1
|
Yang W, Lefebvre V. PTPN11 in cartilage development, adult homeostasis, and diseases. Bone Res 2025; 13:53. [PMID: 40379623 DOI: 10.1038/s41413-025-00425-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2025] [Revised: 03/07/2025] [Accepted: 03/17/2025] [Indexed: 05/19/2025] Open
Abstract
The SH2 domain-containing protein tyrosine phosphatase 2 (SHP2, also known as PTP2C), encoded by PTPN11, is ubiquitously expressed and has context-specific effects. It promotes RAS/MAPK signaling downstream of receptor tyrosine kinases, cytokine receptors, and extracellular matrix proteins, and was shown in various lineages to modulate cell survival, proliferation, differentiation, and migration. Over the past decade, PTPN11 inactivation in chondrocytes was found to cause metachondromatosis, a rare disorder characterized by multiple enchondromas and osteochondroma-like lesions. Moreover, SHP2 inhibition was found to mitigate osteoarthritis pathogenesis in mice, and abundant but incomplete evidence suggests that SHP2 is crucial for cartilage development and adult homeostasis, during which its expression and activity are tightly regulated transcriptionally and posttranslationally, and by varying sets of functional partners. Fully uncovering SHP2 actions and regulation in chondrocytes is thus fundamental to understanding the mechanisms underlying both rare and common cartilage diseases and to designing effective disease treatments. We here review current knowledge, highlight recent discoveries and controversies, and propose new research directions to answer remaining questions.
Collapse
Affiliation(s)
- Wentian Yang
- Department of Orthopaedic Surgery, Brown University Alpert Medical School and Rhode Island Hospital, Providence, RI, USA.
| | - Véronique Lefebvre
- Division of Orthopaedic Surgery, Department of Surgery, Children's Hospital of Philadelphia, Philadelphia, PA, USA.
| |
Collapse
|
2
|
Xue C, Chu Q, Shi Q, Zeng Y, Lu J, Li L. Wnt signaling pathways in biology and disease: mechanisms and therapeutic advances. Signal Transduct Target Ther 2025; 10:106. [PMID: 40180907 PMCID: PMC11968978 DOI: 10.1038/s41392-025-02142-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Revised: 11/13/2024] [Accepted: 12/29/2024] [Indexed: 04/05/2025] Open
Abstract
The Wnt signaling pathway is critically involved in orchestrating cellular functions such as proliferation, migration, survival, and cell fate determination during development. Given its pivotal role in cellular communication, aberrant Wnt signaling has been extensively linked to the pathogenesis of various diseases. This review offers an in-depth analysis of the Wnt pathway, detailing its signal transduction mechanisms and principal components. Furthermore, the complex network of interactions between Wnt cascades and other key signaling pathways, such as Notch, Hedgehog, TGF-β, FGF, and NF-κB, is explored. Genetic mutations affecting the Wnt pathway play a pivotal role in disease progression, with particular emphasis on Wnt signaling's involvement in cancer stem cell biology and the tumor microenvironment. Additionally, this review underscores the diverse mechanisms through which Wnt signaling contributes to diseases such as cardiovascular conditions, neurodegenerative disorders, metabolic syndromes, autoimmune diseases, and cancer. Finally, a comprehensive overview of the therapeutic progress targeting Wnt signaling was given, and the latest progress in disease treatment targeting key components of the Wnt signaling pathway was summarized in detail, including Wnt ligands/receptors, β-catenin destruction complexes, and β-catenin/TCF transcription complexes. The development of small molecule inhibitors, monoclonal antibodies, and combination therapy strategies was emphasized, while the current potential therapeutic challenges were summarized. This aims to enhance the current understanding of this key pathway.
Collapse
Affiliation(s)
- Chen Xue
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Qingfei Chu
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Qingmiao Shi
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yifan Zeng
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Juan Lu
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.
| | - Lanjuan Li
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.
| |
Collapse
|
3
|
Li X, Zhao Y, Jiang G. Aldosterone promotes calcification of vascular smooth muscle cells in mice through the AIF-1/Wnt/β-catenin signaling pathway. Int Urol Nephrol 2025; 57:613-623. [PMID: 39312016 DOI: 10.1007/s11255-024-04213-3] [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: 07/18/2024] [Accepted: 09/18/2024] [Indexed: 01/29/2025]
Abstract
OBJECTIVE This study aimed to investigate the impact of aldosterone on calcification in murine vascular smooth muscle cells (VSMCs) via the allograft inflammatory factor-1 (AIF-1)/Wnt/β-catenin signaling pathway. METHODS Mouse VSMCs were cultured in vitro, and calcification was induced by treatment with 100 nM aldosterone. The level of calcification in mouse VSMCs was evaluated using colorimetric assays to assess ALP activity and qRT-PCR to identify the expression of calcification-related markers, such as Runx2, α-SMA, OCN, and ALP mRNA. Western blot analysis was performed to determine the protein expression levels associated with the Wnt/β-catenin pathway (LRP6, p-LRP6, GSK3β, p-GSK3β, β-catenin) and AIF-1. Plasmid transfection techniques were utilized to either knock down or overexpress AIF-1, and the subsequent alterations in these markers were observed. RESULTS (1) Compared to the control group, the aldosterone treatment group with exhibited a significant increase in ALP. Concurrently, Runx2, OCN, and ALP mRNA levels increased, as did LRP6, p-LRP6, GSK3β, p-GSK3β, β-catenin, and AIF-1 protein levels. Additionally, a significant decrease in the expression of α-SMA mRNA was observed (P < 0.05). (2) The aldosterone + oe-AIF-1 group showed significant increases in ALP activity compared to the aldosterone + oe-NC group, whereas the aldosterone + sh-AIF-1 group showed significant decreases (P < 0.05). (3) The aldosterone + oe-AIF-1 group exhibited significantly upregulated expression of AIF-1, p-LRP6/LRP6, p-GSK3β/GSK3β, and β-catenin proteins relative to the aldosterone + oe-NC group (P < 0.05). This was concurrent with increased mRNA expression of Runx2, OCN, and ALP, and decreased α-SMA mRNA expression (P < 0.05). CONCLUSION Aldosterone affects the calcification process in mouse VSMCs, and the activation of the AIF-1/Wnt/β-catenin signaling pathway is the mechanism behind its action.
Collapse
MESH Headings
- Animals
- Mice
- Wnt Signaling Pathway/drug effects
- Muscle, Smooth, Vascular/cytology
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/pathology
- Muscle, Smooth, Vascular/drug effects
- Aldosterone/pharmacology
- Cells, Cultured
- Vascular Calcification/metabolism
- Vascular Calcification/chemically induced
- Vascular Calcification/pathology
- Core Binding Factor Alpha 1 Subunit/metabolism
- Core Binding Factor Alpha 1 Subunit/genetics
- Myocytes, Smooth Muscle/metabolism
- Myocytes, Smooth Muscle/drug effects
- beta Catenin/metabolism
- Low Density Lipoprotein Receptor-Related Protein-6/metabolism
- Microfilament Proteins/metabolism
- DNA-Binding Proteins/metabolism
Collapse
Affiliation(s)
- Xin Li
- Department of Nephrology, Fourth Affiliated Hospital of Harbin Medical University, Harbin, 150001, Heilongjiang, People's Republic of China
| | - Yingzi Zhao
- Department of Nephrology, Fourth Affiliated Hospital of Harbin Medical University, Harbin, 150001, Heilongjiang, People's Republic of China
| | - Guotao Jiang
- Department of Nephrology, Fourth Affiliated Hospital of Harbin Medical University, Harbin, 150001, Heilongjiang, People's Republic of China.
| |
Collapse
|
4
|
Stellpflug A, Caron J, Fasciano S, Wang B, Wang S. Bone-derived nanoparticles (BNPs) enhance osteogenic differentiation via Notch signaling. NANOSCALE ADVANCES 2025; 7:735-747. [PMID: 39823045 PMCID: PMC11734751 DOI: 10.1039/d4na00797b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/24/2024] [Accepted: 12/26/2024] [Indexed: 01/19/2025]
Abstract
Mesenchymal stem cell (MSC)-based bone tissue regeneration has gained significant attention due to the excellent differentiation capacity and immunomodulatory activity of MSCs. Enhancing osteogenesis regulation is crucial for improving the therapeutic efficacy of MSC-based regeneration. By utilizing the regenerative capacity of bone ECM and the functionality of nanoparticles, we recently engineered bone-based nanoparticles (BNPs) from decellularized porcine bones. The effects of internalization of BNPs on MSC viability, proliferation, and osteogenic differentiation were first investigated and compared at different time points. The phenotypic behaviors, including cell number, proliferation, and differentiation were characterized and compared. By incorporating a LNA/DNA nanobiosensor and MSC live cell imaging, we monitored and compared Notch ligand delta-like 4 (Dll4) expression dynamics in the cytoplasm and nucleus during osteogenic differentiation. Pharmacological interventions are used to inhibit Notch signaling to examine the mechanisms involved. The results suggest that Notch inhibition mediates the osteogenic process, with reduced expression of early and late stage differentiation markers (ALP and calcium mineralization). The internalization of BNPs led to an increase in Dll4 expression, exhibiting a time-dependent pattern that aligned with enhanced cell proliferation and differentiation. Our findings indicate that the observed changes in BNP-treated cells during osteogenic differentiation could be associated with elevated levels of Dll4 mRNA expression. In summary, this study provides new insights into MSC osteogenic differentiation and the molecular mechanisms through which BNPs stimulate this process. The results indicate that BNPs influence osteogenesis by modulating Notch ligand Dll4 expression, demonstrating a potential link between Notch signaling and the proteins present in BNPs.
Collapse
Affiliation(s)
- Austin Stellpflug
- Joint Department of Biomedical Engineering, Marquette University and the Medical College of Wisconsin Milwaukee WI 53226 USA
| | - Justin Caron
- Department of Chemistry, Chemical and Biomedical Engineering, University of New Haven West Haven CT 06516 USA
| | - Samantha Fasciano
- Department of Chemistry, Chemical and Biomedical Engineering, University of New Haven West Haven CT 06516 USA
| | - Bo Wang
- Joint Department of Biomedical Engineering, Marquette University and the Medical College of Wisconsin Milwaukee WI 53226 USA
| | - Shue Wang
- Department of Chemistry, Chemical and Biomedical Engineering, University of New Haven West Haven CT 06516 USA
| |
Collapse
|
5
|
Łuczak JW, Palusińska M, Matak D, Pietrzak D, Nakielski P, Lewicki S, Grodzik M, Szymański Ł. The Future of Bone Repair: Emerging Technologies and Biomaterials in Bone Regeneration. Int J Mol Sci 2024; 25:12766. [PMID: 39684476 DOI: 10.3390/ijms252312766] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2024] [Revised: 11/20/2024] [Accepted: 11/26/2024] [Indexed: 12/18/2024] Open
Abstract
Bone defects and fractures present significant clinical challenges, particularly in orthopedic and maxillofacial applications. While minor bone defects may be capable of healing naturally, those of a critical size necessitate intervention through the use of implants or grafts. The utilization of traditional methodologies, encompassing autografts and allografts, is constrained by several factors. These include the potential for donor site morbidity, the restricted availability of suitable donors, and the possibility of immune rejection. This has prompted extensive research in the field of bone tissue engineering to develop advanced synthetic and bio-derived materials that can support bone regeneration. The optimal bone substitute must achieve a balance between biocompatibility, bioresorbability, osteoconductivity, and osteoinductivity while simultaneously providing mechanical support during the healing process. Recent innovations include the utilization of three-dimensional printing, nanotechnology, and bioactive coatings to create scaffolds that mimic the structure of natural bone and enhance cell proliferation and differentiation. Notwithstanding the advancements above, challenges remain in optimizing the controlled release of growth factors and adapting materials to various clinical contexts. This review provides a comprehensive overview of the current advancements in bone substitute materials, focusing on their biological mechanisms, design considerations, and clinical applications. It explores the role of emerging technologies, such as additive manufacturing and stem cell-based therapies, in advancing the field. Future research highlights the need for multidisciplinary collaboration and rigorous testing to develop advanced bone graft substitutes, improving outcomes and quality of life for patients with complex defects.
Collapse
Affiliation(s)
- Julia Weronika Łuczak
- Department of Molecular Biology, Institute of Genetics and Animal Biotechnology, Polish Academy of Sciences, Postępu 36A, 05-552 Magdalenka, Poland
- Department of Nanobiotechnology, Institute of Biology, Warsaw University of Life Sciences, Ciszewskiego 8, Bldg. 23, 02-786 Warsaw, Poland
| | - Małgorzata Palusińska
- Department of Molecular Biology, Institute of Genetics and Animal Biotechnology, Polish Academy of Sciences, Postępu 36A, 05-552 Magdalenka, Poland
| | - Damian Matak
- European Biomedical Institute, 05-410 Jozefów, Poland
| | - Damian Pietrzak
- Division of Parasitology and Parasitic Diseases, Department of Preclinical Sciences, Institute of Veterinary Medicine, Warsaw University of Life Sciences, 02-786 Warsaw, Poland
| | - Paweł Nakielski
- Department of Biosystems and Soft Matter, Institute of Fundamental Technological Research, Polish Academy of Sciences, Pawińskiego 5B, 02-106 Warsaw, Poland
| | - Sławomir Lewicki
- Institute of Outcomes Research, Maria Sklodowska-Curie Medical Academy, Pl. Żelaznej Bramy 10, 00-136 Warsaw, Poland
| | - Marta Grodzik
- Department of Nanobiotechnology, Institute of Biology, Warsaw University of Life Sciences, Ciszewskiego 8, Bldg. 23, 02-786 Warsaw, Poland
| | - Łukasz Szymański
- Department of Molecular Biology, Institute of Genetics and Animal Biotechnology, Polish Academy of Sciences, Postępu 36A, 05-552 Magdalenka, Poland
- European Biomedical Institute, 05-410 Jozefów, Poland
| |
Collapse
|
6
|
Yamashita E, Negishi S, Kikuta J, Shimizu M, Senpuku H. Effects of Improper Mechanical Force on the Production of Sonic Hedgehog, RANKL, and IL-6 in Human Periodontal Ligament Cells In Vitro. Dent J (Basel) 2024; 12:108. [PMID: 38668020 PMCID: PMC11049549 DOI: 10.3390/dj12040108] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2023] [Revised: 02/28/2024] [Accepted: 04/08/2024] [Indexed: 04/28/2024] Open
Abstract
Improper mechanical stress may induce side effects during orthodontic treatment. If the roots and alveolar bones are extensively resorbed following excess mechanical stress, unplanned tooth mobility and inflammation can occur. Although multiple factors are believed to contribute to the development of side effects, the cause is still unknown. Sonic hedgehog (Shh), one of the hedgehog signals significantly associated with cell growth and cancer development, promotes osteoclast formation in the jawbone. Shh may be associated with root and bone resorptions during orthodontic treatment. In this study, we investigated the relationships between Shh, RANKL, and IL-6 in human periodontal ligament (hPDL) cells exposed to improper mechanical force. Weights were placed on hPDL cells and human gingival fibroblasts (HGFs) for an optimal orthodontic force group (1.0 g/cm2) and a heavy orthodontic force group (4.0 g/cm2). A group with no orthodontic force was used as a control group. Real-time PCR, SDS-PAGE, and Western blotting were performed to examine the effects of orthodontic forces on the expression of Shh, RANKL, and IL-6 at 2, 4, 6, 8, 12, and 24 h after the addition of pressure. The protein expression of Shh was not clearly induced by orthodontic forces of 1.0 and 4.0 g/cm2 compared with the control in HGFs and hPDL cells. In contrast, RANKL and IL-6 gene and protein expression was significantly induced by 1.0 and 4.0 g/cm2 in hPDL cells for forces lasting 6~24 h. However, neither protein was expressed in HGFs. RANKL and IL-6 expressions in response to orthodontic forces and in the control were clearly inhibited by Shh inhibitor RU-SKI 43. Shh did not directly link to RANKL and IL-6 for root and bone resorptions by orthodontic force but was associated with cell activities to be finally guided by the production of cytokines in hPDL cells.
Collapse
Affiliation(s)
- Erika Yamashita
- Department of Orthodontics, Nihon University of School at Matsudo, Matsudo 271-8587, Japan; (E.Y.); (S.N.); (J.K.)
| | - Shinichi Negishi
- Department of Orthodontics, Nihon University of School at Matsudo, Matsudo 271-8587, Japan; (E.Y.); (S.N.); (J.K.)
| | - Jun Kikuta
- Department of Orthodontics, Nihon University of School at Matsudo, Matsudo 271-8587, Japan; (E.Y.); (S.N.); (J.K.)
| | - Mami Shimizu
- Department of Orthodontics, Nihon University of School at Matsudo, Matsudo 271-8587, Japan; (E.Y.); (S.N.); (J.K.)
| | - Hidenobu Senpuku
- Department of Microbiology and Immunology, Nihon University of School at Matsudo, Matsudo 271-8587, Japan
| |
Collapse
|
7
|
Wu Q, Liu H, Yang Q, Qi J, Xi Y, Tang Q, Wang R, Hu J, Li L. Transcriptome-based comparison reveals key genes regulating allometry growth of forelimb and hindlimb bone in duck embryos. Poult Sci 2024; 103:103317. [PMID: 38160613 PMCID: PMC10792745 DOI: 10.1016/j.psj.2023.103317] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2023] [Revised: 10/29/2023] [Accepted: 11/20/2023] [Indexed: 01/03/2024] Open
Abstract
Allometric growth of the forelimb and hindlimb is a widespread phenomenon observed in vertebrates. As a typical precocial bird, ducks exhibit more advanced development of their hindlimbs compared to their forelimbs, enabling them to walk shortly after hatching. This phenomenon is closely associated with the development of long bones in the embryonic stage. However, the molecular mechanism governing the allometric growth of duck forelimb and hindlimb bones is remains elusive. In this study, we employed phenotypic, histological, and gene expression analyses to investigate developmental differences between the humerus (forelimb bone) and tibia/femur (hindlimb bones) in duck embryos. Our results revealed a gradual increase in weight and length disparity between the tibia and humerus from E12 to E28 (embryo age). At E12, endochondral ossification was observed solely in the tibia but not in the humerus. The number of differentially expressed genes (DEGs) gradually increased at H12 vs. T12, H20 vs. T20, and H28 vs. T28 stages consistent with phenotypic variations. A total of 38 DEGs were found across all 3 stages. Protein-protein interaction network analysis demonstrated strong interactions among members of HOXD gene family (HOXD3/8/9/10/11/12), HOXB gene family (HOXB8/9), TBX gene family (TBX4/5/20), HOXA11, SHOX2, and MEIS2. Gene expression profiling indicated higher expression levels for all HOXD genes in the humerus compared to tibia while opposite trends were observed for HOXA/HOXB genes with low or no expression detected in the humerus. These findings suggest distinct roles played by different clusters within HOX gene family during skeletal development regulation of duck embryo's forelimbs versus hind limbs. Notably, TBX4 exhibited high expression levels specifically in tibia whereas TBX5 showed similar patterns exclusively within humerus as seen previously across other species' studies. In summary, this study identified key regulatory genes involved in allometric growth of duck forelimb and hindlimb bones during embryonic development. Skeletal development is a complex physiological process, and further research is needed to elucidate the regulatory role of candidate genes in endochondral ossification.
Collapse
Affiliation(s)
- Qifan Wu
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, 611130, China; Ministry of Agriculture, Forestry and Food Engineering, Yibin University, Yibin, 644000, China
| | - Hehe Liu
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, 611130, China
| | - Qinglan Yang
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, 611130, China
| | - Jingjing Qi
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, 611130, China
| | - Yang Xi
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, 611130, China
| | - Qian Tang
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, 611130, China
| | - Rui Wang
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, 611130, China
| | - Jiwei Hu
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, 611130, China
| | - Liang Li
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, 611130, China.
| |
Collapse
|
8
|
Cao X, Deng S, Liu Q, Wu L, Zhuang X, Ding S. Important Role of the Ihh Signaling Pathway in Initiating Early Cranial Remodeling and Morphological Specialization in Cromileptes altivelis. Animals (Basel) 2023; 13:3840. [PMID: 38136878 PMCID: PMC10740873 DOI: 10.3390/ani13243840] [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: 09/20/2023] [Revised: 12/11/2023] [Accepted: 12/11/2023] [Indexed: 12/24/2023] Open
Abstract
In this study, we identified the important contribution of frontal bone remodeling in shaping the 'sunken head and humpback' appearance in C. altivelis. Our investigation identified a developmental milestone at a total length of 5-6 cm, making the onset of its morphologic specialization in this species. A comparative analysis with closely related species reveals heightened activity in the frontal osteoblasts of the humpback grouper, potentially providing a physiological basis for its remodeling. Furthermore, our findings highlight that a significant upregulation in the expression levels of Ihhb, Ptch1, and Gli2a genes was seen in C. altivelis within the specified developmental stage, indicating an important involvement of the Ihhb-Ptch1-Gli2a signaling pathway in initiating the morphological specialization. We hypothesized that Ihh signaling could be attributed to shifts in mechanical stress, resulting from muscle traction on the frontal bone due to changes in swimming patterns during development. This study not only offers significant insights into unraveling the molecular mechanisms that govern phenotypic specialization and ecological adaptations in the humpback grouper but also serves as a valuable reference for studies on fishes with a controversial morphology and molecular phylogeny.
Collapse
Affiliation(s)
- Xiaoying Cao
- State Key Laboratory of Marine Environment Science, College of Ocean and Earth Sciences, Xiamen University, Xiamen 361102, China
| | - Shunyun Deng
- State Key Laboratory of Marine Environment Science, College of Ocean and Earth Sciences, Xiamen University, Xiamen 361102, China
| | - Quanyin Liu
- State Key Laboratory of Marine Environment Science, College of Ocean and Earth Sciences, Xiamen University, Xiamen 361102, China
| | - Lisheng Wu
- State Key Laboratory of Marine Environment Science, College of Ocean and Earth Sciences, Xiamen University, Xiamen 361102, China
- Xiamen Key Laboratory of Urban Sea Ecological Conservation and Restoration, College of Ocean and Earth Sciences, Xiamen University, Xiamen 361005, China
| | - Xuan Zhuang
- Department of Biological Sciences, University of Arkansas, Fayetteville, AR 72701, USA
| | - Shaoxiong Ding
- State Key Laboratory of Marine Environment Science, College of Ocean and Earth Sciences, Xiamen University, Xiamen 361102, China
- Xiamen Key Laboratory of Urban Sea Ecological Conservation and Restoration, College of Ocean and Earth Sciences, Xiamen University, Xiamen 361005, China
| |
Collapse
|
9
|
Schkoda S, Horman B, Witchey SK, Jansson A, Macari S, Patisaul HB. Skeletal effects following developmental flame-retardant exposure are specific to sex and chemical class in the adult Wistar rat. FRONTIERS IN TOXICOLOGY 2023; 5:1216388. [PMID: 37577032 PMCID: PMC10414991 DOI: 10.3389/ftox.2023.1216388] [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: 05/03/2023] [Accepted: 06/22/2023] [Indexed: 08/15/2023] Open
Abstract
Introduction: Accumulating evidence reveals that endocrine disrupting chemicals (EDCs) can disrupt aspects of metabolic programming, suggesting that skeletal development may be at risk, a possibility that is rarely examined. The commercial flame retardant (FR) mixture, Firemaster 550 (FM 550), has repeatedly been shown to negatively influence metabolic programming, raising concerns that skeletal integrity may consequently be impaired. We have previously shown that gestational and lactational exposure to 1,000 µg FM 550 negatively affected sex-specific skeletal traits in male, but not female, rats assessed at 6 months of age. Whether this outcome is primarily driven by the brominated (BFR) or organophosphate ester (OPFR) portions of the mixture or the effects persist to older ages is unknown. Materials and methods: To address this, in the present study, dams were orally exposed throughout gestation and lactation to either 1,000 μg BFR, 1,000 µg OPFR, or 2,000 µg FM 550. Offspring (n = 8/sex/exposure) were weaned at PND 21 and assessed for femoral cortical and trabecular bone parameters at 8 months of age by high-resolution X-ray micro-computed tomography (micro-CT). Serum levels of serotonin, osteocalcin, alkaline phosphatase, and calcium were quantified. Results: FM 550 affected both sexes, but the females were more appreciably impacted by the OPFRs, while the males were more vulnerable to the BFRs. Conclusion: Although sex specificity was expected due to the sexual dimorphic nature of skeletal physiology, the mechanisms accounting for the male- and female-specific phenotypes remain to be determined. Future work aims to clarify these unresolved issues.
Collapse
Affiliation(s)
- Stacy Schkoda
- Department of Biological Sciences, North Carolina State University, Raleigh, NC, United States
| | - Brian Horman
- Department of Biological Sciences, North Carolina State University, Raleigh, NC, United States
| | - Shannah K. Witchey
- National Toxicology Program, National Institute of Environmental Health Sciences, Research Triangle Park, NC, United States
| | - Anton Jansson
- Analytical Instrumentation Facility, North Carolina State University, Raleigh, NC, United States
| | - Soraia Macari
- Department of Restorative Dentistry, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Heather B. Patisaul
- Department of Biological Sciences, North Carolina State University, Raleigh, NC, United States
- Center for Human Health and the Environment, North Carolina State University, Raleigh, NC, United States
| |
Collapse
|
10
|
Maden M, Polvadore T, Polanco A, Barbazuk WB, Stanley E. Osteoderms in a mammal the spiny mouse Acomys and the independent evolution of dermal armor. iScience 2023; 26:106779. [PMID: 37378333 PMCID: PMC10291248 DOI: 10.1016/j.isci.2023.106779] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Revised: 02/06/2023] [Accepted: 04/25/2023] [Indexed: 06/29/2023] Open
Abstract
Osteoderms are bony plates found in the skin of vertebrates, mostly commonly in reptiles where they have evolved independently multiple times, suggesting the presence of a gene regulatory network that is readily activated and inactivated. They are absent in birds and mammals except for the armadillo. However, we have discovered that in one subfamily of rodents, the Deomyinae, there are osteoderms in the skin of their tails. Osteoderm development begins in the proximal tail skin and is complete 6 weeks after birth. RNA sequencing has identified the gene networks involved in their differentiation. There is a widespread down-regulation of keratin genes and an up-regulation of osteoblast genes and a finely balanced expression of signaling pathways as the osteoderms differentiate. Future comparisons with reptilian osteoderms may allow us to understand how these structures have evolved and why they are so rare in mammals.
Collapse
Affiliation(s)
- Malcolm Maden
- Department of Biology & UF Genetics Institute, University of Florida, Gainesville, FL 32611, USA
| | - Trey Polvadore
- Department of Biology & UF Genetics Institute, University of Florida, Gainesville, FL 32611, USA
| | - Arod Polanco
- Department of Biology & UF Genetics Institute, University of Florida, Gainesville, FL 32611, USA
| | - W. Brad Barbazuk
- Department of Biology & UF Genetics Institute, University of Florida, Gainesville, FL 32611, USA
| | - Edward Stanley
- Florida Museum of Natural History, University of Florida, Museum Road, Gainesville, FL 32611, USA
| |
Collapse
|
11
|
Ismail OM, El-Omar OM, Said UN. Exploring the Role of Urocortin in Osteoporosis. Cureus 2023; 15:e38978. [PMID: 37313093 PMCID: PMC10259878 DOI: 10.7759/cureus.38978] [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] [Accepted: 05/13/2023] [Indexed: 06/15/2023] Open
Abstract
Osteoporosis is a debilitating disease that affects over 200 million people worldwide. Overactive osteoclast activity leads to micro-architectural defects and low bone mass. This culminates in fragility fractures, such as femoral neck fractures. Treatments currently available either are not completely effective or have considerable side effects; thus, there is a need for more effective treatments. The urocortin (Ucn) family, composed of urocortin 1 (Ucn1), urocortin 2 (Ucn2), urocortin 3 (Ucn3), corticotropin-releasing factor (CRF) and corticotropin-releasing factor-binding protein (CRF-BP), exerts a wide range of effects throughout the body. Ucn1 has been shown to inhibit murine osteoclast activity. This review article will aim to bridge the gap between existing knowledge of Ucn and whether it can affect human osteoclasts.
Collapse
Affiliation(s)
- Omar M Ismail
- Faculty of Biology, Medicine and Health, University of Manchester, Manchester, GBR
| | - Omar M El-Omar
- Faculty of Biology, Medicine and Health, University of Manchester, Manchester, GBR
| | - Umar N Said
- Faculty of Biology, Medicine and Health, University of Manchester, Manchester, GBR
| |
Collapse
|
12
|
Li M, Li T, Yin J, Xie C, Zhu J. Evaluation of toxicological effects of bisphenol S with an in vitro human bone marrow mesenchymal stem cell: Implications for bone health. Toxicology 2023; 484:153408. [PMID: 36565802 DOI: 10.1016/j.tox.2022.153408] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2022] [Revised: 12/15/2022] [Accepted: 12/16/2022] [Indexed: 12/24/2022]
Abstract
As the use of bisphenol A (BPA) has been restricted in consumer products, bisphenol S (BPS) is one major alternative to BPA for various materials, leading to growing concerns about its health risks in human beings. However, little is known about the toxic effects of BPS on bone health. We employed human bone marrow mesenchymal stem cells (hBMSCs) for the in vitro assessment of BPS on cell proliferation, differentiation, and self-renewal. Our study revealed that BPS at concentrations of 10-10-10-7 M increased cell viability but induced the morphological changes of hBMSCs. Moreover, BPS decreased ROS generation and increased Nrf2 expression. Furthermore, BPS not only activated ERα/β expression but also increased β-catenin expression and induced the replicative senescence of hBMSCs. Furthermore, we found that the upregulation of β-catenin induced by BPS was mediated, in part, by ER signaling. Overall, our results suggested BPS exposure caused the homeostatic imbalance of hBMSCs.
Collapse
Affiliation(s)
- Mei Li
- The Laboratory Center for Basic Medical Sciences, Nanjing Medical University, Nanjing 211166, China; School of Biomedical Engineering and Informatics, Nanjing Medical University, Nanjing 211166, China
| | - Tenglong Li
- The Laboratory Center for Basic Medical Sciences, Nanjing Medical University, Nanjing 211166, China
| | - Juan Yin
- Department of Laboratory, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Suzhou 215008, China
| | - Chunfeng Xie
- Department of Nutrition and Food Safety, School of Public Health, Nanjing Medical University, Nanjing 211166, China.
| | - Jianyun Zhu
- Department of Laboratory, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Suzhou 215008, China.
| |
Collapse
|
13
|
Cui Y, Yi Q, Sun W, Huang D, Zhang H, Duan L, Shang H, Wang D, Xiong J. Molecular basis and therapeutic potential of myostatin on bone formation and metabolism in orthopedic disease. Biofactors 2023; 49:21-31. [PMID: 32997846 DOI: 10.1002/biof.1675] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Revised: 08/03/2020] [Accepted: 08/04/2020] [Indexed: 12/17/2022]
Abstract
Myostatin, a member of the transforming growth factor-β (TGF-β) superfamily, is a key autocrine/paracrine inhibitor of skeletal muscle growth. Recently, researchers have postulated that myostatin is a negative regulator of bone formation and metabolism. Reportedly, myostatin is highly expressed in the fracture area, affecting the endochondral ossification process during the early stages of fracture healing. Furthermore, myostatin is highly expressed in the synovium of patients with rheumatoid arthritis (RA) and is an effective therapeutic target for interfering with osteoclast formation and joint destruction in RA. Thus, myostatin is a potent anti-osteogenic factor and a direct modulator of osteoclast differentiation. Evaluation of the molecular pathway revealed that myostatin can activate SMAD and mitogen-activated protein kinase signaling pathways, inhibiting the Wnt/β-catenin pathway to synergistically regulate muscle and bone growth and metabolism. In summary, inhibition of myostatin or the myostatin signaling pathway has therapeutic potential in the treatment of orthopedic diseases. This review focused on the effects of myostatin on bone formation and metabolism and discussed the potential therapeutic effects of inhibiting myostatin and its pathways in related orthopedic diseases.
Collapse
Affiliation(s)
- Yinxing Cui
- Guangzhou Medical University, Guangzhou, Guangdong, China
- Guangdong Provincial Research Center for Artificial Intelligence and Digital Orthopedic Technology, Shenzhen Key Laboratory of Tissue Engineering, Shenzhen Laboratory of Digital Orthopedic Engineering, Department of Orthopedics, Shenzhen Second People's Hospital (The First Affiliated Hospital of Shenzhen University, Health Science Center), Shenzhen, Guangdong, China
| | - Qian Yi
- Guangdong Provincial Research Center for Artificial Intelligence and Digital Orthopedic Technology, Shenzhen Key Laboratory of Tissue Engineering, Shenzhen Laboratory of Digital Orthopedic Engineering, Department of Orthopedics, Shenzhen Second People's Hospital (The First Affiliated Hospital of Shenzhen University, Health Science Center), Shenzhen, Guangdong, China
| | - Weichao Sun
- Guangdong Provincial Research Center for Artificial Intelligence and Digital Orthopedic Technology, Shenzhen Key Laboratory of Tissue Engineering, Shenzhen Laboratory of Digital Orthopedic Engineering, Department of Orthopedics, Shenzhen Second People's Hospital (The First Affiliated Hospital of Shenzhen University, Health Science Center), Shenzhen, Guangdong, China
| | - Dixi Huang
- Guangzhou Medical University, Guangzhou, Guangdong, China
- Guangdong Provincial Research Center for Artificial Intelligence and Digital Orthopedic Technology, Shenzhen Key Laboratory of Tissue Engineering, Shenzhen Laboratory of Digital Orthopedic Engineering, Department of Orthopedics, Shenzhen Second People's Hospital (The First Affiliated Hospital of Shenzhen University, Health Science Center), Shenzhen, Guangdong, China
| | - Hui Zhang
- Guangdong Provincial Research Center for Artificial Intelligence and Digital Orthopedic Technology, Shenzhen Key Laboratory of Tissue Engineering, Shenzhen Laboratory of Digital Orthopedic Engineering, Department of Orthopedics, Shenzhen Second People's Hospital (The First Affiliated Hospital of Shenzhen University, Health Science Center), Shenzhen, Guangdong, China
- University of South China, Hengyang, Hunan, China
| | - Li Duan
- Guangdong Provincial Research Center for Artificial Intelligence and Digital Orthopedic Technology, Shenzhen Key Laboratory of Tissue Engineering, Shenzhen Laboratory of Digital Orthopedic Engineering, Department of Orthopedics, Shenzhen Second People's Hospital (The First Affiliated Hospital of Shenzhen University, Health Science Center), Shenzhen, Guangdong, China
| | - Hongxi Shang
- Guangdong Provincial Research Center for Artificial Intelligence and Digital Orthopedic Technology, Shenzhen Key Laboratory of Tissue Engineering, Shenzhen Laboratory of Digital Orthopedic Engineering, Department of Orthopedics, Shenzhen Second People's Hospital (The First Affiliated Hospital of Shenzhen University, Health Science Center), Shenzhen, Guangdong, China
| | - Daping Wang
- Guangdong Provincial Research Center for Artificial Intelligence and Digital Orthopedic Technology, Shenzhen Key Laboratory of Tissue Engineering, Shenzhen Laboratory of Digital Orthopedic Engineering, Department of Orthopedics, Shenzhen Second People's Hospital (The First Affiliated Hospital of Shenzhen University, Health Science Center), Shenzhen, Guangdong, China
| | - Jianyi Xiong
- Guangdong Provincial Research Center for Artificial Intelligence and Digital Orthopedic Technology, Shenzhen Key Laboratory of Tissue Engineering, Shenzhen Laboratory of Digital Orthopedic Engineering, Department of Orthopedics, Shenzhen Second People's Hospital (The First Affiliated Hospital of Shenzhen University, Health Science Center), Shenzhen, Guangdong, China
| |
Collapse
|
14
|
GAO X, SHEN S, NIU Q, MIAO W, HAN Y, HAO Z, AN N, YANG Y, ZHANG Y, ZHANG H, STOREY KB, CHANG H. Differential bone metabolism and protein expression in mice fed a high-fat diet versus Daurian ground squirrels following natural pre-hibernation fattening. J Zhejiang Univ Sci B 2022; 23:1042-1056. [PMID: 36518056 PMCID: PMC9758712 DOI: 10.1631/jzus.b2100798] [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: 12/24/2022]
Abstract
This study compared the effects on bone metabolism and morphology of pathological obesity induced by excessive fat intake in a non-hibernator (mice) versus healthy obesity due to pre-hibernation fattening in a hibernator (ground squirrels). Kunming mice were fed a high-fat diet to provide a model of pathological obesity (OB group). Daurian ground squirrels fattened naturally in their pre-hibernation season (PRE group) were used as a healthy obesity model. Micro-computed tomography (micro-CT) and three-point bending tests were used to determine the microstructure and mechanical properties of bone. Western blots were used to analyze protein expression levels related to bone metabolism (Runt-related transcription factor 2 (RunX2), osteocalcin (OCN), alkaline phosphatase (ALP), osteoprotegerin (OPG), receptor activator of nuclear factor-κB ligand (RANKL), cathepsin K, matrix metallopeptidase 9 (MMP9), patched protein homolog 1 (Ptch1), phosphorylated β-catenin (P-β-catenin), and glycogen synthase kinase-3β (GSK-3β)). Compared with controls, there was no obvious bone loss in the OB mice, and the stiffness of the femur was increased significantly. Compared with summer active squirrels, bone formation was enhanced but the mechanical properties did not change in the PRE group squirrels. In OB mice, western blots showed significantly increased expression levels of all proteins except RunX2, OPG, and Ptch1. PRE ground squirrels showed significantly increased expression of most proteins except OCN and Ptch1, which decreased significantly, and P-β-catenin and OPG, which did not change. In conclusion, for non-hibernating mice, moderate obesity had a certain protective effect on bones, demonstrating two-way regulation, increasing both bone loss and bone formation. For pre-hibernating ground squirrels, the healthy obesity acquired before hibernation had a positive effect on the microstructure of bones, and also enhanced the expression levels of proteins related to bone formation, bone resorption, and Wnt signaling.
Collapse
Affiliation(s)
- Xuli GAO
- Shaanxi Key Laboratory for Animal Conservation, Northwest University, Xi’an710069, China,Key Laboratory of Resource Biology and Biotechnology in Western China (College of Life Sciences, Northwest University), Ministry of Education, Xi’an710069, China
| | - Shenyang SHEN
- Key Laboratory of Resource Biology and Biotechnology in Western China (College of Life Sciences, Northwest University), Ministry of Education, Xi’an710069, China
| | - Qiaohua NIU
- Key Laboratory of Resource Biology and Biotechnology in Western China (College of Life Sciences, Northwest University), Ministry of Education, Xi’an710069, China
| | - Weilan MIAO
- Key Laboratory of Resource Biology and Biotechnology in Western China (College of Life Sciences, Northwest University), Ministry of Education, Xi’an710069, China
| | - Yuting HAN
- Key Laboratory of Resource Biology and Biotechnology in Western China (College of Life Sciences, Northwest University), Ministry of Education, Xi’an710069, China
| | - Ziwei HAO
- Key Laboratory of Resource Biology and Biotechnology in Western China (College of Life Sciences, Northwest University), Ministry of Education, Xi’an710069, China
| | - Ning AN
- Key Laboratory of Resource Biology and Biotechnology in Western China (College of Life Sciences, Northwest University), Ministry of Education, Xi’an710069, China
| | - Yingyu YANG
- Key Laboratory of Resource Biology and Biotechnology in Western China (College of Life Sciences, Northwest University), Ministry of Education, Xi’an710069, China
| | - Yu ZHANG
- Key Laboratory of Resource Biology and Biotechnology in Western China (College of Life Sciences, Northwest University), Ministry of Education, Xi’an710069, China
| | - Han ZHANG
- Key Laboratory of Resource Biology and Biotechnology in Western China (College of Life Sciences, Northwest University), Ministry of Education, Xi’an710069, China
| | - Kenneth B. STOREY
- Department of Biology, Carleton University, Ottawa, ON K1S 5B6, Canada
| | - Hui CHANG
- Shaanxi Key Laboratory for Animal Conservation, Northwest University, Xi’an710069, China,Key Laboratory of Resource Biology and Biotechnology in Western China (College of Life Sciences, Northwest University), Ministry of Education, Xi’an710069, China,Hui CHANG,
| |
Collapse
|
15
|
Vlashi R, Zhang X, Wu M, Chen G. Wnt signaling: essential roles in osteoblast differentiation, bone metabolism and therapeutic implications for bone and skeletal disorders. Genes Dis 2022. [DOI: 10.1016/j.gendis.2022.07.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/16/2022] Open
|
16
|
Thomas S, Jaganathan BG. Signaling network regulating osteogenesis in mesenchymal stem cells. J Cell Commun Signal 2022; 16:47-61. [PMID: 34236594 PMCID: PMC8688675 DOI: 10.1007/s12079-021-00635-1] [Citation(s) in RCA: 66] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Accepted: 06/30/2021] [Indexed: 02/06/2023] Open
Abstract
Osteogenesis is an important developmental event that results in bone formation. Bone forming cells or osteoblasts develop from mesenchymal stem cells (MSCs) through a highly controlled process regulated by several signaling pathways. The osteogenic lineage commitment of MSCs is controlled by cell-cell interactions, paracrine factors, mechanical signals, hormones, and cytokines present in their niche, which activate a plethora of signaling molecules belonging to bone morphogenetic proteins, Wnt, Hedgehog, and Notch signaling. These signaling pathways individually as well as in coordination with other signaling molecules, regulate the osteogenic lineage commitment of MSCs by activating several osteo-lineage specific transcription factors. Here, we discuss the key signaling pathways that regulate osteogenic differentiation of MSCs and the cross-talk between them during osteogenic differentiation. We also discuss how these signaling pathways can be modified for therapy for bone repair and regeneration.
Collapse
Affiliation(s)
- Sachin Thomas
- Stem Cells and Cancer Biology Research Group, Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati, Assam, 781039, India
| | - Bithiah Grace Jaganathan
- Stem Cells and Cancer Biology Research Group, Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati, Assam, 781039, India.
| |
Collapse
|
17
|
New Function of Cholesterol Oxidation Products Involved in Osteoporosis Pathogenesis. Int J Mol Sci 2022; 23:ijms23042020. [PMID: 35216140 PMCID: PMC8876989 DOI: 10.3390/ijms23042020] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2022] [Revised: 02/05/2022] [Accepted: 02/09/2022] [Indexed: 12/13/2022] Open
Abstract
Osteoporosis (OP) is a systemic bone disease characterized by decreased bone strength, microarchitectural changes in bone tissues, and increased risk of fracture. Its occurrence is closely related to various factors such as aging, genetic factors, living habits, and nutritional deficiencies as well as the disturbance of bone homeostasis. The dysregulation of bone metabolism is regarded as one of the key influencing factors causing OP. Cholesterol oxidation products (COPs) are important compounds in the maintenance of bone metabolic homeostasis by participating in several important biological processes such as the differentiation of mesenchymal stem cells, bone formation in osteoblasts, and bone resorption in osteoclasts. The effects of specific COPs on mesenchymal stem cells are mainly manifested by promoting osteoblast genesis and inhibiting adipocyte genesis. This review aims to elucidate the biological roles of COPs in OP development, starting from the molecular mechanisms of OP, pointing out opportunities and challenges in current research, and providing new ideas and perspectives for further studies of OP pathogenesis.
Collapse
|
18
|
Wnt/β-catenin signalling: function, biological mechanisms, and therapeutic opportunities. Signal Transduct Target Ther 2022; 7:3. [PMID: 34980884 PMCID: PMC8724284 DOI: 10.1038/s41392-021-00762-6] [Citation(s) in RCA: 1076] [Impact Index Per Article: 358.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Revised: 06/28/2021] [Accepted: 07/07/2021] [Indexed: 02/06/2023] Open
Abstract
The Wnt/β-catenin pathway comprises a family of proteins that play critical roles in embryonic development and adult tissue homeostasis. The deregulation of Wnt/β-catenin signalling often leads to various serious diseases, including cancer and non-cancer diseases. Although many articles have reviewed Wnt/β-catenin from various aspects, a systematic review encompassing the origin, composition, function, and clinical trials of the Wnt/β-catenin signalling pathway in tumour and diseases is lacking. In this article, we comprehensively review the Wnt/β-catenin pathway from the above five aspects in combination with the latest research. Finally, we propose challenges and opportunities for the development of small-molecular compounds targeting the Wnt signalling pathway in disease treatment.
Collapse
|
19
|
Kim SE, Robles-Lopez K, Cao X, Liu K, Chothani PJ, Bhavani N, Rahman L, Mukhopadhyay S, Wlodarczyk BJ, Finnell RH. Wnt1 Lineage Specific Deletion of Gpr161 Results in Embryonic Midbrain Malformation and Failure of Craniofacial Skeletal Development. Front Genet 2021; 12:761418. [PMID: 34887903 PMCID: PMC8650154 DOI: 10.3389/fgene.2021.761418] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2021] [Accepted: 11/04/2021] [Indexed: 11/13/2022] Open
Abstract
Sonic hedgehog (Shh) signaling regulates multiple morphogenetic processes during embryonic neurogenesis and craniofacial skeletal development. Gpr161 is a known negative regulator of Shh signaling. Nullizygous Gpr161 mice are embryonic lethal, presenting with structural defects involving the neural tube and the craniofacies. However, the lineage specific role of Gpr161 in later embryonic development has not been thoroughly investigated. We studied the Wnt1-Cre lineage specific role of Gpr161 during mouse embryonic development. We observed three major gross morphological phenotypes in Gpr161 cKO (Gpr161 f/f; Wnt1-Cre) fetuses; protrusive tectum defect, encephalocele, and craniofacial skeletal defect. The overall midbrain tissues were expanded and cell proliferation in ventricular zones of midbrain was increased in Gpr161 cKO fetuses, suggesting that protrusive tectal defects in Gpr161 cKO are secondary to the increased proliferation of midbrain neural progenitor cells. Shh signaling activity as well as upstream Wnt signaling activity were increased in midbrain tissues of Gpr161 cKO fetuses. RNA sequencing further suggested that genes in the Shh, Wnt, Fgf and Notch signaling pathways were differentially regulated in the midbrain of Gpr161 cKO fetuses. Finally, we determined that cranial neural crest derived craniofacial bone formation was significantly inhibited in Gpr161 cKO fetuses, which partly explains the development of encephalocele. Our results suggest that Gpr161 plays a distinct role in midbrain development and in the formation of the craniofacial skeleton during mouse embryogenesis.
Collapse
Affiliation(s)
- Sung-Eun Kim
- Department of Pediatrics, Dell Pediatric Research Institute, University of Texas at Austin Dell Medical School, Austin, TX, United States
| | - Karla Robles-Lopez
- Department of Pediatrics, Dell Pediatric Research Institute, University of Texas at Austin Dell Medical School, Austin, TX, United States.,Center for Precision Environmental Health, Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, United States
| | - Xuanye Cao
- Center for Precision Environmental Health, Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, United States
| | - Kristyn Liu
- Department of Pediatrics, Dell Pediatric Research Institute, University of Texas at Austin Dell Medical School, Austin, TX, United States
| | - Pooja J Chothani
- Department of Pediatrics, Dell Pediatric Research Institute, University of Texas at Austin Dell Medical School, Austin, TX, United States
| | - Nikitha Bhavani
- Department of Pediatrics, Dell Pediatric Research Institute, University of Texas at Austin Dell Medical School, Austin, TX, United States
| | - Lauren Rahman
- Department of Pediatrics, Dell Pediatric Research Institute, University of Texas at Austin Dell Medical School, Austin, TX, United States
| | - Saikat Mukhopadhyay
- Department of Cell Biology, University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - Bogdan J Wlodarczyk
- Center for Precision Environmental Health, Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, United States
| | - Richard H Finnell
- Department of Pediatrics, Dell Pediatric Research Institute, University of Texas at Austin Dell Medical School, Austin, TX, United States.,Center for Precision Environmental Health, Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, United States.,Departments of Molecular and Human Genetics and Medicine, Baylor College of Medicine, Houston, TX, United States
| |
Collapse
|
20
|
Cheng B, Jia Y, Wen Y, Hou W, Xu K, Liang C, Cheng S, Liu L, Chu X, Ye J, Yao Y, Zhang F, Xu P. Integrative Analysis of MicroRNA and mRNA Sequencing Data Identifies Novel Candidate Genes and Pathways for Developmental Dysplasia of Hip. Cartilage 2021; 13:1618S-1626S. [PMID: 33522290 PMCID: PMC8804775 DOI: 10.1177/1947603521990859] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
OBJECTIVE Our aim is to explore the candidate pathogenesis genes and pathways of developmental dysplasia of hip (DDH). DESIGN Proliferating primary chondrocytes from hip cartilage were used for total RNA extraction including 5 DDH patients and 5 neck of femur fracture (NOF) subjects. Genome-wide mRNA and microRNA (miRNA) were then sequenced on the Illumina platform (HiSeq2500). Limma package was used for difference analysis of mRNA expression profiles. edgeR was used for difference analysis of miRNA expression profiles. miRanda was used to predict miRNA-target genes. The overlapped DDH associated genes identified by mRNA and miRNA integrative analysis were further compared with the differently expressed genes in hip osteoarthritis (OA) cartilage. RESULTS Differential expression analysis identified 1,833 differently expressed mRNA and 186 differently expressed miRNA for DDH. Integrative analysis of mRNA and miRNA expression profiles identified 175 overlapped candidate genes (differentially expressed genes, DEGs) for DDH, such as VWA1, TMEM119, and SCUBE3. Further gene ontology enrichment analysis detected 111 candidate terms for DDH, such as skeletal system morphogenesis (P = 4.92 × 10-5) and skeletal system development (P = 8.85 × 10-5). Pathway enrichment analysis identified 14 candidate pathways for DDH, such as Hedgehog signaling pathway (P = 4.29 × 10-5) and Wnt signaling pathway (P = 4.42 × 10-2). Among the identified DDH associated candidate genes, we also found some genes were detected in hip OA including EFNA1 and VWA1. CONCLUSIONS We identified multiple novel candidate genes and pathways for DDH, providing novel clues for understanding the molecular mechanism of DDH.
Collapse
Affiliation(s)
- Bolun Cheng
- Key Laboratory of Trace Elements and
Endemic Diseases, Collaborative Innovation Center of Endemic Disease and Health
Promotion for Silk Road Region, School of Public Health, Health Science Center,
Xi’an Jiaotong University, Xi’an, People’s Republic of China
| | - Yumeng Jia
- Key Laboratory of Trace Elements and
Endemic Diseases, Collaborative Innovation Center of Endemic Disease and Health
Promotion for Silk Road Region, School of Public Health, Health Science Center,
Xi’an Jiaotong University, Xi’an, People’s Republic of China,Yumeng Jia, Key Laboratory of Trace Elements
and Endemic Diseases, Collaborative Innovation Center of Endemic Disease and
Health Promotion for Silk Road Region, School of Public Health, Health Science
Center, Xi’an Jiaotong University, No. 76 Yan Ta West Road, Xi’an, 710061,
People’s Republic of China.
| | - Yan Wen
- Key Laboratory of Trace Elements and
Endemic Diseases, Collaborative Innovation Center of Endemic Disease and Health
Promotion for Silk Road Region, School of Public Health, Health Science Center,
Xi’an Jiaotong University, Xi’an, People’s Republic of China
| | - Weikun Hou
- Department of Joint Surgery, Xi’an
Honghui Hospital, Xi’an Jiaotong University Health Science Center, Xi’an, People’s
Republic of China
| | - Ke Xu
- Department of Joint Surgery, Xi’an
Honghui Hospital, Xi’an Jiaotong University Health Science Center, Xi’an, People’s
Republic of China
| | - Chujun Liang
- Key Laboratory of Trace Elements and
Endemic Diseases, Collaborative Innovation Center of Endemic Disease and Health
Promotion for Silk Road Region, School of Public Health, Health Science Center,
Xi’an Jiaotong University, Xi’an, People’s Republic of China
| | - Shiqiang Cheng
- Key Laboratory of Trace Elements and
Endemic Diseases, Collaborative Innovation Center of Endemic Disease and Health
Promotion for Silk Road Region, School of Public Health, Health Science Center,
Xi’an Jiaotong University, Xi’an, People’s Republic of China
| | - Li Liu
- Key Laboratory of Trace Elements and
Endemic Diseases, Collaborative Innovation Center of Endemic Disease and Health
Promotion for Silk Road Region, School of Public Health, Health Science Center,
Xi’an Jiaotong University, Xi’an, People’s Republic of China
| | - Xiaomeng Chu
- Key Laboratory of Trace Elements and
Endemic Diseases, Collaborative Innovation Center of Endemic Disease and Health
Promotion for Silk Road Region, School of Public Health, Health Science Center,
Xi’an Jiaotong University, Xi’an, People’s Republic of China
| | - Jing Ye
- Key Laboratory of Trace Elements and
Endemic Diseases, Collaborative Innovation Center of Endemic Disease and Health
Promotion for Silk Road Region, School of Public Health, Health Science Center,
Xi’an Jiaotong University, Xi’an, People’s Republic of China
| | - Yao Yao
- Key Laboratory of Trace Elements and
Endemic Diseases, Collaborative Innovation Center of Endemic Disease and Health
Promotion for Silk Road Region, School of Public Health, Health Science Center,
Xi’an Jiaotong University, Xi’an, People’s Republic of China
| | - Feng Zhang
- Key Laboratory of Trace Elements and
Endemic Diseases, Collaborative Innovation Center of Endemic Disease and Health
Promotion for Silk Road Region, School of Public Health, Health Science Center,
Xi’an Jiaotong University, Xi’an, People’s Republic of China,Feng Zhang, Key Laboratory of Trace Elements
and Endemic Diseases, Collaborative Innovation Center of Endemic Disease and
Health Promotion for Silk Road Region, School of Public Health, Health Science
Center, Xi’an Jiaotong University, No. 76 Yan Ta West Road, Xi’an, 710061,
People’s Republic of China.
| | - Peng Xu
- Department of Joint Surgery, Xi’an
Honghui Hospital, Xi’an Jiaotong University Health Science Center, Xi’an, People’s
Republic of China,Peng Xu, Department of Joint Surgery, Xi’an
Honghui Hospital, Xi’an Jiaotong University Health Science Center, No. 555 You
Yi East Road, Xi’an, 710000, People’s Repubic of China.
| |
Collapse
|
21
|
Castiella T, Iruzubieta P, Monleón E, Cardiel MJ, Gómez-Vallejo J, Monzón M, Junquera MC. Stromal cells of giant cell tumor of bone show primary cilia in giant cell tumor of bone. Microsc Res Tech 2021; 85:1065-1074. [PMID: 34761465 DOI: 10.1002/jemt.23976] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Revised: 09/04/2021] [Accepted: 10/26/2021] [Indexed: 01/02/2023]
Abstract
Giant cell tumor of bone (GCTB) is a locally aggressive primary bone neoplasm composed by tumoral stromal cells (SCs) and a reactive component that consists of monocytic/histiocytic cells that give rise by fusion to osteoclast-like multinucleated cells. Recently, specific Histone 3.3 mutations have been demonstrated in SCs of GCTB. Many of the pathways related to bone proliferation and regulation depend on the primary cilium, a microtubule-based organelle that protrudes outside the cell and acts as a sensorial antenna. In the present work, we aimed to study the presence and role of primary cilia in GCTB. Ultrastructural, immunohistochemical, and immunofluorescence studies were performed in order to demonstrate, for the first time, that the primary cilium is located in spindle-shaped SCs of GCTB. Moreover, we showed Hedgehog (Hh) signaling pathway activation in these cells. Hence, primary cilia may play a relevant role in GCTB tumorogenesis through Hh signaling activation in SCs. RESEARCH HIGHLIGHTS: Transmission electron microscopy allows describing and differentiating cellular subpopulations in giant cell tumor of bone (GCTB). The primary cilium is present in some tumoral stromal cells of GCTB. Hedgehog signalling is activated in these cells.
Collapse
Affiliation(s)
- Tomás Castiella
- Department of Pathology, Hospital Clínico Universitario Lozano Blesa, Zaragoza, Spain.,Institute for Health Research Aragón (IIS), Zaragoza, Spain
| | - Pablo Iruzubieta
- Department of Human Anatomy and Histology, Faculty of Medicine, University of Zaragoza, Zaragoza, Spain
| | - Eva Monleón
- Department of Human Anatomy and Histology, Faculty of Medicine, University of Zaragoza, Zaragoza, Spain.,Institute for Health Research Aragón (IIS), Zaragoza, Spain
| | - Mª José Cardiel
- Department of Pathology, Hospital Clínico Universitario Lozano Blesa, Zaragoza, Spain
| | - Jesús Gómez-Vallejo
- Department of Traumatology and Orthopaedic Surgery, Hospital Clínico Universitario Lozano Blesa, Zaragoza, Spain
| | - Marta Monzón
- Department of Human Anatomy and Histology, Faculty of Medicine, University of Zaragoza, Zaragoza, Spain.,Institute for Health Research Aragón (IIS), Zaragoza, Spain
| | - Mª Concepción Junquera
- Department of Human Anatomy and Histology, Faculty of Medicine, University of Zaragoza, Zaragoza, Spain.,Institute for Health Research Aragón (IIS), Zaragoza, Spain
| |
Collapse
|
22
|
Qiao L, Liu X, He Y, Zhang J, Huang H, Bian W, Chilufya MM, Zhao Y, Han J. Progress of Signaling Pathways, Stress Pathways and Epigenetics in the Pathogenesis of Skeletal Fluorosis. Int J Mol Sci 2021; 22:ijms222111932. [PMID: 34769367 PMCID: PMC8584317 DOI: 10.3390/ijms222111932] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Revised: 10/27/2021] [Accepted: 10/28/2021] [Indexed: 12/12/2022] Open
Abstract
Fluorine is widely dispersed in nature and has multiple physiological functions. Although it is usually regarded as an essential trace element for humans, this view is not held universally. Moreover, chronic fluorosis, mainly characterized by skeletal fluorosis, can be induced by long-term excessive fluoride consumption. High concentrations of fluoride in the environment and drinking water are major causes, and patients with skeletal fluorosis mainly present with symptoms of osteosclerosis, osteochondrosis, osteoporosis, and degenerative changes in joint cartilage. Etiologies for skeletal fluorosis have been established, but the specific pathogenesis is inconclusive. Currently, active osteogenesis and accelerated bone turnover are considered critical processes in the progression of skeletal fluorosis. In recent years, researchers have conducted extensive studies in fields of signaling pathways (Wnt/β-catenin, Notch, PI3K/Akt/mTOR, Hedgehog, parathyroid hormone, and insulin signaling pathways), stress pathways (oxidative stress and endoplasmic reticulum stress pathways), epigenetics (DNA methylation and non-coding RNAs), and their inter-regulation involved in the pathogenesis of skeletal fluorosis. In this review, we summarised and analyzed relevant findings to provide a basis for comprehensive understandings of the pathogenesis of skeletal fluorosis and hopefully propose more effective prevention and therapeutic strategies.
Collapse
|
23
|
Kirankumar S, Gurusamy N, Rajasingh S, Sigamani V, Vasanthan J, Perales SG, Rajasingh J. Modern approaches on stem cells and scaffolding technology for osteogenic differentiation and regeneration. Exp Biol Med (Maywood) 2021; 247:433-445. [PMID: 34648374 DOI: 10.1177/15353702211052927] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
The process of bone repair has always been a natural mystery. Although bones do repair themselves, supplemental treatment is required for the initiation of the self-regeneration process. Predominantly, surgical procedures are employed for bone regeneration. Recently, cell-based therapy for bone regeneration has proven to be more effective than traditional methods, as it eliminates the immune risk and painful surgeries. In clinical trials, various stem cells, especially mesenchymal stem cells, have shown to be more efficient for the treatment of several bone-related diseases, such as non-union fracture, osteogenesis imperfecta, osteosarcoma, and osteoporosis. Furthermore, the stem cells grown in a suitable three-dimensional scaffold support were found to be more efficient for osteogenesis. It has been shown that the three-dimensional bioscaffolds support and simulate an in vivo environment, which helps in differentiation of stem cells into bone cells. Bone regeneration in patients with bone disorders can be improved through modification of stem cells with several osteogenic factors or using stem cells as carriers for osteogenic factors. In this review, we focused on the various types of stem cells and scaffolds that are being used for bone regeneration. In addition, the molecular mechanisms of various transcription factors, signaling pathways that support bone regeneration and the senescence of the stem cells, which limits bone regeneration, have been discussed.
Collapse
Affiliation(s)
- Shivaani Kirankumar
- Department of Bioscience Research, University of Tennessee Health Science Center, Memphis, TN 38163, USA.,Department of Genetic Engineering, 93104SRM Institute of Science and Technology, Chennai 603203, India
| | - Narasimman Gurusamy
- Department of Bioscience Research, University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | - Sheeja Rajasingh
- Department of Bioscience Research, University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | - Vinoth Sigamani
- Department of Bioscience Research, University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | - Jayavardini Vasanthan
- Department of Bioscience Research, University of Tennessee Health Science Center, Memphis, TN 38163, USA.,Department of Genetic Engineering, 93104SRM Institute of Science and Technology, Chennai 603203, India
| | - Selene G Perales
- Department of Bioscience Research, University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | - Johnson Rajasingh
- Department of Bioscience Research, University of Tennessee Health Science Center, Memphis, TN 38163, USA.,Department of Medicine, University of Tennessee Health Science Center, Memphis, TN 38163, USA.,Department of Microbiology, Immunology and Biochemistry, University of Tennessee Health Science Center, Memphis, TN 38163, USA
| |
Collapse
|
24
|
Primary cilia in hard tissue development and diseases. Front Med 2021; 15:657-678. [PMID: 34515939 DOI: 10.1007/s11684-021-0829-6] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2020] [Accepted: 10/13/2020] [Indexed: 10/20/2022]
Abstract
Bone and teeth are hard tissues. Hard tissue diseases have a serious effect on human survival and quality of life. Primary cilia are protrusions on the surfaces of cells. As antennas, they are distributed on the membrane surfaces of almost all mammalian cell types and participate in the development of organs and the maintenance of homeostasis. Mutations in cilium-related genes result in a variety of developmental and even lethal diseases. Patients with multiple ciliary gene mutations present overt changes in the skeletal system, suggesting that primary cilia are involved in hard tissue development and reconstruction. Furthermore, primary cilia act as sensors of external stimuli and regulate bone homeostasis. Specifically, substances are trafficked through primary cilia by intraflagellar transport, which affects key signaling pathways during hard tissue development. In this review, we summarize the roles of primary cilia in long bone development and remodeling from two perspectives: primary cilia signaling and sensory mechanisms. In addition, the cilium-related diseases of hard tissue and the manifestations of mutant cilia in the skeleton and teeth are described. We believe that all the findings will help with the intervention and treatment of related hard tissue genetic diseases.
Collapse
|
25
|
Comparative genomics provides insights into the aquatic adaptations of mammals. Proc Natl Acad Sci U S A 2021; 118:2106080118. [PMID: 34503999 PMCID: PMC8449357 DOI: 10.1073/pnas.2106080118] [Citation(s) in RCA: 50] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/26/2021] [Indexed: 12/30/2022] Open
Abstract
Divergent lineages can respond to common environmental factors through convergent processes involving shared genomic components or pathways, but the molecular mechanisms are poorly understood. Here, we provide genomic resources and insights into the evolution of mammalian lineages adapting to aquatic life. Our data suggest convergent evolution, for example, in association with thermoregulation through genes associated with a surface heat barrier (NFIA) and internal heat exchange (SEMA3E). Combined with the support of previous reports showing that the UCP1 locus has been lost in many marine mammals independently, our results suggest that the thermostatic strategy of marine mammals shifted from enhancing heat production to limiting heat loss. The ancestors of marine mammals once roamed the land and independently committed to an aquatic lifestyle. These macroevolutionary transitions have intrigued scientists for centuries. Here, we generated high-quality genome assemblies of 17 marine mammals (11 cetaceans and six pinnipeds), including eight assemblies at the chromosome level. Incorporating previously published data, we reconstructed the marine mammal phylogeny and population histories and identified numerous idiosyncratic and convergent genomic variations that possibly contributed to the transition from land to water in marine mammal lineages. Genes associated with the formation of blubber (NFIA), vascular development (SEMA3E), and heat production by brown adipose tissue (UCP1) had unique changes that may contribute to marine mammal thermoregulation. We also observed many lineage-specific changes in the marine mammals, including genes associated with deep diving and navigation. Our study advances understanding of the timing, pattern, and molecular changes associated with the evolution of mammalian lineages adapting to aquatic life.
Collapse
|
26
|
Clayton ZS, Hauffe L, Liu C, Kern M, Hong MY, Brasser SM, Hooshmand S. Chronic ethanol consumption does not reduce true bone density in male Wistar rats. Alcohol 2021; 93:17-23. [PMID: 33662519 DOI: 10.1016/j.alcohol.2021.02.003] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2020] [Revised: 02/04/2021] [Accepted: 02/19/2021] [Indexed: 12/01/2022]
Abstract
Osteoporosis is characterized by reduced bone mineral density (BMD) and increased bone fragility, which may be modified by lifestyle behaviors. In observational studies, chronic moderate ethanol consumption is associated with higher BMD, but results are inconsistent and underlying mechanisms are unknown. To understand the influence of chronic ethanol consumption on true bone density (Archimedes principal), bone mechanical properties (Young's Modulus of bend), and osteogenic gene expression, 12-month-old male Wistar rats were randomly assigned to a control group or ethanol intervention (20% ethanol in drinking water on alternate days) group for 13 weeks and tibiae and femurs were collected. Blood was collected to assess alcohol content and antioxidant enzyme activities. We hypothesized that chronic ethanol consumption would increase true bone density and mechanical properties and increase osteoblastic gene expression and serum antioxidant enzyme activity. Ethanol consumption did not influence femoral or tibial true bone density but did result in lower tibial Young's modulus of bend (p = 0.0002). However, there was no influence of ethanol on other measures of mechanical properties. Femoral pro-osteoclastic gene expression of Dkk1 was lower (p = 0.0006) and pro-osteoblastic gene expression of Ctnnb1 was higher (p = 0.02) with ethanol consumption. We observed no differences in circulating antioxidant activities between groups, other than a tendency for greater (p = 0.08) glutathione peroxidase in the ethanol group. Results showed chronic ethanol consumption did not influence true bone density, only modestly reduced tibial mechanical properties (lower Young's modulus of bend), and moderately impacted expression of genes within the femur known to regulate both osteoblast and osteoclast activities.
Collapse
Affiliation(s)
- Zachary S Clayton
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO, United States
| | - Laura Hauffe
- Institute of Nutrition, Friedrich Schiller University Jena, Jena, Germany
| | - Changqi Liu
- School of Exercise and Nutritional Sciences, San Diego State University, San Diego, CA, United States
| | - Mark Kern
- School of Exercise and Nutritional Sciences, San Diego State University, San Diego, CA, United States
| | - Mee Young Hong
- School of Exercise and Nutritional Sciences, San Diego State University, San Diego, CA, United States
| | - Susan M Brasser
- Department of Psychology, San Diego State University, San Diego, CA, United States
| | - Shirin Hooshmand
- School of Exercise and Nutritional Sciences, San Diego State University, San Diego, CA, United States.
| |
Collapse
|
27
|
Ya J, Xu Y, Wang G, Zhao H. Cadmium induced skeletal underdevelopment, liver cell apoptosis and hepatic energy metabolism disorder in Bufo gargarizans larvae by disrupting thyroid hormone signaling. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2021; 211:111957. [PMID: 33493726 DOI: 10.1016/j.ecoenv.2021.111957] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Revised: 01/14/2021] [Accepted: 01/16/2021] [Indexed: 06/12/2023]
Abstract
Cadmium (Cd) is hazardous to human health and it is also highly detrimental to amphibian life. In this study, Bufo gargarizans larvae were exposed to environmentally relevant Cd concentrations of 5, 100 and 200 μg L-1 from Gosner stage (Gs) 26 to Gs 42 of metamorphic climax about 6 weeks. The results showed thyroid structural injuries and thyroid signaling disruption were induced by high Cd exposure (100 and 200 μg L-1). Moreover, tadpole skeleton including whole body, vertebrata, forelimb and hindlimb was developmentally delayed by high Cd exposure through downregulating the mRNA expressions of genes involved with skeletal ossification and growth pathway. Moreover, liver histopathological injuries were caused by high Cd exposure featured by hepatocytes malformation, nuclear degeneration and increasing melanomacrophage centers. Meanwhile, liver apoptosis rate showed on the rise in a dose-dependent way and Cd stimulated liver apoptosis by upregulating mRNA expressions of genes related to extrinsic and intrinsic apoptosis pathways. Furthermore, high Cd caused hepatic glucometabolism disorder by decreasing the genetic expressions associated with glycolysis and mitochondrial oxidative phosphorylation. In addition, liver lipid metabolism was disrupted by high Cd exposure through downregulating mRNA levels of genes related to fatty oxidation and upregulating mRNA levels of genes related to fatty acid synthesis. We suggested that Cd did great harm to tadpole health by disturbing thyroid function, skeletal growth, liver cell apoptosis signaling and hepatic energy metabolism pathway.
Collapse
Affiliation(s)
- Jing Ya
- College of Life Sciences, Shaanxi Normal University, Xi'an 710119, China
| | - Yifan Xu
- College of Life Sciences, Shaanxi Normal University, Xi'an 710119, China; AP Center, Changzhou Senior High School of Jiangsu Province, Changzhou 213000, China
| | - Gang Wang
- College of Life Sciences, Shaanxi Normal University, Xi'an 710119, China; AP Center, Changzhou Senior High School of Jiangsu Province, Changzhou 213000, China
| | - Hongfeng Zhao
- College of Life Sciences, Shaanxi Normal University, Xi'an 710119, China.
| |
Collapse
|
28
|
Tompkins YH, Su S, Velleman SG, Kim WK. Effects of 20(S)-hydroxycholesterol on satellite cell proliferation and differentiation of broilers. Poult Sci 2021; 100:474-481. [PMID: 33518099 PMCID: PMC7858162 DOI: 10.1016/j.psj.2020.10.032] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2020] [Revised: 09/14/2020] [Accepted: 10/19/2020] [Indexed: 01/30/2023] Open
Abstract
In the modern poultry industry, with increasing product demand, muscle growth rate and meat yield in chickens have tremendously changed. Understanding the regulation of muscle development is important to maintain efficient growth and development in meat-type chickens. 20(S)-hydroxycholesterol (20S) is known as one of the naturally occurring osteogenic cholesterol derivatives due to its ability to induce osteogenic differentiation; however, no studies have evaluated myogenic response to 20S in chicken muscle cells. To determine the use of 20S in vitro for the proliferation and differentiation of chicken satellite cells, satellite cells were isolated from pectoralis major muscle of 4-week-old Ross 708 male chickens and subjected to 0.25, 0.5, and 1.0 μmol of 20S during their proliferation and differentiation stages. Cell proliferation and differentiation were measured every 24 h for 72 h by determining DNA concentration, the activity of creatine kinase, and the expressions of myogenic regulatory transcription factors. Together these results suggested that a lower concentration of 20S did not affect myogenesis but a high concentration of 1.0 μmol 20S can negatively affect proliferation and differentiation in chicken satellite cells.
Collapse
Affiliation(s)
- Yuguo H Tompkins
- Department of Poultry Science, University of Georgia, Athens, USA
| | - Shengchen Su
- Department of Poultry Science, University of Georgia, Athens, USA
| | - Sandra G Velleman
- The Ohio State University, Ohio Agricultural Research and Development Center, Wooster, USA
| | - Woo Kyun Kim
- Department of Poultry Science, University of Georgia, Athens, USA.
| |
Collapse
|
29
|
Brewer N, Fong JT, Zhang D, Ramaswamy G, Shore EM. Gnas Inactivation Alters Subcutaneous Tissues in Progression to Heterotopic Ossification. Front Genet 2021; 12:633206. [PMID: 33574833 PMCID: PMC7870717 DOI: 10.3389/fgene.2021.633206] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2020] [Accepted: 01/04/2021] [Indexed: 11/13/2022] Open
Abstract
Heterotopic ossification (HO), the formation of bone outside of the skeleton, occurs in response to severe trauma and in rare genetic diseases such as progressive osseous heteroplasia (POH). In POH, which is caused by inactivation of GNAS, a gene that encodes the alpha stimulatory subunit of G proteins (Gsα), HO typically initiates within subcutaneous soft tissues before progressing to deeper connective tissues. To mimic POH, we used conditional Gnas-null mice which form HO in subcutaneous tissues upon Gnas inactivation. In response to Gnas inactivation, we determined that prior to detection of heterotopic bone, dermal adipose tissue changed dramatically, with progressively decreased adipose tissue volume and increased density of extracellular matrix over time. Upon depletion of the adipose tissue, heterotopic bone progressively formed in those locations. To investigate the potential relevance of the tissue microenvironment for HO formation, we implanted Gnas-null or control mesenchymal progenitor cells into Gnas-null or control host subcutaneous tissues. We found that mutant cells in a Gnas-null tissue environment induced a robust HO response while little/no HO was detected in control hosts. Additionally, a Gnas-null tissue environment appeared to support the recruitment of control cells to heterotopic bone, although control cell implants were associated with less HO formation compared to mutant cells. Our data support that Gnas inactivation alters the tissue microenvironment to influence mutant and wild-type progenitor cells to contribute to HO formation.
Collapse
Affiliation(s)
- Niambi Brewer
- Department of Orthopedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States.,Center for Research in FOP and Related Disorders, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States.,Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - John T Fong
- Department of Orthopedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States.,Center for Research in FOP and Related Disorders, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Deyu Zhang
- Department of Orthopedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States.,Center for Research in FOP and Related Disorders, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Girish Ramaswamy
- Department of Orthopedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States.,Center for Research in FOP and Related Disorders, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Eileen M Shore
- Department of Orthopedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States.,Center for Research in FOP and Related Disorders, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States.,Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| |
Collapse
|
30
|
Expression Analysis of Susceptibility Genes for Ossification of the Posterior Longitudinal Ligament of the Cervical Spine in Human OPLL-related Tissues and a Spinal Hyperostotic Mouse (ttw/ttw). Spine (Phila Pa 1976) 2020; 45:E1460-E1468. [PMID: 32756283 DOI: 10.1097/brs.0000000000003648] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
STUDY DESIGN Immunohistochemical and real-time reverse transcription-polymerase chain reaction (RT-PCR) analysis. OBJECTIVE The aim of this study was to analyze the expression of five susceptibility genes (RSPO2, HAO1, CCDC91, RHPH9, and STK38L) for human ossification of the posterior longitudinal ligaments (OPLL) identified in a genome-wide association study. SUMMARY OF BACKGROUND DATA Detailed expression and functional studies for the five susceptibility genes are needed to aid in clarification of the etiology and pathogenesis of OPLL. METHODS Immunostaining, cell culture, and real-time RT-PCR were performed on ossified ligament samples collected during anterior cervical decompression for symptomatic OPLL (n = 39 patients) and on control non-OPLL samples (n = 8 patients). Immunohistochemical analysis in spinal hyperostotic mice (ttw/ttw) (n = 25) was also performed. The sample sections were stained for RSPO2, HAO1, CCDC91, RHPH9, STK38L, Runx2, Sox9, and CD90. The mRNA expression levels of the five susceptibility genes were also analyzed in cultured human OPLL and non-OPLL cells subjected to cyclic tensile strain. RESULTS Immunoreactivity for RSPO2 and Sox9 was evident in proliferating chondrocytes in human OPLL tissues and ttw/ttw mice. Application of cyclic tensile strain to cultured human OPLL cells resulted in increases in mRNA levels for RSPO2, HAO1, and CCDC91. However, individual differences in expression in human OPLL-related samples were seen. HAO1-positive cells were detected only in 3- to 6-week-old ttw/ttw mice that did not simultaneously express RSPO2-positive samples. CONCLUSION Among the five susceptibility genes, RSPO2, HAO1, and CCDC91 might be contributory factors in progression of OPLL. RSPO2 may be involved in endochondral ossification, especially in mixed or continuous type OPLL, HAO1 may be an initiation factor for OPLL that is rarely seen in mature human OPLL samples, and CCDC91 may be associated with progression of ossification caused by mechanical stress. These findings provide important insights into the pathogenesis and therapeutic targets for OPLL. LEVEL OF EVIDENCE N/A.
Collapse
|
31
|
Chua K, Virshup DM, Odono EG, Chang KTE, Tan NJH, Hue SSS, Sim AYL, Lee VKM. YJ5 as an immunohistochemical marker of osteogenic lineage. Pathology 2020; 53:229-238. [PMID: 33187685 DOI: 10.1016/j.pathol.2020.07.017] [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: 02/13/2020] [Revised: 07/21/2020] [Accepted: 07/29/2020] [Indexed: 10/23/2022]
Abstract
Overexpression of WLS, an upstream protein in the Wnt pathway, has been implicated in several non-osteogenic tumours. This study represents the first attempt at evaluating WLS expression in various bone and soft tissue tumours using YJ5, a monoclonal antibody specific to WLS, with the aim of elucidating its utility in discerning tumours with aberrant Wnt signalling and as a marker of osteogenic lineage in challenging cases. Tumour tissue sections of 144 bone mass lesions and 63 soft tissue mass lesions were immunostained with the YJ5 antibody following standardised protocols. Subsequent assessment of immunoreactivity segregated cases into one of three groups: absent/weak, moderate, or strong YJ5 immunoreactivity. For the bone tumours, strong YJ5 immunoreactivity was seen in almost all osteosarcomas and chondroblastomas, all osteoblastomas and osteoid osteomas. In contrast, all other cartilaginous tumours, chordomas, aneurysmal bone cysts, chondromyxoid fibromas, most fibrous dysplasias and most giant cell tumours exhibited absent/weak YJ5 immunostaining. For the soft tissue tumours, a more heterogeneous pattern of YJ5 immunoreactivity was observed. Because diffuse and strong YJ5 expression is identified in almost all benign and malignant bone tumours with osteoblastic activity, it can be potentially utilised as an immunohistochemical marker to support osteogenic lineage. If interpreted in the appropriate context, this marker is useful in determining whether a malignant bone tumour is an osteosarcoma, particularly in those subtypes with no or minimal osteoid or unusual morphological features. This marker can also complement SATB2 to denote osteogenic lineage.
Collapse
Affiliation(s)
- Kenon Chua
- Department of Orthopaedic Surgery, Singapore General Hospital, Singapore; Programme in Cancer and Stem Cell Biology, Duke-NUS Medical School, Singapore
| | - David M Virshup
- Programme in Cancer and Stem Cell Biology, Duke-NUS Medical School, Singapore
| | - Eugene G Odono
- Department of Pathology, College of Medicine, University of the Philippines, Manila, Philippines
| | - Kenneth Tou En Chang
- Department of Pathology and Laboratory Medicine, KK Women's and Children's Hospital, Singapore
| | - Nicholas Jin Hong Tan
- Department of Pathology, National University Hospital, National University Health System, Singapore
| | - Susan Swee-Shan Hue
- Department of Pathology, NUH Advance Molecular Pathology Laboratory, Institute of Molecular and Cellular Biology, Singapore
| | - Arthur Yi Loong Sim
- Department of Pathology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | | |
Collapse
|
32
|
In JG, Yin J, Atanga R, Doucet M, Cole RN, DeVine L, Donowitz M, Zachos NC, Blutt SE, Estes MK, Kovbasnjuk O. Epithelial WNT2B and Desert Hedgehog Are Necessary for Human Colonoid Regeneration after Bacterial Cytotoxin Injury. iScience 2020; 23:101618. [PMID: 33089106 PMCID: PMC7559866 DOI: 10.1016/j.isci.2020.101618] [Citation(s) in RCA: 4] [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: 01/31/2020] [Revised: 06/03/2020] [Accepted: 09/24/2020] [Indexed: 01/09/2023] Open
Abstract
Intestinal regeneration and crypt hyperplasia after radiation or pathogen injury relies on Wnt signaling to stimulate stem cell proliferation. Mesenchymal Wnts are essential for homeostasis and regeneration in mice, but the role of epithelial Wnts remains largely uncharacterized. Using the enterohemorrhagic E. coli-secreted cytotoxin EspP to induce injury to human colonoids, we evaluated a simplified, epithelial regeneration model that lacks mesenchymal Wnts. Here, we demonstrate that epithelial-produced WNT2B is upregulated following injury and essential for regeneration. Hedgehog signaling, specifically activation via the ligand Desert Hedgehog (DHH), but not Indian or Sonic Hedgehog, is another driver of regeneration and modulates WNT2B expression. These findings highlight the importance of epithelial WNT2B and DHH in regulating human colonic regeneration after injury.
Collapse
Affiliation(s)
- Julie G. In
- Department of Internal Medicine, Division of Gastroenterology and Hepatology, University of New Mexico, Albuquerque, NM 87131, USA
- Department of Medicine, Division of Gastroenterology and Hepatology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Jianyi Yin
- Department of Medicine, Division of Gastroenterology and Hepatology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Roger Atanga
- Department of Internal Medicine, Division of Gastroenterology and Hepatology, University of New Mexico, Albuquerque, NM 87131, USA
| | - Michele Doucet
- Department of Medicine, Division of Gastroenterology and Hepatology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Robert N. Cole
- Department of Biological Chemistry, Mass Spectrometry and Proteomics Facility, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Lauren DeVine
- Department of Biological Chemistry, Mass Spectrometry and Proteomics Facility, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Mark Donowitz
- Department of Medicine, Division of Gastroenterology and Hepatology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Nicholas C. Zachos
- Department of Medicine, Division of Gastroenterology and Hepatology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Sarah E. Blutt
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Mary K. Estes
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX 77030, USA
- Department of Medicine, Section of Gastroenterology and Hepatology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Olga Kovbasnjuk
- Department of Internal Medicine, Division of Gastroenterology and Hepatology, University of New Mexico, Albuquerque, NM 87131, USA
- Department of Medicine, Division of Gastroenterology and Hepatology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| |
Collapse
|
33
|
Kaempferol promotes proliferation and osteogenic differentiation of periodontal ligament stem cells via Wnt/β-catenin signaling pathway. Life Sci 2020; 258:118143. [DOI: 10.1016/j.lfs.2020.118143] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Accepted: 07/21/2020] [Indexed: 12/20/2022]
|
34
|
Ko FC, Sumner DR. How faithfully does intramembranous bone regeneration recapitulate embryonic skeletal development? Dev Dyn 2020; 250:377-392. [PMID: 32813296 DOI: 10.1002/dvdy.240] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2020] [Revised: 07/19/2020] [Accepted: 08/13/2020] [Indexed: 02/06/2023] Open
Abstract
Postnatal intramembranous bone regeneration plays an important role during a wide variety of musculoskeletal regeneration processes such as fracture healing, joint replacement and dental implant surgery, distraction osteogenesis, stress fracture healing, and repair of skeletal defects caused by trauma or resection of tumors. The molecular basis of intramembranous bone regeneration has been interrogated using rodent models of most of these conditions. These studies reveal that signaling pathways such as Wnt, TGFβ/BMP, FGF, VEGF, and Notch are invoked, reminiscent of embryonic development of membranous bone. Discoveries of several skeletal stem cell/progenitor populations using mouse genetic models also reveal the potential sources of postnatal intramembranous bone regeneration. The purpose of this review is to compare the underlying molecular signals and progenitor cells that characterize embryonic development of membranous bone and postnatal intramembranous bone regeneration.
Collapse
Affiliation(s)
- Frank C Ko
- Department of Cell & Molecular Medicine, Rush University Medical Center, Chicago, Illinois, USA
| | - D Rick Sumner
- Department of Cell & Molecular Medicine, Rush University Medical Center, Chicago, Illinois, USA
| |
Collapse
|
35
|
Salhotra A, Shah HN, Levi B, Longaker MT. Mechanisms of bone development and repair. Nat Rev Mol Cell Biol 2020; 21:696-711. [PMID: 32901139 DOI: 10.1038/s41580-020-00279-w] [Citation(s) in RCA: 607] [Impact Index Per Article: 121.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/23/2020] [Indexed: 12/19/2022]
Abstract
Bone development occurs through a series of synchronous events that result in the formation of the body scaffold. The repair potential of bone and its surrounding microenvironment - including inflammatory, endothelial and Schwann cells - persists throughout adulthood, enabling restoration of tissue to its homeostatic functional state. The isolation of a single skeletal stem cell population through cell surface markers and the development of single-cell technologies are enabling precise elucidation of cellular activity and fate during bone repair by providing key insights into the mechanisms that maintain and regenerate bone during homeostasis and repair. Increased understanding of bone development, as well as normal and aberrant bone repair, has important therapeutic implications for the treatment of bone disease and ageing-related degeneration.
Collapse
Affiliation(s)
- Ankit Salhotra
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, CA, USA.,Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Harsh N Shah
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, CA, USA.,Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Benjamin Levi
- Department of Surgery, University of Michigan, Ann Arbor, MI, USA.
| | - Michael T Longaker
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, CA, USA. .,Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA, USA.
| |
Collapse
|
36
|
Gao R, Shi C, Yang C, Zhao Y, Chen X, Zhou X. Cyclic stretch promotes the ossification of ligamentum flavum by modulating the Indian hedgehog signaling pathway. Mol Med Rep 2020; 22:1119-1128. [PMID: 32626952 PMCID: PMC7339599 DOI: 10.3892/mmr.2020.11200] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2019] [Accepted: 01/29/2020] [Indexed: 11/06/2022] Open
Abstract
The Indian hedgehog (IHH) signaling pathway is an important pathway for bone growth and development. The aim of the present study was to examine the role of the IHH signaling pathway in the development of the ossification of ligamentum flavum (OLF) at the cellular and tissue levels. The expression levels and localization of the osteogenic genes Runt-related transcription factor 2 (RUNX2), Osterix, alkaline phosphatase (ALP), osteocalcin (OCN) and IHH were evaluated in OLF tissues by reverse transcription-quantitative PCR (RT-qPCR) and immunohistochemistry. Non-ossified ligamentum flavum (LF) sections were used as control samples. The tissue explant method was used to obtain cultured LF cells. In addition, OLF cells were subjected to cyclic stretch application for 0, 6, 12 or 24 h. The expression levels of osteogenic genes, and the IHH signaling pathway genes IHH, Smoothened (SMO), GLI family zinc finger 1 (GLI1), GLI2 and GLI3 were evaluated with RT-qPCR and western blotting. Osteogenic differentiation was further evaluated by assessing ALP activity and staining. Moreover, the effect of cyclopamine (Cpn), an IHH signaling inhibitor, on osteogenic differentiation was examined. The RT-qPCR and immunohistochemical results indicated that the mRNA and protein expression levels of RUNX2, Osterix, ALP, OCN and IHH were significantly higher in the OLF group compared with the LF group. Furthermore, application of cyclic stretch to OLF cells resulted in greater ALP activity, and significant increases in mRNA and protein expression levels of RUNX2, Osterix, ALP and OCN in a time-d00ependent manner. Cyclic stretch application also led to significant increases in IHH signaling pathway genes, including IHH, SMO, GLI1 and GLI2, while no significant effect was found on GLI3 expression level. In addition, it was found that Cpn significantly reversed the effect of cyclic stretch on the ALP activity, and the expression levels of RUNX2, Osterix, ALP, OCN, GLI1 and GLI2. Collectively, the present results suggested that the IHH signaling pathway may mediate the effect of cyclic stretch on the OLF cells.
Collapse
Affiliation(s)
- Rui Gao
- Department of Orthopedics, Changzheng Hospital, Second Military Medical University, Shanghai 200003, P.R. China
| | - Changgui Shi
- Department of Orthopedics, Changzheng Hospital, Second Military Medical University, Shanghai 200003, P.R. China
| | - Chengwei Yang
- Department of Orthopedics, Lanzhou General Hospital of PLA, Lanzhou, Gansu 730050, P.R. China
| | - Yin Zhao
- Department of Orthopedics, Changzheng Hospital, Second Military Medical University, Shanghai 200003, P.R. China
| | - Xiongsheng Chen
- Department of Orthopedics, Changzheng Hospital, Second Military Medical University, Shanghai 200003, P.R. China
| | - Xuhui Zhou
- Department of Orthopedics, Changzheng Hospital, Second Military Medical University, Shanghai 200003, P.R. China
| |
Collapse
|
37
|
Hedgehog Activation Regulates Human Osteoblastogenesis. Stem Cell Reports 2020; 15:125-139. [PMID: 32531191 PMCID: PMC7363748 DOI: 10.1016/j.stemcr.2020.05.008] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2020] [Revised: 05/12/2020] [Accepted: 05/13/2020] [Indexed: 12/14/2022] Open
Abstract
Two genetic diseases, Gorlin syndrome and McCune-Albright syndrome (MAS), show completely opposite symptoms in terms of bone mineral density and hedgehog (Hh) activity. In this study, we utilized human induced pluripotent stem cell (iPSC)-based models of the two diseases to understand the roles of Hh signaling in osteogenesis. Gorlin syndrome-derived iPSCs showed increased osteoblastogenesis and mineralization with Hh signaling activation and upregulation of a set of transcription factors in an osteogenic culture, compared with the isogenic control. MAS-specific iPSCs showed poor mineralization with low Hh signaling activity in the osteogenic culture; impaired osteoblastogenesis was restored to the normal level by treatment with an Hh signaling-activating small molecule. These data suggest that Hh signaling is a key controller for differentiation of osteoblasts from precursors. This study may pave a path to new drug therapies for genetic abnormalities in calcification caused by dysregulation of Hh signaling. iPSCs from patients with Gorlin syndrome showed enhancement of osteoblastogenesis Distinct transcription factors, including FOXO1 were induced in Gorlin iPSCs McCune-Albright syndrome-specific iPSCs demonstrated a decrease in Hh activity SAG treatment rescued immature calcification in MAS-specific iPSCs
Collapse
|
38
|
Sugita D, Nakajima H, Kokubo Y, Takeura N, Yayama T, Matsumine A. Cyclic tensile strain facilitates ossification of the cervical posterior longitudinal ligament via increased Indian hedgehog signaling. Sci Rep 2020; 10:7231. [PMID: 32350355 PMCID: PMC7190672 DOI: 10.1038/s41598-020-64304-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2019] [Accepted: 04/15/2020] [Indexed: 11/29/2022] Open
Abstract
The pathomechanisms of initiation and progression of ossification of the posterior longitudinal ligament (OPLL) are unclear. Indian hedgehog (Ihh) and related signaling molecules are key factors in normal enchondral ossification. The purpose of this study is to investigate the contribution of mechanical strain to OPLL and the relationship of Ihh with OPLL. Sections of the posterior longitudinal ligament (PLL) were obtained from 49 patients with OPLL and from 7 patients without OPLL. Cultured PLL cells were subjected to 24 hours of cyclic tensile strain. To identify differentially expressed genes associated with cyclic tensile strain, microarray analysis was performed. Kyoto Encyclopedia of Genes and Genomes (KEGG) analysis identified upregulation of various genes, particularly of the Hedgehog signaling pathway; Ihh and related genes had increased expression compared with controls after 24-hour cyclic tensile strain. In immunoblotting analysis, Ihh, Runx2, Sox9, Gli2, Gli3, and smoothened (SMO) had significantly increased expression after 6- or 12-hour cyclic tensile strain. OPLL samples were strongly immunopositive for Ihh, Sox9, Runx2, Gli2, Gli3, and SMO in the ossification front of OPLL. These results suggest that cyclic tensile strain induces abnormal activation of Ihh and related signaling molecules, and this might be important in the ossification process in OPLL.
Collapse
Affiliation(s)
- Daisuke Sugita
- Department of Orthopaedics and Rehabilitation Medicine, Faculty of Medical Sciences University of Fukui, 23-3 Matsuoka Shimoaizuki, Eiheiji-cho, Yoshida-gun, Fukui, 910-1193, Japan
| | - Hideaki Nakajima
- Department of Orthopaedics and Rehabilitation Medicine, Faculty of Medical Sciences University of Fukui, 23-3 Matsuoka Shimoaizuki, Eiheiji-cho, Yoshida-gun, Fukui, 910-1193, Japan.
| | - Yasuo Kokubo
- Department of Orthopaedics and Rehabilitation Medicine, Faculty of Medical Sciences University of Fukui, 23-3 Matsuoka Shimoaizuki, Eiheiji-cho, Yoshida-gun, Fukui, 910-1193, Japan
| | - Naoto Takeura
- Department of Orthopaedics and Rehabilitation Medicine, Faculty of Medical Sciences University of Fukui, 23-3 Matsuoka Shimoaizuki, Eiheiji-cho, Yoshida-gun, Fukui, 910-1193, Japan
| | - Takafumi Yayama
- Department of Orthopaedic Surgery, Shiga University of Medical Science, Setatsukiwachou, Otsu, Shiga, 520-2192, Japan
| | - Akihiko Matsumine
- Department of Orthopaedics and Rehabilitation Medicine, Faculty of Medical Sciences University of Fukui, 23-3 Matsuoka Shimoaizuki, Eiheiji-cho, Yoshida-gun, Fukui, 910-1193, Japan
| |
Collapse
|
39
|
Disruption of Dhcr7 and Insig1/2 in cholesterol metabolism causes defects in bone formation and homeostasis through primary cilium formation. Bone Res 2020; 8:1. [PMID: 31934493 PMCID: PMC6946666 DOI: 10.1038/s41413-019-0078-3] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2019] [Revised: 08/20/2019] [Accepted: 08/26/2019] [Indexed: 02/07/2023] Open
Abstract
Human linkage studies suggest that craniofacial deformities result from either genetic mutations related to cholesterol metabolism or high-cholesterol maternal diets. However, little is known about the precise roles of intracellular cholesterol metabolism in the development of craniofacial bones, the majority of which are formed through intramembranous ossification. Here, we show that an altered cholesterol metabolic status results in abnormal osteogenesis through dysregulation of primary cilium formation during bone formation. We found that cholesterol metabolic aberrations, induced through disruption of either Dhcr7 (which encodes an enzyme involved in cholesterol synthesis) or Insig1 and Insig2 (which provide a negative feedback mechanism for cholesterol biosynthesis), result in osteoblast differentiation abnormalities. Notably, the primary cilia responsible for sensing extracellular cues were altered in number and length through dysregulated ciliary vesicle fusion in Dhcr7 and Insig1/2 mutant osteoblasts. As a consequence, WNT/β-catenin and hedgehog signaling activities were altered through dysregulated primary cilium formation. Strikingly, the normalization of defective cholesterol metabolism by simvastatin, a drug used in the treatment of cholesterol metabolic aberrations, rescued the abnormalities in both ciliogenesis and osteogenesis in vitro and in vivo. Thus, our results indicate that proper intracellular cholesterol status is crucial for primary cilium formation during skull formation and homeostasis.
Collapse
|
40
|
Burke A, Mishra R. Bone translational medicine. TRANSLATIONAL SYSTEMS MEDICINE AND ORAL DISEASE 2020:283-309. [DOI: 10.1016/b978-0-12-813762-8.00012-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
|
41
|
A combination insecticide at sub-lethal dose debilitated the expression pattern of crucial signalling molecules that facilitate craniofacial patterning in domestic chick Gallus domesticus. Neurotoxicol Teratol 2019; 76:106836. [PMID: 31593814 DOI: 10.1016/j.ntt.2019.106836] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2019] [Revised: 09/04/2019] [Accepted: 09/11/2019] [Indexed: 10/25/2022]
Abstract
Pesticides despite being agents that protect the plants and humans from noxious pests, are infamous for their potential to cause detrimental health issues in nontargeted species. In order to ascertain the latter, a set of experiments were conducted by exposing early chick embryos to a widely used combination insecticide (Ci, 50% chlorpyrifos and 5% cypermethrin). The results revealed a myriad of congenital defects pertaining to craniofacial development such as anophthalmia, microphthalmia, exencephaly as well as deformed beak and cranial structures. These teratological manifestations could be attributed to the Ci induced alteration in the titre of major regulators of neurulation and ossification. Therefore, the mRNA and/or the protein level expression pattern of genes which are reported to be involved in the craniofacial development were studied at selected time points of embryonic development. The analysis of the result showed that there have been significant alternations in the expression patterns of the signalling molecules such as SHH, WNTs, CDH1, CDH2, L1CAM, PAX6, HOX, PCNA, GLI3, BMP7, FGF8, GLIs, SOX9, RUNX2, DLX5, COL10A1, CASPASE3 etc. on embryonic days 2, 4 and/or 10. Concurrently, on day 10, whole-mount skeletal staining and biochemical estimation of hydroxyproline were carried out in the cranial tissues of the embryos. The overall result of the current study indicates that exposure to Ci during early development impede the crucial regulatory signals that orchestrate the morphogenesis of cranial neural crest cells thereby hindering the normal progression of neural tube and endochondral ossification which collectively lead to craniofacial dysmorphism in domestic chicks.
Collapse
|
42
|
Yang H, Zhang M, Liu Q, Zhang H, Zhang J, Lu L, Xie M, Chen D, Wang M. Inhibition of Ihh Reverses Temporomandibular Joint Osteoarthritis via a PTH1R Signaling Dependent Mechanism. Int J Mol Sci 2019; 20:ijms20153797. [PMID: 31382618 PMCID: PMC6695690 DOI: 10.3390/ijms20153797] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2019] [Revised: 07/29/2019] [Accepted: 07/30/2019] [Indexed: 02/07/2023] Open
Abstract
The temporomandibular joint (TMJ), which is biomechanically related to dental occlusion, is often insulted by osteoarthritis (OA). This study was conducted to clarify the relationship between Indian hedgehog (Ihh) and parathyroid hormone receptor 1 (PTH1R) signaling in modulating the enhanced chondrocyte terminal differentiation in dental stimulated TMJ osteoarthritic cartilage. A gain- and loss-of-function strategy was used in an in vitro model in which fluid flow shear stress (FFSS) was applied, and in an in vivo model in which the unilateral anterior cross-bite (UAC) stimulation was adopted. Ihh and PTH1R signaling was modulated through treating the isolated chondrocytes with inhibitor/activator and via deleting Smoothened (Smo) and/or Pth1r genes in mice with the promoter gene of type 2 collagen (Col2-CreER) in the tamoxifen-inducible pattern. We found that both FFSS and UAC stimulation promoted the deep zone chondrocytes to undergo terminal differentiation, while cells in the superficial zone were robust. We demonstrated that the terminal differentiation process in deep zone chondrocytes promoted by FFSS and UAC was mediated by the enhanced Ihh signaling and declined PTH1R expression. The FFSS-promoted terminal differentiation was suppressed by administration of the Ihh inhibitor or PTH1R activator. The UAC-promoted chondrocytes terminal differentiation and OA-like lesions were rescued in Smo knockout, but were enhanced in Pth1r knockout mice. Importantly, the relieving effect of Smo knockout mice was attenuated when Pth1r knockout was also applied. Our data suggest a chondrocyte protective effect of suppressing Ihh signaling in TMJ OA cartilage which is dependent on PTH1R signaling.
Collapse
Affiliation(s)
- Hongxu Yang
- State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases & Shaanxi International Joint Research Center for Oral Diseases, Department of Oral Anatomy and Physiology and TMD, School of Stomatology, the Fourth Military Medical University, Xi'an 710032, China
| | - Mian Zhang
- State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases & Shaanxi International Joint Research Center for Oral Diseases, Department of Oral Anatomy and Physiology and TMD, School of Stomatology, the Fourth Military Medical University, Xi'an 710032, China
| | - Qian Liu
- State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases & Shaanxi International Joint Research Center for Oral Diseases, Department of Oral Anatomy and Physiology and TMD, School of Stomatology, the Fourth Military Medical University, Xi'an 710032, China
| | - Hongyun Zhang
- State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases & Shaanxi International Joint Research Center for Oral Diseases, Department of Oral Anatomy and Physiology and TMD, School of Stomatology, the Fourth Military Medical University, Xi'an 710032, China
| | - Jing Zhang
- State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases & Shaanxi International Joint Research Center for Oral Diseases, Department of Oral Anatomy and Physiology and TMD, School of Stomatology, the Fourth Military Medical University, Xi'an 710032, China
| | - Lei Lu
- State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases & Shaanxi International Joint Research Center for Oral Diseases, Department of Oral Anatomy and Physiology and TMD, School of Stomatology, the Fourth Military Medical University, Xi'an 710032, China
| | - Mianjiao Xie
- State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases & Shaanxi International Joint Research Center for Oral Diseases, Department of Oral Anatomy and Physiology and TMD, School of Stomatology, the Fourth Military Medical University, Xi'an 710032, China
| | - Di Chen
- Department of Biochemistry, Rush University Medical Center, Chicago, IL 60612, USA.
| | - Meiqing Wang
- State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases & Shaanxi International Joint Research Center for Oral Diseases, Department of Oral Anatomy and Physiology and TMD, School of Stomatology, the Fourth Military Medical University, Xi'an 710032, China.
| |
Collapse
|
43
|
Zhang Y, Hu C. WIF-1 and Ihh Expression and Clinical Significance in Patients With Lung Squamous Cell Carcinoma and Adenocarcinoma. Appl Immunohistochem Mol Morphol 2019; 26:454-461. [PMID: 27801732 DOI: 10.1097/pai.0000000000000449] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
This study investigated the expression of wingless-type inhibitory factor-1 (WIF-1) and Ihh protein in tumor tissues and their clinical significance in patients with lung squamous cell carcinoma and adenocarcinoma. The expression of WIF-1 and Ihh protein in 74 squamous cell carcinomas and 76 adenocarcinomas was measured by immunohistochemistry. The percentage of positive WIF-1 protein expression was significantly higher, while positive Ihh protein expression was significantly lower in patients with well-differentiated lung squamous cell carcinoma and adenocarcinoma, tumor node metastasis (TNM) stage I disease, and lymph node metastasis than that in patients with poorly differentiated tumor, TNM stage III disease, and lymph node metastasis (P<0.05, <0.01). Kaplan-Meier survival analysis showed that TNM stage and lymph node metastasis were significantly associated with the mean overall survival of patients with lung squamous cell carcinoma and adenocarcinoma (P<0.05 or <0.01). Patients with lung squamous cell carcinoma (P=0.037) and adenocarcinoma (P=0.001) with positive Ihh protein expression survived significantly shorter than patients with negative Ihh protein expression. In contrast, no significant difference in mean survival was observed in patients with lung squamous cell carcinoma and adenocarcinoma with positive and negative WIF-1 protein expression (P>0.05). Ihh is a marker for poor prognosis in patients with lung squamous cell carcinoma and adenocarcinoma. WIF-1 is not a predictive marker for lung cancer.
Collapse
Affiliation(s)
- Yue Zhang
- Department of Oncology, Second Xiangya Hospital, Central South University, Changsha, Hunan, P.R. China
| | | |
Collapse
|
44
|
Bone growth as the main determinant of mouse digit tip regeneration after amputation. Sci Rep 2019; 9:9720. [PMID: 31273239 PMCID: PMC6609708 DOI: 10.1038/s41598-019-45521-4] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2019] [Accepted: 05/08/2019] [Indexed: 01/08/2023] Open
Abstract
Regeneration is classically demonstrated in mammals using mice digit tip. In this study, we compared different amputation plans and show that distally amputated digits regrow with morphology close to normal but fail to regrow the fat pad. Proximally amputated digits do not regrow the phalangeal bone, but the remaining structures (nail, skin and connective tissue), all with intrinsic regenerative capacity, re-establishing integrity indistinguishably in distally and proximally amputated digits. Thus, we suggest that the bone growth promoted by signals and progenitor cells not removed by distal amputations is responsible for the re-establishment of a drastically different final morphology after distal or proximal digit tip amputations. Despite challenging the use of mouse digit tip as a model system for limb regeneration in mammals, these findings evidence a main role of bone growth in digit tip regeneration and suggest that mechanisms that promote joint structures formation should be the main goal of regenerative medicine for limb and digit regrowth.
Collapse
|
45
|
Chen C, Zheng H, Qi S. Genistein and Silicon Synergistically Protects Against Ovariectomy-Induced Bone Loss Through Upregulating OPG/RANKL Ratio. Biol Trace Elem Res 2019; 188:441-450. [PMID: 30014283 DOI: 10.1007/s12011-018-1433-8] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/03/2018] [Accepted: 07/03/2018] [Indexed: 10/28/2022]
Abstract
We have reported that genistein (Gen) and silicon (Si) have synergistic effects on ovariectomy-induced bone loss in rat; however, the potential mechanisms behind this effect were not fully clarified yet. This study was performed to evaluate the bone protective mechanisms of concomitant intake of genistein and silicon in ovariectomized rat by OPG/RANKL axis. Three-month-old Sprague-Dawley female rats were subjected to ovariectomy (OVX) or sham surgery; after surgery, the OVX rats were randomly divided into five groups: OVX-Gen, OVX-Si, OVX-Gen-Si, OVX-E, and OVX. Genistein, silicon, and 17β-estradiol supplementation were started after ovariectomy and continued for 10 weeks. The results showed that genistein and silicon treatment increased the bone mineral density (BMD) of ovariectomized rats. In addition, the BMD of the tibia and femur were highest in the OVX-Gen-Si group compared with OVX-Gen and OVX-Si group (p < 0.05). After 10 weeks treatment with genistein and silicon, the bone structure of ovariectomized rats was recovered, there was no difference of bone histomorphometric parameters between OVX-Gen-Si, OVX-E, and SHAM group (p > 0.05), and there was no difference in the concentration of serum ALP, Ca, P, OPG, and RANKL between OVX-Gen-Si, SHAM, and OVX-E groups (p > 0.05). RT-PCR showed that genistein and silicon treatment could effectively increase the OPG mRNA expression and decreased the RANKL mRNA expression compared to that of the OVX group (p < 0.05), the OPG/RANKL mRNA ratios were significantly decreased in the OVX group (p < 0.05), and it was nearly to normal in the OVX-Gen-Si group. Immunohistochemical staining results showed that genistein and silicon supplementation could effectively increase the protein expression of OPG and decrease the protein expression of RANKL in bone tissues; there were no significant differences in OPG and RANKL positive expression areas between OVX-Gen-Si, SHAM, and OVX-E group (p > 0.05). The results above indicate that genistein and silicon supplementation can effectively reduce RANKL, increase OPG levels, and OPG/RANKL ratios in the serum and bone tissue of ovariectomized rats; this is the main mechanism by which genistein and silicon play a bone protective role in ovariectomized rats.
Collapse
Affiliation(s)
- Chen Chen
- Chinese-German Joint Laboratory for Natural Product Research, College of Biological Science and Engineering, Qinling-Bashan Mountains Bioresources Comprehensive Development C.I.C, Shaanxi University of Technology, Chaoyang Road, Hantai District, Hanzhong, 723000, Shaanxi Province, China
| | - Hongxing Zheng
- Chinese-German Joint Laboratory for Natural Product Research, College of Biological Science and Engineering, Qinling-Bashan Mountains Bioresources Comprehensive Development C.I.C, Shaanxi University of Technology, Chaoyang Road, Hantai District, Hanzhong, 723000, Shaanxi Province, China.
| | - Shanshan Qi
- Vitamin D research institute, Shaanxi University of Technology, Chaoyang Road, Hantai District, Hanzhong, 723000, Shaanxi Province, China.
| |
Collapse
|
46
|
Zeng Q, Wang Y, Gao J, Yan Z, Li Z, Zou X, Li Y, Wang J, Guo Y. miR-29b-3p regulated osteoblast differentiation via regulating IGF-1 secretion of mechanically stimulated osteocytes. Cell Mol Biol Lett 2019; 24:11. [PMID: 30915127 PMCID: PMC6416934 DOI: 10.1186/s11658-019-0136-2] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2018] [Accepted: 01/23/2019] [Indexed: 12/31/2022] Open
Abstract
Background Mechanical loading is an essential factor for bone formation. A previous study indicated that mechanical tensile strain of 2500 microstrain (με) at 0.5 Hz for 8 h promoted osteogenesis and corresponding mechanoresponsive microRNAs (miRs) were identified in osteoblasts. However, in osteocytes, it has not been identified which miRs respond to the mechanical strain, and it is not fully understood how the mechanoresponsive miRs regulate osteoblast differentiation. Methods Mouse MLO-Y4 osteocytes were applied to the same mechanical tensile strain in vitro. Using molecular and biochemical methods, IGF-1 (insulin-like growth factor-1), PGE2 (prostaglandin E2), OPG (osteoprotegerin) and NOS (nitric oxide synthase) activities of the cells were assayed. MiR microarray and reverse transcription-quantitative polymerase chain reaction (RT-qPCR) assays were applied to select and validate differentially expressed miRs, and the target genes of these miRs were then predicted. MC3T3-E1 osteoblasts were stimulated by the mechanical tensile strain, and the miR-29b-3p expression was detected with miR microarray and RT-qPCR. Additionally, the effect of miR-29b-3p on IFG-1 secretion of osteocytes and the influence of conditioned medium of osteocytes transfected with miR-29b-3p on osteoblast differentiation were investigated. Results The mechanical strain increased secretions of IGF-1 and PGE2, elevated OPG expression and NOS activities, and resulted in altered expression of the ten miRs, and possible target genes for these differentially expressed miRs were revealed through bioinformatics. Among the ten miRs, miR-29b-3p were down-regulated, and miR-29b-3p overexpression decreased the IGF-1 secretion of osteocytes. The mechanical strain did not change expression of osteoblasts' miR-29b-3p. In addition, the conditioned medium of mechanically strained osteocytes promoted osteoblast differentiation, and the conditioned medium of osteocytes transfected with miR-29b-3p mimic inhibited osteoblast differentiation. Conclusions In osteocytes (but not osteoblasts), miR-29b-3p was responsive to the mechanical tensile strain and regulated osteoblast differentiation via regulating IGF-1 secretion of mechanically strained osteocytes.
Collapse
Affiliation(s)
- Qiangcheng Zeng
- 1key laboratory of Functional Bioresource Utilization in University of Shandong, Shandong Key Laboratory of Biophysics, Dezhou University, Dezhou, 253023 China
| | - Yang Wang
- 2Department of Biomedical Engineering, College of Biotechnology, Guilin Medical University, No. 1 Zhiyuan Road, Lingui District, Guilin City, 541100 Guangxi China.,3Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing, 400044 China
| | - Jie Gao
- 1key laboratory of Functional Bioresource Utilization in University of Shandong, Shandong Key Laboratory of Biophysics, Dezhou University, Dezhou, 253023 China.,Medical Department, Secondary Renmin Hospital of Dezhou, Dezhou, 253023 Shangdong China
| | - Zhixiong Yan
- 2Department of Biomedical Engineering, College of Biotechnology, Guilin Medical University, No. 1 Zhiyuan Road, Lingui District, Guilin City, 541100 Guangxi China
| | - Zhenghua Li
- 1key laboratory of Functional Bioresource Utilization in University of Shandong, Shandong Key Laboratory of Biophysics, Dezhou University, Dezhou, 253023 China
| | - Xianqiong Zou
- 2Department of Biomedical Engineering, College of Biotechnology, Guilin Medical University, No. 1 Zhiyuan Road, Lingui District, Guilin City, 541100 Guangxi China
| | - Yanan Li
- 2Department of Biomedical Engineering, College of Biotechnology, Guilin Medical University, No. 1 Zhiyuan Road, Lingui District, Guilin City, 541100 Guangxi China
| | - Jiahui Wang
- 2Department of Biomedical Engineering, College of Biotechnology, Guilin Medical University, No. 1 Zhiyuan Road, Lingui District, Guilin City, 541100 Guangxi China
| | - Yong Guo
- 1key laboratory of Functional Bioresource Utilization in University of Shandong, Shandong Key Laboratory of Biophysics, Dezhou University, Dezhou, 253023 China.,2Department of Biomedical Engineering, College of Biotechnology, Guilin Medical University, No. 1 Zhiyuan Road, Lingui District, Guilin City, 541100 Guangxi China
| |
Collapse
|
47
|
Wang L, Huang J, Moore DC, Song Y, Ehrlich MG, Yang W. SHP2 regulates intramembranous ossification by modifying the TGFβ and BMP2 signaling pathway. Bone 2019; 120:327-335. [PMID: 30471432 PMCID: PMC6360127 DOI: 10.1016/j.bone.2018.11.014] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/04/2018] [Revised: 11/07/2018] [Accepted: 11/20/2018] [Indexed: 02/05/2023]
Abstract
SHP2 is a ubiquitously expressed protein tyrosine phosphatase, which is involved in many signaling pathways to regulate the skeletal development. In endochondral ossification, SHP2 is known to modify the osteogenic fate of osteochondroprogenitors and to impair the osteoblastic transdifferentiation of hypertrophic chondrocytes. However, how SHP2 regulates osteoblast differentiation in intramembranous ossification remains incompletely understood. To address this question, we generated a mouse model to ablate SHP2 in the Prrx1-expressing mesenchymal progenitors by using "Cre-loxP"-mediated gene excision and examined the development of calvarial bone, in which the main process of bone formation is intramembranous ossification. Phenotypic characterization showed that SHP2 mutants have severe defects in calvarial bone formation. Cell lineage tracing and in situ hybridization data showed less osteoblast differentiation of mesenchymal cells and reduced osteogenic genes expression, respectively. Further mechanistic studies revealed enhanced TGFβ and suppressed BMP2 signaling in SHP2 ablated mesenchymal progenitors and their derivatives. Our study uncovered the critical role of SHP2 in osteoblast differentiation through intramembranous ossification and might provide a potential target to treat craniofacial skeleton disorders.
Collapse
Affiliation(s)
- Lijun Wang
- Department of Orthopaedics, Brown University Alpert Medical School and Rhode Island Hospital, Providence, RI 02903, United States of America
| | - Jiahui Huang
- Department of Orthopaedics, Brown University Alpert Medical School and Rhode Island Hospital, Providence, RI 02903, United States of America
| | - Douglas C Moore
- Department of Orthopaedics, Brown University Alpert Medical School and Rhode Island Hospital, Providence, RI 02903, United States of America
| | - Yueming Song
- Department of Orthopedic Surgery, West China Hospital of Sichuan University, Chengdu 610041, P.R. China
| | - Michael G Ehrlich
- Department of Orthopaedics, Brown University Alpert Medical School and Rhode Island Hospital, Providence, RI 02903, United States of America
| | - Wentian Yang
- Department of Orthopaedics, Brown University Alpert Medical School and Rhode Island Hospital, Providence, RI 02903, United States of America.
| |
Collapse
|
48
|
Liu H, Xu J, Jiang R. Mkx-Deficient Mice Exhibit Hedgehog Signaling-Dependent Ectopic Ossification in the Achilles Tendons. J Bone Miner Res 2019; 34:557-569. [PMID: 30458056 PMCID: PMC6535142 DOI: 10.1002/jbmr.3630] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/07/2018] [Revised: 10/25/2018] [Accepted: 11/06/2018] [Indexed: 01/02/2023]
Abstract
Heterotopic ossification is the abnormal formation of mineralized bone in skin, muscle, tendon, or other soft tissues. Tendon ossification often occurs from acute tendon injury or chronic tendon degeneration, for which current treatment relies heavily on surgical removal of the ectopic bony tissues. Unfortunately, surgery creates additional trauma, which often causes recurrence of heterotopic ossification. The molecular mechanisms of heterotopic ossification are not well understood. Previous studies demonstrate that Mkx is a transcription factor crucial for postnatal tendon fibril growth. Here we report that Mkx-/- mutant mice exhibit ectopic ossification in the Achilles tendon within 1 month after birth and the tendon ossification deteriorates with age. Genetic lineage labeling revealed that the tendon ossification in Mkx-/- mice resulted from aberrant differentiation of tendon progenitor cells. Furthermore, tissue-specific inactivation of Mkx in tendon cells postnatally resulted in a similar ossification phenotype, indicating that Mkx plays a key role in tendon tissue homeostasis. Moreover, we show that Hedgehog signaling is ectopically activated at early stages of tendon ossification and that tissue-specific inactivation of Smoothened, which encodes the obligatory transducer of Hedgehog signaling, in the tendon cell lineage prevented or dramatically reduced tendon ossification in Mkx-/- mice. Together, these studies establish a new genetic mouse model of tendon ossification and provide new insight into its pathogenic mechanisms. © 2018 American Society for Bone and Mineral Research.
Collapse
Affiliation(s)
- Han Liu
- Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Jingyue Xu
- Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Rulang Jiang
- Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA.,Division of Plastic Surgery, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA.,Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA.,Shriners Hospitals for Children-Cincinnati, Cincinnati, OH, USA
| |
Collapse
|
49
|
|
50
|
McKenzie JA, Maschhoff C, Liu X, Migotsky N, Silva MJ, Gardner MJ. Activation of hedgehog signaling by systemic agonist improves fracture healing in aged mice. J Orthop Res 2019; 37:51-59. [PMID: 29663560 PMCID: PMC6226344 DOI: 10.1002/jor.24017] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/21/2017] [Accepted: 04/04/2018] [Indexed: 02/04/2023]
Abstract
Fracture healing is a complex process of many coordinated biological pathways. This system can go awry resulting in nonunion, which leads to significant patient morbidity. The Hedgehog (Hh) signaling pathway is upregulated in fracture healing. We hypothesized that the Hh signaling pathway can be pharmacologically modulated to positively affect fracture healing. Diaphyseal femur fractures were created in elderly mice (18 months, C57BL/6 females), which have a blunted and delayed healing response compared to younger mice, and were stabilized with intramedullary pins. To activate the Hh pathway we targeted the receptor Smoothened using an agonist (Hh-Ag1.5 [Hh-Ag]) and compared this to a vehicle control. Expression of Hh target genes were significantly increased in the fracture callus of the agonist group compared to controls, indicating pathway activation. Expression of osteogenic and chondrogenic-related genes was greatly upregulated in fracture callus versus intact femora, although Hh agonist treatment did not consistently enhance this response. Blindly graded, radiographic callus healing scores were significantly higher in the Hh-Ag groups at post operative day (POD) 14, indicating earlier callus bridging. On microCT, Hh-Ag treatment led to greater callus volume (+40%) and bone volume (+25%) at POD21. By day 14, callus vascularity, as assessed by 3D microCT angiography vessel volume, was 85% greater in the Hh-Ag group. Finally, mechanical strength of the calluses in the Hh-Ag groups was significantly greater than in the control groups at POD21. In conclusion, systemic administration of a Hh agonist appears to improve the osseous and vascular healing responses in a mouse fracture healing-impaired model. © 2018 Orthopaedic Research Society. Published by Wiley Periodicals, Inc. J Orthop Res.
Collapse
Affiliation(s)
| | | | - Xiaochen Liu
- Washington University Orthopedics, St. Louis, MO
| | | | | | - Michael J. Gardner
- Washington University Orthopedics, St. Louis, MO,Stanford University Orthopedics, Stanford, CA
| |
Collapse
|