1
|
Lange M, Babczyk P, Tobiasch E. Exosomes: A New Hope for Angiogenesis-Mediated Bone Regeneration. Int J Mol Sci 2024; 25:5204. [PMID: 38791243 PMCID: PMC11120942 DOI: 10.3390/ijms25105204] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Revised: 04/29/2024] [Accepted: 05/08/2024] [Indexed: 05/26/2024] Open
Abstract
Bone is a metabolically dynamic structure that is generally remodeled throughout the lifetime of an individual but often causes problems with increasing age. A key player for bone development and homeostasis, but also under pathological conditions, is the bone vasculature. This complex system of arteries, veins, and capillaries forms distinct structures where each subset of endothelial cells has important functions. Starting with the basic process of angiogenesis and bone-specific blood vessel formation, coupled with initial bone formation, the importance of different vascular structures is highlighted with respect to how these structures are maintained or changed during homeostasis, aging, and pathological conditions. After exemplifying the current knowledge on bone vasculature, this review will move on to exosomes, a novel hotspot of scientific research. Exosomes will be introduced starting from their discovery via current isolation procedures and state-of-the-art characterization to their role in bone vascular development, homeostasis, and bone regeneration and repair while summarizing the underlying signal transduction pathways. With respect to their role in these processes, especially mesenchymal stem cell-derived extracellular vesicles are of interest, which leads to a discussion on patented applications and an update on ongoing clinical trials. Taken together, this review provides an overview of bone vasculature and bone regeneration, with a major focus on how exosomes influence this intricate system, as they might be useful for therapeutic purposes in the near future.
Collapse
Affiliation(s)
- Martin Lange
- Cardiovascular Research Center and Department of Cellular and Molecular Physiology, Yale University School of Medicine, New Haven, CT 06510, USA
| | - Patrick Babczyk
- Department of Natural Sciences, University Bonn-Rhein-Sieg, D-53559 Rheinbach, Germany
| | - Edda Tobiasch
- Department of Natural Sciences, University Bonn-Rhein-Sieg, D-53559 Rheinbach, Germany
| |
Collapse
|
2
|
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
|
3
|
Reyes M, Firat D, Hanna P, Khan M, Bruce M, Shvedova M, Kobayashi T, Schipani E, Gardella TJ, Jüppner H. Substantially Delayed Maturation of Growth Plate Chondrocytes in "Humanized" PTH1R Mice with the H223R Mutation of Jansen's Disease. JBMR Plus 2023; 7:e10802. [PMID: 37808400 PMCID: PMC10556264 DOI: 10.1002/jbm4.10802] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Revised: 07/05/2023] [Accepted: 07/11/2023] [Indexed: 10/10/2023] Open
Abstract
Activating parathyroid hormone (PTH)/PTH-related Peptide (PTHrP) receptor (PTH1R) mutations causes Jansen's metaphyseal chondrodysplasia (JMC), a rare disease characterized by growth plate abnormalities, short stature, and PTH-independent hypercalcemia. Previously generated transgenic JMC mouse models, in which the human PTH1R allele with the H223R mutation (H223R-PTH1R) is expressed in osteoblasts via type Ia1 collagen or DMP1 promoters cause excess bone mass, while expression of the mutant allele via the type IIa1 collagen promoter results in only minor growth plate changes. Thus, neither transgenic JMC model adequately recapitulates the human disease. We therefore generated "humanized" JMC mice in which the H223R-PTH1R allele was expressed via the endogenous mouse Pth1r promoter and, thus, in all relevant target tissues. Founders with the H223R allele typically died within 2 months without reproducing; several mosaic male founders, however, lived longer and produced F1 H223R-PTH1R offspring, which were small and exhibited marked growth plate abnormalities. Serum calcium and phosphate levels of the mutant mice were not different from wild-type littermates, but serum PTH and P1NP were reduced significantly, while CTX-1 and CTX-2 were slightly increased. Histological and RNAscope analyses of the mutant tibial growth plates revealed markedly expanded zones of type II collagen-positive, proliferating/prehypertrophic chondrocytes, abundant apoptotic cells in the growth plate center and a progressive reduction of type X collagen-positive hypertrophic chondrocytes and primary spongiosa. The "humanized" H223R-PTH1R mice are likely to provide a more suitable model for defining the JMC phenotype and for assessing potential treatment options for this debilitating disease of skeletal development and mineral ion homeostasis. © 2023 The Authors. JBMR Plus published by Wiley Periodicals LLC on behalf of American Society for Bone and Mineral Research.
Collapse
Affiliation(s)
- Monica Reyes
- Endocrine UnitMassachusetts General Hospital and Harvard Medical SchoolBostonMAUSA
| | - Damla Firat
- Endocrine UnitMassachusetts General Hospital and Harvard Medical SchoolBostonMAUSA
| | - Patrick Hanna
- Endocrine UnitMassachusetts General Hospital and Harvard Medical SchoolBostonMAUSA
| | - Mohd Khan
- Department of Orthopedic SurgeryUniversity of Pennsylvania, Perelman Medical SchoolPhiladelphiaPAUSA
| | - Michael Bruce
- Endocrine UnitMassachusetts General Hospital and Harvard Medical SchoolBostonMAUSA
| | - Maria Shvedova
- Endocrine UnitMassachusetts General Hospital and Harvard Medical SchoolBostonMAUSA
| | - Tatsuya Kobayashi
- Endocrine UnitMassachusetts General Hospital and Harvard Medical SchoolBostonMAUSA
| | - Ernestina Schipani
- Department of Orthopedic SurgeryUniversity of Pennsylvania, Perelman Medical SchoolPhiladelphiaPAUSA
| | - Thomas J. Gardella
- Endocrine UnitMassachusetts General Hospital and Harvard Medical SchoolBostonMAUSA
| | - Harald Jüppner
- Endocrine UnitMassachusetts General Hospital and Harvard Medical SchoolBostonMAUSA
- Pediatric Nephrology UnitMassachusetts General Hospital and Harvard Medical SchoolBostonMAUSA
| |
Collapse
|
4
|
Yang Y, Wu Y, Yang D, Neo SH, Kadir ND, Goh D, Tan JX, Denslin V, Lee EH, Yang Z. Secretive derived from hypoxia preconditioned mesenchymal stem cells promote cartilage regeneration and mitigate joint inflammation via extracellular vesicles. Bioact Mater 2023; 27:98-112. [PMID: 37006826 PMCID: PMC10063382 DOI: 10.1016/j.bioactmat.2023.03.017] [Citation(s) in RCA: 19] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2022] [Revised: 02/23/2023] [Accepted: 03/22/2023] [Indexed: 03/31/2023] Open
Abstract
Secretome derived from mesenchymal stem cells (MSCs) have profound effects on tissue regeneration, which could become the basis of future MSCs therapies. Hypoxia, as the physiologic environment of MSCs, has great potential to enhance MSCs paracrine therapeutic effect. In our study, the paracrine effects of secretome derived from MSCs preconditioned in normoxia and hypoxia was compared through both in vitro functional assays and an in vivo rat osteochondral defect model. Specifically, the paracrine effect of total EVs were compared to that of soluble factors to characterize the predominant active components in the hypoxic secretome. We demonstrated that hypoxia conditioned medium, as well as the corresponding EVs, at a relatively low dosage, were efficient in promoting the repair of critical-sized osteochondral defects and mitigated the joint inflammation in a rat osteochondral defect model, relative to their normoxia counterpart. In vitro functional test shows enhancement through chondrocyte proliferation, migration, and matrix deposition, while inhibit IL-1β-induced chondrocytes senescence, inflammation, matrix degradation, and pro-inflammatory macrophage activity. Multiple functional proteins, as well as a change in EVs' size profile, with enrichment of specific EV-miRNAs were detected with hypoxia preconditioning, implicating complex molecular pathways involved in hypoxia pre-conditioned MSCs secretome generated cartilage regeneration.
Collapse
|
5
|
Huang J, Han Q, Cai M, Zhu J, Li L, Yu L, Wang Z, Fan G, Zhu Y, Lu J, Zhou G. Effect of Angiogenesis in Bone Tissue Engineering. Ann Biomed Eng 2022; 50:898-913. [PMID: 35525871 DOI: 10.1007/s10439-022-02970-9] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2021] [Accepted: 04/17/2022] [Indexed: 12/20/2022]
Abstract
The reconstruction of large skeletal defects is still a tricky challenge in orthopedics. The newly formed bone tissue migrates sluggishly from the periphery to the center of the scaffold due to the restrictions of exchange of oxygen and nutrition impotent cells osteogenic differentiation. Angiogenesis plays an important role in bone reconstruction and more and more studies on angiogenesis in bone tissue engineering had been published. Promising advances of angiogenesis in bone tissue engineering by scaffold designs, angiogenic factor delivery, in vivo prevascularization and in vitro prevascularization are discussed in detail. Among all the angiogenesis mode, angiogenic factor delivery is the common methods of angiogenesis in bone tissue engineering and possible research directions in the future.
Collapse
Affiliation(s)
- Jianhao Huang
- Department of Orthopedics, Jinling Hospital, The First School of Clinical Medicine, Southern Medical University, Nanjing, 210002, People's Republic of China
| | - Qixiu Han
- Department of Orthopedics, Jinling Hospital, Nanjing Medical University, Nanjing, 210002, People's Republic of China
| | - Meng Cai
- Department of Orthopedics, Jinling Hospital, School of Medicine, Southeast University, Nanjing, 210002, People's Republic of China
| | - Jie Zhu
- Department of Orthopedics, Jinling Hospital, Nanjing Medical University, Nanjing, 210002, People's Republic of China
| | - Lan Li
- State Key Laboratory of Pharmaceutical Biotechnology, Department of Sports Medicine and Adult Reconstructive Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, 210008, People's Republic of China
| | - Lingfeng Yu
- Department of Orthopedics, Jinling Hospital, School of Medicine, Nanjing University, Nanjing, 210002, People's Republic of China
| | - Zhen Wang
- Department of Orthopedics, Jinling Hospital, Nanjing Medical University, Nanjing, 210002, People's Republic of China
| | - Gentao Fan
- Department of Orthopedics, Nanjing Jinling Hospital, 305 Zhongshan East Road, Nanjing, 210002, People's Republic of China
| | - Yan Zhu
- Department of Orthopedics, Nanjing Jinling Hospital, 305 Zhongshan East Road, Nanjing, 210002, People's Republic of China
| | - Jingwei Lu
- Department of Orthopedics, Jinling Hospital, Nanjing Medical University, Nanjing, 210002, People's Republic of China. .,Department of Orthopedics, Nanjing Jinling Hospital, 305 Zhongshan East Road, Nanjing, 210002, People's Republic of China.
| | - Guangxin Zhou
- Department of Orthopedics, Jinling Hospital, The First School of Clinical Medicine, Southern Medical University, Nanjing, 210002, People's Republic of China. .,Department of Orthopedics, Jinling Hospital, School of Medicine, Nanjing University, Nanjing, 210002, People's Republic of China. .,Department of Orthopedics, Nanjing Jinling Hospital, 305 Zhongshan East Road, Nanjing, 210002, People's Republic of China. .,The First School of Clinical Medicine, Southern Medical University, Guangzhou, 510515, People's Republic of China.
| |
Collapse
|
6
|
Jiang Z, Johnson CP, Nykänen O, Nissi M, Lau YK, Wu M, Casal ML, Smith LJ. Epiphyseal cartilage canal architecture and extracellular matrix remodeling in mucopolysaccharidosis VII dogs at the onset of postnatal growth. Connect Tissue Res 2021; 62:698-708. [PMID: 33334202 PMCID: PMC8272733 DOI: 10.1080/03008207.2020.1865939] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Purpose: Mucopolysaccharidosis (MPS) VII is a genetic, lysosomal storage disease characterized by abnormal accumulation of glycosaminoglycans in cells and tissues. MPS VII patients exhibit multiple failures of endochondral ossification during postnatal growth, including markedly delayed cartilage-to-bone conversion in the vertebrae and long bones. Cartilage canals provide the template for vascularization at the onset of secondary ossification. The objective of this study was to investigate whether abnormal cartilage canal architecture and enzyme-mediated extracellular matrix (ECM) remodeling contribute to delayed cartilage-to-bone conversion in MPS VII.Materials and Methods: The epiphyseal cartilage canal networks of 9-day-old healthy control and MPS VII-affected dog vertebrae were characterized using high-resolution, contrast-free quantitative susceptibility mapping magnetic resonance imaging. Relative expression levels of matrix metalloproteinases (MMPs) 9, 13 and 14 were examined using immunohistochemistry, while tartrate-resistant acid phosphatase (TRAP) and alkaline phosphatase (ALP) were examined using in situ enzyme staining.Results: Interestingly, the density, number, connectivity and thickness of cartilage canals was not significantly different between MPS VII and control vertebrae. Immunohistochemistry revealed diminished MMP-9, but normal MMP-13 and 14 expression by epiphyseal cartilage chondrocytes, while ALP and TRAP enzyme expression by chondrocytes and chondroclasts, respectively, were both diminished in MPS VII.Conclusions: Our findings suggest that while the epiphyseal cartilage canal network in MPS VII is normal at the onset of secondary ossification, expression of enzymes required for cartilage resorption and replacement with mineralized ECM, and initiation of angiogenesis, is impaired.
Collapse
Affiliation(s)
- Zhirui Jiang
- Department of Neurosurgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA,Deparment of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Casey P. Johnson
- Department of Veterinary Clinical Sciences, University of Minnesota, Saint Paul, MN, USA,Center for Magnetic Resonance Research, University of Minnesota, Minneapolis, MN, USA
| | - Olli Nykänen
- Department of Applied Physics, University of Eastern Finland, Kuopio, Finland
| | - Mikko Nissi
- Department of Applied Physics, University of Eastern Finland, Kuopio, Finland,Department of Diagnostic Radiology, University of Oulu, Oulu, Finland
| | - Yian Khai Lau
- Department of Neurosurgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA,Deparment of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Meilun Wu
- Department of Neurosurgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA,Deparment of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Margret L. Casal
- Department of Clinical Sciences and Advanced Medicine, School of Veterinary Medicine, University of Pennsylvania, Philadelphia PA, USA
| | - Lachlan J. Smith
- Department of Neurosurgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA,Deparment of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA,Correspondence: Lachlan J. Smith, Ph.D., Associate Professor, Department of Neurosurgery, University of Pennsylvania, 371 Stemmler Hall, 3450 Hamilton Walk, Philadelphia, PA, 19104 USA, , Phone: +1 215 746 2169, Fax: +1 215 573 2133
| |
Collapse
|
7
|
The endothelium-bone axis in development, homeostasis and bone and joint disease. Nat Rev Rheumatol 2021; 17:608-620. [PMID: 34480164 DOI: 10.1038/s41584-021-00682-3] [Citation(s) in RCA: 86] [Impact Index Per Article: 28.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/30/2021] [Indexed: 01/20/2023]
Abstract
Blood vessels form a versatile transport network that is best known for its critical roles in processes such as tissue oxygenation, metabolism and immune surveillance. The vasculature also provides local, often organ-specific, molecular signals that control the behaviour of other cell types in their vicinity during development, homeostasis and regeneration, and also in disease processes. In the skeletal system, the local vasculature is actively involved in both bone formation and resorption. In addition, blood vessels participate in inflammatory processes and contribute to the pathogenesis of diseases that affect the joints, such as rheumatoid arthritis and osteoarthritis. This Review summarizes the current understanding of the architecture, angiogenic growth and functional properties of the bone vasculature. The effects of ageing and pathological conditions, including arthritis and osteoporosis, are also discussed.
Collapse
|
8
|
Apelgren P, Amoroso M, Säljö K, Montelius M, Lindahl A, Stridh Orrhult L, Gatenholm P, Kölby L. Vascularization of tissue engineered cartilage - Sequential in vivo MRI display functional blood circulation. Biomaterials 2021; 276:121002. [PMID: 34274777 DOI: 10.1016/j.biomaterials.2021.121002] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 06/21/2021] [Accepted: 06/28/2021] [Indexed: 12/24/2022]
Abstract
Establishing functional circulation in bioengineered tissue after implantation is vital for the delivery of oxygen and nutrients to the cells. Native cartilage is avascular and thrives on diffusion, which in turn depends on proximity to circulation. Here, we investigate whether a gridded three-dimensional (3D) bioprinted construct would allow ingrowth of blood vessels and thus prove a functional concept for vascularization of bioengineered tissue. Twenty 10 × 10 × 3-mm 3Dbioprinted nanocellulose constructs containing human nasal chondrocytes or cell-free controls were subcutaneously implanted in 20 nude mice. Over the next 3 months, the mice were sequentially imaged with a 7 T small-animal MRI system, and the diffusion and perfusion parameters were analyzed. The chondrocytes survived and proliferated, and the shape of the constructs was well preserved. The diffusion coefficient was high and well preserved over time. The perfusion and diffusion patterns shown by MRI suggested that blood vessels develop over time in the 3D bioprinted constructs; the vessels were confirmed by histology and immunohistochemistry. We conclude that 3D bioprinted tissue with a gridded structure allows ingrowth of blood vessels and has the potential to be vascularized from the host. This is an essential step to take bioengineered tissue from the bench to clinical practice.
Collapse
Affiliation(s)
- Peter Apelgren
- Department of Plastic Surgery, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden; Region Västra Götaland, Sahlgrenska University Hospital, Department of Plastic Surgery, Gothenburg, Sweden.
| | - Matteo Amoroso
- Department of Plastic Surgery, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden; Region Västra Götaland, Sahlgrenska University Hospital, Department of Plastic Surgery, Gothenburg, Sweden
| | - Karin Säljö
- Department of Plastic Surgery, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden; Region Västra Götaland, Sahlgrenska University Hospital, Department of Plastic Surgery, Gothenburg, Sweden
| | - Mikael Montelius
- Department of Radiation Physics, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Anders Lindahl
- Department of Clinical Chemistry and Transfusion Medicine, Institute of Biomedicine, Sahlgrenska Academy, Gothenburg, Sweden
| | - Linnea Stridh Orrhult
- 3D Bioprinting Centre, Department of Chemistry and Chemical Engineering, Chalmers University of Technology, Gothenburg, Sweden
| | - Paul Gatenholm
- 3D Bioprinting Centre, Department of Chemistry and Chemical Engineering, Chalmers University of Technology, Gothenburg, Sweden
| | - Lars Kölby
- Department of Plastic Surgery, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden; Region Västra Götaland, Sahlgrenska University Hospital, Department of Plastic Surgery, Gothenburg, Sweden
| |
Collapse
|
9
|
Caron MMJ, Castermans TMR, van Rietbergen B, Haartmans MJJ, van Rhijn LW, Witlox AMA, Welting TJM. Impairment of Cyclo-oxygenase-2 Function Results in Abnormal Growth Plate Development and Bone Microarchitecture but Does Not Affect Longitudinal Growth of the Long Bones in Skeletally Immature Mice. Cartilage 2021; 12:387-398. [PMID: 30880429 PMCID: PMC8236650 DOI: 10.1177/1947603519833149] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
OBJECTIVE Despite the general awareness that cyclo-oxygenase-2 (COX-2) is crucial for endochondral ossification, the role of COX-2 in skeletal development is largely unknown. We hypothesized that inhibition or genetic loss of COX-2 leads to impaired growth plate development and consequently impaired postnatal development of the long bones. DESIGN Skeletally immature (5 weeks old) B6;129S-Ptgs2tm1Jed/J wildtype mice were treated for 10 weeks with celecoxib (daily oral administration 10 mg/kg) or placebo and compared with B6;129S-Ptgs2tm1Jed/J homozygous knockout mice (n = 12 per group). RESULTS Fifteen weeks postnatally, no significant difference in growth plate (zone) thickness was found between groups. However, significantly higher proteoglycan content and lower expression levels of collagen type II and X staining in the growth plates of celecoxib-treated mice, and to a lesser extent in COX-2 knockout mice. In addition, a significantly decreased cell number and cell size were observed in the hypertrophic zone of the growth plates of both experimental groups. Micro-computed tomography analysis of the subchondral bone region directly beneath the growth plate showed significantly higher bone density and trabecular thickness, following celecoxib treatment. Despite the detected differences in growth plate extracellular matrix composition and subchondral bone morphology, no difference was found in the length of the tibia in celecoxib-treated mice or COX-2 knockout mice. CONCLUSIONS Genetic loss of COX-2 or treatment with celecoxib did not result in detectable differences in gross murine formation of the tibia or femur. However, there were notable phenotypic features detected in the maturation of the growth plate (hypertrophic zone and subchondral bone) as a result of the celecoxib treatment.
Collapse
Affiliation(s)
- Marjolein M. J. Caron
- Department of Orthopedic Surgery, CAPHRI Care and Public Health Research Institute, Maastricht University Medical Center, Maastricht, the Netherlands,Marjolein M. J. Caron, Department of Orthopedic Surgery, CAPHRI Care and Public Health Research Institute, Maastricht University Medical Center. P.O. Box 5800, 6202 AZ, Maastricht, the Netherlands.
| | - Tessy M. R. Castermans
- Department of Orthopedic Surgery, CAPHRI Care and Public Health Research Institute, Maastricht University Medical Center, Maastricht, the Netherlands
| | - Bert van Rietbergen
- Department of Orthopedic Surgery, CAPHRI Care and Public Health Research Institute, Maastricht University Medical Center, Maastricht, the Netherlands,Orthopaedic Biomechanics, Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, the Netherlands
| | - Mirella J. J. Haartmans
- Department of Orthopedic Surgery, CAPHRI Care and Public Health Research Institute, Maastricht University Medical Center, Maastricht, the Netherlands
| | - Lodewijk W. van Rhijn
- Department of Orthopedic Surgery, CAPHRI Care and Public Health Research Institute, Maastricht University Medical Center, Maastricht, the Netherlands
| | - Adhiambo M. A. Witlox
- Department of Orthopedic Surgery, CAPHRI Care and Public Health Research Institute, Maastricht University Medical Center, Maastricht, the Netherlands
| | - Tim J. M. Welting
- Department of Orthopedic Surgery, CAPHRI Care and Public Health Research Institute, Maastricht University Medical Center, Maastricht, the Netherlands
| |
Collapse
|
10
|
Chen M, Li Y, Huang X, Gu Y, Li S, Yin P, Zhang L, Tang P. Skeleton-vasculature chain reaction: a novel insight into the mystery of homeostasis. Bone Res 2021; 9:21. [PMID: 33753717 PMCID: PMC7985324 DOI: 10.1038/s41413-021-00138-0] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Revised: 11/18/2020] [Accepted: 12/16/2020] [Indexed: 02/01/2023] Open
Abstract
Angiogenesis and osteogenesis are coupled. However, the cellular and molecular regulation of these processes remains to be further investigated. Both tissues have recently been recognized as endocrine organs, which has stimulated research interest in the screening and functional identification of novel paracrine factors from both tissues. This review aims to elaborate on the novelty and significance of endocrine regulatory loops between bone and the vasculature. In addition, research progress related to the bone vasculature, vessel-related skeletal diseases, pathological conditions, and angiogenesis-targeted therapeutic strategies are also summarized. With respect to future perspectives, new techniques such as single-cell sequencing, which can be used to show the cellular diversity and plasticity of both tissues, are facilitating progress in this field. Moreover, extracellular vesicle-mediated nuclear acid communication deserves further investigation. In conclusion, a deeper understanding of the cellular and molecular regulation of angiogenesis and osteogenesis coupling may offer an opportunity to identify new therapeutic targets.
Collapse
Affiliation(s)
- Ming Chen
- Department of Orthopedics, Chinese PLA General Hospital, Beijing, China
- National Clinical Research Center for Orthopedics, Sports Medicine & Rehabilitation, Beijing, China
| | - Yi Li
- Department of Orthopedics, Chinese PLA General Hospital, Beijing, China
- National Clinical Research Center for Orthopedics, Sports Medicine & Rehabilitation, Beijing, China
| | - Xiang Huang
- Department of Orthopedics, Chinese PLA General Hospital, Beijing, China
- National Clinical Research Center for Orthopedics, Sports Medicine & Rehabilitation, Beijing, China
| | - Ya Gu
- Department of Orthopedics, Chinese PLA General Hospital, Beijing, China
- National Clinical Research Center for Orthopedics, Sports Medicine & Rehabilitation, Beijing, China
| | - Shang Li
- Department of Orthopedics, Chinese PLA General Hospital, Beijing, China
- National Clinical Research Center for Orthopedics, Sports Medicine & Rehabilitation, Beijing, China
| | - Pengbin Yin
- Department of Orthopedics, Chinese PLA General Hospital, Beijing, China.
- National Clinical Research Center for Orthopedics, Sports Medicine & Rehabilitation, Beijing, China.
| | - Licheng Zhang
- Department of Orthopedics, Chinese PLA General Hospital, Beijing, China.
- National Clinical Research Center for Orthopedics, Sports Medicine & Rehabilitation, Beijing, China.
| | - Peifu Tang
- Department of Orthopedics, Chinese PLA General Hospital, Beijing, China.
- National Clinical Research Center for Orthopedics, Sports Medicine & Rehabilitation, Beijing, China.
| |
Collapse
|
11
|
Blumer MJF. Bone tissue and histological and molecular events during development of the long bones. Ann Anat 2021; 235:151704. [PMID: 33600952 DOI: 10.1016/j.aanat.2021.151704] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2020] [Revised: 01/07/2021] [Accepted: 01/27/2021] [Indexed: 12/20/2022]
Abstract
The bones are of mesenchymal or ectomesenchymal origin, form the skeleton of most vertebrates, and are essential for locomotion and organ protection. As a living tissue they are highly vascularized and remodelled throughout life to maintain intact. Bones consist of osteocytes entrapped in a mineralized extracellular matrix, and via their elaborated network of cytoplasmic processes they do not only communicate with each other but also with the cells on the bone surface (bone lining cells). Bone tissue develops through a series of fine-tuned processes, and there are two modes of bone formation, referred to either as intramembranous or endochondral ossification. In intramembranous ossification, bones develop directly from condensations of mesenchymal cells, and the flat bones of the skull, the clavicles and the perichondral bone cuff develop via this process. The bones of the axial (ribs and vertebrae) and the appendicular skeleton (e.g. upper and lower limbs) form through endochondral ossification where mesenchyme turns into a cartilaginous intermediate with the shape of the future skeletal element that is gradually replaced by bone. Endochondral ossification occurs in all vertebrate taxa and its onset involves differentiation of the chondrocytes, mineralization of the extracellular cartilage matrix and vascularization of the intermediate, followed by disintegration and resorption of the cartilage, bone formation, and finally - after complete ossification of the cartilage model - the establishment of an avascular articular cartilage. The epiphyseal growth plate regulates the longitudinal growth of the bones, achieved by a balanced proliferation and elimination of chondrocytes, and the question whether the late hypertrophic chondrocytes die or transform into osteogenic cells is still being hotly debated. The complex processes leading to endochondral ossification have been studied for over a century, and this review aims to give an overview of the histological and molecular events, arising from the long bones' (e.g. femur, tibia) development. The fate of the hypertrophic chondrocytes will be discussed in the light of new findings obtained from cell tracking studies.
Collapse
Affiliation(s)
- Michael J F Blumer
- Department of Anatomy, Histology and Embryology, Institute of Clinical and Functional Anatomy, Medical University Innsbruck, Müllerstrasse 59, A-6010 Innsbruck, Austria.
| |
Collapse
|
12
|
van Gastel N, Carmeliet G. Metabolic regulation of skeletal cell fate and function in physiology and disease. Nat Metab 2021; 3:11-20. [PMID: 33398192 DOI: 10.1038/s42255-020-00321-3] [Citation(s) in RCA: 54] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/14/2020] [Accepted: 11/10/2020] [Indexed: 01/28/2023]
Abstract
The skeleton is diverse in its functions, which include mechanical support, movement, blood cell production, mineral storage and endocrine regulation. This multifaceted role is achieved through an interplay of osteoblasts, chondrocytes, bone marrow adipocytes and stromal cells, all generated from skeletal stem cells. Emerging evidence shows the importance of cellular metabolism in the molecular control of the skeletal system. The different skeletal cell types not only have distinct metabolic demands relating to their particular functions but also are affected by microenvironmental constraints. Specific metabolites control skeletal stem cell maintenance, direct lineage allocation and mediate cellular communication. Here, we discuss recent findings on the roles of cellular metabolism in determining skeletal stem cell fate, coordinating osteoblast and chondrocyte function, and organizing stromal support of haematopoiesis. We also consider metabolic dysregulation in skeletal ageing and degenerative diseases, and provide an outlook on how the field may evolve in the coming years.
Collapse
Affiliation(s)
- Nick van Gastel
- de Duve Institute, Brussels, Belgium.
- Department of Stem Cell and Regenerative Biology, Harvard Stem Cell Institute, Harvard University, Cambridge, MA, USA.
| | - Geert Carmeliet
- Laboratory of Clinical and Experimental Endocrinology, Department of Chronic Diseases and Metabolism, KU Leuven, Leuven, Belgium.
| |
Collapse
|
13
|
Liebig BE, Kisiday JD, Bahney CS, Ehrhart NP, Goodrich LR. The platelet-rich plasma and mesenchymal stem cell milieu: A review of therapeutic effects on bone healing. J Orthop Res 2020; 38:2539-2550. [PMID: 32589800 PMCID: PMC8354210 DOI: 10.1002/jor.24786] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/08/2020] [Revised: 06/15/2020] [Accepted: 06/24/2020] [Indexed: 02/04/2023]
Abstract
Platelet-rich plasma is autologous plasma that contains concentrated platelets compared to whole blood. It is relatively inexpensive to produce, can be easily isolated from whole blood, and can be administered while the patient is in the operating room. Further, because platelet-rich plasma is an autologous therapy, there is minimal risk for adverse reactions to the patient. Platelet-rich plasma has been used to promote bone regeneration due to its abundance of concentrated growth factors that are essential to wound healing. In this review, we summarize the methods for producing platelet-rich plasma and the history of its use in bone regeneration. We also summarize the growth factor profiles derived from platelet-rich plasma, with emphasis on those factors that play a direct role in promoting bone repair within the local fracture environment. In addition, we discuss the potential advantages of combining platelet-rich plasma with mesenchymal stem cells, a multipotent cell type often obtained from bone marrow or fat, to improve craniofacial and long bone regeneration. We detail what is currently known about how platelet-rich plasma influences mesenchymal stem cells in vitro, and then highlight the clinical outcomes of administering platelet-rich plasma and mesenchymal stem cells as a combination therapy to promote bone regeneration in vivo.
Collapse
Affiliation(s)
- Bethany E. Liebig
- Department of Clinical Sciences, Orthopaedic Research Center, Translational Medicine Institute, College of Veterinary Medicine, Colorado State University, Fort Collins, Colorado
| | - John D. Kisiday
- Department of Clinical Sciences, Orthopaedic Research Center, Translational Medicine Institute, College of Veterinary Medicine, Colorado State University, Fort Collins, Colorado
| | - Chelsea S. Bahney
- Center for Regenerative Sports Medicine, Steadman Philippon Research Institute, Vail, Colorado
| | - Nicole P. Ehrhart
- Department of Clinical Sciences, Flint Animal Cancer Center, College of Veterinary Medicine, Colorado State University, Fort Collins, Colorado
| | - Laurie R. Goodrich
- Department of Clinical Sciences, Orthopaedic Research Center, Translational Medicine Institute, College of Veterinary Medicine, Colorado State University, Fort Collins, Colorado
| |
Collapse
|
14
|
Time Dependency of Non-Thermal Oxygen Plasma and Ultraviolet Irradiation on Cellular Attachment and mRNA Expression of Growth Factors in Osteoblasts on Titanium and Zirconia Surfaces. Int J Mol Sci 2020; 21:ijms21228598. [PMID: 33202662 PMCID: PMC7697706 DOI: 10.3390/ijms21228598] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2020] [Revised: 11/08/2020] [Accepted: 11/11/2020] [Indexed: 12/21/2022] Open
Abstract
Ultraviolet (UV) light and non-thermal plasma (NTP) are promising chair-side surface treatment methods to overcome the time-dependent aging of dental implant surfaces. After showing the efficiency of UV light and NTP treatment in restoring the biological activity of titanium and zirconia surfaces in vitro, the objective of this study was to define appropriate processing times for clinical use. Titanium and zirconia disks were treated by UV light and non-thermal oxygen plasma with increasing duration. Non-treated disks were set as controls. Murine osteoblast-like cells (MC3T3-E1) were seeded onto the treated or non-treated disks. After 2 and 24 h of incubation, the viability of cells on surfaces was assessed using an MTS assay. mRNA expression of vascular endothelial growth factor (VEGF) and hepatocyte growth factor (HGF) were assessed using real-time reverse transcription polymerase chain reaction analysis. Cellular morphology and attachment were observed using confocal microscopy. The viability of MC3T3-E1 was significantly increased in 12 min UV-light treated and 1 min oxygen NTP treated groups. VEGF relative expression reached the highest levels on 12 min UV-light and 1 min NTP treated surfaces of both disks. The highest levels of HGF relative expression were reached on 12 min UV light treated zirconia surfaces. However, cells on 12 and 16 min UV-light and NTP treated surfaces of both materials had a more widely spread cytoskeleton compared to control groups. Twelve min UV-light and one min non-thermal oxygen plasma treatment on titanium and zirconia may be the favored times in terms of increasing the viability, mRNA expression of growth factors and cellular attachment in MC3T3-E1 cells.
Collapse
|
15
|
Pleiotropic actions of Vitamin D in composite musculoskeletal trauma. Injury 2020; 51:2099-2109. [PMID: 32624209 DOI: 10.1016/j.injury.2020.06.023] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/06/2020] [Revised: 05/21/2020] [Accepted: 06/13/2020] [Indexed: 02/02/2023]
Abstract
Composite tissue injuries are the result of high energy impacts caused by motor vehicle accidents, gunshot wounds or blasts. These are highly traumatic injuries characterized by wide-spread, penetrating wounds affecting the entire musculoskeletal system, and are generally defined by frank volumetric muscle loss with concomitant segmental bone defects. At the tissue level, the breadth of damage to multiple tissue systems, and potential for infection from penetration, have been shown to lead to an exaggerated, often chronic inflammatory response with subsequent dysregulation of normal musculoskeletal healing mechanisms. Aside from the direct effects of inflammation on myogenesis and osteogenesis, frank muscle loss has been shown to directly impair fracture union and ultimately contribute to failed wound regeneration. Care for these injuries requires extensive surgical intervention and acute care strategies. However, often these interventions do not adequately mitigate inflammation or promote proper musculoskeletal injury repair and force amputation of the limb. Therefore, identification of factors that can promote tissue regeneration and mitigate inflammation could be key to restoring wound healing after composite tissue injury. One such factor that may directly affect both inflammation and tissue regeneration in response to these multi-tissue injuries may be Vitamin D. Beyond traditional roles, the pleiotropic and localized actions of Vitamin D are increasingly being recognized in most aspects of wound healing in complex tissue injuries - e.g., regulation of inflammation, myogenesis, fracture callus mineralization and remodeling. Conversely, pre-existing Vitamin D deficiency leads to musculoskeletal dysfunction, increased fracture risk or fracture non-unions, decreased strength/function and reduced capacity to heal wounds through increased inflammation. This Vitamin D deficient state requires acute supplementation in order to quickly restore circulating levels to an optimal level, thereby facilitating a robust wound healing response. Herein, the purpose of this review is to address the roles and critical functions of Vitamin D throughout the wound healing process. Findings from this review suggest that careful monitoring and/or supplementation of Vitamin D may be critical for wound regeneration in composite tissue injuries.
Collapse
|
16
|
Collagen IX deficiency leads to premature vascularization and ossification of murine femoral heads through an imbalance of pro- and antiangiogenic factors. Osteoarthritis Cartilage 2020; 28:988-999. [PMID: 32283184 DOI: 10.1016/j.joca.2020.03.015] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/12/2019] [Revised: 02/20/2020] [Accepted: 03/30/2020] [Indexed: 02/02/2023]
Abstract
OBJECTIVE The vascular invasion of cartilage is an essential process in the endochondral ossification of long bones. In contrast, vascularization of articular cartilage constitutes a pathological mechanism in the development of osteoarthritis. Polymorphisms of Col9a1 have been described as risk factors for hip osteoarthritis (OA) and the loss of collagen IX is known to lead to premature OA of the hip joint in mice but the underlying mechanism is so far unknown. DESIGN To understand the contribution of collagen IX to OA development in the hip joint, we analyzed the early development of murine Col9a1-/- femoral heads between newborn stage and 16 weeks of age. RESULTS We found significantly accelerated ossification of the femoral heads in the absence of collagen IX as well as premature vascular and osteoclast invasion, even though hypertrophic differentiation was delayed. The loss of collagen IX led to anatomically altered femoral heads lacking the epiphyseal tubercle. Interestingly, this region was found to contain highest levels of the antiangiogenic protein thrombospondin-1 (TSP-1). Hence, TSP-1 levels were strongly reduced in the Col9a1-/- femoral heads. In addition, antiangiogenic matrilin-1 was found to be decreased, while proangiogenic active MMP-9 levels were increased in the collagen IX deficient mice compared to wildtype controls. CONCLUSION We conclude that collagen IX protects against premature vascularization and cartilage to bone transition in femoral heads by increasing the levels of antiangiogenic TSP-1 and matrilin-1 and decreasing the levels of proangiogenic active MMP-9.
Collapse
|
17
|
Pal S, Rashid M, Singh SK, Porwal K, Singh P, Mohamed R, Gayen JR, Wahajuddin M, Chattopadhyay N. Skeletal restoration by phosphodiesterase 5 inhibitors in osteopenic mice: Evidence of osteoanabolic and osteoangiogenic effects of the drugs. Bone 2020; 135:115305. [PMID: 32126313 DOI: 10.1016/j.bone.2020.115305] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/23/2019] [Revised: 02/12/2020] [Accepted: 02/28/2020] [Indexed: 12/16/2022]
Abstract
Phosphodiesterases (PDEs) hydrolyze cyclic nucleotides and thereby regulate diverse cellular functions. The reports on the skeletal effects of PDE inhibitors are conflicting. Here, we screened 17 clinically used non-xanthine PDE inhibitors (selective and non-selective) using mouse calvarial osteoblasts (MCO) where the readout was osteoblast differentiation. From this screen, we identified sildenafil and vardenafil (both PDE5 inhibitors) having the least osteogenic EC50. Both drugs significantly increased vascular endothelial growth factor (VEGF) and VEGF receptor 2 (VEGFR2) expressions in MCO and the nitric oxide synthase inhibitor L-NAME completely blocked VEGF expression induced by these drugs. Sunitinib, a tyrosine receptor kinase inhibitor that also blocks VEGFR2 blocked sildenafil-/vardenafil-induced osteoblast differentiation. At half of their human equivalent doses, i.e. 6.0 mg/kg sildenafil and 2.5 mg/kg vardenafil, the maximum bone marrow level of sildenafil was 32% and vardenafil was 21% of their blood levels. At these doses, both drugs enhanced bone regeneration at the femur osteotomy site and completely restored bone mass, microarchitecture, and strength in OVX mice. Furthermore, both drugs increased surface referent bone formation and serum bone formation marker (P1NP) without affecting the resorption marker (CTX-1). Both drugs increased the expression of VEGF and VEGFR2 in bones and osteoblasts and increased skeletal vascularity. Sunitinib completely blocked the bone restorative and vascular effects of sildenafil and vardenafil in OVX mice. Taken together, our study suggested that sildenafil and vardenafil at half of their adult human doses completely reversed osteopenia in OVX mice by an osteogenic mechanism that was associated with enhanced skeletal vascularity.
Collapse
Affiliation(s)
- Subhashis Pal
- Division of Endocrinology, Central Drug Research Institute (CDRI), Council of Scientific and Industrial Research (CSIR), Lucknow 226031, India
| | - Mamunur Rashid
- Pharmaceutics & Pharmacokinetics Division, CDRI-CSIR, Lucknow 226031, India
| | | | - Konica Porwal
- Division of Endocrinology, Central Drug Research Institute (CDRI), Council of Scientific and Industrial Research (CSIR), Lucknow 226031, India
| | - Priya Singh
- Division of Endocrinology, Central Drug Research Institute (CDRI), Council of Scientific and Industrial Research (CSIR), Lucknow 226031, India
| | - Riyazuddin Mohamed
- Pharmaceutics & Pharmacokinetics Division, CDRI-CSIR, Lucknow 226031, India
| | - Jiaur R Gayen
- Pharmaceutics & Pharmacokinetics Division, CDRI-CSIR, Lucknow 226031, India
| | | | - Naibedya Chattopadhyay
- Division of Endocrinology, Central Drug Research Institute (CDRI), Council of Scientific and Industrial Research (CSIR), Lucknow 226031, India.
| |
Collapse
|
18
|
Abstract
PURPOSE OF REVIEW Bone elongation is a complex process driven by multiple intrinsic (hormones, growth factors) and extrinsic (nutrition, environment) variables. Bones grow in length by endochondral ossification in cartilaginous growth plates at ends of developing long bones. This review provides an updated overview of the important factors that influence this process. RECENT FINDINGS Insulin-like growth factor-1 (IGF-1) is the major hormone required for growth and a drug for treating pediatric skeletal disorders. Temperature is an underrecognized environmental variable that also impacts linear growth. This paper reviews the current state of knowledge regarding the interaction of IGF-1 and environmental factors on bone elongation. Understanding how internal and external variables regulate bone lengthening is essential for developing and improving treatments for an array of bone elongation disorders. Future studies may benefit from understanding how these unique relationships could offer realistic new approaches for increasing bone length in different growth-limiting conditions.
Collapse
Affiliation(s)
- Holly L Racine
- Department of Natural Sciences and Mathematics, West Liberty University, West Liberty, WV, 26074, USA
| | - Maria A Serrat
- Department of Biomedical Sciences, Joan C. Edwards School of Medicine, Marshall University, 1 John Marshall Drive, Huntington, WV, 25755, USA.
- Department of Clinical and Translational Sciences, Joan C. Edwards School of Medicine, Marshall University, Huntington, WV, 25755, USA.
- Department of Orthopaedics, Joan C. Edwards School of Medicine, Marshall University, Huntington, WV, 25755, USA.
| |
Collapse
|
19
|
Toosi S, Behravan J. Osteogenesis and bone remodeling: A focus on growth factors and bioactive peptides. Biofactors 2020; 46:326-340. [PMID: 31854489 DOI: 10.1002/biof.1598] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/18/2019] [Accepted: 11/30/2019] [Indexed: 12/14/2022]
Abstract
Bone is one of the most frequently transplanted tissues. The bone structure and its physiological function and stem cells biology were known to be closely related to each other for many years. Bone is considered a home to the well-known systems of postnatal mesenchymal stem cells (MSCs). These bone resident MSCs provide a range of growth factors (GF) and cytokines to support cell growth following injury. These GFs include a group of proteins and peptides produced by different cells which are regulators of important cell functions such as division, migration, and differentiation. GF signaling controls the formation and development of the MSCs condensation and plays a critical role in regulating osteogenesis, chondrogenesis, and bone/mineral homeostasis. Thus, a combination of both MSCs and GFs receives high expectations in regenerative medicine, particularly in bone repair applications. It is known that the delivery of exogenous GFs to the non-union bone fracture site remarkably improves healing results. Here we present updated information on bone tissue engineering with a specific focus on GF characteristics and their application in cellular functions and tissue healing. Moreover, the interrelation of GFs with the damaged bone microenvironment and their mechanistic functions are discussed.
Collapse
Affiliation(s)
- Shirin Toosi
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical, Mashhad, Iran
- Food and Drug Administration, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Javad Behravan
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical, Mashhad, Iran
- School of Pharmacy, University of Waterloo, Waterloo, Ontario, Canada
| |
Collapse
|
20
|
Hypoxia, hypoxia-inducible transcription factors and oxygen-sensing prolyl hydroxylases in bone development and homeostasis. Curr Opin Nephrol Hypertens 2020; 28:328-335. [PMID: 30985337 DOI: 10.1097/mnh.0000000000000508] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
PURPOSE OF REVIEW To summarize the role of hypoxia signaling in skeletal cells. RECENT FINDINGS Hypoxia occurs at several stages during bone development. Skeletal cells, like chondrocytes and osteoblasts, respond to this challenge by stabilizing the hypoxia inducible transcription factor HIF, which induces the expression of angiogenic factors and promotes glycolysis. The increased delivery of oxygen and nutrients, together with metabolic adaptations, prevent chondrocyte cell death in the growth plate and promote bone formation by osteoblasts. However, excessive HIF levels have to be avoided during bone development as the resulting metabolic maladaptations cause skeletal dysplasia. Recent studies show that HIF also targets other genes to increase bone mass: it decreases osteoclastogenesis by increasing osteoprotegerin expression and represses sclerostin expression by epigenetic mechanisms, resulting in increased bone formation and decreased resorption. Moreover, increased HIF signaling in osteolineage cells promotes primary and metastatic breast tumor growth, and induces erythropoietin (EPO) production, resulting in polycythemia. Finally, HIF can directly or indirectly through increasing EPO levels, induce the expression and processing of FGF23 and may thereby affect mineral homeostasis and vitamin D metabolism. SUMMARY HIF signaling in skeletal cells not only affects their behavior but also influences erythropoiesis and possibly mineral homeostasis.
Collapse
|
21
|
Lipid availability determines fate of skeletal progenitor cells via SOX9. Nature 2020; 579:111-117. [PMID: 32103177 PMCID: PMC7060079 DOI: 10.1038/s41586-020-2050-1] [Citation(s) in RCA: 141] [Impact Index Per Article: 35.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2016] [Accepted: 01/08/2020] [Indexed: 12/22/2022]
Abstract
The avascular nature of cartilage makes it a unique tissue1–4, but whether and how the absence of nutrient supply regulates chondrogenesis remains unknown. Here, we show that obstruction of vascular invasion during bone healing favours chondrogenic over osteogenic differentiation of skeletal progenitor cells. Unexpectedly, this process is driven by a decreased availability of extracellular lipids. When lipids are scarce, skeletal progenitors activate FoxO transcription factors, which bind to the Sox9 promoter and increase its expression. Besides initiating chondrogenesis, SOX9 acts as a regulator of cellular metabolism by suppressing fatty acid oxidation, and thus adapts the cells to an avascular life. Our results define lipid scarcity as an important determinant of chondrogenic commitment, reveal a role for FoxOs during lipid starvation, and identify SOX9 as a critical metabolic mediator. These data highlight the importance of the nutritional microenvironment in the specification of skeletal cell fate.
Collapse
|
22
|
Mesenchymal VEGFA induces aberrant differentiation in heterotopic ossification. Bone Res 2019; 7:36. [PMID: 31840004 PMCID: PMC6904752 DOI: 10.1038/s41413-019-0075-6] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2019] [Revised: 08/11/2019] [Accepted: 08/26/2019] [Indexed: 12/19/2022] Open
Abstract
Heterotopic ossification (HO) is a debilitating condition characterized by the pathologic formation of ectopic bone. HO occurs commonly following orthopedic surgeries, burns, and neurologic injuries. While surgical excision may provide palliation, the procedure is often burdened with significant intra-operative blood loss due to a more robust contribution of blood supply to the pathologic bone than to native bone. Based on these clinical observations, we set out to examine the role of vascular signaling in HO. Vascular endothelial growth factor A (VEGFA) has previously been shown to be a crucial pro-angiogenic and pro-osteogenic cue during normal bone development and homeostasis. Our findings, using a validated mouse model of HO, demonstrate that HO lesions are highly vascular, and that VEGFA is critical to ectopic bone formation, despite lacking a contribution of endothelial cells within the developing anlagen.
Collapse
|
23
|
Rumney RMH, Lanham SA, Kanczler JM, Kao AP, Thiagarajan L, Dixon JE, Tozzi G, Oreffo ROC. In vivo delivery of VEGF RNA and protein to increase osteogenesis and intraosseous angiogenesis. Sci Rep 2019; 9:17745. [PMID: 31780671 PMCID: PMC6882814 DOI: 10.1038/s41598-019-53249-4] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2019] [Accepted: 10/22/2019] [Indexed: 12/12/2022] Open
Abstract
Deficient bone vasculature is a key component in pathological conditions ranging from developmental skeletal abnormalities to impaired bone repair. Vascularisation is dependent upon vascular endothelial growth factor (VEGF), which drives both angiogenesis and osteogenesis. The aim of this study was to examine the efficacy of blood vessel and bone formation following transfection with VEGF RNA or delivery of recombinant human VEGF165 protein (rhVEGF165) across in vitro and in vivo model systems. To quantify blood vessels within bone, an innovative approach was developed using high-resolution X-ray computed tomography (XCT) to generate quantifiable three-dimensional reconstructions. Application of rhVEGF165 enhanced osteogenesis, as evidenced by increased human osteoblast-like MG-63 cell proliferation in vitro and calvarial bone thickness following in vivo administration. In contrast, transfection with VEGF RNA triggered angiogenic effects by promoting VEGF protein secretion from MG-63VEGF165 cells in vitro, which resulted in significantly increased angiogenesis in the chorioallantoic (CAM) assay in ovo. Furthermore, direct transfection of bone with VEGF RNA in vivo increased intraosseous vascular branching. This study demonstrates the importance of continuous supply as opposed to a single high dose of VEGF on angiogenesis and osteogenesis and, illustrates the potential of XCT in delineating in 3D, blood vessel connectivity in bone.
Collapse
Affiliation(s)
- Robin M H Rumney
- Bone and Joint Research Group, Centre for Human Development, Stem Cells and Regeneration, Faculty of Medicine, Southampton University, Southampton, SO16 6YD, United Kingdom.
- School of Pharmacy and Biomedical Sciences, University of Portsmouth, St Michael's Building, White Swan Road, Portsmouth, PO1 2DT, United Kingdom.
| | - Stuart A Lanham
- Bone and Joint Research Group, Centre for Human Development, Stem Cells and Regeneration, Faculty of Medicine, Southampton University, Southampton, SO16 6YD, United Kingdom
| | - Janos M Kanczler
- Bone and Joint Research Group, Centre for Human Development, Stem Cells and Regeneration, Faculty of Medicine, Southampton University, Southampton, SO16 6YD, United Kingdom
| | - Alexander P Kao
- Zeiss Global Centre, School of Mechanical and Design Engineering, University of Portsmouth, Portsmouth, PO1 3DJ, United Kingdom
| | - Lalitha Thiagarajan
- Wolfson Centre for Stem Cells, Tissue Engineering and Modelling (STEM), Centre of Biomolecular Sciences, School of Pharmacy, University of Nottingham, Nottingham, NG7 2RD, United Kingdom
| | - James E Dixon
- Wolfson Centre for Stem Cells, Tissue Engineering and Modelling (STEM), Centre of Biomolecular Sciences, School of Pharmacy, University of Nottingham, Nottingham, NG7 2RD, United Kingdom
| | - Gianluca Tozzi
- Zeiss Global Centre, School of Mechanical and Design Engineering, University of Portsmouth, Portsmouth, PO1 3DJ, United Kingdom
| | - Richard O C Oreffo
- Bone and Joint Research Group, Centre for Human Development, Stem Cells and Regeneration, Faculty of Medicine, Southampton University, Southampton, SO16 6YD, United Kingdom
| |
Collapse
|
24
|
Deckelbaum RA, Lobov IB, Cheung E, Halasz G, Rajamani S, Lerner J, Tong C, Li Z, Boland P, Dominguez M, Hughes V, Yancopoulos GD, Murphy AJ, Thurston G, Cao J, Romano C, Gale NW. The potassium channel Kcne3 is a VEGFA-inducible gene selectively expressed by vascular endothelial tip cells. Angiogenesis 2019; 23:179-192. [PMID: 31754927 PMCID: PMC7160073 DOI: 10.1007/s10456-019-09696-8] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2019] [Accepted: 11/02/2019] [Indexed: 12/22/2022]
Abstract
Angiogenesis is largely driven by motile endothelial tip-cells capable of invading avascular tissue domains and enabling new vessel formation. Highly responsive to Vascular Endothelial Growth-Factor-A (VEGFA), endothelial tip-cells also suppress angiogenic sprouting in adjacent stalk cells, and thus have been a primary therapeutic focus in addressing neovascular pathologies. Surprisingly, however, there remains a paucity of specific endothelial tip-cell markers. Here, we employ transcriptional profiling and a lacZ reporter allele to identify Kcne3 as an early and selective endothelial tip-cell marker in multiple angiogenic contexts. In development, Kcne3 expression initiates during early phases of angiogenesis (E9) and remains specific to endothelial tip-cells, often adjacent to regions expressing VEGFA. Consistently, Kcne3 activation is highly responsive to exogenous VEGFA but maintains tip-cell specificity throughout normal retinal angiogenesis. We also demonstrate endothelial tip-cell selectivity of Kcne3 in several injury and tumor models. Together, our data show that Kcne3 is a unique marker of sprouting angiogenic tip-cells and offers new opportunities for investigating and targeting this cell type.
Collapse
Affiliation(s)
- Ron A Deckelbaum
- Department of Pre-Therapeutic Target Discovery, Regeneron Pharmaceuticals Inc, Tarrytown, NY, 10591, USA.
| | | | - Eunice Cheung
- Department of Ophthalmology, Regeneron Pharmaceuticals Inc, Tarrytown, NY, 10591, USA
| | - Gabor Halasz
- Department of Bioinformatics, Regeneron Pharmaceuticals Inc, Tarrytown, NY, 10591, USA
| | - Saathyaki Rajamani
- Department of Pre-Therapeutic Target Discovery, Regeneron Pharmaceuticals Inc, Tarrytown, NY, 10591, USA
| | - Julia Lerner
- Department of Pre-Therapeutic Target Discovery, Regeneron Pharmaceuticals Inc, Tarrytown, NY, 10591, USA
| | - Chunxiang Tong
- Department of Pre-Therapeutic Target Discovery, Regeneron Pharmaceuticals Inc, Tarrytown, NY, 10591, USA
| | - Zhe Li
- Department of Oncology & Angiogenesis, Regeneron Pharmaceuticals Inc., Tarrytown, NY, 10591, USA
| | - Patricia Boland
- Department of Oncology & Angiogenesis, Regeneron Pharmaceuticals Inc., Tarrytown, NY, 10591, USA
| | - Melissa Dominguez
- Department of Pre-Therapeutic Target Discovery, Regeneron Pharmaceuticals Inc, Tarrytown, NY, 10591, USA
| | - Virginia Hughes
- Department of Pre-Therapeutic Target Discovery, Regeneron Pharmaceuticals Inc, Tarrytown, NY, 10591, USA
| | - George D Yancopoulos
- Department of Research, Regeneron Pharmaceuticals Inc, Tarrytown, NY, 10591, USA
| | - Andrew J Murphy
- Department of Research, Regeneron Pharmaceuticals Inc, Tarrytown, NY, 10591, USA
| | - Gavin Thurston
- Department of Oncology & Angiogenesis, Regeneron Pharmaceuticals Inc., Tarrytown, NY, 10591, USA
| | - Jingtai Cao
- Department of Ophthalmology, Regeneron Pharmaceuticals Inc, Tarrytown, NY, 10591, USA
| | - Carmelo Romano
- Department of Ophthalmology, Regeneron Pharmaceuticals Inc, Tarrytown, NY, 10591, USA
| | - Nicholas W Gale
- Department of Pre-Therapeutic Target Discovery, Regeneron Pharmaceuticals Inc, Tarrytown, NY, 10591, USA
| |
Collapse
|
25
|
Buettmann EG, McKenzie JA, Migotsky N, Sykes DA, Hu P, Yoneda S, Silva MJ. VEGFA From Early Osteoblast Lineage Cells (Osterix+) Is Required in Mice for Fracture Healing. J Bone Miner Res 2019; 34:1690-1706. [PMID: 31081125 PMCID: PMC6744295 DOI: 10.1002/jbmr.3755] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/18/2018] [Revised: 04/23/2019] [Accepted: 05/01/2019] [Indexed: 12/21/2022]
Abstract
Bone formation via intramembranous and endochondral ossification is necessary for successful healing after a wide range of bone injuries. The pleiotropic cytokine, vascular endothelial growth factor A (VEGFA) has been shown, via nonspecific pharmacologic inhibition, to be indispensable for angiogenesis and ossification following bone fracture and cortical defect repair. However, the importance of VEGFA expression by different cell types during bone healing is not well understood. We sought to determine the role of VEGFA from different osteoblast cell subsets following clinically relevant models of bone fracture and cortical defect. Ubiquitin C (UBC), Osterix (Osx), or Dentin matrix protein 1 (Dmp1) Cre-ERT2 mice (male and female) containing floxed VEGFA alleles (VEGFAfl/fl ) were either given a femur full fracture, ulna stress fracture, or tibia cortical defect at 12 weeks of age. All mice received tamoxifen continuously starting 2 weeks before bone injury and throughout healing. UBC Cre-ERT2 VEGFAfl/fl (UBC cKO) mice, which were used to mimic nonspecific inhibition, had minimal bone formation and impaired angiogenesis across all bone injury models. UBC cKO mice also exhibited impaired periosteal cell proliferation during full fracture, but not stress fracture repair. Osx Cre-ERT2 VEGFAfl/fl (Osx cKO) mice, but not Dmp1 Cre-ERT2 VEGFAfl/fl (Dmp1 cKO) mice, showed impaired periosteal bone formation and angiogenesis in models of full fracture and stress fracture. Neither Osx cKO nor Dmp1 cKO mice demonstrated significant impairments in intramedullary bone formation and angiogenesis following cortical defect. These data suggest that VEGFA from early osteolineage cells (Osx+), but not mature osteoblasts/osteocytes (Dmp1+), is critical at the time of bone injury for rapid periosteal angiogenesis and woven bone formation during fracture repair. Whereas VEGFA from another cell source, not from the osteoblast cell lineage, is necessary at the time of injury for maximum cortical defect intramedullary angiogenesis and osteogenesis. © 2019 American Society for Bone and Mineral Research.
Collapse
Affiliation(s)
- Evan G Buettmann
- Department of Orthopaedic Surgery, Washington University in St. Louis, St. Louis, MO, USA.,Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, MO, USA
| | - Jennifer A McKenzie
- Department of Orthopaedic Surgery, Washington University in St. Louis, St. Louis, MO, USA
| | - Nicole Migotsky
- Department of Orthopaedic Surgery, Washington University in St. Louis, St. Louis, MO, USA.,Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, MO, USA
| | - David Aw Sykes
- Department of Biology, Washington University in St. Louis, St. Louis, MO, USA
| | - Pei Hu
- Department of Orthopaedic Surgery, Washington University in St. Louis, St. Louis, MO, USA
| | - Susumu Yoneda
- Department of Orthopaedic Surgery, Washington University in St. Louis, St. Louis, MO, USA
| | - Matthew J Silva
- Department of Orthopaedic Surgery, Washington University in St. Louis, St. Louis, MO, USA.,Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, MO, USA
| |
Collapse
|
26
|
Tong W, Tower RJ, Chen C, Wang L, Zhong L, Wei Y, Sun H, Cao G, Jia H, Pacifici M, Koyama E, Enomoto-Iwamoto M, Qin L. Periarticular Mesenchymal Progenitors Initiate and Contribute to Secondary Ossification Center Formation During Mouse Long Bone Development. Stem Cells 2019; 37:677-689. [PMID: 30681752 DOI: 10.1002/stem.2975] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2018] [Revised: 12/19/2018] [Accepted: 12/27/2018] [Indexed: 12/16/2022]
Abstract
Long bone development involves the embryonic formation of a primary ossification center (POC) in the incipient diaphysis followed by postnatal development of a secondary ossification center (SOC) at each epiphysis. Studies have elucidated major basic mechanisms of POC development, but relatively little is known about SOC development. To gain insights into SOC formation, we used Col2-Cre Rosa-tdTomato (Col2/Tomato) reporter mice and found that their periarticular region contained numerous Tomato-positive lineage cells expressing much higher Tomato fluorescence (termed TomatoH ) than underlying epiphyseal chondrocytes (termed TomatoL ). With time, the TomatoH cells became evident at the SOC invagination site and cartilage canal, increased in number in the expanding SOC, and were present as mesenchymal lineage cells in the subchondral bone. These data were verified in two mouse lineage tracing models, Col2-CreER Rosa-tdTomato and Gli1-CreER Rosa-tdTomato. In vitro tests showed that the periarticular TomatoH cells from Col2/Tomato mice contained mesenchymal progenitors with multidifferentiation abilities. During canal initiation, the cells expressed vascular endothelial growth factor (VEGF) and migrated into epiphyseal cartilage ahead of individual or clusters of endothelial cells, suggesting a unique role in promoting vasculogenesis. Later during SOC expansion, chondrocytes in epiphyseal cartilage expressed VEGF, and angiogenic blood vessels preceded TomatoH cells. Gene expression analyses of microdissected samples revealed upregulation of MMPs in periarticular cells at the invagination site and suggested potential roles for novel kinase and growth factor signaling pathways in regulating SOC canal initiation. In summary, our data indicate that the periarticular region surrounding epiphyseal cartilage contains mesenchymal progenitors that initiate SOC development and form subchondral bone. Stem Cells 2019;37:677-689.
Collapse
Affiliation(s)
- Wei Tong
- Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA.,Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, People's Republic of China
| | - Robert J Tower
- Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Chider Chen
- Department of Anatomy and Cell Biology, School of Dental Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Luqiang Wang
- Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Leilei Zhong
- Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Yulong Wei
- Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA.,Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, People's Republic of China
| | - Hao Sun
- Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Gaoyuan Cao
- Rutgers Institute for Translational Medicine and Science, Rutgers University, New Brunswick, New Jersey, USA
| | - Haoruo Jia
- Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Maurizio Pacifici
- Translational Research Program in Pediatric Orthopaedics, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - Eiki Koyama
- Translational Research Program in Pediatric Orthopaedics, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - Motomi Enomoto-Iwamoto
- Department of Orthopaedic Surgery, School of Medicine, University of Maryland, Baltimore, Maryland, USA
| | - Ling Qin
- Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| |
Collapse
|
27
|
Stiers PJ, Stegen S, van Gastel N, Van Looveren R, Torrekens S, Carmeliet G. Inhibition of the Oxygen Sensor PHD2 Enhances Tissue-Engineered Endochondral Bone Formation. J Bone Miner Res 2019; 34:333-348. [PMID: 30452097 DOI: 10.1002/jbmr.3599] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/21/2018] [Revised: 09/10/2018] [Accepted: 09/26/2018] [Indexed: 12/29/2022]
Abstract
Tissue engineering holds great promise for bone regenerative medicine, but clinical translation remains challenging. An important factor is the low cell survival after implantation, primarily caused by the lack of functional vasculature at the bone defect. Interestingly, bone development and repair initiate predominantly via an avascular cartilage template, indicating that chondrocytes are adapted to limited vascularization. Given these advantageous properties of chondrocytes, we questioned whether tissue-engineered cartilage intermediates implanted ectopically in mice are able to form bone, even when the volume size increases. Here, we show that endochondral ossification proceeds efficiently when implant size is limited (≤30 mm3 ), but chondrogenesis and matrix synthesis are impaired in the center of larger implants, leading to a fibrotic core. Increasing the level of angiogenic growth factors does not improve this outcome, because this strategy enhances peripheral bone formation, but disrupts the conversion of cartilage into bone in the center, resulting in a fibrotic core, even in small implants. On the other hand, activation of hypoxia signaling in cells before implantation stimulates chondrogenesis and matrix production, which culminates in enhanced bone formation throughout the entire implant. Together, our results show that induction of angiogenesis alone may lead to adverse effects during endochondral bone repair, whereas activation of hypoxia signaling represents a superior therapeutic strategy to improve endochondral bone regeneration in large tissue-engineered implants. © 2018 American Society for Bone and Mineral Research.
Collapse
Affiliation(s)
- Pieter-Jan Stiers
- Laboratory of Clinical and Experimental Endocrinology, Department of Chronic Diseases, Metabolism and Ageing, KU Leuven, Leuven, Belgium.,Prometheus, Division of Skeletal Tissue Engineering, KU Leuven, Leuven, Belgium
| | - Steve Stegen
- Laboratory of Clinical and Experimental Endocrinology, Department of Chronic Diseases, Metabolism and Ageing, KU Leuven, Leuven, Belgium.,Prometheus, Division of Skeletal Tissue Engineering, KU Leuven, Leuven, Belgium
| | - Nick van Gastel
- Laboratory of Clinical and Experimental Endocrinology, Department of Chronic Diseases, Metabolism and Ageing, KU Leuven, Leuven, Belgium.,Prometheus, Division of Skeletal Tissue Engineering, KU Leuven, Leuven, Belgium
| | - Riet Van Looveren
- Laboratory of Clinical and Experimental Endocrinology, Department of Chronic Diseases, Metabolism and Ageing, KU Leuven, Leuven, Belgium
| | - Sophie Torrekens
- Laboratory of Clinical and Experimental Endocrinology, Department of Chronic Diseases, Metabolism and Ageing, KU Leuven, Leuven, Belgium
| | - Geert Carmeliet
- Laboratory of Clinical and Experimental Endocrinology, Department of Chronic Diseases, Metabolism and Ageing, KU Leuven, Leuven, Belgium.,Prometheus, Division of Skeletal Tissue Engineering, KU Leuven, Leuven, Belgium
| |
Collapse
|
28
|
Stegen S, Laperre K, Eelen G, Rinaldi G, Fraisl P, Torrekens S, Van Looveren R, Loopmans S, Bultynck G, Vinckier S, Meersman F, Maxwell PH, Rai J, Weis M, Eyre DR, Ghesquière B, Fendt SM, Carmeliet P, Carmeliet G. HIF-1α metabolically controls collagen synthesis and modification in chondrocytes. Nature 2019; 565:511-515. [PMID: 30651640 PMCID: PMC7195049 DOI: 10.1038/s41586-019-0874-3] [Citation(s) in RCA: 177] [Impact Index Per Article: 35.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2017] [Accepted: 12/05/2018] [Indexed: 12/24/2022]
Abstract
Endochondral ossification, an important process in vertebrate bone formation, is highly dependent on correct functioning of growth plate chondrocytes1. Proliferation of these cells determines longitudinal bone growth and the matrix deposited provides a scaffold for future bone formation. However, these two energy-dependent anabolic processes occur in an avascular environment1,2. In addition, the centre of the expanding growth plate becomes hypoxic, and local activation of the hypoxia-inducible transcription factor HIF-1α is necessary for chondrocyte survival by unidentified cell-intrinsic mechanisms3-6. It is unknown whether there is a requirement for restriction of HIF-1α signalling in the other regions of the growth plate and whether chondrocyte metabolism controls cell function. Here we show that prolonged HIF-1α signalling in chondrocytes leads to skeletal dysplasia by interfering with cellular bioenergetics and biosynthesis. Decreased glucose oxidation results in an energy deficit, which limits proliferation, activates the unfolded protein response and reduces collagen synthesis. However, enhanced glutamine flux increases α-ketoglutarate levels, which in turn increases proline and lysine hydroxylation on collagen. This metabolically regulated collagen modification renders the cartilaginous matrix more resistant to protease-mediated degradation and thereby increases bone mass. Thus, inappropriate HIF-1α signalling results in skeletal dysplasia caused by collagen overmodification, an effect that may also contribute to other diseases involving the extracellular matrix such as cancer and fibrosis.
Collapse
Affiliation(s)
- Steve Stegen
- Laboratory of Clinical and Experimental Endocrinology, Department of Chronic Diseases, Metabolism and Ageing, KU Leuven, Leuven, Belgium
| | - Kjell Laperre
- Laboratory of Clinical and Experimental Endocrinology, Department of Chronic Diseases, Metabolism and Ageing, KU Leuven, Leuven, Belgium
| | - Guy Eelen
- Laboratory of Angiogenesis and Vascular Biology, VIB Center for Cancer Biology, VIB, Leuven, Belgium
- Laboratory of Angiogenesis and Vascular Metabolism, Department of Oncology and Leuven Cancer Institute (LKI), KU Leuven, Leuven, Belgium
| | - Gianmarco Rinaldi
- Laboratory of Cellular Metabolism and Metabolic Regulation, VIB Center for Cancer Biology, VIB, Leuven, Belgium
- Laboratory of Cellular Metabolism and Metabolic Regulation, Department of Oncology and Leuven Cancer Institute (LKI), KU Leuven, Leuven, Belgium
| | - Peter Fraisl
- Laboratory of Angiogenesis and Vascular Biology, VIB Center for Cancer Biology, VIB, Leuven, Belgium
- Laboratory of Angiogenesis and Vascular Metabolism, Department of Oncology and Leuven Cancer Institute (LKI), KU Leuven, Leuven, Belgium
| | - Sophie Torrekens
- Laboratory of Clinical and Experimental Endocrinology, Department of Chronic Diseases, Metabolism and Ageing, KU Leuven, Leuven, Belgium
| | - Riet Van Looveren
- Laboratory of Clinical and Experimental Endocrinology, Department of Chronic Diseases, Metabolism and Ageing, KU Leuven, Leuven, Belgium
| | - Shauni Loopmans
- Laboratory of Clinical and Experimental Endocrinology, Department of Chronic Diseases, Metabolism and Ageing, KU Leuven, Leuven, Belgium
| | - Geert Bultynck
- Laboratory of Molecular and Cellular Signalling, Department of Cellular and Molecular Medicine, KU Leuven, Leuven, Belgium
| | - Stefan Vinckier
- Laboratory of Angiogenesis and Vascular Biology, VIB Center for Cancer Biology, VIB, Leuven, Belgium
- Laboratory of Angiogenesis and Vascular Metabolism, Department of Oncology and Leuven Cancer Institute (LKI), KU Leuven, Leuven, Belgium
| | | | - Patrick H Maxwell
- Cambridge Institute for Medical Research, University of Cambridge, Cambridge, UK
| | - Jyoti Rai
- Department of Orthopaedics, University of Washington, Seattle, WA, USA
| | - MaryAnn Weis
- Department of Orthopaedics, University of Washington, Seattle, WA, USA
| | - David R Eyre
- Department of Orthopaedics, University of Washington, Seattle, WA, USA
| | - Bart Ghesquière
- Metabolomics Expertise Center, Department of Oncology, KU Leuven/VIB Center for Cancer Biology Leuven, Leuven, Belgium
| | - Sarah-Maria Fendt
- Laboratory of Cellular Metabolism and Metabolic Regulation, VIB Center for Cancer Biology, VIB, Leuven, Belgium
- Laboratory of Cellular Metabolism and Metabolic Regulation, Department of Oncology and Leuven Cancer Institute (LKI), KU Leuven, Leuven, Belgium
| | - Peter Carmeliet
- Laboratory of Angiogenesis and Vascular Biology, VIB Center for Cancer Biology, VIB, Leuven, Belgium
- Laboratory of Angiogenesis and Vascular Metabolism, Department of Oncology and Leuven Cancer Institute (LKI), KU Leuven, Leuven, Belgium
- State Key Laboratory of Ophtalmology, Zhongshan Ophtalmic Center, Sun Yat-Sen University, Guangzhou, China
| | - Geert Carmeliet
- Laboratory of Clinical and Experimental Endocrinology, Department of Chronic Diseases, Metabolism and Ageing, KU Leuven, Leuven, Belgium.
| |
Collapse
|
29
|
Lenas P. The Thermodynamics of Development in Bioartificial Tissue Design. Trends Biotechnol 2018; 36:1116-1126. [PMID: 30297153 DOI: 10.1016/j.tibtech.2018.06.006] [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: 03/05/2018] [Revised: 06/13/2018] [Accepted: 06/18/2018] [Indexed: 12/30/2022]
Abstract
The fabrication of bioartificial tissues with authentic structures that could assure their clinical efficacy remains a challenging problem. A new paradigm has emerged that designs bioartificial tissues as intermediate in development tissue forms, which can inherently progress autonomously on developmental pathways, self-organizing their cells into tissue structures as in their in vivo development. Biological processes involved in energy exchange between co-developing tissues are responsible for cell organization into the thermodynamically robust cellular patterns of tissue structures. Bioartificial tissue design rules that aim towards in vitro recapitulation of these processes can ensure the thermodynamic operation of developing tissues, leading to formation of the cellular patterns of tissue structures.
Collapse
Affiliation(s)
- Petros Lenas
- College of Science, Harbin Institute of Technology (Shenzhen), Shenzhen 518055, China.
| |
Collapse
|
30
|
Stegen S, Carmeliet G. The skeletal vascular system - Breathing life into bone tissue. Bone 2018; 115:50-58. [PMID: 28844835 DOI: 10.1016/j.bone.2017.08.022] [Citation(s) in RCA: 73] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/17/2017] [Accepted: 08/23/2017] [Indexed: 11/30/2022]
Abstract
During bone development, homeostasis and repair, a dense vascular system provides oxygen and nutrients to highly anabolic skeletal cells. Characteristic for the vascular system in bone is the serial organization of two capillary systems, each typified by specific morphological and physiological features. Especially the arterial capillaries mediate the growth of the bone vascular system, serve as a niche for skeletal and hematopoietic progenitors and couple angiogenesis to osteogenesis. Endothelial cells and osteoprogenitor cells interact not only physically, but also communicate to each other by secretion of growth factors. A vital angiogenic growth factor is vascular endothelial growth factor and its expression in skeletal cells is controlled by osteogenic transcription factors and hypoxia signaling, whereas the secretion of angiocrine factors by endothelial cells is regulated by Notch signaling, blood flow and possibly hypoxia. Bone loss and impaired fracture repair are often associated with reduced and disorganized blood vessel network and therapeutic targeting of the angiogenic response may contribute to enhanced bone regeneration.
Collapse
Affiliation(s)
- Steve Stegen
- Laboratory of Clinical and Experimental Endocrinology, Department of Chronic Diseases, Metabolism and Ageing, KU Leuven, 3000 Leuven, Belgium; Prometheus, Division of Skeletal Tissue Engineering, KU Leuven, 3000 Leuven, Belgium
| | - Geert Carmeliet
- Laboratory of Clinical and Experimental Endocrinology, Department of Chronic Diseases, Metabolism and Ageing, KU Leuven, 3000 Leuven, Belgium; Prometheus, Division of Skeletal Tissue Engineering, KU Leuven, 3000 Leuven, Belgium.
| |
Collapse
|
31
|
Watson EC, Adams RH. Biology of Bone: The Vasculature of the Skeletal System. Cold Spring Harb Perspect Med 2018; 8:cshperspect.a031559. [PMID: 28893838 DOI: 10.1101/cshperspect.a031559] [Citation(s) in RCA: 75] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Blood vessels are essential for the distribution of oxygen, nutrients, and immune cells, as well as the removal of waste products. In addition to this conventional role as a versatile conduit system, the endothelial cells forming the innermost layer of the vessel wall also possess important signaling capabilities and can control growth, patterning, homeostasis, and regeneration of the surrounding organ. In the skeletal system, blood vessels regulate developmental and regenerative bone formation as well as hematopoiesis by providing vascular niches for hematopoietic stem cells. Here we provide an overview of blood vessel architecture, growth and properties in the healthy, aging, and diseased skeletal system.
Collapse
Affiliation(s)
- Emma C Watson
- Max Planck Institute for Molecular Biomedicine, Department of Tissue Morphogenesis, and University of Münster, Faculty of Medicine, D-48149 Münster, Germany
| | - Ralf H Adams
- Max Planck Institute for Molecular Biomedicine, Department of Tissue Morphogenesis, and University of Münster, Faculty of Medicine, D-48149 Münster, Germany
| |
Collapse
|
32
|
Xu J, Qiu X, Liang Z, Smiley-Jewell S, Lu F, Yu M, Pinkerton KE, Zhao D, Shi B. Exposure to tobacco smoke increases bone loss in spontaneously hypertensive rats. Inhal Toxicol 2018; 30:229-238. [PMID: 30257116 DOI: 10.1080/08958378.2018.1506838] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2018] [Revised: 06/27/2018] [Accepted: 07/23/2018] [Indexed: 01/12/2023]
Abstract
PURPOSE To define if exposure to tobacco smoke (TS) could induce reduction of bone mass and impairment of bone architecture, features observed in osteoporosis in normotensive rats and the influence of TS exposure on the osteoporotic features exhibited in the spontaneously hypertensive (SH) rats. METHODS Normotensive Wistar Kyoto (WKY) and SH rats were exposed to filtered air or TS for 8 weeks, then their proximal femurs were extracted for micro-computed tomography (micro-CT) assessment, histological and immune-histological examinations to quantify the adverse influence of TS exposure on the bone mass and density, as well as bone architecture. RESULTS We found that TS exposure not only induced significant decreases in bone mineral density (BMD), bone volume (BV), cortical and trabecular thickness (Ct.Th and Tb.Th), trabecular surface area (Tb.Ar), expression of hypoxia-inducible factor-1α (HIF-1α) in the trabecular marrow, delayed ossification of cartilage, as well as statistical increases in trabecular separation (Tb.SP) and the number of trabecular marrow adipocytes in both WKY and SH rats, but also exacerbated multiple features of osteoporosis exhibited in SH rats, including decreased BMD, Ct.Th, Tb.Ar, HIF-1α expression, delayed cartilage ossification, and increased Tb.SP. CONCLUSIONS Our results show that TS exposure can reduce bone mass and impair bone architecture and exacerbate multiple features of osteoporosis exhibited in SH rats.
Collapse
Affiliation(s)
- Jingyi Xu
- a Department of Endocrinology , First Affiliated Hospital of Xi'an Jiaotong University , Xi'an , China
| | - Xing Qiu
- b Department of Orthopaedics , Affiliated Zhongshan Hospital of Dalian University , Dalian , China
| | - Zhou Liang
- b Department of Orthopaedics , Affiliated Zhongshan Hospital of Dalian University , Dalian , China
| | | | - Faqiang Lu
- b Department of Orthopaedics , Affiliated Zhongshan Hospital of Dalian University , Dalian , China
| | - Mang Yu
- d Center for Clinical Sciences Research , Stanford University School of Medicine , Stanford , CA , USA
| | - Kent E Pinkerton
- c Center for Health and the Environment , University of California , Davis CA , USA
| | - Dewei Zhao
- b Department of Orthopaedics , Affiliated Zhongshan Hospital of Dalian University , Dalian , China
| | - Bingyin Shi
- a Department of Endocrinology , First Affiliated Hospital of Xi'an Jiaotong University , Xi'an , China
| |
Collapse
|
33
|
Stiers PJ, van Gastel N, Moermans K, Stockmans I, Carmeliet G. An Ectopic Imaging Window for Intravital Imaging of Engineered Bone Tissue. JBMR Plus 2018; 2:92-102. [PMID: 30283894 PMCID: PMC6124161 DOI: 10.1002/jbm4.10028] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/11/2017] [Revised: 12/11/2017] [Accepted: 12/12/2017] [Indexed: 01/16/2023] Open
Abstract
Tissue engineering is a promising branch of regenerative medicine, but its clinical application remains limited because thorough knowledge of the in vivo repair processes in these engineered implants is limited. Common techniques to study the different phases of bone repair in mice are destructive and thus not optimal to gain insight into the dynamics of this process. Instead, multiphoton‐intravital microscopy (MP‐IVM) allows visualization of (sub)cellular processes at high resolution and frequency over extended periods of time when combined with an imaging window that permits optical access to implants in vivo. In this study, we have developed and validated an ectopic imaging window that can be placed over a tissue‐engineered construct implanted in mice. This approach did not interfere with the biological processes of bone regeneration taking place in these implants, as evidenced by histological and micro–computed tomography (μCT)‐based comparison to control ectopic implants. The ectopic imaging window permitted tracking of individual cells over several days in vivo. Furthermore, the use of fluorescent reporters allowed visualization of the onset of angiogenesis and osteogenesis in these constructs. Taken together, this novel imaging window will facilitate further analysis of the spatiotemporal regulation of cellular processes in bone tissue–engineered implants and provides a powerful tool to enhance the therapeutic potential of bone tissue engineering. © 2017 The Authors JBMR Plus published by Wiley Periodicals, Inc. on behalf of American Society for Bone and Mineral Research.
Collapse
Affiliation(s)
- Pieter-Jan Stiers
- Laboratory of Clinical and Experimental Endocrinology Department of Chronic Diseases, Metabolism and Ageing KU Leuven Leuven Belgium.,Prometheus Division of Skeletal Tissue Engineering KU Leuven Leuven Belgium
| | - Nick van Gastel
- Laboratory of Clinical and Experimental Endocrinology Department of Chronic Diseases, Metabolism and Ageing KU Leuven Leuven Belgium.,Prometheus Division of Skeletal Tissue Engineering KU Leuven Leuven Belgium
| | - Karen Moermans
- Laboratory of Clinical and Experimental Endocrinology Department of Chronic Diseases, Metabolism and Ageing KU Leuven Leuven Belgium
| | - Ingrid Stockmans
- Laboratory of Clinical and Experimental Endocrinology Department of Chronic Diseases, Metabolism and Ageing KU Leuven Leuven Belgium
| | - Geert Carmeliet
- Laboratory of Clinical and Experimental Endocrinology Department of Chronic Diseases, Metabolism and Ageing KU Leuven Leuven Belgium.,Prometheus Division of Skeletal Tissue Engineering KU Leuven Leuven Belgium
| |
Collapse
|
34
|
Pan Y, Chen J, Yu Y, Dai K, Wang J, Liu C. Enhancement of BMP-2-mediated angiogenesis and osteogenesis by 2-N,6-O-sulfated chitosan in bone regeneration. Biomater Sci 2018; 6:431-439. [DOI: 10.1039/c7bm01006k] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Sulfated polysaccharides are attractive semi-synthesized materials that can be used as a mimic of heparan sulfate to modulate the protein activity and other physiological processes.
Collapse
Affiliation(s)
- Yuanzhong Pan
- The State Key Laboratory of Bioreactor Engineering
- East China University of Science and Technology
- Shanghai
- 200237 People's Republic of China
- Engineering Research Center for Biomedical Materials of Ministry of Education
| | - Jie Chen
- The State Key Laboratory of Bioreactor Engineering
- East China University of Science and Technology
- Shanghai
- 200237 People's Republic of China
- Engineering Research Center for Biomedical Materials of Ministry of Education
| | - Yuanman Yu
- The State Key Laboratory of Bioreactor Engineering
- East China University of Science and Technology
- Shanghai
- 200237 People's Republic of China
- Engineering Research Center for Biomedical Materials of Ministry of Education
| | - Kai Dai
- The State Key Laboratory of Bioreactor Engineering
- East China University of Science and Technology
- Shanghai
- 200237 People's Republic of China
- Engineering Research Center for Biomedical Materials of Ministry of Education
| | - Jing Wang
- The State Key Laboratory of Bioreactor Engineering
- East China University of Science and Technology
- Shanghai
- 200237 People's Republic of China
- Engineering Research Center for Biomedical Materials of Ministry of Education
| | - Changsheng Liu
- The State Key Laboratory of Bioreactor Engineering
- East China University of Science and Technology
- Shanghai
- 200237 People's Republic of China
- Engineering Research Center for Biomedical Materials of Ministry of Education
| |
Collapse
|
35
|
Stiers PJ, van Gastel N, Moermans K, Stockmans I, Carmeliet G. Regulatory elements driving the expression of skeletal lineage reporters differ during bone development and adulthood. Bone 2017; 105:154-162. [PMID: 28863946 DOI: 10.1016/j.bone.2017.08.029] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/09/2017] [Revised: 08/28/2017] [Accepted: 08/28/2017] [Indexed: 01/06/2023]
Abstract
To improve bone healing or regeneration more insight in the fate and role of the different skeletal cell types is required. Mouse models for fate mapping and lineage tracing of skeletal cells, using stage-specific promoters, have advanced our understanding of bone development, a process that is largely recapitulated during bone repair. However, validation of these models is often only performed during development, whereas proof of the activity and specificity of the used promoters during the bone regenerative process is limited. Here, we show that the regulatory elements of the 6kb collagen type II promoter are not adequate to drive gene expression during bone repair. Similarly, the 2.3kb promoter of collagen type I lacks activity in adult mice, but the 3.2kb promoter is suitable. Furthermore, Cre-mediated fate mapping allows the visualization of progeny, but this label retention may hinder to distinguish these cells from ones with active expression of the marker at later time points. Together, our results show that the lineage-specific regulatory elements driving gene expression during bone development differ from those required later in life and during bone repair, and justify validation of lineage-specific cell tracing and gene silencing strategies during fracture healing and bone regenerative applications.
Collapse
Affiliation(s)
- Pieter-Jan Stiers
- Laboratory of Clinical and Experimental Endocrinology, Department of Clinical and Experimental Medicine, KU Leuven, Leuven, Belgium; Prometheus, Division of Skeletal Tissue Engineering, KU Leuven, Leuven, Belgium
| | - Nick van Gastel
- Laboratory of Clinical and Experimental Endocrinology, Department of Clinical and Experimental Medicine, KU Leuven, Leuven, Belgium; Prometheus, Division of Skeletal Tissue Engineering, KU Leuven, Leuven, Belgium
| | - Karen Moermans
- Laboratory of Clinical and Experimental Endocrinology, Department of Clinical and Experimental Medicine, KU Leuven, Leuven, Belgium
| | - Ingrid Stockmans
- Laboratory of Clinical and Experimental Endocrinology, Department of Clinical and Experimental Medicine, KU Leuven, Leuven, Belgium
| | - Geert Carmeliet
- Laboratory of Clinical and Experimental Endocrinology, Department of Clinical and Experimental Medicine, KU Leuven, Leuven, Belgium; Prometheus, Division of Skeletal Tissue Engineering, KU Leuven, Leuven, Belgium.
| |
Collapse
|
36
|
Subarnbhesaj A, Miyauchi M, Chanbora C, Mikuriya A, Nguyen PT, Furusho H, Ayuningtyas NF, Fujita M, Toratani S, Takechi M, Niida S, Takata T. Roles of VEGF-Flt-1 signaling in malignant behaviors of oral squamous cell carcinoma. PLoS One 2017; 12:e0187092. [PMID: 29149180 PMCID: PMC5693288 DOI: 10.1371/journal.pone.0187092] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2017] [Accepted: 10/15/2017] [Indexed: 12/11/2022] Open
Abstract
Background Vascular endothelial growth factor (VEGF) is a highly specific signaling protein for vascular endothelial cells that plays a critical role in tumor growth and invasion through angiogenesis, and may contribute to cell migration and activation of pre-osteoclasts, osteoclasts and some tumor cells. Objectives We aimed to clarify the detailed roles of VEGF-Flt-1 signaling in bone invasion of oral squamous cell carcinoma (OSCC) cells. Results Forty-two (42) of 54 cases with gingival SCC (77.8%) strongly expressed VEGF, and had a significantly increased number of Flt-1+ osteoclasts (p<0.01) and more aggressive bone invasion (p<0.05). PlGF, a ligand of Flt-1, induced osteoclastogenesis in single culture of bone marrow cells (BMCs), and inhibition of Flt-1-signaling by VEGF tyrosine kinase inhibitor and It’s down stream (Akt and ERK1/2) inhibitos reduced osteoclastogenesis in PlGF-stimulated BMCs (p<0.01). In molecular level, PlGF stimulation significantly upregulated RANKL expression in Flt-1-expressing HSC2 cells via phosphorylation of Akt and ERK1/2. In the co-culture of VEGF-producing HSC2 cells and BMCs, number of TRAP-positive osteoclasts markedly increased (p<0.01). The osteoclastogenesis was significantly inhibited by RANKL-neutralizing antibody (p<0.01) as well as by VEGF tyrosine kinase inhibitor (p<0.01) and it’s downstream (Akt and ERK1/2) inhibitors (p<0.01, p<0.05, respectively). Conclusion VEGF-Flt-1 signaling induces osteoclastogenesis in OSCC through two possible ways: 1) VEGF produced from OSCC cells can directly stimulate the Flt-1 pathway in preosteoclasts to induce migration to future bone resorbing area and differentiation into osteoclasts, and 2) VEGF-Flt-1 signaling upregulates RANKL expression in OSCC cells, which indirectly leads to osteoclast differentiation. Therefore, blocking of the VEGF-Flt-1 signaling may help inhibit bone invasion of OSCC.
Collapse
Affiliation(s)
- Ajiravudh Subarnbhesaj
- Department of Oral and Maxillofacial Pathobiology, School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Mutsumi Miyauchi
- Department of Oral and Maxillofacial Pathobiology, School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
- * E-mail: (MM); (TT)
| | - Chea Chanbora
- Department of Oral and Maxillofacial Pathobiology, School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Aki Mikuriya
- Department of Oral and Maxillofacial Pathobiology, School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Phuong Thao Nguyen
- Department of Global Dental Medicine and Pharmacy at Ho Chi Minh city, Integrated Health Sciences, School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Hisako Furusho
- Department of Oral and Maxillofacial Pathobiology, School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Nurina Febriyanti Ayuningtyas
- Department of Oral and Maxillofacial Pathobiology, School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Minoru Fujita
- Department of Oral and Maxillofacial Radiology, Institute of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Shigeaki Toratani
- Department of Molecular Oral Medicine and Maxillofacial Surgery, Institute of Biomedical and Health Sciences, Hiroshima University, Hiroshima University, Hiroshima, Japan
| | - Masaaki Takechi
- Department of Oral and Maxillofacial Surgery, Institute of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Shumpei Niida
- Biobank, Medical Genome Center, National Center for Geriatrics and Gerontology, Obu, Japan
| | - Takashi Takata
- Department of Oral and Maxillofacial Pathobiology, School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
- * E-mail: (MM); (TT)
| |
Collapse
|
37
|
Abstract
In addition to their conventional role as a conduit system for gases, nutrients, waste products or cells, blood vessels in the skeletal system play active roles in controlling multiple aspects of bone formation and provide niches for hematopoietic stem cells that reside within the bone marrow. In addition, recent studies have highlighted roles for blood vessels during bone healing. Here, we provide an overview of the architecture of the bone vasculature and discuss how blood vessels form within bone, how their formation is modulated, and how they function during development and fracture repair.
Collapse
Affiliation(s)
- Kishor K Sivaraj
- Max-Planck-Institute for Molecular Biomedicine, Department of Tissue Morphogenesis and University of Münster, Faculty of Medicine, Münster D-48149, Germany
| | - Ralf H Adams
- Max-Planck-Institute for Molecular Biomedicine, Department of Tissue Morphogenesis and University of Münster, Faculty of Medicine, Münster D-48149, Germany
| |
Collapse
|
38
|
Hu K, Olsen BR. Vascular endothelial growth factor control mechanisms in skeletal growth and repair. Dev Dyn 2017; 246:227-234. [PMID: 27750398 PMCID: PMC5354946 DOI: 10.1002/dvdy.24463] [Citation(s) in RCA: 84] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2016] [Accepted: 10/01/2016] [Indexed: 01/04/2023] Open
Abstract
Vascular endothelial growth factor A (VEGF) is a critical regulator of vascular development and postnatal angiogenesis and homeostasis, and it is essential for bone development and repair. Blood vessels serve both as structural templates for bone formation and they provide essential cells, growth factors and minerals needed for synthesis and mineralization, as well as turnover, of the extracellular matrix in bone. Through its regulation of angiogenesis, VEGF contributes to coupling of osteogenesis to angiogenesis, and it directly controls the differentiation and function of osteoblasts and osteoclasts. In this review, we summarize the properties of VEGF and its receptors that are relevant to bone formation and repair; the roles of VEGF during development of endochondral and membranous bones; and the contributions of VEGF to bone healing during different phases of bone repair. Finally, we discuss contributions of altered VEGF function in inherited disorders with bone defects as part of their phenotypes, and we speculate on what will be required before therapeutic strategies based on VEGF modulation can be developed for clinical use to treat patients with bone growth disorders and/or compromised bone repair. Developmental Dynamics 246:227-234, 2017. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Kai Hu
- Department of Developmental Biology, Harvard School of Dental Medicine, Boston, Massachusetts
| | - Bjorn R Olsen
- Department of Developmental Biology, Harvard School of Dental Medicine, Boston, Massachusetts
| |
Collapse
|
39
|
The role of vasculature in bone development, regeneration and proper systemic functioning. Angiogenesis 2017; 20:291-302. [PMID: 28194536 PMCID: PMC5511612 DOI: 10.1007/s10456-017-9541-1] [Citation(s) in RCA: 301] [Impact Index Per Article: 43.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2016] [Accepted: 02/09/2017] [Indexed: 12/14/2022]
Abstract
Bone is a richly vascularized connective tissue. As the main source of oxygen, nutrients, hormones, neurotransmitters and growth factors delivered to the bone cells, vasculature is indispensable for appropriate bone development, regeneration and remodeling. Bone vasculature also orchestrates the process of hematopoiesis. Blood supply to the skeletal system is provided by the networks of arteries and arterioles, having distinct molecular characteristics and localizations within the bone structures. Blood vessels of the bone develop through the process of angiogenesis, taking place through different, bone-specific mechanisms. Impaired functioning of the bone blood vessels may be associated with the occurrence of some skeletal and systemic diseases, i.e., osteonecrosis, osteoporosis, atherosclerosis or diabetes mellitus. When a disease or trauma-related large bone defects appear, bone grafting or bone tissue engineering-based strategies are required. However, a successful bone regeneration in both approaches largely depends on a proper blood supply. In this paper, we review the most recent data on the functions, molecular characteristics and significance of the bone blood vessels, with a particular emphasis on the role of angiogenesis and blood vessel functioning in bone development and regeneration, as well as the consequences of its impairment in the course of different skeletal and systemic diseases.
Collapse
|
40
|
Nishida T, Kubota S, Takigawa M. Cell Biological Assays for Measuring Chondrogenic Activities of CCN2 Protein. Methods Mol Biol 2017; 1489:219-237. [PMID: 27734380 DOI: 10.1007/978-1-4939-6430-7_21] [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] [Indexed: 06/06/2023]
Abstract
Growth-plate chondrocytes undergo proliferation, maturation, hypertrophic differentiation, and calcification; and these processes can be reproduced in vitro in a chondrocyte culture system. Using this system, we have shown that CCN family protein 2/connective tissue growth factor (CCN2/CTGF) promotes all stages of proliferation, maturation, hypertrophic differentiation, and calcification, thus suggesting that CCN2 is a multifunctional growth factor for chondrocytes and plays important roles in chondrocyte proliferation and differentiation. In this chapter, we describe how to evaluate CCN2 functions in these processes occurring in cultured chondrocytes. Evaluation strategies for cell proliferation include measuring DNA synthesis by [3H]-thymidine incorporation, cellular metabolic activity, and cell number with a hemocytometer. Next, evaluation strategies to assess maturation are analysis of the gene expression of markers of mature chondrocytes, and examination of proteoglycan and collagen synthesis by using radioactive compounds. In addition, cytohistochemical detection of glycosaminoglycans (GAGs), such as chondroitin sulfate, by use of alcian blue and toluidine blue staining is useful to evaluate chondrocyte maturation. These methods can be also used for evaluation of physiological functions of CCN2 in permanent chondrocytes such as articular and auricular chondrocytes, which do not calcify under physiological conditions. Next, evaluation of hypertrophic differentiation is performed by detecting type X collagen, which is specific marker of hypertrophic chondrocytes, and by measuring alkaline phosphatase (ALP) activity. Finally, evaluation of calcification is performed by detecting matrix calcification by use of alizarin red staining and by examining the incorporation of 45Ca into cartilaginous matrix. These methods would be useful for the evaluation not only of CCN2 but also of its derivatives and other CCN proteins.
Collapse
Affiliation(s)
- Takashi Nishida
- Department of Biochemistry and Molecular Dentistry, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, 2-5-1, Shikata-cho, Kita-ku, Okayama, 700-8525, Japan.
| | - Satoshi Kubota
- Advanced Research Center for Oral and Craniofacial Sciences, Okayama University Dental School/Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, 2-5-1, Shikata-cho, Kita-ku, Okayama, 700-8525, Japan
- Department of Membrane Biochemistry, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Masaharu Takigawa
- Advanced Research Center for Oral and Craniofacial Sciences, Okayama University Dental School/Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, 2-5-1, Shikata-cho, Kita-ku, Okayama, 700-8525, Japan.
| |
Collapse
|
41
|
|
42
|
Ramasamy SK, Kusumbe AP, Schiller M, Zeuschner D, Bixel MG, Milia C, Gamrekelashvili J, Limbourg A, Medvinsky A, Santoro MM, Limbourg FP, Adams RH. Blood flow controls bone vascular function and osteogenesis. Nat Commun 2016; 7:13601. [PMID: 27922003 PMCID: PMC5150650 DOI: 10.1038/ncomms13601] [Citation(s) in RCA: 231] [Impact Index Per Article: 28.9] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2016] [Accepted: 10/19/2016] [Indexed: 12/17/2022] Open
Abstract
While blood vessels play important roles in bone homeostasis and repair, fundamental aspects of vascular function in the skeletal system remain poorly understood. Here we show that the long bone vasculature generates a peculiar flow pattern, which is important for proper angiogenesis. Intravital imaging reveals that vessel growth in murine long bone involves the extension and anastomotic fusion of endothelial buds. Impaired blood flow leads to defective angiogenesis and osteogenesis, and downregulation of Notch signalling in endothelial cells. In aged mice, skeletal blood flow and endothelial Notch activity are also reduced leading to decreased angiogenesis and osteogenesis, which is reverted by genetic reactivation of Notch. Blood flow and angiogenesis in aged mice are also enhanced on administration of bisphosphonate, a class of drugs frequently used for the treatment of osteoporosis. We propose that blood flow and endothelial Notch signalling are key factors controlling ageing processes in the skeletal system. Formation of new blood vessels and bone is coupled. Here the authors show that blood flow represents a key regulator of angiogenesis and endothelial Notch signalling in the bone, and that reactivation of Notch signalling in the endothelium of aged mice rejuvenates the bone.
Collapse
Affiliation(s)
- Saravana K Ramasamy
- Faculty of Medicine, Department of Tissue Morphogenesis, Max-Planck-Institute for Molecular Biomedicine and University of Münster, D-48149 Münster, Germany.,Research group Integrative Skeletal Physiology, Institute of Clinical Sciences, Imperial College London, Hammersmith Hospital Campus, Du Cane Road, London W12 0NN, UK
| | - Anjali P Kusumbe
- Faculty of Medicine, Department of Tissue Morphogenesis, Max-Planck-Institute for Molecular Biomedicine and University of Münster, D-48149 Münster, Germany.,Research group Tissue and Tumor Microenvironments, Kennedy Institute of Rheumatology, University of Oxford, Oxford OX3 7LY, UK
| | - Maria Schiller
- Faculty of Medicine, Department of Tissue Morphogenesis, Max-Planck-Institute for Molecular Biomedicine and University of Münster, D-48149 Münster, Germany
| | - Dagmar Zeuschner
- Electron Microscopy Unit, Max-Planck-Institute for Molecular Biomedicine, D-48149 Münster, Germany
| | - M Gabriele Bixel
- Faculty of Medicine, Department of Tissue Morphogenesis, Max-Planck-Institute for Molecular Biomedicine and University of Münster, D-48149 Münster, Germany
| | - Carlo Milia
- VIB Vesalius Research Center, KU Leuven, 3000 Leuven, Belgium
| | - Jaba Gamrekelashvili
- Department of Nephrology and Hypertension, Hannover Medical School, D-30625 Hannover, Germany
| | - Anne Limbourg
- Department of Plastic and Reconstructive Surgery, Hannover Medical School, D-30625 Hannover, Germany
| | - Alexander Medvinsky
- Research group Ontogeny of Haematopoietic Stem Cells, MRC Centre for Regenerative Medicine, University of Edinburgh, Edinburgh EH16 4UU, Scotland
| | - Massimo M Santoro
- VIB Vesalius Research Center, KU Leuven, 3000 Leuven, Belgium.,Molecular Biotechnology Center, Department of Molecular Biotechnology and Health Sciences, University of Torino, 10126 Torino, Italy
| | - Florian P Limbourg
- Department of Nephrology and Hypertension, Hannover Medical School, D-30625 Hannover, Germany
| | - Ralf H Adams
- Faculty of Medicine, Department of Tissue Morphogenesis, Max-Planck-Institute for Molecular Biomedicine and University of Münster, D-48149 Münster, Germany
| |
Collapse
|
43
|
Eelen G, Verlinden L, Maes C, Beullens I, Gysemans C, Paik JH, DePinho RA, Bouillon R, Carmeliet G, Verstuyf A. Forkhead box O transcription factors in chondrocytes regulate endochondral bone formation. J Steroid Biochem Mol Biol 2016; 164:337-343. [PMID: 26232637 DOI: 10.1016/j.jsbmb.2015.07.015] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/30/2015] [Accepted: 07/26/2015] [Indexed: 01/14/2023]
Abstract
The differentiation of embryonic mesenchymal cells into chondrocytes and the subsequent formation of a cartilaginous scaffold that enables the formation of long bones are hallmarks of endochondral ossification. During this process, chondrocytes undergo a remarkable sequence of events involving proliferation, differentiation, hypertrophy and eventually apoptosis. Forkhead Box O (FoxO) transcription factors (TFs) are well-known regulators of such cellular processes. Although FoxO3a was previously shown to be regulated by 1,25-dihydroxyvitamin D3 in osteoblasts, a possible role for this family of TFs in chondrocytes during endochondral ossification remains largely unstudied. By crossing Collagen2-Cre mice with FoxO1lox/lox;FoxO3alox/lox;FoxO4lox/lox mice, we generated mice in which the three main FoxO isoforms were deleted in growth plate chondrocytes (chondrocyte triple knock-out; CTKO). Intriguingly, CTKO neonates showed a distinct elongation of the hypertrophic zone of the growth plate. CTKO mice had increased overall body and tail length at eight weeks of age and suffered from severe skeletal deformities at older ages. CTKO chondrocytes displayed decreased expression of genes involved in redox homeostasis. These observations illustrate the importance of FoxO signaling in chondrocytes during endochondral ossification.
Collapse
Affiliation(s)
- G Eelen
- Clinical and Experimental Endocrinology, KU Leuven, B-3000 Leuven, Belgium
| | - L Verlinden
- Clinical and Experimental Endocrinology, KU Leuven, B-3000 Leuven, Belgium
| | - C Maes
- Clinical and Experimental Endocrinology, KU Leuven, B-3000 Leuven, Belgium
| | - I Beullens
- Clinical and Experimental Endocrinology, KU Leuven, B-3000 Leuven, Belgium
| | - C Gysemans
- Clinical and Experimental Endocrinology, KU Leuven, B-3000 Leuven, Belgium
| | - J-H Paik
- Department of Pathology and Laboratory Medicine, Weill Cornell Medical College, New York City, NY, USA
| | - R A DePinho
- Department of Cancer Biology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - R Bouillon
- Clinical and Experimental Endocrinology, KU Leuven, B-3000 Leuven, Belgium
| | - G Carmeliet
- Clinical and Experimental Endocrinology, KU Leuven, B-3000 Leuven, Belgium
| | - A Verstuyf
- Clinical and Experimental Endocrinology, KU Leuven, B-3000 Leuven, Belgium.
| |
Collapse
|
44
|
Wen X, Li X, Tang Y, Tang J, Zhou S, Xie Y, Guo J, Yang J, Du X, Su N, Chen L. Chondrocyte FGFR3 Regulates Bone Mass by Inhibiting Osteogenesis. J Biol Chem 2016; 291:24912-24921. [PMID: 27729453 DOI: 10.1074/jbc.m116.730093] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2016] [Revised: 09/24/2016] [Indexed: 12/13/2022] Open
Abstract
Chondrogenesis can regulate bone formation. Fibroblast growth factor receptor 3, highly expressed in chondrocytes, is a negative regulator of bone growth. To investigate whether chondrocyte FGFR3 regulates osteogenesis, thereby contributing to postnatal bone formation and bone remodeling, mice with conditional knock-out of Fgfr3 in chondrocytes (mutant (MUT)) were generated. MUT mice displayed overgrowth of bone with lengthened growth plates. Bone mass of MUT mice was significantly increased at both 1 month and 4 months of age. Histological analysis showed that osteoblast number and bone formation were remarkably enhanced after deletion of Fgfr3 in chondrocytes. Chondrocyte-osteoblast co-culture assay further revealed that Fgfr3 deficiency in chondrocytes promoted differentiation and mineralization of osteoblasts by up-regulating the expressions of Ihh, Bmp2, Bmp4, Bmp7, Wnt4, and Tgf-β1, as well as down-regulating Nog expression. In addition, osteoclastogenesis was also impaired in MUT mice with decreased number of osteoclasts lining trabecular bone, which may be related to the reduced ratio of Rankl to Opg in Fgfr3-deficient chondrocytes. This study reveals that chondrocyte FGFR3 is involved in the regulation of bone formation and bone remodeling by a paracrine mechanism.
Collapse
Affiliation(s)
- Xuan Wen
- From the Department of Rehabilitation Medicine, Center of Bone Metabolism and Repair, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Research Institute of Surgery, Daping Hospital, Third Military Medical University, Chongqing 400042
| | - Xiaogang Li
- From the Department of Rehabilitation Medicine, Center of Bone Metabolism and Repair, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Research Institute of Surgery, Daping Hospital, Third Military Medical University, Chongqing 400042.,the 305 Hospital of Chinese People's Liberation Army, Beijing 100017, and
| | - Yubin Tang
- From the Department of Rehabilitation Medicine, Center of Bone Metabolism and Repair, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Research Institute of Surgery, Daping Hospital, Third Military Medical University, Chongqing 400042.,the Department of Emergency Treatment, Lanzhou General Hospital, Lanzhou Command, Chinese People's Liberation Army, Lanzhou 730050, China
| | - Junzhou Tang
- From the Department of Rehabilitation Medicine, Center of Bone Metabolism and Repair, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Research Institute of Surgery, Daping Hospital, Third Military Medical University, Chongqing 400042
| | - Siru Zhou
- From the Department of Rehabilitation Medicine, Center of Bone Metabolism and Repair, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Research Institute of Surgery, Daping Hospital, Third Military Medical University, Chongqing 400042
| | - Yangli Xie
- From the Department of Rehabilitation Medicine, Center of Bone Metabolism and Repair, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Research Institute of Surgery, Daping Hospital, Third Military Medical University, Chongqing 400042
| | - Jingyuan Guo
- From the Department of Rehabilitation Medicine, Center of Bone Metabolism and Repair, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Research Institute of Surgery, Daping Hospital, Third Military Medical University, Chongqing 400042
| | - Jing Yang
- From the Department of Rehabilitation Medicine, Center of Bone Metabolism and Repair, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Research Institute of Surgery, Daping Hospital, Third Military Medical University, Chongqing 400042
| | - Xiaolan Du
- From the Department of Rehabilitation Medicine, Center of Bone Metabolism and Repair, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Research Institute of Surgery, Daping Hospital, Third Military Medical University, Chongqing 400042
| | - Nan Su
- From the Department of Rehabilitation Medicine, Center of Bone Metabolism and Repair, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Research Institute of Surgery, Daping Hospital, Third Military Medical University, Chongqing 400042,
| | - Lin Chen
- From the Department of Rehabilitation Medicine, Center of Bone Metabolism and Repair, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Research Institute of Surgery, Daping Hospital, Third Military Medical University, Chongqing 400042,
| |
Collapse
|
45
|
Kaplan O, Zárubová J, Mikulová B, Filová E, Bártová J, Bačáková L, Brynda E. Enhanced Mitogenic Activity of Recombinant Human Vascular Endothelial Growth Factor VEGF121 Expressed in E. coli Origami B (DE3) with Molecular Chaperones. PLoS One 2016; 11:e0163697. [PMID: 27716773 PMCID: PMC5055331 DOI: 10.1371/journal.pone.0163697] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2016] [Accepted: 09/13/2016] [Indexed: 12/22/2022] Open
Abstract
We describe the production of a highly-active mutant VEGF variant, α2-PI1-8-VEGF121, which contains a substrate sequence for factor XIIIa at the aminoterminus designed for incorporation into a fibrin gel. The α2-PI1-8-VEGF121 gene was synthesized, cloned into a pET-32a(+) vector and expressed in Escherichia coli Origami B (DE3) host cells. To increase the protein folding and the solubility, the resulting thioredoxin-α2-PI1-8-VEGF121 fusion protein was co-expressed with recombinant molecular chaperones GroES/EL encoded by independent plasmid pGro7. The fusion protein was purified from the soluble fraction of cytoplasmic proteins using affinity chromatography. After cleavage of the thioredoxin fusion part with thrombin, the target protein was purified by a second round of affinity chromatography. The yield of purified α2-PI1-8-VEGF121 was 1.4 mg per liter of the cell culture. The α2-PI1-8-VEGF121 expressed in this work increased the proliferation of endothelial cells 3.9-8.7 times in comparison with commercially-available recombinant VEGF121. This very high mitogenic activity may be caused by co-expression of the growth factor with molecular chaperones not previously used in VEGF production. At the same time, α2-PI1-8-VEGF121 did not elicit considerable inflammatory activation of human endothelial HUVEC cells and human monocyte-like THP-1 cells.
Collapse
Affiliation(s)
- Ondřej Kaplan
- Institute of Macromolecular Chemistry, Czech Academy of Sciences, CZ-162 06, Prague, Czech Republic
- Institute of Physiology, Czech Academy of Sciences, CZ-142 20, Prague, Czech Republic
- * E-mail:
| | - Jana Zárubová
- Institute of Physiology, Czech Academy of Sciences, CZ-142 20, Prague, Czech Republic
| | - Barbora Mikulová
- Institute of Physiology, Czech Academy of Sciences, CZ-142 20, Prague, Czech Republic
- Faculty of Science, Charles University in Prague, CZ-128 40, Prague, Czech Republic
| | - Elena Filová
- Institute of Physiology, Czech Academy of Sciences, CZ-142 20, Prague, Czech Republic
| | - Jiřina Bártová
- School of Dental Medicine, General University Hospital in Prague, CZ-128 08, Prague, Czech Republic
| | - Lucie Bačáková
- Institute of Physiology, Czech Academy of Sciences, CZ-142 20, Prague, Czech Republic
| | - Eduard Brynda
- Institute of Macromolecular Chemistry, Czech Academy of Sciences, CZ-162 06, Prague, Czech Republic
| |
Collapse
|
46
|
Hu K, Olsen BR. The roles of vascular endothelial growth factor in bone repair and regeneration. Bone 2016; 91:30-8. [PMID: 27353702 PMCID: PMC4996701 DOI: 10.1016/j.bone.2016.06.013] [Citation(s) in RCA: 365] [Impact Index Per Article: 45.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/01/2016] [Revised: 06/22/2016] [Accepted: 06/23/2016] [Indexed: 02/08/2023]
Abstract
Vascular endothelial growth factor-A (VEGF) is one of the most important growth factors for regulation of vascular development and angiogenesis. Since bone is a highly vascularized organ and angiogenesis plays an important role in osteogenesis, VEGF also influences skeletal development and postnatal bone repair. Compromised bone repair and regeneration in many patients can be attributed to impaired blood supply; thus, modulation of VEGF levels in bones represents a potential strategy for treating compromised bone repair and improving bone regeneration. This review (i) summarizes the roles of VEGF at different stages of bone repair, including the phases of inflammation, endochondral ossification, intramembranous ossification during callus formation and bone remodeling; (ii) discusses different mechanisms underlying the effects of VEGF on osteoblast function, including paracrine, autocrine and intracrine signaling during bone repair; (iii) summarizes the role of VEGF in the bone regenerative procedure, distraction osteogenesis; and (iv) reviews evidence for the effects of VEGF in the context of repair and regeneration techniques involving the use of scaffolds, skeletal stem cells and growth factors.
Collapse
Affiliation(s)
- Kai Hu
- Department of Developmental Biology, Harvard School of Dental Medicine, Boston, MA, USA.
| | - Bjorn R Olsen
- Department of Developmental Biology, Harvard School of Dental Medicine, Boston, MA, USA.
| |
Collapse
|
47
|
Yamamoto H, Rundqvist H, Branco C, Johnson RS. Autocrine VEGF Isoforms Differentially Regulate Endothelial Cell Behavior. Front Cell Dev Biol 2016; 4:99. [PMID: 27709112 PMCID: PMC5030275 DOI: 10.3389/fcell.2016.00099] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2016] [Accepted: 08/26/2016] [Indexed: 01/06/2023] Open
Abstract
Vascular endothelial growth factor A (VEGF) is involved in all the essential biology of endothelial cells, from proliferation to vessel function, by mediating intercellular interactions and monolayer integrity. It is expressed as three major alternative spliced variants. In mice, these are VEGF120, VEGF164, and VEGF188, each with different affinities for extracellular matrices and cell surfaces, depending on the inclusion of heparin-binding sites, encoded by exons 6 and 7. To determine the role of each VEGF isoform in endothelial homeostasis, we compared phenotypes of primary endothelial cells isolated from lungs of mice expressing single VEGF isoforms in normoxic and hypoxic conditions. The differential expression and distribution of VEGF isoforms affect endothelial cell functions, such as proliferation, adhesion, migration, and integrity, which are dependent on the stability of and affinity to VEGF receptor 2 (VEGFR2). We found a correlation between autocrine VEGF164 and VEGFR2 stability, which is also associated with increased expression of proteins involved in cell adhesion. Endothelial cells expressing only VEGF188, which localizes to extracellular matrices or cell surfaces, presented a mesenchymal morphology and weakened monolayer integrity. Cells expressing only VEGF120 lacked stable VEGFR2 and dysfunctional downstream processes, rendering the cells unviable. Endothelial cells expressing these different isoforms in isolation also had differing rates of apoptosis, proliferation, and signaling via nitric oxide (NO) synthesis. These data indicate that autocrine signaling of each VEGF isoform has unique functions on endothelial homeostasis and response to hypoxia, due to both distinct VEGF distribution and VEGFR2 stability, which appears to be, at least partly, affected by differential NO production. This study demonstrates that each autocrine VEGF isoform has a distinct effect on downstream functions, namely VEGFR2-regulated endothelial cell homeostasis in normoxia and hypoxia.
Collapse
Affiliation(s)
- Hideki Yamamoto
- Department of Physiology, Development and Neuroscience, University of CambridgeCambridge, UK
| | - Helene Rundqvist
- Department of Cell and Molecular Biology, Karolinska InstitutetStockholm, Sweden
| | - Cristina Branco
- Department of Physiology, Development and Neuroscience, University of CambridgeCambridge, UK
| | - Randall S. Johnson
- Department of Physiology, Development and Neuroscience, University of CambridgeCambridge, UK
- Department of Cell and Molecular Biology, Karolinska InstitutetStockholm, Sweden
| |
Collapse
|
48
|
Stiers PJ, van Gastel N, Carmeliet G. Targeting the hypoxic response in bone tissue engineering: A balance between supply and consumption to improve bone regeneration. Mol Cell Endocrinol 2016; 432:96-105. [PMID: 26768117 DOI: 10.1016/j.mce.2015.12.024] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/28/2015] [Revised: 12/22/2015] [Accepted: 12/31/2015] [Indexed: 12/19/2022]
Abstract
Bone tissue engineering is a promising therapeutic alternative for bone grafting of large skeletal defects. It generally comprises an ex vivo engineered combination of a carrier structure, stem/progenitor cells and growth factors. However, the success of these regenerative implants largely depends on how well implanted cells will adapt to the hostile and hypoxic host environment they encounter after implantation. In this review, we will discuss how hypoxia signalling may be used to improve bone regeneration in a tissue-engineered construct. First, hypoxia signalling induces angiogenesis which increases the survival of the implanted cells as well as stimulates bone formation. Second, hypoxia signalling has also angiogenesis-independent effects on mesenchymal cells in vitro, offering exciting new possibilities to improve tissue-engineered bone regeneration in vivo. In addition, studies in other fields have shown that benefits of modulating hypoxia signalling include enhanced cell survival, proliferation and differentiation, culminating in a more potent regenerative implant. Finally, the stimulation of endochondral bone formation as a physiological pathway to circumvent the harmful effects of hypoxia will be briefly touched upon. Thus, angiogenic dependent and independent processes may counteract the deleterious hypoxic effects and we will discuss several therapeutic strategies that may be combined to withstand the hypoxia upon implantation and improve bone regeneration.
Collapse
Affiliation(s)
- Pieter-Jan Stiers
- Laboratory of Clinical and Experimental Endocrinology, Department of Clinical and Experimental Medicine, KU Leuven, Leuven, Belgium; Prometheus, Division of Skeletal Tissue Engineering, KU Leuven, Leuven, Belgium
| | - Nick van Gastel
- Laboratory of Clinical and Experimental Endocrinology, Department of Clinical and Experimental Medicine, KU Leuven, Leuven, Belgium; Prometheus, Division of Skeletal Tissue Engineering, KU Leuven, Leuven, Belgium
| | - Geert Carmeliet
- Laboratory of Clinical and Experimental Endocrinology, Department of Clinical and Experimental Medicine, KU Leuven, Leuven, Belgium; Prometheus, Division of Skeletal Tissue Engineering, KU Leuven, Leuven, Belgium.
| |
Collapse
|
49
|
Ramasamy SK, Kusumbe AP, Itkin T, Gur-Cohen S, Lapidot T, Adams RH. Regulation of Hematopoiesis and Osteogenesis by Blood Vessel-Derived Signals. Annu Rev Cell Dev Biol 2016; 32:649-675. [PMID: 27576121 DOI: 10.1146/annurev-cellbio-111315-124936] [Citation(s) in RCA: 100] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
In addition to their conventional role as a versatile transport system, blood vessels provide signals controlling organ development, regeneration, and stem cell behavior. In the skeletal system, certain capillaries support perivascular osteoprogenitor cells and thereby control bone formation. Blood vessels are also a critical component of niche microenvironments for hematopoietic stem cells. Here we discuss key pathways and factors controlling endothelial cell behavior in bone, the role of vessels in osteogenesis, and the nature of vascular stem cell niches in bone marrow.
Collapse
Affiliation(s)
- Saravana K Ramasamy
- Department of Tissue Morphogenesis, Max Planck Institute for Molecular Biomedicine, D-48169 Münster, Germany; .,Faculty of Medicine, University of Münster, D-48149 Münster, Germany
| | - Anjali P Kusumbe
- Department of Tissue Morphogenesis, Max Planck Institute for Molecular Biomedicine, D-48169 Münster, Germany; .,Faculty of Medicine, University of Münster, D-48149 Münster, Germany
| | - Tomer Itkin
- Department of Immunology, The Weizmann Institute of Science, Rehovot, 76100, Israel;
| | - Shiri Gur-Cohen
- Department of Immunology, The Weizmann Institute of Science, Rehovot, 76100, Israel;
| | - Tsvee Lapidot
- Department of Immunology, The Weizmann Institute of Science, Rehovot, 76100, Israel;
| | - Ralf H Adams
- Department of Tissue Morphogenesis, Max Planck Institute for Molecular Biomedicine, D-48169 Münster, Germany; .,Faculty of Medicine, University of Münster, D-48149 Münster, Germany
| |
Collapse
|
50
|
Marsano A, Medeiros da Cunha CM, Ghanaati S, Gueven S, Centola M, Tsaryk R, Barbeck M, Stuedle C, Barbero A, Helmrich U, Schaeren S, Kirkpatrick JC, Banfi A, Martin I. Spontaneous In Vivo Chondrogenesis of Bone Marrow-Derived Mesenchymal Progenitor Cells by Blocking Vascular Endothelial Growth Factor Signaling. Stem Cells Transl Med 2016; 5:1730-1738. [PMID: 27460852 DOI: 10.5966/sctm.2015-0321] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2015] [Accepted: 06/09/2016] [Indexed: 11/16/2022] Open
Abstract
: Chondrogenic differentiation of bone marrow-derived mesenchymal stromal/stem cells (MSCs) can be induced by presenting morphogenetic factors or soluble signals but typically suffers from limited efficiency, reproducibility across primary batches, and maintenance of phenotypic stability. Considering the avascular and hypoxic milieu of articular cartilage, we hypothesized that sole inhibition of angiogenesis can provide physiological cues to direct in vivo differentiation of uncommitted MSCs to stable cartilage formation. Human MSCs were retrovirally transduced to express a decoy soluble vascular endothelial growth factor (VEGF) receptor-2 (sFlk1), which efficiently sequesters endogenous VEGF in vivo, seeded on collagen sponges and immediately implanted ectopically in nude mice. Although naïve cells formed vascularized fibrous tissue, sFlk1-MSCs abolished vascular ingrowth into engineered constructs, which efficiently and reproducibly developed into hyaline cartilage. The generated cartilage was phenotypically stable and showed no sign of hypertrophic evolution up to 12 weeks. In vitro analyses indicated that spontaneous chondrogenic differentiation by blockade of angiogenesis was related to the generation of a hypoxic environment, in turn activating the transforming growth factor-β pathway. These findings suggest that VEGF blockade is a robust strategy to enhance cartilage repair by endogenous or grafted mesenchymal progenitors. This article outlines the general paradigm of controlling the fate of implanted stem/progenitor cells by engineering their ability to establish specific microenvironmental conditions rather than directly providing individual morphogenic cues. SIGNIFICANCE Chondrogenic differentiation of mesenchymal stromal/stem cells (MSCs) is typically targeted by morphogen delivery, which is often associated with limited efficiency, stability, and robustness. This article proposes a strategy to engineer MSCs with the capacity to establish specific microenvironmental conditions, supporting their own targeted differentiation program. Sole blockade of angiogenesis mediated by transduction for sFlk-1, without delivery of additional morphogens, is sufficient for inducing MSC chondrogenic differentiation. The findings represent a relevant step forward in the field because the method allowed reducing interdonor variability in MSC differentiation efficiency and, importantly, onset of a stable, nonhypertrophic chondrocyte phenotype.
Collapse
Affiliation(s)
- Anna Marsano
- Department of Biomedicine, University of Basel, Basel, Switzerland
- Department of Surgery, University Hospital Basel, Basel, Switzerland
| | - Carolina M Medeiros da Cunha
- Department of Biomedicine, University of Basel, Basel, Switzerland
- Department of Surgery, University Hospital Basel, Basel, Switzerland
| | - Shahram Ghanaati
- Institute of Pathology, University Medical Center Mainz, Mainz, Germany
- Department for Oral, Craniomaxillofacial and Facial Plastic Surgery, University Frankfurt am Main, Frankfurt, Germany
| | - Sinan Gueven
- Department of Biomedicine, University of Basel, Basel, Switzerland
- Department of Surgery, University Hospital Basel, Basel, Switzerland
- Izmir Biomedicine and Genome Center, Dokuz Eylul University Health Campus, Izmir, Turkey
| | - Matteo Centola
- Department of Biomedicine, University of Basel, Basel, Switzerland
- Department of Surgery, University Hospital Basel, Basel, Switzerland
| | - Roman Tsaryk
- Institute of Pathology, University Medical Center Mainz, Mainz, Germany
| | - Mike Barbeck
- Institute of Pathology, University Medical Center Mainz, Mainz, Germany
- Department for Oral, Craniomaxillofacial and Facial Plastic Surgery, University Frankfurt am Main, Frankfurt, Germany
| | - Chiara Stuedle
- Department of Biomedicine, University of Basel, Basel, Switzerland
- Department of Surgery, University Hospital Basel, Basel, Switzerland
| | - Andrea Barbero
- Department of Biomedicine, University of Basel, Basel, Switzerland
- Department of Surgery, University Hospital Basel, Basel, Switzerland
| | - Uta Helmrich
- Department of Biomedicine, University of Basel, Basel, Switzerland
- Department of Surgery, University Hospital Basel, Basel, Switzerland
| | - Stefan Schaeren
- Department of Surgery, University Hospital Basel, Basel, Switzerland
| | | | - Andrea Banfi
- Department of Biomedicine, University of Basel, Basel, Switzerland
- Department of Surgery, University Hospital Basel, Basel, Switzerland
| | - Ivan Martin
- Department of Biomedicine, University of Basel, Basel, Switzerland
- Department of Surgery, University Hospital Basel, Basel, Switzerland
| |
Collapse
|