1
|
Hamilton CM, Winter MJ, Ball JS, Trznadel M, Margiotta-Casaluci L, Owen SF, Tyler CR. Exposure effects of synthetic glucocorticoid drugs on skeletal developmental and immune cell function in zebrafish. THE SCIENCE OF THE TOTAL ENVIRONMENT 2024; 955:176781. [PMID: 39395483 DOI: 10.1016/j.scitotenv.2024.176781] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Revised: 09/06/2024] [Accepted: 10/05/2024] [Indexed: 10/14/2024]
Abstract
Synthetic glucocorticoids (GCs) are used to treat a wide range of human health conditions and as such are frequently detected in the aquatic environment. This, together with the highly conserved nature of the glucocorticoid system across vertebrates means that the potential for biological effects of GCs in fish is relatively high. Here, we found that exposure of zebrafish (Danio rerio) to environmentally relevant concentrations of 4 of the most widely used synthetic GCs (beclomethasone dipropionate, budesonide, fluticasone propionate, and prednisolone), from 0 to 4 days post fertilisation (dpf), resulted in no effects on embryo-larval development or bone and cartilage formation. However, after exposure to equivalents of human therapeutic plasma levels, developmental abnormalities were observed that included pericardial oedema, blood pooling and alterations in jaw cartilage. Furthermore, using a double transgenic zebrafish osteoblast and chondrocyte reporter line, exposure up to 10 dpf resulted in alterations to lower jaw cartilage and bone development for all compounds at, and above, human therapeutic plasma concentrations. In the case of beclomethasone dipropionate, a reduction in lower jaw intercranial distance was observed at the environmentally relevant concentration of 0.1 μg/L. Using further transgenic reporter lines with fluorescently tagged neutrophils and macrophages, we also show exposure of embryo-larvae (0-4 dpf) to the GCs tested resulted in altered immune cell migration, but only at relatively high exposure concentrations. Collectively, our findings show GC exposure impacts embryo-larval zebrafish development, immune function, and skeletal formation, but predominantly at concentrations greater than those currently reported for the aquatic environment. Despite this, however, it is suggested that studies with longer exposure times, and to mixtures of multiple GCs (many GCs act via the same mechanism of action) are warranted before we can confidently assert that these commonly detected contaminants do not pose a risk to fish in the wild.
Collapse
Affiliation(s)
- Charles M Hamilton
- Biosciences, Faculty of Health and Life Sciences, University of Exeter, Exeter, Devon EX4 4QD, UK
| | - Matthew J Winter
- Biosciences, Faculty of Health and Life Sciences, University of Exeter, Exeter, Devon EX4 4QD, UK
| | - Jonathan S Ball
- Biosciences, Faculty of Health and Life Sciences, University of Exeter, Exeter, Devon EX4 4QD, UK
| | - Maciej Trznadel
- Biosciences, Faculty of Health and Life Sciences, University of Exeter, Exeter, Devon EX4 4QD, UK
| | - Luigi Margiotta-Casaluci
- Institute of Pharmaceutical Science, Faculty of Life Sciences & Medicine, King's College London, London SE1 9NH, UK
| | | | - Charles R Tyler
- Biosciences, Faculty of Health and Life Sciences, University of Exeter, Exeter, Devon EX4 4QD, UK.
| |
Collapse
|
2
|
Savage MO. Linear growth in children and adolescents with congenital adrenal hyperplasia. Curr Opin Pediatr 2024; 36:463-466. [PMID: 38747200 DOI: 10.1097/mop.0000000000001361] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 07/04/2024]
Abstract
PURPOSE OF REVIEW Congenital adrenal hyperplasia (CAH) is a relatively common disorder and one of the most challenging conditions seen by pediatric endocrinologists. Poor linear growth in CAH has been recognized for many years. There are new insights to explain this abnormality and shed light on strategies to promote normal growth. RECENT FINDINGS Published data suggest that the dose of hydrocortisone during two critical periods of rapid growth, namely infancy and at puberty, has a fundamental effect on growth velocity, and by definition adult height. To prevent over-treatment, hydrocortisone dosage should remain within the range of 10-15 mg/m 2 body surface area per day. Precursor steroids such as 17-hydroxy progesterone (17OHP) should not be suppressed to undetectable levels. In fact, 17OHP should always be measurable, as complete suppression suggests over-treatment. SUMMARY CAH is a challenging disorder. High-quality compliance within the consultation setting, with the patient seeing the same specialist at every visit, will be rewarded by improved long-term growth potential. Quality auxological monitoring can avoid phases of growth suppression. New therapy with CRH receptor antagonists may lead to a more nuanced approach by allowing fine tuning of hydrocortisone replacement without the need to suppress ACTH secretion.
Collapse
Affiliation(s)
- Martin O Savage
- Centre for Endocrinology, William Harvey Research Institute, Barts and the London School of Medicine & Dentistry, Queen Mary, University of London, London, UK
| |
Collapse
|
3
|
Marrero - Berrios I, Salter SE, Hirday R, Rabolli CP, Tan A, Hung CT, Schloss RS, Yarmush ML. In vitro inflammatory multi-cellular model of osteoarthritis. OSTEOARTHRITIS AND CARTILAGE OPEN 2024; 6:100432. [PMID: 38288345 PMCID: PMC10823137 DOI: 10.1016/j.ocarto.2023.100432] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2023] [Accepted: 12/26/2023] [Indexed: 01/31/2024] Open
Abstract
Objective Osteoarthritis (OA) is a chronic joint disease, with limited treatment options, characterized by inflammation and matrix degradation, and resulting in severe pain or disability. Progressive inflammatory interaction among key cell types, including chondrocytes and macrophages, leads to a cascade of intra- and inter-cellular events which culminate in OA induction. In order to investigate these interactions, we developed a multi-cellular in vitro OA model, to characterize OA progression, and identify and evaluate potential OA therapeutics in response to mediators representing graded levels of inflammatory severity. Methods We compared macrophages, chondrocytes and their co-culture responses to "low" Interleukin-1 (IL-1) or "high" IL-1/tumor necrosis factor (IL-1/TNF) levels of inflammation. We also investigated response changes following the administration of dexamethasone (DEX) or mesenchymal stromal cell (MSC) treatment via a combination of gene expression and secretory changes, reflecting not only inflammation, but also chondrocyte function. Results Inflamed chondrocytes presented an osteoarthritic-like phenotype characterized by high gene expression of pro-inflammatory cytokines and chemokines, up-regulation of ECM degrading proteases, and down-regulation of chondrogenic genes. Our results indicate that while MSC treatment attenuates macrophage inflammation directly, it does not reduce chondrocyte inflammatory responses, unless macrophages are present as well. DEX however, can directly attenuate chondrocyte inflammation. Conclusions Our results highlight the importance of considering multi-cellular interactions when studying complex systems such as the articular joint. In addition, our approach, using a panel of both inflammatory and chondrocyte functional genes, provides a more comprehensive approach to investigate disease biomarkers, and responses to treatment.
Collapse
Affiliation(s)
| | - S. Elina Salter
- Department of Biomedical Engineering, Rutgers University, Piscataway, NJ, USA
| | - Rishabh Hirday
- Department of Biomedical Engineering, Rutgers University, Piscataway, NJ, USA
| | - Charles P. Rabolli
- Department of Biomedical Engineering, Rutgers University, Piscataway, NJ, USA
| | - Andrea Tan
- Department of Biomedical Engineering, Columbia University, New York, NY, USA
| | - Clark T. Hung
- Department of Biomedical Engineering, Columbia University, New York, NY, USA
| | - Rene S. Schloss
- Department of Biomedical Engineering, Rutgers University, Piscataway, NJ, USA
| | - Martin L. Yarmush
- Department of Biomedical Engineering, Rutgers University, Piscataway, NJ, USA
| |
Collapse
|
4
|
Hua J, Huang J, Li G, Lin S, Cui L. Glucocorticoid induced bone disorders in children: Research progress in treatment mechanisms. Front Endocrinol (Lausanne) 2023; 14:1119427. [PMID: 37082116 PMCID: PMC10111257 DOI: 10.3389/fendo.2023.1119427] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/09/2023] [Accepted: 03/20/2023] [Indexed: 04/22/2023] Open
Abstract
Long-term or supra-physiological dose of glucocorticoid (GC) application in clinic can lead to impaired bone growth and osteoporosis. The side effects of GC on the skeletal system are particularly serious in growing children, potentially causing growth retardation or even osteoporotic fractures. Children's bone growth is dependent on endochondral ossification of growth plate chondrocytes, and excessive GC can hinder the development of growth plate and longitudinal bone growth. Despite the availability of drugs for treating osteoporosis, they have failed to effectively prevent or treat longitudinal bone growth and development disorders caused by GCs. As of now, there is no specific drug to mitigate these severe side effects. Traditional Chinese Medicine shows potential as an alternative to the current treatments by eliminating the side effects of GC. In summary, this article comprehensively reviews the research frontiers concerning growth and development disorders resulting from supra-physiological levels of GC and discusses the future research and treatment directions for optimizing steroid therapy. This article may also provide theoretical and experimental insight into the research and development of novel drugs to prevent GC-related side effects.
Collapse
Affiliation(s)
- Junying Hua
- Guangdong Provincial Key Laboratory of Research and Development of Natural Drugs, School of Pharmacy, Guangdong Medical University, Zhanjiang, China
| | - Jianping Huang
- Department of Prosthodontics, Yonsei University College of Dentistry, Seoul, Republic of Korea
| | - Gang Li
- Musculoskeletal Research Laboratory, Department of Orthopaedics & Traumatology, Faculty of Medicine, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong, Hong Kong SAR, China
- Stem Cells and Regenerative Medicine Laboratory, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong, Hong Kong SAR, China
| | - Sien Lin
- Musculoskeletal Research Laboratory, Department of Orthopaedics & Traumatology, Faculty of Medicine, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong, Hong Kong SAR, China
- Stem Cells and Regenerative Medicine Laboratory, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong, Hong Kong SAR, China
- Orthopaedic Center, Affiliated Hospital of Guangdong Medical University, Guangdong Medical University, Zhanjiang, China
- *Correspondence: Liao Cui, ; Sien Lin,
| | - Liao Cui
- Guangdong Provincial Key Laboratory of Research and Development of Natural Drugs, School of Pharmacy, Guangdong Medical University, Zhanjiang, China
- *Correspondence: Liao Cui, ; Sien Lin,
| |
Collapse
|
5
|
Naik AA, Sivaramakrishnan V. Systems analysis of steroid induced osteonecrosis shows role for heme and vitamin D in pathogenesis. GENE REPORTS 2021. [DOI: 10.1016/j.genrep.2021.101383] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
6
|
Sanchez C, Hemmer K, Krömmelbein N, Seilheimer B, Dubuc JE, Antoine C, Henrotin Y. Reduction of Matrix Metallopeptidase 13 and Promotion of Chondrogenesis by Zeel T in Primary Human Osteoarthritic Chondrocytes. Front Pharmacol 2021; 12:635034. [PMID: 34045958 PMCID: PMC8144641 DOI: 10.3389/fphar.2021.635034] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2020] [Accepted: 02/26/2021] [Indexed: 12/21/2022] Open
Abstract
Objectives: Zeel T (Ze14) is a multicomponent medicinal product. Initial preclinical data suggested a preventive effect on cartilage degradation. Clinical observational studies demonstrated that Ze14 reduced symptoms of osteoarthritis (OA), including stiffness and pain. This study aimed to explore these effects further to better understand the mode of action of Ze14 on human OA chondrocytes in vitro. Methods: Primary chondrocytes were obtained from the knees of 19 OA patients and cultured either as monolayers or in alginate beads. The cultures were treated with 20% or 10% (v/v) Ze14 or placebo. For RNA-seq, reads were generated with Illumina NextSeq5000 sequencer and aligned to the human reference genome (UCSC hg19). Differential expression analysis between Ze14 and placebo was performed in R using the DESeq2 package. Protein quantification by ELISA was performed on selected genes from the culture medium and/or the cellular fractions of primary human OA chondrocyte cultures. Results: In monolayer cultures, Ze14 20% (v/v) significantly modified the expression of 13 genes in OA chondrocytes by at least 10% with an adjusted p-value < 0.05: EGR1, FOS, NR4A1, DUSP1, ZFP36, ZFP36L1, NFKBIZ, and CCN1 were upregulated and ATF7IP, TXNIP, DEPP1, CLEC3A, and MMP13 were downregulated after 24 h Ze14 treatment. Ze14 significantly increased (mean 2.3-fold after 24 h, p = 0.0444 and 72 h, p = 0.0239) the CCN1 protein production in human OA chondrocytes. After 72 h, Ze14 significantly increased type II collagen pro-peptide production by mean 27% (p = 0.0147). For both time points CCN1 production by OA chondrocytes was correlated with aggrecan (r = 0.66, p = 0.0004) and type II collagen pro-peptide (r = 0.64, p = 0.0008) production. In alginate beads cultures, pro-MMP-13 was decreased by Ze14 from day 7-14 (from -16 to -25%, p < 0.05) and from day 17-21 (-22%, p = 0.0331) in comparison to controls. Conclusion: Ze14 significantly modified the expression of DUSP1, DEPP1, ZFP36/ZFP36L1, and CLEC3A, which may reduce MMP13 expression and activation. Protein analysis confirmed that Ze14 significantly reduced the production of pro-MMP-13. As MMP-13 is involved in type II collagen degradation, Ze14 may limit cartilage degradation. Ze14 also promoted extracellular matrix formation arguably through CCN1 production, a growth factor well correlated with type II collagen and aggrecan production.
Collapse
Affiliation(s)
- Christelle Sanchez
- MusculoSKeletal Innovative Research Lab, University of Liège, Center for Interdisciplinary Research on Medicines, Liège, Belgium
| | | | | | | | - Jean-Emile Dubuc
- MusculoSKeletal Innovative Research Lab, University of Liège, Center for Interdisciplinary Research on Medicines, Liège, Belgium.,Division of Orthopedics and Musculoskeletal Trauma, Cliniques Universitaires de St Luc, Brussels, Belgium
| | | | - Yves Henrotin
- MusculoSKeletal Innovative Research Lab, University of Liège, Center for Interdisciplinary Research on Medicines, Liège, Belgium.,Artialis SA, Liège, Belgium.,Physical Therapy and Rehabilitation Department, Princess Paola Hospital, Vivalia, Marche-en-Famenne, Belgium
| |
Collapse
|
7
|
van Eegher S, Perez-Lozano ML, Toillon I, Valour D, Pigenet A, Citadelle D, Bourrier C, Courtade-Gaïani S, Grégoire L, Cléret D, Malbos S, Nourissat G, Sautet A, Lafage-Proust MH, Pastoureau P, Rolland-Valognes G, De Ceuninck F, Berenbaum F, Houard X. The differentiation of prehypertrophic into hypertrophic chondrocytes drives an OA-remodeling program and IL-34 expression. Osteoarthritis Cartilage 2021; 29:257-268. [PMID: 33301945 DOI: 10.1016/j.joca.2020.10.013] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/11/2019] [Revised: 09/16/2020] [Accepted: 10/08/2020] [Indexed: 02/02/2023]
Abstract
OBJECTIVES We hypothesize that chondrocytes from the deepest articular cartilage layer are pivotal in maintaining cartilage integrity and that the modification of their prehypertrophic phenotype to a hypertrophic phenotype will drive cartilage degradation in osteoarthritis. DESIGN Murine immature articular chondrocytes (iMACs) were successively cultured into three different culture media to induce a progressive hypertrophic differentiation. Chondrocyte were phenotypically characterized by whole-genome microarray analysis. The expression of IL-34 and its receptors PTPRZ1 and CSF1R in chondrocytes and in human osteoarthritis tissues was assessed by RT-qPCR, ELISA and immunohistochemistry. The expression of bone remodeling and angiogenesis factors and the cell response to IL-1β and IL-34 were investigated by RT-qPCR and ELISA. RESULTS Whole-genome microarray analysis showed that iMACs, prehypertrophic and hypertrophic chondrocytes each displayed a specific phenotype. IL-1β induced a stronger catabolic effect in prehypertrophic chondrocytes than in iMACs. Hypertrophic differentiation of prehypertrophic chondrocytes increased Bmp-2 (95%CI [0.78; 1.98]), Bmp-4 (95%CI [0.89; 1.59]), Cxcl12 (95%CI [2.19; 5.41]), CCL2 (95%CI [3.59; 11.86]), Mmp 3 (95%CI [10.29; 32.14]) and Vegf mRNA expression (95%CI [0.20; 1.74]). Microarray analysis identified IL-34, PTPRZ1 and CSFR1 as being strongly overexpressed in hypertrophic chondrocytes. IL-34 was released by human osteoarthritis cartilage; its receptors were expressed in human osteoarthritis tissues. IL-34 stimulated CCL2 and MMP13 in osteoblasts and hypertrophic chondrocytes but not in iMACs or prehypertrophic chondrocytes. CONCLUSION Our results identify prehypertrophic chondrocytes as being potentially pivotal in the control of cartilage and subchondral bone integrity. Their differentiation into hypertrophic chondrocytes initiates a remodeling program in which IL-34 may be involved.
Collapse
Affiliation(s)
- S van Eegher
- Sorbonne Université, INSERM, Centre de Recherche Saint-Antoine (CRSA), F-75012, Paris, France
| | - M-L Perez-Lozano
- Sorbonne Université, INSERM, Centre de Recherche Saint-Antoine (CRSA), F-75012, Paris, France
| | - I Toillon
- Sorbonne Université, INSERM, Centre de Recherche Saint-Antoine (CRSA), F-75012, Paris, France
| | - D Valour
- Servier Research Institute, F-78290, Croissy-sur-Seine, France
| | - A Pigenet
- Sorbonne Université, INSERM, Centre de Recherche Saint-Antoine (CRSA), F-75012, Paris, France
| | - D Citadelle
- Sorbonne Université, INSERM, Centre de Recherche Saint-Antoine (CRSA), F-75012, Paris, France
| | - C Bourrier
- Servier Research Institute, F-78290, Croissy-sur-Seine, France
| | | | - L Grégoire
- Soladis, 94 Rue Saint-Lazare, F-75009, Paris, France
| | - D Cléret
- Université de Lyon - Université Jean Monnet, INSERM U1059, Faculté de Médecine, F-42270, Saint-Priest en Jarez, France
| | - S Malbos
- Sorbonne Université, INSERM, Centre de Recherche Saint-Antoine (CRSA), F-75012, Paris, France
| | - G Nourissat
- Sorbonne Université, INSERM, Centre de Recherche Saint-Antoine (CRSA), F-75012, Paris, France; Clinique Maussins-Nollet, Ramsay Générale de Santé, F-75019, Paris, France
| | - A Sautet
- Department of Orthopaedic Surgery and Traumatology, APHP Saint-Antoine Hospital, F-75012, Paris, France
| | - M-H Lafage-Proust
- Université de Lyon - Université Jean Monnet, INSERM U1059, Faculté de Médecine, F-42270, Saint-Priest en Jarez, France
| | - P Pastoureau
- Servier Research Institute, F-78290, Croissy-sur-Seine, France
| | | | - F De Ceuninck
- Servier Research Institute, F-78290, Croissy-sur-Seine, France
| | - F Berenbaum
- Sorbonne Université, INSERM, Centre de Recherche Saint-Antoine (CRSA), F-75012, Paris, France; Sorbonne Université, INSERM CRSA, AP-HP Hopital Saint Antoine, Paris.
| | - X Houard
- Sorbonne Université, INSERM, Centre de Recherche Saint-Antoine (CRSA), F-75012, Paris, France
| |
Collapse
|
8
|
Hao Y, Lu C, Zhang B, Xu Z, Guo H, Zhang G. Identifying the Potential Differentially Expressed miRNAs and mRNAs in Osteonecrosis of the Femoral Head Based on Integrated Analysis. Clin Interv Aging 2021; 16:187-202. [PMID: 33542623 PMCID: PMC7851582 DOI: 10.2147/cia.s289479] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2020] [Accepted: 12/22/2020] [Indexed: 12/17/2022] Open
Abstract
Purpose Osteonecrosis of the femoral head is a common disease of the hip that leads to severe pain or joint disability. We aimed to identify potential differentially expressed miRNAs and mRNAs in osteonecrosis of the femoral head. Methods The data of miRNA and mRNA were firstly downloaded from the database. Secondly, the regulatory network of miRNAs-mRNAs was constructed, followed by function annotation of mRNAs. Thirdly, an in vitro experiment was applied to validate the expression of miRNAs and targeted mRNAs. Finally, GSE123568 dataset was used for electronic validation and diagnostic analysis of targeted mRNAs. Results Several regulatory interaction pairs between miRNA and mRNAs were identified, such as hsa-miR-378c-WNT3A/DACT1/CSF1, hsa-let-7a-5p-RCAN2/IL9R, hsa-miR-28-5p-RELA, hsa-miR-3200-5p-RELN, and hsa-miR-532-5p-CLDN18/CLDN10. Interestingly, CLDN10, CLDN18, CSF1, DACT1, IL9R, RCAN2, RELN, and WNT3A had the diagnostic value for osteonecrosis of the femoral head. Wnt signaling pathway (involved WNT3A), chemokine signaling pathway (involved RELA), focal adhesion and ECM-receptor interaction (involved RELN), cell adhesion molecules (CAMs) (involved CLDN18 and CLDN10), cytokine-cytokine receptor interaction, and hematopoietic cell lineage (involved CSF1 and IL9R) were identified. Conclusion The identified differentially expressed miRNAs and mRNAs may be involved in the pathology of osteonecrosis of the femoral head.
Collapse
Affiliation(s)
- Yangquan Hao
- Department of Osteonecrosis and Joint Reconstruction, Honghui Hospital Xian Jiao Tong University Health Science Center, Xian, Shaanxi 710068, People's Republic of China
| | - Chao Lu
- Department of Osteonecrosis and Joint Reconstruction, Honghui Hospital Xian Jiao Tong University Health Science Center, Xian, Shaanxi 710068, People's Republic of China
| | - Baogang Zhang
- Department of Osteonecrosis and Joint Reconstruction, Honghui Hospital Xian Jiao Tong University Health Science Center, Xian, Shaanxi 710068, People's Republic of China
| | - Zhaochen Xu
- Department of Osteonecrosis and Joint Reconstruction, Honghui Hospital Xian Jiao Tong University Health Science Center, Xian, Shaanxi 710068, People's Republic of China
| | - Hao Guo
- Department of Osteonecrosis and Joint Reconstruction, Honghui Hospital Xian Jiao Tong University Health Science Center, Xian, Shaanxi 710068, People's Republic of China
| | - Gaokui Zhang
- Department of Osteonecrosis and Joint Reconstruction, Honghui Hospital Xian Jiao Tong University Health Science Center, Xian, Shaanxi 710068, People's Republic of China
| |
Collapse
|
9
|
Rockel JS, Wu B, Nakamura S, Rossomacha E, Espin-Garcia O, Gandhi R, Kapoor M. TAT-Beclin-1 induces severe synovial hyperplasia and does not protect from injury-induced osteoarthritis in mice. Osteoarthritis Cartilage 2020; 28:1394-1400. [PMID: 32683043 DOI: 10.1016/j.joca.2020.07.001] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/29/2020] [Revised: 06/21/2020] [Accepted: 07/06/2020] [Indexed: 02/02/2023]
Abstract
OBJECT Autophagy maintains cartilage homeostasis and is compromised during osteoarthritis (OA), contributing to cartilage degeneration. We sought to determine if D-isomer TAT-Beclin-1, a potent inducer of autophagy, could attenuate post-traumatic OA in mice. METHODS 10-week-old mice underwent destabilization of the medial meniscus (DMM) surgery to induce post-traumatic OA, or sham surgery (control), and injected intra-articularly with D-isomer TAT-Beclin-1 (0.5-2 mg/kg) or PBS 1 week post-surgery for up to 9 weeks. Mice were sacrificed at 2 or 10 weeks post-surgery. Knee joint sections were evaluated by histopathology for cartilage degeneration and synovitis, and immunostaining for key markers of autophagy (LC3B), cell proliferation (nuclear Ki67), activated fibroblasts (αSMA), and cells of hematopoietic origin (CD45). RESULTS All D-isomer TAT-Beclin-1-treated DMM mice had no difference in the degree of cartilage degeneration compared to PBS-injected DMM mice. Surprisingly, all D-isomer TAT-Beclin-1-treated mice exhibited substantial synovial hyperplasia, with increased cellularity and ECM deposition (fibrosis-like phenotype), as compared to PBS-injected mice. Synovial effects of D-isomer TAT-Beclin-1 were dose- and injection frequency-dependent. An increased percentage of cells positive for LC3B and nuclear Ki67 were found in the synovial intima early after injection, which persisted after frequent injections. CONCLUSIONS D-isomer TAT-Beclin-1 did not attenuate cartilage degeneration, but rather induced synovial hyperplasia associated with increased expression of key markers of autophagy and cell proliferation and a fibrosis-like phenotype, independent of markers of fibroblast activation or persistent hematopoietic-origin cell infiltration. These data suggest that, if not tissue-targeted, caution should be taken using autophagy activators due to diverse cellular responses in the joint.
Collapse
Affiliation(s)
- J S Rockel
- Arthritis Program, Krembil Research Institute, University Health Network, Toronto, ON, Canada
| | - B Wu
- Arthritis Program, Krembil Research Institute, University Health Network, Toronto, ON, Canada
| | - S Nakamura
- Arthritis Program, Krembil Research Institute, University Health Network, Toronto, ON, Canada
| | - E Rossomacha
- Arthritis Program, Krembil Research Institute, University Health Network, Toronto, ON, Canada
| | - O Espin-Garcia
- Arthritis Program, Krembil Research Institute, University Health Network, Toronto, ON, Canada; Department of Biostatistics, Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - R Gandhi
- Arthritis Program, Krembil Research Institute, University Health Network, Toronto, ON, Canada; Department of Surgery and Department of Laboratory Medicine and Pathobiology, University of Toronto, ON, Canada
| | - M Kapoor
- Arthritis Program, Krembil Research Institute, University Health Network, Toronto, ON, Canada; Department of Surgery and Department of Laboratory Medicine and Pathobiology, University of Toronto, ON, Canada.
| |
Collapse
|
10
|
Minnetti M, Caiulo S, Ferrigno R, Baldini-Ferroli B, Bottaro G, Gianfrilli D, Sbardella E, De Martino MC, Savage MO. Abnormal linear growth in paediatric adrenal diseases: Pathogenesis, prevalence and management. Clin Endocrinol (Oxf) 2020; 92:98-108. [PMID: 31747461 DOI: 10.1111/cen.14131] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/24/2019] [Revised: 11/10/2019] [Accepted: 11/18/2019] [Indexed: 12/13/2022]
Abstract
Abnormal adrenal function can interfere with linear growth, potentially causing either acceleration or impairment of growth in paediatric patients. These abnormalities can be caused by direct effects of adrenal hormones, particularly glucocorticoids and sex steroids, or be mediated by indirect mechanisms such as the disturbance of the growth hormone-insulin-like growth factor-1 axis and aromatization of androgens to oestrogens. The early diagnosis and optimal treatment of adrenal disorders can prevent or minimize growth disturbance and facilitate improved height gain. Mechanisms of growth disturbance in the following abnormal states will be discussed; hypercortisolaemia, hyperandrogenaemia and obesity. Prevalence and features of growth disturbance will be discussed in ACTH-dependent and ACTH-independent Cushing's syndrome, adrenocortical tumours, premature adrenarche, congenital adrenal hyperplasia and adrenal insufficiency disorders. Recommendations for management have been included.
Collapse
Affiliation(s)
- Marianna Minnetti
- Department of Experimental Medicine, Sapienza University of Rome, Rome, Italy
| | - Silvana Caiulo
- Department of Pediatrics, IRCCS San Raffaele Hospital, Milan, Italy
| | - Rosario Ferrigno
- Dipartimento di Medicina Clinica e Chirurgia, Federico II University, Naples, Italy
| | - Barbara Baldini-Ferroli
- Dipartimento Pediatrico Universitario Ospedaliero, Bambino Gesu' Children's Hospital, Rome, Italy
| | - Giorgia Bottaro
- Dipartimento Pediatrico Universitario Ospedaliero, Bambino Gesu' Children's Hospital, Rome, Italy
| | - Daniele Gianfrilli
- Department of Experimental Medicine, Sapienza University of Rome, Rome, Italy
| | - Emilia Sbardella
- Department of Experimental Medicine, Sapienza University of Rome, Rome, Italy
| | | | - Martin O Savage
- Endocrinology Centre, William Harvey Research Institute, Barts and the London School of Medicine and Dentistry, London, UK
| |
Collapse
|
11
|
Abstract
While glucocorticoids have been used for over 50 years to treat rheumatoid and osteoarthritis pain, the prescription of glucocorticoids remains controversial because of potentially harmful side effects at the molecular, cellular and tissue levels. One member of the glucocorticoid family, dexamethasone (DEX) has recently been demonstrated to rescue cartilage matrix loss and chondrocyte viability in animal studies and cartilage explant models of tissue injury and post-traumatic osteoarthritis, suggesting the possibility of DEX as a disease-modifying drug if used appropriately. However, the literature on the effects of DEX on cartilage reveals conflicting results on the drug's safety, depending on the dose and duration of DEX exposure as well as the model system used. Overall, DEX has been shown to protect against arthritis-related changes in cartilage structure and function, including matrix loss, inflammation and cartilage viability. These beneficial effects are not always observed in model systems using initially healthy cartilage or isolated chondrocytes, where many studies have reported significant increases in chondrocyte apoptosis. It is crucially important to understand under what conditions DEX may be beneficial or harmful to cartilage and other joint tissues and to determine potential for safe use of this glucocorticoid in the clinic as a disease-modifying drug.
Collapse
Affiliation(s)
- R. Black
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - A. J. Grodzinsky
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA,Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA,Department of Electrical Engineering and Computer Science, Massachusetts Institute of Technology, Cambridge, MA, USA,Address for correspondence: Prof. Al Grodzinsky, MIT, Centre for Biomedical Engineering, 500 Technology Square, Cambridge, MA, 02139, USA.
| |
Collapse
|
12
|
Hernandez Cordero AI, Carbonetto P, Riboni Verri G, Gregory JS, Vandenbergh DJ, P Gyekis J, Blizard DA, Lionikas A. Replication and discovery of musculoskeletal QTLs in LG/J and SM/J advanced intercross lines. Physiol Rep 2019; 6. [PMID: 29479840 PMCID: PMC6430048 DOI: 10.14814/phy2.13561] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2017] [Revised: 12/02/2017] [Accepted: 12/05/2017] [Indexed: 12/14/2022] Open
Abstract
The genetics underlying variation in health‐related musculoskeletal phenotypes can be investigated in a mouse model. Quantitative trait loci (QTLs) affecting musculoskeletal traits in the LG/J and SM/J strain lineage remain to be refined and corroborated. The aim of this study was to map muscle and bone traits in males (n = 506) of the 50th filial generation of advanced intercross lines (LG/SM AIL) derived from the two strains. Genetic contribution to variation in all musculoskeletal traits was confirmed; the SNP heritability of muscle mass ranged between 0.46 and 0.56; and the SNP heritability of tibia length was 0.40. We used two analytical software, GEMMA and QTLRel, to map the underlying QTLs. GEMMA required substantially less computation and recovered all the QTLs identified by QTLRel. Seven significant QTLs were identified for muscle weight (Chr 1, 7, 11, 12, 13, 15, and 16), and two for tibia length, (Chr 1 and 13). Each QTL explained 4–5% of phenotypic variation. One muscle and both bone loci replicated previous findings; the remaining six were novel. Positional candidates for the replicated QTLs were prioritized based on in silico analyses and gene expression in muscle tissue. In summary, we replicated existing QTLs and identified novel QTLs affecting muscle weight, and replicated bone length QTLs in LG/SM AIL males. Heritability estimates substantially exceed the cumulative effect of the QTLs, hence a richer genetic architecture contributing to muscle and bone variability could be uncovered with a larger sample size.
Collapse
Affiliation(s)
- Ana I Hernandez Cordero
- School of Medicine, Medical Sciences and Nutrition, University of Aberdeen, Aberdeen, United Kingdom
| | - Peter Carbonetto
- Research Computing Center and Department of Human Genetics, University of Chicago, Chicago, Illinois
| | - Gioia Riboni Verri
- School of Medicine, Medical Sciences and Nutrition, University of Aberdeen, Aberdeen, United Kingdom
| | - Jennifer S Gregory
- School of Medicine, Medical Sciences and Nutrition, University of Aberdeen, Aberdeen, United Kingdom
| | - David J Vandenbergh
- Department of Biobehavioral Health, The Penn State Institute for the Neurosciences, Molecular and Cellular Integrative Biosciences Program, The Pennsylvania State University, University Park, Pennsylvania
| | - Joseph P Gyekis
- Department of Biobehavioral Health, The Pennsylvania State University, University Park, Pennsylvania
| | - David A Blizard
- Department of Biobehavioral Health, The Penn State Institute for the Neurosciences, Molecular and Cellular Integrative Biosciences Program, The Pennsylvania State University, University Park, Pennsylvania
| | - Arimantas Lionikas
- School of Medicine, Medical Sciences and Nutrition, University of Aberdeen, Aberdeen, United Kingdom
| |
Collapse
|
13
|
Savvidou O, Milonaki M, Goumenos S, Flevas D, Papagelopoulos P, Moutsatsou P. Glucocorticoid signaling and osteoarthritis. Mol Cell Endocrinol 2019; 480:153-166. [PMID: 30445185 DOI: 10.1016/j.mce.2018.11.001] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/11/2018] [Revised: 10/03/2018] [Accepted: 11/11/2018] [Indexed: 01/15/2023]
Abstract
Glucocorticoids are steroid hormones synthesized and released by the adrenal cortex. Their main function is to maintain cell homeostasis through a variety of signaling pathways, responding to changes in an organism's environment or developmental status. Mimicking the actions of natural glucocorticoids, synthetic glucocorticoids have been recruited to treat many diseases that implicate glucocorticoid receptor signaling such as osteoarthritis. In osteoarthritis, synthetic glucocorticoids aim to alleviate inflammation and pain. The variation of patients' response and the possibility of complications associated with their long-term use have led to a need for a better understanding of glucocorticoid receptor signaling in osteoarthritis. In this review, we performed a literature search in the molecular pathways that link the osteoarthritic joint to the glucocorticoid receptor signaling. We hope that this information will advance research in the field and propose new molecular targets for the development of more optimized therapies for osteoarthritis.
Collapse
Affiliation(s)
- Olga Savvidou
- First Department of Orthopaedics, National and Kapodistrian University of Athens, School of Medicine, "ATTIKON" University Hospital, Athens, Greece.
| | - Mandy Milonaki
- Department of Clinical Biochemistry, National and Kapodistrian University of Athens, School of Medicine, "ATTIKON" University Hospital, Athens, Greece.
| | - Stavros Goumenos
- First Department of Orthopaedics, National and Kapodistrian University of Athens, School of Medicine, "ATTIKON" University Hospital, Athens, Greece.
| | - Dimitrios Flevas
- First Department of Orthopaedics, National and Kapodistrian University of Athens, School of Medicine, "ATTIKON" University Hospital, Athens, Greece.
| | - Panayiotis Papagelopoulos
- First Department of Orthopaedics, National and Kapodistrian University of Athens, School of Medicine, "ATTIKON" University Hospital, Athens, Greece.
| | - Paraskevi Moutsatsou
- Department of Clinical Biochemistry, National and Kapodistrian University of Athens, School of Medicine, "ATTIKON" University Hospital, Athens, Greece.
| |
Collapse
|
14
|
Lesage R, Kerkhofs J, Geris L. Computational Modeling and Reverse Engineering to Reveal Dominant Regulatory Interactions Controlling Osteochondral Differentiation: Potential for Regenerative Medicine. Front Bioeng Biotechnol 2018; 6:165. [PMID: 30483498 PMCID: PMC6243751 DOI: 10.3389/fbioe.2018.00165] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2018] [Accepted: 10/22/2018] [Indexed: 01/11/2023] Open
Abstract
The specialization of cartilage cells, or chondrogenic differentiation, is an intricate and meticulously regulated process that plays a vital role in both bone formation and cartilage regeneration. Understanding the molecular regulation of this process might help to identify key regulatory factors that can serve as potential therapeutic targets, or that might improve the development of qualitative and robust skeletal tissue engineering approaches. However, each gene involved in this process is influenced by a myriad of feedback mechanisms that keep its expression in a desirable range, making the prediction of what will happen if one of these genes defaults or is targeted with drugs, challenging. Computer modeling provides a tool to simulate this intricate interplay from a network perspective. This paper aims to give an overview of the current methodologies employed to analyze cell differentiation in the context of skeletal tissue engineering in general and osteochondral differentiation in particular. In network modeling, a network can either be derived from mechanisms and pathways that have been reported in the literature (knowledge-based approach) or it can be inferred directly from the data (data-driven approach). Combinatory approaches allow further optimization of the network. Once a network is established, several modeling technologies are available to interpret dynamically the relationships that have been put forward in the network graph (implication of the activation or inhibition of certain pathways on the evolution of the system over time) and to simulate the possible outcomes of the established network such as a given cell state. This review provides for each of the aforementioned steps (building, optimizing, and modeling the network) a brief theoretical perspective, followed by a concise overview of published works, focusing solely on applications related to cell fate decisions, cartilage differentiation and growth plate biology. Particular attention is paid to an in-house developed example of gene regulatory network modeling of growth plate chondrocyte differentiation as all the aforementioned steps can be illustrated. In summary, this paper discusses and explores a series of tools that form a first step toward a rigorous and systems-level modeling of osteochondral differentiation in the context of regenerative medicine.
Collapse
Affiliation(s)
- Raphaelle Lesage
- Prometheus, Division of Skeletal Tissue Engineering Leuven, KU Leuven, Leuven, Belgium.,Biomechanics Section, KU Leuven, Leuven, Belgium
| | - Johan Kerkhofs
- Prometheus, Division of Skeletal Tissue Engineering Leuven, KU Leuven, Leuven, Belgium.,Biomechanics Section, KU Leuven, Leuven, Belgium
| | - Liesbet Geris
- Prometheus, Division of Skeletal Tissue Engineering Leuven, KU Leuven, Leuven, Belgium.,Biomechanics Section, KU Leuven, Leuven, Belgium.,Biomechanics Research Unit, GIGA in silico Medicine, University of Liège, Liège, Belgium
| |
Collapse
|
15
|
Han LP, Wang CP, Han SL. Overexpression of RBM10 induces osteosarcoma cell apoptosis and inhibits cell proliferation and migration. Med Sci (Paris) 2018; 34 Focus issue F1:81-86. [PMID: 30403180 DOI: 10.1051/medsci/201834f114] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Osteosarcoma is the most common malignant bone tumor with high incidence in adolescence and poor prognosis. RBM10, a member of RBPs, was reported to be a tumor suppressor in many kinds of cancers. However, the roles of RBM10 in osteosarcoma remain unknown. In this study, we found that overexpression of RBM10 decreased osteosarcoma cell proliferation and colony formation in soft agar, and inhibited osteosarcoma cell migration and invasion. Our results also revealed that RBM10 overexpression induced osteosarcoma cell apoptosis via the inhibition of Bcl-2, the activation of caspase-3, and the transcription and production of TNF-α. Our results indicated that RBM10 acts as a tumor suppressor in osteosarcoma. This could enable to define a new strategy for diagnosis and treatment of patients with osteosarcoma.
Collapse
Affiliation(s)
- Li-Ping Han
- Department of Orthopaedics, Zaozhuang Municipal Hospital, 41 Longtou Rd, Zaozhuang 277100, Shandong, P.R. China
| | - Cun-Ping Wang
- Department of Orthopaedics, Zaozhuang Municipal Hospital, 41 Longtou Rd, Zaozhuang 277100, Shandong, P.R. China
| | - Si-Lin Han
- Department of Orthopaedics, Zaozhuang Municipal Hospital, 41 Longtou Rd, Zaozhuang 277100, Shandong, P.R. China
| |
Collapse
|
16
|
Sun MMG, Beier F, Ratneswaran A. Nuclear receptors as potential drug targets in osteoarthritis. Curr Opin Pharmacol 2018; 40:81-86. [PMID: 29626714 DOI: 10.1016/j.coph.2018.03.008] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2018] [Accepted: 03/20/2018] [Indexed: 12/19/2022]
Abstract
Osteoarthritis is amongst the major causes of disability worldwide, but no medications that can slow or stop progression of this disorder have been identified. Recent evidence suggests roles for a variety of members of the nuclear receptor family of ligand-activated transcription factors in various forms of osteoarthritis. Since nuclear receptors are amongst the major classes of drug targets, these studies suggest that modulators of nuclear receptor activity might provide novel strategies to treat osteoarthritis. This review focuses on recent advances in our understanding of the role of nuclear receptors in osteoarthritis onset and progression, as well as their therapeutic implications. Future studies should continue to examine the possible roles of additional nuclear receptors in the pathophysiology of different types of osteoarthritis.
Collapse
Affiliation(s)
- Margaret Man-Ger Sun
- Department of Physiology and Pharmacology, Schulich School of Medicine & Dentistry, The University of Western Ontario, London, ON, Canada N6A 5C1; Bone & Joint Institute, The University of Western Ontario, London, ON, Canada N6A 5C1
| | - Frank Beier
- Department of Physiology and Pharmacology, Schulich School of Medicine & Dentistry, The University of Western Ontario, London, ON, Canada N6A 5C1; Bone & Joint Institute, The University of Western Ontario, London, ON, Canada N6A 5C1.
| | - Anusha Ratneswaran
- Department of Physiology and Pharmacology, Schulich School of Medicine & Dentistry, The University of Western Ontario, London, ON, Canada N6A 5C1; Bone & Joint Institute, The University of Western Ontario, London, ON, Canada N6A 5C1
| |
Collapse
|
17
|
Loiselle JJ, Sutherland LC. RBM10: Harmful or helpful-many factors to consider. J Cell Biochem 2018; 119:3809-3818. [PMID: 29274279 PMCID: PMC5901003 DOI: 10.1002/jcb.26644] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2017] [Accepted: 12/20/2017] [Indexed: 11/25/2022]
Abstract
RBM10 is an RNA binding motif (RBM) protein expressed in most, if not all, human and animal cells. Interest in RBM10 is rapidly increasing and its clinical importance is highlighted by its identification as the causative agent of TARP syndrome, a developmental condition that significantly impacts affected children. RBM10's cellular functions are beginning to be explored, with initial studies demonstrating a tumor suppressor role. Very recently, however, contradictory results have emerged, suggesting a tumor promoter role for RBM10. In this review, we describe the current state of knowledge on RBM10, and address this dichotomy in RBM10 function. Furthermore, we discuss what may be regulating RBM10 function, particularly the importance of RBM10 alternative splicing, and the relationship between RBM10 and its paralogue, RBM5. As RBM10‐related work is gaining momentum, it is critical that the various aspects of RBM10 molecular biology revealed by recent studies be considered moving forward. It is only if these recent advances in RBM10 structure and function are considered that a clearer insight into RBM10 function, and the disease states with which RBM10 mutation is associated, will be gained.
Collapse
Affiliation(s)
- Julie J Loiselle
- Health Sciences North Research Institute (HSNRI), Sudbury, Ontario, Canada
| | | |
Collapse
|
18
|
Zou C, Li W, Pan Y, Khan ZA, Li J, Wu X, Wang Y, Deng L, Liang G, Zhao Y. 11β-HSD1 inhibition ameliorates diabetes-induced cardiomyocyte hypertrophy and cardiac fibrosis through modulation of EGFR activity. Oncotarget 2017; 8:96263-96275. [PMID: 29221204 PMCID: PMC5707098 DOI: 10.18632/oncotarget.22015] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2017] [Accepted: 09/20/2017] [Indexed: 12/20/2022] Open
Abstract
11β-HSD1 has been recognized as a potential therapeutic target for type 2 diabetes. Recent studies have shown that hyperglycemia leads to activation of 11β-HSD1, increasing the intracellular glucocorticoid levels. Excess glucocorticoids may lead to the clinical manifestations of cardiac injury. Therefore, the aim of this study is to investigate whether 11β-HSD1 activation contributes to the development of diabetic cardiomyopathy. To investigate the role of 11β-HSD1, we administered a selective 11β-HSD1 inhibitor in type 1 and type 2 murine models of diabetes and in cultured cardiomyocytes. Our results show that diabetes increases cortisone levels in heart tissues. 11β-HSD1 inhibitor decreased cortisone levels and ameliorated all structural and functional features of diabetic cardiomyopathy including fibrosis and hypertrophy. We also show that high levels of glucose caused cardiomyocyte hypertrophy and increased matrix protein deposition in culture. Importantly, inhibition of 11β-HSD1 attenuated these changes. Moreover, we show that 11β-HSD1 activation mediates these changes through modulating EGFR phosphorylation and activity. Our findings demonstrate that 11β-HSD1 contributes to the development of diabetic cardiomyopathy through activation of glucocorticoid and EGFR signaling pathway. These results suggest that inhibition of 11β-HSD1 might be a therapeutic strategy for diabetic cardiomyopathy, which is independent of its effects on glucose homeostasis.
Collapse
Affiliation(s)
- Chunpeng Zou
- Chemical Biology Research Center, School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, Zhejiang, China.,Department of Ultrasonography, The Second Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Weixin Li
- Chemical Biology Research Center, School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Yong Pan
- Chemical Biology Research Center, School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, Zhejiang, China.,Affiliated Yueqing Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Zia A Khan
- Department of Pathology and Laboratory Medicine, Western University, London, Ontario, Canada
| | - Jieli Li
- Chemical Biology Research Center, School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Xixi Wu
- Chemical Biology Research Center, School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Yi Wang
- Chemical Biology Research Center, School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Liancheng Deng
- Chemical Biology Research Center, School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Guang Liang
- Chemical Biology Research Center, School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, Zhejiang, China.,Affiliated Yueqing Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Yunjie Zhao
- Chemical Biology Research Center, School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, Zhejiang, China.,Affiliated Yueqing Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China
| |
Collapse
|
19
|
Loiselle JJ, Roy JG, Sutherland LC. RBM10 promotes transformation-associated processes in small cell lung cancer and is directly regulated by RBM5. PLoS One 2017; 12:e0180258. [PMID: 28662214 PMCID: PMC5491171 DOI: 10.1371/journal.pone.0180258] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2017] [Accepted: 06/13/2017] [Indexed: 11/19/2022] Open
Abstract
Lung cancers are the leading cause of cancer-related deaths worldwide, with small cell lung cancer (SCLC) being the most aggressive type. At the time of diagnosis, SCLC has usually already metastasized, and an astonishing 95% of patients eventually succumb to the disease. This highlights the need for more effective SCLC screening and treatment options. Interestingly, the earliest and most frequent genetic alteration associated with lung cancers involves a lesion in the region to which the RNA binding protein RBM5 maps. We have recently shown that a decrease in RBM5 expression may be a key step in SCLC development, as RBM5 regulated many transformation-associated processes in SCLC cells. RBM5 is structurally and functionally similar to another RNA binding protein, RBM10. Both proteins have tumor-suppressor properties in a variety of cancer cell lines, and it has been suggested that RBM5 expression can influence RBM10. Due to their similarities, and the recent evidence that RBM10 is mutated in up to 21% of lung cancers, we hypothesized that RBM10 would share RBM5's tumor-suppressor properties in SCLC. Using transcriptome analysis and functional assays, we show, however, that RBM10's function was opposite to what we hypothesized; in the endogenously RBM5-null GLC20 SCLC cell line, RBM10 actually promoted cell proliferation and other transformation-associated processes. Using RNA immunoprecipitation followed by next generation sequencing (RIP-Seq) and Western blotting, we demonstrate that RBM5 post-transcriptionally regulated RBM10 expression via direct interaction with specific RBM10 splice variants. We propose a working model describing the impact of this interaction on cellular processes. Our results provide evidence that RBM10 expression, in RBM5-null tumors, may contribute to tumor growth and metastasis. Measurement of both RBM10 and RBM5 expression in clinical samples may therefore hold prognostic and/or potentially predictive value.
Collapse
Affiliation(s)
- Julie J. Loiselle
- Biomolecular Sciences Program, Laurentian University, Sudbury, Ontario, Canada
| | - Justin G. Roy
- Department of Chemistry and Biochemistry, Laurentian University, Sudbury, Ontario, Canada
| | - Leslie C. Sutherland
- Biomolecular Sciences Program, Laurentian University, Sudbury, Ontario, Canada
- Department of Chemistry and Biochemistry, Laurentian University, Sudbury, Ontario, Canada
- Health Sciences North Research Institute (HSNRI), Sudbury, Ontario, Canada
| |
Collapse
|
20
|
Ratneswaran A, Sun MMG, Dupuis H, Sawyez C, Borradaile N, Beier F. Nuclear receptors regulate lipid metabolism and oxidative stress markers in chondrocytes. J Mol Med (Berl) 2017; 95:431-444. [PMID: 28070626 PMCID: PMC5357281 DOI: 10.1007/s00109-016-1501-5] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2016] [Revised: 11/14/2016] [Accepted: 12/20/2016] [Indexed: 01/21/2023]
Abstract
Abstract Joint homeostasis failure can result in osteoarthritis (OA). Currently, there are no treatments to alter disease progression in OA, but targeting early changes in cellular behavior has great potential. Recent data show that nuclear receptors contribute to the pathogenesis of OA and could be viable therapeutic targets, but their molecular mechanisms in cartilage are incompletely understood. This study examines global changes in gene expression after treatment with agonists for four nuclear receptor implicated in OA (LXR, PPARδ, PPARγ, and RXR). Murine articular chondrocytes were treated with agonists for LXR, PPARδ, PPARγ, or RXR and underwent microarray, qPCR, and cellular lipid analyses to evaluate changes in gene expression and lipid profile. Immunohistochemistry was conducted to compare two differentially expressed targets (Txnip, Gsta4) in control and cartilage-specific PPARδ knockout mice subjected to surgical destabilization of the medial meniscus (DMM). Nuclear receptor agonists induced different gene expression profiles with many responses affecting lipid metabolism. LXR activation downregulated gene expression of proteases involved in OA, whereas RXR agonism decreased expression of ECM components and increased expression of Mmp13. Functional assays indicate increases in cell triglyceride accumulation after PPARγ, LXR, and RXR agonism but a decrease after PPARδ agonism. PPARδ and RXR downregulate the antioxidant Gsta4, and PPARδ upregulates Txnip. Wild-type, but not PPARδ-deficient mice, display increased staining for Txnip after DMM. Collectively, these data demonstrate that nuclear receptor activation in chondrocytes primarily affects lipid metabolism. In the case of PPARδ, this change might lead to increased oxidative stress, possibly contributing to OA-associated changes. Key message Nuclear receptors regulate metabolic genes in chondrocytes. Nuclear receptors affect triglyceride levels. PPARδ mediates regulation of oxidative stress markers. Nuclear receptors are promising therapeutic targets for osteoarthritis.
Electronic supplementary material The online version of this article (doi:10.1007/s00109-016-1501-5) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Anusha Ratneswaran
- Department of Physiology and Pharmacology, Schulich School of Medicine & Dentistry, University of Western Ontario, London, ON, N6A 5C1, Canada.,Western Bone & Joint Institute, University of Western Ontario, London, ON, Canada
| | - Margaret Man-Ger Sun
- Department of Physiology and Pharmacology, Schulich School of Medicine & Dentistry, University of Western Ontario, London, ON, N6A 5C1, Canada.,Western Bone & Joint Institute, University of Western Ontario, London, ON, Canada
| | - Holly Dupuis
- Department of Physiology and Pharmacology, Schulich School of Medicine & Dentistry, University of Western Ontario, London, ON, N6A 5C1, Canada.,Western Bone & Joint Institute, University of Western Ontario, London, ON, Canada
| | - Cynthia Sawyez
- Department of Physiology and Pharmacology, Schulich School of Medicine & Dentistry, University of Western Ontario, London, ON, N6A 5C1, Canada
| | - Nica Borradaile
- Department of Physiology and Pharmacology, Schulich School of Medicine & Dentistry, University of Western Ontario, London, ON, N6A 5C1, Canada
| | - Frank Beier
- Department of Physiology and Pharmacology, Schulich School of Medicine & Dentistry, University of Western Ontario, London, ON, N6A 5C1, Canada. .,Western Bone & Joint Institute, University of Western Ontario, London, ON, Canada.
| |
Collapse
|
21
|
Muqbil I, Aboukameel A, Elloul S, Carlson R, Senapedis W, Baloglu E, Kauffman M, Shacham S, Bhutani D, Zonder J, Azmi AS, Mohammad RM. Anti-tumor activity of selective inhibitor of nuclear export (SINE) compounds, is enhanced in non-Hodgkin lymphoma through combination with mTOR inhibitor and dexamethasone. Cancer Lett 2016; 383:309-317. [PMID: 27693556 PMCID: PMC5584550 DOI: 10.1016/j.canlet.2016.09.016] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2016] [Revised: 09/05/2016] [Accepted: 09/06/2016] [Indexed: 11/21/2022]
Abstract
In previous studies we demonstrated that targeting the nuclear exporter protein exportin-1 (CRM1/XPO1) by a selective inhibitor of nuclear export (SINE) compound is a viable therapeutic strategy against Non-Hodgkin Lymphoma (NHL). Our studies along with pre-clinical work from others led to the evaluation of the lead SINE compound, selinexor, in a phase 1 trial in patients with CLL or NHL (NCT02303392). Continuing our previous work, we studied combinations of selinexor-dexamethasone (DEX) and selinexor-everolimus (EVER) in NHL. Combination of selinexor with DEX or EVER resulted in enhanced cytotoxicity in WSU-DLCL2 and WSU-FSCCL cells which was consistent with enhanced apoptosis. Molecular analysis showed enhancement in the activation of apoptotic signaling and down-regulation of XPO1. This enhancement is consistent with the mechanism of action of these drugs in that both selinexor and DEX antagonize NF-κB (p65) and mTOR (EVER target) is an XPO1 cargo protein. SINE compounds, KPT-251 and KPT-276, showed activities similar to CHOP (cyclophosphamide-hydroxydaunorubicin-oncovin-prednisone) regimen in subcutaneous and disseminated NHL xenograft models in vivo. In both animal models the anti-lymphoma activity of selinexor is enhanced through combination with DEX or EVER. The in vivo activity of selinexor and related SINE compounds relative to 'standard of care' treatment is consistent with the objective responses observed in Phase I NHL patients treated with selinexor. Our pre-clinical data provide a rational basis for testing these combinations in Phase II NHL trials.
Collapse
MESH Headings
- Acrylamides/pharmacology
- Active Transport, Cell Nucleus/drug effects
- Animals
- Antineoplastic Combined Chemotherapy Protocols/pharmacology
- Apoptosis/drug effects
- Cell Line, Tumor
- Cell Survival/drug effects
- Cyclophosphamide/pharmacology
- Dexamethasone/pharmacology
- Dose-Response Relationship, Drug
- Doxorubicin/pharmacology
- Drug Synergism
- Everolimus/pharmacology
- Humans
- Hydrazines/pharmacology
- Karyopherins/antagonists & inhibitors
- Karyopherins/metabolism
- Lymphoma, Non-Hodgkin/drug therapy
- Lymphoma, Non-Hodgkin/enzymology
- Lymphoma, Non-Hodgkin/pathology
- Mice, Inbred ICR
- Mice, SCID
- Oxadiazoles/pharmacology
- Prednisone/pharmacology
- Protein Kinase Inhibitors/pharmacology
- Receptors, Cytoplasmic and Nuclear/antagonists & inhibitors
- Receptors, Cytoplasmic and Nuclear/metabolism
- Signal Transduction/drug effects
- TOR Serine-Threonine Kinases/antagonists & inhibitors
- TOR Serine-Threonine Kinases/metabolism
- Thiazoles/pharmacology
- Time Factors
- Transcription Factor RelA/antagonists & inhibitors
- Transcription Factor RelA/metabolism
- Triazoles/pharmacology
- Tumor Burden/drug effects
- Vincristine/pharmacology
- Xenograft Model Antitumor Assays
- Exportin 1 Protein
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | - Asfar S Azmi
- Department of Oncology, Wayne State University, USA
| | | |
Collapse
|
22
|
Hartmann K, Koenen M, Schauer S, Wittig-Blaich S, Ahmad M, Baschant U, Tuckermann JP. Molecular Actions of Glucocorticoids in Cartilage and Bone During Health, Disease, and Steroid Therapy. Physiol Rev 2016; 96:409-47. [PMID: 26842265 DOI: 10.1152/physrev.00011.2015] [Citation(s) in RCA: 141] [Impact Index Per Article: 17.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Cartilage and bone are severely affected by glucocorticoids (GCs), steroid hormones that are frequently used to treat inflammatory diseases. Major complications associated with long-term steroid therapy include impairment of cartilaginous bone growth and GC-induced osteoporosis. Particularly in arthritis, GC application can increase joint and bone damage. Contrarily, endogenous GC release supports cartilage and bone integrity. In the last decade, substantial progress in the understanding of the molecular mechanisms of GC action has been gained through genome-wide binding studies of the GC receptor. These genomic approaches have revolutionized our understanding of gene regulation by ligand-induced transcription factors in general. Furthermore, specific inactivation of GC signaling and the GC receptor in bone and cartilage cells of rodent models has enabled the cell-specific effects of GCs in normal tissue homeostasis, inflammatory bone diseases, and GC-induced osteoporosis to be dissected. In this review, we summarize the current view of GC action in cartilage and bone. We further discuss future research directions in the context of new concepts for optimized steroid therapies with less detrimental effects on bone.
Collapse
Affiliation(s)
- Kerstin Hartmann
- Institute for Comparative Molecular Endocrinology, University of Ulm, Ulm, Germany; and Division of Endocrinology, Diabetes, and Bone Diseases, Department of Medicine III, Technische Universität Dresden, Dresden, Germany
| | - Mascha Koenen
- Institute for Comparative Molecular Endocrinology, University of Ulm, Ulm, Germany; and Division of Endocrinology, Diabetes, and Bone Diseases, Department of Medicine III, Technische Universität Dresden, Dresden, Germany
| | - Sebastian Schauer
- Institute for Comparative Molecular Endocrinology, University of Ulm, Ulm, Germany; and Division of Endocrinology, Diabetes, and Bone Diseases, Department of Medicine III, Technische Universität Dresden, Dresden, Germany
| | - Stephanie Wittig-Blaich
- Institute for Comparative Molecular Endocrinology, University of Ulm, Ulm, Germany; and Division of Endocrinology, Diabetes, and Bone Diseases, Department of Medicine III, Technische Universität Dresden, Dresden, Germany
| | - Mubashir Ahmad
- Institute for Comparative Molecular Endocrinology, University of Ulm, Ulm, Germany; and Division of Endocrinology, Diabetes, and Bone Diseases, Department of Medicine III, Technische Universität Dresden, Dresden, Germany
| | - Ulrike Baschant
- Institute for Comparative Molecular Endocrinology, University of Ulm, Ulm, Germany; and Division of Endocrinology, Diabetes, and Bone Diseases, Department of Medicine III, Technische Universität Dresden, Dresden, Germany
| | - Jan P Tuckermann
- Institute for Comparative Molecular Endocrinology, University of Ulm, Ulm, Germany; and Division of Endocrinology, Diabetes, and Bone Diseases, Department of Medicine III, Technische Universität Dresden, Dresden, Germany
| |
Collapse
|
23
|
Huang BJ, Huey DJ, Hu JC, Athanasiou KA. Engineering biomechanically functional neocartilage derived from expanded articular chondrocytes through the manipulation of cell-seeding density and dexamethasone concentration. J Tissue Eng Regen Med 2016; 11:2323-2332. [PMID: 27138113 DOI: 10.1002/term.2132] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2015] [Revised: 11/17/2015] [Accepted: 12/10/2015] [Indexed: 11/07/2022]
Abstract
Recent work has established methods to engineer self-assembled, scaffold-free neocartilage from an expanded articular chondrocyte (AC) cell source. In continuing such work, the objective of the present study was to investigate the effects of cell-seeding density and dexamethasone concentration on these neocartilage constructs. Neocartilage discs (5 mm diameter) were formed by self-assembling passaged leporine articular chondrocytes into non-adherent agarose moulds. The cell-seeding densities (2, 3, 4, 5 and 6 million cells/construct) and dexamethasone concentrations (10 and 100 nm) in the culture medium were varied in a full-factorial study. After 4 weeks, the neocartilage constructs were assessed for morphological, biochemical and biomechanical properties. The cell-seeding density profoundly affected neocartilage properties. The two dexamethasone concentrations explored did not induce overall significant differences. Constructs formed using lower cell-seeding densities possessed much higher biochemical and biomechanical properties than constructs seeded with higher cell densities. Notably, the 2 million cells/construct group formed hyaline-like neocartilage with a collagen wet weight (WW) content of ~7% and a Young's modulus of ~4 MPa, representing the high end of values achieved in self-assembled neocartilage. Excitingly, the mechanical properties of these constructs were on a par with that of native cartilage tissues tested under similar conditions. Through optimization of cell-seeding density, this study shows for the first time the use of expanded ACs to form homogeneous self-assembled neocartilage with exceptionally high tensile strength. With such functional properties, these engineered neocartilage constructs provide a promising alternative for treating articular lesions. Copyright © 2016 John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- Brian J Huang
- Department of Biomedical Engineering, University of California, Davis, CA, USA
| | - Daniel J Huey
- Department of Biomedical Engineering, University of California, Davis, CA, USA
| | - Jerry C Hu
- Department of Biomedical Engineering, University of California, Davis, CA, USA
| | - Kyriacos A Athanasiou
- Department of Biomedical Engineering, University of California, Davis, CA, USA.,Department of Orthopedic Surgery, University of California, Davis, CA, USA
| |
Collapse
|
24
|
Wong SC, Dobie R, Altowati MA, Werther GA, Farquharson C, Ahmed SF. Growth and the Growth Hormone-Insulin Like Growth Factor 1 Axis in Children With Chronic Inflammation: Current Evidence, Gaps in Knowledge, and Future Directions. Endocr Rev 2016; 37:62-110. [PMID: 26720129 DOI: 10.1210/er.2015-1026] [Citation(s) in RCA: 92] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Growth failure is frequently encountered in children with chronic inflammatory conditions like juvenile idiopathic arthritis, inflammatory bowel disease, and cystic fibrosis. Delayed puberty and attenuated pubertal growth spurt are often seen during adolescence. The underlying inflammatory state mediated by proinflammatory cytokines, prolonged use of glucocorticoid, and suboptimal nutrition contribute to growth failure and pubertal abnormalities. These factors can impair growth by their effects on the GH-IGF axis and also directly at the level of the growth plate via alterations in chondrogenesis and local growth factor signaling. Recent studies on the impact of cytokines and glucocorticoid on the growth plate further advanced our understanding of growth failure in chronic disease and provided a biological rationale of growth promotion. Targeting cytokines using biological therapy may lead to improvement of growth in some of these children, but approximately one-third continue to grow slowly. There is increasing evidence that the use of relatively high-dose recombinant human GH may lead to partial catch-up growth in chronic inflammatory conditions, although long-term follow-up data are currently limited. In this review, we comprehensively review the growth abnormalities in children with juvenile idiopathic arthritis, inflammatory bowel disease, and cystic fibrosis, systemic abnormalities of the GH-IGF axis, and growth plate perturbations. We also systematically reviewed all the current published studies of recombinant human GH in these conditions and discussed the role of recombinant human IGF-1.
Collapse
Affiliation(s)
- S C Wong
- Developmental Endocrinology Research Group (S.C.W., M.A.A., S.F.A.), University of Glasgow, Royal Hospital for Children, Glasgow G51 4TF, United Kingdom; Division of Developmental Biology (R.D., C.F.), Roslin Institute, University of Edinburgh, Midlothian EH25 9RG, United Kingdom; and Hormone Research (G.A.W.), Murdoch Children's Research Institute, Melbourne, VIC 3052, Australia
| | - R Dobie
- Developmental Endocrinology Research Group (S.C.W., M.A.A., S.F.A.), University of Glasgow, Royal Hospital for Children, Glasgow G51 4TF, United Kingdom; Division of Developmental Biology (R.D., C.F.), Roslin Institute, University of Edinburgh, Midlothian EH25 9RG, United Kingdom; and Hormone Research (G.A.W.), Murdoch Children's Research Institute, Melbourne, VIC 3052, Australia
| | - M A Altowati
- Developmental Endocrinology Research Group (S.C.W., M.A.A., S.F.A.), University of Glasgow, Royal Hospital for Children, Glasgow G51 4TF, United Kingdom; Division of Developmental Biology (R.D., C.F.), Roslin Institute, University of Edinburgh, Midlothian EH25 9RG, United Kingdom; and Hormone Research (G.A.W.), Murdoch Children's Research Institute, Melbourne, VIC 3052, Australia
| | - G A Werther
- Developmental Endocrinology Research Group (S.C.W., M.A.A., S.F.A.), University of Glasgow, Royal Hospital for Children, Glasgow G51 4TF, United Kingdom; Division of Developmental Biology (R.D., C.F.), Roslin Institute, University of Edinburgh, Midlothian EH25 9RG, United Kingdom; and Hormone Research (G.A.W.), Murdoch Children's Research Institute, Melbourne, VIC 3052, Australia
| | - C Farquharson
- Developmental Endocrinology Research Group (S.C.W., M.A.A., S.F.A.), University of Glasgow, Royal Hospital for Children, Glasgow G51 4TF, United Kingdom; Division of Developmental Biology (R.D., C.F.), Roslin Institute, University of Edinburgh, Midlothian EH25 9RG, United Kingdom; and Hormone Research (G.A.W.), Murdoch Children's Research Institute, Melbourne, VIC 3052, Australia
| | - S F Ahmed
- Developmental Endocrinology Research Group (S.C.W., M.A.A., S.F.A.), University of Glasgow, Royal Hospital for Children, Glasgow G51 4TF, United Kingdom; Division of Developmental Biology (R.D., C.F.), Roslin Institute, University of Edinburgh, Midlothian EH25 9RG, United Kingdom; and Hormone Research (G.A.W.), Murdoch Children's Research Institute, Melbourne, VIC 3052, Australia
| |
Collapse
|
25
|
Loiselle JJ, Tessier SJ, Sutherland LC. Post-transcriptional regulation of Rbm5 expression in undifferentiated H9c2 myoblasts. In Vitro Cell Dev Biol Anim 2015; 52:327-336. [PMID: 26659391 PMCID: PMC4833810 DOI: 10.1007/s11626-015-9976-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2015] [Accepted: 11/04/2015] [Indexed: 12/20/2022]
Abstract
We previously examined the expression of Rbm5 during myoblast differentiation and found significantly more protein in the early stages of skeletal myoblast differentiation than during the later stages. We decided to determine if this elevated level was necessary for differentiation. Our hypothesis was that if high levels of Rbm5 protein expression were necessary for the initiation of skeletal myoblast differentiation, then inhibition of expression would prevent differentiation. Our long-term objective is to inhibit Rbm5 expression and examine the effect on H9c2 differentiation. Towards this end, stable knockdown clones and transient knockdown populations were generated. Expression analyses in H9c2 myoblasts demonstrated significant Rbm5 messenger RNA (mRNA) inhibition but, surprisingly, no effect on RBM5 protein levels. Expression of the Rbm5 paralogue Rbm10 was examined in order to (a) ensure no off-target knockdown effect, and (b) investigate any possible compensatory effects. RBM10 protein levels were found to be elevated, in both the clonal and transiently transfected populations. These results suggest that myoblast RBM5 expression is regulated by a process that includes RNA sequestration and/or controlled translation, and that (a) RBM5 function is compensated for by RBM10, and/or (b) RBM5 regulates RBM10 expression. We have developed a model to describe our findings, and suggest further experiments for testing its validity. Since upregulation of Rbm10 might compensate for downregulated Rbm5, and consequently might mask any potential knockdown effect, it could lead to incorrect conclusions regarding the importance of Rbm5 for differentiation. It is therefore imperative to determine how both RBM5 and RBM10 protein expression is regulated.
Collapse
Affiliation(s)
- Julie J Loiselle
- Biomolecular Sciences Program, Laurentian University, 935 Ramsey Lake Road, Sudbury, ON, P3E 2C6, Canada. .,AMRIC, Health Sciences North, 41 Ramsey Lake Road, Sudbury, ON, P3E 5J1, Canada.
| | - Sarah J Tessier
- Department of Biology, Laurentian University, 935 Ramsey Lake Road, Sudbury, ON, P3E 2C6, Canada
| | - Leslie C Sutherland
- Biomolecular Sciences Program, Laurentian University, 935 Ramsey Lake Road, Sudbury, ON, P3E 2C6, Canada.,AMRIC, Health Sciences North, 41 Ramsey Lake Road, Sudbury, ON, P3E 5J1, Canada.,Department of Chemistry and Biochemistry, Laurentian University, 935 Ramsey Lake Road, Sudbury, ON, P3E 2C6, Canada.,Division of Medical Sciences, Northern Ontario School of Medicine, Laurentian University, 935 Ramsey Lake Road, Sudbury, ON, P3E 2C6, Canada.,Department of Medicine, Division of Medical Oncology, University of Ottawa, Ottawa, ON, Canada
| |
Collapse
|
26
|
McCusker CD, Athippozhy A, Diaz-Castillo C, Fowlkes C, Gardiner DM, Voss SR. Positional plasticity in regenerating Amybstoma mexicanum limbs is associated with cell proliferation and pathways of cellular differentiation. BMC DEVELOPMENTAL BIOLOGY 2015; 15:45. [PMID: 26597593 PMCID: PMC4657325 DOI: 10.1186/s12861-015-0095-4] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/21/2015] [Accepted: 11/16/2015] [Indexed: 01/07/2023]
Abstract
Background The endogenous ability to dedifferentiate, re-pattern, and re-differentiate adult cells to repair or replace damaged or missing structures is exclusive to only a few tetrapod species. The Mexican axolotl is one example of these species, having the capacity to regenerate multiple adult structures including their limbs by generating a group of progenitor cells, known as the blastema, which acquire pattern and differentiate into the missing tissues. The formation of a limb regenerate is dependent on cells in the connective tissues that retain memory of their original position in the limb, and use this information to generate the pattern of the missing structure. Observations from recent and historic studies suggest that blastema cells vary in their potential to pattern distal structures during the regeneration process; some cells are plastic and can be reprogrammed to obtain new positional information while others are stable. Our previous studies showed that positional information has temporal and spatial components of variation; early bud (EB) and apical late bud (LB) blastema cells are plastic while basal-LB cells are stable. To identify the potential cellular and molecular basis of this variation, we compared these three cell populations using histological and transcriptional approaches. Results Histologically, the basal-LB sample showed greater tissue organization than the EB and apical-LB samples. We also observed that cell proliferation was more abundant in EB and apical-LB tissue when compared to basal-LB and mature stump tissue. Lastly, we found that genes associated with cellular differentiation were expressed more highly in the basal-LB samples. Conclusions Our results characterize histological and transcriptional differences between EB and apical-LB tissue compared to basal-LB tissue. Combined with our results from a previous study, we hypothesize that the stability of positional information is associated with tissue organization, cell proliferation, and pathways of cellular differentiation. Electronic supplementary material The online version of this article (doi:10.1186/s12861-015-0095-4) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
| | - Antony Athippozhy
- Department of Biology, Spinal Cord and Brain Injury Research Center, University of Kentucky, Lexington, KY, 40506, USA.
| | - Carlos Diaz-Castillo
- Department of Developmental and Cellular Biology, University of California, Irvine, CA, 92602, USA.
| | - Charless Fowlkes
- Donald Bren School of Information and Computer Science, University of California, Irvine, CA, 92602, USA.
| | - David M Gardiner
- Department of Developmental and Cellular Biology, University of California, Irvine, CA, 92602, USA.
| | - S Randal Voss
- Department of Biology, Spinal Cord and Brain Injury Research Center, University of Kentucky, Lexington, KY, 40506, USA.
| |
Collapse
|
27
|
Markway BD, Cho H, Zilberman-Rudenko J, Holden P, McAlinden A, Johnstone B. Hypoxia-inducible factor 3-alpha expression is associated with the stable chondrocyte phenotype. J Orthop Res 2015; 33:1561-70. [PMID: 26174816 DOI: 10.1002/jor.22930] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/26/2015] [Accepted: 04/20/2015] [Indexed: 02/04/2023]
Abstract
The hypoxia-inducible factors HIF-1α and HIF-2α are important regulators of the chondrocyte phenotype but little is known about HIF-3α in cartilage. The objective of this study was to characterize HIF-3α (HIF3A) expression during chondrocyte differentiation in vitro and in native cartilage tissues. HIF3A, COL10A1, and MMP13 were quantified in mesenchymal stem cells (MSCs) and articular chondrocytes from healthy and osteoarthritic (OA) tissue in three-dimensional cultures and in human embryonic epiphyses and adult articular cartilage. HIF3A was found to have an inverse association with hypertrophic markers COL10A1 and MMP13 in chondrogenic cells and tissues. In healthy chondrocytes, HIF3A was induced by dexamethasone and increased during redifferentiation. By comparison, HIF3A expression was extremely low in chondrogenically differentiated MSCs expressing high levels of COL10A1 and MMP13. HIF3A was also lower in redifferentiated OA chondrocytes than in healthy chondrocytes. In human embryonic epiphyseal tissue, HIF3A expression was lowest in the hypertrophic zone. Distinct splice patterns were also found in embryonic cartilage when compared with adult articular cartilage and redifferentiated chondrocytes. These in vitro and in vivo findings suggest that HIF3A levels are indicative of the hypertrophic state of chondrogenic cells and one or more splice variants may be important regulators of the chondrocyte phenotype.
Collapse
Affiliation(s)
- Brandon D Markway
- Department of Orthopaedics and Rehabilitation, Oregon Health & Science University, Portland, Oregon
| | - Holly Cho
- Department of Orthopaedics and Rehabilitation, Oregon Health & Science University, Portland, Oregon
| | | | - Paul Holden
- Department of Orthopaedics and Rehabilitation, Oregon Health & Science University, Portland, Oregon
| | - Audrey McAlinden
- Department of Orthopaedic Surgery, Musculoskeletal Research Center, Washington University, St. Louis, Missouri
| | - Brian Johnstone
- Department of Orthopaedics and Rehabilitation, Oregon Health & Science University, Portland, Oregon
| |
Collapse
|
28
|
Chatzopoulou A, Roy U, Meijer AH, Alia A, Spaink HP, Schaaf MJM. Transcriptional and metabolic effects of glucocorticoid receptor α and β signaling in zebrafish. Endocrinology 2015; 156:1757-69. [PMID: 25756310 DOI: 10.1210/en.2014-1941] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
In humans and zebrafish, 2 glucocorticoid (GC) receptor (GR) splice variants exist: the canonical GR α-isoform (GRα), and the GRβ. In the present study, we have used the zebrafish model system in order to reveal genes affected by each of these 2 receptor isoforms. By injecting zebrafish embryos with different splice-blocking morpholinos, we could knock down both GR isoforms or could target the alternative splicing of the GR pre-mRNA in favor of the GRβ. In addition, specific GRβ overexpression was achieved by injecting mRNA. Embryos were treated with the synthetic GC dexamethasone, and transcriptome analysis was performed. Two distinct gene clusters were found that were regulated by GRα: one that was regulated by GRα under basal conditions (presence of endogenous cortisol only), and one that was regulated upon increased activation of GRα (using a pharmacological dose of dexamathasone). GRβ may act as a dominant-negative inhibitor of GRα when GRβ is overexpressed and the GRα expression level is knocked down simultaneously. However, without GRα knockdown, no evidence for this activity was found. In addition, the data indicate regulation of gene transcription through other mechanisms of action by GRβ. We also investigated the concentrations of several metabolites using nuclear magnetic resonance spectroscopy. We found that dexamethasone treatment and knockdown of GRα together with overexpression of GRβ had opposite effects on glucose, amino acid, and fatty acid levels. Thus, we have shed new light on the molecular mechanisms of GC-induced effects on metabolism, which are known to increase the risk of obesity, hyperglycemia, and diabetes.
Collapse
Affiliation(s)
- Antonia Chatzopoulou
- Department of Animal Sciences and Health (A.C., A.H.M., H.P.S., M.J.M.S.), Institute of Biology, Leiden University, 2333CC Leiden, The Netherlands; Department of Biophysical Organic Chemistry/Solid State NMR (U.R., A.A.), Leiden Institute of Chemistry, Leiden University, 2333CC Leiden, The Netherlands; and Institute of Medical Physics and Biophysics (A.A.), University of Leipzig, D-04107 Leipzig, Germany
| | | | | | | | | | | |
Collapse
|
29
|
Joshi T, Johnson M, Newton R, Giembycz M. An analysis of glucocorticoid receptor-mediated gene expression in BEAS-2B human airway epithelial cells identifies distinct, ligand-directed, transcription profiles with implications for asthma therapeutics. Br J Pharmacol 2015; 172:1360-78. [PMID: 25393397 PMCID: PMC4337707 DOI: 10.1111/bph.13014] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2014] [Revised: 10/08/2014] [Accepted: 11/05/2014] [Indexed: 01/02/2023] Open
Abstract
BACKGROUND AND PURPOSE International asthma guidelines recommend that inhaled glucocorticoids be used as a monotherapy in all patients with mild to moderate disease because of their ability to suppress airways inflammation. Current evidence suggests that the therapeutic benefit of glucocorticoids is due to the transactivation and transrepression of anti-inflammatory and pro-inflammatory genes respectively. However, the extent to which clinically relevant glucocorticoids are equivalent in their ability to modulate gene expression is unclear. EXPERIMENTAL APPROACH A pharmacodynamics investigation of glucocorticoid receptor (GR)-mediated gene transactivation in BEAS-2B human airway epithelial cells was performed using a glucocorticoid response element luciferase reporter coupled with an analysis of glucocorticoid-inducible genes encoding proteins with anti-inflammatory and adverse-effect potential. KEY RESULTS Using transactivation as a functionally relevant output, a given glucocorticoid displayed a unique, gene expression 'fingerprint' where intrinsic efficacy and GR density were essential determinants. We showed that depending on the gene selected for analysis, a given glucocorticoid can behave as an antagonist, partial agonist, full agonist or even 'super agonist'. In the likely event that different, tissue-dependent gene expression profiles are reproduced in vivo, then the anti-inflammatory and adverse-effect potential of many glucocorticoids currently available as asthma therapeutics may not be equivalent. CONCLUSIONS AND IMPLICATIONS The generation of gene expression 'fingerprints' in target and off-target human tissues could assist the rational design of GR agonists with improved therapeutic ratios. This approach could identify compounds that are useful in the management of severe asthma and other inflammatory disorders where systemic exposure is desirable.
Collapse
Affiliation(s)
- T Joshi
- Airways Inflammation Research Group, Department of Physiology and Pharmacology, Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of CalgaryCalgary, AB, Canada
| | - M Johnson
- GlaxoSmithKline Research and DevelopmentUxbridge, Middlesex, UK
| | - R Newton
- Department of Cell Biology and Anatomy, Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of CalgaryCalgary, AB, Canada
| | - M Giembycz
- Airways Inflammation Research Group, Department of Physiology and Pharmacology, Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of CalgaryCalgary, AB, Canada
| |
Collapse
|
30
|
Tu J, Henneicke H, Zhang Y, Stoner S, Cheng TL, Schindeler A, Chen D, Tuckermann J, Cooper MS, Seibel MJ, Zhou H. Disruption of glucocorticoid signaling in chondrocytes delays metaphyseal fracture healing but does not affect normal cartilage and bone development. Bone 2014; 69:12-22. [PMID: 25193158 PMCID: PMC4284102 DOI: 10.1016/j.bone.2014.08.016] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2014] [Revised: 08/18/2014] [Accepted: 08/23/2014] [Indexed: 01/23/2023]
Abstract
States of glucocorticoid excess are associated with defects in chondrocyte function. Most prominently there is a reduction in linear growth but delayed healing of fractures that require endochondral ossification to also occur. In contrast, little is known about the role of endogenous glucocorticoids in chondrocyte function. As glucocorticoids exert their cellular actions through the glucocorticoid receptor (GR), we aimed to elucidate the role of endogenous glucocorticoids in chondrocyte function in vivo through characterization of tamoxifen-inducible chondrocyte-specific GR knockout (chGRKO) mice in which the GR was deleted at various post-natal ages. Knee joint architecture, cartilage structure, growth plates, intervertebral discs, long bone length and bone micro-architecture were similar in chGRKO and control mice at all ages. Analysis of fracture healing in chGRKO and control mice demonstrated that in metaphyseal fractures, chGRKO mice formed a larger cartilaginous callus at 1 and 2 week post-surgery, as well as a smaller amount of well-mineralized bony callus at the fracture site 4 week post-surgery, when compared to control mice. In contrast, chondrocyte-specific GR knockout did not affect diaphyseal fracture healing. We conclude that endogenous GC signaling in chondrocytes plays an important role during metaphyseal fracture healing but is not essential for normal long bone growth.
Collapse
Affiliation(s)
- Jinwen Tu
- Bone Research Program, ANZAC Research Institute, University of Sydney, Sydney, Australia
| | - Holger Henneicke
- Bone Research Program, ANZAC Research Institute, University of Sydney, Sydney, Australia
| | - Yaqing Zhang
- Bone Research Program, ANZAC Research Institute, University of Sydney, Sydney, Australia
| | - Shihani Stoner
- Bone Research Program, ANZAC Research Institute, University of Sydney, Sydney, Australia
| | - Tegan L Cheng
- Orthopaedic Research & Biotechnology Unit, The Children's Hospital at Westmead, Sydney, Australia
| | - Aaron Schindeler
- Orthopaedic Research & Biotechnology Unit, The Children's Hospital at Westmead, Sydney, Australia
| | - Di Chen
- Tissue Department of Biochemistry, Rush University Medical Center, USA
| | - Jan Tuckermann
- Institute of General Zoology and Endocrinology, University of Ulm, Ulm, Germany
| | - Mark S Cooper
- Department of Endocrinology & Metabolism, Concord Hospital, Sydney, Australia
| | - Markus J Seibel
- Bone Research Program, ANZAC Research Institute, University of Sydney, Sydney, Australia; Department of Endocrinology & Metabolism, Concord Hospital, Sydney, Australia
| | - Hong Zhou
- Bone Research Program, ANZAC Research Institute, University of Sydney, Sydney, Australia.
| |
Collapse
|
31
|
Loiselle JJ, Sutherland LC. Differential downregulation of Rbm5 and Rbm10 during skeletal and cardiac differentiation. In Vitro Cell Dev Biol Anim 2013; 50:331-9. [DOI: 10.1007/s11626-013-9708-z] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2013] [Accepted: 10/11/2013] [Indexed: 12/30/2022]
|
32
|
Lewis R, May H, Mobasheri A, Barrett-Jolley R. Chondrocyte channel transcriptomics: do microarray data fit with expression and functional data? Channels (Austin) 2013; 7:459-67. [PMID: 23995703 PMCID: PMC4042480 DOI: 10.4161/chan.26071] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
To date, a range of ion channels have been identified in chondrocytes using a number of different techniques, predominantly electrophysiological and/or biomolecular; each of these has its advantages and disadvantages. Here we aim to compare and contrast the data available from biophysical and microarray experiments. This letter analyses recent transcriptomics datasets from chondrocytes, accessible from the European Bioinformatics Institute (EBI). We discuss whether such bioinformatic analysis of microarray datasets can potentially accelerate identification and discovery of ion channels in chondrocytes. The ion channels which appear most frequently across these microarray datasets are discussed, along with their possible functions. We discuss whether functional or protein data exist which support the microarray data. A microarray experiment comparing gene expression in osteoarthritis and healthy cartilage is also discussed and we verify the differential expression of 2 of these genes, namely the genes encoding large calcium-activated potassium (BK) and aquaporin channels.
Collapse
Affiliation(s)
- Rebecca Lewis
- Musculoskeletal Biology; Institute of Ageing and Chronic Disease; Faculty of Health & Life Sciences; University of Liverpool; Liverpool, UK; The D-BOARD European Consortium for Biomarker Discovery
| | - Hannah May
- Musculoskeletal Biology; Institute of Ageing and Chronic Disease; Faculty of Health & Life Sciences; University of Liverpool; Liverpool, UK
| | - Ali Mobasheri
- Arthritis Research UK Centre for Sport, Exercise and Osteoarthritis; Arthritis Research UK Pain Centre; Medical Research Council and Arthritis Research UK Centre for Musculoskeletal Ageing Research; The University of Nottingham; Queen's Medical Centre; Nottingham, UK; School of Life Sciences; University of Bradford; Bradford, UK; Center for Excellence in Genomic Medicine Research (CEGMR); King Fahad Medical Research Center (KFMRC); King AbdulAziz University; Jeddah, Saudi Arabia; The D-BOARD European Consortium for Biomarker Discovery
| | - Richard Barrett-Jolley
- Musculoskeletal Biology; Institute of Ageing and Chronic Disease; Faculty of Health & Life Sciences; University of Liverpool; Liverpool, UK; The D-BOARD European Consortium for Biomarker Discovery
| |
Collapse
|
33
|
Prykhozhij SV, Marsico A, Meijsing SH. Zebrafish Expression Ontology of Gene Sets (ZEOGS): a tool to analyze enrichment of zebrafish anatomical terms in large gene sets. Zebrafish 2013; 10:303-15. [PMID: 23656298 DOI: 10.1089/zeb.2012.0865] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
The zebrafish (Danio rerio) is an established model organism for developmental and biomedical research. It is frequently used for high-throughput functional genomics experiments, such as genome-wide gene expression measurements, to systematically analyze molecular mechanisms. However, the use of whole embryos or larvae in such experiments leads to a loss of the spatial information. To address this problem, we have developed a tool called Zebrafish Expression Ontology of Gene Sets (ZEOGS) to assess the enrichment of anatomical terms in large gene sets. ZEOGS uses gene expression pattern data from several sources: first, in situ hybridization experiments from the Zebrafish Model Organism Database (ZFIN); second, it uses the Zebrafish Anatomical Ontology, a controlled vocabulary that describes connected anatomical structures; and third, the available connections between expression patterns and anatomical terms contained in ZFIN. Upon input of a gene set, ZEOGS determines which anatomical structures are overrepresented in the input gene set. ZEOGS allows one for the first time to look at groups of genes and to describe them in terms of shared anatomical structures. To establish ZEOGS, we first tested it on random gene selections and on two public microarray datasets with known tissue-specific gene expression changes. These tests showed that ZEOGS could reliably identify the tissues affected, whereas only very few enriched terms to none were found in the random gene sets. Next we applied ZEOGS to microarray datasets of 24 and 72 h postfertilization zebrafish embryos treated with beclomethasone, a potent glucocorticoid. This analysis resulted in the identification of several anatomical terms related to glucocorticoid-responsive tissues, some of which were stage-specific. Our studies highlight the ability of ZEOGS to extract spatial information from datasets derived from whole embryos, indicating that ZEOGS could be a useful tool to automatically analyze gene expression pattern features of any large zebrafish gene set.
Collapse
|
34
|
Huey DJ, Hu JC, Athanasiou KA. Chondrogenically tuned expansion enhances the cartilaginous matrix-forming capabilities of primary, adult, leporine chondrocytes. Cell Transplant 2012; 22:331-40. [PMID: 23044188 DOI: 10.3727/096368912x657648] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
When expanded through passage, chondrocytes lose their ability to produce high-quality cartilaginous matrix. This study attempts to improve the properties of constructs formed with expanded chondrocytes through alterations in the expansion protocol and the ratio of primary to expanded chondrocytes used to form cartilage constructs. A chondrogenically tuned expansion protocol provided similar monolayer growth rates as those obtained using serum-containing medium and enhanced cartilaginous properties of resultant constructs. Various ratios of primary to chondrogenically expanded chondrocytes were then self-assembled to form neocartilage. Biochemical analysis showed that constructs formed with only expanded cells had twice the GAG per wet weight and collagen II/collagen I ratio compared to constructs formed with primary chondrocytes. Biomechanically, compressive properties of constructs formed with only passaged cells matched the instantaneous modulus and exceeded the relaxation modulus of constructs formed with only primary cells. These counterintuitive results show that, by applying proper expansion and three-dimensional culture techniques, the cartilage-forming potential of adult chondrocytes expanded through passage can be enhanced over that of primary cells.
Collapse
Affiliation(s)
- Daniel J Huey
- Department of Biomedical Engineering, University of California-Davis, Davis, CA 95616, USA
| | | | | |
Collapse
|
35
|
Chindelevitch L, Loh PR, Enayetallah A, Berger B, Ziemek D. Assessing statistical significance in causal graphs. BMC Bioinformatics 2012; 13:35. [PMID: 22348444 PMCID: PMC3307026 DOI: 10.1186/1471-2105-13-35] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2011] [Accepted: 02/20/2012] [Indexed: 12/20/2022] Open
Abstract
Background Causal graphs are an increasingly popular tool for the analysis of biological datasets. In particular, signed causal graphs--directed graphs whose edges additionally have a sign denoting upregulation or downregulation--can be used to model regulatory networks within a cell. Such models allow prediction of downstream effects of regulation of biological entities; conversely, they also enable inference of causative agents behind observed expression changes. However, due to their complex nature, signed causal graph models present special challenges with respect to assessing statistical significance. In this paper we frame and solve two fundamental computational problems that arise in practice when computing appropriate null distributions for hypothesis testing. Results First, we show how to compute a p-value for agreement between observed and model-predicted classifications of gene transcripts as upregulated, downregulated, or neither. Specifically, how likely are the classifications to agree to the same extent under the null distribution of the observed classification being randomized? This problem, which we call "Ternary Dot Product Distribution" owing to its mathematical form, can be viewed as a generalization of Fisher's exact test to ternary variables. We present two computationally efficient algorithms for computing the Ternary Dot Product Distribution and investigate its combinatorial structure analytically and numerically to establish computational complexity bounds. Second, we develop an algorithm for efficiently performing random sampling of causal graphs. This enables p-value computation under a different, equally important null distribution obtained by randomizing the graph topology but keeping fixed its basic structure: connectedness and the positive and negative in- and out-degrees of each vertex. We provide an algorithm for sampling a graph from this distribution uniformly at random. We also highlight theoretical challenges unique to signed causal graphs; previous work on graph randomization has studied undirected graphs and directed but unsigned graphs. Conclusion We present algorithmic solutions to two statistical significance questions necessary to apply the causal graph methodology, a powerful tool for biological network analysis. The algorithms we present are both fast and provably correct. Our work may be of independent interest in non-biological contexts as well, as it generalizes mathematical results that have been studied extensively in other fields.
Collapse
Affiliation(s)
- Leonid Chindelevitch
- Computational Sciences Center of Emphasis, Pfizer Worldwide Research & Development, Cambridge, MA, USA
| | | | | | | | | |
Collapse
|
36
|
Wang K, Bacon ML, Tessier JJ, Rintala-Maki ND, Tang V, Sutherland LC. RBM10 Modulates Apoptosis and Influences TNF-α Gene Expression. J Cell Death 2012; 5:1-19. [PMID: 26446321 PMCID: PMC4583097 DOI: 10.4137/jcd.s9073] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023] Open
Abstract
Recent evidence suggests that protein encoded by the RNA Binding Motif 10 (RBM10) gene has the ability to modulate apoptosis. The objective of this study was to test this hypothesis by manipulating RBM10 expression levels and examining the downstream consequences. The results showed that transient overexpression of RBM10 correlated with significantly elevated levels of tumour necrosis factor alpha (TNF-α) mRNA and soluble TNF-α (sTNF-α) protein, and increased apoptosis (phosphatidyl serine exposure on the outer cell membrane and nuclear condensation). Stable RNA interference-mediated RBM10 knockdown clones were less susceptible to TNF-α-mediated apoptosis, and had decreased sTNF-α protein levels. Elevated levels of TNF-α associated with RBM10 overexpression resulted from increased TNF-α transcription, not TNF-α mRNA stabilization. These results suggest that RBM10 has the ability to modulate apoptosis, and that it does so via a mechanism involving alterations to TNFR super family-mediated signaling. These data provide the first direct evidence that human RBM10 can function as an apoptosis modulator and cytokine expression regulator.
Collapse
Affiliation(s)
- Ke Wang
- Tumour Biology Group, North East Cancer Centre, Health Sciences North, Laurentian University, Sudbury, Ontario, Canada
| | - Mackensey L Bacon
- Tumour Biology Group, North East Cancer Centre, Health Sciences North, Laurentian University, Sudbury, Ontario, Canada
| | - Julie J Tessier
- Tumour Biology Group, North East Cancer Centre, Health Sciences North, Laurentian University, Sudbury, Ontario, Canada
| | - Nina D Rintala-Maki
- Tumour Biology Group, North East Cancer Centre, Health Sciences North, Laurentian University, Sudbury, Ontario, Canada
| | - Vanessa Tang
- Tumour Biology Group, North East Cancer Centre, Health Sciences North, Laurentian University, Sudbury, Ontario, Canada
| | - Leslie C Sutherland
- Tumour Biology Group, North East Cancer Centre, Health Sciences North, Laurentian University, Sudbury, Ontario, Canada ; Division of Medical Sciences, Northern Ontario School of Medicine, Laurentian University, Sudbury, Ontario, Canada ; Biomolecular Sciences Program, Laurentian University, Sudbury, Ontario, Canada ; Department of Chemistry and Biochemistry, Laurentian University, Sudbury, Ontario, Canada ; Department of Medicine, Division of Medical Oncology, University of Ottawa, Ontario, Canada
| |
Collapse
|
37
|
Woods A, James CG, Wang G, Dupuis H, Beier F. Control of chondrocyte gene expression by actin dynamics: a novel role of cholesterol/Ror-alpha signalling in endochondral bone growth. J Cell Mol Med 2011; 13:3497-516. [PMID: 20196782 PMCID: PMC4516504 DOI: 10.1111/j.1582-4934.2009.00684.x] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Elucidating the signalling pathways that regulate chondrocyte differentiation, such as the actin cytoskeleton and Rho GTPases, during development is essential for understanding of pathological conditions of cartilage, such as chondrodysplasias and osteoarthritis. Manipulation of actin dynamics in tibia organ cultures isolated from E15.5 mice results in pronounced enhancement of endochondral bone growth and specific changes in growth plate architecture. Global changes in gene expression were examined of primary chondrocytes isolated from embryonic tibia, treated with the compounds cytochalasin D, jasplakinolide (actin modifiers) and the ROCK inhibitor Y27632. Cytochalasin D elicited the most pronounced response and induced many features of hypertrophic chondrocyte differentiation. Bioinformatics analyses of microarray data and expression validation by real-time PCR and immunohistochemistry resulted in the identification of the nuclear receptor retinoid related orphan receptor-α (Ror-α) as a novel putative regulator of chondrocyte hypertrophy. Expression of Ror-α target genes, (Lpl, fatty acid binding protein 4 [Fabp4], Cd36 and kruppel-like factor 5 [Klf15]) were induced during chondrocyte hypertrophy and by cytochalasin D and are cholesterol dependent. Stimulation of Ror-α by cholesterol results in increased bone growth and enlarged, rounded cells, a phenotype similar to chondrocyte hypertrophy and to the changes induced by cytochalasin D, while inhibition of cholesterol synthesis by lovastatin inhibits cytochalasin D induced bone growth. Additionally, we show that in a mouse model of cartilage specific (Col2-Cre) Rac1, inactivation results in increased Hif-1α (a regulator of Rora gene expression) and Ror-α+ cells within hypertrophic growth plates. We provide evidence that cholesterol signalling through increased Ror-α expression stimulates chondrocyte hypertrophy and partially mediates responses of cartilage to actin dynamics.
Collapse
Affiliation(s)
- Anita Woods
- CIHR Group in Skeletal Development and Remodeling, Department of Physiology and Pharmacology, Schulich School of Medicine and Dentistry, The University of Western Ontario, London, Ontario, Canada
| | | | | | | | | |
Collapse
|
38
|
Taneda T, Zhu W, Cao Q, Watanabe H, Yamaguchi Y, Handa H, Wada T. Erythropoiesis is regulated by the transcription elongation factor Foggy/Spt5 through gata1 gene regulation. Genes Cells 2011; 16:231-42. [DOI: 10.1111/j.1365-2443.2010.01481.x] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
|
39
|
Abstract
Glucocorticoids have a direct, inhibitory effect on the growth plate, as demonstrated by in vivo and organ culture studies. Glucocorticoids slow longitudinal bone growth by inhibiting chondrocyte proliferation, hypertrophy, and cartilage matrix synthesis. The molecular mediators of these effects are poorly understood. Glucocorticoids also delay growth plate senescence. The decreased rate of senescence appears to be a consequence of the growth inhibition and, in particular, may occur because glucocorticoids slow proliferation of stem-like cells in the resting zone and therefore conserve the limited proliferative capacity of these cells. This slowing of senescence appears to explain the phenomenon of catch-up growth following transient glucocorticoid exposure. After the exposure, the growth plate is less senescent, and therefore grows more rapidly than is normal for age. Glucocorticoids cause growth inhibition and subsequent catch-up growth not only in terms of longitudinal bone growth at the growth plate but also in terms of cross-sectional bone growth at the periosteum. Whether the underlying mechanisms are analogous to those at the growth plate is not known.
Collapse
|
40
|
Rauch A, Seitz S, Baschant U, Schilling AF, Illing A, Stride B, Kirilov M, Mandic V, Takacz A, Schmidt-Ullrich R, Ostermay S, Schinke T, Spanbroek R, Zaiss MM, Angel PE, Lerner UH, David JP, Reichardt HM, Amling M, Schütz G, Tuckermann JP. Glucocorticoids suppress bone formation by attenuating osteoblast differentiation via the monomeric glucocorticoid receptor. Cell Metab 2010; 11:517-31. [PMID: 20519123 DOI: 10.1016/j.cmet.2010.05.005] [Citation(s) in RCA: 305] [Impact Index Per Article: 21.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/14/2009] [Revised: 12/25/2009] [Accepted: 05/04/2010] [Indexed: 01/10/2023]
Abstract
Development of osteoporosis severely complicates long-term glucocorticoid (GC) therapy. Using a Cre-transgenic mouse line, we now demonstrate that GCs are unable to repress bone formation in the absence of glucocorticoid receptor (GR) expression in osteoblasts as they become refractory to hormone-induced apoptosis, inhibition of proliferation, and differentiation. In contrast, GC treatment still reduces bone formation in mice carrying a mutation that only disrupts GR dimerization, resulting in bone loss in vivo, enhanced apoptosis, and suppressed differentiation in vitro. The inhibitory GC effects on osteoblasts can be explained by a mechanism involving suppression of cytokines, such as interleukin 11, via interaction of the monomeric GR with AP-1, but not NF-kappaB. Thus, GCs inhibit cytokines independent of GR dimerization and thereby attenuate osteoblast differentiation, which accounts, in part, for bone loss during GC therapy.
Collapse
Affiliation(s)
- Alexander Rauch
- Tissue-specific hormone action, Leibniz Institute for Age Research, Fritz Lipmann Institute, 07745 Jena, Germany
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Abstract
An analysis of mRNA expression in T47D breast cancer cells treated with the synthetic progestin R5020 revealed a subset of progesterone receptor (PR) target genes that are enriched for E2F binding sites. Following up on this observation, we determined that PR-B acts in both direct and indirect manners to positively upregulate E2F1 expression in T47D cells. The direct effects of PR on E2F1 expression were confirmed by chromatin immunoprecipitation (ChIP) analysis, which indicated that the agonist-bound receptor was recruited to several enhancer elements proximal to the E2F1 transcript. However, we also noted that cycloheximide partially inhibits R5020 induction of E2F1 expression, indicating that the ligand-dependent actions of PR on this gene may involve additional indirect regulatory pathways. In support of this hypothesis, we demonstrated that treatment with R5020 significantly increases both hyperphosphorylation of Rb and recruitment of E2F1 to its own promoter, thus activating a positive feedback loop that further amplifies its transcription. Furthermore, we established that PR-mediated induction of Krüppel-like factor 15 (KLF15), which can bind to GC-rich DNA within the E2F1 promoter, is required for maximal induction of E2F1 expression by progestins. Taken together, these results suggest a new paradigm for multimodal regulation of target gene expression by PR.
Collapse
|
42
|
Woods A, James CG, Wang G, Dupuis H, Beier F. Control of chondrocyte gene expression by actin dynamics: a novel role of cholesterol/Ror-α signalling in endochondral bone growth. J Cell Mol Med 2010. [DOI: 10.1111/j.1582-4934.2008.00684.x] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023] Open
|
43
|
Ulici V, James CG, Hoenselaar KD, Beier F. Regulation of gene expression by PI3K in mouse growth plate chondrocytes. PLoS One 2010; 5:e8866. [PMID: 20111593 PMCID: PMC2810323 DOI: 10.1371/journal.pone.0008866] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2009] [Accepted: 01/02/2010] [Indexed: 02/02/2023] Open
Abstract
Background Endochondral ossification, the process through which long bones are formed, involves chondrocyte proliferation and hypertrophic differentiation in the cartilage growth plate. In a previous publication we showed that pharmacological inhibition of the PI3K signaling pathway results in reduced endochondral bone growth, and in particular, shortening of the hypertrophic zone in a tibia organ culture system. In this current study we aimed to investigate targets of the PI3K signaling pathway in hypertrophic chondrocytes. Methodology/Principal Findings Through the intersection of two different microarray analyses methods (classical single gene analysis and GSEA) and two different chondrocyte differentiation systems (primary chondrocytes treated with a pharmacological inhibitor of PI3K and microdissected growth plates), we were able to identify a high number of genes grouped in GSEA functional categories regulated by the PI3K signaling pathway. Genes such as Phlda2 and F13a1 were down-regulated upon PI3K inhibition and showed increased expression in the hypertrophic zone compared to the proliferative/resting zone of the growth plate. In contrast, other genes including Nr4a1 and Adamts5 were up-regulated upon PI3K inhibition and showed reduced expression in the hypertrophic zone. Regulation of these genes by PI3K signaling was confirmed by quantitative RT-PCR. We focused on F13a1 as an interesting target because of its known role in chondrocyte hypertrophy and osteoarthritis. Mouse E15.5 tibiae cultured with LY294002 (PI3K inhibitor) for 6 days showed decreased expression of factor XIIIa in the hypertrophic zone compared to control cultures. Conclusions/Significance Discovering targets of signaling pathways in hypertrophic chondrocytes could lead to targeted therapy in osteoarthritis and a better understanding of the cartilage environment for tissue engineering.
Collapse
Affiliation(s)
- Veronica Ulici
- CIHR Group in Skeletal Development and Remodeling, Department of Physiology and Pharmacology, University of Western Ontario, London, Ontario, Canada
| | - Claudine G. James
- CIHR Group in Skeletal Development and Remodeling, Department of Physiology and Pharmacology, University of Western Ontario, London, Ontario, Canada
| | - Katie D. Hoenselaar
- CIHR Group in Skeletal Development and Remodeling, Department of Physiology and Pharmacology, University of Western Ontario, London, Ontario, Canada
| | - Frank Beier
- CIHR Group in Skeletal Development and Remodeling, Department of Physiology and Pharmacology, University of Western Ontario, London, Ontario, Canada
- * E-mail:
| |
Collapse
|
44
|
James CG, Stanton LA, Agoston H, Ulici V, Underhill TM, Beier F. Genome-wide analyses of gene expression during mouse endochondral ossification. PLoS One 2010; 5:e8693. [PMID: 20084171 PMCID: PMC2805713 DOI: 10.1371/journal.pone.0008693] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2009] [Accepted: 12/13/2009] [Indexed: 12/24/2022] Open
Abstract
Background Endochondral ossification is a complex process involving a series of events that are initiated by the establishment of a chondrogenic template and culminate in its replacement through the coordinated activity of osteoblasts, osteoclasts and endothelial cells. Comprehensive analyses of in vivo gene expression profiles during these processes are essential to obtain a complete understanding of the regulatory mechanisms involved. Methodology/Principal Findings To address these issues, we completed a microarray screen of three zones derived from manually segmented embryonic mouse tibiae. Classification of genes differentially expressed between each respective zone, functional categorization as well as characterization of gene expression patterns, cytogenetic loci, signaling pathways and functional motifs both confirmed reported data and provided novel insights into endochondral ossification. Parallel comparisons of the microdissected tibiae data set with our previously completed micromass culture screen further corroborated the suitability of micromass cultures for modeling gene expression in chondrocyte development. The micromass culture system demonstrated striking similarities to the in vivo microdissected tibiae screen; however, the micromass system was unable to accurately distinguish gene expression differences in the hypertrophic and mineralized zones of the tibia. Conclusions/Significance These studies allow us to better understand gene expression patterns in the growth plate and endochondral bones and provide an important technical resource for comparison of gene expression in diseased or experimentally-manipulated cartilages. Ultimately, this work will help to define the genomic context in which genes are expressed in long bones and to understand physiological and pathological ossification.
Collapse
Affiliation(s)
- Claudine G. James
- CIHR Group in Skeletal Development and Remodelling, Department of Physiology and Pharmacology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, Canada
| | - Lee-Anne Stanton
- CIHR Group in Skeletal Development and Remodelling, Department of Physiology and Pharmacology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, Canada
| | - Hanga Agoston
- CIHR Group in Skeletal Development and Remodelling, Department of Physiology and Pharmacology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, Canada
| | - Veronica Ulici
- CIHR Group in Skeletal Development and Remodelling, Department of Physiology and Pharmacology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, Canada
- * E-mail: (VU); (FB)
| | - T. Michael Underhill
- Department of Cellular and Physiological Sciences, University of British Columbia, Vancouver, British Columbia, Canada
| | - Frank Beier
- CIHR Group in Skeletal Development and Remodelling, Department of Physiology and Pharmacology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, Canada
- * E-mail: (VU); (FB)
| |
Collapse
|
45
|
Newton R, Leigh R, Giembycz MA. Pharmacological strategies for improving the efficacy and therapeutic ratio of glucocorticoids in inflammatory lung diseases. Pharmacol Ther 2009; 125:286-327. [PMID: 19932713 DOI: 10.1016/j.pharmthera.2009.11.003] [Citation(s) in RCA: 78] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2009] [Accepted: 11/02/2009] [Indexed: 10/20/2022]
Abstract
Glucocorticoids are widely used to treat various inflammatory lung diseases. Acting via the glucocorticoid receptor (GR), they exert clinical effects predominantly by modulating gene transcription. This may be to either induce (transactivate) or repress (transrepress) gene transcription. However, certain individuals, including those who smoke, have certain asthma phenotypes, chronic obstructive pulmonary disease (COPD) or some interstitial diseases may respond poorly to the beneficial effects of glucocorticoids. In these cases, high dose, often oral or parental, glucocorticoids are typically prescribed. This generally leads to adverse effects that compromise clinical utility. There is, therefore, a need to enhance the clinical efficacy of glucocorticoids while minimizing adverse effects. In this context, a long-acting beta(2)-adrenoceptor agonist (LABA) can enhance the clinical efficacy of an inhaled corticosteroid (ICS) in asthma and COPD. Furthermore, LABAs can augment glucocorticoid-dependent gene expression and this action may account for some of the benefits of LABA/ICS combination therapies when compared to ICS given as a monotherapy. In addition to metabolic genes and other adverse effects that are induced by glucocorticoids, there are many other glucocorticoid-inducible genes that have significant anti-inflammatory potential. We therefore advocate a move away from the search for ligands of GR that dissociate transactivation from transrepression. Instead, we submit that ligands should be functionally screened by virtue of their ability to induce or repress biologically-relevant genes in target tissues. In this review, we discuss pharmacological methods by which selective GR modulators and "add-on" therapies may be exploited to improve the clinical efficacy of glucocorticoids while reducing potential adverse effects.
Collapse
Affiliation(s)
- Robert Newton
- Department of Cell Biology and Anatomy, Airway Inflammation Group, Institute of Infection, Immunity and Inflammation, Faculty of Medicine, University of Calgary, Calgary, Alberta, Canada.
| | | | | |
Collapse
|
46
|
Stewart MD, Wong J. Nuclear receptor repression: regulatory mechanisms and physiological implications. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2009; 87:235-59. [PMID: 20374706 DOI: 10.1016/s1877-1173(09)87007-5] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
The ability to associate with corepressors and to inhibit transcription is an intrinsic property of most members of the nuclear receptor (NR) superfamily. NRs induce transcriptional repression by recruiting multiprotein corepressor complexes. Nuclear receptor corepressor (NCoR) and silencing mediator of retinoic and thyroid receptors (SMRT) are the most well characterized corepressor complexes and mediate repression for virtually all NRs. In turn, corepressor complexes repress transcription because they either contain or associate with chromatin modifying enzymes. These include histone deacetylases, histone H3K4 demethylases, histone H3K9 or H3K27 methyltransferases, and ATP-dependent chromatin remodeling factors. Two types of NR-interacting corepressors exist. Ligand-independent corepressors, like NCoR/SMRT, bind to unliganded or antagonist-bound NRs, whereas ligand-dependent corepressors (LCoRs) associate with NRs in the presence of agonist. Therefore, LCoRs may serve to attenuate NR-mediated transcriptional activation. Somewhat unexpectedly, classical coactivators may also function as "corepressors" to mediate repression by agonist-bound NRs. In this chapter, we will discuss the various modes and mechanisms of repression by NRs as well as discuss the known physiological functions of NR-mediated repression.
Collapse
Affiliation(s)
- M David Stewart
- Department of Genetics, University of Texas M.D. Anderson Cancer Center, Houston, Texas 77030, USA
| | | |
Collapse
|
47
|
Ho STB, Yang Z, Hui HPJ, Oh KWS, Choo BHA, Lee EH. A serum free approach towards the conservation of chondrogenic phenotype during in vitro cell expansion. Growth Factors 2009; 27:321-33. [PMID: 19626506 DOI: 10.1080/08977190903137595] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
OBJECTIVE Functionally viable chondrocytes in sufficient quantity is crucial for the success of matrix associated autologous chondrocyte implantation. This is difficult with conventional methods as chondrocytes dedifferentiate during 2D expansion with the loss of their chondrogenic phenotype. Moreover, established protocols are dependent on the use of serum which is not without its drawbacks. This study sought to address the issue by evaluating the feasibility of serum free, growth factors supplemented chondrocyte media with extracellular matrix (ECM) coatings. DESIGN Passage 2 human chondrocytes were cultured in serum supplemented media or serum free media with collagen I or fibronectin coatings. Cell attachment and proliferation were assessed in these conditions. The cells were redifferentiated via pellet cultures for 7 and 14 days before being subjected to histological and gene expression analysis. RESULTS The serum-free, growth factor cocktail supplemented with ECM coating improved long-term chondrocyte proliferation with enhanced basal Sox 9 expression. Upon induction, the redifferentiated chondrocytes expressed aggrecan and collagen II especially so for the cells plated on collagen coated surfaces. The chondrocytic phenotype was better conserved under the serum free conditions but the loss of the hyaline cartilage characteristics was not completely halted given the expression of collagen I. These essential cartilage markers were, however, reduced or absented for cells expanded with serum. Moreover, serum cultures displayed a higher tendency of undergoing hypertrophy given the stronger collagen X gene expression. CONCLUSION The advocated technique promoted cell expansion with respect to conventional serum supplemented cultures while reducing the loss of the chondrogenic phenotype. This demonstrates the feasibility and potential of the novel concomitant use of serum free media and ECM coatings in the expansion of chondrocytes for cartilage regenerative applications.
Collapse
Affiliation(s)
- Saey Tuan Barnabas Ho
- Department of Orthopaedic Surgery, Yong Loo Lin School of Medical, National University of Singapore, Singapore, Singapore
| | | | | | | | | | | |
Collapse
|
48
|
Owen HC, Ahmed SF, Farquharson C. Chondrocyte p21(WAF1/CIP1) expression is increased by dexamethasone but does not contribute to dexamethasone-induced growth retardation in vivo. Calcif Tissue Int 2009; 85:326-34. [PMID: 19727539 DOI: 10.1007/s00223-009-9276-0] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/30/2009] [Accepted: 07/29/2009] [Indexed: 01/03/2023]
Abstract
It has been shown that cell cycle genes play an important role in the coordination of chondrocyte proliferation and differentiation. The inhibitory effects of glucocorticoids (GCs) on chondrocyte proliferation are consistent with GCs disrupting cell cycle progression and promoting cell cycle exit. Cyclin-dependent kinase inhibitors (CDKIs) force cells to exit the cell cycle and differentiate, and studies have shown that expression of the CDKI p21(CIP1/WAF1) is increased in terminally differentiated cells. In this study, p21 mRNA and protein expression was increased during chondrocyte differentiation and after exposure to dexamethasone (Dex, 10(-6 )M) in murine chondrogenic ATDC5 cells. In 4-week-old mice lacking a functional p21 gene, Dex caused a reduction in body weight compared to saline control null mice, but this was consistent with the reduction in body weight observed in Dex-treated wild-type littermates. In addition, p21 ablation had no effect on the reduction in width of the growth plate or reduced mineral apposition rate in Dex-treated mice. However, an alteration in growth rate and epiphyseal structure is evident when comparing p21(-/-) and wild-type mice. These findings suggest that p21 does not directly contribute to GC-induced growth retardation in vivo but is involved in the maintenance of the growth plate.
Collapse
Affiliation(s)
- H C Owen
- Bone Biology Group, Division of Developmental Biology, The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Roslin, Edinburgh, Midlothian, EH25 9PS, UK
| | | | | |
Collapse
|
49
|
Solomon LA, Li JR, Bérubé NG, Beier F. Loss of ATRX in chondrocytes has minimal effects on skeletal development. PLoS One 2009; 4:e7106. [PMID: 19774083 PMCID: PMC2744333 DOI: 10.1371/journal.pone.0007106] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2009] [Accepted: 08/25/2009] [Indexed: 11/19/2022] Open
Abstract
Background Mutations in the human ATRX gene cause developmental defects, including skeletal deformities and dwarfism. ATRX encodes a chromatin remodeling protein, however the role of ATRX in skeletal development is currently unknown. Methodology/Principal Findings We induced Atrx deletion in mouse cartilage using the Cre-loxP system, with Cre expression driven by the collagen II (Col2a1) promoter. Growth rate, body size and weight, and long bone length did not differ in AtrxCol2cre mice compared to control littermates. Histological analyses of the growth plate did not reveal any differences between control and mutant mice. Expression patterns of Sox9, a transcription factor required for cartilage morphogenesis, and p57, a marker of cell cycle arrest and hypertrophic chondrocyte differentiation, was unaffected. However, loss of ATRX in cartilage led to a delay in the ossification of the hips in some mice. We also observed hindlimb polydactily in one out of 61 mutants. Conclusions/Significance These findings indicate that ATRX is not directly required for development or growth of cartilage in the mouse, suggesting that the short stature in ATR-X patients is caused by defects in cartilage-extrinsic mechanisms.
Collapse
Affiliation(s)
- Lauren A. Solomon
- Department of Biochemistry, University of Western Ontario, London, Ontario, Canada
- Children's Health Research Institute, Victoria Research Laboratories, London, Ontario, Canada
- Skeletal Biology Group, University of Western Ontario, London, Ontario, Canada
| | - Jennifer R. Li
- Department of Biochemistry, University of Western Ontario, London, Ontario, Canada
- Children's Health Research Institute, Victoria Research Laboratories, London, Ontario, Canada
- Skeletal Biology Group, University of Western Ontario, London, Ontario, Canada
| | - Nathalie G. Bérubé
- Department of Biochemistry, University of Western Ontario, London, Ontario, Canada
- Children's Health Research Institute, Victoria Research Laboratories, London, Ontario, Canada
- Department of Paediatrics, University of Western Ontario, London, Ontario, Canada
- * E-mail: (NB); (FB)
| | - Frank Beier
- Department of Biochemistry, University of Western Ontario, London, Ontario, Canada
- Skeletal Biology Group, University of Western Ontario, London, Ontario, Canada
- Department of Paediatrics, University of Western Ontario, London, Ontario, Canada
- Department of Physiology and Pharmacology, University of Western Ontario, London, Ontario, Canada
- * E-mail: (NB); (FB)
| |
Collapse
|
50
|
Polar lipid remodeling and increased sulfatide expression are associated with the glioma therapeutic candidates, wild type p53 elevation and the topoisomerase-1 inhibitor, irinotecan. Glycoconj J 2009; 27:27-38. [PMID: 19557511 DOI: 10.1007/s10719-009-9249-6] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2008] [Revised: 03/10/2009] [Accepted: 05/29/2009] [Indexed: 10/20/2022]
Abstract
We report changes in gene and polar lipid expression induced by adenovirus-delivered wild-type (wt) p53 gene and chemotherapy of U87 MG glioblastoma cells, a treatment known to trigger apoptosis and cell cycle arrest. Sulfatides (sulfonated glycolipids) were most highly modulated by wild-type p53 treatment; however, no changes were observed in expression levels of mRNA for genes involved in sulfatide metabolism, indicating post-transcriptional control of sulfatide synthesis. Modulation of the aglycones of GD1 and GM1b was observed in wild-type p53-treated cells. The treatment also leads to an increase in phospholipids such as phosphatidyl inositols, phosphatidyl serines, phosphatidyl glycerols, and phosphatidyl ethanolamines, especially hydroxylated phospholipids. These dramatic changes in the composition of cellular glycolipids in response to p53 gene expression and cytotoxic chemotherapy treatment indicate the large role that they play in cell signaling. The use of the human glioma cell line U87 appears to be an excellent model system both in tissue culture and in intracranial murine xenograft models to further characterize the role of sulfatides in modulating glioma responsivity to therapeutic agents.
Collapse
|