1
|
Cabiati M, Vozzi F, Ceccherini E, Guiducci L, Persiani E, Gisone I, Sgalippa A, Cecchettini A, Del Ry S. Exploring Bone Morphogenetic Protein-2 and -4 mRNA Expression and Their Receptor Assessment in a Dynamic In Vitro Model of Vascular Calcification. Cells 2024; 13:2091. [PMID: 39768183 PMCID: PMC11674890 DOI: 10.3390/cells13242091] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2024] [Revised: 12/10/2024] [Accepted: 12/16/2024] [Indexed: 01/11/2025] Open
Abstract
BACKGROUND Vascular calcification (VC) is a dynamic, tightly regulated process driven by cellular activity and resembling the mechanisms of bone formation, with specific molecules playing pivotal roles in its progression. We aimed to investigate the involvement of the bone morphogenic proteins (BMP-2, BMP-4, BMPR-1a/1b, and BMPR-2) system in this process. Our study used an advanced in vitro model that simulates the biological environment of the vascular wall, assessing the ability of a phosphate mixture to induce the osteoblastic switch in human coronary artery smooth muscle cells (HCASMCs). METHODS HCASMCs were grown in mono- and co-culture with human coronary artery endothelial cells (HCAECs) in a double-flow bioreactor (LiveBox2 and IVTech), allowing static and dynamic conditions through a peristaltic pump. The VC was stimulated by incubation in a calcifying medium for 7 days. A BMP system Real-Time PCR was performed at the end of each experiment. RESULTS In monocultures, BMP-2 expression increased in calcified HCASMCs in static (p = 0.01) and dynamic conditions. BMP-4 and the biological receptors were expressed in all the experimental settings, increasing mainly in dynamic flow conditions. In co-cultures, we observed a marked increase in BMP-2 and BMP-4, BMPR-1a (p = 0.04 and p = 0.01, respectively), and BMPR-2 (p = 0.001) in the calcifying setting mostly in dynamic conditions. CONCLUSIONS The increase in BMP-2/4 in co-culture suggests that these genes might promote the switch towards an osteogenic-like phenotype, data also supported by the rise of both BMPR-1a and BMPR-2. Thus, our findings provide insights into the mechanisms by which dynamic co-culture modulates the BMP system activation in an environment mimicking in vivo VC's cellular and mechanical characteristics.
Collapse
Affiliation(s)
- Manuela Cabiati
- Institute of Clinical Physiology IFC-CNR, Via Giuseppe Moruzzi 1, 56124 Pisa, Italy; (F.V.); (E.C.); (L.G.); (E.P.); (I.G.); (S.D.R.)
| | - Federico Vozzi
- Institute of Clinical Physiology IFC-CNR, Via Giuseppe Moruzzi 1, 56124 Pisa, Italy; (F.V.); (E.C.); (L.G.); (E.P.); (I.G.); (S.D.R.)
| | - Elisa Ceccherini
- Institute of Clinical Physiology IFC-CNR, Via Giuseppe Moruzzi 1, 56124 Pisa, Italy; (F.V.); (E.C.); (L.G.); (E.P.); (I.G.); (S.D.R.)
| | - Letizia Guiducci
- Institute of Clinical Physiology IFC-CNR, Via Giuseppe Moruzzi 1, 56124 Pisa, Italy; (F.V.); (E.C.); (L.G.); (E.P.); (I.G.); (S.D.R.)
| | - Elisa Persiani
- Institute of Clinical Physiology IFC-CNR, Via Giuseppe Moruzzi 1, 56124 Pisa, Italy; (F.V.); (E.C.); (L.G.); (E.P.); (I.G.); (S.D.R.)
| | - Ilaria Gisone
- Institute of Clinical Physiology IFC-CNR, Via Giuseppe Moruzzi 1, 56124 Pisa, Italy; (F.V.); (E.C.); (L.G.); (E.P.); (I.G.); (S.D.R.)
| | - Agnese Sgalippa
- Health Science Interdisciplinary Center, Sant’Anna School of Advanced Studies, 56100 Pisa, Italy;
| | - Antonella Cecchettini
- Institute of Clinical Physiology IFC-CNR, Via Giuseppe Moruzzi 1, 56124 Pisa, Italy; (F.V.); (E.C.); (L.G.); (E.P.); (I.G.); (S.D.R.)
- Department of Clinical and Experimental Medicine, University of Pisa, 56100 Pisa, Italy
| | - Silvia Del Ry
- Institute of Clinical Physiology IFC-CNR, Via Giuseppe Moruzzi 1, 56124 Pisa, Italy; (F.V.); (E.C.); (L.G.); (E.P.); (I.G.); (S.D.R.)
- Department of Clinical and Experimental Medicine, University of Pisa, 56100 Pisa, Italy
| |
Collapse
|
2
|
Gadomski SJ, Mui BW, Gorodetsky R, Paravastu SS, Featherall J, Li L, Haffey A, Kim JC, Kuznetsov SA, Futrega K, Lazmi-Hailu A, Merling RK, Martin D, McCaskie AW, Robey PG. Time- and cell-specific activation of BMP signaling restrains chondrocyte hypertrophy. iScience 2024; 27:110537. [PMID: 39193188 PMCID: PMC11347861 DOI: 10.1016/j.isci.2024.110537] [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: 04/14/2023] [Revised: 02/29/2024] [Accepted: 07/15/2024] [Indexed: 08/29/2024] Open
Abstract
Stem cell therapies for degenerative cartilage disease are limited by an incomplete understanding of hyaline cartilage formation and maintenance. Human bone marrow stromal cells/skeletal stem cells (hBMSCs/SSCs) produce stable hyaline cartilage when attached to hyaluronic acid-coated fibrin microbeads (HyA-FMBs), yet the mechanism remains unclear. In vitro, hBMSC/SSC/HyA-FMB organoids exhibited reduced BMP signaling early in chondrogenic differentiation, followed by restoration of BMP signaling in chondrogenic IGFBP5 + /MGP + cells. Subsequently, human-induced pluripotent stem cell (hiPSC)-derived sclerotome cells were established (BMP inhibition) and then treated with transforming growth factor β (TGF-β) -/+ BMP2 and growth differentiation factor 5 (GDF5) (BMP signaling activation). TGF-β alone elicited a weak chondrogenic response, but TGF-β/BMP2/GDF5 led to delamination of SOX9 + aggregates (chondrospheroids) with high expression of COL2A1, ACAN, and PRG4 and minimal expression of COL10A1 and ALP in vitro. While transplanted hBMSCs/SSCs/HyA-FMBs did not heal articular cartilage defects in immunocompromised rodents, chondrospheroid-derived cells/HyA-FMBs formed non-hypertrophic cartilage that persisted until at least 5 months in vivo.
Collapse
Affiliation(s)
- Stephen J. Gadomski
- Skeletal Biology Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Department of Health and Human Services, Bethesda, MD 20892, USA
- NIH Oxford-Cambridge Scholars Program in Partnership with Medical University of South Carolina, Charleston, SC 29425, USA
- Wellcome-MRC Cambridge Stem Cell Institute, Cambridge CB2 0AW, UK
| | - Byron W.H. Mui
- Skeletal Biology Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Department of Health and Human Services, Bethesda, MD 20892, USA
- Wellcome-MRC Cambridge Stem Cell Institute, Cambridge CB2 0AW, UK
- NIH Oxford-Cambridge Scholars Program in Partnership with Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- NIH Medical Research Scholars Program, National Institutes of Health, Bethesda, MD 20892, USA
| | - Raphael Gorodetsky
- Lab of Biotechnology and Radiobiology, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | - Sriram S. Paravastu
- Skeletal Biology Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Department of Health and Human Services, Bethesda, MD 20892, USA
- NIH Medical Research Scholars Program, National Institutes of Health, Bethesda, MD 20892, USA
| | - Joseph Featherall
- Skeletal Biology Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Department of Health and Human Services, Bethesda, MD 20892, USA
- NIH Medical Research Scholars Program, National Institutes of Health, Bethesda, MD 20892, USA
| | - Li Li
- National Institute of Dental and Craniofacial Research Imaging Core, National Institutes of Health, Bethesda, MD 20892, USA
| | - Abigail Haffey
- Skeletal Biology Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Department of Health and Human Services, Bethesda, MD 20892, USA
- National Institute of Dental and Craniofacial Research Summer Internship Program, National Institutes of Health, Bethesda, MD 20892, USA
| | - Jae-Chun Kim
- Skeletal Biology Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Department of Health and Human Services, Bethesda, MD 20892, USA
- National Institute of Dental and Craniofacial Research Summer Dental Student Program, National Institutes of Health, Bethesda, MD 20892, USA
| | - Sergei A. Kuznetsov
- Skeletal Biology Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Department of Health and Human Services, Bethesda, MD 20892, USA
| | - Kathryn Futrega
- Skeletal Biology Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Department of Health and Human Services, Bethesda, MD 20892, USA
| | - Astar Lazmi-Hailu
- Lab of Biotechnology and Radiobiology, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | - Randall K. Merling
- Skeletal Biology Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Department of Health and Human Services, Bethesda, MD 20892, USA
| | - NIDCD/NIDCR Genomics and Computational Biology Core,
- Genomics and Computational Biology Core, National Institute on Deafness and Other Communication Disorders, 35A Convent Drive, Room 1F-103, Bethesda, MD 20892, USA
- Genomics and Computational Biology Core, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD 20892, USA
| | - Daniel Martin
- Genomics and Computational Biology Core, National Institute on Deafness and Other Communication Disorders, 35A Convent Drive, Room 1F-103, Bethesda, MD 20892, USA
- Genomics and Computational Biology Core, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD 20892, USA
| | - Andrew W. McCaskie
- Wellcome-MRC Cambridge Stem Cell Institute, Cambridge CB2 0AW, UK
- Department of Surgery, School of Clinical Medicine, University of Cambridge, Cambridge CB2 0QQ, UK
| | - Pamela G. Robey
- Skeletal Biology Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Department of Health and Human Services, Bethesda, MD 20892, USA
| |
Collapse
|
3
|
Chantiri M, Nammour S, El Toum S, Zeinoun T. Histological and Immunohistochemical Evaluation of Rh-BMP2: Effect on Gingival Healing Acceleration and Proliferation of Human Epithelial Cells. Life (Basel) 2024; 14:459. [PMID: 38672730 PMCID: PMC11051349 DOI: 10.3390/life14040459] [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: 02/01/2024] [Revised: 03/12/2024] [Accepted: 03/25/2024] [Indexed: 04/28/2024] Open
Abstract
This study aims to histologically and immunohistochemically evaluate the effect recombinant human bone morphogenetic protein (rh-BMP2) injected in gingival tissue has on the acceleration of the epithelial migration from the wound edges and epithelial cell proliferation after implant surgery. MATERIAL AND METHODS The study includes 20 patients who underwent bilateral implant surgeries in the premolar-molar region of the mandible, followed by guided bone regeneration. Each patient received an implant in both locations, but rh-BMP2 was only on the right side. At 9 days from the surgery, a gingival biopsy was performed 3 mm distally to the last implant. In total, 20 samples were collected from the left side (control group #1) and 20 from right (test group #1). This was repeated at a 4-month interval during healing abutment placements. Tissues were processed and stained with hematoxylin-eosin and then immunohistochemically for the expression of Ki-67 and further histological examination. RESULT Complete closure of the epithelium with new cell formation was observed in the 55% test group and 20% control group after 9 days. At 4 months, although 100% samples of all groups had complete epithelial closure, the test group showed that the epithelial cells were more organized and mature due to the increased number of blood vessels. The average number of new epithelial cells was 17.15 ± 7.545 and 16.12 ± 7.683 cells per mm in test group, respectively, at 9 days and 4 months and 10.99 ± 5.660 and 10.95 ± 5.768 in control groups. CONCLUSION Evident from histological observations, rh-BMP-2 can accelerate the closure of gingival wounds, the healing process of epithelial gingival tissue, and the formation of epithelial cells in patients undergoing dental implant treatment.
Collapse
Affiliation(s)
- Mansour Chantiri
- Department of Periodontology, Faculty of Dental Medicine, Lebanese University, Beirut 27798, Lebanon;
| | - Samir Nammour
- Department of Dental Sciences, Faculty of Medicine, University of Liege, 4000 Liege, Belgium;
| | - Sami El Toum
- Department of Oral Medicine and Maxillofacial Radiology, Faculty of Dental Medicine, Lebanese University, Beirut 27798, Lebanon;
| | - Toni Zeinoun
- Department of Oral and Maxillo-Facial Surgery, Faculty of Dental Medicine, Lebanese University, Beirut 27798, Lebanon
| |
Collapse
|
4
|
Zhang G, Chu M, Yang H, Li H, Shi J, Feng P, Wang S, Pan Z. Expression, Polymorphism, and Potential Functional Sites of the BMPR1A Gene in the Sheep Horn. Genes (Basel) 2024; 15:376. [PMID: 38540434 PMCID: PMC10970624 DOI: 10.3390/genes15030376] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2024] [Revised: 03/15/2024] [Accepted: 03/16/2024] [Indexed: 06/14/2024] Open
Abstract
Sheep horns are composed of bone and sheaths, and the BMPR1A gene is required for cartilage and osteogenic differentiation. Therefore, the BMPR1A gene may have a function related to the sheep horn, but its relationship with the sheep horn remains unclear. In this study, we first utilized RNA sequencing (RNA-seq) data to investigate the expression of the BMPR1A gene in different tissues and breeds of sheep. Second, whole-genome sequencing (WGS) data were used to explore the functional sites of the BMPR1A gene. Lastly, the allele-specific expression of the BMPR1A gene was explored. Our results indicate that BMPR1A gene expression is significantly higher in the normal horn groups than in the scurred groups. Importantly, this trend is consistent across several sheep breeds. Therefore, this finding suggests that the BMPR1A gene may be related to horn type. A total of 43 Single-Nucleotide Polymorphisms (SNPs) (F-statistics > 0.15) and 10 allele-specific expressions (ASEs) exhibited difference between the large and small horn populations. It is probable that these sites significantly impact the size of sheep horns. Compared to other polled species, we discovered ten amino acid sites that could influence horn presence. By combining RNA-seq and WGS functional loci results, we identified a functional site at position 40574836 on chromosome 25 that is both an SNP and exhibits allele-specific expression. In conclusion, we demonstrated that the BMPR1A gene is associated with horn type and identified some important functional sites which can be used as molecular markers in the breeding of sheep horns.
Collapse
Affiliation(s)
- Guoqing Zhang
- School of Chemistry and Chemical Engineering, University of Jinan, Jinan 250022, China;
- State Key Laboratory of Animal Biotech Breeding, Institute of Animal Science, Chinese Academy of Agricultural Sciences (CAAS), Beijing 100193, China; (M.C.); (H.Y.); (H.L.); (J.S.); (P.F.)
| | - Mingxing Chu
- State Key Laboratory of Animal Biotech Breeding, Institute of Animal Science, Chinese Academy of Agricultural Sciences (CAAS), Beijing 100193, China; (M.C.); (H.Y.); (H.L.); (J.S.); (P.F.)
| | - Hao Yang
- State Key Laboratory of Animal Biotech Breeding, Institute of Animal Science, Chinese Academy of Agricultural Sciences (CAAS), Beijing 100193, China; (M.C.); (H.Y.); (H.L.); (J.S.); (P.F.)
| | - Hao Li
- State Key Laboratory of Animal Biotech Breeding, Institute of Animal Science, Chinese Academy of Agricultural Sciences (CAAS), Beijing 100193, China; (M.C.); (H.Y.); (H.L.); (J.S.); (P.F.)
| | - Jianxin Shi
- State Key Laboratory of Animal Biotech Breeding, Institute of Animal Science, Chinese Academy of Agricultural Sciences (CAAS), Beijing 100193, China; (M.C.); (H.Y.); (H.L.); (J.S.); (P.F.)
| | - Pingjie Feng
- State Key Laboratory of Animal Biotech Breeding, Institute of Animal Science, Chinese Academy of Agricultural Sciences (CAAS), Beijing 100193, China; (M.C.); (H.Y.); (H.L.); (J.S.); (P.F.)
| | - Shoufeng Wang
- School of Chemistry and Chemical Engineering, University of Jinan, Jinan 250022, China;
| | - Zhangyuan Pan
- State Key Laboratory of Animal Biotech Breeding, Institute of Animal Science, Chinese Academy of Agricultural Sciences (CAAS), Beijing 100193, China; (M.C.); (H.Y.); (H.L.); (J.S.); (P.F.)
| |
Collapse
|
5
|
Sadowska JM, Power RN, Genoud KJ, Matheson A, González-Vázquez A, Costard L, Eichholz K, Pitacco P, Hallegouet T, Chen G, Curtin CM, Murphy CM, Cavanagh B, Zhang H, Kelly DJ, Boccaccini AR, O'Brien FJ. A Multifunctional Scaffold for Bone Infection Treatment by Delivery of microRNA Therapeutics Combined With Antimicrobial Nanoparticles. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2307639. [PMID: 38009631 DOI: 10.1002/adma.202307639] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Revised: 11/18/2023] [Indexed: 11/29/2023]
Abstract
Treating bone infections and ensuring bone repair is one of the greatest global challenges of modern orthopedics, made complex by antimicrobial resistance (AMR) risks due to long-term antibiotic treatment and debilitating large bone defects following infected tissue removal. An ideal multi-faceted solution would will eradicate bacterial infection without long-term antibiotic use, simultaneously stimulating osteogenesis and angiogenesis. Here, a multifunctional collagen-based scaffold that addresses these needs by leveraging the potential of antibiotic-free antimicrobial nanoparticles (copper-doped bioactive glass, CuBG) to combat infection without contributing to AMR in conjunction with microRNA-based gene therapy (utilizing an inhibitor of microRNA-138) to stimulate both osteogenesis and angiogenesis, is developed. CuBG scaffolds reduce the attachment of gram-positive bacteria by over 80%, showcasing antimicrobial functionality. The antagomiR-138 nanoparticles induce osteogenesis of human mesenchymal stem cells in vitro and heal a large load-bearing defect in a rat femur when delivered on the scaffold. Combining both promising technologies results in a multifunctional antagomiR-138-activated CuBG scaffold inducing hMSC-mediated osteogenesis and stimulating vasculogenesis in an in vivo chick chorioallantoic membrane model. Overall, this multifunctional scaffold catalyzes killing mechanisms in bacteria while inducing bone repair through osteogenic and angiogenic coupling, making this platform a promising multi-functional strategy for treating and repairing complex bone infections.
Collapse
Affiliation(s)
- Joanna M Sadowska
- Tissue Engineering Research Group, Dept. of Anatomy and Regenerative Medicine, Royal College of Surgeons in Ireland (RCSI) University of Medicine and Health Sciences, Dublin, D02 YN77, Ireland
| | - Rachael N Power
- Tissue Engineering Research Group, Dept. of Anatomy and Regenerative Medicine, Royal College of Surgeons in Ireland (RCSI) University of Medicine and Health Sciences, Dublin, D02 YN77, Ireland
| | - Katelyn J Genoud
- Tissue Engineering Research Group, Dept. of Anatomy and Regenerative Medicine, Royal College of Surgeons in Ireland (RCSI) University of Medicine and Health Sciences, Dublin, D02 YN77, Ireland
- Advanced Materials and Bioengineering Research Centre (AMBER), Royal College of Surgeons in Ireland (RCSI) University of Medicine and Health Sciences and Trinity College Dublin (TCD), Dublin, D02 W085, Ireland
| | - Austyn Matheson
- Tissue Engineering Research Group, Dept. of Anatomy and Regenerative Medicine, Royal College of Surgeons in Ireland (RCSI) University of Medicine and Health Sciences, Dublin, D02 YN77, Ireland
- Advanced Materials and Bioengineering Research Centre (AMBER), Royal College of Surgeons in Ireland (RCSI) University of Medicine and Health Sciences and Trinity College Dublin (TCD), Dublin, D02 W085, Ireland
| | - Arlyng González-Vázquez
- Tissue Engineering Research Group, Dept. of Anatomy and Regenerative Medicine, Royal College of Surgeons in Ireland (RCSI) University of Medicine and Health Sciences, Dublin, D02 YN77, Ireland
| | - Lara Costard
- Tissue Engineering Research Group, Dept. of Anatomy and Regenerative Medicine, Royal College of Surgeons in Ireland (RCSI) University of Medicine and Health Sciences, Dublin, D02 YN77, Ireland
| | - Kian Eichholz
- Advanced Materials and Bioengineering Research Centre (AMBER), Royal College of Surgeons in Ireland (RCSI) University of Medicine and Health Sciences and Trinity College Dublin (TCD), Dublin, D02 W085, Ireland
- Trinity Centre for Biomedical Engineering, Trinity College Dublin (TCD), Dublin, D02 R590, Ireland
| | - Pierluca Pitacco
- Advanced Materials and Bioengineering Research Centre (AMBER), Royal College of Surgeons in Ireland (RCSI) University of Medicine and Health Sciences and Trinity College Dublin (TCD), Dublin, D02 W085, Ireland
- Trinity Centre for Biomedical Engineering, Trinity College Dublin (TCD), Dublin, D02 R590, Ireland
| | - Tanguy Hallegouet
- Tissue Engineering Research Group, Dept. of Anatomy and Regenerative Medicine, Royal College of Surgeons in Ireland (RCSI) University of Medicine and Health Sciences, Dublin, D02 YN77, Ireland
- University of Strasbourg, Strasbourg, 67412, France
| | - Gang Chen
- Microsurgical Research and Training Facility (MRTF), Royal College of Surgeons in Ireland (RCSI) University of Medicine and Health Sciences, Dublin, D02 YN77, Ireland
| | - Caroline M Curtin
- Tissue Engineering Research Group, Dept. of Anatomy and Regenerative Medicine, Royal College of Surgeons in Ireland (RCSI) University of Medicine and Health Sciences, Dublin, D02 YN77, Ireland
- Advanced Materials and Bioengineering Research Centre (AMBER), Royal College of Surgeons in Ireland (RCSI) University of Medicine and Health Sciences and Trinity College Dublin (TCD), Dublin, D02 W085, Ireland
- Trinity Centre for Biomedical Engineering, Trinity College Dublin (TCD), Dublin, D02 R590, Ireland
| | - Ciara M Murphy
- Tissue Engineering Research Group, Dept. of Anatomy and Regenerative Medicine, Royal College of Surgeons in Ireland (RCSI) University of Medicine and Health Sciences, Dublin, D02 YN77, Ireland
- Advanced Materials and Bioengineering Research Centre (AMBER), Royal College of Surgeons in Ireland (RCSI) University of Medicine and Health Sciences and Trinity College Dublin (TCD), Dublin, D02 W085, Ireland
- Trinity Centre for Biomedical Engineering, Trinity College Dublin (TCD), Dublin, D02 R590, Ireland
| | - Brenton Cavanagh
- Cellular and Molecular Imaging Core, Royal College of Surgeons in Ireland (RCSI) University of Medicine and Health Sciences, Dublin, D02 YN77, Ireland
| | - Huijun Zhang
- Institute of Biomaterials, Friedrich-Alexander University Erlangen-Nuremberg, 91056, Erlangen, Germany
| | - Daniel J Kelly
- Tissue Engineering Research Group, Dept. of Anatomy and Regenerative Medicine, Royal College of Surgeons in Ireland (RCSI) University of Medicine and Health Sciences, Dublin, D02 YN77, Ireland
- Advanced Materials and Bioengineering Research Centre (AMBER), Royal College of Surgeons in Ireland (RCSI) University of Medicine and Health Sciences and Trinity College Dublin (TCD), Dublin, D02 W085, Ireland
- Trinity Centre for Biomedical Engineering, Trinity College Dublin (TCD), Dublin, D02 R590, Ireland
| | - Aldo R Boccaccini
- Institute of Biomaterials, Friedrich-Alexander University Erlangen-Nuremberg, 91056, Erlangen, Germany
| | - Fergal J O'Brien
- Tissue Engineering Research Group, Dept. of Anatomy and Regenerative Medicine, Royal College of Surgeons in Ireland (RCSI) University of Medicine and Health Sciences, Dublin, D02 YN77, Ireland
- Advanced Materials and Bioengineering Research Centre (AMBER), Royal College of Surgeons in Ireland (RCSI) University of Medicine and Health Sciences and Trinity College Dublin (TCD), Dublin, D02 W085, Ireland
- Trinity Centre for Biomedical Engineering, Trinity College Dublin (TCD), Dublin, D02 R590, Ireland
| |
Collapse
|
6
|
Wu M, Wu S, Chen W, Li YP. The roles and regulatory mechanisms of TGF-β and BMP signaling in bone and cartilage development, homeostasis and disease. Cell Res 2024; 34:101-123. [PMID: 38267638 PMCID: PMC10837209 DOI: 10.1038/s41422-023-00918-9] [Citation(s) in RCA: 98] [Impact Index Per Article: 98.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2023] [Accepted: 12/15/2023] [Indexed: 01/26/2024] Open
Abstract
Transforming growth factor-βs (TGF-βs) and bone morphometric proteins (BMPs) belong to the TGF-β superfamily and perform essential functions during osteoblast and chondrocyte lineage commitment and differentiation, skeletal development, and homeostasis. TGF-βs and BMPs transduce signals through SMAD-dependent and -independent pathways; specifically, they recruit different receptor heterotetramers and R-Smad complexes, resulting in unique biological readouts. BMPs promote osteogenesis, osteoclastogenesis, and chondrogenesis at all differentiation stages, while TGF-βs play different roles in a stage-dependent manner. BMPs and TGF-β have opposite functions in articular cartilage homeostasis. Moreover, TGF-β has a specific role in maintaining the osteocyte network. The precise activation of BMP and TGF-β signaling requires regulatory machinery at multiple levels, including latency control in the matrix, extracellular antagonists, ubiquitination and phosphorylation in the cytoplasm, nucleus-cytoplasm transportation, and transcriptional co-regulation in the nuclei. This review weaves the background information with the latest advances in the signaling facilitated by TGF-βs and BMPs, and the advanced understanding of their diverse physiological functions and regulations. This review also summarizes the human diseases and mouse models associated with disordered TGF-β and BMP signaling. A more precise understanding of the BMP and TGF-β signaling could facilitate the development of bona fide clinical applications in treating bone and cartilage disorders.
Collapse
Affiliation(s)
- Mengrui Wu
- Department of Cell and Developmental Biology, College of Life Sciences, Zhejiang University, Hangzhou, Zhejiang, China.
| | - Shali Wu
- Department of Cell and Developmental Biology, College of Life Sciences, Zhejiang University, Hangzhou, Zhejiang, China
| | - Wei Chen
- Division in Cellular and Molecular Medicine, Department of Pathology and Laboratory Medicine, Tulane University School of Medicine, Tulane University, New Orleans, LA, USA
| | - Yi-Ping Li
- Division in Cellular and Molecular Medicine, Department of Pathology and Laboratory Medicine, Tulane University School of Medicine, Tulane University, New Orleans, LA, USA.
| |
Collapse
|
7
|
Miguez PA, de Paiva Gonçalves V, Musskopf ML, Rivera-Concepcion A, McGaughey S, Yu C, Lee DJ, Tuin SA, Ali A. Mitigation of BMP-induced inflammation in craniofacial bone regeneration and improvement of bone parameters by dietary hesperidin. Sci Rep 2024; 14:2602. [PMID: 38297106 PMCID: PMC10830467 DOI: 10.1038/s41598-024-52566-7] [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: 09/13/2023] [Accepted: 01/20/2024] [Indexed: 02/02/2024] Open
Abstract
Based on anti-inflammatory and osteogenic properties of hesperidin (HE), we hypothesized its systemic administration could be a cost-effective method of improving BMP-induced bone regeneration. Sprague-Dawley rats were allocated into 4 groups (n = 10/group): a 5-mm critical-sized mandible defect + collagen scaffold or, scaffold + 1 µg of BMP2 with and without dietary HE at 100 mg/kg. HE was administered by oral gavage 4 weeks prior to surgeries until euthanasia at day 7 or 14 post-surgery. The healing tissue within the defect collected at day 7 was subjected to gene expression analysis. Mandibles harvested at day 14 were subjected to microcomputed tomography and histology. HE + BMP2-treated rats had a statistically significant decrease in expression of inflammatory genes compared to BMP2 alone. The high-dose BMP2 alone caused cystic-like regeneration with incomplete defect closure. HE + BMP2 showed virtually complete bone fusion. Collagen fibril birefringence pattern (red color) under polarized light indicated high organization in BMP2-induced newly formed bone (NFB) in HE-supplemented group (p < 0.05). Clear changes in osteocyte lacunae as well as a statistically significant increase in osteoclasts were found around NFB in HE-treated rats. A significant increase in trabecular volume and thickness, and trabecular and cortical density was found in femurs of HE-supplemented rats (p < 0.05). Our findings show, for the first time, that dietary HE has a remarkable modulatory role in the function of locally delivered high-dose BMP2 in bone regeneration possibly via control of inflammation, osteogenesis, changes in osteocyte and osteoclast function and collagen maturation in regenerated and native bone. In conclusion, HE had a significant skeletal bone sparing effect and the ability to provide a more effective BMP-induced craniofacial regeneration.
Collapse
Affiliation(s)
- Patricia A Miguez
- Division of Comprehensive Oral Health - Periodontology, Adams School of Dentistry, University of North Carolina at Chapel Hill, CB# 7455, Rm 4610, Koury Oral Health Sciences, 385 S. Columbia St., Chapel Hill, NC, 27599-7455, USA.
| | - Vinícius de Paiva Gonçalves
- Oral and Craniofacial Health Sciences, Adams School of Dentistry, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Marta L Musskopf
- Division of Comprehensive Oral Health - Periodontology, Adams School of Dentistry, University of North Carolina at Chapel Hill, CB# 7455, Rm 4610, Koury Oral Health Sciences, 385 S. Columbia St., Chapel Hill, NC, 27599-7455, USA
| | | | - Skylar McGaughey
- Adams School of Dentistry, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Christina Yu
- Adams School of Dentistry, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Dong Joon Lee
- Oral and Craniofacial Health Sciences, Adams School of Dentistry, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Stephen A Tuin
- Oral and Craniofacial Health Sciences, Adams School of Dentistry, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Aya Ali
- Division of Comprehensive Oral Health - Periodontology, Adams School of Dentistry, University of North Carolina at Chapel Hill, CB# 7455, Rm 4610, Koury Oral Health Sciences, 385 S. Columbia St., Chapel Hill, NC, 27599-7455, USA
| |
Collapse
|
8
|
Kuchynsky K, Stevens P, Hite A, Xie W, Diop K, Tang S, Pietrzak M, Khan S, Walter B, Purmessur D. Transcriptional profiling of human cartilage endplate cells identifies novel genes and cell clusters underlying degenerated and non-degenerated phenotypes. Arthritis Res Ther 2024; 26:12. [PMID: 38173036 PMCID: PMC10763221 DOI: 10.1186/s13075-023-03220-6] [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: 08/30/2023] [Accepted: 11/22/2023] [Indexed: 01/05/2024] Open
Abstract
BACKGROUND Low back pain is a leading cause of disability worldwide and is frequently attributed to intervertebral disc (IVD) degeneration. Though the contributions of the adjacent cartilage endplates (CEP) to IVD degeneration are well documented, the phenotype and functions of the resident CEP cells are critically understudied. To better characterize CEP cell phenotype and possible mechanisms of CEP degeneration, bulk and single-cell RNA sequencing of non-degenerated and degenerated CEP cells were performed. METHODS Human lumbar CEP cells from degenerated (Thompson grade ≥ 4) and non-degenerated (Thompson grade ≤ 2) discs were expanded for bulk (N=4 non-degenerated, N=4 degenerated) and single-cell (N=1 non-degenerated, N=1 degenerated) RNA sequencing. Genes identified from bulk RNA sequencing were categorized by function and their expression in non-degenerated and degenerated CEP cells were compared. A PubMed literature review was also performed to determine which genes were previously identified and studied in the CEP, IVD, and other cartilaginous tissues. For single-cell RNA sequencing, different cell clusters were resolved using unsupervised clustering and functional annotation. Differential gene expression analysis and Gene Ontology, respectively, were used to compare gene expression and functional enrichment between cell clusters, as well as between non-degenerated and degenerated CEP samples. RESULTS Bulk RNA sequencing revealed 38 genes were significantly upregulated and 15 genes were significantly downregulated in degenerated CEP cells relative to non-degenerated cells (|fold change| ≥ 1.5). Of these, only 2 genes were previously studied in CEP cells, and 31 were previously studied in the IVD and other cartilaginous tissues. Single-cell RNA sequencing revealed 11 unique cell clusters, including multiple chondrocyte and progenitor subpopulations with distinct gene expression and functional profiles. Analysis of genes in the bulk RNA sequencing dataset showed that progenitor cell clusters from both samples were enriched in "non-degenerated" genes but not "degenerated" genes. For both bulk- and single-cell analyses, gene expression and pathway enrichment analyses highlighted several pathways that may regulate CEP degeneration, including transcriptional regulation, translational regulation, intracellular transport, and mitochondrial dysfunction. CONCLUSIONS This thorough analysis using RNA sequencing methods highlighted numerous differences between non-degenerated and degenerated CEP cells, the phenotypic heterogeneity of CEP cells, and several pathways of interest that may be relevant in CEP degeneration.
Collapse
Affiliation(s)
- Kyle Kuchynsky
- Department of Biomedical Engineering, The Ohio State University, 3016 Fontana Laboratories, 140 W. 19th Ave, Columbus, OH, 43210, USA
| | - Patrick Stevens
- Department of Biomedical Informatics, The Ohio State University, Columbus, OH, USA
| | - Amy Hite
- Department of Biomedical Informatics, The Ohio State University, Columbus, OH, USA
| | - William Xie
- Department of Chemistry and Biochemistry, The Ohio State University, Columbus, OH, USA
| | - Khady Diop
- Department of Biomedical Engineering, The Ohio State University, 3016 Fontana Laboratories, 140 W. 19th Ave, Columbus, OH, 43210, USA
| | - Shirley Tang
- Department of Biomedical Engineering, The Ohio State University, 3016 Fontana Laboratories, 140 W. 19th Ave, Columbus, OH, 43210, USA
| | - Maciej Pietrzak
- Department of Biomedical Informatics, The Ohio State University, Columbus, OH, USA
- The James Comprehensive Cancer Center, The Ohio State University, Columbus, OH, USA
| | - Safdar Khan
- Department of Orthopaedics, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Benjamin Walter
- Department of Biomedical Engineering, The Ohio State University, 3016 Fontana Laboratories, 140 W. 19th Ave, Columbus, OH, 43210, USA
| | - Devina Purmessur
- Department of Biomedical Engineering, The Ohio State University, 3016 Fontana Laboratories, 140 W. 19th Ave, Columbus, OH, 43210, USA.
- Department of Orthopaedics, The Ohio State University Wexner Medical Center, Columbus, OH, USA.
| |
Collapse
|
9
|
Tölle J, Koch A, Schlicht K, Finger D, Kaehler W, Höppner M, Graetz C, Dörfer C, Schulte DM, Fawzy El-Sayed K. Effect of Hyperbaric Oxygen and Inflammation on Human Gingival Mesenchymal Stem/Progenitor Cells. Cells 2023; 12:2479. [PMID: 37887323 PMCID: PMC10605813 DOI: 10.3390/cells12202479] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2023] [Revised: 10/06/2023] [Accepted: 10/11/2023] [Indexed: 10/28/2023] Open
Abstract
The present study explores for the first time the effect of hyperbaric oxygen (HBO) on gingival mesenchymal stem cells' (G-MSCs) gene expression profile, intracellular pathway activation, pluripotency, and differentiation potential under an experimental inflammatory setup. G-MSCs were isolated from five healthy individuals (n = 5) and characterized. Single (24 h) or double (72 h) HBO stimulation (100% O2, 3 bar, 90 min) was performed under experimental inflammatory [IL-1β (1 ng/mL)/TNF-α (10 ng/mL)/IFN-γ (100 ng/mL)] and non-inflammatory micro-environment. Next Generation Sequencing and KEGG pathway enrichment analysis, G-MSCs' pluripotency gene expression, Wnt-/β-catenin pathway activation, proliferation, colony formation, and differentiation were investigated. G-MSCs demonstrated all mesenchymal stem/progenitor cells' characteristics. The beneficial effect of a single HBO stimulation was evident, with anti-inflammatory effects and induction of differentiation (TLL1, ID3, BHLHE40), proliferation/cell survival (BMF, ID3, TXNIP, PDK4, ABL2), migration (ABL2) and osteogenic differentiation (p < 0.05). A second HBO stimulation at 72 h had a detrimental effect, significantly increasing the inflammation-induced cellular stress and ROS accumulation through HMOX1, BHLHE40, and ARL4C amplification and pathway enrichment (p < 0.05). Results outline a positive short-term single HBO anti-inflammatory, regenerative, and differentiation stimulatory effect on G-MSCs. A second (72 h) stimulation is detrimental to the same properties. The current results could open new perspectives in the clinical application of short-termed HBO induction in G-MSCs-mediated periodontal reparative/regenerative mechanisms.
Collapse
Affiliation(s)
- Johannes Tölle
- Clinic for Conservative Dentistry and Periodontology, School of Dental Medicine, Christian-Albrechts-University, 24105 Kiel, Germany; (J.T.); (D.F.); (C.G.); (C.D.)
| | - Andreas Koch
- German Naval Medical Institute, 24119 Kiel, Germany; (A.K.); (W.K.)
| | - Kristina Schlicht
- Institute of Diabetes and Clinical Metabolic Research, University Hospital Schleswig-Holstein, 24105 Kiel, Germany; (K.S.); (D.M.S.)
| | - Dirk Finger
- Clinic for Conservative Dentistry and Periodontology, School of Dental Medicine, Christian-Albrechts-University, 24105 Kiel, Germany; (J.T.); (D.F.); (C.G.); (C.D.)
| | - Wataru Kaehler
- German Naval Medical Institute, 24119 Kiel, Germany; (A.K.); (W.K.)
| | - Marc Höppner
- Institute of Clinical Molecular Biology, School of Medicine, Christian-Albrechts-University, 24105 Kiel, Germany;
| | - Christian Graetz
- Clinic for Conservative Dentistry and Periodontology, School of Dental Medicine, Christian-Albrechts-University, 24105 Kiel, Germany; (J.T.); (D.F.); (C.G.); (C.D.)
| | - Christof Dörfer
- Clinic for Conservative Dentistry and Periodontology, School of Dental Medicine, Christian-Albrechts-University, 24105 Kiel, Germany; (J.T.); (D.F.); (C.G.); (C.D.)
| | - Dominik M. Schulte
- Institute of Diabetes and Clinical Metabolic Research, University Hospital Schleswig-Holstein, 24105 Kiel, Germany; (K.S.); (D.M.S.)
- Division of Endocrinology, Diabetes and Clinical Nutrition, Department of Internal Medicine I, University Hospital Schleswig-Holstein, 24105 Kiel, Germany
| | - Karim Fawzy El-Sayed
- Clinic for Conservative Dentistry and Periodontology, School of Dental Medicine, Christian-Albrechts-University, 24105 Kiel, Germany; (J.T.); (D.F.); (C.G.); (C.D.)
- Oral Medicine and Periodontology Department, Faculty of Dentistry, Cairo University, Cairo 12613, Egypt
| |
Collapse
|
10
|
Guo R, Gu T, Xiao Y, Xiao T, Liu Q, Li Z, Yu J. Hsa-miR-27b-5p suppresses the osteogenic and odontogenic differentiation of stem cells from human exfoliated deciduous teeth via targeting BMPR1A: An ex vivo study. Int Endod J 2023; 56:1284-1300. [PMID: 37485765 DOI: 10.1111/iej.13959] [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: 02/10/2023] [Revised: 07/18/2023] [Accepted: 07/19/2023] [Indexed: 07/25/2023]
Abstract
AIM Recently, miR-27b-5p was shown to be abundantly expressed in extracellular vehicles (EVs) from the inflammatory microenvironment. This study determined the role of miR-27b-5p in regulating osteogenic and odontogenic differentiation of stem cells from human exfoliated deciduous teeth (SHEDs) and further examined the regulatory mechanism of bone morphogenetic protein receptor type-1A (BMPR1A). METHODOLOGY Characteristics of SHEDs and SHEDs-EVs derived from SHEDs were evaluated respectively. The expression of miR-27b-5p in SHEDs and EVs was detected during osteo-induction. Mechanically, SHEDs were treated with miR-27b-5p mimics or an inhibitor, and the osteogenic/odontogenic differentiation and proliferation were assessed. Bioinformatic analysis and luciferase reporter were utilized for target gene prediction and verification. Finally, BMPR1A-overexpressed plasmids were transfected into SHEDs to investigate the participation of the BMPR1A/SMAD4 pathway. Data were analysed using Student's t-test, one-way analysis of variance and Chi-square test. RESULTS MiR-27b-5p was expressed in both SHEDs and EVs and was significantly increased at the initial stage of differentiation and then decreased in a time-dependent manner (p < .01). Upregulation of miR-27b-5p significantly suppressed osteogenic/odontogenic differentiation of SHEDs and inhibited proliferation (p < .05), whereas inhibition of miR-27b-5p enhanced the differentiation (p < .05). Dual-luciferase reporter assay and pull-down assay confirmed the binding site between miR-27b-5p and BMPR1A (p < .05). The overexpression of BMPR1A rescued the effect of miR-27b-5p, while contributed to the decrease of pluripotency (p < .05). Additionally, miR-27b-5p maintained pluripotency in BMPR1A-overexpressed SHEDs (p < .05). CONCLUSIONS MiR-27b-5p in SHEDs/EVs was inversely associated with differentiation and suppressed the osteogenic and odontogenic differentiation of SHEDs and maintained the pluripotency of SHEDs partly by shuttering BMPR1A-targeting BMP signalling. Theoretically, inhibition of miR-27b-5p represents a potential strategy to promote osteanagenesis and dentinogenesis. However, miR-27b-5p capsuled EVs might maintain cell pluripotency and self-renewal for non-cell-targeted therapy.
Collapse
Affiliation(s)
- Rong Guo
- Department of Endodontics, The Affiliated Stomatological Hospital of Nanjing Medical University, Nanjing, China
- Jiangsu Province Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing, China
- Jiangsu Province Engineering Research Center of Stomatological Translational Medicine, Nanjing Medical University, Nanjing, China
| | - Tingjie Gu
- Department of Endodontics, The Affiliated Stomatological Hospital of Nanjing Medical University, Nanjing, China
- Jiangsu Province Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing, China
- Jiangsu Province Engineering Research Center of Stomatological Translational Medicine, Nanjing Medical University, Nanjing, China
| | - Ya Xiao
- Department of Endodontics, The Affiliated Stomatological Hospital of Nanjing Medical University, Nanjing, China
- Jiangsu Province Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing, China
- Jiangsu Province Engineering Research Center of Stomatological Translational Medicine, Nanjing Medical University, Nanjing, China
| | - Tong Xiao
- Department of Endodontics, The Affiliated Stomatological Hospital of Nanjing Medical University, Nanjing, China
- Jiangsu Province Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing, China
- Jiangsu Province Engineering Research Center of Stomatological Translational Medicine, Nanjing Medical University, Nanjing, China
| | - Qian Liu
- Jiangsu Province Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing, China
- Jiangsu Province Engineering Research Center of Stomatological Translational Medicine, Nanjing Medical University, Nanjing, China
- Department of Paediatric Dentistry, The Affiliated Stomatological Hospital of Nanjing Medical University, Nanjing, China
| | - Zehan Li
- Department of Endodontics, The Affiliated Stomatological Hospital of Nanjing Medical University, Nanjing, China
- Jiangsu Province Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing, China
- Jiangsu Province Engineering Research Center of Stomatological Translational Medicine, Nanjing Medical University, Nanjing, China
| | - Jinhua Yu
- Department of Endodontics, The Affiliated Stomatological Hospital of Nanjing Medical University, Nanjing, China
- Jiangsu Province Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing, China
- Jiangsu Province Engineering Research Center of Stomatological Translational Medicine, Nanjing Medical University, Nanjing, China
| |
Collapse
|
11
|
Ruan X, Gu J, Chen M, Zhao F, Aili M, Zhang D. Multiple roles of ALK3 in osteoarthritis. Bone Joint Res 2023; 12:397-411. [PMID: 37394235 PMCID: PMC10315222 DOI: 10.1302/2046-3758.127.bjr-2022-0310.r1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 07/04/2023] Open
Abstract
Osteoarthritis (OA) is a chronic degenerative joint disease characterized by progressive cartilage degradation, synovial membrane inflammation, osteophyte formation, and subchondral bone sclerosis. Pathological changes in cartilage and subchondral bone are the main processes in OA. In recent decades, many studies have demonstrated that activin-like kinase 3 (ALK3), a bone morphogenetic protein receptor, is essential for cartilage formation, osteogenesis, and postnatal skeletal development. Although the role of bone morphogenetic protein (BMP) signalling in articular cartilage and bone has been extensively studied, many new discoveries have been made in recent years around ALK3 targets in articular cartilage, subchondral bone, and the interaction between the two, broadening the original knowledge of the relationship between ALK3 and OA. In this review, we focus on the roles of ALK3 in OA, including cartilage and subchondral bone and related cells. It may be helpful to seek more efficient drugs or treatments for OA based on ALK3 signalling in future.
Collapse
Affiliation(s)
- Xianchun Ruan
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Jinning Gu
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
- Department of Orthodontics, Peking University School and Hospital of Stomatology, Beijing, China
| | - Mingyang Chen
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Fulin Zhao
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Munire Aili
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Demao Zhang
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
- Institute of Biomedical Engineering, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, China
| |
Collapse
|
12
|
Ruan X, Zhang Z, Aili M, Luo X, Wei Q, Zhang D, Bai M. Activin receptor-like kinase 3: a critical modulator of development and function of mineralized tissues. Front Cell Dev Biol 2023; 11:1209817. [PMID: 37457289 PMCID: PMC10347416 DOI: 10.3389/fcell.2023.1209817] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2023] [Accepted: 06/22/2023] [Indexed: 07/18/2023] Open
Abstract
Mineralized tissues, such as teeth and bones, pose significant challenges for repair due to their hardness, low permeability, and limited blood flow compared to soft tissues. Bone morphogenetic proteins (BMPs) have been identified as playing a crucial role in mineralized tissue formation and repair. However, the application of large amounts of exogenous BMPs may cause side effects such as inflammation. Therefore, it is necessary to identify a more precise molecular target downstream of the ligands. Activin receptor-like kinase 3 (ALK3), a key transmembrane receptor, serves as a vital gateway for the transmission of BMP signals, triggering cellular responses. Recent research has yielded new insights into the regulatory roles of ALK3 in mineralized tissues. Experimental knockout or mutation of ALK3 has been shown to result in skeletal dysmorphisms and failure of tooth formation, eruption, and orthodontic tooth movement. This review summarizes the roles of ALK3 in mineralized tissue regulation and elucidates how ALK3-mediated signaling influences the physiology and pathology of teeth and bones. Additionally, this review provides a reference for recommended basic research and potential future treatment strategies for the repair and regeneration of mineralized tissues.
Collapse
Affiliation(s)
- Xianchun Ruan
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
- National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Zhaowei Zhang
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
- National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Munire Aili
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
- National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Xiang Luo
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
- National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
- West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu, China
| | - Qiang Wei
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
- National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Demao Zhang
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
- National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
- West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu, China
| | - Mingru Bai
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
- National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
- Department of Cariology and Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| |
Collapse
|
13
|
Gan J, Deng X, Le Y, Lai J, Liao X. The Development of Naringin for Use against Bone and Cartilage Disorders. Molecules 2023; 28:3716. [PMID: 37175126 PMCID: PMC10180405 DOI: 10.3390/molecules28093716] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2023] [Revised: 04/17/2023] [Accepted: 04/24/2023] [Indexed: 05/15/2023] Open
Abstract
Bone and cartilage disorders are the leading causes of musculoskeletal disability. There is no absolute cure for all bone and cartilage disorders. The exploration of natural compounds for the potential therapeutic use against bone and cartilage disorders is proving promising. Among these natural chemicals, naringin, a flavanone glycoside, is a potential candidate due to its multifaceted pharmacological activities in bone and cartilage tissues. Emerging studies indicate that naringin may promote osteogenic differentiation, inhibit osteoclast formation, and exhibit protective effects against osteoporosis in vivo and in vitro. Many signaling pathways, such as BMP-2, Wnt/β-catenin, and VEGF/VEGFR, participate in the biological actions of naringin in mediating the pathological development of osteoporosis. In addition, the anti-inflammatory, anti-oxidative stress, and anti-apoptosis abilities of naringin also demonstrate its beneficial effects against bone and cartilage disorders, including intervertebral disc degeneration, osteoarthritis, rheumatoid arthritis, bone and cartilage tumors, and tibial dyschondroplasia. Naringin exhibits protective effects against bone and cartilage disorders. However, more efforts are still needed due to, at least in part, the uncertainty of drug targets. Further biological and pharmacological evaluations of naringin and its applications in bone tissue engineering, particularly its therapeutic effects against osteoporosis, might result in developing potential drug candidates.
Collapse
Affiliation(s)
- Juwen Gan
- Department of Pulmonary and Critical Care Medicine, Ganzhou People’s Hospital, Ganzhou 341000, China
| | - Xiaolan Deng
- Department of Pharmacy, Haikou Affiliated Hospital, Xiangya School of Medicine, Central South University, Haikou 570208, China
| | - Yonghong Le
- Department of Pulmonary and Critical Care Medicine, Ganzhou People’s Hospital, Ganzhou 341000, China
| | - Jun Lai
- Department of Pharmacy, Ganzhou People’s Hospital, Ganzhou 341000, China
| | - Xiaofei Liao
- Department of Pharmacy, Ganzhou People’s Hospital, Ganzhou 341000, China
| |
Collapse
|
14
|
Gigout A, Werkmann D, Menges S, Brenneis C, Henson F, Cowan KJ, Musil D, Thudium CS, Gühring H, Michaelis M, Kleinschmidt-Doerr K. R399E, A Mutated Form of Growth and Differentiation Factor 5, for Disease Modification of Osteoarthritis. Arthritis Rheumatol 2023; 75:375-386. [PMID: 36054172 DOI: 10.1002/art.42343] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Revised: 07/05/2022] [Accepted: 08/31/2022] [Indexed: 11/10/2022]
Abstract
OBJECTIVE To preclinically characterize a mutant form of growth and differentiation factor 5, R399E, with reduced osteogenic properties as a potential disease-modifying osteoarthritis (OA) drug. METHODS Cartilage, synovium, and meniscus samples from patients with OA were used to evaluate anabolic and antiinflammatory properties of R399E. In the rabbit joint instability model, 65 rabbits underwent transection of the anterior cruciate ligament plus partial meniscectomy. Three intraarticular (IA) R399E doses were administered biweekly 6 times, and static incapacitance was determined to assess joint pain. OA was evaluated 13 weeks after surgery. In sheep, medial meniscus transection was performed to induce OA, dynamic weight bearing was measured in-life, and OA was assessed after 13 weeks. RESULTS Intermittent exposure to R399E (1 week per month) was sufficient to induce cell proliferation and release of anabolic markers in 3-dimensional chondrocyte cultures. R399E also inhibited the release of interleukin-1β (IL-1β), IL-6, and prostaglandin E2 from cartilage with synovium, meniscal cell, and synoviocyte cultures. In rabbits, the mean difference (95% confidence interval [95% CI]) in weight bearing for R399E compared to vehicle was -5.8 (95% confidence interval [95% CI] -9.54, -2.15), -7.2 (95% CI -10.93, -3.54), and -7.7 (95% CI -11.49, -3.84) for the 0.6, 6, and 60 μg doses, respectively, 6 hours after the first IA injection, and was statistically significant through the entire study for all doses. Cartilage surface structure improved with the 6-μg dose. Structural and symptomatic improvement with the same dose was confirmed in the sheep model of OA. CONCLUSION R399E influences several pathologic processes contributing to OA, highlighting its potential as a disease-modifying therapy.
Collapse
Affiliation(s)
- Anne Gigout
- Merck Healthcare KGaA 64293, Darmstadt, Germany
| | | | | | | | - Frances Henson
- Division of Trauma and Orthopaedic Surgery, University of Cambridge, Cambridge, UK
| | | | | | | | | | | | | |
Collapse
|
15
|
Tian S, Li W, Zhong Z, Wang F, Xiao Q. Genome-wide re-sequencing data reveals the genetic diversity and population structure of Wenchang chicken in China. Anim Genet 2023; 54:328-337. [PMID: 36639920 DOI: 10.1111/age.13293] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Revised: 11/14/2022] [Accepted: 12/31/2022] [Indexed: 01/15/2023]
Abstract
Wenchang (WC) chicken, the only indigenous chicken breed listed in Chinese genetic resources in Hainan province, is well known for its excellent meat quality and is sold all over southeast Asia. In recent years, the number of WC has decreased sharply with considerable variability in the quality at market. To explore the genetic diversity and population structure of WC chickens, the whole-genome data of 235 WC individuals from three conservation farms were obtained using the Illumina 150 bp paired-end platform and used in conjunction with the sequencing data from 123 individuals from other chicken breeds (including eight Chinese indigenous chicken breeds and three foreign or commercial breeds) downloaded from a public database. A total of 12 111 532 SNPs were identified, of which 11 541 878 SNPs were identified in WC. The results of gene enrichment analyses revealed that the SNPs harbored in WC genomes are mainly related to environmental adaptation, disease resistance and meat quality traits. Genetic diversity statistics, quantified by expected heterozygosity, observed heterozygosity, linkage disequilibrium, nucleotide diversity and fixation statistics, indicated that WC displays high genetic diversity compared with other Chinese indigenous chicken breeds. Genetic structure analyses showed that each population displayed great differentiation between WC and the other breeds, indicating the uniqueness of WC. In conclusion, the results of our study provide the first genomic overview of genetic variants, genetic diversity and population structure of WC from three conservation farms. This information will be valuable for the future breeding and conservation of WC and other surveyed populations.
Collapse
Affiliation(s)
- Shuaishuai Tian
- Hainan Key Laboratory of Tropical Animal Reproduction and Breeding and Epidemic Disease Research, College of Animal Science and Technology, Hainan University, Haikou, China
| | - Wei Li
- Department of Animal Science, College of Animal Sciences, Zhejiang University, Hangzhou, China
| | - Ziqi Zhong
- Hainan Key Laboratory of Tropical Animal Reproduction and Breeding and Epidemic Disease Research, College of Animal Science and Technology, Hainan University, Haikou, China
| | - Feifan Wang
- Hainan Key Laboratory of Tropical Animal Reproduction and Breeding and Epidemic Disease Research, College of Animal Science and Technology, Hainan University, Haikou, China
| | - Qian Xiao
- Hainan Key Laboratory of Tropical Animal Reproduction and Breeding and Epidemic Disease Research, College of Animal Science and Technology, Hainan University, Haikou, China
| |
Collapse
|
16
|
Nun N, Joy A. Fabrication and Bioactivity of Peptide-Conjugated Biomaterial Tissue Engineering Constructs. Macromol Rapid Commun 2023; 44:e2200342. [PMID: 35822458 DOI: 10.1002/marc.202200342] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2022] [Revised: 06/22/2022] [Indexed: 01/11/2023]
Abstract
Tissue engineering combines materials engineering, cells and biochemical factors to improve, restore or replace various types of biological tissues. A nearly limitless combination of these strategies can be combined, providing a means to augment the function of a number of biological tissues such as skin tissue, neural tissue, bones, and cartilage. Compounds such as small molecule therapeutics, proteins, and even living cells have been incorporated into tissue engineering constructs to influence biological processes at the site of implantation. Peptides have been conjugated to tissue engineering constructs to circumvent limitations associated with conjugation of proteins or incorporation of cells. This review highlights various contemporary examples in which peptide conjugation is used to overcome the disadvantages associated with the inclusion of other bioactive compounds. This review covers several peptides that are commonly used in the literature as well as those that do not appear as frequently to provide a broad scope of the utility of the peptide conjugation technique for designing constructs capable of influencing the repair and regeneration of various bodily tissues. Additionally, a brief description of the construct fabrication techniques encountered in the covered examples and their advantages in various tissue engineering applications is provided.
Collapse
Affiliation(s)
- Nicholas Nun
- School of Polymer Science and Polymer Engineering, The University of Akron, Akron, OH, 44321, USA
| | - Abraham Joy
- School of Polymer Science and Polymer Engineering, The University of Akron, Akron, OH, 44321, USA
| |
Collapse
|
17
|
Kuhns BD, Reuter JM, Hansen VL, Soles GL, Jonason JH, Ackert-Bicknell CL, Wu CL, Giordano BD. Whole-genome RNA sequencing identifies distinct transcriptomic profiles in impingement cartilage between patients with femoroacetabular impingement and hip osteoarthritis. J Orthop Res 2022. [PMID: 36463522 DOI: 10.1002/jor.25485] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Accepted: 11/17/2022] [Indexed: 12/05/2022]
Abstract
Femoroacetabular impingement (FAI) has a strong clinical association with the development of hip osteoarthritis (OA); however, the pathobiological mechanisms underlying the transition from focal impingement to global joint degeneration remain poorly understood. The purpose of this study is to use whole-genome RNA sequencing to identify and subsequently validate differentially expressed genes (DEGs) in femoral head articular cartilage samples from patients with FAI and hip OA secondary to FAI. Thirty-seven patients were included in the study with whole-genome RNA sequencing performed on 10 gender-matched patients in the FAI and OA cohorts and the remaining specimens were used for validation analyses. We identified a total of 3531 DEGs between the FAI and OA cohorts with multiple targets for genes implicated in canonical OA pathways. Quantitative reverse transcription-polymerase chain reaction validation confirmed increased expression of FGF18 and WNT16 in the FAI samples, while there was increased expression of MMP13 and ADAMTS4 in the OA samples. Expression levels of FGF18 and WNT16 were also higher in FAI samples with mild cartilage damage compared to FAI samples with severe cartilage damage or OA cartilage. Our study further expands the knowledge regarding distinct genetic reprogramming in the cartilage between FAI and hip OA patients. We independently validated the results of the sequencing analysis and found increased expression of anabolic markers in patients with FAI and minimal histologic cartilage damage, suggesting that anabolic signaling may be increased in early FAI with a transition to catabolic and inflammatory gene expression as FAI progresses towards more severe hip OA. Clinical significance:Cam-type FAI has a strong clinical association with hip OA; however, the cellular pathophysiology of disease progression remains poorly understood. Several previous studies have demonstrated increased expression of inflammatory markers in FAI cartilage samples, suggesting the involvement of these inflammatory pathways in the disease progression. Our study further expands the knowledge regarding distinct genetic reprogramming in the cartilage between FAI and hip OA patients. In addition to differences in inflammatory gene expression, we also identified differential expression in multiple pathways involved in hip OA progression.
Collapse
Affiliation(s)
- Benjamin D Kuhns
- Center for Regenerative and Personalized Medicine, Steadman-Philippon Research Institute, Vail, Colorado, USA
| | - John M Reuter
- Department of Orthopaedics and Rehabilitation, University of Rochester Medical Center, Rochester, New York, USA
| | - Victoria L Hansen
- Department of Orthopaedics and Rehabilitation, University of Rochester Medical Center, Rochester, New York, USA
| | - Gillian L Soles
- Department of Orthopedic Surgery, University of California Davis Health System, Sacramento, California, USA
| | - Jennifer H Jonason
- Department of Orthopaedics and Rehabilitation, University of Rochester Medical Center, Rochester, New York, USA
| | - Cheryl L Ackert-Bicknell
- Colorado Program for Musculoskeletal Research, Department of Orthopedics, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Chia-Lung Wu
- Department of Orthopaedics and Rehabilitation, University of Rochester Medical Center, Rochester, New York, USA
| | - Brian D Giordano
- Department of Orthopaedics and Rehabilitation, University of Rochester Medical Center, Rochester, New York, USA
| |
Collapse
|
18
|
Xie M, Dai H, Gu Q, Xiao C, Wang H, Lei Y, Wu C, Li X, Lin B, Li S. Identification of genes contributing to cisplatin resistance in osteosarcoma cells. FEBS Open Bio 2022; 13:164-173. [PMID: 36408691 PMCID: PMC9808595 DOI: 10.1002/2211-5463.13524] [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: 07/27/2022] [Revised: 11/08/2022] [Accepted: 11/18/2022] [Indexed: 11/22/2022] Open
Abstract
Osteosarcomas are prevalent in children and young adults and have a high recurrence rate. Cisplatin, doxorubicin, and methotrexate are common adjuvant chemotherapy drugs for treatment of osteosarcoma, but multidrug resistance is a growing problem. Therefore, understanding the molecular mechanisms of chemotherapy resistance in osteosarcoma cells is crucial for developing new therapeutic approaches and ultimately improving the prognosis of osteosarcoma patients. To identify genes associated with cisplatin resistance in osteosarcoma, we screened a large-scale mutant library generated by transfecting human osteosarcoma cells with a piggyBac (PB) transposon-based gene activation vector. Several candidate genes were identified by using Splinkerette-PCR paired with Next Generation Sequencing. We created a disease-free survival predictor model, which includes ZNF720, REEP3, CNNM2, and CGREF1, using TARGET (Therapeutically Applicable Research to Generate Effective Treatments) datasets. Additionally, the results of our enrichment analysis between the Four_genes_high group and Low_group suggested that these four genes may participate in cisplatin resistance in osteosarcoma through cross talk between various signaling pathways, especially the signaling pathway related to bone formation. These data may help guide future studies into chemotherapy for osteosarcoma.
Collapse
Affiliation(s)
- Mingzhong Xie
- The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical UniversityLuzhouChina
| | - Haoping Dai
- The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical UniversityLuzhouChina
| | - Qingwen Gu
- The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical UniversityLuzhouChina
| | - Changming Xiao
- The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical UniversityLuzhouChina
| | - Haozhong Wang
- The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical UniversityLuzhouChina
| | - Yang Lei
- The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical UniversityLuzhouChina
| | - Chunxiao Wu
- The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical UniversityLuzhouChina
| | - Xuening Li
- The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical UniversityLuzhouChina
| | - Birong Lin
- The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical UniversityLuzhouChina
| | - Sen Li
- The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical UniversityLuzhouChina
| |
Collapse
|
19
|
Niu Z, Su G, Li T, Yu H, Shen Y, Zhang D, Liu X. Vascular Calcification: New Insights Into BMP Type I Receptor A. Front Pharmacol 2022; 13:887253. [PMID: 35462911 PMCID: PMC9019578 DOI: 10.3389/fphar.2022.887253] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Accepted: 03/21/2022] [Indexed: 11/13/2022] Open
Abstract
Vascular calcification (VC) is a complex ectopic calcification process and an important indicator of increased risk for diabetes, atherosclerosis, chronic kidney disease, and other diseases. Therefore, clarifying the pathogenesis of VC is of great clinical significance. Numerous studies have shown that the onset and progression of VC are similar to bone formation. Members of the bone morphogenetic protein (BMP) family of proteins are considered key molecules in the progression of vascular calcification. BMP type I receptor A (BMPR1A) is a key receptor of BMP factors acting on the cell membrane, is widely expressed in various tissues and cells, and is an important “portal” for BMP to enter cells and exert their biological effect. In recent years, many discoveries have been made regarding the occurrence and treatment of ectopic ossification-related diseases involving BMP signaling targets. Studies have confirmed that BMPR1A is involved in osteogenic differentiation and that its high expression in vascular endothelial cells and smooth muscle cells can lead to vascular calcification. This article reviews the role of BMPR1A in vascular calcification and the possible underlying molecular mechanisms to provide clues for the clinical treatment of such diseases.
Collapse
Affiliation(s)
- Zhixing Niu
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Guanyue Su
- West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, China
| | - Tiantian Li
- West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, China
| | - Hongchi Yu
- West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, China
| | - Yang Shen
- West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, China
| | - Demao Zhang
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
- West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, China
- *Correspondence: Demao Zhang, ; Xiaoheng Liu,
| | - Xiaoheng Liu
- West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, China
- *Correspondence: Demao Zhang, ; Xiaoheng Liu,
| |
Collapse
|
20
|
In silico effect of Korean medicinal phytocompounds on gene targets of osteoarthritis. ADVANCES IN TRADITIONAL MEDICINE 2021. [DOI: 10.1007/s13596-021-00616-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
21
|
Zhang Y, Liu T, Hu X, Wang M, Wang J, Zou B, Tan P, Cui T, Dou Y, Ning L, huang Y, Rao S, Wang D, Zhao X. CellCall: integrating paired ligand-receptor and transcription factor activities for cell-cell communication. Nucleic Acids Res 2021; 49:8520-8534. [PMID: 34331449 PMCID: PMC8421219 DOI: 10.1093/nar/gkab638] [Citation(s) in RCA: 148] [Impact Index Per Article: 37.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Revised: 06/24/2021] [Accepted: 07/16/2021] [Indexed: 12/14/2022] Open
Abstract
With the dramatic development of single-cell RNA sequencing (scRNA-seq) technologies, the systematic decoding of cell-cell communication has received great research interest. To date, several in-silico methods have been developed, but most of them lack the ability to predict the communication pathways connecting the insides and outsides of cells. Here, we developed CellCall, a toolkit to infer inter- and intracellular communication pathways by integrating paired ligand-receptor and transcription factor (TF) activity. Moreover, CellCall uses an embedded pathway activity analysis method to identify the significantly activated pathways involved in intercellular crosstalk between certain cell types. Additionally, CellCall offers a rich suite of visualization options (Circos plot, Sankey plot, bubble plot, ridge plot, etc.) to present the analysis results. Case studies on scRNA-seq datasets of human testicular cells and the tumor immune microenvironment demonstrated the reliable and unique functionality of CellCall in intercellular communication analysis and internal TF activity exploration, which were further validated experimentally. Comparative analysis of CellCall and other tools indicated that CellCall was more accurate and offered more functions. In summary, CellCall provides a sophisticated and practical tool allowing researchers to decipher intercellular communication and related internal regulatory signals based on scRNA-seq data. CellCall is freely available at https://github.com/ShellyCoder/cellcall.
Collapse
Affiliation(s)
- Yang Zhang
- Department of Bioinformatics, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Tianyuan Liu
- Department of Bioinformatics, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Xuesong Hu
- State Key Laboratory of Organ Failure Research, Department of Developmental Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Mei Wang
- State Key Laboratory of Organ Failure Research, Department of Developmental Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Jing Wang
- Department of Bioinformatics, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Bohao Zou
- Department of Statistics, University of California Davis, Davis, CA, USA
| | - Puwen Tan
- Department of Bioinformatics, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Tianyu Cui
- Department of Bioinformatics, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Yiying Dou
- Department of Bioinformatics, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Lin Ning
- Department of Bioinformatics, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Yan huang
- Department of Bioinformatics, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Shuan Rao
- Department of Thoracic Surgery, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Dong Wang
- Department of Bioinformatics, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Xiaoyang Zhao
- State Key Laboratory of Organ Failure Research, Department of Developmental Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
- Guangdong Key Laboratory of Construction and Detection in Tissue Engineering, Southern Medical University, Guangzhou 510515, China
- Department of Gynecology, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China
| |
Collapse
|
22
|
Jiang H, Jia P. MiR-153-3p inhibits osteogenic differentiation of periodontal ligament stem cells through KDM6A-induced demethylation of H3K27me3. J Periodontal Res 2020; 56:379-387. [PMID: 33368310 DOI: 10.1111/jre.12830] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2020] [Revised: 11/23/2020] [Accepted: 12/03/2020] [Indexed: 12/11/2022]
Abstract
BACKGROUND AND OBJECTIVE Periodontal ligament stem cells (PDLSCs) have potential for osteogenic differentiation and show a great foreground in treating bone diseases. Histone three lysine 27 (H3K27) demethylase lysine demethylase 6A (KDM6A) is a critical epigenetic modifier and plays an important role in regulating osteogenic differentiation. Multiple microRNAs have been found to play important roles in osteogenesis. The aim of this study was to explore the mechanisms underlying the roles of miR-153-3p and KDM6A in PDLSC osteogenesis. METHODS The levels of the osteogenic markers alkaline phosphatase (ALP), runt-related transcription factor 2 (Runx2), and osteopontin (OPN) were measured by western blotting. Osteoblast activity and mineral deposition were detected by ALP and Alizarin red S (ARS) staining. The levels of miR-153-3p and KDM6A were measured by quantitative real-time PCR (qRT-PCR). A luciferase reporter assay was used to confirm the interaction between KDM6A and miR-153-3p. Gain-of-function and loss-of-function assays were performed to identify the roles of miR-153-3p and KDM6A in the osteogenic differentiation of PDLSCs. RESULTS In osteogenic PDLSCs, the expression of KDM6A, ALP, Runx2, and OPN was upregulated, whereas that of miR-153-3p was downregulated. miR-153-3p downregulation or KDM6A overexpression promoted the osteogenic differentiation of PDLSCs, as demonstrated by increases in ALP activity, matrix mineralization, and ALP, Runx2, and OPN expression. KDM6A was confirmed to be a target of miR-153-3p, and KDM6A overexpression reversed the inhibitory effect of miR-153-3p mimic on PDLSC osteogenesis. KDM6A promoted ALP, Runx2, and OPN expression through the demethylation of H3K27me3 on the promoter regions of these genes. CONCLUSION miR-153-3p inhibited PDLSC osteogenesis by targeting KDM6A and inhibiting ALP, Runx2, and OPN transcription. These findings provide latent hope for PDLSCs application in periodontal therapy.
Collapse
Affiliation(s)
- Hao Jiang
- Department of Stomatology, The Fifth Central Hospital of Tianjin, Tianjin, China
| | - Peizeng Jia
- Department of Orthodontics, Peking University School of Stomatology, Beijing, China
| |
Collapse
|
23
|
First person – Tanja Mang. J Cell Sci 2020. [DOI: 10.1242/jcs.253005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
ABSTRACT
First Person is a series of interviews with the first authors of a selection of papers published in Journal of Cell Science, helping early-career researchers promote themselves alongside their papers. Tanja Mang is first author on ‘BMPR1A is necessary for chondrogenesis and osteogenesis, whereas BMPR1B prevents hypertrophic differentiation’, published in JCS. Tanja conducted the research described in this article while a PhD student in Dr Anne Gigout's lab at Merck KGaA, Darmstadt, Germany, where she worked on the development of relevant preclinical cell culture models to provide systems for testing drugs and identifying specific biomarkers.
Collapse
|