1
|
Kim SH, Shin HL, Son TH, Kim D, Kim HG, Cho JH, Choi SW. The Biphasic Activity of Auricularia Auricula-Judae Extract on Bone Homeostasis through Inhibition of Osteoclastogenesis and Modulation of Osteogenic Activity. J Microbiol Biotechnol 2024; 34:2576-2585. [PMID: 39467699 PMCID: PMC11729361 DOI: 10.4014/jmb.2408.08055] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2024] [Revised: 10/08/2024] [Accepted: 10/13/2024] [Indexed: 10/30/2024]
Abstract
Osteoporosis arises from the disturbance of bone homeostasis, a process regulated by osteoblasts and osteoclasts. The treatment and prevention of bone metabolic disorders resulting from an imbalance in bone homeostasis require the use of agents that effectively promote both bone formation and anti-resorptive effects. Therefore, an investigation was carried out to determine the potential of the edible mushroom Auricularia auricula-judae in modulating bone remodeling by inhibiting RANKL-induced osteoclastogenesis and enhancing BMP-2-stimulated osteoblast differentiation. Moreover, this study assessed the mode of action of the Auricularia auricula-judae extracts. The staining of tartrate-resistant acid phosphatase (TRAP), a marker for osteoclast activity, demonstrated that Auricularia auricula-judae water extract (AAJWE) inhibited the formation of multinucleated osteoclasts while exhibiting no cytotoxic effects. The study demonstrated that AAJWE reduced RANKL-induced osteoclast differentiation by inhibiting c-Fos/NFATc1 through the inhibition of ERK and JNK phosphorylation during the RANKL-induced osteoclast differentiation. Moreover, AAJWE exhibited a dose-dependent induction of ALP expression in the presence of BMP-2 during osteoblast differentiation. The AAJWE strengthened BMP-2-induced osteogenesis through the activation of Runx2 and Smad phosphorylation. Therefore, AAJWE emerges as a promising candidate for both prevention and therapy owing to its biphasic effect, which aids in the preservation of bone homeostasis.
Collapse
Affiliation(s)
- Shin-Hye Kim
- Forest Biomaterials Research Center, National Institute of Forest Science (NIFoS), Korea Forest Service (KFS), Jinju 52817, Republic of Korea
| | - Hye-Lim Shin
- Forest Biomaterials Research Center, National Institute of Forest Science (NIFoS), Korea Forest Service (KFS), Jinju 52817, Republic of Korea
- Department of Biological Sciences, Jeonbuk National University, Jeonju 54896, Republic of Korea
| | - Tae Hyun Son
- Forest Biomaterials Research Center, National Institute of Forest Science (NIFoS), Korea Forest Service (KFS), Jinju 52817, Republic of Korea
| | - Dongsoo Kim
- Forest Biomaterials Research Center, National Institute of Forest Science (NIFoS), Korea Forest Service (KFS), Jinju 52817, Republic of Korea
| | - Hwan-Gyu Kim
- Department of Biological Sciences, Jeonbuk National University, Jeonju 54896, Republic of Korea
| | - Jae-Han Cho
- Postharvest Research Division, National Institute of Horticultural and Herbal Science (NIHHS), Rural Development Administration (RDA), Wanju 55365, Republic of Korea
| | - Sik-Won Choi
- Forest Biomaterials Research Center, National Institute of Forest Science (NIFoS), Korea Forest Service (KFS), Jinju 52817, Republic of Korea
| |
Collapse
|
2
|
Zhang Y, Feng X, Zheng B, Liu Y. Regulation and mechanistic insights into tensile strain in mesenchymal stem cell osteogenic differentiation. Bone 2024; 187:117197. [PMID: 38986825 DOI: 10.1016/j.bone.2024.117197] [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: 04/12/2024] [Revised: 06/24/2024] [Accepted: 07/07/2024] [Indexed: 07/12/2024]
Abstract
Bone marrow mesenchymal stem cells (BMSCs) are integral to bone remodeling and homeostasis, as they are capable of differentiating into osteogenic and adipogenic lineages. This differentiation is substantially influenced by mechanosensitivity, particularly to tensile strain, which is a prevalent mechanical stimulus known to enhance osteogenic differentiation. This review specifically examines the effects of various cyclic tensile stress (CTS) conditions on BMSC osteogenesis. It delves into the effects of different loading devices, magnitudes, frequencies, elongation levels, dimensionalities, and coculture conditions, providing a comparative analysis that aids identification of the most conducive parameters for the osteogenic differentiation of BMSCs. Subsequently, this review delineates the signaling pathways activated by CTS, such as Wnt/β-catenin, BMP, Notch, MAPK, PI3K/Akt, and Hedgehog, which are instrumental in mediating the osteogenic differentiation of BMSCs. Through a detailed examination of these pathways, this study elucidates the intricate mechanisms whereby tensile strain promotes osteogenic differentiation, offering valuable guidance for optimizing therapeutic strategies aimed at enhancing bone regeneration.
Collapse
Affiliation(s)
- Yongxin Zhang
- Department of Orthodontics, School and Hospital of Stomatology, Liaoning Provincial Key Laboratory of Oral Disease, China Medical University, Shenyang 110002, China; Shenyang Clinical Medical Research Center of Orthodontic Disease, China
| | - Xu Feng
- Department of Orthodontics, School and Hospital of Stomatology, Liaoning Provincial Key Laboratory of Oral Disease, China Medical University, Shenyang 110002, China; Shenyang Clinical Medical Research Center of Orthodontic Disease, China
| | - Bowen Zheng
- Department of Orthodontics, School and Hospital of Stomatology, Liaoning Provincial Key Laboratory of Oral Disease, China Medical University, Shenyang 110002, China; Shenyang Clinical Medical Research Center of Orthodontic Disease, China.
| | - Yi Liu
- Department of Orthodontics, School and Hospital of Stomatology, Liaoning Provincial Key Laboratory of Oral Disease, China Medical University, Shenyang 110002, China; Shenyang Clinical Medical Research Center of Orthodontic Disease, China.
| |
Collapse
|
3
|
Yadav P, Bandyopadhayaya S, Soni S, Saini S, Sharma LK, Shrivastava SK, Mandal CC. Simvastatin prevents BMP-2 driven cell migration and invasion by suppressing oncogenic DNMT1 expression in breast cancer cells. Gene 2023; 882:147636. [PMID: 37442305 DOI: 10.1016/j.gene.2023.147636] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2023] [Revised: 07/08/2023] [Accepted: 07/10/2023] [Indexed: 07/15/2023]
Abstract
Both epigenetic and genetic changes in the cancer genome act simultaneously to promote tumor development and metastasis. Aberrant DNA methylation, a prime epigenetic event, is often observed in various cancer types. The elevated DNA methyltransferase 1 (DNMT1) enzyme creates DNA hypermethylation at CpG islands to drive oncogenic potential. This study emphasized to decipher the molecular mechanism of endogenous regulation of DNMT1 expression for finding upstream signaling molecules. Cancer database analyses found an upregulated DNMT1 expression in most cancer types including breast cancer. Overexpression of DNMT1 showed an increased cell migration, invasion, and stemness potential whereas 5-azacytidine (DNMT1 inhibitor) and siRNA mediated knockdown of DNMT1 exhibited inhibition of such cancer activities in breast cancer MDA-MB-231 and MCF-7 cells. Infact, cancer database analyses further found a positive correlation of DNMT1 transcript with both cholesterol pathway regulatory genes and BMP signaling molecules. Experimental observations documented that the cholesterol-lowering drug, simvastatin decreased DNMT1 transcript as well as protein, whereas BMP-2 treatment increased DNMT1 expression in breast cancer cells. In addition, expression of various key cholesterol regulatory genes was found to be upregulated in response to BMP-2 treatment. Moreover, simvastatin inhibited BMP-2 induced DNMT1 expression in breast cancer cells. Thus, this study for the first time reveals that both BMP-2 signaling and cholesterol pathways could regulate endogenous DNMT1 expression in cancer cells.
Collapse
Affiliation(s)
- Pooja Yadav
- Department of Biochemistry, School of Life Sciences, Central University of Rajasthan, Ajmer 305817, Rajasthan, India
| | - Shreetama Bandyopadhayaya
- Department of Biochemistry, School of Life Sciences, Central University of Rajasthan, Ajmer 305817, Rajasthan, India
| | - Sneha Soni
- Department of Biochemistry, School of Life Sciences, Central University of Rajasthan, Ajmer 305817, Rajasthan, India
| | - Sunil Saini
- Department of Biophysics, All India Institute of Medical Sciences, New Delhi, India
| | - Lokendra K Sharma
- Department of Molecular Medicine and Biotechnology, Sanjay Gandhi Post Graduate Institute of Medical Sciences, Lucknow, U.P., India
| | - Sandeep K Shrivastava
- Centre for Innovation, Research & Development, Dr. B. Lal Clinical Laboratory Pvt Ltd. Jaipur, Rajasthan, India
| | - Chandi C Mandal
- Department of Biochemistry, School of Life Sciences, Central University of Rajasthan, Ajmer 305817, Rajasthan, India.
| |
Collapse
|
4
|
Reyes Fernandez PC, Wright CS, Farach-Carson MC, Thompson WR. Examining Mechanisms for Voltage-Sensitive Calcium Channel-Mediated Secretion Events in Bone Cells. Calcif Tissue Int 2023; 113:126-142. [PMID: 37261463 PMCID: PMC11008533 DOI: 10.1007/s00223-023-01097-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Accepted: 05/16/2023] [Indexed: 06/02/2023]
Abstract
In addition to their well-described functions in cell excitability, voltage-sensitive calcium channels (VSCCs) serve a critical role in calcium (Ca2+)-mediated secretion of pleiotropic paracrine and endocrine factors, including those produced in bone. Influx of Ca2+ through VSCCs activates intracellular signaling pathways to modulate a variety of cellular processes that include cell proliferation, differentiation, and bone adaptation in response to mechanical stimuli. Less well understood is the role of VSCCs in the control of bone and calcium homeostasis mediated through secreted factors. In this review, we discuss the various functions of VSCCs in skeletal cells as regulators of Ca2+ dynamics and detail how these channels might control the release of bioactive factors from bone cells. Because VSCCs are druggable, a better understanding of the multiple functions of these channels in the skeleton offers the opportunity for developing new therapies to enhance and maintain bone and to improve systemic health.
Collapse
Affiliation(s)
- Perla C Reyes Fernandez
- Department of Physical Therapy, School of Health and Human Sciences, Indiana University, Indianapolis, IN, 46202, USA
- Center for Musculoskeletal Health, Indiana University, Indianapolis, IN, 46202, USA
| | - Christian S Wright
- Department of Physical Therapy, School of Health and Human Sciences, Indiana University, Indianapolis, IN, 46202, USA
- Center for Musculoskeletal Health, Indiana University, Indianapolis, IN, 46202, USA
| | - Mary C Farach-Carson
- Department of Diagnostic and Biomedical Sciences, School of Dentistry, The University of Texas Health Science Center at Houston, Houston, TX, 77054, USA
- Departments of BioSciences and Bioengineering, Rice University, Houston, TX, 77005, USA
| | - William R Thompson
- Department of Physical Therapy, School of Health and Human Sciences, Indiana University, Indianapolis, IN, 46202, USA.
- Center for Musculoskeletal Health, Indiana University, Indianapolis, IN, 46202, USA.
- Department of Anatomy, Cell Biology and Physiology, School of Medicine, Indiana University, Indianapolis, IN, 46202, USA.
| |
Collapse
|
5
|
BMP Signaling Pathway in Dentin Development and Diseases. Cells 2022; 11:cells11142216. [PMID: 35883659 PMCID: PMC9317121 DOI: 10.3390/cells11142216] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Revised: 07/08/2022] [Accepted: 07/12/2022] [Indexed: 12/27/2022] Open
Abstract
BMP signaling plays an important role in dentin development. BMPs and antagonists regulate odontoblast differentiation and downstream gene expression via canonical Smad and non-canonical Smad signaling pathways. The interaction of BMPs with their receptors leads to the formation of complexes and the transduction of signals to the canonical Smad signaling pathway (for example, BMP ligands, receptors, and Smads) and the non-canonical Smad signaling pathway (for example, MAPKs, p38, Erk, JNK, and PI3K/Akt) to regulate dental mesenchymal stem cell/progenitor proliferation and differentiation during dentin development and homeostasis. Both the canonical Smad and non-canonical Smad signaling pathways converge at transcription factors, such as Dlx3, Osx, Runx2, and others, to promote the differentiation of dental pulp mesenchymal cells into odontoblasts and downregulated gene expressions, such as those of DSPP and DMP1. Dysregulated BMP signaling causes a number of tooth disorders in humans. Mutation or knockout of BMP signaling-associated genes in mice results in dentin defects which enable a better understanding of the BMP signaling networks underlying odontoblast differentiation and dentin formation. This review summarizes the recent advances in our understanding of BMP signaling in odontoblast differentiation and dentin formation. It includes discussion of the expression of BMPs, their receptors, and the implicated downstream genes during dentinogenesis. In addition, the structures of BMPs, BMP receptors, antagonists, and dysregulation of BMP signaling pathways associated with dentin defects are described.
Collapse
|
6
|
Sharma T, Kapoor A, Mandal CC. Duality of bone morphogenetic proteins in cancer: A comprehensive analysis. J Cell Physiol 2022; 237:3127-3163. [DOI: 10.1002/jcp.30785] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Revised: 04/06/2022] [Accepted: 04/29/2022] [Indexed: 11/06/2022]
Affiliation(s)
- Tanu Sharma
- Department of Biochemistry, School of Life Sciences Central University of Rajasthan Ajmer Rajasthan India
| | - Anmol Kapoor
- Department of Biochemistry, School of Life Sciences Central University of Rajasthan Ajmer Rajasthan India
| | - Chandi C. Mandal
- Department of Biochemistry, School of Life Sciences Central University of Rajasthan Ajmer Rajasthan India
| |
Collapse
|
7
|
Roos CM, Zhang B, Hagler MA, Arghami A, Miller JD. MnSOD protects against vascular calcification independent of changes in vascular function in hypercholesterolemic mice. Atherosclerosis 2021; 331:31-37. [PMID: 34147244 DOI: 10.1016/j.atherosclerosis.2021.06.005] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Revised: 05/05/2021] [Accepted: 06/04/2021] [Indexed: 11/16/2022]
Abstract
BACKGROUND AND AIMS The overall goal of this study was to determine the effects of MnSOD-deficiency on vascular structure and function in hypercholesterolemic mice. Previous work suggested that increases in mitochondrial-derived reactive oxygen species (ROS) can exacerbate vascular dysfunction and atherosclerosis. It remains unknown, however, how MnSOD-deficiency and local compensatory mechanisms impact atherosclerotic plaque composition. METHODS AND RESULTS We used a hypercholesterolemic mouse model (ldlr-/-/ApoB100/100; LA), either wild-type for MnSOD (LA-MnSOD+/+) or MnSOD-haploinsufficient (LA-MnSOD+/-), that was fed a western diet for either 3 or 6 months. Consistent with previous reports, reductions of MnSOD did not significantly worsen hypercholesterolemia-induced endothelial dysfunction in the aorta. Critically, dramatic impairment of vascular function with Nox2 inhibition or catalase pretreatment suggested the presence of a significant NO-independent vasodilatory mechanism in LA-MnSOD+/- mice (e.g. H2O2). Despite remarkably well-preserved overall vascular relaxation, loss of mitochondrial antioxidant capacity in LA-MnSOD+/- mice significantly increased osteogenic signalling and vascular calcification compared to the LA-MnSOD+/+ littermates. CONCLUSIONS Collectively, these data are the first to suggest that loss of mitochondrial antioxidant capacity in hypercholesterolemic mice results in dramatic upregulation of NADPH oxidase-derived H2O2. While this appears to be adaptive in the context of preserving overall endothelium-dependent relaxation and vascular function, these increases in ROS appear to be remarkably maladaptive and deleterious in the context of vascular calcification.
Collapse
Affiliation(s)
| | - Bin Zhang
- Departments of Surgery, Mayo Clinic, Rochester, MN, USA
| | | | - Arman Arghami
- Departments of Surgery, Mayo Clinic, Rochester, MN, USA
| | - Jordan D Miller
- Departments of Surgery, Mayo Clinic, Rochester, MN, USA; Physiology & Biomedical Engineering, Mayo Clinic, Rochester, MN, USA.
| |
Collapse
|
8
|
Mandal CC. Osteolytic metastasis in breast cancer: effective prevention strategies. Expert Rev Anticancer Ther 2020; 20:797-811. [PMID: 32772585 DOI: 10.1080/14737140.2020.1807950] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
INTRODUCTION Breast cancer is the most common cancer in women throughout the world. Patients who are diagnosed early generally have better prognosis and survivability. Indeed, advanced stage breast cancer often develops osteolytic metastases, leading to bone destruction. Although there are select drugs available to treat bone metastatic disease, these drugs have shown limited success. AREA COVERED This paper emphasizes updated mechanisms of bone remodeling and osteolytic bone metastases of breast cancer. This article also aims to explore the potential of novel natural and synthetic therapeutics in the effective prevention of breast cancer-induced osteolysis and osteolytic metastases of breast cancer. EXPERT OPINION Targeting TGFβ and BMP signaling pathways, along with osteoclast activity, appears to be a promising therapeutic strategy in the prevention of breast cancer-induced osteolytic bone destruction and metastatic growth at bone metastatic niches. Pilot studies in animal models suggest various natural and synthetic compounds and monoclonal antibodies as putative therapeutics in the prevention of breast cancer stimulated osteolytic activity. However, comprehensive pre-clinical studies demonstrating the PK/PD and in-depth understanding of molecular mechanism(s) by which these potential molecules exhibit anti-tumor growth and anti-osteolytic activity are still required to develop effective therapies against breast cancer-induced osteolytic bone disease.
Collapse
Affiliation(s)
- Chandi C Mandal
- Department of Biochemistry, School of Life Sciences, Central University of Rajasthan , Ajmer, India
| |
Collapse
|
9
|
Sharma T, Mandal CC. Omega-3 fatty acids in pathological calcification and bone health. J Food Biochem 2020; 44:e13333. [PMID: 32548903 DOI: 10.1111/jfbc.13333] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2020] [Revised: 05/21/2020] [Accepted: 05/21/2020] [Indexed: 01/19/2023]
Abstract
Omega-3 fatty acids (ω-3FAs) such as Docosahexaenoic acid (DHA) and Eicosapentanoic acid (EPA), are active ingredient of fish oil, which have larger health benefits against various diseases including cardiovascular, neurodegenerative, cancers and bone diseases. Substantial studies documented a preventive role of omega-3 fatty acids in pathological calcification like vascular calcification and microcalcification in cancer tissues. In parallel, these fatty acids improve bone quality probably by preventing bone decay and augmenting bone mineralization. This study also addresses that the functions of ω-3FAs not only depend on tissue types, but also work through different molecular mechanisms for preventing pathological calcification in various tissues and improving bone health. PRACTICAL APPLICATIONS: Practical applications of the current study are to improve the knowledge about the supplementation of omega-3 fatty acids. This study infers that supplementation of omega-3 fatty acids aids in bone preservation in elder females at the risk of osteoporosis and also, on the contrary, omega-3 fatty acids interfere with pathological calcification of vascular cells and cancer cells. Omega-3 supplementation should be given to the cardiac patients because of its cardio protective role. In line with this, omega-3 supplementation should be included with chemotherapy for cancer patients as it can prevent osteoblastic potential of breast cancer patients, responsible for pathological mineralization, and blocks off target toxicities. Administration of omega-3 fatty acid with chemotherapy will not only improve survival of cancer patients, but also improve the bone quality. Thus, this study allows a better understanding on omega-3 fatty acids in combating pathological complications such as osteoporosis, vascular calcification, and breast microcalcification.
Collapse
Affiliation(s)
- Tanu Sharma
- Department of Biochemistry, School of Life Sciences, Central University of Rajasthan, Ajmer, India
| | - Chandi C Mandal
- Department of Biochemistry, School of Life Sciences, Central University of Rajasthan, Ajmer, India
| |
Collapse
|
10
|
Omi M, Kulkarni AK, Raichur A, Fox M, Uptergrove A, Zhang H, Mishina Y. BMP-Smad Signaling Regulates Postnatal Crown Dentinogenesis in Mouse Molar. JBMR Plus 2020; 4:e10249. [PMID: 32149267 PMCID: PMC7017888 DOI: 10.1002/jbm4.10249] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/05/2019] [Revised: 10/14/2019] [Accepted: 10/24/2019] [Indexed: 12/13/2022] Open
Abstract
Dentinogenesis, a formation of dentin by odontoblasts, is an essential process during tooth development. Bone morphogenetic proteins (BMPs) are one of the most crucial growth factors that contribute to dentin formation. However, it is still unclear how BMP signaling pathways regulate postnatal crown and root dentinogenesis. BMPs transduce signals through canonical Smad and non-Smad signaling pathways including p38 and ERK signaling pathways. To investigate the roles of Smad and non-Smad signaling pathways in dentinogenesis, we conditionally deleted Bmpr1a, which encodes the type 1A receptor for BMPs, to remove both Smad and non-Smad pathways in Osterix-expressing cells. We also expressed a constitutively activated form of Bmpr1a (caBmpr1a) to increase Smad1/5/9 signaling activity without altered non-Smad activity in odontoblasts. To understand the function of BMP signaling during postnatal dentin formation, Cre activity was induced at the day of birth. Our results showed that loss of BmpR1A in odontoblasts resulted in impaired dentin formation and short molar roots at postnatal day 21. Bmpr1a cKO mice displayed a reduction of dentin matrix production compared to controls associated with increased cell proliferation and reduced Osx and Dspp expression. In contrast, caBmpr1a mutant mice that show increased Smad1/5/9 signaling activity resulted in no overt tooth phenotype. To further dissect the functions of each signaling activity, we generated Bmpr1a cKO mice also expressing caBmpr1a to restore only Smad1/5/9 signaling activity. Restoring Smad activity in the compound mutant mice rescued impaired crown dentin formation in the Bmpr1a cKO mice; however, impaired root dentin formation and short roots were not changed. These results suggest that BMP-Smad signaling in odontoblasts is responsible for crown dentin formation, while non-Smad signaling may play a major role in root dentin formation and elongation. © 2019 The Authors. JBMR Plus published by Wiley Periodicals, Inc. on behalf of American Society for Bone and Mineral Research. © 2019 The Authors. JBMR Plus published by Wiley Periodicals, Inc. on behalf of American Society for Bone and Mineral Research.
Collapse
Affiliation(s)
- Maiko Omi
- Department of Biologic and Materials Sciences and ProsthodonticsUniversity of Michigan School of DentistryAnn ArborMIUSA
| | - Anshul K Kulkarni
- Department of Biologic and Materials Sciences and ProsthodonticsUniversity of Michigan School of DentistryAnn ArborMIUSA
| | - Anagha Raichur
- Department of Biologic and Materials Sciences and ProsthodonticsUniversity of Michigan School of DentistryAnn ArborMIUSA
| | - Mason Fox
- Department of Biologic and Materials Sciences and ProsthodonticsUniversity of Michigan School of DentistryAnn ArborMIUSA
| | - Amber Uptergrove
- Department of Biologic and Materials Sciences and ProsthodonticsUniversity of Michigan School of DentistryAnn ArborMIUSA
| | - Honghao Zhang
- Department of Biologic and Materials Sciences and ProsthodonticsUniversity of Michigan School of DentistryAnn ArborMIUSA
| | - Yuji Mishina
- Department of Biologic and Materials Sciences and ProsthodonticsUniversity of Michigan School of DentistryAnn ArborMIUSA
| |
Collapse
|
11
|
Dos Santos BP, Garbay B, Fenelon M, Rosselin M, Garanger E, Lecommandoux S, Oliveira H, Amédée J. Development of a cell-free and growth factor-free hydrogel capable of inducing angiogenesis and innervation after subcutaneous implantation. Acta Biomater 2019; 99:154-167. [PMID: 31425892 DOI: 10.1016/j.actbio.2019.08.028] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2019] [Revised: 08/01/2019] [Accepted: 08/15/2019] [Indexed: 12/18/2022]
Abstract
Despite significant progress in the field of biomaterials for bone repair, the lack of attention to the vascular and nervous networks within bone implants could be one of the main reasons for the delayed or impaired recovery of bone defects. The design of innovative biomaterials should improve the host capacity of healing to restore a functional tissue, taking into account that the nerve systems closely interact with blood vessels in the bone tissue. The aim of this work is to develop a cell-free and growth factor-free hydrogel capable to promote angiogenesis and innervation. To this end, we have used elastin-like polypeptides (ELPs), poly(ethylene glycol) (PEG) and increasing concentrations of the adhesion peptide IKVAV (25% (w/w) representing 1.7 mM and 50% (w/w) representing 4.1 mM) to formulate and produce hydrogels. When characterized in vitro, hydrogels have fine-tunable rheological properties, microporous structure and are biocompatible. At the biological level, 50% IKVAV composition up-regulated Runx2, Osx, Spp1, Vegfa and Bmp2 in mesenchymal stromal cells and Tek in endothelial cells, and sustained the formation of long neurites in sensory neurons. When implanted subcutaneously in mice, hydrogels induced no signals of major inflammation and the 50% IKVAV composition induced higher vessel density and formation of nervous terminations in the peripheral tissue. This novel composite has important features for tissue engineering, showing higher osteogenic, angiogenic and innervation potential in vitro, being not inflammatory in vivo, and inducing angiogenesis and innervation subcutaneously. STATEMENT OF SIGNIFICANCE: One of the main limitations in the field of tissue engineering remains the sufficient vascularization and innervation during tissue repair. In this scope, the development of advanced biomaterials that can support these processes is of crucial importance. Here, we formulated different compositions of Elastin-like polypeptide-based hydrogels bearing the IKVAV adhesion sequence. These compositions showed controlled mechanical properties, and were degradable in vitro. Additionally, we could identify in vitro a composition capable to promote neurite formation and to modulate endothelial and mesenchymal stromal cells gene expression, in view of angiogenesis and osteogenesis, respectively. When tested in vivo, it showed no signs of major inflammation and induced the formation of a highly vascularized and innervated neotissue. In this sense, our approach represents a potential advance in the development of new strategies to promote tissue regeneration, taking into account both angiogenesis and innervation.
Collapse
Affiliation(s)
- Bruno Paiva Dos Santos
- Tissue Bioengineering Laboratory (BioTis), Inserm U1026, University of Bordeaux, Bordeaux, France.
| | - Bertrand Garbay
- Univ. Bordeaux, CNRS, Bordeaux INP, LCPO, UMR 5629, F-33600 Pessac, France
| | - Mathilde Fenelon
- Tissue Bioengineering Laboratory (BioTis), Inserm U1026, University of Bordeaux, Bordeaux, France; CHU Bordeaux, Department of Oral Surgery, F-33076 Bordeaux, France
| | - Marie Rosselin
- Univ. Bordeaux, CNRS, Bordeaux INP, LCPO, UMR 5629, F-33600 Pessac, France
| | - Elisabeth Garanger
- Univ. Bordeaux, CNRS, Bordeaux INP, LCPO, UMR 5629, F-33600 Pessac, France
| | | | - Hugo Oliveira
- Tissue Bioengineering Laboratory (BioTis), Inserm U1026, University of Bordeaux, Bordeaux, France
| | - Joëlle Amédée
- Tissue Bioengineering Laboratory (BioTis), Inserm U1026, University of Bordeaux, Bordeaux, France
| |
Collapse
|
12
|
Takeno A, Kanazawa I, Tanaka KI, Notsu M, Sugimoto T. Phloretin Suppresses Bone Morphogenetic Protein-2-Induced Osteoblastogenesis and Mineralization via Inhibition of Phosphatidylinositol 3-kinases/Akt Pathway. Int J Mol Sci 2019; 20:ijms20102481. [PMID: 31137461 PMCID: PMC6566987 DOI: 10.3390/ijms20102481] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2019] [Revised: 05/15/2019] [Accepted: 05/16/2019] [Indexed: 12/15/2022] Open
Abstract
Phloretin has pleiotropic effects, including glucose transporter (GLUT) inhibition. We previously showed that phloretin promoted adipogenesis of bone marrow stromal cell (BMSC) line ST2 independently of GLUT1 inhibition. This study investigated the effect of phloretin on osteoblastogenesis of ST2 cells and osteoblastic MC3T3-E1 cells. Treatment with 10 to 100 µM phloretin suppressed mineralization and expression of osteoblast differentiation markers, such as alkaline phosphatase (ALP), osteocalcin (OCN), type 1 collagen, runt-related transcription factor 2 (Runx2), and osterix (Osx), while increased adipogenic markers, peroxisome proliferator-activated receptor γ (PPARγ), CCAAT/enhancer-binding protein α (C/EBPα), fatty acid-binding protein 4, and adiponectin. Phloretin also inhibited mineralization and decreased osteoblast differentiation markers of MC3T3-E1 cells. Phloretin suppressed phosphorylation of Akt in ST2 cells. In addition, treatment with a phosphatidylinositol 3-kinase (PI3K)/Akt inhibitor, LY294002, suppressed the mineralization and the expression of osteoblast differentiation markers other than ALP. GLUT1 silencing by siRNA did not affect mineralization, although it decreased the expression of OCN and increased the expression of ALP, Runx2, and Osx. The effects of GLUT1 silencing on osteoblast differentiation markers and mineralization were inconsistent with those of phloretin. Taken together, these findings suggest that phloretin suppressed osteoblastogenesis of ST2 and MC3T3-E1 cells by inhibiting the PI3K/Akt pathway, suggesting that the effects of phloretin may not be associated with glucose uptake inhibition.
Collapse
Affiliation(s)
- Ayumu Takeno
- Internal Medicine 1, Shimane University Faculty of Medicine, 89-1, Enya-cho, Izumo, Shimane 693-8501, Japan.
| | - Ippei Kanazawa
- Internal Medicine 1, Shimane University Faculty of Medicine, 89-1, Enya-cho, Izumo, Shimane 693-8501, Japan.
| | - Ken-Ichiro Tanaka
- Internal Medicine 1, Shimane University Faculty of Medicine, 89-1, Enya-cho, Izumo, Shimane 693-8501, Japan.
| | - Masakazu Notsu
- Internal Medicine 1, Shimane University Faculty of Medicine, 89-1, Enya-cho, Izumo, Shimane 693-8501, Japan.
| | - Toshitsugu Sugimoto
- Internal Medicine 1, Shimane University Faculty of Medicine, 89-1, Enya-cho, Izumo, Shimane 693-8501, Japan.
| |
Collapse
|
13
|
Choi YH, Han Y, Jin SW, Lee GH, Kim GS, Lee DY, Chung YC, Lee KY, Jeong HG. Pseudoshikonin I enhances osteoblast differentiation by stimulating Runx2 and Osterix. J Cell Biochem 2018; 119:748-757. [PMID: 28657691 DOI: 10.1002/jcb.26238] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2017] [Accepted: 06/26/2017] [Indexed: 12/24/2022]
Abstract
Pseudoshikonin I (PSI), a novel biomaterial isolated from Lithospermi radix, has been recognized as an herbal medicine for the treatment of infectious and inflammatory diseases. Bone remodeling maintains a balance through bone resorption (osteoclastogenesis) and bone formation (osteoblastogenesis). Bone formation is generally attributed to osteoblasts. However, the effects of PSI on the bone are not well known. In this study, we found that the ethanol extracts of PSI induced osteoblast differentiation by increasing the expression of bone morphogenic protein 4 (BMP 4). PSI positively regulates the transcriptional expression and osteogenic activity of osteoblast-specific transcription factors such as Runx2 and Osterix. To identify the signaling pathways that mediate PSI-induced osteoblastogenesis, we examined the effects of serine-threonine kinase inhibitors that are known regulators of Osterix and Runx2. PSI-induced upregulation of Osterix and Runx2 was suppressed by treatment with AKT and PKA inhibitors. These results suggest that PSI enhances osteoblast differentiation by stimulating Osterix and Runx2 via the AKT and PKA signaling pathways. Thus, the activation of Runx2 and Osterix is modulated by PSI, thereby demonstrating its potential as a treatment target for bone disease.
Collapse
Affiliation(s)
- You Hee Choi
- College of Pharmacy, Chonnam National University, Gwangju, Republic of Korea
| | - Younho Han
- College of Pharmacy, Chonnam National University, Gwangju, Republic of Korea
- Department of Oral Pharmacology, College of Dentistry, Wonkwang University, Iksan, Republic of Korea
| | - Sun Woo Jin
- College of Pharmacy, Chungnam National University, Daejeon, Republic of Korea
| | - Gi Ho Lee
- College of Pharmacy, Chungnam National University, Daejeon, Republic of Korea
| | - Geum Soog Kim
- Department of Herbal Crop Research, National Institute of Horticultural and Herbal Science, RDA, Eumseong, Republic of Korea
| | - Dae Young Lee
- Department of Herbal Crop Research, National Institute of Horticultural and Herbal Science, RDA, Eumseong, Republic of Korea
| | - Young Chul Chung
- Department of Food Science, International University of Korea, Jinju, Republic of Korea
| | - Kwang Youl Lee
- College of Pharmacy, Chonnam National University, Gwangju, Republic of Korea
| | - Hye Gwang Jeong
- College of Pharmacy, Chungnam National University, Daejeon, Republic of Korea
| |
Collapse
|
14
|
Sepulveda H, Villagra A, Montecino M. Tet-Mediated DNA Demethylation Is Required for SWI/SNF-Dependent Chromatin Remodeling and Histone-Modifying Activities That Trigger Expression of the Sp7 Osteoblast Master Gene during Mesenchymal Lineage Commitment. Mol Cell Biol 2017; 37:e00177-17. [PMID: 28784721 PMCID: PMC5615189 DOI: 10.1128/mcb.00177-17] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2017] [Revised: 05/15/2017] [Accepted: 07/22/2017] [Indexed: 12/22/2022] Open
Abstract
Here we assess histone modification, chromatin remodeling, and DNA methylation processes that coordinately control the expression of the bone master transcription factor Sp7 (osterix) during mesenchymal lineage commitment in mammalian cells. We find that Sp7 gene silencing is mediated by DNA methyltransferase1/3 (DNMT1/3)-, histone deacetylase 1/2/4 (HDAC1/2/4)-, Setdb1/Suv39h1-, and Ezh1/2-containing complexes. In contrast, Sp7 gene activation involves changes in histone modifications, accompanied by decreased nucleosome enrichment and DNA demethylation mediated by SWI/SNF- and Tet1/Tet2-containing complexes, respectively. Inhibition of DNA methylation triggers changes in the histone modification profile and chromatin-remodeling events leading to Sp7 gene expression. Tet1/Tet2 silencing prevents Sp7 expression during osteoblast differentiation as it impairs DNA demethylation and alters the recruitment of histone methylase (COMPASS)-, histone demethylase (Jmjd2a/Jmjd3)-, and SWI/SNF-containing complexes to the Sp7 promoter. The dissection of these interconnected epigenetic mechanisms that govern Sp7 gene activation reveals a hierarchical process where regulatory components mediating DNA demethylation play a leading role.
Collapse
Affiliation(s)
- Hugo Sepulveda
- Center for Biomedical Research, Faculty of Biological Sciences and Faculty of Medicine, Universidad Andres Bello, Santiago, Chile
- FONDAP Center for Genome Regulation, Faculty of Biological Sciences and Faculty of Medicine, Universidad Andres Bello, Santiago, Chile
| | - Alejandro Villagra
- Department of Biochemistry and Molecular Medicine, School of Medicine and Health Sciences, The George Washington University, Washington, DC, USA
| | - Martin Montecino
- Center for Biomedical Research, Faculty of Biological Sciences and Faculty of Medicine, Universidad Andres Bello, Santiago, Chile
- FONDAP Center for Genome Regulation, Faculty of Biological Sciences and Faculty of Medicine, Universidad Andres Bello, Santiago, Chile
| |
Collapse
|
15
|
Chowdhury K, Sharma A, Kumar S, Gunjan GK, Nag A, Mandal CC. Colocynth Extracts Prevent Epithelial to Mesenchymal Transition and Stemness of Breast Cancer Cells. Front Pharmacol 2017; 8:593. [PMID: 28928657 PMCID: PMC5591978 DOI: 10.3389/fphar.2017.00593] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2017] [Accepted: 08/16/2017] [Indexed: 12/12/2022] Open
Abstract
Modern treatment strategies provide better overall survival in cancer patients, primarily by controlling tumor growth. However, off-target and systemic toxicity, tumor recurrence, and resistance to therapy are still inadvertent hurdles in current treatment regimens. Similarly, metastasis is another deadly threat to patients suffering from cancer. This has created an urgent demand to come up with new drugs having anti-metastatic potential and minimum side effects. Thus, this study was aimed at exploring the anti-proliferative and anti-metastatic potential of colocynth medicinal plant. Results from MTT assay, morphological visualization of cells and scratch assay indicated a role of ethanol and acetone extracts of fruit pulp of the colocynth plant in inhibiting cell viability, enhancing cell cytotoxicity and preventing cell migration in various cancer cell types, including breast cancer cell lines MCF-7 and MDA-MB-231, and cervical cancer cell line SiHa, subsequently having a low cytotoxic effect on mononuclear PBMC and macrophage J774A cells. Our study in metastatic MDA-MB-231 cells showed that both ethanol and acetone pulp extracts decreased transcript levels of the anti-apoptotic genes BCL2 and BCLXL, and a reverse effect was observed for the pro-apoptotic genes BAX and caspase 3. Additionally, enhanced caspase 3 activity and downregulated BCL2 protein were seen, indicating a role of these extracts in inducing apoptotic activity. Moreover, MDA-MB-231 cells treated with both these extracts demonstrated up-regulation of the epithelial gene keratin 19 and down-regulation of the mesenchymal genes, vimentin, N-cadherin, Zeb1 and Zeb2 compared to control, suggesting a suppressive impact of these extracts in epithelial to mesenchymal transition (EMT). In addition, these extracts inhibited colony and sphere formation with simultaneous reduction in the transcript level of the stemness associated genes, BMI-1 and CD44. It was also found that both the plant extracts exhibited synergistic potential with the chemotherapeutic drug doxorubicin to inhibit cancer viability. Furthermore, GC-MS/MS analysis revealed the presence of certain novel compounds in both the extracts that are responsible for the anti-cancer role of the extracts. Overall, the results of this report suggest, for the first time, that colocynth fruit pulp extracts may block the proliferative as well as metastatic activity of breast cancer cells.
Collapse
Affiliation(s)
- Kaushik Chowdhury
- Department of Biochemistry, School of Life Sciences, Central University of RajasthanAjmer, India
| | - Ankit Sharma
- Department of Biochemistry, School of Life Sciences, Central University of RajasthanAjmer, India
| | - Suresh Kumar
- Department of Biochemistry, School of Life Sciences, Central University of RajasthanAjmer, India
| | - Gyanesh K Gunjan
- Department of Biochemistry, School of Life Sciences, Central University of RajasthanAjmer, India
| | - Alo Nag
- Department of Biochemistry, University of DelhiNew Delhi, India
| | - Chandi C Mandal
- Department of Biochemistry, School of Life Sciences, Central University of RajasthanAjmer, India
| |
Collapse
|
16
|
Chowdhury K, Sharma A, Sharma T, Kumar S, Mandal CC. Simvastatin and MBCD Inhibit Breast Cancer-Induced Osteoclast Activity by Targeting Osteoclastogenic Factors. Cancer Invest 2017; 35:403-413. [PMID: 28463564 DOI: 10.1080/07357907.2017.1309548] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Previous reports have documented that cholesterol-lowering simvastatin prevented osteolytic metastasis of breast cancer in animal model in which cancer cells were placed into blood circulation. Thus, simvastatin treatment might have a preventive effect in inhibiting osteoclast activity of metastatic bone microenvironment. This study documented that both simvastatin and MBCD (cholesterol depleting drug) blocked the breast cancer-induced TRAP and MMP activity, and expressions of various osteoclastogenic genes (TRAP, Cathepsin K, and NFATc1) in pre-osteoclast RAW264.7 cells, and osteoclastogenic CSF-1 and RANKL expressions in breast cancer MCF-7 cells. Thus, these findings unravel a molecular mechanism of simvastatin-/MBCD-mediated inhibition of breast cancer-driven osteoclast activity.
Collapse
Affiliation(s)
- Kaushik Chowdhury
- a Department of Biochemistry, School of Life Sciences , Central University of Rajasthan , Ajmer , India
| | - Ankit Sharma
- a Department of Biochemistry, School of Life Sciences , Central University of Rajasthan , Ajmer , India
| | - Tanu Sharma
- a Department of Biochemistry, School of Life Sciences , Central University of Rajasthan , Ajmer , India
| | - Suresh Kumar
- a Department of Biochemistry, School of Life Sciences , Central University of Rajasthan , Ajmer , India
| | - Chandi C Mandal
- a Department of Biochemistry, School of Life Sciences , Central University of Rajasthan , Ajmer , India
| |
Collapse
|
17
|
Furue K, Sena K, Sakoda K, Nakamura T, Noguchi K. Involvement of the phosphoinositide 3-kinase/Akt signaling pathway in bone morphogenetic protein 9-stimulated osteogenic differentiation and stromal cell-derived factor 1 production in human periodontal ligament fibroblasts. Eur J Oral Sci 2017; 125:119-126. [DOI: 10.1111/eos.12336] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/28/2016] [Indexed: 12/23/2022]
Affiliation(s)
- Kirara Furue
- Department of Periodontology; Kagoshima University Graduate School of Medical and Dental Sciences; Kagoshima Japan
| | - Kotaro Sena
- Department of Periodontology; Kagoshima University Graduate School of Medical and Dental Sciences; Kagoshima Japan
| | - Kenji Sakoda
- Department of Periodontology; Kagoshima University Graduate School of Medical and Dental Sciences; Kagoshima Japan
| | - Toshiaki Nakamura
- Department of Periodontology; Kagoshima University Graduate School of Medical and Dental Sciences; Kagoshima Japan
| | - Kazuyuki Noguchi
- Department of Periodontology; Kagoshima University Graduate School of Medical and Dental Sciences; Kagoshima Japan
| |
Collapse
|
18
|
Lauzon MA, Drevelle O, Daviau A, Faucheux N. Effects of BMP-9 and BMP-2 on the PI3K/Akt Pathway in MC3T3-E1 Preosteoblasts. Tissue Eng Part A 2016; 22:1075-85. [PMID: 27477105 DOI: 10.1089/ten.tea.2016.0151] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
The bone morphogenetic proteins (BMPs), which are involved in bone formation and repair, play an important role in tissue engineering. For example, BMP-9 and BMP-2, which are members of different BMP subfamilies, are osteoinductive factors. However, several studies have recently shown that BMP-9 is more osteogenic than BMP-2. We have previously shown that fetal bovine serum (FBS) strongly enhances the osteoblast differentiation of murine preosteoblasts (MC3T3-E1) to BMP-9 but not to BMP-2. This effect is mimicked by IGF-2, which primarily activates the PI3K/Akt pathway, but how Akt phosphorylation sites are implicated in such differentiation is unclear. The effects of BMP-9 and BMP-2 with or without FBS or IGF-2 on Akt phosphorylation sites and subsequent osteoblastic differentiation were determined, respectively, by western blot analysis and alkaline phosphatase activity measurements. The involvement of phosphorylated Akt at Thr308 and/or Ser473 on BMP-mediated osteoblast differentiation was further studied using specific inhibitors. In MC3T3-E1 incubated with or without FBS, BMP-9 and BMP-2 activate Akt on Ser473 and Thr308 very differently in a time and dose-dependent manner. Using inhibitors specific to each Akt phosphorylation site, we showed that both Ser473 and Thr308 must be phosphorylated for BMP-9 and/or IGF-2-induced osteoblast differentiation, whereas BMP-2 requires phosphorylation of only Ser473. Furthermore, cells stimulated with BMP-2 in the presence of FBS require the phosphorylation of Akt at Ser473 and the dephosphorylation of Akt at Thr308 to increase the osteoblast differentiation with alkaline phosphatase activity similar to that of BMP-9 plus FBS. These results provide a better understanding into how BMP-9 induces osteoblast differentiation and its synergy with IGF-2 at the signaling level. This knowledge is essential for preparing the serum-free osteogenic media required for bone tissue engineering or developing growth factor delivery systems to improve bone formation.
Collapse
Affiliation(s)
- Marc-Antoine Lauzon
- 1 Department of Chemical and Biotechnological Engineering, Université de Sherbrooke , Sherbrooke, Canada
| | - Olivier Drevelle
- 1 Department of Chemical and Biotechnological Engineering, Université de Sherbrooke , Sherbrooke, Canada .,2 Department of Chemical Engineering, École Polytechnique de Montréal , Montréal, Canada
| | - Alex Daviau
- 1 Department of Chemical and Biotechnological Engineering, Université de Sherbrooke , Sherbrooke, Canada
| | - Nathalie Faucheux
- 1 Department of Chemical and Biotechnological Engineering, Université de Sherbrooke , Sherbrooke, Canada
| |
Collapse
|
19
|
Sharma T, Radosevich JA, Pachori G, Mandal CC. A Molecular View of Pathological Microcalcification in Breast Cancer. J Mammary Gland Biol Neoplasia 2016; 21:25-40. [PMID: 26769216 DOI: 10.1007/s10911-015-9349-9] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/10/2015] [Accepted: 12/30/2015] [Indexed: 12/11/2022] Open
Abstract
Breast microcalcification is a potential diagnostic indicator for non-palpable breast cancers. Microcalcification type I (calcium oxalate) is restricted to benign tissue, whereas type II (calcium hydroxyapatite) occurs both in benign as well as in malignant lesions. Microcalcification is a pathological complication of the mammary gland. Over the past few decades, much attention has been paid to exploit this property, which forms the basis for advances in diagnostic procedures and imaging techniques. The mechanism of its formation is still poorly understood. Hence, in this paper, we have attempted to address the molecular mechanism of microcalcification in breast cancer. The central theme of this communication is "how a subpopulation of heterogeneous breast tumor cells attains an osteoblast-like phenotype, and what activities drive the process of pathophysiological microcalcification, especially at the invasive or infiltrating front of breast tumors". The role of bone morphogenetic proteins (BMPs) and tumor associated macrophages (TAMs) along with epithelial to mesenchymal transition (EMT) in manipulating this pathological process has been highlighted. Therefore, this review offers a novel insight into the mechanism underlying the development of microcalcification in breast carcinomas.
Collapse
Affiliation(s)
- Tanu Sharma
- Department of Biochemistry, Central University of Rajasthan, NH-8, Bandarsindri, Kishangarh, Ajmer, Rajasthan, 305817, India
| | - James A Radosevich
- Department of Oral Medicine and Diagnostic Sciences, College of Dentistry, University of Illinois at Chicago, Chicago, IL, 60612, USA
| | - Geeta Pachori
- Department of Pathology, J.L.N Medical College, Ajmer, Rajasthan, 305001, India
| | - Chandi C Mandal
- Department of Biochemistry, Central University of Rajasthan, NH-8, Bandarsindri, Kishangarh, Ajmer, Rajasthan, 305817, India.
| |
Collapse
|
20
|
He YD, Sui BD, Li M, Huang J, Chen S, Wu LA. Site-specific function and regulation of Osterix in tooth root formation. Int Endod J 2016; 49:1124-1131. [PMID: 26599722 DOI: 10.1111/iej.12585] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2015] [Accepted: 11/16/2015] [Indexed: 02/06/2023]
Abstract
Congenital diseases of tooth roots, in terms of developmental abnormalities of short and thin root phenotypes, can lead to loss of teeth. A more complete understanding of the genetic molecular pathways and biological processes controlling tooth root formation is required. Recent studies have revealed that Osterix (Osx), a key mesenchymal transcriptional factor participating in both the processes of osteogenesis and odontogenesis, plays a vital role underlying the mechanisms of developmental differences between root and crown. During tooth development, Osx expression has been identified from late embryonic to postnatal stages when the tooth root develops, particularly in odontoblasts and cementoblasts to promote their differentiation and mineralization. Furthermore, the site-specific function of Osx in tooth root formation has been confirmed, because odontoblastic Osx-conditional knockout mice demonstrate primarily short and thin root phenotypes with no apparent abnormalities in the crown (Journal of Bone and Mineral Research 30, 2014 and 742, Journal of Dental Research 94, 2015 and 430). These findings suggest that Osx functions to promote odontoblast and cementoblast differentiation and root elongation only in root, but not in crown formation. Mechanistic research shows regulatory networks of Osx expression, which can be controlled through manipulating the epithelial BMP signalling, mesenchymal Runx2 expression and cellular phosphorylation levels, indicating feasible routes of promoting Osx expression postnatally (Journal of Cellular Biochemistry 114, 2013 and 975). In this regard, a promising approach might be available to regenerate the congenitally diseased root and that regenerative therapy would be the best choice for patients with developmental tooth diseases.
Collapse
Affiliation(s)
- Y D He
- Department of Pediatric Dentistry, School of Stomatology, The Fourth Military Medical University, Xi'an, China.,State Key Laboratory of Military Stomatology, School of Stomatology, The Fourth Military Medical University, Xi'an, China
| | - B D Sui
- State Key Laboratory of Military Stomatology, School of Stomatology, The Fourth Military Medical University, Xi'an, China.,Research and Development Center for Tissue Engineering, School of Stomatology, The Fourth Military Medical University, Xi'an, China
| | - M Li
- State Key Laboratory of Military Stomatology, School of Stomatology, The Fourth Military Medical University, Xi'an, China
| | - J Huang
- Department of Pediatric Dentistry, School of Stomatology, The Fourth Military Medical University, Xi'an, China.,State Key Laboratory of Military Stomatology, School of Stomatology, The Fourth Military Medical University, Xi'an, China.,Department of Anatomy, Histology & Embryology, Basic Medical College, The Fourth Military Medical University, Xi'an, China
| | - S Chen
- Department of Pediatric Dentistry, School of Stomatology, The Fourth Military Medical University, Xi'an, China.,Department of Developmental Dentistry, The University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - L A Wu
- Department of Pediatric Dentistry, School of Stomatology, The Fourth Military Medical University, Xi'an, China.,State Key Laboratory of Military Stomatology, School of Stomatology, The Fourth Military Medical University, Xi'an, China
| |
Collapse
|
21
|
Mandal CC, Das F, Ganapathy S, Harris SE, Choudhury GG, Ghosh-Choudhury N. Bone Morphogenetic Protein-2 (BMP-2) Activates NFATc1 Transcription Factor via an Autoregulatory Loop Involving Smad/Akt/Ca2+ Signaling. J Biol Chem 2015; 291:1148-61. [PMID: 26472929 DOI: 10.1074/jbc.m115.668939] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2015] [Indexed: 11/06/2022] Open
Abstract
Bone remodeling is controlled by dual actions of osteoclasts (OCs) and osteoblasts (OBs). The calcium-sensitive nuclear factor of activated T cells (NFAT) c1 transcription factor, as an OC signature gene, regulates differentiation of OCs downstream of bone morphogenetic protein-2 (BMP-2)-stimulated osteoblast-coded factors. To analyze a functional link between BMP-2 and NFATc1, we analyzed bones from OB-specific BMP-2 knock-out mice for NFATc1 expression by immunohistochemical staining and found significant reduction in NFATc1 expression. This indicated a requirement of BMP-2 for NFATc1 expression in OBs. We showed that BMP-2, via the receptor-specific Smad pathway, regulates expression of NFATc1 in OBs. Phosphatidylinositol 3-kinase/Akt signaling acting downstream of BMP-2 also drives NFATc1 expression and transcriptional activation. Under the basal condition, NFATc1 is phosphorylated. Activation of NFAT requires dephosphorylation by the calcium-dependent serine/threonine phosphatase calcineurin. We examined the role of calcium in BMP-2-stimulated regulation of NFATc1 in osteoblasts. 1,2Bis(2aminophenoxy)ethaneN,N,N',N'-tetraacetic acid acetoxymethyl ester, an inhibitor of intracellular calcium abundance, blocked BMP-2-induced transcription of NFATc1. Interestingly, BMP-2 induced calcium release from intracellular stores and increased calcineurin phosphatase activity, resulting in NFATc1 nuclear translocation. Cyclosporin A, which inhibits calcineurin upstream of NFATc1, blocked BMP-2-induced NFATc1 mRNA and protein expression. Expression of NFATc1 directly increased its transcription and VIVIT peptide, an inhibitor of NFATc1, suppressed BMP-2-stimulated NFATc1 transcription, confirming its autoregulation. Together, these data show a role of NFATc1 downstream of BMP-2 in mouse bone development and provide novel evidence for the presence of a cross-talk among Smad, phosphatidylinositol 3-kinase/Akt, and Ca(2+) signaling for BMP-2-induced NFATc1 expression through an autoregulatory loop.
Collapse
Affiliation(s)
| | | | | | - Stephen E Harris
- Periodontics, University of Texas Health Science Center, San Antonio, Texas 78229
| | - Goutam Ghosh Choudhury
- Medicine, and From Veterans Affairs Research and Geriatric Research Education and Clinical Center, South Texas Veterans Health Care System and
| | | |
Collapse
|
22
|
Mehta J, Asthana S, Mandal CC, Saxena S. A molecular analysis provides novel insights into androgen receptor signalling in breast cancer. PLoS One 2015; 10:e0120622. [PMID: 25781993 PMCID: PMC4364071 DOI: 10.1371/journal.pone.0120622] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2014] [Accepted: 02/05/2015] [Indexed: 01/25/2023] Open
Abstract
BACKGROUND Androgen Receptor (AR) is an essential transcription factor for the development of secondary sex characteristics, spermatogenesis and carcinogenesis. Recently AR has been implicated in the development and progression of breast and prostate cancers. Although some of the functions of the AR are known but the mechanistic details of these divergent processes are still not clear. Therefore understanding the regulatory mechanisms of the functioning of the AR in ER-/AR+ breast cancer will provide many novel targets for the purpose of therapeutic intervention. METHODS/RESULTS Using bioinformatics tools, we have identified 75 AR targets having prominent roles in cell cycle, apoptosis and metabolism. Herein, we validated 10 genes as AR targets by studying the regulation of these genes in MDA-MB-453 cell line on stimulation by androgens like 5α-dihydrotestosterone (DHT), using RT-qPCR and ChIP assay. It was observed that all the identified genes involved in cell cycle except MAD1L1 were found to be up regulated whereas expression of apoptosis related genes was decreased in response to DHT treatment. We performed an exhaustive, rigid-body docking between individual ARE and DNA binding domain (DBD) of the AR protein and it was found that novel residues K567, K588, K591 and R592 are involved in the process of DNA binding. To verify these specific DNA-protein interactions electrostatic energy term calculations for each residue was determined using the linearized Poisson-Boltzmann equation. Our experimental data showed that treatment of breast cancer cells with DHT promotes cell proliferation and decreases apoptosis. It was observed that bicalutamide treatment was able to reverse the effect of DHT. CONCLUSION Taken together, our results provide new insights into the mechanism by which AR promotes breast cancer progression. Moreover our work proposes to use bicalutamide along with taxanes as novel therapy for the treatment of TNBCs, which are positive for downstream AR signalling.
Collapse
Affiliation(s)
- Jatin Mehta
- National Institute of Pathology, ICMR, Safdarjang Hospital, New Delhi, India
| | - Shailendra Asthana
- National Institute of Pathology, ICMR, Safdarjang Hospital, New Delhi, India
| | | | - Sunita Saxena
- National Institute of Pathology, ICMR, Safdarjang Hospital, New Delhi, India
- * E-mail:
| |
Collapse
|
23
|
Lee SH, Jeong HM, Han Y, Cheong H, Kang BY, Lee KY. Prolyl isomerase Pin1 regulates the osteogenic activity of Osterix. Mol Cell Endocrinol 2015; 400:32-40. [PMID: 25463757 DOI: 10.1016/j.mce.2014.11.017] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/31/2014] [Revised: 10/27/2014] [Accepted: 11/20/2014] [Indexed: 02/07/2023]
Abstract
Osterix is an essential transcription factor for osteoblast differentiation and bone formation. The mechanism of regulation of Osterix by post-translational modification remains unknown. Peptidyl-prolyl isomerase 1 (Pin1) catalyzes the isomerization of pSer/Thr-Pro bonds and induces a conformational change in its substrates, subsequently regulating diverse cellular processes. In this study, we demonstrated that Pin1 interacts with Osterix and influences its protein stability and transcriptional activity. This regulation is likely due to the suppression of poly-ubiquitination-mediated proteasomal degradation of Osterix. Collectively, our data demonstrate that Pin1 is a novel regulator of Osterix and may play an essential role in the regulation of osteogenic differentiation.
Collapse
Affiliation(s)
- Sung Ho Lee
- College of Pharmacy & Research Institute of Drug Development, Chonnam National University, Gwangju 500-757, Republic of Korea
| | - Hyung Min Jeong
- College of Pharmacy & Research Institute of Drug Development, Chonnam National University, Gwangju 500-757, Republic of Korea
| | - Younho Han
- College of Pharmacy & Research Institute of Drug Development, Chonnam National University, Gwangju 500-757, Republic of Korea
| | - Heesun Cheong
- Research Institute, National Cancer Center, 323 Ilsan-ro, Ilsandong-gu, Goyang-si, Gyeonggi-do 410-769, Republic of Korea
| | - Bok Yun Kang
- College of Pharmacy & Research Institute of Drug Development, Chonnam National University, Gwangju 500-757, Republic of Korea
| | - Kwang Youl Lee
- College of Pharmacy & Research Institute of Drug Development, Chonnam National University, Gwangju 500-757, Republic of Korea.
| |
Collapse
|
24
|
Mandal CC. High Cholesterol Deteriorates Bone Health: New Insights into Molecular Mechanisms. Front Endocrinol (Lausanne) 2015; 6:165. [PMID: 26557105 PMCID: PMC4617053 DOI: 10.3389/fendo.2015.00165] [Citation(s) in RCA: 69] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/29/2015] [Accepted: 10/08/2015] [Indexed: 01/09/2023] Open
Abstract
Many epidemiological studies show a positive connection between cardiovascular diseases and risk of osteoporosis, suggesting a role of hyperlipidemia and/or hypercholesterolemia in regulating osteoporosis. The majority of the studies indicated a correlation between high cholesterol and high LDL-cholesterol level with low bone mineral density, a strong predictor of osteoporosis. Similarly, bone metastasis is a serious complication of cancer for patients. Several epidemiological and basic studies have established that high cholesterol is associated with increased cancer risk. Moreover, osteoporotic bone environment predisposes the cancer cells for metastatic growth in the bone microenvironment. This review focuses on how cholesterol and cholesterol-lowering drugs (statins) regulate the functions of bone residential osteoblast and osteoclast cells to augment or to prevent bone deterioration. Moreover, this study provides an insight into molecular mechanisms of cholesterol-mediated bone deterioration. It also proposes a potential mechanism by which cellular cholesterol boosts cancer-induced bone metastasis.
Collapse
Affiliation(s)
- Chandi C. Mandal
- Department of Biochemistry, School of Life Sciences, Central University of Rajasthan, Rajasthan, India
- *Correspondence: Chandi C. Mandal,
| |
Collapse
|
25
|
Ghosh-Choudhury N, Mandal CC, Das F, Ganapathy S, Ahuja S, Ghosh Choudhury G. c-Abl-dependent molecular circuitry involving Smad5 and phosphatidylinositol 3-kinase regulates bone morphogenetic protein-2-induced osteogenesis. J Biol Chem 2013; 288:24503-17. [PMID: 23821550 DOI: 10.1074/jbc.m113.455733] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
Skeletal remodeling consists of timely formation and resorption of bone by osteoblasts and osteoclasts in a quantitative manner. Patients with chronic myeloid leukemia receiving inhibitors of c-Abl tyrosine kinase often show reduced bone remodeling due to impaired osteoblast and osteoclast function. BMP-2 plays a significant role in bone generation and resorption by contributing to the formation of mature osteoblasts and osteoclasts. The effects of c-Abl on BMP-2-induced bone remodeling and the underlying mechanisms are not well studied. Using a pharmacological inhibitor and expression of a dominant negative mutant of c-Abl, we show an essential role of this tyrosine kinase in the development of bone nodules containing mature osteoblasts and formation of multinucleated osteoclasts in response to BMP-2. Calvarial osteoblasts prepared from c-Abl null mice showed the absolute requirement of this tyrosine kinase in maturation of osteoblasts and osteoclasts. Activation of phosphatidylinositol 3-kinase (PI 3-kinase)/Akt signaling by BMP-2 leads to osteoblast differentiation. Remarkably, inhibition of c-Abl significantly suppressed BMP-2-stimulated PI 3-kinase activity and its downstream Akt phosphorylation. Interestingly, c-Abl regulated BMP-2-induced osteoclastogenic CSF-1 expression. More importantly, we identified the requirements of c-Abl in BMP-2 autoregulation and the expressions of alkaline phosphatase and osterix that are necessary for osteoblast differentiation. c-Abl contributed to BMP receptor-specific Smad-dependent transcription of CSF-1, osterix, and BMP-2. Finally, c-Abl associates with BMP receptor IA and regulates phosphorylation of Smad in response to BMP-2. We propose that activation of c-Abl is an important step, which induces into two signaling pathways involving noncanonical PI 3-kinase and canonical Smads to integrate BMP-2-induced osteogenesis.
Collapse
Affiliation(s)
- Nandini Ghosh-Choudhury
- Veterans Affairs Research, South Texas Veterans Health Care System, San Antonio, Texas 78229, USA.
| | | | | | | | | | | |
Collapse
|
26
|
Luo Q, Huang Y, Deng X, Zhang J, Li X, Zhao S, Li X. Polyelectrolyte multilayer coating with two regulatory molecules on titanium: construction and its biological effects. Nanomedicine (Lond) 2013; 8:739-55. [PMID: 23384699 DOI: 10.2217/nnm.12.151] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Aim: This study aimed at constructing a novel disulfide-crosslinked collagen I/hyaluronic acid polyelectrolyte multilayer (PEM) coating incorporated with bFGF and arginine–glycine–aspartic acid on titanium via the layer-by-layer technique, and evaluating its biological effects. Materials & methods: The surface topography and components, thickness, degradation behaviors and bFGF release profiles of the PEM coating were investigated. The effects of the PEM coating on osteoprogenitor cell growth and bone implant interfacial binding strength in the rabbit femur model were evaluated separately. Results: The formation of disulfide bonds improved the stability of the PEM coating, resulting in a coating that can release bFGF in a slow and sustained manner. Biological evaluations revealed that the resultant PEM coating on titanium promoted various cell behaviors and enhanced the binding strength. Conclusion: The employed cotreatment regimen enabled bFGF and arginine–glycine–aspartic acid to have a synergistic effect on the cell responses, which, in turn, improved the osseointegration. Original submitted 26 January 2012; Revised submitted 10 June 2012; Published online 5 February 2013
Collapse
Affiliation(s)
- Qiaojie Luo
- Department of Oral & Maxillofacial Surgery, The Affiliated Stomatology Hospital, College of Medicine, Zhejiang University, 395 Yan’an Road, Hangzhou 310006, China
| | - Ying Huang
- Department of Oral & Maxillofacial Surgery, The Affiliated Stomatology Hospital, College of Medicine, Zhejiang University, 395 Yan’an Road, Hangzhou 310006, China
| | - Xuliang Deng
- School & Hospital of Stomatology, Peking University, Beijing 100871, China
| | - Jianxiang Zhang
- Department of Pharmaceutics, College of Pharmacy, Third Military Medical University, Chongqing 400038, China
| | - Xiaohui Li
- Department of Pharmaceutics, College of Pharmacy, Third Military Medical University, Chongqing 400038, China
| | - Shifang Zhao
- Department of Oral & Maxillofacial Surgery, The Affiliated Stomatology Hospital, College of Medicine, Zhejiang University, 395 Yan’an Road, Hangzhou 310006, China
| | - Xiaodong Li
- Department of Oral & Maxillofacial Surgery, The Affiliated Stomatology Hospital, College of Medicine, Zhejiang University, 395 Yan’an Road, Hangzhou 310006, China.
| |
Collapse
|
27
|
McGonnell IM, Grigoriadis AE, Lam EWF, Price JS, Sunters A. A specific role for phosphoinositide 3-kinase and AKT in osteoblasts? Front Endocrinol (Lausanne) 2012; 3:88. [PMID: 22833734 PMCID: PMC3400941 DOI: 10.3389/fendo.2012.00088] [Citation(s) in RCA: 76] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/30/2012] [Accepted: 06/29/2012] [Indexed: 12/25/2022] Open
Abstract
The phosphoinositide 3-kinase and AKT (protein kinase B) signaling pathway (PI3K/AKT) plays a central role in the control of cell survival, growth, and proliferation throughout the body. With regard to bone, and particularly in osteoblasts, there is an increasing amount of evidence that the many signaling molecules exert some of their bone-specific effects in part via selectively activating some of the generic effects of the PI3K/AKT pathway in osteoblasts. There is further data demonstrating that PI3K/AKT has the capacity to specifically cross-talk with other signaling pathways and transcriptional networks controlling bone cells' development in order to fine-tune the osteoblast phenotype. There is also evidence that perturbations in the PI3K/AKT pathway may well be responsible for certain bone pathologies. In this review, we discuss some of these findings and suggest that the PI3K/AKT pathway is a central nexus in the extensive network of extracellular signaling pathways that control the osteoblast.
Collapse
Affiliation(s)
- Imelda M. McGonnell
- Department of Veterinary Basic Sciences, The Royal Veterinary College,London, UK
| | - Agamemnon E. Grigoriadis
- Department of Craniofacial Development and Stem Cell Biology, King’s College London, Guy’s Hospital,London, UK
| | - Eric W.-F. Lam
- Department of Surgery and Cancer, Imperial College London, Hammersmith Hospital,London, UK
| | - Joanna S. Price
- School of Veterinary Sciences, University of Bristol,Bristol, UK
| | - Andrew Sunters
- Department of Veterinary Basic Sciences, The Royal Veterinary College,London, UK
- *Correspondence: Andrew Sunters, Department of Veterinary Basic Sciences, The Royal Veterinary College, Royal College Street, Camden, London NW1 0TU, UK. e-mail:
| |
Collapse
|
28
|
Guntur AR, Rosen CJ, Naski MC. N-cadherin adherens junctions mediate osteogenesis through PI3K signaling. Bone 2012; 50:54-62. [PMID: 21964322 PMCID: PMC3251172 DOI: 10.1016/j.bone.2011.09.036] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/14/2011] [Revised: 08/22/2011] [Accepted: 09/07/2011] [Indexed: 01/18/2023]
Abstract
During endochondral ossification, the cartilage is surrounded by a layer of cells that constitute the perichondrium. Communication between osteoblasts in the perichondrium via N-cadherin adherens junctions is essential for endochondral bone growth. We observed that adherens junction molecule N-cadherin and its interacting partners p120, β-catenin and PTEN are expressed by cells present in the perichondrium. To study if N-cadherin mediated adherens junctions play a role in mediating signal transduction events during bone development, we utilized MC3T3E1 preosteoblasts plated at sub confluent (low) and confluent (high) densities to mimic adherens junction formation. When MC3T3E1 cells were plated at high density we observed an increase in phosphorylation of AKTSer473 and its downstream target GSK3Ser9, which coincided with an increase in Osterix, Osteomodulin and Osteoglycin gene expression. Using immunofluorescence, we identified N-cadherin, p120 and β-catenin localized at the membrane of MC3T3E1 cells. Treatment of confluent MC3T3E1 cells with an N-cadherin junction inhibitor-EGTA and a PI3K inhibitor LY294002 resulted in reduction of phosphorylation levels of AKT and GSK3 and expression of Osterix, Osteomodulin and Osteoglycin. Furthermore, utilizing an N-cadherin blocking antibody resulted in reduced AKT signaling and Osterix gene expression, suggesting that osteoblast junction formation is linked to activation of PI3K signaling, which leads to osteoblast differentiation. To further explore the strength of this linkage, we utilized a conditional knockout approach using Dermo1cre to delete β-catenin and PTEN, two important proteins known to be essential for adherens junctions and PI3K signaling, respectively. In the absence of β-catenin, we observed a decrease in adherens junctions and AKT signaling in the perichondrium. PTEN deletion, on the other hand, increased the number of cells expressing N-cadherin in the perichondrium. These observations show that N-cadherin mediated junctions between osteoblasts are needed for osteoblast gene transcription.
Collapse
Affiliation(s)
- Anyonya R Guntur
- Department of Biochemistry University of Texas Health Science Center at San Antonio, 7703 Floyd Curl Drive, San Antonio, TX 78229, USA.
| | | | | |
Collapse
|
29
|
Guntur AR, Rosen CJ. The skeleton: a multi-functional complex organ: new insights into osteoblasts and their role in bone formation: the central role of PI3Kinase. J Endocrinol 2011; 211:123-30. [PMID: 21673026 PMCID: PMC3348869 DOI: 10.1530/joe-11-0175] [Citation(s) in RCA: 91] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
Studies on bone development, formation and turnover have grown exponentially over the last decade in part because of the utility of genetic models. One area that has received considerable attention has been the phosphatidylinositol 3-kinase (PI3K) signaling pathway, which has emerged as a major survival network for osteoblasts. Genetic engineering has enabled investigators to study downstream effectors of PI3K by directly overexpressing activated forms of AKT in cells of the skeletal lineage or deleting Pten that leads to a constitutively active AKT. The results from these studies have provided novel insights into bone development and remodeling, critical processes in the lifelong maintenance of skeletal health. This paper reviews those data in relation to recent advances in osteoblast biology and their potential relevance to chronic disorders of the skeleton and their treatment.
Collapse
Affiliation(s)
- Anyonya R Guntur
- The Musculoskeletal Laboratory, Maine Medical Center Research Institute, Center for Clinical and Translational Research, Scarborough, Maine 04074, USA
| | | |
Collapse
|
30
|
Maeno T, Moriishi T, Yoshida CA, Komori H, Kanatani N, Izumi SI, Takaoka K, Komori T. Early onset of Runx2 expression caused craniosynostosis, ectopic bone formation, and limb defects. Bone 2011; 49:673-82. [PMID: 21807129 DOI: 10.1016/j.bone.2011.07.023] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/09/2011] [Revised: 07/07/2011] [Accepted: 07/13/2011] [Indexed: 11/23/2022]
Abstract
RUNX2 is an essential transcription factor for osteoblast differentiation, because osteoblast differentiation is completely blocked in Runx2-deficient mice. However, it remains to be clarified whether RUNX2 is sufficient for osteoblast differentiation during embryogenesis. To address this issue, Runx2 transgenic mice were generated under the control of the Prrx1 promoter, which directs the transgene expression to mesenchymal cells before the onset of bone development. The transgene expression was detected in the cranium, limb buds, and the region from the mandible to anterior chest wall. The skull became small and the limbs were shortened depending on the levels of the transgene expression. Early onset of Runx2 expression in the cranial mesenchyme induced mineralization on E13.0, when no mineralization was observed in wild-type mice, and resulted in craniosynostosis as shown by the closure of sutures and fontanelles on E18.5. Col1a1 and Spp1 expressions were detected in the mineralized regions on E12.5-13.5. The limb bones were hypoplastic and fused, and ectopic bones were formed in the hands and feet. Col2a1 expression was inhibited but Col1a1 expression was induced in the limb buds on E12.5. In the anterior chest wall, ectopic bones were formed through the process of intramembranous ossification, interrupting the formation of cartilaginous anlagen of sternal manubrium. These findings indicate that RUNX2 is sufficient to direct mesenchymal cells to osteoblasts and lead to intramembranous bone formation during embryogenesis; Runx2 inhibits chondrocyte differentiation at an early stage; and that Runx2 expression at appropriate level, times and spaces during embryogenesis is essential for skeletal development.
Collapse
Affiliation(s)
- Takafumi Maeno
- Department of Cell Biology, Unit of Basic Medical Sciences, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki 852-8588, Japan
| | | | | | | | | | | | | | | |
Collapse
|
31
|
Bustos-Valenzuela JC, Fujita A, Halcsik E, Granjeiro JM, Sogayar MC. Unveiling novel genes upregulated by both rhBMP2 and rhBMP7 during early osteoblastic transdifferentiation of C2C12 cells. BMC Res Notes 2011; 4:370. [PMID: 21943021 PMCID: PMC3196718 DOI: 10.1186/1756-0500-4-370] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2010] [Accepted: 09/26/2011] [Indexed: 01/24/2023] Open
Abstract
FINDINGS We set out to analyse the gene expression profile of pre-osteoblastic C2C12 cells during osteodifferentiation induced by both rhBMP2 and rhBMP7 using DNA microarrays. Induced and repressed genes were intercepted, resulting in 1,318 induced genes and 704 repressed genes by both rhBMP2 and rhBMP7. We selected and validated, by RT-qPCR, 24 genes which were upregulated by rhBMP2 and rhBMP7; of these, 13 are related to transcription (Runx2, Dlx1, Dlx2, Dlx5, Id1, Id2, Id3, Fkhr1, Osx, Hoxc8, Glis1, Glis3 and Cfdp1), four are associated with cell signalling pathways (Lrp6, Dvl1, Ecsit and PKCδ) and seven are associated with the extracellular matrix (Ltbp2, Grn, Postn, Plod1, BMP1, Htra1 and IGFBP-rP10). The novel identified genes include: Hoxc8, Glis1, Glis3, Ecsit, PKCδ, LrP6, Dvl1, Grn, BMP1, Ltbp2, Plod1, Htra1 and IGFBP-rP10. BACKGROUND BMPs (bone morphogenetic proteins) are members of the TGFβ (transforming growth factor-β) super-family of proteins, which regulate growth and differentiation of different cell types in various tissues, and play a critical role in the differentiation of mesenchymal cells into osteoblasts. In particular, rhBMP2 and rhBMP7 promote osteoinduction in vitro and in vivo, and both proteins are therapeutically applied in orthopaedics and dentistry. CONCLUSION Using DNA microarrays and RT-qPCR, we identified both previously known and novel genes which are upregulated by rhBMP2 and rhBMP7 during the onset of osteoblastic transdifferentiation of pre-myoblastic C2C12 cells. Subsequent studies of these genes in C2C12 and mesenchymal or pre-osteoblastic cells should reveal more details about their role during this type of cellular differentiation induced by BMP2 or BMP7. These studies are relevant to better understanding the molecular mechanisms underlying osteoblastic differentiation and bone repair.
Collapse
Affiliation(s)
- Juan C Bustos-Valenzuela
- Chemistry Institute, Department of Biochemistry, Cell and Molecular Therapy Centre (NUCEL), University of São Paulo, Avenida Prof, Lineu Prestes, 748 Bloco 9S, São Paulo, SP 05508-000, Brazil.
| | | | | | | | | |
Collapse
|
32
|
Choi YH, Jeong HM, Jin YH, Li H, Yeo CY, Lee KY. Akt phosphorylates and regulates the osteogenic activity of Osterix. Biochem Biophys Res Commun 2011; 411:637-41. [PMID: 21777568 DOI: 10.1016/j.bbrc.2011.07.009] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2011] [Accepted: 07/01/2011] [Indexed: 12/15/2022]
Abstract
Osterix (Osx), a zinc-finger transcription factor is required for osteoblast differentiation and new bone formation during embryonic development. Akt is a member of the serine/threonine-specific protein kinase and plays important roles in osteoblast differentiation. The function of Osterix can be also modulated by post-translational modification. But, the precise molecular signaling mechanisms between Osterix and Akt are not known. In this study, we investigated the potential regulation of Osterix function by Akt in osteoblast differentiation. We found that Akt phosphorylates Osterix and that Akt activation increases protein stability, osteogenic activity and transcriptional activity of Osterix. We also found that BMP-2 increases the protein level of Osterix in an Akt activity-dependent manner. These results suggest that Akt activity enhances the osteogenic function of Osterix, at least in part, through protein stabilization and that BMP-2 regulates the osteogenic function of Osterix, at least in part, through Akt.
Collapse
Affiliation(s)
- You Hee Choi
- College of Pharmacy and Research Institute of Drug development, Chonnam National University, Gwangju 500-757, Republic of Korea
| | | | | | | | | | | |
Collapse
|
33
|
Mandal CC, Ghosh-Choudhury N, Yoneda T, Choudhury GG, Ghosh-Choudhury N. Simvastatin prevents skeletal metastasis of breast cancer by an antagonistic interplay between p53 and CD44. J Biol Chem 2011; 286:11314-27. [PMID: 21199873 DOI: 10.1074/jbc.m110.193714] [Citation(s) in RCA: 100] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Substantial data from clinical trials and epidemiological studies show promising results for use of statins in many cancers, including mammary carcinoma. Breast tumor primarily metastasizes to bone to form osteolytic lesions, causing severe pain and pathological fracture. Here, we report that simvastatin acts as an inhibitor of osteolysis in a mouse model of breast cancer skeletal metastasis of human mammary cancer cell MDA-MB-231, which expresses the mutant p53R280K. Simvastatin and lovastatin attenuated migration and invasion of MDA-MB-231 and BT-20 breast tumor cells in culture. Acquisition of phenotype to express the cancer stem cell marker, CD44, leads to invasive potential of the tumor cells. Interestingly, statins significantly decreased the expression of CD44 protein via a transcriptional mechanism. shRNA-mediated down-regulation of CD44 markedly reduced the migration and invasion of breast cancer cells in culture. We identified that in the MDA-MB-231 cells, simvastatin elevated the levels of mutated p53R280K, which was remarkably active as a transcription factor. shRNA-derived inhibition of mutant p53R280K augmented the expression of CD44, leading to increased migration and invasion. Finally, we demonstrate an inverse correlation between expression of p53 and CD44 in the tumors of mice that received simvastatin. Our results reveal a unique function of statins, which foster enhanced expression of mutant p53R280K to prevent breast cancer cell metastasis to bone.
Collapse
Affiliation(s)
- Chandi Charan Mandal
- Department of Pathology, University of Texas Health Science Center, San Antonio, San Antonio, Texas 78229, USA
| | | | | | | | | |
Collapse
|