young University, Provo, UT, USA
| | - Korance Goodwin
- Department of Physiology and Developmental Biology, College of Life Sciences, Brigham Young University, Provo, UT, USA
| | - Timothy Leishman
- Department of Physiology and Developmental Biology, College of Life Sciences, Brigham Young University, Provo, UT, USA
| | - Michelle Graf
- Department of Physiology and Developmental Biology, College of Life Sciences, Brigham Young University, Provo, UT, USA
| |
Collapse
|
21
|
Abstract
Osteochondral (OC) lesions are a major cause of chronic musculoskeletal pain and functional disability, which reduces the quality of life of the patients and entails high costs to the society. Currently, there are no effective treatments, so in vitro and in vivo disease models are critically important to obtain knowledge about the causes and to develop effective treatments for OC injuries. In vitro models are essential to clarify the causes of the disease and the subsequent design of the first barrier to test potential therapeutics. On the other hand, in vivo models are anatomically more similar to humans allowing to reproduce the pattern and progression of the lesion in a controlled scene and offering the opportunity to study the symptoms and responses to new treatments. Moreover, in vivo models are the most suitable preclinical model, being a fundamental and a mandatory step to ensure the successful transfer to clinical trials. Both in vitro and in vitro models have a number of advantages and limitation, and the choice of the most appropriate model for each study depends on many factors, such as the purpose of the study, handling or the ease to obtain, and cost, among others. In this chapter, we present the main in vitro and in vivo OC disease models that have been used over the years in the study of origin, progress, and treatment approaches of OC defects.
Collapse
|
22
|
Lawrence EA, Kague E, Aggleton JA, Harniman RL, Roddy KA, Hammond CL. The mechanical impact of col11a2 loss on joints; col11a2 mutant zebrafish show changes to joint development and function, which leads to early-onset osteoarthritis. Philos Trans R Soc Lond B Biol Sci 2018; 373:rstb.2017.0335. [PMID: 30249781 PMCID: PMC6158203 DOI: 10.1098/rstb.2017.0335] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/03/2018] [Indexed: 12/12/2022] Open
Abstract
Collagen is the major structural component of cartilage, and mutations in the genes encoding type XI collagen are associated with severe skeletal dysplasias (fibrochondrogenesis and Stickler syndrome) and early-onset osteoarthritis (OA). The impact of the lack of type XI collagen on cell behaviour and mechanical performance during skeleton development is unknown. We studied a zebrafish mutant for col11a2 and evaluated cartilage, bone development and mechanical properties to address this. We show that in col11a2 mutants, type II collagen is made but is prematurely degraded in maturing cartilage and ectopically expressed in the joint. These changes are correlated with increased stiffness of both bone and cartilage; quantified using atomic force microscopy. In the mutants, the skeletal rudiment terminal region in the jaw joint is broader and the interzone smaller. These differences in shape and material properties impact on joint function and mechanical performance, which we modelled using finite element analyses. Finally, we show that col11a2 heterozygous carriers reach adulthood but show signs of severe early-onset OA. Taken together, our data demonstrate a key role for type XI collagen in maintaining the properties of cartilage matrix; which when lost leads to alterations to cell behaviour that give rise to joint pathologies.This article is part of the Theo Murphy meeting issue 'Mechanics of development'.
Collapse
Affiliation(s)
- Elizabeth A Lawrence
- School of Physiology, Pharmacology and Neuroscience, University of Bristol, Bristol BS8 1TD, UK
| | - Erika Kague
- School of Physiology, Pharmacology and Neuroscience, University of Bristol, Bristol BS8 1TD, UK
| | - Jessye A Aggleton
- School of Anthropology and Archaeology, University of Bristol, Bristol BS8 1UU, UK
| | | | - Karen A Roddy
- School of Physiology, Pharmacology and Neuroscience, University of Bristol, Bristol BS8 1TD, UK
| | - Chrissy L Hammond
- School of Physiology, Pharmacology and Neuroscience, University of Bristol, Bristol BS8 1TD, UK
| |
Collapse
|
23
|
Holyoak DT, Otero M, Armar NS, Ziemian SN, Otto A, Cullinane D, Wright TM, Goldring SR, Goldring MB, van der Meulen MC. Collagen XI mutation lowers susceptibility to load-induced cartilage damage in mice. J Orthop Res 2018; 36:711-720. [PMID: 28898438 PMCID: PMC8813548 DOI: 10.1002/jor.23731] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/01/2017] [Accepted: 08/31/2017] [Indexed: 02/04/2023]
Abstract
Interactions among risk factors for osteoarthritis (OA) are not well understood. We investigated the combined impact of two prevalent risk factors: mechanical loading and genetically abnormal cartilage tissue properties. We used cyclic tibial compression to simulate mechanical loading in the cho/+ (Col11a1 haploinsufficient) mouse, which has abnormal collagen fibrils in cartilage due to a point mutation in the Col11a1 gene. We hypothesized that the mutant collagen would not alter phenotypic bone properties and that cho/+ mice, which develop early onset OA, would develop enhanced load-induced cartilage damage compared to their littermates. To test our hypotheses, we applied cyclic compression to the left tibiae of 6-month-old cho/+ male mice and wild-type (WT) littermates for 1, 2, and 6 weeks at moderate (4.5 N) and high (9.0 N) peak load magnitudes. We then characterized load-induced cartilage and bone changes by histology, microcomputed tomography, and immunohistochemistry. Prior to loading, cho/+ mice had less dense, thinner cortical bone compared to WT littermates. In addition, in loaded and non-loaded limbs, cho/+ mice had thicker cartilage. With high loads, cho/+ mice experienced less load-induced cartilage damage at all time points and displayed decreased matrix metalloproteinase (MMP)-13 levels compared to WT littermates. The thinner, less dense cortical bone and thicker cartilage were unexpected and may have contributed to the reduced severity of load-induced cartilage damage in cho/+ mice. Furthermore, the spontaneous proteoglycan loss resulting from the mutant collagen XI was not additive to cartilage damage from mechanical loading, suggesting that these risk factors act through independent pathways. © 2017 Orthopaedic Research Society. Published by Wiley Periodicals, Inc. J Orthop Res 36:711-720, 2018.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Timothy M. Wright
- Cornell University, Ithaca, NY,Hospital for Special Surgery, New York, NY,Weill Cornell Medical College, New York, NY
| | - Steven R. Goldring
- Hospital for Special Surgery, New York, NY,Weill Cornell Medical College, New York, NY
| | - Mary B. Goldring
- Hospital for Special Surgery, New York, NY,Weill Cornell Medical College, New York, NY
| | | |
Collapse
|
24
|
Vogiatzi MG, Li D, Tian L, Garifallou JP, Kim CE, Hakonarson H, Levine MA. A novel dominant COL11A1 mutation in a child with Stickler syndrome type II is associated with recurrent fractures. Osteoporos Int 2018; 29:247-251. [PMID: 28971234 DOI: 10.1007/s00198-017-4229-3] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/10/2017] [Accepted: 09/14/2017] [Indexed: 10/18/2022]
Abstract
UNLABELLED This case describes a child with blindness, recurrent low-impact fractures, low bone mass, and intermittent joint pain who was found to have a novel missense mutation in COL11A1, consistent with Stickler syndrome type II. The case illustrates the phenotypic variability of the syndrome, which may include increased fragility in childhood. INTRODUCTION Stickler syndrome type II is an autosomal dominant disorder caused by mutations in the gene that encodes the type XI collagen chain α1 (COL11A1). Manifestations include craniofacial dysmorphology and ocular abnormalities that may lead to blindness, hearing loss, and skeletal anomalies that range from joint pain and arthritis to scoliosis and hypermobility. METHODS Herein, we describe a child who carried the presumed diagnosis of osteoporosis-pseudoglioma syndrome because of the combined findings of recurrent low-impact fractures due to low bone mass and blindness. The child also suffered from joint pain but had no facial dysmorphism or hearing loss. RESULTS Targeted sequencing and deletion analysis of the LRP5, COL1A1, and COL1A2 genes failed to identify any mutations, and whole exome sequence analysis revealed a novel missense mutation (c.3032C>A:p.P1011Q) in COL11A1, consistent with Stickler type II. CONCLUSION This case highlights the phenotypic variability of Stickler type II, broadens the list of differential diagnosis of increased bone fragility in childhood, and highlights utility of unbiased genetic testing towards establishing the correct diagnosis in children with frequent fractures.
Collapse
Affiliation(s)
- M G Vogiatzi
- Division of Endocrinology and Diabetes, Children's Hospital of Philadelphia, 3401 Civic Center Blvd., Suite 11NW 30, Philadelphia, PA, 19104, USA.
| | - D Li
- Center for Applied Genomics, The Children's Hospital of Philadelphia, Philadelphia, PA, 19104, USA
| | - L Tian
- Center for Applied Genomics, The Children's Hospital of Philadelphia, Philadelphia, PA, 19104, USA
| | - J P Garifallou
- Center for Applied Genomics, The Children's Hospital of Philadelphia, Philadelphia, PA, 19104, USA
| | - C E Kim
- Center for Applied Genomics, The Children's Hospital of Philadelphia, Philadelphia, PA, 19104, USA
| | - H Hakonarson
- Center for Applied Genomics, The Children's Hospital of Philadelphia, Philadelphia, PA, 19104, USA
| | - M A Levine
- Division of Endocrinology and Diabetes, Children's Hospital of Philadelphia, 3401 Civic Center Blvd., Suite 11NW 30, Philadelphia, PA, 19104, USA
| |
Collapse
|
25
|
Matrisome Profiling During Intervertebral Disc Development And Ageing. Sci Rep 2017; 7:11629. [PMID: 28912585 PMCID: PMC5599645 DOI: 10.1038/s41598-017-11960-0] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2017] [Accepted: 09/01/2017] [Indexed: 12/19/2022] Open
Abstract
Intervertebral disc (IVD) degeneration is often the cause of low back pain. Degeneration occurs with age and is accompanied by extracellular matrix (ECM) depletion, culminating in nucleus pulpous (NP) extrusion and IVD destruction. The changes that occur in the disc with age have been under investigation. However, a thorough study of ECM profiling is needed, to better understand IVD development and age-associated degeneration. As so, iTRAQ LC-MS/MS analysis of foetus, young and old bovine NPs, was performed to define the NP matrisome. The enrichment of Collagen XII and XIV in foetus, Fibronectin and Prolargin in elder NPs and Collagen XI in young ones was independently validated. This study provides the first matrisome database of healthy discs during development and ageing, which is key to determine the pathways and processes that maintain disc homeostasis. The factors identified may help to explain age-associated IVD degeneration or constitute putative effectors for disc regeneration.
Collapse
|
26
|
Shin SU, Lee J, Kim JH, Kim WH, Song SE, Chu A, Kim HS, Han W, Ryu HS, Moon WK. Gene expression profiling of calcifications in breast cancer. Sci Rep 2017; 7:11427. [PMID: 28900139 PMCID: PMC5595962 DOI: 10.1038/s41598-017-11331-9] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2017] [Accepted: 08/22/2017] [Indexed: 12/21/2022] Open
Abstract
We investigated the gene expression profiles of calcifications in breast cancer. Gene expression analysis of surgical specimen was performed using Affymetrix GeneChip® Human Gene 2.0 ST arrays in 168 breast cancer patients. The mammographic calcifications were reviewed by three radiologists and classified into three groups according to malignancy probability: breast cancers without suspicious calcifications; breast cancers with low-to-intermediate suspicious calcifications; and breast cancers with highly suspicious calcifications. To identify differentially expressed genes (DEGs) between these three groups, a one-way analysis of variance was performed with post hoc comparisons with Tukey's honest significant difference test. To explore the biological significance of DEGs, we used DAVID for gene ontology analysis and BioLattice for clustering analysis. A total of 2551 genes showed differential expression among the three groups. ERBB2 genes are up-regulated in breast cancers with highly suspicious calcifications (fold change 2.474, p < 0.001). Gene ontology analysis revealed that the immune, defense and inflammatory responses were decreased in breast cancers with highly suspicious calcifications compared to breast cancers without suspicious calcifications (p from 10-23 to 10-8). The clustering analysis also demonstrated that the immune system is associated with mammographic calcifications (p < 0.001). Our study showed calcifications in breast cancers are associated with high levels of mRNA expression of ERBB2 and decreased immune system activity.
Collapse
Affiliation(s)
- Sung Ui Shin
- Department of Radiology, Seoul National University Hospital and Seoul National University College of Medicine, Seoul, Korea
| | - Jeonghoon Lee
- Division of Biomedical Informatics, Seoul National University College of Medicine, Seoul, Korea
| | - Ju Han Kim
- Division of Biomedical Informatics, Seoul National University College of Medicine, Seoul, Korea
| | - Won Hwa Kim
- Department of Radiology, Seoul National University Hospital and Seoul National University College of Medicine, Seoul, Korea
| | - Sung Eun Song
- Department of Radiology, Seoul National University Hospital and Seoul National University College of Medicine, Seoul, Korea
| | - Ajung Chu
- Department of Radiology, Seoul National University Hospital and Seoul National University College of Medicine, Seoul, Korea
| | - Hoe Suk Kim
- Department of Radiology, Seoul National University Hospital and Seoul National University College of Medicine, Seoul, Korea
| | - Wonshik Han
- Department of Surgery, Seoul National University Hospital, Seoul, Korea
| | - Han Suk Ryu
- Department of Pathology, Seoul National University Hospital, Seoul, Korea
| | - Woo Kyung Moon
- Department of Radiology, Seoul National University Hospital and Seoul National University College of Medicine, Seoul, Korea.
| |
Collapse
|
27
|
Hughes A, Oxford AE, Tawara K, Jorcyk CL, Oxford JT. Endoplasmic Reticulum Stress and Unfolded Protein Response in Cartilage Pathophysiology; Contributing Factors to Apoptosis and Osteoarthritis. Int J Mol Sci 2017; 18:ijms18030665. [PMID: 28335520 PMCID: PMC5372677 DOI: 10.3390/ijms18030665] [Citation(s) in RCA: 75] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2017] [Revised: 03/15/2017] [Accepted: 03/16/2017] [Indexed: 12/11/2022] Open
Abstract
Chondrocytes of the growth plate undergo apoptosis during the process of endochondral ossification, as well as during the progression of osteoarthritis. Although the regulation of this process is not completely understood, alterations in the precisely orchestrated programmed cell death during development can have catastrophic results, as exemplified by several chondrodystrophies which are frequently accompanied by early onset osteoarthritis. Understanding the mechanisms that underlie chondrocyte apoptosis during endochondral ossification in the growth plate has the potential to impact the development of therapeutic applications for chondrodystrophies and associated early onset osteoarthritis. In recent years, several chondrodysplasias and collagenopathies have been recognized as protein-folding diseases that lead to endoplasmic reticulum stress, endoplasmic reticulum associated degradation, and the unfolded protein response. Under conditions of prolonged endoplasmic reticulum stress in which the protein folding load outweighs the folding capacity of the endoplasmic reticulum, cellular dysfunction and death often occur. However, unfolded protein response (UPR) signaling is also required for the normal maturation of chondrocytes and osteoblasts. Understanding how UPR signaling may contribute to cartilage pathophysiology is an essential step toward therapeutic modulation of skeletal disorders that lead to osteoarthritis.
Collapse
Affiliation(s)
- Alexandria Hughes
- Department of Biological Sciences, Boise State University, Boise, ID 83725, USA.
- Biomolecular Research Center, Boise State University, Boise, ID 83725, USA.
| | - Alexandra E Oxford
- Department of Biological Sciences, Boise State University, Boise, ID 83725, USA.
- Biomolecular Research Center, Boise State University, Boise, ID 83725, USA.
| | - Ken Tawara
- Biomolecular Sciences Graduate Program, Boise State University, Boise, ID 83725, USA.
- Biomolecular Research Center, Boise State University, Boise, ID 83725, USA.
| | - Cheryl L Jorcyk
- Department of Biological Sciences, Boise State University, Boise, ID 83725, USA.
- Biomolecular Sciences Graduate Program, Boise State University, Boise, ID 83725, USA.
- Biomolecular Research Center, Boise State University, Boise, ID 83725, USA.
| | - Julia Thom Oxford
- Department of Biological Sciences, Boise State University, Boise, ID 83725, USA.
- Biomolecular Sciences Graduate Program, Boise State University, Boise, ID 83725, USA.
- Biomolecular Research Center, Boise State University, Boise, ID 83725, USA.
| |
Collapse
|
28
|
Dluzen DF, Noren Hooten N, Evans MK. Extracellular RNA in aging. WILEY INTERDISCIPLINARY REVIEWS-RNA 2016; 8. [PMID: 27531497 DOI: 10.1002/wrna.1385] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/13/2016] [Revised: 07/07/2016] [Accepted: 07/09/2016] [Indexed: 12/16/2022]
Abstract
Since the discovery of extracellular RNA (exRNA) in circulation and other bodily fluids, there has been considerable effort to catalog and assess whether exRNAs can be used as markers for health and disease. A variety of exRNA species have been identified including messenger RNA and noncoding RNA such as microRNA (miRNA), small nucleolar RNA, transfer RNA, and long noncoding RNA. Age-related changes in exRNA abundance have been observed, and it is likely that some of these transcripts play a role in aging. In this review, we summarize the current state of exRNA profiling in various body fluids and discuss age-related changes in exRNA abundance that have been identified in humans and other model organisms. miRNAs, in particular, are a major focus of current research and we will highlight and discuss the potential role that specific miRNAs might play in age-related phenotypes and disease. We will also review challenges facing this emerging field and various strategies that can be used for the validation and future use of exRNAs as markers of aging and age-related disease. WIREs RNA 2017, 8:e1385. doi: 10.1002/wrna.1385 For further resources related to this article, please visit the WIREs website.
Collapse
Affiliation(s)
- Douglas F Dluzen
- Laboratory of Epidemiology and Population Sciences, National Institute on Aging, National Institutes of Health, Baltimore, MD, USA
| | - Nicole Noren Hooten
- Laboratory of Epidemiology and Population Sciences, National Institute on Aging, National Institutes of Health, Baltimore, MD, USA
| | - Michele K Evans
- Laboratory of Epidemiology and Population Sciences, National Institute on Aging, National Institutes of Health, Baltimore, MD, USA
| |
Collapse
|