1
|
Dong W, Yang C, Guo D, Jia M, Wang Y, Wang J. PTX3-assembled pericellular hyaluronan matrix enhances endochondral ossification during fracture healing and heterotopic ossification. Bone 2025; 192:117385. [PMID: 39732447 DOI: 10.1016/j.bone.2024.117385] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/11/2024] [Revised: 12/23/2024] [Accepted: 12/24/2024] [Indexed: 12/30/2024]
Abstract
Endochondral ossification (EO) is a pivotal process during fracture healing and traumatic heterotopic ossification (HO), involving the cartilaginous matrix synthesis and mineralization. Unlike the extracellular matrix, the hyaluronan (HA)-rich pericellular matrix (PCM) directly envelops chondrocytes, serving as the frontline for extracellular signal reception and undergoing dynamic remodeling. Pentraxin 3 (PTX3), a secreted glycoprotein, facilitates HA matrix assembly and remodeling. However, it remains unclear whether PTX3 affects EO by regulating HA-rich PCM assembly of chondrocytes, thereby impacting fracture healing and traumatic HO. This study demonstrates that PTX3 deficiency impairs fracture healing and inhibits traumatic HO, but dose not affect growth plate development in mice. PTX3 expression is up-regulated during chondrocyte matrix synthesis and maturation and is localized in the HA-rich PCM. PTX3 promotes the assembly of HA-rich PCM in a serum- and TSG6-dependent manner, fostering CD44 receptor clustering, activating the FAK/AKT signaling pathway, and promoting chondrocyte matrix synthesis and maturation. Local injection of PTX3/TSG6 matrix protein mixture effectively promotes fracture healing in mice. In conclusion, PTX3-assembled HA-rich PCM promotes chondrocyte matrix synthesis and maturation via CD44/FAK/AKT signaling. This mechanism facilitates EO during fracture healing and traumatic HO in mice.
Collapse
Affiliation(s)
- Wei Dong
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan 430079, China
| | - Chang Yang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan 430079, China
| | - Donghua Guo
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan 430079, China
| | - Meie Jia
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan 430079, China
| | - Yan Wang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan 430079, China
| | - Jiawei Wang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan 430079, China.
| |
Collapse
|
2
|
Jahan N, Islam S, Sivasundaram K, Ota A, Naito M, Kuroda J, Watanabe H. Role of versican in extracellular matrix formation: analysis in 3D culture. Am J Physiol Cell Physiol 2025; 328:C245-C257. [PMID: 39656505 DOI: 10.1152/ajpcell.00495.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2024] [Revised: 10/28/2024] [Accepted: 11/20/2024] [Indexed: 01/01/2025]
Abstract
Three-dimensional (3-D) cell culture creates an environment that allows cells to grow and interact with the surrounding extracellular framework. Versican plays a pivotal role in forming the provisional matrix, but it is still unclear how this proteoglycan affects the formation of the extracellular matrix. Here, we established a 3-D culture system using fibrin gel, which enables a long-term culture up to a month. With this system, we characterized fibroblasts obtained from the newborn knock-in homozygotes, termed R/R, expressing a disintegrin and metalloproteinase with thrombospondin motif (ADAMTS)-resistant versican and wild-type mice. R/R fibroblasts showed higher levels of versican deposition than wild-type, demonstrating that the initial ADAMTS-cleavage site is involved in versican turnover. These fibroblasts exhibited faster proliferation and myofibroblastic differentiation, concomitant with higher levels of transforming growth factor β-signaling. R/R fibroblast culture had higher deposition levels of fibronectin, type I and V collagens, and fibrillin-1, especially at the late stages of culture. These results suggest that versican expressed by dermal fibroblasts facilitates the extracellular matrix formation, at least by affecting fibroblast behavior.NEW & NOTEWORTHY We established a 3-D-culture system useful for analyzing fibroblast behavior and matrix formation. The initial cleavage site by ADAMTSs in versican core protein is mainly involved in versican turnover. Accumulating versican facilitates fibroblast proliferation and myofibroblastic differentiation in an autocrine or paracrine manner. Accumulating versican promotes the deposition of fibronectin and collagens.
Collapse
Affiliation(s)
- Nushrat Jahan
- Institute for Molecular Science of Medicine, Aichi Medical University, Nagakute, Japan
| | - Shamima Islam
- Institute for Molecular Science of Medicine, Aichi Medical University, Nagakute, Japan
| | - Karnan Sivasundaram
- Department of Biochemistry, School of Medicine, Aichi Medical University, Nagakute, Japan
| | - Akinobu Ota
- Department of Biochemistry, School of Medicine, Aichi Medical University, Nagakute, Japan
| | - Munekazu Naito
- Department of Anatomy, School of Medicine, Aichi Medical University, Nagakute, Japan
| | - Junpei Kuroda
- Laboratory of Pattern Formation, Graduate School of Frontier Biosciences, Osaka University, Suita, Japan
| | - Hideto Watanabe
- Institute for Molecular Science of Medicine, Aichi Medical University, Nagakute, Japan
| |
Collapse
|
3
|
Arbee S, Himeno T, Miura-Yura E, Kato M, Islam S, Jahan N, Kamiya H, Watanabe H. Versican maintains the homeostasis of adipose tissues and regulates energy metabolism. Biochem Biophys Res Commun 2024; 727:150309. [PMID: 38936224 DOI: 10.1016/j.bbrc.2024.150309] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Revised: 06/14/2024] [Accepted: 06/21/2024] [Indexed: 06/29/2024]
Abstract
Versican is a large chondroitin sulfate proteoglycan in the extracellular matrix. It plays a pivotal role in the formation of the provisional matrix. S100a4, previously known as fibroblast-specific protein, functions as a calcium channel-binding protein. To investigate the role of versican expressed in fibroblasts, we generated conditional knockout mice in which versican expression is deleted in cells expressing S100a4. We found that S100a4 is expressed in adipose tissues, and these mice exhibit obesity under a normal diet, which becomes apparent as early as five months. The white adipose tissues of these mice exhibited decreased expression levels of S100a4 and versican and hypertrophy of adipocytes. qRT-PCR showed a reduced level of UCP1 in their white adipose tissues, indicating that the basic energy metabolism is diminished. These results suggest that versican in adipose tissues maintains the homeostasis of adipose tissues and regulates energy metabolism.
Collapse
Affiliation(s)
- Shahida Arbee
- Insitute for Molecular Science of Medicine, Aichi Medical University, 1-1 Yazakokarimata, Nagakute, Aichi, 480-1195, Japan
| | - Tatsuhito Himeno
- Division of Diabetes, Department of Internal Medicine, Aichi Medical University, 1-1 Yazakokarimata, Nagakute, Aichi, 480-1195, Japan
| | - Emiri Miura-Yura
- Division of Diabetes, Department of Internal Medicine, Aichi Medical University, 1-1 Yazakokarimata, Nagakute, Aichi, 480-1195, Japan
| | - Makoto Kato
- Division of Diabetes, Department of Internal Medicine, Aichi Medical University, 1-1 Yazakokarimata, Nagakute, Aichi, 480-1195, Japan
| | - Shamima Islam
- Insitute for Molecular Science of Medicine, Aichi Medical University, 1-1 Yazakokarimata, Nagakute, Aichi, 480-1195, Japan
| | - Nushrat Jahan
- Insitute for Molecular Science of Medicine, Aichi Medical University, 1-1 Yazakokarimata, Nagakute, Aichi, 480-1195, Japan
| | - Hideki Kamiya
- Division of Diabetes, Department of Internal Medicine, Aichi Medical University, 1-1 Yazakokarimata, Nagakute, Aichi, 480-1195, Japan
| | - Hideto Watanabe
- Insitute for Molecular Science of Medicine, Aichi Medical University, 1-1 Yazakokarimata, Nagakute, Aichi, 480-1195, Japan.
| |
Collapse
|
4
|
Mead TJ, Bhutada S, Foulcer SJ, Peruzzi N, Nelson CM, Seifert DE, Larkin J, Tran-Lundmark K, Filmus J, Apte SS. Combined genetic-pharmacologic inactivation of tightly linked ADAMTS proteases in temporally specific windows uncovers distinct roles for versican proteolysis and glypican-6 in cardiac development. Matrix Biol 2024; 131:1-16. [PMID: 38750698 PMCID: PMC11526477 DOI: 10.1016/j.matbio.2024.05.003] [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: 12/21/2023] [Revised: 05/07/2024] [Accepted: 05/09/2024] [Indexed: 05/22/2024]
Abstract
Extracellular matrix remodeling mechanisms are understudied in cardiac development and congenital heart defects. We show that matrix-degrading metalloproteases ADAMTS1 and ADAMTS5, are extensively co-expressed during mouse cardiac development. The mouse mutants of each gene have mild cardiac anomalies, however, their combined genetic inactivation to elicit cooperative roles is precluded by tight gene linkage. Therefore, we coupled Adamts1 inactivation with pharmacologic ADAMTS5 blockade to uncover stage-specific cooperative roles and investigated their potential substrates in mouse cardiac development. ADAMTS5 blockade was achieved in Adamts1 null mouse embryos using an activity-blocking monoclonal antibody during distinct developmental windows spanning myocardial compaction or cardiac septation and outflow tract rotation. Synchrotron imaging, RNA in situ hybridization, immunofluorescence microscopy and electron microscopy were used to determine the impact on cardiac development and compared to Gpc6 and ADAMTS-cleavage resistant versican mutants. Mass spectrometry-based N-terminomics was used to seek relevant substrates. Combined inactivation of ADAMTS1 and ADAMTS5 prior to 12.5 days of gestation led to dramatic accumulation of versican-rich cardiac jelly and inhibited formation of compact and trabecular myocardium, which was also observed in mice with ADAMTS cleavage-resistant versican. Combined inactivation after 12.5 days impaired outflow tract development and ventricular septal closure, generating a tetralogy of Fallot-like defect. N-terminomics of combined ADAMTS knockout and control hearts identified a cleaved glypican-6 peptide only in the controls. ADAMTS1 and ADAMTS5 expression in cells was associated with specific glypican-6 cleavages. Paradoxically, combined ADAMTS1 and ADAMTS5 inactivation reduced cardiac glypican-6 and outflow tract Gpc6 transcription. Notably, Gpc6-/- hearts demonstrated similar rotational defects as combined ADAMTS inactivated hearts and both had reduced hedgehog signaling. Thus, versican proteolysis in cardiac jelly at the canonical Glu441-Ala442 site is cooperatively mediated by ADAMTS1 and ADAMTS5 and required for proper ventricular cardiomyogenesis, whereas, reduced glypican-6 after combined ADAMTS inactivation impairs hedgehog signaling, leading to outflow tract malrotation.
Collapse
Affiliation(s)
- Timothy J Mead
- Department of Biomedical Engineering, Cleveland Clinic Lerner Research Institute, Cleveland, OH, USA; Department of Pediatrics, Case Western Reserve University School of Medicine, Cleveland, OH, USA; University Hospitals Rainbow Babies and Children's Hospital, Cleveland, OH, USA.
| | - Sumit Bhutada
- Department of Biomedical Engineering, Cleveland Clinic Lerner Research Institute, Cleveland, OH, USA
| | - Simon J Foulcer
- Department of Biomedical Engineering, Cleveland Clinic Lerner Research Institute, Cleveland, OH, USA
| | - Niccolò Peruzzi
- Department of Experimental Medical Science, and Wallenberg Center for Molecular Medicine Lund University and The Pediatric Heart Center, Skane University Hospital, Lund, Sweden
| | - Courtney M Nelson
- Department of Biomedical Engineering, Cleveland Clinic Lerner Research Institute, Cleveland, OH, USA
| | - Deborah E Seifert
- Department of Pediatrics, Case Western Reserve University School of Medicine, Cleveland, OH, USA
| | | | - Karin Tran-Lundmark
- Department of Experimental Medical Science, and Wallenberg Center for Molecular Medicine Lund University and The Pediatric Heart Center, Skane University Hospital, Lund, Sweden
| | - Jorge Filmus
- Sunnybrook Research Institute and Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada
| | - Suneel S Apte
- Department of Biomedical Engineering, Cleveland Clinic Lerner Research Institute, Cleveland, OH, USA.
| |
Collapse
|
5
|
Liu Y, Jia F, Li K, Liang C, Lin X, Geng W, Li Y. Critical signaling molecules in the temporomandibular joint osteoarthritis under different magnitudes of mechanical stimulation. Front Pharmacol 2024; 15:1419494. [PMID: 39055494 PMCID: PMC11269110 DOI: 10.3389/fphar.2024.1419494] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Accepted: 06/14/2024] [Indexed: 07/27/2024] Open
Abstract
The mechanical stress environment in the temporomandibular joint (TMJ) is constantly changing due to daily mandibular movements. Therefore, TMJ tissues, such as condylar cartilage, the synovial membrane and discs, are influenced by different magnitudes of mechanical stimulation. Moderate mechanical stimulation is beneficial for maintaining homeostasis, whereas abnormal mechanical stimulation leads to degeneration and ultimately contributes to the development of temporomandibular joint osteoarthritis (TMJOA), which involves changes in critical signaling molecules. Under abnormal mechanical stimulation, compensatory molecules may prevent degenerative changes while decompensatory molecules aggravate. In this review, we summarize the critical signaling molecules that are stimulated by moderate or abnormal mechanical loading in TMJ tissues, mainly in condylar cartilage. Furthermore, we classify abnormal mechanical stimulation-induced molecules into compensatory or decompensatory molecules. Our aim is to understand the pathophysiological mechanism of TMJ dysfunction more deeply in the ever-changing mechanical environment, and then provide new ideas for discovering effective diagnostic and therapeutic targets in TMJOA.
Collapse
Affiliation(s)
| | | | | | | | | | - Wei Geng
- Department of Dental Implant Center, Beijing Stomatological Hospital, School of Stomatology, Capital Medical University, Beijing, China
| | - Yanxi Li
- Department of Dental Implant Center, Beijing Stomatological Hospital, School of Stomatology, Capital Medical University, Beijing, China
| |
Collapse
|
6
|
Li MD, Lu JW, Zhang F, Lei WJ, Pan F, Lin YK, Ling LJ, Myatt L, Wang WS, Sun K. ADAMTS4 is a crucial proteolytic enzyme for versican cleavage in the amnion at parturition. Commun Biol 2024; 7:301. [PMID: 38461223 PMCID: PMC10924920 DOI: 10.1038/s42003-024-06007-w] [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: 09/27/2023] [Accepted: 03/03/2024] [Indexed: 03/11/2024] Open
Abstract
Hyalectan cleavage may play an important role in extracellular matrix remodeling. However, the proteolytic enzyme responsible for hyalectan degradation for fetal membrane rupture at parturition remains unknown. Here, we reveal that versican (VCAN) is the major hyalectan in the amnion, where its cleavage increases at parturition with spontaneous rupture of membrane. We further reveal that ADAMTS4 is a crucial proteolytic enzyme for VCAN cleavage in the amnion. Inflammatory factors may enhance VCAN cleavage by inducing ADAMTS4 expression and inhibiting ADAMTS4 endocytosis in amnion fibroblasts. In turn, versikine, the VCAN cleavage product, induces inflammatory factors in amnion fibroblasts, thereby forming a feedforward loop between inflammation and VCAN degradation. Mouse studies show that intra-amniotic injection of ADAMTS4 induces preterm birth along with increased VCAN degradation and proinflammatory factors abundance in the fetal membranes. Conclusively, there is enhanced VCAN cleavage by ADAMTS4 in the amnion at parturition, which can be reenforced by inflammation.
Collapse
Affiliation(s)
- Meng-Die Li
- Center for Reproductive Medicine, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, PR China
- Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Shanghai, PR China
| | - Jiang-Wen Lu
- Center for Reproductive Medicine, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, PR China
- Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Shanghai, PR China
| | - Fan Zhang
- Center for Reproductive Medicine, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, PR China
- Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Shanghai, PR China
| | - Wen-Jia Lei
- Center for Reproductive Medicine, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, PR China
- Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Shanghai, PR China
| | - Fan Pan
- Center for Reproductive Medicine, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, PR China
- Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Shanghai, PR China
| | - Yi-Kai Lin
- Center for Reproductive Medicine, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, PR China
- Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Shanghai, PR China
| | - Li-Jun Ling
- Department of Obstetrics, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai, PR China
| | - Leslie Myatt
- Department of Obstetrics and Gynecology, Oregon Health & Science University, Portland, OR, USA
| | - Wang-Sheng Wang
- Center for Reproductive Medicine, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, PR China.
- Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Shanghai, PR China.
| | - Kang Sun
- Center for Reproductive Medicine, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, PR China.
- Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Shanghai, PR China.
| |
Collapse
|
7
|
Sao K, Risbud MV. Proteoglycan Dysfunction: A Common Link Between Intervertebral Disc Degeneration and Skeletal Dysplasia. Neurospine 2024; 21:162-178. [PMID: 38569642 PMCID: PMC10992626 DOI: 10.14245/ns.2347342.671] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Revised: 02/04/2024] [Accepted: 02/23/2024] [Indexed: 04/05/2024] Open
Abstract
Proteoglycans through their sulfated glycosaminoglycans regulate cell-matrix signaling during tissue development, regeneration, and degeneration processes. Large extracellular proteoglycans such as aggrecan, versican, and perlecan are especially important for the structural integrity of the intervertebral disc and cartilage during development. In these tissues, proteoglycans are responsible for hydration, joint flexibility, and the absorption of mechanical loads. Loss or reduction of these molecules can lead to disc degeneration and skeletal dysplasia, evident from loss of disc height or defects in skeletal development respectively. In this review, we discuss the common proteoglycans found in the disc and cartilage and elaborate on various murine models and skeletal dysplasias in humans to highlight how their absence and/or aberrant expression causes accelerated disc degeneration and developmental defects.
Collapse
Affiliation(s)
- Kimheak Sao
- Graduate Program in Cell Biology and Regenerative Medicine, Jefferson College of Life Sciences, Thomas Jefferson University, Philadelphia, PA, USA
- Department of Orthopaedic Surgery, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA, USA
| | - Makarand V. Risbud
- Graduate Program in Cell Biology and Regenerative Medicine, Jefferson College of Life Sciences, Thomas Jefferson University, Philadelphia, PA, USA
- Department of Orthopaedic Surgery, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA, USA
| |
Collapse
|
8
|
Alcaide-Ruggiero L, Cugat R, Domínguez JM. Proteoglycans in Articular Cartilage and Their Contribution to Chondral Injury and Repair Mechanisms. Int J Mol Sci 2023; 24:10824. [PMID: 37446002 DOI: 10.3390/ijms241310824] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Revised: 06/17/2023] [Accepted: 06/23/2023] [Indexed: 07/15/2023] Open
Abstract
Proteoglycans are vital components of the extracellular matrix in articular cartilage, providing biomechanical properties crucial for its proper functioning. They are key players in chondral diseases, specifically in the degradation of the extracellular matrix. Evaluating proteoglycan molecules can serve as a biomarker for joint degradation in osteoarthritis patients, as well as assessing the quality of repaired tissue following different treatment strategies for chondral injuries. Despite ongoing research, understanding osteoarthritis and cartilage repair remains unclear, making the identification of key molecules essential for early diagnosis and effective treatment. This review offers an overview of proteoglycans as primary molecules in articular cartilage. It describes the various types of proteoglycans present in both healthy and damaged cartilage, highlighting their roles. Additionally, the review emphasizes the importance of assessing proteoglycans to evaluate the quality of repaired articular tissue. It concludes by providing a visual and narrative description of aggrecan distribution and presence in healthy cartilage. Proteoglycans, such as aggrecan, biglycan, decorin, perlecan, and versican, significantly contribute to maintaining the health of articular cartilage and the cartilage repair process. Therefore, studying these proteoglycans is vital for early diagnosis, evaluating the quality of repaired cartilage, and assessing treatment effectiveness.
Collapse
Affiliation(s)
- Lourdes Alcaide-Ruggiero
- Departamento de Medicina y Cirugía Animal, Facultad de Veterinaria, Universidad de Córdoba, Hospital Clínico Veterinario, Campus de Rabanales, Ctra. Madrid-Cádiz Km 396, 14014 Córdoba, Spain
- Fundación García-Cugat, Plaza Alfonso Comín 5-7, 08023 Barcelona, Spain
| | - Ramón Cugat
- Fundación García-Cugat, Plaza Alfonso Comín 5-7, 08023 Barcelona, Spain
- Instituto Cugat y Mutualidad de Futbolistas Españoles, Delegación Catalana, 08023 Barcelona, Spain
| | - Juan Manuel Domínguez
- Departamento de Medicina y Cirugía Animal, Facultad de Veterinaria, Universidad de Córdoba, Hospital Clínico Veterinario, Campus de Rabanales, Ctra. Madrid-Cádiz Km 396, 14014 Córdoba, Spain
- Fundación García-Cugat, Plaza Alfonso Comín 5-7, 08023 Barcelona, Spain
| |
Collapse
|
9
|
Scott AK, Casas E, Schneider SE, Swearingen AR, Van Den Elzen CL, Seelbinder B, Barthold JE, Kugel JF, Stern JL, Foster KJ, Emery NC, Brumbaugh J, Neu CP. Mechanical memory stored through epigenetic remodeling reduces cell therapeutic potential. Biophys J 2023; 122:1428-1444. [PMID: 36871159 PMCID: PMC10147835 DOI: 10.1016/j.bpj.2023.03.004] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2022] [Revised: 01/31/2023] [Accepted: 03/01/2023] [Indexed: 03/06/2023] Open
Abstract
Understanding how cells remember previous mechanical environments to influence their fate, or mechanical memory, informs the design of biomaterials and therapies in medicine. Current regeneration therapies, such as cartilage regeneration procedures, require 2D cell expansion processes to achieve large cell populations critical for the repair of damaged tissues. However, the limit of mechanical priming for cartilage regeneration procedures before inducing long-term mechanical memory following expansion processes is unknown, and mechanisms defining how physical environments influence the therapeutic potential of cells remain poorly understood. Here, we identify a threshold to mechanical priming separating reversible and irreversible effects of mechanical memory. After 16 population doublings in 2D culture, expression levels of tissue-identifying genes in primary cartilage cells (chondrocytes) are not recovered when transferred to 3D hydrogels, while expression levels of these genes were recovered for cells only expanded for eight population doublings. Additionally, we show that the loss and recovery of the chondrocyte phenotype correlates with a change in chromatin architecture, as shown by structural remodeling of the trimethylation of H3K9. Efforts to disrupt the chromatin architecture by suppressing or increasing levels of H3K9me3 reveal that only with increased levels of H3K9me3 did the chromatin architecture of the native chondrocyte phenotype partially return, along with increased levels of chondrogenic gene expression. These results further support the connection between the chondrocyte phenotype and chromatin architecture, and also reveal the therapeutic potential of inhibitors of epigenetic modifiers as disruptors of mechanical memory when large numbers of phenotypically suitable cells are required for regeneration procedures.
Collapse
Affiliation(s)
- Adrienne K Scott
- Paul M. Rady Department of Mechanical Engineering, University of Colorado Boulder, Boulder, Colorado
| | - Eduard Casas
- Department of Molecular, Cellular & Developmental Biology, University of Colorado Boulder, Boulder, Colorado
| | - Stephanie E Schneider
- Paul M. Rady Department of Mechanical Engineering, University of Colorado Boulder, Boulder, Colorado
| | - Alison R Swearingen
- Department of Molecular, Cellular & Developmental Biology, University of Colorado Boulder, Boulder, Colorado
| | - Courtney L Van Den Elzen
- Department of Ecology and Evolutionary Biology, University of Colorado Boulder, Boulder, Colorado
| | - Benjamin Seelbinder
- Paul M. Rady Department of Mechanical Engineering, University of Colorado Boulder, Boulder, Colorado
| | - Jeanne E Barthold
- Paul M. Rady Department of Mechanical Engineering, University of Colorado Boulder, Boulder, Colorado
| | - Jennifer F Kugel
- Department of Biochemistry, University of Colorado Boulder, Boulder, Colorado
| | - Josh Lewis Stern
- Department of Biochemistry, University of Colorado Boulder, Boulder, Colorado; Biochemistry and Molecular Genetics, O'Neal Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, Alabama
| | - Kyla J Foster
- Department of Biochemistry, University of Colorado Boulder, Boulder, Colorado
| | - Nancy C Emery
- Department of Ecology and Evolutionary Biology, University of Colorado Boulder, Boulder, Colorado
| | - Justin Brumbaugh
- Department of Molecular, Cellular & Developmental Biology, University of Colorado Boulder, Boulder, Colorado
| | - Corey P Neu
- Paul M. Rady Department of Mechanical Engineering, University of Colorado Boulder, Boulder, Colorado; Biomedical Engineering Program, University of Colorado Boulder, Boulder, Colorado; BioFrontiers Institute, University of Colorado Boulder, Boulder, Colorado.
| |
Collapse
|
10
|
Zhang H, Si P, Kong Q, Ma J. Transcriptome reveals the toxicity and genetic response of zebrafish to naphthenic acids and benzo[a]pyrene at ambient concentrations. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2023; 253:114700. [PMID: 36863161 DOI: 10.1016/j.ecoenv.2023.114700] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/16/2022] [Revised: 02/14/2023] [Accepted: 02/26/2023] [Indexed: 06/18/2023]
Abstract
Naphthenic acids (NAs) are typical contaminants in heavily crude oil. Benzo[a]pyrene (B[a]P) is also a component of crude oil, but their combined effects have not been systematically explored. In this study, zebrafish (Danio rerio) were used as the test organisms, and behavioral indicators and enzyme activities were used as toxicity indicators. Combined with the effects of environmental concentrations, the toxic effects of low concentrations of commercially available NAs (0.5 mg/LNA) and benzo[a]pyrene (0.8 μg/LBaP) at single and compound exposures (0.5 mg/LNA and 0.8 μg/LBaP) were assayed in zebrafish, and transcriptome sequencing technology was used to explore the molecular mechanism of the two compounds affecting zebrafish from the molecular biology level. Sensitive molecular markers that could indicate the presence of contaminants were screened. The results showed that (1) zebrafish in the NA and BaP exposure groups exhibited increased locomotor behavior, and the mixed exposure group exhibited inhibition of locomotor behavior. Oxidative stress biomarkers showed increased activity under single exposure and decreased activity under the mixed exposure. (2) NA stress led to changes in the activity of transporters and the intensity of energy metabolism; BaP directly stimulates the pathway of actin production. When the two compounds are combined, the excitability of neurons in the central nervous system is decreased, and the actin-related genes are down-regulated. (3) After BaP and Mix treatments, genes were enriched in the cytokine-receptor interaction and actin signal pathway, while NA increased the toxic effect on the mixed treatment group. In general, the interaction between NA and BaP has a synergistic effect on the transcription of zebrafish nerve and motor behavior-related genes, resulting in increased toxicity under combined exposure. The changes in expression of various zebrafish genes are manifested in the changes in the normal movement behavior of zebrafish and the intensification of oxidative stress in the apparent behavior and physiological indicators. CAPSULE ABSTRACT: We investigated the toxicity and genetic alterations caused by NA, B[a]P, and their mixtures in zebrafish in an aquatic environment using transcriptome sequencing technology and comprehensive behavioral analysis. These changes involved energy metabolism, the generation of muscle cells, and the nervous system.
Collapse
Affiliation(s)
- Huanxin Zhang
- College of Geography and Environment, Shandong Normal University, Jinan 250000, China.
| | - Panpan Si
- College of Geography and Environment, Shandong Normal University, Jinan 250000, China
| | - Qiang Kong
- College of Geography and Environment, Shandong Normal University, Jinan 250000, China
| | - Jinyue Ma
- College of Geography and Environment, Shandong Normal University, Jinan 250000, China
| |
Collapse
|
11
|
Yuan Y, Zhu Q, Yao X, Shi Z, Wen J. Maternal circulating metabolic biomarkers and their prediction performance for gestational diabetes mellitus related macrosomia. BMC Pregnancy Childbirth 2023; 23:113. [PMID: 36788507 PMCID: PMC9926775 DOI: 10.1186/s12884-023-05440-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Accepted: 02/09/2023] [Indexed: 02/16/2023] Open
Abstract
INTRODUCTION Gestational diabetes mellitus (GDM), a metabolism-related pregnancy complication, is significantly associated with an increased risk of macrosomia. We hypothesized that maternal circulating metabolic biomarkers differed between women with GDM and macrosomia (GDM-M) and women with GDM and normal neonatal weight (GDM-N), and had good prediction performance for GDM-M. METHODS Plasma samples from 44 GDM-M and 44 GDM-N were analyzed using Olink Proseek multiplex metabolism assay targeting 92 biomarkers. Combined different clinical characteristics and Olink markers, LASSO regression was used to optimize variable selection, and Logistic regression was applied to build a predictive model. Nomogram was developed based on the selected variables visually. Receiver operating characteristic (ROC) curve, calibration plot, and clinical impact curve were used to validate the model. RESULTS We found 4 metabolism-related biomarkers differing between groups [CLUL1 (Clusterin-like protein 1), VCAN (Versican core protein), FCRL1 (Fc receptor-like protein 1), RNASE3 (Eosinophil cationic protein), FDR < 0.05]. Based on the different clinical characteristics and Olink markers, a total of nine predictors, namely pre-pregnancy body mass index (BMI), weight gain at 24 gestational weeks (gw), parity, oral glucose tolerance test (OGTT) 2 h glucose at 24 gw, high-density lipoprotein (HDL) and low-density lipoprotein (LDL) at 24 gw, and plasma expression of CLUL1, VCAN and RNASE3 at 24 gw, were identified by LASSO regression. The model constructed using these 9 predictors displayed good prediction performance for GDM-M, with an area under the ROC of 0.970 (sensitivity = 0.955, specificity = 0.886), and was well calibrated (P Hosmer-Lemeshow test = 0.897). CONCLUSION The Model included pre-pregnancy BMI, weight gain at 24 gw, parity, OGTT 2 h glucose at 24 gw, HDL and LDL at 24 gw, and plasma expression of CLUL1, VCAN and RNASE3 at 24 gw had good prediction performance for predicting macrosomia in women with GDM.
Collapse
Affiliation(s)
- Yingdi Yuan
- grid.460072.7Department of Pediatrics, The First People’s Hospital of Lianyungang, Xuzhou Medical University Affiliated Hospital of Lianyungang (Lianyungang Clinical College of Nanjing Medical University), Lianyungang, China ,grid.459791.70000 0004 1757 7869Nanjing Maternity and Child Health Care Institute, Women’s Hospital of Nanjing Medical University, Nanjing Maternity and Child Health Care Hospital, Nanjing, China
| | - Qingyi Zhu
- grid.459791.70000 0004 1757 7869Nanjing Maternity and Child Health Care Institute, Women’s Hospital of Nanjing Medical University, Nanjing Maternity and Child Health Care Hospital, Nanjing, China ,grid.459791.70000 0004 1757 7869Department of Obstetrics, Women’s Hospital of Nanjing Medical University, Nanjing Maternity and Child Health Care Hospital, Nanjing, China
| | - Xiaodie Yao
- grid.459791.70000 0004 1757 7869Nanjing Maternity and Child Health Care Institute, Women’s Hospital of Nanjing Medical University, Nanjing Maternity and Child Health Care Hospital, Nanjing, China
| | - Zhonghua Shi
- Department of Obstetrics, Women's Hospital of Nanjing Medical University, Nanjing Maternity and Child Health Care Hospital, Nanjing, China.
| | - Juan Wen
- Nanjing Maternity and Child Health Care Institute, Women's Hospital of Nanjing Medical University, Nanjing Maternity and Child Health Care Hospital, Nanjing, China.
| |
Collapse
|
12
|
Sagae Y, Horie A, Yanai A, Ohara T, Nakakita B, Kitawaki Y, Okunomiya A, Tani H, Yamaguchi K, Hamanishi J, Lydon JP, Daikoku T, Watanabe H, Mandai M. Versican provides the provisional matrix for uterine spiral artery dilation and fetal growth. Matrix Biol 2023; 115:16-31. [PMID: 36423736 DOI: 10.1016/j.matbio.2022.11.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Revised: 11/17/2022] [Accepted: 11/20/2022] [Indexed: 11/23/2022]
Abstract
The extracellular matrix (ECM) in the endometrium plays a crucial role in mammalian pregnancy. We have shown that versican secreted from the endometrial epithelium promotes embryo implantation. Versican is a proteoglycan, a major player in the provisional matrix, and versikine, its N-terminal fragment cleaved by ADAMTS proteinases, serves as a bioactive molecule. Here, since versican expression in the placenta was dynamically altered in humans and mice, we investigated the role of versican in pregnancy using uterine-specific Vcan deletion mice (uKO mice) and ADAMTS-resistant versican expressing mice (V1R mice). uKO mice exhibited insufficient spiral artery dilation, followed by fetal growth restriction and maternal hypertension. Further analysis revealed impaired proliferation of tissue-resident natural killer cells required for spiral artery dilation. V1R mice showed the same results as the control, eliminating the involvement of versikine. Our results provide a new concept that versican, one factor of ECM, contributes to placentation and following fetal growth.
Collapse
Affiliation(s)
- Yusuke Sagae
- Department of Gynecology and Obstetrics, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Akihito Horie
- Department of Gynecology and Obstetrics, Kyoto University Graduate School of Medicine, Kyoto, Japan.
| | - Akihiro Yanai
- Department of Gynecology and Obstetrics, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Tsutomu Ohara
- Department of Gynecology and Obstetrics, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Baku Nakakita
- Department of Gynecology and Obstetrics, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Yoshimi Kitawaki
- Department of Gynecology and Obstetrics, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Asuka Okunomiya
- Department of Gynecology and Obstetrics, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Hirohiko Tani
- Department of Gynecology and Obstetrics, Shizuoka General Hospital, Shizuoka, Japan
| | - Ken Yamaguchi
- Department of Gynecology and Obstetrics, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Junzo Hamanishi
- Department of Gynecology and Obstetrics, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - John P Lydon
- Department of Pathology and Immunology, Center for Drug Discovery, Center for Reproductive Medicine, Baylor College of Medicine, Houston, TX, United States of America
| | - Takiko Daikoku
- Division of Animal Disease Model, Research Center for Experimental Modeling of Human Disease, Kanazawa University, Kanazawa, Japan
| | - Hideto Watanabe
- Institute for Molecular Science of Medicine, Aichi Medical University, Nagakute, Japan
| | - Masaki Mandai
- Department of Gynecology and Obstetrics, Kyoto University Graduate School of Medicine, Kyoto, Japan
| |
Collapse
|
13
|
Yanagi N, Kato S, Fukazawa T, Kubo T. Cellular responses in the FGF10-mediated improvement of hindlimb regenerative capacity in Xenopus laevis revealed by single-cell transcriptomics. Dev Growth Differ 2022; 64:266-278. [PMID: 35642106 PMCID: PMC11520959 DOI: 10.1111/dgd.12795] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Revised: 05/16/2022] [Accepted: 05/24/2022] [Indexed: 12/28/2022]
Abstract
Xenopus laevis tadpoles possess regenerative capacity in their hindlimb buds at early developmental stages (stages ~52-54); they can regenerate complete hindlimbs with digits after limb bud amputation. However, they gradually lose their regenerative capacity as metamorphosis proceeds. Tadpoles in late developmental stages regenerate fewer digits (stage ~56), or only form cartilaginous spike without digits or joints (stage ~58 or later) after amputation. Previous studies have shown that administration of fibroblast growth factor 10 (FGF10) in late-stage (stage 56) tadpole hindlimb buds after amputation can improve their regenerative capacity, which means that the cells responding to FGF10 signaling play an important role in limb bud regeneration. In this study, we performed single-cell RNA sequencing (scRNA-seq) of hindlimb buds that were amputated and administered FGF10 by implanting FGF10-soaked beads at a late stage (stage 56), and explored cell clusters exhibiting a differential gene expression pattern compared with that in controls treated with phosphate-buffered saline. The scRNA-seq data showed expansion of fgf8-expressing cells in the cluster of the apical epidermal cap of FGF10-treated hindlimb buds, which was reported previously, indicating that the administration of FGF10 was successful. On analysis, in addition to the epidermal cluster, a subset of myeloid cells and a newly identified cluster of steap4-expressing cells showed remarkable differences in their gene expression profiles between the FGF10- or phosphate-buffered saline-treatment conditions, suggesting a possible role of these clusters in improving the regenerative capacity of hindlimbs via FGF10 administration.
Collapse
Affiliation(s)
- Nodoka Yanagi
- Department of Biological Sciences, Graduate School of ScienceThe University of TokyoTokyoJapan
| | - Sumika Kato
- Department of Biological Sciences, Graduate School of ScienceThe University of TokyoTokyoJapan
| | - Taro Fukazawa
- Department of Biological Sciences, Graduate School of ScienceThe University of TokyoTokyoJapan
| | - Takeo Kubo
- Department of Biological Sciences, Graduate School of ScienceThe University of TokyoTokyoJapan
| |
Collapse
|
14
|
Yang Z, Yi P, Liu Z, Zhang W, Mei L, Feng C, Tu C, Li Z. Stem Cell-Laden Hydrogel-Based 3D Bioprinting for Bone and Cartilage Tissue Engineering. Front Bioeng Biotechnol 2022; 10:865770. [PMID: 35656197 PMCID: PMC9152119 DOI: 10.3389/fbioe.2022.865770] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2022] [Accepted: 04/18/2022] [Indexed: 12/30/2022] Open
Abstract
Tremendous advances in tissue engineering and regenerative medicine have revealed the potential of fabricating biomaterials to solve the dilemma of bone and articular defects by promoting osteochondral and cartilage regeneration. Three-dimensional (3D) bioprinting is an innovative fabrication technology to precisely distribute the cell-laden bioink for the construction of artificial tissues, demonstrating great prospect in bone and joint construction areas. With well controllable printability, biocompatibility, biodegradability, and mechanical properties, hydrogels have been emerging as an attractive 3D bioprinting material, which provides a favorable biomimetic microenvironment for cell adhesion, orientation, migration, proliferation, and differentiation. Stem cell-based therapy has been known as a promising approach in regenerative medicine; however, limitations arise from the uncontrollable proliferation, migration, and differentiation of the stem cells and fortunately could be improved after stem cells were encapsulated in the hydrogel. In this review, our focus was centered on the characterization and application of stem cell-laden hydrogel-based 3D bioprinting for bone and cartilage tissue engineering. We not only highlighted the effect of various kinds of hydrogels, stem cells, inorganic particles, and growth factors on chondrogenesis and osteogenesis but also outlined the relationship between biophysical properties like biocompatibility, biodegradability, osteoinductivity, and the regeneration of bone and cartilage. This study was invented to discuss the challenge we have been encountering, the recent progress we have achieved, and the future perspective we have proposed for in this field.
Collapse
Affiliation(s)
- Zhimin Yang
- Department of Orthopedics, The Second Xiangya Hospital, Central South University, Changsha, China
- Hunan Key Laboratory of Tumor Models and Individualized Medicine, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Ping Yi
- Department of Dermatology, The Second Xiangya Hospital, Central South University, Hunan Key Laboratory of Medical Epigenomics, Changsha, China
| | - Zhongyue Liu
- Department of Orthopedics, The Second Xiangya Hospital, Central South University, Changsha, China
- Hunan Key Laboratory of Tumor Models and Individualized Medicine, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Wenchao Zhang
- Department of Orthopedics, The Second Xiangya Hospital, Central South University, Changsha, China
- Hunan Key Laboratory of Tumor Models and Individualized Medicine, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Lin Mei
- Department of Orthopedics, The Second Xiangya Hospital, Central South University, Changsha, China
- Hunan Key Laboratory of Tumor Models and Individualized Medicine, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Chengyao Feng
- Department of Orthopedics, The Second Xiangya Hospital, Central South University, Changsha, China
- Hunan Key Laboratory of Tumor Models and Individualized Medicine, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Chao Tu
- Department of Orthopedics, The Second Xiangya Hospital, Central South University, Changsha, China
- Hunan Key Laboratory of Tumor Models and Individualized Medicine, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Zhihong Li
- Department of Orthopedics, The Second Xiangya Hospital, Central South University, Changsha, China
- Hunan Key Laboratory of Tumor Models and Individualized Medicine, The Second Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
15
|
Schwartz NB, Domowicz MS. Roles of Chondroitin Sulfate Proteoglycans as Regulators of Skeletal Development. Front Cell Dev Biol 2022; 10:745372. [PMID: 35465334 PMCID: PMC9026158 DOI: 10.3389/fcell.2022.745372] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2021] [Accepted: 03/21/2022] [Indexed: 11/29/2022] Open
Abstract
The extracellular matrix (ECM) is critically important for most cellular processes including differentiation, morphogenesis, growth, survival and regeneration. The interplay between cells and the ECM often involves bidirectional signaling between ECM components and small molecules, i.e., growth factors, morphogens, hormones, etc., that regulate critical life processes. The ECM provides biochemical and contextual information by binding, storing, and releasing the bioactive signaling molecules, and/or mechanical information that signals from the cell membrane integrins through the cytoskeleton to the nucleus, thereby influencing cell phenotypes. Using these dynamic, reciprocal processes, cells can also remodel and reshape the ECM by degrading and re-assembling it, thereby sculpting their environments. In this review, we summarize the role of chondroitin sulfate proteoglycans as regulators of cell and tissue development using the skeletal growth plate model, with an emphasis on use of naturally occurring, or created mutants to decipher the role of proteoglycan components in signaling paradigms.
Collapse
Affiliation(s)
- Nancy B. Schwartz
- Department of Pediatrics, Biological Sciences Division, The University of Chicago, Chicago, IL, United States
- Department of Biochemistry and Molecular Biology, Biological Sciences Division, The University of Chicago, Chicago, IL, United States
- *Correspondence: Nancy B. Schwartz,
| | - Miriam S. Domowicz
- Department of Pediatrics, Biological Sciences Division, The University of Chicago, Chicago, IL, United States
| |
Collapse
|
16
|
Abstract
Aggrecan (Acan) and versican (Vcan) are large chondroitin sulfate proteoglycans of the extracellular matrix. They share the same structural domains at both N and C-termini. The N-terminal G1 domain binds hyaluronan (HA), forms an HA-rich matrix, and regulates HA-mediated signaling. The C-terminal G3 domain binds other extracellular matrix molecules and forms a supramolecular structure that stores TGFb and BMPs and regulates their signaling. EGF-like motifs in the G3 domain may directly act like an EGF ligand. Both Acan and Vcan are present in cartilage, intervertebral disc, brain, heart, and aorta. Their localizations are essentially reciprocal. This review describes their structural domains, expression patterns and functions, and regulation of their expression.
Collapse
Affiliation(s)
- Hideto Watanabe
- Institute for Molecular Science of Medicine, Aichi Medical University, Nagakute, Aichi, Japan
| |
Collapse
|
17
|
Islam S, Jahan N, Shahida A, Karnan S, Watanabe H. Accumulation of versican and lack of versikine ameliorate acute colitis. Matrix Biol 2022; 107:59-76. [DOI: 10.1016/j.matbio.2022.02.004] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2021] [Revised: 02/05/2022] [Accepted: 02/09/2022] [Indexed: 12/11/2022]
|
18
|
Rohde D, Vandoorne K, Lee IH, Grune J, Zhang S, McAlpine CS, Schloss MJ, Nayar R, Courties G, Frodermann V, Wojtkiewicz G, Honold L, Chen Q, Schmidt S, Iwamoto Y, Sun Y, Cremer S, Hoyer FF, Iborra-Egea O, Muñoz-Guijosa C, Ji F, Zhou B, Adams RH, Wythe JD, Hidalgo J, Watanabe H, Jung Y, van der Laan AM, Piek JJ, Kfoury Y, Désogère PA, Vinegoni C, Dutta P, Sadreyev RI, Caravan P, Bayes-Genis A, Libby P, Scadden DT, Lin CP, Naxerova K, Swirski FK, Nahrendorf M. Bone marrow endothelial dysfunction promotes myeloid cell expansion in cardiovascular disease. NATURE CARDIOVASCULAR RESEARCH 2022; 1:28-44. [PMID: 35747128 PMCID: PMC9216333 DOI: 10.1038/s44161-021-00002-8] [Citation(s) in RCA: 42] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/09/2021] [Accepted: 10/27/2021] [Indexed: 12/13/2022]
Abstract
Abnormal hematopoiesis advances cardiovascular disease by generating excess inflammatory leukocytes that attack the arteries and the heart. The bone marrow niche regulates hematopoietic stem cell proliferation and hence the systemic leukocyte pool, but whether cardiovascular disease affects the hematopoietic organ's microvasculature is unknown. Here we show that hypertension, atherosclerosis and myocardial infarction (MI) instigate endothelial dysfunction, leakage, vascular fibrosis and angiogenesis in the bone marrow, altogether leading to overproduction of inflammatory myeloid cells and systemic leukocytosis. Limiting angiogenesis with endothelial deletion of Vegfr2 (encoding vascular endothelial growth factor (VEGF) receptor 2) curbed emergency hematopoiesis after MI. We noted that bone marrow endothelial cells assumed inflammatory transcriptional phenotypes in all examined stages of cardiovascular disease. Endothelial deletion of Il6 or Vcan (encoding versican), genes shown to be highly expressed in mice with atherosclerosis or MI, reduced hematopoiesis and systemic myeloid cell numbers in these conditions. Our findings establish that cardiovascular disease remodels the vascular bone marrow niche, stimulating hematopoiesis and production of inflammatory leukocytes.
Collapse
Affiliation(s)
- David Rohde
- Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Department of Cardiology, Angiology and Pneumology, Heidelberg University Hospital, Heidelberg, Germany
- These authors contributed equally: David Rohde, Katrien Vandoorne
| | - Katrien Vandoorne
- Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Biomedical Engineering Faculty, Technion-Israel Institute of Technology, Haifa, Israel
- These authors contributed equally: David Rohde, Katrien Vandoorne
| | - I-Hsiu Lee
- Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Jana Grune
- Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Shuang Zhang
- Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Cameron S. McAlpine
- Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Maximilian J. Schloss
- Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Ribhu Nayar
- Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Gabriel Courties
- Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Vanessa Frodermann
- Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Gregory Wojtkiewicz
- Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Lisa Honold
- Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Qi Chen
- Max Planck Institute for Molecular Biomedicine, Muenster, Germany
| | - Stephen Schmidt
- Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Yoshiko Iwamoto
- Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Yuan Sun
- Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Sebastian Cremer
- Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Friedrich F. Hoyer
- Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | | | | | - Fei Ji
- Department of Genetics, Harvard Medical School, Boston, MA, USA
- Department of Molecular Biology, Massachusetts General Hospital, Boston, MA, USA
| | - Bin Zhou
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, Shanghai, China
| | - Ralf H. Adams
- Max Planck Institute for Molecular Biomedicine, Muenster, Germany
| | - Joshua D. Wythe
- Cardiovascular Research Institute, Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, TX, USA
| | - Juan Hidalgo
- Institute of Neurosciences and Department of Cellular Biology, Physiology and Immunology, Universitat Autonoma de Barcelona, Barcelona, Spain
| | - Hideto Watanabe
- Institute for Molecular Science of Medicine, Aichi Medical University, Aichi, Japan
| | - Yookyung Jung
- Wellman Center for Photomedicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Anja M. van der Laan
- Heart Center, Department of Cardiology, Amsterdam University Medical Center, University of Amsterdam, Amsterdam, the Netherlands
| | - Jan J. Piek
- Heart Center, Department of Cardiology, Amsterdam University Medical Center, University of Amsterdam, Amsterdam, the Netherlands
| | - Youmna Kfoury
- Center for Regenerative Medicine and Cancer Center, Massachusetts General Hospital, Boston, MA, USA
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA, USA
| | - Pauline A. Désogère
- Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, USA
| | - Claudio Vinegoni
- Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Partha Dutta
- Pittsburgh Heart, Lung, Blood and Vascular Medicine Institute, Division of Cardiology, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Ruslan I. Sadreyev
- Department of Molecular Biology, Massachusetts General Hospital, Boston, MA, USA
- Department of Pathology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Peter Caravan
- Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, USA
| | | | - Peter Libby
- Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA, USA
| | - David T. Scadden
- Center for Regenerative Medicine and Cancer Center, Massachusetts General Hospital, Boston, MA, USA
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA, USA
| | - Charles P. Lin
- Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Institute for Molecular Science of Medicine, Aichi Medical University, Aichi, Japan
| | - Kamila Naxerova
- Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Filip K. Swirski
- Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Matthias Nahrendorf
- Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Cardiovascular Research Center, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Department of Internal Medicine I, University Hospital Wuerzburg, Wuerzburg, Germany
| |
Collapse
|
19
|
Tang S, Chen P, Zhang H, Weng H, Fang Z, Chen C, Peng G, Gao H, Hu K, Chen J, Chen L, Chen X. Comparison of Curative Effect of Human Umbilical Cord-Derived Mesenchymal Stem Cells and Their Small Extracellular Vesicles in Treating Osteoarthritis. Int J Nanomedicine 2021; 16:8185-8202. [PMID: 34938076 PMCID: PMC8687685 DOI: 10.2147/ijn.s336062] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Accepted: 11/04/2021] [Indexed: 01/15/2023] Open
Abstract
Introduction Human umbilical cord-derived mesenchymal stem cells (hUC-MSCs) and their small extracellular vesicles (hUC-MSC-sEVs) have shown attractive prospects applying in regenerative medicine. This study aimed to compare the therapeutic effects of two agents on osteoarthritis (OA) and investigate underlying mechanism using proteomics. Methods In vitro, the proliferation and migration abilities of chondrocytes treated with hUC-MSCs or hUC-MSC-sEVs were detected by Cell Counting Kit-8 assay and scratch wound assay. In vivo, hUC-MSCs (a single dose of 5 × 105) or hUC-MSC-sEVs (30 μg/time) were injected into the knee joints of anterior cruciate ligament transection-induced OA model. Hematoxylin and eosin, Safranin O/Fast Green staining were used to observe cartilage degeneration. The levels of cartilage matrix metabolic molecules (Collagen II, MMP13 and ADAMTS5) and macrophage polarization markers (CD14, IL-1β, IL-10 and CD206) were assessed by immunohistochemistry. Finally, proteomics analysis was performed to characterize the proteinaceous contents of two agents. Results In vitro data showed that hUC-MSC-sEVs were taken up by chondrocytes. A total of 15 μg/mL of sEVs show the greatest proliferative and migratory capacities among all groups. In the animal study, hUC-MSCs and hUC-MSC-sEVs alleviated cartilage damage. This effect was mediated via maintaining cartilage homeostasis, as was confirmed by upregulation of the COL II and downregulation of the MMP13 and ADAMTS5. Moreover, the M1 macrophage markers (CD14) were significantly reduced, while the M2 macrophage markers (CD206 and IL-10) were increased in the hUC-MSCs and hUC-MSC-sEVs relative to the untreated group. Mechanistically, we found that many proteins connected to cartilage repair were more abundant in sEVs. Notably, compared to hUC-MSCs, the upregulated proteins in sEVs were mostly involved in the regulation of immune effector process, extracellular matrix organization, PI3K-AKT signaling pathways, and Rap1 signaling pathway. Conclusion Our study indicated that hUC-MSC-sEVs protect cartilage from damage and many cartilage repair-related proteins are probably involved in the restoration process. These data suggest the promising potential of hUC-MSC-sEVs as a therapeutic agent for OA.
Collapse
Affiliation(s)
- Shijie Tang
- Department of Plastic Surgery, Fujian Medical University Union Hospital, Fuzhou, 350001, People's Republic of China.,Department of Plastic Surgery and Regenerative Medicine Institute, Fujian Medical University, Fuzhou, 350001, People's Republic of China.,Engineering Research Center of Tissue and Organ Regeneration, Fujian Province University, Fuzhou, 350001, People's Republic of China.,Oncology Institution, Fujian Medical University, Fuzhou, 350004, People's Republic of China
| | - Penghong Chen
- Department of Plastic Surgery, Fujian Medical University Union Hospital, Fuzhou, 350001, People's Republic of China.,Department of Plastic Surgery and Regenerative Medicine Institute, Fujian Medical University, Fuzhou, 350001, People's Republic of China.,Engineering Research Center of Tissue and Organ Regeneration, Fujian Province University, Fuzhou, 350001, People's Republic of China.,Oncology Institution, Fujian Medical University, Fuzhou, 350004, People's Republic of China
| | - Haoruo Zhang
- Department of Plastic Surgery, Fujian Medical University Union Hospital, Fuzhou, 350001, People's Republic of China.,Department of Plastic Surgery and Regenerative Medicine Institute, Fujian Medical University, Fuzhou, 350001, People's Republic of China.,Engineering Research Center of Tissue and Organ Regeneration, Fujian Province University, Fuzhou, 350001, People's Republic of China.,Oncology Institution, Fujian Medical University, Fuzhou, 350004, People's Republic of China
| | - Haiyan Weng
- Department of Plastic Surgery, Fujian Medical University Union Hospital, Fuzhou, 350001, People's Republic of China.,Department of Plastic Surgery and Regenerative Medicine Institute, Fujian Medical University, Fuzhou, 350001, People's Republic of China.,Engineering Research Center of Tissue and Organ Regeneration, Fujian Province University, Fuzhou, 350001, People's Republic of China.,Oncology Institution, Fujian Medical University, Fuzhou, 350004, People's Republic of China
| | - Zhuoqun Fang
- Department of Plastic Surgery, Fujian Medical University Union Hospital, Fuzhou, 350001, People's Republic of China.,Department of Plastic Surgery and Regenerative Medicine Institute, Fujian Medical University, Fuzhou, 350001, People's Republic of China.,Engineering Research Center of Tissue and Organ Regeneration, Fujian Province University, Fuzhou, 350001, People's Republic of China.,Oncology Institution, Fujian Medical University, Fuzhou, 350004, People's Republic of China
| | - Caixiang Chen
- Department of Plastic Surgery, Fujian Medical University Union Hospital, Fuzhou, 350001, People's Republic of China.,Department of Plastic Surgery and Regenerative Medicine Institute, Fujian Medical University, Fuzhou, 350001, People's Republic of China.,Engineering Research Center of Tissue and Organ Regeneration, Fujian Province University, Fuzhou, 350001, People's Republic of China.,Oncology Institution, Fujian Medical University, Fuzhou, 350004, People's Republic of China
| | - Guohao Peng
- Department of Plastic Surgery, Fujian Medical University Union Hospital, Fuzhou, 350001, People's Republic of China.,Department of Plastic Surgery and Regenerative Medicine Institute, Fujian Medical University, Fuzhou, 350001, People's Republic of China.,Engineering Research Center of Tissue and Organ Regeneration, Fujian Province University, Fuzhou, 350001, People's Republic of China.,Oncology Institution, Fujian Medical University, Fuzhou, 350004, People's Republic of China
| | - Hangqi Gao
- Department of Plastic Surgery, Fujian Medical University Union Hospital, Fuzhou, 350001, People's Republic of China.,Department of Plastic Surgery and Regenerative Medicine Institute, Fujian Medical University, Fuzhou, 350001, People's Republic of China.,Engineering Research Center of Tissue and Organ Regeneration, Fujian Province University, Fuzhou, 350001, People's Republic of China.,Oncology Institution, Fujian Medical University, Fuzhou, 350004, People's Republic of China
| | - Kailun Hu
- Department of Plastic Surgery, Fujian Medical University Union Hospital, Fuzhou, 350001, People's Republic of China.,Department of Plastic Surgery and Regenerative Medicine Institute, Fujian Medical University, Fuzhou, 350001, People's Republic of China.,Engineering Research Center of Tissue and Organ Regeneration, Fujian Province University, Fuzhou, 350001, People's Republic of China.,Oncology Institution, Fujian Medical University, Fuzhou, 350004, People's Republic of China
| | - Jinghua Chen
- Department of Pharmaceutical Analysis, the School of Pharmacy, Fujian Medical University, Fuzhou, 350100, People's Republic of China
| | - Liangwan Chen
- Engineering Research Center of Tissue and Organ Regeneration, Fujian Province University, Fuzhou, 350001, People's Republic of China.,Department of Cardiac Surgery, Fujian Medical University Union Hospital, Fuzhou, 350001, People's Republic of China
| | - Xiaosong Chen
- Department of Plastic Surgery, Fujian Medical University Union Hospital, Fuzhou, 350001, People's Republic of China.,Department of Plastic Surgery and Regenerative Medicine Institute, Fujian Medical University, Fuzhou, 350001, People's Republic of China.,Engineering Research Center of Tissue and Organ Regeneration, Fujian Province University, Fuzhou, 350001, People's Republic of China
| |
Collapse
|
20
|
Proteoglycans and Diseases of Soft Tissues. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1348:127-138. [PMID: 34807417 DOI: 10.1007/978-3-030-80614-9_5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Proteoglycans consist of protein cores to which at least one glycosaminoglycan chain is attached. They play important roles in the physiology and biomechanical function of tendons, ligaments, cardiovascular system, and other systems through their involvement in regulation of assembly and maintenance of extracellular matrix, and through their participation in cell proliferation together with growth factors. They can be divided into two main groups, small and large proteoglycans. The small proteoglycans are also known as small leucine-rich proteoglycans (SLRPs) which are encoded by 18 genes and are further subclassified into Classes I-V. Several members of Class I and II, such as decorin and biglycan from Class I, and Class II fibromodulin and lumican, are known to regulate collagen fibrillogenesis. Decorin limits the diameter of collagen fibrils during fibrillogenesis. The function of biglycan in fibrillogenesis is similar to that of decorin. Though biomechanical function of tendon is compromised in decorin-deficient mice, decorin can substitute for lack of biglycan in biglycan-deficient mice. New data also indicate an important role for biglycan in disorders of the cardiovascular system, including aortic valve stenosis and aortic dissection. Two members of the Class II of SLRPs, fibromodulin and lumican bind to the same site within the collagen molecule and can substitute for each other in fibromodulin- or lumican-deficient mice.Aggrecan and versican are the major representatives of the large proteoglycans. Though they are mainly found in the cartilage where they provide resilience and toughness, they are present also in tensile portions of tendons and, in slightly different biochemical form in fibrocartilage. Degradation by aggrecanase is responsible for the appearance of different forms of aggrecan and versican in different parts of the tendon where these cleaved forms play different roles. In addition, they are important components of the ventricularis of cardiac valves. Mutations in the gene for versican or in the gene for elastin (which binds to versican ) lead to severe disruptions of normal developmental of the heart at least in mice.
Collapse
|
21
|
Martin DR, Santamaria S, Koch CD, Ahnström J, Apte SS. Identification of novel ADAMTS1, ADAMTS4 and ADAMTS5 cleavage sites in versican using a label-free quantitative proteomics approach. J Proteomics 2021; 249:104358. [PMID: 34450332 PMCID: PMC8713443 DOI: 10.1016/j.jprot.2021.104358] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2021] [Revised: 07/20/2021] [Accepted: 08/16/2021] [Indexed: 01/08/2023]
Abstract
The chondroitin sulfate proteoglycan versican is important for embryonic development and several human disorders. The versican V1 splice isoform is widely expressed and cleaved by ADAMTS proteases at a well-characterized site, Glu441-Ala442. Since ADAMTS proteases cleave the homologous proteoglycan aggrecan at multiple sites, we hypothesized that additional cleavage sites existed within versican. We report a quantitative label-free approach that ranks abundance of liquid chromatography-tandem mass spectrometry (LC-MS/MS)-identified semi-tryptic peptides after versican digestion by ADAMTS1, ADAMTS4 and ADAMTS5 to identify site-specific cleavages. Recombinant purified versican V1 constructs were digested with the recombinant full-length proteases, using catalytically inactive mutant proteases in control digests. Semi-tryptic peptide abundance ratios determined by LC-MS/MS in ADAMTS:control digests were compared to the mean of all identified peptides to obtain a z-score by which outlier peptides were ranked, using semi-tryptic peptides identifying Glu441 -Ala442 cleavage as the benchmark. Tryptic peptides with higher abundance in control digests supported cleavage site identification. We identified several novel cleavage sites supporting the ADAMTS1/4/5 cleavage site preference for a P1-Glu residue in proteoglycan substrates. Digestion of proteins in vitro and application of this z-score approach is potentially widely applicable for mapping protease cleavage sites using label-free proteomics. SIGNIFICANCE: Versican abundance and turnover are relevant to the pathogenesis of several human disorders. Versican is cleaved by A Disintegrin-like And Metalloprotease with Thrombospondin type 1 motifs (ADAMTS) family members at Glu441-Ala442, generating a bioactive proteoform called versikine, but additional cleavage sites and the site-specificity of individual ADAMTS proteases is unexplored. Here, we used a label-free proteomics strategy to identify versican cleavage sites for 3 ADAMTS proteases, applying a novel z-score-based statistical approach to compare the protease digests of versican to controls (digests with inactive protease) using the known protease cleavage site as a benchmark. We identified 21 novel cleavage sites that had a comparable z-score to the benchmark. Given the functional significance of versikine, they represent potentially significant cleavages and helped to refine a substrate site preference for each protease.The z-score approach is potentially widely applicable for discovery of site-specific cleavages within an purified protein or small ensemble of proteins using any protease.
Collapse
Affiliation(s)
- Daniel R Martin
- Department of Biomedical Engineering, Cleveland Clinic Lerner Research Institute, Cleveland, OH 44195, USA
| | - Salvatore Santamaria
- Department of Immunology and Inflammation, 5th Floor Commonwealth Building, Hammersmith Hospital Campus, Du Cane Road, W12 0NN London, United Kingdom
| | - Christopher D Koch
- Department of Biomedical Engineering, Cleveland Clinic Lerner Research Institute, Cleveland, OH 44195, USA
| | - Josefin Ahnström
- Department of Immunology and Inflammation, 5th Floor Commonwealth Building, Hammersmith Hospital Campus, Du Cane Road, W12 0NN London, United Kingdom
| | - Suneel S Apte
- Department of Biomedical Engineering, Cleveland Clinic Lerner Research Institute, Cleveland, OH 44195, USA.
| |
Collapse
|
22
|
Nguyen TH, Duong CM, Nguyen XH, Than UTT. Mesenchymal Stem Cell-Derived Extracellular Vesicles for Osteoarthritis Treatment: Extracellular Matrix Protection, Chondrocyte and Osteocyte Physiology, Pain and Inflammation Management. Cells 2021; 10:2887. [PMID: 34831109 PMCID: PMC8616200 DOI: 10.3390/cells10112887] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2021] [Revised: 10/22/2021] [Accepted: 10/24/2021] [Indexed: 12/11/2022] Open
Abstract
Osteoarthritis (OA) is a common degenerative disease that can lead to persistent pain and motion restriction. In the last decade, stem cells, particularly mesenchymal stem cells (MSCs), have been explored as a potential alternative OA therapy due to their regenerative capacity. Furthermore, it has been shown that trophic factors enveloped in extracellular vesicles (EVs), including exosomes, are a crucial aspect of MSC-based treatment for OA. Evidently, EVs derived from different MSC sources might rescue the OA phenotype by targeting many biological processes associated with cartilage extracellular matrix (ECM) degradation and exerting protective effects on different joint cell types. Despite this advancement, different studies employing EV treatment for OA have revealed reverse outcomes depending on the EV cargo, cell source, and pathological condition. Hence, in this review, we aim to summarize and discuss the possible effects of MSC-derived EVs based on recent findings at different stages of OA development, including effects on cartilage ECM, chondrocyte biology, osteocytes and bone homeostasis, inflammation, and pain management. Additionally, we discuss further strategies and technical advances for manipulating EVs to specifically target OA to bring the therapy closer to clinical use.
Collapse
Affiliation(s)
- Thu Huyen Nguyen
- Department of Bioscience, University of Milan, 20133 Milan, Italy;
- Vinmec Research Institute of Stem Cell and Gene Technology, Vinmec Healthcare System, Hanoi 100000, Vietnam; (C.M.D.); (X.-H.N.)
| | - Chau Minh Duong
- Vinmec Research Institute of Stem Cell and Gene Technology, Vinmec Healthcare System, Hanoi 100000, Vietnam; (C.M.D.); (X.-H.N.)
- Department of Biology, Clark University, Worcester, MA 01610, USA
| | - Xuan-Hung Nguyen
- Vinmec Research Institute of Stem Cell and Gene Technology, Vinmec Healthcare System, Hanoi 100000, Vietnam; (C.M.D.); (X.-H.N.)
- Vinmec Research Institute of Applied Sciences and Regenerative Medicine, Vinmec Healthcare System, Hanoi 100000, Vietnam
- College of Health Sciences, VinUniversity, Hanoi 100000, Vietnam
| | - Uyen Thi Trang Than
- Vinmec Research Institute of Stem Cell and Gene Technology, Vinmec Healthcare System, Hanoi 100000, Vietnam; (C.M.D.); (X.-H.N.)
- Vinmec Research Institute of Applied Sciences and Regenerative Medicine, Vinmec Healthcare System, Hanoi 100000, Vietnam
| |
Collapse
|
23
|
Isolation and Purification of Versican and Analysis of Versican Proteolysis. Methods Mol Biol 2021. [PMID: 34626407 DOI: 10.1007/978-1-0716-1398-6_43] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/02/2023]
Abstract
Versican is a widely distributed chondroitin sulfate proteoglycan that forms large complexes with the glycosaminoglycan hyaluronan (HA). As a consequence of HA binding to its receptor CD44 and interactions of the versican C-terminal globular (G3) domain with a variety of extracellular matrix proteins, versican is a key component of well-defined networks in pericellular matrix and extracellular matrix. Versican is crucial for several developmental processes in the embryo ranging from cardiac development to digit separation, and there is an increasing interest in its roles in cancer and inflammation. Versican proteolysis by ADAMTS proteases is highly regulated, occurs at specific peptide bonds, and is relevant to several physiological and disease mechanisms. In this chapter, methods are described for the isolation and detection of intact and cleaved versican in tissues using morphologic and biochemical techniques. These, together with the methodologies for purification and analysis of recombinant versican and an N-terminal versican fragment named versikine that are provided here, are likely to facilitate further progress on the biology of versican and its proteolysis.
Collapse
|
24
|
Stone RN, Frahs SM, Hardy MJ, Fujimoto A, Pu X, Keller-Peck C, Oxford JT. Decellularized Porcine Cartilage Scaffold; Validation of Decellularization and Evaluation of Biomarkers of Chondrogenesis. Int J Mol Sci 2021; 22:6241. [PMID: 34207917 PMCID: PMC8230108 DOI: 10.3390/ijms22126241] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2021] [Revised: 06/01/2021] [Accepted: 06/04/2021] [Indexed: 12/21/2022] Open
Abstract
Osteoarthritis is a major concern in the United States and worldwide. Current non-surgical and surgical approaches alleviate pain but show little evidence of cartilage restoration. Cell-based treatments may hold promise for the regeneration of hyaline cartilage-like tissue at the site of injury or wear. Cell-cell and cell-matrix interactions have been shown to drive cell differentiation pathways. Biomaterials for clinically relevant applications can be generated from decellularized porcine auricular cartilage. This material may represent a suitable scaffold on which to seed and grow chondrocytes to create new cartilage. In this study, we used decellularization techniques to create an extracellular matrix scaffold that supports chondrocyte cell attachment and growth in tissue culture conditions. Results presented here evaluate the decellularization process histologically and molecularly. We identified new and novel biomarker profiles that may aid future cartilage decellularization efforts. Additionally, the resulting scaffold was characterized using scanning electron microscopy, fluorescence microscopy, and proteomics. Cellular response to the decellularized scaffold was evaluated by quantitative real-time PCR for gene expression analysis.
Collapse
Affiliation(s)
- Roxanne N. Stone
- Interdisciplinary Studies Program, Boise State University, Boise, ID 83725, USA;
- Biomolecular Research Center, Boise State University, Boise, ID 83725, USA; (S.M.F.); (M.J.H.); (A.F.); (X.P.); (C.K.-P.)
| | - Stephanie M. Frahs
- Biomolecular Research Center, Boise State University, Boise, ID 83725, USA; (S.M.F.); (M.J.H.); (A.F.); (X.P.); (C.K.-P.)
- Center of Biomedical Research Excellence in Matrix Biology, Boise State University, Boise, ID 83725, USA
- Biomolecular Sciences Graduate Programs, Boise State University, Boise, ID 83725, USA
| | - Makenna J. Hardy
- Biomolecular Research Center, Boise State University, Boise, ID 83725, USA; (S.M.F.); (M.J.H.); (A.F.); (X.P.); (C.K.-P.)
- Center of Biomedical Research Excellence in Matrix Biology, Boise State University, Boise, ID 83725, USA
- Biomolecular Sciences Graduate Programs, Boise State University, Boise, ID 83725, USA
| | - Akina Fujimoto
- Biomolecular Research Center, Boise State University, Boise, ID 83725, USA; (S.M.F.); (M.J.H.); (A.F.); (X.P.); (C.K.-P.)
- Center of Biomedical Research Excellence in Matrix Biology, Boise State University, Boise, ID 83725, USA
| | - Xinzhu Pu
- Biomolecular Research Center, Boise State University, Boise, ID 83725, USA; (S.M.F.); (M.J.H.); (A.F.); (X.P.); (C.K.-P.)
- Center of Biomedical Research Excellence in Matrix Biology, Boise State University, Boise, ID 83725, USA
- Biomolecular Sciences Graduate Programs, Boise State University, Boise, ID 83725, USA
| | - Cynthia Keller-Peck
- Biomolecular Research Center, Boise State University, Boise, ID 83725, USA; (S.M.F.); (M.J.H.); (A.F.); (X.P.); (C.K.-P.)
- Center of Biomedical Research Excellence in Matrix Biology, Boise State University, Boise, ID 83725, USA
| | - Julia Thom Oxford
- Biomolecular Research Center, Boise State University, Boise, ID 83725, USA; (S.M.F.); (M.J.H.); (A.F.); (X.P.); (C.K.-P.)
- Center of Biomedical Research Excellence in Matrix Biology, Boise State University, Boise, ID 83725, USA
- Biomolecular Sciences Graduate Programs, Boise State University, Boise, ID 83725, USA
- Department of Biological Sciences, Boise State University, Boise, ID 83725, USA
| |
Collapse
|
25
|
Thompson SD, Pichika R, Lieber RL, Lavasani M. Systemic transplantation of adult multipotent stem cells prevents articular cartilage degeneration in a mouse model of accelerated ageing. Immun Ageing 2021; 18:27. [PMID: 34098983 PMCID: PMC8183038 DOI: 10.1186/s12979-021-00239-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2020] [Accepted: 05/26/2021] [Indexed: 01/13/2023]
Abstract
BACKGROUND Osteoarthritis (OA) is one of the most prevalent joint diseases of advanced age and is a leading cause of disability worldwide. Ageing is a major risk factor for the articular cartilage (AC) degeneration that leads to OA, and the age-related decline in regenerative capacity accelerates OA progression. Here we demonstrate that systemic transplantation of a unique population of adult multipotent muscle-derived stem/progenitor cells (MDSPCs), isolated from young wild-type mice, into Zmpste24-/- mice (a model of Hutchinson-Gilford progeria syndrome, a condition marked by accelerated ageing), prevents ageing-related homeostatic decline of AC. RESULTS MDSPC treatment inhibited expression of cartilage-degrading factors such as pro-inflammatory cytokines and extracellular matrix-proteinases, whereas pro-regenerative markers associated with cartilage mechanical support and tensile strength, cartilage resilience, chondrocyte proliferation and differentiation, and cartilage growth, were increased. Notably, MDSPC transplantation also increased the expression level of genes known for their key roles in immunomodulation, autophagy, stress resistance, pro-longevity, and telomere protection. Our findings also indicate that MDSPC transplantation increased proteoglycan content by regulating chondrocyte proliferation. CONCLUSIONS Together, these findings demonstrate the ability of systemically transplanted young MDSPCs to preserve a healthy homeostasis and promote tissue regeneration at the molecular and tissue level in progeroid AC. These results highlight the therapeutic potential of systemically delivered multipotent adult stem cells to prevent age-associated AC degeneration.
Collapse
Affiliation(s)
- Seth D Thompson
- Shirley Ryan Abilitylab (Formerly the Rehabilitation Institute of Chicago), 355 E. Erie St, IL, 60611, Chicago, USA
- Department of Physical Medicine and Rehabilitation, Northwestern University, Chicago, USA
- Northwestern University Interdepartmental Neuroscience (NUIN) Graduate Program, Northwestern University, Chicago, USA
| | - Rajeswari Pichika
- Shirley Ryan Abilitylab (Formerly the Rehabilitation Institute of Chicago), 355 E. Erie St, IL, 60611, Chicago, USA
- Department of Physical Medicine and Rehabilitation, Northwestern University, Chicago, USA
| | - Richard L Lieber
- Shirley Ryan Abilitylab (Formerly the Rehabilitation Institute of Chicago), 355 E. Erie St, IL, 60611, Chicago, USA
- Department of Physical Medicine and Rehabilitation, Northwestern University, Chicago, USA
| | - Mitra Lavasani
- Shirley Ryan Abilitylab (Formerly the Rehabilitation Institute of Chicago), 355 E. Erie St, IL, 60611, Chicago, USA.
- Department of Physical Medicine and Rehabilitation, Northwestern University, Chicago, USA.
- Northwestern University Interdepartmental Neuroscience (NUIN) Graduate Program, Northwestern University, Chicago, USA.
| |
Collapse
|
26
|
Higuchi T, Suzuki D, Watanabe T, Fanhchaksai K, Ota K, Yokoo K, Furukawa H, Watanabe H. Versican contributes to ligament formation of knee joints. PLoS One 2021; 16:e0250366. [PMID: 33886644 PMCID: PMC8061984 DOI: 10.1371/journal.pone.0250366] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2020] [Accepted: 04/06/2021] [Indexed: 11/30/2022] Open
Abstract
Versican is a large proteoglycan in the extracellular matrix. During embryonic stages, it plays a crucial role in the development of cartilage, heart, and dermis. Previously, we reported that Prx1-Vcan conditional knockout mice, lacking Vcan expression in mesenchymal condensation areas of the limb bud, show the impaired joint formation and delayed cartilage development. Here, we investigated their phenotype in adults and found that they develop swelling of the knee joint. Histologically, their newborn joint exhibited impaired formation of both anterior and posterior cruciate ligaments. Immunostaining revealed a decrease in scleraxis-positive cells in both articular cartilage and ligament of Prx1-Vcan knee joint, spotty patterns of type I collagen, and the presence of type II collagen concomitant with the absence of versican expression. These results suggest that versican expression during the perinatal period is required for cruciate ligaments’ formation and that its depletion affects joint function in later ages.
Collapse
Affiliation(s)
- Tomoko Higuchi
- Department of Plastic Surgery, Aichi Medical University, Nagakute, Japan
| | - Daisuke Suzuki
- Department of Health Sciences, Hokkaido Chitose College of Rehabilitation, Chitose, Japan
| | - Takafumi Watanabe
- Laboratory of Veterinary Anatomy, School of Veterinary Medicine, Rakuno Gakuen University, Ebetsu, Japan
| | - Kanda Fanhchaksai
- Institute for Molecular Science of Medicine, Aichi Medical University, Nagakute, Japan
| | - Keiko Ota
- Institute for Molecular Science of Medicine, Aichi Medical University, Nagakute, Japan
| | - Kazuhisa Yokoo
- Department of Plastic Surgery, Aichi Medical University, Nagakute, Japan
| | - Hiroshi Furukawa
- Department of Plastic Surgery, Aichi Medical University, Nagakute, Japan
| | - Hideto Watanabe
- Institute for Molecular Science of Medicine, Aichi Medical University, Nagakute, Japan
- * E-mail:
| |
Collapse
|
27
|
Hong CC, Tang AT, Detter MR, Choi JP, Wang R, Yang X, Guerrero AA, Wittig CF, Hobson N, Girard R, Lightle R, Moore T, Shenkar R, Polster SP, Goddard LM, Ren AA, Leu NA, Sterling S, Yang J, Li L, Chen M, Mericko-Ishizuka P, Dow LE, Watanabe H, Schwaninger M, Min W, Marchuk DA, Zheng X, Awad IA, Kahn ML. Cerebral cavernous malformations are driven by ADAMTS5 proteolysis of versican. J Exp Med 2021; 217:151938. [PMID: 32648916 PMCID: PMC7537394 DOI: 10.1084/jem.20200140] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2020] [Revised: 03/30/2020] [Accepted: 05/20/2020] [Indexed: 12/18/2022] Open
Abstract
Cerebral cavernous malformations (CCMs) form following loss of the CCM protein complex in brain endothelial cells due to increased endothelial MEKK3 signaling and KLF2/4 transcription factor expression, but the downstream events that drive lesion formation remain undefined. Recent studies have revealed that CCM lesions expand by incorporating neighboring wild-type endothelial cells, indicative of a cell nonautonomous mechanism. Here we find that endothelial loss of ADAMTS5 reduced CCM formation in the neonatal mouse model. Conversely, endothelial gain of ADAMTS5 conferred early lesion genesis in the absence of increased KLF2/4 expression and synergized with KRIT1 loss of function to create large malformations. Lowering versican expression reduced CCM burden, indicating that versican is the relevant ADAMTS5 substrate and that lesion formation requires proteolysis but not loss of this extracellular matrix protein. These findings identify endothelial secretion of ADAMTS5 and cleavage of versican as downstream mechanisms of CCM pathogenesis and provide a basis for the participation of wild-type endothelial cells in lesion formation.
Collapse
Affiliation(s)
- Courtney C Hong
- Department of Medicine and Cardiovascular Institute, University of Pennsylvania, Philadelphia, PA
| | - Alan T Tang
- Department of Medicine and Cardiovascular Institute, University of Pennsylvania, Philadelphia, PA
| | - Matthew R Detter
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, NC
| | - Jaesung P Choi
- Centenary Institute, Sydney Medical School, University of Sydney, Sydney, Australia
| | - Rui Wang
- Department of Pharmacology, School of Basic Medical Sciences, Tianjian Medical University, Tianjin, China
| | - Xi Yang
- Department of Pharmacology, School of Basic Medical Sciences, Tianjian Medical University, Tianjin, China
| | - Andrea A Guerrero
- Department of Medicine and Cardiovascular Institute, University of Pennsylvania, Philadelphia, PA
| | - Carl F Wittig
- Department of Medicine and Cardiovascular Institute, University of Pennsylvania, Philadelphia, PA
| | - Nicholas Hobson
- Neurovascular Surgery Program, Section of Neurosurgery, Department of Surgery, The University of Chicago School of Medicine and Biological Sciences, Chicago, IL
| | - Romuald Girard
- Neurovascular Surgery Program, Section of Neurosurgery, Department of Surgery, The University of Chicago School of Medicine and Biological Sciences, Chicago, IL
| | - Rhonda Lightle
- Neurovascular Surgery Program, Section of Neurosurgery, Department of Surgery, The University of Chicago School of Medicine and Biological Sciences, Chicago, IL
| | - Thomas Moore
- Neurovascular Surgery Program, Section of Neurosurgery, Department of Surgery, The University of Chicago School of Medicine and Biological Sciences, Chicago, IL
| | - Robert Shenkar
- Neurovascular Surgery Program, Section of Neurosurgery, Department of Surgery, The University of Chicago School of Medicine and Biological Sciences, Chicago, IL
| | - Sean P Polster
- Neurovascular Surgery Program, Section of Neurosurgery, Department of Surgery, The University of Chicago School of Medicine and Biological Sciences, Chicago, IL
| | - Lauren M Goddard
- Department of Medicine and Cardiovascular Institute, University of Pennsylvania, Philadelphia, PA
| | - Aileen A Ren
- Department of Medicine and Cardiovascular Institute, University of Pennsylvania, Philadelphia, PA
| | - N Adrian Leu
- Department of Biomedical Sciences, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA
| | - Stephanie Sterling
- Department of Biomedical Sciences, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA
| | - Jisheng Yang
- Department of Medicine and Cardiovascular Institute, University of Pennsylvania, Philadelphia, PA
| | - Li Li
- Department of Medicine and Cardiovascular Institute, University of Pennsylvania, Philadelphia, PA
| | - Mei Chen
- Department of Medicine and Cardiovascular Institute, University of Pennsylvania, Philadelphia, PA
| | | | - Lukas E Dow
- Department of Medicine, Weill-Cornell Medicine, New York, NY
| | - Hideto Watanabe
- Institute for Molecular Science of Medicine, Aichi Medical University, Aichi, Japan
| | - Markus Schwaninger
- Institute of Experimental and Clinical Pharmacology and Toxicology, University of Lubeck, Lubeck, Germany
| | - Wang Min
- Interdepartmental Program in Vascular Biology and Therapeutics, Department of Pathology, Yale University School of Medicine, New Haven, CT
| | - Douglas A Marchuk
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, NC
| | - Xiangjian Zheng
- Centenary Institute, Sydney Medical School, University of Sydney, Sydney, Australia.,Department of Pharmacology, School of Basic Medical Sciences, Tianjian Medical University, Tianjin, China
| | - Issam A Awad
- Neurovascular Surgery Program, Section of Neurosurgery, Department of Surgery, The University of Chicago School of Medicine and Biological Sciences, Chicago, IL
| | - Mark L Kahn
- Department of Medicine and Cardiovascular Institute, University of Pennsylvania, Philadelphia, PA
| |
Collapse
|
28
|
Giannasi C, Niada S, Magagnotti C, Ragni E, Andolfo A, Brini AT. Comparison of two ASC-derived therapeutics in an in vitro OA model: secretome versus extracellular vesicles. Stem Cell Res Ther 2020; 11:521. [PMID: 33272318 PMCID: PMC7711257 DOI: 10.1186/s13287-020-02035-5] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Accepted: 11/18/2020] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND In the last years, several clinical trials have proved the safety and efficacy of adipose-derived stem/stromal cells (ASC) in contrasting osteoarthritis (OA). Since ASC act mainly through paracrine mechanisms, their secretome (conditioned medium, CM) represents a promising therapeutic alternative. ASC-CM is a complex cocktail of proteins, nucleic acids, and lipids released as soluble factors and/or conveyed into extracellular vesicles (EV). Here, we investigate its therapeutic potential in an in vitro model of OA. METHODS Human articular chondrocytes (CH) were induced towards an OA phenotype by 10 ng/ml TNFα in the presence of either ASC-CM or EV, both deriving from 5 × 105 cells, to evaluate the effect on hypertrophic, catabolic, and inflammatory markers. RESULTS Given the same number of donor cells, our data reveal a higher therapeutic potential of ASC-CM compared to EV alone that was confirmed by its enrichment in chondroprotective factors among which TIMP-1 and -2 stand out. In details, only ASC-CM significantly decreased MMP activity (22% and 29% after 3 and 6 days) and PGE2 expression (up to 40% at day 6) boosted by the inflammatory cytokine. Conversely, both treatments down-modulated of ~ 30% the hypertrophic marker COL10A1. CONCLUSIONS These biological and molecular evidences of ASC-CM beneficial action on CH with an induced OA phenotype may lay the basis for its future clinical translation as a cell-free therapeutic in the management of OA.
Collapse
Affiliation(s)
- Chiara Giannasi
- Laboratorio di Applicazioni Biotecnologiche, IRCCS Istituto Ortopedico Galeazzi, Milan, Italy.
| | - Stefania Niada
- Laboratorio di Applicazioni Biotecnologiche, IRCCS Istituto Ortopedico Galeazzi, Milan, Italy
| | - Cinzia Magagnotti
- Proteomics and Metabolomics Facility (ProMeFa), IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Enrico Ragni
- Laboratorio di Biotecnologie Applicate all'Ortopedia, IRCCS Istituto Ortopedico Galeazzi, Milan, Italy
| | - Annapaola Andolfo
- Proteomics and Metabolomics Facility (ProMeFa), IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Anna Teresa Brini
- Laboratorio di Applicazioni Biotecnologiche, IRCCS Istituto Ortopedico Galeazzi, Milan, Italy.,Department of Biomedical, Surgical and Dental Sciences, Università degli Studi di Milano, Milan, Italy
| |
Collapse
|
29
|
Yuan J, Jia J, Wu T, Liu X, Hu S, Zhang J, Ding R, Pang C, Cheng X. Comprehensive evaluation of differential long non-coding RNA and gene expression in patients with cartilaginous endplate degeneration of cervical vertebra. Exp Ther Med 2020; 20:260. [PMID: 33199985 PMCID: PMC7664616 DOI: 10.3892/etm.2020.9390] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2020] [Accepted: 07/31/2020] [Indexed: 02/07/2023] Open
Abstract
Long non-coding RNAs (lncRNAs) are emerging as key regulators in gene expression; however, little is currently known regarding their role in cartilaginous endplate (CE) degeneration (CED) of cervical vertebra. The present study aimed to investigate the expression levels of lncRNAs and analyze their potential functions in CED of cervical vertebra in patients with cervical fracture and cervical spondylosis. Human competitive endogenous RNA (ceRNA) array was used to analyze lncRNA and mRNA expression levels in CE samples from patients with cervical fracture and cervical spondylosis, who received anterior cervical discectomy and fusion. Differentially expressed lncRNAs (DELs) or differentially expressed genes (DEGs) were identified and functionally analyzed, using Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analyses. An lncRNA-microRNA(miRNA)-mRNA ceRNA regulatory network was constructed based on the DELs and DEGs, and the ceRNA network was visualized using Cytoscape 3.7.2 software. In total, one downregulated mRNA, one upregulated miRNA and five downstream regulated lncRNAs were identified using reverse transcription-quantitative PCR in CED and healthy CE samples. A total of 369 lncRNAs and 246 mRNAs were identified as differentially expressed in CE. The GO and KEGG analyses demonstrated that the majority of GO and KEGG enrichments were associated with CED. Furthermore, a ceRNA network was established, including 168 putative miRNA response elements, 189 upregulated and 37 downregulated lncRNAs and 47 upregulated and 10dow regulated DEGs. The present study analyzed the function of DEGs in the ceRNA network and filtered out the same items as in DEG-function enrichment analysis. These results provide a new perspective for an improved understanding of ceRNA-mediated gene regulation in cervical spondylosis, and provide a novel theoretical basis for further studies on the function of lncRNA in cervical spondylosis. However, further experiments are required to validate the results of the present study.
Collapse
Affiliation(s)
- Jinghong Yuan
- Department of Orthopedics, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
- Institute of Orthopedics of Jiangxi Province, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
- Institute of Minimally Invasive Orthopedics of Nanchang University, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Jingyu Jia
- Department of Orthopedics, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
- Institute of Orthopedics of Jiangxi Province, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
- Institute of Minimally Invasive Orthopedics of Nanchang University, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Tianlong Wu
- Department of Orthopedics, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
- Institute of Orthopedics of Jiangxi Province, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
- Institute of Minimally Invasive Orthopedics of Nanchang University, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Xijuan Liu
- Department of Pediatrics, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Shen Hu
- Department of Orthopedics, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Jian Zhang
- Department of Orthopedics, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Rui Ding
- Department of Orthopedics, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Chongzhi Pang
- Department of Orthopedics, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Xigao Cheng
- Department of Orthopedics, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
- Institute of Orthopedics of Jiangxi Province, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
- Institute of Minimally Invasive Orthopedics of Nanchang University, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
- Correspondence to: Professor Xigao Cheng, Department of Orthopedics, The Second Affiliated Hospital of Nanchang University, 1 Minde Road, Donghu, Nanchang, Jiangxi 330006, P.R. China
| |
Collapse
|
30
|
Islam S, Watanabe H. Versican: A Dynamic Regulator of the Extracellular Matrix. J Histochem Cytochem 2020; 68:763-775. [PMID: 33131383 DOI: 10.1369/0022155420953922] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Versican is a large chondroitin sulfate/dermatan sulfate proteoglycan belonging to the aggrecan/lectican family. In adults, this proteoglycan serves as a structural macromolecule of the extracellular matrix in the brain and large blood vessels. In contrast, versican is transiently expressed at high levels during development and under pathological conditions when the extracellular matrix dramatically changes, including in the inflammation and repair process. There are many reports showing the upregulation of versican in cancer, which correlates with cancer aggressiveness. Versican has four classical splice variants, and all the variants contain G1 and G3 domains at N- and C-termini, respectively. There are two glycosaminoglycan attachment domains CSα and CSβ. The largest V0 variant contains both CSα and CSβ, V1 contains CSβ, V2 contains CSα, and the shortest G3 variant has neither of them. Versican degradation is initiated by cleavage at a site in the CSβ domain by ADAMTS (a disintegrin and metalloproteinase with thrombospondin motifs) proteinases. The N-terminal fragment containing the G1 domain has been reported to exert various biological functions, although its mechanisms of action have not yet been elucidated. In this review, we describe the role of versican in inflammation and cancer and also address the biological function of versikine.
Collapse
Affiliation(s)
- Shamima Islam
- Institute for Molecular Science of Medicine, Aichi Medical University, Nagakute, Japan
| | - Hideto Watanabe
- Institute for Molecular Science of Medicine, Aichi Medical University, Nagakute, Japan
| |
Collapse
|
31
|
Bian Q, Cheng YH, Wilson JP, Su EY, Kim DW, Wang H, Yoo S, Blackshaw S, Cahan P. A single cell transcriptional atlas of early synovial joint development. Development 2020; 147:dev.185777. [PMID: 32580935 DOI: 10.1242/dev.185777] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2019] [Accepted: 06/09/2020] [Indexed: 12/14/2022]
Abstract
Synovial joint development begins with the formation of the interzone, a region of condensed mesenchymal cells at the site of the prospective joint. Recently, lineage-tracing strategies have revealed that Gdf5-lineage cells native to and from outside the interzone contribute to most, if not all, of the major joint components. However, there is limited knowledge of the specific transcriptional and signaling programs that regulate interzone formation and fate diversification of synovial joint constituents. To address this, we have performed single cell RNA-Seq analysis of 7329 synovial joint progenitor cells from the developing murine knee joint from E12.5 to E15.5. By using a combination of computational analytics, in situ hybridization and in vitro characterization of prospectively isolated populations, we have identified the transcriptional profiles of the major developmental paths for joint progenitors. Our freely available single cell transcriptional atlas will serve as a resource for the community to uncover transcriptional programs and cell interactions that regulate synovial joint development.
Collapse
Affiliation(s)
- Qin Bian
- Institute for Cell Engineering, Johns Hopkins School of Medicine, Baltimore MD 21205, USA.,Department of Biomedical Engineering, Johns Hopkins School of Medicine, Baltimore MD 21205, USA
| | - Yu-Hao Cheng
- Institute for Cell Engineering, Johns Hopkins School of Medicine, Baltimore MD 21205, USA.,Department of Molecular Biology and Genetics, Johns Hopkins School of Medicine, Baltimore MD 21205, USA
| | - Jordan P Wilson
- Institute for Cell Engineering, Johns Hopkins School of Medicine, Baltimore MD 21205, USA
| | - Emily Y Su
- Department of Biomedical Engineering, Johns Hopkins School of Medicine, Baltimore MD 21205, USA
| | - Dong Won Kim
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins School of Medicine, Baltimore MD 21205, USA
| | - Hong Wang
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins School of Medicine, Baltimore MD 21205, USA
| | - Sooyeon Yoo
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins School of Medicine, Baltimore MD 21205, USA
| | - Seth Blackshaw
- Institute for Cell Engineering, Johns Hopkins School of Medicine, Baltimore MD 21205, USA.,Solomon H. Snyder Department of Neuroscience, Johns Hopkins School of Medicine, Baltimore MD 21205, USA
| | - Patrick Cahan
- Institute for Cell Engineering, Johns Hopkins School of Medicine, Baltimore MD 21205, USA .,Department of Biomedical Engineering, Johns Hopkins School of Medicine, Baltimore MD 21205, USA.,Department of Molecular Biology and Genetics, Johns Hopkins School of Medicine, Baltimore MD 21205, USA
| |
Collapse
|
32
|
Anthwal N, Fenelon JC, Johnston SD, Renfree MB, Tucker AS. Transient role of the middle ear as a lower jaw support across mammals. eLife 2020; 9:e57860. [PMID: 32600529 PMCID: PMC7363448 DOI: 10.7554/elife.57860] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2020] [Accepted: 06/17/2020] [Indexed: 12/14/2022] Open
Abstract
Mammals articulate their jaws using a novel joint between the dentary and squamosal bones. In eutherian mammals, this joint forms in the embryo, supporting feeding and vocalisation from birth. In contrast, marsupials and monotremes exhibit extreme altriciality and are born before the bones of the novel mammalian jaw joint form. These mammals need to rely on other mechanisms to allow them to feed. Here, we show that this vital function is carried out by the earlier developing, cartilaginous incus of the middle ear, abutting the cranial base to form a cranio-mandibular articulation. The nature of this articulation varies between monotremes and marsupials, with juvenile monotremes retaining a double articulation, similar to that of the fossil mammaliaform Morganucodon, while marsupials use a versican-rich matrix to stabilise the jaw against the cranial base. These findings provide novel insight into the evolution of mammals and the changing relationship between the jaw and ear.
Collapse
Affiliation(s)
- Neal Anthwal
- Centre for Craniofacial and Regenerative Biology, King's College LondonLondonUnited Kingdom
| | - Jane C Fenelon
- School of BioSciences, University of MelbourneVictoriaAustralia
| | - Stephen D Johnston
- School of Agriculture and Food Sciences, University of QueenslandGattonAustralia
| | | | - Abigail S Tucker
- Centre for Craniofacial and Regenerative Biology, King's College LondonLondonUnited Kingdom
| |
Collapse
|
33
|
Liguori GR, Liguori TTA, de Moraes SR, Sinkunas V, Terlizzi V, van Dongen JA, Sharma PK, Moreira LFP, Harmsen MC. Molecular and Biomechanical Clues From Cardiac Tissue Decellularized Extracellular Matrix Drive Stromal Cell Plasticity. Front Bioeng Biotechnol 2020; 8:520. [PMID: 32548106 PMCID: PMC7273975 DOI: 10.3389/fbioe.2020.00520] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2019] [Accepted: 05/01/2020] [Indexed: 01/09/2023] Open
Abstract
Decellularized-organ-derived extracellular matrix (dECM) has been used for many years in tissue engineering and regenerative medicine. The manufacturing of hydrogels from dECM allows to make use of the pro-regenerative properties of the ECM and, simultaneously, to shape the material in any necessary way. The objective of the present project was to investigate differences between cardiovascular tissues (left ventricle, mitral valve, and aorta) with respect to generating dECM hydrogels and their interaction with cells in 2D and 3D. The left ventricle, mitral valve, and aorta of porcine hearts were decellularized using a series of detergent treatments (SDS, Triton-X 100 and deoxycholate). Mass spectrometry-based proteomics yielded the ECM proteins composition of the dECM. The dECM was digested with pepsin and resuspended in PBS (pH 7.4). Upon warming to 37°C, the suspension turns into a gel. Hydrogel stiffness was determined for samples with a dECM concentration of 20 mg/mL. Adipose tissue-derived stromal cells (ASC) and a combination of ASC with human pulmonary microvascular endothelial cells (HPMVEC) were cultured, respectively, on and in hydrogels to analyze cellular plasticity in 2D and vascular network formation in 3D. Differentiation of ASC was induced with 10 ng/mL of TGF-β1 and SM22α used as differentiation marker. 3D vascular network formation was evaluated with confocal microscopy after immunofluorescent staining of PECAM-1. In dECM, the most abundant protein was collagen VI for the left ventricle and mitral valve and elastin for the aorta. The stiffness of the hydrogel derived from the aorta (6,998 ± 895 Pa) was significantly higher than those derived from the left ventricle (3,384 ± 698 Pa) and the mitral valve (3,233 ± 323 Pa) (One-way ANOVA, p = 0.0008). Aorta-derived dECM hydrogel drove non-induced (without TGF-β1) differentiation, while hydrogels derived from the left ventricle and mitral valve inhibited TGF-β1-induced differentiation. All hydrogels supported vascular network formation within 7 days of culture, but ventricular dECM hydrogel demonstrated more robust vascular networks, with thicker and longer vascular structures. All the three main cardiovascular tissues, myocardium, valves, and large arteries, could be used to fabricate hydrogels from dECM, and these showed an origin-dependent influence on ASC differentiation and vascular network formation.
Collapse
Affiliation(s)
- Gabriel Romero Liguori
- Department of Pathology and Medical Biology, University Medical Center Groningen, University of Groningen, Groningen, Netherlands.,Instituto do Coração (InCor), Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil
| | - Tácia Tavares Aquinas Liguori
- Department of Pathology and Medical Biology, University Medical Center Groningen, University of Groningen, Groningen, Netherlands.,Instituto do Coração (InCor), Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil
| | - Sérgio Rodrigues de Moraes
- Instituto do Coração (InCor), Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil
| | - Viktor Sinkunas
- Instituto do Coração (InCor), Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil
| | - Vincenzo Terlizzi
- Department of Pathology and Medical Biology, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Joris A van Dongen
- Department of Pathology and Medical Biology, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Prashant K Sharma
- Department of Biomedical Engineering, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Luiz Felipe Pinho Moreira
- Instituto do Coração (InCor), Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil
| | - Martin Conrad Harmsen
- Department of Pathology and Medical Biology, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| |
Collapse
|
34
|
Li Y, Ma Q, Li P, Wang J, Wang M, Fan Y, Wang T, Wang C, Wang T, Zhao B. Proteomics reveals different pathological processes of adipose tissue, liver, and skeletal muscle under insulin resistance. J Cell Physiol 2020; 235:6441-6461. [PMID: 32115712 DOI: 10.1002/jcp.29658] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2019] [Accepted: 02/12/2020] [Indexed: 12/17/2022]
Abstract
Type 2 diabetes mellitus is the most common type of diabetes, and insulin resistance (IR) is its core pathological mechanism. Proteomics is an ingenious and promising Omics technology that can comprehensively describe the global protein expression profiling of body or specific tissue, and is widely applied to the study of molecular mechanisms of diseases. In this paper, we focused on insulin target organs: adipose tissue, liver, and skeletal muscle, and analyzed the different pathological processes of IR in these three tissues based on proteomics research. By literature studies, we proposed that the main pathological processes of IR among target organs were diverse, which showed unique characteristics and focuses. We further summarized the differential proteins in target organs which were verified to be related to IR, and discussed the proteins that may play key roles in the emphasized pathological processes, aiming at discovering potentially specific differential proteins of IR, and providing new ideas for pathological mechanism research of IR.
Collapse
Affiliation(s)
- Yaqi Li
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Quantao Ma
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Pengfei Li
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Jingkang Wang
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Min Wang
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Yuanyuan Fan
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Tieshan Wang
- Beijing Research Institute of Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Chunguo Wang
- Beijing Research Institute of Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Ting Wang
- Beijing Research Institute of Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Baosheng Zhao
- Beijing Research Institute of Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| |
Collapse
|
35
|
Tsiapalis D, De Pieri A, Spanoudes K, Sallent I, Kearns S, Kelly JL, Raghunath M, Zeugolis DI. The synergistic effect of low oxygen tension and macromolecular crowding in the development of extracellular matrix-rich tendon equivalents. Biofabrication 2020; 12:025018. [PMID: 31855856 DOI: 10.1088/1758-5090/ab6412] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Cellular therapies play an important role in tendon tissue engineering, with tenocytes being the most prominent and potent cell population available. However, for the development of a rich extracellular matrix tenocyte-assembled tendon equivalent, prolonged in vitro culture is required, which is associated with phenotypic drift. Recapitulation of tendon tissue microenvironment in vitro with cues that enhance and accelerate extracellular matrix synthesis and deposition, whilst maintaining tenocyte phenotype, may lead to functional cell therapies. Herein, we assessed the synergistic effect of low oxygen tension (enhances extracellular matrix synthesis) and macromolecular crowding (enhances extracellular matrix deposition) in human tenocyte culture. Protein analysis demonstrated that human tenocytes at 2% oxygen tension and with 50 μg ml-1 carrageenan (macromolecular crowder used) significantly increased synthesis and deposition of collagen types I, III, V and VI. Gene analysis at day 7 illustrated that human tenocytes at 2% oxygen tension and with 50 μg ml-1 carrageenan significantly increased the expression of prolyl 4-hydroxylase subunit alpha 1, procollagen-lysine 2- oxoglutarate 5-dioxygenase 2, scleraxis, tenomodulin and elastin, whilst chondrogenic (e.g. runt-related transcription factor 2, cartilage oligomeric matrix protein, aggrecan) and osteogenic (e.g. secreted phosphoprotein 1, bone gamma-carboxyglutamate protein) trans-differentiation markers were significantly down-regulated or remained unchanged. Collectively, our data clearly illustrates the beneficial synergistic effect of low oxygen tension and macromolecular crowding in the accelerated development of tissue equivalents.
Collapse
Affiliation(s)
- Dimitrios Tsiapalis
- Regenerative, Modular & Developmental Engineering Laboratory (REMODEL), Biomedical Sciences Building, National University of Ireland Galway (NUI Galway), Galway, Ireland. Science Foundation Ireland (SFI) Centre for Research in Medical Devices (CÚRAM), Biomedical Sciences Building, National University of Ireland Galway (NUI Galway), Galway, Ireland
| | | | | | | | | | | | | | | |
Collapse
|
36
|
Casanova MR, Reis RL, Martins A, Neves NM. Fibronectin Bound to a Fibrous Substrate Has Chondrogenic Induction Properties. Biomacromolecules 2020; 21:1368-1378. [DOI: 10.1021/acs.biomac.9b01546] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Affiliation(s)
- Marta R. Casanova
- 3B’s Research Group, I3Bs—Research Institute on Biomaterials, Biodegradables and Biomimetics of University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark—Parque de Ciência e Tecnologia, Zona Industrial da Gandra, Barco, 4805-017 Guimarães, Portugal
- ICVS/3B’s—PT Government Associate Laboratory, 4805-017 Braga/Guimarães, Portugal
| | - Rui L. Reis
- 3B’s Research Group, I3Bs—Research Institute on Biomaterials, Biodegradables and Biomimetics of University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark—Parque de Ciência e Tecnologia, Zona Industrial da Gandra, Barco, 4805-017 Guimarães, Portugal
- ICVS/3B’s—PT Government Associate Laboratory, 4805-017 Braga/Guimarães, Portugal
- The Discoveries Centre for Regenerative and Precision Medicine, Headquarters at University of Minho, Avepark, Barco, 4805-017 Guimarães, Portugal
| | - Albino Martins
- 3B’s Research Group, I3Bs—Research Institute on Biomaterials, Biodegradables and Biomimetics of University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark—Parque de Ciência e Tecnologia, Zona Industrial da Gandra, Barco, 4805-017 Guimarães, Portugal
- ICVS/3B’s—PT Government Associate Laboratory, 4805-017 Braga/Guimarães, Portugal
| | - Nuno M. Neves
- 3B’s Research Group, I3Bs—Research Institute on Biomaterials, Biodegradables and Biomimetics of University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark—Parque de Ciência e Tecnologia, Zona Industrial da Gandra, Barco, 4805-017 Guimarães, Portugal
- ICVS/3B’s—PT Government Associate Laboratory, 4805-017 Braga/Guimarães, Portugal
- The Discoveries Centre for Regenerative and Precision Medicine, Headquarters at University of Minho, Avepark, Barco, 4805-017 Guimarães, Portugal
| |
Collapse
|
37
|
Islam S, Chuensirikulchai K, Khummuang S, Keratibumrungpong T, Kongtawelert P, Kasinrerk W, Hatano S, Nagamachi A, Honda H, Watanabe H. Accumulation of versican facilitates wound healing: Implication of its initial ADAMTS-cleavage site. Matrix Biol 2019; 87:77-93. [PMID: 31669737 DOI: 10.1016/j.matbio.2019.10.006] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2019] [Revised: 10/20/2019] [Accepted: 10/21/2019] [Indexed: 11/16/2022]
Abstract
Versican is a large chondroitin sulfate/dermatan sulfate proteoglycan in the extracellular matrix, and is expressed at high levels in tissues during development and remodeling in pathological conditions. Its core protein is cleaved at a region close to the N-terminal end of CSβ domain by several members of a disintegrin and metalloproteinase with thrombospondin motifs (ADAMTS) family, i.e., ADAMTS-1, 4, 5, 9, 15, and 20. Here, using a CRISPR/Cas9 system, we generated knock-in mice (V1R), which express an ADAMTS cleavage-resistant versican. Some V1R homozygote mice, termed R/R, exhibit syndactyly and organ hemorrhage. In wound healing experiments, R/R wound shows accumulation of versican and activated TGFβ-signaling in the early stage, leading to faster healing than wild type wound. Immunostaining for Ki67, CD31, smooth muscle α-actin, periostin demonstrates higher levels of overall cell proliferation and an increased number of endothelial cells and myofibroblasts. Immunostaining for CD11b and qRT-PCR for macrophage markers revealed increased levels of inflammatory cell infiltration, especially those of M1 macrophages. Cultured R/R dermal fibroblasts revealed increased deposition of versican, type I and III collagens, and hyaluronan, and upregulation of Smad2/3 signaling. Taken together, these results demonstrate that the cleavage site determines versican turnover and that versican plays a central role in the provisional matrix during the wound repair.
Collapse
Affiliation(s)
- Shamima Islam
- Institute for Molecular Science of Medicine, Aichi Medical University, 1-1 Yazakokarimata, Nagakute, Aichi 480-1195, Japan
| | - Kantinan Chuensirikulchai
- Institute for Molecular Science of Medicine, Aichi Medical University, 1-1 Yazakokarimata, Nagakute, Aichi 480-1195, Japan; Biomedical Technology Research Center, Division of Clinical Immunology, Department of Medical Technology, Faculty of Associated Medical Sciences, Chiang Mai University, Chiang Mai, Thailand
| | - Saichit Khummuang
- Institute for Molecular Science of Medicine, Aichi Medical University, 1-1 Yazakokarimata, Nagakute, Aichi 480-1195, Japan; Biomedical Technology Research Center, Division of Clinical Immunology, Department of Medical Technology, Faculty of Associated Medical Sciences, Chiang Mai University, Chiang Mai, Thailand
| | - Tanyaporn Keratibumrungpong
- Institute for Molecular Science of Medicine, Aichi Medical University, 1-1 Yazakokarimata, Nagakute, Aichi 480-1195, Japan; Thailand Excellence Center for Tissue Engineering and Stem Cells, Department of Biochemistry, Faculty of Medicine, Chiang Mai University, Chiang Mai, 50200, Thailand
| | - Prachya Kongtawelert
- Thailand Excellence Center for Tissue Engineering and Stem Cells, Department of Biochemistry, Faculty of Medicine, Chiang Mai University, Chiang Mai, 50200, Thailand
| | - Watchara Kasinrerk
- Biomedical Technology Research Center, Division of Clinical Immunology, Department of Medical Technology, Faculty of Associated Medical Sciences, Chiang Mai University, Chiang Mai, Thailand
| | - Sonoko Hatano
- Institute for Molecular Science of Medicine, Aichi Medical University, 1-1 Yazakokarimata, Nagakute, Aichi 480-1195, Japan
| | - Akiko Nagamachi
- Department of Molecular Oncology and Leukemia Program Project, Research Institute for Radiation Biology and Medicine, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima, Japan
| | - Hiroaki Honda
- Field of Human Disease Models, Major in Advanced Life Sciences and Medicine, Institute of Laboratory Animals, Tokyo Women's Medical University, 8-1 Kawada-cho, Shinjuku-ku, Tokyo 162-8666, Japan
| | - Hideto Watanabe
- Institute for Molecular Science of Medicine, Aichi Medical University, 1-1 Yazakokarimata, Nagakute, Aichi 480-1195, Japan.
| |
Collapse
|
38
|
Bahbahani H, Musa HH, Wragg D, Shuiep ES, Almathen F, Hanotte O. Genome Diversity and Signatures of Selection for Production and Performance Traits in Dromedary Camels. Front Genet 2019; 10:893. [PMID: 31608121 PMCID: PMC6761857 DOI: 10.3389/fgene.2019.00893] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2019] [Accepted: 08/23/2019] [Indexed: 12/20/2022] Open
Abstract
Dromedary camels (Camelus dromedarius) are single-humped animals found throughout the deserts of Africa, the Arabian Peninsula, and the southwest of Asia. This well-adapted species is mainly used for milk and meat production, although some specific types exhibit superior running performance and are used in racing competitions. However, neither performance nor production camels are bred under intensive genomic selection programs with specific aims to improve these traits. In this study, the full genome sequence data of six camels from the Arabian Peninsula and the genotyping-by-sequencing data of 44 camels (29 packing and 15 racing) from Sudan were analyzed to assess their genome diversities, relationships, and candidate signatures of positive selection. Genome ADMIXTURE and principle component analyses indicate clear geographic separation between the Sudanese and the Arabian Peninsula camels, but with no population-specific genetic distinction within populations. Camel samples from the Arabian Peninsula show higher mean heterozygosity (0.560 ± 0.003) than those from Sudan (0.347 ± 0.003). Analyses of signatures of selection, using pooled heterozygosity (Hp) approach, in the Sudanese camels revealed 176, 189, and 308 candidate regions under positive selection in the combined and packing and racing camel populations, respectively. These regions host genes that might be associated with adaptation to arid environment, dairy traits, energy homeostasis, and chondrogenesis. Eight regions show high genetic differentiation, based on Fst analysis, between the Sudanese packing and racing camel types. Genes associated with chondrogenesis, energy balance, and urinary system development were found within these regions. Our results advocate for further detailed investigation of the genome of the dromedary camel to identify and characterize genes and variants associated with their valuable phenotypic traits. The results of which may support the development of breeding programs to improve the production and performance traits of this unique domesticated species.
Collapse
Affiliation(s)
- Hussain Bahbahani
- Department of Biological Sciences, Faculty of Science, Kuwait University, Kuwait City, Kuwait
| | - Hassan H Musa
- Department of Medical Microbiology, Faculty of Medical Laboratory Sciences, University of Khartoum, Khartoum North, Sudan
| | - David Wragg
- Centre for Tropical Livestock Genetics and Health, The Roslin Institute, Edinburgh, United Kingdom
| | - Eltahir S Shuiep
- Department of Animal Production, Faculty of Agricultural and Environmental Sciences, University of Gadarif, Gadarif State, Sudan
| | - Faisal Almathen
- Department of Public Health, College of Veterinary Medicine, King Faisal University, Al-Hasa, Saudi Arabia
| | - Olivier Hanotte
- LiveGene, International Livestock Research Institute (ILRI), Addis Ababa, Ethiopia
| |
Collapse
|
39
|
Karamanos NK, Piperigkou Z, Theocharis AD, Watanabe H, Franchi M, Baud S, Brézillon S, Götte M, Passi A, Vigetti D, Ricard-Blum S, Sanderson RD, Neill T, Iozzo RV. Proteoglycan Chemical Diversity Drives Multifunctional Cell Regulation and Therapeutics. Chem Rev 2018; 118:9152-9232. [PMID: 30204432 DOI: 10.1021/acs.chemrev.8b00354] [Citation(s) in RCA: 246] [Impact Index Per Article: 35.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Affiliation(s)
- Nikos K. Karamanos
- Biochemistry, Biochemical Analysis & Matrix Pathobiology Research Group, Laboratory of Biochemistry, Department of Chemistry, University of Patras, Patras 26110, Greece
- Foundation for Research and Technology-Hellas (FORTH)/Institute of Chemical Engineering Sciences (ICE-HT), Patras 26110, Greece
| | - Zoi Piperigkou
- Biochemistry, Biochemical Analysis & Matrix Pathobiology Research Group, Laboratory of Biochemistry, Department of Chemistry, University of Patras, Patras 26110, Greece
- Foundation for Research and Technology-Hellas (FORTH)/Institute of Chemical Engineering Sciences (ICE-HT), Patras 26110, Greece
| | - Achilleas D. Theocharis
- Biochemistry, Biochemical Analysis & Matrix Pathobiology Research Group, Laboratory of Biochemistry, Department of Chemistry, University of Patras, Patras 26110, Greece
| | - Hideto Watanabe
- Institute for Molecular Science of Medicine, Aichi Medical University, Aichi 480-1195, Japan
| | - Marco Franchi
- Department for Life Quality Studies, University of Bologna, Rimini 47100, Italy
| | - Stéphanie Baud
- Université de Reims Champagne-Ardenne, Laboratoire SiRMa, CNRS UMR MEDyC 7369, Faculté de Médecine, 51 rue Cognacq Jay, Reims 51100, France
| | - Stéphane Brézillon
- Université de Reims Champagne-Ardenne, Laboratoire de Biochimie Médicale et Biologie Moléculaire, CNRS UMR MEDyC 7369, Faculté de Médecine, 51 rue Cognacq Jay, Reims 51100, France
| | - Martin Götte
- Department of Gynecology and Obstetrics, Münster University Hospital, Münster 48149, Germany
| | - Alberto Passi
- Department of Medicine and Surgery, University of Insubria, Varese 21100, Italy
| | - Davide Vigetti
- Department of Medicine and Surgery, University of Insubria, Varese 21100, Italy
| | - Sylvie Ricard-Blum
- University Claude Bernard Lyon 1, CNRS, UMR 5246, Institute of Molecular and Supramolecular Chemistry and Biochemistry, Villeurbanne 69622, France
| | - Ralph D. Sanderson
- Department of Pathology, Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, Alabama 35294, United States
| | - Thomas Neill
- Department of Pathology, Anatomy and Cell Biology, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, Pennsylvania 10107, United States
| | - Renato V. Iozzo
- Department of Pathology, Anatomy and Cell Biology, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, Pennsylvania 10107, United States
| |
Collapse
|
40
|
Ankala A, Jain N, Hubbard B, Alexander JJ, Shankar SP. Is exon 8 the most critical or the only dispensable exon of the VCAN gene? Insights into VCAN variants and clinical spectrum of Wagner syndrome. Am J Med Genet A 2018; 176:1778-1783. [PMID: 30055036 DOI: 10.1002/ajmg.a.38855] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2017] [Revised: 04/26/2018] [Accepted: 05/01/2018] [Indexed: 11/10/2022]
Abstract
Wagner syndrome and erosive vitreoretinopathy together constitute the phenotypic continuum of an autosomal dominant vitreoretinopathy, with clinical findings typically isolated to the eye. The disease is caused by pathogenic variants in the VCAN gene and all such variants reported to date are those that plausibly result in haploinsufficiency of exon 8 containing vcan transcripts. Here, we report the molecular findings and long-term follow-up of a 16-year-old female with a history of retinal detachments and pigmentary retinal changes. Next-generation sequencing and microarray analysis of 141 genes established a diagnosis of Wagner syndrome in this individual, by detection of an 11.7 kilobase (kb) deletion encompassing exon 8 of VCAN. In light of the emerging functions and roles of versican protein in human disease, we discuss how variants within exon 8 of the VCAN gene can be compared to those in exon 2 of the COL2A1 gene that cause atypical Stickler syndrome and propose that variants in other regions of the gene can be expected to present with a more systemic disease. The distinctive facial features and atypical gastrointestinal symptoms observed in this long-term follow-up study support the possibility that individuals with VCAN-related vitreoretinopathy may have extra-ocular clinical features.
Collapse
Affiliation(s)
- Arunkanth Ankala
- Department of Human Genetics, Emory University School of Medicine, Atlanta, Georgia.,EGL Genetic Diagnostics, LLC, Atlanta, Georgia
| | - Nieraj Jain
- Department of Ophthalmology, Emory University, Atlanta, Georgia
| | - Baker Hubbard
- Department of Ophthalmology, Emory University, Atlanta, Georgia
| | - John J Alexander
- Department of Human Genetics, Emory University School of Medicine, Atlanta, Georgia.,EGL Genetic Diagnostics, LLC, Atlanta, Georgia
| | - Suma P Shankar
- Department of Human Genetics, Emory University School of Medicine, Atlanta, Georgia.,Department of Ophthalmology, Emory University, Atlanta, Georgia.,Division of Genomic Medicine, Department of Pediatrics, UC Davis School of Medicine, Sacramento, California
| |
Collapse
|
41
|
Hatano S, Nagai N, Sugiura N, Tsuchimoto J, Isogai Z, Kimata K, Ota A, Karnan S, Hosokawa Y, Watanabe H. Versican A-subdomain is required for its adequate function in dermal development. Connect Tissue Res 2018; 59:178-190. [PMID: 28488903 DOI: 10.1080/03008207.2017.1324432] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Versican, a large chondroitin sulfate (CS) proteoglycan, serves as a structural macromolecule of the extracellular matrix (ECM) and regulates cell behavior. We determined the function of versican in dermal development using VcanΔ3/Δ3 mutant mice expressing versican with deleted A-subdomain of the N-terminal G1 domain. The mutant versican showed a decreased hyaluronan (HA)-binding ability and failed to accumulate in the ECM. In the early developmental stage, VcanΔ3/Δ3 dermis showed a decrease in versican expression as compared with WT. As development proceeded, versican expression further decreased to a barely detectable level, and VcanΔ3/Δ3 mice died at the neonatal period (P0). At P0, VcanΔ3/Δ3 dermis exhibited an impaired ECM structure and decreased cell density. While the level of collagen deposition was similar in both genotypes, collagen biosynthesis significantly decreased in VcanΔ3/Δ3 fibroblasts as compared with that in wild type (WT). Transforming growth factor β (TGFβ) signaling mediated through the Smad2/3-dependent pathway was down-regulated in VcanΔ3/Δ3 fibroblasts and a reduced TGFβ storage in the ECM was observed. Microarray analysis revealed a decrease in the expression levels of transcription factors, early growth response (Egr) 2 and 4, which act downstream of TGFβ signaling. Thus, our results suggest that A-subdomain is necessary for adequate versican expression in dermis and that versican is involved in the formation of the ECM and regulation of TGFβ signaling.
Collapse
Affiliation(s)
- Sonoko Hatano
- a Institute for Molecular Science of Medicine, Aichi Medical University , Aichi , Japan
| | - Naoko Nagai
- a Institute for Molecular Science of Medicine, Aichi Medical University , Aichi , Japan
| | - Nobuo Sugiura
- a Institute for Molecular Science of Medicine, Aichi Medical University , Aichi , Japan
| | - Jun Tsuchimoto
- a Institute for Molecular Science of Medicine, Aichi Medical University , Aichi , Japan
| | - Zenzo Isogai
- b Department of Advanced Medicine , National Center for Geriatrics and Gerontology , Aichi , Japan
| | - Koji Kimata
- a Institute for Molecular Science of Medicine, Aichi Medical University , Aichi , Japan
| | - Akinobu Ota
- c Department of Biochemistry , Aichi Medical University School of Medicine , Aichi , Japan
| | - Sivasundaram Karnan
- c Department of Biochemistry , Aichi Medical University School of Medicine , Aichi , Japan
| | - Yoshitaka Hosokawa
- c Department of Biochemistry , Aichi Medical University School of Medicine , Aichi , Japan
| | - Hideto Watanabe
- a Institute for Molecular Science of Medicine, Aichi Medical University , Aichi , Japan
| |
Collapse
|
42
|
Glucocorticoids Improve Myogenic Differentiation In Vitro by Suppressing the Synthesis of Versican, a Transitional Matrix Protein Overexpressed in Dystrophic Skeletal Muscles. Int J Mol Sci 2017; 18:ijms18122629. [PMID: 29211034 PMCID: PMC5751232 DOI: 10.3390/ijms18122629] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2017] [Revised: 11/24/2017] [Accepted: 11/27/2017] [Indexed: 12/17/2022] Open
Abstract
In Duchenne muscular dystrophy (DMD), a dysregulated extracellular matrix (ECM) directly exacerbates pathology. Glucocorticoids are beneficial therapeutics in DMD, and have pleiotropic effects on the composition and processing of ECM proteins in other biological contexts. The synthesis and remodelling of a transitional versican-rich matrix is necessary for myogenesis; whether glucocorticoids modulate this transitional matrix is not known. Here, versican expression and processing were examined in hindlimb and diaphragm muscles from mdx dystrophin-deficient mice and C57BL/10 wild type mice. V0/V1 versican (Vcan) mRNA transcripts and protein levels were upregulated in dystrophic compared to wild type muscles, especially in the more severely affected mdx diaphragm. Processed versican (versikine) was detected in wild type and dystrophic muscles, and immunoreactivity was highly associated with newly regenerated myofibres. Glucocorticoids enhanced C2C12 myoblast fusion by modulating the expression of genes regulating transitional matrix synthesis and processing. Specifically, Tgfβ1, Vcan and hyaluronan synthase-2 (Has2) mRNA transcripts were decreased by 50% and Adamts1 mRNA transcripts were increased three-fold by glucocorticoid treatment. The addition of exogenous versican impaired myoblast fusion, whilst glucocorticoids alleviated this inhibition in fusion. In dystrophic mdx muscles, versican upregulation correlated with pathology. We propose that versican is a novel and relevant target gene in DMD, given its suppression by glucocorticoids and that in excess it impairs myoblast fusion, a process key for muscle regeneration.
Collapse
|
43
|
Michaelis M, Lucius R, Wiltfang J, Birkenfeld F. Determination of VEGF, collagen type 1 and versican in the discus articularis of the temporomandibular joint in relation to dental status. J Craniomaxillofac Surg 2017; 46:67-74. [PMID: 29198579 DOI: 10.1016/j.jcms.2017.09.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2017] [Revised: 07/08/2017] [Accepted: 09/14/2017] [Indexed: 10/18/2022] Open
Abstract
The aim of this study was the investigation and comparison of the presence of vascular endothelial growth factor (VEGF), collagen type 1 and the proteoglycan versican in the discus articularis in relation to dental status (full dentition [1], partial dentition [2] and edentulous [3]). The right disci articulares were removed from 17 donated bodies (6 with full dentition, 5 with partial dentition and 6 edentulous). The specimens were immunohistochemically stained for VEGF, collagen type 1 and versican. Semiquantitative analysis of the disci was conducted within the groups based on the intensity of immunoreactivity of VEGF, collagen type 1 and versican. In addition, a pairwise comparison was carried out between the three experimental groups. The results revealed significantly higher immunoreactivity for VEGF and versican in groups 2 and 3 than in group 1. Conversely, determination of immunoreactivity was significantly higher in group 1 for collagen type 1 than in the other two groups. These results indicate an elevated presence of the proteoglycan versican and the neoangiogenesis factor VEGF when the occlusal supporting zone has been lost. By contrast, detection of collagen type 1 is reduced. The loss of collagen type 1 and rise in versican and VEGF suggest increasing degeneration when the supporting zone is lost due to the loss of teeth.
Collapse
Affiliation(s)
- Malte Michaelis
- Institute of Anatomy (Head: Prof. Dr. Ralph Lucius), Christian-Albrechts-University of Kiel, Olshausenstraße 40, 24118 Kiel, Germany
| | - Ralph Lucius
- Institute of Anatomy (Head: Prof. Dr. Ralph Lucius), Christian-Albrechts-University of Kiel, Olshausenstraße 40, 24118 Kiel, Germany
| | - Jörg Wiltfang
- Department of Oral and Maxillofacial Surgery (Head: Prof. Dr. Dr. Jörg Wiltfang), University Hospital Schleswig-Holstein, Arnold-Heller-Straße 16, 24105 Kiel, Germany
| | - Falk Birkenfeld
- Department of Oral and Maxillofacial Surgery (Head: Prof. Dr. Dr. Jörg Wiltfang), University Hospital Schleswig-Holstein, Arnold-Heller-Straße 16, 24105 Kiel, Germany.
| |
Collapse
|
44
|
Zillikens MC, Demissie S, Hsu YH, Yerges-Armstrong LM, Chou WC, Stolk L, Livshits G, Broer L, Johnson T, Koller DL, Kutalik Z, Luan J, Malkin I, Ried JS, Smith AV, Thorleifsson G, Vandenput L, Hua Zhao J, Zhang W, Aghdassi A, Åkesson K, Amin N, Baier LJ, Barroso I, Bennett DA, Bertram L, Biffar R, Bochud M, Boehnke M, Borecki IB, Buchman AS, Byberg L, Campbell H, Campos Obanda N, Cauley JA, Cawthon PM, Cederberg H, Chen Z, Cho NH, Jin Choi H, Claussnitzer M, Collins F, Cummings SR, De Jager PL, Demuth I, Dhonukshe-Rutten RAM, Diatchenko L, Eiriksdottir G, Enneman AW, Erdos M, Eriksson JG, Eriksson J, Estrada K, Evans DS, Feitosa MF, Fu M, Garcia M, Gieger C, Girke T, Glazer NL, Grallert H, Grewal J, Han BG, Hanson RL, Hayward C, Hofman A, Hoffman EP, Homuth G, Hsueh WC, Hubal MJ, Hubbard A, Huffman KM, Husted LB, Illig T, Ingelsson E, Ittermann T, Jansson JO, Jordan JM, Jula A, Karlsson M, Khaw KT, Kilpeläinen TO, Klopp N, Kloth JSL, Koistinen HA, Kraus WE, Kritchevsky S, Kuulasmaa T, Kuusisto J, Laakso M, Lahti J, Lang T, Langdahl BL, Launer LJ, Lee JY, Lerch MM, Lewis JR, Lind L, Lindgren C, Liu Y, Liu T, Liu Y, Ljunggren Ö, Lorentzon M, Luben RN, Maixner W, McGuigan FE, Medina-Gomez C, Meitinger T, Melhus H, Mellström D, Melov S, Michaëlsson K, Mitchell BD, Morris AP, Mosekilde L, Newman A, Nielson CM, O'Connell JR, Oostra BA, Orwoll ES, Palotie A, Parker SCJ, Peacock M, Perola M, Peters A, Polasek O, Prince RL, Räikkönen K, Ralston SH, Ripatti S, Robbins JA, Rotter JI, Rudan I, Salomaa V, Satterfield S, Schadt EE, Schipf S, Scott L, Sehmi J, Shen J, Soo Shin C, Sigurdsson G, Smith S, Soranzo N, Stančáková A, Steinhagen-Thiessen E, Streeten EA, Styrkarsdottir U, Swart KMA, Tan ST, Tarnopolsky MA, Thompson P, Thomson CA, Thorsteinsdottir U, Tikkanen E, Tranah GJ, Tuomilehto J, van Schoor NM, Verma A, Vollenweider P, Völzke H, Wactawski-Wende J, Walker M, Weedon MN, Welch R, Wichmann HE, Widen E, Williams FMK, Wilson JF, Wright NC, Xie W, Yu L, Zhou Y, Chambers JC, Döring A, van Duijn CM, Econs MJ, Gudnason V, Kooner JS, Psaty BM, Spector TD, Stefansson K, Rivadeneira F, Uitterlinden AG, Wareham NJ, Ossowski V, Waterworth D, Loos RJF, Karasik D, Harris TB, Ohlsson C, Kiel DP. Large meta-analysis of genome-wide association studies identifies five loci for lean body mass. Nat Commun 2017; 8:80. [PMID: 28724990 PMCID: PMC5517526 DOI: 10.1038/s41467-017-00031-7] [Citation(s) in RCA: 111] [Impact Index Per Article: 13.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2015] [Accepted: 05/02/2017] [Indexed: 12/25/2022] Open
Abstract
Lean body mass, consisting mostly of skeletal muscle, is important for healthy aging. We performed a genome-wide association study for whole body (20 cohorts of European ancestry with n = 38,292) and appendicular (arms and legs) lean body mass (n = 28,330) measured using dual energy X-ray absorptiometry or bioelectrical impedance analysis, adjusted for sex, age, height, and fat mass. Twenty-one single-nucleotide polymorphisms were significantly associated with lean body mass either genome wide (p < 5 × 10-8) or suggestively genome wide (p < 2.3 × 10-6). Replication in 63,475 (47,227 of European ancestry) individuals from 33 cohorts for whole body lean body mass and in 45,090 (42,360 of European ancestry) subjects from 25 cohorts for appendicular lean body mass was successful for five single-nucleotide polymorphisms in/near HSD17B11, VCAN, ADAMTSL3, IRS1, and FTO for total lean body mass and for three single-nucleotide polymorphisms in/near VCAN, ADAMTSL3, and IRS1 for appendicular lean body mass. Our findings provide new insight into the genetics of lean body mass.Lean body mass is a highly heritable trait and is associated with various health conditions. Here, Kiel and colleagues perform a meta-analysis of genome-wide association studies for whole body lean body mass and find five novel genetic loci to be significantly associated.
Collapse
Affiliation(s)
- M Carola Zillikens
- Department of Internal Medicine, Erasmus MC, Rotterdam, 3000, The Netherlands
- Netherlands Genomics Initiative (NGI)-sponsored Netherlands Consortium for Healthy Aging (NCHA), Leiden, 2593, The Netherlands
| | - Serkalem Demissie
- Department of Biostatistics, Boston University School of Public Health, Boston, MA, 02118, USA
| | - Yi-Hsiang Hsu
- Hebrew SeniorLife, Institute for Aging Research, Roslindale, MA, 02131, USA
- Harvard Medical School, Boston, MA, 02115, USA
- Molecular and Integrative Physiological Sciences Program, Harvard School of Public Health, Boston, MA, 02115, USA
| | - Laura M Yerges-Armstrong
- Program in Personalized and Genomic Medicine, and Department of Medicine, Division of Endocrinology, Diabetes and Nutrition, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
| | - Wen-Chi Chou
- Hebrew SeniorLife, Institute for Aging Research, Roslindale, MA, 02131, USA
- Harvard Medical School, Boston, MA, 02115, USA
- Broad Institute, Cambridge, MA, 02142, USA
| | - Lisette Stolk
- Department of Internal Medicine, Erasmus MC, Rotterdam, 3000, The Netherlands
- Netherlands Genomics Initiative (NGI)-sponsored Netherlands Consortium for Healthy Aging (NCHA), Leiden, 2593, The Netherlands
| | - Gregory Livshits
- Sackler Faculty of Medicine, Department of Anatomy and Anthropology, Tel Aviv University, Tel Aviv, 6997801, Israel
- Department of Twin Research and Genetic Epidemiology, King's College London, St Thomas' Campus, London, WC2R 2LS, UK
| | - Linda Broer
- Department of Epidemiology, Erasmus MC, Rotterdam, 3000, The Netherlands
| | - Toby Johnson
- Department of Medical Genetics, University of Lausanne, Lausanne, 1011, Switzerland
- Swiss Institute of Bioinformatics, Lausanne, 1015, Switzerland
- Centre Hospitalier Universitaire (CHUV), University Institute for Social and Preventive Medicine, Lausanne, 1010, Switzerland
| | - Daniel L Koller
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Zoltán Kutalik
- Department of Medical Genetics, University of Lausanne, Lausanne, 1011, Switzerland
- Swiss Institute of Bioinformatics, Lausanne, 1015, Switzerland
- Centre Hospitalier Universitaire (CHUV), University Institute for Social and Preventive Medicine, Lausanne, 1010, Switzerland
| | - Jian'an Luan
- MRC Epidemiology Unit, University of Cambridge School of Clinical Medicine, Cambridge Biomedical Campus, Cambridge, CB2 OQQ, UK
| | - Ida Malkin
- Sackler Faculty of Medicine, Department of Anatomy and Anthropology, Tel Aviv University, Tel Aviv, 6997801, Israel
| | - Janina S Ried
- Institute of Epidemiology II, Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg, 85764, Germany
| | - Albert V Smith
- Icelandic Heart Association, Kopavogur, 201, Iceland
- Faculty of Medicine, University of Iceland, Reykjavik, 101, Iceland
| | | | - Liesbeth Vandenput
- Department of Internal Medicine, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, SE-405 30, Sweden
| | - Jing Hua Zhao
- MRC Epidemiology Unit, University of Cambridge School of Clinical Medicine, Cambridge Biomedical Campus, Cambridge, CB2 OQQ, UK
| | - Weihua Zhang
- Department Epidemiology and Biostatistics, School of Public Health, Imperial College, London, SW7 2AZ, UK
- Cardiology Department, Ealing Hospital NHS Trust, Middlesex, UB1 3HW, UK
| | - Ali Aghdassi
- Department of Medicine A, University of Greifswald, Greifswald, 17489, Germany
| | - Kristina Åkesson
- Department of Clinical Sciences, Lund University, Malmö, 22362, Sweden
- Department of Orthopedics, Skåne University Hospital, Malmö, S-205 02, Sweden
| | - Najaf Amin
- Department of Epidemiology, Erasmus MC, Rotterdam, 3000, The Netherlands
| | - Leslie J Baier
- Phoenix Epidemiology and Clinical Research Branch, National Institute of Diabetes and Digestive and Kidney Diseases, NIH, Phoenix, AZ, 85014, USA
| | - Inês Barroso
- Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton, CB10 1SA, UK
- NIHR Cambridge Biomedical Research Centre, Institute of Metabolic Science, Addenbrooke's Hospital, Cambridge, CB2 OQQ, UK
- Institute of Metabolic Science, Addenbrooke's Hospital, University of Cambridge Metabolic Research Laboratories, Cambridge, CB2 OQQ, UK
| | - David A Bennett
- Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, IL, 60612, USA
| | - Lars Bertram
- Lübeck Interdisciplinary Platform for Genome Analytics, Institutes of Neurogenetics and Experimental & Integrative Genomics, University of Lübeck, Lübeck, 23562, Germany
- School of Public Health, Faculty of Medicine, Imperial College London, London, W6 8RP, UK
| | - Rainer Biffar
- Centre of Oral Health, Department of Prosthetic Dentistry, Gerodontology and Biomaterials, University of Greifswald, Greifswald, 17489, Germany
| | - Murielle Bochud
- Centre Hospitalier Universitaire (CHUV), University Institute for Social and Preventive Medicine, Lausanne, 1010, Switzerland
| | - Michael Boehnke
- Department of Biostatistics and Center for Statistical Genetics, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Ingrid B Borecki
- Division of Statistical Genomics, Department of Genetics, Washington University School of Medicine, St Louis, MO, 63110, USA
- Division of Biostatistics, Washington University School of Medicine, St Louis, MO, 63110, USA
| | - Aron S Buchman
- Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, IL, 60612, USA
| | - Liisa Byberg
- Department of Surgical Sciences, Uppsala University, Uppsala, 75185, Sweden
| | - Harry Campbell
- Usher Institute of Population Health Sciences and Informatics, University of Edinburgh, Edinburgh, Scotland, EH8 9AG, UK
| | | | - Jane A Cauley
- Department of Epidemiology Graduate School of Public Health, University of Pittsburgh, Pittsburgh, PA, 15261, USA
| | - Peggy M Cawthon
- California Pacific Medical Center Research Institute, San Francisco, CA, 94107, USA
| | - Henna Cederberg
- Department of Medicine, University of Eastern Finland and Kuopio University Hospital, Kuopio, 70210, Finland
| | - Zhao Chen
- Mel and Enid Zuckerman College of Public Health, University of Arizona, Tucson, AZ, 85714, USA
| | - Nam H Cho
- Department of Preventive Medicine, Ajou University School of Medicine, Youngtong-Gu, Suwon, 16499, Korea
| | - Hyung Jin Choi
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul, 03080, Korea
- Department of Internal Medicine, Chungbuk National University Hospital, Cheongju Si, Korea
| | - Melina Claussnitzer
- Hebrew SeniorLife, Institute for Aging Research, Roslindale, MA, 02131, USA
- Harvard Medical School, Boston, MA, 02115, USA
- Broad Institute, Cambridge, MA, 02142, USA
- Computer Science and Artificial Intelligence Laboratory, MIT, Cambridge, MA, 02139, USA
- Institute of Human Genetics, MRI, Technische Universität München, Munich, 81675, Germany
- Beth Israel Deaconess Medical Center, Boston, MA, 02215, USA
| | - Francis Collins
- Medical Genomics and Metabolic Genetics Branch, National Human Genome Research Institute, Bethesda, MD, 20892, USA
| | - Steven R Cummings
- California Pacific Medical Center Research Institute, San Francisco, CA, 94107, USA
| | - Philip L De Jager
- Harvard Medical School, Boston, MA, 02115, USA
- Program in Translational NeuroPsychiatric Genomics, Department of Neurology, Brigham and Women's Hospital, Boston, MA, 02115, USA
- Program in Medical and Population Genetics, Broad Institute, Cambridge, MA, 02142, USA
| | - Ilja Demuth
- Lipid Clinic at the Interdisciplinary Metabolism Center, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, 13353, Germany
- Institute of Medical and Human Genetics, Charité - Universitätsmedizin Berlin, Berlin, 13353, Germany
| | | | - Luda Diatchenko
- Alan Edwards Centre for Research on Pain, McGill University, Montreal, H3A 0G1, Canada
- Regional Center for Neurosensory Disorders, School of Dentistry, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | | | - Anke W Enneman
- Department of Internal Medicine, Erasmus MC, Rotterdam, 3000, The Netherlands
| | - Mike Erdos
- Medical Genomics and Metabolic Genetics Branch, National Human Genome Research Institute, Bethesda, MD, 20892, USA
| | - Johan G Eriksson
- Department of General Practice and Primary Health Care, University of Helsinki, Helsinki, 00014, Finland
- Unit of General Practice, Helsinki University Central Hospital, Helsinki, 00014, Finland
- Folkhalsan Research Centre, Helsinki, 00250, Finland
- Vasa Central Hospital, Vasa, 65130, Finland
- National Institute for Health and Welfare, Helsinki, 00271, Finland
| | - Joel Eriksson
- Department of Internal Medicine, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, SE-405 30, Sweden
| | - Karol Estrada
- Department of Internal Medicine, Erasmus MC, Rotterdam, 3000, The Netherlands
- Department of Epidemiology, Erasmus MC, Rotterdam, 3000, The Netherlands
| | - Daniel S Evans
- California Pacific Medical Center Research Institute, San Francisco, CA, 94107, USA
| | - Mary F Feitosa
- Division of Statistical Genomics, Department of Genetics, Washington University School of Medicine, St Louis, MO, 63110, USA
| | - Mao Fu
- Program in Personalized and Genomic Medicine, and Department of Medicine, Division of Endocrinology, Diabetes and Nutrition, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
| | - Melissa Garcia
- Laboratory of Epidemiology and Population Sciences, Intramural Research Program, National Institute for Aging, Bethesda, MD, 20892, USA
| | - Christian Gieger
- Institute of Epidemiology II, Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg, 85764, Germany
- Research Unit of Molecular Epidemiology, Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg, 85764, Germany
- Institute of Genetic Epidemiology, Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg, 85764, Germany
| | - Thomas Girke
- Institute for Integrative Genome Biology, University of California, Riverside, CA, 92521, USA
- Department of Botany and Plant Sciences, University of California, Riverside, CA, 92521, USA
| | - Nicole L Glazer
- Departments of Medicine and Epidemiology, Boston University School of Medicine and Public Health, Boston, MA, 02118, USA
| | - Harald Grallert
- Institute of Epidemiology II, Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg, 85764, Germany
- Research Unit of Molecular Epidemiology, Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg, 85764, Germany
- Department of Botany and Plant Sciences, University of California, Riverside, CA, 92521, USA
- German Center for Diabetes Research (DZD), Neuherberg, Germany
- CCG Type 2 Diabetes, Helmholtz Zentrum München, Neuherberg, 85764, Germany
- CCG Nutrigenomics and Type 2 Diabetes. Helmholtz Zentrum München, Neuherberg, 85764, Germany
| | - Jagvir Grewal
- Cardiology Department, Ealing Hospital NHS Trust, Middlesex, UB1 3HW, UK
- National Heart and Lung Institute, Imperial College London, London, SW3 6LY, UK
| | - Bok-Ghee Han
- Center for Genome Science, National Institute of Health, Osong Health Technology Administration Complex, Chungcheongbuk-do, 28159, Korea
| | - Robert L Hanson
- Phoenix Epidemiology and Clinical Research Branch, National Institute of Diabetes and Digestive and Kidney Diseases, NIH, Phoenix, AZ, 85014, USA
| | - Caroline Hayward
- MRC Human Genetics Unit, IGMM, University of Edinburgh, Edinburgh, Scotland, EH4 2XU, UK
| | - Albert Hofman
- Netherlands Genomics Initiative (NGI)-sponsored Netherlands Consortium for Healthy Aging (NCHA), Leiden, 2593, The Netherlands
- Department of Epidemiology, Erasmus MC, Rotterdam, 3000, The Netherlands
| | - Eric P Hoffman
- Department of Pharmaceutical Sciences, SUNY Binghamton, Binghamton, NY, 13902, USA
| | - Georg Homuth
- Interfaculty Institute for Genetics and Functional Genomics, University of Greifswald, Greifswald, 17487, Germany
| | - Wen-Chi Hsueh
- Phoenix Epidemiology and Clinical Research Branch, National Institute of Diabetes and Digestive and Kidney Diseases, NIH, Phoenix, AZ, 85014, USA
| | - Monica J Hubal
- Department of Exercise and Nutrition Sciences, George Washington University, Washington, DC, 20052, USA
- Research Center for Genetic Medicine, Children's National Medical Center, Washington, DC, 20052, USA
| | - Alan Hubbard
- Division of Biostatistics, School of Public Health, University of California, Berkeley, CA, 94720, USA
| | - Kim M Huffman
- Division of Rheumatology, Department of Medicine, Duke Molecular Physiology Institute, Duke University School of Medicine, Durham, NC, 27710, USA
| | - Lise B Husted
- Endocrinology and Internal Medicine, Aarhus University Hospital, Aarhus, DK 8000, Denmark
| | - Thomas Illig
- Research Unit of Molecular Epidemiology, Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg, 85764, Germany
- Department of Human Genetics, Hannover Medical School, Hannover, 30625, Germany
- Hannover Unified Biobank, Hannover Medical School, Hannover, 30625, Germany
| | - Erik Ingelsson
- Department of Medical Sciences, Uppsala University, Uppsala, 75185, Sweden
- Division of Cardiovascular Medicine, Department of Medicine, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Till Ittermann
- Institute for Community Medicine, University of Greifswald, Greifswald, 17489, Germany
| | - John-Olov Jansson
- Department of Physiology, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, SE 405 30, Sweden
| | - Joanne M Jordan
- Thurston Arthritis Research Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27517, USA
| | - Antti Jula
- National Institute for Health and Welfare, Helsinki, 00271, Finland
| | - Magnus Karlsson
- Department of Clinical Sciences and Orthopaedics, Lund University, Skåne University Hospital SUS, Malmö, 22362, Sweden
| | - Kay-Tee Khaw
- Department of Public Health and Primary Care, University of Cambridge, Cambridge, CB1 8RN, UK
| | - Tuomas O Kilpeläinen
- MRC Epidemiology Unit, University of Cambridge School of Clinical Medicine, Cambridge Biomedical Campus, Cambridge, CB2 OQQ, UK
- The Novo Nordisk Foundation Center for Basic Metabolic Research, Section of Metabolic Genetics, University of Copenhagen, Copenhagen, 2100, Denmark
- Department of Environmental Medicine and Public Health, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Norman Klopp
- Research Unit of Molecular Epidemiology, Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg, 85764, Germany
- Hannover Unified Biobank, Hannover Medical School, Hannover, 30625, Germany
| | | | - Heikki A Koistinen
- Department of Medicine, University of Helsinki and Helsinki University Central Hospital, Helsinki, 00029, Finland
- Endocrinology, Abdominal Center, University of Helsinki and Helsinki University Central Hospital, Helsinki, 00029, Finland
- Department of Health, National Institute for Health and Welfare, Helsinki, 00271, Finland
- Minerva Foundation Institute for Medical Research, Helsinki, 00290, Finland
| | - William E Kraus
- Division of Cardiology, Department of Medicine, Duke Molecular Physiology Institute, Duke University School of Medicine, Durham, NC, 27710, USA
| | - Stephen Kritchevsky
- Sticht Center on Aging, Wake Forest School of Medicine, Winston-Salem, NC, 27157, USA
| | - Teemu Kuulasmaa
- Department of Medicine, University of Eastern Finland and Kuopio University Hospital, Kuopio, 70210, Finland
| | - Johanna Kuusisto
- Department of Medicine, University of Eastern Finland and Kuopio University Hospital, Kuopio, 70210, Finland
| | - Markku Laakso
- Department of Medicine, University of Eastern Finland and Kuopio University Hospital, Kuopio, 70210, Finland
| | - Jari Lahti
- Institute of Behavioural Sciences, University of Helsinki, Helsinki, FI00014, Finland
| | - Thomas Lang
- University of California San Francisco, San Francisco, CA, 94143, USA
| | - Bente L Langdahl
- Endocrinology and Internal Medicine, Aarhus University Hospital, Aarhus, DK 8000, Denmark
| | - Lenore J Launer
- Laboratory of Epidemiology and Population Sciences, Intramural Research Program, National Institute for Aging, Bethesda, MD, 20892, USA
| | - Jong-Young Lee
- Center for Genome Science, National Institute of Health, Osong Health Technology Administration Complex, Chungcheongbuk-do, 28159, Korea
| | - Markus M Lerch
- Department of Medicine A, University of Greifswald, Greifswald, 17489, Germany
| | - Joshua R Lewis
- School of Medicine and Pharmacology, University of Western Australia, Perth, 6009, Australia
- Centre for Kidney Research, School of Public Health, University of Sydney, Sydney, 2006, Australia
| | - Lars Lind
- Department of Medical Sciences, Uppsala University, Uppsala, 75185, Sweden
| | - Cecilia Lindgren
- Wellcome Trust Centre for Human Genetics, Oxford University, Oxford, OX3 7BN, UK
| | - Yongmei Liu
- Department of Epidemiology and Prevention, Wake Forest School of Medicine, Winston-Salem, NC, 27517, USA
| | - Tian Liu
- Max Planck Institute for Molecular Genetics, Berlin, 14195, Germany
- Max Planck Institute for Human Development, Berlin, 14195, Germany
| | - Youfang Liu
- Thurston Arthritis Research Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27517, USA
| | - Östen Ljunggren
- Department of Medical Sciences, Uppsala University, Uppsala, 75185, Sweden
| | - Mattias Lorentzon
- Department of Internal Medicine, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, SE-405 30, Sweden
| | - Robert N Luben
- Department of Public Health and Primary Care, University of Cambridge, Cambridge, CB1 8RN, UK
| | - William Maixner
- Regional Center for Neurosensory Disorders, School of Dentistry, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Fiona E McGuigan
- Department of Clinical Sciences, Lund University, Malmö, 22362, Sweden
| | - Carolina Medina-Gomez
- Department of Internal Medicine, Erasmus MC, Rotterdam, 3000, The Netherlands
- Department of Epidemiology, Erasmus MC, Rotterdam, 3000, The Netherlands
| | - Thomas Meitinger
- Institute of Human Genetics, MRI, Technische Universität München, Munich, 81675, Germany
- Institute of Human Genetics, Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg, 85764, Germany
| | - Håkan Melhus
- Department of Medical Sciences, Uppsala University, Uppsala, 75185, Sweden
| | - Dan Mellström
- Department of Internal Medicine, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, SE-405 30, Sweden
| | - Simon Melov
- Buck Institute for Research on Aging, Novato, CA, 94945, USA
- Leonard Davis School of Gerontology, University of Southern California, LA, CA, 90089, USA
| | - Karl Michaëlsson
- Department of Surgical Sciences, Uppsala University, Uppsala, 75185, Sweden
| | - Braxton D Mitchell
- Program in Personalized and Genomic Medicine, and Department of Medicine, Division of Endocrinology, Diabetes and Nutrition, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
- Geriatrics Research and Education Clinical Center, Baltimore Veterans Administration Medical Center, Baltimore, MD, 21201, USA
| | - Andrew P Morris
- Wellcome Trust Centre for Human Genetics, Oxford University, Oxford, OX3 7BN, UK
- Institute of Translational Medicine, University of Liverpool, Liverpool, L69 3BX, UK
| | - Leif Mosekilde
- Endocrinology and Internal Medicine, Aarhus University Hospital, Aarhus, DK 8000, Denmark
| | - Anne Newman
- Center for Aging and Population Health, University of Pittsburgh, Pittsburgh, PA, 15261, USA
| | | | - Jeffrey R O'Connell
- Program in Personalized and Genomic Medicine, and Department of Medicine, Division of Endocrinology, Diabetes and Nutrition, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
| | - Ben A Oostra
- Department of Clinical Genetics, Erasmus MC, Rotterdam, 300 CA, The Netherlands
- Centre for Medical Systems Biology and Netherlands Consortium on Healthy Aging, Leiden, RC2300, The Netherlands
| | - Eric S Orwoll
- Oregon Health & Science University, Portland, OR, 97239, USA
| | - Aarno Palotie
- Institute for Molecular Medicine Finland (FIMM), University of Helsinki, Helsinki, 00251, Finland
- Department of Medical Genetics, University of Helsinki and University Central Hospital, Helsinki, FI00014, Finland
| | - Stephen C J Parker
- Human Genetics and Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Munro Peacock
- Department of Medicine, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Markus Perola
- National Institute for Health and Welfare, Helsinki, 00271, Finland
- Institute for Molecular Medicine Finland (FIMM), University of Helsinki, Helsinki, 00251, Finland
- Diabetes and Obesity Research Program, University of Helsinki, Helsinki, FI00014, Finland
- Estonian Genome Center, University of Tartu, Tartu, Estonia
| | - Annette Peters
- Institute of Epidemiology II, Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg, 85764, Germany
- Research Unit of Molecular Epidemiology, Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg, 85764, Germany
| | - Ozren Polasek
- Faculty of Medicine, Department of Public Health, University of Split, Split, 21000, Croatia
| | - Richard L Prince
- School of Medicine and Pharmacology, University of Western Australia, Perth, 6009, Australia
- Department of Endocrinology and Diabetes, Sir Charles Gardiner Hospital, Perth, 6009, Australia
| | - Katri Räikkönen
- Institute of Behavioural Sciences, University of Helsinki, Helsinki, FI00014, Finland
| | - Stuart H Ralston
- Molecular Medicine Centre, MRC Institute of Genetics and Molecular Medicine, Western General Hospital, Edinburgh, Scotland, EH4 2XU, UK
| | - Samuli Ripatti
- Institute for Molecular Medicine Finland (FIMM), University of Helsinki, Helsinki, 00251, Finland
- Hjelt Institute, University of Helsinki, Helsinki, Finland
- Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton, CB10 1SA, UK
| | - John A Robbins
- Department of Medicine, University of California at Davis, Sacramento, CA, 95817, USA
| | - Jerome I Rotter
- Institute for Translational Genomic and Population Sciences, Los Angeles Biomedical Research Institute and Department of Pediatrics, Harbor UCLA Medical Center, Torrance, CA, 90502, USA
| | - Igor Rudan
- Usher Institute of Population Health Sciences and Informatics, University of Edinburgh, Edinburgh, Scotland, EH8 9AG, UK
| | - Veikko Salomaa
- National Institute for Health and Welfare, Helsinki, 00271, Finland
| | - Suzanne Satterfield
- Department of Preventive Medicine, University of Tennessee Health Science Center, Memphis, TN, 38163, USA
| | - Eric E Schadt
- Department of Genetics and Genomic Science, Institute of Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Sabine Schipf
- Institute for Community Medicine, University of Greifswald, Greifswald, 17489, Germany
| | - Laura Scott
- Department of Biostatistics and Center for Statistical Genetics, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Joban Sehmi
- Cardiology Department, Ealing Hospital NHS Trust, Middlesex, UB1 3HW, UK
- National Heart and Lung Institute, Imperial College London, London, SW3 6LY, UK
| | - Jian Shen
- Oregon Health & Science University, Portland, OR, 97239, USA
| | - Chan Soo Shin
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul, 03080, Korea
| | - Gunnar Sigurdsson
- Faculty of Medicine, University of Iceland, Reykjavik, 101, Iceland
- Department of Endocrinology and Metabolism, Landspitali, The National University Hospital of Iceland, Reykjavik, 101, Iceland
| | - Shad Smith
- Center for Translational Pain Medicine, Department of Anesthiology, Duke University Medical Center, Durham, NC, 27110, USA
| | - Nicole Soranzo
- Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton, CB10 1SA, UK
| | - Alena Stančáková
- Department of Medicine, University of Eastern Finland and Kuopio University Hospital, Kuopio, 70210, Finland
| | - Elisabeth Steinhagen-Thiessen
- Lipid Clinic at the Interdisciplinary Metabolism Center, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, 13353, Germany
| | - Elizabeth A Streeten
- Program in Personalized and Genomic Medicine, and Department of Medicine, Division of Endocrinology, Diabetes and Nutrition, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
- Geriatric Research and Education Clinical Center (GRECC) - Veterans Administration Medical Center, Baltimore, MD, 21201, USA
| | | | - Karin M A Swart
- Department of Epidemiology and Biostatistics, and the EMGO Institute, VU University Medical Center, Amsterdam, BT1081, The Netherlands
| | - Sian-Tsung Tan
- Cardiology Department, Ealing Hospital NHS Trust, Middlesex, UB1 3HW, UK
- National Heart and Lung Institute, Imperial College London, London, SW3 6LY, UK
| | - Mark A Tarnopolsky
- Department of Medicine, McMaster University Medical Center, Hamilton, ON, Canada, L8N 3Z5
| | - Patricia Thompson
- Department of Pathology, Stony Brook School of Medicine, Stony Brook, NY, 11794, USA
| | - Cynthia A Thomson
- Mel and Enid Zuckerman College of Public Health, University of Arizona, Tucson, AZ, 85714, USA
| | - Unnur Thorsteinsdottir
- Faculty of Medicine, University of Iceland, Reykjavik, 101, Iceland
- deCODE Genetics, Reykjavik, 101, Iceland
| | - Emmi Tikkanen
- National Institute for Health and Welfare, Helsinki, 00271, Finland
- Institute for Molecular Medicine Finland (FIMM), University of Helsinki, Helsinki, 00251, Finland
- Molecular Medicine Centre, MRC Institute of Genetics and Molecular Medicine, Western General Hospital, Edinburgh, Scotland, EH4 2XU, UK
| | - Gregory J Tranah
- California Pacific Medical Center Research Institute, San Francisco, CA, 94107, USA
| | - Jaakko Tuomilehto
- Vasa Central Hospital, Vasa, 65130, Finland
- Department of Neuroscience and Preventive Medicine, Danube-University Krems, Krems, 3500, Austria
- Diabetes Research Group, King Abdulaziz University, Jeddah, 12589, Saudi Arabia
- Dasman Diabetes Institute, Dasman, 15462, Kuwait
| | - Natasja M van Schoor
- Department of Epidemiology and Biostatistics, and the EMGO Institute, VU University Medical Center, Amsterdam, BT1081, The Netherlands
| | - Arjun Verma
- Cardiology Department, Ealing Hospital NHS Trust, Middlesex, UB1 3HW, UK
| | - Peter Vollenweider
- Department of Medicine and Internal Medicine, Centre Hospitalier Universitaire Vaudois (CHUV), Lausanne, CH-1011, Switzerland
| | - Henry Völzke
- Institute for Community Medicine, University of Greifswald, Greifswald, 17489, Germany
| | - Jean Wactawski-Wende
- Department of Epidemiology and Environmental Health, University at Buffalo, State University of New York, Buffalo, NY, 14214, USA
| | - Mark Walker
- Institute of Cellular Medicine, Newcastle University, Newcastle upon Tyne, NE2 4HH, UK
| | - Michael N Weedon
- Genetics of Complex Traits, University of Exeter Medical School, Exeter, EX1 2LU, UK
| | - Ryan Welch
- Department of Biostatistics and Center for Statistical Genetics, University of Michigan, Ann Arbor, MI, 48109, USA
| | - H-Erich Wichmann
- Institute of Epidemiology II, Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg, 85764, Germany
- Institute of Medical Informatics, Biometry and Epidemiology, Chair of Epidemiology, Ludwig-Maximilians-Universität, Munich, 81377, Germany
- Institute of Medical Statistics and Epidemiology, Technical University, Munich, 81675, Germany
| | - Elisabeth Widen
- Institute for Molecular Medicine Finland (FIMM), University of Helsinki, Helsinki, 00251, Finland
| | - Frances M K Williams
- Department of Twin Research and Genetic Epidemiology, King's College London, St Thomas' Campus, London, WC2R 2LS, UK
| | - James F Wilson
- Usher Institute of Population Health Sciences and Informatics, University of Edinburgh, Edinburgh, Scotland, EH8 9AG, UK
- MRC Human Genetics Unit, IGMM, University of Edinburgh, Edinburgh, Scotland, EH4 2XU, UK
| | - Nicole C Wright
- Department of Epidemiology, University of Alabama at Birmingham, Birmingham, AL, 35294, USA
| | - Weijia Xie
- Genetics of Complex Traits, University of Exeter Medical School, Exeter, EX1 2LU, UK
| | - Lei Yu
- Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, IL, 60612, USA
| | - Yanhua Zhou
- Department of Biostatistics, Boston University School of Public Health, Boston, MA, 02118, USA
| | - John C Chambers
- Department Epidemiology and Biostatistics, School of Public Health, Imperial College, London, SW7 2AZ, UK
- Cardiology Department, Ealing Hospital NHS Trust, Middlesex, UB1 3HW, UK
- NIHR Cardiovascular Biomedical Research Unit, Royal Brompton and Harefield NHS Foundation Trust and Imperial College, London, SW3 6NP, UK
- Imperial College Healthcare NHS Trust, London, W2 1NY, UK
| | - Angela Döring
- Institute of Epidemiology II, Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg, 85764, Germany
- Institute of Epidemiology I, Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg, 85764, Germany
| | - Cornelia M van Duijn
- Department of Epidemiology, Erasmus MC, Rotterdam, 3000, The Netherlands
- Centre for Medical Systems Biology and Netherlands Consortium on Healthy Aging, Leiden, RC2300, The Netherlands
| | - Michael J Econs
- Department of Medicine and Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Vilmundur Gudnason
- Icelandic Heart Association, Kopavogur, 201, Iceland
- Faculty of Medicine, University of Iceland, Reykjavik, 101, Iceland
| | - Jaspal S Kooner
- Cardiology Department, Ealing Hospital NHS Trust, Middlesex, UB1 3HW, UK
- National Heart and Lung Institute, Imperial College London, London, SW3 6LY, UK
- Imperial College Healthcare NHS Trust, London, W2 1NY, UK
| | - Bruce M Psaty
- Departments of Medicine, Epidemiology, and Health Services, Cardiovascular Health Research Unit, University of Washington, Seattle, WA, 98101, USA
- Kaiser Permanente Washington Health Research Institute, Washington, Seattle, WA, 98101, USA
| | - Timothy D Spector
- Department of Twin Research and Genetic Epidemiology, King's College London, St Thomas' Campus, London, WC2R 2LS, UK
| | - Kari Stefansson
- Faculty of Medicine, University of Iceland, Reykjavik, 101, Iceland
- deCODE Genetics, Reykjavik, 101, Iceland
| | - Fernando Rivadeneira
- Department of Internal Medicine, Erasmus MC, Rotterdam, 3000, The Netherlands
- Netherlands Genomics Initiative (NGI)-sponsored Netherlands Consortium for Healthy Aging (NCHA), Leiden, 2593, The Netherlands
- Department of Epidemiology, Erasmus MC, Rotterdam, 3000, The Netherlands
| | - André G Uitterlinden
- Department of Internal Medicine, Erasmus MC, Rotterdam, 3000, The Netherlands
- Netherlands Genomics Initiative (NGI)-sponsored Netherlands Consortium for Healthy Aging (NCHA), Leiden, 2593, The Netherlands
- Department of Epidemiology, Erasmus MC, Rotterdam, 3000, The Netherlands
| | - Nicholas J Wareham
- MRC Epidemiology Unit, University of Cambridge School of Clinical Medicine, Cambridge Biomedical Campus, Cambridge, CB2 OQQ, UK
| | - Vicky Ossowski
- Phoenix Epidemiology and Clinical Research Branch, National Institute of Diabetes and Digestive and Kidney Diseases, NIH, Phoenix, AZ, 85014, USA
| | - Dawn Waterworth
- Medical Genetics, GlaxoSmithKline, Philadelphia, PA, 19112, USA
| | - Ruth J F Loos
- MRC Epidemiology Unit, University of Cambridge School of Clinical Medicine, Cambridge Biomedical Campus, Cambridge, CB2 OQQ, UK
- The Charles Bronfman Institute for Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Institute of Child Health and Development, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- The Genetics of Obesity and Related Traits Program, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Department of Preventive Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - David Karasik
- Hebrew SeniorLife, Institute for Aging Research, Roslindale, MA, 02131, USA
- Harvard Medical School, Boston, MA, 02115, USA
- Faculty of Medicine in the Galilee, Bar-Ilan University, Safed, 1311502, Israel
| | - Tamara B Harris
- Laboratory of Epidemiology and Population Sciences, Intramural Research Program, National Institute for Aging, Bethesda, MD, 20892, USA
| | - Claes Ohlsson
- Department of Internal Medicine, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, SE-405 30, Sweden
| | - Douglas P Kiel
- Hebrew SeniorLife, Institute for Aging Research, Roslindale, MA, 02131, USA.
- Harvard Medical School, Boston, MA, 02115, USA.
- Beth Israel Deaconess Medical Center, Boston, MA, 02215, USA.
| |
Collapse
|
45
|
Pauly HM, Place LW, Haut Donahue TL, Kipper MJ. Mechanical Properties and Cell Compatibility of Agarose Hydrogels Containing Proteoglycan Mimetic Graft Copolymers. Biomacromolecules 2017; 18:2220-2229. [DOI: 10.1021/acs.biomac.7b00643] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Affiliation(s)
- Hannah M. Pauly
- School
of Biomedical Engineering, ∥Department of Mechanical Engineering, and ⊥Department of
Chemical and Biological Engineering, Colorado State University, 1370 Campus Delivery, Fort Collins, Colorado United States
| | - Laura W. Place
- School
of Biomedical Engineering, ∥Department of Mechanical Engineering, and ⊥Department of
Chemical and Biological Engineering, Colorado State University, 1370 Campus Delivery, Fort Collins, Colorado United States
| | - Tammy L. Haut Donahue
- School
of Biomedical Engineering, ∥Department of Mechanical Engineering, and ⊥Department of
Chemical and Biological Engineering, Colorado State University, 1370 Campus Delivery, Fort Collins, Colorado United States
| | - Matt J. Kipper
- School
of Biomedical Engineering, ∥Department of Mechanical Engineering, and ⊥Department of
Chemical and Biological Engineering, Colorado State University, 1370 Campus Delivery, Fort Collins, Colorado United States
| |
Collapse
|
46
|
The ADAMTS hyalectanase family: biological insights from diverse species. Biochem J 2017; 473:2011-22. [PMID: 27407170 DOI: 10.1042/bcj20160148] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2016] [Accepted: 03/29/2016] [Indexed: 12/13/2022]
Abstract
The a disintegrin-like and metalloproteinase with thrombospondin type-1 motifs (ADAMTS) family of metzincins are complex secreted proteins that have diverse functions during development. The hyalectanases (ADAMTS1, 4, 5, 8, 9, 15 and 20) are a subset of this family that have enzymatic activity against hyalectan proteoglycans, the processing of which has important implications during development. This review explores the evolution, expression and developmental functions of the ADAMTS family, focusing on the ADAMTS hyalectanases and their substrates in diverse species. This review gives an overview of how the family and their substrates evolved from non-vertebrates to mammals, the expression of the hyalectanases and substrates in different species and their functions during development, and how these functions are conserved across species.
Collapse
|
47
|
Danford ID, Verkuil LD, Choi DJ, Collins DW, Gudiseva HV, Uyhazi KE, Lau MK, Kanu LN, Grant GR, Chavali VRM, O'Brien JM. Characterizing the "POAGome": A bioinformatics-driven approach to primary open-angle glaucoma. Prog Retin Eye Res 2017; 58:89-114. [PMID: 28223208 PMCID: PMC5464971 DOI: 10.1016/j.preteyeres.2017.02.001] [Citation(s) in RCA: 51] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2016] [Revised: 02/03/2017] [Accepted: 02/10/2017] [Indexed: 01/10/2023]
Abstract
Primary open-angle glaucoma (POAG) is a genetically, physiologically, and phenotypically complex neurodegenerative disorder. This study addressed the expanding collection of genes associated with POAG, referred to as the "POAGome." We used bioinformatics tools to perform an extensive, systematic literature search and compiled 542 genes with confirmed associations with POAG and its related phenotypes (normal tension glaucoma, ocular hypertension, juvenile open-angle glaucoma, and primary congenital glaucoma). The genes were classified according to their associated ocular tissues and phenotypes, and functional annotation and pathway analyses were subsequently performed. Our study reveals that no single molecular pathway can encompass the pathophysiology of POAG. The analyses suggested that inflammation and senescence may play pivotal roles in both the development and perpetuation of the retinal ganglion cell degeneration seen in POAG. The TGF-β signaling pathway was repeatedly implicated in our analyses, suggesting that it may be an important contributor to the manifestation of POAG in the anterior and posterior segments of the globe. We propose a molecular model of POAG revolving around TGF-β signaling, which incorporates the roles of inflammation and senescence in this disease. Finally, we highlight emerging molecular therapies that show promise for treating POAG.
Collapse
Affiliation(s)
- Ian D Danford
- Scheie Eye Institute, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Lana D Verkuil
- Scheie Eye Institute, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Daniel J Choi
- Scheie Eye Institute, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - David W Collins
- Scheie Eye Institute, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Harini V Gudiseva
- Scheie Eye Institute, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Katherine E Uyhazi
- Scheie Eye Institute, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Marisa K Lau
- Scheie Eye Institute, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Levi N Kanu
- Scheie Eye Institute, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Gregory R Grant
- Department of Genetics, University of Pennsylvania, Philadelphia, PA, USA, Penn Center for Bioinformatics, University of Pennsylvania, Philadelphia, PA, USA
| | - Venkata R M Chavali
- Scheie Eye Institute, University of Pennsylvania, Philadelphia, PA, 19104, USA.
| | - Joan M O'Brien
- Scheie Eye Institute, University of Pennsylvania, Philadelphia, PA, 19104, USA
| |
Collapse
|
48
|
Kudo T, Nakatani S, Kakizaki M, Arai A, Ishida K, Wada M, Kobata K. Supplemented Chondroitin Sulfate and Hyaluronic Acid Suppress Mineralization of the Chondrogenic Cell Line, ATDC5, via Direct Inhibition of Alkaline Phosphatase. Biol Pharm Bull 2017; 40:2075-2080. [DOI: 10.1248/bpb.b17-00059] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Affiliation(s)
- Toshiya Kudo
- Department of Pharmaceutical Sciences, Josai University
| | | | | | - Ai Arai
- Department of Pharmaceutical Sciences, Josai University
| | | | - Masahiro Wada
- Department of Pharmaceutical Sciences, Josai University
| | - Kenji Kobata
- Department of Pharmaceutical Sciences, Josai University
| |
Collapse
|
49
|
Kang I, Harten IA, Chang MY, Braun KR, Sheih A, Nivison MP, Johnson PY, Workman G, Kaber G, Evanko SP, Chan CK, Merrilees MJ, Ziegler SF, Kinsella MG, Frevert CW, Wight TN. Versican Deficiency Significantly Reduces Lung Inflammatory Response Induced by Polyinosine-Polycytidylic Acid Stimulation. J Biol Chem 2016; 292:51-63. [PMID: 27895126 DOI: 10.1074/jbc.m116.753186] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2016] [Revised: 11/10/2016] [Indexed: 12/16/2022] Open
Abstract
Viral infection is an exacerbating factor contributing to chronic airway diseases, such as asthma, via mechanisms that are still unclear. Polyinosine-polycytidylic acid (poly(I:C)), a Toll-like receptor 3 (TLR3) agonist used as a mimetic to study viral infection, has been shown to elicit inflammatory responses in lungs and to exacerbate pulmonary allergic reactions in animal models. Previously, we have shown that poly(I:C) stimulates lung fibroblasts to accumulate an extracellular matrix (ECM), enriched in hyaluronan (HA) and its binding partner versican, which promotes monocyte adhesion. In the current study, we aimed to determine the in vivo role of versican in mediating inflammatory responses in poly(I:C)-induced lung inflammation using a tamoxifen-inducible versican-deficient mouse model (Vcan-/- mice). In C57Bl/6 mice, poly(I:C) instillation significantly increased accumulation of versican and HA, especially in the perivascular and peribronchial regions, which were enriched in infiltrating leukocytes. In contrast, versican-deficient (Vcan-/-) lungs did not exhibit increases in versican or HA in these regions and had strikingly reduced numbers of leukocytes in the bronchoalveolar lavage fluid and lower expression of inflammatory chemokines and cytokines. Poly(I:C) stimulation of lung fibroblasts isolated from control mice generated HA-enriched cable structures in the ECM, providing a substrate for monocytic cells in vitro, whereas lung fibroblasts from Vcan-/- mice did not. Moreover, increases in proinflammatory cytokine expression were also greatly attenuated in the Vcan-/- lung fibroblasts. These findings provide strong evidence that versican is a critical inflammatory mediator during poly(I:C)-induced acute lung injury and, in association with HA, generates an ECM that promotes leukocyte infiltration and adhesion.
Collapse
Affiliation(s)
| | | | - Mary Y Chang
- the Department of Comparative Medicine and Center for Lung Biology, University of Washington, Seattle, Washington 98109, and
| | | | - Alyssa Sheih
- Immunology Program, Benaroya Research Institute, Seattle, Washington 98101
| | | | | | | | | | | | | | - Mervyn J Merrilees
- the Department of Anatomy and Medical Imaging, School of Medical Sciences, University of Auckland, Auckland 1010, New Zealand
| | - Steven F Ziegler
- Immunology Program, Benaroya Research Institute, Seattle, Washington 98101
| | | | - Charles W Frevert
- the Department of Comparative Medicine and Center for Lung Biology, University of Washington, Seattle, Washington 98109, and
| | | |
Collapse
|
50
|
Saha A, Rolfe R, Carroll S, Kelly DJ, Murphy P. Chondrogenesis of embryonic limb bud cells in micromass culture progresses rapidly to hypertrophy and is modulated by hydrostatic pressure. Cell Tissue Res 2016; 368:47-59. [PMID: 27770257 DOI: 10.1007/s00441-016-2512-9] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2016] [Accepted: 09/17/2016] [Indexed: 12/18/2022]
Abstract
Chondrogenesis in vivo is precisely controlled in time and space. The entire limb skeleton forms from cells at the core of the early limb bud that condense and undergo chondrogenic differentiation. Whether they form stable cartilage at the articular surface of the joint or transient cartilage that progresses to hypertrophy as endochondral bone, replacing the cartilage template of the skeletal rudiment, is spatially controlled over several days in the embryo. Here, we follow the differentiation of cells taken from the early limb bud (embryonic day 11.5), grown in high-density micromass culture and show that a self-organising pattern of evenly spaced cartilage nodules occurs spontaneously in growth medium. Although chondrogenesis is enhanced by addition of BMP6 to the medium, the spatial pattern of nodule formation is disrupted. We show rapid progression of the entire nodule to hypertrophy in culture and therefore loss of the local signals required to direct formation of stable cartilage. Dynamic hydrostatic pressure, which we have previously predicted to be a feature of the forming embryonic joint region, had a stabilising effect on chondrogenesis, reducing expression of hypertrophic marker genes. This demonstrates the use of micromass culture as a relatively simple assay to compare the effect of both biophysical and molecular signals on spatial and temporal control of chondrogenesis that could be used to examine the response of different types of progenitor cell, both adult- and embryo-derived.
Collapse
Affiliation(s)
- Anurati Saha
- Department of Zoology, School of Natural Sciences, Trinity College, Dublin, Ireland
| | - Rebecca Rolfe
- Department of Zoology, School of Natural Sciences, Trinity College, Dublin, Ireland.,Trinity Centre for Bioengineering, School of Engineering, Trinity College, Dublin, Ireland
| | - Simon Carroll
- Trinity Centre for Bioengineering, School of Engineering, Trinity College, Dublin, Ireland
| | - Daniel J Kelly
- Trinity Centre for Bioengineering, School of Engineering, Trinity College, Dublin, Ireland
| | - Paula Murphy
- Department of Zoology, School of Natural Sciences, Trinity College, Dublin, Ireland. .,Trinity Centre for Bioengineering, School of Engineering, Trinity College, Dublin, Ireland.
| |
Collapse
|