1
|
Tao H, Zhu P, Xia W, Chu M, Chen K, Wang Q, Gu Y, Lu X, Bai J, Geng D. The Emerging Role of the Mitochondrial Respiratory Chain in Skeletal Aging. Aging Dis 2024; 15:1784-1812. [PMID: 37815897 PMCID: PMC11272194 DOI: 10.14336/ad.2023.0924] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2023] [Accepted: 09/24/2023] [Indexed: 10/12/2023] Open
Abstract
Maintenance of mitochondrial homeostasis is crucial for ensuring healthy mitochondria and normal cellular function. This process is primarily responsible for regulating processes that include mitochondrial OXPHOS, which generates ATP, as well as mitochondrial oxidative stress, apoptosis, calcium homeostasis, and mitophagy. Bone mesenchymal stem cells express factors that aid in bone formation and vascular growth. Positive regulation of hematopoietic stem cells in the bone marrow affects the differentiation of osteoclasts. Furthermore, the metabolic regulation of cells that play fundamental roles in various regions of the bone, as well as interactions within the bone microenvironment, actively participates in regulating bone integrity and aging. The maintenance of cellular homeostasis is dependent on the regulation of intracellular organelles, thus understanding the impact of mitochondrial functional changes on overall bone metabolism is crucially important. Recent studies have revealed that mitochondrial homeostasis can lead to morphological and functional abnormalities in senescent cells, particularly in the context of bone diseases. Mitochondrial dysfunction in skeletal diseases results in abnormal metabolism of bone-associated cells and a secondary dysregulated microenvironment within bone tissue. This imbalance in the oxidative system and immune disruption in the bone microenvironment ultimately leads to bone dysplasia. In this review, we examine the latest developments in mitochondrial respiratory chain regulation and its impacts on maintenance of bone health. Specifically, we explored whether enhancing mitochondrial function can reduce the occurrence of bone cell deterioration and improve bone metabolism. These findings offer prospects for developing bone remodeling biology strategies to treat age-related degenerative diseases.
Collapse
Affiliation(s)
- Huaqiang Tao
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Jiangsu, China.
| | - Pengfei Zhu
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Jiangsu, China.
| | - Wenyu Xia
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Jiangsu, China.
| | - Miao Chu
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Jiangsu, China.
| | - Kai Chen
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Jiangsu, China.
| | - Qiufei Wang
- Department of Orthopedics, Changshu Hospital Affiliated to Soochow University, First People’s Hospital of Changshu City, Jiangsu, China.
| | - Ye Gu
- Department of Orthopedics, Changshu Hospital Affiliated to Soochow University, First People’s Hospital of Changshu City, Jiangsu, China.
| | - Xiaomin Lu
- Department of Oncology, Affiliated Haian Hospital of Nantong University, Jiangsu, China.
| | - Jiaxiang Bai
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Jiangsu, China.
- Department of Orthopedics, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Anhui, China.
| | - Dechun Geng
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Jiangsu, China.
| |
Collapse
|
2
|
Giordo R, Tulasigeri Totiger S, Caggiari G, Cossu A, Manunta AF, Posadino AM, Pintus G. Protective Effect of Knee Postoperative Fluid on Oxidative-Induced Damage in Human Knee Articular Chondrocytes. Antioxidants (Basel) 2024; 13:188. [PMID: 38397786 PMCID: PMC10886415 DOI: 10.3390/antiox13020188] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Revised: 01/15/2024] [Accepted: 01/30/2024] [Indexed: 02/25/2024] Open
Abstract
The oxidative-stress-elicited deterioration of chondrocyte function is the initial stage of changes leading to the disruption of cartilage homeostasis. These changes entail a series of catabolic damages mediated by proinflammatory cytokines, MMPs, and aggrecanases, which increase ROS generation. Such uncontrolled ROS production, inadequately balanced by the cellular antioxidant capacity, eventually contributes to the development and progression of chondropathies. Several pieces of evidence show that different growth factors, single or combined, as well as anti-inflammatory cytokines and chemokines, can stimulate chondrogenesis and improve cartilage repair and regeneration. In this view, hypothesizing a potential growth-factor-associated action, we investigate the possible protective effect of post-operation knee fluid from patients undergoing prosthesis replacement surgery against ROS-induced damage on normal human knee articular chondrocytes (HKACs). To this end, HKACs were pre-treated with post-operation knee fluid and then exposed to H2O2 to mimic oxidative stress. Intracellular ROS levels were measured by using the molecular probe H2DCFDA; cytosolic and mitochondrial oxidative status were assessed by using HKACs infected with lentiviral particles harboring the redox-sensing green fluorescent protein (roGFP); and cell proliferation was determined by measuring the rate of DNA synthesis with BrdU incorporation. Moreover, superoxide dismutase (SOD), catalase, and glutathione levels from the cell lysates of treated cells were also measured. Postoperative peripheral blood sera from the same patients were used as controls. Our study shows that post-operation knee fluid can counteract H2O2-elicited oxidative stress by decreasing the intracellular ROS levels, preserving the cytosolic and mitochondrial redox status, maintaining the proliferation of oxidatively stressed HKACs, and upregulating chondrocyte antioxidant defense. Overall, our results support and propose an important effect of post-operation knee fluid substances in maintaining HKAC function by mediating cell antioxidative system upregulation and protecting cells from oxidative stress.
Collapse
Affiliation(s)
- Roberta Giordo
- Department of Biomedical Sciences, University of Sassari, Viale San Pietro 43/B, 07100 Sassari, Italy; (R.G.); (S.T.T.); (A.C.)
| | - Smitha Tulasigeri Totiger
- Department of Biomedical Sciences, University of Sassari, Viale San Pietro 43/B, 07100 Sassari, Italy; (R.G.); (S.T.T.); (A.C.)
| | - Gianfilippo Caggiari
- Orthopaedic and Traumatology Department, University Hospital, University of Sassari, Viale San Pietro 43/B, 07100 Sassari, Italy; (G.C.); (A.F.M.)
| | - Annalisa Cossu
- Department of Biomedical Sciences, University of Sassari, Viale San Pietro 43/B, 07100 Sassari, Italy; (R.G.); (S.T.T.); (A.C.)
| | - Andrea Fabio Manunta
- Orthopaedic and Traumatology Department, University Hospital, University of Sassari, Viale San Pietro 43/B, 07100 Sassari, Italy; (G.C.); (A.F.M.)
| | - Anna Maria Posadino
- Department of Biomedical Sciences, University of Sassari, Viale San Pietro 43/B, 07100 Sassari, Italy; (R.G.); (S.T.T.); (A.C.)
| | - Gianfranco Pintus
- Department of Biomedical Sciences, University of Sassari, Viale San Pietro 43/B, 07100 Sassari, Italy; (R.G.); (S.T.T.); (A.C.)
- Department of Medical Laboratory Sciences, College of Health Sciences, Sharjah Institute for Medical Research, University of Sharjah, Sharjah 27272, United Arab Emirates
| |
Collapse
|
3
|
Pinto-Cardoso R, Bessa-Andrês C, Correia-de-Sá P, Bernardo Noronha-Matos J. Could hypoxia rehabilitate the osteochondral diseased interface? Lessons from the interplay of hypoxia and purinergic signals elsewhere. Biochem Pharmacol 2023:115646. [PMID: 37321413 DOI: 10.1016/j.bcp.2023.115646] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2023] [Revised: 06/03/2023] [Accepted: 06/07/2023] [Indexed: 06/17/2023]
Abstract
The osteochondral unit comprises the articular cartilage (90%), subchondral bone (5%) and calcified cartilage (5%). All cells present at the osteochondral unit that is ultimately responsible for matrix production and osteochondral homeostasis, such as chondrocytes, osteoblasts, osteoclasts and osteocytes, can release adenine and/or uracil nucleotides to the local microenvironment. Nucleotides are released by these cells either constitutively or upon plasma membrane damage, mechanical stress or hypoxia conditions. Once in the extracellular space, endogenously released nucleotides can activate membrane-bound purinoceptors. Activation of these receptors is fine-tuning regulated by nucleotides' breakdown by enzymes of the ecto-nucleotidase cascade. Depending on the pathophysiological conditions, both the avascular cartilage and the subchondral bone subsist to significant changes in oxygen tension, which has a tremendous impact on tissue homeostasis. Cell stress due to hypoxic conditions directly influences the expression and activity of several purinergic signalling players, namely nucleotide release channels (e.g. Cx43), NTPDase enzymes and purinoceptors. This review gathers experimental evidence concerning the interplay between hypoxia and the purinergic signalling cascade contributing to osteochondral unit homeostasis. Reporting deviations to this relationship resulting from pathological alterations of articular joints may ultimately unravel novel therapeutic targets for osteochondral rehabilitation. At this point, one can only hypothesize how hypoxia mimetic conditions can be beneficial to the ex vivo expansion and differentiation of osteo- and chondro-progenitors for auto-transplantation and tissue regenerative purposes.
Collapse
Affiliation(s)
- Rui Pinto-Cardoso
- Laboratório de Farmacologia e Neurobiologia; Center for Drug Discovery and Innovative Medicines (MedInUP), Departamento de Imuno-Fisiologia e Farmacologia, Instituto de Ciências Biomédicas Abel Salazar - Universidade do Porto (ICBAS-UP)
| | - Catarina Bessa-Andrês
- Laboratório de Farmacologia e Neurobiologia; Center for Drug Discovery and Innovative Medicines (MedInUP), Departamento de Imuno-Fisiologia e Farmacologia, Instituto de Ciências Biomédicas Abel Salazar - Universidade do Porto (ICBAS-UP)
| | - Paulo Correia-de-Sá
- Laboratório de Farmacologia e Neurobiologia; Center for Drug Discovery and Innovative Medicines (MedInUP), Departamento de Imuno-Fisiologia e Farmacologia, Instituto de Ciências Biomédicas Abel Salazar - Universidade do Porto (ICBAS-UP)
| | - José Bernardo Noronha-Matos
- Laboratório de Farmacologia e Neurobiologia; Center for Drug Discovery and Innovative Medicines (MedInUP), Departamento de Imuno-Fisiologia e Farmacologia, Instituto de Ciências Biomédicas Abel Salazar - Universidade do Porto (ICBAS-UP).
| |
Collapse
|
4
|
Donnenfield JI, Proffen BL, Fleming BC, Murray MM. Responding to ACL Injury and its Treatments: Comparative Gene Expression between Articular Cartilage and Synovium. Bioengineering (Basel) 2023; 10:527. [PMID: 37237597 PMCID: PMC10215325 DOI: 10.3390/bioengineering10050527] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Revised: 04/20/2023] [Accepted: 04/24/2023] [Indexed: 05/28/2023] Open
Abstract
The relationship between cartilage and synovium is a rapidly growing area of osteoarthritis research. However, to the best of our knowledge, the relationships in gene expression between these two tissues have not been explored in mid-stage disease development. The current study compared the transcriptomes of these two tissues in a large animal model one year following posttraumatic osteoarthritis induction and multiple surgical treatment modalities. Thirty-six Yucatan minipigs underwent transection of the anterior cruciate ligament. Subjects were randomized to no further intervention, ligament reconstruction, or ligament repair augmented with an extracellular matrix (ECM) scaffold, followed by RNA sequencing of the articular cartilage and synovium at 52 weeks after harvest. Twelve intact contralateral knees served as controls. Across all treatment modalities, the primary difference in the transcriptomes was that the articular cartilage had greater upregulation of genes related to immune activation compared to the synovium-once baseline differences between cartilage and synovium were adjusted for. Oppositely, synovium featured greater upregulation of genes related to Wnt signaling compared to articular cartilage. After adjusting for expression differences between cartilage and synovium seen following ligament reconstruction, ligament repair with an ECM scaffold upregulated pathways related to ion homeostasis, tissue remodeling, and collagen catabolism in cartilage relative to synovium. These findings implicate inflammatory pathways within cartilage in the mid-stage development of posttraumatic osteoarthritis, independent of surgical treatment. Moreover, use of an ECM scaffold may exert a chondroprotective effect over gold-standard reconstruction through preferentially activating ion homeostatic and tissue remodeling pathways within cartilage.
Collapse
Affiliation(s)
- Jonah I. Donnenfield
- Division of Sports Medicine, Department of Orthopaedic Surgery, Boston Children’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Benedikt L. Proffen
- Division of Sports Medicine, Department of Orthopaedic Surgery, Boston Children’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Braden C. Fleming
- Department of Orthopaedics, Warren Alpert Medical School of Brown University/Rhode Island Hospital, Providence, RI 02903, USA
| | - Martha M. Murray
- Division of Sports Medicine, Department of Orthopaedic Surgery, Boston Children’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| |
Collapse
|
5
|
Zeng CY, Wang XF, Hua FZ. HIF-1α in Osteoarthritis: From Pathogenesis to Therapeutic Implications. Front Pharmacol 2022; 13:927126. [PMID: 35865944 PMCID: PMC9294386 DOI: 10.3389/fphar.2022.927126] [Citation(s) in RCA: 33] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2022] [Accepted: 06/10/2022] [Indexed: 11/13/2022] Open
Abstract
Osteoarthritis is a common age-related joint degenerative disease. Pain, swelling, brief morning stiffness, and functional limitations are its main characteristics. There are still no well-established strategies to cure osteoarthritis. Therefore, better clarification of mechanisms associated with the onset and progression of osteoarthritis is critical to provide a theoretical basis for the establishment of novel preventive and therapeutic strategies. Chondrocytes exist in a hypoxic environment, and HIF-1α plays a vital role in regulating hypoxic response. HIF-1α responds to cellular oxygenation decreases in tissue regulating survival and growth arrest of chondrocytes. The activation of HIF-1α could regulate autophagy and apoptosis of chondrocytes, decrease inflammatory cytokine synthesis, and regulate the chondrocyte extracellular matrix environment. Moreover, it could maintain the chondrogenic phenotype that regulates glycolysis and the mitochondrial function of osteoarthritis, resulting in a denser collagen matrix that delays cartilage degradation. Thus, HIF-1α is likely to be a crucial therapeutic target for osteoarthritis via regulating chondrocyte inflammation and metabolism. In this review, we summarize the mechanism of hypoxia in the pathogenic mechanisms of osteoarthritis, and focus on a series of therapeutic treatments targeting HIF-1α for osteoarthritis. Further clarification of the regulatory mechanisms of HIF-1α in osteoarthritis may provide more useful clues to developing novel osteoarthritis treatment strategies.
Collapse
Affiliation(s)
- Chu-Yang Zeng
- Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University, Nanchang, China
- Department of Rehabilitation Medicine, The Third Hospital of Hebei Medical University, Shijiazhuang, China
| | - Xi-Feng Wang
- Department of Anesthesiology, The First Affiliated Hospital of Nanchang University, Nanchang, China
- *Correspondence: Xi-Feng Wang, ; Fu-Zhou Hua,
| | - Fu-Zhou Hua
- Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University, Nanchang, China
- *Correspondence: Xi-Feng Wang, ; Fu-Zhou Hua,
| |
Collapse
|
6
|
von Mentzer U, Corciulo C, Stubelius A. Biomaterial Integration in the Joint: Pathological Considerations, Immunomodulation, and the Extracellular Matrix. Macromol Biosci 2022; 22:e2200037. [PMID: 35420256 DOI: 10.1002/mabi.202200037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2022] [Revised: 03/30/2022] [Indexed: 11/08/2022]
Abstract
Defects of articular joints are becoming an increasing societal burden due to a persistent increase in obesity and aging. For some patients suffering from cartilage erosion, joint replacement is the final option to regain proper motion and limit pain. Extensive research has been undertaken to identify novel strategies enabling earlier intervention to promote regeneration and cartilage healing. With the introduction of decellularized extracellular matrix (dECM), researchers have tapped into the potential for increased tissue regeneration by designing biomaterials with inherent biochemical and immunomodulatory signals. Compared to conventional and synthetic materials, dECM-based materials invoke a reduced foreign body response. It is therefore highly beneficial to understand the interplay of how these native tissue-based materials initiate a favorable remodeling process by the immune system. Yet, such an understanding also demands increasing considerations of the pathological environment and remodeling processes, especially for materials designed for early disease intervention. This knowledge would avoid rejection and help predict complications in conditions with inflammatory components such as arthritides. This review outlines general issues facing biomaterial integration and emphasizes the importance of tissue-derived macromolecular components in regulating essential homeostatic, immunological, and pathological processes to increase biomaterial integration for patients suffering from joint degenerative diseases. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Ula von Mentzer
- Division of Chemical Biology, Department of Biology and Biological Engineering, Chalmers University of Technology, Kemivägen 10, Gothenburg, 41296, Sweden
| | - Carmen Corciulo
- Centre for Bone and Arthritis Research, Department of Rheumatology and Inflammation, Sahlgrenska Academy at the University of Gothenburg, Guldhedsgatan 10A, Gothenburg, 41296, Sweden
| | - Alexandra Stubelius
- Division of Chemical Biology, Department of Biology and Biological Engineering, Chalmers University of Technology, Kemivägen 10, Gothenburg, 41296, Sweden
| |
Collapse
|
7
|
Elucidating the role of hypoxia-inducible factor in rheumatoid arthritis. Inflammopharmacology 2022; 30:737-748. [PMID: 35364736 DOI: 10.1007/s10787-022-00974-4] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Accepted: 03/08/2022] [Indexed: 12/12/2022]
Abstract
Rheumatoid arthritis (RA) is a chronic multifactorial disease, provocative, and degenerative autoimmune condition that impacts millions of individuals around the globe. As a result of this understanding, anti-inflammatory drugs have been created, perhaps widely effective (like steroids) and highly specialized methods (including anti-TNF antibody) using biological therapies (including TNF inhibitors). Despite this, the connections between inflammatory response, articular development, and intracellular responsiveness to changes in oxygen concentration are undervalued in rheumatoid arthritis. Hypoxia, or a lack of oxygen, is thought to cause enhanced synovial angiogenesis in RA, which is mediated by some of the hypoxia-inducible factors like vascular endothelial growth factor (VEGF). Substantial genetic alterations occur when the HIF regulatory factors signaling cycle is activated, allowing organelles, tissues, and species to acclimatize to decreasing oxygen saturation. The most well-characterized hypoxia-responsive transcripts are the angiogenic stimulant VEGF, whose production is greatly elevated by hypoxia in several types of cells, especially RA synovium fibroblasts. Blocking vascular endothelial growth factors has been demonstrated to be helpful in murine models of rheumatism, indicating how hypoxia could trigger the angiogenesis process, resulting in the progression of RA. These mechanisms highlight the intimate affiliation amongst hypoxia, angiogenesis, and inflammation in rheumatoid arthritis. This review will look at how hypoxia activates molecular pathways and how other pathways involving inflammatory signals develop and sustain synovitis in rheumatoid arthritis.
Collapse
|
8
|
Pei YA, Pei M. Hypoxia Modulates Regenerative Potential of Fetal Stem Cells. APPLIED SCIENCES (BASEL, SWITZERLAND) 2022; 12:363. [PMID: 36660242 PMCID: PMC9846719 DOI: 10.3390/app12010363] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 06/17/2023]
Abstract
Adult mesenchymal stem cells (MSCs) are prone to senescence, which limits the scope of their use in tissue engineering and regeneration and increases the likelihood of post-implantation failure. As a robust alternative cell source, fetal stem cells can prevent an immune reaction and senescence. However, few studies use this cell type. In this study, we sought to characterize fetal cells' regenerative potential in hypoxic conditions. Specifically, we examined whether hypoxic exposure during the expansion and differentiation phases would affect human fetal nucleus pulposus cell (NPC) and fetal synovium-derived stem cell (SDSC) plasticity and three-lineage differentiation potential. We concluded that fetal NPCs represent the most promising cell source for chondrogenic differentiation, as they are more responsive and display stronger phenotypic stability, particularly when expanded and differentiated in hypoxic conditions. Fetal SDSCs have less potential for chondrogenic differentiation compared to their adult counterpart. This study also indicated that fetal SDSCs exhibit a discrepancy in adipogenic and osteogenic differentiation in response to hypoxia.
Collapse
Affiliation(s)
- Yixuan Amy Pei
- Stem Cell and Tissue Engineering Laboratory, Department of Orthopaedics, West Virginia University, Morgantown, WV 26506, USA
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Ming Pei
- Stem Cell and Tissue Engineering Laboratory, Department of Orthopaedics, West Virginia University, Morgantown, WV 26506, USA
- WVU Cancer Institute, West Virginia University, Morgantown, WV 26506, USA
| |
Collapse
|
9
|
Yue D, Du L, Zhang B, Wu H, Yang Q, Wang M, Pan J. Time-dependently Appeared Microenvironmental Changes and Mechanism after Cartilage or Joint Damage and the Influences on Cartilage Regeneration. Organogenesis 2021; 17:85-99. [PMID: 34806543 DOI: 10.1080/15476278.2021.1991199] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Cartilage and joint damage easily degenerates cartilage and turns into osteoarthritis (OA), which seriously affects human life and work, and has no cure currently. The temporal and spatial changes of multiple microenvironments upon the damage of cartilage and joint are noticed, including the emergences of inflammation, bone remodeling, blood vessels, and nerves, as well as alterations of extracellular and pericellular matrix, oxygen tension, biomechanics, underneath articular cartilage tissues, and pH value. This review summarizes the existing literatures on microenvironmental changes, mechanisms, and their negative effects on cartilage regeneration following cartilage and joint damage. We conclude that time-dependently rebuilding the multiple normal microenvironments of damaged cartilage is the key for cartilage regeneration after systematic studies for the timing and correlations of various microenvironment changes.
Collapse
Affiliation(s)
- Danyang Yue
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, Bioengineering College, Chongqing University, Chongqing, PR China
| | - Lin Du
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, Bioengineering College, Chongqing University, Chongqing, PR China
| | - Bingbing Zhang
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, Bioengineering College, Chongqing University, Chongqing, PR China
| | - Huan Wu
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, Bioengineering College, Chongqing University, Chongqing, PR China
| | - Qiong Yang
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, Bioengineering College, Chongqing University, Chongqing, PR China
| | - Min Wang
- Orthopedic Department, Xinqiao Hospital, Army Medical University, Chongqing, PR China
| | - Jun Pan
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, Bioengineering College, Chongqing University, Chongqing, PR China
| |
Collapse
|
10
|
Jeyaraman M, Muthu S, Gangadaran P, Ranjan R, Jeyaraman N, Prajwal GS, Mishra PC, Rajendran RL, Ahn BC. Osteogenic and Chondrogenic Potential of Periosteum-Derived Mesenchymal Stromal Cells: Do They Hold the Key to the Future? Pharmaceuticals (Basel) 2021; 14:ph14111133. [PMID: 34832915 PMCID: PMC8618036 DOI: 10.3390/ph14111133] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2021] [Revised: 11/02/2021] [Accepted: 11/04/2021] [Indexed: 02/05/2023] Open
Abstract
The periosteum, with its outer fibrous and inner cambium layer, lies in a dynamic environment with a niche of pluripotent stem cells for their reparative needs. The inner cambium layer is rich in mesenchymal progenitors, osteogenic progenitors, osteoblasts, and fibroblasts in a scant collagen matrix environment. Their role in union and remodeling of fracture is well known. However, the periosteum as a source of mesenchymal stem cells has not been explored in detail. Moreover, with the continuous expansion of techniques, newer insights have been acquired into the roles and regulation of these periosteal cells. From a therapeutic standpoint, the periosteum as a source of tissue engineering has gained much attraction. Apart from its role in bone repair, analysis of the bone-forming potential of periosteum-derived stem cells is lacking. Hence, this article elucidates the role of the periosteum as a potential source of mesenchymal stem cells along with their capacity for osteogenic and chondrogenic differentiation for therapeutic application in the future.
Collapse
Affiliation(s)
- Madhan Jeyaraman
- Department of Orthopaedics, School of Medical Sciences and Research, Sharda University, Greater Noida 201306, Uttar Pradesh, India; (M.J.); (R.R.)
- Department of Biotechnology, School of Engineering and Technology, Sharda University, Greater Noida 201310, Uttar Pradesh, India
- International Association of Stem Cell and Regenerative Medicine (IASRM), Greater Kailash, New Delhi 110048, Uttar Pradesh, India;
| | - Sathish Muthu
- Department of Biotechnology, School of Engineering and Technology, Sharda University, Greater Noida 201310, Uttar Pradesh, India
- International Association of Stem Cell and Regenerative Medicine (IASRM), Greater Kailash, New Delhi 110048, Uttar Pradesh, India;
- Department of Orthopaedics, Government Medical College and Hospital, Dindigul 624304, Tamil Nadu, India
- Correspondence: (S.M.); (R.L.R.); (B.-C.A.); Tel.: +82-53-420-4914 (R.L.R.); +82-53-420-5583 (B.-C.A.)
| | - Prakash Gangadaran
- BK21 FOUR KNU Convergence Educational Program of Biomedical Sciences for Creative Future Talents, Department of Biomedical Sciences, School of Medicine, Kyungpook National University, Daegu 41944, Korea;
- Department of Nuclear Medicine, School of Medicine, Kyungpook National University, Kyungpook National University Hospital, Daegu 41944, Korea
| | - Rajni Ranjan
- Department of Orthopaedics, School of Medical Sciences and Research, Sharda University, Greater Noida 201306, Uttar Pradesh, India; (M.J.); (R.R.)
| | - Naveen Jeyaraman
- Department of Orthopaedics, Atlas Hospitals, Tiruchirappalli 620002, Tamil Nadu, India;
| | | | - Prabhu Chandra Mishra
- International Association of Stem Cell and Regenerative Medicine (IASRM), Greater Kailash, New Delhi 110048, Uttar Pradesh, India;
| | - Ramya Lakshmi Rajendran
- Department of Nuclear Medicine, School of Medicine, Kyungpook National University, Kyungpook National University Hospital, Daegu 41944, Korea
- Correspondence: (S.M.); (R.L.R.); (B.-C.A.); Tel.: +82-53-420-4914 (R.L.R.); +82-53-420-5583 (B.-C.A.)
| | - Byeong-Cheol Ahn
- BK21 FOUR KNU Convergence Educational Program of Biomedical Sciences for Creative Future Talents, Department of Biomedical Sciences, School of Medicine, Kyungpook National University, Daegu 41944, Korea;
- Department of Nuclear Medicine, School of Medicine, Kyungpook National University, Kyungpook National University Hospital, Daegu 41944, Korea
- Correspondence: (S.M.); (R.L.R.); (B.-C.A.); Tel.: +82-53-420-4914 (R.L.R.); +82-53-420-5583 (B.-C.A.)
| |
Collapse
|
11
|
Castro CM, Corciulo C, Friedman B, Li Z, Jacob S, Fenyo D, Cronstein BN. Adenosine A2A receptor null chondrocyte transcriptome resembles that of human osteoarthritic chondrocytes. Purinergic Signal 2021; 17:439-448. [PMID: 33973110 PMCID: PMC8410926 DOI: 10.1007/s11302-021-09788-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2021] [Accepted: 04/08/2021] [Indexed: 11/25/2022] Open
Abstract
Adenosine signaling plays a critical role in the maintenance of articular cartilage and may serve as a novel therapeutic for osteoarthritis (OA), a highly prevalent and morbid disease without effective therapeutics in the current market. Mice lacking adenosine A2A receptors (A2AR) develop spontaneous OA by 16 weeks of age, a finding relevant to human OA since loss of adenosine signaling due to diminished adenosine production (NT5E deficiency) also leads to development of OA in mice and humans. To better understand the mechanism by which A2AR and adenosine generation protect from OA development, we examined differential gene expression in neonatal chondrocytes from WT and A2AR null mice. Analysis of differentially expressed genes was analyzed by KEGG pathway analysis, and oPOSSUM and the flatiron database were used to identify transcription factor binding enrichment, and tissue-specific network analyses and patterns were compared to gene expression patterns in chondrocytes from patients with OA. There was a differential expression of 2211 genes (padj<0.05). Pathway enrichment analysis revealed that pro-inflammatory changes, increased metalloprotease, reduced matrix organization, and homeostasis are upregulated in A2AR null chondrocytes. Moreover, stress responses, including autophagy and HIF-1 signaling, seem to be important drivers of OA and bear marked resemblance to the human OA transcriptome. Although A2AR null mice are born with grossly intact articular cartilage, we identify here the molecular foundations for early-onset OA in these mice, further establishing their role as models for human disease and the potential use of adenosine as a treatment for human disease.
Collapse
Affiliation(s)
- Cristina M. Castro
- Department of Medicine, Beth Israel Deaconess Medical Center (BIDMC), Boston, MA USA
| | - Carmen Corciulo
- Division of Translational Medicine, NYUGSOM, New York, NY USA
- Centre for Bone and Arthritis Research, Department of Internal Medicine and Clinical Nutritional, Institute of Medicine, Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| | - Benjamin Friedman
- Department of Medicine, Division of Rheumatology, NYUGSOM, New York, NY USA
| | - Zhi Li
- Institute for Systems Genetics, NYU Langone Health, New York, NY USA
- Department of Biochemistry and Molecular Pharmacology, NYU Langone Health, New York, NY USA
| | - Samson Jacob
- Institute for Systems Genetics, NYU Langone Health, New York, NY USA
| | - David Fenyo
- Institute for Systems Genetics, NYU Langone Health, New York, NY USA
- Department of Biochemistry and Molecular Pharmacology, NYU Langone Health, New York, NY USA
| | - Bruce N. Cronstein
- Department of Medicine, Division of Rheumatology, NYUGSOM, New York, NY USA
| |
Collapse
|
12
|
Razmara E, Bitaraf A, Karimi B, Babashah S. Functions of the SNAI family in chondrocyte-to-osteocyte development. Ann N Y Acad Sci 2021; 1503:5-22. [PMID: 34403146 DOI: 10.1111/nyas.14668] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 06/22/2021] [Accepted: 07/02/2021] [Indexed: 12/12/2022]
Abstract
Different cellular mechanisms contribute to osteocyte development. And while critical roles for members of the zinc finger protein SNAI family (SNAIs) have been discussed in cancer-related models, there are few reviews summarizing their importance for chondrocyte-to-osteocyte development. To help fill this gap, we review the roles of SNAIs in the development of mature osteocytes from chondrocytes, including the regulation of chondro- and osteogenesis through different signaling pathways and in programmed cell death. We also discuss how epigenetic factors-including DNA methylation, histone methylation and acetylation, and noncoding RNAs-contribute differently to both chondrocyte and osteocyte development. To better grasp the important roles of SNAIs in bone development, we also review genotype-phenotype correlations in different animal models. We end with comments about the possible importance of the SNAI family in cartilage/bone development and the potential applications for therapeutic goals.
Collapse
Affiliation(s)
- Ehsan Razmara
- Australian Regenerative Medicine Institute, Monash University, Clayton, Victoria, Australia
| | - Amirreza Bitaraf
- Department of Molecular Genetics, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran
| | - Behnaz Karimi
- Hematology/Oncology and Stem Cell Transplantation Research Center, Tehran University of Medical Science, Tehran, Iran
| | - Sadegh Babashah
- Department of Molecular Genetics, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran
| |
Collapse
|
13
|
Tarantino R, Chiu LLY, Weber JF, Yat Tse M, Bardana DD, Pang SC, Waldman SD. Effect of nutrient metabolism on cartilaginous tissue formation. Biotechnol Bioeng 2021; 118:4119-4128. [PMID: 34265075 DOI: 10.1002/bit.27888] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Revised: 06/25/2021] [Accepted: 07/09/2021] [Indexed: 11/09/2022]
Abstract
A major shortcoming in cartilage tissue engineering is the low biosynthetic response of chondrocytes. While different strategies have been investigated, a novel approach may be to control nutrient metabolism. Although known for their anaerobic metabolism, chondrocytes are more synthetically active under conditions that elicit mixed aerobic-anaerobic metabolism. Here, we postulate this metabolic switch induces HIF-1α signaling resulting in improved growth. Transition to different metabolic states can result in the pooling of metabolites, several of which can stabilize HIF-1α by interfering with PHD2. Chondrocytes cultured under increased media availability accelerated tissue deposition with the greatest effect occurring at 2 ml/106 cells. Under higher media availability, metabolism switched from anaerobic to mixed aerobic-anaerobic. Around this transition, maximal changes in PHD2 activity, HIF-1α expression, and HIF-1 target gene expression were observed. Loss-of-function studies using YC-1 confirmed the involvement of HIF-1. Lastly, targeted metabolomic studies revealed that intracellular lactate and succinate correlated with PHD2 activity. This study demonstrates that cartilaginous tissue formation can be regulated by nutrient metabolism and that this response is mediated through changes in HIF-1α signaling. By harnessing this newly identified metabolic switch, engineered cartilage implants may be developed without the need for sophisticated methods which could aid translation to the clinic.
Collapse
Affiliation(s)
- Roberto Tarantino
- Department of Chemical Engineering, Ryerson University, Toronto, Ontario, Canada.,Li Ka Shing Knowledge Institute, St. Michael's Hospital, Toronto, Ontario, Canada
| | - Loraine L Y Chiu
- Li Ka Shing Knowledge Institute, St. Michael's Hospital, Toronto, Ontario, Canada
| | - Joanna F Weber
- Li Ka Shing Knowledge Institute, St. Michael's Hospital, Toronto, Ontario, Canada
| | - Man Yat Tse
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Ontario, Canada
| | - Davide D Bardana
- Department of Surgery, Queen's University, Kingston, Ontario, Canada
| | - Stephen C Pang
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Ontario, Canada
| | - Stephen D Waldman
- Department of Chemical Engineering, Ryerson University, Toronto, Ontario, Canada.,Li Ka Shing Knowledge Institute, St. Michael's Hospital, Toronto, Ontario, Canada
| |
Collapse
|
14
|
Baddam P, Young D, Dunsmore G, Nie C, Eaton F, Elahi S, Jovel J, Adesida AB, Dufour A, Graf D. Nasal Septum Deviation as the Consequence of BMP-Controlled Changes to Cartilage Properties. Front Cell Dev Biol 2021; 9:696545. [PMID: 34249945 PMCID: PMC8265824 DOI: 10.3389/fcell.2021.696545] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2021] [Accepted: 05/24/2021] [Indexed: 11/29/2022] Open
Abstract
The nasal septum cartilage is a specialized hyaline cartilage important for normal midfacial growth. Abnormal midfacial growth is associated with midfacial hypoplasia and nasal septum deviation (NSD). However, the underlying genetics and associated functional consequences of these two anomalies are poorly understood. We have previously shown that loss of Bone Morphogenetic Protein 7 (BMP7) from neural crest (BMP7 ncko ) leads to midfacial hypoplasia and subsequent septum deviation. In this study we elucidate the cellular and molecular abnormalities underlying NSD using comparative gene expression, quantitative proteomics, and immunofluorescence analysis. We show that reduced cartilage growth and septum deviation are associated with acquisition of elastic cartilage markers and share similarities with osteoarthritis (OA) of the knee. The genetic reduction of BMP2 in BMP7 ncko mice was sufficient to rescue NSD and suppress elastic cartilage markers. To our knowledge this investigation provides the first genetic example of an in vivo cartilage fate switch showing that this is controlled by the relative balance of BMP2 and BMP7. Cellular and molecular changes similar between NSD and knee OA suggest a related etiology underlying these cartilage abnormalities.
Collapse
Affiliation(s)
- Pranidhi Baddam
- School of Dentistry, University of Alberta, Edmonton, AB, Canada
| | - Daniel Young
- Department of Physiology and Pharmacology, University of Calgary, Calgary, AB, Canada
| | - Garett Dunsmore
- Department of Medical Microbiology and Immunology, University of Alberta, Edmonton, AB, Canada
| | - Chunpeng Nie
- School of Dentistry, University of Alberta, Edmonton, AB, Canada
| | - Farah Eaton
- School of Dentistry, University of Alberta, Edmonton, AB, Canada
| | - Shokrollah Elahi
- School of Dentistry, University of Alberta, Edmonton, AB, Canada
| | - Juan Jovel
- Department of Medicine, University of Alberta, Edmonton, AB, Canada
| | | | - Antoine Dufour
- Department of Physiology and Pharmacology, University of Calgary, Calgary, AB, Canada
| | - Daniel Graf
- School of Dentistry, University of Alberta, Edmonton, AB, Canada
| |
Collapse
|
15
|
Jacob B, Jüllig M, Middleditch M, Payne L, Broom N, Sarojini V, Thambyah A. Protein Levels and Microstructural Changes in Localized Regions of Early Cartilage Degeneration Compared with Adjacent Intact Cartilage. Cartilage 2021; 12:192-210. [PMID: 30486653 PMCID: PMC7970373 DOI: 10.1177/1947603518809401] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
OBJECTIVE It was hypothesized that the respective protein profiles of bovine cartilage from sites of localized mild to moderate (GI to GII) degeneration versus adjacent sites of intact tissue would vary in accordance with the tissue microstructural changes associated with a pre-osteoarthritic state. METHODS A total of 15 bovine patellae were obtained for this study. Paired samples of tissue were collected from the lateral region of each patella. If the patella contained a site of degeneration, a paired tissue set involved taking one sample each from the degenerated site and the intact tissue adjacent to it. Sufficient tissue was collected to facilitate 2 arms of investigation: microstructural imaging and proteome analysis. The microstructural analysis used a bespoke tissue preparation technique imaged with differential interference contrast optical microscopy to assess fibrillar scale destructuring and underlying bone spicule formation. An iTRAQ-based proteome analysis was performed using liquid chromatography-tandem mass spectrometry to identify the differential levels of proteins across the intact and degenerated cartilage and further, the results were validated with multiple reaction monitoring assay. RESULTS In the healthy cartilage pairs, there was no significant variation in protein profiles between 2 adjacent sample sites. In pairs of tissue that contained a sample of GI/GII tissue, there were both significant microstructural changes as well as the difference in abundance levels of 24 proteins. CONCLUSIONS From the known functions of the 24 proteins, found to be strongly aligned with the specific microstructural changes observed, a unique "proteins ensemble" involved in the initiation and progression of early cartilage degeneration is proposed.
Collapse
Affiliation(s)
- Bincy Jacob
- School of Biological Sciences, The
University of Auckland, Auckland, New Zealand
| | - Mia Jüllig
- School of Biological Sciences, The
University of Auckland, Auckland, New Zealand
| | - Martin Middleditch
- School of Biological Sciences, The
University of Auckland, Auckland, New Zealand
| | - Leo Payne
- School of Biological Sciences, The
University of Auckland, Auckland, New Zealand
| | - Neil Broom
- Department of Chemical and Materials
Engineering, Experimental Tissue Mechanics Laboratory, University of Auckland,
Auckland, New Zealand
| | | | - Ashvin Thambyah
- Department of Chemical and Materials
Engineering, Experimental Tissue Mechanics Laboratory, University of Auckland,
Auckland, New Zealand,Ashvin Thambyah, Department of Chemical and
Materials Engineering, Experimental Tissue Mechanics Laboratory, University of
Auckland, 20 Symonds Street, Auckland, 1010, New Zealand.
| |
Collapse
|
16
|
Ziliotto M, Rodrigues RM, Chies JAB. Controlled hypobaric hypoxia increases immunological tolerance by modifying HLA-G expression, a potential therapy to inflammatory diseases. Med Hypotheses 2020; 140:109664. [PMID: 32155542 DOI: 10.1016/j.mehy.2020.109664] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2020] [Revised: 02/26/2020] [Accepted: 03/03/2020] [Indexed: 12/22/2022]
Abstract
One of the most striking characteristics of human beings is the incredible capacity to adapt to different environments. This capacity allowed humans to spread all over our planet, occupying habitats as diverse as deserts, tropical forests or tundra regions. Interactions with the environment, climate, food and water availability shaped our evolution and define our survival. Essential to human life, oxygen availability also controls human dispersion and adaptation. For example, low oxygen availability can lead to physiological adaptations in populations living in highlands. Moreover, the consequences of differential oxygen availability (or even exposure to hypoxia) are evident in process as fine-tuned controlled as gene regulation. Physiological responses to fluctuations in oxygen availability are crucial already from the early days of life, since the human fetal environment is characterized by hypoxia. Hypoxia-Inducible Factors (HIFs) act as major regulators of pathways involved in glycolysis, erythropoiesis, angiogenesis, cell proliferation and stem cells function. Here we explore the physiological consequences of hypoxia in the human organism. In this sense, and considering the existence of HIF sequences in promoter regions of genes important to immune regulation, we hypothesize that exposure to induced hypoxia through the use of hypobaric chambers can be used as a complementary therapy to control chronic inflammation in several diseases characterized by systemic inflammatory conditions. Among these inflammatory conditions we highlight autoimmune diseases and chronic inflammation in HIV infected individuals under antiretroviral treatment. Several experiments, including arthritis animal models, the evaluation of athletes that already use hypobaric chambers to induce erythropoiesis, and the potential consequences of hypoxia as an immunotolerogenic inducer in the HIV infection context are approached and discussed here.
Collapse
Affiliation(s)
- Marina Ziliotto
- Laboratory of Immunobiology and Immunogenetics, Department of Genetics, Universidade Federal do Rio Grande do Sul - UFRGS, Porto Alegre, Rio Grande do Sul, Brazil
| | - Raul Marques Rodrigues
- Laboratory of Immunobiology and Immunogenetics, Department of Genetics, Universidade Federal do Rio Grande do Sul - UFRGS, Porto Alegre, Rio Grande do Sul, Brazil
| | - José Artur Bogo Chies
- Laboratory of Immunobiology and Immunogenetics, Department of Genetics, Universidade Federal do Rio Grande do Sul - UFRGS, Porto Alegre, Rio Grande do Sul, Brazil.
| |
Collapse
|
17
|
Liang R, Zhao J, Li B, Cai P, Loh XJ, Xu C, Chen P, Kai D, Zheng L. Implantable and degradable antioxidant poly(ε-caprolactone)-lignin nanofiber membrane for effective osteoarthritis treatment. Biomaterials 2020; 230:119601. [DOI: 10.1016/j.biomaterials.2019.119601] [Citation(s) in RCA: 49] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2019] [Revised: 11/01/2019] [Accepted: 11/01/2019] [Indexed: 12/31/2022]
|
18
|
Van de Walle A, Faissal W, Wilhelm C, Luciani N. Role of growth factors and oxygen to limit hypertrophy and impact of high magnetic nanoparticles dose during stem cell chondrogenesis. Comput Struct Biotechnol J 2018; 16:532-542. [PMID: 30524668 PMCID: PMC6260287 DOI: 10.1016/j.csbj.2018.10.014] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2018] [Revised: 10/25/2018] [Accepted: 10/26/2018] [Indexed: 12/26/2022] Open
Abstract
Due to an unmet clinical need of curative treatments for osteoarthritic patients, tissue engineering strategies that propose the development of cartilage tissue replacements from stem cells have emerged. Some of these strategies are based on the internalization of magnetic nanoparticles into stem cells to then initiate the chondrogenesis via magnetic compaction. A major difficulty is to drive the chondrogenic differentiation of the cells such as they produce an extracellular matrix free of hypertrophic collagen. An additional difficulty has to be overcome when nanoparticles are used, knowing that a high dose of nanoparticles can limit the chondrogenesis. We here propose a gene-based analysis of the effects of chemical factors (growth factors, hypoxia) on the chondrogenic differentiation of human mesenchymal stem cells both with and without nanoparticles. We focus on the synthesis of two of the most important constituents present in the cartilaginous extracellular matrix (Collagen II and Aggrecan) and on the expression of collagen X, the signature of hypertrophic cartilage, in order to provide a quantitative index of the type of cartilage produced (i.e. hyaline, hypertrophic). We demonstrate that by applying specific environmental conditions, gene expression can be directed toward the production of hyaline cartilage, with limited hypertrophy. Besides, a combination of the growth factors IGF-1, TGF-β3, with a hypoxic conditioning remarkably reduced the impact of high nanoparticles concentration.
Collapse
Affiliation(s)
| | | | - Claire Wilhelm
- Corresponding authors at: Laboratoire MSC, UMR 7057 CNRS, University Paris Diderot, France.
| | - Nathalie Luciani
- Corresponding authors at: Laboratoire MSC, UMR 7057 CNRS, University Paris Diderot, France.
| |
Collapse
|
19
|
Bicho D, Pina S, Oliveira JM, Reis RL. In Vitro Mimetic Models for the Bone-Cartilage Interface Regeneration. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2018; 1059:373-394. [PMID: 29736583 DOI: 10.1007/978-3-319-76735-2_17] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
In embryonic development, pure cartilage structures are in the basis of bone-cartilage interfaces. Despite this fact, the mature bone and cartilage structures can vary greatly in composition and function. Nevertheless, they collaborate in the osteochondral region to create a smooth transition zone that supports the movements and forces resulting from the daily activities. In this sense, all the hierarchical organization is involved in the maintenance and reestablishment of the equilibrium in case of damage. Therefore, this interface has attracted a great deal of interest in order to understand the mechanisms of regeneration or disease progression in osteoarthritis. With that purpose, in vitro tissue models (either static or dynamic) have been studied. Static in vitro tissue models include monocultures, co-cultures, 3D cultures, and ex vivo cultures, mostly cultivated in flat surfaces, while dynamic models involve the use of bioreactors and microfluidic systems. The latter have emerged as alternatives to study the cellular interactions in a more authentic manner over some disadvantages of the static models. The current alternatives of in vitro mimetic models for bone-cartilage interface regeneration are overviewed and discussed herein.
Collapse
Affiliation(s)
- Diana Bicho
- 3B's Research Group - Biomaterials, Biodegradables and Biomimetics, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, University of Minho, Barco, Guimarães, Portugal.
- ICVS/3B's - PT Government Associate Laboratory, Braga, Guimarães, Portugal.
| | - Sandra Pina
- ICVS/3B's - PT Government Associate Laboratory, Braga, Guimarães, Portugal
| | - J Miguel Oliveira
- ICVS/3B's - PT Government Associate Laboratory, Braga, Guimarães, Portugal
- The Discoveries Centre for Regenerative and Precision Medicine, Headquarters at University of Minho, Guimarães, Portugal
| | - Rui L Reis
- ICVS/3B's - PT Government Associate Laboratory, Braga, Guimarães, Portugal
- The Discoveries Centre for Regenerative and Precision Medicine, Headquarters at University of Minho, Guimarães, Portugal
| |
Collapse
|
20
|
Zhou J, Yue D, Bai Y, Kong F, Pan J. Map and correlate intracellular calcium response and matrix deposition in cartilage under physiological oxygen tensions. J Cell Physiol 2017; 233:4949-4960. [PMID: 29215706 DOI: 10.1002/jcp.26326] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2017] [Accepted: 11/29/2017] [Indexed: 12/28/2022]
Abstract
Face to the limited repair capability of cartilage, we intended to find out signaling responsible for its matrix synthesis. Since spontaneous calcium response likes a label of cell status, here it was mapped in fresh and 24 hr cultured in situ chondrocytes under oxygen tensions of 20%, 5%, and 1% as well as mimic hypoxia conditions. The calcium source was traced using ethylene glycol-bis (β-aminoethyl ether)-N,N,N',N'-tetraacetic acid (EGTA) and thapsigargin (TG) to treat cartilage. Their relative matrix of type II collagen (COLL-II) and glycosaminoglycan (GAG) were quantified after cultured for 3 and 7 days. We disclosed the specific fingerprint of calcium response and matrix deposition along the histological zones under various oxygen tensions, from which the effects of hyperoxia, normoxia, and hypoxia conditions on as well as the optimal oxygen tensions for maintenance of various zones of cartilage or chondrocytes were derived and obtained. Our results revealed that cytoplasm calcium was conducive to synthesize COLL-II but detrimental to synthesize GAG. These results provide correlation in addition to details of intracellular calcium response and matrix deposition in in situ cartilage along its histological zones under physiological oxygen tensions.
Collapse
Affiliation(s)
- Jin Zhou
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, College of Bioengineering, Chongqing University, Chongqing, China
| | - Danyang Yue
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, College of Bioengineering, Chongqing University, Chongqing, China
| | - Yuying Bai
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, College of Bioengineering, Chongqing University, Chongqing, China
| | - Fei Kong
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, College of Bioengineering, Chongqing University, Chongqing, China
| | - Jun Pan
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, College of Bioengineering, Chongqing University, Chongqing, China
| |
Collapse
|
21
|
Yuan T, Luo H, Guo L, Fan H, Liang J, Fan Y, Zhang X. In vivo immunological properties research on mesenchymal stem cells based engineering cartilage by a dialyzer pocket model. JOURNAL OF MATERIALS SCIENCE. MATERIALS IN MEDICINE 2017; 28:150. [PMID: 28831637 DOI: 10.1007/s10856-017-5955-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/14/2017] [Accepted: 08/01/2017] [Indexed: 06/07/2023]
Abstract
As the seed cells, the immune properties of the mesenchymal stem cells are important for the tissue engineering restoring effect. But the in vivo research model is lacking. In the study, based on a dialyzer pocket model, changes in immunological properties and the differentiation of seeded mesenchymal stem cells (MSCs) in collagen hydrogel were studied in muscle and articular cavity implantation, respectively. The results showed that collagen hydrogel can induce MSCs to form cartilage tissue, followed by alteration of immunological properties. In muscle implantation, relatively low expression of major histocompatibility complex (MHC) molecules and low level of one-way mixed lymphocyte reactions (MLR) on the seeded MSCs were observed, but only a little cartilage tissue formed. In articular cavity implantation, more cartilage tissue formed, but higher MHC expressions and MLR level were found. Results indicated that the immunomodulation and the cartilage formation of the seeded MSCs will be impacted by the scaffold and the environment of the in vivo implanted site. The dialyzer pocket model can be used for the in vivo research for the MSC-based strategy of the tissue engineering, especially for the optimization of the immunomodulation.
Collapse
Affiliation(s)
- Tun Yuan
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, China
| | - Hongrong Luo
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, China
| | - Likun Guo
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, China
| | - Hongsong Fan
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, China
| | - Jie Liang
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, China.
| | - Yujiang Fan
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, China.
| | - Xingdong Zhang
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, China
| |
Collapse
|
22
|
Cleven AHG, Suijker J, Agrogiannis G, Briaire-de Bruijn IH, Frizzell N, Hoekstra AS, Wijers-Koster PM, Cleton-Jansen AM, Bovée JVMG. IDH1 or - 2 mutations do not predict outcome and do not cause loss of 5-hydroxymethylcytosine or altered histone modifications in central chondrosarcomas. Clin Sarcoma Res 2017; 7:8. [PMID: 28484589 PMCID: PMC5418698 DOI: 10.1186/s13569-017-0074-6] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2017] [Accepted: 04/19/2017] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Mutations in isocitrate dehydrogenase (IDH)1 or -2 are found in ~50% of conventional central chondrosarcomas and in up to 87% of their assumed benign precursors enchondromas. The mutant enzyme acquires the activity to convert α-ketoglutarate into the oncometabolite d-2-hydroxyglutarate (d-2-HG), which competitively inhibits α-ketoglutarate dependent enzymes such as histone- and DNA demethylases. METHODS We therefore evaluated the effect of IDH1 or -2 mutations on histone modifications (H3K4me3, H3K9me3 and H3K27me3), chromatin remodeler ATRX expression, DNA modifications (5-hmC and 5-mC), and TET1 subcellular localization in a genotyped cohort (IDH, succinate dehydrogenase (SDH) and fumarate hydratase (FH)) of enchondromas and central chondrosarcomas (n = 101) using immunohistochemistry. RESULTS IDH1 or -2 mutations were found in 60.8% of the central cartilaginous tumours, while mutations in FH and SDH were absent. The mutation status did not correlate with outcome. Chondrosarcomas are strongly positive for the histone modifications H3K4me3, H3K9me3 and H3K27me3, which was independent of the IDH1 or -2 mutation status. Two out of 36 chondrosarcomas (5.6%) show complete loss of ATRX. Levels of 5-hmC and 5-mC are highly variable in central cartilaginous tumours and are not associated with mutation status. In tumours with loss of 5-hmC, expression of TET1 was more prominent in the cytoplasm than the nucleus (p = 0.0001). CONCLUSIONS In summary, in central chondrosarcoma IDH1 or -2 mutations do not affect immunohistochemical levels of 5-hmC, 5mC, trimethylation of H3K4, -K9 and K27 and outcome, as compared to wildtype.
Collapse
Affiliation(s)
- Arjen H G Cleven
- Department of Pathology, Leiden University Medical Center, L1-Q, P.O. Box 9600, 2300 RC Leiden, The Netherlands
| | - Johnny Suijker
- Department of Pathology, Leiden University Medical Center, L1-Q, P.O. Box 9600, 2300 RC Leiden, The Netherlands
| | - Georgios Agrogiannis
- 1st Department of Pathology, Laikon General Hospital, Athens University School of Medicine, Athens, Greece
| | - Inge H Briaire-de Bruijn
- Department of Pathology, Leiden University Medical Center, L1-Q, P.O. Box 9600, 2300 RC Leiden, The Netherlands
| | - Norma Frizzell
- Department of Pharmacology, Physiology & Neuroscience, School of Medicine, University of South Carolina, Columbia, USA
| | - Attje S Hoekstra
- Department of Human Genetics, Leiden University Medical Center, Leiden, The Netherlands
| | - Pauline M Wijers-Koster
- Department of Pathology, Leiden University Medical Center, L1-Q, P.O. Box 9600, 2300 RC Leiden, The Netherlands
| | - Anne-Marie Cleton-Jansen
- Department of Pathology, Leiden University Medical Center, L1-Q, P.O. Box 9600, 2300 RC Leiden, The Netherlands
| | - Judith V M G Bovée
- Department of Pathology, Leiden University Medical Center, L1-Q, P.O. Box 9600, 2300 RC Leiden, The Netherlands
| |
Collapse
|
23
|
Ratcliffe P, Koivunen P, Myllyharju J, Ragoussis J, Bovée JV, Batinic-Haberle I, Vinatier C, Trichet V, Robriquet F, Oliver L, Gardie B. Update on hypoxia-inducible factors and hydroxylases in oxygen regulatory pathways: from physiology to therapeutics. HYPOXIA 2017; 5:11-20. [PMID: 28352643 PMCID: PMC5359007 DOI: 10.2147/hp.s127042] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
The “Hypoxia Nantes 2016” organized its second conference dedicated to the field of hypoxia research. This conference focused on “the role of hypoxia under physiological conditions as well as in cancer” and took place in Nantes, France, in October 6–7, 2016. The main objective of this conference was to bring together a large group of scientists from different spheres of hypoxia. Recent advances were presented and discussed around different topics: genomics, physiology, musculoskeletal, stem cells, microenvironment and cancer, and oxidative stress. This review summarizes the major highlights of the meeting.
Collapse
Affiliation(s)
- Peter Ratcliffe
- Target Discovery Institute, University of Oxford; The Francis Crick Institute, London, UK
| | - Peppi Koivunen
- Oulu Center for Cell-Matrix Research, Biocenter Oulu and Faculty of Biochemistry and Molecular Medicine, University of Oulu, Oulu, Finland
| | - Johanna Myllyharju
- Oulu Center for Cell-Matrix Research, Biocenter Oulu and Faculty of Biochemistry and Molecular Medicine, University of Oulu, Oulu, Finland
| | - Jiannis Ragoussis
- McGill University and Genome Quebec Innovation Centre, McGill University, Montreal, Canada
| | - Judith Vmg Bovée
- Department of Pathology, Leiden University Medical Center, Leiden, the Netherlands
| | - Ines Batinic-Haberle
- Department of Radiation Oncology, Duke University School of Medicine, Durham, NC, USA
| | - Claire Vinatier
- INSERM UMR 1229, Regenerative Medicine and Skeleton-RMeS, Team STEP, University of Nantes, UFR Odontology
| | | | | | - Lisa Oliver
- CRCINA, INSERM, Université de Nantes, Nantes
| | - Betty Gardie
- CRCINA, INSERM, Université de Nantes, Nantes; Ecole Pratique des Hautes Etudes, PSL Research University, Paris, France
| |
Collapse
|
24
|
The Signaling Pathways Involved in Chondrocyte Differentiation and Hypertrophic Differentiation. Stem Cells Int 2016; 2016:2470351. [PMID: 28074096 PMCID: PMC5198191 DOI: 10.1155/2016/2470351] [Citation(s) in RCA: 94] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2016] [Accepted: 11/22/2016] [Indexed: 12/19/2022] Open
Abstract
Chondrocytes communicate with each other mainly via diffusible signals rather than direct cell-to-cell contact. The chondrogenic differentiation of mesenchymal stem cells (MSCs) is well regulated by the interactions of varieties of growth factors, cytokines, and signaling molecules. A number of critical signaling molecules have been identified to regulate the differentiation of chondrocyte from mesenchymal progenitor cells to their terminal maturation of hypertrophic chondrocytes, including bone morphogenetic proteins (BMPs), SRY-related high-mobility group-box gene 9 (Sox9), parathyroid hormone-related peptide (PTHrP), Indian hedgehog (Ihh), fibroblast growth factor receptor 3 (FGFR3), and β-catenin. Except for these molecules, other factors such as adenosine, O2 tension, and reactive oxygen species (ROS) also have a vital role in cartilage formation and chondrocyte maturation. Here, we outlined the complex transcriptional network and the function of key factors in this network that determine and regulate the genetic program of chondrogenesis and chondrocyte differentiation.
Collapse
|
25
|
Kean TJ, Mera H, Whitney GA, MacKay DL, Awadallah A, Fernandes RJ, Dennis JE. Disparate response of articular- and auricular-derived chondrocytes to oxygen tension. Connect Tissue Res 2016; 57:319-33. [PMID: 27128439 PMCID: PMC4984267 DOI: 10.1080/03008207.2016.1182996] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
PURPOSE/AIM To determine the effect of reduced (5%) oxygen tension on chondrogenesis of auricular-derived chondrocytes. Currently, many cell and tissue culture experiments are performed at 20% oxygen with 5% carbon dioxide. Few cells in the body are subjected to this supra-physiological oxygen tension. Chondrocytes and their mesenchymal progenitors are widely reported to have greater chondrogenic expression when cultured at low, more physiological, oxygen tension (1-7%). Although generally accepted, there is still some controversy, and different culture methods, species, and outcome metrics cloud the field. These results are, however, articular chondrocyte biased and have not been reported for auricular-derived chondrocytes. MATERIALS AND METHODS Auricular and articular chondrocytes were isolated from skeletally mature New Zealand White rabbits, expanded in culture and differentiated in high density cultures with serum-free chondrogenic media. Cartilage tissue derived from aggregate cultures or from the tissue engineered sheets were assessed for biomechanical, glycosaminoglycan, collagen, collagen cross-links, and lysyl oxidase activity and expression. RESULTS Our studies show increased proliferation rates for both auricular and articular chondrocytes at low (5%) O2 versus standard (20%) O2. In our scaffold-free chondrogenic cultures, low O2 was found to increase articular chondrocyte accumulation of glycosaminoglycan, but not cross-linked type II collagen, or total collagen. Conversely, auricular chondrocytes accumulated less glycosaminoglycan, cross-linked type II collagen and total collagen under low oxygen tension. CONCLUSIONS This study highlights the dramatic difference in response to low O2 of chondrocytes isolated from different anatomical sites. Low O2 is beneficial for articular-derived chondrogenesis but detrimental for auricular-derived chondrogenesis.
Collapse
Affiliation(s)
- Thomas J. Kean
- Matrix Biology Program, Benaroya Research Institute at Virginia Mason, Seattle, WA, USA,Department of Orthopedics, Case Western Reserve University, Cleveland, OH, USA,Department of Orthopedic Surgery, Baylor College of Medicine, Houston, TX, USA
| | - Hisashi Mera
- Matrix Biology Program, Benaroya Research Institute at Virginia Mason, Seattle, WA, USA,Department of Orthopedics, Case Western Reserve University, Cleveland, OH, USA,Department of Health and Sports Sciences, Mukogawa Women’s University, Hyogo, Japan
| | - G. Adam Whitney
- Matrix Biology Program, Benaroya Research Institute at Virginia Mason, Seattle, WA, USA,Department of Orthopedics, Case Western Reserve University, Cleveland, OH, USA
| | - Danielle L. MacKay
- Matrix Biology Program, Benaroya Research Institute at Virginia Mason, Seattle, WA, USA,Department of Orthopedic Surgery, Baylor College of Medicine, Houston, TX, USA
| | - Amad Awadallah
- Department of Orthopedics, Case Western Reserve University, Cleveland, OH, USA
| | - Russell J. Fernandes
- Department of Orthopaedics and Sports Medicine, University of Washington, Seattle, WA, USA
| | - James E. Dennis
- Matrix Biology Program, Benaroya Research Institute at Virginia Mason, Seattle, WA, USA,Department of Orthopedics, Case Western Reserve University, Cleveland, OH, USA,Department of Orthopedic Surgery, Baylor College of Medicine, Houston, TX, USA
| |
Collapse
|
26
|
Hua S, Dias TH. Hypoxia-Inducible Factor (HIF) as a Target for Novel Therapies in Rheumatoid Arthritis. Front Pharmacol 2016; 7:184. [PMID: 27445820 PMCID: PMC4921475 DOI: 10.3389/fphar.2016.00184] [Citation(s) in RCA: 83] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2016] [Accepted: 06/10/2016] [Indexed: 12/19/2022] Open
Abstract
Hypoxia is an important micro-environmental characteristic of rheumatoid arthritis (RA). Hypoxia-inducible factors (HIF) are key transcriptional factors that are highly expressed in RA synovium to regulate the adaptive responses to this hypoxic milieu. Accumulating evidence supports hypoxia and HIFs in regulating a number of important pathophysiological characteristics of RA, including synovial inflammation, angiogenesis, and cartilage destruction. Experimental and clinical data have confirmed the upregulation of both HIF-1α and HIF-2α in RA. This review will focus on the differential expression of HIFs within the synovial joint and its functional behavior in different cell types to regulate RA progression. Potential development of new therapeutic strategies targeting HIF-regulated pathways at sites of disease in RA will also be addressed.
Collapse
Affiliation(s)
- Susan Hua
- School of Biomedical Sciences and Pharmacy, University of NewcastleCallaghan, NSW, Australia; Hunter Medical Research InstituteNew Lambton Heights, NSW, Australia
| | - Thilani H Dias
- School of Biomedical Sciences and Pharmacy, University of Newcastle Callaghan, NSW, Australia
| |
Collapse
|
27
|
Tyner H, Patel R. Propionibacterium acnes biofilm - A sanctuary for Staphylococcus aureus? Anaerobe 2016; 40:63-7. [PMID: 27241780 DOI: 10.1016/j.anaerobe.2016.05.014] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2016] [Revised: 05/16/2016] [Accepted: 05/26/2016] [Indexed: 10/21/2022]
Abstract
The purpose of this study was to measure the effect of combined culture of Propionibacterium acnes and Staphylococcus aureus on biofilm formation under different oxygen concentrations. We measured planktonic growth and biofilm formation of P. acnes and S. aureus alone and together under aerobic and anaerobic conditions. Both P. acnes and S. aureus grew under anaerobic conditions. When grown under anaerobic conditions, P. acnes with or without S. aureus formed a denser biomass biofilm than did S. aureus alone. Viable S. aureus was recovered from a16-day old combined P. acnes and S. aureus biofilm, but not a monomicrobial S. aureus biofilm.
Collapse
Affiliation(s)
- Harmony Tyner
- Division of Infectious Diseases, Department of Medicine, USA
| | - Robin Patel
- Division of Infectious Diseases, Department of Medicine, USA; Division Clinical Microbiology, Department of Laboratory Medicine and Pathology, USA.
| |
Collapse
|
28
|
Cisewski SE, Zhang L, Kuo J, Wright GJ, Wu Y, Kern MJ, Yao H. The effects of oxygen level and glucose concentration on the metabolism of porcine TMJ disc cells. Osteoarthritis Cartilage 2015; 23:1790-6. [PMID: 26033165 PMCID: PMC4577453 DOI: 10.1016/j.joca.2015.05.021] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/17/2014] [Revised: 04/30/2015] [Accepted: 05/21/2015] [Indexed: 02/02/2023]
Abstract
OBJECTIVE To determine the combined effect of oxygen level and glucose concentration on cell viability, ATP production, and matrix synthesis of temporomandibular joint (TMJ) disc cells. DESIGN TMJ disc cells were isolated from pigs aged 6-8 months and cultured in a monolayer. Cell cultures were preconditioned for 48 h with 0, 1.5, 5, or 25 mM glucose DMEM under 1%, 5%, 10%, or 21% O2 level, respectively. The cell viability was measured using the WST-1 assay. ATP production was determined using the Luciferin-Luciferase assay. Collagen and proteoglycan synthesis were determined by measuring the incorporation of [2, 3-(3)H] proline and [(35)S] sulfate into the cells, respectively. RESULTS TMJ disc cell viability significantly decreased (P < 0.0001) without glucose. With glucose present, decreased oxygen levels significantly increased viability (P < 0.0001), while a decrease in glucose concentration significantly decreased viability (P < 0.0001). With glucose present, decreasing oxygen levels significantly reduced ATP production (P < 0.0001) and matrix synthesis (P < 0.0001). A decreased glucose concentration significantly decreased collagen synthesis (P < 0.0001). The interaction between glucose and oxygen was significant in regards to cell viability (P < 0.0001), ATP production (P = 0.00015), and collagen (P = 0.0002) and proteoglycan synthesis (P < 0.0001). CONCLUSIONS Although both glucose and oxygen are important, glucose is the limiting nutrient for TMJ disc cell survival. At low oxygen levels, the production of ATP, collagen, and proteoglycan are severely inhibited. These results suggest that steeper nutrient gradients may exist in the TMJ disc and it may be vulnerable to pathological events that impede nutrient supply.
Collapse
Affiliation(s)
| | - Lixia Zhang
- Department of Oral Health Sciences, Medical University of South Carolina (MUSC), Charleston, SC
| | - Jonathan Kuo
- Department of Bioengineering, Clemson University, Clemson, SC
| | | | - Yongren Wu
- Department of Bioengineering, Clemson University, Clemson, SC
| | - Michael J. Kern
- Department of Oral Health Sciences, Medical University of South Carolina (MUSC), Charleston, SC
| | - Hai Yao
- Department of Bioengineering, Clemson University, Clemson, SC
,Department of Oral Health Sciences, Medical University of South Carolina (MUSC), Charleston, SC
| |
Collapse
|
29
|
Collins JA, Moots RJ, Clegg PD, Milner PI. Resveratrol and N-acetylcysteine influence redox balance in equine articular chondrocytes under acidic and very low oxygen conditions. Free Radic Biol Med 2015; 86:57-64. [PMID: 25998424 PMCID: PMC4562226 DOI: 10.1016/j.freeradbiomed.2015.05.008] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/16/2014] [Revised: 05/05/2015] [Accepted: 05/08/2015] [Indexed: 12/16/2022]
Abstract
Mature articular cartilage is an avascular tissue characterized by a low oxygen environment. In joint disease, acidosis and further reductions in oxygen levels occur, compromising cartilage integrity.This study investigated how acidosis and very low oxygen levels affect components of the cellular redox system in equine articular chondrocytesand whether the antioxidants resveratrol and N-acetylcysteine could modulate this system. We used articular chondrocytes isolated from nondiseased equine joints and cultured them in a 3-D alginate bead system for 48h in <1, 2, 5, and 21% O2 at pH 7.2 or 6.2 in the absence or presence of the proinflammatory cytokine, interleukin-1β (10ng/ml).In addition, chondrocytes were cultured with resveratrol (10µM) or N-acetylcysteine (NAC) (2mM).Cell viability, glycosaminoglycan (GAG) release, mitochondrial membrane potential (ΔΨm), reactive oxygen species (ROS), GSH:GSSG ratio, and SOD1 and SOD2 protein expression were measured. Very low levels of oxygen (<1%), acidosis (pH 6.2), and exposure to IL-1β led to reductions in cell viability, increased GAG release, alterations in ΔΨm and ROS levels, and reduced GSH:GSSG ratio. In addition, SOD1 and SOD2 protein expressions were reduced. Both resveratrol and NAC partially restored ΔΨm and ROS levels and prevented GAG release and cell loss and normalized SOD1 and SOD2 protein expression. In particular NAC was highly effective at restoring the GSH:GSSG ratio.These results show that the antioxidants resveratrol and N-acetylcysteine can counteract the redox imbalance in articular chondrocytes induced by low oxygen and acidic conditions.
Collapse
Affiliation(s)
- John A Collins
- Institute of Ageing and Chronic Disease, University of Liverpool, Leahurst Campus, Cheshire, UK, CH64 7TE
| | - Robert J Moots
- Institute of Ageing and Chronic Disease, University of Liverpool, University Hospital Aintree, Liverpool, UK, L9 7AL
| | - Peter D Clegg
- Institute of Ageing and Chronic Disease, University of Liverpool, Leahurst Campus, Cheshire, UK, CH64 7TE
| | - Peter I Milner
- Institute of Ageing and Chronic Disease, University of Liverpool, Leahurst Campus, Cheshire, UK, CH64 7TE.
| |
Collapse
|
30
|
In vitro engineering of human 3D chondrosarcoma: a preclinical model relevant for investigations of radiation quality impact. BMC Cancer 2015; 15:579. [PMID: 26253487 PMCID: PMC4529727 DOI: 10.1186/s12885-015-1590-5] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2015] [Accepted: 07/31/2015] [Indexed: 12/22/2022] Open
Abstract
Background The benefit of better ballistic and higher efficiency of carbon ions for cancer treatment (hadron-therapy) is asserted since decades, especially for unresectable or resistant tumors like sarcomas. However, hadron-therapy with carbon ions stays underused and raises some concerns about potential side effects for patients. Chondrosarcoma is a cartilaginous tumor, chemo- and radiation-resistant, that lacks reference models for basic and pre-clinical studies in radiation-biology. Most studies about cellular effects of ionizing radiation, including hadrons, were performed under growth conditions dramatically different from human homeostasis. Tridimensional in vitro models are a fair alternative to animal models to approach tissue and tumors microenvironment. Methods By using a collagen matrix, standardized culture conditions, physiological oxygen tension and a well defined chondrosarcoma cell line, we developed a pertinent in vitro 3D model for hadron-biology studies. Low- and high-Linear Energy Transfer (LET) ionizing radiations from GANIL facilities of ~1 keV/μm and 103 ± 4 keV/μm were used respectively, at 2 Gy single dose. The impact of radiation quality on chondrosarcoma cells cultivated in 3D was analyzed on cell death, cell proliferation and DNA repair. Results A fair distribution of chondrosarcoma cells was observed in the whole 3D scaffold. Moreover, LET distribution in depth, for ions, was calculated and found acceptable for radiation-biology studies using this kind of scaffold. No difference in cell toxicity was observed between low- and high-LET radiations but a higher rate of proliferation was displayed following high-LET irradiation. Furthermore, 3D models presented a higher and longer induction of H2AX phosphorylation after 2 Gy of high-LET compared to low-LET radiations. Conclusions The presented results show the feasibility and usefulness of our 3D chondrosarcoma model in the study of the impact of radiation quality on cell fate. The observed changes in our tissue-like model after ionizing radiation exposure may explain some discrepancies between radiation-biology studies and clinical data. Electronic supplementary material The online version of this article (doi:10.1186/s12885-015-1590-5) contains supplementary material, which is available to authorized users.
Collapse
|
31
|
Torrero JI, Martínez C. New developments in the treatment of osteoarthritis - focus on biologic agents. Open Access Rheumatol 2015; 7:33-43. [PMID: 27790043 PMCID: PMC5045124 DOI: 10.2147/oarrr.s50058] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
Osteoarthritis (OA) is one of the most common diseases around the world. Medical, social, and financial consequences oblige clinicians, surgeons, and researchers to focus on finding the best treatment option, to eradicate and stop this degenerative joint disease, in order to avoid surgical options which in many instances are over-indicated. Noninvasive treatments, such as anti-inflammatory drugs, physiotherapy, orthotic devices, dietary supplements, have demonstrated lack of effectiveness. The possibility to perform intra-articular injections with hyaluronic acid, corticosteroids, or the newest but criticized treatment based on platelet-rich plasma (PRP) has changed the management of OA disease. The use of PRP has led to many differences in treatment since there is a lack of consensus about protocols, indications, number of doses, cost-effectiveness, and duration of the treatment. Many publications have suggested efficacy in tendon injuries, but when PRP has been indicated to treat cartilage injuries, things are more inconsistent. Some authors have reported their experience treating OA with PRP, and it seems that, if well indicated, it is an option as a supplementary therapy. Therefore, we need to understand that OA is a mechanical disease which not only produces changes in radiographs, but also affects the quality of life. Pathogenesis of OA has been well explained, providing us new knowledge and future possibilities to improve the clinical approach. From basic science to surgery, there is a great field we all need to contribute to, because the general population is aging and total joint replacements should not be the only solution for OA. So herein is an actual review of the developments for treating OA with biologics, intended to be useful for the population inside orthopedics who could be called bio-orthopedists, since OA is a molecular homeostasis disbalance between catabolism and anabolism triggered by mechanical stress.
Collapse
Affiliation(s)
| | - Carlos Martínez
- University of Illinois Hospital and Health Sciences System, Chicago, IL, USA
| |
Collapse
|
32
|
Directing Chondrogenesis of Primary Chondrocytes by Exposure to Glucose Concentrations. JOURNAL OF BIOMIMETICS BIOMATERIALS AND BIOMEDICAL ENGINEERING 2015. [DOI: 10.4028/www.scientific.net/jbbbe.24.30] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Since articular cartilage is avascular, both nutrient supply and metabolic waste excretion depend on diffusion. However, the major cause of the progression of articular cartilage defect is the poor inherent regenerative capacity of chondrocytes which limits the process of cartilage tissue repair. Creation of nutrient gradients in in vitro cell culture, however, can provide a clue on zonal distributions of cells and glycosaminoglycan synthesis throughout the tissue engineered cartilage. We hypothesized that glucose gradient, in combination with growth factors, could induce differences in matrix distributions for articular cartilage regeneration. Chondrocytes were harvested from bovine cartilage and expanded in monolayers. First, either p0 or p2 chondrocytes were differentiated in serum-free chondrogenic medium containing different glucose concentrations supplemented with TGFβ3/dex or IGF-1under hypoxic or normoxic conditions for 7 days in monolayer. The results indicate that cellular metabolism, cell numbers and glycosaminoglycan (GAG) content increased with increase in glucose concentration in all conditions. Aggrecan (AGC) expression consistently increased with decreasing glucose concentration in both normoxic and hypoxic conditions. COL II and COL I expressions increased with increasing glucose concentration up to 5mmol/L. The expression of COMP increased with increasing glucose concentration under hypoxic conditions and interestingly showed an opposite trend under normoxic conditions. However, comparing the chondrogenic capacity of p0 and p2 cells in the different glucose concentrations did not show differences, but the potential of p2 cells was in general lower compared to p0. Hypoxia had stimulatory effects on matrix production compared to normoxia in both passages. Therefore, supplemented glucose concentration in monolayer could induce differences in matrix production, but the chondrogenic potential remained equal. Therefore, this information could be use to a create gradients through a tissue-engineered cartilage.
Collapse
|
33
|
Saintigny Y, Cruet-Hennequart S, Hamdi DH, Chevalier F, Lefaix JL. Impact of Therapeutic Irradiation on Healthy Articular Cartilage. Radiat Res 2015; 183:135-46. [DOI: 10.1667/rr13928.1] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
|
34
|
Alexander PG, Gottardi R, Lin H, Lozito TP, Tuan RS. Three-dimensional osteogenic and chondrogenic systems to model osteochondral physiology and degenerative joint diseases. Exp Biol Med (Maywood) 2014; 239:1080-95. [PMID: 24994814 DOI: 10.1177/1535370214539232] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Tissue engineered constructs have the potential to function as in vitro pre-clinical models of normal tissue function and disease pathogenesis for drug screening and toxicity assessment. Effective high throughput assays demand minimal systems with clearly defined performance parameters. These systems must accurately model the structure and function of the human organs and their physiological response to different stimuli. Musculoskeletal tissues present unique challenges in this respect, as they are load-bearing, matrix-rich tissues whose functionality is intimately connected to the extracellular matrix and its organization. Of particular clinical importance is the osteochondral junction, the target tissue affected in degenerative joint diseases, such as osteoarthritis (OA), which consists of hyaline articular cartilage in close interaction with subchondral bone. In this review, we present an overview of currently available in vitro three-dimensional systems for bone and cartilage tissue engineering that mimic native physiology, and the utility and limitations of these systems. Specifically, we address the need to combine bone, cartilage and other tissues to form an interactive microphysiological system (MPS) to fully capture the biological complexity and mechanical functions of the osteochondral junction of the articular joint. The potential applications of three-dimensional MPSs for musculoskeletal biology and medicine are highlighted.
Collapse
Affiliation(s)
- Peter G Alexander
- Center for Cellular and Molecular Engineering, Department of Orthopaedic Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA 15219, USA McGowan Institute for Regenerative Medicine, University of Pittsburgh, PA, 15219 USA
| | - Riccardo Gottardi
- Center for Cellular and Molecular Engineering, Department of Orthopaedic Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA 15219, USA McGowan Institute for Regenerative Medicine, University of Pittsburgh, PA, 15219 USA Ri.MED Foundation, Palermo, I-90133 Italy
| | - Hang Lin
- Center for Cellular and Molecular Engineering, Department of Orthopaedic Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA 15219, USA McGowan Institute for Regenerative Medicine, University of Pittsburgh, PA, 15219 USA
| | - Thomas P Lozito
- Center for Cellular and Molecular Engineering, Department of Orthopaedic Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA 15219, USA McGowan Institute for Regenerative Medicine, University of Pittsburgh, PA, 15219 USA
| | - Rocky S Tuan
- Center for Cellular and Molecular Engineering, Department of Orthopaedic Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA 15219, USA McGowan Institute for Regenerative Medicine, University of Pittsburgh, PA, 15219 USA Department of Bioengineering, University of Pittsburgh Swanson School of Engineering, Pittsburgh, PA 15261, USA Department of Mechanical Engineering and Materials Science, University of Pittsburgh Swanson School of Engineering, Pittsburgh, PA 15261, USA
| |
Collapse
|
35
|
Juranek I, Stern R, Soltes L. Hyaluronan peroxidation is required for normal synovial function: an hypothesis. Med Hypotheses 2014; 82:662-6. [PMID: 24655797 DOI: 10.1016/j.mehy.2014.02.024] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2013] [Revised: 02/14/2014] [Accepted: 02/23/2014] [Indexed: 12/11/2022]
Abstract
Despite widespread use of antioxidants, reactive oxygen species have important functions in normal tissues. Herein, we present an example of a physiological role for free radicals, and in particular, reactive oxygen species, that are suppressed by anti-oxidants. Free radicals catalyze the degradation of hyaluronan in synovial fluid, a tissue in which hyaluronidase activity is barely detectable. Articular cartilage requires a low oxygen environment. The process of hyaluronan peroxidation consumes significant amounts of molecular oxygen, thus keeping the tension of oxygen in the joint at a low but physiologically critical level. One concern is the change in physical activity between day and night, with periods of joint hyperemia and ischemia, respectively. Increased oxygen and the resulting oxidative stress would lead to chondrocyte dysfunction and cartilage damage. A mechanism for keeping oxygen levels low is required. We postulate that a mechanism indeed exists for the removal of excess oxygen. High-molar-mass hyaluronan turnover in synovial fluid utilizes peroxidative degradation, during which oxygen is massively consumed. The peroxidation itself may be initiated by hydrogen peroxide, which is produced by chondrocyte mitochondria, that can diffuse into the synovial fluid. The resulting decrease in available oxygen down-regulates hyaluronan peroxidation. This in turn prevents excessive oxygen consumption. It appears that free radicals and reactive oxygen species may be components of normal physiology, particularly in the synovial fluid of joints and articular cartilage. It is suggested therefore that indiscriminate use of anti-oxidants, vigorously promoted currently by health professionals and the health industry, be approached with caution.
Collapse
Affiliation(s)
- I Juranek
- Institute of Experimental Pharmacology and Toxicology, Slovak Academy of Sciences, Dubravska Cesta 9, SK-84104 Bratislava, Slovakia
| | - R Stern
- Department of Basic Biomedical Sciences, Touro College of Osteopathic Medicine, 230 West-125th St., New York, NY 10027, USA.
| | - L Soltes
- Institute of Experimental Pharmacology and Toxicology, Slovak Academy of Sciences, Dubravska Cesta 9, SK-84104 Bratislava, Slovakia
| |
Collapse
|
36
|
Collins J, Moots R, Winstanley R, Clegg P, Milner P. Oxygen and pH-sensitivity of human osteoarthritic chondrocytes in 3-D alginate bead culture system. Osteoarthritis Cartilage 2013; 21:1790-8. [PMID: 23850530 PMCID: PMC3807787 DOI: 10.1016/j.joca.2013.06.028] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2013] [Revised: 06/25/2013] [Accepted: 06/27/2013] [Indexed: 02/02/2023]
Abstract
OBJECTIVE To identify the effect of alterations in physical parameters such as oxygen and pH on processes associated with cellular redox balance in osteoarthritic chondrocytes. METHOD Human osteoarthritic chondrocytes (HOAC) were isolated from total knee arthroplasty samples and cultured in 3-D alginate beads in four different oxygen tensions (<1%, 2%, 5% and 21% O2), at pH 7.2 and 6.2 and in the presence or absence of 10 ng/ml, interleukin-1β (IL-1β). Cell viability, media glycosaminoglycan (GAG) levels, media nitrate/nitrate levels, active matrix metalloproteinase (MMP)-13 and intracellular adenosine triphosphate (ATPi) were measured over a 96-h time course. Intracellular reactive oxygen species (ROS), mitochondrial membrane potential, intracellular pH and reduced/oxidised glutathione (GSH/GSSG) were additionally measured after 48-h incubation under these experimental conditions. RESULTS Hypoxia (2% O2) and anoxia (<1% O2), acidosis (pH 6.2) and 10 ng/ml IL-1β reduced HOAC cell viability and increased GAG media levels. Acidosis and IL-1β increased nitrite/nitrate release, but increases were moderate at 2% O2 and significantly reduced at <1% O2. ATPi was significantly reduced following hypoxia and anoxia and acidosis. At 48 h cellular ROS levels were increased by acidosis and IL-1β but reduced in hypoxia and anoxia. Mitochondrial membrane potential was reduced in low oxygen, acidosis and IL-1β. Anoxia also resulted in intracellular acidosis. GSH/GSSG ratio was reduced in low oxygen conditions, acidosis and IL-1β. CONCLUSIONS This study shows that oxygen and pH affect elements of the redox system in HOAC including cellular anti-oxidants, mitochondrial membrane potential and ROS levels.
Collapse
Affiliation(s)
- J.A. Collins
- Department of Musculoskeletal Biology, Institute of Ageing and Chronic Disease, University of Liverpool, Leahurst Campus, Neston, Cheshire CH64 7TE, UK
| | - R.J. Moots
- Department of Musculoskeletal Biology, Institute of Ageing and Chronic Disease, University of Liverpool, University Hospital, Aintree, Liverpool L9 7AL, UK
| | - R. Winstanley
- Department of Musculoskeletal Biology, Institute of Ageing and Chronic Disease, University of Liverpool, Leahurst Campus, Neston, Cheshire CH64 7TE, UK
| | - P.D. Clegg
- Department of Musculoskeletal Biology, Institute of Ageing and Chronic Disease, University of Liverpool, Leahurst Campus, Neston, Cheshire CH64 7TE, UK
| | - P.I. Milner
- Department of Musculoskeletal Biology, Institute of Ageing and Chronic Disease, University of Liverpool, Leahurst Campus, Neston, Cheshire CH64 7TE, UK,Address correspondence and reprint requests to: P.I. Milner, Department of Musculoskeletal Biology, Institute of Ageing and Chronic Disease, University of Liverpool, Leahurst Campus, Neston, Cheshire CH64 7TE, UK. Tel: 44-151-7946041; Fax: 44-151-7946034.
| |
Collapse
|
37
|
Parathyroid hormone-related protein is induced by hypoxia and promotes expression of the differentiated phenotype of human articular chondrocytes. Clin Sci (Lond) 2013; 125:461-70. [PMID: 23662774 DOI: 10.1042/cs20120610] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
PTHrP (parathyroid hormone-related protein) is crucial for normal cartilage development and long bone growth and acts to delay chondrocyte hypertrophy and terminal differentiation in the growth plate. After growth plate closure adult HACs (human articular chondrocytes) still produce PTHrP, suggesting a possible role for this factor in the permanent articular cartilage. However, the expression regulation and function of PTHrP in the permanent articular cartilage is unknown. Human articular cartilage is an avascular tissue and functions in a hypoxic environment. The resident chondrocytes have adapted to hypoxia and use it to drive their tissue-specific functions. In the present study, we explored directly in normal articular chondrocytes isolated from a range of human donors the effect of hypoxia on PTHrP expression and whether PTHrP can regulate the expression of the permanent articular chondrocyte phenotype. We show that in HACs PTHrP is up-regulated by hypoxia in a HIF (hypoxia-inducible factor)-1α and HIF-2α-dependent manner. Using recombinant PTHrP, siRNA-mediated depletion of endogenous PTHrP and by blocking signalling through its receptor [PTHR1 (PTHrP receptor 1)], we show that hypoxia-induced PTHrP is a positive regulator of the key cartilage transcription factor SOX9 [SRY (sex determining region on the Y chromosome)-box 9], leading to increased COL2A1 (collagen type II, α1) expression. Our findings thus identify PTHrP as a potential factor for cartilage repair therapies through its ability to promote the differentiated HAC phenotype.
Collapse
|
38
|
Abstract
The degenerative joint disease osteoarthritis (OA) is the most prevalent form of chronic musculoskeletal disease worldwide, and it commonly afflicts the elderly population. OA-induced impairment of joint function can debilitate normal physical activity, and in more severe cases, it can lead to complete joint destruction and loss of independence or even mobility. The pathophysiology of OA remains to be fully elucidated, despite the extensive research efforts into this complex disease. Studies have revealed that reactive oxygen species (ROS) can contribute to the onset and progression of OA by inducing indispensable chondrocyte death and matrix degradation. However, ROS are also key components of many normal physiological processes, and at moderate levels, they act as indispensable second messengers. This review focuses on the dual role of ROS in cartilage, with the aim of gaining insights into how ROS can be regulated such that its beneficial effects are maintained and its detrimental effects are eliminated.
Collapse
|
39
|
Stacey MW, Grubbs J, Asmar A, Pryor J, Elsayed-Ali H, Cao W, Beskok A, Dutta D, Darby DA, Fecteau A, Werner A, Kelly RE. Decorin expression, straw-like structure, and differentiation of human costal cartilage. Connect Tissue Res 2012; 53:415-21. [PMID: 22490077 DOI: 10.3109/03008207.2012.684113] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Costal cartilage is much understudied compared with the load-bearing cartilages. Abnormally grown costal cartilages are associated with the inherited chest wall deformities pectus excavatum and pectus carinatum resulting in sunken and pigeon chests, respectively. A lack of understanding of the ultrastructural and molecular biology of costal cartilage is a major confounder in predicting causes and outcomes of these disorders. This study analyzed the structure of marginal human costal cartilage (ribs 6-10) through scanning electron and atomic force microscopes and identified the presence of straw-like structures running longitudinally. We also demonstrated that chondrocytes tend to occur singly or as doublets and that centrally located cells produce high levels of aggrecan compared with more peripherally located cells measured using immunohistochemistry. Gene expression from mRNA extracted from cartilage showed high levels of decorin expression, likely associated with the large, complex tubular structures running through this cartilage type. COL2A1, ACAN, and TIMP1 also showed higher levels of expression compared with ACTB. Analysis of gene expression ratios demonstrate that costal cartilage is under differentiated compared with published ratios for articular cartilage, likely due to the vastly different biomechanical environments of each cartilage type. Further studies need to establish whether findings described here from the costal margins are significantly different than the cartilage of the "true ribs" and how these values change with age.
Collapse
Affiliation(s)
- M W Stacey
- Frank Reidy Research Center for Bioelectrics, Old Dominion University, Norfolk, VA 23508, USA.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Kang H, Peng J, Lu S, Liu S, Zhang L, Huang J, Sui X, Zhao B, Wang A, Xu W, Luo Z, Guo Q. In vivo cartilage repair using adipose-derived stem cell-loaded decellularized cartilage ECM scaffolds. J Tissue Eng Regen Med 2012; 8:442-53. [PMID: 22674864 DOI: 10.1002/term.1538] [Citation(s) in RCA: 98] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2011] [Revised: 02/29/2012] [Accepted: 04/18/2012] [Indexed: 12/12/2022]
Abstract
We have previously reported a natural, human cartilage ECM (extracellular matrix)-derived three-dimensional (3D) porous acellular scaffold for in vivo cartilage tissue engineering in nude mice. However, the in vivo repair effects of this scaffold are still unknown. The aim of this study was to further explore the feasibility of application of cell-loaded scaffolds, using autologous adipose-derived stem cells (ADSCs), for cartilage defect repair in rabbits. A defect 4 mm in diameter was created on the patellar groove of the femur in both knees, and was repaired with the chondrogenically induced ADSC-scaffold constructs (group A) or the scaffold alone (group B); defects without treatment were used as controls (group C). The results showed that in group A all defects were fully filled with repair tissue and at 6 months post-surgery most of the repair site was filled with hyaline cartilage. In contrast, in group B all defects were partially filled with repair tissue, but only half of the repair tissue was hyaline cartilage. Defects were only filled with fibrotic tissue in group C. Indeed, histological grading score analysis revealed that an average score in group A was higher than in groups B and C. GAG and type II collagen content and biomechanical property detection showed that the group A levels approached those of normal cartilage. In conclusion, ADSC-loaded cartilage ECM scaffolds induced cartilage repair tissue comparable to native cartilage in terms of mechanical properties and biochemical components.
Collapse
Affiliation(s)
- Hongjun Kang
- Key Laboratory of PLA, Chinese PLA General Hospital, Beijing, People's Republic of China
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Ziskoven C, Jäger M, Zilkens C, Bloch W, Brixius K, Krauspe R. Oxidative stress in secondary osteoarthritis: from cartilage destruction to clinical presentation? Orthop Rev (Pavia) 2011; 2:e23. [PMID: 21808712 PMCID: PMC3143971 DOI: 10.4081/or.2010.e23] [Citation(s) in RCA: 72] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/21/2010] [Accepted: 11/29/2010] [Indexed: 12/21/2022] Open
Abstract
Due to an increasing life expectance, osteoarthritis (OA) is one of the most common chronic diseases. Although strong efforts have been made to regenerate degenerated joint cartilage, OA is a progressive and irreversible disease up to date. Among other factors the dysbalance between free radical burden and cellular scavenging mechanisms defined as oxidative stress is a relevant part of OA pathogenesis. Here, only little data are available about the mediation and interaction between different joint compartments. The article provides a review of the current literature regarding the influence of oxidative stress on cellular aging, senescence and apoptosis in different joint compartments (cartilage, synovial tissue and subchondral bone). Free radical exposure is known to promote cellular senescence and apoptosis. Radical oxygen species (ROS) involvement in inflammation, fibrosis control and pain nociception has been proven. The data from literature indicates a link between free radical burden and OA pathogenesis mediating local tissue reactions between the joint compartments. Hence, oxidative stress is likely not only to promote cartilage destruction but also to be involved in inflammative transformation, promoting the transition from clinically silent cartilage destruction to apparent OA. ROS induced by exogenous factors such as overload, trauma, local intraarticular lesion and consecutive synovial inflammation cause cartilage degradation. In the affected joint, free radicals mediate disease progression. The interrelationship between oxidative stress and OA etiology might provide a novel approach to the comprehension and therefore modification of disease progression and symptom control.
Collapse
Affiliation(s)
- Christoph Ziskoven
- Orthopedic Department, Heinrich-Heine University Medical School, Düsseldorf, Germany
| | | | | | | | | | | |
Collapse
|
42
|
Hirose J, Yamabe S, Takada K, Okamoto N, Nagai R, Mizuta H. Immunohistochemical distribution of advanced glycation end products (AGEs) in human osteoarthritic cartilage. Acta Histochem 2011; 113:613-8. [PMID: 20656335 DOI: 10.1016/j.acthis.2010.06.007] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2010] [Revised: 06/21/2010] [Accepted: 06/22/2010] [Indexed: 01/11/2023]
Abstract
Advanced glycation end products (AGEs) may be associated with osteoarthritis (OA), because the accumulation of AGEs in articular cartilage are among the most striking age-related changes. AGEs modify the tissue protein structure and function and stimulate the cellular responses mediated by a specific receptor for AGEs (RAGE). This study investigated the localization of AGEs in degenerated cartilage using newly identified epitope-specific antibodies to determine the linkage between the distribution of AGEs and the development and progression of OA. Osteochondral specimens of the tibial plateau from OA patients were immunostained by specific antibodies against N(ε)-(carboxymethyl)lysine (CML), N(ε)-(carboxyethyl)lysine (CEL), pentosidine, GA-pyridine, and RAGE. The immunohistochemical distribution of these epitopes was evaluated during cartilage degeneration. The immunoreactivity (IR) of AGEs and RAGE was stronger in cells rather than in the extracellular matrix. Higher IR of cellular CML and CEL was observed in both mild and severe OA cartilage in comparison to macroscopically intact cartilage. There was a strong association between GA-pyridine and RAGE in the pattern of increasing IR with the OA grade. These IR patterns of AGEs varying with cartilage degeneration indicate that AGE modified proteins are associated with cartilage degeneration. The coincidental up-regulation of GA-pyridine and RAGE suggests that GA-pyridine is the most significant AGE for cartilage degeneration via the RAGE pathway.
Collapse
Affiliation(s)
- Jun Hirose
- Department of Orthopaedic Surgery, Kumamoto University Hospital, Honjo, Japan.
| | | | | | | | | | | |
Collapse
|
43
|
Terkeltaub R, Yang B, Lotz M, Liu-Bryan R. Chondrocyte AMP-activated protein kinase activity suppresses matrix degradation responses to proinflammatory cytokines interleukin-1β and tumor necrosis factor α. ACTA ACUST UNITED AC 2011; 63:1928-37. [PMID: 21400477 DOI: 10.1002/art.30333] [Citation(s) in RCA: 133] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
OBJECTIVE Interleukin-1β (IL-1β) and tumor necrosis factor α (TNFα) stimulate chondrocyte matrix catabolic responses, thereby compromising cartilage homeostasis in osteoarthritis (OA). AMP-activated protein kinase (AMPK), which regulates energy homeostasis and cellular metabolism, also exerts antiinflammatory effects in multiple tissues. This study was undertaken to test the hypothesis that AMPK activity limits chondrocyte matrix catabolic responses to IL-1β and TNFα. METHODS Expression of AMPK subunits was examined, and AMPKα activity was ascertained by the phosphorylation status of AMPKα Thr(172) in human knee articular chondrocytes and cartilage by Western blotting and immunohistochemistry, respectively. Procatabolic responses to IL-1β and TNFα, such as release of glycosaminoglycan, nitric oxide, and matrix metalloproteinases 3 and 13 were determined by dimethylmethylene blue assay, Griess reaction, and Western blotting, respectively, in cartilage explants and chondrocytes with and without knockdown of AMPKα by small interfering RNA. RESULTS Normal human knee articular chondrocytes expressed AMPKα1, α2, β1, β2, and γ1 subunits. AMPK activity was constitutively present in normal articular chondrocytes and cartilage, but decreased in OA articular chondrocytes and cartilage and in normal chondrocytes treated with IL-1β and TNFα. Knockdown of AMPKα resulted in enhanced catabolic responses to IL-1β and TNFα in chondrocytes. Moreover, AMPK activators suppressed cartilage/chondrocyte procatabolic responses to IL-1β and TNFα and the capacity of TNFα and CXCL8 (IL-8) to induce type X collagen expression. CONCLUSION Our findings indicate that AMPK activity is reduced in OA cartilage and in chondrocytes following treatment with IL-1β or TNFα. AMPK activators attenuate dephosphorylation of AMPKα and procatabolic responses in chondrocytes induced by these cytokines. These observations suggest that maintenance of AMPK activity supports cartilage homeostasis by protecting cartilage matrix from inflammation-induced degradation.
Collapse
Affiliation(s)
- Robert Terkeltaub
- VA San Diego Medical Center and University of California, San Diego, CA, USA
| | | | | | | |
Collapse
|
44
|
Botter SM, Zar M, van Osch GJVM, van Steeg H, Dollé MET, Hoeijmakers JHJ, Weinans H, van Leeuwen JPTM. Analysis of osteoarthritis in a mouse model of the progeroid human DNA repair syndrome trichothiodystrophy. AGE (DORDRECHT, NETHERLANDS) 2011; 33:247-260. [PMID: 20820927 PMCID: PMC3168596 DOI: 10.1007/s11357-010-9175-3] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/28/2010] [Accepted: 08/18/2010] [Indexed: 05/29/2023]
Abstract
The increasing average age in developed societies is paralleled by an increase in the prevalence of many age-related diseases such as osteoarthritis (OA), which is characterized by deformation of the joint due to cartilage damage and increased turnover of subchondral bone. Consequently, deficiency in DNA repair, often associated with premature aging, may lead to increased pathology of these two tissues. To examine this possibility, we analyzed the bone and cartilage phenotype of male and female knee joints derived from 52- to 104-week-old WT C57Bl/6 and trichothiodystrophy (TTD) mice, who carry a defect in the nucleotide excision repair pathway and display many features of premature aging. Using micro-CT, we found bone loss in all groups of 104-week-old compared to 52-week-old mice. Cartilage damage was mild to moderate in all mice. Surprisingly, female TTD mice had less cartilage damage, proteoglycan depletion, and osteophytosis compared to WT controls. OA severity in males did not significantly differ between genotypes, although TTD males had less osteophytosis. These results indicate that in premature aging TTD mice age-related changes in cartilage were not more severe compared to WT mice, in striking contrast with bone and many other tissues. This segmental aging character may be explained by a difference in vasculature and thereby oxygen load in cartilage and bone. Alternatively, a difference in impact of an anti-aging response, previously found to be triggered by accumulation of DNA damage, might help explain why female mice were protected from cartilage damage. These findings underline the exceptional segmental nature of progeroid conditions and provide an explanation for pro- and anti-aging features occurring in the same individual.
Collapse
Affiliation(s)
- Sander M. Botter
- Department of Internal Medicine, Erasmus MC, University Medical Centre Rotterdam, EE585, P.O. Box 2040, 3000 CA Rotterdam, The Netherlands
- Department of Orthopaedics, Erasmus MC, University Medical Centre Rotterdam, Rotterdam, The Netherlands
| | - Michel Zar
- Department of Orthopaedics, Erasmus MC, University Medical Centre Rotterdam, Rotterdam, The Netherlands
| | - Gerjo J. V. M van Osch
- Department of Orthopaedics, Erasmus MC, University Medical Centre Rotterdam, Rotterdam, The Netherlands
- Department of Otorhinolaryngology, Erasmus MC, University Medical Centre Rotterdam, Rotterdam, The Netherlands
| | - Harry van Steeg
- National Institute of Public Health and the Environment, Laboratory for Health Protection Research, Bilthoven, The Netherlands
| | - Martijn E. T. Dollé
- National Institute of Public Health and the Environment, Laboratory for Health Protection Research, Bilthoven, The Netherlands
| | - Jan H. J. Hoeijmakers
- Department of Cell biology and Genetics, Medical Genetics Centre, Erasmus MC, University Medical Centre Rotterdam, Rotterdam, The Netherlands
| | - Harrie Weinans
- Department of Orthopaedics, Erasmus MC, University Medical Centre Rotterdam, Rotterdam, The Netherlands
| | - Johannes P. T. M. van Leeuwen
- Department of Internal Medicine, Erasmus MC, University Medical Centre Rotterdam, EE585, P.O. Box 2040, 3000 CA Rotterdam, The Netherlands
| |
Collapse
|
45
|
Ziskoven C, Jäger M, Kircher J, Patzer T, Bloch W, Brixius K, Krauspe R. Physiology and pathophysiology of nitrosative and oxidative stress in osteoarthritic joint destruction. Can J Physiol Pharmacol 2011; 89:455-66. [PMID: 21793696 DOI: 10.1139/y11-055] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Osteoarthritis (OA) is one of the most common chronic diseases, with increasing importance due to increased life expectancy. On a cellular level, the pathophysiology of joint function impairment and ultimate destruction associated with OA remains poorly understood. Free radicals are highly reactive molecules involved in both normal intracellular signal transduction and degenerative cellular processes. An imbalance between the free radical burden and cellular scavenging mechanisms, defined as oxidative stress, has been identified as a relevant factor in OA pathogenesis. This literature review elucidates the involvement of nitrosative and oxidative stress in cellular ageing in joints, cell senescence, and apoptosis. Free radical exposure is known to promote cellular senescence and apoptosis, and the involvement of radical oxygen species (ROS) in inflammation, fibrosis control, and pain nociception has been proven. A relatively novel approach to OA pathophysiology considers the joint to be a dynamic system consisting of 3, continuously interacting compartments, cartilage, synovial tissue, and subchondral bone. Current knowledge concerning free radical involvement in paracrine signalling in OA is reviewed. The interrelationship between oxidative imbalances and OA pathophysiology may provide a novel approach to the comprehension, and therefore modification, of OA disease progression and symptom control.
Collapse
Affiliation(s)
- Christoph Ziskoven
- Orthopedic Department, Heinrich-Heine University Medical School, Düsseldorf, Germany
| | | | | | | | | | | | | |
Collapse
|
46
|
Schultz M, Jin W, Waheed A, Moed BR, Sly W, Zhang Z. Expression profile of carbonic anhydrases in articular cartilage. Histochem Cell Biol 2011; 136:145-51. [PMID: 21739214 DOI: 10.1007/s00418-011-0836-9] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/26/2011] [Indexed: 12/30/2022]
Abstract
Carbonic anhydrases (CAs), which catalyze the reversible reaction of carbonate hydration, are important for cartilage homeostasis. The full spectrum of CA activity of all 13 isoenzymes in articular cartilage is unknown. This study quantified the mRNA profile of CAs in rat articular cartilage, using quantitative polymerase chain reactions. Among the 13 functional CAs, CAs II, III, Vb, IX, XII and XIII were significantly expressed at mRNA level by the chondrocytes in articular cartilage. To verify these significantly expressed CAs in articular cartilage at protein level, immunohistochemistry was performed. While CAs III, Vb and XII distributed in the full-thickness of cartilage, including the calcified zone of cartilage, CA II was mainly localized in the proliferative zone of cartilage. CA IX was limited in the superficial zone of cartilage and CA XIII expressed in the superficial and partially mid zone. These results provide a framework for understanding individual CAs as well as the integrated CA family in cartilage biology, including matrix mineralization.
Collapse
Affiliation(s)
- Melissa Schultz
- Center for Anatomical Science and Education, Saint Louis University, St. Louis, MO, USA
| | | | | | | | | | | |
Collapse
|
47
|
Abstract
Oxygen is not only an obviously important substrate, but it is also a regulatory signal that controls expression of a specific genetic program. Crucial mediator of the adaptive response of cells to hypoxia is the family of Hypoxia-Inducible Transcription Factors (HIFs).The fetal growth plate, which is an avascular structure of mesenchymal origin, has a unique out-in gradient of oxygenation. HIF-1alpha is necessary for chondrogenesis in vivo by controlling a complex homeostatic response that allows chondrocytes to survive and differentiate in a hypoxic environment. Moreover, HIFs are also essential in osteogenesis and joint development. This brief Perspective summarizes the critical role of HIFs in endochondral bone development.
Collapse
|
48
|
White R, Gibson JS. The effect of oxygen tension on calcium homeostasis in bovine articular chondrocytes. J Orthop Surg Res 2010; 5:27. [PMID: 20420658 PMCID: PMC2873548 DOI: 10.1186/1749-799x-5-27] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/21/2009] [Accepted: 04/26/2010] [Indexed: 01/11/2023] Open
Abstract
Background Articular chondrocytes normally experience a lower O2 tension compared to that seen by many other tissues. This level may fall further in joint disease. Ionic homeostasis is essential for chondrocyte function but, at least in the case of H+ ions, it is sensitive to changes in O2 levels. Ca2+ homeostasis is also critical but the effect of changes in O2 tension has not been investigated on this parameter. Here we define the effect of hypoxia on Ca2+ homeostasis in bovine articular chondrocytes. Methods Chondrocytes from articular cartilage slices were isolated enzymatically using collagenase. Cytoplasmic Ca2+ levels ([Ca2+]i) were followed fluorimetrically using Fura-2 to determine the effect of changes in O2 tension. The effects of ion substitution (replacing extracellular Na+ with NMDG+ and chelating Ca2+ with EGTA) were tested. Levels of reactive oxygen species (ROS) and the mitochondrial membrane potential were measured and correlated with [Ca2+]i. Results A reduction in O2 tension from 20% to 1% for 16-18 h caused [Ca2+]i to approximately double, reaching 105 ± 23 nM (p < 0.001). Ion substitutions indicated that Na+/Ca2+ exchange activity was not inhibited at low O2 levels. At 1% O2, ROS levels fell and mitochondria depolarised. Restoring ROS levels (with an oxidant H2O2, a non-specific ROS generator Co2+ or the mitochondrial complex II inhibitor antimycin A) concomitantly reduced [Ca2+]i. Conclusions O2 tension exerts a significant effect on [Ca2+]i. The proposed mechanism involves ROS from mitochondria. Findings emphasise the importance of using realistic O2 tensions when studying the physiology and pathology of articular cartilage and the potential interactions between O2, ROS and Ca2+.
Collapse
Affiliation(s)
- Rachel White
- Department of Veterinary Medicine, University of Cambridge, Madingley Road, Cambridge, CB3 OES, UK.
| | | |
Collapse
|
49
|
Cillero-Pastor B, Ruiz-Romero C, Caramés B, López-Armada MJ, Blanco FJ. Proteomic analysis by two-dimensional electrophoresis to identify the normal human chondrocyte proteome stimulated by tumor necrosis factor alpha and interleukin-1beta. ACTA ACUST UNITED AC 2010; 62:802-14. [PMID: 20131227 DOI: 10.1002/art.27265] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
OBJECTIVE To determine the intracellular proteome of normal human chondrocytes stimulated with interleukin-1beta (IL-1beta) and tumor necrosis factor alpha (TNFalpha) and to ascertain differences in the protein expression patterns of these 2 cytokines. METHODS Normal human knee cartilage chondrocytes were incubated for 48 hours without stimulation or stimulated with IL-1beta (5 ng/ml) or with TNFalpha (10 ng/ml). For each culture condition, protein extracts from 4 normal subjects were pooled and resolved using 2-dimensional electrophoresis. Protein spots were visualized with Sypro stain, and qualitative and quantitative analyses were performed using PDQuest software. Protein spots were then identified by mass spectrometry, using matrix-assisted laser desorption ionization-time-of-flight/time-of-flight technology. RESULTS We identified 37 spots by mass spectrometry (MS) or by MS/MS, corresponding to 35 different proteins. In IL-1beta-stimulated chondrocytes, IL-1beta was found to modulate 22 proteins, as compared with unstimulated chondrocytes. All of these proteins except connective tissue growth factor (CCND2) were up-regulated. Proteins involved in cellular metabolism and energy (23%) that were up-regulated or induced by IL-1beta included nicotinamide phosphoribosyltransferase, long-chain fatty acid-coenzyme A ligase 4, delta-aminolevulinic acid dehydratase, triosephosphate isomerase, and an isoform of glyceraldehyde-3-phosphate dehydrogenase. In TNFalpha-stimulated chondrocytes, TNFalpha was found to modulate 20 proteins, as compared with unstimulated chondrocytes. All of these except chitinase 3-like 1 (cartilage glycoprotein 39), proteasome activator complex subunit 2, and G3PDH, were up-regulated. Eighteen proteins were differently modulated by IL-1beta and TNFalpha. Of these, 45% were related to metabolism. CONCLUSION IL-1beta and TNFalpha induce different profiles of intracellular protein expression in healthy human chondrocytes. Most of the proteins that are differently regulated are proteins that are implicated in the generation of cellular energy and in glycolysis.
Collapse
|
50
|
Oxygen tension regulates the expression of ANK (progressive ankylosis) in an HIF-1-dependent manner in growth plate chondrocytes. J Bone Miner Res 2009; 24:1869-78. [PMID: 19419319 PMCID: PMC2765931 DOI: 10.1359/jbmr.090512] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
The proximal promoter region of ANK, a gene that codes for a protein that regulates the transport of inorganic pyrophosphate, contains two hypoxia responsive elements (HREs); therefore, we studied the expression and function of ANK at different oxygen tensions. ATDC5 and N1511 clonal chondrocytic cells were cultured in either hypoxia (2% O(2)) or normoxia (21% O(2)). Transcript and protein levels of ANK were depressed in hypoxic conditions, as were levels of extracellular pyrophosphate (ePPi). To determine whether HIF-1 was involved in the oxemic response, Hif-1alpha knockdown cells were exposed to varying oxygen conditions and ANK expression was assessed. Knockdown of Hif-1alpha resulted in low levels of expression of ANK in hypoxia and normoxia. Chromatin immunoprecipitation (ChIP) assays explored the binding of Hif-1alpha to ANK HREs and showed that Hif-1alpha is able to bind to the HREs of ANK more avidly in normoxia than in hypoxia. Furthermore, functional studies of Hif-1alpha activity using luciferase reporter assays of wildtype and mutagenized HREs showed that only HRE-1 binds Hif-1alpha in normoxia. Expression of ANK in growth plate and articular cartilage was low in hypoxic regions of the tissues, and higher levels of ANK expression were observed in the synovium and meniscus in regions that have a normally higher oxygen tension. The data suggest that ANK expression and function in vitro and in vivo are repressed in hypoxic environments and that the effect is regulated by HIF-1.
Collapse
|