1
|
Huang G, Li W, Zhong Y, Liao W, Zhang Z. Mendelian randomization to evaluate the causal relationship between liver enzymes and the risk of six specific bone and joint-related diseases. Front Immunol 2023; 14:1195553. [PMID: 37662902 PMCID: PMC10469508 DOI: 10.3389/fimmu.2023.1195553] [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: 03/28/2023] [Accepted: 07/24/2023] [Indexed: 09/05/2023] Open
Abstract
Background Studies of liver dysfunction in relation to bone and joint-related diseases are scarce, and its causality remains unclear. Our objective was to investigate whether serum liver enzymes are causally associated with bone and joint-related diseases using Mendelian randomization (MR) designs. Methods Genetic data on serum liver enzymes (alkaline phosphatase (ALP); alanine transaminase (ALT); gamma-glutamyl transferase (GGT)) and six common bone and joint-related diseases (rheumatoid arthritis (RA), osteoporosis, osteoarthritis (OA), ankylosing spondylitis, psoriatic arthritis, and gout) were derived from independent genome-wide association studies of European ancestry. The inverse variance-weighted (IVW) method was applied for the main causal estimate. Complementary sensitivity analyses and reverse causal analyses were utilized to confirm the robustness of the results. Results Using the IVW method, the positive causality between ALP and the risk of osteoporosis diagnosed by bone mineral density (BMD) at different sites was indicated (femoral neck, lumbar spine, and total body BMD, odds ratio (OR) [95% CI], 0.40 [0.23-0.69], 0.35 [0.19-0.67], and 0.33 [0.22-0.51], respectively). ALP was also linked to a higher risk of RA (OR [95% CI], 6.26 [1.69-23.51]). Evidence of potential harmful effects of higher levels of ALT on the risk of hip and knee OA was acquired (OR [95% CI], 2.48 [1.39-4.41] and 3.07 [1.49-6.30], respectively). No causal relationship was observed between GGT and these bone and joint-related diseases. The study also found that BMD were all negatively linked to ALP levels (OR [95% CI] for TBMD, FN-BMD, and LS-BMD: 0.993 [0.991-0.995], 0.993 [0.988-0.998], and 0.993 [0.989, 0.998], respectively) in the reverse causal analysis. The results were replicated via sensitivity analysis in the validation process. Conclusions Our study revealed a significant association between liver function and bone and joint-related diseases.
Collapse
Affiliation(s)
- Guiwu Huang
- Department of Joint Surgery, The First Affiliated Hospital of Sun Yat-sen University, Sun Yat-sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Orthopedics and Traumatology, The First Affiliated Hospital of Sun Yat-sen University, Sun Yat-sen University, Guangzhou, China
| | - Wenchang Li
- Department of Joint Surgery, The First Affiliated Hospital of Sun Yat-sen University, Sun Yat-sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Orthopedics and Traumatology, The First Affiliated Hospital of Sun Yat-sen University, Sun Yat-sen University, Guangzhou, China
| | - Yonglie Zhong
- Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Weiming Liao
- Department of Joint Surgery, The First Affiliated Hospital of Sun Yat-sen University, Sun Yat-sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Orthopedics and Traumatology, The First Affiliated Hospital of Sun Yat-sen University, Sun Yat-sen University, Guangzhou, China
| | - Zhiqi Zhang
- Department of Joint Surgery, The First Affiliated Hospital of Sun Yat-sen University, Sun Yat-sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Orthopedics and Traumatology, The First Affiliated Hospital of Sun Yat-sen University, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
2
|
Gong XF, Li XP, Zhang LX, Center JR, Bliuc D, Shi Y, Wang HB, He L, Wu XB. Current status and distribution of hip fractures among older adults in China. Osteoporos Int 2021; 32:1785-1793. [PMID: 33655399 DOI: 10.1007/s00198-021-05849-y] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/07/2020] [Accepted: 01/12/2021] [Indexed: 12/13/2022]
Abstract
UNLABELLED China is a middle-risk country for hip fracture at present, which differs from previous data that it was low-risk. By 2050, the total number of hip fractures in people older than 65 years is predicted to be 1.3 million. INTRODUCTION To assess hip fracture incidence in China and examine the heterogeneity of hip fracture in seven geographical regions of China. METHODS There were 238,230 hip fracture patients aged 65 years or older from 2013 to 2016 from a large national in-patients database (HQMS) involving 30.6 million hospitalizations. Taking into account the total national hospitalization rate per calendar year, we estimated the incidence of hip fracture per 100,000 residents older than 65 years in China overall and in seven geographical Chinese regions. RESULTS The proportion of men and women older than 65 years with hip fractures was 1.00:1.95. Between 2013 and 2016, the number of hip fractures per 100,000 people age 65+ was 278. China has vast territories; the number of hip fractures per 100,000 people over 65 years old was 202 in Northeast China and 374 in Northwest China. Northwest has higher altitude, lower population density, is less developed with lower urbanization than Northeast China which is low altitude, and highly urbanized. CONCLUSIONS China should no longer be regarded as a low-risk country for hip fracture. By 2050, the total number of hip fractures in people older than 65 years in China is predicted to be 1.3 million. Higher altitude areas had higher hip fracture rates than lower altitude, higher urbanized areas.
Collapse
Affiliation(s)
- X F Gong
- Department of Orthopaedic Trauma, Beijing Jishuitan Hospital, the 4th Medical College of Peking University, Beijing, China
- Department of Orthopaedic and Trauma, Lhasa People's Hospital, Lhasa, Tibet, China
| | - X P Li
- Department of Geriatrics, Beijing Jishuitan Hospital, the 4th Medical College of Peking University, Beijing, China.
| | - L X Zhang
- Renal Division, Department of Medicine, Peking University First Hospital, Peking University Institute of Nephrology, Beijing, China
| | - J R Center
- Bone and Mineral Research Program, Garvan Institute of Medical Research, St Vincent's Hospital, Sydney, Australia
| | - D Bliuc
- Bone and Mineral Research Program, Garvan Institute of Medical Research, St Vincent's Hospital, Sydney, Australia
| | - Y Shi
- China Standard Medical Information Research Center, Shenzhen, Guangdong, China
| | - H B Wang
- Clinical Trial Unit, First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - L He
- Department of Orthopaedic Trauma, Beijing Jishuitan Hospital, the 4th Medical College of Peking University, Beijing, China
| | - X B Wu
- Department of Orthopaedic Trauma, Beijing Jishuitan Hospital, the 4th Medical College of Peking University, Beijing, China.
| |
Collapse
|
3
|
Zhang X, He J, Wang W. Progress in the use of mesenchymal stromal cells for osteoarthritis treatment. Cytotherapy 2021; 23:459-470. [PMID: 33736933 DOI: 10.1016/j.jcyt.2021.01.008] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2020] [Revised: 12/20/2020] [Accepted: 01/29/2021] [Indexed: 12/26/2022]
Abstract
LITERATURE REVIEW OF MSCS IN THE TREATMENT OF OSTEOARTHRITIS IN THE PAST FIVE YEARS: Osteoarthritis (OA) is one of the most common chronic joint diseases, with prominent symptoms caused by many factors. However, current medical interventions for OA have resulted in poor clinical outcomes, demonstrating that there are huge unmet medical needs in this area. Cell therapy has opened new avenues of OA treatment. Different sources of mesenchymal stromal cells (MSCs) may have different phenotypes and cellular functions. Pre-clinical and clinical studies have demonstrated the feasibility, safety and efficacy of MSC therapy. Mitogen-activated protein kinase, Wnt and Notch signaling pathways are involved in the chondrogenesis of MSC-mediated treatments. MSCs may also exert effective immunoregulatory and paracrine effects to stimulate tissue repair. Therapy with extracellular vesicles containing cytokines, which are secreted by MSCs, might be a potential treatment for OA.
Collapse
Affiliation(s)
- Xiaotian Zhang
- Department of Orthopedics, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Jiyin He
- Department of Orthopedics, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Wen Wang
- Clinical Development, IASO Biotherapeutics Co., Ltd., Shanghai, China.
| |
Collapse
|
4
|
Elzakra N, Kim Y. HIF-1α Metabolic Pathways in Human Cancer. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1280:243-260. [PMID: 33791987 DOI: 10.1007/978-3-030-51652-9_17] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Oxygen is directly involved in many key pathophysiological processes. Oxygen deficiency, also known as hypoxia, could have adverse effects on mammalian cells, with ischemia in vital tissues being the most significant (Michiels C. Physiological and pathological responses to hypoxia. Am J Pathol 164(6): 1875-1882, 2004); therefore, timely adaptive responses to variations in oxygen availability are essential for cellular homeostasis and survival. The most critical molecular event in hypoxic response is the activation and stabilization of a transcriptional factor termed hypoxia-induced factor-1 (HIF-1) that is responsible for the upregulation of many downstream effector genes, collectively known as hypoxia-responsive genes. Multiple key biological pathways such as proliferation, energy metabolism, invasion, and metastasis are governed by these genes; thus, HIF-1-mediated pathways are equally pivotal in both physiology and pathology.As we gain knowledge on the molecular mechanisms underlying the regulation of HIF-1, a great focus has been placed on elucidating the cellular function of HIF-1, particularly the role of HIF-1 in cancer pathogenesis pathways such as proliferation, invasion, angiogenesis, and metastasis. In cancer, HIF-1 is directly involved in the shift of cancer tissues from oxidative phosphorylation to aerobic glycolysis, a phenomenon known as the Warburg effect. Although targeting HIF-1 as a cancer therapy seems like an extremely rational approach, owing to the complex network of its downstream effector genes, the development of specific HIF-1 inhibitors with fewer side effects and more specificity has not been achieved. Therefore, in this review, we provide a brief background about the function of HIF proteins in hypoxia response with a special emphasis on the unique role played by HIF-1α in cancer growth and invasiveness, in the hypoxia response context.
Collapse
Affiliation(s)
- Naseim Elzakra
- School of Dentistry, University of California Los Angeles, Los Angeles, CA, USA.
| | - Yong Kim
- School of Dentistry, University of California Los Angeles, Los Angeles, CA, USA. .,Laboratory of Stem Cell and Cancer Epigenetics, Center for Oral Oncology Research, UCLA School of Dentistry, Los Angeles, CA, USA. .,UCLA's Jonsson Comprehensive Cancer Center, Los Angeles, CA, USA. .,Broad Stem Cell Research Institute, Los Angeles, CA, USA.
| |
Collapse
|
5
|
Culibrk RA, Hahn MS. The Role of Chronic Inflammatory Bone and Joint Disorders in the Pathogenesis and Progression of Alzheimer's Disease. Front Aging Neurosci 2020; 12:583884. [PMID: 33364931 PMCID: PMC7750365 DOI: 10.3389/fnagi.2020.583884] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Accepted: 11/06/2020] [Indexed: 12/12/2022] Open
Abstract
Late-onset Alzheimer's Disease (LOAD) is a devastating neurodegenerative disorder that causes significant cognitive debilitation in tens of millions of patients worldwide. Throughout disease progression, abnormal secretase activity results in the aberrant cleavage and subsequent aggregation of neurotoxic Aβ plaques in the cerebral extracellular space and hyperphosphorylation and destabilization of structural tau proteins surrounding neuronal microtubules. Both pathologies ultimately incite the propagation of a disease-associated subset of microglia-the principle immune cells of the brain-characterized by preferentially pro-inflammatory cytokine secretion and inhibited AD substrate uptake capacity, which further contribute to neuronal degeneration. For decades, chronic neuroinflammation has been identified as one of the cardinal pathophysiological driving features of AD; however, despite a number of works postulating the underlying mechanisms of inflammation-mediated neurodegeneration, its pathogenesis and relation to the inception of cognitive impairment remain obscure. Moreover, the limited clinical success of treatments targeting specific pathological features in the central nervous system (CNS) illustrates the need to investigate alternative, more holistic approaches for ameliorating AD outcomes. Accumulating evidence suggests significant interplay between peripheral immune activity and blood-brain barrier permeability, microglial activation and proliferation, and AD-related cognitive decline. In this work, we review a narrow but significant subset of chronic peripheral inflammatory conditions, describe how these pathologies are associated with the preponderance of neuroinflammation, and posit that we may exploit peripheral immune processes to design interventional, preventative therapies for LOAD. We then provide a comprehensive overview of notable treatment paradigms that have demonstrated considerable merit toward treating these disorders.
Collapse
Affiliation(s)
| | - Mariah S. Hahn
- Department of Biomedical Engineering, Rensselaer Polytechnic Institute, Troy, NY, United States
| |
Collapse
|
6
|
Mohanram Y, Zhang J, Tsiridis E, Yang XB. Comparing bone tissue engineering efficacy of HDPSCs, HBMSCs on 3D biomimetic ABM-P-15 scaffolds in vitro and in vivo. Cytotechnology 2020; 72:715-730. [PMID: 32820463 PMCID: PMC7548016 DOI: 10.1007/s10616-020-00414-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2020] [Accepted: 07/19/2020] [Indexed: 02/06/2023] Open
Abstract
Human bone marrow mesenchymal stem cells (HBMSCs) has been the gold standard for bone regeneration. However, the low proliferation rate and long doubling time limited its clinical applications. This study aims to compare the bone tissue engineering efficacy of human dental pulp stem cells (HDPSCs) with HBMSCs in 2D, and 3D anorganic bone mineral (ABM) coated with a biomimetic collagen peptide (ABM-P-15) for improving bone-forming speed and efficacy in vitro and in vivo. The multipotential of both HDPSCs and HBMSCs have been compared in vitro. The bone formation of HDPSCs on ABM-P-15 was tested using in vivo model. The osteogenic potential of the cells was confirmed by alkaline phosphatase (ALP) and immunohistological staining for osteogenic markers. Enhanced ALP, collagen, lipid droplet, or glycosaminoglycans production were visible in HDPSCs and HBMSCs after osteogenic, adipogenic and chondrogenic induction. HDPSC showed stronger ALP staining compared to HBMSCs. Confocal images showed more viable HDPSCs on both ABM-P-15 and ABM scaffolds compared to HBMSCs on similar scaffolds. ABM-P-15 enhanced cell attachment/spreading/bridging formation on ABM-P-15 scaffolds and significantly increased quantitative ALP specific activities of the HDPSCs and HBMSCs. After 8 weeks in vivo implantation in diffusion chamber model, the HDPSCs on ABM-P-15 scaffolds showed extensive high organised collagenous matrix formation that was positive for COL-I and OCN compared to ABM alone. In conclusion, the HDPSCs have a higher proliferation rate and better osteogenic capacity, which indicated the potential of combining HDPSCs with ABM-P-15 scaffolds for improving bone regeneration speed and efficacy.
Collapse
Affiliation(s)
- Yamuna Mohanram
- Biomaterials & Tissue Engineering Group, Department of Oral Biology, School of Dentistry, University of Leeds, Level 7, Wellcome Trust Brenner Building, St. James's University Hospital, Leeds, LS9 7TF, UK
| | - Jingying Zhang
- Biomaterials & Tissue Engineering Group, Department of Oral Biology, School of Dentistry, University of Leeds, Level 7, Wellcome Trust Brenner Building, St. James's University Hospital, Leeds, LS9 7TF, UK.,The Second Clinical Medical College, Guangdong Medical University, Dongguan, 523808, Guangdong, China
| | - Eleftherios Tsiridis
- Academic Orthopaedic Department, Aristotle University Medical School, 54124, Thessaloniki, Greece
| | - Xuebin B Yang
- Biomaterials & Tissue Engineering Group, Department of Oral Biology, School of Dentistry, University of Leeds, Level 7, Wellcome Trust Brenner Building, St. James's University Hospital, Leeds, LS9 7TF, UK.
| |
Collapse
|
7
|
URDEITX PAU, FARZANEH SOLMAZ, MOUSAVI SJAMALEDDIN, DOWEIDAR MOHAMEDH. ROLE OF OXYGEN CONCENTRATION IN THE OSTEOBLASTS BEHAVIOR: A FINITE ELEMENT MODEL. J MECH MED BIOL 2020. [DOI: 10.1142/s0219519419500647] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Oxygen concentration plays a key role in cell survival and viability. Besides, it has important effects on essential cellular biological processes such as cell migration, differentiation, proliferation and apoptosis. Therefore, the prediction of the cellular response to the alterations of the oxygen concentration can help significantly in the advances of cell culture research. Here, we present a 3D computational mechanotactic model to simulate all the previously mentioned cell processes under different oxygen concentrations. With this model, three cases have been studied. Starting with mesenchymal stem cells within an extracellular matrix with mechanical properties suitable for its differentiation into osteoblasts, and under different oxygen conditions to evaluate their behavior under normoxia, hypoxia and anoxia. The obtained results, which are consistent with the experimental observations, indicate that cells tend to migrate toward zones with higher oxygen concentration where they accelerate their differentiation and proliferation. This technique can be employed to control cell migration toward fracture zones to accelerate the healing process. Besides, as expected, to avoid cell apoptosis under conditions of anoxia and to avoid the inhibition of the differentiation and proliferation processes under conditions of hypoxia, the state of normoxia should be maintained throughout the entire cell-culture process.
Collapse
Affiliation(s)
- PAU URDEITX
- Mechanical Engineering Department, School of Engineering and Architecture (EINA), University of Zaragoza, Zaragoza, Spain
- Aragón Institute of Engineering Research (I3A), University of Zaragoza, Zaragoza, Spain
- Biomedical Research Networking Center in Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Zaragoza, Spain
| | - SOLMAZ FARZANEH
- Mines Saint-Etienne, Univ Lyon, Univ Jean Monnet, INSERM, U 1059, Sainbiose, Centre CIS, F - 42023, Saint-Etienne, France
| | - S. JAMALEDDIN MOUSAVI
- Mines Saint-Etienne, Univ Lyon, Univ Jean Monnet, INSERM, U 1059, Sainbiose, Centre CIS, F - 42023, Saint-Etienne, France
| | - MOHAMED H. DOWEIDAR
- Mechanical Engineering Department, School of Engineering and Architecture (EINA), University of Zaragoza, Zaragoza, Spain
- Aragón Institute of Engineering Research (I3A), University of Zaragoza, Zaragoza, Spain
- Biomedical Research Networking Center in Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Zaragoza, Spain
| |
Collapse
|
8
|
Leslie SK, Cohen DJ, Boyan BD, Schwartz Z. Production of osteogenic and angiogenic factors by microencapsulated adipose stem cells varies with culture conditions. J Biomed Mater Res B Appl Biomater 2019; 108:1857-1867. [PMID: 31872938 DOI: 10.1002/jbm.b.34527] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2019] [Revised: 10/28/2019] [Accepted: 11/16/2019] [Indexed: 01/06/2023]
Abstract
Growth factors produced by stem cells aid in the bone repair process. We investigated the ability of encapsulated rat adipose-derived stem cells (rASCs) treated with osteogenic media (OM) to produce growth factors, and determined the optimal combination of OM components that will lead to the production of both osteogenic and angiogenic factors. Our results demonstrate that microencapsulated stem cells were able to produce vascular endothelial growth factor (VEGF), fibroblast growth factor-2, and bone morphogenetic protein-2 (BMP2) necessary for bone regeneration. OM led to the reduction of angiogenic factors; however, the removal of dexamethasone restored angiogenic factor production. Additionally, we determined whether the effect of dexamethasone on VEGF and BMP2 varied among rat, rabbit, mouse, and humans. Dexamethasone led to a reduction in VEGF levels in ASCs derived from rats, mice, and humans, while this reduction was absent in rabbit ASCs (rbASCs). Human ASCs (hASCs) from donors of different race and sex showed a similar response to dexamethasone with secreted VEGF levels. BMP2 levels secreted by rbASCs, mouse ASCs (mASCs), and hASCs were independent of the media treatments, while rASCs responded differently in the surrounding media and within the microbeads. In conclusion, microencapsulated ASCs can be treated to produce osteogenic and angiogenic factors for tissue regeneration applications, but outcomes may vary with culture conditions.
Collapse
Affiliation(s)
- Shirae K Leslie
- College of Engineering, Virginia Commonwealth University, Richmond, Virginia
| | - David Joshua Cohen
- College of Engineering, Virginia Commonwealth University, Richmond, Virginia
| | - Barbara D Boyan
- College of Engineering, Virginia Commonwealth University, Richmond, Virginia.,Wallace H. Coulter Department of Biomedical Engineering at Georgia Tech and Emory University, Georgia Institute of Technology, Atlanta, Georgia
| | - Zvi Schwartz
- College of Engineering, Virginia Commonwealth University, Richmond, Virginia.,Department of Periodontics, University of Texas Health Science Center at San Antonio, San Antonio, Texas
| |
Collapse
|
9
|
George A, Ellis M, Gill HS. Hypoxia-inducible factor (HIF): how to improve osseointegration in hip arthroplasty secondary to avascular necrosis in sickle cell disease. EFORT Open Rev 2019; 4:567-575. [PMID: 31598335 PMCID: PMC6771077 DOI: 10.1302/2058-5241.4.180030] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Many studies in the literature have been carried out to evaluate the various cellular and molecular processes involved in osteogenesis. Angiogenesis and bone formation work closely together in this group of disorders. Hypoxia-inducible factor (HIF) which is stimulated in tissue hypoxia triggers a cascade of molecular processes that helps manage this physiological deficiency. However, there still remains a paucity of knowledge with regard to how sickle cell bone pathology, in particular avascular necrosis, could be altered when it comes to osseointegration at the molecular level. Hypoxia-inducible factor has been identified as key in mediating how cells adapt to molecular oxygen levels. The aim of this review is to further elucidate the physiology of hypoxia-inducible factor with its various pathways and to establish what role this factor could play in altering the disease pathophysiology of avascular necrosis caused by sickle cell disease and in improving osseointegration. This review article also seeks to propose certain research methodology frameworks in exploring how osseointegration could be improved in sickle cell disease patients with total hip replacements and how it could eventually reduce their already increased risk of undergoing revision surgery.
Cite this article: EFORT Open Rev 2019;4:567-575. DOI: 10.1302/2058-5241.4.180030
Collapse
Affiliation(s)
- Akintunde George
- Centre for Integrated Bioprocessing Research, Department of Chemical Engineering, University of Bath, Bath, UK
| | - Marianne Ellis
- Centre for Integrated Bioprocessing Research, Department of Chemical Engineering, University of Bath, Bath, UK
| | | |
Collapse
|
10
|
Tan L, Zhang Y, Huang Y, Luo Y, Liu Y. Preservation of alveolar ridge after tooth extraction with hypoxia-inducible factor-1α protein in a dog model. Exp Ther Med 2019; 17:2913-2920. [PMID: 30936961 PMCID: PMC6434234 DOI: 10.3892/etm.2019.7301] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2018] [Accepted: 10/26/2018] [Indexed: 12/12/2022] Open
Abstract
Hypoxia-inducible factor (HIF)-1α protein, which is upregulated by hypoxia, serves an important role in angiogenesis during osteogenesis. The aim of the present study was to investigate the effect of HIF-1α on alveolar ridge preservation in a dog tooth extraction model. Six beagle dogs were used in the present study. The second and fourth premolar teeth of the lower jaws on both sides were extracted. Two unilateral extraction sockets were randomly selected and filled with Bio-Oss and Bio-Oss + HIF-1α. The contralateral sockets remained unfilled and served as the negative control. Micro-computed tomography examination and histological staining were performed to examine the difference of new bone formation among the three groups. Western blotting and reverse transcription-quantitative polymerase chain reaction analysis were used to detect the expression levels of osteogenesis- and angiogenesis-associated genes in the bone tissues of the three groups. Twelve weeks post-surgery, trabecular bone formation in the Bio-Oss + HIF-1α group was significantly increased compared with the other groups. The expression levels of osteogenesis-associated genes (runt-related transcription factor 2, osteoblast-specific transcription factor osterix and osteocalcin) and angiogenesis-associated genes (HIF-1α and vascular endothelial growth factor) were all significantly increased in the Bio-Oss + HIF-1α group compared with the other two groups (P<0.05). The present results indicated that Bio-Oss with HIF-1α can promote osteogenesis and angiogenesis in vivo and may be used as an effective treatment for the preservation of the alveolar ridge.
Collapse
Affiliation(s)
- Luanjun Tan
- Department of Stomatology, Shanghai East Hospital Affiliated with Tongji University, Shanghai 200120, P.R. China
| | - Yingdi Zhang
- Department of Stomatology, Shanghai East Hospital Affiliated with Tongji University, Shanghai 200120, P.R. China
| | - Yuanliang Huang
- Department of Stomatology, Shanghai East Hospital Affiliated with Tongji University, Shanghai 200120, P.R. China
| | - Yuan Luo
- Oral Biomedical Engineering Laboratory, Shanghai Stomatological Hospital, Fudan University, Shanghai 200031, P.R. China.,Department of Oral Surgery, Shanghai Stomatological Hospital, Fudan University, Shanghai 200031, P.R. China
| | - Yuehua Liu
- Department of Orthodontics, Shanghai Stomatological Hospital, Fudan University, Shanghai 200031, P.R. China.,Oral Biomedical Engineering Laboratory, Shanghai Stomatological Hospital, Fudan University, Shanghai 200031, P.R. China
| |
Collapse
|
11
|
Petkus DL, Murray-Kolb LE, De Souza MJ. The Unexplored Crossroads of the Female Athlete Triad and Iron Deficiency: A Narrative Review. Sports Med 2018; 47:1721-1737. [PMID: 28290159 DOI: 10.1007/s40279-017-0706-2] [Citation(s) in RCA: 56] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Despite the severity and prevalence of iron deficiency in exercising women, few published reports have explored how iron deficiency interacts with another prevalent and severe condition in exercising women: the 'female athlete triad.' This review aims to describe how iron deficiency may interact with each component of the female athlete triad, that is, energy status, reproductive function, and bone health. The effects of iron deficiency on energy status are discussed in regards to thyroid function, metabolic fuel availability, eating behaviors, and energy expenditure. The interactions between iron deficiency and reproductive function are explored by discussing the potentially impaired fertility and hyperprolactinemia due to iron deficiency and the alterations in iron metabolism due to menstrual blood loss and estrogen exposure. The interaction of iron deficiency with bone health may occur via dysregulation of the growth hormone/insulin-like growth factor-1 axis, hypoxia, and hypothyroidism. Based on these discussions, several future directions for research are presented.
Collapse
Affiliation(s)
- Dylan L Petkus
- Department of Kinesiology, The Pennsylvania State University, 104 Noll Laboratory, University Park, PA, 16802, USA
| | - Laura E Murray-Kolb
- Department of Nutritional Sciences, The Pennsylvania State University, University Park, PA, USA
| | - Mary Jane De Souza
- Department of Kinesiology, The Pennsylvania State University, 104 Noll Laboratory, University Park, PA, 16802, USA.
| |
Collapse
|
12
|
Wang D, Liu Y, Yang X, Zhou J. Hypoxic preconditioning enhances cell hypoxia tolerance and correlated lncRNA and mRNA analysis. Life Sci 2018; 208:46-54. [DOI: 10.1016/j.lfs.2018.07.014] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2018] [Revised: 06/23/2018] [Accepted: 07/07/2018] [Indexed: 01/04/2023]
|
13
|
Aaron RK, Racine JR, Voisinet A, Evangelista P, Dyke JP. Subchondral bone circulation in osteoarthritis of the human knee. Osteoarthritis Cartilage 2018; 26:940-944. [PMID: 29723635 DOI: 10.1016/j.joca.2018.04.003] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/15/2017] [Revised: 03/31/2018] [Accepted: 04/09/2018] [Indexed: 02/02/2023]
Abstract
OBJECTIVE The hypothesis of this study is that human subchondral bone exhibits abnormal patterns of perfusion in osteoarthritis (OA) that can be characterized by kinetic parameters of blood flow using dynamic contrast enhanced (DCE) MRI. DESIGN Fifteen subjects with advanced OA of the knee and seven control subjects without OA were studied at 1.5 T with DCE-MRI. Region of interest (ROIs) analysis of pharmacokinetic perfusion parameters were used to examine initial uptake and washout of the contrast agent in the lateral tibial plateau. RESULTS Arterial and venous perfusion kinetics were abnormal in subchondral OA bone compared to those of normal controls. Time-intensity curves (TIC) exhibited delayed contrast clearance in OA knees compared to normal. Quantitatively, changes were observed in the kinetic parameters, kep, Akep, and kel. The mean kep and Akep were reduced in OA, compared to normal bone, indicating a reduction of arterial inflow and delayed signal enhancement. The kel in OA bone was lower than in normal bone, the negative kel indicating a reduction in venous outflow. The area under the TIC (AUC60) indicated greater residual contrast in OA bone. CONCLUSIONS DCE-MRI can quantitatively assess subchondral bone perfusion kinetics in human OA and identify heterogeneous regions of perfusion deficits. The results are consistent with venous stasis in OA, reflecting venous outflow obstruction, and can affect intraosseous pressure, reduce arterial inflow, reduce oxygen content, and may contribute to altered cell signaling in, and the pathophysiology of, OA.
Collapse
Affiliation(s)
- R K Aaron
- Department of Orthopaedics, Warren Alpert Medical School of Brown University, Providence, RI, USA.
| | - J R Racine
- Department of Orthopaedics, Warren Alpert Medical School of Brown University, Providence, RI, USA.
| | - A Voisinet
- Department of Orthopaedics, Warren Alpert Medical School of Brown University, Providence, RI, USA.
| | - P Evangelista
- Department of Radiology, Warren Alpert Medical School of Brown University, Providence, RI, USA.
| | - J P Dyke
- Citigroup Biomedical Imaging Center, Weill Cornell Medicine, New York, NY, USA; Department of Radiology, Weill Cornell Medicine, New York, NY, USA.
| |
Collapse
|
14
|
Li Y, Wang J, Wang Y, Du W, Wang S. Transplantation of copper-doped calcium polyphosphate scaffolds combined with copper (II) preconditioned bone marrow mesenchymal stem cells for bone defect repair. J Biomater Appl 2018; 32:738-753. [PMID: 29295641 DOI: 10.1177/0885328217739456] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Calcium polyphosphate is a bioactive ceramic that possesses similar mineral components to bone and possess good physicochemical properties. However, pure calcium polyphosphate scaffold is brittle, and it is insufficient in promoting vascularization and osteoinductivity. This study was conducted to assess whether copper (Cu) incorporated into calcium polyphosphate could improve the scaffolds' inherent shortcomings. In the experiments, Cu-calcium polyphosphate scaffolds' mechanical strength, biocompatibility, and biodegradability were researched primarily. And then, hypoxia-inducible factor 1-alpha expression along with angiogenesis and osteogenesis potential when the scaffolds treated with the bone marrow mesenchymal stem cells (BMMSCs) were analyzed in vitro. In in vivo studies, the Cu-calcium polyphosphate scaffolds combined with the liquid extract preconditioned BMMSCs were implanted into animal model to repair the bone defects. Meanwhile, we also evaluate the expression of angiogenic and osteogenic factors. For comparison, Cu-calcium polyphosphate, calcium polyphosphate, and blank control groups were designed. According to the results, proper content of Cu incorporated with calcium polyphosphate (0.1% Cu-calcium polyphosphate) did not significantly change the scaffold's degradation velocity, but it obtained higher compress mechanical strength and Cu-doped scaffolds were less brittle. Besides, these scaffolds incorporated with Cu showed better cytocompatibility and cell proliferation activity. Moreover, after Cu was doped, the hypoxia-inducible factor 1-alpha expression was up-regulated with the angiogenic and osteogenic factors levels increased both in in vitro and in vivo study. The bone defect healing capacity was accessed, using Cu-calcium polyphosphate combined with preconditioned BMMSCs further enhanced new bone formation and improved hypoxia-inducible factor 1-alpha, alkaline phosphatase, osteocalcin, and vascular endothelial growth factor expression. In conclusion, doped Cu into calcium polyphosphate was an alternative strategy for improving calcium polyphosphate's mechanical property and promoting the osteogenesis and angiogenesis potential. Using Cu-calcium polyphosphate scaffolds combined with Cu preconditioned BMMSCs to treat bone defect could enhance bone defect healing.
Collapse
Affiliation(s)
- Yanhong Li
- 74713 Lanzhou University Second Hospital , Lanzhou, P.R. China
| | - Jing Wang
- 74713 Lanzhou University Second Hospital , Lanzhou, P.R. China
| | - Yuliang Wang
- 74713 Lanzhou University Second Hospital , Lanzhou, P.R. China
| | - Wenjia Du
- 74713 Lanzhou University Second Hospital , Lanzhou, P.R. China
| | - Shuanke Wang
- 74713 Lanzhou University Second Hospital , Lanzhou, P.R. China
| |
Collapse
|
15
|
Walters G, Pountos I, Giannoudis PV. The cytokines and micro-environment of fracture haematoma: Current evidence. J Tissue Eng Regen Med 2017; 12:e1662-e1677. [PMID: 29047220 DOI: 10.1002/term.2593] [Citation(s) in RCA: 66] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2017] [Revised: 08/31/2017] [Accepted: 10/09/2017] [Indexed: 12/18/2022]
Abstract
Fracture haematoma formation is the first and foremost important stage of fracture healing. It orchestrates the inflammatory and cellular processes leading to the formation of callus and the restoration of the continuity of the bone. Evidence suggests that blocking this initial stage could lead to an impairment of the overall bone healing process. This review aims to analyse the existing evidence of molecular contributions to bone healing within fracture haematoma and to determine the potential to modify the molecular response to fracture in the haematoma with the aim of improving union times. A comprehensive search of literature documenting fracture haematoma cytokine content was performed. Suitable papers according to prespecified criteria were identified and analysed according to Preferred Reporting Items for Systematic Reviews and Meta-analyses guidelines. A total of 89 manuscripts formed the basis of this analysis. Low oxygen tension, high acidity, and high calcium characterised initially the fracture haematoma micro-environment. In addition, a number of cytokines have been measured with concentrations significantly higher than those found in peripheral circulation. Growth factors have also been isolated, with an observed increase in bone morphogenetic proteins, platelet-derived growth factor, and transforming growth factor. Although molecular modification of fracture haematoma has been attempted, more research is required to determine a suitable biological response modifier leading to therapeutic effects. The cytokine content of fracture haematoma gives insight into processes occurring in the initial stages of fracture healing. Manipulation of signalling molecules represents a promising pathway to target future therapies aiming to upregulate the osteogenesis.
Collapse
Affiliation(s)
- Gavin Walters
- Academic Department of Trauma and Orthopaedics, School of Medicine, University of Leeds, Leeds, UK
| | - Ippokratis Pountos
- Academic Department of Trauma and Orthopaedics, School of Medicine, University of Leeds, Leeds, UK
| | - Peter V Giannoudis
- Academic Department of Trauma and Orthopaedics, School of Medicine, University of Leeds, Leeds, UK
- NIHR Leeds Biomedical Research Unit, Chapel Allerton Hospital, Leeds, UK
| |
Collapse
|
16
|
Contribution of Circulatory Disturbances in Subchondral Bone to the Pathophysiology of Osteoarthritis. Curr Rheumatol Rep 2017; 19:49. [DOI: 10.1007/s11926-017-0660-x] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
|
17
|
Miclau KR, Brazina SA, Bahney CS, Hankenson KD, Hunt TK, Marcucio RS, Miclau T. Stimulating Fracture Healing in Ischemic Environments: Does Oxygen Direct Stem Cell Fate during Fracture Healing? Front Cell Dev Biol 2017; 5:45. [PMID: 28523266 PMCID: PMC5416746 DOI: 10.3389/fcell.2017.00045] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2017] [Accepted: 04/12/2017] [Indexed: 01/27/2023] Open
Abstract
Bone fractures represent an enormous societal and economic burden as one of the most prevalent causes of disability worldwide. Each year, nearly 15 million people are affected by fractures in the United States alone. Data indicate that the blood supply is critical for fracture healing; as data indicate that concomitant bone and vascular injury are major risk factors for non-union. However, the various role(s) that the vasculature plays remains speculative. Fracture stabilization dictates stem cell fate choices during repair. In stabilized fractures stem cells differentiate directly into osteoblasts and heal the injury by intramembranous ossification. In contrast, in non-stable fractures stem cells differentiate into chondrocytes and the bone heals through endochondral ossification, where a cartilage template transforms into bone as the chondrocytes transform into osteoblasts. One suggested role of the vasculature has been to participate in the stem cell fate decisions due to delivery of oxygen. In stable fractures, the blood vessels are thought to remain intact and promote osteogenesis, while in non-stable fractures, continual disruption of the vasculature creates hypoxia that favors formation of cartilage, which is avascular. However, recent data suggests that non-stable fractures are more vascularized than stable fractures, that oxygen does not appear associated with differentiation of stem cells into chondrocytes and osteoblasts, that cartilage is not hypoxic, and that oxygen, not sustained hypoxia, is required for angiogenesis. These unexpected results, which contrast other published studies, are indicative of the need to better understand the complex, spatio-temporal regulation of vascularization and oxygenation in fracture healing. This work has also revealed that oxygen, along with the promotion of angiogenesis, may be novel adjuvants that can stimulate healing in select patient populations.
Collapse
Affiliation(s)
- Katherine R Miclau
- Department of Orthopaedic Surgery, University of CaliforniaSan Francisco, CA, USA.,Zuckerberg San Francisco General Hospital, Orthopaedic Trauma InstituteSan Francisco, CA, USA.,Harvard CollegeCambridge, MA, USA
| | - Sloane A Brazina
- Department of Orthopaedic Surgery, University of CaliforniaSan Francisco, CA, USA.,Zuckerberg San Francisco General Hospital, Orthopaedic Trauma InstituteSan Francisco, CA, USA
| | - Chelsea S Bahney
- Department of Orthopaedic Surgery, University of CaliforniaSan Francisco, CA, USA.,Zuckerberg San Francisco General Hospital, Orthopaedic Trauma InstituteSan Francisco, CA, USA
| | - Kurt D Hankenson
- Department of Small Animal Clinical Science and Department of Physiology, Michigan State UniversityEast Lansing, MI, USA.,Department of Orthopaedic Surgery, University of PennsylvaniaPhiladelphia, PA, USA
| | - Thomas K Hunt
- Department of Surgery, University of CaliforniaSan Francisco, CA, USA
| | - Ralph S Marcucio
- Department of Orthopaedic Surgery, University of CaliforniaSan Francisco, CA, USA.,Zuckerberg San Francisco General Hospital, Orthopaedic Trauma InstituteSan Francisco, CA, USA
| | - Theodore Miclau
- Department of Orthopaedic Surgery, University of CaliforniaSan Francisco, CA, USA.,Zuckerberg San Francisco General Hospital, Orthopaedic Trauma InstituteSan Francisco, CA, USA
| |
Collapse
|
18
|
Li Q, Liu R, Zhao J, Lu Q. N-methyl pyrrolidone (NMP) ameliorates the hypoxia-reduced osteoblast differentiation via inhibiting the NF-κB signaling. J Toxicol Sci 2017; 41:701-9. [PMID: 27665779 DOI: 10.2131/jts.41.701] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
Ischemic-hypoxic condition for local osteoblasts and bone mesenchymal stem cells during bone fracture inhibits bone repairing. N-methyl pyrrolidone (NMP) has been approved as a safe and biologically inactive small chemical molecule, and might be useful for bone fracture repairing. In the present study, we investigated the effect of NMP on the hypoxia-reduced cellular viability and the expression of differentiation-associated markers, such as bone morphogenetic protein 2 (BMP-2), propeptide of type I procollagen I (PINP), alkaline phosphatase (ALP) or runt-related transcription factor 2 (Runx2) in the osteoblasts, and then we examined the molecular mechanism underlining such effect in the human osteoblastic hFOB 1.19 cells. Our results demonstrated that NMP significantly blocked the hypoxia-induced cell viability reduction and inhibited the hypoxia-caused expression downregulation of BMP-2, PINP, ALP and Runx2 in hFOB 1.19 cells. Then we confirmed the involvement of nuclear factor κB (NF-κB) pathway in the regulation by NMP on the hypoxia-mediated the reduction of osteoblast differentiation. The upregulated expression and transcriptional activity of NF-κB, while the downregulated inhibitory κB expression by the hypoxia treatment was reversed by the treatment with 10 mM NMP. In conclusion, our study found a protective role of NMP in osteoblast differentiation in response to hypoxia, and such protection was through inhibiting the NF-κB signaling. This suggests that NMP might be a protective agent in bone fracture repairing.
Collapse
Affiliation(s)
- Qiang Li
- Department of Orthopedics, the Affiliated Hospital of Inner Mongolia Medical University, China
| | | | | | | |
Collapse
|
19
|
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
|
20
|
Qiu Y, Chen Y, Zeng T, Guo W, Zhou W, Yang X. EGCG ameliorates the hypoxia-induced apoptosis and osteogenic differentiation reduction of mesenchymal stem cells via upregulating miR-210. Mol Biol Rep 2016; 43:183-93. [DOI: 10.1007/s11033-015-3936-0] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2015] [Accepted: 12/09/2015] [Indexed: 02/04/2023]
|
21
|
Fan L, Zhang C, Yu Z, Shi Z, Dang X, Wang K. Transplantation of hypoxia preconditioned bone marrow mesenchymal stem cells enhances angiogenesis and osteogenesis in rabbit femoral head osteonecrosis. Bone 2015; 81:544-553. [PMID: 26363339 DOI: 10.1016/j.bone.2015.09.005] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/21/2015] [Revised: 09/06/2015] [Accepted: 09/08/2015] [Indexed: 12/17/2022]
Abstract
PURPOSE Osteonecrosis of the femoral head may be a disease resulting from abnormal proliferation or differentiation of mesenchymal stem cells. The present investigation explored the novel strategy of hypoxia-preconditioned BMMSCs to reverse the impairment of osteonecrosis BMMSCs and enhance the therapeutic potential of hypoxia-treated BMMSC transplantation. METHODS BMMSCs from the anterior superior iliac spine region of osteonecrosis rabbit were cultured under 20% O2 or 2% O2 conditions. Normal BMMSCs were cultured under 20% O2 condition as control. Growth factors secreted were examined by enzyme-linked immunosorbent assay. 20% O2 or 2% O2 BMMSCs were injected into the femoral head of rabbits after core decompression. Cell viability and apoptosis were assessed in vitro, and TUNEL staining of the femoral head was analyzed after transplantation. Angiogenesis (capillary-like structure formation, CD31 immunohistochemical staining and ink infusion angiography) and osteogenesis (Alizarin red-S staining, micro-CT scanning and OCN immunohistochemical staining) tests were conducted as well. RESULTS 2% O2 exposure up-regulated growth factor secretion in BMMSCs. Apoptosis in 2% O2 group was lower when compared with that in 20% O2 osteonecrosis group. Cell viability in 2% O2 was significantly higher when compared with that in 20% O2 osteonecrosis group. Growth factor secretion, cell viability, apoptosis, capillary-like structure formation, Alizarin red-S staining, and ALP staining showed no difference between the 2% O2 BMMSC and normal BMMSC groups. Transplantation of 2% O2 versus 20% O2 mesenchymal stem cells after core decompression resulted in an increase in angiogenesis function and a decrease in local tissue apoptosis. Our study also found that osteogenesis function was improved after hypoxic stem cell transplantation. CONCLUSION Hypoxic preconditioning of BMMSCs is an effective means of reversing the impairment of osteonecrosis BMMSCs, promoting their regenerative capability and therapeutic potential for the treatment of osteonecrosis.
Collapse
Affiliation(s)
- Lihong Fan
- Department of Orthopedics, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710004, China.
| | - Chen Zhang
- Department of Orthopedics, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710004, China.
| | - Zefeng Yu
- Department of Orthopedics, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710004, China.
| | - Zhibin Shi
- Department of Orthopedics, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710004, China.
| | - Xiaoqian Dang
- Department of Orthopedics, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710004, China.
| | - Kunzheng Wang
- Department of Orthopedics, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710004, China.
| |
Collapse
|
22
|
Dyke JP, Synan M, Ezell P, Ballon D, Racine J, Aaron RK. Characterization of bone perfusion by dynamic contrast-enhanced magnetic resonance imaging and positron emission tomography in the Dunkin-Hartley guinea pig model of advanced osteoarthritis. J Orthop Res 2015; 33:366-72. [PMID: 25410523 PMCID: PMC4346481 DOI: 10.1002/jor.22768] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/17/2014] [Accepted: 10/11/2014] [Indexed: 02/04/2023]
Abstract
This study characterizes changes in subchondral bone circulation in OA and examines relationships to bone structure and cartilage degeneration in Dunkin-Hartley guinea pigs. We have used dynamic contrast-enhanced MRI (DCE-MRI) and PET, with pharmacokinetic modeling, to characterize subchondral bone perfusion. Assessments are made of perfusion kinetics and vascular permeability by MRI, and blood volume and flow, and radionuclide incorporation into bone, by PET. These parameters are compared to cartilage lesion severity and bone histomorphometry. Assessments of intraosseous thrombi are made morphologically. Prolonged signal enhancement during the clearance phase of MRI correlated with OA severity and suggested venous stasis. Vascular permeability was not increased indicating that transvascular migration of contrast agent was not responsible for signal enhancement. Intraosseous thrombi were not observed. Decreased perfusion associated with severe OA was confirmed by PET and was associated with reduced radionuclide incorporation and osteoporosis. MRI and PET can be used to characterize kinetic parameters of circulation in OA and correlate them with subchondral bone metabolism of interest to the pathophysiology of OA. The significance of these observations may lie in alterations induced in the expression of cytokines by OA osteoblasts that are related to bone remodeling and cartilage breakdown.
Collapse
Affiliation(s)
- Jonathan P. Dyke
- Department of Radiology, Weill Cornell Medical College, New York, NY,Citigroup Biomedical Imaging Center, Weill Cornell Medical College, New York, NY
| | - Michael Synan
- Citigroup Biomedical Imaging Center, Weill Cornell Medical College, New York, NY
| | - Paula Ezell
- Department of Comparative Medicine, Medical University of South Carolina, Charleston, SC
| | - Douglas Ballon
- Department of Radiology, Weill Cornell Medical College, New York, NY,Citigroup Biomedical Imaging Center, Weill Cornell Medical College, New York, NY
| | - Jennifer Racine
- Department of Orthopedics, The Warren Alpert Medical School of Brown University, Providence, RI
| | - Roy K. Aaron
- Department of Orthopedics, The Warren Alpert Medical School of Brown University, Providence, RI
| |
Collapse
|
23
|
Liu T, Zou W, Shi G, Xu J, Zhang F, Xiao J, Wang Y. Hypoxia-induced MTA1 promotes MC3T3 osteoblast growth but suppresses MC3T3 osteoblast differentiation. Eur J Med Res 2015; 20:10. [PMID: 25644400 PMCID: PMC4324858 DOI: 10.1186/s40001-015-0084-x] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2014] [Accepted: 01/09/2015] [Indexed: 01/27/2023] Open
Abstract
Background Bone fracture is one of the most common physical injuries in which gene expression and the microenvironment are reprogramed to facilitate the recovery process. Methods By specific siRNA transfection and MTT assay, we evaluated the effects of metastasis-associated gene 1 (MTA1) in osteoblast growth. To show the role of MTA1 in osteoblast under hypoxia conditions, by overexpressing and silencing MTA1 expression, we performed mineral deposition and alkaline phosphatase activity assay to observe the differentiation status of osteoblast cells. Real-time PCR and Western blot assays were adopted to detect the expression of certain target genes. Results Here, we reported that hypoxia-induced MTA1 expression through hypoxia-induced factor 1 alpha (HIF-1α) and stimulated the growth of osteoblast MC3T3 cells. Silencing of MTA1 through specific siRNA suppressed MC3T3 cell growth and elicited cell differentiation and induced alkaline phosphatase activation and the upregulation of bone morphogenetic protein-2 and osteocalcin. Conclusions We found that MTA1 was regulated by HIF-1α in hypoxia circumstance to suppress osteoblast differentiation. These findings provide new insights for bone fracture healing and new strategies to develop potential targets to promote fracture healing. Electronic supplementary material The online version of this article (doi:10.1186/s40001-015-0084-x) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Tielong Liu
- Department of Orthopaedics, Shanghai Changzheng Hospital, 415, Fengyang Road, Shanghai, 200003, China.
| | - Weiwei Zou
- Department of Medical Imaging, Shanghai Changzheng Hospital, Shanghai, 200003, China.
| | - Guodong Shi
- Department of Orthopaedics, Shanghai Changzheng Hospital, 415, Fengyang Road, Shanghai, 200003, China.
| | - Jian Xu
- Suqian Worker's Hospital, Suqian, Jiangsu, Province 223800, China.
| | - Fei Zhang
- Center Hospital of Ningbo Development Zone, Ningbo, Zhejiang, Province 315800, China.
| | - Jianru Xiao
- Department of Orthopaedics, Shanghai Changzheng Hospital, 415, Fengyang Road, Shanghai, 200003, China.
| | - Yan Wang
- Department of Orthopaedics, The General Hospital of People's Liberation Army, 28, Fuxing Road, Haidian District, Beijing, 100853, China.
| |
Collapse
|
24
|
Chang J, Jackson SG, Wardale J, Jones SW. Hypoxia modulates the phenotype of osteoblasts isolated from knee osteoarthritis patients, leading to undermineralized bone nodule formation. Arthritis Rheumatol 2014; 66:1789-99. [PMID: 24574272 PMCID: PMC4282481 DOI: 10.1002/art.38403] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2013] [Accepted: 02/06/2014] [Indexed: 02/06/2023]
Abstract
Objective To investigate the role of hypoxia in the pathology of osteoarthritic (OA) bone by exploring its effect on the phenotype of isolated primary osteoblasts from patients with knee OA. Methods OA bone samples were collected at the time of elective joint replacement surgery for knee or hip OA. Normal bone samples were collected postmortem from cadaver donors. Primary osteoblasts were isolated from knee OA bone chips and cultured under normoxic or hypoxic (2% O2) conditions. Alkaline phosphatase activity was quantified using an enzymatic assay, and osteopontin and prostaglandin E2 (PGE2) production was assayed by enzyme-linked immunosorbent assay. Total RNA was extracted from bone and osteoblasts, and gene expression was profiled by quantitative reverse transcription–polymerase chain reaction. Results Human OA bone tissue sections stained positively for carbonic anhydrase IX, a biomarker of hypoxia, and exhibited differential expression of genes that mediate the vasculature and blood coagulation as compared to those found in normal bone. Culture of primary osteoblasts isolated from knee OA bone under hypoxic conditions profoundly affected the osteoblast phenotype, including the expression of genes that mediate bone matrix, bone remodeling, and bone vasculature. Hypoxia also increased the expression of cyclooxygenase 2 and the production of PGE2 by OA osteoblasts. Osteoblast expression of type II collagen α1 chain, angiopoietin-like 4, and insulin-like growth factor binding protein 1 was shown to be mediated by hypoxia-inducible factor 1α. Chronic hypoxia reduced osteoblast- mineralized bone nodule formation. Conclusion These findings demonstrate that hypoxia can induce pathologic changes in osteoblast functionality consistent with an OA phenotype, providing evidence that hypoxia is a key driver of OA pathology.
Collapse
Affiliation(s)
- Joan Chang
- University of Copenhagen, Copenhagen, Denmark
| | | | | | | |
Collapse
|
25
|
Wan L, Zhang F, He Q, Tsang WP, Lu L, Li Q, Wu Z, Qiu G, Zhou G, Wan C. EPO promotes bone repair through enhanced cartilaginous callus formation and angiogenesis. PLoS One 2014; 9:e102010. [PMID: 25003898 PMCID: PMC4087003 DOI: 10.1371/journal.pone.0102010] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2014] [Accepted: 06/13/2014] [Indexed: 12/13/2022] Open
Abstract
Erythropoietin (EPO)/erythropoietin receptor (EPOR) signaling is involved in the development and regeneration of several non-hematopoietic tissues including the skeleton. EPO is identified as a downstream target of the hypoxia inducible factor-α (HIF-α) pathway. It is shown that EPO exerts a positive role in bone repair, however, the underlying cellular and molecular mechanisms remain unclear. In the present study we show that EPO and EPOR are expressed in the proliferating, pre-hypertrophic and hypertrophic zone of the developing mouse growth plates as well as in the cartilaginous callus of the healing bone. The proliferation rate of chondrocytes is increased under EPO treatment, while this effect is decreased following siRNA mediated knockdown of EPOR in chondrocytes. EPO treatment increases biosynthesis of proteoglycan, accompanied by up-regulation of chondrogenic marker genes including SOX9, SOX5, SOX6, collagen type 2, and aggrecan. The effects are inhibited by knockdown of EPOR. Blockage of the endogenous EPO in chondrocytes also impaired the chondrogenic differentiation. In addition, EPO promotes metatarsal endothelial sprouting in vitro. This coincides with the in vivo data that local delivery of EPO increases vascularity at the mid-stage of bone healing (day 14). In a mouse femoral fracture model, EPO promotes cartilaginous callus formation at days 7 and 14, and enhances bone healing at day 28 indexed by improved X-ray score and micro-CT analysis of microstructure of new bone regenerates, which results in improved biomechanical properties. Our results indicate that EPO enhances chondrogenic and angiogenic responses during bone repair. EPO's function on chondrocyte proliferation and differentiation is at least partially mediated by its receptor EPOR. EPO may serve as a therapeutic agent to facilitate skeletal regeneration.
Collapse
Affiliation(s)
- Lin Wan
- Ministry of Education Key Laboratory for Regenerative Medicine, School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Fengjie Zhang
- Ministry of Education Key Laboratory for Regenerative Medicine, School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
- School of Biomedical Sciences Core Laboratory, Shenzhen Research Institute, The Chinese University of Hong Kong, Shenzhen, China
| | - Qiling He
- Departments of Microbiology and Pathology, University of Alabama at Birmingham, Birmingham, Alabama, United States of America
| | - Wing Pui Tsang
- Ministry of Education Key Laboratory for Regenerative Medicine, School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
- School of Biomedical Sciences Core Laboratory, Shenzhen Research Institute, The Chinese University of Hong Kong, Shenzhen, China
| | - Li Lu
- Ministry of Education Key Laboratory for Regenerative Medicine, School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
- Guangdong Key Laboratory of Pharmaceutical Bioactive Substances, New Drug Function Research Center, School of Life Science and Biopharmacy, Guangdong Pharmaceutical University, Guangzhou, China
| | - Qingnan Li
- Guangdong Key Laboratory of Pharmaceutical Bioactive Substances, New Drug Function Research Center, School of Life Science and Biopharmacy, Guangdong Pharmaceutical University, Guangzhou, China
| | - Zhihong Wu
- Department of Orthopaedics, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| | - Guixing Qiu
- Department of Orthopaedics, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| | - Guangqian Zhou
- The Center for Anti-Ageing and Regenerative Medicine, Medical School, Shenzhen University, Shenzhen, China
| | - Chao Wan
- Ministry of Education Key Laboratory for Regenerative Medicine, School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
- School of Biomedical Sciences Core Laboratory, Shenzhen Research Institute, The Chinese University of Hong Kong, Shenzhen, China
- * E-mail:
| |
Collapse
|
26
|
Lee AMC, Morrison JL, Botting KJ, Shandala T, Xian CJ. Effects of Maternal Hypoxia during Pregnancy on Bone Development in Offspring: A Guinea Pig Model. Int J Endocrinol 2014; 2014:916918. [PMID: 24949010 PMCID: PMC4052078 DOI: 10.1155/2014/916918] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/10/2014] [Revised: 04/09/2014] [Accepted: 04/10/2014] [Indexed: 01/21/2023] Open
Abstract
Low birth weight is associated with reduced bone mass and density in adult life. However, effects of maternal hypoxia (MH) on offspring bone development are not known. Objective. The current study investigated the effects of fetal growth restriction induced by MH during the last half of gestation on bone structure and volume in the offspring of the fetus near term and the pup in adolescence. Methods. During 35-62-day gestation (term, 69d), guinea pigs were housed in room air (21% O2; control) or 12% O2 (MH). Offspring femur and tibia were collected at 62d gestation and 120d after birth. Results. MH decreased fetal birth weight but did not affect osteogenic potential pools in the fetal bone marrow. Histological analysis showed no effects of MH on tibial growth plate thickness in either fetal or postnatal offspring, although there was increased VEGF mRNA expression in the growth plate of postnatal offspring. MH did not change primary spongiosa height but lowered collagen-1 mRNA expression in postnatal offspring. There was increased mRNA expression of adipogenesis-related gene (FABP4) in bone from the MH postnatal offspring. Conclusion. MH during late gestation did not change the pool of osteogenic cells before birth or growth plate heights before and after birth. However, MH reduced expression of bone formation marker (collagen-1) and increased expression of fat formation marker (FABP4) in postnatal offspring bone.
Collapse
Affiliation(s)
- Alice M. C. Lee
- Sansom Institute for Health Research, School of Pharmacy and Medical Sciences, University of South Australia, City East Campus, GPO Box 2471, Adelaide, SA 5001, Australia
| | - Janna L. Morrison
- Sansom Institute for Health Research, School of Pharmacy and Medical Sciences, University of South Australia, City East Campus, GPO Box 2471, Adelaide, SA 5001, Australia
| | - Kimberley J. Botting
- Sansom Institute for Health Research, School of Pharmacy and Medical Sciences, University of South Australia, City East Campus, GPO Box 2471, Adelaide, SA 5001, Australia
- Discipline of Physiology, School of Medical Sciences, University of Adelaide, Adelaide, SA 5005, Australia
| | - Tetyana Shandala
- Sansom Institute for Health Research, School of Pharmacy and Medical Sciences, University of South Australia, City East Campus, GPO Box 2471, Adelaide, SA 5001, Australia
| | - Cory J. Xian
- Sansom Institute for Health Research, School of Pharmacy and Medical Sciences, University of South Australia, City East Campus, GPO Box 2471, Adelaide, SA 5001, Australia
- *Cory J. Xian:
| |
Collapse
|
27
|
Wang X, Schröder HC, Feng Q, Draenert F, Müller WEG. The deep-sea natural products, biogenic polyphosphate (Bio-PolyP) and biogenic silica (Bio-Silica), as biomimetic scaffolds for bone tissue engineering: fabrication of a morphogenetically-active polymer. Mar Drugs 2013; 11:718-46. [PMID: 23528950 PMCID: PMC3705367 DOI: 10.3390/md11030718] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2013] [Revised: 02/04/2013] [Accepted: 02/06/2013] [Indexed: 12/12/2022] Open
Abstract
Bone defects in human, caused by fractures/nonunions or trauma, gain increasing impact and have become a medical challenge in the present-day aging population. Frequently, those fractures require surgical intervention which ideally relies on autografts or suboptimally on allografts. Therefore, it is pressing and likewise challenging to develop bone substitution materials to heal bone defects. During the differentiation of osteoblasts from their mesenchymal progenitor/stem cells and of osteoclasts from their hemopoietic precursor cells, a lineage-specific release of growth factors and a trans-lineage homeostatic cross-talk via signaling molecules take place. Hence, the major hurdle is to fabricate a template that is functioning in a way mimicking the morphogenetic, inductive role(s) of the native extracellular matrix. In the last few years, two naturally occurring polymers that are produced by deep-sea sponges, the biogenic polyphosphate (bio-polyP) and biogenic silica (bio-silica) have also been identified as promoting morphogenetic on both osteoblasts and osteoclasts. These polymers elicit cytokines that affect bone mineralization (hydroxyapatite formation). In this manner, bio-silica and bio-polyP cause an increased release of BMP-2, the key mediator activating the anabolic arm of the hydroxyapatite forming cells, and of RANKL. In addition, bio-polyP inhibits the progression of the pre-osteoclasts to functionally active osteoclasts. Based on these findings, new bioinspired strategies for the fabrication of bone biomimetic templates have been developed applying 3D-printing techniques. Finally, a strategy is outlined by which these two morphogenetically active polymers might be used to develop a novel functionally active polymer.
Collapse
Affiliation(s)
- Xiaohong Wang
- ERC Advanced Investigator Grant Research Group at Institute for Physiological Chemistry, University Medical Center of the Johannes Gutenberg University, Duesbergweg 6, D-55128 Mainz, Germany; E-Mail:
- National Research Center for Geoanalysis, Chinese Academy of Geological Sciences, 26 Baiwanzhuang Dajie, 100037 Beijing, China
| | - Heinz C. Schröder
- ERC Advanced Investigator Grant Research Group at Institute for Physiological Chemistry, University Medical Center of the Johannes Gutenberg University, Duesbergweg 6, D-55128 Mainz, Germany; E-Mail:
| | - Qingling Feng
- Department of Materials Science and Engineering, Tsinghua University, 100084 Beijing, China; E-Mail:
| | - Florian Draenert
- Department and Clinic for Oral and Maxillofacial Surgery, Baldingerstraße, D-35033 Marburg, Germany; E-Mail:
| | - Werner E. G. Müller
- ERC Advanced Investigator Grant Research Group at Institute for Physiological Chemistry, University Medical Center of the Johannes Gutenberg University, Duesbergweg 6, D-55128 Mainz, Germany; E-Mail:
| |
Collapse
|
28
|
Inorganic Polymers: Morphogenic Inorganic Biopolymers for Rapid Prototyping Chain. BIOMEDICAL INORGANIC POLYMERS 2013; 54:235-59. [DOI: 10.1007/978-3-642-41004-8_9] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
|
29
|
Pattappa G, Thorpe SD, Jegard NC, Heywood HK, de Bruijn JD, Lee DA. Continuous and uninterrupted oxygen tension influences the colony formation and oxidative metabolism of human mesenchymal stem cells. Tissue Eng Part C Methods 2012; 19:68-79. [PMID: 22731854 DOI: 10.1089/ten.tec.2011.0734] [Citation(s) in RCA: 95] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Mesenchymal stem cells (MSCs) are an attractive cell source for tissue engineering applications due to their multipotentiality and increased expansion potential compared to mature cells. However, the full potential of MSCs for cellular therapies is not realised, due, in part, to premature proliferative senescence and impaired differentiation capacity following expansion under 20% oxygen. Bone marrow MSCs reside under reduced oxygen levels (4%-7% oxygen), thus this study investigates the effects of uninterrupted physiological oxygen tensions (2%, 5%) on MSC expansion and subsequent differentiation. Expansion potential was evaluated from colony formation efficiency, population-doubling rates, and cellular senescence. Colony formation was significantly reduced under 5% oxygen compared to 2% and 20% oxygen. Population-doubling time was initially shorter with 20% oxygen, but subsequently no significant differences in doubling time were detected between the oxygen conditions. MSCs expanded with 20% oxygen contained a greater proportion of senescent cells than those under physiological oxygen levels, indicated by a three to fourfold increase in β-galactosidase staining. This may be related to the approximately twofold enhanced mitochondrial oxygen consumption under this culture condition. Chondrogenic differentiation was achieved following expansion at each oxygen condition. However, osteogenesis was only achieved for cells expanded and differentiated at 20% oxygen, indicated by alkaline phosphatase activity and alizarin red staining. These studies demonstrate that uninterrupted hypoxia may enhance long-term MSC expansion, but results in a population with impaired osteogenic differentiation potential. Thus, novel differentiation conditions are required to enable differentiation to nonchondrogenic lineages using hypoxia-cultured MSCs.
Collapse
Affiliation(s)
- Girish Pattappa
- School of Engineering and Materials Science, Queen Mary University of London, London, United Kingdom
| | | | | | | | | | | |
Collapse
|
30
|
Darrington E, Zhong M, Vo BH, Khan SA. Vascular endothelial growth factor A, secreted in response to transforming growth factor-β1 under hypoxic conditions, induces autocrine effects on migration of prostate cancer cells. Asian J Androl 2012; 14:745-51. [PMID: 22705563 DOI: 10.1038/aja.2011.197] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Hypoxia and transforming growth factor-β1 (TGF-β1) increase vascular endothelial growth factor A (VEGFA) expression in a number of malignancies. This effect of hypoxia and TGF-β1 might be responsible for tumor progression and metastasis of advanced prostate cancer. In the present study, TGF-β1 was shown to induce VEGFA(165) secretion from both normal cell lines (HPV7 and RWPE1) and prostate cancer cell lines (DU145 and PC3). Conversely, hypoxia-stimulated VEGFA(165) secretion was observed only in prostate cancer cell lines. Hypoxia induced TGF-β1 expression in PC3 prostate cancer cells, and the TGF-β type I receptor (ALK5) kinase inhibitor partially blocked hypoxia-mediated VEGFA(165) secretion. This effect of hypoxia provides a novel mechanism to increase VEGFA expression in prostate cancer cells. Although autocrine signaling of VEGFA has been implicated in prostate cancer progression and metastasis, the associated mechanism is poorly characterized. VEGFA activity is mediated via VEGF receptor (VEGFR) 1 (Flt-1) and 2 (Flk-1/KDR). Whereas VEGFR-1 mRNA was detected in normal prostate epithelial cells, VEGFR-2 mRNA and VEGFR protein were expressed only in PC3 cells. VEGFA(165) treatment induced phosphorylation of extracellular signal-regulated kinase 1/2 (ERK1/2) in PC3 cells but not in HPV7 cells, suggesting that the autocrine function of VEGFA may be uniquely associated with prostate cancer. Activation of VEGFR-2 by VEGFA(165) was shown to enhance migration of PC3 cells. A similar effect was also observed with endogenous VEGFA induced by TGF-β1 and hypoxia. These findings illustrate that an autocrine loop of VEGFA via VEGFR-2 is critical for the tumorigenic effects of TGF-β1 and hypoxia on metastatic prostate cancers.
Collapse
Affiliation(s)
- Eric Darrington
- Center for Cancer Research and Therapeutic Development, Clark Atlanta University, Atlanta, GA 30314, USA
| | | | | | | |
Collapse
|
31
|
Low-level laser therapy induces the expressions of BMP-2, osteocalcin, and TGF-β1 in hypoxic-cultured human osteoblasts. Lasers Med Sci 2012; 28:543-50. [PMID: 22552925 DOI: 10.1007/s10103-012-1109-0] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2011] [Accepted: 04/16/2012] [Indexed: 02/04/2023]
Abstract
The aim of this study was to examine the effect of low-level laser therapy (LLLT) on the cell viability and the expression of hypoxia-inducible factor-1s (HIF-1s), bone morphogenic protein-2 (BMP-2), osteocalcin, type I collagen, transforming growth factor-β1 (TGF-β1), and Akt in hypoxic-cultured human osteoblasts. Human fetal osteoblast cells (cell line 1.19) were cultured under 1 % oxygen tension for 72 h. Cell cultures were divided into two groups. At the experimental side, low-level laser (808 nm, GaAlAs diode) was applied at 0, 24, and 48 h. After irradiation, each cell culture was incubated 24 h more under hypoxia. Total energy was 1.2, 2.4, and 3.6 J/cm(2), respectively. Non-irradiated cultures served as controls. Comparisons between the two groups were analyzed by t test; a p value <0.05 was considered statistically significant. Hypoxia resulted in a decrease in the expression of type I collagen, osteocalcin, and TGF-β1 (p < 0.001, p < 0.001, and p < 0.01, respectively). Cell viability and BMP-2 expression were not decreased by hypoxic condition. On the other hand, LLLT on hypoxic-cultured osteoblast promoted the expression of BMP-2, osteocalcin, and TGF-β1 (p < 0.05, p < 0.01, and p < 0.001, respectively). Cell proliferation was also increased time-dependently. However, hypoxia decreased in type I collagen expression (p < 0.001), and LLLT did not affect type I collagen expression in hypoxic-cultured osteoblasts. Furthermore, LLLT inhibited HIF-1 and Akt expression in hypoxic conditioned osteoblasts. We concluded that LLLT induces the expression of BMP-2, osteocalcin, and TGF- β1 in 1 % hypoxic-cultured human osteoblasts.
Collapse
|
32
|
Nicolaije C, Koedam M, van Leeuwen JPTM. Decreased oxygen tension lowers reactive oxygen species and apoptosis and inhibits osteoblast matrix mineralization through changes in early osteoblast differentiation. J Cell Physiol 2012; 227:1309-18. [PMID: 21604266 DOI: 10.1002/jcp.22841] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
Accumulating data show that oxygen tension can have an important effect on cell function and fate. We used the human pre-osteoblastic cell line SV-HFO, which forms a mineralizing extracellular matrix, to study the effect of low oxygen tension (2%) on osteoblast differentiation and mineralization. Mineralization was significantly reduced by 60-70% under 2% oxygen, which was paralleled by lower intracellular levels of reactive oxygen species (ROS) and apoptosis. Following this reduction in ROS the cells switched to a lower level of protection by down-regulating their antioxidant enzyme expression. The downside of this is that it left the cells more vulnerable to a subsequent oxidative challenge. Total collagen content was reduced in the 2% oxygen cultures and expression of matrix genes and matrix-metabolizing enzymes was significantly affected. Alkaline phosphatase activity and RNA expression as well as RUNX2 expression were significantly reduced under 2% oxygen. Time phase studies showed that high oxygen in the first phase of osteoblast differentiation and prior to mineralization is crucial for optimal differentiation and mineralization. Switching to 2% or 20% oxygen only during mineralization phase did not change the eventual level of mineralization. In conclusion, this study shows the significance of oxygen tension for proper osteoblast differentiation, extra cellular matrix (ECM) formation, and eventual mineralization. We demonstrated that the major impact of oxygen tension is in the early phase of osteoblast differentiation. Low oxygen in this phase leaves the cells in a premature differentiation state that cannot provide the correct signals for matrix maturation and mineralization.
Collapse
Affiliation(s)
- Claudia Nicolaije
- Department of Internal Medicine, Erasmus Medical Center, Rotterdam, The Netherlands
| | | | | |
Collapse
|
33
|
Wang Y, Li J, Wang Y, Lei L, Jiang C, An S, Zhan Y, Cheng Q, Zhao Z, Wang J, Jiang L. Effects of hypoxia on osteogenic differentiation of rat bone marrow mesenchymal stem cells. Mol Cell Biochem 2011; 362:25-33. [PMID: 22198287 DOI: 10.1007/s11010-011-1124-7] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2011] [Accepted: 10/12/2011] [Indexed: 02/05/2023]
Abstract
Bone reconstruction is essential in orthodontic treatment that caters to the correction of malocclusion by bone reconstruction. Mesenchymal stem cells (MSCs) have been demonstrated a great potency of osteogenesis. The aim of this study was to investigate the effect of hypoxia on the rat bone marrow MSCs (rBMSCs) in vitro during osteogenesis. In this study, we found that temporary exposure of rBMSCs after osteogenic induction for 7 days to hypoxia (2% oxygen) led to a marked decrease in ALPase activity and the expression of osteocalcin and Runt related transcription factor 2/core binding factor a1 (Runx2/Cbfa1). Meanwhile, we found that exposure to hypoxia led to an early and transient increase in the level of phosphorylated ERK1/2 but had no obvious effects on mitogen-activated protein kinase (p38 MAPK) level. Based on these results, we concluded that hypoxia could inhibit osteogenic differentiation of rBMSCs possibly through MEK-ERK 1/2, while p38 MAPK may not participate in this regulation. Further exploration into the mechanisms of hypoxia on osteogenesis would surely provide reliable evidence for clinical practice.
Collapse
Affiliation(s)
- Yating Wang
- State Key Laboratory of Oral Diseases, West China College of Stomatology, West China Hospital of Stomatology, Sichuan University, Chengdu, People's Republic of China
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Pattappa G, Heywood HK, de Bruijn JD, Lee DA. The metabolism of human mesenchymal stem cells during proliferation and differentiation. J Cell Physiol 2011; 226:2562-70. [PMID: 21792913 DOI: 10.1002/jcp.22605] [Citation(s) in RCA: 227] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Human mesenchymal stem cells (MSCs) reside under hypoxic conditions in vivo, between 4% and 7% oxygen. Differentiation of MSCs under hypoxic conditions results in inhibited osteogenesis, while chondrogenesis is unaffected. The reasons for these results may be associated with the inherent metabolism of the cells. The present investigation measured the oxygen consumption, glucose consumption and lactate production of MSCs during proliferation and subsequent differentiation towards the osteogenic and chondrogenic lineages. MSCs expanded under normoxia had an oxygen consumption rate of ∼98 fmol/cell/h, 75% of which was azide-sensitive, suggesting that these cells derive a significant proportion of ATP from oxidative phosphorylation in addition to glycolysis. By contrast, MSCs differentiated towards the chondrogenic lineage using pellet culture had significantly reduced oxygen consumption after 24 h in culture, falling to ∼12 fmol/cell/h after 21 days, indicating a shift towards a predominantly glycolytic metabolism. By comparison, MSCs retained an oxygen consumption rate of ∼98 fmol/cell/h over 21 days of osteogenic culture conditions, indicating that these cells had a more oxidative energy metabolism than the chondrogenic cultures. In conclusion, osteogenic and chondrogenic MSC cultures appear to adopt the balance of oxidative phosphorylation and glycolysis reported for the respective mature cell phenotypes. The addition of TGF-β to chondrogenic pellet cultures significantly enhanced glycosaminoglycan accumulation, but caused no significant effect on cellular oxygen consumption. Thus, the differences between the energy metabolism of chondrogenic and osteogenic cultures may be associated with the culture conditions and not necessarily their respective differentiation.
Collapse
Affiliation(s)
- Girish Pattappa
- School of Engineering and Materials Science, Queen Mary University of London, London, UK
| | | | | | | |
Collapse
|
35
|
Jonitz A, Lochner K, Lindner T, Hansmann D, Marrot A, Bader R. Oxygen consumption, acidification and migration capacity of human primary osteoblasts within a three-dimensional tantalum scaffold. JOURNAL OF MATERIALS SCIENCE. MATERIALS IN MEDICINE 2011; 22:2089-2095. [PMID: 21744104 DOI: 10.1007/s10856-011-4384-6] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/18/2011] [Accepted: 06/25/2011] [Indexed: 05/31/2023]
Abstract
A major clinical problem within synthetic, large-scaled scaffolds is the insufficient nutrient supply resulting in inhomogeneous cell proliferation and differentiation. The aim of this study was to analyse pH value, oxygen consumption and migration of human osteoblasts within a 3D tantalum scaffold, clinically used for larger bone defects. After 24 h the oxygen concentration within the scaffold decreased significantly and remained low during incubation. Monitoring of the pH value inside the tantalum scaffold showed a slightly acidification under static culture conditions. However, cell migration within the 3D scaffold was detected. Hence, in clinical application it can be assumed that porous tantalum scaffolds can be settled by osteoblasts under critical oxygen and nutrient supply. In general, monitoring of cell migration, oxygen consumption and acidification can be a suitable instrument for creating advanced 3D bone scaffolds.
Collapse
Affiliation(s)
- Anika Jonitz
- Department of Orthopaedics, University of Rostock, Rostock, Germany.
| | | | | | | | | | | |
Collapse
|
36
|
Influence of thrombophlebitis on TGF-β1 and its signaling pathway in the vein wall. Folia Histochem Cytobiol 2011; 48:542-8. [PMID: 21478096 DOI: 10.2478/v10042-010-0041-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Extensive extracellular matrix remodeling of the vein wall is involved in varicose veins pathogenesis. This process is controlled by numerous factors, including peptide growth factors. The aim of the study was to evaluate influence of thrombophlebitis on TGF-β1 and its signaling pathway in the vein wall. TGF-β1 mRNAlevels, growth factor content and its expression were evaluated by RT-PCR, ELISA, and western blot methods, respectively, in the walls of normal veins, varicose veins and varicose veins complicated by thrombophlebitis. Western blot analysis was used to assess TGF-β receptor type II (TGF-β RII) and p-Smad2/3 protein expression in the investigated material. Unchanged mRNA levels of TGF-β1, decreased TGF-β1 content, as well as decreased expression of latent and active forms of TGF-β1 were found in varicose veins. Increased expression of TGF-β RII and p-Smad2/3 were found in varicose veins. Thrombophlebitis led to increased protein expression of the TGF-β1 active form and p-Smad2/3 in the vein wall compared to varicose veins. TGF-β1 may play a role in the disease pathogenesis because of increased expression and activation of its receptor in the wall of varicose veins. Thrombophlebitis accelerates activation of TGF-β1 and activity of its receptor in the varicose vein wall.
Collapse
|
37
|
Tuzlakoglu K, Santos MI, Neves N, Reis RL. Design of nano- and microfiber combined scaffolds by electrospinning of collagen onto starch-based fiber meshes: a man-made equivalent of natural extracellular matrix. Tissue Eng Part A 2010; 17:463-73. [PMID: 20825361 DOI: 10.1089/ten.tea.2010.0178] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Mimicking the structural organization and biologic function of natural extracellular matrix has been one of the main goals of tissue engineering. Nevertheless, the majority of scaffolding materials for bone regeneration highlights biochemical functionality in detriment of mechanical properties. In this work we present a rather innovative construct that combines in the same structure electrospun type I collagen nanofibers with starch-based microfibers. These combined structures were obtained by a two-step methodology and structurally consist in a type I collagen nano-network incorporated on a macro starch-based support. The morphology of the developed structures was assessed by several microscopy techniques and the collagenous nature of the nano-network was confirmed by immunohistochemistry. In addition, and especially regarding the requirements of large bone defects, we also successfully introduced the concept of layer by layer, as a way to produce thicker structures. In an attempt to recreate bone microenvironment, the design and biochemical composition of the combined structures also envisioned bone-forming cells and endothelial cells (ECs). The inclusion of a type I collagen nano-network induced a stretched morphology and improved the metabolic activity of osteoblasts. Regarding ECs, the presence of type I collagen on the combined structures provided adhesive support and obviated the need of precoating with fibronectin. It was also importantly observed that ECs on the nano-network organized into circular structures, a three-dimensional arrangement distinct from that observed for osteoblasts and resembling the microcappillary-like organizations formed during angiogenesis. By providing simultaneously physical and chemical cues for cells, the herein-proposed combined structures hold a great potential in bone regeneration as a man-made equivalent of extracellular matrix.
Collapse
Affiliation(s)
- Kadriye Tuzlakoglu
- 3B's Research Group-Biomaterials, Biodegradables and Biomimetics, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, University of Minho, Guimarães, Portugal.
| | | | | | | |
Collapse
|
38
|
Arnett TR. Acidosis, hypoxia and bone. Arch Biochem Biophys 2010; 503:103-9. [PMID: 20655868 DOI: 10.1016/j.abb.2010.07.021] [Citation(s) in RCA: 186] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2010] [Revised: 07/17/2010] [Accepted: 07/20/2010] [Indexed: 10/19/2022]
Abstract
Bone homeostasis is profoundly affected by local pH and oxygen tension. It has long been recognised that the skeleton contains a large reserve of alkaline mineral (hydroxyapatite), which is ultimately available to neutralise metabolic H(+) if acid-base balance is not maintained within narrow limits. Bone cells are extremely sensitive to the direct effects of pH: acidosis inhibits mineral deposition by osteoblasts but it activates osteoclasts to resorb bone and other mineralised tissues. These reciprocal responses act to maximise the availability of OH(-) ions from hydroxyapatite in solution, where they can buffer excess H(+). The mechanisms by which bone cells sense small pH changes are likely to be complex, involving ion channels and receptors in the cell membrane, as well as direct intracellular effects. The importance of oxygen tension in the skeleton has also long been known. Recent work shows that hypoxia blocks the growth and differentiation of osteoblasts (and thus bone formation), whilst strongly stimulating osteoclast formation (and thus bone resorption). Surprisingly, the resorptive function of osteoclasts is unimpaired in hypoxia. In vivo, tissue hypoxia is usually accompanied by acidosis due to reduced vascular perfusion and increased glycolytic metabolism. Thus, disruption of the blood supply can engender a multiple negative impact on bone via the direct actions of reduced pO(2) and pH on bone cells. These observations may contribute to our understanding of the bone disturbances that occur in numerous settings, including ageing, inflammation, fractures, tumours, anaemias, kidney disease, diabetes, respiratory disease and smoking.
Collapse
Affiliation(s)
- Timothy R Arnett
- Department of Cell & Developmental Biology, University College London, London WC1E 6BT, UK.
| |
Collapse
|
39
|
Santos MI, Reis RL. Vascularization in bone tissue engineering: physiology, current strategies, major hurdles and future challenges. Macromol Biosci 2010; 10:12-27. [PMID: 19688722 DOI: 10.1002/mabi.200900107] [Citation(s) in RCA: 299] [Impact Index Per Article: 21.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
The lack of a functional vascular supply has, to a large extent, hampered the whole range of clinical applications of 'successful' laboratory-based bone tissue engineering strategies. To the present, grafts have been dependent on post-implant vascularization, which jeopardizes graft integration and often leads to its failure. For this reason, the development of strategies that could effectively induce the establishment of a microcirculation in the engineered constructs has become a major goal for the tissue engineering research community. This review addresses the role and importance of the development of a vascular network in bone tissue engineering and provides an overview of the most up to date research efforts to develop such a network.
Collapse
Affiliation(s)
- Marina I Santos
- 3B's Research Group - Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, 4806-909 Taipas, Guimarães, Portugal.
| | | |
Collapse
|
40
|
Volkmer E, Kallukalam BC, Maertz J, Otto S, Drosse I, Polzer H, Bocker W, Stengele M, Docheva D, Mutschler W, Schieker M. Hypoxic preconditioning of human mesenchymal stem cells overcomes hypoxia-induced inhibition of osteogenic differentiation. Tissue Eng Part A 2010; 16:153-64. [PMID: 19642854 DOI: 10.1089/ten.tea.2009.0021] [Citation(s) in RCA: 82] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
Osteogenic differentiation of human mesenchymal stem cells (hMSCs) into osteoblasts is a prerequisite for subsequent bone formation. Numerous studies have explored osteogenic differentiation under standard tissue culture conditions, which usually employ 21% of oxygen. However, bone precursor cells such as hMSCs reside in stem cell niches of low-oxygen atmospheres. Furthermore, they are subjected to low oxygen concentrations when cultured on three-dimensional scaffolds in vitro, and even more so after transplantation when vascularization has yet to be established. Similarly, hMSCs are exposed to low oxygen in the fracture microenvironment following bony injury. Recent studies revealed that hypoxic preconditioning improves cellular engraftment and survival in low-oxygen atmospheres. In our study we investigated the osteogenic differentiation potential of hMSCs under 2% O(2) (hypoxia) in comparison to a standard tissue culture oxygen atmosphere of 21% (normoxia). We assessed the osteogenic differentiation of hMSCs following hypoxic preconditioning to address whether this pretreatment is beneficial for subsequent differentiation processes as well. To validate our findings we carefully characterized the extent of hypoxia exerted and its effect on cell survival and proliferation. We found that hMSCs proliferate better if cultured under 2% of oxygen. We confirmed that osteogenic differentiation of hMSCs is indeed inhibited if osteogenic induction is carried out under constant hypoxia. Finally, we showed for the first time that hypoxic preconditioning of hMSCs prior to osteogenic induction restores osteogenic differentiation of hMSCs under hypoxic conditions. Collectively, our results indicate that maintaining constant levels of oxygen improves the osteogenic potential of hMSCs and suggest that low oxygen concentrations may preserve the stemness of hMSCs. In addition, our data support the hypothesis that if low-oxygen atmospheres are expected at the site of implantation, hypoxic pretreatment may be beneficial for the cells' subsequent in vivo performance.
Collapse
Affiliation(s)
- Elias Volkmer
- Experimental Surgery and Regenerative Medicine, Department of Surgery, Ludwig-Maximilians-University, Munich, Germany
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Pallotta I, Lovett M, Rice W, Kaplan DL, Balduini A. Bone marrow osteoblastic niche: a new model to study physiological regulation of megakaryopoiesis. PLoS One 2009; 4:e8359. [PMID: 20027303 PMCID: PMC2793008 DOI: 10.1371/journal.pone.0008359] [Citation(s) in RCA: 66] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2009] [Accepted: 11/23/2009] [Indexed: 12/20/2022] Open
Abstract
Background The mechanism by which megakaryocytes (Mks) proliferate, differentiate, and release platelets into circulation are not well understood. Growing evidence indicates that a complex regulatory mechanism, involving cellular interactions, composition of the extracellular matrix and physical parameters such as oxygen tension, may contribute to the quiescent or permissive microenvironment related to Mk differentiation and maturation within the bone marrow. Methodology/Principal Findings Differentiating human mesenchymal stem cells (hMSCs) into osteoblasts (hOSTs), we established an in vitro model for the osteoblastic niche. We demonstrated for the first time that the combination of HSCs, Mks and hypoxia sustain and promote bone formation by increasing type I collagen release from hOSTs and enhancing its fibrillar organization, as revealed by second harmonic generation microscopy. Through co-culture, we demonstrated that direct cell-cell contact modulates Mk maturation and differentiation. In particular we showed that low oxygen tension and direct interaction of hematopoietic stem cells (HSCs) with hOSTs inhibits Mk maturation and proplatelet formation (PPF). This regulatory mechanism was dependent on the fibrillar structure of type I collagen released by hOSTs and on the resulting engagement of the alpha2beta1 integrin. In contrast, normoxic conditions and the direct interaction of HSCs with undifferentiated hMSCs promoted Mk maturation and PPF, through a mechanism involving the VCAM-1 pathway. Conclusions/Significance By combining cellular, physical and biochemical parameters, we mimicked an in vitro model of the osteoblastic niche that provides a physiological quiescent microenvironment where Mk differentiation and PPF are prevented. These findings serve as an important step in developing suitable in vitro systems to use for the study and manipulation of Mk differentiation and maturation in both normal and diseased states.
Collapse
Affiliation(s)
- Isabella Pallotta
- Department of Biomedical Engineering, Tufts University, Medford, Massachusetts, United States of America
- Department of Biochemistry, University of Pavia, Pavia, Italy
| | - Michael Lovett
- Department of Biomedical Engineering, Tufts University, Medford, Massachusetts, United States of America
| | - William Rice
- Department of Biomedical Engineering, Tufts University, Medford, Massachusetts, United States of America
| | - David L. Kaplan
- Department of Biomedical Engineering, Tufts University, Medford, Massachusetts, United States of America
- * E-mail: (DLK); (AB)
| | - Alessandra Balduini
- Department of Biomedical Engineering, Tufts University, Medford, Massachusetts, United States of America
- Department of Biochemistry, University of Pavia, Pavia, Italy
- * E-mail: (DLK); (AB)
| |
Collapse
|
42
|
Lee JY, Taub PJ, Wang L, Clark A, Zhu LL, Maharam ER, Leong DJ, Ramcharan M, Li Z, Liu Z, Ma YZ, Sun L, Zaidi M, Majeska RJ, Sun HB. Identification of CITED2 as a negative regulator of fracture healing. Biochem Biophys Res Commun 2009; 387:641-5. [PMID: 19607804 DOI: 10.1016/j.bbrc.2009.07.029] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2009] [Accepted: 07/08/2009] [Indexed: 01/18/2023]
Abstract
The transcription regulator CITED2 (CBP/p300-Interacting-Transactivator-with-ED-rich-tail-2) is known to suppress genes mediating angiogenesis and extracellular matrix (ECM) remodeling. However, it is unclear whether CITED2 has a role in controlling skeletal repair or remodeling. We tested the hypothesis that CITED2 functions in bone fracture healing by suppressing the expression of genes critical to ECM remodeling, angiogenesis and osteogenesis, importantly the matrix metalloproteinases (MMPs). Three hours following mandibular osteotomy or sham surgery of adult rats, osteotomy fronts were harvested and the expression of CITED2 and genes associated with fracture healing was ascertained by quantitative PCR. In parallel, gain-of-function studies examined the effect of overexpressing CITED2 on the expression and activity of several MMPs. In the fractured mandible, CITED2 expression was inversely related to the expression of MMP-2, -3, -9, -13, VEGF, HIF-1alpha, M-CSF, RANK-L, and OPG. Consistent with this, the over-expression of CITED2 in osteoblasts inhibited the expression and activity of MMP-2, -3, -9, and -13. Taken together, the studies suggest that CITED2 is a critical upstream regulator of fracture healing. The suppression of CITED2 early after fracture may allow an optimal initiation of the healing response.
Collapse
Affiliation(s)
- Jonathan Y Lee
- Leni and Peter W. May Department of Orthopaedics, Mount Sinai School of Medicine, NY, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Oliveira SM, Amaral IF, Barbosa MA, Teixeira CC. Engineering endochondral bone: in vitro studies. Tissue Eng Part A 2009; 15:625-34. [PMID: 18759672 PMCID: PMC2744199 DOI: 10.1089/ten.tea.2008.0051] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2008] [Accepted: 06/06/2008] [Indexed: 11/12/2022] Open
Abstract
Chitosan scaffolds have been shown to possess biological and mechanical properties suitable for tissue engineering and clinical applications. In the present work, chitosan sponges were evaluated regarding their ability to support cartilage cell proliferation and maturation, which are the first steps in endochondral bone formation. Chitosan sponges were seeded with chondrocytes isolated from chicken embryo sterna. Chondrocyte/chitosan constructs were cultured for 20 days, and treated with retinoic acid (RA) to induce chondrocyte maturation and matrix synthesis. At different time points, samples were collected for microscopic, histological, biochemical, and mechanical analyses. Results show chondrocyte attachment, proliferation, and abundant matrix synthesis, completely obliterating the pores of the sponges. RA treatment caused chondrocyte hypertrophy, characterized by the presence of type X collagen in the extracellular matrix and increased alkaline phosphatase activity. In addition, hypertrophy markedly changed the mechanical properties of the chondrocyte/chitosan constructs. In conclusion, we have developed chitosan sponges with adequate pore structure and mechanical properties to serve as a support for hypertrophic chondrocytes. In parallel studies, we have evaluated the ability of this mature cartilage scaffold to induce endochondral ossification.
Collapse
Affiliation(s)
- Serafim M. Oliveira
- Department of Mechanical Engineering, ESTV—Escola Superior de Tecnologia de Viseu, Viseu, Portugal
- Divisão de Biomateriais, INEB—Instituto de Engenharia Biomédica, Porto, Portugal
- Department of Metallurgy and Materials, FEUP—Faculdade de Engenharia da Universidade do Porto, Porto, Portugal
- Department of Basic Science and Craniofacial Biology, New York University College of Dentistry, New York, New York
| | - Isabel F. Amaral
- Divisão de Biomateriais, INEB—Instituto de Engenharia Biomédica, Porto, Portugal
| | - Mário A. Barbosa
- Divisão de Biomateriais, INEB—Instituto de Engenharia Biomédica, Porto, Portugal
- Department of Metallurgy and Materials, FEUP—Faculdade de Engenharia da Universidade do Porto, Porto, Portugal
| | - Cristina C. Teixeira
- Department of Basic Science and Craniofacial Biology, New York University College of Dentistry, New York, New York
| |
Collapse
|
44
|
Kaneko K, Furuyama K, Aburatani H, Shibahara S. Hypoxia induces erythroid-specific 5-aminolevulinate synthase expression in human erythroid cells through transforming growth factor-β signaling. FEBS J 2009; 276:1370-82. [DOI: 10.1111/j.1742-4658.2009.06878.x] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
45
|
Potier E, Ferreira E, Dennler S, Mauviel A, Oudina K, Logeart-Avramoglou D, Petite H. Desferrioxamine-driven upregulation of angiogenic factor expression by human bone marrow stromal cells. J Tissue Eng Regen Med 2008; 2:272-8. [PMID: 18512268 DOI: 10.1002/term.92] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Bone marrow stromal cells (BMSCs) are the subject of intense research because of their biological properties and potential use for the repair of damaged tissues. Success of BMSC-based therapies, however, relies on a number of methodological improvements, including the establishment of a vascular network providing nutrients and oxygen to the transplanted cells and ensuring their immediate survival and long-term functionality. We described a method to enhance the autocrine expression of angiogenic factors by BMSCs. For this purpose, human BMSCs were treated with desferrioxamine (DFX). No PDGF-BB, VEGF-R1 or -R2 mRNA expression was detected under any of the conditions tested. mRNA and protein expression levels of TGFbeta1 were similar in BMSCs, whether they were exposed to DFX (50 microM) or to control conditions under normoxia for 48 h. In comparison with the results obtained with control conditions under normoxia, exposure of BMSCs to DFX for 48 h resulted in upregulation of bFGF at the protein (26-fold) but not at the mRNA levels and VEGF at both the mRNA (1.5-fold) and protein levels (4.5-fold). In comparison with the results obtained with control conditions under hypoxia, DFX induced a 50% increase in VEGF secretion but led to the same level of hypoxia inducible factor-1alpha protein expression (a transduction factor involved in angiogenic factor expression and known to be activated by DFX). Exposure of BMSCs to DFX resulted in oversecretion of angiogenic factors, suggesting that DFX-treated BMSCs could be used to supply angiogenic factors.
Collapse
Affiliation(s)
- Esther Potier
- Université Denis Diderot Paris VII, Laboratoire de Recherches Orthopédiques (B2OA), UMR CNRS 7052, Paris, France.
| | | | | | | | | | | | | |
Collapse
|
46
|
Metabolic and Functional Characterization of Human Adipose-Derived Stem Cells in Tissue Engineering. Plast Reconstr Surg 2008; 122:725-738. [DOI: 10.1097/prs.0b013e318180ec9f] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
|
47
|
Hyperbaric Oxygen Inhibits Growth But Not Differentiation of Normal and Irradiated Osteoblasts. J Craniofac Surg 2008; 19:757-65. [DOI: 10.1097/scs.0b013e31816aac19] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
|
48
|
Immunohistochemical and in-situ hybridization study of hypoxia inducible factor-1alpha and angiopoietin-1 in a rabbit model of mandibular distraction osteogenesis. Int J Oral Maxillofac Surg 2008; 37:554-60. [PMID: 18339517 DOI: 10.1016/j.ijom.2008.01.015] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2007] [Revised: 11/13/2007] [Accepted: 01/23/2008] [Indexed: 11/20/2022]
Abstract
The endogenous release of angiogenic factors in a rabbit mandibular distraction osteogenesis model was investigated. The spatial and temporal expression of hypoxia inducible factor-1alpha (HIF-1alpha) and angiopoietin-1 (Ang-1) was compared at different phases. The lengthened calluses were harvested on post-osteotomy days 13, 20, 34 and 48, and then stained with haematoxylin & eosin. Immunohistochemical (IHC) and in-situ hybridization (ISH) examination of HIF-1alpha and Ang-1 staining was performed. The ossification in the distracted gap was predominantly intramembranous and slightly endochondral. Expression of HIF-1alpha and Ang-1 was mainly detected in the cytoplasm of fibroblast-like cells, osteoblasts and immature osteocytes on day 13 and 20, but declined with bone maturation. HIF-1alpha was also detected in the nuclei of some osteoblasts. These results suggest that the production of HIF-1alpha and Ang-1 in the distracted gap may contribute to new bone formation during gradual distraction of the mandible.
Collapse
|
49
|
Hiraga T, Kizaka-Kondoh S, Hirota K, Hiraoka M, Yoneda T. Hypoxia and hypoxia-inducible factor-1 expression enhance osteolytic bone metastases of breast cancer. Cancer Res 2007; 67:4157-63. [PMID: 17483326 DOI: 10.1158/0008-5472.can-06-2355] [Citation(s) in RCA: 176] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Hypoxia is a common feature of solid tumors and is associated with their malignant phenotype. The transcription factor hypoxia-inducible factor-1 (HIF-1) is a major regulator of adaptation to hypoxia and is implicated in the malignant progression of cancers. Here, we studied whether hypoxia and HIF-1 expression contribute to the development of bone metastases using a well-characterized animal model of bone metastasis in MDA-MB-231 human breast cancer cells. To study the role of hypoxia in bone metastases, we tested the effects of the fusion protein (TOP3), the oxygen-dependent degradation domain of HIF-1alpha fused with HIV-TAT, and procaspase-3. TOP3 selectively induced apoptosis in hypoxic tumor cells in vitro and significantly reduced bone metastases in vivo. We next examined the role of HIF-1 in bone metastases by establishing MDA-MB-231 cells overexpressing constitutively active or dominant-negative HIF-1alpha (MDA/CA-HIF or MDA/DN-HIF, respectively). Bone metastases of MDA/CA-HIF were significantly increased with elevated number of CD31-positive blood vessels. In contrast, bone metastases were significantly reduced in MDA/DN-HIF. Because the progression of osteolytic bone metastases is due in part to the imbalance between bone formation and bone resorption, we examined the effects of hypoxia and HIF-1 on the differentiation of osteoblasts and osteoclasts. Hypoxia and CA-HIF overexpression markedly inhibited osteoblastic differentiation, whereas hypoxia increased osteoclast-like cell formation. In conclusion, these results suggest that tumor-associated hypoxia and HIF-1 expression promote the progression of bone metastases in breast cancer. Our results also suggest that hypoxia and HIF-1 lead to the development of osteolytic bone metastases by suppressing osteoblast differentiation and promoting osteoclastogenesis.
Collapse
Affiliation(s)
- Toru Hiraga
- Department of Biochemistry, Graduate School of Dentistry, Osaka University, Suita, Osaka, Japan
| | | | | | | | | |
Collapse
|
50
|
Potier E, Ferreira E, Andriamanalijaona R, Pujol JP, Oudina K, Logeart-Avramoglou D, Petite H. Hypoxia affects mesenchymal stromal cell osteogenic differentiation and angiogenic factor expression. Bone 2007; 40:1078-87. [PMID: 17276151 DOI: 10.1016/j.bone.2006.11.024] [Citation(s) in RCA: 223] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/14/2006] [Revised: 11/17/2006] [Accepted: 11/22/2006] [Indexed: 01/02/2023]
Abstract
Mesenchymal stromal cells (MSCs) seeded onto biocompatible scaffolds have been proposed for repairing bone defects. When transplanted in vivo, MSCs (expanded in vitro in 21% O(2)) undergo temporary oxygen deprivation due to the lack of pre-existing blood vessels within these scaffolds. In the present study, the effects of temporary (48 h) exposure to hypoxia (<or=1% O(2)) on primary human MSC survival and osteogenic potential were investigated. Temporary exposure of MSCs to hypoxia had no effect on MSC survival, but resulted in (i) persistent (up to 14 days post exposure) down-regulation of cbfa-1/Runx2, osteocalcin and type I collagen and (ii) permanent (up to 28 days post exposure) up-regulation of osteopontin mRNA expressions. Since angiogenesis is known to contribute crucially to alleviating hypoxia, the effects of temporary hypoxia on angiogenic factor expression by MSCs were also assessed. Temporary hypoxia led to a 2-fold increase in VEGF expression at both the mRNA and protein levels. Other growth factors and cytokines secreted by MSCs under control conditions (namely bFGF, TGFbeta1 and IL-8) were not affected by temporary exposure to hypoxia. All in all, these results indicate that temporary exposure of MSCs to hypoxia leads to limited stimulation of angiogenic factor secretion but to persistent down-regulation of several osteoblastic markers, which suggests that exposure of MSCs transplanted in vivo to hypoxia may affect their bone forming potential. These findings prompt for the development of appropriate cell culture or in vivo transplantation conditions preserving the full osteogenic potential of MSCs.
Collapse
Affiliation(s)
- Esther Potier
- Laboratoire de Recherches Orthopédiques (B2OA), UMR CNRS 7052, Faculté de Médecine Lariboisière-Saint-Louis, 10 Avenue de Verdun, 75010 Paris, France
| | | | | | | | | | | | | |
Collapse
|