Computational prediction of small molecules with predicted binding to FGFR3 and testing biological effects in bone cells

2021 ◽  
pp. 153537022110021
Author(s):  
Subburaman Mohan ◽  
Karthikeyan Muthusamy ◽  
Selvaraman Nagamani ◽  
Chandrasekhar Kesavan

Activating anabolic receptor-mediated signaling is essential for stimulating new bone formation and for promoting bone healing in humans. Fibroblast growth factor receptor (FGFR) 3 is reported to be an important positive regulator of osteogenesis. Presently, recombinant proteins are used to stimulate FGFR3 function but have limitations for therapy due to expense and stability. Therefore, there is a need for identification of novel small molecules binding to FGFR3 that promote biological function. In silico molecular docking and high-throughput virtual screening on zinc database identified seven compounds predicted to bind to an active site within the βCʹ-βE loop, specific to FGFR3. All seven compounds fall within an acceptable range of ADME/T properties. Four compounds showed a 30–65% oral absorption rate. Density functional theory analysis revealed a high HOMO-LUMO gap, reflecting high molecular stability for compounds 14977614 and 13509082. Five compounds exhibited mutagenicity, while the other three compounds presented irritability. Computational mutagenesis predicted that mutating G322 affected compound binding to FGFR3. Molecular dynamics simulation revealed compound 14977614 is stable in binding to FGFR3. Furthermore, compound 14977614, with an oral absorption rate of 60% and high molecular stability, produced significant increases in both proliferation and differentiation of bone marrow stromal cells in vitro. Anti-FGFR3 treatment completely blocked the stimulatory effect of 14977614 on BMSC proliferation. Ex vivo treatment of mouse calvaria in organ culture for seven days with 14977614 increased mineralization and expression levels of bone formation markers. In conclusion, computational analyses identified seven compounds that bind to the FGFR3, and in vitro studies showed that compound 14977614 exerts significant biological effects on osteogenic cells.

2021 ◽  
Vol 14 (4) ◽  
pp. 289
Author(s):  
Sana Ansari ◽  
Bregje W. M. de de Wildt ◽  
Michelle A. M. Vis ◽  
Carolina E. de de Korte ◽  
Keita Ito ◽  
...  

Bone is a complex organ maintained by three main cell types: osteoblasts, osteoclasts, and osteocytes. During bone formation, osteoblasts deposit a mineralized organic matrix. Evidence shows that bone cells release extracellular vesicles (EVs): nano-sized bilayer vesicles, which are involved in intercellular communication by delivering their cargoes through protein–ligand interactions or fusion to the plasma membrane of the recipient cell. Osteoblasts shed a subset of EVs known as matrix vesicles (MtVs), which contain phosphatases, calcium, and inorganic phosphate. These vesicles are believed to have a major role in matrix mineralization, and they feature bone-targeting and osteo-inductive properties. Understanding their contribution in bone formation and mineralization could help to target bone pathologies or bone regeneration using novel approaches such as stimulating MtV secretion in vivo, or the administration of in vitro or biomimetically produced MtVs. This review attempts to discuss the role of MtVs in biomineralization and their potential application for bone pathologies and bone regeneration.


Author(s):  
Ashis Kumar Goswami ◽  
Hemanta Kumar Sharma ◽  
Neelutpal Gogoi ◽  
Ankita Kashyap ◽  
Bhaskar Jyoti Gogoi

Background: Malaria is caused by different species of Plasmodium; among which P. falciparum is the most severe. Coptis teeta is an ethnomedicinal plant of enormous importance for tribes of north east India. Objective: In this study, the anti malarial activity of the methanol extracts of Coptis teeta was evaluated in vitro and lead identification via in silico study. Method: On the basis of the in vitro results, in silico analysis by application of different modules of Discovery Studio 2018 was performed on multiple targets of P. falciparum taking into consideration some of the compounds reported from C. teeta. Results: The IC50 of the methanol extract of Coptis teeta 0.08 µg/ml in 3D7 strain and 0.7 µg/ml in Dd2 strain of P. falciparum. From the docking study, noroxyhydrastatine was observed to have better binding affinity in comparison to chloroquine. The binding of noroxyhydrastinine with dihydroorotate dehydrogenase was further validated by molecular dynamics simulation and was observed to be significantly stable in comparison to the co-crystal inhibitor. During simulations it was observed that noroxyhydrastinine retained the interactions, giving strong indications of its effectiveness against the P. falciparum proteins and stability in the binding pocket. From the Density-functional theory analysis, the band gap energy of noroxyhydrastinine was found to be 0.186 Ha indicating a favourable interaction. Conclusion: The in silico analysis as an addition to the in vitro results provide strong evidence of noroxyhydrastinine as an anti malarial agent.


Blood ◽  
2013 ◽  
Vol 122 (21) ◽  
pp. 881-881 ◽  
Author(s):  
Michaela R Reagan ◽  
Archana Swami ◽  
Pamela A Basto ◽  
Yuji Mishima ◽  
Jinhe Liu ◽  
...  

Abstract Introduction The bone marrow (BM) niche is known to exert a protective effect on lymphoid tumors, such as multiple myeloma (MM), where mesenchymal stem cell interactions with clonal plasma cells increase tumor proliferation and survival. However, certain cells within the BM milieu, such as mature osteoblasts and osteocytes, have demonstrated the potential to inhibit tumor growth; utilizing these cells presents a promising new anti-cancer approach. Hence, designing better methods of bone-specific delivery for both direct cancer cell treatment and indirect treatment through the modulation of bone cells may result in a potent, two-pronged anti-cancer strategy. Our work aimed to develop a novel system to target both MM and bone cells to induce greater osteogenesis and hamper tumor growth. Methods PEG–PLGA nanoparticles (NPs) coupled to alendronate (“bone-targeted”) or alone (“non-targeted”) were formulated and loaded with bortezomib (“BTZ-NPs”) or left empty (“BTZ-free”). NPs were characterized for their physiochemical properties, including size (using dynamic light scattering; surface charges (Zeta potential); and bone affinity (using hydroxyapatite binding). NPs were engineered with different formulation methods and those with the optimal physiochemical characteristics and drug encapsulation efficiency were used for further studies. BTZ release kinetics were analyzed using HPLC. Anti-MM effects were assessed in vitro using MTT, bioluminescence (BLI) and Annexin V/PI apoptosis flow cytometry analysis on MM1S cells. In vivo, efficacy was measured by mouse weight, BLI and survival after i.v. cancer cell injections in mice. Cellular uptake was assessed in vitro by flow cytometry and in vivo biodistribution was assessed using fluorescent whole body and fixed section imaging. Bone specificity was assessed in vitro by co-culture of bone-targeted and non-targeted NPs with bone chips or hydroxyapatite using fluorescence and TEM imaging. In an in vivo model of myeloma treatment, female Nod/SCID beige mice were injected i.v. with 4 × 106 Luc+/GFP+ MM1S cells and, at day 21, treated with a) BTZ, b) BTZ-bone-targeted NPs, c) BTZ-non-targeted NPs or d) BTZ-free bone-targeted NPs. Using an in vivo model of pre-treatment for cancer prevention, mice were pre-treated with i.p. injections of BTZ-bone-targeted NPs and appropriate controls thrice weekly for 3 weeks. They were then injected i.v. with Luc+/GFP+ 5TGM1 or MM1S cells and monitored for BLI and survival. Static and dynamic bone histomorphometry and μCT were used to assess effects of pre-treatment on bone formation and osteolysis prevention. Results Our biodegradable, NPs had uniform size distribution within the range of 100 to 200 nm based on the type of formulation, with a zeta potential of ±5mV. Bone- targeted NPs showed high affinity towards bone mineral in vitro and better skeletal accumulation in vivo compared to non-targeted NPs. NPs were easily up-taken by cells in vitro, and BTZ release kinetics showed a burst followed by a sustained-release pattern over 60 hrs. BTZ-NPs induced apoptosis in MM cells in vitro. Importantly, BTZ-bone-targeted-NP pre-treated mice showed significantly less tumor burden (BLI) and longer survival than free drug or drug-free bone-targeted NPs, thus demonstrating a tumor-inhibiting effect unique to the BTZ-bone-targeted-NPs. Pre-treatment with BTZ increased bone formation in tibias and femurs, as measured by μCT of bone volume/total volume, and trabecular thickness and number, suggesting that increased bone volume may inhibit MM. In a second mouse model, both BTZ-bone-targeted NPs and BTZ-free NPs were equally able to reduce tumor growth in vivo when given after tumor formation. Conclusion Bone-targeted nanoparticles hold great potential for clinical applications in delivering chemotherapies to bone marrow niches, reducing off-target effects, increasing local drug concentrations, and lengthening the therapeutic window. BTZ-bone-targeted NPs are able to slow tumor growth and increase survival in mice when used as a pre-treatment. This may result, at least in part, from BTZ-induced increased bone formation. These findings indicate that BTZ-bone-targeted NPs exert a chemopreventive effect in MM in vivo, thus suggesting their potential use in the clinical setting. Disclosures: Basto: BIND Therapeutics: Patent licensed by BIND, Patent licensed by BIND Patents & Royalties. Farokhzad:BIND Therapeutics: Employment, Equity Ownership; Selecta Biosciences: Employment, Equity Ownership. Ghobrial:Onyx: Membership on an entity’s Board of Directors or advisory committees; BMS: Membership on an entity’s Board of Directors or advisory committees; BMS: Research Funding; Sanofi: Research Funding; Novartis: Membership on an entity’s Board of Directors or advisory committees.


2020 ◽  
Author(s):  
Yun Gong ◽  
Junxiao Yang ◽  
Xiaohua Li ◽  
Cui Zhou ◽  
Yu Chen ◽  
...  

AbstractOsteoblasts are multifunctional bone cells, which play essential roles in bone formation, angiogenesis regulation, as well as maintenance of hematopoiesis. Although both in vivo and in vitro studies on mice have identified several potential osteoblast subtypes based on their different transition stages or biological responses to external stimuli, the categorization of primary osteoblast subtypes in vivo in humans has not yet been achieved. Here, we used single-cell RNA sequencing (scRNA-seq) to perform a systematic cellular taxonomy dissection of freshly isolated human osteoblasts. Based on the gene expression patterns and cell lineage reconstruction, we identified three distinct cell clusters including preosteoblasts, mature osteoblasts, and an undetermined rare osteoblast subpopulation. This novel subtype was mainly characterized by the nuclear receptor subfamily 4 group A member 1 and 2 (NR4A1 and NR4A2), and its existence was confirmed by immunofluorescence staining. Trajectory inference analysis suggested that the undetermined cluster, together with the preosteoblasts, are involved in the regulation of osteoblastogenesis and also give rise to mature osteoblasts. Investigation of the biological processes and signaling pathways enriched in each subpopulation revealed that in addition to bone formation, preosteoblasts and undetermined osteoblasts may also regulate both angiogenesis and hemopoiesis. Finally, we demonstrated that there are systematic differences between the transcriptional profiles of human osteoblasts in vivo and mouse osteoblasts both in vivo and in vitro, highlighting the necessity for studying bone physiological processes in humans rather than solely relying on mouse models. Our findings provide novel insights into the cellular heterogeneity and potential biological functions of human primary osteoblasts at the single-cell level, which is an important and necessary step to further dissect the biological roles of osteoblasts in bone metabolism under various (patho-) physiological conditions.


Author(s):  
Y. X. Qin ◽  
S. Zhang ◽  
J. Cheng

Mechanotransduction has demonstrated potentials for tissue adaptation in vivo and in vitro. It is well documented that ultrasound, as a mechanical signal, can produce a wide variety of biological effects in vitro and in vivo[1]. For example, pulsed ultrasound can be used to accelerate the rate of bone fracture healing noninvasively. Although a wide range of studies have been done, mechanism for this therapeutic effect on bone healing is currently unknown and still under active investigation. In our previous studies, we have developed methodology allowed in vitro manipulating osteoblastic cells using acoustic radiation force (ARF) generated by ultrasound without the effects of acoustic streaming and ultrasound-induced temperature rise. Furthermore, we also confirmed that ARF modulated intracellular Ca2+ transient in MC3T3-E1 osteoblast-like cells in a strain and frequency-dependent manner. A potential mechanism by which bone cells may sense ultrasound is through their structures such as primary cilia and cytoskeletons. The purpose of the current study was to evaluate the hypothesis that acoustic radiation force can regulate the activities of the primary cilium and the cytoskeleton of the cells, which act as the mechanotransductive signals to mediate Ca2+ flux, as a pathway in response to cyclic loading.


2013 ◽  
Vol 200 (4) ◽  
pp. 537-549 ◽  
Author(s):  
Joachim Albers ◽  
Johannes Keller ◽  
Anke Baranowsky ◽  
Frank Timo Beil ◽  
Philip Catala-Lehnen ◽  
...  

Although Wnt signaling is considered a key regulatory pathway for bone formation, inactivation of β-catenin in osteoblasts does not affect their activity but rather causes increased osteoclastogenesis due to insufficient production of osteoprotegerin (Opg). By monitoring the expression pattern of all known genes encoding Wnt receptors in mouse tissues and bone cells we identified Frizzled 8 (Fzd8) as a candidate regulator of bone remodeling. Fzd8-deficient mice displayed osteopenia with normal bone formation and increased osteoclastogenesis, but this phenotype was not associated with impaired Wnt signaling or Opg production by osteoblasts. The deduced direct negative influence of canonical Wnt signaling on osteoclastogenesis was confirmed in vitro and through the generation of mice lacking β-catenin in the osteoclast lineage. Here, we observed increased bone resorption despite normal Opg production and a resistance to the anti-osteoclastogenic effect of Wnt3a. These results demonstrate that Fzd8 and β-catenin negatively regulate osteoclast differentiation independent of osteoblasts and that canonical Wnt signaling controls bone resorption by two different mechanisms.


2012 ◽  
Vol 100B (3) ◽  
pp. 850-861 ◽  
Author(s):  
J. David Eick ◽  
Cielo Barragan-Adjemian ◽  
Jennifer Rosser ◽  
Jennifer R. Melander ◽  
Vladimir Dusevich ◽  
...  
Keyword(s):  

2019 ◽  
Vol 116 (11) ◽  
pp. 4855-4860 ◽  
Author(s):  
Anne M. Arnold ◽  
Brian D. Holt ◽  
Leila Daneshmandi ◽  
Cato T. Laurencin ◽  
Stefanie A. Sydlik

Synthetic, resorbable scaffolds for bone regeneration have potential to transform the clinical standard of care. Here, we demonstrate that functional graphenic materials (FGMs) could serve as an osteoinductive scaffold: recruiting native cells to the site of injury and promoting differentiation into bone cells. By invoking a Lewis acid-catalyzed Arbuzov reaction, we are able to functionalize graphene oxide (GO) to produce phosphate graphenes (PGs) with unprecedented control of functional group density, mechanical properties, and counterion identity. In aqueous environments, PGs release inducerons, including Ca2+ and PO43−. Calcium phosphate graphene (CaPG) intrinsically induces osteogenesis in vitro and in the presence of bone marrow stromal cells (BMSCs), can induce ectopic bone formation in vivo. Additionally, an FGM can be made by noncovalently loading GO with the growth factor recombinant human bone morphogenetic protein 2 (rhBMP-2), producing a scaffold that induces ectopic bone formation with or without BMSCs. The FGMs reported here are intrinsically inductive scaffolds with significant potential to revolutionize the regeneration of bone.


2021 ◽  
Author(s):  
Yue Zhou ◽  
Hao Jia ◽  
Aihua Hu ◽  
Rangru Liu ◽  
Xiangzhou Zeng ◽  
...  

Abstract Background: Promoting bone marrow mesenchymal stem cell (BMSC) osteoblastic differentiation is a promising therapeutic strategy for osteoporosis (OP). The present study demonstrates that miR-483-5p inhibits the osteogenic differentiation of BMSCs. Therefore, selectively delivering the nanoparticles carrying antagomir-483-5p (miR-483-5p inhibitor) to BMSCs is expected to become an effective treatment drug for OP.Methods: Real-time PCR assays were used to analyse miR-483-5p, ALP and Bglap levels in BMSCs of ovariectomized and aged osteoporotic mice. To selectively and efficiently deliver antagomir-483-5p to BMSCs in vivo, immunoglobulin G and poloxamer-188 were used to encapsulate the functional small molecules, and BMSC-targeting aptamer was employed to confirm the direction of the nanoparticles. Luciferase assays were used to determine the target genes of miR-483-5p. Western blot assays and immunohistochemistry staining were used to detect the targets in vitro and vivo.Results: miR-483-5p levels were increased in BMSCs of ovariectomized and aged osteoporotic mice. Inhibition of miR-483-5p levels in BMSCs by antagomir-483-5p in vitro promoted the expression of bone formation markers, such as ALP and Bglap. The FAM-BMSC-aptamer-nanoparticles carrying antagomir-483-5p were taken up by BMSCs, resulting in stimulation of BMSC osteoblastic differentiation in vitro and osteoporosis prevention in vivo. Furthermore, our research demonstrated that mitogen-activated protein kinase 1 (MAPK1) and SMAD family member 5 (Smad5) were direct targets of miR-483-5p in regulating BMSC osteoblastic differentiation and osteoporosis pathological processes. Conclusions: The important therapeutic role of FAM-BMSC-aptamer-nanoparticles carrying antagomir-483-5p in osteoporosis was established in our study. These nanoparticles are novel candidate for the clinical prevention and treatment of osteoporosis. The optimized targeted drug delivery platform for small molecules will provide new ideas for the treatment of clinical diseases.


Sign in / Sign up

Export Citation Format

Share Document