1
|
Ouimet F, Patel D, Tsontakis M, Samson C, Forbes SL. Establishing the volatile organic compound profile and detection capabilities of human remain detection dogs to human bones. Forensic Sci Int Synerg 2025; 10:100566. [PMID: 39760049 PMCID: PMC11699303 DOI: 10.1016/j.fsisyn.2024.100566] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2024] [Revised: 11/28/2024] [Accepted: 12/04/2024] [Indexed: 01/07/2025]
Abstract
The detection of skeletal remains using human remain detection dogs (HRD) is often reported anecdotally by handlers to be a challenge. Limited studies have been conducted to determine the volatile organic compounds (VOCs) emitted from bones, particularly when there is limited organic matter remaining. This study aimed to determine the VOCs emitted from dry, weathered bones and examine the detection performance of HRD dogs on these bones when used as training aids. The VOCs of four different bones (clavicle, rib, humerus, and vertebrae) from three cadavers were collected using sorbent tubes and analyzed using comprehensive two-dimensional gas chromatography‒time-of-flight mass spectrometry (GC × GC‒TOFMS). Subsequently, the responses of the HRD dogs to the bone samples were recorded over two separate two-day trials. A total of 296 VOCs were detected and classified into chemical classes, with aromatics and linear aliphatics being the most abundant classes. Several differences in the chemical class distribution were observed between the bone types, but the number and intensity of the VOCs were similar between the bone samples. During the HRD dog training, a higher false detection rate was observed on the first day of each trial; however, the detection rate improved to 100 % on the second day of each trial. Although the dogs are capable of detecting bones, they require exposure to and training with a diverse range of skeletal remains to enhance their efficiency. This is necessary due to the variations in the types and intensity of VOCs compared to earlier decomposition stages involving soft tissue.
Collapse
Affiliation(s)
- Frédérique Ouimet
- Département de Chimie, Biochimie et Physique, Université du Québec à Trois-Rivières, 3351 boul. des Forges, Trois-Rivières, Québec, G8Z 4M3, Canada
- Department of Chemistry and Biochemistry, University of Windsor, 401 Sunset Avenue, Windsor, Ontario, N9B 3P4, Canada
| | - Darshil Patel
- Département de Chimie, Biochimie et Physique, Université du Québec à Trois-Rivières, 3351 boul. des Forges, Trois-Rivières, Québec, G8Z 4M3, Canada
- Department of Chemistry and Biochemistry, University of Windsor, 401 Sunset Avenue, Windsor, Ontario, N9B 3P4, Canada
| | - Marissa Tsontakis
- Département de Chimie, Biochimie et Physique, Université du Québec à Trois-Rivières, 3351 boul. des Forges, Trois-Rivières, Québec, G8Z 4M3, Canada
| | - Clifford Samson
- Canine Unit, Ontario Provincial Police, 777 Memorial Ave, Orillia, Ontario, L3V 7V3, Canada
| | - Shari L. Forbes
- Département de Chimie, Biochimie et Physique, Université du Québec à Trois-Rivières, 3351 boul. des Forges, Trois-Rivières, Québec, G8Z 4M3, Canada
- Department of Chemistry and Biochemistry, University of Windsor, 401 Sunset Avenue, Windsor, Ontario, N9B 3P4, Canada
| |
Collapse
|
2
|
Chen Y, Guo B, Zhang Y, Bao X, Li D, Lin J. Injectable hypoxia-preconditioned human exfoliated deciduous teeth stem cells encapsulated within GelMA-AMP microspheres for bone regeneration in periodontitis. Colloids Surf B Biointerfaces 2025; 247:114452. [PMID: 39689590 DOI: 10.1016/j.colsurfb.2024.114452] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2024] [Revised: 11/21/2024] [Accepted: 12/09/2024] [Indexed: 12/19/2024]
Abstract
Periodontitis, an inflammatory and infectious disease resulting from dental plaque, affects tooth-supporting tissues and interconnects with various systemic conditions. Advancing periodontal tissue regeneration stands as pivotal in periodontitis treatment. Presently, odontogenic stem cells garner substantial interest for dental pulp functional tissue regeneration. Essential to stem cell delivery success is the design of suitable drug delivery vehicles. Hence, this research introduces novel injectable antimicrobial peptide-grafted methacryloyl gelatin (GelMA-AMP) microspheres, housing hypoxia-inducible factor (HIF-1α), ensuring sustained release to stimulate osteogenic differentiation of deciduous dental pulp stem cells (SHEDs). Through in vitro co-culturing and preparation of HIF-1α@GelMA-AMP@SHED cell spheres, they were injected and transplanted into periodontal pockets for periodontitis treatment. Results display microsphere sizes averaging 93.92 ± 6.00 μm, akin to human dental pulp, showcasing commendable cytocompatibility and in vitro antibacterial properties. Additionally, GelMA-AMP microspheres carrying HIF-1α enhance cell adhesion, proliferation, and extracellular matrix protein secretion. Notably, HIF-1α@GelMA-AMP@SHED microspheres were effective in reducing periodontal inflammation in vivo and promoting vascularization and tissue regeneration in the periodontal region. Consequently, the application potential of HIF-1α@GelMA-AMP@SHED hydrogel microspheres in periodontitis treatment appears promising.
Collapse
Affiliation(s)
- Yu Chen
- Department of Stomatology, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China; Department of Stomatology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou 510000, China
| | - Bing Guo
- Department of Stomatology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou 510000, China
| | - Yanqing Zhang
- Department of Stomatology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou 510000, China
| | - Xinyu Bao
- Department of Stomatology, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Dan Li
- Department of Stomatology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou 510000, China
| | - Jun Lin
- Department of Stomatology, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China.
| |
Collapse
|
3
|
Wang J, Zhou D, Li R, Sheng S, Li G, Sun Y, Wang P, Mo Y, Liu H, Chen X, Geng Z, Zhang Q, Jing Y, Bai L, Xu K, Su J. Protocol for engineering bone organoids from mesenchymal stem cells. Bioact Mater 2025; 45:388-400. [PMID: 39687559 PMCID: PMC11647664 DOI: 10.1016/j.bioactmat.2024.11.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Revised: 11/12/2024] [Accepted: 11/12/2024] [Indexed: 12/18/2024] Open
Abstract
Bone organoids are emerging as powerful tools for studying bone development and related diseases. However, the simplified design of current methods somewhat limits their application potential, as these methods produce single-tissue organoids that fail to replicate the bone microarchitecture or achieve effective mineralization. To address this issue, we propose a three-dimensional (3D) construction strategy for generating mineralized bone structures using bone marrow-derived mesenchymal stem cells (BMSCs). By mixing BMSCs with hydrogel to create a bone matrix-mimicking bioink and employing projection-based light-curing 3D printing technology, we constructed 3D-printed structures, which were then implanted subcutaneously into nude mice, away from the native bone microenvironment. Even without external stimulation, these implants spontaneously formed mineralized bone domains. With long-term culture, these structures gradually matured into fully differentiated bone tissue, completing both mineralization and vascularization. This in vivo bone organoid model offers a novel platform for studying bone development, exploring congenital diseases, testing drugs, and developing therapeutic applications.
Collapse
Affiliation(s)
- Jian Wang
- Department of Orthopedics, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
- Trauma Orthopedics Center, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
- Institute of Musculoskeletal Injury and Translational Medicine of Organoids, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
- Institute of Translational Medicine, Shanghai University, Shanghai, 200444, China
- School of Medicine, Shanghai University, Shanghai, 200444, China
- National Center for Translational Medicine SHU Branch, Shanghai University, Shanghai, 200444, China
| | - Dongyang Zhou
- Institute of Translational Medicine, Shanghai University, Shanghai, 200444, China
- National Center for Translational Medicine SHU Branch, Shanghai University, Shanghai, 200444, China
| | - Ruiyang Li
- Department of Orthopedics, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
- Trauma Orthopedics Center, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
- Institute of Musculoskeletal Injury and Translational Medicine of Organoids, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
| | - Shihao Sheng
- Department of Orthopedics, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
- Trauma Orthopedics Center, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
- Institute of Musculoskeletal Injury and Translational Medicine of Organoids, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
| | - Guangfeng Li
- Institute of Translational Medicine, Shanghai University, Shanghai, 200444, China
- School of Medicine, Shanghai University, Shanghai, 200444, China
- National Center for Translational Medicine SHU Branch, Shanghai University, Shanghai, 200444, China
- Department of Orthopedics, Shanghai Zhongye Hospital, Shanghai, 200941, China
| | - Yue Sun
- Institute of Translational Medicine, Shanghai University, Shanghai, 200444, China
- School of Medicine, Shanghai University, Shanghai, 200444, China
- National Center for Translational Medicine SHU Branch, Shanghai University, Shanghai, 200444, China
| | - Peng Wang
- Department of Orthopedics, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
- Trauma Orthopedics Center, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
- Institute of Musculoskeletal Injury and Translational Medicine of Organoids, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
| | - Yulin Mo
- Institute of Translational Medicine, Shanghai University, Shanghai, 200444, China
- School of Medicine, Shanghai University, Shanghai, 200444, China
- National Center for Translational Medicine SHU Branch, Shanghai University, Shanghai, 200444, China
| | - Han Liu
- Institute of Translational Medicine, Shanghai University, Shanghai, 200444, China
- National Center for Translational Medicine SHU Branch, Shanghai University, Shanghai, 200444, China
| | - Xiao Chen
- Department of Orthopedics, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
- Trauma Orthopedics Center, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
- Institute of Musculoskeletal Injury and Translational Medicine of Organoids, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
- Institute of Translational Medicine, Shanghai University, Shanghai, 200444, China
- National Center for Translational Medicine SHU Branch, Shanghai University, Shanghai, 200444, China
| | - Zhen Geng
- Institute of Translational Medicine, Shanghai University, Shanghai, 200444, China
- National Center for Translational Medicine SHU Branch, Shanghai University, Shanghai, 200444, China
| | - Qin Zhang
- Institute of Translational Medicine, Shanghai University, Shanghai, 200444, China
- National Center for Translational Medicine SHU Branch, Shanghai University, Shanghai, 200444, China
| | - Yingying Jing
- Institute of Translational Medicine, Shanghai University, Shanghai, 200444, China
- National Center for Translational Medicine SHU Branch, Shanghai University, Shanghai, 200444, China
| | - Long Bai
- Institute of Translational Medicine, Shanghai University, Shanghai, 200444, China
- National Center for Translational Medicine SHU Branch, Shanghai University, Shanghai, 200444, China
| | - Ke Xu
- Institute of Translational Medicine, Shanghai University, Shanghai, 200444, China
- National Center for Translational Medicine SHU Branch, Shanghai University, Shanghai, 200444, China
| | - Jiacan Su
- Department of Orthopedics, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
- Trauma Orthopedics Center, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
- Institute of Musculoskeletal Injury and Translational Medicine of Organoids, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
- Institute of Translational Medicine, Shanghai University, Shanghai, 200444, China
- National Center for Translational Medicine SHU Branch, Shanghai University, Shanghai, 200444, China
| |
Collapse
|
4
|
Perepletchikova D, Malashicheva A. Communication between endothelial cells and osteoblasts in regulation of bone homeostasis: Notch players. Stem Cell Res Ther 2025; 16:56. [PMID: 39920854 PMCID: PMC11806792 DOI: 10.1186/s13287-025-04176-x] [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: 07/25/2024] [Accepted: 01/23/2025] [Indexed: 02/09/2025] Open
Abstract
Endothelial cells coat blood vessels and release molecular signals to affect the fate of other cells. Endothelial cells can adjust their behavior in response to the changing microenvironmental conditions. During bone regeneration, bone tissue cells release factors that promote blood vessel growth. Notch is a key signaling that regulates cell fate decisions in many tissues and plays an important role in bone tissue development and homeostasis. Understanding the interplay between angiogenesis and osteogenesis is currently a focus of research efforts in order to facilitate and improve osteogenesis when needed. Our review explores the cellular and molecular mechanisms including Notch-dependent endothelial-MSC communication that drive osteogenesis-angiogenesis processes and their effects on bone remodeling and repair.
Collapse
Affiliation(s)
| | - Anna Malashicheva
- Institute of Cytology Russian Academy of Science, St. Petersburg, Russia, 194064.
| |
Collapse
|
5
|
Andreasen CM, Wölfel EM, Ejersted C, Andersen TL, Frost M. Type 2 diabetes patients exhibit delayed but coupled bone remodelling, maintaining cortical porosity comparable to healthy controls: A histomorphometric analysis of trans-iliac bone biopsies. Bone 2025; 193:117412. [PMID: 39884487 DOI: 10.1016/j.bone.2025.117412] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/14/2024] [Revised: 01/06/2025] [Accepted: 01/26/2025] [Indexed: 02/01/2025]
Abstract
OBJECTIVE Fracture risk is increased in longstanding type 2 diabetes (T2D). High-resolution peripheral quantitative CT scans have demonstrated higher cortical porosity in T2D complicated by microvascular disease (MVD). We investigated if cortical bone resorption is followed by inadequate bone formation in individuals with T2D complicated by MVD. METHODS Thirty-five adult men and women with T2D were recruited from outpatient clinics and through public advertisement. All participants had at least one previous measure of c-peptide >700, a negative GAD antibody test, and 13 had known microvascular disease status. Trans iliac crest bone biopsies were collected for histomorphometric analysis. Glucose control was assessed using HbA1c. Additionally, trans iliac bone specimens from 10 individuals without T2D were included as controls. RESULTS Following quality assessment, samples from 30 T2D and 10 controls were used for histomorphometric analyses of cortical bone remodelling. The final study population included 23 men and 7 postmenopausal women with a mean age of 65.8 years for the T2D-MVD group (CI95% 61.2-70.3) and 65.2 years in the T2D + MVD group (CI95% 59.6-70.9), and a mean T2D disease duration of 16.9 years. Seventeen had MVD (57 %). The controls included 5 men and 5 women with a mean age of 64.7 years (CI95% 58.5-70.9). The area, diameter, and density of cortical pores were the same in cases with and without MVD, but the pore diameter was lower than controls. While T2D had significantly more eroded-formative pores compared to controls, there were no significant differences in the proportion of eroded and formative pores between the groups. In quiescent pores/osteons, the osteon diameter and wall thickness were larger in T2D groups than controls. CONCLUSION Cortical bone porosity was not increased in individuals with T2D complicated by MVD. However, an enhanced prevalence of eroded-formative pores and increased osteon diameter concur with a slightly prolonged reversal-resorption phase in T2D irrespective of the presence of MVD.
Collapse
Affiliation(s)
- C M Andreasen
- Molecular Bone Histology Laboratory (MBH lab), Research Unit of Pathology, Dept. of Clinical Research, University of Southern Denmark, DK-5000 Odense C, Denmark; Department of Pathology, Odense University Hospital, DK-5000 Odense C, Denmark.
| | - E M Wölfel
- Molecular Endocrinology Laboratory (KMEB), Department of Endocrinology, Odense University Hospital, DK-5000 Odense C, Denmark
| | - C Ejersted
- Molecular Endocrinology Laboratory (KMEB), Department of Endocrinology, Odense University Hospital, DK-5000 Odense C, Denmark
| | - T L Andersen
- Molecular Bone Histology Laboratory (MBH lab), Research Unit of Pathology, Dept. of Clinical Research, University of Southern Denmark, DK-5000 Odense C, Denmark; Department of Pathology, Odense University Hospital, DK-5000 Odense C, Denmark
| | - M Frost
- Molecular Endocrinology Laboratory (KMEB), Department of Endocrinology, Odense University Hospital, DK-5000 Odense C, Denmark; Steno Diabetes Centre Odense, Odense University Hospital, DK-5000 Odense C, Denmark
| |
Collapse
|
6
|
Lee EJ, Lialios P, Curtis M, Williams J, Kim Y, Salipante P, Hudson S, Esch MB, Levi M, Kitlinska J, Alimperti S. Glucocorticoids Alter Bone Microvascular Barrier via MAPK/Connexin43 Mechanisms. Adv Healthc Mater 2025:e2404302. [PMID: 39831839 DOI: 10.1002/adhm.202404302] [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: 11/02/2024] [Revised: 12/30/2024] [Indexed: 01/22/2025]
Abstract
Glucocorticoids (GCs) are standard-of-care treatments for inflammatory and immune disorders, and their long-term use increases the risk of osteoporosis. Although GCs decrease bone functionality, their role in bone microvasculature is incompletely understood. Herein, the study investigates the mechanisms of bone microvascular barrier function via osteoblast-endothelial interactions in response to GCs. The animal data shows that prednisolone (Psl) downregulated the osteoblast function and microvessel number and size. To investigate the role of GCs in bone endothelial barrier function further, a bicellular microfluidic in vitro system is developed and utilized, which consists of three-dimensional (3D) perfusable microvascular structures embedded in collagen I/osteoblast matrix. Interestingly, it is demonstrated that GCs significantly inhibit osteogenesis and microvascular barrier function by interfering with endothelial-osteoblast interactions. This effect is triggered by MAPK-induced phosphorylation of connexin43 (Cx43) at Ser282. Collectively, this study sheds light on microvascular function in bone disorders, as osteoporosis, and permits to capture dynamic changes in endothelial-bone interactions under GCs by dissecting the MAPK/Cx43 mechanism and proposing this as a potential target for bone diseases.
Collapse
Affiliation(s)
- Eun-Jin Lee
- Department of Biochemistry and Molecular and Cellular Biology, School of Medicine, Georgetown University, Washington, DC, 20057, USA
- Biological and Biomedical Engineering Center, School of Medicine, Georgetown University, Washington, DC, 20057, USA
- Microsystems and Nanotechnology Division, Physical Measurement Laboratory, National Institute of Standards and Technology, Gaithersburg, MD, 20899, USA
- Department of Chemistry and Biochemistry, College of Computer, Mathematical and Natural Sciences, University of Maryland, College Park, MD, 20742, USA
| | - Peter Lialios
- Department of Biochemistry and Molecular and Cellular Biology, School of Medicine, Georgetown University, Washington, DC, 20057, USA
- Biological and Biomedical Engineering Center, School of Medicine, Georgetown University, Washington, DC, 20057, USA
| | - Micaila Curtis
- Department of Biochemistry and Molecular and Cellular Biology, School of Medicine, Georgetown University, Washington, DC, 20057, USA
- Biological and Biomedical Engineering Center, School of Medicine, Georgetown University, Washington, DC, 20057, USA
| | - James Williams
- Department of Biochemistry and Molecular and Cellular Biology, School of Medicine, Georgetown University, Washington, DC, 20057, USA
| | - Yoontae Kim
- Department of Biochemistry and Molecular and Cellular Biology, School of Medicine, Georgetown University, Washington, DC, 20057, USA
- Biological and Biomedical Engineering Center, School of Medicine, Georgetown University, Washington, DC, 20057, USA
| | - Paul Salipante
- Materials Science and Engineering Division, Material Measurement Laboratory, National Institute of Standards and Technology, Gaithersburg, MD, 20899, USA
| | - Steven Hudson
- Materials Science and Engineering Division, Material Measurement Laboratory, National Institute of Standards and Technology, Gaithersburg, MD, 20899, USA
| | - Mandy B Esch
- Microsystems and Nanotechnology Division, Physical Measurement Laboratory, National Institute of Standards and Technology, Gaithersburg, MD, 20899, USA
| | - Moshe Levi
- Department of Biochemistry and Molecular and Cellular Biology, School of Medicine, Georgetown University, Washington, DC, 20057, USA
- Biological and Biomedical Engineering Center, School of Medicine, Georgetown University, Washington, DC, 20057, USA
| | - Joanna Kitlinska
- Department of Biochemistry and Molecular and Cellular Biology, School of Medicine, Georgetown University, Washington, DC, 20057, USA
- Biological and Biomedical Engineering Center, School of Medicine, Georgetown University, Washington, DC, 20057, USA
| | - Stella Alimperti
- Department of Biochemistry and Molecular and Cellular Biology, School of Medicine, Georgetown University, Washington, DC, 20057, USA
| |
Collapse
|
7
|
Park H, Trupiano SP, Medarev SL, Ghosh P, Caldwell JT, Yarrow JF, Muller-Delp JM. Aerobic exercise training-induced bone and vascular adaptations in mice lacking adiponectin. Bone 2025; 190:117272. [PMID: 39369833 PMCID: PMC11795456 DOI: 10.1016/j.bone.2024.117272] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/29/2024] [Revised: 09/23/2024] [Accepted: 10/03/2024] [Indexed: 10/08/2024]
Abstract
Adiponectin regulates lipid and glucose metabolism, and insulin sensitivity in various target organs; however, the effects of adiponectin on bone health remain controversial. Exercise training can enhance bone density, bone microarchitecture, and blood flow. This study aimed to elucidate the role of adiponectin in adaptations of bone microarchitecture and bone vasculature in response to aerobic exercise training. Adult male C57BL/6 wild-type (WT) and homozygous adiponectin knockout (AdipoKO) mice were either treadmill exercise trained or remained sedentary for 8-10 weeks. The trabecular structures of the distal femoral metaphysis, a weight-bearing bone, and the mandible, a non-weight-bearing bone, were examined using microcomputed tomography. The femoral principal nutrient arteries were isolated to assess vasoreactivity (vasodilation and vasoconstriction) and structural remodeling. At the femoral metaphysis, impaired trabecular bone structures, including reduced connectivity density and increased trabecular spacing, were observed in AdipoKO mice compared to WT mice. In addition, nitric oxide-mediated, endothelium-dependent vasodilation was substantially reduced, and wall-to-lumen ratio was significantly increased in the femoral principal nutrient artery of AdipoKO mice. Interestingly, although exercise training-induced enhancements in trabecular connectivity density were observed at the femoral metaphysis of both WT and AdipoKO, increased vasoconstrictor responses were only observed in the femoral principal nutrient artery of WT mice, not in the AdipoKO mice. In mandibular trabecular bone, exercise training increased trabecular bone volume fraction (BV/TV, %) and intersection surface in the mandible of both WT and AdipoKO mice. These findings indicate that adiponectin is crucial for maintaining normal bone microarchitecture and vasculature. Although the absence of adiponectin compromises bone vascular adaptation to exercise training in mice, some exercise training-induced alterations in bone microarchitecture occurred in the absence of adiponectin, suggesting contribution of compensatory mechanisms during exercise training.
Collapse
Affiliation(s)
- Hyerim Park
- Department of Health, Nutrition, and Food Sciences, Florida State University, Tallahassee, FL, USA
| | - Samuel P Trupiano
- Department of Biomedical Sciences, Florida State University, Tallahassee, FL, USA
| | - Steven L Medarev
- Department of Biomedical Sciences, Florida State University, Tallahassee, FL, USA
| | - Payal Ghosh
- Department of Health, Nutrition, and Food Sciences, Florida State University, Tallahassee, FL, USA
| | - Jacob T Caldwell
- Department of Biomedical Sciences, Florida State University, Tallahassee, FL, USA; Department of Exercise and Sport Science, University of Wisconsin-La Crosse, La Crosse, WI, USA
| | - Joshua F Yarrow
- Eastern Colorado Geriatrics Research, Education, and Clinical Center, Rocky Mountain Regional Veterans Affairs Medical Center, VA Eastern Colorado Health Care System, Aurora, CO, USA
| | - Judy M Muller-Delp
- Department of Biomedical Sciences, Florida State University, Tallahassee, FL, USA.
| |
Collapse
|
8
|
Zhong C, Tang Z, Yu X, Wang L, Ren C, Qin L, Zhou P. Advances in the Construction and Application of Bone-on-a-Chip Based on Microfluidic Technologies. J Biomed Mater Res B Appl Biomater 2024; 112:e35502. [PMID: 39555794 DOI: 10.1002/jbm.b.35502] [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: 06/19/2024] [Revised: 10/19/2024] [Accepted: 10/28/2024] [Indexed: 11/19/2024]
Abstract
Bone-on-a-chip (BOC) models that based on microfluidic technology have widely applied to understand bone physiology and the pathogenesis of related diseases. In this review, we provide an overview of bone biology and related diseases, explain the advantages and applications of microfluidic technology in the construction of BOC models, and summarize their progress in physiology, pathology, and drug development. Finally, we discussed the problems to be solved and the future directions of microfluidic technology and BOC platforms, so as to provide a reference for researchers to design better BOC models.
Collapse
Affiliation(s)
- Chang Zhong
- Key Laboratory of Dental Maxillofacial Reconstruction and Biological Intelligence Manufacturing, School and Hospital of Stomatology, Lanzhou University, Lanzhou, China
| | - Zihui Tang
- Key Laboratory of Dental Maxillofacial Reconstruction and Biological Intelligence Manufacturing, School and Hospital of Stomatology, Lanzhou University, Lanzhou, China
| | - Xin Yu
- Key Laboratory of Dental Maxillofacial Reconstruction and Biological Intelligence Manufacturing, School and Hospital of Stomatology, Lanzhou University, Lanzhou, China
| | - Lu Wang
- Key Laboratory of Dental Maxillofacial Reconstruction and Biological Intelligence Manufacturing, School and Hospital of Stomatology, Lanzhou University, Lanzhou, China
| | - Chenyuan Ren
- Key Laboratory of Dental Maxillofacial Reconstruction and Biological Intelligence Manufacturing, School and Hospital of Stomatology, Lanzhou University, Lanzhou, China
| | - Liying Qin
- School of Stomatology, Gansu Health Vocational College, Lanzhou, China
| | - Ping Zhou
- Key Laboratory of Dental Maxillofacial Reconstruction and Biological Intelligence Manufacturing, School and Hospital of Stomatology, Lanzhou University, Lanzhou, China
| |
Collapse
|
9
|
Labelle-Dumais C, Mazur C, Kaya S, Obata Y, Lee B, Acevedo C, Alliston T, Gould DB. Skeletal pathology in mouse models of Gould syndrome is partially alleviated by genetically reducing TGFβ signaling. Matrix Biol 2024; 133:1-13. [PMID: 39097038 DOI: 10.1016/j.matbio.2024.07.005] [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: 05/25/2024] [Revised: 07/30/2024] [Accepted: 07/31/2024] [Indexed: 08/05/2024]
Abstract
Skeletal defects are hallmark features of many extracellular matrix (ECM) and collagen-related disorders. However, a biological function in bone has never been defined for the highly evolutionarily conserved type IV collagen. Collagen type IV alpha 1 (COL4A1) and alpha 2 (COL4A2) form α1α1α2 (IV) heterotrimers that represent a fundamental basement membrane constituent present in every organ of the body, including the skeleton. COL4A1 and COL4A2 mutations cause Gould syndrome, a variable and clinically heterogenous multisystem disorder generally characterized by the presence of cerebrovascular disease with ocular, renal, and muscular manifestations. We have previously identified elevated TGFβ signaling as a pathological insult resulting from Col4a1 mutations and demonstrated that reducing TGFβ signaling ameliorate ocular and cerebrovascular phenotypes in Col4a1 mutant mouse models of Gould syndrome. In this study, we describe the first characterization of skeletal defects in Col4a1 mutant mice that include a developmental delay in osteogenesis and structural, biomechanical and vascular alterations of mature bones. Using distinct mouse models, we show that allelic heterogeneity influences the presentation of skeletal pathology resulting from Col4a1 mutations. Importantly, we found that TGFβ target gene expression is elevated in developing bones from Col4a1 mutant mice and show that genetically reducing TGFβ signaling partially ameliorates skeletal manifestations. Collectively, these findings identify a novel and unsuspected role for type IV collagen in bone biology, expand the spectrum of manifestations associated with Gould syndrome to include skeletal abnormalities, and implicate elevated TGFβ signaling in skeletal pathogenesis in Col4a1 mutant mice.
Collapse
Affiliation(s)
- Cassandre Labelle-Dumais
- Departments of Ophthalmology, University of California San Francisco, San Francisco, CA 94143, USA
| | - Courtney Mazur
- Department of Orthopaedic Surgery, University of California San Francisco, San Francisco, CA, 94143, USA; UC Berkeley/UCSF Graduate Program in Bioengineering, San Francisco, CA 94143, USA
| | - Serra Kaya
- Department of Orthopaedic Surgery, University of California San Francisco, San Francisco, CA, 94143, USA
| | - Yoshihiro Obata
- Department of Mechanical and Aerospace Engineering, University of California San Diego, San Diego, CA 92093, USA
| | - Bryson Lee
- Departments of Ophthalmology, University of California San Francisco, San Francisco, CA 94143, USA
| | - Claire Acevedo
- Department of Orthopaedic Surgery, University of California San Francisco, San Francisco, CA, 94143, USA; Materials Science Division, Lawrence Berkeley National Laboratory, Berkeley, CA 94720, USA; Department of Mechanical and Aerospace Engineering, University of California San Diego, San Diego, CA 92093, USA
| | - Tamara Alliston
- Department of Orthopaedic Surgery, University of California San Francisco, San Francisco, CA, 94143, USA; UC Berkeley/UCSF Graduate Program in Bioengineering, San Francisco, CA 94143, USA
| | - Douglas B Gould
- Departments of Ophthalmology, University of California San Francisco, San Francisco, CA 94143, USA; Department of Anatomy, Institute for Human Genetics, Bakar Aging Research Institute, and Cardiovascular Research Institute, University of California San Francisco, San Francisco, CA 94143, USA.
| |
Collapse
|
10
|
Dei Rossi G, Vergani LM, Buccino F. A Novel Triad of Bio-Inspired Design, Digital Fabrication, and Bio-Derived Materials for Personalised Bone Repair. MATERIALS (BASEL, SWITZERLAND) 2024; 17:5305. [PMID: 39517582 PMCID: PMC11547793 DOI: 10.3390/ma17215305] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/08/2024] [Revised: 10/22/2024] [Accepted: 10/28/2024] [Indexed: 11/16/2024]
Abstract
The emerging paradigm of personalised bone repair embodies a transformative triad comprising bio-inspired design, digital fabrication, and the exploration of innovative materials. The increasing average age of the population, alongside the rising incidence of fractures associated with age-related conditions such as osteoporosis, necessitates the development of customised, efficient, and minimally invasive treatment modalities as alternatives to conventional methods (e.g., autografts, allografts, Ilizarov distraction, and bone fixators) typically employed to promote bone regeneration. A promising innovative technique involves the use of cellularised scaffolds incorporating mesenchymal stem cells (MSCs). The selection of materials-ranging from metals and ceramics to synthetic or natural bio-derived polymers-combined with a design inspired by natural sources (including bone, corals, algae, shells, silk, and plants) facilitates the replication of geometries, architectures, porosities, biodegradation capabilities, and mechanical properties conducive to physiological bone regeneration. To mimic internal structures and geometries for construct customisation, scaffolds can be designed using Computer-aided Design (CAD) and fabricated via 3D-printing techniques. This approach not only enables precise control over external shapes and internal architectures but also accommodates the use of diverse materials that improve biological performance and provide economic advantages. Finally, advanced numerical models are employed to simulate, analyse, and optimise the complex processes involved in personalised bone regeneration, with computational predictions validated against experimental data and in vivo studies to ascertain the model's ability to predict the recovery of bone shape and function.
Collapse
Affiliation(s)
- Greta Dei Rossi
- Department of Mechanical Engineering (DMEC), Politecnico di Milano, Via La Masa 1, 20156 Milano, Italy; (G.D.R.); (F.B.)
| | - Laura Maria Vergani
- Department of Mechanical Engineering (DMEC), Politecnico di Milano, Via La Masa 1, 20156 Milano, Italy; (G.D.R.); (F.B.)
- IRCCS Orthopedic Institute Galeazzi, Via Cristina Belgioioso 173, 20157 Milan, Italy
| | - Federica Buccino
- Department of Mechanical Engineering (DMEC), Politecnico di Milano, Via La Masa 1, 20156 Milano, Italy; (G.D.R.); (F.B.)
- IRCCS Orthopedic Institute Galeazzi, Via Cristina Belgioioso 173, 20157 Milan, Italy
| |
Collapse
|
11
|
Castañón-Cortés LG, Bravo-Vázquez LA, Santoyo-Valencia G, Medina-Feria S, Sahare P, Duttaroy AK, Paul S. Current advances in the development of microRNA-integrated tissue engineering strategies: a cornerstone of regenerative medicine. Front Bioeng Biotechnol 2024; 12:1484151. [PMID: 39479296 PMCID: PMC11521876 DOI: 10.3389/fbioe.2024.1484151] [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: 08/21/2024] [Accepted: 10/07/2024] [Indexed: 11/02/2024] Open
Abstract
Regenerative medicine is an innovative scientific field focused on repairing, replacing, or regenerating damaged tissues and organs to restore their normal functions. A central aspect of this research arena relies on the use of tissue-engineered scaffolds, which serve as structural supports that mimic the extracellular matrix, providing an environment that orchestrates cell growth and tissue formation. Remarkably, the therapeutic efficacy of these scaffolds can be improved by harnessing the properties of other molecules or compounds that have crucial roles in healing and regeneration pathways, such as phytochemicals, enzymes, transcription factors, and non-coding RNAs (ncRNAs). In particular, microRNAs (miRNAs) are a class of tiny (20-24 nt), highly conserved ncRNAs that play a critical role in the regulation of gene expression at the post-transcriptional level. Accordingly, miRNAs are involved in a myriad of biological processes, including cell differentiation, proliferation, and apoptosis, as well as tissue regeneration, angiogenesis, and osteogenesis. On this basis, over the past years, a number of research studies have demonstrated that miRNAs can be integrated into tissue-engineered scaffolds to create advanced therapeutic platforms that precisely modulate cellular behavior and offer a controlled and targeted release of miRNAs to optimize tissue repair and regeneration. Therefore, in this current review, we discuss the most recent advances in the development of miRNA-loaded tissue-engineered scaffolds and provide an overview of the future outlooks that should be aborded in this area of study in order to lay the groundwork for the clinical translation of these tissue engineering approaches.
Collapse
Affiliation(s)
| | | | | | - Sara Medina-Feria
- School of Engineering and Sciences, Tecnologico de Monterrey, Queretaro, Mexico
| | - Padmavati Sahare
- School of Engineering and Sciences, Institute of Advanced Materials for Sustainable Manufacturing, Tecnologico de Monterrey, Queretaro, Mexico
| | - Asim K. Duttaroy
- Department of Nutrition, Institute of Basic Medical Sciences, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Sujay Paul
- School of Engineering and Sciences, Tecnologico de Monterrey, Queretaro, Mexico
| |
Collapse
|
12
|
Dousti M, Parsa S, Sani F, Bagherzadeh E, Zamanzadeh Z, Dara M, Sani M, Azarpira N. Enhancing bone regeneration: Unleashing the potential of magnetic nanoparticles in a microtissue model. J Cell Mol Med 2024; 28:e70040. [PMID: 39219020 PMCID: PMC11366680 DOI: 10.1111/jcmm.70040] [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: 06/01/2024] [Revised: 08/13/2024] [Accepted: 08/16/2024] [Indexed: 09/04/2024] Open
Abstract
Bone tissue engineering addresses the limitations of autologous resources and the risk of allograft disease transmission in bone diseases. In this regard, engineered three-dimensional (3D) models emerge as biomimetic alternatives to natural tissues, replicating intracellular communication. Moreover, the unique properties of super-paramagnetic iron oxide nanoparticles (SPIONs) were shown to promote bone regeneration via enhanced osteogenesis and angiogenesis in bone models. This study aimed to investigate the effects of SPION on both osteogenesis and angiogenesis and characterized a co-culture of Human umbilical vein endothelial cells (HUVEC) and MG-63 cells as a model of bone microtissue. HUVECs: MG-63s with a ratio of 4:1 demonstrated the best results among other cell ratios, and 50 μg/mL of SPION was the optimum concentration for maximum survival, cell migration and mineralization. In addition, the data from gene expression illustrated that the expression of osteogenesis-related genes, including osteopontin, osteocalcin, alkaline phosphatase, and collagen-I, as well as the expression of the angiogenesis-related marker, CD-31, and the tube formation, is significantly elevated when the 50 μg/mL concentration of SPION is applied to the microtissue samples. SPION application in a designed 3D bone microtissue model involving a co-culture of osteoblast and endothelial cells resulted in increased expression of specific markers related to angiogenesis and osteogenesis. This includes the design of a novel biomimetic model to boost blood compatibility and biocompatibility of primary materials while promoting osteogenic activity in microtissue bone models. Moreover, this can improve interaction with surrounding tissues and broaden the knowledge to promote superior-performance implants, preventing device failure.
Collapse
Affiliation(s)
- Maryam Dousti
- Shiraz Institute for Stem Cell and Regenerative MedicineShiraz University of Medical ScienceShirazIran
- Department of Genetics, Faculty of Biological Sciences and TechnologyShahid Ashrafi Esfahani UniversityIsfahanIran
| | - Shima Parsa
- Shiraz Institute for Stem Cell and Regenerative MedicineShiraz University of Medical ScienceShirazIran
| | - Farnaz Sani
- Shiraz Institute for Stem Cell and Regenerative MedicineShiraz University of Medical ScienceShirazIran
| | | | - Zahra Zamanzadeh
- Department of Genetics, Faculty of Biological Sciences and TechnologyShahid Ashrafi Esfahani UniversityIsfahanIran
| | - Mahintaj Dara
- Stem Cells Technology Research CenterShiraz University of Medical SciencesShirazIran
| | - Mahsa Sani
- Shiraz Institute for Stem Cell and Regenerative MedicineShiraz University of Medical ScienceShirazIran
- Tissue Engineering Department, School of Advanced Medical Science and TechnologyShiraz University of Medical ScienceShirazIran
| | - Negar Azarpira
- Shiraz Institute for Stem Cell and Regenerative MedicineShiraz University of Medical ScienceShirazIran
- Transplant Research CenterShiraz University of Medical ScienceShirazIran
| |
Collapse
|
13
|
Hu S, Liang Y, Pan X. Exosomes: A promising new strategy for treating osteoporosis in the future. J Drug Deliv Sci Technol 2024; 97:105571. [DOI: 10.1016/j.jddst.2024.105571] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
|
14
|
Yang L, Li D, Yan Y, Yang Q, Li L, Zha Y. Microvascular permeability and texture analysis of bone marrow in diabetic rabbits with critical limb ischemia based on dynamic contrast-enhanced magnetic resonance imaging. J Diabetes Investig 2024; 15:584-593. [PMID: 38240456 PMCID: PMC11060156 DOI: 10.1111/jdi.14145] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/11/2023] [Revised: 12/16/2023] [Accepted: 12/21/2023] [Indexed: 05/01/2024] Open
Abstract
BACKGROUND Early on in the development of diabetes, skeletal muscles can exhibit microarchitectural changes that can be detected using texture analysis (TA) based on volume transfer constant (Ktrans) maps. Nevertheless, there have been few studies and thus we evaluated microvascular permeability and the TA of the bone marrow in diabetics with critical limb ischemia (CLI). METHODS Eighteen male rabbits were randomly assigned equally into an operation group with hindlimb ischemia and diabetes, a sham-operated group with diabetes only, and a control group. Dynamic contrast enhanced magnetic resonance imaging (DCE-MRI) was performed on all rabbits at predetermined intervals (1, 5, 10, 15, 20, and 25 days post-surgery). The pharmacokinetic model was used to generate the permeability parameters, while the textural parameters were derived from the Ktrans map. Data analysis methods included the independent sample t-test, Mann-Whitney U test, repeated-measures analysis of variance, and Pearson correlation tests. RESULTS The Ktrans values reached a minimum on day 1 after ischemia induction, then gradually recovered, but remained lower than those of the sham-operated group. The volume fraction only showed a significant difference between the operation group and the sham-operated group on day 5 post-surgery, but not in the extravascular extracellular space volume fraction at all time points. A significantly reduced Ktrans on day 1, a decreased number of bone trabeculae (Tb.N), and the area of bone trabeculae (Tb.Ar), and an increased microvessel density on day 25 in the operation group compared with the sham-operated group were observed. At each time point, there was a discernible difference between the two groups in the mean value, mean of positive pixels, and sumAverage. CONCLUSIONS The early stages of diabetic bone marrow with CLI can be evaluated by DCE-MRI for microvascular permeability. Texture analysis based on DCE-MRI could act as an imaging discriminator and new radiological analysis tool for critical limb ischemia in diabetes mellitus.
Collapse
Affiliation(s)
- Liu Yang
- Department of RadiologyRenmin Hospital of Wuhan UniversityNo. 238 Jiefang RoadWuhan430060Wuchang DistrictChina
- Department of RadiologyUnion Hospital, Tongji Medical College, Huazhong University of Science and Technology1277 Jiefang AvenueWuhan430022China
| | - Donghang Li
- Department of Thoracic SurgeryRenmin Hospital of Wuhan UniversityNo.238 Jiefang RoadWuhan430060Wuchang DistrictChina
| | - Yuchen Yan
- Department of RadiologyRenmin Hospital of Wuhan UniversityNo. 238 Jiefang RoadWuhan430060Wuchang DistrictChina
| | - Qi Yang
- Department of RadiologyRenmin Hospital of Wuhan UniversityNo. 238 Jiefang RoadWuhan430060Wuchang DistrictChina
| | - Liang Li
- Department of RadiologyRenmin Hospital of Wuhan UniversityNo. 238 Jiefang RoadWuhan430060Wuchang DistrictChina
| | - Yunfei Zha
- Department of RadiologyRenmin Hospital of Wuhan UniversityNo. 238 Jiefang RoadWuhan430060Wuchang DistrictChina
| |
Collapse
|
15
|
Poulos MG, Ramalingam P, Winiarski A, Gutkin MC, Katsnelson L, Carter C, Pibouin-Fragner L, Eichmann A, Thomas JL, Miquerol L, Butler JM. Complementary and Inducible creER T2 Mouse Models for Functional Evaluation of Endothelial Cell Subtypes in the Bone Marrow. Stem Cell Rev Rep 2024; 20:1135-1149. [PMID: 38438768 PMCID: PMC11087254 DOI: 10.1007/s12015-024-10703-9] [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] [Accepted: 02/21/2024] [Indexed: 03/06/2024]
Abstract
In the adult bone marrow (BM), endothelial cells (ECs) are an integral component of the hematopoietic stem cell (HSC)-supportive niche, which modulates HSC activity by producing secreted and membrane-bound paracrine signals. Within the BM, distinct vascular arteriole, transitional, and sinusoidal EC subtypes display unique paracrine expression profiles and create anatomically-discrete microenvironments. However, the relative contributions of vascular endothelial subtypes in supporting hematopoiesis is unclear. Moreover, constitutive expression and off-target activity of currently available endothelial-specific and endothelial-subtype-specific murine cre lines potentially confound data analysis and interpretation. To address this, we describe two tamoxifen-inducible cre-expressing lines, Vegfr3-creERT2 and Cx40-creERT2, that efficiently label sinusoidal/transitional and arteriole endothelium respectively in adult marrow, without off-target activity in hematopoietic or perivascular cells. Utilizing an established mouse model in which cre-dependent recombination constitutively-activates MAPK signaling within adult endothelium, we identify arteriole ECs as the driver of MAPK-mediated hematopoietic dysfunction. These results define complementary tamoxifen-inducible creERT2-expressing mouse lines that label functionally-discrete and non-overlapping sinusoidal/transitional and arteriole EC populations in the adult BM, providing a robust toolset to investigate the differential contributions of vascular subtypes in maintaining hematopoietic homeostasis.
Collapse
Affiliation(s)
- Michael G Poulos
- Department of Medicine, University of Florida Health Cancer Center, Gainesville, FL, 32610, USA
- Division of Hematology/Oncology, University of Florida, 1333 Center Drive, BH-022D, Gainesville, FL, 32610, USA
| | - Pradeep Ramalingam
- Department of Medicine, University of Florida Health Cancer Center, Gainesville, FL, 32610, USA
- Division of Hematology/Oncology, University of Florida, 1333 Center Drive, BH-022D, Gainesville, FL, 32610, USA
| | - Agatha Winiarski
- Department of Medicine, University of Florida Health Cancer Center, Gainesville, FL, 32610, USA
| | - Michael C Gutkin
- Ansary Stem Cell Institute, Division of Regenerative Medicine, Department of Medicine, Weill Cornell Medicine, New York, NY, 10065, USA
| | - Lizabeth Katsnelson
- Ansary Stem Cell Institute, Division of Regenerative Medicine, Department of Medicine, Weill Cornell Medicine, New York, NY, 10065, USA
| | - Cody Carter
- Department of Medicine, University of Florida Health Cancer Center, Gainesville, FL, 32610, USA
| | | | - Anne Eichmann
- Université de Paris Cité, Inserm, PARCC, 75015, Paris, France
- Department of Molecular and Cellular Physiology, Yale University School of Medicine, New Haven, CT, 06510, USA
- Cardiovascular Research Center, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT, 06511, USA
| | - Jean-Leon Thomas
- Department of Neurology, Yale University School of Medicine, New Haven, CT, 06511, USA
- Paris Brain Institute, Université Pierre et Marie Curie Paris, 06 UMRS1127, Sorbonne Université, Paris Brain Institute, Paris, France
| | - Lucile Miquerol
- Aix-Marseille Université, CNRS UMR 7288, IBDM, 13288, Marseille, France
| | - Jason M Butler
- Department of Medicine, University of Florida Health Cancer Center, Gainesville, FL, 32610, USA.
- Ansary Stem Cell Institute, Division of Regenerative Medicine, Department of Medicine, Weill Cornell Medicine, New York, NY, 10065, USA.
- Division of Hematology/Oncology, University of Florida, 1333 Center Drive, BH-022D, Gainesville, FL, 32610, USA.
| |
Collapse
|
16
|
Haider MT, Freytag V, Krause L, Spethmann T, Gosau T, Beine MC, Knies C, Schröder-Schwarz J, Horn M, Riecken K, Lange T. Comparison of ex vivo bioluminescence imaging, Alu-qPCR and histology for the quantification of spontaneous lung and bone metastases in subcutaneous xenograft mouse models. Clin Exp Metastasis 2024; 41:103-115. [PMID: 38353934 PMCID: PMC10972982 DOI: 10.1007/s10585-024-10268-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2023] [Accepted: 01/16/2024] [Indexed: 03/28/2024]
Abstract
Bioluminescence imaging (BLI) is a non-invasive state-of-the-art-method for longitudinal tracking of tumor cells in mice. The technique is commonly used to determine bone metastatic burden in vivo and also suitable ex vivo to detect even smallest bone micro-metastases in spontaneous metastasis xenograft models. However, it is unclear to which extent ex vivo BLI correlates with alternative methods for metastasis quantification. Here, we compared ex vivo BLI, human DNA-based Alu-qPCR, and histology for the quantification of bone vs. lung metastases, which are amongst the most common sites of metastasis in prostate cancer (PCa) patients and spontaneous PCa xenograft models. Data from 93 immunodeficient mice were considered, each of which were subcutaneously injected with luciferase/RGB-labeled human PCa PC-3 cells. The primary tumors were resected at ~ 0.75 cm³ and mice were sacrificed ~ 3 weeks after surgery and immediately examined by ex vivo BLI. Afterwards, the right lungs and hind limbs with the higher BLI signal (BLIHi bone) were processed for histology, whereas the left lung lobes and hind limbs with the lower BLI signal (BLILo bone) were prepared for Alu-qPCR. Our data demonstrate remarkable differences in the correlation coefficients of the different methods for lung metastasis detection (r ~ 0.8) vs. bone metastasis detection (r ~ 0.4). However, the BLI values of the BLIHi and BLILo bones correlated very strongly (r ~ 0.9), indicating that the method per se was reliable under identical limitations; the overall level of metastasis to contralateral bones was astonishingly similar. Instead, the level of lung metastasis only weakly to moderately correlated with the level of bone metastasis formation. Summarized, we observed a considerable discrepancy between ex vivo BLI and histology/Alu-qPCR in the quantification of bone metastases, which was not observed in the case of lung metastases. Future studies using ex vivo BLI for bone metastasis quantification should combine multiple methods to accurately determine metastatic load in bone samples.
Collapse
Affiliation(s)
- Marie-Therese Haider
- Institute of Anatomy and Experimental Morphology, University Medical Center Hamburg- Eppendorf, Martinistrasse 52, Hamburg, 20246, Germany
| | - Vera Freytag
- Institute of Anatomy and Experimental Morphology, University Medical Center Hamburg- Eppendorf, Martinistrasse 52, Hamburg, 20246, Germany
| | - Linda Krause
- Institute of Medical Biometry and Epidemiology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Tanja Spethmann
- Institute of Anatomy and Experimental Morphology, University Medical Center Hamburg- Eppendorf, Martinistrasse 52, Hamburg, 20246, Germany
| | - Tobias Gosau
- Institute of Anatomy and Experimental Morphology, University Medical Center Hamburg- Eppendorf, Martinistrasse 52, Hamburg, 20246, Germany
| | - Mia C Beine
- Institute of Anatomy and Experimental Morphology, University Medical Center Hamburg- Eppendorf, Martinistrasse 52, Hamburg, 20246, Germany
| | - Christine Knies
- Institute of Anatomy and Experimental Morphology, University Medical Center Hamburg- Eppendorf, Martinistrasse 52, Hamburg, 20246, Germany
| | - Jennifer Schröder-Schwarz
- Institute of Anatomy and Experimental Morphology, University Medical Center Hamburg- Eppendorf, Martinistrasse 52, Hamburg, 20246, Germany
| | - Michael Horn
- Core Facility In Vivo Optical Imaging, University Cancer Center Hamburg, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Mildred Scheel Cancer Career Center, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Kristoffer Riecken
- Research Department Cell and Gene Therapy, Department of Stem Cell Transplantation, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Tobias Lange
- Institute of Anatomy and Experimental Morphology, University Medical Center Hamburg- Eppendorf, Martinistrasse 52, Hamburg, 20246, Germany.
- Institute of Anatomy I, University Hospital Jena, Teichgraben 7, Jena, 07743, Germany.
- Comprehensive Cancer Center Central Germany (CCCG), Ulm, Germany.
| |
Collapse
|
17
|
Schevenels G, Cabochette P, America M, Vandenborne A, De Grande L, Guenther S, He L, Dieu M, Christou B, Vermeersch M, Germano RFV, Perez-Morga D, Renard P, Martin M, Vanlandewijck M, Betsholtz C, Vanhollebeke B. A brain-specific angiogenic mechanism enabled by tip cell specialization. Nature 2024; 628:863-871. [PMID: 38570687 PMCID: PMC11041701 DOI: 10.1038/s41586-024-07283-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2023] [Accepted: 03/07/2024] [Indexed: 04/05/2024]
Abstract
Vertebrate organs require locally adapted blood vessels1,2. The gain of such organotypic vessel specializations is often deemed to be molecularly unrelated to the process of organ vascularization. Here, opposing this model, we reveal a molecular mechanism for brain-specific angiogenesis that operates under the control of Wnt7a/b ligands-well-known blood-brain barrier maturation signals3-5. The control mechanism relies on Wnt7a/b-dependent expression of Mmp25, which we find is enriched in brain endothelial cells. CRISPR-Cas9 mutagenesis in zebrafish reveals that this poorly characterized glycosylphosphatidylinositol-anchored matrix metalloproteinase is selectively required in endothelial tip cells to enable their initial migration across the pial basement membrane lining the brain surface. Mechanistically, Mmp25 confers brain invasive competence by cleaving meningeal fibroblast-derived collagen IV α5/6 chains within a short non-collagenous region of the central helical part of the heterotrimer. After genetic interference with the pial basement membrane composition, the Wnt-β-catenin-dependent organotypic control of brain angiogenesis is lost, resulting in properly patterned, yet blood-brain-barrier-defective cerebrovasculatures. We reveal an organ-specific angiogenesis mechanism, shed light on tip cell mechanistic angiodiversity and thereby illustrate how organs, by imposing local constraints on angiogenic tip cells, can select vessels matching their distinctive physiological requirements.
Collapse
Affiliation(s)
- Giel Schevenels
- Laboratory of Neurovascular Signaling, Department of Molecular Biology, ULB Neuroscience Institute, Université libre de Bruxelles (ULB), Gosselies, Belgium
| | - Pauline Cabochette
- Laboratory of Neurovascular Signaling, Department of Molecular Biology, ULB Neuroscience Institute, Université libre de Bruxelles (ULB), Gosselies, Belgium
| | - Michelle America
- Laboratory of Neurovascular Signaling, Department of Molecular Biology, ULB Neuroscience Institute, Université libre de Bruxelles (ULB), Gosselies, Belgium
| | - Arnaud Vandenborne
- Laboratory of Neurovascular Signaling, Department of Molecular Biology, ULB Neuroscience Institute, Université libre de Bruxelles (ULB), Gosselies, Belgium
| | - Line De Grande
- Laboratory of Neurovascular Signaling, Department of Molecular Biology, ULB Neuroscience Institute, Université libre de Bruxelles (ULB), Gosselies, Belgium
| | - Stefan Guenther
- Max Planck Institute for Heart and Lung Research, ECCPS Bioinformatics and Deep Sequencing Platform, Bad Nauheim, Germany
| | - Liqun He
- Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, Uppsala University, Uppsala, Sweden
| | - Marc Dieu
- Mass Spectrometry Facility (MaSUN), University of Namur, Namur, Belgium
| | - Basile Christou
- Laboratory of Neurovascular Signaling, Department of Molecular Biology, ULB Neuroscience Institute, Université libre de Bruxelles (ULB), Gosselies, Belgium
| | - Marjorie Vermeersch
- Center for Microscopy and Molecular Imaging (CMMI), Université libre de Bruxelles (ULB), Gosselies, Belgium
| | - Raoul F V Germano
- Laboratory of Neurovascular Signaling, Department of Molecular Biology, ULB Neuroscience Institute, Université libre de Bruxelles (ULB), Gosselies, Belgium
| | - David Perez-Morga
- Laboratory of Neurovascular Signaling, Department of Molecular Biology, ULB Neuroscience Institute, Université libre de Bruxelles (ULB), Gosselies, Belgium
- Center for Microscopy and Molecular Imaging (CMMI), Université libre de Bruxelles (ULB), Gosselies, Belgium
| | - Patricia Renard
- Mass Spectrometry Facility (MaSUN), University of Namur, Namur, Belgium
| | - Maud Martin
- Laboratory of Neurovascular Signaling, Department of Molecular Biology, ULB Neuroscience Institute, Université libre de Bruxelles (ULB), Gosselies, Belgium
| | - Michael Vanlandewijck
- Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, Uppsala University, Uppsala, Sweden
- Department of Medicine (Huddinge), Karolinska Institutet, Huddinge, Sweden
| | - Christer Betsholtz
- Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, Uppsala University, Uppsala, Sweden
- Department of Medicine (Huddinge), Karolinska Institutet, Huddinge, Sweden
| | - Benoit Vanhollebeke
- Laboratory of Neurovascular Signaling, Department of Molecular Biology, ULB Neuroscience Institute, Université libre de Bruxelles (ULB), Gosselies, Belgium.
| |
Collapse
|
18
|
Li C, Chen G, Wang Y, Xu W, Hu M. Indirect co-culture of osteoblasts and endothelial cells in vitro based on a biomimetic 3D composite hydrogel scaffold to promote the proliferation and differentiation of osteoblasts. PLoS One 2024; 19:e0298689. [PMID: 38527040 PMCID: PMC10962808 DOI: 10.1371/journal.pone.0298689] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2023] [Accepted: 01/30/2024] [Indexed: 03/27/2024] Open
Abstract
The field of orthopedics has long struggled with the challenge of repairing and regenerating bone defects, which involves a complex process of osteogenesis requiring coordinated interactions among different types of cells. The crucial role of endothelial cells and osteoblasts in bone vascularization and osteogenesis underscores the importance of their intimate interaction. However, efforts to bioengineer bone tissue have been impeded by the difficulty in establishing proper angiogenesis and osteogenesis in tissue structures. This study presents a novel approach to bone tissue engineering, involving a three-dimensional composite hydrogel scaffold composed of sodium alginate microspheres encapsulated in type I collagen. Using this scaffold, a three-dimensional indirect co-culture system was established for osteoblasts and endothelial cells to evaluate the osteogenic differentiation potential of osteoblasts. Results demonstrate that the non-contact co-culture system of endothelial cells and osteoblasts constructed by the composite hydrogel scaffold loaded with microspheres holds promise for bone tissue engineering. The innovative concept of an indirect co-culture system presents exciting prospects for conducting intercellular communication studies and offers a valuable in vitro tissue platform to investigate tissue regeneration.
Collapse
Affiliation(s)
- Cheng Li
- Department of Orthopedics, Jiangsu Provincial People's Hospital, Nanjing, Jiangsu, China
| | - Guanghui Chen
- Department of Orthopedics, Dongguan Tungwah Hospital, Dongguan, Guangdong, China
| | - Yangyang Wang
- Guangdong Medical University, Zhanjiang, Guangdong, China
| | - Wenwu Xu
- Department of Orthopedics, Dongguan Tungwah Hospital, Dongguan, Guangdong, China
| | - Minghui Hu
- Department of Orthopedics, DongGuan SongShan Lake Tungwah Hospital, Dongguan, Guangdong, China
| |
Collapse
|
19
|
Eazer J, Barsoum M, Smith C, Hotta K, Behnke B, Holmes C, Caldwell J, Ghosh P, Reid-Foley E, Park H, Delp M, Muller-Delp J. Adaptations of bone and bone vasculature to muscular stretch training. JBMR Plus 2024; 8:ziad019. [PMID: 38741608 PMCID: PMC11090128 DOI: 10.1093/jbmrpl/ziad019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Revised: 11/16/2023] [Accepted: 12/07/2023] [Indexed: 05/16/2024] Open
Abstract
The magnitude of bone formation and remodeling is linked to both the magnitude of strain placed on the bone and the perfusion of bone. It was previously reported that an increase in bone perfusion and bone density occurs in the femur of old rats with moderate aerobic exercise training. This study determined the acute and chronic effects of static muscle stretching on bone blood flow and remodeling. Old male Fischer 344 rats were randomized to either a naive or stretch-trained group. Static stretching of ankle flexor muscles was achieved by placement of a dorsiflexion splint on the left ankle for 30 min/d, 5d/wk for 4wk. The opposite hindlimb served as a contralateral control (nonstretched) limb. Bone blood flow was assessed during and after acute stretching in naive rats, and at rest and during exercise in stretch-trained rats. Vascular reactivity of the nutrient artery of the proximal tibia was also assessed in stretch-trained rats. MicroCT analysis was used to assess bone volume and micro-architecture of the trabecular bone of both tibias near that growth plate. In naive rats, static stretching increased blood flow to the proximal tibial metaphasis. Blood flow to the proximal tibial metaphysis during treadmill exercise was higher in the stretched limb after 4 wk of daily stretching. Daily stretching also increased tibial bone weight and increased total volume in both the proximal and distal tibial metaphyses. In the trabecular bone immediately below the proximal tibial growth plate, total volume and bone volume increased, but bone volume/total volume was unchanged and trabecular connectivity decreased. In contrast, intravascular volume increased in this region of the bone. These data suggest that blood flow to the tibia increases during bouts of static stretching of the hindlimb muscles, and that 4 wk of daily muscle stretching leads to bone remodeling and an increase in intravascular volume of the tibial bone.
Collapse
Affiliation(s)
- Julia Eazer
- Department of Biomedical Sciences, Florida State University, Tallahassee, Fl, 32304, United States
| | - Mina Barsoum
- Department of Chemical Engineering, Florida State University, Tallahassee, Fl, 32304, United States
| | - Cole Smith
- Department of Biomedical Sciences, Florida State University, Tallahassee, Fl, 32304, United States
| | - Kazuki Hotta
- Department of Biomedical Sciences, Florida State University, Tallahassee, Fl, 32304, United States
- Department of Physical Therapy, Niigata University of Health and Welfare, Niigata, Japan
| | - Brad Behnke
- Department of Kinesiology & Johnson Cancer Research Center, Kansas State University, Manhattan, KS, 66506, United States
| | - Christina Holmes
- Department of Biomedical Engineering, Florida State University, Tallahassee, FL, 32310 United States
| | - Jacob Caldwell
- Department of Biomedical Sciences, Florida State University, Tallahassee, Fl, 32304, United States
| | - Payal Ghosh
- Department of Nutrition and Integrative Physiology, Florida State University, Tallahassee, Fl, 32304, United States
| | - Emily Reid-Foley
- Department of Biomedical Sciences, Florida State University, Tallahassee, Fl, 32304, United States
| | - Hyerim Park
- Department of Nutrition and Integrative Physiology, Florida State University, Tallahassee, Fl, 32304, United States
| | - Michael Delp
- Department of Nutrition and Integrative Physiology, Florida State University, Tallahassee, Fl, 32304, United States
| | - Judy Muller-Delp
- Department of Biomedical Sciences, Florida State University, Tallahassee, Fl, 32304, United States
| |
Collapse
|
20
|
Granat MM, Eifler-Zydel J, Kolmas J. Statins-Their Role in Bone Tissue Metabolism and Local Applications with Different Carriers. Int J Mol Sci 2024; 25:2378. [PMID: 38397055 PMCID: PMC10888549 DOI: 10.3390/ijms25042378] [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: 01/04/2024] [Revised: 02/13/2024] [Accepted: 02/15/2024] [Indexed: 02/25/2024] Open
Abstract
Statins, widely prescribed for lipid disorders, primarily target 3-hydroxy-3-methylglutaryl-coenzyme A (HMG-CoA) reductase competitively and reversibly, resulting in reduced low-density lipoprotein cholesterol (LDL-C). This mechanism proves effective in lowering the risk of lipid-related diseases such as ischemic cerebrovascular and coronary artery diseases. Beyond their established use, statins are under scrutiny for potential applications in treating bone diseases. The focus of research centers mainly on simvastatin, a lipophilic statin demonstrating efficacy in preventing osteoporosis and aiding in fracture and bone defect healing. Notably, these effects manifest at elevated doses (20 mg/kg/day) of statins, posing challenges for systematic administration due to their limited bone affinity. Current investigations explore intraosseous statin delivery facilitated by specialized carriers. This paper outlines various carrier types, characterizing their structures and underscoring various statins' potential as local treatments for bone diseases.
Collapse
Affiliation(s)
- Marcin Mateusz Granat
- Department of Clinical and Experimental Pharmacology, Faculty of Medicine, Medical University of Warsaw, Banacha 1 Str., 02-097 Warsaw, Poland;
| | - Joanna Eifler-Zydel
- Department of Pharmaceutical Chemistry and Biomaterials, Faculty of Pharmacy, Medical University of Warsaw, Banacha 1 Str., 02-097 Warsaw, Poland;
| | - Joanna Kolmas
- Department of Pharmaceutical Chemistry and Biomaterials, Faculty of Pharmacy, Medical University of Warsaw, Banacha 1 Str., 02-097 Warsaw, Poland;
| |
Collapse
|
21
|
Xu HK, Liu JX, Zhou ZK, Zheng CX, Sui BD, Yuan Y, Kong L, Jin Y, Chen J. Osteoporosis under psychological stress: mechanisms and therapeutics. LIFE MEDICINE 2024; 3:lnae009. [PMID: 39872391 PMCID: PMC11749647 DOI: 10.1093/lifemedi/lnae009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Accepted: 03/06/2024] [Indexed: 01/30/2025]
Abstract
Psychological stress has been associated with the onset of several diseases, including osteoporosis. However, the underlying pathogenic mechanism remains unknown, and effective therapeutic strategies are still unavailable. Growing evidence suggests that the sympathetic nervous system regulates bone homeostasis and vascular function under psychological stress, as well as the coupling of osteogenesis and angiogenesis in bone development, remodeling, and regeneration. Furthermore, extracellular vesicles (EVs), particularly mesenchymal stem cell extracellular vesicles (MSC-EVs), have emerged as prospecting therapies for stimulating angiogenesis and bone regeneration. We summarize the role of sympathetic regulation in bone homeostasis and vascular function in response to psychological stress and emphasize the relationship between vessels and bone. Finally, we suggest using MSC-EVs as a promising therapeutic method for treating osteoporosis in psychological stress.
Collapse
Affiliation(s)
- Hao-Kun Xu
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi International Joint Research Center for Oral Diseases, Center for Tissue Engineering, School of Stomatology, The Fourth Military Medical University, Xi’an 710032, China
- Department of Oral Anatomy and Physiology, School of Stomatology, The Fourth Military Medical University, Xi’an 710032, China
| | - Jie-Xi Liu
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi International Joint Research Center for Oral Diseases, Center for Tissue Engineering, School of Stomatology, The Fourth Military Medical University, Xi’an 710032, China
| | - Ze-Kai Zhou
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi International Joint Research Center for Oral Diseases, Center for Tissue Engineering, School of Stomatology, The Fourth Military Medical University, Xi’an 710032, China
- School of Basic Medicine, The Fourth Military Medical University, Xi’an 710032, China
| | - Chen-Xi Zheng
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi International Joint Research Center for Oral Diseases, Center for Tissue Engineering, School of Stomatology, The Fourth Military Medical University, Xi’an 710032, China
| | - Bing-Dong Sui
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi International Joint Research Center for Oral Diseases, Center for Tissue Engineering, School of Stomatology, The Fourth Military Medical University, Xi’an 710032, China
| | - Yuan Yuan
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi International Joint Research Center for Oral Diseases, Center for Tissue Engineering, School of Stomatology, The Fourth Military Medical University, Xi’an 710032, China
- Exercise Immunology Center, Wuhan Sports University, Wuhan 430079, China
| | - Liang Kong
- Department of Oral and Maxillofacial Surgery, School of Stomatology, The Fourth Military Medical University, Xi’an 710032, China
| | - Yan Jin
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi International Joint Research Center for Oral Diseases, Center for Tissue Engineering, School of Stomatology, The Fourth Military Medical University, Xi’an 710032, China
| | - Ji Chen
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi International Joint Research Center for Oral Diseases, Center for Tissue Engineering, School of Stomatology, The Fourth Military Medical University, Xi’an 710032, China
- Department of Oral Implantology, School of Stomatology, The Fourth Military Medical University, Xi’an 710032, China
| |
Collapse
|
22
|
Shahriar SMS, Polavoram NS, Andrabi SM, Su Y, Lee D, Tran HQ, Schindler SJ, Xie J. Transforming layered 2D mats into multiphasic 3D nanofiber scaffolds with tailored gradient features for tissue regeneration. BMEMAT 2024; 2:e12065. [PMID: 38586163 PMCID: PMC10997325 DOI: 10.1002/bmm2.12065] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/05/2023] [Accepted: 11/27/2023] [Indexed: 04/09/2024]
Abstract
Multiphasic scaffolds with tailored gradient features hold significant promise for tissue regeneration applications. Herein, this work reports the transformation of two-dimensional (2D) layered fiber mats into three dimensional (3D) multiphasic scaffolds using a 'solids-of-revolution' inspired gas-foaming expansion technology. These scaffolds feature precise control over fiber alignment, pore size, and regional structure. Manipulating nanofiber mat layers and Pluronic F127 concentrations allows further customization of pore size and fiber alignment within different scaffold regions. The cellular response to multiphasic scaffolds demonstrates the number of cells migrated and proliferated onto the scaffolds are mainly dependent on the pore size rather than fiber alignment. In vivo subcutaneous implantation of multiphasic scaffolds to rats reveals substantial cell infiltration, neo tissue formation, collagen deposition, and new vessel formation within scaffolds, greatly surpassing the capabilities of traditional nanofiber mats. Histological examination indicates the importance of optimizing pore size and fiber alignment for promotion of cell infiltration and tissue regeneration. Overall, these scaffolds have potential applications in tissue modeling, studying tissue-tissue interactions, interface tissue engineering, and high-throughput screening for optimized tissue regeneration.
Collapse
Affiliation(s)
- S. M. Shatil Shahriar
- Department of Surgery – Transplant and Mary & Dick Holland Regenerative Medicine Program, College of Medicine, University of Nebraska Medical Center, Omaha, NE, 68198, USA
- Eppley Institute for Research in Cancer and Allied Diseases, College of Medicine, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Navatha Shree Polavoram
- Department of Surgery – Transplant and Mary & Dick Holland Regenerative Medicine Program, College of Medicine, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Syed Muntazir Andrabi
- Department of Surgery – Transplant and Mary & Dick Holland Regenerative Medicine Program, College of Medicine, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Yajuan Su
- Department of Surgery – Transplant and Mary & Dick Holland Regenerative Medicine Program, College of Medicine, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Donghee Lee
- Department of Surgery – Transplant and Mary & Dick Holland Regenerative Medicine Program, College of Medicine, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Huy Quang Tran
- Department of Surgery – Transplant and Mary & Dick Holland Regenerative Medicine Program, College of Medicine, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Samantha J. Schindler
- Department of Surgery – Transplant and Mary & Dick Holland Regenerative Medicine Program, College of Medicine, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Jingwei Xie
- Department of Surgery – Transplant and Mary & Dick Holland Regenerative Medicine Program, College of Medicine, University of Nebraska Medical Center, Omaha, NE, 68198, USA
- Department of Mechanical and Materials Engineering, University of Nebraska Lincoln, Lincoln, NE, 68588, USA
| |
Collapse
|
23
|
Boubaker F, Teixeira PAG, Hossu G, Douis N, Gillet P, Blum A, Gillet R. In vivo depiction of cortical bone vascularization with ultra-high resolution-CT and deep learning algorithm reconstruction using osteoid osteoma as a model. Diagn Interv Imaging 2024; 105:26-32. [PMID: 37482455 DOI: 10.1016/j.diii.2023.07.001] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2023] [Revised: 06/24/2023] [Accepted: 07/12/2023] [Indexed: 07/25/2023]
Abstract
PURPOSE The purpose of this study was to evaluate the ability to depict in vivo bone vascularization using ultra-high-resolution (UHR) computed tomography (CT) with deep learning reconstruction (DLR) and hybrid iterative reconstruction algorithm, compared to simulated conventional CT, using osteoid osteoma as a model. MATERIALS AND METHODS Patients with histopathologically proven cortical osteoid osteoma who underwent UHR-CT between October 2019 and October 2022 were retrospectively included. Images were acquired with a 1024 × 1024 matrix and reconstructed with DLR and hybrid iterative reconstruction algorithm. To simulate conventional CT, images with a 512 × 512 matrix were also reconstructed. Two radiologists (R1, R2) independently evaluated the number of blood vessels entering the nidus and crossing the bone cortex, as well as vessel identification and image quality with a 5-point scale. Standard deviation (SD) of attenuation in the adjacent muscle and that of air were used as image noise and recorded. RESULTS Thirteen patients with 13 osteoid osteomas were included. There were 11 men and two women with a mean age of 21.8 ± 9.1 (SD) years. For both readers, UHR-CT with DLR depicted more nidus vessels (11.5 ± 4.3 [SD] (R1) and 11.9 ± 4.6 [SD] (R2)) and cortical vessels (4 ± 3.8 [SD] and 4.3 ± 4.1 [SD], respectively) than UHR-CT with hybrid iterative reconstruction (10.5 ± 4.3 [SD] and 10.4 ± 4.6 [SD], and 4.1 ± 3.8 [SD] and 4.3 ± 3.8 [SD], respectively) and simulated conventional CT (5.3 ± 2.2 [SD] and 6.4 ± 2.5 [SD], 2 ± 1.2 [SD] and 2.4 ± 1.6 [SD], respectively) (P < 0.05). UHR-CT with DLR provided less image noise than simulated conventional CT and UHR-CT with hybrid iterative reconstruction (P < 0.05). UHR-CT with DLR received the greatest score and simulated conventional CT the lowest score for vessel identification and image quality. CONCLUSION UHR-CT with DLR shows less noise than UHR-CT with hybrid iterative reconstruction and significantly improves cortical bone vascularization depiction compared to simulated conventional CT.
Collapse
Affiliation(s)
- Fatma Boubaker
- Guilloz Imaging Department, Central Hospital, University Hospital Center of Nancy, 54000, Nancy, France
| | - Pedro Augusto Gondim Teixeira
- Guilloz Imaging Department, Central Hospital, University Hospital Center of Nancy, 54000, Nancy, France; Université de Lorraine, INSERM, IADI, 54000, Nancy, France; Université de Lorraine, CIC, Innovation Technologique, University Hospital Center of Nancy, 54000, Nancy, France
| | - Gabriela Hossu
- Université de Lorraine, INSERM, IADI, 54000, Nancy, France; Université de Lorraine, CIC, Innovation Technologique, University Hospital Center of Nancy, 54000, Nancy, France
| | - Nicolas Douis
- Guilloz Imaging Department, Central Hospital, University Hospital Center of Nancy, 54000, Nancy, France
| | - Pierre Gillet
- Université de Lorraine, CNRS, IMoPA, 54000, Nancy, France
| | - Alain Blum
- Guilloz Imaging Department, Central Hospital, University Hospital Center of Nancy, 54000, Nancy, France; Université de Lorraine, INSERM, IADI, 54000, Nancy, France; Université de Lorraine, CIC, Innovation Technologique, University Hospital Center of Nancy, 54000, Nancy, France
| | - Romain Gillet
- Guilloz Imaging Department, Central Hospital, University Hospital Center of Nancy, 54000, Nancy, France; Université de Lorraine, INSERM, IADI, 54000, Nancy, France; Université de Lorraine, CIC, Innovation Technologique, University Hospital Center of Nancy, 54000, Nancy, France.
| |
Collapse
|
24
|
Canalis E, Mocarska M, Schilling L, Jafar-Nejad P, Carrer M. Antisense oligonucleotides targeting a NOTCH3 mutation in male mice ameliorate the cortical osteopenia of lateral meningocele syndrome. Bone 2023; 177:116898. [PMID: 37704069 PMCID: PMC10591917 DOI: 10.1016/j.bone.2023.116898] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Revised: 09/06/2023] [Accepted: 09/08/2023] [Indexed: 09/15/2023]
Abstract
Lateral Meningocele Syndrome (LMS) is a monogenic disorder associated with NOTCH3 pathogenic variants that result in the stabilization of NOTCH3 and a gain-of-function. A mouse model (Notch3em1Ecan) harboring a 6691-TAATGA mutation in the Notch3 locus that results in a functional outcome analogous to LMS exhibits cancellous and cortical bone osteopenia. We tested Notch3 antisense oligonucleotides (ASOs) specific to the Notch36691-TAATGA mutation for their effects on Notch3 downregulation and on the osteopenia of Notch3em1Ecan mice. Twenty-four mouse Notch3 mutant ASOs were designed and tested for toxic effects in vivo, and 12 safe ASOs were tested for their impact on the downregulation of Notch36691-TAATGA and Notch3 mRNA in osteoblast cultures from Notch3em1Ecan mice. Three ASOs downregulated Notch3 mutant transcripts specifically and were tested in vivo for their effects on the bone microarchitecture of Notch3em1Ecan mice. All three ASOs were well tolerated. One of these ASOs had more consistent effects in vivo and was studied in detail. The Notch3 mutant ASO downregulated Notch3 mutant transcripts in osteoblasts and bone marrow stromal cells and had no effect on other Notch receptors. The subcutaneous administration of Notch3 mutant ASO at 50 mg/Kg decreased Notch36691-TAATGA mRNA in bone without apparent toxicity; microcomputed tomography demonstrated that the ASO ameliorated the cortical osteopenia of Notch3em1Ecan mice but not the cancellous bone osteopenia. In conclusion, a Notch3 ASO that downregulates Notch3 mutant expression specifically ameliorates the cortical osteopenia in Notch3em1Ecan mice. ASOs may become useful strategies in the management of monogenic disorders affecting the skeleton.
Collapse
Affiliation(s)
- Ernesto Canalis
- Department of Orthopaedic Surgery, UConn Health, Farmington, CT, USA; Department of Medicine, UConn Health, Farmington, CT, USA; UConn Musculoskeletal Institute, UConn Health, Farmington, CT, USA.
| | - Magda Mocarska
- UConn Musculoskeletal Institute, UConn Health, Farmington, CT, USA
| | - Lauren Schilling
- UConn Musculoskeletal Institute, UConn Health, Farmington, CT, USA
| | | | | |
Collapse
|
25
|
Fu F, Luo H, Du Y, Chen Y, Tian K, Pan J, Li J, Wang N, Bao R, Jin H, Tong P, Ruan H, Wu C. AR/PCC herb pair inhibits osteoblast pyroptosis to alleviate diabetes-related osteoporosis by activating Nrf2/Keap1 pathway. J Cell Mol Med 2023; 27:3601-3613. [PMID: 37621124 PMCID: PMC10660633 DOI: 10.1111/jcmm.17928] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Revised: 07/28/2023] [Accepted: 08/04/2023] [Indexed: 08/26/2023] Open
Abstract
Osteoporosis is a prevalent complication of diabetes, characterized by systemic metabolic impairment of bone mass and microarchitecture, particularly in the spine. Anemarrhenae Rhizoma/Phellodendri Chinensis Cortex (AR/PCC) herb pair has been extensively employed in Traditional Chinese Medicine to manage diabetes; however, its potential to ameliorate diabetic osteoporosis (DOP) has remained obscure. Herein, we explored the protective efficacy of AR/PCC herb pair against DOP using a streptozotocin (STZ)-induced rat diabetic model. Our data showed that AR/PCC could effectively reduce the elevated fasting blood glucose and reverse the osteoporotic phenotype of diabetic rats, resulting in significant improvements in vertebral trabecular area percentage, trabecular thickness and trabecular number, while reducing trabecular separation. Specifically, AR/PCC herb pair improved impaired osteogenesis, nerve ingrowth and angiogenesis. More importantly, it could mitigate the aberrant activation of osteoblast pyroptosis in the vertebral bodies of diabetic rats by reducing increased expressions of Nlrp3, Asc, Caspase1, Gsdmd and IL-1β. Mechanistically, AR/PCC activated antioxidant pathway through the upregulation of the antioxidant response protein Nrf2, while concurrently decreasing its negative feedback regulator Keap1. Collectively, our in vivo findings demonstrate that AR/PCC can inhibit osteoblast pyroptosis and alleviate STZ-induced rat DOP, suggesting its potential as a therapeutic agent for mitigating DOP.
Collapse
Affiliation(s)
- Fangda Fu
- Institute of Orthopaedics and TraumatologyThe First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Traditional Chinese Medicine)HangzhouChina
| | - Huan Luo
- Department of Pharmacy, The Second Affiliated Hospital, School of MedicineZhejiang UniversityHangzhouChina
| | - Yu Du
- The First Affiliated Hospital of Zhejiang Chinese Medical UniversityHangzhouChina
| | - Yuying Chen
- The Fourth Clinical Medical College of Zhejiang Chinese Medical UniversityHangzhouChina
| | - Kun Tian
- Department of OrthopaedicsThe First Affiliated Hospital of Zhejiang Chinese Medical UniversityHangzhouChina
| | - Jin Pan
- Department of Architecture, School of ArchitectureChina Academy of ArtHangzhouChina
| | - Jian Li
- Department of OrthopaedicsHangzhou Ninth People's HospitalHangzhouChina
| | - Nani Wang
- Department of MedicineZhejiang Academy of Traditional Chinese MedicineHangzhouChina
| | - Ronghua Bao
- Hangzhou Fuyang Hospital of TCM Orthopedics and TraumatologyHangzhouChina
| | - Hongting Jin
- Institute of Orthopaedics and TraumatologyThe First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Traditional Chinese Medicine)HangzhouChina
| | - Peijian Tong
- Institute of Orthopaedics and TraumatologyThe First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Traditional Chinese Medicine)HangzhouChina
| | - Hongfeng Ruan
- Institute of Orthopaedics and TraumatologyThe First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Traditional Chinese Medicine)HangzhouChina
| | - Chengliang Wu
- Institute of Orthopaedics and TraumatologyThe First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Traditional Chinese Medicine)HangzhouChina
| |
Collapse
|
26
|
Yu Z, Jiang X, Yin J, Han L, Xiong C, Huo Z, Xu J, Shang J, Xi K, Nong L, Huang Y, Zhou X. CK1ε drives osteogenic differentiation of bone marrow mesenchymal stem cells via activating Wnt/β-catenin pathway. Aging (Albany NY) 2023; 15:10193-10212. [PMID: 37787983 PMCID: PMC10599756 DOI: 10.18632/aging.205067] [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: 05/06/2023] [Accepted: 09/08/2023] [Indexed: 10/04/2023]
Abstract
The treatment of bone defects is a difficult problem in orthopedics. At present, the treatment mainly relies on autologous or allogeneic bone transplantation, which may lead to some complications such as foreign body rejection, local infection, pain, or numbness at the bone donor site. Local injection of conservative therapy to treat bone defects is one of the research hotspots at present. Bone marrow mesenchymal stem cells (BMSCs) can self-renew, significantly proliferate, and differentiate into various types of cells. Although it has been reported that CK1ε could mediate the Wnt/β-catenin pathway, leading to the development of the diseases, whether CK1ε plays a role in bone regeneration through the Wnt/β-catenin pathway has rarely been reported. The purpose of this study was to investigate whether CK1ε was involved in the osteogenic differentiation (OD) of BMSCs through the Wnt/β-catenin pathway and explore the mechanism. We used quantitative reverse transcription-polymerase chain reaction (qRT-qPCR), Western blots, immunofluorescence, alkaline phosphatase, and alizarin red staining to detect the effect of CK1ε on the OD of BMSCs and the Wnt/β-catenin signaling pathway. CK1ε was highly expressed in BMSCs with OD, and our study further demonstrated that CK1ε might promote the OD of BMSCs by activating DLV2 phosphorylation, initiating Wnt signaling downstream, and activating β-catenin nuclear transfer. In addition, by locally injecting a CK1ε-carrying adeno-associated virus (AAV5- CK1ε) into a femoral condyle defect rat model, the overexpression of CK1ε significantly promoted bone repair. Our data show that CK1ε was involved in the regulation of OD by mediating Wnt/β-catenin. This may provide a new strategy for the treatment of bone defects.
Collapse
Affiliation(s)
- Zhentang Yu
- Department of Orthopedics, The Affiliated Changzhou Second People’s Hospital of Nanjing Medical University, Changzhou 213000, China
- Department of Orthopedics, Yibin Integrated Traditional Chinese and Western Medicine Hospital, Yibin 644104, China
- Department of Graduate School, Dalian Medical University, Dalian 116000, China
- Changzhou Medical Center, Nanjing Medical University, Changzhou 213000, China
| | - Xijia Jiang
- Department of Orthopedics, The Affiliated Changzhou Second People’s Hospital of Nanjing Medical University, Changzhou 213000, China
- Changzhou Medical Center, Nanjing Medical University, Changzhou 213000, China
| | - Jianjian Yin
- Department of Orthopedics, The Affiliated Changzhou Second People’s Hospital of Nanjing Medical University, Changzhou 213000, China
- Changzhou Medical Center, Nanjing Medical University, Changzhou 213000, China
| | - Lei Han
- Department of Orthopedics, The Affiliated Changzhou Second People’s Hospital of Nanjing Medical University, Changzhou 213000, China
- Department of Graduate School, Dalian Medical University, Dalian 116000, China
- Changzhou Medical Center, Nanjing Medical University, Changzhou 213000, China
| | - Chengwei Xiong
- Department of Orthopedics, The Affiliated Changzhou Second People’s Hospital of Nanjing Medical University, Changzhou 213000, China
- Department of Graduate School, Dalian Medical University, Dalian 116000, China
- Changzhou Medical Center, Nanjing Medical University, Changzhou 213000, China
| | - Zhennan Huo
- Department of Orthopedics, The Affiliated Changzhou Second People’s Hospital of Nanjing Medical University, Changzhou 213000, China
- Department of Graduate School, Dalian Medical University, Dalian 116000, China
- Changzhou Medical Center, Nanjing Medical University, Changzhou 213000, China
| | - Jie Xu
- Department of Orthopedics, The Affiliated Changzhou Second People’s Hospital of Nanjing Medical University, Changzhou 213000, China
- Changzhou Medical Center, Nanjing Medical University, Changzhou 213000, China
| | - Jingjing Shang
- Department of Pharmacy, The Affiliated Changzhou Second People’s Hospital of Nanjing Medical University, Changzhou 213000, China
| | - Kun Xi
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Orthopedic Institute, Soochow University, Suzhou 215006, China
| | - Luming Nong
- Department of Orthopedics, The Affiliated Changzhou Second People’s Hospital of Nanjing Medical University, Changzhou 213000, China
- Changzhou Medical Center, Nanjing Medical University, Changzhou 213000, China
| | - Yong Huang
- Department of Orthopedics, The Affiliated Changzhou Second People’s Hospital of Nanjing Medical University, Changzhou 213000, China
- Changzhou Medical Center, Nanjing Medical University, Changzhou 213000, China
| | - Xindie Zhou
- Department of Orthopedics, The Affiliated Changzhou Second People’s Hospital of Nanjing Medical University, Changzhou 213000, China
- Department of Orthopedics, Gonghe County Hospital of Traditional Chinese Medicine, Hainan Tibetan Autonomous Prefecture, Qinghai 811800, China
- Changzhou Medical Center, Nanjing Medical University, Changzhou 213000, China
| |
Collapse
|
27
|
Dong Y, Chen Y, Ma G, Cao H. The role of E3 ubiquitin ligases in bone homeostasis and related diseases. Acta Pharm Sin B 2023; 13:3963-3987. [PMID: 37799379 PMCID: PMC10547920 DOI: 10.1016/j.apsb.2023.06.016] [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: 02/09/2022] [Revised: 05/16/2023] [Accepted: 05/18/2023] [Indexed: 10/07/2023] Open
Abstract
The ubiquitin-proteasome system (UPS) dedicates to degrade intracellular proteins to modulate demic homeostasis and functions of organisms. These enzymatic cascades mark and modifies target proteins diversly through covalently binding ubiquitin molecules. In the UPS, E3 ubiquitin ligases are the crucial constituents by the advantage of recognizing and presenting proteins to proteasomes for proteolysis. As the major regulators of protein homeostasis, E3 ligases are indispensable to proper cell manners in diverse systems, and they are well described in physiological bone growth and bone metabolism. Pathologically, classic bone-related diseases such as metabolic bone diseases, arthritis, bone neoplasms and bone metastasis of the tumor, etc., were also depicted in a UPS-dependent manner. Therefore, skeletal system is versatilely regulated by UPS and it is worthy to summarize the underlying mechanism. Furthermore, based on the current status of treatment, normal or pathological osteogenesis and tumorigenesis elaborated in this review highlight the clinical significance of UPS research. As a strategy possibly remedies the limitations of UPS treatment, emerging PROTAC was described comprehensively to illustrate its potential in clinical application. Altogether, the purpose of this review aims to provide more evidence for exploiting novel therapeutic strategies based on UPS for bone associated diseases.
Collapse
Affiliation(s)
| | | | - Guixing Ma
- Department of Biochemistry, School of Medicine, Southern University of Science and Technology, Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Key University Laboratory of Metabolism and Health of Guangdong, Shenzhen 518055, China
| | - Huiling Cao
- Department of Biochemistry, School of Medicine, Southern University of Science and Technology, Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Key University Laboratory of Metabolism and Health of Guangdong, Shenzhen 518055, China
| |
Collapse
|
28
|
Quadri N, Upadhyai P. Primary cilia in skeletal development and disease. Exp Cell Res 2023; 431:113751. [PMID: 37574037 DOI: 10.1016/j.yexcr.2023.113751] [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: 07/07/2023] [Revised: 08/09/2023] [Accepted: 08/11/2023] [Indexed: 08/15/2023]
Abstract
Primary cilia are non-motile, microtubule-based sensory organelle present in most vertebrate cells with a fundamental role in the modulation of organismal development, morphogenesis, and repair. Here we focus on the role of primary cilia in embryonic and postnatal skeletal development. We examine evidence supporting its involvement in physiochemical and developmental signaling that regulates proliferation, patterning, differentiation and homeostasis of osteoblasts, chondrocytes, and their progenitor cells in the skeleton. We discuss how signaling effectors in mechanotransduction and bone development, such as Hedgehog, Wnt, Fibroblast growth factor and second messenger pathways operate at least in part at the primary cilium. The relevance of primary cilia in bone formation and maintenance is underscored by a growing list of rare genetic skeletal ciliopathies. We collate these findings and summarize the current understanding of molecular factors and mechanisms governing primary ciliogenesis and ciliary function in skeletal development and disease.
Collapse
Affiliation(s)
- Neha Quadri
- Department of Medical Genetics, Kasturba Medical College, Manipal, Manipal Academy of Higher Education, Manipal, India
| | - Priyanka Upadhyai
- Department of Medical Genetics, Kasturba Medical College, Manipal, Manipal Academy of Higher Education, Manipal, India.
| |
Collapse
|
29
|
Hao RC, Li ZL, Wang FY, Tang J, Li PL, Yin BF, Li XT, Han MY, Mao N, Liu B, Ding L, Zhu H. Single-cell transcriptomic analysis identifies a highly replicating Cd168 + skeletal stem/progenitor cell population in mouse long bones. J Genet Genomics 2023; 50:702-712. [PMID: 37075860 DOI: 10.1016/j.jgg.2023.04.004] [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: 01/23/2023] [Revised: 04/08/2023] [Accepted: 04/09/2023] [Indexed: 04/21/2023]
Abstract
Skeletal stem/progenitor cells (SSPCs) are tissue-specific stem/progenitor cells localized within skeletons and contribute to bone development, homeostasis, and regeneration. However, the heterogeneity of SSPC populations in mouse long bones and their respective regenerative capacity remain to be further clarified. In this study, we perform integrated analysis using single-cell RNA sequencing (scRNA-seq) datasets of mouse hindlimb buds, postnatal long bones, and fractured long bones. Our analyses reveal the heterogeneity of osteochondrogenic lineage cells and recapitulate the developmental trajectories during mouse long bone growth. In addition, we identify a novel Cd168+ SSPC population with highly replicating capacity and osteochondrogenic potential in embryonic and postnatal long bones. Moreover, the Cd168+ SSPCs can contribute to newly formed skeletal tissues during fracture healing. Furthermore, the results of multicolor immunofluorescence show that Cd168+ SSPCs reside in the superficial zone of articular cartilage as well as in growth plates of postnatal mouse long bones. In summary, we identify a novel Cd168+ SSPC population with regenerative potential in mouse long bones, which adds to the knowledge of the tissue-specific stem cells in skeletons.
Collapse
Affiliation(s)
- Rui-Cong Hao
- Basic Medical College of Anhui Medical University, Hefei, Anhui 230032, China; Beijing Institute of Radiation Medicine, Beijing 100850, China
| | - Zhi-Ling Li
- Beijing Institute of Radiation Medicine, Beijing 100850, China
| | - Fei-Yan Wang
- Basic Medical College of Anhui Medical University, Hefei, Anhui 230032, China; Beijing Institute of Radiation Medicine, Beijing 100850, China
| | - Jie Tang
- Beijing Institute of Radiation Medicine, Beijing 100850, China
| | - Pei-Lin Li
- Beijing Institute of Radiation Medicine, Beijing 100850, China
| | - Bo-Feng Yin
- Beijing Institute of Radiation Medicine, Beijing 100850, China
| | - Xiao-Tong Li
- Beijing Institute of Radiation Medicine, Beijing 100850, China
| | - Meng-Yue Han
- Basic Medical College of Anhui Medical University, Hefei, Anhui 230032, China; Beijing Institute of Radiation Medicine, Beijing 100850, China
| | - Ning Mao
- Beijing Institute of Basic Medical Sciences, Beijing 100850, China
| | - Bing Liu
- State Key Laboratory of Experimental Hematology, Department of Hematology, Fifth Medical Center of Chinese PLA General Hospital, Beijing 100071, China
| | - Li Ding
- Basic Medical College of Anhui Medical University, Hefei, Anhui 230032, China; Air Force Medical Center, PLA, Beijing 100142, China.
| | - Heng Zhu
- Basic Medical College of Anhui Medical University, Hefei, Anhui 230032, China; Beijing Institute of Radiation Medicine, Beijing 100850, China.
| |
Collapse
|
30
|
Rinderknecht H, Mayer A, Histing T, Ehnert S, Nüssler A. Herbal Extracts of Ginseng and Maqui Berry Show Only Minimal Effects on an In Vitro Model of Early Fracture Repair of Smokers. Foods 2023; 12:2960. [PMID: 37569229 PMCID: PMC10419284 DOI: 10.3390/foods12152960] [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: 07/11/2023] [Revised: 07/28/2023] [Accepted: 07/29/2023] [Indexed: 08/13/2023] Open
Abstract
Smoking is a major risk factor for delayed fracture healing, affecting several aspects of early fracture repair, including inflammation, osteogenesis, and angiogenesis. Panax ginseng (GE) and maqui berry extract (MBE) were shown in our previous studies to reduce smoke-induced cellular damage in late bone-healing in vitro models. We aimed here to analyze their effects on the early fracture repair of smokers in a 3D co-culture model of fracture hematomas and endothelial cells. Both extracts did not alter the cellular viability at concentrations of up to 100 µg/mL. In early fracture repair in vitro, they were unable to reduce smoking-induced inflammation and induce osteo- or chondrogenicity. Regarding angiogenesis, smoking-induced stress in HUVECs could not be counteracted by both extracts. Furthermore, smoking-impaired tube formation was not restored by GE but was harmed by MBE. However, GE promoted angiogenesis initiation under smoking conditions via the Angpt/Tie2 axis. To summarize, cigarette smoking strikingly affected early fracture healing processes in vitro, but herbal extracts at the applied doses had only a limited effect. Since both extracts were shown before to be very effective in later stages of fracture healing, our data suggest that their early use immediately after fracture does not appear to negatively impact later beneficial effects.
Collapse
Affiliation(s)
| | | | | | | | - Andreas Nüssler
- Siegfried-Weller Institute for Trauma Research, BG Trauma Center, University of Tuebingen, Schnarrenbergstrasse 95, 72070 Tuebingen, Germany; (H.R.); (A.M.); (T.H.); (S.E.)
| |
Collapse
|
31
|
Gillet R, Boubaker F, Hossu G, Thay A, Gillet P, Blum A, Teixeira PAG. Computed Tomography Bone Imaging: Pushing the Boundaries in Clinical Practice. Semin Musculoskelet Radiol 2023; 27:397-410. [PMID: 37748463 DOI: 10.1055/s-0043-1768451] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/27/2023]
Abstract
Bone microarchitecture has several clinical implications over and above estimating bone strength. Computed tomography (CT) analysis mainly uses high-resolution peripheral quantitative CT and micro-CT, research imaging techniques, most often limited to peripheral skeleton assessment. Ultra-high-resolution (UHR) CT and photon-counting detector CT, two commercially available techniques, provide images that can approach the spatial resolution of the trabeculae, bringing bone microarchitecture analysis into clinical practice and improving depiction of bone vascularization, tumor matrix, and cortical and periosteal bone. This review presents bone microarchitecture anatomy, principles of analysis, reference measurements, and an update on the performance and potential clinical applications of these new CT techniques. We also share our clinical experience and technical considerations using an UHR-CT device.
Collapse
Affiliation(s)
- Romain Gillet
- Guilloz Imaging Department, Central Hospital, University Hospital Center of Nancy, Nancy, France
- Université de Lorraine, CIC, Innovation Technologique, University Hospital Center of Nancy, Nancy, France
- Université de Lorraine, INSERM, IADI, Nancy, France
| | - Fatma Boubaker
- Guilloz Imaging Department, Central Hospital, University Hospital Center of Nancy, Nancy, France
| | - Gabriela Hossu
- Université de Lorraine, CIC, Innovation Technologique, University Hospital Center of Nancy, Nancy, France
- Université de Lorraine, INSERM, IADI, Nancy, France
| | | | | | - Alain Blum
- Guilloz Imaging Department, Central Hospital, University Hospital Center of Nancy, Nancy, France
- Université de Lorraine, CIC, Innovation Technologique, University Hospital Center of Nancy, Nancy, France
- Université de Lorraine, INSERM, IADI, Nancy, France
| | - Pedro Augusto Gondim Teixeira
- Guilloz Imaging Department, Central Hospital, University Hospital Center of Nancy, Nancy, France
- Université de Lorraine, CIC, Innovation Technologique, University Hospital Center of Nancy, Nancy, France
- Université de Lorraine, INSERM, IADI, Nancy, France
| |
Collapse
|
32
|
Luo G, Sun Z, Liu H, Yuan Z, Wang W, Tu B, Li J, Fan C. Verteporfin attenuates trauma-induced heterotopic ossification of Achilles tendon by inhibiting osteogenesis and angiogenesis involving YAP/β-catenin signaling. FASEB J 2023; 37:e23057. [PMID: 37367700 DOI: 10.1096/fj.202300568r] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2023] [Revised: 06/05/2023] [Accepted: 06/13/2023] [Indexed: 06/28/2023]
Abstract
Heterotopic ossification occurs as a pathological ossification condition characterized by ectopic bone formation within soft tissues following trauma. Vascularization has long been established to fuel skeletal ossification during tissue development and regeneration. However, the feasibility of vascularization as a target of heterotopic ossification prevention remained to be further clarified. Here, we aimed to identify whether verteporfin as a widely used FDA-approved anti-vascularization drug could effectively inhibit trauma-induced heterotopic ossification formation. In the current study, we found that verteporfin not only dose dependently inhibited the angiogenic activity of human umbilical vein endothelial cells (HUVECs) but also the osteogenic differentiation of tendon stem cells (TDSCs). Moreover, YAP/β-catenin signaling axis was downregulated by the verteporfin. Application of lithium chloride, an agonist of β-catenin, recovered TDSCs osteogenesis and HUVECs angiogenesis that was inhibited by verteporfin. In vivo, verteporfin attenuated heterotopic ossification formation by decelerating osteogenesis and the vessels densely associated with osteoprogenitors formation, which could also be readily reversed by lithium chloride, as revealed by histological analysis and Micro-CT scan in a murine burn/tenotomy model. Collectively, this study confirmed the therapeutic effect of verteporfin on angiogenesis and osteogenesis in trauma-induced heterotopic ossification. Our study sheds light on the anti-vascularization strategy with verteporfin as a candidate treatment for heterotopic ossification prevention.
Collapse
Affiliation(s)
- Gang Luo
- Department of Orthopaedics, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, PR China
| | - Ziyang Sun
- Department of Orthopaedics, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, PR China
| | - Hang Liu
- Department of Orthopaedics, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, PR China
| | - Zhengqiang Yuan
- Department of Orthopaedics, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, PR China
| | - Wei Wang
- Department of Orthopaedics, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, PR China
| | - Bing Tu
- Department of Orthopaedics, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, PR China
| | - Juehong Li
- Department of Orthopaedics, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, PR China
| | - Cunyi Fan
- Department of Orthopaedics, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, PR China
- Shanghai Engineering Research Center for Orthopaedic Material Innovation and Tissue Regeneration, Shanghai, PR China
| |
Collapse
|
33
|
Watanabe H, Maishi N, Hoshi-Numahata M, Nishiura M, Nakanishi-Kimura A, Hida K, Iimura T. Skeletal-Vascular Interactions in Bone Development, Homeostasis, and Pathological Destruction. Int J Mol Sci 2023; 24:10912. [PMID: 37446097 DOI: 10.3390/ijms241310912] [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: 06/12/2023] [Revised: 06/28/2023] [Accepted: 06/28/2023] [Indexed: 07/15/2023] Open
Abstract
Bone is a highly vascularized organ that not only plays multiple roles in supporting the body and organs but also endows the microstructure, enabling distinct cell lineages to reciprocally interact. Recent studies have uncovered relevant roles of the bone vasculature in bone patterning, morphogenesis, homeostasis, and pathological bone destruction, including osteoporosis and tumor metastasis. This review provides an overview of current topics in the interactive molecular events between endothelial cells and bone cells during bone ontogeny and discusses the future direction of this research area to find novel ways to treat bone diseases.
Collapse
Affiliation(s)
- Haruhisa Watanabe
- Department of Pharmacology, Faculty and Graduate School of Dental Medicine, Hokkaido University, N13 W7, Sapporo 060-8586, Hokkaido, Japan
| | - Nako Maishi
- Department of Vascular Biology, Faculty and Graduate School of Dental Medicine, Hokkaido University, N13 W7, Sapporo 060-8586, Hokkaido, Japan
| | - Marie Hoshi-Numahata
- Department of Pharmacology, Faculty and Graduate School of Dental Medicine, Hokkaido University, N13 W7, Sapporo 060-8586, Hokkaido, Japan
| | - Mai Nishiura
- Department of Pharmacology, Faculty and Graduate School of Dental Medicine, Hokkaido University, N13 W7, Sapporo 060-8586, Hokkaido, Japan
| | - Atsuko Nakanishi-Kimura
- Department of Pharmacology, Faculty and Graduate School of Dental Medicine, Hokkaido University, N13 W7, Sapporo 060-8586, Hokkaido, Japan
| | - Kyoko Hida
- Department of Vascular Biology, Faculty and Graduate School of Dental Medicine, Hokkaido University, N13 W7, Sapporo 060-8586, Hokkaido, Japan
| | - Tadahiro Iimura
- Department of Pharmacology, Faculty and Graduate School of Dental Medicine, Hokkaido University, N13 W7, Sapporo 060-8586, Hokkaido, Japan
| |
Collapse
|
34
|
Wu N, Gao H, Wang X, Pei X. Surface Modification of Titanium Implants by Metal Ions and Nanoparticles for Biomedical Application. ACS Biomater Sci Eng 2023; 9:2970-2990. [PMID: 37184344 DOI: 10.1021/acsbiomaterials.2c00722] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/16/2023]
Abstract
Implant surface modification can improve osseointegration and reduce peri-implant inflammation. Implant surfaces are modified with metals because of their excellent mechanical properties and significant functions. Metal surface modification is divided into metal ions and nanoparticle surface modification. These two methods function by adding a finishing metal to the surface of the implant, and both play a role in promoting osteogenic, angiogenic, and antibacterial properties. Based on this, the nanostructural surface changes confer stronger antibacterial and cellular affinity to the implant surface. The current paper reviews the forms, mechanisms, and applications of nanoparticles and metal ion modifications to provide a foundation for the surface modification of implants.
Collapse
Affiliation(s)
- Nan Wu
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Prosthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, China
| | - Hongyu Gao
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Prosthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, China
| | - Xu Wang
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Prosthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, China
| | - Xibo Pei
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Prosthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, China
| |
Collapse
|
35
|
Buzkova P, Cauley JA, Fink HA, Robbins JA, Mukamal KJ, Barzilay JI. Age-Related Factors Associated With The Risk of Hip Fracture. Endocr Pract 2023; 29:478-483. [PMID: 36889582 PMCID: PMC10258141 DOI: 10.1016/j.eprac.2023.03.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Revised: 02/25/2023] [Accepted: 03/01/2023] [Indexed: 03/08/2023]
Abstract
OBJECTIVE Advancing age is a powerful risk factor for hip fractures. The biological mechanisms through which aging impacts the risk of hip fractures have not been well studied. METHODS Biological factors associated with "advancing age" that help to explain how aging is associated with the risk of hip fractures are reviewed. The findings are based on analyses of the Cardiovascular Health Study, an ongoing observational study of adults aged ≥65 years with 25 years of follow-up. RESULTS The following 5 age-related factors were found to be significantly associated with the risk of hip fractures: (1) microvascular disease of the kidneys (albuminuria and/or elevated urine-albumin-to-creatinine ratio) and brain (abnormal white matter disease on brain magnetic resonance imaging); (2) increased serum levels of carboxymethyl-lysine, an advanced glycation end product that reflects glycation and oxidative stress; (3) reduced parasympathetic tone, as derived from 24-hour Holter monitoring; (4) carotid artery atherosclerosis in the absence of clinical cardiovascular disease; and (5) increased transfatty acid levels in the blood. Each of these factors was associated with a 10% to 25% increased risk of fractures. These associations were independent of traditional risk factors for hip fractures. CONCLUSION Several factors associated with older age help to explain how "aging" may be associated with the risk of hip fractures. These same factors may also explain the high risk of mortality following hip fractures.
Collapse
Affiliation(s)
- Petra Buzkova
- Department of Biostatistics, School of Public Health, University of Washington, Seattle, WA
| | - Jane A Cauley
- Department of Epidemiology, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, PA
| | - Howard A Fink
- Geriatric Research Education and Clinical Center, VA Health Care System, Minneapolis, MN
| | - John A Robbins
- Department of Medicine, University of California, Irvine, CA
| | - Kenneth J Mukamal
- Department of Medicine, Beth Israel Deaconess Medical Center, Brookline, MA
| | - Joshua I Barzilay
- Division of Endocrinology, Kaiser Permanente of Georgia, and Department of Endocrinology, Emory University School of Medicine, Atlanta, GA.
| |
Collapse
|
36
|
Guo Z, Li J, Tan J, Sun S, Yan Q, Qin H. Exosomal miR-214-3p from senescent osteoblasts accelerates endothelial cell senescence. J Orthop Surg Res 2023; 18:391. [PMID: 37248458 DOI: 10.1186/s13018-023-03859-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Accepted: 05/16/2023] [Indexed: 05/31/2023] Open
Abstract
BACKGROUND Osteoporosis is a common systemic bone disease that leads to bone fragility and increases the risk of fracture. However, the pathogenesis of osteoporosis is considered to be highly complex. The exosomes can regulate the communication between cells. The specific mechanism of information transmission between osteoblasts and endothelial cells is worthy of further study. METHODS Exosomes were isolated and verified from senescent osteoblasts. The source and properties of exosomes were determined by TEM, particle size analysis and western blot. We established the co-culture model of endothelial cells and senescent osteoblasts. We used qRT-PCR to identify differentially expressed miRNAs. The functional changes of vascular endothelial cells were verified by cell transfection. β-Galactosidase cell senescence assay, Hoechst cell apoptosis assay, Ki67 cell proliferation assay and Transwell migration assay were used to verify cell senescence, apoptosis, proliferation, and migration. The potential target gene of miRNA was detected by bio-informatics pathway and double luciferase report. RESULTS We discovered that senescent osteoblasts could promote the senescence and apoptosis of vascular endothelial cells and inhibit their proliferation and migration. miR-214-3p was upregulated in senescent osteoblast-derived exosomes. miR-214-3p could effectively promote senescence and apoptosis of endothelial cells and inhibit proliferation and migration ability. L1CAM is a miR-214-3p direct target gene determined by bio-informatics and double luciferase report. CONCLUSIONS In conclusion, senescent osteoblast-derived exosomes can accelerate endothelial cell senescence through miR-214-3p/L1CAM pathway. Our experiments reveal the role of exosomes in the skeletal microenvironment.
Collapse
Affiliation(s)
- Zhen Guo
- Department of Physiology, Guangxi Medical University, Nanning, People's Republic of China
| | - Jing Li
- Department of Physiology, Guangxi Medical University, Nanning, People's Republic of China
| | - Jiyong Tan
- Key Laboratory of Longevity and Aging-Related Diseases, Ministry of Education, Guangxi Medical University, Nanning, People's Republic of China
| | - Sainan Sun
- Department of Physiology, Guangxi Medical University, Nanning, People's Republic of China
| | - Qing Yan
- Department of Physiology, Guangxi Medical University, Nanning, People's Republic of China
| | - Hao Qin
- Department of Orthopedics, Guigang City People's Hospital, No. 1 Zhongshan Middle Road, Gangbei District, Guigang, 537100, Guangxi, People's Republic of China.
| |
Collapse
|
37
|
Andreasen CM, El-Masri BM, MacDonald B, Laursen KS, Nielsen MH, Thomsen JS, Delaisse JM, Andersen TL. Local coordination between intracortical bone remodeling and vascular development in human juvenile bone. Bone 2023; 173:116787. [PMID: 37150243 DOI: 10.1016/j.bone.2023.116787] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/21/2023] [Revised: 05/02/2023] [Accepted: 05/02/2023] [Indexed: 05/09/2023]
Abstract
Although failure to establish a vascular network has been associated with many skeletal disorders, little is known about what drives development of vasculature in the intracortical bone compartments. Here, we show that intracortical bone resorption events are coordinated with development of the vasculature. We investigated the prevalence of vascular structures at different remodeling stages as well as their 3D organization using proximal femoral cortical bone from 5 girls and 6 boys (aged 6-15 years). A 2D analysis revealed that non-quiescent intracortical pores contained more vascular structures than quiescent pores (p < 0.0001). Type 2 pores, i.e., remodeling of existing pores, had a higher density of vascular structures than type 1 pores, i.e., de novo created pores (p < 0.05). Furthermore, pores at the eroded-formative remodeling stage, had more vascular structures than pores at any other remodeling stage (p < 0.05). A 3D reconstruction of an intracortical remodeling event showed that osteoclasts in the advancing tip of the cutting cone as well as preosteoclasts in the lumen expressed vascular endothelial growth factor-A (VEGFA), while VEGFA-receptors 1 and 2 mainly were expressed in endothelial cells in the adjacent vasculature. Consequently, we propose that the progression of the vascular network in intracortical remodeling events is driven by osteoclasts expressing VEGFA. Moreover, the vasculature is continuously reconfigured according to the demands of the remodeling events at the surrounding bone surfaces.
Collapse
Affiliation(s)
- Christina Møller Andreasen
- Department of Pathology, Odense University Hospital, Odense, Denmark; Molecular Bone Histology lab, Pathology Research Unit, Department of Clinical Research, University of Southern Denmark, Denmark; Department of Molecular Medicine, University of Southern Denmark, Denmark.
| | - Bilal Mohamad El-Masri
- Department of Pathology, Odense University Hospital, Odense, Denmark; Molecular Bone Histology lab, Pathology Research Unit, Department of Clinical Research, University of Southern Denmark, Denmark; Department of Molecular Medicine, University of Southern Denmark, Denmark.
| | - Birgit MacDonald
- Department of Pathology, Odense University Hospital, Odense, Denmark; Molecular Bone Histology lab, Pathology Research Unit, Department of Clinical Research, University of Southern Denmark, Denmark; Clinical Cell Biology, Vejle Hospital - Lillebaelt Hospital, Institute of Regional Health Research, University of Southern Denmark, Denmark
| | - Kaja Søndergaard Laursen
- Clinical Cell Biology, Vejle Hospital - Lillebaelt Hospital, Institute of Regional Health Research, University of Southern Denmark, Denmark; Molecular Bone Histology lab, Department of Forensic Medicine, Aarhus University, Aarhus, Denmark.
| | - Malene Hykkelbjerg Nielsen
- Department of Pathology, Odense University Hospital, Odense, Denmark; Molecular Bone Histology lab, Pathology Research Unit, Department of Clinical Research, University of Southern Denmark, Denmark; Department of Molecular Medicine, University of Southern Denmark, Denmark.
| | | | - Jean-Marie Delaisse
- Department of Pathology, Odense University Hospital, Odense, Denmark; Molecular Bone Histology lab, Pathology Research Unit, Department of Clinical Research, University of Southern Denmark, Denmark; Department of Molecular Medicine, University of Southern Denmark, Denmark; Clinical Cell Biology, Vejle Hospital - Lillebaelt Hospital, Institute of Regional Health Research, University of Southern Denmark, Denmark.
| | - Thomas Levin Andersen
- Department of Pathology, Odense University Hospital, Odense, Denmark; Molecular Bone Histology lab, Pathology Research Unit, Department of Clinical Research, University of Southern Denmark, Denmark; Department of Molecular Medicine, University of Southern Denmark, Denmark; Clinical Cell Biology, Vejle Hospital - Lillebaelt Hospital, Institute of Regional Health Research, University of Southern Denmark, Denmark; Molecular Bone Histology lab, Department of Forensic Medicine, Aarhus University, Aarhus, Denmark.
| |
Collapse
|
38
|
You J, Liu M, Li M, Zhai S, Quni S, Zhang L, Liu X, Jia K, Zhang Y, Zhou Y. The Role of HIF-1α in Bone Regeneration: A New Direction and Challenge in Bone Tissue Engineering. Int J Mol Sci 2023; 24:ijms24098029. [PMID: 37175732 PMCID: PMC10179302 DOI: 10.3390/ijms24098029] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2023] [Revised: 04/22/2023] [Accepted: 04/25/2023] [Indexed: 05/15/2023] Open
Abstract
The process of repairing significant bone defects requires the recruitment of a considerable number of cells for osteogenesis-related activities, which implies the consumption of a substantial amount of oxygen and nutrients. Therefore, the limited supply of nutrients and oxygen at the defect site is a vital constraint that affects the regenerative effect, which is closely related to the degree of a well-established vascular network. Hypoxia-inducible factor (HIF-1α), which is an essential transcription factor activated in hypoxic environments, plays a vital role in vascular network construction. HIF-1α, which plays a central role in regulating cartilage and bone formation, induces vascular invasion and differentiation of osteoprogenitor cells to promote and maintain extracellular matrix production by mediating the adaptive response of cells to changes in oxygen levels. However, the application of HIF-1α in bone tissue engineering is still controversial. As such, clarifying the function of HIF-1α in regulating the bone regeneration process is one of the urgent issues that need to be addressed. This review provides insight into the mechanisms of HIF-1α action in bone regeneration and related recent advances. It also describes current strategies for applying hypoxia induction and hypoxia mimicry in bone tissue engineering, providing theoretical support for the use of HIF-1α in establishing a novel and feasible bone repair strategy in clinical settings.
Collapse
Affiliation(s)
- Jiaqian You
- Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, Hospital of Stomatology, Jilin University, Changchun 130021, China
- School of Stomatology, Jilin University, Changchun 130021, China
| | - Manxuan Liu
- School of Stomatology, Jilin University, Changchun 130021, China
| | - Minghui Li
- School of Stomatology, Jilin University, Changchun 130021, China
| | - Shaobo Zhai
- School of Stomatology, Jilin University, Changchun 130021, China
| | - Sezhen Quni
- School of Stomatology, Jilin University, Changchun 130021, China
| | - Lu Zhang
- School of Stomatology, Jilin University, Changchun 130021, China
| | - Xiuyu Liu
- School of Stomatology, Jilin University, Changchun 130021, China
| | - Kewen Jia
- School of Stomatology, Jilin University, Changchun 130021, China
| | - Yidi Zhang
- Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, Hospital of Stomatology, Jilin University, Changchun 130021, China
- School of Stomatology, Jilin University, Changchun 130021, China
| | - Yanmin Zhou
- Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, Hospital of Stomatology, Jilin University, Changchun 130021, China
- School of Stomatology, Jilin University, Changchun 130021, China
| |
Collapse
|
39
|
Hosseini FS, Abedini AA, Chen F, Whitfield T, Ude CC, Laurencin CT. Oxygen-Generating Biomaterials for Translational Bone Regenerative Engineering. ACS APPLIED MATERIALS & INTERFACES 2023; 15:50721-50741. [PMID: 36988393 DOI: 10.1021/acsami.2c20715] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/19/2023]
Abstract
Successful regeneration of critical-size defects remains one of the significant challenges in regenerative engineering. These large-scale bone defects are difficult to regenerate and are often reconstructed with matrices that do not provide adequate oxygen levels to stem cells involved in the regeneration process. Hypoxia-induced necrosis predominantly occurs in the center of large matrices since the host tissue's local vasculature fails to provide sufficient nutrients and oxygen. Indeed, utilizing oxygen-generating materials can overcome the central hypoxic region, induce tissue in-growth, and increase the quality of life for patients with extensive tissue damage. This article reviews recent advances in oxygen-generating biomaterials for translational bone regenerative engineering. We discussed different oxygen-releasing and delivery methods, fabrication methods for oxygen-releasing matrices, biology, oxygen's role in bone regeneration, and emerging new oxygen delivery methods that could potentially be used for bone regenerative engineering.
Collapse
Affiliation(s)
- Fatemeh S Hosseini
- Connecticut Convergence Institute for Translation in Regenerative Engineering, UConn Health, Farmington, Connecticut 06030, United States
- Raymond and Beverly Sackler Center for Biomedical, Biological, Physical and Engineering Sciences, UConn Health, Farmington, Connecticut 06030, United States
- Department of Skeletal Biology and Regeneration, UConn Health, Farmington, Connecticut 06030, United States
- Department of Orthopedic Surgery, UConn Health, Farmington, Connecticut 06030, United States
| | - Amir Abbas Abedini
- Connecticut Convergence Institute for Translation in Regenerative Engineering, UConn Health, Farmington, Connecticut 06030, United States
- Raymond and Beverly Sackler Center for Biomedical, Biological, Physical and Engineering Sciences, UConn Health, Farmington, Connecticut 06030, United States
- Department of Materials Science and Engineering, University of Connecticut, Storrs, Connecticut 06269, United States
| | - Feiyang Chen
- Connecticut Convergence Institute for Translation in Regenerative Engineering, UConn Health, Farmington, Connecticut 06030, United States
| | - Taraje Whitfield
- Connecticut Convergence Institute for Translation in Regenerative Engineering, UConn Health, Farmington, Connecticut 06030, United States
- Raymond and Beverly Sackler Center for Biomedical, Biological, Physical and Engineering Sciences, UConn Health, Farmington, Connecticut 06030, United States
- Department of Skeletal Biology and Regeneration, UConn Health, Farmington, Connecticut 06030, United States
| | - Chinedu C Ude
- Connecticut Convergence Institute for Translation in Regenerative Engineering, UConn Health, Farmington, Connecticut 06030, United States
- Raymond and Beverly Sackler Center for Biomedical, Biological, Physical and Engineering Sciences, UConn Health, Farmington, Connecticut 06030, United States
- Department of Orthopedic Surgery, UConn Health, Farmington, Connecticut 06030, United States
| | - Cato T Laurencin
- Connecticut Convergence Institute for Translation in Regenerative Engineering, UConn Health, Farmington, Connecticut 06030, United States
- Raymond and Beverly Sackler Center for Biomedical, Biological, Physical and Engineering Sciences, UConn Health, Farmington, Connecticut 06030, United States
- Department of Skeletal Biology and Regeneration, UConn Health, Farmington, Connecticut 06030, United States
- Department of Orthopedic Surgery, UConn Health, Farmington, Connecticut 06030, United States
- Department of Biomedical Engineering, University of Connecticut, Storrs, Connecticut 06269, United States
- Department of Chemical and Bimolecular Engineering, University of Connecticut, Storrs, Connecticut 06269, United States
- Department of Materials Science and Engineering, University of Connecticut, Storrs, Connecticut 06269, United States
| |
Collapse
|
40
|
de Wildt BWM, Cramer EEA, de Silva LS, Ito K, Gawlitta D, Hofmann S. Evaluating material-driven regeneration in a tissue engineered human in vitro bone defect model. Bone 2023; 166:116597. [PMID: 36280106 DOI: 10.1016/j.bone.2022.116597] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/15/2022] [Revised: 10/07/2022] [Accepted: 10/18/2022] [Indexed: 11/16/2022]
Abstract
Advanced in vitro human bone defect models can contribute to the evaluation of materials for in situ bone regeneration, addressing both translational and ethical concerns regarding animal models. In this study, we attempted to develop such a model to study material-driven regeneration, using a tissue engineering approach. By co-culturing human umbilical vein endothelial cells (HUVECs) with human bone marrow-derived mesenchymal stromal cells (hBMSCs) on silk fibroin scaffolds with in vitro critically sized defects, the growth of vascular-like networks and three-dimensional bone-like tissue was facilitated. After a model build-up phase of 28 days, materials were artificially implanted and HUVEC and hBMSC migration, cell-material interactions, and osteoinduction were evaluated 14 days after implantation. The materials physiologically relevant for bone regeneration included a platelet gel as blood clot mimic, cartilage spheres as soft callus mimics, and a fibrin gel as control. Although the in vitro model was limited in the evaluation of immune responses, hallmarks of physiological bone regeneration were observed in vitro. These included the endothelial cell chemotaxis induced by the blood clot mimic and the mineralization of the soft callus mimic. Therefore, the present in vitro model could contribute to an improved pre-clinical evaluation of biomaterials while reducing the need for animal experiments.
Collapse
Affiliation(s)
- Bregje W M de Wildt
- Orthopaedic Biomechanics, Department of Biomedical Engineering, Institute for Complex Molecular Systems (ICMS), Eindhoven University of Technology, Eindhoven, the Netherlands
| | - Esther E A Cramer
- Orthopaedic Biomechanics, Department of Biomedical Engineering, Institute for Complex Molecular Systems (ICMS), Eindhoven University of Technology, Eindhoven, the Netherlands
| | - Leanne S de Silva
- Department of Oral and Maxillofacial Surgery & Special Dental Care, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands; Regenerative Medicine Center Utrecht, Utrecht, the Netherlands
| | - Keita Ito
- Orthopaedic Biomechanics, Department of Biomedical Engineering, Institute for Complex Molecular Systems (ICMS), Eindhoven University of Technology, Eindhoven, the Netherlands
| | - Debby Gawlitta
- Department of Oral and Maxillofacial Surgery & Special Dental Care, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands; Regenerative Medicine Center Utrecht, Utrecht, the Netherlands
| | - Sandra Hofmann
- Orthopaedic Biomechanics, Department of Biomedical Engineering, Institute for Complex Molecular Systems (ICMS), Eindhoven University of Technology, Eindhoven, the Netherlands.
| |
Collapse
|
41
|
Ma Q, Miri Z, Haugen HJ, Moghanian A, Loca D. Significance of mechanical loading in bone fracture healing, bone regeneration, and vascularization. J Tissue Eng 2023; 14:20417314231172573. [PMID: 37251734 PMCID: PMC10214107 DOI: 10.1177/20417314231172573] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2023] [Accepted: 04/13/2023] [Indexed: 05/31/2023] Open
Abstract
In 1892, J.L. Wolff proposed that bone could respond to mechanical and biophysical stimuli as a dynamic organ. This theory presents a unique opportunity for investigations on bone and its potential to aid in tissue repair. Routine activities such as exercise or machinery application can exert mechanical loads on bone. Previous research has demonstrated that mechanical loading can affect the differentiation and development of mesenchymal tissue. However, the extent to which mechanical stimulation can help repair or generate bone tissue and the related mechanisms remain unclear. Four key cell types in bone tissue, including osteoblasts, osteoclasts, bone lining cells, and osteocytes, play critical roles in responding to mechanical stimuli, while other cell lineages such as myocytes, platelets, fibroblasts, endothelial cells, and chondrocytes also exhibit mechanosensitivity. Mechanical loading can regulate the biological functions of bone tissue through the mechanosensor of bone cells intraosseously, making it a potential target for fracture healing and bone regeneration. This review aims to clarify these issues and explain bone remodeling, structure dynamics, and mechano-transduction processes in response to mechanical loading. Loading of different magnitudes, frequencies, and types, such as dynamic versus static loads, are analyzed to determine the effects of mechanical stimulation on bone tissue structure and cellular function. Finally, the importance of vascularization in nutrient supply for bone healing and regeneration was further discussed.
Collapse
Affiliation(s)
- Qianli Ma
- Department of Biomaterials, Institute
of Clinical Dentistry, University of Oslo, Norway
- Department of Immunology, School of
Basic Medicine, Fourth Military Medical University, Xi’an, PR China
| | - Zahra Miri
- Department of Materials Engineering,
Isfahan University of Technology, Isfahan, Iran
| | - Håvard Jostein Haugen
- Department of Biomaterials, Institute
of Clinical Dentistry, University of Oslo, Norway
| | - Amirhossein Moghanian
- Department of Materials Engineering,
Imam Khomeini International University, Qazvin, Iran
| | - Dagnjia Loca
- Rudolfs Cimdins Riga Biomaterials
Innovations and Development Centre, Institute of General Chemical Engineering,
Faculty of Materials Science and Applied Chemistry, Riga Technical University, Riga,
Latvia
- Baltic Biomaterials Centre of
Excellence, Headquarters at Riga Technical University, Riga, Latvia
| |
Collapse
|
42
|
Qin Y, Chen ZH, Wu JJ, Zhang ZY, Yuan ZD, Guo DY, Chen MN, Li X, Yuan FL. Circadian clock genes as promising therapeutic targets for bone loss. Biomed Pharmacother 2023; 157:114019. [PMID: 36423544 DOI: 10.1016/j.biopha.2022.114019] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2022] [Revised: 11/11/2022] [Accepted: 11/13/2022] [Indexed: 11/22/2022] Open
Abstract
The circadian clock regulates many key physiological processes such as the sleep-wake cycle, hormone release, cardiovascular health, glucose metabolism and body temperature. Recent evidence has suggested a critical role of the circadian system in controlling bone metabolism. Here we review the connection between bone metabolism and the biological clock, and the roles of these mechanisms in bone loss. We also analyze the regulatory effects of clock-related genes on signaling pathways and transcription factors in osteoblasts and osteoclasts. Additionally, osteocytes and endothelial cells (ECs) regulated by the circadian clock are also discussed in our review. Furthermore, we also summarize the regulation of circadian clock genes by some novel modulators, which provides us with a new insight into a potential strategy to prevent and treat bone diseases such as osteoporosis by targeting circadian genes.
Collapse
Affiliation(s)
- Yi Qin
- Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Zhong-Hua Chen
- Institute of Integrated Chinese and Western Medicine, The Hospital Affiliated to Jiangnan University, Wuxi, Jiangsu 214041, China
| | - Jun-Jie Wu
- Institute of Integrated Chinese and Western Medicine, The Hospital Affiliated to Jiangnan University, Wuxi, Jiangsu 214041, China
| | - Zhen-Yu Zhang
- Institute of Integrated Chinese and Western Medicine, The Hospital Affiliated to Jiangnan University, Wuxi, Jiangsu 214041, China
| | - Zheng-Dong Yuan
- Institute of Integrated Chinese and Western Medicine, The Hospital Affiliated to Jiangnan University, Wuxi, Jiangsu 214041, China
| | - Dan-Yang Guo
- Institute of Integrated Chinese and Western Medicine, The Hospital Affiliated to Jiangnan University, Wuxi, Jiangsu 214041, China
| | - Meng-Nan Chen
- Institute of Integrated Chinese and Western Medicine, The Hospital Affiliated to Jiangnan University, Wuxi, Jiangsu 214041, China
| | - Xia Li
- Institute of Integrated Chinese and Western Medicine, The Hospital Affiliated to Jiangnan University, Wuxi, Jiangsu 214041, China.
| | - Feng-Lai Yuan
- Institute of Integrated Chinese and Western Medicine, The Hospital Affiliated to Jiangnan University, Wuxi, Jiangsu 214041, China.
| |
Collapse
|
43
|
Feng H, Jiang B, Xing W, Sun J, Greenblatt MB, Zou W. Skeletal stem cells: origins, definitions, and functions in bone development and disease. LIFE MEDICINE 2022; 1:276-293. [PMID: 36811112 PMCID: PMC9938638 DOI: 10.1093/lifemedi/lnac048] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Accepted: 11/04/2022] [Indexed: 12/13/2022]
Abstract
Skeletal stem cells (SSCs) are tissue-specific stem cells that can self-renew and sit at the apex of their differentiation hierarchy, giving rise to mature skeletal cell types required for bone growth, maintenance, and repair. Dysfunction in SSCs is caused by stress conditions like ageing and inflammation and is emerging as a contributor to skeletal pathology, such as the pathogenesis of fracture nonunion. Recent lineage tracing experiments have shown that SSCs exist in the bone marrow, periosteum, and resting zone of the growth plate. Unraveling their regulatory networks is crucial for understanding skeletal diseases and developing therapeutic strategies. In this review, we systematically introduce the definition, location, stem cell niches, regulatory signaling pathways, and clinical applications of SSCs.
Collapse
Affiliation(s)
- Heng Feng
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, CAS Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai 200031, China
| | - Bo Jiang
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, CAS Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai 200031, China
| | - Wenhui Xing
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, CAS Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai 200031, China
| | - Jun Sun
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY 10065, USA
| | - Matthew B Greenblatt
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY 10065, USA
- Research Division, Hospital for Special Surgery, New York, NY 10065, USA
| | - Weiguo Zou
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, CAS Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai 200031, China
- Institute of Microsurgery on Extremities, Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, Shanghai 200233, China
| |
Collapse
|
44
|
Schilling K, Zhai Y, Zhou Z, Zhou B, Brown E, Zhang X. High-resolution imaging of the osteogenic and angiogenic interface at the site of murine cranial bone defect repair via multiphoton microscopy. eLife 2022; 11:e83146. [PMID: 36326085 PMCID: PMC9678361 DOI: 10.7554/elife.83146] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Accepted: 10/31/2022] [Indexed: 11/05/2022] Open
Abstract
The spatiotemporal blood vessel formation and specification at the osteogenic and angiogenic interface of murine cranial bone defect repair were examined utilizing a high-resolution multiphoton-based imaging platform in conjunction with advanced optical techniques that allow interrogation of the oxygen microenvironment and cellular energy metabolism in living animals. Our study demonstrates the dynamic changes of vessel types, that is, arterial, venous, and capillary vessel networks at the superior and dura periosteum of cranial bone defect, suggesting a differential coupling of the vessel type with osteoblast expansion and bone tissue deposition/remodeling during repair. Employing transgenic reporter mouse models that label distinct types of vessels at the site of repair, we further show that oxygen distributions in capillary vessels at the healing site are heterogeneous as well as time- and location-dependent. The endothelial cells coupling to osteoblasts prefer glycolysis and are less sensitive to microenvironmental oxygen changes than osteoblasts. In comparison, osteoblasts utilize relatively more OxPhos and potentially consume more oxygen at the site of repair. Taken together, our study highlights the dynamics and functional significance of blood vessel types at the site of defect repair, opening up opportunities for further delineating the oxygen and metabolic microenvironment at the interface of bone tissue regeneration.
Collapse
Affiliation(s)
- Kevin Schilling
- Center for Musculoskeletal Research, University of Rochester, School of Medicine and DentistryRochesterUnited States
- Department of Biomedical Engineering, University of RochesterRochesterUnited States
| | - Yuankun Zhai
- Center for Musculoskeletal Research, University of Rochester, School of Medicine and DentistryRochesterUnited States
| | - Zhuang Zhou
- Center for Musculoskeletal Research, University of Rochester, School of Medicine and DentistryRochesterUnited States
| | - Bin Zhou
- Shanghai Institutes for Biological SciencesShanghaiChina
| | - Edward Brown
- Department of Biomedical Engineering, University of RochesterRochesterUnited States
| | - Xinping Zhang
- Center for Musculoskeletal Research, University of Rochester, School of Medicine and DentistryRochesterUnited States
| |
Collapse
|
45
|
Naiche LA, Villa SR, Kitajewski JK. Endothelial Cell Fate Determination: A Top Notch Job in Vascular Decision-Making. Cold Spring Harb Perspect Med 2022; 12:a041183. [PMID: 35288401 PMCID: PMC9619357 DOI: 10.1101/cshperspect.a041183] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
As vascular networks form, endothelial cells (ECs) undergo cell fate decisions that determine whether they become tip or stalk cells of the developing vascular plexus or mature into arterial, venous, or lymphatic endothelium. EC fate decisions are coordinated with neighboring cells to initiate sprouting, maintain endothelial barrier, or ensure appropriate specialization of vessels. We describe mechanisms that control EC fate at specific steps in these processes, with an emphasis on the role of the Notch signaling pathway. Specific EC fate determination steps that are highlighted are tip/stalk selection during sprouting angiogenesis, venous-arterial specification, arteriogenesis, lymphatic vessel specification, and lymphatic valve formation.
Collapse
Affiliation(s)
- L A Naiche
- Department of Physiology and Biophysics, University of Illinois Chicago, Chicago, Illinois 60612, USA
| | - Stephanie R Villa
- Department of Physiology and Biophysics, University of Illinois Chicago, Chicago, Illinois 60612, USA
| | - Jan K Kitajewski
- Department of Physiology and Biophysics, University of Illinois Chicago, Chicago, Illinois 60612, USA
- University of Illinois Cancer Center, Chicago, Illinois 60612, USA
| |
Collapse
|
46
|
Tiffany AS, Harley BAC. Growing Pains: The Need for Engineered Platforms to Study Growth Plate Biology. Adv Healthc Mater 2022; 11:e2200471. [PMID: 35905390 PMCID: PMC9547842 DOI: 10.1002/adhm.202200471] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Revised: 07/11/2022] [Indexed: 01/27/2023]
Abstract
Growth plates, or physis, are highly specialized cartilage tissues responsible for longitudinal bone growth in children and adolescents. Chondrocytes that reside in growth plates are organized into three distinct zones essential for proper function. Modeling key features of growth plates may provide an avenue to develop advanced tissue engineering strategies and perspectives for cartilage and bone regenerative medicine applications and a platform to study processes linked to disease progression. In this review, a brief introduction of the growth plates and their role in skeletal development is first provided. Injuries and diseases of the growth plates as well as physiological and pathological mechanisms associated with remodeling and disease progression are discussed. Growth plate biology, namely, its architecture and extracellular matrix organization, resident cell types, and growth factor signaling are then focused. Next, opportunities and challenges for developing 3D biomaterial models to study aspects of growth plate biology and disease in vitro are discussed. Finally, opportunities for increasingly sophisticated in vitro biomaterial models of the growth plate to study spatiotemporal aspects of growth plate remodeling, to investigate multicellular signaling underlying growth plate biology, and to develop platforms that address key roadblocks to in vivo musculoskeletal tissue engineering applications are described.
Collapse
Affiliation(s)
- Aleczandria S. Tiffany
- Department of Chemical and Biomolecular EngineeringUniversity of Illinois at Urbana‐ChampaignUrbanaIL61801USA
| | - Brendan A. C. Harley
- Department of Chemical and Biomolecular EngineeringUniversity of Illinois at Urbana‐ChampaignUrbanaIL61801USA
- Carl R. Woese Institute for Genomic BiologyUniversity of Illinois at Urbana‐ChampaignUrbanaIL61801USA
| |
Collapse
|
47
|
Qin Q, Liu Y, Yang Z, Aimaijiang M, Ma R, Yang Y, Zhang Y, Zhou Y. Hypoxia-Inducible Factors Signaling in Osteogenesis and Skeletal Repair. Int J Mol Sci 2022; 23:ijms231911201. [PMID: 36232501 PMCID: PMC9569554 DOI: 10.3390/ijms231911201] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Revised: 09/10/2022] [Accepted: 09/14/2022] [Indexed: 11/23/2022] Open
Abstract
Sufficient oxygen is required to maintain normal cellular and physiological function, such as a creature’s development, breeding, and homeostasis. Lately, some researchers have reported that both pathological hypoxia and environmental hypoxia might affect bone health. Adaptation to hypoxia is a pivotal cellular event in normal cell development and differentiation and in pathological settings such as ischemia. As central mediators of homeostasis, hypoxia-inducible transcription factors (HIFs) can allow cells to survive in a low-oxygen environment and are essential for the regulation of osteogenesis and skeletal repair. From this perspective, we summarized the role of HIF-1 and HIF-2 in signaling pathways implicated in bone development and skeletal repair and outlined the molecular mechanism of regulation of downstream growth factors and protein molecules such as VEGF, EPO, and so on. All of these present an opportunity for developing therapies for bone regeneration.
Collapse
|
48
|
Cheng L, Yang F, Chen X, Kang J, Li J, Zhang Y, Liu J, Li J, Ma J, Duan J. Identification of Novel Compound Heterozygous Variants of MMP9 in Fetus With Metaphyseal Anadysplasia Type 2. Front Genet 2022; 13:938457. [PMID: 36035187 PMCID: PMC9411662 DOI: 10.3389/fgene.2022.938457] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2022] [Accepted: 06/14/2022] [Indexed: 11/13/2022] Open
Abstract
Matrix metalloproteinase 9 (MMP9) is an important member of the matrix metalloproteinase family and plays a key role in balancing extracellular matrix proteins. Studies have shown that the homozygous mutations in MMP9 can lead to metaphyseal anadysplasia type 2 (MANDP2, OMIM#613073). The clinical phenotype of this disease is limited and there were only five reported cases of MANDP2 associated with homozygous MMP9 mutations from three families. In this study, we described a case of a fetus with skeletal system malformation. The main clinical manifestations include the short bilateral femur, absence of right fibula, and curved ipsilateral tibia with short length. Importantly, two novel compound heterozygous variants of the MMP9 gene (NM_004,994.3: c.151C > T and c.929del) were found through the trio whole exome sequencing and Sanger sequencing. This is the first report that identified the compound heterozygous variants of the MMP9 gene associated with metaphyseal dysplasia type 2.
Collapse
Affiliation(s)
- Lin Cheng
- Department of Obstetrics and Gynecology, Zhongnan Hospital of Wuhan University, Wuhan, China
- Hubei Clinical Research Center for Prenatal Diagnosis and Birth Health, Wuhan, China
- Wuhan Clinical Research Center for Reproductive Science and Birth Health, Wuhan, China
| | - Fan Yang
- Department of Ultrasound Imaging, Hubei Maternal and Child Health Hospital, Wuhan, China
| | - Xinlin Chen
- Department of Ultrasound Imaging, Hubei Maternal and Child Health Hospital, Wuhan, China
| | - Jiawei Kang
- Department of Obstetrics and Gynecology, Zhongnan Hospital of Wuhan University, Wuhan, China
- Hubei Clinical Research Center for Prenatal Diagnosis and Birth Health, Wuhan, China
- Wuhan Clinical Research Center for Reproductive Science and Birth Health, Wuhan, China
| | - Jiafu Li
- Department of Obstetrics and Gynecology, Zhongnan Hospital of Wuhan University, Wuhan, China
- Hubei Clinical Research Center for Prenatal Diagnosis and Birth Health, Wuhan, China
- Wuhan Clinical Research Center for Reproductive Science and Birth Health, Wuhan, China
| | - Yuanzhen Zhang
- Department of Obstetrics and Gynecology, Zhongnan Hospital of Wuhan University, Wuhan, China
- Hubei Clinical Research Center for Prenatal Diagnosis and Birth Health, Wuhan, China
- Wuhan Clinical Research Center for Reproductive Science and Birth Health, Wuhan, China
| | - Juan Liu
- Department of Obstetrics and Gynecology, Zhongnan Hospital of Wuhan University, Wuhan, China
- Hubei Clinical Research Center for Prenatal Diagnosis and Birth Health, Wuhan, China
- Wuhan Clinical Research Center for Reproductive Science and Birth Health, Wuhan, China
| | - Jin Li
- Hubei Clinical Research Center for Prenatal Diagnosis and Birth Health, Wuhan, China
- Wuhan Clinical Research Center for Reproductive Science and Birth Health, Wuhan, China
- Department of Laboratory Medicine, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Jianhong Ma
- Department of Obstetrics and Gynecology, Zhongnan Hospital of Wuhan University, Wuhan, China
- Hubei Clinical Research Center for Prenatal Diagnosis and Birth Health, Wuhan, China
- Wuhan Clinical Research Center for Reproductive Science and Birth Health, Wuhan, China
- *Correspondence: Jianhong Ma, ; Jie Duan,
| | - Jie Duan
- Department of Obstetrics and Gynecology, Zhongnan Hospital of Wuhan University, Wuhan, China
- Hubei Clinical Research Center for Prenatal Diagnosis and Birth Health, Wuhan, China
- Wuhan Clinical Research Center for Reproductive Science and Birth Health, Wuhan, China
- *Correspondence: Jianhong Ma, ; Jie Duan,
| |
Collapse
|
49
|
IL-6 and Leptin Are Potential Biomarkers for Osteoporotic Fracture Risk Assessment and Prediction of Postmenopausal Women with Low Bone Mass: A Follow-Up Study Using a Regional Sample Cohort. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:8691830. [PMID: 35993023 PMCID: PMC9385352 DOI: 10.1155/2022/8691830] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/04/2022] [Accepted: 06/24/2022] [Indexed: 11/17/2022]
Abstract
Osteoporotic fracture, a major complication which is known as the outcome postmenopausal osteoporosis, seriously threatens the health of postmenopausal women. At present, the traditional osteoporotic fracture prediction methods are characterized by inconvenient application and time-consuming statistical results, while predictive serum biomarkers can make up for this shortcoming. Accurate and advanced risk prediction of osteoporotic fracture is meaningful to early prevention and intervention, effectively avoiding the risk of this disease and the secondary fracture in the surgical treatment. In this study, based on the BEYOND cohort, a 2-year follow-up study was conducted after subjects participated to survey if OF occurred. Independent sample t-test and Mann–Whitney U-test were used to analyze the differences of bone metabolism biomarkers between the OF and non-OF group. Cox proportional hazard model was used to screen the potential biomarkers might be used to predict OF risk. ROC curves and AUCs were used to analyze the predictive accuracy, and the Delong's test was used to compare the differences between the AUCs. 15 postmenopausal women with low bone mass and OF were found, and other 60 subjects without OF were matched with 1 : 4, age, and BMI classification as control group. The serum IL-6 (OR = 1.139, 95%CI = 1.058 − 1.226) and leptin (OR = 0.921, 95%CI = 0.848 − 1.000) were found as OF risk predictive biomarkers for postmenopausal women with low bone mass with high accuracy (IL − 6 = 0.871) (leptin = 0.813) and accuracy enhanced when they were combined (AUC = 0.898). The results of Delong's test showed that the difference of AUC between leptin and IL-6&Leptin was meaningful (P = 0.024) but meaningless between IL-6 and leptin (P = 0.436), IL-6 and IL-6&Leptin (P = 0.606). To sum up, IL-6 and leptin are the predictive biomarkers of OF for postmenopausal women with low bone mass. The IL-6 can improve the prediction accuracy of leptin (P = 0.024), but not vice versa (P = 0.606). Trial Information. Registered on the Chinese Clinical Trial Registry already. (Registration Number: ChiCTR-SOC-17013090).
Collapse
|
50
|
Wang Q, Zhang Y, Zhang E, Xing X, Chen Y, Nie K, Yuan H, Su MY, Lang N. A Multiparametric Method Based on Clinical and CT-Based Radiomics to Predict the Expression of p53 and VEGF in Patients With Spinal Giant Cell Tumor of Bone. Front Oncol 2022; 12:894696. [PMID: 35800059 PMCID: PMC9253421 DOI: 10.3389/fonc.2022.894696] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2022] [Accepted: 05/19/2022] [Indexed: 11/13/2022] Open
Abstract
PurposeThis project aimed to assess the significance of vascular endothelial growth factor (VEGF) and p53 for predicting progression-free survival (PFS) in patients with spinal giant cell tumor of bone (GCTB) and to construct models for predicting these two biomarkers based on clinical and computer tomography (CT) radiomics to identify high-risk patients for improving treatment.Material and MethodsA retrospective study was performed from April 2009 to January 2019. A total of 80 patients with spinal GCTB who underwent surgery in our institution were identified. VEGF and p53 expression and clinical and general imaging information were collected. Multivariate Cox regression models were used to verify the prognostic factors. The radiomics features were extracted from the regions of interest (ROIs) in preoperative CT, and then important features were selected by the SVM to build classification models, evaluated by 10-fold crossvalidation. The clinical variables were processed using the same method to build a conventional model for comparison.ResultsThe immunohistochemistry of 80 patients was obtained: 49 with high-VEGF and 31 with low-VEGF, 68 with wild-type p53, and 12 with mutant p53. p53 and VEGF were independent prognostic factors affecting PFS found in multivariate Cox regression analysis. For VEGF, the Spinal Instability Neoplastic Score (SINS) was greater in the high than low groups, p < 0.001. For p53, SINS (p = 0.030) and Enneking stage (p = 0.017) were higher in mutant than wild-type groups. The VEGF radiomics model built using 3 features achieved an area under the curve (AUC) of 0.88, and the p53 radiomics model built using 4 features had an AUC of 0.79. The conventional model built using SINS, and the Enneking stage had a slightly lower AUC of 0.81 for VEGF and 0.72 for p53.Conclusionp53 and VEGF are associated with prognosis in patients with spinal GCTB, and the radiomics analysis based on preoperative CT provides a feasible method for the evaluation of these two biomarkers, which may aid in choosing better management strategies.
Collapse
Affiliation(s)
- Qizheng Wang
- Department of Radiology, Peking University Third Hospital, Beijing, China
| | - Yang Zhang
- Department of Radiological Sciences, University of California Irvine, Irvine, CA, United States
- Department of Radiation Oncology, Robert Wood Johnson Medical School, New Brunswick, NJ, United States
| | - Enlong Zhang
- Department of Radiology, Peking University International Hospital, Beijing, China
| | - Xiaoying Xing
- Department of Radiology, Peking University Third Hospital, Beijing, China
| | - Yongye Chen
- Department of Radiology, Peking University Third Hospital, Beijing, China
| | - Ke Nie
- Department of Radiation Oncology, Robert Wood Johnson Medical School, New Brunswick, NJ, United States
| | - Huishu Yuan
- Department of Radiology, Peking University Third Hospital, Beijing, China
| | - Min-Ying Su
- Department of Radiological Sciences, University of California Irvine, Irvine, CA, United States
- Department of Medical Imaging and Radiological Sciences, Kaohsiung Medical University, Kaohsiung, Taiwan
- *Correspondence: Ning Lang, ; Min-Ying Su,
| | - Ning Lang
- Department of Radiology, Peking University Third Hospital, Beijing, China
- *Correspondence: Ning Lang, ; Min-Ying Su,
| |
Collapse
|