1
|
Tan Z, Shek HT, Li Z, Xia L, He Y, Chen P, Wong JSH, Gao B, Chan D, To MKT. An inducible mouse model of osteogenesis imperfecta type V reveals aberrant osteogenesis caused by Ifitm5 c.-14C>T mutation. J Bone Miner Res 2025; 40:577-590. [PMID: 39908237 PMCID: PMC12103725 DOI: 10.1093/jbmr/zjaf022] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/11/2024] [Revised: 01/14/2025] [Accepted: 01/18/2025] [Indexed: 02/07/2025]
Abstract
Osteogenesis imperfecta (OI) type V is typically characterized by radial head dislocation, calcification of interosseous membrane, and hyperplastic callus. It is caused by the c.-14C>T mutation in the 5' UTR of IFITM5 gene, adding 5 amino acids (MALEP) to the N-terminal of IFITM5 protein. Previous studies have suggested a neomorphic function of the MALEP-IFITM5 protein. However, the underlying mechanisms remain unclear due to embryonic lethality in previous mouse models. Therefore, we developed an inducible mouse model (Ifitm5flox c.-14C>T) that could be induced by Cre expressed at different developmental stages to explore the pathogenic effects of the neomorphic MALEP-IFITM5. The mutant Ifitm5 allele could be regulated by the endogenous regulatory elements after Cre recombination, maintaining its spatiotemporal expression pattern and physiological level. Specifically, Prx1-Cre; Ifitm5flox c.-14C>T mutant mice were born with fractures in all limbs, showing impaired ossification and enhanced chondrogenesis associated with increased SOX9 abundance. Analyses of single-cell RNA sequencing data revealed arrested osteogenesis in Prx1-Cre; Ifitm5flox c.-14C>T mouse. A major population of cells expressing both osteogenic and chondrogenic signature genes was identified in the mutant mouse. Reduced expression of SP7 and SOST in the cortical regions of mutant mice confirmed delayed osteocyte maturation and compromised osteogenesis. Elevated bone marrow adipocytes were found in the adult mutant mice. Ectopic chondrogenesis and SOX9 expression were also observed in the perichondrium regions of Col1a1-Cre; Ifitm5flox c.-14C>T and Ocn-Cre; Ifitm5flox c.-14C>T mutant mice. The inducible Ifitm5flox c.-14C>T mouse model and integrated single-cell transcriptomic analyses elucidated that ectopic expression of SOX9 and disrupted homeostatic balance among osteogenesis, chondrogenesis, and adipogenesis may contribute to the pathogenesis caused by MALEP-IFITM5, helping to gain deeper insights into the molecular mechanisms of type V OI.
Collapse
Affiliation(s)
- Zhijia Tan
- Department of Orthopaedics and Traumatology, The University of Hong Kong-Shenzhen Hospital, Shenzhen 518053, China
- Shenzhen Clinical Research Centre for Rare Diseases, The University of Hong Kong-Shenzhen Hospital, Shenzhen 518053, China
- Department of Orthopaedics and Traumatology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Hiu Tung Shek
- Shenzhen Clinical Research Centre for Rare Diseases, The University of Hong Kong-Shenzhen Hospital, Shenzhen 518053, China
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Zeluan Li
- Shenzhen Clinical Research Centre for Rare Diseases, The University of Hong Kong-Shenzhen Hospital, Shenzhen 518053, China
- Department of Orthopaedics and Traumatology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Linjian Xia
- Clinical, Translational and Basic Research Laboratory, The University of Hong Kong-Shenzhen Hospital, Shenzhen 518053, China
| | - Yanni He
- Department of Orthopaedics and Traumatology, The University of Hong Kong-Shenzhen Hospital, Shenzhen 518053, China
- Shenzhen Clinical Research Centre for Rare Diseases, The University of Hong Kong-Shenzhen Hospital, Shenzhen 518053, China
| | - Peikai Chen
- Department of Orthopaedics and Traumatology, The University of Hong Kong-Shenzhen Hospital, Shenzhen 518053, China
- Shenzhen Clinical Research Centre for Rare Diseases, The University of Hong Kong-Shenzhen Hospital, Shenzhen 518053, China
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Janus Siu Him Wong
- Department of Orthopaedics and Traumatology, The University of Hong Kong-Shenzhen Hospital, Shenzhen 518053, China
- Shenzhen Clinical Research Centre for Rare Diseases, The University of Hong Kong-Shenzhen Hospital, Shenzhen 518053, China
- Department of Orthopaedics and Traumatology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Bo Gao
- Department of Orthopaedics and Traumatology, The University of Hong Kong-Shenzhen Hospital, Shenzhen 518053, China
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China
| | - Danny Chan
- Department of Orthopaedics and Traumatology, The University of Hong Kong-Shenzhen Hospital, Shenzhen 518053, China
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Michael Kai Tsun To
- Department of Orthopaedics and Traumatology, The University of Hong Kong-Shenzhen Hospital, Shenzhen 518053, China
- Shenzhen Clinical Research Centre for Rare Diseases, The University of Hong Kong-Shenzhen Hospital, Shenzhen 518053, China
- Department of Orthopaedics and Traumatology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| |
Collapse
|
2
|
Garmendia Urdalleta A, Witte-Bouma J, Kops N, Lolli A, Farrell E. Osteoclast Incorporation in an In Vitro 3D Model of Endochondral Ossification. Tissue Eng Part A 2025. [PMID: 40279297 DOI: 10.1089/ten.tea.2024.0281] [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: 04/27/2025] Open
Abstract
In vitro models aim to recapitulate human physiological processes, improving upon and replacing the need for animal-based models. Modeling bone formation via endochondral ossification in vitro is a very complex process due to the large number of cell types involved. Most current models are limited to mimicking the initial stages of the process (i.e., cartilage template formation and mineralization of the matrix), using a single cell type. Chondroclasts/osteoclasts are key players in cartilage resorption during endochondral ossification, but their introduction into in vitro models has thus far proven challenging. In this study, we aimed toward a new level of model complexity by introducing human monocyte-derived osteoclasts into 3D in vitro-cultured cartilage templates undergoing mineralization. Chondrogenic and mineralized chondrogenic pellets were formed from human pediatric bone marrow stromal cells and cultured in the presence of transforming growth factor-β3 (TGF-β) and TGF-β/β-glycerophosphate, respectively. These pellets have the capacity to form bone if implanted in vivo. To identify suitable in vitro co-culture conditions and investigate cell interactions, pellets were co-cultured with CD14+ monocytes in an indirect (transwell) or direct setting for up to 14 days, and osteoclastogenesis was assessed by means of histological stainings, osteoclast counting, and gene expression analysis. Upon direct co-culture, we achieved effective osteoclast formation in situ in regions of both mineralized and unmineralized cartilages. Notably, in vitro-generated osteoclasts showed the ability to form tunnels in the chondrogenic matrix and infiltrate the mineralized matrix. Addition of osteoclasts in human in vitro models of endochondral ossification increases the physiological relevance of these models. This will allow for the development of robust 3D human in vitro systems for the study of bone formation, disease modeling, and drug discovery, further reducing the need for animal models in the future.
Collapse
Affiliation(s)
- Amaia Garmendia Urdalleta
- Department of Oral and Maxillofacial Surgery, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Janneke Witte-Bouma
- Department of Oral and Maxillofacial Surgery, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Nicole Kops
- Department of Orthopaedics and Sports Medicine, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Andrea Lolli
- Department of Oral and Maxillofacial Surgery, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Eric Farrell
- Department of Oral and Maxillofacial Surgery, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| |
Collapse
|
3
|
Fang J, Wang S. Bibliometric analysis of research trends and emerging insights of osteoarthritis and chondrocyte hypertrophy. Front Surg 2025; 12:1538339. [PMID: 40276315 PMCID: PMC12018320 DOI: 10.3389/fsurg.2025.1538339] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2024] [Accepted: 03/31/2025] [Indexed: 04/26/2025] Open
Abstract
Background This study aims to systematically analyze the intersection of OA and chondrocyte hypertrophy using bibliometric methods, providing an quantitative and comprehensive overview of the current research status and emerging trends in this field. Methods Relevant publications were retrieved from the Web of Science Core Collection database using the search query TS = ("chondrocyte* hypertroph*" OR "hypertrophic chondrocyte*" OR "cartilage hypertroph*") AND ("osteoarthriti*" OR "OA" OR "degenerative arthritis"). Several bibliometric tools, including Vosviewer, CiteSpace, the R package (bibliometrix), and Excel 2021, were utilized to systematically analyze the publications on the intersection of chondrocyte hypertrophy and OA. Results A total of 639 publications, published between 1995 and 2025, were identified. The findings indicate a steady global increase in research on OA and chondrocyte hypertrophy, with an increasing number of studies being published in high-impact journals, suggesting a promising developmental trajectory. China and the United States are leading in this field. OSTEOARTHRITIS AND CARTILAGE is identified as the core journal in this area, while ANNALS OF THE RHEUMATIC DISEASES has the highest impact factor among the top publishing journals. Keyword analysis reveals that research hotspots primarily focus on stem cells, tissue engineering, cartilage repair, inflammation, oxidative stress, autophagy, apoptosis, senescence, and related bioactive factors. Conclusion This study elucidates the current research status and trends at the intersection of OA and chondrocyte hypertrophy, providing crucial references for future research. Future studies should continue to focus on these potential therapeutic approaches, key phenotypes, and regulatory mechanisms, enhance international cooperation to develop more effective strategies and treatments for OA.
Collapse
Affiliation(s)
- Jiajin Fang
- School of Clinical Medicine, Weifang Medical University, Weifang, Shandong, China
- Department of Sports Medical and Rehabilitation, Peking University Shenzhen Hospital, Shenzhen, Guangdong, China
| | - Shuai Wang
- Pain Management Department, Zhejiang Provincial Hospital of Traditional Chinese Medicine, Hangzhou, China
| |
Collapse
|
4
|
Andreev D, Porschitz P. Emerging Roles of Eosinophils in Bone. Curr Osteoporos Rep 2025; 23:17. [PMID: 40183859 PMCID: PMC11971228 DOI: 10.1007/s11914-025-00913-6] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 03/11/2025] [Indexed: 04/05/2025]
Abstract
PURPOSE OF THE REVIEW Eosinophils are traditionally known for their role in immune defense against parasites and their involvement in various immunopathologies, including eosinophilic airway diseases, eosinophilic dermatoses, and gastrointestinal disorders. However, recent findings from our group and other leading laboratories have broadened this perspective, revealing that eosinophils also play crucial roles in tissue development, homeostasis, and regeneration. This review aims to highlight the regulatory functions of eosinophils within the bone niche and emphasize the importance of further research into their role in bone biology. RECENT FINDINGS Growing evidence suggests that eosinophils are key regulators of bone metabolism, extending beyond their established roles in immunity and inflammation. They contribute to bone homeostasis by inhibiting osteoclast differentiation, helping to prevent excessive bone resorption in osteoporosis and inflammatory arthritis. Additionally, eosinophils may promote osteoblast-mediated bone formation, modulate the mesenchymal and hematopoietic stem cell niche, and contribute to the bone microenvironment by affecting vascularization and extracellular matrix composition. However, their impact may vary under pathological conditions. Patients with eosinophilic disorders are often at an increased risk of osteoporosis and fragility fractures, though this is largely attributed to disease-related treatments rather than eosinophil activity itself. Despite emerging insights into the role of eosinophils in bone biology, the underlying mechanisms remain incompletely understood. Further research is essential to elucidate how eosinophils influence bone physiology and pathology.
Collapse
Affiliation(s)
- Darja Andreev
- Center for Regenerative Therapies Dresden (CRTD), Technische Universität (TU) Dresden, 01307, Dresden, Germany.
| | - Pauline Porschitz
- Center for Regenerative Therapies Dresden (CRTD), Technische Universität (TU) Dresden, 01307, Dresden, Germany
| |
Collapse
|
5
|
Seehawer M, Polyak K. Epigenetic drivers of metalloproteinases and metastasis. Trends Cell Biol 2025:S0962-8924(25)00044-3. [PMID: 40089451 DOI: 10.1016/j.tcb.2025.02.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2024] [Revised: 02/18/2025] [Accepted: 02/19/2025] [Indexed: 03/17/2025]
Abstract
Metalloproteinases (MPs) are crucial for development and homeostasis due to their diverse physiological functions, from the cellular to the organismal level. Their activity is tightly regulated at multiple levels, including epigenetic regulation through DNA methylation and histone modifications. Aberrant MP expression can result in pathological events, involving extracellular matrix remodeling, which can facilitate cancer cell invasion and dissemination. As clinical testing of MP inhibitors has been limited by toxicity, alternative approaches are needed. Epigenetically-driven MP expression is often specific to cancer cells, giving an enticing possibility for cancer cell-specific targeting. Moreover, aberrant epigenetic activity can also drive other metastatic events. Therefore, targeting the epigenetic regulators of MP expression may be a promising alternative approach for the prevention and treatment of metastatic disease.
Collapse
Affiliation(s)
- Marco Seehawer
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA; Department of Medicine, Brigham and Women's Hospital, Boston, MA 02115, USA; Department of Medicine, Harvard Medical School, Boston, MA 02115, USA
| | - Kornelia Polyak
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA; Department of Medicine, Brigham and Women's Hospital, Boston, MA 02115, USA; Department of Medicine, Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|
6
|
Taye N, Karoulias SZ, Balic Z, Wang LW, Willard BB, Martin D, Richard D, Okamoto AS, Capellini TD, Apte SS, Hubmacher D. Combined ADAMTS10 and ADAMTS17 inactivation exacerbates bone shortening and compromises extracellular matrix formation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.23.634616. [PMID: 39896540 PMCID: PMC11785165 DOI: 10.1101/2025.01.23.634616] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 02/04/2025]
Abstract
Weill-Marchesani syndrome (WMS) is characterized by severe short stature, short hands and feet (brachydactyly), joint contractures, tight skin, and heart valve, eye, and skin anomalies. Whereas recessive WMS is caused by mutations in ADAMTS10, ADAMTS17, or LTBP2, dominant WMS is caused by mutations in FBN1 (encoding fibrillin-1). Since bone growth is driven by chondrocyte proliferation and hypertrophy in the growth plates, the genetics of WMS suggests that the affected ECM proteins act within the same pathway to regulate chondrocyte and growth plate function. Here, we investigated the role of the secreted ADAMTS proteases ADAMTS10 and ADAMTS17 in growth plate function and ECM formation. We generated Adamts10;Adamts17 double knockout (DKO) mice, which showed significant postnatal lethality compared to single Adamts10 or Adamts17 KO mice. Importantly, we observed severe bone shortening DKO mice, which correlated with a narrower hypertrophic zone in their growth plates. ADAMTS17 substrates identified by N-terminomics and yeast two-hybrid screening identified the ECM proteins fibronectin and collagen VI (COL6). However, validation experiments did not reveal direct proteolysis of either fibronectin or COL6 by ADAMTS17. We then investigated ECM formation in primary ADAMTS10- and ADAMTS17-deficient skin fibroblasts and observed compromised fibronectin deposition concomitant with aberrant intracellular accumulation of fibrillin-1. These findings support a role for ADAMTS17 in ECM protein secretion and assembly. Collectively, our data suggest that ADAMTS10 and ADAMTS17 regulate bone growth by regulating chondrocyte hypertrophy or hypertrophic chondrocyte turnover. Mechanistically, ADAMTS17 appears to be a critical regulator of ECM protein secretion or pericellular matrix assembly, whereas ADAMTS10 likely modulates ECM formation at later stages, possibly regulating the spatio-temporal deposition of fibrillin isoforms.
Collapse
Affiliation(s)
- Nandaraj Taye
- Orthopedic Research Laboratories, Leni & Peter W. May Department of Orthopedics, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Stylianos Z. Karoulias
- Orthopedic Research Laboratories, Leni & Peter W. May Department of Orthopedics, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Zerina Balic
- Orthopedic Research Laboratories, Leni & Peter W. May Department of Orthopedics, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Lauren W. Wang
- Department of Biomedical Engineering, Cleveland Clinic Lerner Research Institute, Cleveland, OH, 44195, USA
- Department of Orthopaedic Surgery, Cleveland Clinic Orthopaedic and Rheumatologic Institute, Cleveland, OH, 44195, USA
| | - Belinda B. Willard
- Proteomics and Metabolomics Core, Cleveland Clinic Lerner Research Institute, Cleveland, OH, 44195, USA
| | - Daniel Martin
- Department of Biomedical Engineering, Cleveland Clinic Lerner Research Institute, Cleveland, OH, 44195, USA
| | - Daniel Richard
- Human Evolutionary Biology, Harvard University, Cambridge, MA, 02138, USA
| | | | - Terence D. Capellini
- Human Evolutionary Biology, Harvard University, Cambridge, MA, 02138, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, 02142, USA
| | - Suneel S. Apte
- Department of Biomedical Engineering, Cleveland Clinic Lerner Research Institute, Cleveland, OH, 44195, USA
- Department of Orthopaedic Surgery, Cleveland Clinic Orthopaedic and Rheumatologic Institute, Cleveland, OH, 44195, USA
| | - Dirk Hubmacher
- Orthopedic Research Laboratories, Leni & Peter W. May Department of Orthopedics, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Mindich Child Health and Development Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| |
Collapse
|
7
|
Minges P, Eder M, Eder AC. Dual-Labeled Small Peptides in Cancer Imaging and Fluorescence-Guided Surgery: Progress and Future Perspectives. Pharmaceuticals (Basel) 2025; 18:143. [PMID: 40005958 PMCID: PMC11858487 DOI: 10.3390/ph18020143] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2024] [Revised: 01/14/2025] [Accepted: 01/17/2025] [Indexed: 02/27/2025] Open
Abstract
Dual-labeled compounds that combine radiolabeling and fluorescence labeling represent a significant advancement in precision oncology. Their clinical implementation enhances patient care and outcomes by leveraging the high sensitivity of radioimaging for tumor detection and taking advantage of fluorescence-based optical visualization for surgical guidance. Non-invasive radioimaging facilitates immediate identification of both primary tumors and metastases, while fluorescence imaging assists in decision-making during surgery by offering a spatial distinction between malignant and non-malignant tissue. These advancements hold promise for enhancing patient outcomes and personalization of cancer treatment. The development of dual-labeled molecular probes targeting various cancer biomarkers is crucial in addressing the heterogeneity inherent in cancer pathology and recent studies had already demonstrated the impact of dual-labeled compounds in surgical decision-making (NCT03699332, NCT03407781). This review focuses on the development and application of small dual-labeled peptides in the imaging and treatment of various cancer types. It summarizes the biomarkers targeted to date, tracing their development from initial discovery to the latest advancements in peptidomimetics. Through comprehensive analysis of recent preclinical and clinical studies, the review demonstrates the potential of these dual-labeled peptides to improve tumor detection, localization, and resection. Additionally, it highlights the evolving landscape of dual-modality imaging, emphasizing its critical role in advancing personalized and effective cancer therapy. This synthesis of current research underscores the promise of dual-labeled peptides in enhancing diagnostic accuracy and therapeutic outcomes in oncology.
Collapse
Affiliation(s)
- Paul Minges
- Department of Nuclear Medicine, University Medical Center Freiburg, Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany; (P.M.); (M.E.)
- Department of Radiopharmaceutical Development, German Cancer Consortium (DKTK), Partner Site Freiburg, Freiburg, Germany and German Cancer Research Center, 69120 Heidelberg, Germany
| | - Matthias Eder
- Department of Nuclear Medicine, University Medical Center Freiburg, Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany; (P.M.); (M.E.)
- Department of Radiopharmaceutical Development, German Cancer Consortium (DKTK), Partner Site Freiburg, Freiburg, Germany and German Cancer Research Center, 69120 Heidelberg, Germany
| | - Ann-Christin Eder
- Department of Nuclear Medicine, University Medical Center Freiburg, Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany; (P.M.); (M.E.)
- Department of Radiopharmaceutical Development, German Cancer Consortium (DKTK), Partner Site Freiburg, Freiburg, Germany and German Cancer Research Center, 69120 Heidelberg, Germany
| |
Collapse
|
8
|
Li J, Sun N, Hu S, Zuo Z. Chronic high fat diet-induced cerebrovascular remodeling impairs recovery of blood flow after cerebral ischemia in mice. J Cereb Blood Flow Metab 2025:271678X251313723. [PMID: 39819094 PMCID: PMC11748376 DOI: 10.1177/0271678x251313723] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/22/2024] [Revised: 09/28/2024] [Accepted: 12/19/2024] [Indexed: 01/19/2025]
Abstract
Obesity and associated metabolic disturbances worsen brain ischemia outcome. High fat diet (HFD)-fed mice are obese and have cerebrovascular remodeling and worsened brain ischemia outcome. We determined whether HFD-induced cerebrovascular remodeling impaired reperfusion to the ischemic penumbra. Six-week-old C57BL/6J or matrix metalloprotease-9 knockout (MMP-9-/-) mice were on HFD or regular diet (RD) for 12 to 14 months before a 60-min left middle cerebral arterial occlusion (MCAO). Photoacoustic microscopy was performed at left cerebral frontal cortex. HFD increased cerebrovascular density and tortuosity in C57BL/6J mice but not in MMP-9-/- mice. Blood flow to the ischemic penumbra slowly recovered but did not reach the baseline 2 h after MCAO in RD-fed mice. Oxygen extraction fraction was increased to maintain cerebral metabolic rate of oxygen (CMRO2) throughout brain ischemia and reperfusion period. This blood flow recovery was worsened in HFD-fed mice, leading to decreased CMRO2. MMP-9-/- attenuated these HFD effects. HFD increased MMP-9 activity and interleukin 1β. Pyrrolidine dithiocarbamate, an anti-inflammatory agent, abolished the HFD effects. Interleukin 1β increased MMP-9 activity. In summary, HFD induces cerebrovascular remodeling, leading to worsened recovery of blood supply to the ischemic penumbra to contribute to poor outcome after brain ischemia. Neuroinflammation may activate MMP-9 in HFD-fed mice.
Collapse
Affiliation(s)
- Jun Li
- Department of Anesthesiology, University of Virginia, Charlottesville, VA, USA
| | - Naidi Sun
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA, USA
- Department of Biomedical Engineering, Washington University in St Louis, St. Louis, MO, USA
| | - Song Hu
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA, USA
- Department of Biomedical Engineering, Washington University in St Louis, St. Louis, MO, USA
| | - Zhiyi Zuo
- Department of Anesthesiology, University of Virginia, Charlottesville, VA, USA
| |
Collapse
|
9
|
Mao W, Zheng Y, Zhang W, Yin J, Liu Z, He P, Hou G, Huang G, Chen H, Lin J, Xu J, Li A, Qin S. Enocyanin promotes osteogenesis and bone regeneration by inhibiting MMP9. Int J Mol Med 2025; 55:9. [PMID: 39513591 PMCID: PMC11554379 DOI: 10.3892/ijmm.2024.5450] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Accepted: 08/29/2024] [Indexed: 11/15/2024] Open
Abstract
Enocyanin (ENO), an anthocyanin extracted from grapes, has been shown to exert inhibitory effects on acid phosphatase and inflammation; however, its role in osteogenesis and bone formation is currently unknown. The present study aimed to investigate the effects of ENO on osteogenesis in vitro and bone formation in vivo, and to explore the rudimentary mechanisms. KusaO cells were employed to evaluate the osteogenic role of ENO in vitro by Alizarin red S staining, ALP staining, quantitative PCR and western blotting, and an in vivo analysis of the therapeutic effects of ENO on a femoral fracture model was performed using stereo microscope, micro‑CT and histological staining. To further investigate the underlying mechanisms, mRNA sequencing was employed to investigate the changes in gene expression and the downstream pathways after ENO treatment. The results showed that ENO could promote the osteogenic differentiation of KusaO cells in vitro and bone fracture regeneration in vivo. Mechanistically, ENO was highly related to bone formation, including the 'Wnt signalling pathway', 'bone development' and 'bone mineralization'. In addition, matrix metalloproteinase 9 (MMP9) was identified as one of the targets of ENO in its promotional role in osteogenesis. In conclusion, ENO may represent a therapeutic candidate for bone regeneration in bone fractures by regulating osteogenesis and bone formation via MMP9.
Collapse
Affiliation(s)
- Wei Mao
- Guangzhou Institute of Traumatic Surgery, Department of Orthopaedics, Guangzhou Red Cross Hospital, Medical College, Jinan University, Guangzhou, Guangdong 510220, P.R. China
| | - Yinfeng Zheng
- Guangzhou Institute of Traumatic Surgery, Department of Orthopaedics, Guangzhou Red Cross Hospital, Medical College, Jinan University, Guangzhou, Guangdong 510220, P.R. China
| | - Wencong Zhang
- Guangzhou Institute of Traumatic Surgery, Department of Orthopaedics, Guangzhou Red Cross Hospital, Medical College, Jinan University, Guangzhou, Guangdong 510220, P.R. China
| | - Jinrong Yin
- Guangzhou Institute of Traumatic Surgery, Department of Orthopaedics, Guangzhou Red Cross Hospital, Medical College, Jinan University, Guangzhou, Guangdong 510220, P.R. China
| | - Zhiyi Liu
- Guangzhou Institute of Traumatic Surgery, Department of Orthopaedics, Guangzhou Red Cross Hospital, Medical College, Jinan University, Guangzhou, Guangdong 510220, P.R. China
| | - Peiliang He
- Guangzhou Institute of Traumatic Surgery, Department of Orthopaedics, Guangzhou Red Cross Hospital, Medical College, Jinan University, Guangzhou, Guangdong 510220, P.R. China
| | - Guodong Hou
- Guangzhou Institute of Traumatic Surgery, Department of Orthopaedics, Guangzhou Red Cross Hospital, Medical College, Jinan University, Guangzhou, Guangdong 510220, P.R. China
| | - Guowei Huang
- Guangzhou Institute of Traumatic Surgery, Department of Orthopaedics, Guangzhou Red Cross Hospital, Medical College, Jinan University, Guangzhou, Guangdong 510220, P.R. China
| | - Huan Chen
- Guangzhou Institute of Traumatic Surgery, Department of Orthopaedics, Guangzhou Red Cross Hospital, Medical College, Jinan University, Guangzhou, Guangdong 510220, P.R. China
| | - Junyan Lin
- Guangzhou Institute of Traumatic Surgery, Department of Orthopaedics, Guangzhou Red Cross Hospital, Medical College, Jinan University, Guangzhou, Guangdong 510220, P.R. China
| | - Jiake Xu
- School of Biomedical Science, The University of Western Australia, Perth 6009, Australia
- Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong 518055, P.R. China
| | - Aiguo Li
- Guangzhou Institute of Traumatic Surgery, Department of Orthopaedics, Guangzhou Red Cross Hospital, Medical College, Jinan University, Guangzhou, Guangdong 510220, P.R. China
| | - Shengnan Qin
- Guangzhou Institute of Traumatic Surgery, Department of Orthopaedics, Guangzhou Red Cross Hospital, Medical College, Jinan University, Guangzhou, Guangdong 510220, P.R. China
- School of Biomedical Science, The University of Western Australia, Perth 6009, Australia
- Monoclonal Antibody Facility, Harry Perkins Institute of Medical Research, Perth 6009, Australia
| |
Collapse
|
10
|
Li J, Li K, Zhang Y, Li X, Wang H. Regulation mechanism of endochondral ossification in Rana zhenhaiensis during metamorphosis based on histomorphology and transcriptome analyses. COMPARATIVE BIOCHEMISTRY AND PHYSIOLOGY. PART D, GENOMICS & PROTEOMICS 2024; 52:101286. [PMID: 38996694 DOI: 10.1016/j.cbd.2024.101286] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Revised: 06/28/2024] [Accepted: 06/29/2024] [Indexed: 07/14/2024]
Abstract
Endochondral ossification plays a crucial role in the limb development of amphibians. This study explored the ossification sequence in the hindlimb of Rana zhenhaiensis tadpoles and the correlation between thyroid hormones (THs) and endochondral ossification via histomorphology and transcriptional analyses. Our results suggest that ossification of the femur and tibiofibula was initiated during the period of high THs activity (metamorphosis climax). In addition, the results of differentially expressed gene analyses in the hindlimb and tail showed that systemic factors, transcription factors, and locally secreted factors interacted with each other during the metamorphosis climax to regulate the occurrence of endochondral ossification. These results will enrich the morphological data of anurans and provide scientific reference for the evolutionary history of vertebrates.
Collapse
Affiliation(s)
- Jiayi Li
- College of Life Science, Shaanxi Normal University, Xi'an 710119, China
| | - Kaiyue Li
- College of Life Science, Shaanxi Normal University, Xi'an 710119, China
| | - Yue Zhang
- College of Life Science, Shaanxi Normal University, Xi'an 710119, China
| | - Xinyi Li
- College of Life Science, Shaanxi Normal University, Xi'an 710119, China
| | - Hongyuan Wang
- College of Life Science, Shaanxi Normal University, Xi'an 710119, China.
| |
Collapse
|
11
|
Liu M, Zhang H, Li Y, Huang D, Zuo H, Yang J, Chen Z. Loss of MMP9 disturbs cranial suture fusion via suppressing cell proliferation, chondrogenesis and osteogenesis in mice. Matrix Biol 2024; 134:93-106. [PMID: 39374863 DOI: 10.1016/j.matbio.2024.10.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Revised: 10/04/2024] [Accepted: 10/04/2024] [Indexed: 10/09/2024]
Abstract
Cranial sutures function as growth centers for calvarial bones. Abnormal suture closure will cause permanent cranium deformities. MMP9 is a member of the gelatinases that degrades components of the extracellular matrix. MMP9 has been reported to regulate bone development and remodeling. However, the function of MMP9 in cranial suture development is still unknown. Here, we identified that the expression of Mmp9 was specifically elevated during fusion of posterior frontal (PF) suture compared with other patent sutures in mice. Interestingly, inhibition of MMP9 ex vivo or knockout of Mmp9 in mice (Mmp9-/-) disturbed the fusion of PF suture. Histological analysis showed that knockout of Mmp9 resulted in wider distance between osteogenic fronts, suppressed cell condensation and endocranial bone formation in PF suture. Proliferation, chondrogenesis and osteogenesis of suture cells were decreased in Mmp9-/- mice, leading to the PF suture defects. Moreover, transcriptome analysis of PF suture revealed upregulated ribosome biogenesis and downregulated IGF signaling associated with abnormal closure of PF suture in Mmp9-/- mice. Inhibition of the ribosome biogenesis partially rescued PF suture defects caused by Mmp9 knockout. Altogether, these results indicate that MMP9 is critical for the fusion of cranial sutures, thus suggesting MMP9 as a potential therapeutic target for cranial suture diseases.
Collapse
Affiliation(s)
- Ming Liu
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School and Hospital of Stomatology, Wuhan University, Wuhan 430079, China
| | - Hanshu Zhang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School and Hospital of Stomatology, Wuhan University, Wuhan 430079, China
| | - Yuanyuan Li
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School and Hospital of Stomatology, Wuhan University, Wuhan 430079, China
| | - Delan Huang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School and Hospital of Stomatology, Wuhan University, Wuhan 430079, China
| | - Huanyan Zuo
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School and Hospital of Stomatology, Wuhan University, Wuhan 430079, China
| | - Jingwen Yang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School and Hospital of Stomatology, Wuhan University, Wuhan 430079, China; Department of Pediatric Dentistry, School and Hospital of Stomatology, Wuhan University, Wuhan 430079, China.
| | - Zhi Chen
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School and Hospital of Stomatology, Wuhan University, Wuhan 430079, China; Department of Cariology and Endodontics, School & Hospital of Stomatology, Wuhan University, Wuhan 430079, China.
| |
Collapse
|
12
|
Kalev-Altman R, Fraggi-Rankis V, Monsonego-Ornan E, Sela-Donenfeld D. The bone Gla protein osteocalcin is expressed in cranial neural crest cells. BMC Res Notes 2024; 17:329. [PMID: 39501347 PMCID: PMC11539830 DOI: 10.1186/s13104-024-06990-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Accepted: 10/28/2024] [Indexed: 11/08/2024] Open
Abstract
BACKGROUND Osteocalcin is a small protein abundant in the bone extracellular-matrix, that serves as a marker for mature osteoblasts. To become activated, osteocalcin undergoes a specific post-translational carboxylation. Osteocalcin is expressed at advanced stages of embryogenesis and after birth, when bone formation takes place. Neural crest cells (NCCs) are a unique cell population that evolves during early stages of development. While initially NCCs populate the dorsal neural-tube, later they undergo epithelial-to-mesenchymal-transition and migrate throughout the embryo in highly-regulated manner. NCCs give rise to multiple cell types including neurons and glia of the peripheral nervous system, chromaffin cells and skin melanocytes. Remarkably, in the head region, NCCs give rise to cartilage and bone. FINDING Here we report that osteocalcin is detected in cranial NCCs. Analysis of chick embryos at stages of cranial NCC migration revealed that osteocalcin mRNA and protein is expressed in pre-migratory and migratory NCCs in-vivo and ex-vivo. Addition of warfarin, an inhibitor of osteocalcin carboxylation, onto neural-tube explants, reduced the amount of NCC migration. These results provide the first evidence of osteocalcin presence in cranial NCCs, much before they give rise to craniofacial skeleton, and propose its possible involvement in the regulation of NCC migration.
Collapse
Affiliation(s)
- Rotem Kalev-Altman
- Koret School of Veterinary Medicine, The Robert H. Smith Faculty of Agriculture, Food and Environment, The Hebrew University, Rehovot, 76100, Israel
- The Institute of Biochemistry and Nutrition, The Robert H. Smith Faculty of Agriculture, Food and Environment, The Hebrew University, Rehovot, 76100, Israel
- The Section of Genetic Medicine, The Department of Medicine, The University of Chicago, Chicago, IL, 60637, USA
| | - Veatriki Fraggi-Rankis
- Koret School of Veterinary Medicine, The Robert H. Smith Faculty of Agriculture, Food and Environment, The Hebrew University, Rehovot, 76100, Israel
- Smart Assays Biotechnologies Ltd, Weizmann Science Park, Ness Ziona, 7414003, Israel
| | - Efrat Monsonego-Ornan
- The Institute of Biochemistry and Nutrition, The Robert H. Smith Faculty of Agriculture, Food and Environment, The Hebrew University, Rehovot, 76100, Israel
| | - Dalit Sela-Donenfeld
- Koret School of Veterinary Medicine, The Robert H. Smith Faculty of Agriculture, Food and Environment, The Hebrew University, Rehovot, 76100, Israel.
| |
Collapse
|
13
|
Zhou Z, Mao X, Jiang C, Li W, Zhou T, Liu M, Sun S, Wang M, Dong N, Wu Q, Zhou H. Deficiencies in corin and atrial natriuretic peptide-mediated signaling impair endochondral ossification in bone development. Commun Biol 2024; 7:1380. [PMID: 39443661 PMCID: PMC11500007 DOI: 10.1038/s42003-024-07077-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Accepted: 10/14/2024] [Indexed: 10/25/2024] Open
Abstract
Corin is a protease that activates atrial natriuretic peptide (ANP), a hormone in cardiovascular homeostasis. Structurally, ANP is similar to C-type natriuretic peptide (CNP) crucial in bone development. Here, we examine the role of corin and ANP in chondrocyte differentiation and bone formation. We show that in Corin and Nppa (encoding ANP) knockout (KO) mice, chondrocyte differentiation is impaired, resulting in shortened limb long bones. In adult mice, Corin and Nppa deficiency impairs bone density and microarchitecture. Molecular studies in cartilages from newborn Corin and Nppa KO mice and in cultured chondrocytes indicate that corin and ANP act in chondrocytes via cGMP-dependent protein kinase G signaling to inhibit mitogen-activated protein kinase phosphorylation and stimulate glycogen synthase kinase-3β phosphorylation and β-catenin upregulation. These results indicate that corin and ANP signaling regulates chondrocyte differentiation in bone development and homeostasis, suggesting that enhancing ANP signaling may improve bone quality in patients with osteoporosis.
Collapse
Affiliation(s)
- Zibin Zhou
- Department of Orthopedics, the Second Affiliated Hospital of Soochow University, Suzhou, China
- Cyrus Tang Hematology Center, Collaborative Innovation Center of Hematology, State Key Laboratory of Radiation Medicine and Prevention, Soochow University, Suzhou, China
| | - Xiaoyu Mao
- Department of Orthopedics, the Second Affiliated Hospital of Soochow University, Suzhou, China
- Cyrus Tang Hematology Center, Collaborative Innovation Center of Hematology, State Key Laboratory of Radiation Medicine and Prevention, Soochow University, Suzhou, China
| | - Chun Jiang
- Department of Orthopedics, the Second Affiliated Hospital of Soochow University, Suzhou, China
- Cyrus Tang Hematology Center, Collaborative Innovation Center of Hematology, State Key Laboratory of Radiation Medicine and Prevention, Soochow University, Suzhou, China
| | - Wenguo Li
- Cyrus Tang Hematology Center, Collaborative Innovation Center of Hematology, State Key Laboratory of Radiation Medicine and Prevention, Soochow University, Suzhou, China
| | - Tiantian Zhou
- Cyrus Tang Hematology Center, Collaborative Innovation Center of Hematology, State Key Laboratory of Radiation Medicine and Prevention, Soochow University, Suzhou, China
| | - Meng Liu
- Cyrus Tang Hematology Center, Collaborative Innovation Center of Hematology, State Key Laboratory of Radiation Medicine and Prevention, Soochow University, Suzhou, China
| | - Shijin Sun
- Cyrus Tang Hematology Center, Collaborative Innovation Center of Hematology, State Key Laboratory of Radiation Medicine and Prevention, Soochow University, Suzhou, China
| | - Mengting Wang
- Cyrus Tang Hematology Center, Collaborative Innovation Center of Hematology, State Key Laboratory of Radiation Medicine and Prevention, Soochow University, Suzhou, China
- NHC Key Laboratory of Thrombosis and Hemostasis, Jiangsu Institute of Hematology, the First Affiliated Hospital of Soochow University, Suzhou, China
| | - Ningzheng Dong
- Cyrus Tang Hematology Center, Collaborative Innovation Center of Hematology, State Key Laboratory of Radiation Medicine and Prevention, Soochow University, Suzhou, China.
- NHC Key Laboratory of Thrombosis and Hemostasis, Jiangsu Institute of Hematology, the First Affiliated Hospital of Soochow University, Suzhou, China.
| | - Qingyu Wu
- Cyrus Tang Hematology Center, Collaborative Innovation Center of Hematology, State Key Laboratory of Radiation Medicine and Prevention, Soochow University, Suzhou, China.
| | - Haibin Zhou
- Department of Orthopedics, the Second Affiliated Hospital of Soochow University, Suzhou, China.
| |
Collapse
|
14
|
Geister E, Ard D, Patel H, Findley A, DeSouza G, Martin L, Knox H, Gavara N, Lugea A, Sabbatini ME. The Role of Twist1 in Chronic Pancreatitis-Associated Pancreatic Stellate Cells. THE AMERICAN JOURNAL OF PATHOLOGY 2024; 194:1879-1897. [PMID: 39032603 PMCID: PMC11423762 DOI: 10.1016/j.ajpath.2024.06.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/23/2023] [Revised: 06/18/2024] [Accepted: 06/28/2024] [Indexed: 07/23/2024]
Abstract
In healthy pancreas, pancreatic stellate cells (PaSCs) synthesize the basement membrane, which is mainly composed of type IV collagen and laminin. In chronic pancreatitis (CP), PaSCs are responsible for the production of a rigid extracellular matrix (ECM) that is mainly composed of fibronectin and type I/III collagen. Reactive oxygen species evoke the formation of the rigid ECM by PaSCs. One source of reactive oxygen species is NADPH oxidase (Nox) enzymes. Nox1 up-regulates the expression of Twist1 and matrix metalloproteinase-9 (MMP-9) in PaSCs from mice with CP. This study determined the functional relationship between Twist1 and MMP-9, and other PaSC-produced proteins, and the extent to which Twist1 regulates digestion of ECM proteins in CP. Twist1 induced the expression of MMP-9 in mouse PaSCs. The action of Twist1 was not selective to MMP-9 because Twist1 induced the expression of types I and IV collagen, fibronectin, transforming growth factor, and α-smooth muscle actin. Luciferase assay indicated that Twist1 in human primary PaSCs increased the expression of MMP-9 at the transcriptional level in an NF-κB dependent manner. The digestion of type I/III collagen by MMP-9 secreted by PaSCs from mice with CP depended on Twist1. Thus, Twist1 in PaSCs from mice with CP induced rigid ECM production and MMP-9 transcription in an NF-κB-dependent mechanism that selectively displayed proteolytic activity toward type I/III collagen.
Collapse
Affiliation(s)
- Emma Geister
- Department of Biological Sciences, Augusta University, Augusta, Georgia
| | - Dalton Ard
- Department of Biological Sciences, Augusta University, Augusta, Georgia
| | - Heer Patel
- Department of Biological Sciences, Augusta University, Augusta, Georgia
| | - Alyssa Findley
- Department of Biological Sciences, Augusta University, Augusta, Georgia
| | - Godfrey DeSouza
- Department of Biological Sciences, Augusta University, Augusta, Georgia
| | - Lyndsay Martin
- Department of Biological Sciences, Augusta University, Augusta, Georgia
| | - Henry Knox
- Department of Biological Sciences, Augusta University, Augusta, Georgia
| | - Natasha Gavara
- Department of Biological Sciences, Augusta University, Augusta, Georgia
| | - Aurelia Lugea
- Cedars-Sinai Medical Center, Los Angeles, California
| | | |
Collapse
|
15
|
Todorova VK, Azhar G, Stone A, Malapati SJ, Che Y, Zhang W, Makhoul I, Wei JY. Neutrophil Biomarkers Can Predict Cardiotoxicity of Anthracyclines in Breast Cancer. Int J Mol Sci 2024; 25:9735. [PMID: 39273682 PMCID: PMC11395913 DOI: 10.3390/ijms25179735] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2024] [Revised: 08/29/2024] [Accepted: 08/31/2024] [Indexed: 09/15/2024] Open
Abstract
Doxorubicin (DOX), a commonly used anticancer agent, causes cardiotoxicity that begins with the first dose and may progress to heart failure years after treatment. An inflammatory response associated with neutrophil recruitment has been recognized as a mechanism of DOX-induced cardiotoxicity. This study aimed to validate mRNA expression of the previously identified biomarkers of DOX-induced cardiotoxicity, PGLYRP1, CAMP, MMP9, and CEACAM8, and to assay their protein expression in the peripheral blood of breast cancer patients. Blood samples from 40 breast cancer patients treated with DOX-based chemotherapy were collected before and after the first chemotherapy cycle and > 2 years after treatment. The protein and gene expression of PGLYRP1/Tag7, CAMP/LL37, MMP9/gelatinase B, and CEACAM8/CD66b were determined using ELISA and reverse transcription-quantitative polymerase chain reaction (RT-qPCR). Receiver operating characteristic (ROC) curve analysis was used to determine the diagnostic value of each candidate biomarker. Patients with cardiotoxicity (n = 20) had significantly elevated levels of PGLYRP1, CAMP, MMP9, and CEACAM8 at baseline, after the first dose of DOX-based chemotherapy, and at > 2 years after treatment relative to patients without cardiotoxicity (n = 20). The first dose of DOX induced significantly higher levels of all examined biomarkers in both groups of patients. At > 2 years post treatment, the levels of all but MMP9 dropped below the baseline. There was a good correlation between the expression of mRNA and the target proteins. We demonstrate that circulating levels of PGLYRP1, CAMP, MMP9, and CEACAM8 can predict the cardiotoxicity of DOX. This novel finding may be of value in the early identification of patients at risk for cardiotoxicity.
Collapse
Affiliation(s)
- Valentina K Todorova
- Division of Hematology/Oncology, Department of Internal Medicine, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
- Department of Geriatrics, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
- Winthrop P. Rockefeller Cancer Institute, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
- Central Arkansas Veterans Healthcare System, Little Rock, AR 72205, USA
| | - Gohar Azhar
- Department of Geriatrics, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Annjanette Stone
- Central Arkansas Veterans Healthcare System, Little Rock, AR 72205, USA
| | - Sindhu J Malapati
- Division of Hematology/Oncology, Department of Internal Medicine, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
- Winthrop P. Rockefeller Cancer Institute, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Yingni Che
- Department of Geriatrics, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Wei Zhang
- Department of Mathematics and Statistics, University of Arkansas at Little Rock, Little Rock, AR 72205, USA
| | - Issam Makhoul
- Division of Hematology/Oncology, Department of Internal Medicine, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
- Winthrop P. Rockefeller Cancer Institute, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Jeanne Y Wei
- Department of Geriatrics, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| |
Collapse
|
16
|
Chen F, Zhao J, Meng F, He F, Ni J, Fu Y. The vascular contribution of apolipoprotein E to Alzheimer's disease. Brain 2024; 147:2946-2965. [PMID: 38748848 DOI: 10.1093/brain/awae156] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2023] [Revised: 03/23/2024] [Accepted: 04/21/2024] [Indexed: 09/04/2024] Open
Abstract
Alzheimer's disease, the most prevalent form of dementia, imposes a substantial societal burden. The persistent inadequacy of disease-modifying drugs targeting amyloid plaques and neurofibrillary tangles suggests the contribution of alternative pathogenic mechanisms. A frequently overlooked aspect is cerebrovascular dysfunction, which may manifest early in the progression of Alzheimer's disease pathology. Mounting evidence underscores the pivotal role of the apolipoprotein E gene, particularly the apolipoprotein ε4 allele as the strongest genetic risk factor for late-onset Alzheimer's disease, in the cerebrovascular pathology associated with Alzheimer's disease. In this review, we examine the evidence elucidating the cerebrovascular impact of both central and peripheral apolipoprotein E on the pathogenesis of Alzheimer's disease. We present a novel three-hit hypothesis, outlining potential mechanisms that shed light on the intricate relationship among different pathogenic events. Finally, we discuss prospective therapeutics targeting the cerebrovascular pathology associated with apolipoprotein E and explore their implications for future research endeavours.
Collapse
Affiliation(s)
- Feng Chen
- Department of Neurology, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Jing Zhao
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and Institute of Neuroscience, Soochow University, Suzhou 215123, China
| | - Fanxia Meng
- Department of Neurology, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Fangping He
- Department of Neurology, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Jie Ni
- Department of Neurology, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Yuan Fu
- Department of Neurology, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| |
Collapse
|
17
|
Bianchi E, Bhattacharya B, Bowling AJ, Pence HE, Mundy PC, Jones G, Muriana A, Grever WE, Pappas-Garton A, Sriram S, LaRocca J, Bondesson M. Applications of Zebrafish Embryo Models to Predict Developmental Toxicity for Agrochemical Product Development. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2024; 72:18132-18145. [PMID: 39087946 DOI: 10.1021/acs.jafc.4c00970] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/02/2024]
Abstract
The development of safe crop protection products is a complex process that traditionally relies on intensive animal use for hazard identification. Methods that capture toxicity in early stages of agrochemical discovery programs enable a more efficient and sustainable product development pipeline. Here, we explored whether the zebrafish model can be leveraged to identify mammalian-relevant toxicity. We used transgenic zebrafish to assess developmental toxicity following exposures to known mammalian teratogens and captured larval morphological malformations, including bone and vascular perturbations. We further applied toxicogenomics to identify common biomarker signatures of teratogen exposure. The results show that the larval malformation assay predicted teratogenicity with 82.35% accuracy, 87.50% specificity, and 77.78% sensitivity. Similar and slightly lower accuracies were obtained with the vascular and bone assays, respectively. A set of 20 biomarkers were identified that efficiently segregated teratogenic chemicals from nonteratogens. In conclusion, zebrafish are valuable, robust, and cost-effective models for toxicity testing in the early stages of product development.
Collapse
Affiliation(s)
- Enrica Bianchi
- Corteva Agriscience, Indianapolis, Indiana 46268, United States
| | | | | | - Heather E Pence
- Corteva Agriscience, Indianapolis, Indiana 46268, United States
| | - Paige C Mundy
- Corteva Agriscience, Indianapolis, Indiana 46268, United States
| | - Gabe Jones
- Corteva Agriscience, Indianapolis, Indiana 46268, United States
| | | | | | | | | | - Jessica LaRocca
- Corteva Agriscience, Indianapolis, Indiana 46268, United States
| | - Maria Bondesson
- Department of Intelligent Systems Engineering, Indiana University, Bloomington, Indiana 47408, United States
| |
Collapse
|
18
|
Yang F, Lee G, Fan Y. Navigating tumor angiogenesis: therapeutic perspectives and myeloid cell regulation mechanism. Angiogenesis 2024; 27:333-349. [PMID: 38580870 PMCID: PMC11303583 DOI: 10.1007/s10456-024-09913-z] [Citation(s) in RCA: 22] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2023] [Accepted: 03/04/2024] [Indexed: 04/07/2024]
Abstract
Sustained angiogenesis stands as a hallmark of cancer. The intricate vascular tumor microenvironment fuels cancer progression and metastasis, fosters therapy resistance, and facilitates immune evasion. Therapeutic strategies targeting tumor vasculature have emerged as transformative for cancer treatment, encompassing anti-angiogenesis, vessel normalization, and endothelial reprogramming. Growing evidence suggests the dynamic regulation of tumor angiogenesis by infiltrating myeloid cells, such as macrophages, myeloid-derived suppressor cells (MDSCs), and neutrophils. Understanding these regulatory mechanisms is pivotal in paving the way for successful vasculature-targeted cancer treatments. Therapeutic interventions aimed to disrupt myeloid cell-mediated tumor angiogenesis may reshape tumor microenvironment and overcome tumor resistance to radio/chemotherapy and immunotherapy.
Collapse
Affiliation(s)
- Fan Yang
- Department of Radiation Oncology, University of Pennsylvania, Philadelphia, PA, 19104, USA.
- Department of Obstetrics and Gynecology, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China.
- Shanghai Key Laboratory of Gynecologic Oncology, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China.
| | - Gloria Lee
- Department of Radiation Oncology, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Yi Fan
- Department of Radiation Oncology, University of Pennsylvania, Philadelphia, PA, 19104, USA.
| |
Collapse
|
19
|
Rodrigues KE, Pontes MHB, Cantão MBS, Prado AF. The role of matrix metalloproteinase-9 in cardiac remodeling and dysfunction and as a possible blood biomarker in heart failure. Pharmacol Res 2024; 206:107285. [PMID: 38942342 DOI: 10.1016/j.phrs.2024.107285] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Revised: 06/15/2024] [Accepted: 06/23/2024] [Indexed: 06/30/2024]
Abstract
Heart failure (HF) is the leading cause of morbidity and mortality in cardiovascular diseases, being responsible for many hospitalizations annually. HF is considered a public health problem with significant economic and social impact, which makes searches essential for strategies that improve the ability to predict and diagnose HF. In this way, biomarkers can help in risk stratification for a more personalized approach to patients with HF. Preclinical and clinical evidence shows the participation of matrix metalloproteinase 9 (MMP-9) in the HF process. In this review, we will demonstrate the critical role that MMP-9 plays in cardiac remodeling and dysfunction. We will also show its importance as a blood biomarker in acute and chronic HF patients.
Collapse
Affiliation(s)
- Keuri Eleutério Rodrigues
- Biodiversity and Biotechnology Post Graduate Program - BIONORTE, Institute of Biological Sciences, Federal University of Para, Belem, Brazil; Cardiovascular System Pharmacology and Toxicology Laboratory, Institute of Biological Sciences, Federal University of Para, Belem, Brazil
| | - Maria Helena Barbosa Pontes
- Cardiovascular System Pharmacology and Toxicology Laboratory, Institute of Biological Sciences, Federal University of Para, Belem, Brazil; Pharmacology and Biochemistry Post Graduate Program - FARMABIO, Institute of Biological Sciences, Federal University of Para, Belem, Brazil
| | - Manoel Benedito Sousa Cantão
- Cardiovascular System Pharmacology and Toxicology Laboratory, Institute of Biological Sciences, Federal University of Para, Belem, Brazil; Pharmacology and Biochemistry Post Graduate Program - FARMABIO, Institute of Biological Sciences, Federal University of Para, Belem, Brazil
| | - Alejandro Ferraz Prado
- Biodiversity and Biotechnology Post Graduate Program - BIONORTE, Institute of Biological Sciences, Federal University of Para, Belem, Brazil; Cardiovascular System Pharmacology and Toxicology Laboratory, Institute of Biological Sciences, Federal University of Para, Belem, Brazil; Pharmacology and Biochemistry Post Graduate Program - FARMABIO, Institute of Biological Sciences, Federal University of Para, Belem, Brazil.
| |
Collapse
|
20
|
Yao Q, He L, Bao C, Yan X, Ao J. The role of TNF-α in osteoporosis, bone repair and inflammatory bone diseases: A review. Tissue Cell 2024; 89:102422. [PMID: 39003912 DOI: 10.1016/j.tice.2024.102422] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2023] [Revised: 05/17/2024] [Accepted: 05/23/2024] [Indexed: 07/16/2024]
Abstract
Tumour necrosis factor alpha (TNF-α) is a pleiotropic cytokine synthesised primarily by mononuclear cells; it has a potent pro-inflammatory effect, playing a crucial role in metabolic, immune, and inflammatory diseases. This cytokine has been studied in various biological systems. In bone tissue, TNF-α plays an integral role in skeletal disorders such as osteoporosis, fracture repair and rheumatoid arthritis through its involvement in regulating the balance between osteoblasts and osteoclasts, mediating inflammatory responses, promoting angiogenesis and exacerbating synovial proliferation. The biological effect TNF-α exerts in this context is determined by a combination of the signalling pathway it activates, the type of receptor it binds, and the concentration and duration of exposure. This review summarises the participation and pathophysiological role of TNF-α in osteoporosis, bone damage repair, chronic immunoinflammatory bone disease and spinal cord injury, and discusses its main mechanisms.
Collapse
Affiliation(s)
| | - Li He
- Affiliated Hospital of Zunyi Medical University, China.
| | | | - Xuhang Yan
- Affiliated Hospital of Zunyi Medical University, China.
| | - Jun Ao
- Affiliated Hospital of Zunyi Medical University, China.
| |
Collapse
|
21
|
Gheorghe SR, Crăciun AM, Ilyés T, Tisa IB, Sur L, Lupan I, Samasca G, Silaghi CN. Converging Mechanisms of Vascular and Cartilaginous Calcification. BIOLOGY 2024; 13:565. [PMID: 39194503 DOI: 10.3390/biology13080565] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/29/2024] [Revised: 07/19/2024] [Accepted: 07/24/2024] [Indexed: 08/29/2024]
Abstract
Physiological calcification occurs in bones and epiphyseal cartilage as they grow, whereas ectopic calcification occurs in blood vessels, cartilage, and soft tissues. Although it was formerly thought to be a passive and degenerative process associated with aging, ectopic calcification has been identified as an active cell-mediated process resembling osteogenesis, and an increasing number of studies have provided evidence for this paradigm shift. A significant association between vascular calcification and cardiovascular risk has been demonstrated by various studies, which have shown that arterial calcification has predictive value for future coronary events. With respect to cartilaginous calcification, calcium phosphate or hydroxyapatite crystals can form asymptomatic deposits in joints or periarticular tissues, contributing to the pathophysiology of osteoarthritis, rheumatoid arthritis, ankylosing spondylitis, tendinitis, and bursitis. The risk factors and sequence of events that initiate ectopic calcification, as well as the mechanisms that prevent the development of this pathology, are still topics of debate. Consequently, in this review, we focus on the nexus of the mechanisms underlying vascular and cartilaginous calcifications, trying to circumscribe the similarities and disparities between them to provide more clarity in this regard.
Collapse
Affiliation(s)
- Simona R Gheorghe
- Department of Medical Biochemistry, Iuliu Hatieganu University of Medicine and Pharmacy, 400349 Cluj-Napoca, Romania
| | - Alexandra M Crăciun
- Department of Medical Biochemistry, Iuliu Hatieganu University of Medicine and Pharmacy, 400349 Cluj-Napoca, Romania
| | - Tamás Ilyés
- Department of Medical Biochemistry, Iuliu Hatieganu University of Medicine and Pharmacy, 400349 Cluj-Napoca, Romania
| | - Ioana Badiu Tisa
- Department of Pediatrics III, Iuliu Hatieganu University of Medicine and Pharmacy, 400217 Cluj-Napoca, Romania
| | - Lucia Sur
- Department of Pediatrics I, Iuliu Hatieganu University of Medicine and Pharmacy, 400370 Cluj-Napoca, Romania
| | - Iulia Lupan
- Department of Molecular Biology, Babes-Bolyai University, 400084 Cluj-Napoca, Romania
| | - Gabriel Samasca
- Department of Immunology, Iuliu Hatieganu University of Medicine and Pharmacy, 400162 Cluj-Napoca, Romania
| | - Ciprian N Silaghi
- Department of Medical Biochemistry, Iuliu Hatieganu University of Medicine and Pharmacy, 400349 Cluj-Napoca, Romania
| |
Collapse
|
22
|
Yuan L, Wei J, Xiao S, Jin S, Xia X, Liu H, Liu J, Hu J, Zuo Y, Li Y, Yang F, Li J. Nano-laponite encapsulated coaxial fiber scaffold promotes endochondral osteogenesis. Regen Biomater 2024; 11:rbae080. [PMID: 39055302 PMCID: PMC11269679 DOI: 10.1093/rb/rbae080] [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: 02/21/2024] [Revised: 05/23/2024] [Accepted: 06/15/2024] [Indexed: 07/27/2024] Open
Abstract
Osteoinductive supplements without side effects stand out from the growth factors and drugs widely used in bone tissue engineering. Lithium magnesium sodium silicate hydrate (laponite) nanoflake is a promising bioactive component for bone regeneration, attributed to its inherent biosafety and effective osteoinductivity. Up to now, the in vivo osteogenic potential and mechanisms of laponite-encapsulated fibrous membranes remain largely unexplored. This study presents a unique method for homogeneously integrating high concentrations of laponite RDS into a polycaprolactone (PCL) matrix by dispersing laponite RDS sol into the polymer solution. Subsequently, a core-shell fibrous membrane (10RP-PG), embedding laponite-loaded PCL in its core, was crafted using coaxial electrospinning. The PCL core's slow degradation and the shell's gradient degradation enabled the sustained release of bioactive ions (Si and Mg) from laponite. In vivo studies on a critical-sized calvarial bone defect model demonstrated that the 10RP-PG membrane markedly enhanced bone formation and remodeling by accelerating the process of endochondral ossification. Further transcriptome analysis suggested that osteogenesis in the 10RP-PG membrane is driven by Mg and Si from endocytosed laponite, activating pathways related to ossification and endochondral ossification, including Hippo, Wnt and Notch. The fabricated nanocomposite fibrous membranes hold great promise in the fields of critical-sized bone defect repair.
Collapse
Affiliation(s)
- Li Yuan
- Research Center for Nano-Biomaterials, Analytical and Testing Center, Sichuan University, Chengdu 610064, China
| | - Jiawei Wei
- Research Center for Nano-Biomaterials, Analytical and Testing Center, Sichuan University, Chengdu 610064, China
| | - Shiqi Xiao
- Research Center for Nano-Biomaterials, Analytical and Testing Center, Sichuan University, Chengdu 610064, China
| | - Shue Jin
- Research Center for Nano-Biomaterials, Analytical and Testing Center, Sichuan University, Chengdu 610064, China
| | - Xue Xia
- Research Center for Nano-Biomaterials, Analytical and Testing Center, Sichuan University, Chengdu 610064, China
| | - Huan Liu
- Research Center for Nano-Biomaterials, Analytical and Testing Center, Sichuan University, Chengdu 610064, China
| | - Jiangshan Liu
- Research Center for Nano-Biomaterials, Analytical and Testing Center, Sichuan University, Chengdu 610064, China
| | - Jiaxin Hu
- Research Center for Nano-Biomaterials, Analytical and Testing Center, Sichuan University, Chengdu 610064, China
| | - Yi Zuo
- Research Center for Nano-Biomaterials, Analytical and Testing Center, Sichuan University, Chengdu 610064, China
| | - Yubao Li
- Research Center for Nano-Biomaterials, Analytical and Testing Center, Sichuan University, Chengdu 610064, China
| | - Fang Yang
- Department of Dentistry—Regenerative Biomaterials, Research Institute for Medical Innovation, Nijmegen, 6525EX, The Netherlands
| | - Jidong Li
- Research Center for Nano-Biomaterials, Analytical and Testing Center, Sichuan University, Chengdu 610064, China
| |
Collapse
|
23
|
Lounev V, Groppe JC, Brewer N, Wentworth KL, Smith V, Xu M, Schomburg L, Bhargava P, Al Mukaddam M, Hsiao EC, Shore EM, Pignolo RJ, Kaplan FS. Matrix metalloproteinase-9 deficiency confers resilience in fibrodysplasia ossificans progressiva in a man and mice. J Bone Miner Res 2024; 39:382-398. [PMID: 38477818 DOI: 10.1093/jbmr/zjae029] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/18/2024] [Revised: 02/11/2024] [Accepted: 02/13/2024] [Indexed: 03/14/2024]
Abstract
Single case studies of extraordinary disease resilience may provide therapeutic insight into conditions for which no definitive treatments exist. An otherwise healthy 35-year-old man (patient-R) with the canonical pathogenic ACVR1R206H variant and the classic congenital great toe malformation of fibrodysplasia ossificans progressiva (FOP) had extreme paucity of post-natal heterotopic ossification (HO) and nearly normal mobility. We hypothesized that patient-R lacked a sufficient post-natal inflammatory trigger for HO. A plasma biomarker survey revealed a reduction in total matrix metalloproteinase-9 (MMP-9) compared to healthy controls and individuals with quiescent FOP. Whole exome sequencing identified compound heterozygous variants in MMP-9 (c.59C > T, p.A20V and c.493G > A, p.D165N). Structural analysis of the D165N variant predicted both decreased MMP-9 secretion and activity that were confirmed by enzyme-linked immunosorbent assay and gelatin zymography. Further, human proinflammatory M1-like macrophages expressing either MMP-9 variant produced significantly less Activin A, an obligate ligand for HO in FOP, compared to wildtype controls. Importantly, MMP-9 inhibition by genetic, biologic, or pharmacologic means in multiple FOP mouse models abrogated trauma-induced HO, sequestered Activin A in the extracellular matrix (ECM), and induced regeneration of injured skeletal muscle. Our data suggest that MMP-9 is a druggable node linking inflammation to HO, orchestrates an existential role in the pathogenesis of FOP, and illustrates that a single patient's clinical phenotype can reveal critical molecular mechanisms of disease that unveil novel treatment strategies.
Collapse
Affiliation(s)
- Vitali Lounev
- Department of Orthopaedic Surgery, The University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, United States
- The Center for Research in FOP and Related Disorders, The University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, United States
| | - Jay C Groppe
- Department of Biomedical Sciences, Texas A & M University College of Dentistry, Dallas, TX 75246-2013, United States
| | - Niambi Brewer
- Department of Orthopaedic Surgery, The University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, United States
- The Center for Research in FOP and Related Disorders, The University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, United States
| | - Kelly L Wentworth
- Department of Medicine, Division of Endocrinology and Metabolism, Zuckerberg San Francisco General Hospital, University of California, San Francisco, CA 94143-0794, United States
- Department of Medicine, University of California, San Francisco, CA 94143-0794, United States
| | | | - Meiqi Xu
- Department of Orthopaedic Surgery, The University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, United States
- The Center for Research in FOP and Related Disorders, The University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, United States
| | - Lutz Schomburg
- Institute for Experimental Endocrinology, Charite University Hospital, D-10115 Berlin, Germany
| | | | - Mona Al Mukaddam
- Department of Orthopaedic Surgery, The University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, United States
- The Center for Research in FOP and Related Disorders, The University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, United States
- Department of Medicine, The University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, United States
| | - Edward C Hsiao
- Department of Medicine, University of California, San Francisco, CA 94143-0794, United States
- Division of Endocrinology and Metabolism, The Institute for Human Genetics, the Program in Craniofacial Biology, University of California, San Francisco, CA 94143-0794, United States
| | - Eileen M Shore
- Department of Orthopaedic Surgery, The University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, United States
- The Center for Research in FOP and Related Disorders, The University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, United States
- Department of Genetics, The University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, United States
| | - Robert J Pignolo
- Department of Medicine, Mayo Clinic College of Medicine, Mayo Clinic, Rochester, MN 55905, United States
| | - Frederick S Kaplan
- Department of Orthopaedic Surgery, The University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, United States
- The Center for Research in FOP and Related Disorders, The University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, United States
- Department of Medicine, The University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, United States
| |
Collapse
|
24
|
Abdollahi F, Saghatchi M, Paryab A, Malek Khachatourian A, Stephens ED, Toprak MS, Badv M. Angiogenesis in bone tissue engineering via ceramic scaffolds: A review of concepts and recent advancements. BIOMATERIALS ADVANCES 2024; 159:213828. [PMID: 38479240 DOI: 10.1016/j.bioadv.2024.213828] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/10/2023] [Revised: 03/08/2024] [Accepted: 03/08/2024] [Indexed: 04/05/2024]
Abstract
Due to organ donor shortages, long transplant waitlists, and the complications/limitations associated with auto and allotransplantation, biomaterials and tissue-engineered models are gaining attention as feasible alternatives for replacing and reconstructing damaged organs and tissues. Among various tissue engineering applications, bone tissue engineering has become a promising strategy to replace or repair damaged bone. We aimed to provide an overview of bioactive ceramic scaffolds in bone tissue engineering, focusing on angiogenesis and the effect of different biofunctionalization strategies. Different routes to angiogenesis, including chemical induction through signaling molecules immobilized covalently or non-covalently, in situ secretion of angiogenic growth factors, and the degradation of inorganic scaffolds, are described. Physical induction mechanisms are also discussed, followed by a review of methods for fabricating bioactive ceramic scaffolds via microfabrication methods, such as photolithography and 3D printing. Finally, the strengths and weaknesses of the commonly used methodologies and future directions are discussed.
Collapse
Affiliation(s)
- Farnoosh Abdollahi
- Department of Dentistry, Kashan University of Medical Science, Kashan, Iran
| | - Mahshid Saghatchi
- School of Metallurgy & Materials Engineering, Iran University of Science and Technology, Tehran, Iran
| | - Amirhosein Paryab
- Department of Materials Science & Engineering, Sharif University of Technology, Tehran, Iran
| | | | - Emma D Stephens
- Department of Biomedical Engineering, University of Calgary, 2500 University Drive NW, Calgary, Alberta T2N 1N4, Canada
| | - Muhammet S Toprak
- Department of Applied Physics, Biomedical and X-ray Physics, KTH Royal Institute of Technology, SE 10691 Stockholm, Sweden
| | - Maryam Badv
- Department of Biomedical Engineering, University of Calgary, 2500 University Drive NW, Calgary, Alberta T2N 1N4, Canada; Libin Cardiovascular Institute, University of Calgary, 3330 Hospital Drive NW, Calgary, AB T2N 4N1, Canada
| |
Collapse
|
25
|
Pignolo RJ, Kaplan FS, Wang H. Cell Senescence in Heterotopic Ossification. Biomolecules 2024; 14:485. [PMID: 38672501 PMCID: PMC11047966 DOI: 10.3390/biom14040485] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Revised: 04/08/2024] [Accepted: 04/15/2024] [Indexed: 04/28/2024] Open
Abstract
The formation of bone outside the normal skeleton, or heterotopic ossification (HO), occurs through genetic and acquired mechanisms. Fibrodysplasia ossificans progressiva (FOP), the most devastating genetic condition of HO, is due to mutations in the ACVR1/ALK2 gene and is relentlessly progressive. Acquired HO is mostly precipitated by injury or orthopedic surgical procedures but can also be associated with certain conditions related to aging. Cellular senescence is a hallmark of aging and thought to be a tumor-suppressive mechanism with characteristic features such as irreversible growth arrest, apoptosis resistance, and an inflammatory senescence-associated secretory phenotype (SASP). Here, we review possible roles for cellular senescence in HO and how targeting senescent cells may provide new therapeutic approaches to both FOP and acquired forms of HO.
Collapse
Affiliation(s)
- Robert J. Pignolo
- Department of Medicine, Section of Geriatric Medicine & Gerontology, Mayo Clinic, Rochester, MN 55905, USA
- Divisions of Endocrinology and Hospital Internal Medicine, Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN 55905, USA
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, MN 55905, USA;
| | - Frederick S. Kaplan
- Department of Orthopaedic Surgery, The Perelman School of Medicine of the University of Pennsylvania, Philadelphia, PA 19104, USA;
- Department of Medicine, The Perelman School of Medicine of the University of Pennsylvania, Philadelphia, PA 19104, USA
- The Center for Research in FOP and Related Disorders, The Perelman School of Medicine of the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Haitao Wang
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, MN 55905, USA;
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN 55905, USA
| |
Collapse
|
26
|
Xiang Q, Qu L, Lei H, Duan Z, Zhu C, Yuwen W, Ma X, Fan D. Expression of Multicopy Tandem Recombinant Ginseng Hexapeptide in Bacillus subtilis and the Evaluation of Antiaging Activity. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2024; 72:7266-7278. [PMID: 38523338 DOI: 10.1021/acs.jafc.3c09158] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/26/2024]
Abstract
Ginseng oligopeptides are naturally occurring small-molecule peptides extracted from ginseng that exhibit positive effects on health and longevity. However, the current industrial production of ginseng oligopeptides primarily relies on plant extraction and chemical synthesis. In this study, we proposed a novel genetic engineering approach to produce active ginseng peptides through multicopy tandem insertion (5 and 15 times). The recombinant ginseng peptides were successfully produced from engineered Bacillus subtilis with an increasing yield from 356.55 to 2900 mg/L as the repeats multiple. Additionally, an oxidative stress-induced aging model caused by H2O2 was established to evaluate whether the recombinant ginseng peptides, without enzymatic hydrolysis into individual peptides, also have positive effects on antiaging. The results demonstrated that all two kinds of recombinant ginseng peptides could also delay cellular aging through various mechanisms, such as inhibiting cell cycle arrest, suppressing the expression of pro-inflammatory factors, and enhancing cellular antioxidant capacity.
Collapse
Affiliation(s)
- Qingyu Xiang
- Engineering Research Center of Western Resource Innovation Medicine Green Manufacturing, Ministry of Education, School of Chemical Engineering, Northwest University, Xi'an 710069, China
- Shaanxi R&D Center of Biomaterials and Fermentation Engineering, School of Chemical Engineering, Northwest University, Xi'an 710069, China
- Biotech. & Biomed. Research Institute, Northwest University, Xi'an 710069, China
| | - Linlin Qu
- Engineering Research Center of Western Resource Innovation Medicine Green Manufacturing, Ministry of Education, School of Chemical Engineering, Northwest University, Xi'an 710069, China
- Shaanxi R&D Center of Biomaterials and Fermentation Engineering, School of Chemical Engineering, Northwest University, Xi'an 710069, China
- Biotech. & Biomed. Research Institute, Northwest University, Xi'an 710069, China
| | - Huan Lei
- Engineering Research Center of Western Resource Innovation Medicine Green Manufacturing, Ministry of Education, School of Chemical Engineering, Northwest University, Xi'an 710069, China
- Shaanxi R&D Center of Biomaterials and Fermentation Engineering, School of Chemical Engineering, Northwest University, Xi'an 710069, China
- Biotech. & Biomed. Research Institute, Northwest University, Xi'an 710069, China
| | - Zhiguang Duan
- Engineering Research Center of Western Resource Innovation Medicine Green Manufacturing, Ministry of Education, School of Chemical Engineering, Northwest University, Xi'an 710069, China
- Shaanxi R&D Center of Biomaterials and Fermentation Engineering, School of Chemical Engineering, Northwest University, Xi'an 710069, China
- Biotech. & Biomed. Research Institute, Northwest University, Xi'an 710069, China
| | - Chenhui Zhu
- Engineering Research Center of Western Resource Innovation Medicine Green Manufacturing, Ministry of Education, School of Chemical Engineering, Northwest University, Xi'an 710069, China
- Shaanxi R&D Center of Biomaterials and Fermentation Engineering, School of Chemical Engineering, Northwest University, Xi'an 710069, China
- Biotech. & Biomed. Research Institute, Northwest University, Xi'an 710069, China
| | - Weigang Yuwen
- Shaanxi Gaint Biotechnology Co., Ltd, Xi'an 710065, Shaanxi, China
| | - Xiaoxuan Ma
- Engineering Research Center of Western Resource Innovation Medicine Green Manufacturing, Ministry of Education, School of Chemical Engineering, Northwest University, Xi'an 710069, China
- Shaanxi R&D Center of Biomaterials and Fermentation Engineering, School of Chemical Engineering, Northwest University, Xi'an 710069, China
- Biotech. & Biomed. Research Institute, Northwest University, Xi'an 710069, China
| | - Daidi Fan
- Engineering Research Center of Western Resource Innovation Medicine Green Manufacturing, Ministry of Education, School of Chemical Engineering, Northwest University, Xi'an 710069, China
- Shaanxi R&D Center of Biomaterials and Fermentation Engineering, School of Chemical Engineering, Northwest University, Xi'an 710069, China
- Biotech. & Biomed. Research Institute, Northwest University, Xi'an 710069, China
| |
Collapse
|
27
|
Rashid H, Smith CM, Convers V, Clark K, Javed A. Runx2 deletion in hypertrophic chondrocytes impairs osteoclast mediated bone resorption. Bone 2024; 181:117014. [PMID: 38218304 PMCID: PMC10922707 DOI: 10.1016/j.bone.2024.117014] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Revised: 12/27/2023] [Accepted: 01/05/2024] [Indexed: 01/15/2024]
Abstract
Deletion of Runx2 gene in proliferating chondrocytes results in complete failure of endochondral ossification and perinatal lethality. We reported recently that mice with Runx2 deletion specifically in hypertrophic chondrocytes (HCs) using the Col10a1-Cre transgene survive and exhibit enlarged growth plates due to decreased HC apoptosis and cartilage resorption. Bulk of chondrogenesis occurs postnatally, however, the role of Runx2 in HCs during postnatal chondrogenesis is unknown. Despite limb dwarfism, adult homozygous (Runx2HC/HC) mice showed a significant increase in length of growth plate and articular cartilage. Consistent with doubling of the hypertrophic zone, collagen type X expression was increased in Runx2HC/HC mice. In sharp contrast, expression of metalloproteinases and aggrecanases were markedly decreased. Impaired cartilage degradation was evident by the retention of significant amount of safranin-O positive cartilage. Histomorphometry and μCT uncovered increased trabecular bone mass with a significant increase in BV/TV ratio, trabecular number, thickness, and a decrease in trabecular space in Runx2HC/HC mice. To identify if this is due to increased bone synthesis, expression of osteoblast differentiation markers was evaluated and found to be comparable amongst littermates. Histomorphometry confirmed similar number of osteoblasts in the littermates. Furthermore, dynamic bone synthesis showed no differences in mineral apposition or bone formation rates. Surprisingly, three-point-bending test revealed Runx2HC/HC bones to be structurally less strong. Interestingly, both the number and surface of osteoclasts were markedly reduced in Runx2HC/HC littermates. Rankl and IL-17a ligands that promote osteoclast differentiation were markedly reduced in Runx2HC/HC mice. Bone marrow cultures were performed to independently establish Runx2 and hypertrophic chondrocytes role in osteoclast development. The culture from the Runx2HC/HC mice formed significantly fewer and smaller osteoclasts. The expression of mature osteoclast markers, Ctsk and Mmp9, were significantly reduced in the cultures from Runx2HC/HC mice. Thus, Runx2 functions extend beyond embryonic development and chondrocyte hypertrophy by regulating cartilage degradation, osteoclast differentiation, and bone resorption during postnatal endochondral ossification.
Collapse
Affiliation(s)
- Harunur Rashid
- Department of Oral and Maxillofacial Surgery, Institute of Oral Health Research, School of Dentistry, University of Alabama at Birmingham, Birmingham 35233, AL, USA
| | - Caris M Smith
- Department of Oral and Maxillofacial Surgery, Institute of Oral Health Research, School of Dentistry, University of Alabama at Birmingham, Birmingham 35233, AL, USA
| | - Vashti Convers
- Department of Oral and Maxillofacial Surgery, Institute of Oral Health Research, School of Dentistry, University of Alabama at Birmingham, Birmingham 35233, AL, USA
| | - Katelynn Clark
- Department of Oral and Maxillofacial Surgery, Institute of Oral Health Research, School of Dentistry, University of Alabama at Birmingham, Birmingham 35233, AL, USA
| | - Amjad Javed
- Department of Oral and Maxillofacial Surgery, Institute of Oral Health Research, School of Dentistry, University of Alabama at Birmingham, Birmingham 35233, AL, USA.
| |
Collapse
|
28
|
Yongzhen L, Yan G, Jing L, Chenyan R, Chuanqing M, Yun S, Weihui C. Embryonic inhibition of colony-stimulating factor 1 receptor induces enlarged cartilaginous zone of the midpalatal suture in postnatal mice. Orthod Craniofac Res 2024; 27:276-286. [PMID: 37904627 DOI: 10.1111/ocr.12724] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2023] [Revised: 09/03/2023] [Accepted: 10/16/2023] [Indexed: 11/01/2023]
Abstract
OBJECTIVES The midpalatal suture acts as the growth centre of the maxilla. Colony-stimulating factor 1 receptor (CSF1R) is essential for osteoclastogenesis. Deletion of CSF1R, and its ligand, results in significant craniofacial phenotypes but has not been studied in detail in the midpalatal suture. MATERIALS AND METHODS Pregnant ICR mice were treated with the CSF1R inhibitor PLX5622 at embryo Day 14.5 (E14.5) to E17.5. Pups at E18.5, postnatal Day 3 (P3) and P7 were collected for skeletal and histological staining. Osteoclasts were labelled using TRAP staining. PHH3 and TUNEL were employed to detect cell proliferation and apoptosis. Sox9, Ihh, and Col10a1 and Runx2, Col1a1, and DMP1 were used to detect chondrogenic differentiation and osteogenic differentiation, respectively. CD31, MMP9 and CTSK were utilized to assess vascular invasion and osteoclast secretion enzymes, respectively. RESULTS Embryonic inhibition of CSF1R resulted in a depletion of TRAP-positive cells and an enlarged cartilage zone of the midpalatal suture of postnatal mice. Compared to those in the control group, Sox9, Ihh, Col10a1, Runx2 and Col1a1 were upregulated, whereas TUNEL and DMP1 were decreased in this zone. In the trabecular region, Col10a1 was upregulated, while TUNEL, Col1a1 and DMP1 were downregulated. Moreover, the expression of MMP9, CTSK and CD31 was decreased, and invasion into the cartilage zone was delayed. CONCLUSIONS Embryonic inhibition of CSF1R led to an abnormally enlarged cartilaginous zone in the midpalatal suture, potentially due to delayed endochondral ossification caused by the depletion of osteoclasts. Additionally, we established a novel model of midpalatal suture dysplasia, offering prospects for future research.
Collapse
Affiliation(s)
- Lai Yongzhen
- Department of Oral and Cranio-maxillofacial Science, Fujian Medical university Union Hospital, Fuzhou, China
- Stomatological Key Laboratory of Fujian College and University, Fuzhou, China
| | - Guo Yan
- Stomatological Key Laboratory of Fujian College and University, Fuzhou, China
| | - Liu Jing
- Department of Stomatology, Fujian Maternal and Child Health Hospital, Fuzhou, China
| | - Ren Chenyan
- Stomatological Key Laboratory of Fujian College and University, Fuzhou, China
| | - Mao Chuanqing
- Department of Oral and Cranio-maxillofacial Science, Fujian Medical university Union Hospital, Fuzhou, China
| | - Shi Yun
- Stomatological Key Laboratory of Fujian College and University, Fuzhou, China
| | - Chen Weihui
- Department of Oral and Cranio-maxillofacial Science, Fujian Medical university Union Hospital, Fuzhou, China
- Stomatological Key Laboratory of Fujian College and University, Fuzhou, China
| |
Collapse
|
29
|
Zhao Z, Sun X, Tu P, Ma Y, Guo Y, Zhang Y, Liu M, Wang L, Chen X, Si L, Li G, Pan Y. Mechanisms of vascular invasion after cartilage injury and potential engineering cartilage treatment strategies. FASEB J 2024; 38:e23559. [PMID: 38502020 DOI: 10.1096/fj.202302391rr] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2023] [Revised: 02/26/2024] [Accepted: 03/01/2024] [Indexed: 03/20/2024]
Abstract
Articular cartilage injury is one of the most common diseases in orthopedic clinics. Following an articular cartilage injury, an inability to resist vascular invasion can result in cartilage calcification by newly formed blood vessels. This process ultimately leads to the loss of joint function, significantly impacting the patient's quality of life. As a result, developing anti-angiogenic methods to repair damaged cartilage has become a popular research topic. Despite this, tissue engineering, as an anti-angiogenic strategy in cartilage injury repair, has not yet been adequately investigated. This exhaustive literature review mainly focused on the process and mechanism of vascular invasion in articular cartilage injury repair and summarized the major regulatory factors and signaling pathways affecting angiogenesis in the process of cartilage injury. We aimed to discuss several potential methods for engineering cartilage repair with anti-angiogenic strategies. Three anti-angiogenic tissue engineering methods were identified, including administering angiogenesis inhibitors, applying scaffolds to manage angiogenesis, and utilizing in vitro bioreactors to enhance the therapeutic properties of cultured chondrocytes. The advantages and disadvantages of each strategy were also analyzed. By exploring these anti-angiogenic tissue engineering methods, we hope to provide guidance for researchers in related fields for future research and development in cartilage repair.
Collapse
Affiliation(s)
- Zitong Zhao
- School of Integrated Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, P.R. China
- Laboratory of New Techniques of Restoration and Reconstruction of Orthopedics and Traumatology, Nanjing University of Chinese Medicine, Nanjing, P.R. China
| | - Xiaoxian Sun
- Laboratory of New Techniques of Restoration and Reconstruction of Orthopedics and Traumatology, Nanjing University of Chinese Medicine, Nanjing, P.R. China
- Affiliated Hospital, Nanjing University of Chinese Medicine, Nanjing, P.R. China
| | - Pengcheng Tu
- Affiliated Hospital, Nanjing University of Chinese Medicine, Nanjing, P.R. China
| | - Yong Ma
- School of Integrated Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, P.R. China
- Laboratory of New Techniques of Restoration and Reconstruction of Orthopedics and Traumatology, Nanjing University of Chinese Medicine, Nanjing, P.R. China
- Affiliated Hospital, Nanjing University of Chinese Medicine, Nanjing, P.R. China
- Jiangsu CM Clinical Innovation Center of Degenerative Bone & Joint Disease, Wuxi TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Wuxi, P.R. China
| | - Yang Guo
- Laboratory of New Techniques of Restoration and Reconstruction of Orthopedics and Traumatology, Nanjing University of Chinese Medicine, Nanjing, P.R. China
- Affiliated Hospital, Nanjing University of Chinese Medicine, Nanjing, P.R. China
- Jiangsu CM Clinical Innovation Center of Degenerative Bone & Joint Disease, Wuxi TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Wuxi, P.R. China
| | - Yafeng Zhang
- Jiangsu CM Clinical Innovation Center of Degenerative Bone & Joint Disease, Wuxi TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Wuxi, P.R. China
| | - Mengmin Liu
- School of Integrated Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, P.R. China
- Laboratory of New Techniques of Restoration and Reconstruction of Orthopedics and Traumatology, Nanjing University of Chinese Medicine, Nanjing, P.R. China
| | - Lining Wang
- School of Integrated Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, P.R. China
- Laboratory of New Techniques of Restoration and Reconstruction of Orthopedics and Traumatology, Nanjing University of Chinese Medicine, Nanjing, P.R. China
| | - Xinyu Chen
- Affiliated Hospital, Nanjing University of Chinese Medicine, Nanjing, P.R. China
| | - Lin Si
- Affiliated Hospital, Nanjing University of Chinese Medicine, Nanjing, P.R. China
| | - Guangguang Li
- Orthopedics and traumatology department, Yixing Traditional Chinese Medicine Hospital, Yixing, P.R. China
| | - Yalan Pan
- Laboratory of New Techniques of Restoration and Reconstruction of Orthopedics and Traumatology, Nanjing University of Chinese Medicine, Nanjing, P.R. China
| |
Collapse
|
30
|
Pijet B, Kostrzewska-Księzyk A, Pijet-Kucicka M, Kaczmarek L. Matrix Metalloproteinase-9 Contributes to Epilepsy Development after Ischemic Stroke in Mice. Int J Mol Sci 2024; 25:896. [PMID: 38255970 PMCID: PMC10815104 DOI: 10.3390/ijms25020896] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Revised: 01/04/2024] [Accepted: 01/08/2024] [Indexed: 01/24/2024] Open
Abstract
Epilepsy, a neurological disorder affecting over 50 million individuals globally, is characterized by an enduring predisposition and diverse consequences, both neurobiological and social. Acquired epilepsy, constituting 30% of cases, often results from brain-damaging injuries like ischemic stroke. With one third of epilepsy cases being resistant to existing drugs and without any preventive therapeutics for epileptogenesis, identifying anti-epileptogenic targets is crucial. Stroke being a leading cause of acquired epilepsy, particularly in the elderly, prompts the need for understanding post-stroke epileptogenesis. Despite the challenges in studying stroke-evoked epilepsy in rodents due to poor long-term survival rates, in this presented study the use of an animal care protocol allowed for comprehensive investigation. We highlight the role of matrix metalloproteinase-9 (MMP-9) in post-stroke epileptogenesis, emphasizing MMP-9 involvement in mouse models and its potential as a therapeutic target. Using a focal Middle Cerebral Artery occlusion model, this study demonstrates MMP-9 activation following ischemia, influencing susceptibility to seizures. MMP-9 knockout reduces epileptic features, while overexpression exacerbates them. The findings show that MMP-9 is a key player in post-stroke epileptogenesis, presenting opportunities for future therapies and expanding our understanding of acquired epilepsy.
Collapse
Affiliation(s)
- Barbara Pijet
- Laboratory of Neurobiology, Braincity, Nencki Institute of Experimental Biology, Pasteura 3, 02-093 Warsaw, Poland; (A.K.-K.)
| | | | | | | |
Collapse
|
31
|
Al Maruf DSA, Xin H, Cheng K, Garcia AG, Mohseni-Dargah M, Ben-Sefer E, Tomaskovic-Crook E, Crook JM, Clark JR. Bioengineered cartilaginous grafts for repairing segmental mandibular defects. J Tissue Eng 2024; 15. [DOI: 10.1177/20417314241267017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2025] Open
Abstract
Reconstructing critical-sized craniofacial bone defects is a global healthcare challenge. Current methods, like autologous bone transplantation, face limitations. Bone tissue engineering offers an alternative to autologous bone, with traditional approaches focusing on stimulating osteogenesis via the intramembranous ossification (IMO) pathway. However, IMO falls short in addressing larger defects, particularly in clinical scenarios where there is insufficient vascularisation. This review explores redirecting bone regeneration through endochondral ossification (ECO), a process observed in long bone healing stimulated by hypoxic conditions. Despite its promise, gaps exist in applying ECO to bone tissue engineering experiments, requiring the elucidation of key aspects such as cell sources, biomaterials and priming protocols. This review discusses various scaffold biomaterials and cellular sources for chondrogenesis and hypertrophic chondrocyte priming, mirroring the ECO pathway. The review highlights challenges in current endochondral priming and proposes alternative approaches. Emphasis is on segmental mandibular defect repair, offering insights for future research and clinical application. This concise review aims to advance bone tissue engineering by addressing critical gaps in ECO strategies.
Collapse
Affiliation(s)
- D S Abdullah Al Maruf
- Integrated Prosthetics and Reconstruction, Department of Head and Neck Surgery, Chris O’Brien Lifehouse, Camperdown, NSW, Australia
- Central Clinical School, Faculty of Medicine and Health, The University of Sydney, Camperdown, NSW, Australia
| | - Hai Xin
- Central Clinical School, Faculty of Medicine and Health, The University of Sydney, Camperdown, NSW, Australia
| | - Kai Cheng
- Royal Prince Alfred Institute of Academic Surgery, Sydney Local Health District, Camperdown, NSW, Australia
| | - Alejandro Garcia Garcia
- Cell, Tissue and Organ Engineering Laboratory, Biomedical Centre (BMC), Department of Clinical Sciences Lund, Stem Cell Centre, Lund University, Lund, Sweden
| | - Masoud Mohseni-Dargah
- Integrated Prosthetics and Reconstruction, Department of Head and Neck Surgery, Chris O’Brien Lifehouse, Camperdown, NSW, Australia
| | - Eitan Ben-Sefer
- Integrated Prosthetics and Reconstruction, Department of Head and Neck Surgery, Chris O’Brien Lifehouse, Camperdown, NSW, Australia
- Arto Hardy Biomedical Innovation Hub, Chris O`Brien Lifehouse, Camperdown, NSW, Australia
- School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Camperdown, NSW, Australia
| | - Eva Tomaskovic-Crook
- Arto Hardy Biomedical Innovation Hub, Chris O`Brien Lifehouse, Camperdown, NSW, Australia
- School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Camperdown, NSW, Australia
- Sarcoma and Surgical Research Centre, Chris O’Brien Lifehouse, Camperdown, NSW, Australia
- ARC Centre of Excellence for Electromaterials Science, The University of Wollongong, Wollongong, NSW, Australia
- Intelligent Polymer Research Institute, AIIM Facility, The University of Wollongong, Wollongong, NSW, Australia
| | - Jeremy Micah Crook
- Arto Hardy Biomedical Innovation Hub, Chris O`Brien Lifehouse, Camperdown, NSW, Australia
- School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Camperdown, NSW, Australia
- Sarcoma and Surgical Research Centre, Chris O’Brien Lifehouse, Camperdown, NSW, Australia
- ARC Centre of Excellence for Electromaterials Science, The University of Wollongong, Wollongong, NSW, Australia
- Intelligent Polymer Research Institute, AIIM Facility, The University of Wollongong, Wollongong, NSW, Australia
| | - Jonathan Robert Clark
- Integrated Prosthetics and Reconstruction, Department of Head and Neck Surgery, Chris O’Brien Lifehouse, Camperdown, NSW, Australia
- Central Clinical School, Faculty of Medicine and Health, The University of Sydney, Camperdown, NSW, Australia
- Royal Prince Alfred Institute of Academic Surgery, Sydney Local Health District, Camperdown, NSW, Australia
| |
Collapse
|
32
|
Djaziri N, Burel C, Abbad L, Bakey Z, Piedagnel R, Lelongt B. Cleavage of periostin by MMP9 protects mice from kidney cystic disease. PLoS One 2023; 18:e0294922. [PMID: 38039285 PMCID: PMC10691688 DOI: 10.1371/journal.pone.0294922] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Accepted: 11/12/2023] [Indexed: 12/03/2023] Open
Abstract
The matrix metalloproteinase MMP9 influences cellular morphology and function, and plays important roles in organogenesis and disease. It exerts both protective and deleterious effects in renal pathology, depending upon its specific substrates. To explore new functions for MMP9 in kidney cysts formation and disease progression, we generated a mouse model by breeding juvenile cystic kidney (jck) mice with MMP9 deficient mice. Specifically, we provide evidence that MMP9 is overexpressed in cystic tissue where its enzymatic activity is increased 7-fold. MMP9 deficiency in cystic kidney worsen cystic kidney diseases by decreasing renal function, favoring cyst expansion and fibrosis. In addition, we find that periostin is a new critical substrate for MMP9 and in its absence periostin accumulates in cystic lining cells. As periostin promotes renal cyst growth and interstitial fibrosis in polycystic kidney diseases, we propose that the control of periostin by MMP9 and its associated intracellular signaling pathways including integrins, integrin-linked kinase and focal adhesion kinase confers to MMP9 a protective effect on the severity of the disease.
Collapse
Affiliation(s)
- Nabila Djaziri
- Sorbonne Université, Paris, France
- Institut National de la Santé et de la Recherche Médicale (INSERM) Unité Mixte de Recherche (UMR), Paris, France
| | - Cindy Burel
- Sorbonne Université, Paris, France
- Institut National de la Santé et de la Recherche Médicale (INSERM) Unité Mixte de Recherche (UMR), Paris, France
| | - Lilia Abbad
- Sorbonne Université, Paris, France
- Institut National de la Santé et de la Recherche Médicale (INSERM) Unité Mixte de Recherche (UMR), Paris, France
| | - Zeineb Bakey
- Sorbonne Université, Paris, France
- Institut National de la Santé et de la Recherche Médicale (INSERM) Unité Mixte de Recherche (UMR), Paris, France
| | - Rémi Piedagnel
- Sorbonne Université, Paris, France
- Institut National de la Santé et de la Recherche Médicale (INSERM) Unité Mixte de Recherche (UMR), Paris, France
| | - Brigitte Lelongt
- Sorbonne Université, Paris, France
- Institut National de la Santé et de la Recherche Médicale (INSERM) Unité Mixte de Recherche (UMR), Paris, France
| |
Collapse
|
33
|
Chen N, Wu RW, Lam Y, Chan WC, Chan D. Hypertrophic chondrocytes at the junction of musculoskeletal structures. Bone Rep 2023; 19:101698. [PMID: 37485234 PMCID: PMC10359737 DOI: 10.1016/j.bonr.2023.101698] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Revised: 06/12/2023] [Accepted: 07/01/2023] [Indexed: 07/25/2023] Open
Abstract
Hypertrophic chondrocytes are found at unique locations at the junction of skeletal tissues, cartilage growth plate, articular cartilage, enthesis and intervertebral discs. Their role in the skeleton is best understood in the process of endochondral ossification in development and bone fracture healing. Chondrocyte hypertrophy occurs in degenerative conditions such as osteoarthritis. Thus, the role of hypertrophic chondrocytes in skeletal biology and pathology is context dependent. This review will focus on hypertrophic chondrocytes in endochondral ossification, in which they exist in a transient state, but acting as a central regulator of differentiation, mineralization, vascularization and conversion to bone. The amazing journey of a chondrocyte from being entrapped in the extracellular matrix environment to becoming proliferative then hypertrophic will be discussed. Recent studies on the dynamic changes and plasticity of hypertrophic chondrocytes have provided new insights into how we view these cells, not as terminally differentiated but as cells that can dedifferentiate to more progenitor-like cells in a transition to osteoblasts and adipocytes, as well as a source of skeletal stem and progenitor cells residing in the bone marrow. This will provide a foundation for studies of hypertrophic chondrocytes at other skeletal sites in development, tissue maintenance, pathology and therapy.
Collapse
Affiliation(s)
- Ning Chen
- School of Biomedical Sciences, The University of Hong Kong, Hong Kong, China
| | - Robin W.H. Wu
- School of Biomedical Sciences, The University of Hong Kong, Hong Kong, China
| | - Yan Lam
- School of Biomedical Sciences, The University of Hong Kong, Hong Kong, China
| | - Wilson C.W. Chan
- School of Biomedical Sciences, The University of Hong Kong, Hong Kong, China
- Department of Orthopaedics Surgery and Traumatology, The University of Hong Kong-Shenzhen Hospital (HKU-SZH), Shenzhen 518053, China
| | - Danny Chan
- School of Biomedical Sciences, The University of Hong Kong, Hong Kong, China
| |
Collapse
|
34
|
Li X, Li J, Li K, Zhang Z, Wang H. Effects of perchlorate and exogenous T4 exposures on body condition and endochondral ossification of Rana chensinensis tadpoles. AQUATIC TOXICOLOGY (AMSTERDAM, NETHERLANDS) 2023; 265:106767. [PMID: 37972501 DOI: 10.1016/j.aquatox.2023.106767] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/03/2023] [Revised: 10/14/2023] [Accepted: 11/10/2023] [Indexed: 11/19/2023]
Abstract
Perchlorate, as an endocrine-disrupting chemical (EDC), is largely produced and used in the military, fireworks, fertilizers, and other industries and widely exists in water. Although perchlorate is known to destroy the normal function of thyroid hormones (THs) in amphibians and interfere with their growth and development, the impact of TH levels caused by sodium perchlorate (NaClO4) on endochondral ossification and skeletal development is poorly investigated, and the underlying molecular mechanism has not been clarified. The present study aimed to explore the potential effects of NaClO4 and exogenous thyroxine (T4) on the skeletal development of Rana chensinensis tadpoles and elucidate the related molecular mechanisms. Our results showed that histological changes occurred to the femur and tibia-fibula of tadpoles raised in 250 μg/L NaClO4 and 5 μg/L exogenous T4, and the length of their hindlimbs was significantly reduced. In addition, exogenous T4 exposure significantly interfered with the expression of Dio3, TRβ, MMP9, MMP13, and Runx2, inhibiting the endochondral ossification process. Therefore, we provide robust evidence that the changes in TH levels caused by NaClO4 and exogenous T4 will adversely affect the endochondral ossification and skeletal development of R. chensinensis tadpoles.
Collapse
Affiliation(s)
- Xinyi Li
- College of Life Science, Shaanxi Normal University, Xi'an 710119, China
| | - Jiayi Li
- College of Life Science, Shaanxi Normal University, Xi'an 710119, China
| | - Kaiyue Li
- College of Life Science, Shaanxi Normal University, Xi'an 710119, China
| | - Zhiqin Zhang
- Basic Experimental Teaching Center, Shaanxi Normal University, Xi'an 710119, China
| | - Hongyuan Wang
- College of Life Science, Shaanxi Normal University, Xi'an 710119, China.
| |
Collapse
|
35
|
Kalev-Altman R, Becker G, Levy T, Penn S, Shpigel NY, Monsonego-Ornan E, Sela-Donenfeld D. Mmp2 Deficiency Leads to Defective Parturition and High Dystocia Rates in Mice. Int J Mol Sci 2023; 24:16822. [PMID: 38069145 PMCID: PMC10706207 DOI: 10.3390/ijms242316822] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2023] [Revised: 11/14/2023] [Accepted: 11/22/2023] [Indexed: 12/18/2023] Open
Abstract
Parturition is the final and essential step for mammalian reproduction. While the uterus is quiescent during pregnancy, fundamental changes arise in the myometrial contractility, inducing fetal expulsion. Extracellular matrix (ECM) remodeling is fundamental for these events. The gelatinases subgroup of matrix metalloproteinases (MMPs), MMP2 and MMP9, participate in uterine ECM remodeling throughout pregnancy and parturition. However, their loss-of-function effect is unknown. Here, we determined the result of eliminating Mmp2 and/or Mmp9 on parturition in vivo, using single- and double-knockout (dKO) mice. The dystocia rates were measured in each genotype, and uterine tissue was collected from nulliparous synchronized females at the ages of 2, 4, 9 and 12 months. Very high percentages of dystocia (40-55%) were found in the Mmp2-/- and dKO females, contrary to the Mmp9-/- and wild-type females. The histological analysis of the uterus and cervix revealed that Mmp2-/- tissues undergo marked structural alterations, including highly enlarged myometrial, endometrial and luminal cavity. Increased collagen deposition was also demonstrated, suggesting a mechanism of extensive fibrosis in the Mmp2-/- myometrium, which may result in dystocia. Overall, this study describes a new role for MMP2 in myometrium remodeling during mammalian parturition process, highlighting a novel cause for dystocia due to a loss in MMP2 activity in the uterine tissue.
Collapse
Affiliation(s)
- Rotem Kalev-Altman
- The Koret School of Veterinary Medicine, The RH Smith Faculty of Agriculture, Food and Environment, The Hebrew University of Jerusalem, Rehovot 7610001, Israel (N.Y.S.)
- The Institute of Biochemistry, Food Science and Nutrition, The RH Smith Faculty of Agriculture, Food and Environment, The Hebrew University of Jerusalem, Rehovot 7610001, Israel (E.M.-O.)
| | - Gal Becker
- The Institute of Biochemistry, Food Science and Nutrition, The RH Smith Faculty of Agriculture, Food and Environment, The Hebrew University of Jerusalem, Rehovot 7610001, Israel (E.M.-O.)
| | - Tamar Levy
- The Koret School of Veterinary Medicine, The RH Smith Faculty of Agriculture, Food and Environment, The Hebrew University of Jerusalem, Rehovot 7610001, Israel (N.Y.S.)
| | - Svetlana Penn
- The Institute of Biochemistry, Food Science and Nutrition, The RH Smith Faculty of Agriculture, Food and Environment, The Hebrew University of Jerusalem, Rehovot 7610001, Israel (E.M.-O.)
| | - Nahum Y. Shpigel
- The Koret School of Veterinary Medicine, The RH Smith Faculty of Agriculture, Food and Environment, The Hebrew University of Jerusalem, Rehovot 7610001, Israel (N.Y.S.)
| | - Efrat Monsonego-Ornan
- The Institute of Biochemistry, Food Science and Nutrition, The RH Smith Faculty of Agriculture, Food and Environment, The Hebrew University of Jerusalem, Rehovot 7610001, Israel (E.M.-O.)
| | - Dalit Sela-Donenfeld
- The Koret School of Veterinary Medicine, The RH Smith Faculty of Agriculture, Food and Environment, The Hebrew University of Jerusalem, Rehovot 7610001, Israel (N.Y.S.)
| |
Collapse
|
36
|
Alruwaili N, Kandhi S, Froogh G, Kelly MR, Sun D, Wolin MS. Superoxide-Mediated Upregulation of MMP9 Participates in BMPR2 Destabilization and Pulmonary Hypertension Development. Antioxidants (Basel) 2023; 12:1961. [PMID: 38001814 PMCID: PMC10669489 DOI: 10.3390/antiox12111961] [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: 09/18/2023] [Revised: 10/25/2023] [Accepted: 10/31/2023] [Indexed: 11/26/2023] Open
Abstract
BACKGROUND AND AIMS we previously reported in studies on organoid-cultured bovine pulmonary arteries that pulmonary hypertension (PH) conditions of exposure to hypoxia or endothelin-1 caused a loss of a cartilage oligomeric matrix protein (COMP) stabilization of bone morphogenetic protein receptor-2 (BMPR2) function, a known key process contributing to pulmonary hypertension development. Based on subsequent findings, these conditions were associated with an extracellular superoxide-mediated increase in matrix metalloproteinase 9 (MMP-9) expression. We investigated if this contributed to PH development using mice deficient in MMP9. RESULTS wild-type (WT) mice exposed to Sugen/Hypoxia (SuHx) to induce PH had increased levels of MMP9 in their lungs. Hemodynamic measures from MMP9 knockout mice (MMP9 KO) indicated they had attenuated PH parameters compared to WT mice based on an ECHO assessment of pulmonary artery pressure, right ventricular systolic pressure, and Fulton index hypertrophy measurements. In vitro vascular reactivity studies showed impaired endothelium-dependent and endothelium-independent NO-associated vasodilatory responses in the pulmonary arteries of SuHx mice and decreased lung levels of COMP and BMPR2 expression. These changes were attenuated in MMP9 KO mice potentially through preserving COMP-dependent stabilization of BMPR2. INNOVATION this study supports a new function of superoxide in increasing MMP9 and the associated impairment of BMPR2 in promoting PH development which could be a target for future therapies. CONCLUSION superoxide, through promoting increases in MMP9, mediates BMPR2 depletion and its consequent control of vascular function in response to PH mediators and the SuHx mouse model of PH.
Collapse
Affiliation(s)
- Norah Alruwaili
- Department of Physiology, New York Medical College, Valhalla, NY 10595, USA; (N.A.); (D.S.)
- Department of Basic Sciences, College of Science and Health Professions, King Saud Bin Abdulaziz for Health Sciences, Riyadh 11481, Saudi Arabia
| | - Sharath Kandhi
- Department of Physiology, New York Medical College, Valhalla, NY 10595, USA; (N.A.); (D.S.)
| | - Ghezal Froogh
- Department of Physiology, New York Medical College, Valhalla, NY 10595, USA; (N.A.); (D.S.)
| | - Melissa R. Kelly
- Department of Physiology, New York Medical College, Valhalla, NY 10595, USA; (N.A.); (D.S.)
| | - Dong Sun
- Department of Physiology, New York Medical College, Valhalla, NY 10595, USA; (N.A.); (D.S.)
| | - Michael S. Wolin
- Department of Physiology, New York Medical College, Valhalla, NY 10595, USA; (N.A.); (D.S.)
| |
Collapse
|
37
|
Jordan TJ, Chen J, Li N, Burette S, Wan L, Chen L, Culton DA, Geng S, Googe P, Thomas NE, Diaz LA, Liu Z. The Eotaxin-1/CCR3 Axis and Matrix Metalloproteinase-9 Are Critical in Anti-NC16A IgE-Induced Bullous Pemphigoid. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2023; 211:1216-1223. [PMID: 37672029 PMCID: PMC10592335 DOI: 10.4049/jimmunol.2300080] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Accepted: 07/13/2023] [Indexed: 09/07/2023]
Abstract
Bullous pemphigoid (BP) is the most common autoimmune bullous skin disease of humans and is characterized by eosinophilic inflammation and circulating and tissue-bound IgG and IgE autoantibodies directed against two hemidesmosomal proteins: BP180 and BP230. The noncollagenous 16A domain (NC16A) of BP180 has been found to contain major epitopes recognized by autoantibodies in BP. We recently established the pathogenicity of anti-NC16A IgE through passive transfer of patient-derived autoantibodies to double-humanized mice that express the human high-affinity IgE receptor, FcεRI, and human NC16A domain (FcεRI/NC16A). In this model, anti-NC16A IgEs recruit eosinophils to mediate tissue injury and clinical disease in FcεRI/NC16A mice. The objective of this study was to characterize the molecular and cellular events that underlie eosinophil recruitment and eosinophil-dependent tissue injury in anti-NC16A IgE-induced BP. We show that anti-NC16A IgEs significantly increase levels of key eosinophil chemoattractants, eotaxin-1 and eotaxin-2, as well as the proteolytic enzyme matrix metalloproteinase-9 (MMP-9) in the lesional skin of FcεRI/NC16A mice. Importantly, neutralization of eotaxin-1, but not eotaxin-2, and blockade of the main eotaxin receptor, CCR3, drastically reduce anti-NC16A IgE-induced disease activity. We further show that anti-NC16A IgE/NC16A immune complexes induce the release of MMP-9 from eosinophils, and that MMP-9-deficient mice are resistant to anti-NC16A IgE-induced BP. Lastly, we find significantly increased levels of eotaxin-1, eotaxin-2, and MMP-9 in blister fluids of BP patients. Taken together, this study establishes the eotaxin-1/CCR3 axis and MMP-9 as key players in anti-NC16A IgE-induced BP and candidate therapeutic targets for future drug development and testing.
Collapse
Affiliation(s)
- Tyler J.M. Jordan
- Departments of Dermatology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Department of Clinical Sciences, North Carolina State University, Raleigh, NC 27607, USA
| | - Jinbo Chen
- Departments of Dermatology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Department of Dermatology, Wuhan No. 1 Hospital, Wuhan 430022, China
| | - Ning Li
- Departments of Dermatology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Susan Burette
- Departments of Dermatology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Li Wan
- Department of Dermatology, Wuhan No. 1 Hospital, Wuhan 430022, China
- Dermatology Hospital of Southern Medical University, Guangzhou 510091, China
| | - Liuqing Chen
- Department of Dermatology, Wuhan No. 1 Hospital, Wuhan 430022, China
| | - Donna A. Culton
- Departments of Dermatology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Songmei Geng
- Department of Dermatology, The Second Affiliated Hospital, School of Medicine, Xi’an Jiaotong University, Xi’an, Shaanxi 710004, China
| | - Paul Googe
- Departments of Dermatology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Nancy E. Thomas
- Departments of Dermatology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Luis A. Diaz
- Departments of Dermatology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Zhi Liu
- Departments of Dermatology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| |
Collapse
|
38
|
Cho CY, Kang SH, Kim BC, Kim TK, Kim JH, Kim M, Sohn Y, Jung HS. Gleditsiae fructus regulates osteoclastogenesis by inhibiting the c‑Fos/NFATc1 pathway and alleviating bone loss in an ovariectomy model. Mol Med Rep 2023; 28:187. [PMID: 37594079 PMCID: PMC10463233 DOI: 10.3892/mmr.2023.13074] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Accepted: 07/21/2023] [Indexed: 08/19/2023] Open
Abstract
Medical and economic developments have allowed the human lifespan to extend and, as a result, the elderly population has increased worldwide. Osteoporosis is a common geriatric disease that has no symptoms and even a small impact can cause fractures in patients, leading to a serious deterioration in the quality of life. Osteoporosis treatment typically involves bisphosphonates and selective estrogen receptor modulators. However, these treatments are known to cause severe side effects, such as mandibular osteonecrosis and breast cancer, if used for an extended period of time. Therefore, it is essential to develop therapeutic agents from natural products that have fewer side effects. Gleditsiae fructus (GF) is a dried or immature fruit of Gleditsia sinensis Lam. and is composed of various triterpenoid saponins. The anti‑inflammatory effect of GF has been confirmed in various diseases, and since the anti‑inflammatory effect plays a major role in inhibiting osteoclast differentiation, GF was expected to be effective in osteoclast differentiation and menopausal osteoporosis; however, to the best of our knowledge, it has not yet been studied. Therefore, the present study was designed to examine the effect of GF on osteoclastogenesis and to investigate the mechanism underlying inhibition of osteoclast differentiation. The effects of GF on osteoclastogenesis were determined in vitro by tartrate‑resistant acid phosphatase (TRAP) staining, pit formation assays, filamentous actin (F‑actin) ring formation assays, western blotting and reverse transcription‑quantitative PCR analyses. Furthermore, the administration of GF to an animal model exhibiting menopausal osteoporosis allowed for the analysis of alterations in the bone microstructure of the femur using micro‑CT. Additionally, assessments of femoral tissue and serum were conducted. The present study revealed that the administration of GF resulted in a reduction in osteoclast levels, F‑actin rings, TRAP activity and pit area. Furthermore, GF showed a dose‑dependent suppression of nuclear factor of activated T‑cells cytoplasmic, c‑Fos and other osteoclastogenesis‑related markers.
Collapse
Affiliation(s)
- Chang-Young Cho
- Department of Anatomy, College of Korean Medicine, Kyung Hee University, Seoul 02-447, Republic of Korea
| | - Se Hwang Kang
- Department of Anatomy, College of Korean Medicine, Kyung Hee University, Seoul 02-447, Republic of Korea
| | - Byung-Chan Kim
- Department of Anatomy, College of Korean Medicine, Kyung Hee University, Seoul 02-447, Republic of Korea
| | - Tae-Kyu Kim
- Department of Anatomy, College of Korean Medicine, Kyung Hee University, Seoul 02-447, Republic of Korea
| | - Jae-Hyun Kim
- Department of Anatomy, College of Korean Medicine, Kyung Hee University, Seoul 02-447, Republic of Korea
| | - Minsun Kim
- Department of Anatomy, College of Korean Medicine, Kyung Hee University, Seoul 02-447, Republic of Korea
| | - Youngjoo Sohn
- Department of Anatomy, College of Korean Medicine, Kyung Hee University, Seoul 02-447, Republic of Korea
| | - Hyuk-Sang Jung
- Department of Anatomy, College of Korean Medicine, Kyung Hee University, Seoul 02-447, Republic of Korea
| |
Collapse
|
39
|
Yang MC, Nakamura M, Kageyama Y, Igari Y, Sasano Y. Age-Related Gene and Protein Expression in Mouse Mandibular Condyle Analyzed by Cap Analysis of Gene Expression and Immunohistochemistry. Gerontology 2023; 69:1295-1306. [PMID: 37769633 DOI: 10.1159/000533921] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2022] [Accepted: 08/29/2023] [Indexed: 10/03/2023] Open
Abstract
INTRODUCTION Aging, an inevitable physiological process, leads to morphological and histological degenerative changes in the mandibular condylar cartilage (MCC); however, the molecular mechanism has not yet been elucidated, and little information is available on age-related factors. Therefore, this study was designed to identify age-related factors by investigating the age-related differentially expressed genes (DEGs) and localization of their translated protein expression in the mandibular condyle. METHODS Mandibular condyles were collected from 10- and 50-week-old mice. Total RNA was extracted from the samples and then analyzed using cap analysis of gene expression (CAGE) to identify age-related DEGs. Gene ontology (GO) enrichment analysis was performed to determine which biological processes were most affected by aging in terms of gene expression using Metascape. The mandibular condyle samples were processed for histology to investigate morphological changes caused by aging and for immunohistochemistry to localize the protein expression encoded by age-related genes identified with CAGE. Semi-quantitative immunohistochemistry was performed to assess age-related extracellular matrix (ECM) protein levels in the MCC. The histological sections were also used for Alcian blue histochemistry to detect glycosaminoglycans (GAGs). RESULTS GO enrichment analysis revealed that the genes related to "extracellular matrix organization," including Acan, Col1a1, Col1a2, Col2a1, Mmp3, Mmp9, and Mmp13, were most differentially expressed in the aged mandibular condyle. Among these seven genes, Mmp3 was upregulated, and the others were downregulated with aging. Histological examination showed the age-related morphological and histological changes in the MCC. Immunohistochemical investigation showed the localization of matrix metalloproteinases (MMPs)-3, -9, and -13 and their substrate proteins, aggrecan, type I collagen, and type II collagen, in the mandibular condyle at 10 and 50 weeks, indicating different localizations between the young and the aged. In the aged MCC, semi-quantitative immunohistochemistry showed a significant decrease in the aggrecan protein level, and Alcian blue histochemistry showed a decrease in GAGs. CONCLUSION MMP-3, MMP-9, and MMP-13 contribute to the remodeling of the ECM of the MCC and subchondral bone during aging by degrading ECM proteins at specific times and sites under the regulation of their production and secretion.
Collapse
Affiliation(s)
- Mu-Chen Yang
- Division of Craniofacial Development and Tissue Biology, Tohoku University Graduate School of Dentistry, Sendai, Japan
| | - Megumi Nakamura
- Division of Craniofacial Development and Tissue Biology, Tohoku University Graduate School of Dentistry, Sendai, Japan
| | - Yoko Kageyama
- Division of Aging and Geriatric Dentistry, Tohoku University Graduate School of Dentistry, Sendai, Japan
| | - Yohei Igari
- Division of Aging and Geriatric Dentistry, Tohoku University Graduate School of Dentistry, Sendai, Japan
| | - Yasuyuki Sasano
- Division of Craniofacial Development and Tissue Biology, Tohoku University Graduate School of Dentistry, Sendai, Japan
| |
Collapse
|
40
|
Yadav PS, Papaioannou G, Kobelski MM, Demay MB. Phosphate-induced activation of VEGFR2 leads to caspase-9-mediated apoptosis of hypertrophic chondrocytes. iScience 2023; 26:107548. [PMID: 37636062 PMCID: PMC10450517 DOI: 10.1016/j.isci.2023.107548] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2023] [Revised: 06/21/2023] [Accepted: 08/02/2023] [Indexed: 08/29/2023] Open
Abstract
Low circulating phosphate (Pi) leads to rickets, characterized by expansion of the hypertrophic chondrocytes (HCs) in the growth plate due to impaired HC apoptosis. Studies in HCs demonstrate that Pi activates the Raf/MEK/ERK1/2 and mitochondrial apoptotic pathways. To determine how Pi activates these pathways, a small-molecule screen was undertaken to identify inhibitors of Pi-induced ERK1/2 phosphorylation in HCs. Vascular endothelial growth factor receptor 2 (VEGFR2) was identified as a target. In vitro studies in HCs demonstrate that VEGFR2 inhibitors block Pi-induced pERK1/2 and caspase-9 cleavage. Like Pi, rhVEGF activates ERK1/2 and caspase-9 in HCs and induces phosphorylation of VEGFR2, confirming that Pi activates this signaling pathway in HCs. Chondrocyte-specific depletion of VEGFR2 leads to an increase in HCs, impaired vascular invasion, and a decrease in HC apoptosis. Thus, these studies define a role for VEGFR2 in transducing Pi signals and mediating its effects on growth plate maturation.
Collapse
Affiliation(s)
- Prem Swaroop Yadav
- Endocrine Unit, Massachusetts General Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Garyfallia Papaioannou
- Endocrine Unit, Massachusetts General Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | | | - Marie B. Demay
- Endocrine Unit, Massachusetts General Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| |
Collapse
|
41
|
Markina E, Tyrina E, Ratushnyy A, Andreeva E, Buravkova L. Heterotypic Cell Culture from Mouse Bone Marrow under Simulated Microgravity: Lessons for Stromal Lineage Functions. Int J Mol Sci 2023; 24:13746. [PMID: 37762048 PMCID: PMC10531336 DOI: 10.3390/ijms241813746] [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: 07/31/2023] [Revised: 08/25/2023] [Accepted: 08/30/2023] [Indexed: 09/29/2023] Open
Abstract
Muscle and skeleton structures are considered most susceptible to negative factors of spaceflights, namely microgravity. Three-dimensional clinorotation is a ground-based simulation of microgravity. It provides an opportunity to elucidate the effects of microgravity at the cellular level. The extracellular matrix (ECM) content, transcriptional profiles of genes encoding ECM and remodelling molecules, and secretory profiles were investigated in a heterotypic primary culture of bone marrow cells after 14 days of 3D clinorotation. Simulated microgravity negatively affected stromal lineage cells, responsible for bone tissue formation. This was evidenced by the reduced ECM volume and stromal cell numbers, including multipotent mesenchymal stromal cells (MSCs). ECM genes encoding proteins responsible for matrix stiffness and cell-ECM contacts were downregulated. In a heterotypic population of bone marrow cells, the upregulation of genes encoding ECM degrading molecules and the formation of a paracrine profile that can stimulate ECM degradation, may be mechanisms of osteodegenerative events that develop in real spaceflight.
Collapse
Affiliation(s)
- Elena Markina
- Cell Physiology Laboratory, Institute of Biomedical Problems, Russian Academy of Sciences, 123007 Moscow, Russia; (E.T.); (A.R.); (L.B.)
| | | | | | - Elena Andreeva
- Cell Physiology Laboratory, Institute of Biomedical Problems, Russian Academy of Sciences, 123007 Moscow, Russia; (E.T.); (A.R.); (L.B.)
| | | |
Collapse
|
42
|
Yang X, Zhou T, Wang X, Xia Y, Cao X, Cheng X, Cao Y, Ma P, Ma H, Qin A, Zhao J. Loss of DDRGK1 impairs IRE1α UFMylation in spondyloepiphyseal dysplasia. Int J Biol Sci 2023; 19:4709-4725. [PMID: 37781516 PMCID: PMC10539710 DOI: 10.7150/ijbs.82765] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2023] [Accepted: 08/27/2023] [Indexed: 10/03/2023] Open
Abstract
Spondyloepiphyseal dysplasia (SEMD) is a rare disease in which cartilage growth is disrupted, and the DDRGK1 mutation is one of the causative genes. In our study, we established Ddrgk1fl/fl, Col2a1-ERT Cre mice, which showed a thickened hypertrophic zone (HZ) in the growth plate, simulating the previous reported SEMD pathology in vivo. Instead of the classical modulation mechanism towards SOX9, our further mechanism study found that DDRGK1 stabilizes the stress sensor endoplasmic reticulum-to-nucleus signaling 1 (IRE1α) to maintain endoplasmic reticulum (ER) homoeostasis. The loss of DDRGK1 decreased the UFMylation and subsequently led to increased ubiquitylation-mediated IRE1α degradation, causing ER dysfunction and activating the PERK/CHOP/Caspase3 apoptosis pathway. Further DDRGK1 K268R-mutant mice revealed the importance of K268 UFMylation site in IRE1α degradation and subsequent ER dysfunction. In conclusion, DDRGK1 stabilizes IRE1α to ameliorate ER stress and following apoptosis in chondrocytes, which finally promote the normal chondrogenesis.
Collapse
Affiliation(s)
- Xiao Yang
- Shanghai Key Laboratory of Orthopedic Implants, Department of Orthopedics, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Tangjun Zhou
- Shanghai Key Laboratory of Orthopedic Implants, Department of Orthopedics, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xin Wang
- Shanghai Key Laboratory of Orthopedic Implants, Department of Orthopedics, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ying Xia
- Institute of Precision Medicine, The Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiankun Cao
- Shanghai Key Laboratory of Orthopedic Implants, Department of Orthopedics, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiaofei Cheng
- Shanghai Key Laboratory of Orthopedic Implants, Department of Orthopedics, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yu Cao
- Shanghai Key Laboratory of Orthopedic Implants, Department of Orthopedics, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Institute of Precision Medicine, The Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Peixiang Ma
- Shanghai Key Laboratory of Orthopedic Implants, Department of Orthopedics, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Hui Ma
- Shanghai Key Laboratory of Orthopedic Implants, Department of Orthopedics, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - An Qin
- Shanghai Key Laboratory of Orthopedic Implants, Department of Orthopedics, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jie Zhao
- Shanghai Key Laboratory of Orthopedic Implants, Department of Orthopedics, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
43
|
Weng T, Yang M, Zhang W, Jin R, Xia S, Zhang M, Wu P, He X, Han C, Zhao X, Wang X. Dual gene-activated dermal scaffolds regulate angiogenesis and wound healing by mediating the coexpression of VEGF and angiopoietin-1. Bioeng Transl Med 2023; 8:e10562. [PMID: 37693053 PMCID: PMC10487340 DOI: 10.1002/btm2.10562] [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: 06/30/2022] [Revised: 05/22/2023] [Accepted: 06/01/2023] [Indexed: 09/12/2023] Open
Abstract
The vascularization of dermal substitutes is a key challenge in efforts to heal deep skin defects. In this study, dual gene-activated dermal scaffolds (DGADSs-1) were fabricated by loading nanocomposite particles of polyethylenimine (PEI)/multiple plasmid DNAs (pDNAs) encoding vascular endothelial growth factor and angiopoietin-1 at a ratio of 1:1. In a similar manner, DGADSs-2 were loaded with a chimeric plasmid encoding both VEGF and Ang-1. In vitro studies showed that both types of DGADSs released PEI/pDNA nanoparticles in a sustained manner; they demonstrated effective transfection ability, leading to upregulated expression of VEGF and Ang-1. Furthermore, both types of DGADSs promoted fibroblast proliferation and blood vessel formation, although DGADSs-1 showed a more obvious promotion effect. A rat full-thickness skin defect model showed that split-thickness skin transplanted using a one-step method could achieve full survival at the 12th day after surgery in both DGADSs-1 and DGADSs-2 groups, and the vascularization time of dermal substitutes was significantly shortened. Compared with the other three groups of scaffolds, the DGADSs-1 group had significantly greater cell infiltration, collagen deposition, neovascularization, and vascular maturation, all of which promoted wound healing. Thus, compared with single-gene-activated dermal scaffolds, DGADSs show greater potential for enhancing angiogenesis. DGADSs with different loading modes also exhibited differences in terms of angiogenesis; the effect of loading two genes (DGADSs-1) was better than the effect of loading a chimeric gene (DGADSs-2). In summary, DGADSs, which continuously upregulate VEGF and Ang-1 expression, offer a new functional tissue-engineered dermal substitute with the ability to activate vascularization.
Collapse
Affiliation(s)
- Tingting Weng
- Department of Burns & Wound Care CentreSecond Affiliated Hospital of Zhejiang University School of MedicineHangzhouChina
- The Key Laboratory of Severe Trauma and Burns of Zhejiang ProvinceHangzhouChina
- Department of Burn and Plastic SurgeryChildren's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, National Children's Regional Medical CenterHangzhouChina
| | - Min Yang
- Department of Burns & Wound Care CentreSecond Affiliated Hospital of Zhejiang University School of MedicineHangzhouChina
- The Key Laboratory of Severe Trauma and Burns of Zhejiang ProvinceHangzhouChina
| | - Wei Zhang
- Department of Burns & Wound Care CentreSecond Affiliated Hospital of Zhejiang University School of MedicineHangzhouChina
- The Key Laboratory of Severe Trauma and Burns of Zhejiang ProvinceHangzhouChina
| | - Ronghua Jin
- Department of Burns & Wound Care CentreSecond Affiliated Hospital of Zhejiang University School of MedicineHangzhouChina
- The Key Laboratory of Severe Trauma and Burns of Zhejiang ProvinceHangzhouChina
| | - Sizhan Xia
- Department of Burns & Wound Care CentreSecond Affiliated Hospital of Zhejiang University School of MedicineHangzhouChina
- The Key Laboratory of Severe Trauma and Burns of Zhejiang ProvinceHangzhouChina
| | - Manjia Zhang
- The First Clinical Medical College, Zhejiang Chinese Medical UniversityHangzhouChina
| | - Pan Wu
- Department of Burns & Wound Care CentreSecond Affiliated Hospital of Zhejiang University School of MedicineHangzhouChina
- The Key Laboratory of Severe Trauma and Burns of Zhejiang ProvinceHangzhouChina
| | - Xiaojie He
- Department of Burns & Wound Care CentreSecond Affiliated Hospital of Zhejiang University School of MedicineHangzhouChina
- The Key Laboratory of Severe Trauma and Burns of Zhejiang ProvinceHangzhouChina
| | - Chunmao Han
- Department of Burns & Wound Care CentreSecond Affiliated Hospital of Zhejiang University School of MedicineHangzhouChina
- The Key Laboratory of Severe Trauma and Burns of Zhejiang ProvinceHangzhouChina
| | - Xiong Zhao
- Department of Burn and Plastic SurgeryChildren's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, National Children's Regional Medical CenterHangzhouChina
| | - Xingang Wang
- Department of Burns & Wound Care CentreSecond Affiliated Hospital of Zhejiang University School of MedicineHangzhouChina
- The Key Laboratory of Severe Trauma and Burns of Zhejiang ProvinceHangzhouChina
| |
Collapse
|
44
|
Khoswanto C. Role of matrix metalloproteinases in bone regeneration: Narrative review. J Oral Biol Craniofac Res 2023; 13:539-543. [PMID: 37351418 PMCID: PMC10282173 DOI: 10.1016/j.jobcr.2023.06.002] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Revised: 05/14/2023] [Accepted: 06/07/2023] [Indexed: 06/24/2023] Open
Abstract
Background Matrix metalloproteinases (MMPs) not only work as enzymes but also as degrading enzymes that have been shown to play an important function in extracellular matrix (ECM) regeneration, including bone regeneration. To generate new bone tissue, bone regeneration or repair relies on a series of regulated processes in which MMPs play an important role. Bone cells express the MMPs in an active state, and these MMPs are assumed to have a crucial role, not only for the viability and functionality of osteoclasts, osteoblasts, and osteocytes but also for the formation and development of chondrocytes. Objective This study aimed to review and present the roles of matrix metalloproteinases in bone regeneration. Methods An analysis of the scientific literature on the topics of matrix metalloproteinases in bone regeneration was done on PubMed and Google Scholar. Search results were screened for articles that described or investigated the impacts matrix metalloproteinases have on bones in relation to dentistry. The journals' cited papers were also assessed for relevance and included if they complied with the criteria for inclusion. Accessibility to the full document was one of the prerequisites for admission. Result Bone regeneration are intricate ongoing processes involving numerous MMPs, especially MMP 2, 9 and 13. MMP-2 appears to alter bone growth through influencing osteoclast and osteoblast activity and proliferation, MMP-9-deficient animals have abnormal bone formation exclusively during endochondral ossification, MMP 13 is responsible for osteoclast receptor activation, has been linked to the breakdown bone resorption. Conclusions MMP 2, 9, and 13 play a major protective role in osteogenesis and bone regeneration.
Collapse
Affiliation(s)
- Christian Khoswanto
- Department of Oral Biology Faculty of Dentistry, Airlangga University. Jln. Mayjend. Prof. Dr. Moestopo No. 47, Surabaya, 60132, Indonesia
| |
Collapse
|
45
|
Al-Roub A, Akhter N, Al-Rashed F, Wilson A, Alzaid F, Al-Mulla F, Sindhu S, Ahmad R. TNFα induces matrix metalloproteinase-9 expression in monocytic cells through ACSL1/JNK/ERK/NF-kB signaling pathways. Sci Rep 2023; 13:14351. [PMID: 37658104 PMCID: PMC10474281 DOI: 10.1038/s41598-023-41514-6] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2023] [Accepted: 08/28/2023] [Indexed: 09/03/2023] Open
Abstract
Studies have established the association between increased plasma levels of matrix metalloproteinase (MMP)-9 and adipose tissue inflammation. Tumor necrosis factor α (TNFα) was elevated in obesity and is involved in the induction of MMP-9 in monocytic cells. However, the underlying molecular mechanism was incompletely understood. As per our recent report, TNFα mediates inflammatory responses through long-chain acyl-CoA synthetase 1 (ACSL1). Therefore, we further investigated the role of ACSL1 in TNFα-mediated MMP-9 secretion in monocytic cells. THP-1 cells and primary monocytes were used to study MMP-9 expression. mRNA and protein levels of MMP-9 were determined by qRT-PCR and ELISA, respectively. Signaling pathways were studied using Western blotting, inhibitors, and NF-kB/AP1 reporter cells. We found that THP-1 cells and primary human monocytes displayed increased MMP-9 mRNA expression and protein secretion after incubation with TNFα. ACSL1 inhibition using triacsin C significantly reduced the expression of MMP-9 in the THP-1 cells. However, the inhibition of β-oxidation and ceramide biosynthesis did not affect the TNFα-induced MMP-9 production. Using small interfering RNA-mediated ACSL1 knockdown, we further confirmed that TNFα-induced MMP-9 expression/secretion was significantly reduced in ACSL1-deficient cells. TNFα-mediated MMP-9 expression was also significantly reduced by the inhibition of ERK1/ERK2, JNK, and NF-kB. We further observed that TNFα induced phosphorylation of SAPK/JNK (p54/46), ERK1/2 (p44/42 MAPK), and NF-kB p65. ACSL1 inhibition reduced the TNFα-mediated phosphorylation of SAPK/JNK, c-Jun, ERK1/2, and NF-kB. In addition, increased NF-κB/AP-1 activity was inhibited in triacsin C treated cells. Altogether, our findings suggest that ACSL1/JNK/ERK/NF-kB axis plays an important role in the regulation of MMP-9 induced by TNFα in monocytic THP-1 cells.
Collapse
Affiliation(s)
- Areej Al-Roub
- Immunology and Microbiology Department, Dasman Diabetes Institute, Kuwait City, Kuwait
| | - Nadeem Akhter
- Immunology and Microbiology Department, Dasman Diabetes Institute, Kuwait City, Kuwait
| | - Fatema Al-Rashed
- Immunology and Microbiology Department, Dasman Diabetes Institute, Kuwait City, Kuwait
| | - Ajit Wilson
- Immunology and Microbiology Department, Dasman Diabetes Institute, Kuwait City, Kuwait
| | - Fawaz Alzaid
- Bioenergetic Department, Dasman Diabetes Institute, 15462, Dasman, Kuwait
- Enfants Malades (INEM), INSERM U1151/CNRS UMRS8253, IMMEDIAB, Université de Paris Cité, 75015, Paris, France
| | - Fahd Al-Mulla
- Genetics and Bioinformatics, Dasman Diabetes Institute, 15462, Dasman, Kuwait
| | - Sardar Sindhu
- Immunology and Microbiology Department, Dasman Diabetes Institute, Kuwait City, Kuwait
- Animal and Imaging Core Facility, Dasman Diabetes Institute, 15462, Dasman, Kuwait
| | - Rasheed Ahmad
- Immunology and Microbiology Department, Dasman Diabetes Institute, Kuwait City, Kuwait.
| |
Collapse
|
46
|
Abstract
Angiogenesis in the bone is unique and involves distinctive signals. Whether they are created through intramembranous ossification or endochondral ossification, bones are highly vascularized tissues. Long bones undergo a sequence of processes known as endochondral osteogenesis. Angiogenesis occurs during the creation of endochondral bone and is mediated by a variety of cells and factors. An initially avascular cartilage template is invaded by blood vessels from the nearby subchondral bone thanks to the secreted angiogenic chemicals by hypertrophic chondrocytes. Vascular endothelial growth factor (VEGF), one of several angiogenic molecules, is a significant regulator of blood vessel invasion, cartilage remodeling, and ossification of freshly created bone matrix; chondrocyte proliferation and hypertrophy are facilitated by the production of VEGFA and VEGF receptor-2 (VEGFR-2), which is stimulated by fibroblast growth factors (FGFs). NOTCH signaling controls blood capillaries formation during bone maturation and regeneration, while hypoxia-inducible factor 1 alpha (HIF1-a) promotes chondrocyte development by switching to anaerobic metabolism. To control skeletal remodeling and repair, osteogenic cells release angiogenic factors, whereas endothelial cells secrete angiocrine factors. One of the better instances of functional blood vessels specialization for certain organs is the skeletal system. A subpopulation of capillary endothelial cells in the bone regulate the activity of osteoprogenitor cells, which in turn affects bone formation during development and adult homeostasis. Angiogenesis and osteogenesis are strictly connected, and their crosstalk is essential to guarantee bone formation and to maintain bone homeostasis. Additionally, pathological processes including inflammation, cancer, and aging include both bone endothelial cells and angiocrine factors. Therefore, the study and understanding of these mechanisms is fundamental, because molecules and factors involved may represent key targets for novel and advanced therapies.
Collapse
Affiliation(s)
- Domenico Ribatti
- Department of Translational Biomedicine and Neurosciences, University of Bari Medical School, Bari, Italy
| | | |
Collapse
|
47
|
Shen F, Huang X, He G, Shi Y. The emerging studies on mesenchymal progenitors in the long bone. Cell Biosci 2023; 13:105. [PMID: 37301964 DOI: 10.1186/s13578-023-01039-x] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2022] [Accepted: 05/01/2023] [Indexed: 06/12/2023] Open
Abstract
Mesenchymal progenitors (MPs) are considered to play vital roles in bone development, growth, bone turnover, and repair. In recent years, benefiting from advanced approaches such as single-cell sequence, lineage tracing, flow cytometry, and transplantation, multiple MPs are identified and characterized in several locations of bone, including perichondrium, growth plate, periosteum, endosteum, trabecular bone, and stromal compartment. However, although great discoveries about skeletal stem cells (SSCs) and progenitors are present, it is still largely obscure how the varied landscape of MPs from different residing sites diversely contribute to the further differentiation of osteoblasts, osteocytes, chondrocytes, and other stromal cells in their respective destiny sites during development and regeneration. Here we discuss recent findings on MPs' origin, differentiation, and maintenance during long bone development and homeostasis, providing clues and models of how the MPs contribute to bone development and repair.
Collapse
Affiliation(s)
- Fangyuan Shen
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Xiaobin Huang
- Department of Oral and Maxillofacial Surgery/Pharmacology, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Guangxu He
- Department of Orthopedics, The Second Xiangya Hospital, Central South University, NO. 139 Middle Renmin Road, Changsha, Hunan, China.
| | - Yu Shi
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China.
| |
Collapse
|
48
|
Mai W, Liu Q, Li J, Zheng M, Yan F, Liu H, Lei Y, Xu J, Xu J. Comprehensive analysis of the oncogenic and immunological role of FAP and identification of the ceRNA network in human cancers. Aging (Albany NY) 2023; 15:3738-3758. [PMID: 37166418 PMCID: PMC10449273 DOI: 10.18632/aging.204707] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Accepted: 04/22/2023] [Indexed: 05/12/2023]
Abstract
Fibroblast activation protein-alpha (FAP) is a transmembrane serine protease involving in tissue remodeling. Previous studies report that FAP is highly expressed in certain tumors and participated in oncogenesis. However, there is still lack of systematic and in-depth analysis of FAP based on clinical big data. Here, we comprehensively map the FAP expression profile, prognostic outcome, genetic alteration, immune infiltration across over 30 types of human cancers through multiple datasets including TCGA, CPTAC, and cBioPortal. We find that FAP is up-regulated in most cancer types, and increased FAP expression is associated with advanced pathological stages or poor prognosis in several cancers. Furthermore, FAP is significantly correlated with the infiltration of cancer-associated fibroblasts, macrophages, myeloid dendritic cells, as well as endothelia cells. Immunosuppressive checkpoint proteins or cytokines expression, microsatellite instability and tumor mutational burden analysis also indicate the regulation role of FAP in tumor progression. Gene enrichment analysis demonstrates that ECM-receptor interaction as well as extracellular matrix and structure process are linked to the potential mechanism of FAP in tumor pathogenesis. The ceRNA network is also constructed and identified the involvement of LINC00707/hsa-miR-30e-5p/FAP, LINC02535/hsa-miR-30e-5p/FAP, LINC02535/hsa-miR-30d-5p/FAP, as well as AC026356.1/hsa-miR-30d-5p/FAP axis in tumor progression. In conclusion, our study offers new insights into the oncogenic and immunological role of FAP from a pan-cancer perspective, providing new clues for developing novel targeted anti-tumor strategies.
Collapse
Affiliation(s)
- Weiqian Mai
- School of Medicine, School of Life Science and Engineering, Foshan University, Foshan 528000, China
| | - Qingyou Liu
- School of Medicine, School of Life Science and Engineering, Foshan University, Foshan 528000, China
| | - Jiasheng Li
- School of Medicine, School of Life Science and Engineering, Foshan University, Foshan 528000, China
| | - Mincheng Zheng
- Integrative Medicine Research Center, School of Basic Medical Sciences, Guangzhou University of Chinese Medicine, University Town, Guangzhou 510006, China
| | - Fuman Yan
- Department of Physiology, School of Basic Medical Sciences, Guangzhou University of Chinese Medicine, University Town, Guangzhou 510006, China
| | - Hui Liu
- School of Medicine, School of Life Science and Engineering, Foshan University, Foshan 528000, China
| | - Yuhe Lei
- Department of Pharmacy, Shenzhen Hospital of Guangzhou University of Chinese Medicine, Shenzhen 518034, China
| | - Jinwen Xu
- Integrative Medicine Research Center, School of Basic Medical Sciences, Guangzhou University of Chinese Medicine, University Town, Guangzhou 510006, China
- Department of Physiology, School of Basic Medical Sciences, Guangzhou University of Chinese Medicine, University Town, Guangzhou 510006, China
| | - Jiean Xu
- Integrative Medicine Research Center, School of Basic Medical Sciences, Guangzhou University of Chinese Medicine, University Town, Guangzhou 510006, China
- Department of Physiology, School of Basic Medical Sciences, Guangzhou University of Chinese Medicine, University Town, Guangzhou 510006, China
| |
Collapse
|
49
|
Rashid DJ, Sheheen JR, Huey T, Surya K, Sanders JB, Horner JR, Voyich J, Chapman SC. Nonpathological inflammation drives the development of an avian flight adaptation. Proc Natl Acad Sci U S A 2023; 120:e2219757120. [PMID: 37126698 PMCID: PMC10175837 DOI: 10.1073/pnas.2219757120] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2022] [Accepted: 03/18/2023] [Indexed: 05/03/2023] Open
Abstract
The development of modern birds provides a window into the biology of their dinosaur ancestors. We investigated avian postnatal development and found that sterile inflammation drives formation of the pygostyle, a compound structure resulting from bone fusion in the tail. Inflammation is generally induced by compromised tissue integrity, but here is involved in normal bone development. Transcriptome profiling and immuno/histochemistry reveal a robust inflammatory response that resembles bone fracture healing. The data suggest the involvement of necroptosis and multiple immune cell types, notably heterophils (the avian equivalent of neutrophils). Additionally, nucleus pulposus structures, heretofore unknown in birds, are involved in disc remodeling. Anti-inflammatory corticosteroid treatment inhibited vertebral fusion, substantiating the crucial role of inflammation in the ankylosis process. This study shows that inflammation can drive developmental skeletogenesis, in this case leading to the formation of a flight-adapted tail structure on the evolutionary path to modern avians.
Collapse
Affiliation(s)
- Dana J. Rashid
- Department of Microbiology and Cell Biology, Montana State University, Bozeman, MT59717
| | - Joseph R. Sheheen
- Department of Biological Sciences, Clemson University, Clemson, SC29634
| | - Tori Huey
- Department of Biological Sciences, Clemson University, Clemson, SC29634
| | - Kevin Surya
- Molecular Biosciences Program, Montana State University, Bozeman, MT59717
| | | | - John R. Horner
- Schmid College of Science and Technology, Chapman University, Orange, CA92866
| | - Jovanka Voyich
- Department of Microbiology and Cell Biology, Montana State University, Bozeman, MT59717
| | - Susan C. Chapman
- Department of Biological Sciences, Clemson University, Clemson, SC29634
| |
Collapse
|
50
|
Waker CA, Hwang AE, Bowman-Gibson S, Chandiramani CH, Linkous B, Stone ML, Keoni CI, Kaufman MR, Brown TL. Mouse models of preeclampsia with preexisting comorbidities. Front Physiol 2023; 14:1137058. [PMID: 37089425 PMCID: PMC10117893 DOI: 10.3389/fphys.2023.1137058] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Accepted: 03/17/2023] [Indexed: 04/08/2023] Open
Abstract
Preeclampsia is a pregnancy-specific condition and a leading cause of maternal and fetal morbidity and mortality. It is thought to occur due to abnormal placental development or dysfunction, because the only known cure is delivery of the placenta. Several clinical risk factors are associated with an increased incidence of preeclampsia including chronic hypertension, diabetes, autoimmune conditions, kidney disease, and obesity. How these comorbidities intersect with preeclamptic etiology, however, is not well understood. This may be due to the limited number of animal models as well as the paucity of studies investigating the impact of these comorbidities. This review examines the current mouse models of chronic hypertension, pregestational diabetes, and obesity that subsequently develop preeclampsia-like symptoms and discusses how closely these models recapitulate the human condition. Finally, we propose an avenue to expand the development of mouse models of preeclampsia superimposed on chronic comorbidities to provide a strong foundation needed for preclinical testing.
Collapse
Affiliation(s)
- Christopher A. Waker
- Department of Neuroscience, Cell Biology and Physiology, Boonshoft School of Medicine, Wright State University, Dayton, OH, United States
| | - Amy E. Hwang
- Department of Neuroscience, Cell Biology and Physiology, Boonshoft School of Medicine, Wright State University, Dayton, OH, United States
| | - Scout Bowman-Gibson
- Department of Neuroscience, Cell Biology and Physiology, Boonshoft School of Medicine, Wright State University, Dayton, OH, United States
| | - Chandni H. Chandiramani
- Department of Neuroscience, Cell Biology and Physiology, Boonshoft School of Medicine, Wright State University, Dayton, OH, United States
- Department of Obstetrics and Gynecology, Boonshoft School of Medicine, Wright State University, Dayton, OH, United States
| | - Bryce Linkous
- Department of Neuroscience, Cell Biology and Physiology, Boonshoft School of Medicine, Wright State University, Dayton, OH, United States
| | - Madison L. Stone
- Department of Neuroscience, Cell Biology and Physiology, Boonshoft School of Medicine, Wright State University, Dayton, OH, United States
| | - Chanel I. Keoni
- Department of Neuroscience, Cell Biology and Physiology, Boonshoft School of Medicine, Wright State University, Dayton, OH, United States
| | - Melissa R. Kaufman
- Department of Neuroscience, Cell Biology and Physiology, Boonshoft School of Medicine, Wright State University, Dayton, OH, United States
| | - Thomas L. Brown
- Department of Neuroscience, Cell Biology and Physiology, Boonshoft School of Medicine, Wright State University, Dayton, OH, United States
- Department of Obstetrics and Gynecology, Boonshoft School of Medicine, Wright State University, Dayton, OH, United States
- *Correspondence: Thomas L. Brown,
| |
Collapse
|