1
|
Li Y, Wang Y, Liu Q, Lv S, Wang Y, Zhang H, Zhao Q, Shang L. Kaempferol promotes osteogenic differentiation in bone marrow mesenchymal stem cells by inhibiting CAV-1. J Orthop Surg Res 2024; 19:678. [PMID: 39434162 PMCID: PMC11495062 DOI: 10.1186/s13018-024-05174-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Accepted: 10/13/2024] [Indexed: 10/23/2024] Open
Abstract
OBJECTIVE Our study focused on the effects and molecular mechanisms of kaempferol, a major active component of Eucommia ulmoides Oliver (EUO), on the osteogenic differentiation of bone marrow mesenchymal stem cells (BMSCs). METHODS Target molecules for EUO, osteoarthritis, and osteogenic differentiation were identified through network pharmacology analysis. BMSCs were isolated and treated with various concentrations of kaempferol. Optimal concentration was determined through MTT assays. Osteogenic differentiation was assessed using alkaline phosphatase (ALP) and Alizarin Red S staining, while osteogenic markers (Collagen I, RUNX2, and OPN) and CAV-1 expression were analyzed using RT-qPCR and Western blot. The effects of combined treatment with kaempferol and an overexpression vector for CAV-1 (oe-CAV-1) on osteogenic differentiation were also observed. RESULTS Network pharmacology analysis identified kaempferol as the primary active component influencing CAV-1 targeted in subsequent experiments. It was found that 10 µM kaempferol was optimal for treating BMSCs. Post-treatment, significant increases in ALP activity and calcium deposition were observed, along with elevated expression of osteogenic markers, and decreased CAV-1. Overexpression of CAV-1 significantly reversed the promotive effects of kaempferol on BMSC osteogenic differentiation, effectively inhibiting the process. CONCLUSION Collectively, kaempferol promotes osteogenic differentiation in BMSCs by inhibiting CAV-1 expression.
Collapse
Affiliation(s)
- Yingxue Li
- 7th Ward, Department of Rheumatology and Immunology, Xi'an No.5 Hospital, Xi'an, 710082, Shaanxi, P.R. China
| | - Ying Wang
- 5th Ward, Department of Rheumatology and Immunology, Xi'an No.5 Hospital, Xi'an, 710082, Shaanxi, P.R. China
| | - Qian Liu
- 5th Ward, Department of Rheumatology and Immunology, Xi'an No.5 Hospital, Xi'an, 710082, Shaanxi, P.R. China
| | - Shuiying Lv
- 7th Ward, Department of Rheumatology and Immunology, Xi'an No.5 Hospital, Xi'an, 710082, Shaanxi, P.R. China
| | - Yali Wang
- Department of Traditional Chinese Medicine, Xi'an International Medical Center Hospital, No. 777, Xitai Road, High-tech Zone, Xi'an, 710117, Shaanxi, P.R. China
| | - Huanhuan Zhang
- Chinese Medicine Pharmacy, Xi'an International Medical Center Hospital, Xi'an, 710117, Shaanxi, P.R. China
| | - Qiuhong Zhao
- Department of Pharmacy, Xi'an International Medical Center Hospital, Xi'an, 710117, Shaanxi, P.R. China
| | - Lei Shang
- Department of Traditional Chinese Medicine, Xi'an International Medical Center Hospital, No. 777, Xitai Road, High-tech Zone, Xi'an, 710117, Shaanxi, P.R. China.
| |
Collapse
|
2
|
Liu S, Zhao L, Peng Y, Liu X, Yan W, Zhang L, Zhang J. Obesity induced caveolin-1 impairs osteogenesis via activating mitophagy and inhibiting Sirt1 signaling. Bone 2024; 186:117146. [PMID: 38844017 DOI: 10.1016/j.bone.2024.117146] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/24/2023] [Revised: 06/01/2024] [Accepted: 06/03/2024] [Indexed: 07/08/2024]
Abstract
Obesity has become a major global health problem and the effect on bone formation has received increasing attention. However, the interaction between obesity and bone metabolism is complex and still not fully understood. Here, we show that caveolin-1 (Cav1), a membrane scaffold protein involved in regulating a variety of cellular processes, plays a key regulatory role as a bridge connecting obesity and bone metabolism. High-fat diet (HFD)-induced obese C57BL/6J mouse displayed a significant increase in Cav1 expression and lower osteogenic activity; In vitro treatment of osteoblastic MC3T3-E1 cells with 1 mM free fatty acids (FFA) significantly promoted Cav1 expression and PINK1/Parkin regulated mitophagy, but inhibited the expression of osteogenic marker genes. Conversely, reduced expression of the Cav1 gene prevented these effects. Both endogenous oxidative stress and Sirt1 pathway were also significantly reduced after Cav1 knockdown in FFA-treated cells. Finally, Cav1-Sirt1 docking and co-immunoprecipitation results showed that Cav1 interacted with Sirt1 and FFA enhanced the interaction. Taken together, these results suggest that obesity impairs bone development and formation through up-regulation of the Cav1 gene, which lead to inhibition of Sirt1/FOXO1 and Sirt1/PGC-1α signaling pathways through interacting with Sirt1 molecule, and an increase of mitophagy level.
Collapse
Affiliation(s)
- Shuai Liu
- Bioengineering College, Zhuhai campus of Zunyi Medical University, Zhuhai, Guangdong, China
| | - Lixia Zhao
- Bioengineering College, Zhuhai campus of Zunyi Medical University, Zhuhai, Guangdong, China
| | - Yanqiu Peng
- Bioengineering College, Zhuhai campus of Zunyi Medical University, Zhuhai, Guangdong, China
| | - Xing Liu
- Bioengineering College, Zhuhai campus of Zunyi Medical University, Zhuhai, Guangdong, China
| | - Wenmin Yan
- Bioengineering College, Zhuhai campus of Zunyi Medical University, Zhuhai, Guangdong, China
| | - Lizi Zhang
- Bioengineering College, Zhuhai campus of Zunyi Medical University, Zhuhai, Guangdong, China
| | - Jian Zhang
- Bioengineering College, Zhuhai campus of Zunyi Medical University, Zhuhai, Guangdong, China.
| |
Collapse
|
3
|
Bakhshian Nik A, Kaiser K, Sun P, Khomtchouk BB, Hutcheson JD. Altered Caveolin-1 Dynamics Result in Divergent Mineralization Responses in Bone and Vascular Calcification. Cell Mol Bioeng 2023; 16:299-308. [PMID: 37811003 PMCID: PMC10550882 DOI: 10.1007/s12195-023-00779-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Accepted: 08/08/2023] [Indexed: 10/10/2023] Open
Abstract
Introduction Though vascular smooth muscle cells adopt an osteogenic phenotype during pathological vascular calcification, clinical studies note an inverse correlation between bone mineral density and arterial mineral-also known as the calcification paradox. Both processes are mediated by extracellular vesicles (EVs) that sequester calcium and phosphate. Calcifying EV formation in the vasculature requires caveolin-1 (CAV1), a membrane scaffolding protein that resides in membrane invaginations (caveolae). Of note, caveolin-1-deficient mice, however, have increased bone mineral density. We hypothesized that caveolin-1 may play divergent roles in calcifying EV formation from vascular smooth muscle cells (VSMCs) and osteoblasts (HOBs). Methods Primary human coronary artery VSMCs and osteoblasts were cultured for up to 28 days in an osteogenic media. CAV1 expression was knocked down using siRNA. Methyl β-cyclodextrin (MβCD) and a calpain inhibitor were used, respectively, to disrupt and stabilize the caveolar domains in VSMCs and HOBs. Results CAV1 genetic variation demonstrates significant inverse relationships between bone-mineral density (BMD) and coronary artery calcification (CAC) across two independent epidemiological cohorts. Culture in osteogenic (OS) media increased calcification in HOBs and VSMCs. siRNA knockdown of CAV1 abrogated VSMC calcification with no effect on osteoblast mineralization. MβCD-mediated caveolae disruption led to a 3-fold increase of calcification in VSMCs treated with osteogenic media (p < 0.05) but hindered osteoblast mineralization (p < 0.01). Conversely, stabilizing caveolae by calpain inhibition prevented VSMC calcification (p < 0.05) without affecting osteoblast mineralization. There was no significant difference in CAV1 content between lipid domains from HOBs cultured in OS and control media. Conclusion Our data indicate fundamental cellular-level differences in physiological and pathophysiological mineralization mediated by CAV1 dynamics. This is the first study to suggest that divergent mechanisms in calcifying EV formation may play a role in the calcification paradox. Supplementary Information The online version contains supplementary material available at 10.1007/s12195-023-00779-7.
Collapse
Affiliation(s)
- Amirala Bakhshian Nik
- Department of Biomedical Engineering, Florida International University, 10555 W Flagler St, EC 2612, Miami, FL 33174 USA
| | - Katherine Kaiser
- Department of Biomedical Engineering, Florida International University, 10555 W Flagler St, EC 2612, Miami, FL 33174 USA
| | - Patrick Sun
- Department of BioHealth Informatics, Luddy School of Informatics, Computing, and Engineering, Indiana University, 535 W Michigan St, IT 477, Indianapolis, IN 46202 USA
| | - Bohdan B. Khomtchouk
- Department of BioHealth Informatics, Luddy School of Informatics, Computing, and Engineering, Indiana University, 535 W Michigan St, IT 477, Indianapolis, IN 46202 USA
- Krannert Cardiovascular Research Center, Indiana University School of Medicine, Indianapolis, IN USA
- Center for Computational Biology & Bioinformatics, Indiana University School of Medicine, Indianapolis, IN USA
- Center for Diabetes and Metabolic Diseases, Indiana University School of Medicine, Indianapolis, IN USA
| | - Joshua D. Hutcheson
- Department of Biomedical Engineering, Florida International University, 10555 W Flagler St, EC 2612, Miami, FL 33174 USA
- Biomolecular Sciences Institute, Florida International University, Miami, FL USA
| |
Collapse
|
4
|
Chen J, Wen J, Fu Y, Li X, Huang J, Guan X, Zhou Y. A bifunctional bortezomib-loaded porous nano-hydroxyapatite/alginate scaffold for simultaneous tumor inhibition and bone regeneration. J Nanobiotechnology 2023; 21:174. [PMID: 37264410 DOI: 10.1186/s12951-023-01940-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Accepted: 05/26/2023] [Indexed: 06/03/2023] Open
Abstract
Treatments of osteolytic lesions due to malignant metastasis remain one of the major clinical challenges. The residual tumor cells after surgical resections and an acidic tumor microenvironment are unfavorable for osteogenic induction. Bortezomib (BTZ), a proteasome inhibitor used in chemotherapy, also has an osteogenic potential in concentration- and Ca2+-dependent manners. In this study, controlled delivery of BTZ in a novel bifunctional scaffold based on nano-hydroxyapatite (nHA) and sodium alginate (SA) nanocomposite, namely BTZ/nHA@SA, has been explored. By smartly adjusting microenvironments, a sustainable release of Ca2+ from nHA could be achieved, which was not only able to cross-link SA but also to regulate the switch between the dual functions of tumor inhibition and bone regeneration of BTZ to promote the osteogenic pathway. The freeze-dried BTZ/nHA@SA scaffold has excellent interconnectivity, is capable to promote the attachment and proliferation of mouse embryonic osteoblast precursor cells, as well as effectively induces breast cancer cell death in vitro. Furthermore, in vivo, studies using a mouse tumor model and a rabbit femoral defect model showed that the BTZ/nHA@SA scaffold could promote tumor ablation, and also enhance bone repair. Therefore, the BTZ/nHA@SA scaffold has unique dual functions of inhibiting tumor recurrence and promoting bone tissue regeneration simultaneously. This smart bi-functional scaffold offers a promising novel approach for oncological treatments by synchronously orchestrating tumor inhibition and tissue regeneration for the repair of neoplastic bone defects.
Collapse
Affiliation(s)
- Jiafei Chen
- The Affiliated Hospital of Stomatology, School of Stomatology, Zhejiang University of Medicine, and Key Laboratory of Oral Biomedical Research of Zhejiang Province, Hangzhou, 310006, Zhejiang, China
| | - Junru Wen
- The Affiliated Hospital of Stomatology, School of Stomatology, Zhejiang University of Medicine, and Key Laboratory of Oral Biomedical Research of Zhejiang Province, Hangzhou, 310006, Zhejiang, China
| | - Yike Fu
- State Key Laboratory of Silicon Materials, School of Materials Science and Engineering, Zhejiang University, Hangzhou, 310027, P.R. China
- ZJU-Hangzhou Global Scientific and Technological Innovation Center, Zhejiang University, Hangzhou, 311200, P.R. China
| | - Xiang Li
- State Key Laboratory of Silicon Materials, School of Materials Science and Engineering, Zhejiang University, Hangzhou, 310027, P.R. China.
- ZJU-Hangzhou Global Scientific and Technological Innovation Center, Zhejiang University, Hangzhou, 311200, P.R. China.
| | - Jie Huang
- Department of Mechanical Engineering, University College London, London, WC1E 7JE, UK
| | - Xiaoxu Guan
- The Affiliated Hospital of Stomatology, School of Stomatology, Zhejiang University of Medicine, and Key Laboratory of Oral Biomedical Research of Zhejiang Province, Hangzhou, 310006, Zhejiang, China.
| | - Yi Zhou
- The Affiliated Hospital of Stomatology, School of Stomatology, Zhejiang University of Medicine, and Key Laboratory of Oral Biomedical Research of Zhejiang Province, Hangzhou, 310006, Zhejiang, China.
| |
Collapse
|
5
|
Bakhshian Nik A, Ng HH, Ashbrook SK, Sun P, Iacoviello F, Shearing PR, Bertazzo S, Mero D, Khomtchouk BB, Hutcheson JD. Epidermal growth factor receptor inhibition prevents vascular calcifying extracellular vesicle biogenesis. Am J Physiol Heart Circ Physiol 2023; 324:H553-H570. [PMID: 36827229 PMCID: PMC10042607 DOI: 10.1152/ajpheart.00280.2022] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Revised: 02/02/2023] [Accepted: 02/17/2023] [Indexed: 02/25/2023]
Abstract
Chronic kidney disease (CKD) increases the risk of cardiovascular disease, including vascular calcification, leading to higher mortality. The release of calcifying extracellular vesicles (EVs) by vascular smooth muscle cells (VSMCs) promotes ectopic mineralization of vessel walls. Caveolin-1 (CAV1), a structural protein in the plasma membrane, plays a major role in calcifying EV biogenesis in VSMCs. Epidermal growth factor receptor (EGFR) colocalizes with and influences the intracellular trafficking of CAV1. Using a diet-induced mouse model of CKD followed by a high-phosphate diet to promote vascular calcification, we assessed the potential of EGFR inhibition to prevent vascular calcification. Furthermore, we computationally analyzed 7,651 individuals in the Multi-Ethnic Study of Atherosclerosis (MESA) and Framingham cohorts to assess potential correlations between coronary artery calcium and single-nucleotide polymorphisms (SNPs) associated with elevated serum levels of EGFR. Mice with CKD developed widespread vascular calcification, associated with increased serum levels of EGFR. In both the CKD mice and human VSMC culture, EGFR inhibition significantly reduced vascular calcification by mitigating the release of CAV1-positive calcifying EVs. EGFR inhibition also increased bone mineral density in CKD mice. Individuals in the MESA and Framingham cohorts with SNPs associated with increased serum EGFR exhibit elevated coronary artery calcium. Given that EGFR inhibitors exhibit clinical safety and efficacy in other pathologies, the current data suggest that EGFR may represent an ideal target to prevent pathological vascular calcification in CKD.NEW & NOTEWORTHY Here, we investigate the potential of epidermal growth factor receptor (EGFR) inhibition to prevent vascular calcification, a leading indicator of and contributor to cardiovascular morbidity and mortality. EGFR interacts and affects the trafficking of the plasma membrane scaffolding protein caveolin-1. Previous studies reported a key role for caveolin-1 in the development of specialized extracellular vesicles that mediate vascular calcification; however, no role of EGFR has been reported. We demonstrated that EGFR inhibition modulates caveolin-1 trafficking and hinders calcifying extracellular vesicle formation, which prevents vascular calcification. Given that EGFR inhibitors are clinically approved for other indications, this may represent a novel therapeutic strategy for vascular calcification.
Collapse
Affiliation(s)
- Amirala Bakhshian Nik
- Department of Biomedical Engineering, Florida International University, Miami, Florida, United States
| | - Hooi Hooi Ng
- Department of Biomedical Engineering, Florida International University, Miami, Florida, United States
- Department of Human and Molecular Genetics, Herbert Wertheim College of Medicine, Florida International University, Miami, Florida, United States
| | - Sophie K Ashbrook
- Department of Biomedical Engineering, Florida International University, Miami, Florida, United States
| | - Patrick Sun
- Department of BioHealth Informatics, Luddy School of Informatics, Computing, and Engineering, Indiana University, Indianapolis, Indiana, United States
| | - Francesco Iacoviello
- Department of Chemical Engineering, University College London, London, United Kingdom
| | - Paul R Shearing
- Department of Chemical Engineering, University College London, London, United Kingdom
| | - Sergio Bertazzo
- Department of Medical Physics and Biomedical Engineering, University College London, London, United Kingdom
| | - Deniel Mero
- Dock Therapeutics, Inc., Middletown, Delaware, United States
| | - Bohdan B Khomtchouk
- Department of BioHealth Informatics, Luddy School of Informatics, Computing, and Engineering, Indiana University, Indianapolis, Indiana, United States
- Krannert Cardiovascular Research Center, Indiana University School of Medicine, Indianapolis, Indiana, United States
- Center for Computational Biology and Bioinformatics, Indiana University School of Medicine, Indianapolis, Indiana, United States
| | - Joshua D Hutcheson
- Department of Biomedical Engineering, Florida International University, Miami, Florida, United States
- Biomolecular Sciences Institute, Florida International University, Miami, Florida, United States
| |
Collapse
|
6
|
Human skeletal dysplasia causing L596P-mutant alters the conserved amino acid pattern at the lipid-water-Interface of TRPV4. BIOCHIMICA ET BIOPHYSICA ACTA. BIOMEMBRANES 2023; 1865:184085. [PMID: 36403799 DOI: 10.1016/j.bbamem.2022.184085] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/10/2022] [Revised: 11/01/2022] [Accepted: 11/02/2022] [Indexed: 11/18/2022]
Abstract
TRPV4 is a polymodal and non-selective cation channel that is activated by multiple physical and chemical stimuli. >50 naturally occurring point-mutation of TRPV4 have been identified in human, most of which induce different diseases commonly termed as channelopathies. While, these mutations are either "gain-of-function" or "loss-of-function" in nature, the exact molecular and cellular mechanisms behind such diverse channelopathies are largely unknown. In this work, we analyze the evolutionary conservation of individual amino acids present in the lipid-water-interface (LWI) regions and the relationship of TRPV4 with membrane cholesterol. Our data suggests that the positive-negative charges and hydrophobic-hydrophilic amino acids form "specific patterns" in the LWI region which remain conserved throughout the vertebrate evolution and thus suggesting for the specific microenvironment where TRPV4 remain functional. Notably, Spondylometaphyseal Dysplasia, Kozlowski (SMDK) disease causing L596P mutation disrupts this pattern significantly at the LWI region. L596P mutant also sequesters Caveolin-1 differently, especially in partial cholesterol-depleted (~40 % reduction) conditions. L596P shows altered localization in membrane and enhanced Ca2+-influx properties in cell as well as in filopodia-like structures. We propose that conserved pattern of amino acids is an important parameter for proper localization and functions of TRPV4 in physiological conditions. These findings also offer a new paradigm to analyze the channelopathies caused by mutations in LWI regions of other channels as well.
Collapse
|
7
|
Mehta SM, De Santos DR, Sridhar S, Aguayo VC, Meraz CA, Mikos M, Grande-Allen KJ. Fabricating a Low-Cost, Microscopy-Compatible Mechanical Testing Device. EXPERIMENTAL TECHNIQUES 2022; 46:731-743. [PMID: 39119455 PMCID: PMC11308025 DOI: 10.1007/s40799-021-00513-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Accepted: 09/16/2021] [Indexed: 08/10/2024]
Abstract
Most commercially-available mechanical testing devices are bulky, expensive, and unable to evaluate changes in sample microstructure under load. This leaves a crucial gap in understanding between material structure and bulk mechanical properties. Our objective was to fabricate a mechanical testing device small enough to fit in most upright or inverted microscopy stages and able to position samples to allow for simultaneous mechanical and microstructural characterization. Parts were 3D printed using the hobbyist-friendly Fused Filament Fabrication technique, then assembled with commercial fasteners and translation components to create a mechanical testing device that utilized the deflection of plastic posts to determine sample reaction forces under applied strain. Video of sample deformation was analyzed using a custom processing script to calculate stress and strain behavior in an automated and high-throughput manner. This device was able to perform mechanical characterization with an accuracy comparable to commercial mechanical testing devices for a wide range of nonlinear and viscoelastic samples under dry and hydrated conditions. Additionally, the device showed compatibility with different upright and inverted microscopes and was able to demonstrate accurate mechanical testing results when used with these instruments. We successfully developed a device capable of accurately testing a majority of soft materials in the field of Biomedical Engineering with the ability to perform additional microstructural characterization using microscopy at a price point of $600.
Collapse
Affiliation(s)
- S M Mehta
- Department of Bioengineering, Rice University, Houston, TX, 77005
| | - D R De Santos
- Department of Mechanical Engineering, University of Texas at El Paso, El Paso, TX 79968
| | - S Sridhar
- Department of Bioengineering, Rice University, Houston, TX, 77005
| | - V C Aguayo
- Department of Biomedical Engineering, Texas A&M, College Station, TX 77843
| | - C A Meraz
- Department of Mechanical Engineering, University of Texas at El Paso, El Paso, TX 79968
| | - M Mikos
- Department of Bioengineering, Rice University, Houston, TX, 77005
| | - K J Grande-Allen
- Department of Bioengineering, Rice University, Houston, TX, 77005
| |
Collapse
|
8
|
Wang L, Feng Y, Zhang C, Chen X, Huang H, Li W, Zhang J, Liu Y. Upregulation of OGT by Caveolin-1 promotes hepatocellular carcinoma cell migration and invasion. Cell Biol Int 2021; 45:2251-2263. [PMID: 34288245 DOI: 10.1002/cbin.11673] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2020] [Revised: 05/26/2021] [Accepted: 07/03/2021] [Indexed: 11/08/2022]
Abstract
Caveolin-1 (CAV1), a major structural protein of caveolae, is reported to exert a positive regulatory effect on tumor growth and to play a crucial role in hepatocellular carcinoma (HCC) cell metastasis by regulating glycosyltransferase expression and cellular glycosylation. However, the role of CAV1 in modulating protein glycosylation and tumor metastasis remains to be further elucidated. In the present study, we showed that CAV1 promoted the expression of O-GlcNAc transferase (OGT), which catalyzed the addition of O-GlcNAc residues to a variety of nuclear and cytoplasmic proteins. In human HCC cell lines with different metastatic potentials, high levels of OGT and cellular O-GlcNAcylation were associated with CAV1 expression and cell metastasis. Overexpression of CAV1 increased the levels of OGT and O-GlcNAcylation, and cell migration was also increased. Furthermore, CAV1 was found to reduce the expression of Runt-related transcription factor 2 (RUNX2) in HCC cells. Subsequently, this effect resulted in the attenuation of the RUNX2-induced transcription of microRNA24 (miR24), a microRNA previously shown to suppress OGT mRNA expression by targeting its 3' untranslated regions (UTR). Finally, we demonstrated that CAV1 induced cellular O-GlcNAcylation and HCC cell invasion. This study provides evidence of CAV1-mediated increases in OGT expression and O-GlcNAcylation. These data provide insight into a novel mechanism underlying HCC metastasis and suggest a novel strategy for the treatment of HCC.
Collapse
Affiliation(s)
- Lingyan Wang
- Department of Biochemistry, School of Life and Pharmaceutical Sciences, Dalian University of Technology, Panjin, China
| | - Yuan Feng
- Department of Biochemistry, School of Life and Pharmaceutical Sciences, Dalian University of Technology, Panjin, China
| | - Cheng Zhang
- Department of Biochemistry, School of Life and Pharmaceutical Sciences, Dalian University of Technology, Panjin, China
| | - Xixi Chen
- Department of Biochemistry, School of Life and Pharmaceutical Sciences, Dalian University of Technology, Panjin, China
| | - Huang Huang
- Department of Biochemistry, School of Life and Pharmaceutical Sciences, Dalian University of Technology, Panjin, China
| | - Wenli Li
- Department of Biochemistry, School of Life and Pharmaceutical Sciences, Dalian University of Technology, Panjin, China
| | - Jianing Zhang
- Department of Biochemistry, School of Life and Pharmaceutical Sciences, Dalian University of Technology, Panjin, China
| | - Yubo Liu
- Department of Biochemistry, School of Life and Pharmaceutical Sciences, Dalian University of Technology, Panjin, China
| |
Collapse
|
9
|
Li Z, MacDougald OA. Preclinical models for investigating how bone marrow adipocytes influence bone and hematopoietic cellularity. Best Pract Res Clin Endocrinol Metab 2021; 35:101547. [PMID: 34016532 PMCID: PMC8458229 DOI: 10.1016/j.beem.2021.101547] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Laboratory mice are a crucial preclinical model system for investigating bone marrow adipocyte (BMAd)-bone and BMAd-hematopoiesis interactions. In this review, we evaluate the suitability of mice to model common human diseases related to osteopenia or hematopoietic disorders, point out consistencies and discrepancies among different studies, and provide insights into model selection. Species, age, sex, skeletal site, and treatment protocol should all be considered when designing future studies.
Collapse
Affiliation(s)
- Ziru Li
- Department of Molecular & Integrative Physiology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Ormond A MacDougald
- Department of Molecular & Integrative Physiology, University of Michigan Medical School, Ann Arbor, MI, USA; Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI, USA.
| |
Collapse
|
10
|
Yemanyi F, Baidouri H, Burns AR, Raghunathan V. Dexamethasone and Glucocorticoid-Induced Matrix Temporally Modulate Key Integrins, Caveolins, Contractility, and Stiffness in Human Trabecular Meshwork Cells. Invest Ophthalmol Vis Sci 2021; 61:16. [PMID: 33170205 PMCID: PMC7686803 DOI: 10.1167/iovs.61.13.16] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Purpose To determine the temporal effects of dexamethasone (DEX) and glucocorticoid-induced matrix (GIM) on integrins/integrin adhesomes, caveolins, cytoskeletal-related proteins, and stiffness in human trabecular meshwork (hTM) cells. Methods Primary hTM cells were plated on plastic dishes (TCP), treated with vehicle (Veh) or 100 nM DEX in 1% serum media for 1, 3, 5, and 7 day(s). Concurrently, hTM cells were also plated on vehicle control matrices (VehMs) and GIMs for similar time points; VehMs and GIMs had been generated from chronic cultures of Veh-/DEX-stimulated hTM cells and characterized biochemically. Subsets of cells prior to plating on TCP or VehMs / GIMs served as baseline. Protein expression of mechanoreceptors, cytoskeletal-related proteins, and elastic moduli of hTM cells were determined. Results Compared with Veh, DEX temporally overexpressed αV, β3, and β5 integrins from day 3 to day 7, and integrin linked kinase at day 7, in hTM cells. However, DEX decreased β1 integrin at day 1 and day 7, while increasing Cavin1 at day 7, in a time-independent manner. Further, DEX temporally upregulated α-smooth muscle actin(α-SMA) and RhoA at day 7 and day 5, respectively; while temporally downregulating Cdc42 at day 3 and day 7 in hTM cells. Conversely, GIM showed increased immunostaining of fibronectin extra-domain A and B isoforms. Compared with VehM, GIM temporally increased αV integrin, Cavin1, and RhoA from day 3 to day 7, at day 3 and day 7, and at day 5, respectively, in hTM cells. Further, GIM overexpressed α-SMA at day 3 and day 7, and stiffened hTM cells from day 1 to day 7, in a time-independent fashion. Conclusions Our data highlight crucial mechanoreceptors, integrin adhesomes, and actin-related proteins that may temporally sustain fibrotic phenotypes precipitated by DEX and/or GIM in hTM cells.
Collapse
Affiliation(s)
- Felix Yemanyi
- Department of Basic Sciences, University of Houston College of Optometry, Houston, Texas, United States
| | - Hasna Baidouri
- Department of Basic Sciences, University of Houston College of Optometry, Houston, Texas, United States
| | - Alan R Burns
- Department of Basic Sciences, University of Houston College of Optometry, Houston, Texas, United States
| | - VijayKrishna Raghunathan
- Department of Basic Sciences, University of Houston College of Optometry, Houston, Texas, United States.,Department of Biomedical Engineering, Cullen College of Engineering, University of Houston, Houston, Texas, United States
| |
Collapse
|
11
|
Li Y, Liu G, Xiao F, Gu W, Gao Z, Wu Y, Wang P, Shi M, Yang M, Zhong Z, Liu B. Dual Role of Caveolin-1 in β-Catenin Signaling During Fracture Healing Induced by Low-Intensity Pulsed Ultrasound in Rabbits. J BIOMATER TISS ENG 2021. [DOI: 10.1166/jbt.2021.2553] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
We did this research to observe the effect of LIPUS on long bone fracture repair and caveolin-1, β-catenin signaling expression in the radius defects of rabbits, to explore its possible molecular mechanisms. 24 male New Zealand rabbits with bilateral radial bone defects
were divided into 4 groups randomly, n = 6. The right side had daily LIPUS exposure for 20 minutes, while the left received sham treatment. After 7, 14, 21, 28 days, respectively, fracture healing was observed by X-ray imaging and Dual Energy X-ray Absorptiometry (DXA) scan, specimens
were harvested for histology, immunohistochemistry, and gene expression analysis. We found that LIPUS brought forward endochondral ossification, increased the bone callus size without changes in Bone Mineral Density (BMD). The caveolin-1 expression increased first then decreased, while the
β-catenin kept growing during the process. These demonstrated that caveolin-1 participated in fracture healing accelerated by LIPUS, which was speculated to play a dual role in β-catenin signaling expression.
Collapse
Affiliation(s)
- Yun Li
- Department of Rehabilitation Medicine, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Guanghua Liu
- Department of Rehabilitation Medicine, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Feng Xiao
- Department of Pathology, Shanghai Seventh People’s Hospital, Shanghai 200137, China
| | - Wenqin Gu
- Department of Rehabilitation Medicine, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Zhengdong Gao
- Department of Radiology, Fenglin Community Health Service Center, Xuhui District, Shanghai 200030, China
| | - Yiming Wu
- Department of Rehabilitation Medicine, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Ping Wang
- Department of Rehabilitation Medicine, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Mingfang Shi
- Department of Rehabilitation Medicine, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Mingzhen Yang
- Department of Rehabilitation Medicine, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Zongye Zhong
- Department of Rehabilitation Medicine, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Bangzhong Liu
- Department of Rehabilitation Medicine, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| |
Collapse
|
12
|
Lolo FN, Jiménez-Jiménez V, Sánchez-Álvarez M, Del Pozo MÁ. Tumor-stroma biomechanical crosstalk: a perspective on the role of caveolin-1 in tumor progression. Cancer Metastasis Rev 2021; 39:485-503. [PMID: 32514892 DOI: 10.1007/s10555-020-09900-y] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Tumor stiffening is a hallmark of malignancy that actively drives tumor progression and aggressiveness. Recent research has shed light onto several molecular underpinnings of this biomechanical process, which has a reciprocal crosstalk between tumor cells, stromal fibroblasts, and extracellular matrix remodeling at its core. This dynamic communication shapes the tumor microenvironment; significantly determines disease features including therapeutic resistance, relapse, or metastasis; and potentially holds the key for novel antitumor strategies. Caveolae and their components emerge as integrators of different aspects of cell function, mechanotransduction, and ECM-cell interaction. Here, we review our current knowledge on the several pivotal roles of the essential caveolar component caveolin-1 in this multidirectional biomechanical crosstalk and highlight standing questions in the field.
Collapse
Affiliation(s)
- Fidel Nicolás Lolo
- Mechanoadaptation and Caveolae Biology Lab, Cell and Developmental Biology Area, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | - Víctor Jiménez-Jiménez
- Mechanoadaptation and Caveolae Biology Lab, Cell and Developmental Biology Area, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | - Miguel Sánchez-Álvarez
- Mechanoadaptation and Caveolae Biology Lab, Cell and Developmental Biology Area, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | - Miguel Ángel Del Pozo
- Mechanoadaptation and Caveolae Biology Lab, Cell and Developmental Biology Area, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain.
| |
Collapse
|
13
|
Yemanyi F, Vranka J, Raghunathan VK. Crosslinked Extracellular Matrix Stiffens Human Trabecular Meshwork Cells Via Dysregulating β-catenin and YAP/TAZ Signaling Pathways. Invest Ophthalmol Vis Sci 2021; 61:41. [PMID: 32832971 PMCID: PMC7452853 DOI: 10.1167/iovs.61.10.41] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Purpose The purpose of this study was to determine whether genipin-induced crosslinked cell-derived matrix (XCDM) precipitates fibrotic phenotypes in human trabecular meshwork (hTM) cells by dysregulating β-catenin and Yes-associated protein (YAP)/ transcriptional coactivator with PDZ-binding motif (TAZ) signaling pathways. Methods Cell-derived matrices were treated with control or genipin for 5 hours to obtain respective uncrosslinked (CDM) and XCDMs and characterized. hTM cells were seeded on these matrices with/without Wnt pathway modulators in serum-free media for 24 hours. Elastic modulus, gene, and protein (whole cell and subcellular fractions) expressions of signaling mediators and targets of Wnt/β-catenin and YAP/TAZ pathways were determined. Results At the highest genipin concentration (10% XCDM), XCDM had increased immunostaining of N-ε(γ-glutamyl)-lysine crosslinks, appeared morphologically fused, and was stiffer (5.3-fold, P < 0.001). On 10% XCDM, hTM cells were 7.8-fold (P < 0.001) stiffer, total β-catenin was unchanged, pβ-catenin was elevated, and pGSK3β was suppressed. Although 10% XCDM had no effect on cytoplasmic β-catenin levels, it reduced nuclear β-catenin, cadherin 11, and key Wnt target genes/proteins. The 10% XCDM increased total TAZ, decreased pTAZ, and increased cytoplasmic TAZ levels in hTM cells. The 10% XCDM increased total YAP, reduced nuclear YAP levels, and critical YAP/TAZ target genes/proteins. Wnt activation rescued hTM cells from 10% XCDM-induced stiffening associated with increased nuclear β-catenin. Conclusions Increased cytoplasmic TAZ may inhibit β-catenin from its nuclear shuttling or regulating cadherin 11 important for aqueous homeostasis. Elevated cytoplasmic TAZ may inhibit YAP's probable homeostatic function in the nucleus. Together, TAZ's cytoplasmic localization may be an important downstream event of how increased TM extracellular matrix (ECM) crosslinking may cause increased stiffness and ocular hypertension in vivo. However, Wnt pathway activation may ameliorate ocular hypertensive phenotypes induced by crosslinked ECM.
Collapse
Affiliation(s)
- Felix Yemanyi
- Department of Basic Sciences, College of Optometry, University of Houston, Houston, TX, United States
| | - Janice Vranka
- Casey Eye Institute, Oregon Health and Science University, Portland, OR, United States
| | - Vijay Krishna Raghunathan
- Department of Basic Sciences, College of Optometry, University of Houston, Houston, TX, United States.,Department of Biomedical Engineering, Cullen College of Engineering, University of Houston, Houston, TX, United States
| |
Collapse
|
14
|
McGrath C, Little-Letsinger SE, Sankaran JS, Sen B, Xie Z, Styner MA, Zong X, Chen W, Rubin J, Klett EL, Coleman RA, Styner M. Exercise Increases Bone in SEIPIN Deficient Lipodystrophy, Despite Low Marrow Adiposity. Front Endocrinol (Lausanne) 2021; 12:782194. [PMID: 35145475 PMCID: PMC8822583 DOI: 10.3389/fendo.2021.782194] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Accepted: 12/20/2021] [Indexed: 01/12/2023] Open
Abstract
Exercise, typically beneficial for skeletal health, has not yet been studied in lipodystrophy, a condition characterized by paucity of white adipose tissue, with eventual diabetes, and steatosis. We applied a mouse model of global deficiency of Bscl2 (SEIPIN), required for lipid droplet formation. Male twelve-week-old B6 knockouts (KO) and wild type (WT) littermates were assigned six-weeks of voluntary, running exercise (E) versus non-exercise (N=5-8). KO weighed 14% less than WT (p=0.01) and exhibited an absence of epididymal adipose tissue; KO liver Plin1 via qPCR was 9-fold that of WT (p=0.04), consistent with steatosis. Bone marrow adipose tissue (BMAT), unlike white adipose, was measurable, although 40.5% lower in KO vs WT (p=0.0003) via 9.4T MRI/advanced image analysis. SEIPIN ablation's most notable effect marrow adiposity was in the proximal femoral diaphysis (-56% KO vs WT, p=0.005), with relative preservation in KO-distal-femur. Bone via μCT was preserved in SEIPIN KO, though some quality parameters were attenuated. Running distance, speed, and time were comparable in KO and WT. Exercise reduced weight (-24% WT-E vs WT p<0.001) but not in KO. Notably, exercise increased trabecular BV/TV in both (+31%, KO-E vs KO, p=0.004; +14%, WT-E vs WT, p=0.006). The presence and distribution of BMAT in SEIPIN KO, though lower than WT, is unexpected and points to a uniqueness of this depot. That trabecular bone increases were achievable in both KO and WT, despite a difference in BMAT quantity/distribution, points to potential metabolic flexibility during exercise-induced skeletal anabolism.
Collapse
Affiliation(s)
- Cody McGrath
- Department of Medicine, Division of Endocrinology & Metabolism, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Sarah E. Little-Letsinger
- Department of Medicine, Division of Endocrinology & Metabolism, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Jeyantt Srinivas Sankaran
- Department of Medicine, Division of Endocrinology & Metabolism, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Buer Sen
- Department of Medicine, Division of Endocrinology & Metabolism, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Zhihui Xie
- Department of Medicine, Division of Endocrinology & Metabolism, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Martin A. Styner
- Department of Psychiatry, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
- Department of Computer Science, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Xiaopeng Zong
- Biomedical Research Imaging Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Weiqin Chen
- Department of Physiology, Medical College of Georgia at Augusta University, Augusta, GA, United States
| | - Janet Rubin
- Department of Medicine, Division of Endocrinology & Metabolism, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
- North Carolina Diabetes Research Center (NCDRC), Chapel Hill, NC, United States
| | - Eric L. Klett
- Department of Medicine, Division of Endocrinology & Metabolism, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
- North Carolina Diabetes Research Center (NCDRC), Chapel Hill, NC, United States
- Department of Nutrition, Gillings School of Global Public Health, UNC, Chapel Hill, NC, United States
| | - Rosalind A. Coleman
- Department of Nutrition, Gillings School of Global Public Health, UNC, Chapel Hill, NC, United States
| | - Maya Styner
- Department of Medicine, Division of Endocrinology & Metabolism, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
- North Carolina Diabetes Research Center (NCDRC), Chapel Hill, NC, United States
- *Correspondence: Maya Styner,
| |
Collapse
|
15
|
Zhou YM, Yang YY, Jing YX, Yuan TJ, Sun LH, Tao B, Liu JM, Zhao HY. BMP9 Reduces Bone Loss in Ovariectomized Mice by Dual Regulation of Bone Remodeling. J Bone Miner Res 2020; 35:978-993. [PMID: 31914211 DOI: 10.1002/jbmr.3957] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/22/2019] [Revised: 12/22/2019] [Accepted: 12/28/2019] [Indexed: 12/18/2022]
Abstract
Bone remodeling is dynamic and is tightly regulated through bone resorption dominated by osteoclasts and bone formation dominated by osteoblasts. Imbalances in this process can cause various pathological conditions, such as osteoporosis. Bone morphogenetic protein 9 (BMP9), a biomolecule produced and secreted by the liver, has many pharmacological effects, including anti-liver fibrosis, antitumor, anti-heart failure, and antidiabetic activities. However, the effects of BMP9 on the regulation of osteoblast and osteoclast functions and the underlying molecular mechanism(s) have not yet been investigated. In this study, BMP9 increased the expression of osteoblastogenic gene markers, such as ALP, Cola1, OCN, RUNX2, and OSX, and ALP activity in MC3T3-E1 cells by upregulating LGR6 and activating the Wnt/β-catenin pathway. BMP9 also suppressed receptor activator of nuclear factor-κB (NF-κB) ligand (RANKL)-induced osteoclast differentiation of bone marrow macrophages (BMMs) by inhibiting the Akt-NF-κB-NFATc1 pathway. More importantly, in an ovariectomy (OVX) mouse model, BMP9 attenuated bone loss and improved bone biomechanical properties in vivo by increasing bone-forming activity and suppressing bone resorption activity. Accordingly, our current work highlights the dual regulatory effects that BMP9 exerts on bone remodeling by promoting bone anabolic activity and inhibiting osteoclast differentiation in OVX mice. © 2020 American Society for Bone and Mineral Research.
Collapse
Affiliation(s)
- Yan-Man Zhou
- Department of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Institute of Endocrine and Metabolic Diseases, Shanghai Clinical Center for Endocrine and Metabolic Diseases, Shanghai, China
| | - Yu-Ying Yang
- Department of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Institute of Endocrine and Metabolic Diseases, Shanghai Clinical Center for Endocrine and Metabolic Diseases, Shanghai, China
| | - Yi-Xuan Jing
- Department of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Institute of Endocrine and Metabolic Diseases, Shanghai Clinical Center for Endocrine and Metabolic Diseases, Shanghai, China
| | - Tian-Jiao Yuan
- Department of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Institute of Endocrine and Metabolic Diseases, Shanghai Clinical Center for Endocrine and Metabolic Diseases, Shanghai, China
| | - Li-Hao Sun
- Department of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Institute of Endocrine and Metabolic Diseases, Shanghai Clinical Center for Endocrine and Metabolic Diseases, Shanghai, China
| | - Bei Tao
- Department of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Institute of Endocrine and Metabolic Diseases, Shanghai Clinical Center for Endocrine and Metabolic Diseases, Shanghai, China
| | - Jian-Min Liu
- Department of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Institute of Endocrine and Metabolic Diseases, Shanghai Clinical Center for Endocrine and Metabolic Diseases, Shanghai, China
| | - Hong-Yan Zhao
- Department of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Institute of Endocrine and Metabolic Diseases, Shanghai Clinical Center for Endocrine and Metabolic Diseases, Shanghai, China
| |
Collapse
|
16
|
Liang LL, Su ZB. In vitro effect of caveolin-1 as a slow-release material on bone-tendon junction healing: A comparative study. Kaohsiung J Med Sci 2019; 35:175-182. [PMID: 30887723 DOI: 10.1002/kjm2.12022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2017] [Accepted: 02/11/2019] [Indexed: 11/07/2022] Open
Abstract
Bone tendon junction injury is hard to cure because of its special anatomical structure, and the treatment applied for bone-tendon junction injury cannot result in the perfect vascular regeneration and restoration of the fibrocartilage zone. In this article, we aim to explore the effect of caveolin-1 as a slow-release material on bone-tendon junction healing. Seventy-two New Zealand rabbits were randomly selected and assigned into the experimental, sham-operated and control groups (n = 24). Caveolin-1 microspheres and microcapsule were developed as drug delivery system. At the 4th, 8th, and 12th weeks after surgery, quadriceps muscle patella-patellar tendon (QMPPT) was obtained from each rabbit to observe the tendon-to-bone tunnel healing, and X-ray examination, histological examination and biomechanical testing were applied for evaluating new bone formation. As the X-ray showed, caveolin-1 increased the new bone area at each time point. At the 4th and 8th weeks after surgery, the rabbit treated with caveolin-1 slow release material showed repair of fibrocartilage. According to the biomechanical results, the cross-sectional area, breaking load and ultimate tensile strength were increased along with time. At the same time point, caveolin-1 increased the ultimate tensile strength. Our study demonstrates that caveolin-1 as a slow-release material could accelerate bone-tendon junction healing by promoting the formation of the transition zone.
Collapse
Affiliation(s)
- Lin-Lin Liang
- Department of Clinical Laboratory, The Second People's Hospital in Jiulongpo District Chongqing, Chongqing, China
| | - Zheng-Bing Su
- Department of Orthopedics, The Second Affiliated Hospital of Army Medical University, Chongqing, China
| |
Collapse
|
17
|
Sarem M, Otto O, Tanaka S, Shastri VP. Cell number in mesenchymal stem cell aggregates dictates cell stiffness and chondrogenesis. Stem Cell Res Ther 2019; 10:10. [PMID: 30630531 PMCID: PMC6329065 DOI: 10.1186/s13287-018-1103-y] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2018] [Revised: 12/03/2018] [Accepted: 12/09/2018] [Indexed: 01/09/2023] Open
Abstract
Background Although mesenchymal stem/stromal cell (MSC) chondrogenic differentiation has been thoroughly investigated, the rudiments for enhancing chondrogenesis have remained largely dependent on external cues. Focus to date has been on extrinsic variables such as soluble signals, culture conditions (bioreactors), and mechanical stimulation. However, the role of intrinsic mechanisms of MSC programming-based mechanobiology remains to be explored. Since aggregation of MSCs, a prerequisite for chondrogenesis, generates tension within the cell agglomerate, we inquired if the initial number of cells forming the aggregate (aggregate cell number (ACN)) can impact chondrogenesis. Methods Aggregates of varying ACN were formed using well-established centrifugation approach. Progression of chondrogenic differentiation in the aggregates was assessed over 3 weeks in presence and absence of transforming growth factor-beta 1 (TGF-β1). Mechanical properties of the cells were characterized using high-throughput real-time deformability cytometry (RT-DC), and gene expression was analyzed using Affymetrix gene array. Expression of molecular markers linked to chondrogenesis was assessed using western blot and immunofluorescence. Results Reducing ACN from 500 k to 70 k lead to activation and acceleration of the chondrogenic differentiation, independent of soluble chondro-inductive factors, which involves changes to β-catenin-dependent TCF/LEF transcriptional activity and expression of anti-apoptotic protein survivin. RT-DC analysis revealed that stiffness and size of cells within aggregates are modulated by ACN. A direct correlation between progression of chondrogenesis and emergence of stiffer cell phenotype was found. Affymetrix gene array analysis revealed a downregulation of genes associated with lipid synthesis and regulation, which could account for observed changes in cell stiffness. Immunofluorescence and western blot analysis revealed that increasing ACN upregulates the expression of lipid raft protein caveolin-1, a β-catenin binding partner, and downregulates the expression of N-cadherin. As a demonstration of the relevance of these findings in MSC-based strategies for skeletal repair, it is shown that implanting aggregates within collagenous matrix not only decreases the necessity for high cell numbers but also leads to marked improvement in the quality of the deposited tissue. Conclusions This study presents a simple and donor-independent strategy to enhance the efficiency of MSC chondrogenic differentiation and identifies changes in cell mechanics coincident with MSC chondrogenesis with potential translational applications. Electronic supplementary material The online version of this article (10.1186/s13287-018-1103-y) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Melika Sarem
- Institute for Macromolecular Chemistry, University of Freiburg, Stefan-Meier Str.31, 79104, Freiburg, Germany.,BIOSS Centre for Biological Signaling Studies, University of Freiburg, 79104, Freiburg, Germany.,Helmholtz Virtual Institute on Multifunctional Biomaterials for Medicine, Kantstr. 55, 14513, Teltow, Germany
| | - Oliver Otto
- Centre for Innovation Competence - Humoral Immune Response in Cardiovascular Diseases, University of Greifswald, Fleischmannstr. 42-44, 17489, Greifswald, Germany
| | - Simon Tanaka
- Computational Biology Group, D-BSSE, ETH Zürich, Mattenstrasse 26, 4058, Basel, Switzerland
| | - V Prasad Shastri
- Institute for Macromolecular Chemistry, University of Freiburg, Stefan-Meier Str.31, 79104, Freiburg, Germany. .,BIOSS Centre for Biological Signaling Studies, University of Freiburg, 79104, Freiburg, Germany. .,Helmholtz Virtual Institute on Multifunctional Biomaterials for Medicine, Kantstr. 55, 14513, Teltow, Germany.
| |
Collapse
|
18
|
Yang R, Wang J, Zhou Z, Qi S, Ruan S, Lin Z, Xin Q, Lin Y, Chen X, Xie J. Role of caveolin-1 in epidermal stem cells during burn wound healing in rats. Dev Biol 2018; 445:271-279. [PMID: 30476483 DOI: 10.1016/j.ydbio.2018.11.015] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2018] [Revised: 11/22/2018] [Accepted: 11/22/2018] [Indexed: 12/24/2022]
Abstract
Local transplantation of stem cells has therapeutic effects on skin damage but cannot provide satisfactory wound healing. Studies on the mechanisms underlying the therapeutic effects of stem cells on skin wound healing will be needed. Hence, in the present study, we explored the role of Caveolin-1 in epidermal stem cells (EpiSCs) in the modulation of wound healing. We first isolated EpiSCs from mouse skin tissues and established stable EpiSCs with overexpression of Caveolin-1 using a lentiviral construct. We then evaluated the epidermal growth factor (EGF)-induced cell proliferation ability using cell counting Kit-8 (CCK-8) assay and assessed EpiSC pluripotency by examining Nanog mRNA levels in EpiSCs. Furthermore, we treated mice with skin burn injury using EpiSCs with overexpression of Caveolin-1. Histological examinations were conducted to evaluate re-epithelialization, wound scores, cell proliferation and capillary density in wounds. We found that overexpression of Caveolin-1 in EpiSCs promoted EGF-induced cell proliferation ability and increased wound closure in a mouse model of skin burn injury. Histological evaluation demonstrated that overexpression of Caveolin-1 in EpiSCs promoted re-epithelialization in wounds, enhanced cellularity, and increased vasculature, as well as increased wound scores. Taken together, our results suggested that Caveolin-1 expression in the EpiSCs play a critical role in the regulation of EpiSC proliferation ability and alteration of EpiSC proliferation ability may be an effective approach in promoting EpiSC-based therapy in skin wound healing.
Collapse
Affiliation(s)
- Ronghua Yang
- Department of Burn Surgery, the First People's Hospital of Foshan, Foshan 528000, Guangdong, China
| | - Jingru Wang
- Department of Burn Surgery, the First People's Hospital of Foshan, Foshan 528000, Guangdong, China
| | - Ziheng Zhou
- Department of Burn Surgery, First Affiliated Hospital of Sun Yat-Sen University, Guangzhou 510000, Guangdong, China
| | - Shaohai Qi
- Department of Burn Surgery, First Affiliated Hospital of Sun Yat-Sen University, Guangzhou 510000, Guangdong, China
| | - Shubin Ruan
- Department of Burn Surgery, the First People's Hospital of Foshan, Foshan 528000, Guangdong, China
| | - Zepeng Lin
- Department of Burn Surgery, the First People's Hospital of Foshan, Foshan 528000, Guangdong, China
| | - Qi Xin
- Department of Burn Surgery, the First People's Hospital of Foshan, Foshan 528000, Guangdong, China
| | - Yan Lin
- Department of Burn Surgery, the First People's Hospital of Foshan, Foshan 528000, Guangdong, China
| | - Xiaodong Chen
- Department of Burn Surgery, the First People's Hospital of Foshan, Foshan 528000, Guangdong, China.
| | - Julin Xie
- Department of Burn Surgery, First Affiliated Hospital of Sun Yat-Sen University, Guangzhou 510000, Guangdong, China.
| |
Collapse
|
19
|
Yang R, Wang J, Zhou Z, Qi S, Ruan S, Lin Z, Xin Q, Lin Y, Chen X, Xie J. Curcumin promotes burn wound healing in mice by upregulating caveolin-1 in epidermal stem cells. Phytother Res 2018; 33:422-430. [PMID: 30461085 DOI: 10.1002/ptr.6238] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2018] [Revised: 10/26/2018] [Accepted: 10/27/2018] [Indexed: 01/07/2023]
Abstract
We aimed to explore the effect of curcumin on epidermal stem cells (ESCs) in regulating wound healing and the underlying molecular mechanism. We treated mouse ESCs isolated from skin tissues with curcumin, and then assessed the proliferation ability of cells induced by epidermal growth factor using cell counting kit-8 assay. The pluripotency of ESCs was evaluated as well through examination of Nanog expression in ESCs. Further, mice with skin burns were treated with ESCs with or without curcumin pretreatments. Histological evaluations were then preformed to determine wound scores, cell proliferation, reepithelialization, and capillary density in wounds. Curcumin treatment promoted the proliferative ability of ESCs and conditioned medium from curcumin-treated ESCs enhanced human umbilical vein endothelial cell (HUVEC) tube formation. We also found curcumin treatment elevated caveolin-1 expression in ESCs, which was required for the beneficial effect of curcumin on ESC proliferation and HUVEC tube formation. Next, using a mouse model of burn wound healing, curcumin-treated ESCs exhibited enhanced wound closure, which also required caveolin-1 expression. Our current study demonstrates the beneficial effect of curcumin on burn wound healing in mice, which is mediated by upregulating caveolin-1 in ESCs, and supports the potential therapeutic role of curcumin in ESC-based treatment against skin wound healing.
Collapse
Affiliation(s)
- Ronghua Yang
- Department of Burn Surgery, the First People's Hospital of Foshan, Foshan, China
| | - Jingru Wang
- Department of Burn Surgery, the First People's Hospital of Foshan, Foshan, China
| | - Ziheng Zhou
- Department of Burn Surgery, First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Shaohai Qi
- Department of Burn Surgery, First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Shubin Ruan
- Department of Burn Surgery, the First People's Hospital of Foshan, Foshan, China
| | - Zepeng Lin
- Department of Burn Surgery, the First People's Hospital of Foshan, Foshan, China
| | - Qi Xin
- Department of Burn Surgery, the First People's Hospital of Foshan, Foshan, China
| | - Yan Lin
- Department of Burn Surgery, the First People's Hospital of Foshan, Foshan, China
| | - Xiaodong Chen
- Department of Burn Surgery, the First People's Hospital of Foshan, Foshan, China
| | - Julin Xie
- Department of Burn Surgery, First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| |
Collapse
|
20
|
Caveolin-1 Controls Hyperresponsiveness to Mechanical Stimuli and Fibrogenesis-Associated RUNX2 Activation in Keloid Fibroblasts. J Invest Dermatol 2017; 138:208-218. [PMID: 28899682 DOI: 10.1016/j.jid.2017.05.041] [Citation(s) in RCA: 66] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2016] [Revised: 05/20/2017] [Accepted: 05/30/2017] [Indexed: 11/22/2022]
Abstract
Keloids are pathological scars characterized by excessive extracellular matrix production that are prone to form in body sites with increased skin tension. CAV1, the principal coat protein of caveolae, has been associated with the regulation of cell mechanics, including cell softening and loss of stiffness sensing ability in NIH3T3 fibroblasts. Although CAV1 is present in low amounts in keloid fibroblasts (KFs), the causal association between CAV1 down-regulation and its aberrant responses to mechanical stimuli remain unclear. In this study, atomic force microscopy showed that KFs were softer than normal fibroblasts with a loss of stiffness sensing. The decrease of CAV1 contributed to the hyperactivation of fibrogenesis-associated RUNX2, a transcription factor germane to osteogenesis/chondrogenesis, and increased migratory ability in KFs. Treatment of KFs with trichostatin A, which increased the acetylation level of histone H3, increased CAV1 and decreased RUNX2 and fibronectin. Trichostatin A treatment also resulted in cell stiffening and decreased migratory ability in KFs. Collectively, these results suggest a role for CAV1 down-regulation in linking the aberrant responsiveness to mechanical stimulation and extracellular matrix accumulation with the progression of keloids, findings that may lead to new developments in the prevention and treatment of keloid scarring.
Collapse
|
21
|
Gene expression profiling of bone marrow mesenchymal stem cells from Osteogenesis Imperfecta patients during osteoblast differentiation. Eur J Med Genet 2017; 60:326-334. [DOI: 10.1016/j.ejmg.2017.04.003] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2016] [Revised: 03/22/2017] [Accepted: 04/05/2017] [Indexed: 12/26/2022]
|
22
|
Chen JR, Lazarenko OP, Blackburn ML, Shankar K. Dietary factors during early life program bone formation in female rats. FASEB J 2016; 31:376-387. [PMID: 27733448 DOI: 10.1096/fj.201600703r] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2016] [Accepted: 09/28/2016] [Indexed: 01/10/2023]
Abstract
Nutritional status during intrauterine and early postnatal life impacts the risk of chronic diseases; however, evidence for an association between early-life dietary factors and bone health in adults is limited. Soy protein isolate (SPI) may be one such dietary factor that promotes bone accretion during early life with persistent effects into adulthood. In the present study, we fed postnatal day (PND) 24 weanling female rats an SPI diet for 30 d [short-term SPI (ST-SPI)], and on PND 55, we switched SPI diet to control Cas diet until age 6 mo. Rats then underwent either ovariectomy (OVX) or sham surgery and thereafter either continued to be fed an SPI diet or control diet for 1 or 3 wk. We showed significantly increased bone mass in 30-d SPI-fed young rats compared with controls. OVX-induced bone loss was associated with increased osteoblastic cell senescence. On the one hand, both long-term SPI (continuous SPI diet throughout life) and ST-SPI diet only in early life protected against 1 wk post-OVX-associated bone loss. On the other hand, long-term SPI diet diminished the loss of total, trabecular, and cortical bone mineral density, whereas ST-SPI diet only reduced cortical bone mineral density loss 3 wk post-OVX. Persistent and protective effects of SPI diets on OVX-induced bone loss were associated with down-regulation of the caveolin-1/p53-mediated senescence pathway in bone. We recapitulated these results in cell cultures. Reprogramming of cellular senescence signaling by SPI-associated isoflavones in osteoblastic cells may explain the persistent effects of SPI on bone. These results suggest that OVX-induced bone loss, in part, is a result of increased osteoblastic cell senescence, and that ST-SPI diet early in life has modest but persistent programming effects on bone formation to prevent OVX-induced bone loss in adult female rats.-Chen, J.-R., Lazarenko, O. P., Blackburn, M. L., Shankar, K. Dietary factors during early life program bone formation in female rats.
Collapse
Affiliation(s)
- Jin-Ran Chen
- Arkansas Children's Nutrition Center, Little Rock, Arkansas, USA; and .,Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
| | - Oxana P Lazarenko
- Arkansas Children's Nutrition Center, Little Rock, Arkansas, USA; and.,Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
| | - Michael L Blackburn
- Arkansas Children's Nutrition Center, Little Rock, Arkansas, USA; and.,Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
| | - Kartik Shankar
- Arkansas Children's Nutrition Center, Little Rock, Arkansas, USA; and.,Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
| |
Collapse
|
23
|
Bai Y, Wu J, Li D, Morgan EE, Liu J, Zhao X, Walsh A, Saikumar J, Tinkel J, Joe B, Gupta R, Liu L. Differential roles of caveolin-1 in ouabain-induced Na+/K+-ATPase cardiac signaling and contractility. Physiol Genomics 2016; 48:739-748. [PMID: 27519543 PMCID: PMC5243228 DOI: 10.1152/physiolgenomics.00042.2016] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2016] [Accepted: 08/03/2016] [Indexed: 11/22/2022] Open
Abstract
Binding of ouabain to cardiac Na+/K+-ATPase initiates cell signaling and causes contractility in cardiomyocytes. It is widely accepted that caveolins, structural proteins of caveolae, have been implicated in signal transduction. It is known that caveolae play a role in Na+/K+-ATPase functions. Regulation of caveolin-1 in ouabain-mediated cardiac signaling and contractility has never been reported. The aim of this study is to compare ouabain-induced cardiac signaling and contractility in wild-type (WT) and caveolin-1 knockout (cav-1 KO) mice. In contrast with WT cardiomyocytes, ouabain-induced signaling e.g., activation of phosphoinositide 3-kinase-α/Akt and extracellular signal-regulated kinases (ERK)1/2, and hypertrophic growth were significantly reduced in cav-1 KO cardiomyocytes. Interactions of the Na+/K+-ATPase α1-subunit with caveolin-3 and the Na+/K+-ATPase α1-subunit with PI3K-α were also decreased in cav-1 KO cardiomyocytes. The results from cav-1 KO mouse embryonic fibroblasts also proved that cav-1 significantly attenuated ouabain-induced ERK1/2 activation without alteration in protein and cholesterol distribution in caveolae/lipid rafts. Intriguingly, the effect of ouabain induced positive inotropy in vivo (via transient infusion of ouabain, 0.48 nmol/g body wt) was not attenuated in cav-1 KO mice. Furthermore, ouabain (1-100 μM) induced dose-dependent contractility in isolated working hearts from WT and cav-1 KO mice. The effects of ouabain on contractility between WT and cav-1 KO mice were not significantly different. These results demonstrated differential roles of cav-1 in the regulation of ouabain signaling and contractility. Signaling by ouabain, in contrast to contractility, may be a redundant property of Na+/K+-ATPase.
Collapse
Affiliation(s)
- Yan Bai
- Department of Biochemistry and Cancer Biology, College of Medicine and Life Sciences, University of Toledo, Toledo, Ohio; Pediatrics Department of Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, HuBei, China
| | - Jian Wu
- Department of Biochemistry and Cancer Biology, College of Medicine and Life Sciences, University of Toledo, Toledo, Ohio
| | - Daxiang Li
- Department of Biochemistry and Cancer Biology, College of Medicine and Life Sciences, University of Toledo, Toledo, Ohio; State Key Laboratory of Tea Plant Biology and Utilization, Anhui Agricultural University, Hefei, Anhui, China; and
| | - Eric E Morgan
- Center for Hypertension and Personalized Medicine, College of Medicine and Life Sciences, University of Toledo, Toledo, Ohio
| | - Jiang Liu
- Department of Pharmacology, Physiology and Toxicology, JCE School of Medicine, Marshall University, Huntington, West Virginia
| | - Xiaochen Zhao
- Department of Physiology and Pharmacology, College of Medicine and Life Sciences, University of Toledo, Toledo, Ohio
| | - Aaron Walsh
- Department of Biochemistry and Cancer Biology, College of Medicine and Life Sciences, University of Toledo, Toledo, Ohio
| | - Jagannath Saikumar
- Department of Physiology and Pharmacology, College of Medicine and Life Sciences, University of Toledo, Toledo, Ohio
| | - Jodi Tinkel
- Department of Medicine, College of Medicine and Life Sciences, University of Toledo, Toledo, Ohio
| | - Bina Joe
- Department of Physiology and Pharmacology, College of Medicine and Life Sciences, University of Toledo, Toledo, Ohio; Center for Hypertension and Personalized Medicine, College of Medicine and Life Sciences, University of Toledo, Toledo, Ohio
| | - Rajesh Gupta
- Department of Medicine, College of Medicine and Life Sciences, University of Toledo, Toledo, Ohio
| | - Lijun Liu
- Department of Biochemistry and Cancer Biology, College of Medicine and Life Sciences, University of Toledo, Toledo, Ohio; Department of Medicine, College of Medicine and Life Sciences, University of Toledo, Toledo, Ohio; Center for Hypertension and Personalized Medicine, College of Medicine and Life Sciences, University of Toledo, Toledo, Ohio;
| |
Collapse
|
24
|
Chen Y, Wang C, Huang Q, Wu D, Cao J, Xu X, Yang C, Li X. Caveolin-1 Plays an Important Role in the Differentiation of Bone Marrow-Derived Mesenchymal Stem Cells into Cardiomyocytes. Cardiology 2016; 136:40-48. [PMID: 27554796 DOI: 10.1159/000446869] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/08/2016] [Accepted: 05/15/2016] [Indexed: 11/19/2022]
Abstract
OBJECTIVES Accumulating evidence has demonstrated that bone marrow-derived mesenchymal stem cells (BMSCs) may transdifferentiate into cardiomyocytes, making BMSCs a promising source of cardiomyocytes for transplantation. However, little is known about the molecular mechanisms underlying myogenic conversion of BMSCs. METHODS This study was designed to investigate the functional role of caveolin-1 in the cardiomyocyte differentiation of BMSCs and to explore the potential underlying molecular mechanisms. RESULTS BMSC differentiation was induced by treatment with 10 μM 5-azacytidine, and immunofluorescence assay showed that the expression of cardiomyocyte marker cardiac troponin T (cTnT) was significantly increased compared with a control group. Meanwhile, an increased caveolin-1 expression was found during the 5-azacytidine-induced BMSC differentiation. Additionally, the role of caveolin-1 in the differentiation process was then studied by using caveolin-1 siRNAs. We found that silencing caveolin-1 during induction remarkably enhanced the expression of cardiomyocyte marker genes, including cTnT, Nkx2.5 (cardiac-specific transcription factor), α-cardiac actin and α-myosin heavy chain (α-MHC). Moreover, we observed that downregulation of caveolin-1 was accompanied by inhibition of signal transducer and activator of transcription 3 (STAT3) phosphorylation. CONCLUSIONS Taken together, these findings demonstrate that caveolin-1 plays an important role in the differentiation of BMSCs into cardiomyocytes in conjunction with the STAT3 pathway.
Collapse
Affiliation(s)
- Ying Chen
- Department of Cardiology, Wuxi Second People's Hospital of Nanjing Medical University, Wuxi, China
| | | | | | | | | | | | | | | |
Collapse
|
25
|
Lee SY, Yi JK, Yun HM, Bae CH, Cho ES, Lee KS, Kim EC. Expression of Caveolin-1 in Periodontal Tissue and Its Role in Osteoblastic and Cementoblastic Differentiation In Vitro. Calcif Tissue Int 2016; 98:497-510. [PMID: 26686692 DOI: 10.1007/s00223-015-0095-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/02/2015] [Accepted: 11/25/2015] [Indexed: 01/13/2023]
Abstract
It has been previously reported that caveolin-1 (Cav-1) knockout mice exhibit increased bone size and stiffness. However, the expression and role of Cav-1 on periodontal tissue is poorly understood. The aim of this study was to investigate the immunohistochemical expression of Cav-1 in the mouse periodontium and explore the role of Cav-1 on osteoblastic and cementoblastic differentiation in human periodontal ligament cells (hPDLCs), cementoblasts, and osteoblasts. To reveal the molecular mechanisms of Cav-1 activity, associated signaling pathways were also examined. Immunolocalization of Cav-1 was studied in mice periodontal tissue. Differentiation was evaluated by ALP activity, alizarin red S staining, and RT-PCR for marker genes. Signal transduction was analyzed using Western blotting and confocal microscopy. Cav-1 expression was observed in hPDLCs, cementoblasts, and osteoblasts of the periodontium both in vivo and in vitro. Inhibition of Cav-1 expression by methyl-β-cyclodextrin (MβCD) and knockdown of Cav-1 by siRNA promoted osteoblastic and cementoblastic differentiation by increasing ALP activity, calcium nodule formation, and mRNA expression of differentiation markers in hPDLCs, cementoblasts, and osteoblasts. Osteogenic medium-induced BMP-2 and BMP-7 expression, and phosphorylation of Smad1/5/8 were enhanced by MβCD and siRNA knockdown of Cav-1, which was reversed by BMP inhibitor noggin. MβCD and Cav-1 siRNA knockdown increased OM-induced AMPK, Akt, GSK3β, and CREB phosphorylation, which were reversed by Ara-A, a specific AMPK inhibitor. Moreover, OM-induced activation of p38, ERK, JNK, and NF-κB was enhanced by Cav-1 inhibition. This study demonstrates, for the first time, that Cav-1 is expressed in developing periodontal tissue and in vitro in periodontal-related cells. Cav-1 inhibition positively regulates osteoblastic differentiation in hPDLCs, cementoblasts, and osteoblasts via BMP, AMPK, MAPK, and NF-κB pathway. Thus, Cav-1 inhibition may be a novel molecular target for therapeutic approaches in periodontitis or osteolytic disease.
Collapse
Affiliation(s)
- So-Youn Lee
- Department of Oral and Maxillofacial Pathology and Research Center for Tooth and Periodontal Regeneration (MRC), Kyung Hee University, 14 Kyungheedae-ro Dongdaemun-gu, Seoul, 02453, Republic of Korea
| | - Jin-Kyu Yi
- Department of Conservative Dentistry, Kyung Hee University, Seoul, Republic of Korea
| | - Hyung-Mun Yun
- Department of Oral and Maxillofacial Pathology and Research Center for Tooth and Periodontal Regeneration (MRC), Kyung Hee University, 14 Kyungheedae-ro Dongdaemun-gu, Seoul, 02453, Republic of Korea
| | - Cheol-Hyeon Bae
- Cluster for Craniofacial Development and Regeneration Research, Institute of Oral Biosciences, School of Dentistry, Chonbuk National University, Jeonju, Republic of Korea
| | - Eui-Sic Cho
- Cluster for Craniofacial Development and Regeneration Research, Institute of Oral Biosciences, School of Dentistry, Chonbuk National University, Jeonju, Republic of Korea
| | - Kook-Sun Lee
- Division of Dentistry, Department of Oral and Maxillofacial Radiology, Graduate School, Kyung Hee University, Seoul, Republic of Korea
| | - Eun-Cheol Kim
- Department of Oral and Maxillofacial Pathology and Research Center for Tooth and Periodontal Regeneration (MRC), Kyung Hee University, 14 Kyungheedae-ro Dongdaemun-gu, Seoul, 02453, Republic of Korea.
| |
Collapse
|
26
|
Zhu C, Zheng XF, Yang YH, Li B, Wang YR, Jiang SD, Jiang LS. LGR4 acts as a key receptor for R-spondin 2 to promote osteogenesis through Wnt signaling pathway. Cell Signal 2016; 28:989-1000. [PMID: 27140682 DOI: 10.1016/j.cellsig.2016.04.010] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2015] [Revised: 04/01/2016] [Accepted: 04/27/2016] [Indexed: 02/01/2023]
Abstract
R-spondin proteins are identified as secreted agonists of the canonical Wnt/β-catenin signaling pathway, and leucine-rich repeat-containing G-protein-coupled receptors (LGR) are recognized as R-spondin receptors. The potential role of R-spondin 2 (Rspo2) and LGR4 in mediating osteogenesis remains poorly understood. In our in vitro experiments, we found that Rspo2 could promote osteogenesis through activating the Wnt signaling pathway in MC3T3-E1 cells. However, this effect of Rsop2 disappeared in the cells with functional disruption of LGR4. Meanwhile, Rspo2 significantly inhibited osteoclastogenesis and this effect of Rspo2 was dependent on the presence of osteoblasts with normal function of LGR4. In our in vivo experiments, we found that application of exogenous Rspo2 rescued the bone loss and improved the microarchitecture of bone in OVX mice. Rspo2 could be a positive regulator of bone metabolism through activating the canonical Wnt/β-catenin signaling, and LGR4 acted as a key receptor for Rspo2 to promote osteogenesis.
Collapse
Affiliation(s)
- Chao Zhu
- Department of Orthopedic Surgery, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200092, China
| | - Xin-Feng Zheng
- Department of Orthopedic Surgery, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200092, China
| | - Yue-Hua Yang
- Department of Orthopedic Surgery, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200092, China
| | - Bo Li
- Department of Orthopedic Surgery, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200092, China
| | - Yu-Ren Wang
- Department of Orthopedic Surgery, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200092, China
| | - Sheng-Dan Jiang
- Department of Orthopedic Surgery, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200092, China.
| | - Lei-Sheng Jiang
- Department of Orthopedic Surgery, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200092, China.
| |
Collapse
|
27
|
Uzer G, Fuchs RK, Rubin J, Thompson WR. Concise Review: Plasma and Nuclear Membranes Convey Mechanical Information to Regulate Mesenchymal Stem Cell Lineage. Stem Cells 2016; 34:1455-63. [PMID: 26891206 DOI: 10.1002/stem.2342] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2015] [Revised: 12/14/2015] [Accepted: 12/29/2015] [Indexed: 12/21/2022]
Abstract
Numerous factors including chemical, hormonal, spatial, and physical cues determine stem cell fate. While the regulation of stem cell differentiation by soluble factors is well-characterized, the role of mechanical force in the determination of lineage fate is just beginning to be understood. Investigation of the role of force on cell function has largely focused on "outside-in" signaling, initiated at the plasma membrane. When interfaced with the extracellular matrix, the cell uses integral membrane proteins, such as those found in focal adhesion complexes to translate force into biochemical signals. Akin to these outside-in connections, the internal cytoskeleton is physically linked to the nucleus, via proteins that span the nuclear membrane. Although structurally and biochemically distinct, these two forms of mechanical coupling influence stem cell lineage fate and, when disrupted, often lead to disease. Here we provide an overview of how mechanical coupling occurs at the plasma and nuclear membranes. We also discuss the role of force on stem cell differentiation, with focus on the biochemical signals generated at the cell membrane and the nucleus, and how those signals influence various diseases. While the interaction of stem cells with their physical environment and how they respond to force is complex, an understanding of the mechanical regulation of these cells is critical in the design of novel therapeutics to combat diseases associated with aging, cancer, and osteoporosis. Stem Cells 2016;34:1455-1463.
Collapse
Affiliation(s)
- Gunes Uzer
- Department of Medicine, University of North Carolina, Chapel Hill, North Carolina, USA
| | - Robyn K Fuchs
- School of Health and Rehabilitation Sciences, Department of Physical Therapy, Indiana University, Indianapolis, Indiana, USA
| | - Janet Rubin
- Department of Medicine, University of North Carolina, Chapel Hill, North Carolina, USA
| | - William R Thompson
- School of Health and Rehabilitation Sciences, Department of Physical Therapy, Indiana University, Indianapolis, Indiana, USA
| |
Collapse
|
28
|
Baker N, Sohn J, Tuan RS. Promotion of human mesenchymal stem cell osteogenesis by PI3-kinase/Akt signaling, and the influence of caveolin-1/cholesterol homeostasis. Stem Cell Res Ther 2015; 6:238. [PMID: 26626726 PMCID: PMC4667507 DOI: 10.1186/s13287-015-0225-8] [Citation(s) in RCA: 81] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2015] [Revised: 01/08/2015] [Accepted: 11/05/2015] [Indexed: 11/21/2022] Open
Abstract
Introduction Stem cells are considered an important resource for tissue repair and regeneration. Their utilization in regenerative medicine will be aided by mechanistic insight into their responsiveness to external stimuli. It is likely that, similar to all other cells, an initial determinant of stem cell responsiveness to external stimuli is the organization of signaling molecules in cell membrane rafts. The clustering of signaling molecules in these cholesterol-rich membrane microdomains can affect the activity, specificity, cross-talk and amplification of cell signaling. Membrane rafts fall into two broad categories, non-caveolar and caveolar, based on the absence or presence, respectively, of caveolin scaffolding proteins. We have recently demonstrated that caveolin-1 (Cav-1) expression increases during, and knockdown of Cav-1 expression enhances, osteogenic differentiation of human bone marrow derived mesenchymal stem cells (MSCs). The increase in Cav-1 expression observed during osteogenesis is likely a negative feedback mechanism. We hypothesize that focal adhesion signaling pathways such as PI3K/Akt signaling may be negatively regulated by Cav-1 during human MSC osteogenesis. Methods Human bone marrow MSCs were isolated from femoral heads obtained after total hip arthroplasty. MSCs were incubated in standard growth medium alone or induced to osteogenically differentiate by the addition of supplements (β-glycerophosphate, ascorbic acid, dexamethasone, and 1,25-dihydroxyvitamin D3). The activation of and requirement for PI3K/Akt signaling in MSC osteogenesis were assessed by immunoblotting for phosphorylated Akt, and treatment with the PI3K inhibitor LY294002 and Akt siRNA, respectively. The influences of Cav-1 and cholesterol membrane rafts on PI3K/Akt signaling were investigated by treatment with Cav-1 siRNA, methyl-β-cyclodextrin, or cholesterol oxidase, followed by cellular sub-fractionation and/or immunoblotting for phosphorylated Akt. Results LY294002 and Akt siRNA inhibited MSC osteogenesis. Methyl-β-cyclodextrin, which disrupts all membrane rafts, inhibited osteogenesis. Conversely, Cav-1 siRNA and cholesterol oxidase, which displaces Cav-1 from caveolae, enhanced Akt signaling induced by osteogenic supplements. In control cells, phosphorylated Akt began to accumulate in caveolae after 10 days of osteogenic differentiation. Conclusions PI3K/Akt signaling is a key pathway required for human MSC osteogenesis, and it is likely that localization of active Akt in non-caveolar and caveolar membrane rafts positively and negatively contributes to osteogenesis, respectively.
Collapse
Affiliation(s)
- Natasha Baker
- Center for Cellular and Molecular Engineering, Department of Orthopaedic Surgery, University of Pittsburgh School of Medicine, 450 Technology Drive, Room 221, Pittsburgh, PA, 15219-3143, USA. .,Present address: Kenneth P. Dietrich School of Arts and Sciences, Department of Biological Sciences, University of Pittsburgh, 4249 Fifth Avenue, Pittsburgh, PA, 15260, USA.
| | - Jihee Sohn
- Center for Cellular and Molecular Engineering, Department of Orthopaedic Surgery, University of Pittsburgh School of Medicine, 450 Technology Drive, Room 221, Pittsburgh, PA, 15219-3143, USA.
| | - Rocky S Tuan
- Center for Cellular and Molecular Engineering, Department of Orthopaedic Surgery, University of Pittsburgh School of Medicine, 450 Technology Drive, Room 221, Pittsburgh, PA, 15219-3143, USA.
| |
Collapse
|
29
|
Wang S, Kaartinen MT. Cellular Factor XIIIA Transglutaminase Localizes in Caveolae and Regulates Caveolin-1 Phosphorylation, Homo-oligomerization and c-Src Signaling in Osteoblasts. J Histochem Cytochem 2015; 63:829-41. [PMID: 26231113 DOI: 10.1369/0022155415597964] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2015] [Accepted: 07/05/2015] [Indexed: 11/22/2022] Open
Abstract
Transglutaminases (TGs) are a family of widely distributed enzymes that catalyze protein crosslinking by forming a covalent isopeptide bond between the substrate proteins. We have shown that MC3T3-E1 osteoblasts express Factor XIII-A (FXIII-A), and that the extracellular crosslinking activity of FXIII-A is involved in regulating matrix secretion and deposition. In this study, we have investigated the localization and potential role of intracellular FXIII-A. Conventional immunofluorescence microscopy and TIRF microscopy analyses showed that FXIII-A co-localizes with caveolin-1 in specialized membrane structures, caveolae, in differentiating osteoblasts. The caveolae-disrupting agent methyl-β-cyclodextrin abolished FXIII-A staining and co-localization with caveolin-1 from the osteoblast plasma membrane. The presence of FXIII-A in caveolae was confirmed by preparing caveolae-enriched cellular fractions using sucrose density gradient ultracentrifugation followed by western blotting. Despite this association of FXIII-A with caveolae, there was no detectable transglutaminase activity in caveolae, as measured by monodansylcadaverine incorporation. TG inhibitor NC9--which can alter TG enzyme conformation--localized to caveolae and displaced FXIII-A from these structures when added to the osteoblast cultures. The decreased FXIII-A levels in caveolae after NC9 treatment increased c-Src activation, which resulted in caveolin-1 phosphorylation, homo-oligomerization and Akt phosphorylation, suggesting cellular FXIII-A has a role in regulating c-Src signaling in osteoblasts.
Collapse
Affiliation(s)
- Shuai Wang
- Division of Biomedical Sciences, Faculty of Dentistry, McGill University, Montreal, QC, Canada. (SW, MTK)
| | - Mari T Kaartinen
- Division of Experimental Medicine, Department of Medicine, Faculty of Medicine, McGill University, Montreal, QC, Canada (MTK),Division of Biomedical Sciences, Faculty of Dentistry, McGill University, Montreal, QC, Canada. (SW, MTK)
| |
Collapse
|
30
|
Lee YD, Yoon SH, Park CK, Lee J, Lee ZH, Kim HH. Caveolin-1 regulates osteoclastogenesis and bone metabolism in a sex-dependent manner. J Biol Chem 2015; 290:6522-30. [PMID: 25561739 DOI: 10.1074/jbc.m114.598581] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Lipid raft microdomains have important roles in various cellular responses. Caveolae are a specialized type of lipid rafts that are stabilized by oligomers of caveolin proteins. Here, we show that caveolin-1 (Cav-1) plays a crucial role in the regulation of osteoclastogenesis. We found that caveolin-1 was dramatically up-regulated by receptor activator of nuclear factor κB ligand (RANKL), the osteoclast differentiation factor. Knockdown of Cav-1 reduced osteoclastogenesis and induction of NFATc1, the master transcription factor for osteoclastogenesis, by RANKL. Consistent with the in vitro results, injection of caveolin-1 siRNA onto mice calvariae showed reduction in RANKL-induced bone resorption and osteoclast formation. Moreover, Cav-1(-/-) female mice had higher bone volume and lower osteoclast number compared with wild type mice. However, Cav-1(-/-) male mice had both osteoclast and osteoblast numbers higher than wild type mice with no difference in bone volume. The sex dependence in the effect of Cav-1 deficiency was partly attributed to decreased receptor activator of nuclear factor κB and increased cFms expression in osteoclast precursors of female and male mice, respectively. Taken together, these data demonstrate that Cav-1 has a complicated but critical role for osteoclastogenesis.
Collapse
Affiliation(s)
- Yong Deok Lee
- From the Department of Cell and Developmental Biology, BK21 Program and Dental Research Institute, Seoul National University, 28 Yeongon-Dong, Chongno-Gu, Seoul 110-749, Korea
| | - Soo-Hyun Yoon
- From the Department of Cell and Developmental Biology, BK21 Program and Dental Research Institute, Seoul National University, 28 Yeongon-Dong, Chongno-Gu, Seoul 110-749, Korea
| | - Cheol Kyu Park
- From the Department of Cell and Developmental Biology, BK21 Program and Dental Research Institute, Seoul National University, 28 Yeongon-Dong, Chongno-Gu, Seoul 110-749, Korea
| | - Jiyeon Lee
- From the Department of Cell and Developmental Biology, BK21 Program and Dental Research Institute, Seoul National University, 28 Yeongon-Dong, Chongno-Gu, Seoul 110-749, Korea
| | - Zang Hee Lee
- From the Department of Cell and Developmental Biology, BK21 Program and Dental Research Institute, Seoul National University, 28 Yeongon-Dong, Chongno-Gu, Seoul 110-749, Korea
| | - Hong-Hee Kim
- From the Department of Cell and Developmental Biology, BK21 Program and Dental Research Institute, Seoul National University, 28 Yeongon-Dong, Chongno-Gu, Seoul 110-749, Korea
| |
Collapse
|
31
|
Abstract
1α,25-dihydroxyvitamin D3 [1α,25(OH)2D3] is crucial for normal skeletal development and bone homeostasis. Protein disulfide isomerase family A, member 3 (PDIA3) mediates 1α,25(OH)2D3 initiated-rapid membrane signaling in several cell types. To understand its role in regulating skeletal development, we generated Pdia3-deficient mice and examined the physiologic consequence of Pdia3-disruption in embryos and Pdia3+/− heterozygotes at different ages. No mice homozygous for the Pdia3-deletion were found at birth nor were there embryos after E12.5, indicating that targeted disruption of the Pdia3 gene resulted in early embryonic lethality. Pdia3-deficiency also resulted in skeletal manifestations as revealed by µCT analysis of the tibias. In comparison to wild type mice, Pdia3 heterozygous mice displayed expanded growth plates associated with decreased tether formation. Histomorphometry also showed that the hypertrophic zone in Pdia3+/− mice was more cellular than seen in wild type growth plates. Metaphyseal trabecular bone in Pdia3+/− mice exhibited an age-dependent phenotype with lower BV/TV and trabecular numbers, which was most pronounced at 15 weeks of age. Bone marrow cells from Pdia3+/− mice exhibited impaired osteoblastic differentiation, based on reduced expression of osteoblast markers and mineral deposition compared to cells from wild type animals. Collectively, our findings provide in vivo evidence that PDIA3 is essential for normal skeletal development. The fact that the Pdia3+/− heterozygous mice share a similar growth plate and bone phenotype to nVdr knockout mice, suggests that PDIA3-mediated rapid membrane signaling might be an alternative mechanism responsible for 1α,25(OH)2D3’s actions in regulating skeletal development.
Collapse
|
32
|
Gangadharan V, Nohe A, Caplan J, Czymmek K, Duncan RL. Caveolin-1 regulates P2X7 receptor signaling in osteoblasts. Am J Physiol Cell Physiol 2014; 308:C41-50. [PMID: 25318104 DOI: 10.1152/ajpcell.00037.2014] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The synthesis of new bone in response to a novel applied mechanical load requires a complex series of cellular signaling events in osteoblasts and osteocytes. The activation of the purinergic receptor P2X(7)R is central to this mechanotransduction signaling cascade. Recently, P2X(7)R have been found to be associated with caveolae, a subset of lipid microdomains found in several cell types. Deletion of caveolin-1 (CAV1), the primary protein constituent of caveolae in osteoblasts, results in increased bone mass, leading us to hypothesize that the P2X(7)R is scaffolded to caveolae in osteoblasts. Thus, upon activation of the P2X(7)R, we postulate that caveolae are endocytosed, thereby modulating the downstream signal. Sucrose gradient fractionation of MC3T3-E1 preosteoblasts showed that CAV1 was translocated to the denser cytosolic fractions upon stimulation with ATP. Both ATP and the more specific P2X(7)R agonist 2'(3')-O-(4-benzoylbenzoyl)ATP (BzATP) induced endocytosis of CAV1, which was inhibited when MC3T3-E1 cells were pretreated with the specific P2X7R antagonist A-839977. The P2X7R cofractionated with CAV1, but, using superresolution structured illumination microscopy, we found only a subpopulation of P2X(7)R in these lipid microdomains on the membrane of MC3T3-E1 cells. Suppression of CAV1 enhanced the intracellular Ca(2+) response to BzATP, suggesting that caveolae regulate P2X(7)R signaling. This proposed mechanism is supported by increased mineralization in CAV1 knockdown MC3T3-E1 cells treated with BzATP. These data suggest that caveolae regulate P2X(7)R signaling upon activation by undergoing endocytosis and potentially carrying with it other signaling proteins, hence controlling the spatiotemporal signaling of P2X(7)R in osteoblasts.
Collapse
Affiliation(s)
- Vimal Gangadharan
- Department of Biological Sciences, University of Delaware, Newark, Delaware; and
| | - Anja Nohe
- Department of Biological Sciences, University of Delaware, Newark, Delaware; and
| | - Jeffrey Caplan
- Department of Biological Sciences, University of Delaware, Newark, Delaware; and Bioimaging Center, Delaware Biotechnology Institute, Newark, Delaware
| | - Kirk Czymmek
- Department of Biological Sciences, University of Delaware, Newark, Delaware; and Bioimaging Center, Delaware Biotechnology Institute, Newark, Delaware
| | - Randall L Duncan
- Department of Biological Sciences, University of Delaware, Newark, Delaware; and Bioimaging Center, Delaware Biotechnology Institute, Newark, Delaware
| |
Collapse
|
33
|
Gredes T, Gedrange T, Hinüber C, Gelinsky M, Kunert-Keil C. Histological and molecular-biological analyses of poly(3-hydroxybutyrate) (PHB) patches for enhancement of bone regeneration. Ann Anat 2014; 199:36-42. [PMID: 24862689 DOI: 10.1016/j.aanat.2014.04.003] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2014] [Revised: 02/17/2014] [Accepted: 04/18/2014] [Indexed: 01/25/2023]
Abstract
Tissue engineered cell-seeded constructs with poly(3)hydroxybutyrate (PHB) induced ectopic bone formation after implantation into the back muscle of rats. The objective of our in vivo study was to evaluate the osteogenic potential of pure PHB patches in surgically created cranial defects. For this, PHB patches were analyzed after implantation in surgically created defects on the cranium of adult male rats. After healing periods of 4, 8 and 12 weeks, the bone tissue specimens containing PHB patches were processed and analyzed histologically as well as molecular-biologically. After 4 weeks, the PHB patches were completely embedded in connective tissue. Eight weeks after PHB insertion, bone regeneration proceeding from bearing bone was found in 50% of all treated animals, whereas all PHB treated cavities showed both bone formation and embedding of the patches in bone 12 weeks after surgery. Furthermore, all slices showed pronounced development of blood vessels. Histomorphometric analysis presented a regenerated bone mean value between 46.4 ± 16.1% and 54.2 ± 19.3% after 4-12 weeks of healing. Caveolin-1 staining in capillary-like structures showed a 1.16-1.38 fold increased expression in PHB treated defects compared to controls. Real-time RT-PCR analyses showed significantly lower expressions of Alpl, Col1a1 and VEGFA in cranium defects after treatment with PHB patches compared to untreated bony defects of the same cranium. Within the limits of the presented animal investigation, it could conclude that the tested PHB patches featured a good biocompatibility and an osteoconductive character.
Collapse
Affiliation(s)
- Tomasz Gredes
- Department of Orthodontics, Carl Gustav Carus Campus, Technische Universität Dresden, Germany.
| | - Tomasz Gedrange
- Department of Orthodontics, Carl Gustav Carus Campus, Technische Universität Dresden, Germany
| | - Claudia Hinüber
- Leibniz Institute of Polymer Research Dresden e. V., Dresden, Germany; Institute of Material Science, Technische Universität Dresden, Germany
| | - Michael Gelinsky
- Centre for Translational Bone, Joint and Soft tissue research, Carl Gustav Carus Campus, Technische Universität, Dresden, Germany
| | - Christiane Kunert-Keil
- Department of Orthodontics, Carl Gustav Carus Campus, Technische Universität Dresden, Germany
| |
Collapse
|
34
|
Zhang J, Lazarenko OP, Blackburn ML, Badger TM, Ronis MJJ, Chen J. Soy protein isolate down‐regulates caveolin‐1 expression to suppress osteoblastic cell senescence pathways. FASEB J 2014; 28:3134-45. [DOI: 10.1096/fj.13-243659] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Affiliation(s)
- Jian Zhang
- Arkansas Children's Nutrition CenterLittle RockArkansasUSA
- Department of PediatricsUniversity of Arkansas for Medical SciencesLittle RockArkansasUSA
| | - Oxana P. Lazarenko
- Arkansas Children's Nutrition CenterLittle RockArkansasUSA
- Department of PediatricsUniversity of Arkansas for Medical SciencesLittle RockArkansasUSA
| | - Michael L. Blackburn
- Arkansas Children's Nutrition CenterLittle RockArkansasUSA
- Department of PediatricsUniversity of Arkansas for Medical SciencesLittle RockArkansasUSA
| | - Thomas M. Badger
- Arkansas Children's Nutrition CenterLittle RockArkansasUSA
- Department of PediatricsUniversity of Arkansas for Medical SciencesLittle RockArkansasUSA
| | - Martin J. J. Ronis
- Arkansas Children's Nutrition CenterLittle RockArkansasUSA
- Department of PediatricsUniversity of Arkansas for Medical SciencesLittle RockArkansasUSA
- Department of Pharmacology and ToxicologyUniversity of Arkansas for Medical SciencesLittle RockArkansasUSA
| | - Jin‐Ran Chen
- Arkansas Children's Nutrition CenterLittle RockArkansasUSA
- Department of PediatricsUniversity of Arkansas for Medical SciencesLittle RockArkansasUSA
| |
Collapse
|
35
|
Abstract
Marrow adipose tissue (MAT) is functionally distinct from both white and brown adipose tissue and can contribute to systemic and skeletal metabolism. MAT formation is a spatially and temporally defined developmental event, suggesting that MAT is an organ that serves important functions and, like other organs, can undergo pathologic change. The well-documented inverse relationship between MAT and bone mineral density has been interpreted to mean that MAT removal is a possible therapeutic target for osteoporosis. However, the bone and metabolic phenotypes of patients with lipodystrophy argues that retention of MAT may actually be beneficial in some circumstances. Furthermore, MAT may exist in two forms, regulated and constitutive, with divergent responses to hematopoietic and nutritional demands. In this review, we discuss the role of MAT in lipodystrophy, bone loss, and metabolism, and highlight our current understanding of this unique adipose tissue depot.
Collapse
Affiliation(s)
- Erica L Scheller
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, Michigan
| | | |
Collapse
|
36
|
Martineau C, Martin-Falstrault L, Brissette L, Moreau R. The atherogenic Scarb1 null mouse model shows a high bone mass phenotype. Am J Physiol Endocrinol Metab 2014; 306:E48-57. [PMID: 24253048 PMCID: PMC3920004 DOI: 10.1152/ajpendo.00421.2013] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Scavenger receptor class B, type I (SR-BI), the Scarb1 gene product, is a receptor associated with cholesteryl ester uptake from high-density lipoproteins (HDL), which drives cholesterol movement from peripheral tissues toward the liver for excretion, and, consequently, Scarb1 null mice are prone to atherosclerosis. Because studies have linked atherosclerosis incidence with osteoporosis, we characterized the bone metabolism in these mice. Bone morphometry was assessed through microcomputed tomography and histology. Marrow stromal cells (MSCs) were used to characterize influence of endogenous SR-BI in cell functions. Total and HDL-associated cholesterol in null mice were increased by 32-60%, correlating with its role in lipoprotein metabolism. Distal metaphyses from 2- and 4-mo-old null mice showed correspondingly 46 and 37% higher bone volume fraction associated with a higher number of trabeculae. Histomorphometric analyses in 2-mo-old null male mice revealed 1.42-fold greater osteoblast surface, 1.37-fold higher percent mineralizing surface, and 1.69-fold enhanced bone formation rate. In vitro assays for MSCs from null mice revealed 37% higher proliferation rate, 48% more alkaline phosphatase activity, 70% greater mineralization potential and a 2-fold osterix (Sp7) expression, yet a 0.5-fold decrease in caveolin-1 (Cav1) expression. Selective uptake levels of HDL-associated cholesteryl oleate and estradiol were similar between MSC from wild-type and Scarb1 null mice, suggesting that its contribution to this process is not its main role in these cells. However, Scarb1 knockout stunted the HDL-dependent regulation of Cav1 genic expression. Scarb1 null mice are not prone to osteoporosis but show higher bone mass associated with enhanced bone formation.
Collapse
Affiliation(s)
- Corine Martineau
- Laboratoire du Métabolisme Osseux, BioMed, Université du Québec à Montréal, Département des Sciences Biologiques, Université du Québec à Montréal, Montréal, Quebec, Canada; and
| | | | | | | |
Collapse
|
37
|
The less-often-traveled surface of stem cells: caveolin-1 and caveolae in stem cells, tissue repair and regeneration. Stem Cell Res Ther 2013; 4:90. [PMID: 23899671 PMCID: PMC3854699 DOI: 10.1186/scrt276] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
Stem cells are an important resource for tissue repair and regeneration. While a great deal of attention has focused on derivation and molecular regulation of stem cells, relatively little research has focused on how the subcellular structure and composition of the cell membrane influences stem cell activities such as proliferation, differentiation and homing. Caveolae are specialized membrane lipid rafts coated with caveolin scaffolding proteins, which can regulate cholesterol transport and the activity of cell signaling receptors and their downstream effectors. Caveolin-1 is involved in the regulation of many cellular processes, including growth, control of mitochondrial antioxidant levels, migration and senescence. These activities are of relevance to stem cell biology, and in this review evidence for caveolin-1 involvement in stem cell biology is summarized. Altered stem and progenitor cell populations in caveolin-1 null mice suggest that caveolin-1 can regulate stem cell proliferation, and in vitro studies with isolated stem cells suggest that caveolin-1 regulates stem cell differentiation. The available evidence leads us to hypothesize that caveolin-1 expression may stabilize the differentiated and undifferentiated stem cell phenotype, and transient downregulation of caveolin-1 expression may be required for transition between the two. Such regulation would probably be critical in regenerative applications of adult stem cells and during tissue regeneration. We also review here the temporal changes in caveolin-1 expression reported during tissue repair. Delayed muscle regeneration in transgenic mice overexpressing caveolin-1 as well as compromised cardiac, brain and liver tissue repair and delayed wound healing in caveolin-1 null mice suggest that caveolin-1 plays an important role in tissue repair, but that this role may be negative or positive depending on the tissue type and the nature of the repair process. Finally, we also discuss how caveolin-1 quiescence-inducing activities and effects on mitochondrial antioxidant levels may influence stem cell aging.
Collapse
|
38
|
Baker N, Zhang G, You Y, Tuan RS. Caveolin-1 regulates proliferation and osteogenic differentiation of human mesenchymal stem cells. J Cell Biochem 2013; 113:3773-87. [PMID: 22807396 DOI: 10.1002/jcb.24252] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Caveolin-1 is a scaffolding protein of cholesterol-rich caveolae lipid rafts in the plasma membrane. In addition to regulating cholesterol transport, caveolin-1 has the ability to bind a diverse array of cell signaling molecules and regulate cell signal transduction in caveolae. Currently, there is little known about the role of caveolin-1 in stem cells. It has been reported that the caveolin-1 null mouse has an expanded population of cells expressing stem cell markers in the gut, mammary gland, and brain, suggestive of a role for caveolin-1 in stem cell regulation. The caveolin-1 null mouse also has increased bone mass and an increased bone formation rate, and its bone marrow-derived mesenchymal stem cells (MSCs) have enhanced osteogenic potential. However, the role of caveolin-1 in human MSC osteogenic differentiation remains unexplored. In this study, we have characterized the expression of caveolin-1 in human bone marrow derived MSCs. We show that caveolin-1 protein is enriched in density gradient-fractionated MSC plasma membrane, consisting of ~100 nm diameter membrane-bound vesicles, and is distributed in a punctate pattern by immunofluoresence localization. Expression of caveolin-1 increases in MSCs induced to undergo osteogenic differentiation, and siRNA-mediated knockdown of caveolin-1 expression enhances MSC proliferation and osteogenic differentiation. Taken together, these findings suggest that caveolin-1 normally acts to regulate the differentiation and renewal of MSCs, and increased caveolin-1 expression during MSC osteogenesis likely acts as a negative feedback to stabilize the cell phenotype.
Collapse
Affiliation(s)
- Natasha Baker
- Cartilage Biology and Orthopaedics Branch, National Institute of Arthritis and Musculoskeletal and Skin Diseases, Bethesda, MD 20892, USA
| | | | | | | |
Collapse
|
39
|
Smolders LA, Meij BP, Onis D, Riemers FM, Bergknut N, Wubbolts R, Grinwis GCM, Houweling M, Groot Koerkamp MJA, van Leenen D, Holstege FCP, Hazewinkel HAW, Creemers LB, Penning LC, Tryfonidou MA. Gene expression profiling of early intervertebral disc degeneration reveals a down-regulation of canonical Wnt signaling and caveolin-1 expression: implications for development of regenerative strategies. Arthritis Res Ther 2013; 15:R23. [PMID: 23360510 PMCID: PMC3672710 DOI: 10.1186/ar4157] [Citation(s) in RCA: 62] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2012] [Accepted: 01/10/2013] [Indexed: 01/07/2023] Open
Abstract
INTRODUCTION Early degeneration of the intervertebral disc (IVD) involves a change in cellular differentiation from notochordal cells (NCs) in the nucleus pulposus (NP) to chondrocyte-like cells (CLCs). The purpose of this study was to investigate the gene expression profiles involved in this process using NP tissue from non-chondrodystrophic and chondrodystrophic dogs, a species with naturally occurring IVD degeneration. METHODS Dual channel DNA microarrays were used to compare 1) healthy NP tissue containing only NCs (NC-rich), 2) NP tissue with a mixed population of NCs and CLCs (Mixed), and 3) NP tissue containing solely CLCs (CLC-rich) in both non-chondrodystrophic and chondrodystrophic dogs. Based on previous reports and the findings of the microarray analyses, canonical Wnt signaling was further evaluated using qPCR of relevant Wnt target genes. We hypothesized that caveolin-1, a regulator of Wnt signaling that showed significant changes in gene expression in the microarray analyses, played a significant role in early IVD degeneration. Caveolin-1 expression was investigated in IVD tissue sections and in cultured NCs. To investigate the significance of Caveolin-1 in IVD health and degeneration, the NP of 3-month-old Caveolin-1 knock-out mice was histopathologically evaluated and compared with the NP of wild-type mice of the same age. RESULTS Early IVD degeneration involved significant changes in numerous pathways, including Wnt/β-catenin signaling. With regard to Wnt/β-catenin signaling, axin2 gene expression was significantly higher in chondrodystrophic dogs compared with non-chondrodystrophic dogs. IVD degeneration involved significant down-regulation of axin2 gene expression. IVD degeneration involved significant down-regulation in Caveolin-1 gene and protein expression. NCs showed abundant caveolin-1 expression in vivo and in vitro, whereas CLCs did not. The NP of wild-type mice was rich in viable NCs, whereas the NP of Caveolin-1 knock-out mice contained chondroid-like matrix with mainly apoptotic, small, rounded cells. CONCLUSIONS Early IVD degeneration involves down-regulation of canonical Wnt signaling and Caveolin-1 expression, which appears to be essential to the physiology and preservation of NCs. Therefore, Caveolin-1 may be regarded an exciting target for developing strategies for IVD regeneration.
Collapse
|
40
|
Chen C, Qiao R, Wei R, Guo Y, Ai H, Ma J, Ren J, Huang L. A comprehensive survey of copy number variation in 18 diverse pig populations and identification of candidate copy number variable genes associated with complex traits. BMC Genomics 2012; 13:733. [PMID: 23270433 PMCID: PMC3543711 DOI: 10.1186/1471-2164-13-733] [Citation(s) in RCA: 81] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2012] [Accepted: 12/15/2012] [Indexed: 01/04/2023] Open
Abstract
Background Copy number variation (CNV) is a major source of structural variants and has been commonly identified in mammalian genome. It is associated with gene expression and may present a major genetic component of phenotypic diversity. Unlike many other mammalian genomes where CNVs have been well annotated, studies of porcine CNV in diverse breeds are still limited. Result Here we used Porcine SNP60 BeadChip and PennCNV algorithm to identify 1,315 putative CNVs belonging to 565 CNV regions (CNVRs) in 1,693 pigs from 18 diverse populations. Total 538 out of 683 CNVs identified in a White Duroc × Erhualian F2 population fit Mendelian transmission and 6 out of 7 randomly selected CNVRs were confirmed by quantitative real time PCR. CNVRs were non-randomly distributed in the pig genome. Several CNV hotspots were found on pig chromosomes 6, 11, 13, 14 and 17. CNV numbers differ greatly among different pig populations. The Duroc pigs were identified to have the most number of CNVs per individual. Among 1,765 transcripts located within the CNVRs, 634 genes have been reported to be copy number variable genes in the human genome. By integrating analysis of QTL mapping, CNVRs and the description of phenotypes in knockout mice, we identified 7 copy number variable genes as candidate genes for phenotypes related to carcass length, backfat thickness, abdominal fat weight, length of scapular, intermuscle fat content of logissimus muscle, body weight at 240 day, glycolytic potential of logissimus muscle, mean corpuscular hemoglobin, mean corpuscular volume and humerus diameter. Conclusion We revealed the distribution of the unprecedented number of 565 CNVRs in pig genome and investigated copy number variable genes as the possible candidate genes for phenotypic traits. These findings give novel insights into porcine CNVs and provide resources to facilitate the identification of trait-related CNVs.
Collapse
Affiliation(s)
- Congying Chen
- Key Laboratory for Animal Biotechnology of Jiangxi Province and the Ministry of Agriculture of China, Jiangxi Agricultural University, Nanchang, 330045, China
| | | | | | | | | | | | | | | |
Collapse
|
41
|
Hermann CD, Lee CSD, Gadepalli S, Lawrence KA, Richards MA, Olivares-Navarrete R, Williams JK, Schwartz Z, Boyan BD. Interrelationship of cranial suture fusion, basicranial development, and resynostosis following suturectomy in twist1(+/-) mice, a murine model of Saethre-Chotzen syndrome. Calcif Tissue Int 2012; 91:255-66. [PMID: 22903506 DOI: 10.1007/s00223-012-9632-3] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/10/2012] [Accepted: 06/07/2012] [Indexed: 01/12/2023]
Abstract
The interrelationships among suture fusion, basicranial development, and subsequent resynostosis in syndromic craniosynostosis have yet to be examined. The objectives of this study were to determine the potential relationship between suture fusion and cranial base development in a model of syndromic craniosynostosis and to assess the effects of the syndrome on resynostosis following suturectomy. To do this, posterior frontal and coronal suture fusion, postnatal development of sphenooccipital synchondrosis, and resynostosis in Twist1(+/+) (WT) and Twist1(+/-) litter-matched mice (a model for Saethre-Chotzen syndrome) were quantified by evaluating μCT images with advanced image-processing algorithms. The coronal suture in Twist(+/-) mice developed, fused, and mineralized at a faster rate than that in normal littermates at postnatal days 6-30. Moreover, premature fusion of the coronal suture in Twist1(+/-) mice preceded alterations in cranial base development. Analysis of synchondrosis showed faster mineralization in Twist(+/-) mice at postnatal days 25-30. In a rapid resynostosis model, there was an inability to fuse both the midline posterior frontal suture and craniotomy defects in 21-day-old Twist(+/-) mice, despite having accelerated mineralization in the posterior frontal suture and defects. This study showed that dissimilarities between Twist1(+/+) and Twist1(+/-) mice are not limited to a fused coronal suture but include differences in fusion of other sutures, the regenerative capacity of the cranial vault, and the development of the cranial base.
Collapse
Affiliation(s)
- Christopher D Hermann
- Wallace H. Coulter Department of Biomedical Engineering, Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA 30332-0363, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Canullo L, Heinemann F, Gedrange T, Biffar R, Kunert-Keil C. Histological evaluation at different times after augmentation of extraction sites grafted with a magnesium-enriched hydroxyapatite: double-blinded randomized controlled trial. Clin Oral Implants Res 2012; 24:398-406. [DOI: 10.1111/clr.12035] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/30/2012] [Indexed: 11/30/2022]
Affiliation(s)
| | - Friedhelm Heinemann
- Department of Prosthodontics, Gerostomatolgy and Biomaterials; University Medicine of Greifswald; Greifswald; Germany
| | - Tomasz Gedrange
- Deptartment of Orthodontics; Technische Universität Dresden; Dresden; Germany
| | - Reiner Biffar
- Department of Prosthodontics, Gerostomatolgy and Biomaterials; University Medicine of Greifswald; Greifswald; Germany
| | | |
Collapse
|
43
|
Boyan BD, Chen J, Schwartz Z. Mechanism of Pdia3-dependent 1α,25-dihydroxy vitamin D3 signaling in musculoskeletal cells. Steroids 2012; 77:892-6. [PMID: 22569272 DOI: 10.1016/j.steroids.2012.04.018] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2012] [Revised: 04/14/2012] [Accepted: 04/25/2012] [Indexed: 12/29/2022]
Abstract
1α,25-Dihydroxy vitamin D3 [1,25(OH)2D3] acts on cells through traditional steroid hormone receptor-mediated gene transcription and by initiating rapid membrane-associated signaling pathways. Two receptors have been implicated in rapid signaling by 1,25(OH)2D3, the classical nuclear vitamin D receptor (VDR) and the more recently identified protein disulfide isomerase, family A, member 3 (Pdia3). Our lab along with other groups has established various tools to investigate the role of these two receptors, including gene knock-out, conditional knock-out, silencing, and over-expression in various model systems (growth plate chondrocytes, osteoblastic cells, chick intestinal epithelial cells, mouse embryoid bodies, extracellular matrix vesicles and isolated cell membranes). The data demonstrate the requirement for Pdia3 in 1,25(OH)2D3 induced phospholipase A2 (PLA2) and protein kinase C (PKC) activation and downstream responses. Pdia3+/- heterozygote mice also exhibit both cartilage and bone defects. VDR is present on the plasma membrane and one VDR-/- mouse strain lacks transcaltachia, although 1,25(OH)2D3 induced PKC activation and transcaltachia are not affected in another VDR-/- mouse strain. In the context of osteoblast differentiation, both receptors are expressed during osteogenic commitment of embryoid bodies and silencing of each causes a more mature osteoblast phenotype in MC3T3-E1 pre-osteoblasts. Pdia3 exists in caveolae, where it interacts with PLA2 activating protein (PLAA) and caveolin-1 to initiate rapid signaling via PLA2, phospholipase C (PLC), PKC, and ultimately the ERK1/2 family of mitogen activated protein kinases (MAPK). Using the growth plate chondrocyte and matrix vesicle models, we have demonstrated that Pdia3-dependent signaling in response to 1,25(OH)2D3 regulates growth plate physiology.
Collapse
Affiliation(s)
- Barbara D Boyan
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA, USA.
| | | | | |
Collapse
|
44
|
Abstract
Caveolins (Cavs) are integrated plasma membrane proteins that are complex signaling regulators with numerous partners and whose activity is highly dependent on cellular context. Cavs are both positive and negative regulators of cell signaling in and/or out of caveolae, invaginated lipid raft domains whose formation is caveolin expression dependent. Caveolins and rafts have been implicated in membrane compartmentalization; proteins and lipids accumulate in these membrane microdomains where they transmit fast, amplified and specific signaling cascades. The concept of plasma membrane organization within functional rafts is still in exploration and sometimes questioned. In this chapter, we discuss the opposing functions of caveolin in cell signaling regulation focusing on the role of caveolin both as a promoter and inhibitor of different signaling pathways and on the impact of membrane domain localization on caveolin functionality in cell proliferation, survival, apoptosis and migration.
Collapse
|
45
|
Thompson WR, Rubin CT, Rubin J. Mechanical regulation of signaling pathways in bone. Gene 2012; 503:179-93. [PMID: 22575727 DOI: 10.1016/j.gene.2012.04.076] [Citation(s) in RCA: 287] [Impact Index Per Article: 22.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2012] [Revised: 03/20/2012] [Accepted: 04/22/2012] [Indexed: 12/21/2022]
Abstract
A wide range of cell types depend on mechanically induced signals to enable appropriate physiological responses. The skeleton is particularly dependent on mechanical information to guide the resident cell population towards adaptation, maintenance and repair. Research at the organ, tissue, cell and molecular levels has improved our understanding of how the skeleton can recognize the functional environment, and how these challenges are translated into cellular information that can site-specifically alter phenotype. This review first considers those cells within the skeleton that are responsive to mechanical signals, including osteoblasts, osteoclasts, osteocytes and osteoprogenitors. This is discussed in light of a range of experimental approaches that can vary parameters such as strain, fluid shear stress, and pressure. The identity of mechanoreceptor candidates is approached, with consideration of integrins, pericellular tethers, focal adhesions, ion channels, cadherins, connexins, and the plasma membrane including caveolar and non-caveolar lipid rafts and their influence on integral signaling protein interactions. Several mechanically regulated intracellular signaling cascades are detailed including activation of kinases (Akt, MAPK, FAK), β-catenin, GTPases, and calcium signaling events. While the interaction of bone cells with their mechanical environment is complex, an understanding of mechanical regulation of bone signaling is crucial to understanding bone physiology, the etiology of diseases such as osteoporosis, and to the development of interventions to improve bone strength.
Collapse
Affiliation(s)
- William R Thompson
- Department of Medicine, University of North Carolina, Chapel Hill, NC 27599, USA.
| | | | | |
Collapse
|
46
|
Zhang J, Lazarenko OP, Wu X, Tong Y, Blackburn ML, Gomez-Acevedo H, Shankar K, Badger TM, Ronis MJJ, Chen JR. Differential effects of short term feeding of a soy protein isolate diet and estrogen treatment on bone in the pre-pubertal rat. PLoS One 2012; 7:e35736. [PMID: 22536432 PMCID: PMC3335011 DOI: 10.1371/journal.pone.0035736] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2011] [Accepted: 03/20/2012] [Indexed: 11/19/2022] Open
Abstract
Background Previous reports suggest that beneficial effects of soy on bone quality are due to the estrogenic actions of isoflavone phytochemicals associated with the protein. However, mechanistic studies comparing the effects of soy diet and estrogens on bone, particularly in rapidly growing animals are lacking. Methodology and Principal Findings We studied the effects of short term feeding of soy protein isolate (SPI) on bone in comparison to the effects of 17β-estradiol (E2) in pre-pubertal rats. Female rats were weaned to one of 4 treatments: 1) a control casein-based diet (CAS); 2) CAS with subcutaneous E2 (10 µg/kg/d) (CAS+E2); 3) a SPI-containing diet (SPI); or 4) SPI with subcutaneous E2 (SPI) or SPI with 10 µg/kg/d E2 (SPI+E2) for 14 days beginning on postnatal day 20. SPI increased while E2 decreased bone turnover compared to CAS. In contrast, both treatments decreased serum sclerostin levels. Microarray analysis of RNA isolated from bone revealed 652 genes regulated by SPI, 491 genes regulated by E2, and 266 genes regulated by both SPI diet and E2 compared to CAS. The expression of caveolin-1, a protein localized in the cell membrane, was down-regulated (p<0.05) in rats fed SPI, but not by E2 compared to rats fed casein. Down-regulation of caveolin-1 by SPI was associated with increased BMP2, Smad and Runx2 expression in bone and osteoblasts (p<0.05). Conclusions/Significance These results suggest SPI and E2 have different effects on bone turnover prior to puberty. Approximately half of the genes are regulated in the same direction by E2 or SPI, but in combination, SPI blocks the estrogen effects and returns the profile towards control levels. In addition, there are E2 specific and SPI-specific gene changes related to regulation of bone formation.
Collapse
Affiliation(s)
- Jian Zhang
- Arkansas Children's Nutrition Center, Little Rock, Arkansas, United States of America
- Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, Arkansas, United States of America
| | - Oxana P. Lazarenko
- Arkansas Children's Nutrition Center, Little Rock, Arkansas, United States of America
- Department of Physiology and Biophysics, University of Arkansas for Medical Sciences, Little Rock, Arkansas, United States of America
| | - Xianli Wu
- Arkansas Children's Nutrition Center, Little Rock, Arkansas, United States of America
- Department of Physiology and Biophysics, University of Arkansas for Medical Sciences, Little Rock, Arkansas, United States of America
| | - Yudong Tong
- Arkansas Children's Nutrition Center, Little Rock, Arkansas, United States of America
| | - Michael L. Blackburn
- Arkansas Children's Nutrition Center, Little Rock, Arkansas, United States of America
- Department of Physiology and Biophysics, University of Arkansas for Medical Sciences, Little Rock, Arkansas, United States of America
| | - Horatio Gomez-Acevedo
- Arkansas Children's Nutrition Center, Little Rock, Arkansas, United States of America
| | - Kartik Shankar
- Arkansas Children's Nutrition Center, Little Rock, Arkansas, United States of America
- Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, Arkansas, United States of America
| | - Thomas M. Badger
- Arkansas Children's Nutrition Center, Little Rock, Arkansas, United States of America
- Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, Arkansas, United States of America
- Department of Physiology and Biophysics, University of Arkansas for Medical Sciences, Little Rock, Arkansas, United States of America
| | - Martin J. J. Ronis
- Arkansas Children's Nutrition Center, Little Rock, Arkansas, United States of America
- Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, Arkansas, United States of America
- Department of Pharmacology and Toxicology, University of Arkansas for Medical Sciences, Little Rock, Arkansas, United States of America
| | - Jin-Ran Chen
- Arkansas Children's Nutrition Center, Little Rock, Arkansas, United States of America
- Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, Arkansas, United States of America
- * E-mail:
| |
Collapse
|
47
|
Hada N, Okayasu M, Ito J, Nakayachi M, Hayashida C, Kaneda T, Uchida N, Muramatsu T, Koike C, Masuhara M, Sato T, Hakeda Y. Receptor activator of NF-κB ligand-dependent expression of caveolin-1 in osteoclast precursors, and high dependency of osteoclastogenesis on exogenous lipoprotein. Bone 2012; 50:226-36. [PMID: 22075210 DOI: 10.1016/j.bone.2011.10.028] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/06/2011] [Revised: 10/22/2011] [Accepted: 10/25/2011] [Indexed: 02/07/2023]
Abstract
Although extensive studies have done much to clarify the molecular mechanisms of osteoclastogenesis during the last ten years, there may still be unknown molecules associated with osteoclast differentiation. Thus, we used fluorescent differential display to screen for genes whose expression is induced by receptor activator of NF-κB ligand (RANKL), a crucial molecule for osteoclast formation. We identified caveolin-1 (Cav-1) as a RANKL-induced gene. Cav-1 is a major structural protein of caveolae and lipid rafts, cholesterol-enriched microdomains in the plasma membrane (PM). The RANKL-induced Cav-1 was immediately conveyed to lipid rafts. Conversely, expression of flotillin-1 (Flot-1), another scaffolding protein of lipid rafts, was reduced during osteoclastogenesis, indicating conversion of Flot-1-predominant rafts into Cav-1-enriched rafts. However, in vitro osteoclastogenesis of precursor cells from Cav-1-null mice was comparable to that of wild-type mice, while Cav-2 expression in the knockout osteoclasts was maintained. Conversely, Cav-2 gene silencing in Cav-1-null osteoclast precursors using siRNA for Cav-2 increased osteoclast formation, suggesting that the Cav-1/Cav-2 complex may act as a negative regulator for osteoclastogenesis. On the other hand, destruction of lipid rafts by removal of cholesterol from the PM by methyl-ß-cyclodextrin (MCD) treatment caused disordered signal transductions for osteoclastogenesis, such as hyperactivation of Erk1/2 and insensitivity of Akt to RANKL stimulus. The abnormal signaling was reproduced by deleting exogenous lipoproteins from the culture medium, which also resulted in reduced osteoclast formation. In addition, the deletion caused delayed expression of nuclear factor of activated T cells c1 (NFATc1), and depressed its activation in the cytosol and inhibited its translocation into nuclei. Simultaneously, the deletion reduced the level of FcRγ, a trigger protein for initiating the calcium signaling needed to activate NFATc1, and decreased Cav-1 in lipid rafts. These findings indicate that the molecular mechanisms of osteoclastogenesis are highly dependent on extracellular lipoprotein and the integrity of lipid rafts, and suggest possible involvement of cholesterol.
Collapse
Affiliation(s)
- Naoto Hada
- Division of Oral Anatomy, Department of Human Development and Fostering, Meikai University School of Dentistry, Sakado, Saitama 350-0283, Japan
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Lee CSD, Chen J, Wang Y, Williams JK, Ranly DM, Schwartz Z, Boyan BD. Coordinated tether formation in anatomically distinct mice growth centers is dependent on a functional vitamin D receptor and is tightly linked to three-dimensional tissue morphology. Bone 2011; 49:419-27. [PMID: 21601024 DOI: 10.1016/j.bone.2011.05.004] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/19/2011] [Revised: 04/27/2011] [Accepted: 05/04/2011] [Indexed: 11/17/2022]
Abstract
Bone bridges linking the epiphysis and metaphysis termed "tethers" have been found in the femoral growth plates of C57Bl/6 mice and are disrupted when the vitamin D receptor (VDR) is ablated. It is unknown if tethers are found in other growth centers, if they are regulated in a comparable manner, or if they have a functional role in skeletal development or stability. To address this, distal femoral growth plates (GPs) and spheno-occipital synchondroses (SOSs) of wild-type C57Bl/6 mice from 2 to 15 weeks of age were analyzed using μCT scans. The GPs and SOSs of VDR+/+ and VDR-/- mice fed regular or rescue diets to restore mineral homeostasis until 10 weeks of age were also scanned. Tethers in GPs and SOSs both thickened and accumulated in number as these growth centers decreased in size. Ablating the VDR made GPs and SOSs rachitic and nearly eliminated tether formation. Rescue diets restored the volume of both growth centers but only partially restored growth center thickness and tether formation, suggesting that lα,25-dihydroxy vitamin D(3) partially regulates tether formation in these growth centers via its receptor. In VDR+/+ mice 2-15 weeks in age, growth center thickness was inversely correlated to animal weight whereas tether phenotype (tether volume/growth center volume, tether number/mm, tether width, tether spacing) was significantly related to animal weight. In both 2-15 week old VDR+/+ and 10 week old VDR+/+ and VDR-/- mice on normal and rescue diets, tether phenotype (tether number/mm, tether spacing) had strikingly similar relationships to growth center thickness. These results show that tethers are present in growth centers in different anatomic and undergo developmental changes in a comparable manner; in both sites, VDR-regulated tether formation is strongly linked to growth center morphology; and tether formation is associated with body weight, suggesting a role in maintaining growth plate stability during growth.
Collapse
Affiliation(s)
- Christopher S D Lee
- Wallace H. Coulter Department of Biomedical Engineering and Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA 30332-0363, USA
| | | | | | | | | | | | | |
Collapse
|
49
|
Abstract
We introduced the mechanosome hypothesis in 2003 as a heuristic model for investigating mechanotransduction in bone (Pavalko et al., J Cell Biochem, 2003, 88(1):104-112). This model suggested specific approaches for investigating how mechanical information is conveyed from the membrane of the sensor bone cell to the target genes and how this transmitted information from the membrane is converted into changes in transcription. The key concepts underlying the mechanosome hypothesis are that load-induced deformation of bone deforms the sensor cell membrane; embedded in the membrane are the focal adhesion and cadherin-catenin complexes, which in turn are physically connected to the chromatin via a solid-state scaffold. The physical stimulation of the membrane launches multiprotein complexes (mechanosomes) from the adhesion platforms while concomitantly tugging target genes into position for contact with the incoming mechanosomes, the carriers of the mechanical information to the nucleus. The mechanosome is comprised of an adhesion-associated protein and a nucleocytoplasmic shuttling transcription factor. Upon arrival at the target gene, mechanosomes alter DNA conformation and thus influence the interactions between trans-acting proteins along the gene, changing gene activity. Here, we update significant progress related to the mechanosome concept since publication of our original hypothesis. The launching of adhesion- and cytoskeletal-associated proteins into the nucleus toward target genes appears to be a common mechanism for regulating cell response to changes in its mechanical microenvironment.
Collapse
Affiliation(s)
- Joseph P. Bidwell
- Department of Anatomy and Cell Biology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Fredrick M. Pavalko
- Department of Cellular and Integrative Physiology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| |
Collapse
|
50
|
Case N, Xie Z, Sen B, Styner M, Zou M, O'Conor C, Horowitz M, Rubin J. Mechanical activation of β-catenin regulates phenotype in adult murine marrow-derived mesenchymal stem cells. J Orthop Res 2010; 28:1531-8. [PMID: 20872592 PMCID: PMC3046385 DOI: 10.1002/jor.21156] [Citation(s) in RCA: 62] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Regulation of skeletal remodeling appears to influence the differentiation of multipotent mesenchymal stem cells (MSC) resident in the bone marrow. As murine marrow cultures are contaminated with hematopoietic cells, they are problematic for studying direct effects of mechanical input. Here we use a modified technique to isolate marrow-derived MSC (mdMSC) from adult mice, yielding a population able to differentiate into adipogenic and osteogenic phenotypes that is devoid of hematopoietic cells. In pure mdMSC populations, a daily strain regimen inhibited adipogenic differentiation, suppressing expression of PPARγ and adiponectin. Strain increased β-catenin and inhibition of adipogenesis required this effect. Under osteogenic conditions, strain activated β-catenin signaling and increased expression of WISP1 and COX2. mdMSC were also generated from mice lacking caveolin-1, a protein known to sequester β-catenin: caveolin-1((-/-)) mdMSC exhibited retarded differentiation along both adipogenic and osteogenic lineages but retained mechanical responses that involved β-catenin activation. Interestingly, caveolin-1((-/-)) mdMSC failed to express bone sialoprotein and did not form mineralized nodules. In summary, mdMSC from adult mice respond to both soluble factors and mechanical input, with mechanical activation of β-catenin influencing phenotype. As such, these cells offer a useful model for studies of direct mechanical regulation of MSC differentiation and function.
Collapse
Affiliation(s)
- Natasha Case
- Department of Medicine, University of North Carolina, Chapel Hill, North Carolina 27599, USA.
| | | | | | | | | | | | | | | |
Collapse
|