81 results on '"Safadi FF"'
Search Results
2. Prostaglandin E2 exacerbates collagen-induced arthritis in mice through the inflammatory interleukin-23/interleukin-17 axis.
- Author
-
Sheibanie AF, Khayrullina T, Safadi FF, and Ganea D
- Abstract
OBJECTIVE: Recently, Th17 cells, a new subset of CD4+ T cells, emerged as major players in inflammation/autoimmunity. Maintenance of the Th17 phenotype requires interleukin-23 (IL-23), whereas the Th1-promoting cytokine IL-12p70 exerts a negative effect on Th17 cell differentiation. The lipid mediator prostaglandin E(2) (PGE(2)) acts primarily as a proinflammatory agent in autoimmune conditions, through mechanisms that remain to be elucidated. The aim of this study was to investigate whether PGE(2) released in inflammatory foci activates resident dendritic cells (DCs) to express IL-23 (at the expense of IL-12) and IL-6, resulting in a shift toward Th17 cell responses. METHODS: The effect of PGE(2) on IL-23 production by DCs and subsequent induction of T cell-derived IL-17 was assessed in vitro and in vivo. The effect of the stable PGE analog misoprostol was evaluated in a murine model of rheumatoid arthritis, in conjunction with IL-23 and IL-17 expression in affected joints and draining lymph nodes. RESULTS: In vivo administration of PGE(2) induced IL-23-dependent IL-17 production. Administration of misoprostol exacerbated collagen-induced arthritis (CIA). CIA exacerbation was associated with increased levels of IL-23p19/p40 messenger RNA and reduced expression of IL-12p35, and with increased levels of the proinflammatory cytokines IL-17, IL-1beta, IL-6, and tumor necrosis factor in the affected joint. Following ex vivo restimulation, draining lymph node cells from misoprostol-treated mice secreted higher levels of IL-17 and lower levels of interferon-gamma. CONCLUSION: Our results indicate that PGE(2) enhances DC-derived IL-6 production and induces a shift in the IL-23/IL-12 balance in favor of IL-23, resulting in increased IL-17 production, presumably through the amplification of self-reactive Th17 cells. [ABSTRACT FROM AUTHOR]
- Published
- 2007
- Full Text
- View/download PDF
3. Navigating the microbial maze: unraveling the connection between gut microbiome and pediatric kidney and urinary tract disease.
- Author
-
Kusumi K, Islam MS, Banker H, Safadi FF, and Raina R
- Subjects
- Child, Humans, Kidney microbiology, Kidney physiopathology, Probiotics administration & dosage, Renal Insufficiency, Chronic microbiology, Renal Insufficiency, Chronic therapy, Dysbiosis complications, Dysbiosis diet therapy, Dysbiosis microbiology, Gastrointestinal Microbiome physiology, Urologic Diseases diet therapy, Urologic Diseases microbiology, Urologic Diseases physiopathology
- Abstract
The gut microbiome is made up of trillions of bacteria, viruses, archaea, and microbes that play a significant role in the maintenance of normal physiology in humans. Recent research has highlighted the effects of the microbiome and its dysbiosis in the pathogenesis and maintenance of kidney disease, especially chronic kidney disease (CKD) and its associated cardiovascular disease. While studies have addressed the kidney-microbiome axis in adults, how dysbiosis may uniquely impact pediatric kidney disease patients is not well-established. This narrative review highlights all relevant studies focusing on the microbiome and pediatric kidney disease that were published between 7/2015 and 7/2023. This review highlights pediatric-specific considerations including growth and bone health as well as emphasizing the need for increased pediatric research. Understanding microbiome-kidney interactions may allow for novel, less invasive interventions such as dietary changes and the use of probiotics to improve preventive care and ameliorate long-term morbidity and mortality in this vulnerable population., Competing Interests: Declarations. Conflict of interest: The authors declare no competing interests., (© 2024. The Author(s), under exclusive licence to International Pediatric Nephrology Association.)
- Published
- 2025
- Full Text
- View/download PDF
4. The expression and function of Gpnmb in lymphatic endothelial cells.
- Author
-
Kronk TA, Solorzano E, Robinson GT, Castor J, Ball HC, and Safadi FF
- Abstract
The lymphatic system functions in fluid homeostasis, lipid absorption and the modulation of the immune response. The role of Gpnmb (osteoactivin), an established osteoinductive molecule with newly identified anti-inflammatory properties, has not been studied in lymphangiogenesis. Here, we demonstrate that Gpnmb increases lymphatic endothelial cell (LEC) migration and lymphangiogenesis marker gene expression in vitro by enhancing pro-autophagic gene expression, while no changes were observed in cell proliferation or viability. In addition, cellular spreading and cytoskeletal reorganization was not altered following Gpnmb treatment. We show that systemic Gpnmb overexpression in vivo leads to increases in lymphatic tubule number per area. Overall, data presented in this study suggest Gpnmb is a positive modulator of lymphangiogenesis., Competing Interests: Declaration of competing interest The authors declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper., (Copyright © 2024. Published by Elsevier B.V.)
- Published
- 2024
- Full Text
- View/download PDF
5. The Lymphatic Endothelial Cell Secretome Inhibits Osteoblast Differentiation and Bone Formation.
- Author
-
Solorzano E, Alejo AL, Ball HC, Robinson GT, Solorzano AL, Safadi R, Douglas J, Kelly M, and Safadi FF
- Subjects
- Infant, Newborn, Humans, Secretome, Osteoblasts, Cell Differentiation, Skull, Endothelial Cells, Osteogenesis, Bone Resorption
- Abstract
Complex lymphatic anomalies (CLAs) are a set of rare diseases with unique osteopathic profiles. Recent efforts have identified how lymphatic-specific somatic activating mutations can induce abnormal lymphatic formations that are capable of invading bone and inducing bone resorption. The abnormal bone resorption in CLA patients has been linked to overactive osteoclasts in areas with lymphatic invasions. Despite these findings, the mechanism associated with progressive bone loss in CLAs remains to be elucidated. In order to determine the role of osteoblasts in CLAs, we sought to assess osteoblast differentiation and bone formation when exposed to the lymphatic endothelial cell secretome. When treated with lymphatic endothelial cell conditioned medium (L-CM), osteoblasts exhibited a significant decrease in proliferation, differentiation, and function. Additionally, L-CM treatment also inhibited bone formation through a neonatal calvaria explant culture. These findings are the first to reveal how osteoblasts may be actively suppressed during bone lymphatic invasion in CLAs.
- Published
- 2023
- Full Text
- View/download PDF
6. Osteopathy in Complex Lymphatic Anomalies.
- Author
-
Solorzano E, Alejo AL, Ball HC, Magoline J, Khalil Y, Kelly M, and Safadi FF
- Subjects
- Bone and Bones, Humans, Lymphangiogenesis, Bone Diseases metabolism, Lymphatic Abnormalities, Lymphatic Vessels metabolism
- Abstract
Complex Lymphatic Anomalies (CLA) are lymphatic malformations with idiopathic bone and soft tissue involvement. The extent of the abnormal lymphatic presentation and boney invasion varies between subtypes of CLA. The etiology of these diseases has proven to be extremely elusive due to their rarity and irregular progression. In this review, we compiled literature on each of the four primary CLA subtypes and discuss their clinical presentation, lymphatic invasion, osseous profile, and regulatory pathways associated with abnormal bone loss caused by the lymphatic invasion. We highlight key proliferation and differentiation pathways shared between lymphatics and bone and how these systems may interact with each other to stimulate lymphangiogenesis and cause bone loss.
- Published
- 2022
- Full Text
- View/download PDF
7. Epigenetic Regulation of Chondrocytes and Subchondral Bone in Osteoarthritis.
- Author
-
Ball HC, Alejo AL, Samson TK, Alejo AM, and Safadi FF
- Abstract
The aim of this review is to provide an updated review of the epigenetic factors involved in the onset and development of osteoarthritis (OA). OA is a prevalent degenerative joint disease characterized by chronic inflammation, ectopic bone formation within the joint, and physical and proteolytic cartilage degradation which result in chronic pain and loss of mobility. At present, no disease-modifying therapeutics exist for the prevention or treatment of the disease. Research has identified several OA risk factors including mechanical stressors, physical activity, obesity, traumatic joint injury, genetic predisposition, and age. Recently, there has been increased interest in identifying epigenetic factors involved in the pathogenesis of OA. In this review, we detail several of these epigenetic modifications with known functions in the onset and progression of the disease. We also review current therapeutics targeting aberrant epigenetic regulation as potential options for preventive or therapeutic treatment.
- Published
- 2022
- Full Text
- View/download PDF
8. A Pre-clinical Standard Operating Procedure for Evaluating Orthobiologics in an In Vivo Rat Spinal Fusion Model.
- Author
-
Alejo AL, McDermott S, Khalil Y, Ball HC, Robinson GT, Solorzano E, Alejo AM, Douglas J, Samson TK, Young JW, and Safadi FF
- Abstract
The rat animal model is a cost effective and reliable model used in spinal pre-clinical research. Complications from various surgical procedures in humans often arise that were based on these pre-clinical animal models. Therefore safe and efficacious pre-clinical animal models are needed to establish continuity into clinical trials. A Standard Operating Procedure (SOP) is a validated method that allows researchers to safely and carefully replicate previously successful surgical techniques. Thus, the aim of this study is to describe in detail the procedures involved in a common rat bilateral posterolateral intertransverse spinal fusion SOP used to test the efficacy and safety different orthobiologics using a collagen-soaked sponge as an orthobiologic carrier. Only two orthobiologics are currently FDA approved for spinal fusion surgery which include recombinant bone morphogenetic protein 2 (rhBMP-2), and I-FACTOR. While there are many additional orthobiologics currently being tested, one way to show their safety profile and gain FDA approval, is to use well established pre-clinical animal models. A preoperative, intraoperative, and postoperative surgical setup including specific anesthesia and euthanasia protocols are outlined. Furthermore, we describe different postoperative methods used to validate the spinal fusion SOP, which include μ CT analysis, histopathology, biomechanical testing, and blood analysis. This SOP can help increase validity, transparency, efficacy, and reproducibly in future rat spinal fusion surgery procedures., Competing Interests: Conflicts of Interest The authors have no conflicts of interest to declare.
- Published
- 2022
- Full Text
- View/download PDF
9. The role of miR-150 regulates bone cell differentiation and function.
- Author
-
Moussa FM, Cook BP, Sondag GR, DeSanto M, Obri MS, McDermott SE, and Safadi FF
- Subjects
- Animals, Cell Differentiation, Mice, Mice, Inbred C57BL, Mice, Knockout, Osteoblasts, Osteoclasts, Osteocytes, Osteogenesis genetics, Bone Resorption genetics, MicroRNAs genetics
- Abstract
Background: mir-RNAs play a role in regulating bone homeostasis. In this study we assessed the functional role of mir-RNA 150 in bone homeostasis. We also assess the effects of miR-150 deficiency on osteoblast and osteoclast differentiation and function using in vivo and in vitro approaches., Methods: Wild type (WT) (C57BL/6J) and miR-150 KO mice were compared for a variety of parameters. Micro-CT imaging was conducted to quantify trabecular bone mass inferior to the distal growth plate of the femur. Von Kossa staining was performed for osteoblast culture mineralization. RT-qPCR, biochemical analysis and bone histomorphometry were utilized for quantification of relevant genes and serum protein measurements. Differentiation and function of osteoblasts and osteoclasts was performed using primarily cultures and assessed the cell autonomous response of mir-RNA-150 on cell differentiation and function., Results: Mir-150 exhibited expression in a variety of tissues and increases progressively with age. Through micro-CT imaging, we found that KO mice presented reduced bone mass at 4, 8, and 16 weeks of age compared to WT mice. Furthermore, histomorphometric analysis revealed increased trabecular separation, decreased bone thickness, and decreased osteoblast number in KO compared to WT mice. Mir-150 deficiency also correlated with higher bone resorption, accompanied with significant increases in CTX-1 serum levels, and a decrease in cell apoptotic rate ex vivo. Additionally, miR-150 KO mice showed increased osteoblast differentiation and decreased osteoclastogenesis ex vivo. Luciferase assay showed increased Osteoactivin/GPNMB expression in miR-150 KO osteoblasts compared to WT cells., Conclusion: Our data suggests that miR-150 influences osteoblast and osteoclast functionality and differentiation; specifically, miR-150 serves as a negative regulator for osteoblasts and a positive regulator for osteoclasts by regulating, at least in part, Osteoactivin/GPNMB expression., (Copyright © 2020. Published by Elsevier Inc.)
- Published
- 2021
- Full Text
- View/download PDF
10. Linking gene expression and phenotypic changes in the developmental and evolutionary origins of osteosclerosis in the ribs of bowhead whales (Balaena mysticetus).
- Author
-
Cooper LN, Ball HC, Vinyard CJ, Safadi FF, George JC, and Thewissen JGM
- Subjects
- Aging, Animals, Epigenesis, Genetic, Osteosclerosis genetics, Osteosclerosis pathology, Ribs metabolism, Gene Expression Regulation, Developmental physiology, Osteosclerosis veterinary, Ribs physiology, Whales genetics, Whales growth & development
- Abstract
Bowhead whales are among the longest-lived mammals with an extreme lifespan of about 211 years. During the first 25 years of their lives, rib bones increase in mineral density and the medulla transitions from compact to trabecular bone. Molecular drivers associated with these phenotypic changes in bone remain unknown. This study assessed expression levels of osteogenic genes from samples of rib bones of bowheads. Samples were harvested from prenatal to 86-year-old whales, representing the first third of the bowhead lifespan. Fetal to 2-year-old bowheads showed expression levels consistent with the rapid deposition of the bone extracellular matrix. Sexually mature animals showed expression levels associated with low rates of osteogenesis and increased osteoclastogenesis. After the first 25 years of life, declines in osteogenesis corresponded with increased expression of EZH2, an epigenetic regulator of osteogenesis. These findings suggest EZH2 may be at least one epigenetic modifier that contributes to the age-related changes in the rib bone phenotype along with the transition from compact to trabecular bone. Ancient cetaceans and their fossil relatives also display these phenotypes, suggesting EZH2 may have shaped the skeleton of whales in evolutionary history., (© 2020 Wiley Periodicals LLC.)
- Published
- 2020
- Full Text
- View/download PDF
11. Transgenic Overexpression of GPNMB Protects Against MPTP-Induced Neurodegeneration.
- Author
-
Budge KM, Neal ML, Richardson JR, and Safadi FF
- Subjects
- Animals, Corpus Striatum pathology, Dopaminergic Neurons pathology, Eye Proteins metabolism, Inflammation chemically induced, Inflammation genetics, Inflammation metabolism, Inflammation pathology, Lipopolysaccharides, MPTP Poisoning metabolism, MPTP Poisoning pathology, Male, Membrane Glycoproteins metabolism, Mice, Mice, Transgenic, Microglia pathology, Corpus Striatum metabolism, Dopaminergic Neurons metabolism, Eye Proteins genetics, MPTP Poisoning genetics, Membrane Glycoproteins genetics, Microglia metabolism
- Abstract
Parkinson's disease (PD) is a progressive neurodegenerative disease highlighted by a marked loss of dopaminergic cell loss and motor disturbances. Currently, there are no drugs that slow the progression of the disease. A myriad of factors have been implicated in the pathogenesis and progression of PD including neuroinflammation. Although anti-inflammatory agents are being evaluated as potential disease-modifying therapies for PD, none has proven effective to date, suggesting that new and novel targets are needed. Glycoprotein nonmetastatic melanoma protein B (GPNMB) is a transmembrane glycoprotein that has recently been shown to reduce inflammation in astrocytes and to be increased in post-mortem PD brain samples. Here we show that transgenic overexpression of GPNMB protects against dopaminergic neurodegeneration in a 1-methyl-4-phenyl-1,2,3,6-tetrahydropridine mouse model of Parkinson's disease. Furthermore, GPNMB overexpression reduces gliosis and prevented microglial morphological changes following MPTP treatment compared with wild-type MPTP-treated mice. Additionally, recombinant GPNMB attenuates LPS-induced inflammation in primary mouse microglia. These results suggest a neuroprotective and anti-inflammatory role for GPNMB and warrant further investigation for GPNMB as a novel therapy for PD.
- Published
- 2020
- Full Text
- View/download PDF
12. A novel regulatory role of TRAPPC9 in L-plastin-mediated osteoclast actin ring formation.
- Author
-
Hussein NJ, Mbimba T, Al-Adlaan AA, Ansari MY, Jaber FA, McDermott S, Kasumov T, and Safadi FF
- Subjects
- Animals, Cell Differentiation, Chromatography, Liquid, Gene Expression Regulation, Male, Mice, Mice, Inbred C57BL, NF-kappa B p50 Subunit metabolism, Osteoclasts cytology, Recombinant Proteins metabolism, Tandem Mass Spectrometry, Vesicular Transport Proteins, Vinculin metabolism, Actins metabolism, Cathepsin K metabolism, Intercellular Signaling Peptides and Proteins metabolism, Membrane Glycoproteins metabolism, Microfilament Proteins metabolism, Osteoclasts metabolism, Podosomes metabolism
- Abstract
Trafficking protein particle complex 9 (TRAPPC9) is a major subunit of the TRAPPII complex. TRAPPC9 has been reported to bind nuclear factor κB kinase subunit β (IKKβ) and NF-kB-inducing kinase (NIK) where it plays a role in the canonical and noncanonical of nuclear factor-κB (NF-kB) signaling pathways, receptively. The role of TRAPPC9 in protein trafficking and cytoskeleton organization in osteoclast (OC) has not been studied yet. In this study, we examined the mRNA expression of TRAPPC9 during OC differentiation. Next, we examined the colocalization of TRAPPC9 with cathepsin-K, known to mediate OC resorption suggesting that TRAPPC9 mediates the trafficking pathway within OC. To identify TRAPPC9 protein partners important for OC-mediated cytoskeleton re-organization, we conducted immunoprecipitation of TRAPPC9 in mature OCs followed by mass spectrometry analysis. Our data showed that TRAPPC9 binds various protein partners. One protein with high recovery rate is L-plastin (LPL). LPL localizes at the podosomes and reported to play a crucial role in actin aggregation thereby actin ring formation and OC function. Although the role of LPL in OC-mediated bone resorption has not fully reported in detail. Here, first, we confirmed the binding of LPL to TRAPPC9 and, then, we investigated the potential regulatory role of TRAPPC9 in LPL-mediated OC cytoskeleton reorganization. We assessed the localization of TRAPPC9 and LPL in OC and found that TRAPPC9 is colocalized with LPL at the periphery of OC. Next, we determined the effect of TRAPPC9 overexpression on LPL recruitment to the actin ring using a viral system. Interestingly, our data showed that TRAPPC9 overexpression promotes the recruitment of LPL to the actin ring when compared with control cultures. In addition, we observed that TRAPPC9 overexpression reorganizes actin clusters/aggregates and regulates vinculin recruitment into the OC periphery to initiate podosome formation., (© 2019 Wiley Periodicals, Inc.)
- Published
- 2020
- Full Text
- View/download PDF
13. Autophagy plays an essential role in bone homeostasis.
- Author
-
Jaber FA, Khan NM, Ansari MY, Al-Adlaan AA, Hussein NJ, and Safadi FF
- Subjects
- Animals, Cell Differentiation physiology, Homeostasis physiology, Humans, Osteoblasts cytology, Osteoclasts cytology, Osteocytes cytology, Autophagy physiology, Bone Remodeling physiology, Bone and Bones cytology, Osteogenesis physiology
- Abstract
Autophagy is very critical for multiple cellular processes. Autophagy plays a critical role in bone cell differentiation and function., (© 2019 Wiley Periodicals, Inc.)
- Published
- 2019
- Full Text
- View/download PDF
14. TRAPPC9: Novel insights into its trafficking and signaling pathways in health and disease (Review).
- Author
-
Mbimba T, Hussein NJ, Najeed A, and Safadi FF
- Subjects
- Animals, Carrier Proteins chemistry, Carrier Proteins genetics, Gene Expression Regulation, Humans, Intercellular Signaling Peptides and Proteins, Protein Binding, Protein Interaction Domains and Motifs, Protein Transport, Structure-Activity Relationship, Carrier Proteins metabolism, Disease Susceptibility, Signal Transduction
- Abstract
Trafficking protein particle complex 9 (TRAPPC9) is a protein subunit of the transport protein particle II (TRAPPII), which has been reported to be important in the trafficking of cargo from the endoplasmic reticulum (ER) to the Golgi, and in intra‑Golgi and endosome‑to‑Golgi transport in yeast cells. In mammalian cells, TRAPPII has been shown to be important in Golgi vesicle tethering and intra‑Golgi transport. TRAPPC9 is considered to be a novel molecule capable of modulating the activation of nuclear factor‑κB (NF‑κB). Mutations in TRAPPC9 have been linked to a rare consanguineous hereditary form of mental retardation, as part of the NF‑κB pathways. In addition, TRAPPC9 has been reported to be involved in breast and colon cancer and liver diseases. The present review highlights the most recent publications on the structure, expression and function of TRAPPC9, and its association with various human diseases.
- Published
- 2018
- Full Text
- View/download PDF
15. Glycoprotein NMB: an Emerging Role in Neurodegenerative Disease.
- Author
-
Budge KM, Neal ML, Richardson JR, and Safadi FF
- Subjects
- Animals, Humans, Immune System metabolism, Membrane Glycoproteins chemistry, Nerve Degeneration pathology, Neurodegenerative Diseases therapy, Membrane Glycoproteins metabolism, Neurodegenerative Diseases metabolism
- Abstract
Neurodegeneration is characterized by severe neuronal loss leading to the cognitive and physical impairments that define various neurodegenerative diseases. Neuroinflammation is one hallmark of neurodegenerative diseases and can ultimately contribute to disease progression. Increased inflammatory cytokines, such as interleukin-6 (IL-6), interleukin-1β (IL-1 β), and tumor necrosis factor-α (TNF-α) are associated with Alzheimer's disease (AD), Parkinson's disease (PD), amyotrophic lateral sclerosis (ALS), and multiple sclerosis (MS). Unfortunately, current therapeutic options lack ability to stop or effectively slow progression of these diseases and are primarily aimed at alleviating symptoms. Thus, it is crucial to discover novel treatment candidates for neurodegenerative diseases. Glycoprotein nonmetastatic melanoma protein B (GPNMB) is a type-I transmembrane glycoprotein first identified in a melanoma cell line. GPNMB augments bone mineral deposition by stimulating osteoblast differentiation. Aside from its anabolic function in the bone, emerging evidence suggests that GPNMB has anti-inflammatory and reparative functions. GPNMB has also been demonstrated to be neuroprotective in an animal model of ALS, cerebral ischemia, and other disease models. Given these discoveries, GPNMB should be investigated as a potential therapeutic option for multiple neurodegenerative diseases.
- Published
- 2018
- Full Text
- View/download PDF
16. The glycoprotein GPNMB attenuates astrocyte inflammatory responses through the CD44 receptor.
- Author
-
Neal ML, Boyle AM, Budge KM, Safadi FF, and Richardson JR
- Subjects
- 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine pharmacology, Analysis of Variance, Animals, Case-Control Studies, Cells, Cultured, Cytokines genetics, Cytokines metabolism, Databases, Chemical, Female, Glial Fibrillary Acidic Protein metabolism, Humans, Inflammation etiology, Male, Mice, Neurotoxins toxicity, Nitric Oxide metabolism, Parkinson Disease complications, RNA, Messenger metabolism, Reactive Oxygen Species metabolism, Signal Transduction drug effects, Anti-Inflammatory Agents therapeutic use, Astrocytes drug effects, Hyaluronan Receptors metabolism, Inflammation drug therapy, Membrane Glycoproteins therapeutic use, Parkinson Disease pathology
- Abstract
Background: Neuroinflammation is one of the hallmarks of neurodegenerative diseases, such as Parkinson's disease (PD). Activation of glial cells, including microglia and astrocytes, is a characteristic of the inflammatory response. Glycoprotein non-metastatic melanoma protein B (GPNMB) is a transmembrane glycoprotein that releases a soluble signaling peptide when cleaved by ADAM10 or other extracellular proteases. GPNMB has demonstrated a neuroprotective role in animal models of ALS and ischemia. However, the mechanism of this protection has not been well established. CD44 is a receptor expressed on astrocytes that can bind GPNMB, and CD44 activation has been demonstrated to reduce NFκB activation and subsequent inflammatory responses in macrophages. GPNMB signaling has not been investigated in models of PD or specifically in astrocytes. More recently, genetic studies have linked polymorphisms in GPNMB with risk for PD. Therefore, it is important to understand the role this signaling protein plays in PD., Methods: We used data mining techniques to evaluate mRNA expression of GPNMB and its receptor CD44 in the substantia nigra of PD and control brains. Immunofluorescence and qPCR techniques were used to assess GPNMB and CD44 levels in mice treated with MPTP. In vitro experiments utilized the immortalized mouse astrocyte cell line IMA2.1 and purified primary mouse astrocytes. The effects of recombinant GPNMB on cytokine-induced astrocyte activation was determined by qPCR, immunofluorescence, and measurement of nitric oxide and reactive oxygen production., Results: Increased GPNMB and CD44 expression was observed in the substantia nigra of human PD brains and in GFAP-positive astrocytes in an animal model of PD. GPNMB treatment attenuated cytokine-induced levels of inducible nitric oxide synthase, nitric oxide, reactive oxygen species, and the inflammatory cytokine IL-6 in an astrocyte cell line and primary mouse astrocytes. Using primary mouse astrocytes from CD44 knockout mice, we found that the anti-inflammatory effects of GPNMB are CD44-mediated., Conclusions: These results demonstrate that GPNMB may exert its neuroprotective effect through reducing astrocyte-mediated neuroinflammation in a CD44-dependent manner, providing novel mechanistic insight into the neuroprotective properties of GPNMB.
- Published
- 2018
- Full Text
- View/download PDF
17. Osteoactivin regulates head and neck squamous cell carcinoma invasion by modulating matrix metalloproteases.
- Author
-
Arosarena OA, Barr EW, Thorpe R, Yankey H, Tarr JT, and Safadi FF
- Subjects
- Carcinoma, Squamous Cell genetics, Carcinoma, Squamous Cell pathology, Cell Line, Tumor, Gene Expression Regulation, Enzymologic, Gene Expression Regulation, Neoplastic, Head and Neck Neoplasms genetics, Head and Neck Neoplasms pathology, Humans, Matrix Metalloproteinases, Secreted genetics, Membrane Glycoproteins genetics, Neoplasm Invasiveness, RNA Interference, RNA, Messenger genetics, RNA, Messenger metabolism, Signal Transduction, Squamous Cell Carcinoma of Head and Neck, Transfection, Carcinoma, Squamous Cell enzymology, Cell Movement, Head and Neck Neoplasms enzymology, Matrix Metalloproteinases, Secreted metabolism, Membrane Glycoproteins metabolism
- Abstract
Nearly 60% of patients with head and neck squamous cell carcinoma (HNSCC) die of metastases or locoregional recurrence. Metastasis is mediated by cancer cell migration and invasion, which are in part dependent on extracellular matrix degradation by matrix metalloproteinases. Osteoactivin (OA) overexpression plays a role in metastases in several malignancies, and has been shown to upregulate matrix metalloproteinase (MMP) expression and activity. To determine how OA modulates MMP expression and activity in HNSCC, and to investigate OA effects on cell invasion, we assessed effects of OA treatment on MMP mRNA and protein expression, as well as gelatinase and caseinolytic activity in HNSCC cell lines. We assessed the effects of OA gene silencing on MMP expression, gelatinase and caseinolytic activity, and cell invasion. OA treatment had differential effects on MMP mRNA expression. OA treatment upregulated MMP-10 expression in UMSCC14a (p = 0.0431) and SCC15 (p < 0.0001) cells, but decreased MMP-9 expression in UMSCC14a cells (p = 0.0002). OA gene silencing decreased MMP-10 expression in UMSCC12 cells (p = 0.0001), and MMP-3 (p = 0.0005) and -9 (p = 0.0036) expression in SCC25 cells. In SCC15 and SCC25 cells, OA treatment increased MMP-2 (p = 0.0408) and MMP-9 gelatinase activity (p < 0.0001), respectively. OA depletion decreased MMP-2 (p = 0.0023) and -9 (p < 0.0001) activity in SCC25 cells. OA treatment increased 70 kDa caseinolytic activity in UMSCC12 cells consistent with tissue type plasminogen activator (p = 0.0078). OA depletion decreased invasive capacity of UMSCC12 cells (p < 0.0001). OA's effects on MMP expression in HNSCC are variable, and may promote cancer cell invasion., (© 2017 Wiley Periodicals, Inc.)
- Published
- 2018
- Full Text
- View/download PDF
18. Identification of Novel Agents for the Treatment of Brain Metastases of Breast Cancer.
- Author
-
Venishetty VK, Geldenhuys WJ, Terell-Hall TB, Griffith JIG, Sondag GR, Safadi FF, and Lockman PR
- Subjects
- Animals, Apoptosis, Brain Neoplasms pathology, Cell Line, Tumor, Female, Humans, Mice, Antineoplastic Agents therapeutic use, Brain Neoplasms drug therapy, Brain Neoplasms secondary, Breast Neoplasms pathology
- Abstract
Background: Brain cancer from metastasized breast cancer has a high mortality rate in women. The treatment of lesions is hampered in large part by the blood-brain barrier (BBB), which prevents adequate distribution of anti-cancer compounds to brain metastases., Method: In this study we used a novel screening method to identify candidate molecules that are well-suited to utilizing the BBB choline transporter for distribution into the brain parenchyma., Results: From our screen we identified two compounds, Ch-1 and Ch-2 that were able to reduce the brain tumor burden in a murine mouse model of brain metastasis of breast cancer. These compounds also significantly increased the survival of mice by more than 10 days. Mechanistic studies indicated that Ch-1 is able to prevent the activation of the pro-survival mitogen-activated kinases (MAPKs) by osteoactivin (OA; Glycoprotein nonmetastatic melanoma protein B GPNMB)., Conclusion: The results from this study show that nutrient transporter virtual screening is a viable novel alternative to traditional drug screening programs to identify anti-cancer compounds for the treatment of brain cancers., (Copyright© Bentham Science Publishers; For any queries, please email at epub@benthamscience.org.)
- Published
- 2017
- Full Text
- View/download PDF
19. Osteoactivin inhibition of osteoclastogenesis is mediated through CD44-ERK signaling.
- Author
-
Sondag GR, Mbimba TS, Moussa FM, Novak K, Yu B, Jaber FA, Abdelmagid SM, Geldenhuys WJ, and Safadi FF
- Subjects
- Animals, Cell Differentiation, Cells, Cultured, Male, Mice, Inbred C57BL, Osteoclasts metabolism, RANK Ligand metabolism, Recombinant Proteins metabolism, Eye Proteins metabolism, Hyaluronan Receptors metabolism, MAP Kinase Signaling System, Membrane Glycoproteins metabolism, Osteoclasts cytology, Signal Transduction
- Abstract
Osteoactivin is a heavily glycosylated protein shown to have a role in bone remodeling. Previous studies from our lab have shown that mutation in Osteoactivin enhances osteoclast differentiation but inhibits their function. To date, a classical receptor and a signaling pathway for Osteoactivin-mediated osteoclast inhibition has not yet been characterized. In this study, we examined the role of Osteoactivin treatment on osteoclastogenesis using bone marrow-derived osteoclast progenitor cells and identify a signaling pathway relating to Osteoactivin function. We reveal that recombinant Osteoactivin treatment inhibited osteoclast differentiation in a dose-dependent manner shown by qPCR, TRAP staining, activity and count. Using several approaches, we show that Osteoactivin binds CD44 in osteoclasts. Furthermore, recombinant Osteoactivin treatment inhibited ERK phosphorylation in a CD44-dependent manner. Finally, we examined the role of Osteoactivin on receptor activator of nuclear factor-κ B ligand (RANKL)-induced osteolysis in vivo. Our data indicate that recombinant Osteoactivin inhibits RANKL-induced osteolysis in vivo and this effect is CD44-dependent. Overall, our data indicate that Osteoactivin is a negative regulator of osteoclastogenesis in vitro and in vivo and that this process is regulated through CD44 and ERK activation.
- Published
- 2016
- Full Text
- View/download PDF
20. Osteoactivin Promotes Migration of Oral Squamous Cell Carcinomas.
- Author
-
Arosarena OA, Dela Cadena RA, Denny MF, Bryant E, Barr EW, Thorpe R, and Safadi FF
- Subjects
- Carcinoma, Squamous Cell genetics, Carcinoma, Squamous Cell pathology, Cell Adhesion, Cell Line, Tumor, Cell Proliferation, Cell Survival, Enzyme Activation, Gene Expression Regulation, Neoplastic, Head and Neck Neoplasms genetics, Head and Neck Neoplasms pathology, Humans, Integrin beta1 metabolism, Membrane Glycoproteins genetics, Mitogen-Activated Protein Kinases metabolism, Mouth Neoplasms genetics, Mouth Neoplasms pathology, Neoplasm Invasiveness, Protein Binding, RNA Interference, RNA, Messenger metabolism, Signal Transduction, Squamous Cell Carcinoma of Head and Neck, Time Factors, Transfection, Carcinoma, Squamous Cell metabolism, Cell Movement, Head and Neck Neoplasms metabolism, Membrane Glycoproteins metabolism, Mouth Neoplasms metabolism
- Abstract
Nearly 50% of patients with oral squamous cell carcinoma (OSCC) die of metastases or locoregional recurrence. Metastasis is mediated by cancer cell adhesion, migration, and invasion. Osteoactivin (OA) overexpression plays a role in metastases in several malignancies. The aims were to determine how integrin interactions modulate OA-induced OSCC cell migration; and to investigate OA effects on cell survival and proliferation. We confirmed OA mRNA and protein overexpression in OSCC cell lines. We assessed OA's interactions with integrins using adhesion inhibition assays, fluorescent immunocytochemistry and co-immunoprecipitation. We investigated OA-mediated activation of mitogen-activated protein kinases (MAPKs) and cell survival. Integrin inhibition effects on OA-mediated cell migration were determined. We assessed effects of OA knock-down on cell migration and proliferation. OA is overexpressed in OSCC cell lines, and serves as a migration-promoting adhesion molecule. OA co-localized with integrin subunits, and co-immunoprecipitated with the subunits. Integrin blocking antibodies, especially those directed against the β1 subunit, inhibited cell adhesion (P = 0.03 for SCC15 cells). Adhesion to OA activated MAPKs in UMSCC14a cells and OA treatment promoted survival of SCC15 cells. Integrin-neutralizing antibodies enhanced cell migration with OA in the extracellular matrix. OA knock-down resulted in decreased proliferation of SCC15 and SCC25 cells, but did not inhibit cell migration. OA in the extracellular matrix promotes OSCC cell adhesion and migration, and may be a novel target in the prevention of HNSCC spread. J. Cell. Physiol. 231: 1761-1770, 2016. © 2015 Wiley Periodicals, Inc., (© 2015 Wiley Periodicals, Inc.)
- Published
- 2016
- Full Text
- View/download PDF
21. Orthosilicic acid, Si(OH)4, stimulates osteoblast differentiation in vitro by upregulating miR-146a to antagonize NF-κB activation.
- Author
-
Zhou X, Moussa FM, Mankoci S, Ustriyana P, Zhang N, Abdelmagid S, Molenda J, Murphy WL, Safadi FF, and Sahai N
- Subjects
- Animals, Humans, Mice, Osteogenesis drug effects, RAW 264.7 Cells, Cell Differentiation drug effects, Mesenchymal Stem Cells metabolism, MicroRNAs biosynthesis, NF-kappa B metabolism, Osteoblasts metabolism, Silicic Acid pharmacology
- Abstract
Unlabelled: Accumulating evidence over the last 40years suggests that silicate from dietary as well as silicate-containing biomaterials is beneficial to bone formation. However, the exact biological role(s) of silicate on bone cells are still unclear and controversial. Here, we report that orthosilicic acid (Si(OH)4) stimulated human mesenchymal stem cells (hMSCs) osteoblastic differentiation in vitro. To elucidate the possible molecular mechanisms, differential microRNA microarray analysis was used to show that Si(OH)4 significantly up-regulated microRNA-146a (miR-146a) expression during hMSC osteogenic differentiation. Si(OH)4 induced miR-146a expression profiling was further validated by quantitative RT-PCR (qRT-PCR), which indicated miR-146a was up-regulated during the late stages of hMSC osteogenic differentiation. Inhibition of miR-146a function by anti-miR-146a suppressed osteogenic differentiation of MC3T3 pre-osteoblasts, whereas Si(OH)4 treatment promoted osteoblast-specific genes transcription, alkaline phosphatase (ALP) production, and mineralization. Furthermore, luciferase reporter assay, Western blotting, enzyme-linked immunosorbent assay (ELISA), and immunofluorescence showed that Si(OH)4 decreased TNFα-induced activation of NF-κB, a signal transduction pathway that inhibits osteoblastic bone formation, through the known miR-146a negative feedback loop. Our studies established a mechanism for Si(OH)4 to promote osteogenesis by antagonizing NF-κB activation via miR-146a, which might be interesting to guide the design of osteo-inductive biomaterials for treatments of bone defects in humans., Statement of Significance: Accumulating evidence over 40years suggests that silicate is beneficial to bone formation. However, the biological role(s) of silicate on bone cells are still unclear and controversial. Here, we report that Si(OH)4, the simplest form of silicate, can stimulate human mesenchymal stem cells osteoblastic differentiation. We identified that miR-146a is the expression signature in bone cells treated with Si(OH)4. Further analysis of miR-146a in bone cells reveals that Si(OH)4 upregulates miR-146a to antagonize the activation of NF-κB. Si(OH)4 was also shown to deactivate the same NF-κB pathway to suppress osteoclast formation. Our findings are important to the development of third-generation cell-and gene affecting biomaterials, and suggest silicate and miR-146a can be used as pharmaceuticals for bone fracture prevention and therapy., (Copyright © 2016 Acta Materialia Inc. Published by Elsevier Ltd. All rights reserved.)
- Published
- 2016
- Full Text
- View/download PDF
22. Macrophage-Associated Osteoactivin/GPNMB Mediates Mesenchymal Stem Cell Survival, Proliferation, and Migration Via a CD44-Dependent Mechanism.
- Author
-
Yu B, Sondag GR, Malcuit C, Kim MH, and Safadi FF
- Subjects
- Animals, Cell Survival physiology, Eye Proteins genetics, Hyaluronan Receptors genetics, MAP Kinase Signaling System physiology, Macrophages cytology, Membrane Glycoproteins genetics, Mesenchymal Stem Cells cytology, Mice, Mice, Knockout, Cell Movement physiology, Cell Proliferation physiology, Eye Proteins metabolism, Hyaluronan Receptors metabolism, Macrophages metabolism, Membrane Glycoproteins metabolism, Mesenchymal Stem Cells metabolism
- Abstract
Although MSCs have been widely recognized to have therapeutic potential in the repair of injured or diseased tissues, it remains unclear how functional activities of mesenchymal stem cells (MSCs) are influenced by the surrounding inflammatory milieu at the site of tissue injury. Macrophages constitute an essential component of innate immunity and have been shown to exhibit a phenotypic plasticity in response to various stimuli, which play a central role in both acute inflammation and wound repair. Osteoactivin (OA)/Glycoprotein non-metastatic melanoma protein B (GPNMB), a transmembrane glycoprotein that plays a role in cell differentiation, survival, and angiogenesis. The objective of this study was to investigate the potential role of OA/GPNMB in macrophage-induced MSC function. We found that reparative M2 macrophages express significantly greater levels of OA/GPNMB than pro-inflammatory M1 macrophages. Furthermore, using loss of function and rescue studies, we demonstrated that M2 macrophages-secreted OA/GPNMB positively regulates the viability, proliferation, and migration of MSCs. More importantly, we demonstrated that OA/GPNMB acts through ERK and AKT signaling pathways in MSCs via CD44, to induce these effects. Taken together, our results provide pivotal insight into the mechanism by which OA/GPNMB contributes to the tissue reparative phenotype of M2 macrophages and positively regulates functional activities of MSCs. J. Cell. Biochem. 117: 1511-1521, 2016. © 2015 Wiley Periodicals, Inc., (© 2015 Wiley Periodicals, Inc.)
- Published
- 2016
- Full Text
- View/download PDF
23. Methods and insights from the characterization of osteoprogenitor cells of bats (Mammalia: Chiroptera).
- Author
-
Ball HC, Moussa FM, Mbimba T, Orman R, Safadi FF, and Cooper LN
- Subjects
- Animals, Bone Marrow Cells cytology, Cell Differentiation genetics, Cell Proliferation, Cells, Cultured, Cellular Reprogramming, Chiroptera, Core Binding Factor Alpha 1 Subunit genetics, Core Binding Factor Alpha 1 Subunit metabolism, Mice, Mice, Inbred C57BL, Osteoblasts cytology, Osteocalcin genetics, Osteocalcin metabolism, Real-Time Polymerase Chain Reaction, Stem Cells metabolism, Transcription Factors genetics, Transcription Factors metabolism, Osteoblasts metabolism, Osteogenesis genetics, Stem Cells cytology
- Abstract
Osteoprogenitor cells contribute to the development and maintenance of skeletal tissues. Bats are unique model taxa whose cellular processes are poorly understood, especially in regards to skeletal biology. Forelimb bones of bats, unlike those of terrestrial mammals, bend during flight and function in controlled deformation. As a first step towards understanding the molecular processes governing deposition of this flexible bone matrix, we provide the first method for isolation and differentiation of cell populations derived from the bone marrow and cortical bone of bats, and compare results with those harvested from C57BL/6J mice. Osteogenic capacity of these cells was assessed via absolute quantitative real-time PCR (qPCR) and through quantification of in vitro mineral deposition. Results indicate the differentiated bone cells of bats display significantly lower gene expression of known osteogenic markers (Runt-related transcription factor (RUNX2), osteocalcin (BGLAP) and osterix (SP7)), and deposit a less-mineralized matrix compared with murine controls. By characterizing the in vitro performance of osteoprogenitor cells throughout differentiation and matrix production, this study lays the ground work for in vitro manipulations of bat stem and osteoprogenitor cells and extends our understanding of the cellular diversity across mammals that occupy different habitats., (Copyright © 2016 The Authors. Published by Elsevier B.V. All rights reserved.)
- Published
- 2016
- Full Text
- View/download PDF
24. Osteoactivin (GPNMB) ectodomain protein promotes growth and invasive behavior of human lung cancer cells.
- Author
-
Oyewumi MO, Manickavasagam D, Novak K, Wehrung D, Paulic N, Moussa FM, Sondag GR, and Safadi FF
- Subjects
- Animals, Apoptosis, Carcinoma, Non-Small-Cell Lung metabolism, Cell Adhesion, Female, Humans, Lung Neoplasms metabolism, Mice, Mice, Nude, Neoplasm Invasiveness, Protein Domains, Tumor Cells, Cultured, Xenograft Model Antitumor Assays, Biomarkers, Tumor metabolism, Carcinoma, Non-Small-Cell Lung pathology, Cell Movement, Cell Proliferation, Lung Neoplasms pathology, Membrane Glycoproteins metabolism
- Abstract
The potential application of GPNMB/OA as a therapeutic target for lung cancer will require a greater understanding of the impact of GPNMB/OA ectodomain (ECD) protein shedding into tumor tissues. Thus, in this work we characterized GPNMB/OA expression and extent of shedding of its ECD protein while evaluating the impact on lung cancer progression using three non-small cell lung cancer (NSCLC) cell lines: A549, SK-MES-1 and calu-6. We observed a direct correlation (R2 = 0.89) between GPNMB/OA expression on NSCLC cells and the extent of GPNMB/OA ECD protein shedding. Meanwhile, siRNA-mediated knockdown of GPNMB/OA in cancer cells significantly reduced GPNMB/OA ECD protein shedding, migration, invasion and adhesion to extracellular matrix materials. Also, exogenous treatment of cancer cells (expressing low GPNMB/OA) with recombinant GPNMB/OA protein (rOA) significantly facilitated cell invasion and migration, but the effects of rOA was negated by inclusion of a selective RGD peptide. Further studies in athymic (nu/nu) mice-bearing calu-6 showed that intratumoral supplementation with rOA effectively facilitated in vivo tumor growth as characterized by a high number of proliferating cells (Ki67 staining) coupled with a low number of apoptotic cells. Taken together, our results accentuate the relevance of GPNMB/OA ECD protein shedding to progression of lung cancer. Thus, strategies that suppress GPNMB/OA expression on lung cancer cells as well as negate shedding of GPNMB/OA ECD protein are worthy of consideration in lung cancer therapeutics.
- Published
- 2016
- Full Text
- View/download PDF
25. Growth and repair factors, osteoactivin, matrix metalloproteinase and heat shock protein 72, increase with resolution of inflammation in musculotendinous tissues in a rat model of repetitive grasping.
- Author
-
Frara N, Abdelmagid SM, Tytell M, Amin M, Popoff SN, Safadi FF, and Barbe MF
- Subjects
- Animals, Cumulative Trauma Disorders prevention & control, Disease Models, Animal, Female, Hand Strength physiology, Inflammation metabolism, Inflammation prevention & control, Intercellular Signaling Peptides and Proteins biosynthesis, Muscle, Skeletal pathology, Rats, Rats, Sprague-Dawley, Tendons pathology, Cumulative Trauma Disorders metabolism, HSP72 Heat-Shock Proteins biosynthesis, Matrix Metalloproteinases biosynthesis, Membrane Glycoproteins biosynthesis, Muscle, Skeletal metabolism, Tendons metabolism
- Abstract
Background: Expression of the growth factor osteoactivin (OA) increases during tissue degeneration and regeneration, fracture repair and after denervation-induced disuse atrophy, concomitant with increased matrix metalloproteinases (MMPs). However, OA's expression with repetitive overuse injuries is unknown. The aim of this study was to evaluate: 1) OA expression in an operant rat model of repetitive overuse; 2) expression of MMPs; 3) inflammatory cytokines indicative of injury or inflammation; and 4) the inducible form of heat shock protein 70 (HSPA1A/HSP72) as the latter is known to increase during metabolic stress and to be involved in cellular repair. Young adult female rats performed a high repetition negligible force (HRNF) food retrieval task for up to 6 weeks and were compared to control rats., Methods: Flexor digitorum muscles and tendons were collected from 22 young adult female rats performing a HRNF reaching task for 3 to 6 weeks, and 12 food restricted control (FRC) rats. OA mRNA levels were assessed by quantitative polymerase chain reaction (qPCR). OA, MMP-1, -2, -3, and -13 and HSP72 protein expression was assayed using Western blotting. Immunohistochemistry and image analysis was used to evaluate OA and HSP72 expression. ELISA was performed for HSP72 and inflammatory cytokines., Results: Flexor digitorum muscles and tendons from 6-week HRNF rats showed increased OA mRNA and protein expression compared to FRC rats. MMP-1, -2 and -3 progressively increased in muscles whereas MMP-1 and -3 increased in tendons with HRNF task performance. HSP72 increased in 6-week HRNF muscles and tendons, compared to controls, and co-localized with OA in the myofiber sarcolemma. IL-1alpha and beta increased transiently in tendons or muscles in HRNF week 3 before resolving in week 6., Conclusion: The simultaneous increases of OA with factors involved in tissue repair (MMPs and HSP72) supports a role of OA in tissue regeneration after repetitive overuse.
- Published
- 2016
- Full Text
- View/download PDF
26. Transgenic Expression of Osteoactivin/gpnmb Enhances Bone Formation In Vivo and Osteoprogenitor Differentiation Ex Vivo.
- Author
-
Frara N, Abdelmagid SM, Sondag GR, Moussa FM, Yingling VR, Owen TA, Popoff SN, Barbe MF, and Safadi FF
- Subjects
- Animals, Bone Density physiology, Bone Remodeling genetics, Bone Resorption metabolism, Cell Differentiation genetics, Eye Proteins genetics, Membrane Glycoproteins genetics, Mice, Transgenic, Osteogenesis genetics, Protein Serine-Threonine Kinases metabolism, Receptor, Transforming Growth Factor-beta Type I, Receptors, Transforming Growth Factor beta metabolism, Bone Remodeling physiology, Bone and Bones metabolism, Cell Differentiation physiology, Eye Proteins metabolism, Membrane Glycoproteins metabolism, Osteoblasts cytology, Osteoclasts cytology
- Abstract
Initial identification of osteoactivin (OA)/glycoprotein non-melanoma clone B (gpnmb) was demonstrated in an osteopetrotic rat model, where OA expression was increased threefold in mutant bones, compared to normal. OA mRNA and protein expression increase during active bone regeneration post-fracture, and primary rat osteoblasts show increased OA expression during differentiation in vitro. To further examine OA/gpnmb as an osteoinductive agent, we characterized the skeletal phenotype of transgenic mouse overexpressing OA/gpnmb under the CMV-promoter (OA-Tg). Western blot analysis showed increased OA/gpnmb in OA-Tg osteoblasts, compared to wild-type (WT). In OA-Tg mouse femurs versus WT littermates, micro-CT analysis showed increased trabecular bone volume and thickness, and cortical bone thickness; histomorphometry showed increased osteoblast numbers, bone formation and mineral apposition rates in OA-Tg mice; and biomechanical testing showed higher peak moment and stiffness. Given that OA/gpnmb is also over-expressed in osteoclasts in OA-Tg mice, we evaluated bone resorption by ELISA and histomorphometry, and observed decreased serum CTX-1 and RANK-L, and decreased osteoclast numbers in OA-Tg, compared to WT mice, indicating decreased bone remodeling in OA-Tg mice. The proliferation rate of OA-Tg osteoblasts in vitro was higher, compared to WT, as was alkaline phosphatase staining and activity, the latter indicating enhanced differentiation of OA-Tg osteoprogenitors. Quantitative RT-PCR analysis showed increased TGF-β1 and TGF-β receptors I and II expression in OA-Tg osteoblasts, compared to WT. Together, these data suggest that OA overexpression has an osteoinductive effect on bone mass in vivo and stimulates osteoprogenitor differentiation ex vivo., (© 2015 Wiley Periodicals, Inc.)
- Published
- 2016
- Full Text
- View/download PDF
27. Mutation in Osteoactivin Promotes Receptor Activator of NFκB Ligand (RANKL)-mediated Osteoclast Differentiation and Survival but Inhibits Osteoclast Function.
- Author
-
Abdelmagid SM, Sondag GR, Moussa FM, Belcher JY, Yu B, Stinnett H, Novak K, Mbimba T, Khol M, Hankenson KD, Malcuit C, and Safadi FF
- Subjects
- Animals, Bone Remodeling, Mice, Mice, Inbred DBA, RANK Ligand metabolism, Signal Transduction, X-Ray Microtomography, Cell Differentiation physiology, Cell Survival physiology, Eye Proteins genetics, Membrane Glycoproteins genetics, Mutation, Osteoclasts cytology, RANK Ligand physiology
- Abstract
We previously reported on the importance of osteoactivin (OA/Gpnmb) in osteogenesis. In this study, we examined the role of OA in osteoclastogenesis, using mice with a nonsense mutation in the Gpnmb gene (D2J) and wild-type controls (D2J/Gpnmb(+)). In these D2J mice, micro-computed tomography and histomorphometric analyses revealed increased cortical thickness, whereas total porosity and eroded surface were significantly reduced in D2J mice compared with wild-type controls, and these results were corroborated by lower serum levels of CTX-1. Contrary to these observations and counterintuitively, temporal gene expression analyses supported up-regulated osteoclastogenesis in D2J mice and increased osteoclast differentiation rates ex vivo, marked by increased number and size. The finding that MAPK was activated in early differentiating and mature D2J osteoclasts and that survival of D2J osteoclasts was enhanced and mediated by activation of the AKT-GSK3β pathway supports this observation. Furthermore, this was abrogated by the addition of recombinant OA to cultures, which restored osteoclastogenesis to wild-type levels. Moreover, mix and match co-cultures demonstrated an induction of osteoclastogenesis in D2J osteoblasts co-cultured with osteoclasts of D2J or wild-type. Last, in functional osteo-assays, we show that bone resorption activity of D2J osteoclasts is dramatically reduced, and these osteoclasts present an abnormal ruffled border over the bone surface. Collectively, these data support a model whereby OA/Gpnmb acts as a negative regulator of osteoclast differentiation and survival but not function by inhibiting the ERK/AKT signaling pathways., (© 2015 by The American Society for Biochemistry and Molecular Biology, Inc.)
- Published
- 2015
- Full Text
- View/download PDF
28. Integrin mediated adhesion of osteoblasts to connective tissue growth factor (CTGF/CCN2) induces cytoskeleton reorganization and cell differentiation.
- Author
-
Hendesi H, Barbe MF, Safadi FF, Monroy MA, and Popoff SN
- Subjects
- Animals, Cell Adhesion, Cell Line, Connective Tissue Growth Factor chemistry, Core Binding Factor Alpha 1 Subunit metabolism, Extracellular Signal-Regulated MAP Kinases metabolism, Focal Adhesion Protein-Tyrosine Kinases metabolism, Mice, Receptors, Vitronectin metabolism, Signal Transduction, Transcriptional Activation, rac GTP-Binding Proteins metabolism, Cell Differentiation, Connective Tissue Growth Factor metabolism, Cytoskeleton metabolism, Integrins metabolism, Osteoblasts cytology, Osteoblasts metabolism
- Abstract
Pre-osteoblast adhesion and interaction with extracellular matrix (ECM) proteins through integrin receptors result in activation of signaling pathways regulating osteoblast differentiation. Connective tissue growth factor (CTGF/CCN2) is a matricellular protein secreted into the ECM. Prior studies in various cell types have shown that cell adhesion to CTGF via integrin receptors results in activation of specific signaling pathways that regulate cell functions, such as differentiation and cytoskeletal reorganization. To date, there are no studies that have examined whether CTGF can serve as an adhesive substrate for osteoblasts. In this study, we used the MC3T3-E1 cell line to demonstrate that CTGF serves as an adhesive matrix for osteoblasts. Anti-integrin blocking experiments and co-immunoprecipitation assays demonstrated that the integrin αvβ1 plays a key role in osteoblast adhesion to a CTGF matrix. Immunofluorescence staining of osteoblasts cultured on a CTGF matrix confirmed actin cytoskeletal reorganization, enhanced spreading, formation of focal adhesions, and activation of Rac1. Alkaline phosphatase (ALP) staining and activity assays, as well as Alizarin red staining demonstrated that osteoblast attachment to CTGF matrix enhanced maturation, bone nodule formation and matrix mineralization. To investigate whether the effect of CTGF on osteoblast differentiation involves integrin-mediated activation of specific signaling pathways, we performed Western blot, chromatin immunoprecipitation (ChIP) and qPCR assays. Osteoblasts cultured on a CTGF matrix showed increased total and phosphorylated (activated) forms of focal adhesion kinase (FAK) and extracellular signal-regulated kinase (ERK). Inhibition of ERK blocked osteogenic differentiation in cells cultured on a CTGF matrix. There was an increase in runt-related transcription factor 2 (Runx2) binding to the osteocalcin gene promoter, and in the expression of osteogenic markers regulated by Runx2. Collectively, the results of this study are the first to demonstrate CTGF serves as a suitable matrix protein, enhancing osteoblast adhesion (via αvβ1 integrin) and promoting cell spreading via cytoskeletal reorganization and Rac1 activation. Furthermore, integrin-mediated activation of ERK signaling resulted in increased osteoblast differentiation accompanied by an increase in Runx2 binding to the osteocalcin promoter and in the expression of osteogenic markers.
- Published
- 2015
- Full Text
- View/download PDF
29. Role of inflammation in the aging bones.
- Author
-
Abdelmagid SM, Barbe MF, and Safadi FF
- Subjects
- Animals, Bone Marrow Cells physiology, Bone and Bones cytology, Bone and Bones immunology, Cumulative Trauma Disorders physiopathology, Humans, Aging physiology, Bone and Bones physiopathology, Cell Differentiation physiology, Inflammation physiopathology, Macrophages physiology, Models, Biological, Osteoblasts physiology, Osteoclasts physiology
- Abstract
Chronic inflammation in aging is characterized by increased inflammatory cytokines, bone loss, decreased adaptation, and defective tissue repair in response to injury. Aging leads to inherent changes in mesenchymal stem cell (MSC) differentiation, resulting in impaired osteoblastogenesis. Also, the pro-inflammatory cytokines increase with aging, leading to enhanced myelopoiesis and osteoclastogenesis. Bone marrow macrophages (BMMs) play pivotal roles in osteoblast differentiation, the maintenance of hematopoietic stem cells (HSCs), and subsequent bone repair. However, during aging, little is known about the role of macrophages in the differentiation and function of MSC and HSC. Aged mammals have higher circulating pro-inflammatory cytokines than young adults, supporting the hypothesis of increased inflammation with aging. This review will aid in the understanding of the potential role(s) of pro-inflammatory (M1) and anti-inflammatory (M2) macrophages in differentiation and function of osteoblasts and osteoclasts in relation to aging., (Copyright © 2014 Elsevier Inc. All rights reserved.)
- Published
- 2015
- Full Text
- View/download PDF
30. Parkinson's disease biomarker: a patent evaluation of WO2013153386.
- Author
-
Geldenhuys WJ, Abdelmagid SM, Gallegos PJ, and Safadi FF
- Subjects
- Biomarkers metabolism, Disease Progression, Dopaminergic Neurons pathology, Humans, Mitochondria pathology, Parkinson Disease physiopathology, Patents as Topic, Phosphorylation, Parkinson Disease diagnosis, Protein Kinases metabolism, Ubiquitin-Protein Ligases metabolism
- Abstract
Introduction: Parkinson's disease (PD) is a neurodegenerative movement disorder resultant from the loss of dopaminergic neurons in the brain. There is an urgent need for effective biomarkers that can be used in the early diagnosis of PD. Mitochondrial dysfunction plays a significant role in PD pathology, which has led to the evaluation of mitophagy markers, PTEN-induced putative kinase 1 (PINK1), and PARKIN as possible biomarkers for the early diagnosis of PD., Areas Covered: The current patent describes the use of phosphorylation of PINK1 and PARKIN as a diagnostic measure. Specifically, Ser65 on PARKIN, which is phosphorylated by PINK1, and the autophosphorylation of PINK1 at Thr257 are described., Expert Opinion: This patent describes a much needed methodology that can easily be adapted in the clinical setting by which a biological sample, such as serum or cerebrospinal fluid, is collected and analyzed for the phosphorylation markers. Here, the phosphorylation activity seen in PINK1 and PARKIN can differentiate between age-matched controls and PD patients. This patent presents a novel diagnostic measure in early PD, as well as determines which medications would have a beneficial effect on a patient's disease progression.
- Published
- 2014
- Full Text
- View/download PDF
31. Osteoactivin promotes osteoblast adhesion through HSPG and αvβ1 integrin.
- Author
-
Moussa FM, Hisijara IA, Sondag GR, Scott EM, Frara N, Abdelmagid SM, and Safadi FF
- Subjects
- 3T3 Cells, Actin Cytoskeleton physiology, Alkaline Phosphatase biosynthesis, Animals, Antibodies immunology, Cell Adhesion, Cell Differentiation, Cell Line, Cell Proliferation, Chlorates pharmacology, Extracellular Signal-Regulated MAP Kinases biosynthesis, Eye Proteins genetics, Eye Proteins immunology, Focal Adhesion Kinase 1 biosynthesis, Focal Adhesions, Heparin pharmacology, Membrane Glycoproteins genetics, Membrane Glycoproteins immunology, Mice, Mice, Inbred C57BL, Osteogenesis physiology, Protein Binding, Rats, Receptors, Vitronectin immunology, Recombinant Proteins, Eye Proteins metabolism, Heparan Sulfate Proteoglycans metabolism, Membrane Glycoproteins metabolism, Osteoblasts physiology, Receptors, Vitronectin metabolism
- Abstract
Osteoactivin (OA), also known as glycoprotein nmb (gpnmb) plays an important role in the regulation of osteoblast differentiation and function. OA induced osteoblast differentiation and function in vitro by stimulating alkaline phosphatase (ALP) activity, osteocalcin production, nodule formation, and matrix mineralization. Recent studies reported a role for OA in cell adhesion and integrin binding. In this study, we demonstrate that recombinant osteoactivin (rOA) as a matricellular protein stimulated adhesion, spreading and differentiation of MC3T3-E1 osteoblast-like cells through binding to αv β1 integrin and heparan sulfated proteoglycans (HSPGs). MC3T3-E1 cell adhesion to rOA was blocked by neutralizing anti-OA or anti-αv and β1 integrin antibodies. rOA stimulated-osteoblast adhesion was also inhibited by soluble heparin and sodium chlorate. Interestingly, rOA stimulated-osteoblast adhesion promoted an increase in FAK and ERK activation, resulting in the formation of focal adhesions, cell spreading and enhanced actin cytoskeleton organization. In addition, differentiation of primary osteoblasts was augmented on rOA coated-wells marked by increased alkaline phosphatase staining and activity. Taken together, these data implicate OA as a matricellular protein that stimulates osteoblast adhesion through binding to αv β1 integrin and cell surface HSPGs, resulting in increased cell spreading, actin reorganization, and osteoblast differentiation with emphasis on the positive role of OA in osteogenesis., (© 2014 Wiley Periodicals, Inc.)
- Published
- 2014
- Full Text
- View/download PDF
32. Osteoactivin induces transdifferentiation of C2C12 myoblasts into osteoblasts.
- Author
-
Sondag GR, Salihoglu S, Lababidi SL, Crowder DC, Moussa FM, Abdelmagid SM, and Safadi FF
- Subjects
- Alkaline Phosphatase biosynthesis, Animals, Cell Line, Core Binding Factor Alpha 1 Subunit biosynthesis, Eye Proteins metabolism, Focal Adhesion Protein-Tyrosine Kinases biosynthesis, Gene Expression Regulation, Developmental genetics, Humans, Membrane Glycoproteins metabolism, Mice, Myoblasts cytology, Osteoblasts cytology, Cell Transdifferentiation genetics, Eye Proteins genetics, Membrane Glycoproteins genetics, Myoblasts metabolism, Osteoblasts metabolism
- Abstract
Osteoactivin (OA) is a novel osteogenic factor important for osteoblast differentiation and function. Previous studies showed that OA stimulates matrix mineralization and transcription of osteoblast specific genes required for differentiation. OA plays a role in wound healing and its expression was shown to increase in post fracture calluses. OA expression was reported in muscle as OA is upregulated in cases of denervation and unloading stress. The regulatory mechanisms of OA in muscle and bone have not yet been determined. In this study, we examined whether OA plays a role in transdifferentiation of C2C12 myoblast into osteoblasts. Infected C2C12 with a retroviral vector overexpressing OA under the CMV promoter were able to transdifferentiate from myoblasts into osteoblasts. Immunofluorescence analysis showed that skeletal muscle marker MF-20 was severely downregulated in cells overexpressing OA and contained significantly less myotubes compared to uninfected control. C2C12 myoblasts overexpressing OA showed an increase in expression of bone specific markers such as alkaline phosphatase and alizarin red staining, and also showed an increase in Runx2 protein expression. We also detected increased levels of phosphorylated focal adhesion kinase (FAK) in C2C12 myoblasts overexpressing OA compared to control. Taken together, our results suggest that OA is able to induce transdifferentiation of myoblasts into osteoblasts through increasing levels of phosphorylated FAK., (© 2013 Wiley Periodicals, Inc.)
- Published
- 2014
- Full Text
- View/download PDF
33. Mutation in osteoactivin decreases bone formation in vivo and osteoblast differentiation in vitro.
- Author
-
Abdelmagid SM, Belcher JY, Moussa FM, Lababidi SL, Sondag GR, Novak KM, Sanyurah AS, Frara NA, Razmpour R, Del Carpio-Cano FE, and Safadi FF
- Subjects
- Alkaline Phosphatase metabolism, Animals, Animals, Newborn, Apoptosis, Bone and Bones metabolism, Bone and Bones pathology, Cell Differentiation genetics, Male, Mice, Mice, Inbred DBA, Mutation, Osteoblasts cytology, Phenotype, Receptors, Transforming Growth Factor beta metabolism, Transforming Growth Factor beta metabolism, Eye Proteins genetics, Membrane Glycoproteins genetics, Osteoblasts physiology, Osteocalcin genetics, Osteogenesis genetics, Signal Transduction
- Abstract
We have previously identified osteoactivin (OA), encoded by Gpnmb, as an osteogenic factor that stimulates osteoblast differentiation in vitro. To elucidate the importance of OA in osteogenesis, we characterized the skeletal phenotype of a mouse model, DBA/2J (D2J) with a loss-of-function mutation in Gpnmb. Microtomography of D2J mice showed decreased trabecular mass, compared to that in wild-type mice [DBA/2J-Gpnmb(+)/SjJ (D2J/Gpnmb(+))]. Serum analysis showed decreases in OA and the bone-formation markers alkaline phosphatase and osteocalcin in D2J mice. Although D2J mice showed decreased osteoid and mineralization surfaces, their osteoblasts were increased in number, compared to D2J/Gpnmb(+) mice. We then examined the ability of D2J osteoblasts to differentiate in culture, where their differentiation and function were decreased, as evidenced by low alkaline phosphatase activity and matrix mineralization. Quantitative RT-PCR analyses confirmed the decreased expression of differentiation markers in D2J osteoblasts. In vitro, D2J osteoblasts proliferated and survived significantly less, compared to D2J/Gpnmb(+) osteoblasts. Next, we investigated whether mutant OA protein induces endoplasmic reticulum stress in D2J osteoblasts. Neither endoplasmic reticulum stress markers nor endoplasmic reticulum ultrastructure were altered in D2J osteoblasts. Finally, we assessed underlying mechanisms that might alter proliferation of D2J osteoblasts. Interestingly, TGF-β receptors and Smad-2/3 phosphorylation were up-regulated in D2J osteoblasts, suggesting that OA contributes to TGF-β signaling. These data confirm the anabolic role of OA in postnatal bone formation., (Copyright © 2014 American Society for Investigative Pathology. Published by Elsevier Inc. All rights reserved.)
- Published
- 2014
- Full Text
- View/download PDF
34. Emerging lung cancer therapeutic targets based on the pathogenesis of bone metastases.
- Author
-
Oyewumi MO, Alazizi A, Wehrung D, Manochakian R, and Safadi FF
- Abstract
Lung cancer is the second most common cancer and the leading cause of cancer related mortality in both men and women. Each year, more people die of lung cancer than of colon, breast, and prostate cancers combined. It is widely accepted that tumor metastasis is a formidable barrier to effective treatment of lung cancer. The bone is one of the frequent metastatic sites for lung cancer occurring in a large number of patients. Bone metastases can cause a wide range of symptoms that could impair quality of life of lung cancer patients and shorten their survival. We strongly believe that molecular targets (tumor-related and bone microenvironment based) that have been implicated in lung cancer bone metastases hold great promise in lung cancer therapeutics. Thus, this paper discusses some of the emerging molecular targets that have provided insights into the cascade of metastases in lung cancer with the focus on bone invasion. It is anticipated that the information gathered might be useful in future efforts of optimizing lung cancer treatment strategies.
- Published
- 2014
- Full Text
- View/download PDF
35. Assembly of the prothrombinase complex on the surface of human foreskin fibroblasts: Implications for connective tissue growth factor.
- Author
-
Rico MC, Rough JJ, Manns JM, Del Carpio-Cano F, Safadi FF, Kunapuli SP, and DeLa Cadena RA
- Subjects
- Basic Helix-Loop-Helix Leucine Zipper Transcription Factors metabolism, Cell Line, Connective Tissue Growth Factor biosynthesis, Connective Tissue Growth Factor genetics, Factor V genetics, Factor VII metabolism, Factor Xa genetics, Fibroblasts enzymology, Foreskin cytology, Gene Expression, Humans, Male, Thrombin biosynthesis, Thromboplastin biosynthesis, Thromboplastin genetics, Thromboplastin metabolism, Transforming Growth Factor beta metabolism, Connective Tissue Growth Factor metabolism, Factor V metabolism, Factor Xa metabolism, Fibroblasts metabolism
- Abstract
Activated factor X (FXa) and thrombin can up-regulate gene expression of connective tissue growth factor (CTGF/CCN2) on fibroblasts. Since tissue factor (TF) is expressed on these cells, we hypothesized that they may assemble the prothrombinase complex leading to CTGF/CCN2 upregulation. In addition, the effect of thrombospondin-1 (TSP1) on this reaction was evaluated. Human foreskin fibroblasts were incubated with purified factor VII (FVII), factor X (FX), factor V (FV), prothrombin and calcium in the presence and absence of TSP1. Generation of FXa and of thrombin were assessed using chromogenic substrates. SMAD pathway phosphorylation was detected via Western-blot analysis. Pre-incubation of fibroblasts with FVII led to its auto-activation by cell-surface expressed TF, which in turn in the presence of FX, FVa, prothrombin and calcium led to FXa (9.7±0.8nM) and thrombin (7.9±0.04 U/mL×10-3) generation. Addition of TSP1 significantly enhanced thrombin (23.3±0.7 U/mL×10-3) but not FXa (8.5±0.6nM) generation. FXa and thrombin generation leads to upregulation of CTGF/CCN2. TSP1 alone upregulated CTGF/CCN2, an effect mediated via activation of transforming growth factor beta (TGFβ) as shown by phosphorylation of the SMAD pathway, an event blunted by using a TGFβ receptor I inhibitor (TGFβRI). FXa- and thrombin-induced upregulation of CTGF/CCN2 was not blocked by TGFβRI. In summary, assembly of the prothrombinase complex occurs on fibroblast's surface leading to serine proteases generation, an event enhanced by TSP1 and associated with CTGF/CCN2 upregulation. These mechanisms may play an important role in human diseases associated with fibrosis., (Copyright © 2011 Elsevier Ltd. All rights reserved.)
- Published
- 2012
- Full Text
- View/download PDF
36. Exploratory study on the effect of osteoactivin on bone formation in the rat critical-size calvarial defect model.
- Author
-
Bateman JP, Safadi FF, Susin C, and Wikesjö UM
- Subjects
- Animals, Bone Diseases pathology, Bone Regeneration drug effects, Craniotomy, Drug Carriers, Gelatin Sponge, Absorbable, Male, Random Allocation, Rats, Rats, Sprague-Dawley, Skull pathology, Time Factors, Bone Diseases drug therapy, Membrane Glycoproteins therapeutic use, Osteogenesis drug effects, Skull drug effects
- Abstract
Background and Objective: Osteoactivin is a novel glycoprotein shown to exhibit an important role in regulating osteoblast differentiation and function. The aim of the present study was to evaluate the potential of osteoactivin to support bone regeneration using an established defect model., Material and Methods: Critical-size, 8-mm-diameter through-and-through calvarial osteotomy defects were created in 60 adult male Sprague-Dawley rats. Test animals received 0.1 mL of osteoactivin in phosphate-buffered saline (50 μg/mL) soak-loaded onto an absorbable collagen sponge. Controls received 0.1 mL of phosphate-buffered saline soak-loaded onto the absorbable collagen sponge or no further intervention (sham-surgery). The animals were euthanized 2 and 4 wk after treatment and histometric analyses were performed., Results: The absorbable collagen sponge control (mean ± standard deviation: 40.9 ± 26.9%) showed borderline significant greater bone fill compared with sham-surgery (22.9 ± 15.8%; p = 0.10) and osteoactivin (20.2 ± 11.8%; p = 0.07) treatments at 2 wk. In contrast, osteoactivin (84.7 ± 15.8%) showed significantly greater bone fill than sham-surgery (28.4 ± 9.6%; p < 0.001) and absorbable collagen sponge (41.8 ± 22.1%; p < 0.001) at 4 wk. No animals receiving sham-surgery or absorbable collagen sponge exhibited complete bone fill at 4 wk while 70% of the animals receiving osteoactivin showed complete bone fill., Conclusion: Osteoactivin demonstrates a significant potential to support bone regeneration/formation. Studies using discriminating large animal models are necessary to explore clinical application for periodontal and craniofacial indications., (© 2011 John Wiley & Sons A/S.)
- Published
- 2012
- Full Text
- View/download PDF
37. Performance of repetitive tasks induces decreased grip strength and increased fibrogenic proteins in skeletal muscle: role of force and inflammation.
- Author
-
Abdelmagid SM, Barr AE, Rico M, Amin M, Litvin J, Popoff SN, Safadi FF, and Barbe MF
- Subjects
- Animals, Anti-Inflammatory Agents, Non-Steroidal pharmacology, Collagen Type I metabolism, Connective Tissue Growth Factor metabolism, Female, Fibrosis, Forelimb drug effects, Forelimb metabolism, Forelimb physiopathology, Ibuprofen pharmacology, Inflammation metabolism, Rats, Rats, Sprague-Dawley, Transforming Growth Factor beta1 metabolism, Hand Strength, Muscle, Skeletal metabolism
- Abstract
Background: This study elucidates exposure-response relationships between performance of repetitive tasks, grip strength declines, and fibrogenic-related protein changes in muscles, and their link to inflammation. Specifically, we examined forearm flexor digitorum muscles for changes in connective tissue growth factor (CTGF; a matrix protein associated with fibrosis), collagen type I (Col1; a matrix component), and transforming growth factor beta 1 (TGFB1; an upstream modulator of CTGF and collagen), in rats performing one of two repetitive tasks, with or without anti-inflammatory drugs., Methodology/results: To examine the roles of force versus repetition, rats performed either a high repetition negligible force food retrieval task (HRNF), or a high repetition high force handle-pulling task (HRHF), for up to 9 weeks, with results compared to trained only (TR-NF or TR-HF) and normal control rats. Grip strength declined with both tasks, with the greatest declines in 9-week HRHF rats. Quantitative PCR (qPCR) analyses of HRNF muscles showed increased expression of Col1 in weeks 3-9, and CTGF in weeks 6 and 9. Immunohistochemistry confirmed PCR results, and also showed greater increases of CTGF and collagen matrix in 9-week HRHF rats than 9-week HRNF rats. ELISA, and immunohistochemistry revealed greater increases of TGFB1 in TR-HF and 6-week HRHF, compared to 6-week HRNF rats. To examine the role of inflammation, results from 6-week HRHF rats were compared to rats receiving ibuprofen or anti-TNF-α treatment in HRHF weeks 4-6. Both treatments attenuated HRHF-induced increases in CTGF and fibrosis by 6 weeks of task performance. Ibuprofen attenuated TGFB1 increases and grip strength declines, matching our prior results with anti-TNFα., Conclusions/significance: Performance of highly repetitive tasks was associated with force-dependent declines in grip strength and increased fibrogenic-related proteins in flexor digitorum muscles. These changes were attenuated, at least short-term, by anti-inflammatory treatments.
- Published
- 2012
- Full Text
- View/download PDF
38. Homeodomain transcription factors regulate BMP-2-induced osteoactivin transcription in osteoblasts.
- Author
-
Singh M, Del Carpio-Cano FE, Monroy MA, Popoff SN, and Safadi FF
- Subjects
- Animals, Blotting, Western, Bone Morphogenetic Protein 2 genetics, Cells, Cultured, Chromatin Immunoprecipitation, Electrophoretic Mobility Shift Assay, Homeodomain Proteins genetics, Membrane Glycoproteins genetics, Promoter Regions, Genetic genetics, RNA, Small Interfering, Rats, Transcription Factors genetics, Transcription, Genetic, Transcriptional Activation genetics, Bone Morphogenetic Protein 2 metabolism, Cell Differentiation genetics, Homeodomain Proteins metabolism, Membrane Glycoproteins biosynthesis, Osteoblasts cytology, Transcription Factors metabolism
- Abstract
Osteoactivin (OA) is required for the differentiation of osteoblast cells. OA expression is stimulated by bone morphogenetic protein-2 (BMP-2). BMP-2 recruits homeodomain transcription factors Dlx3, Dlx5, and Msx2 to selectively activate or repress transcription of osteogenic genes and hence tightly regulate their transcription during osteoblast differentiation. Considering the key roles of Dlx3, Dlx5, and Msx2 in osteoblast differentiation, here we hypothesize that homeodomain proteins regulate BMP-2-induced OA transcription during osteoblast differentiation. Four classical homeodomain binding sites were identified in the proximal 0.96 kb region of rat OA promoter. Deletions and mutagenesis studies of the OA promoter region indicated that all four homeodomain binding sites are crucial for BMP-2-induced OA promoter activity. Simultaneous disruption of homeodomain binding sites at -852 and -843 of the transcription start site of OA gene significantly decreased the BMP-2-induced OA transcription and inhibited binding of Dlx3, Dlx5, and Msx2 proteins to the OA promoter. Dlx3 and Dlx5 proteins were found to activate the OA transcription, whereas, Msx2 suppressed BMP-2-induced OA transcription. Using chromatin immunoprecipitation assays, we demonstrated that the OA promoter is predominantly occupied by Dlx3 and Dlx5 during the proliferation and matrix maturation stages of osteoblast differentiation, respectively. During the matrix mineralization stage, BMP-2 robustly enhanced the recruitment of Dlx5 and to a lesser extent of Dlx3 and Msx2 to the OA promoter region. Collectively, our results show that the BMP-2-induced OA transcription is differentially regulated by Dlx3, Dlx5, and Msx2 during osteoblast differentiation., (Copyright © 2011 Wiley Periodicals, Inc.)
- Published
- 2012
- Full Text
- View/download PDF
39. Comprehensive characterization of mesenchymal stem cells from human placenta and fetal membrane and their response to osteoactivin stimulation.
- Author
-
Raynaud CM, Maleki M, Lis R, Ahmed B, Al-Azwani I, Malek J, Safadi FF, and Rafii A
- Abstract
Mesenchymal stem cells (MSCs) are the most promising seed cells for cell therapy and can be isolated from various sources of human adult tissues such as bone marrow (BM-MSC) and adipose tissue. However, cells from these tissues must be obtained through invasive procedures. We, therefore, characterized MSCs isolated from fresh placenta (Pl-MSC) and fetal membrane (Mb-MSC) through morphological and fluorescent-activated cell sorting (FACS). MSC frequency is higher in membrane than placenta (2.14% ± 0.65 versus 15.67% ± 0.29%). Pl/Mb-MSCs in vitro expansion potential was significantly higher than BM-MSCs. We demonstrated that one of the MSC-specific marker is sufficient for MSC isolation and that culture in specific media is the optimal way for selecting very homogenous MSC population. These MSCs could be differentiated into mesodermal cells expressing cell markers and cytologic staining consistent with mature osteoblasts and adipocytes. Transcriptomic analysis and cytokine arrays demonstrated broad similarity between placenta- and membrane-derived MSCs and only discrete differences with BM-MSCs with enrichment of networks involved in bone differentiation. Pl/Mb-MSCs displayed higher osteogenic differentiation potential than BM-MSC when their response to osteoactivin was evaluated. Fetal-tissue-derived mesenchymal cells may, therefore, be considered as a major source of MSCs to reach clinical scale banking in particular for bone regeneration.
- Published
- 2012
- Full Text
- View/download PDF
40. Comparison of bone morphogenetic protein-2 and osteoactivin for mesenchymal cell differentiation: effects of bolus and continuous administration.
- Author
-
Arosarena OA, Del Carpio-Cano FE, Dela Cadena RA, Rico MC, Nwodim E, and Safadi FF
- Subjects
- Alkaline Phosphatase biosynthesis, Alkaline Phosphatase genetics, Animals, Cell Differentiation genetics, Cell Line, Delayed-Action Preparations, Gene Expression drug effects, Hydrogel, Polyethylene Glycol Dimethacrylate administration & dosage, Mesenchymal Stem Cells cytology, Mice, Osteocalcin biosynthesis, Osteocalcin genetics, Bone Morphogenetic Protein 2 administration & dosage, Cell Differentiation drug effects, Eye Proteins administration & dosage, Membrane Glycoproteins administration & dosage, Mesenchymal Stem Cells drug effects
- Abstract
Current osteoinductive protein therapy utilizes bolus administration of large doses of bone morphogenetic proteins (BMPs), which is costly, and may not replicate normal bone healing. The limited in vivo biologic activity of BMPs requires the investigation of growth factors that may enhance this activity. In this study, we utilized the C3H10T1/2 murine mesenchymal stem cell line to test the hypotheses that osteoactivin (OA) has comparable osteoinductive effects to bone morphogenetic protein-2 (BMP-2), and that sustained administration of either growth factor would result in increased osteoblastic differentiation as compared to bolus administration. Sustained release biodegradable hydrogels were designed, and C3H10T1/2 cells were grown on hydrogels loaded with BMP-2 or OA. Controls were grown on unloaded hydrogels, and positive controls were exposed to bolus growth factor administration. Cells were harvested at several time points to assess osteoblastic differentiation. Alkaline phosphatase (ALP) staining and activity, and gene expression of ALP and osteocalcin were assessed. Treatment with OA or BMP-2 resulted in comparable effects on osteoblastic marker expression. However, cells grown on hydrogels demonstrated osteoblastic differentiation that was not as robust as cells treated with bolus administration. This study shows that OA has comparable effects to BMP-2 on osteoblastic differentiation using both bolus administration and continuous release, and that bolus administration of OA has a more profound effect than administration using hydrogels for sustained release. This study will lead to a better understanding of appropriate delivery methods of osteogenic growth factors like OA for repair of fractures and segmental bone defects., (Copyright © 2011 Wiley-Liss, Inc.)
- Published
- 2011
- Full Text
- View/download PDF
41. Different effects on bone strength and cell differentiation in pre pubertal caloric restriction versus hypothalamic suppression.
- Author
-
Joshi RN, Safadi FF, Barbe MF, Del Carpio-Cano F, Popoff SN, and Yingling VR
- Subjects
- Animals, Body Weight physiology, Bone and Bones diagnostic imaging, Cell Proliferation, Female, Gonadotropin-Releasing Hormone antagonists & inhibitors, Growth and Development, Insulin-Like Growth Factor I metabolism, Lumbar Vertebrae diagnostic imaging, Organ Size, Osteoblasts metabolism, Osteocalcin blood, Rats, Rats, Sprague-Dawley, Uterus anatomy & histology, X-Ray Microtomography, Bone and Bones physiology, Caloric Restriction, Cell Differentiation, Hypothalamus metabolism, Osteoblasts cytology, Sexual Maturation physiology
- Abstract
Hypothalamic amenorrhea and energy restriction during puberty affect peak bone mass accrual. One hypothesis suggests energy restriction alters hypothalamic function resulting in suppressed estradiol levels leading to bone loss. However, both positive and negative results have been reported regarding energy restriction and bone strength. Therefore, the purpose of this study was to investigate energy restriction and hypothalamic suppression during pubertal onset on bone mechanical strength and the osteogenic capacity of bone marrow-derived cells in two models: female rats treated with gonadotropin releasing hormone antagonists (GnRH-a) or 30% energy restriction. At 23 days of age, female Sprague Dawley rats were assigned to three groups: control group (C, n=10), GnRH-a group (n=10), and Energy Restriction (ER, n=12) group. GnRH-a animals received daily injections for 27 days. The animals in the ER group received 70% of the control animals' intake. After sacrifice (50 days of age), body weight, uterine and muscle weights were measured. Bone marrow-derived stromal cells were cultured and assayed for proliferation and differentiation into osteoblasts. Outcome measures included bone strength, bone histomorphometry and architecture, serum IGF-1 and osteocalcin. GnRH-a suppressed uterine weight, decreased osteoblast proliferation, bone strength, trabecular bone volume and architecture compared to control. Elevated serum IGF-1 and osteocalcin levels and body weight were found. The ER model had an increase in osteoblast proliferation compared to the GnRH-a group, similar bone strength relative to body weight and increased trabecular bone volume in the lumbar spine compared to control. The ER animals were smaller but had developed bone strength sufficient for their size. In contrast, suppressed estradiol via hypothalamic suppression resulted in bone strength deficits and trabecular bone volume loss. In summary, our results support the hypothesis that during periods of nutritional stress the increased vertebral bone volume may be an adaptive mechanism to store mineral which differs from suppressed estradiol resulting from hypothalamic suppression., (Copyright © 2011 Elsevier Inc. All rights reserved.)
- Published
- 2011
- Full Text
- View/download PDF
42. The role of connective tissue growth factor (CTGF/CCN2) in skeletogenesis.
- Author
-
Arnott JA, Lambi AG, Mundy C, Hendesi H, Pixley RA, Owen TA, Safadi FF, and Popoff SN
- Subjects
- Animals, Bone Development genetics, Chondrogenesis genetics, Chondrogenesis physiology, Connective Tissue Growth Factor genetics, Eptifibatide, Gene Expression Regulation, Developmental, Humans, Mice, Mice, Transgenic, Neovascularization, Physiologic genetics, Neovascularization, Physiologic physiology, Osteogenesis genetics, Osteogenesis physiology, Transforming Growth Factor beta genetics, Transforming Growth Factor beta metabolism, Bone Development physiology, Connective Tissue Growth Factor physiology, Peptides metabolism
- Abstract
Connective tissue growth factor (CTGF) is a 38 kDa, cysteine rich, extracellular matrix protein composed of 4 domains or modules. CTGF has been shown to regulate a diverse array of cellular functions and has been implicated in more complex biological processes such as angiogenesis, chondrogenesis, and osteogenesis. A role for CTGF in the development and maintenance of skeletal tissues first came to light in studies demonstrating its expression in cartilage and bone cells, which was dramatically increased during skeletal repair or regeneration. The physiological significance of CTGF in skeletogenesis was confirmed in CTGF-null mice, which exhibited multiple skeletal dysmorphisms as a result of impaired growth plate chondrogenesis, angiogenesis, and bone formation/mineralization. Given the emerging importance of CTGF in osteogenesis and chondrogenesis, this review will focus on its expression in skeletal tissues, its effects on osteoblast and chondrocyte differentiation and function, and the skeletal implications of ablation or over-expression of CTGF in knockout or transgenic mouse models, respectively. In addition, this review will examine the role of integrin-mediated signaling and the regulation of CTGF expression as it relates to skeletogenesis. We will emphasize CTGF studies in bone or bone cells, and will identify opportunities for future investigations concerning CTGF and chondrogenesis/osteogenesis.
- Published
- 2011
- Full Text
- View/download PDF
43. Temporal and spatial expression of osteoactivin during fracture repair.
- Author
-
Abdelmagid SM, Barbe MF, Hadjiargyrou M, Owen TA, Razmpour R, Rehman S, Popoff SN, and Safadi FF
- Subjects
- Animals, Chondrogenesis genetics, Eye Proteins biosynthesis, Growth Plate metabolism, Membrane Glycoproteins biosynthesis, Osteoblasts metabolism, RNA, Messenger analysis, Rats, Time Factors, Eye Proteins analysis, Eye Proteins genetics, Femur injuries, Fracture Healing genetics, Membrane Glycoproteins analysis, Membrane Glycoproteins genetics
- Abstract
We previously identified osteoactivin (OA) as a novel secreted osteogenic factor with high expression in developing long bones and calvaria, and that stimulates osteoblast differentiation and matrix mineralization in vitro. In this study, we report on OA mRNA and protein expression in intact long bone and growth plate, and in fracture calluses collected at several time points up to 21 days post-fracture (PF). OA mRNA and protein were highly expressed in osteoblasts localized in the metaphysis of intact tibia, and in hypertrophic chondrocytes localized in growth plate, findings assessed by in situ hybridization and immunohistochemistry, respectively. Using a rat fracture model, Northern blot analysis showed that expression of OA mRNA was significantly higher in day-3 and day-10 PF calluses than in intact rat femurs. Using in situ hybridization, we examined OA mRNA expression during fracture healing and found that OA was temporally regulated, with positive signals seen as early as day-3 PF, reaching a maximal intensity at day-10 PF, and finally declining at day-21 PF. At day-5 PF, which correlates with chondrogenesis, OA mRNA levels were significantly higher in the soft callus than in intact femurs. Similarly, we detected high OA protein immunoexpression throughout the reparative phase of the hard callus compared to intact femurs. Interestingly, the secreted OA protein was also detected within the newly made cartilage matrix and osteoid tissue. Taken together, these results suggest the possibility that OA plays an important role in bone formation and serves as a positive regulator of fracture healing., (© 2010 Wiley-Liss, Inc.)
- Published
- 2010
- Full Text
- View/download PDF
44. Src is a major signaling component for CTGF induction by TGF-beta1 in osteoblasts.
- Author
-
Zhang X, Arnott JA, Rehman S, Delong WG Jr, Sanjay A, Safadi FF, and Popoff SN
- Subjects
- Animals, Cell Line, Connective Tissue Growth Factor genetics, Enzyme Activation, Enzyme Inhibitors metabolism, Extracellular Signal-Regulated MAP Kinases metabolism, Flavonoids metabolism, Osteoblasts cytology, Promoter Regions, Genetic, RNA, Small Interfering genetics, RNA, Small Interfering metabolism, Rats, Rats, Sprague-Dawley, Smad Proteins metabolism, src-Family Kinases antagonists & inhibitors, src-Family Kinases genetics, Connective Tissue Growth Factor metabolism, Osteoblasts physiology, Signal Transduction physiology, Transforming Growth Factor beta1 metabolism, src-Family Kinases metabolism
- Abstract
Connective tissue growth factor (CTGF/CCN2) is induced by transforming growth factor beta1 (TGF-beta1) where it acts as a downstream mediator of TGF-beta1 induced matrix production in osteoblasts. We have shown the requirement of Src, Erk, and Smad signaling for CTGF induction by TGF-beta1 in osteoblasts; however, the potential interaction among these signaling pathways remains undetermined. In this study we demonstrate that TGF-beta1 activates Src kinase in ROS17/2.8 cells and that treatment with the Src family kinase inhibitor PP2 prevents Src activation and CTGF induction by TGF-beta1. Additionally, inhibiting Src activation prevented Erk activation, Smads 2 and 3 activation and nuclear translocation by TGF-beta1, demonstrating that Src is an essential upstream signaling partner of both Erk and Smads in osteoblasts. MAPKs such as Erk can modulate the Smad pathway directly by mediating the phosphorylation of Smads or indirectly through activation/inactivation of required nuclear co-activators that mediate Smad DNA binding. When we treated cells with the Erk inhibitor, PD98059, it inhibited TGF-beta1-induced CTGF protein expression but had no effect on Src activation, Smad activation or Smad nuclear translocation. However PD98059 impaired transcriptional complex formation on the Smad binding element (SBE) of the CTGF promoter, demonstrating that Erk activation was required for SBE transactivation. These data demonstrate that Src is an essential upstream signaling transducer of Erk and Smad signaling with respect to TGF-beta1 in osteoblasts and that Smads and Erk function independently but are both essential for forming a transcriptionally active complex on the CTGF promoter in osteoblasts., ((c) 2010 Wiley-Liss, Inc.)
- Published
- 2010
- Full Text
- View/download PDF
45. The chromatin remodeling factor SRCAP modulates expression of prostate specific antigen and cellular proliferation in prostate cancer cells.
- Author
-
Slupianek A, Yerrum S, Safadi FF, and Monroy MA
- Subjects
- Cell Line, Tumor, Cell Proliferation, Gene Knockdown Techniques, Histones metabolism, Humans, Immunohistochemistry, Male, Promoter Regions, Genetic genetics, Prostate metabolism, Prostate pathology, Prostate-Specific Antigen metabolism, Protein Binding, RNA Interference, RNA, Small Interfering metabolism, Receptors, Androgen genetics, Transcriptional Activation genetics, Adenosine Triphosphatases metabolism, Chromatin Assembly and Disassembly genetics, Gene Expression Regulation, Neoplastic, Prostate-Specific Antigen genetics, Prostatic Neoplasms genetics, Prostatic Neoplasms pathology
- Abstract
The SNF2-related CBP activator protein (SRCAP) serves as a coactivator for several nuclear receptors including the androgen receptor (AR). SRCAP is an ATPase that is the core subunit of a large multiprotein complex and was shown to incorporate the histone variant H2A.Z into nucleosomes. In this report, we demonstrate that SRCAP is expressed in the epithelium of normal prostate and in prostate carcinoma cells, and is associated with AR in the nucleus. Using transient transfection assays we demonstrate that SRCAP activates hormone-dependent transcription of the androgen responsive, prostate specific antigen (PSA)-Luciferase reporter gene in human prostate cells. The in vivo occupancy of SRCAP at the endogenous PSA promoter is demonstrated using chromatin immunoprecipitation assays. ShRNA mediated knockdown of SRCAP resulted in decreased H2A.Z binding at the enhancer region of the PSA promoter and decreased expression of PSA in prostate cancer cells. Furthermore, inhibition of SRCAP expression significantly inhibited androgen dependent prostate cancer cell growth. These data identify SRCAP as a physiologically relevant mediator of PSA expression, and demonstrate that SRCAP plays a role in prostate cancer cell proliferation.
- Published
- 2010
- Full Text
- View/download PDF
46. Functional roles of osteoactivin in normal and disease processes.
- Author
-
Singh M, Del Carpio-Cano F, Belcher JY, Crawford K, Frara N, Owen TA, Popoff SN, and Safadi FF
- Subjects
- Animals, Eye Proteins chemistry, Eye Proteins genetics, Humans, Inflammation physiopathology, Liver physiology, Membrane Glycoproteins chemistry, Membrane Glycoproteins genetics, Muscle, Skeletal physiology, Neoplasms physiopathology, Protein Processing, Post-Translational, Structural Homology, Protein, Bone and Bones physiology, Eye Proteins physiology, Membrane Glycoproteins physiology
- Abstract
Osteoactivin (OA) protein was discovered in bone cells a decade ago. Recent literature suggests that osteoactivin is crucial for the differentiation and functioning of different cell types, including bone-forming osteoblasts and bone-resorbing osteoclast cells. Here, we review the literature to date on various regulatory functions of osteoactivin, as well as its discovery, structure, expression, and function in different tissues and cells. The transcriptional regulation of osteoactivin and its mechanism of action in normal and diseased conditions with special emphasis on bone are also covered in this review. In addition, we touch on the therapeutic potential of osteoactivin in cancer and bone diseases.
- Published
- 2010
- Full Text
- View/download PDF
47. Periostin-like-factor in osteogenesis.
- Author
-
Zhu S, Barbe MF, Liu C, Hadjiargyrou M, Popoff SN, Rani S, Safadi FF, and Litvin J
- Subjects
- Animals, Animals, Newborn, Calcification, Physiologic, Calcium metabolism, Cell Adhesion Molecules genetics, Cell Differentiation, Cell Proliferation, Femur metabolism, Femur pathology, Fracture Healing, Fractures, Bone metabolism, Fractures, Bone physiopathology, Gene Expression Regulation, Muscle Proteins genetics, Osteoblasts cytology, Osteoblasts metabolism, Periosteum cytology, Periosteum metabolism, Rats, Rats, Inbred Lew, Wound Healing, Cell Adhesion Molecules metabolism, Muscle Proteins metabolism, Osteogenesis
- Abstract
Periostin-like-factor (PLF), an isoform related to Periostin, is expressed in bone, heart, and vascular smooth muscle cells. PLF was detected by immunostaining in mesenchymal cells in the periosteum and in osteoblasts lining trabecular bone, suggesting that PLF has a role in osteogenesis. PLF has a signal peptide and is also secreted from osteoblasts in vitro. To study the function of PLF in osteogenesis, we assessed the effect of PLF on osteoblast proliferation and differentiation in vitro and bone formation in vivo. First, to examine whether PLF regulates osteoblast proliferation in vitro, the CyQUANT cell proliferation assay was performed. PLF over-expression by adenovirus resulted in a significantly higher rate of cell proliferation compared to controls. This finding suggests that PLF promotes osteoblast proliferation in vitro. Second, to test whether PLF mediates osteoblast differentiation in vitro, differentiation markers of osteoblasts, were assessed, including alkaline phosphatase staining and activity, von Kossa staining and calcium deposition. Over-expression of PLF resulted in higher expression and activity of alkaline phosphatase and higher amounts of mineralization and calcium deposition compared to controls. These data suggest that PLF promotes osteoblast differentiation in vitro. Third, to investigate the role of PLF in bone formation in vivo, PLF adenovirus was injected into 6-week-old rat femur bone marrow. Over-expression of PLF resulted in increased bone formation within the marrow cavity. Lastly, in a model of fracture healing, PLF expression is robustly upregulated in callus osteoblasts at post-fracture days 7 and 14. Taken together, these findings suggests that PLF induces bone formation in vivo. We conclude that PLF stimulates bone formation in vivo possibly by promoting osteoblast proliferation and differentiation.
- Published
- 2009
- Full Text
- View/download PDF
48. Serum and tissue cytokines and chemokines increase with repetitive upper extremity tasks.
- Author
-
Barbe MF, Elliott MB, Abdelmagid SM, Amin M, Popoff SN, Safadi FF, and Barr AE
- Subjects
- Animals, Cumulative Trauma Disorders pathology, Cumulative Trauma Disorders physiopathology, Disease Models, Animal, Female, Forelimb, Hand Strength physiology, Macrophages pathology, Muscle, Skeletal pathology, Rats, Rats, Sprague-Dawley, Tendon Injuries metabolism, Tendon Injuries pathology, Tendon Injuries physiopathology, Tendons pathology, Chemokines blood, Cumulative Trauma Disorders blood, Muscle, Skeletal metabolism, Tendons metabolism
- Abstract
We investigated inflammation in rats performing a low repetition, negligible force (LRNF) or high repetition, negligible force (HRNF) task of reaching and retrieving food pellets at target rates of two or four reaches/min for 2 h/day, for 6-8 weeks. Serum was assayed for 11 cytokines and chemokines; forelimb tissues for four cytokines. Macrophages were counted in forelimb tissues of LRNF rats to add to results from our previous studies of HRNF rats. In HRNF rats, serum IL-1 alpha, IL-1 beta, TNFalpha, MIP2, MIP3a, and RANTES were elevated in weeks 6 and 8. In contrast, only MIP2 and MIP3a increased in serum of LRNF rats. In 8 week HRNF reach limb tissues, IL-1 alpha, IL-1beta, TNFalpha, and IL-10 increased in distal bones, IL-1 alpha and -beta in muscles, and TNFalpha in tendons. Only IL-10 increased in LRNF reach limb muscles in week 8. Serum IL-1 alpha and MIP2 correlated with macrophages in LRNF loose connective tissues, serum MIP3a and MIP2 correlated negatively with grip strength, while serum TNFalpha, MIP3a, and MIP2 correlated positively with total number of reaches. Thus, several tissue and circulating cytokines/chemokines increase in an exposure dependent manner following short-term performance of repetitive reaching tasks and correlate with macrophage infiltration and decreasing grip strength., ((c) 2008 Orthopaedic Research Society.)
- Published
- 2008
- Full Text
- View/download PDF
49. Osteoactivin, an anabolic factor that regulates osteoblast differentiation and function.
- Author
-
Abdelmagid SM, Barbe MF, Rico MC, Salihoglu S, Arango-Hisijara I, Selim AH, Anderson MG, Owen TA, Popoff SN, and Safadi FF
- Subjects
- Amino Acid Sequence, Animals, Animals, Newborn, Cells, Cultured, Eye Proteins genetics, Eye Proteins metabolism, Gene Expression Profiling, Glycosylation drug effects, Membrane Glycoproteins genetics, Membrane Glycoproteins metabolism, Metabolism genetics, Mice, Mice, Inbred DBA, Mice, Knockout, Models, Biological, Osteoblasts metabolism, Osteogenesis genetics, Protein Transport, Rats, Sequence Homology, Amino Acid, Tretinoin pharmacology, Cell Differentiation genetics, Eye Proteins physiology, Membrane Glycoproteins physiology, Osteoblasts physiology
- Abstract
Osteoactivin (OA) is a novel glycoprotein that is highly expressed during osteoblast differentiation. Using Western blot analysis, our data show that OA protein has two isoforms, one is transmembranous and the other is secreted into the conditioned medium of primary osteoblasts cultures. Fractionation of osteoblast cell compartments showed that the mature, glycosylated OA isoform of 115 kDa is found in the membranous fraction. Both OA isoforms (secreted and transmembrane) are found in the cytoplasmic fraction of osteoblasts. Overexpression of EGFP-tagged OA in osteoblasts showed that OA protein accumulates into vesicles for transportation to the cell membrane. We examined OA protein production in primary osteoblast cultures and found that OA is maximally expressed during the third week of culture (last stage of osteoblast differentiation). Glycosylation studies showed that OA isoform of 115 kDa is highly glycosylated. We also showed that retinoic acid (RA) stimulates the mannosylation of OA protein. In contrast, tunicamycin (TM) strongly inhibited N-glycans incorporation into OA protein. The functional role of the secreted OA isoform was revealed when cultures treated with anti-OA antibody, showed decreased osteoblast differentiation compared to untreated control cultures. Gain-of-function in osteoblasts using the pBABE viral system showed that OA overexpression in osteoblast stimulated their differentiation and function. The availability of a naturally occurring mutant mouse with a truncated OA protein provided further evidence that OA is an important factor for terminal osteoblast differentiation and mineralization. Using bone marrow mesenchymal cells derived from OA mutant and wild-type mice and testing their ability to differentiate into osteoblasts showed that differentiation of OA mutant osteoblasts was significantly reduced compared to wild-type osteoblasts. Collectively, our data suggest that OA acts as a positive regulator of osteoblastogenesis.
- Published
- 2008
- Full Text
- View/download PDF
50. Histopathological observations of a polylactic acid-based device intended for guided bone/tissue regeneration.
- Author
-
Polimeni G, Koo KT, Pringle GA, Agelan A, Safadi FF, and Wikesjö UM
- Subjects
- Animals, Bone Resorption pathology, Foreign-Body Reaction pathology, Guided Tissue Regeneration adverse effects, Guided Tissue Regeneration methods, Longitudinal Studies, Male, Osseointegration drug effects, Polyesters, Rats, Rats, Wistar, Skull drug effects, Skull pathology, Skull surgery, Time Factors, Absorbable Implants adverse effects, Bone Resorption chemically induced, Foreign-Body Reaction chemically induced, Lactic Acid adverse effects, Membranes, Artificial, Polymers adverse effects
- Abstract
Background: Barrier devices have been shown to support alveolar bone and periodontal regeneration, a procedure also known as guided bone/tissue regeneration (GBR/GTR). Popular demand and clinical convenience have raised an interest in bioresorbable barrier devices. Tissue reactions to such bioresorbable devices are, however, generally not well explored., Purpose: The objective of this study was to evaluate short- and long-term tissue reactions following implantation of a bioresorbable polylactic acid (PLA)-based barrier device using a rat model., Materials and Methods: Twenty-one young adult male Sprague-Dawley rats were used. The animals were divided into three groups including 15 animals receiving the PLA device and animals serving as sham surgery (five) or nonoperated (one) controls. Using aseptic techniques, the PLA device was surgically implanted in direct contact with the calvarial bone. Animals receiving the PLA device were sacrificed at 3, 5, 7, and 12 months postsurgery to provide longitudinal histopathological observations of tissue and biomaterials reactions. Control animals were sacrificed at 3 months., Results: Animals were maintained without adverse events. Sham surgery and nonoperated control animals showed no signs of new bone formation or resorption, or signs of inflammatory reactions in adjoining soft tissues. In contrast, extensive amounts of residual biomaterial with evidence of foreign body reactions and bone resorption were observed in animals receiving the PLA device over 12 months., Conclusions: The results suggest that the PLA device may induce bone resorbing foreign body reactions. Importantly, the PLA device does not resorb within a 12-month healing interval. These biomaterials properties may influence new bone formation and maintenance when applying the device for GBR/GTR.
- Published
- 2008
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.