Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Samir M. Abdelmagid is active.

Publication


Featured researches published by Samir M. Abdelmagid.


Experimental Cell Research | 2008

Osteoactivin, an anabolic factor that regulates osteoblast differentiation and function

Samir M. Abdelmagid; Mary F. Barbe; Mario C. Rico; Sibel Salihoglu; Israel Arango-Hisijara; Abdul Hafez Selim; Michael G. Anderson; Thomas A. Owen; Steven N. Popoff; Fayez F. Safadi

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.


Journal of Cellular Physiology | 2007

Osteoactivin acts as downstream mediator of BMP-2 effects on osteoblast function

Samir M. Abdelmagid; Mary F. Barbe; Israel Arango-Hisijara; Thomas A. Owen; Steven N. Popoff; Fayez F. Safadi

Our laboratory previously showed that osteoactivin (OA) is a novel, osteoblast‐related glycoprotein that plays a role in osteoblast differentiation and function. The purpose of this study was to examine the regulation of OA expression by BMP‐2 and the role OA plays as a downstream mediator of BMP‐2 effects in osteoblast function. Using primary osteoblast cultures, we tested different doses of BMP‐2 on the regulation of OA expression during osteoblast development. To test whether Smad‐1 signaling is responsible for BMP‐2 regulation of OA expression, osteoblast cultures were transfected with Smad1 siRNA, treated with 50 ng/ml of BMP‐2 and analyzed by Western blot. BMP‐2 treatment increased OA mRNA and protein expression in a dose‐dependent manner and this upregulation was blocked in Smad1 siRNA transfected cultures. We next examined whether the role of OA as a downstream mediator of BMP‐2 effects on osteoblast differentiation and matrix mineralization. Osteoblast cultures were transfected with OA antisense oligonucleotides and treated with 50 ng/ml of BMP‐2. Cultures transfected with OA antisense oligonucleotides and treated with BMP‐2 showed a reduction of OA expression associated with a significant reduction in early and late differentiation markers induced by BMP‐2. Therefore, OA acts, at least in part, as a downstream mediator of BMP‐2 effects on osteoblast differentiation and matrix mineralization. Our findings suggest that BMP‐2 regulates OA expression through the Smad1 signaling pathway. Our data also emphasize that OA protein acts as a downstream mediator of BMP‐2 effects on osteoblast differentiation and function. J. Cell. Physiol. 210: 26–37, 2007.


Journal of Orthopaedic Research | 2008

Serum and Tissue Cytokines and Chemokines Increase with Repetitive Upper Extremity Tasks

Mary F. Barbe; Melanie B. Elliott; Samir M. Abdelmagid; Mamta Amin; Steven N. Popoff; Fayez F. Safadi; Ann E. Barr

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‐1α, IL‐1β, TNFα, 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‐1α, IL‐1β, TNFα, and IL‐10 increased in distal bones, IL‐1α and ‐β in muscles, and TNFα in tendons. Only IL‐10 increased in LRNF reach limb muscles in week 8. Serum IL‐1α and MIP2 correlated with macrophages in LRNF loose connective tissues, serum MIP3a and MIP2 correlated negatively with grip strength, while serum TNFα, 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.


Life Sciences | 2015

Role of inflammation in the aging bones

Samir M. Abdelmagid; Mary F. Barbe; Fayez F. Safadi

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.


Journal of Cellular Biochemistry | 2010

Temporal and spatial expression of osteoactivin during fracture repair

Samir M. Abdelmagid; Mary F. Barbe; Michael Hadjiargyrou; Thomas A. Owen; Roshanak Razmpour; Saqib Rehman; Steven N. Popoff; Fayez F. Safadi

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. J. Cell. Biochem. 111: 295–309, 2010.


Journal of Cellular Physiology | 2014

Osteoactivin Induces Transdifferentiation of C2C12 Myoblasts Into Osteoblasts

Gregory R. Sondag; Sibel Salihoglu; Suzanne L. Lababidi; Douglas C. Crowder; Fouad M. Moussa; Samir M. Abdelmagid; Fayez F. Safadi

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. J. Cell. Physiol. 229: 955–966, 2014.


Journal of Cellular Biochemistry | 2014

Osteoactivin promotes osteoblast adhesion through HSPG and αvβ1 integrin.

Fouad M. Moussa; Israel Arango Hisijara; Gregory R. Sondag; Ethan M. Scott; Nagat Frara; Samir M. Abdelmagid; Fayez F. Safadi

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 αvβ1 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 αvβ1 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. J. Cell. Biochem. 115: 1243–1253, 2014.


Acta Biomaterialia | 2016

Orthosilicic acid, Si(OH)4, stimulates osteoblast differentiation in vitro by upregulating miR-146a to antagonize NF-κB activation

Xianfeng Zhou; Fouad M. Moussa; Steven Mankoci; Putu Ustriyana; Nianli Zhang; Samir M. Abdelmagid; Jim Molenda; William L. Murphy; Fayez F. Safadi; Nita Sahai

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.


Journal of Biological Chemistry | 2015

Mutation in Osteoactivin Promotes Receptor Activator of NFκB Ligand (RANKL)-mediated Osteoclast Differentiation and Survival but Inhibits Osteoclast Function.

Samir M. Abdelmagid; Gregory R. Sondag; Fouad M. Moussa; Joyce Y. Belcher; Bing Yu; Hilary Stinnett; Kimberly Novak; Thomas Mbimba; Matthew Philip Khol; Kurt D. Hankenson; Christopher Malcuit; Fayez F. Safadi

Background: The importance of osteoactivin has emerged from its role in osteogenesis. Results: Loss-of-function mutation of osteoactivin stimulates osteoclast differentiation and survival, with a defect in bone resorption. Conclusion: Osteoactivin acts as a negative regulator of osteoclastogenesis and a positive regulator of osteoclast function. Significance: This study helps to identify the possible mechanisms and targets of osteoactivin during osteoclast differentiation with impact on bone remodeling. 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.


Experimental and Molecular Medicine | 2016

Osteoactivin inhibition of osteoclastogenesis is mediated through CD44-ERK signaling.

Gregory R. Sondag; Thomas Mbimba; Fouad M. Moussa; Kimberly Novak; Bing Yu; Fatima A Jaber; Samir M. Abdelmagid; Werner J. Geldenhuys; Fayez F. Safadi

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.

Collaboration


Dive into the Samir M. Abdelmagid's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Gregory R. Sondag

Northeast Ohio Medical University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Kimberly Novak

Northeast Ohio Medical University

View shared research outputs
Top Co-Authors

Avatar

Bing Yu

Northeast Ohio Medical University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Joyce Y. Belcher

Northeast Ohio Medical University

View shared research outputs
Researchain Logo
Decentralizing Knowledge