42 results on '"Muthurangan Manikandan"'
Search Results
2. IL-1β-mediated TGFβ/SMAD signaling pathway inactivation impaired ex vivo osteogenic activity of human bone marrow-derived stromal cells
- Author
-
Amer Mahmood, Mona Elsafadi, Muthurangan Manikandan, and Musaad Alfayez
- Subjects
cytokine signaling ,adult stem cells ,osteoblastic differentiation ,tgfβ signaling ,heterogeneous population ,cross-talk ,Biotechnology ,TP248.13-248.65 - Abstract
Bone loss is caused by inflammation and is mediated by pro-inflammatory cytokines that control bone formation and bone resorption. The study aimed to determine the effect of secreted factors from human bone marrow-derived stromal cells (hBMSC) of no-heterotopic bone-forming capacity (hBMSC–Bone) cells on the differentiation potential of hBMSC which capable of creating bone in-vivo (hBMSC+Bone) and dissect the molecular signature of these cells for understanding the complicated relationship of stem cells and microenvironment. hBMSC cultures are heterogenous with respect to differentiation and function. However, the nature of interaction between different cell populations within hBMSC cultures is poorly investigated. We employed two clonal hBMSC lines which exhibit different functional phenotypes based on the presence of either high or low osteoblastic differentiation capacity, bone forming (hBMSC+Bone) and non-bone forming (hBMSC−Bone), and examined their biological interaction. Adding conditioned media (CM) of hBMSC−Bone cultures resulted in suppression of cell proliferation and osteoblasts differentiation of hBMSC+Bone. Microarray analysis of CM-treated hBMSC+Bone revealed significant enrichment of several pathways, including TGFβ signaling. Follow-up experiments corroborated the inhibitory effects on TGFβ signaling as evidenced by decreased SMAD2 phosphorylation and TGFβ-responsive genes (TAGLN, ACTA2 and TPM1). Interestingly, IL1β is highly expressed in hBMSC−Bone and is present in its CM. Incubating hBMSC−Bone with rhIL-1RI rescued the functional phenotype of hBMSC−Bone with respect to cell proliferation and differentiation into osteoblasts, and upregulated TGFβ-responsive genes. These data demonstrated that IL1β-TGFβ signaling is part of the intercellular communication within the heterogenous population of hBMSCs and regulates their commitment to osteoblastic fate. Supplemental data for this article is available online at https://doi.org/10.1080/13102818.2021.1939784 .
- Published
- 2021
- Full Text
- View/download PDF
3. Convergence of TGFβ and BMP signaling in regulating human bone marrow stromal cell differentiation
- Author
-
Mona Elsafadi, Tasneem Shinwari, Sami Al-Malki, Muthurangan Manikandan, Amer Mahmood, Abdullah Aldahmash, Musaad Alfayez, Moustapha Kassem, and Nehad M. Alajez
- Subjects
Medicine ,Science - Abstract
Abstract Targeting regulatory signaling pathways that control human bone marrow stromal (skeletal or mesenchymal) stem cell (hBMSC) differentiation and lineage fate determination is gaining momentum in the regenerative medicine field. Therefore, to identify the central regulatory mechanism of osteoblast differentiation of hBMSCs, the molecular phenotypes of two clonal hBMSC lines exhibiting opposite in vivo phenotypes, namely, bone forming (hBMSC+bone) and non-bone forming (hBMSC−Bone) cells, were studied. Global transcriptome analysis revealed significant downregulation of several TGFβ responsive genes, namely, TAGLN, TMP1, ACTA2, TGFβ2, SMAD6, SMAD9, BMP2, and BMP4 in hBMSC−Bone cells and upregulation on SERPINB2 and NOG. Transcriptomic data was associated with marked reduction in SMAD2 protein phosphorylation, which thereby implies the inactivation of TGFβ and BMP signaling in those cells. Concordantly, activation of TGFβ signaling in hBMSC−Bone cells using either recombinant TGFβ1 protein or knockdown of SERPINB2 TGFβ-responsive gene partially restored their osteoblastic differentiation potential. Similarly, the activation of BMP signaling using exogenous BMP4 or via siRNA-mediated knockdown of NOG partially restored the differentiation phenotype of hBMSC−Bone cells. Concordantly, recombinant NOG impaired ex vivo osteoblastic differentiation of hBMSC+Bone cells, which was associated with SERBINB2 upregulation. Our data suggests the existence of reciprocal relationship between TGFB and BMP signaling that regulates hBMSC lineage commitment and differentiation, whilst provide a plausible strategy for generating osteoblastic committed cells from hBMSCs for clinical applications.
- Published
- 2019
- Full Text
- View/download PDF
4. MicroRNA Expression Profiling on Paired Primary and Lymph Node Metastatic Breast Cancer Revealed Distinct microRNA Profile Associated With LNM
- Author
-
Ramesh Elango, Khalid A. Alsaleh, Radhakrishnan Vishnubalaji, Muthurangan Manikandan, Arwa M. Ali, Nashwa Abd El-Aziz, Abdulrhaman Altheyab, Ammar Al-Rikabi, Musaad Alfayez, Abdullah Aldahmash, and Nehad M. Alajez
- Subjects
breast cancer ,lymph node metastasis ,miRNA signature ,hsa-miR-205-5p ,hsa-miR-214-3p ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Breast cancer (BC) is the foremost cause of cancer-related deaths in women. BC patients are oftentimes presented with lymph node metastasis (LNM), which increases their risk of recurrence. Compelling data have recently implicated microRNAs in promoting BC metastasis. Therefore, the identification of microRNA (miRNA)-based molecular signature associated with LNM could provide an opportunity for a more personalized treatment for BC patients with high risk of LNM. In current study, we performed comprehensive miRNA profiling in matched primary breast and LNM and identified 40 miRNAs, which were differentially expressed in LNM compared to primary tumors. The expression of 14 miRNAs (Up: hsa-miR-155-5p, hsa-miR-150-5p, hsa-miR-146a-5p, hsa-miR-142-5p and down: hsa-miR-200a-3p, hsa-miR-200b-3p, hsa-miR-200c-3p, hsa-miR-205-5p, hsa-miR-210-3p, hsa-miR-214-3p, hsa-miR-141-3p, hsa-miR-127-3p, hsa-miR-125a-5p, and hsa-let-7c-5p) was subsequently validated in a second cohort of 32 breast and 32 matched LNM tumor tissues. Mechanistically, forced expression of hsa-miR-205-5p, or hsa-miR-214-3p epigenetically inhibited MDA-MB-231 cell proliferation, colony formation, and cell migration. Global gene expression profiling on MDA-MB-231 cells overexpressing hsa-miR-205-5p, or hsa-miR-214-3p in combination with in silico target prediction and ingenuity pathway analyses identified multiple bona fide targets for hsa-miR-205-5p, hsa-miR-214-3p affecting cellular proliferation and migration. Interestingly, interrogation of the expression levels of hsa-miR-205 and hsa-miR-214 in the METABRIC breast cancer dataset revealed significantly poor overall survival in patients with downregulated expression of miR-205 [HR = 0.75 (0.61–0.91)], p = 0.003 and hsa-miR-214 [HR = 0.74 (0.59–0.93) p = 0.008]. Our data unraveled the miRNA-transcriptional landscape associated with LNM and provide novel insight on the role of several miRNAs in promoting BC LNM, and suggest their potential utilization in the clinical management of BC patients.
- Published
- 2020
- Full Text
- View/download PDF
5. Stem cell library screen identified ruxolitinib as regulator of osteoblastic differentiation of human skeletal stem cells
- Author
-
Nihal AlMuraikhi, Dalia Ali, Aliah Alshanwani, Radhakrishnan Vishnubalaji, Muthurangan Manikandan, Muhammad Atteya, Abdulaziz Siyal, Musaad Alfayez, Abdullah Aldahmash, Moustapha Kassem, and Nehad M. Alajez
- Subjects
Medicine (General) ,R5-920 ,Biochemistry ,QD415-436 - Abstract
Abstract Background Better understanding of the signaling pathways that regulate human bone marrow stromal stem cell (hBMSC) differentiation into bone-forming osteoblasts is crucial for their clinical use in regenerative medicine. Chemical biology approaches using small molecules targeting specific signaling pathways are increasingly employed to manipulate stem cell differentiation fate. Methods We employed alkaline phosphatase activity and staining assays to assess osteoblast differentiation and Alizarin R staining to assess mineralized matrix formation of cultured hBMSCs. Changes in gene expression were assessed using an Agilent microarray platform, and data normalization and bioinformatics were performed using GeneSpring software. For in vivo ectopic bone formation experiments, hMSCs were mixed with hydroxyapatite–tricalcium phosphate granules and implanted subcutaneously into the dorsal surface of 8-week-old female nude mice. Hematoxylin and eosin staining and Sirius Red staining were used to detect bone formation in vivo. Results We identified several compounds which inhibited osteoblastic differentiation of hMSCs. In particular, we identified ruxolitinib (INCB018424) (3 μM), an inhibitor of JAK-STAT signaling that inhibited osteoblastic differentiation and matrix mineralization of hMSCs in vitro and reduced ectopic bone formation in vivo. Global gene expression profiling of ruxolitinib-treated cells identified 847 upregulated and 822 downregulated mRNA transcripts, compared to vehicle-treated control cells. Bioinformatic analysis revealed differential regulation of multiple genetic pathways, including TGFβ and insulin signaling, endochondral ossification, and focal adhesion. Conclusions We identified ruxolitinib as an important regulator of osteoblast differentiation of hMSCs. It is plausible that inhibition of osteoblast differentiation by ruxolitinib may represent a novel therapeutic strategy for the treatment of pathological conditions caused by accelerated osteoblast differentiation and mineralization.
- Published
- 2018
- Full Text
- View/download PDF
6. Which stem cells to choose for regenerative medicine application: Bone marrow and adipose tissue stromal stem cells – Similarities and differences
- Author
-
Nehad M Alajez, Dalia Al-Ali, Radhakrishnan Vishnubalaji, Muthurangan Manikandan, Musaad Alfayez, Moustapha Kassem, and Abdullah Aldahmash
- Subjects
adipose tissue ,bone marrow ,mesenchymal stromal cells ,osteogenesis ,pathways ,Medicine ,Public aspects of medicine ,RA1-1270 - Abstract
Background: Clinical use of stromal stem cells in regenerative medicine is increasingly recognized as a promising treatment modality for age-related degenerative diseases based on the promising initial results of clinical trials. However, the magnitude of positive effects observed in these trials has been variable which can be explained by the lack of standardization of the stem cell products “cell product.” Bone marrow-derived stromal (also known mesenchymal) stem cells (BM-hMSC) and adipose tissue-hMSC (AD-hMSC) have been used interchangeably in clinical trials employing stromal stem cells as they were thought to be functionally identical. Methods: In the present study, we performed an extensive side-by-side comparison of BM-hMSC and AD-hMSC for their CD marker expression using FACS analysis, molecular phenotype using global mRNA gene expression analysis, and functional studies for their in vitro differentiation capacity to osteoblasts and adipocytes. Results: We observed both stromal cell populations were CD44+ CD13+ CD90+ CD29+ CD105+ CD14− HLDR−. We also observed that they express common genetic signature consisting of 13,667 genes with enrichment in a number of pathways relevant to stem cell biology, for example, focal adhesion, insulin signaling, and mitogen-activated protein kinase signaling. On the other hand, we observed significant differences in their molecular phenotype with 3282 and 1409 genes differentially expression in BM-hMSC and AD-hMSC, respectively. Further analysis revealed higher expression of genes associated with osteoblast differentiation in BM-hMSC and those of adipocyte differentiation in AD-hMSC which correlated with their differential capacity for osteoblast versus adipocyte differentiation, respectively. Conclusion: Our data suggest that the clinical use of MSC in therapy depend on MSC site of origin, and thus, BM-hMSC are better suited for clinical trials aiming at enhancing bone regeneration. We suggest that molecular phenotype of stem cells is relevant approach for stem cell screening before their clinical transplantation.
- Published
- 2018
- Full Text
- View/download PDF
7. MicroRNA-4739 regulates osteogenic and adipocytic differentiation of immortalized human bone marrow stromal cells via targeting LRP3
- Author
-
Mona Elsafadi, Muthurangan Manikandan, Nehad M Alajez, Rimi Hamam, Raed Abu Dawud, Abdullah Aldahmash, Zafar Iqbal, Musaad Alfayez, Moustapha Kassem, and Amer Mahmood
- Subjects
Biology (General) ,QH301-705.5 - Abstract
Understanding the regulatory networks underlying lineage differentiation and fate determination of human bone marrow stromal cells (hBMSC) is a prerequisite for their therapeutic use. The goal of the current study was to unravel the novel role of the low-density lipoprotein receptor-related protein 3 (LRP3) in regulating the osteogenic and adipogenic differentiation of immortalized hBMSCs. Gene expression profiling revealed significantly higher LRP3 levels in the highly osteogenic hBMSC clone imCL1 than in the less osteogenic clone imCL2, as well as a significant upregulation of LRP3 during the osteogenic induction of the imCL1 clone. Data from functional and gene expression assays demonstrated the role of LRP3 as a molecular switch promoting hBMSC lineage differentiation into osteoblasts and inhibiting differentiation into adipocytes. Interestingly, microRNA (miRNA) expression profiling identified miR-4739 as the most under-represented miRNA (−36.11 fold) in imCL1 compared to imCL2. The TargetScan prediction algorithm, combined with functional and biochemical assays, identified LRP3 mRNA as a novel target of miR-4739, with a single potential binding site for miR-4739 located in the LRP3 3′ UTR. Regulation of LRP3 expression by miR-4739 was subsequently confirmed by qRT-PCR, western blotting, and luciferase assays. Over-expression of miR-4739 mimicked the effects of LRP3 knockdown on promoting adipogenic and suppressing osteogenic differentiation of hBMSCs. Hence, we report for the first time a novel biological role for the LRP3/hsa-miR-4739 axis in balancing osteogenic and adipocytic differentiation of hBMSCs. Our data support the potential utilization of miRNA-based therapies in regenerative medicine.
- Published
- 2017
- Full Text
- View/download PDF
8. Notch Signaling Inhibition by LY411575 Attenuates Osteoblast Differentiation and Decreased Ectopic Bone Formation Capacity of Human Skeletal (Mesenchymal) Stem Cells
- Author
-
Nihal AlMuraikhi, Dalia Ali, Radhakrishnan Vishnubalaji, Muthurangan Manikandan, Muhammad Atteya, Abdulaziz Siyal, Musaad Alfayez, Abdullah Aldahmash, Moustapha Kassem, and Nehad M. Alajez
- Subjects
Internal medicine ,RC31-1245 - Abstract
Background. Chemical biology approaches using small molecule inhibitors targeting specific signaling pathways are useful tools to dissect the molecular mechanisms governing stem cell differentiation and for their possible use in therapeutic interventions. Methods. Stem cell signaling small molecule library functional screen was performed employing human bone marrow skeletal (mesenchymal) stem cells (hBMSCs). Alkaline phosphatase (ALP) activity and formation of mineralized matrix visualized by Alizarin red staining were employed as markers for osteoblastic differentiation. Global gene expression profiling was conducted using the Agilent microarray platform, and data normalization and bioinformatics were performed using GeneSpring software. Pathway analyses were conducted using the Ingenuity Pathway Analysis (IPA) tool. In vivo ectopic bone formation was performed using hBMSC mixed with hydroxyapatite–tricalcium phosphate granules that were implanted subcutaneously in 8-week-old female nude mice. Hematoxylin and eosin staining and Sirius red staining were performed to identify bone formation in vivo. Results. Among the tested molecules, LY411575, a potent γ-secretase and Notch signaling inhibitor, exhibited significant inhibitory effects on osteoblastic differentiation of hBMSCs manifested by reduced ALP activity, mineralized matrix formation, and decreased osteoblast-specific gene expression as well as in vivo ectopic bone formation. Global gene expression profiling of LY411575-treated cells revealed changes in multiple signaling pathways, including focal adhesion, insulin, TGFβ, IL6, and Notch signaling, and decreased the expression of genes associated with functional categories of tissue development. Among the affected signaling networks were TGFβ1, SPP1, and ERK regulatory networks. Conclusions. We identified γ-secretase inhibitor (LY411575) as a potent regulator of osteoblastic differentiation of hBMSC that may be useful as a therapeutic option for treating conditions associated with ectopic bone formation.
- Published
- 2019
- Full Text
- View/download PDF
9. Hedgehog Signaling Inhibition by Smoothened Antagonist BMS-833923 Reduces Osteoblast Differentiation and Ectopic Bone Formation of Human Skeletal (Mesenchymal) Stem Cells
- Author
-
Nihal AlMuraikhi, Nuha Almasoud, Sarah Binhamdan, Ghaydaa Younis, Dalia Ali, Muthurangan Manikandan, Radhakrishnan Vishnubalaji, Muhammad Atteya, Abdulaziz Siyal, Musaad Alfayez, Abdullah Aldahmash, Moustapha Kassem, and Nehad M. Alajez
- Subjects
Internal medicine ,RC31-1245 - Abstract
Background. Hedgehog (Hh) signaling is essential for osteoblast differentiation of mesenchymal progenitors during endochondral bone formation. However, the critical role of Hh signaling during adult bone remodeling remains to be elucidated. Methods. A Smoothened (SMO) antagonist/Hedgehog inhibitor, BMS-833923, identified during a functional screening of a stem cell signaling small molecule library, was investigated for its effects on the osteoblast differentiation of human skeletal (mesenchymal) stem cells (hMSC). Alkaline phosphatase (ALP) activity and Alizarin red staining were employed as markers for osteoblast differentiation and in vitro mineralization capacity, respectively. Global gene expression profiling was performed using the Agilent® microarray platform. Effects on in vivo ectopic bone formation were assessed by implanting hMSC mixed with hydroxyapatite-tricalcium phosphate granules subcutaneously in 8-week-old female nude mice, and the amount of bone formed was assessed using quantitative histology. Results. BMS-833923, a SMO antagonist/Hedgehog inhibitor, exhibited significant inhibitory effects on osteoblast differentiation of hMSCs reflected by decreased ALP activity, in vitro mineralization, and downregulation of osteoblast-related gene expression. Similarly, we observed decreased in vivo ectopic bone formation. Global gene expression profiling of BMS-833923-treated compared to vehicle-treated control cells, identified 348 upregulated and 540 downregulated genes with significant effects on multiple signaling pathways, including GPCR, endochondral ossification, RANK-RANKL, insulin, TNF alpha, IL6, and inflammatory response. Further bioinformatic analysis employing Ingenuity Pathway Analysis revealed significant enrichment in BMS-833923-treated cells for a number of functional categories and networks involved in connective and skeletal tissue development and disorders, e.g., NFκB and STAT signaling. Conclusions. We identified SMO/Hedgehog antagonist (BMS-833923) as a powerful inhibitor of osteoblastic differentiation of hMSC that may be useful as a therapeutic option for treating conditions associated with high heterotopic bone formation and mineralization.
- Published
- 2019
- Full Text
- View/download PDF
10. Author Correction: Tankyrase inhibitor XAV-939 enhances osteoblastogenesis and mineralization of human skeletal (mesenchymal) stem cells
- Author
-
Nuha Almasoud, Sarah Binhamdan, Ghaydaa Younis, Hanouf Alaskar, Amal Alotaibi, Muthurangan Manikandan, Musaad Alfayez, Moustapha Kassem, and Nihal AlMuraikhi
- Subjects
Medicine ,Science - Abstract
An amendment to this paper has been published and can be accessed via a link at the top of the paper.
- Published
- 2021
- Full Text
- View/download PDF
11. TGFβ1-Induced Differentiation of Human Bone Marrow-Derived MSCs Is Mediated by Changes to the Actin Cytoskeleton
- Author
-
Mona Elsafadi, Muthurangan Manikandan, Sami Almalki, Mohammad Mobarak, Muhammad Atteya, Zafar Iqbal, Jamil Amjad Hashmi, Sameerah Shaheen, Nehad Alajez, Musaad Alfayez, Moustapha Kassem, Raed Abu Dawud, and Amer Mahmood
- Subjects
Internal medicine ,RC31-1245 - Abstract
TGFβ is a potent regulator of several biological functions in many cell types, but its role in the differentiation of human bone marrow-derived skeletal stem cells (hMSCs) is currently poorly understood. In the present study, we demonstrate that a single dose of TGFβ1 prior to induction of osteogenic or adipogenic differentiation results in increased mineralized matrix or increased numbers of lipid-filled mature adipocytes, respectively. To identify the mechanisms underlying this TGFβ-mediated enhancement of lineage commitment, we compared the gene expression profiles of TGFβ1-treated hMSC cultures using DNA microarrays. In total, 1932 genes were upregulated, and 1298 genes were downregulated. Bioinformatics analysis revealed that TGFβl treatment was associated with an enrichment of genes in the skeletal and extracellular matrix categories and the regulation of the actin cytoskeleton. To investigate further, we examined the actin cytoskeleton following treatment with TGFβ1 and/or cytochalasin D. Interestingly, cytochalasin D treatment of hMSCs enhanced adipogenic differentiation but inhibited osteogenic differentiation. Global gene expression profiling revealed a significant enrichment of pathways related to osteogenesis and adipogenesis and of genes regulated by both TGFβ1 and cytochalasin D. Our study demonstrates that TGFβ1 enhances hMSC commitment to either the osteogenic or adipogenic lineages by reorganizing the actin cytoskeleton.
- Published
- 2018
- Full Text
- View/download PDF
12. Romidepsin Promotes Osteogenic and Adipocytic Differentiation of Human Mesenchymal Stem Cells through Inhibition of Histondeacetylase Activity
- Author
-
Dalia Ali, Elna P. Chalisserry, Muthurangan Manikandan, Rimi Hamam, Musaad Alfayez, Moustapha Kassem, Abdullah Aldahmash, and Nehad M. Alajez
- Subjects
Internal medicine ,RC31-1245 - Abstract
Bone marrow mesenchymal stem cells (BMSCs) are adult multipotent stem cells that can differentiate into mesodermal lineage cells, including adipocytes and osteoblasts. However, the epigenetic mechanisms governing the lineage-specific commitment of BMSCs into adipocytes or osteoblasts are under investigation. Herein, we investigated the epigenetic effect of romidepsin, a small molecule dual inhibitor targeting HDAC1 and HDAC2 identified through an epigenetic library functional screen. BMSCs exposed to romidepsin (5 nM) exhibited enhanced adipocytic and osteoblastic differentiation. Global gene expression and signaling pathway analyses of differentially expressed genes revealed a strong enrichment of genes involved in adipogenesis and osteogenesis in romidepsin-treated BMSCs during induction into adipocytes or osteoblasts, respectively. Pharmacological inhibition of FAK signaling during adipogenesis or inhibition of FAK or TGFβ signaling during osteogenesis diminished the biological effects of romidepsin on BMSCs. The results of chromatin immunoprecipitation combined with quantitative polymerase chain reaction indicated a significant increase in H3K9Ac epigenetic markers in the promoter regions of peroxisome proliferator-activated receptor gamma (PPARγ) and KLF15 (related to adipogenesis) or SP7 (Osterix) and alkaline phosphatase (ALP) (related to osteogenesis) in romidepsin-treated BMSCs. Our data indicated that romidepsin is a novel in vitro modulator of adipocytic and osteoblastic differentiation of BMSCs.
- Published
- 2018
- Full Text
- View/download PDF
13. Characterization of Cellular and Molecular Heterogeneity of Bone Marrow Stromal Cells
- Author
-
Mona Elsafadi, Muthurangan Manikandan, Muhammad Atteya, Jamil Amjad Hashmi, Zafar Iqbal, Abdullah Aldahmash, Musaad Alfayez, Moustapha Kassem, and Amer Mahmood
- Subjects
Internal medicine ,RC31-1245 - Abstract
Human bone marrow-derived stromal stem cells (hBMSC) exhibit multiple functions, including differentiation into skeletal cells (progenitor function), hematopoiesis support, and immune regulation (nonprogenitor function). We have previously demonstrated the presence of morphological and functional heterogeneity of hBMSC cultures. In the present study, we characterized in detail two hTERT-BMSC clonal cell populations termed here CL1 and CL2 that represent an opposing phenotype with respect to morphology, markers expression: alkaline phosphatase (ALP) and CD146, and ex vivo differentiation potential. CL1 differentiated readily to osteoblasts, adipocytes, and chondrocytes as shown by expression of lineage specific genes and proteins. Whole genome transcriptome profiling of CL1 versus CL2 revealed enrichment in CL1 of bone-, mineralization-, and skeletal muscle-related genes, for example, ALP, POSTN, IGFBP5 BMP4, and CXCL12. On the other hand, CL2 transcriptome was enriched in immune modulatory genes, for example, CD14, CD99, NOTCH3, CXCL6, CFB, and CFI. Furthermore, gene expression microarray analysis of osteoblast differentiated CL1 versus CL2 showed significant upregulation in CL1 of bone development and osteoblast differentiation genes which included several homeobox genes: TBX15, HOXA2 and HOXA10, and IGF1, FGFR3, BMP6, MCAM, ITGA10, IGFBP5, and ALP. siRNA-based downregulation of the ALP gene in CL1 impaired osteoblastic and adipocytic differentiation. Our studies demonstrate the existence of molecular and functional heterogeneity in cultured hBMSC. ALP can be employed to identify osteoblastic and adipocytic progenitor cells in the heterogeneous hBMSC cultures.
- Published
- 2016
- Full Text
- View/download PDF
14. Novel Chitosan-Gelatin Scaffold with Valproic Acid Augments In Vitro Osteoblast Differentiation of Mesenchymal Stem Cells.
- Author
-
Alghofaily, Maha, Alsalleeh, Fahd, Alssum, Lamees, Muthurangan, Manikandan, Alfayez, Musaad, Weir, Michael D., and Xu, Hockin H. K.
- Subjects
RUNX proteins ,MESENCHYMAL stem cell differentiation ,VALPROIC acid ,HISTONE deacetylase inhibitors ,MESENCHYMAL stem cells ,TISSUE scaffolds - Abstract
The study aimed to develop a biodegradable scaffold incorporating valproic acid (VPA) for improved human bone marrow-derived mesenchymal stem cell (hBMSC) proliferation, differentiation, and bone mineral synthesis. A chitosan–gelatin (CH-G) scaffold was fabricated and loaded with varying concentrations of VPA (1, 3, 5 mM/L). In vitro studies assessed drug release, cell proliferation, morphology, mineralization, and gene expression. VPA was rapidly released from the scaffold, with over 90% cumulative release within seven days. Cells cultured on VPA-loaded scaffolds exhibited significantly enhanced proliferation and mineralization compared to the control. VPA treatment upregulated osteocalcin and runt-related transcription factor 2 (Runx-2) expression, key markers of osteogenic differentiation. The CH-G scaffold, particularly with 1 mM/L VPA, demonstrates excellent biocompatibility and promotes hBMSC-mediated bone regeneration. This novel approach holds promise for future applications in bone tissue engineering. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
15. Inhibition of GSK-3β Enhances Osteoblast Differentiation of Human Mesenchymal Stem Cells through Wnt Signalling Overexpressing Runx2
- Author
-
AlMuraikhi, Nihal, primary, Binhamdan, Sarah, additional, Alaskar, Hanouf, additional, Alotaibi, Amal, additional, Tareen, Sumaiya, additional, Muthurangan, Manikandan, additional, and Alfayez, Musaad, additional
- Published
- 2023
- Full Text
- View/download PDF
16. Profiling of G-Protein Coupled Receptors in Adipose Tissue and Differentiating Adipocytes Offers a Translational Resource for Obesity/Metabolic Research
- Author
-
Al Mahri, Saeed, primary, Okla, Meshail, additional, Rashid, Mamoon, additional, Malik, Shuja Shafi, additional, Iqbal, Jahangir, additional, Al Ibrahim, Maria, additional, Dairi, Ghida, additional, Mahmood, Amer, additional, Muthurangan, Manikandan, additional, Yaqinuddin, Ahmed, additional, and Mohammad, Sameer, additional
- Published
- 2023
- Full Text
- View/download PDF
17. Silver Nanoparticles Alone or in Combination with Calcium Hydroxide Modulate the Viability, Attachment, Migration, and Osteogenic Differentiation of Human Mesenchymal Stem Cells
- Author
-
Algazlan, Almaha S., primary, Almuraikhi, Nihal, additional, Muthurangan, Manikandan, additional, Balto, Hanan, additional, and Alsalleeh, Fahd, additional
- Published
- 2022
- Full Text
- View/download PDF
18. IL-1β-mediated TGFβ/SMAD signaling pathway inactivation impaired ex vivo osteogenic activity of human bone marrow-derived stromal cells
- Author
-
Muthurangan Manikandan, Amer Mahmood, Mona Elsafadi, and Musaad Alfayez
- Subjects
Stromal cell ,Chemistry ,Human bone ,Inflammation ,SMAD ,heterogeneous population ,adult stem cells ,Bone resorption ,medicine ,Cancer research ,tgfβ signaling ,cytokine signaling ,cross-talk ,medicine.symptom ,Signal transduction ,osteoblastic differentiation ,Ex vivo ,TP248.13-248.65 ,Adult stem cell ,Biotechnology - Abstract
Bone loss is caused by inflammation and is mediated by pro-inflammatory cytokines that control bone formation and bone resorption. The study aimed to determine the effect of secreted factors from human bone marrow-derived stromal cells (hBMSC) of no-heterotopic bone-forming capacity (hBMSC–Bone) cells on the differentiation potential of hBMSC which capable of creating bone in-vivo (hBMSC+Bone) and dissect the molecular signature of these cells for understanding the complicated relationship of stem cells and microenvironment. hBMSC cultures are heterogenous with respect to differentiation and function. However, the nature of interaction between different cell populations within hBMSC cultures is poorly investigated. We employed two clonal hBMSC lines which exhibit different functional phenotypes based on the presence of either high or low osteoblastic differentiation capacity, bone forming (hBMSC+Bone) and non-bone forming (hBMSC−Bone), and examined their biological interaction. Adding conditioned media (CM) of hBMSC−Bone cultures resulted in suppression of cell proliferation and osteoblasts differentiation of hBMSC+Bone. Microarray analysis of CM-treated hBMSC+Bone revealed significant enrichment of several pathways, including TGFβ signaling. Follow-up experiments corroborated the inhibitory effects on TGFβ signaling as evidenced by decreased SMAD2 phosphorylation and TGFβ-responsive genes (TAGLN, ACTA2 and TPM1). Interestingly, IL1β is highly expressed in hBMSC−Bone and is present in its CM. Incubating hBMSC−Bone with rhIL-1RI rescued the functional phenotype of hBMSC−Bone with respect to cell proliferation and differentiation into osteoblasts, and upregulated TGFβ-responsive genes. These data demonstrated that IL1β-TGFβ signaling is part of the intercellular communication within the heterogenous population of hBMSCs and regulates their commitment to osteoblastic fate. Supplemental data for this article is available online at https://doi.org/10.1080/13102818.2021.1939784 .
- Published
- 2021
19. Transgelin is a poor prognostic factor associated with advanced colorectal cancer (CRC) stage promoting tumor growth and migration in a TGFβ-dependent manner
- Author
-
Tasneem Shinwari, Mona Elsafadi, Nehad M. Alajez, Mohammad Mobarak, Radhakrishnan Vishnubalaji, Muthurangan Manikandan, Moustapha Kassem, Abdullah Aldahmash, Amer Mahmood, Musaad Alfayez, and Sami Almalki
- Subjects
Cancer Research ,Angiogenesis ,Colorectal cancer ,medicine.medical_treatment ,Immunology ,Mice, Nude ,Muscle Proteins ,Biology ,p38 Mitogen-Activated Protein Kinases ,Actin cytoskeleton organization ,Article ,Targeted therapy ,Transforming Growth Factor beta1 ,Cellular and Molecular Neuroscience ,Cell growth ,Cell Movement ,Databases, Genetic ,medicine ,Animals ,Humans ,Gene Regulatory Networks ,Neoplasm Invasiveness ,lcsh:QH573-671 ,Cell Proliferation ,Neoplasm Staging ,lcsh:Cytology ,Microfilament Proteins ,Cell migration ,Cell Biology ,medicine.disease ,HCT116 Cells ,Tumor Burden ,Colon cancer ,Gene Expression Regulation, Neoplastic ,Tumor progression ,Cancer cell ,Cancer research ,Female ,Colorectal Neoplasms ,HT29 Cells ,Transforming growth factor ,Signal Transduction - Abstract
Colorectal cancer (CRC) is the fourth most common cancer type globally. Investigating the signaling pathways that maintain cancer cell phenotype can identify new biomarkers for targeted therapy. Aberrant transforming growth factor-β (TGFβ) signaling has been implicated in CRC progression, however, the exact mechanism by which TGFβ exerts its function is still being unraveled. Herein, we investigated TAGLN expression, prognostic value, and its regulation by TGFβ in CRC. While TAGLN was generally found to be downregulated in CRC, elevated expression of TAGLN was associated with advanced CRC stage and predicted poor overall survival (hazard ratio (HR) = 1.8, log-rank test P-value = 0.014) and disease-free survival (HR = 1.6, log-rank test P-value = 0.046), hence implicating TAGLN as poor prognostic factor in CRC. Forced expression of TAGLN was associated with enhanced CRC cell proliferation, clonogenic growth, cell migration and in vivo tumor formation in immunocompromised mice, while targeted depletion of TAGLN exhibited opposing biological effects. Global gene expression profiling of TAGLN-overexpressing or TAGLN-deficient CRC cell lines revealed deregulation of multiple cancer-related genes and signaling pathways. Transmission electron microscopy (TEM) revealed ultrastructural changes due to loss of TAGLN, including disruption of actin cytoskeleton organization and aberrant actin filament distribution. Hierarchical clustering, principle component, and ingenuity pathway analyses revealed distinct molecular profile associated with TAGLNhigh CRC patients with remarkable activation of a number of mechanistic networks, including SMARCA4, TGFβ1, and P38 MAPK. The P38 MAPK was the top predicted upstream regulator network promoting cell movement through regulation of several intermediate molecules, including TGFβ1. Concordantly, functional categories associated with cellular movement and angiogenesis were also enriched in TAGLNhigh CRC, supporting a model for the molecular mechanisms linking TGFβ-induced upregulation of TAGLN and CRC tumor progression and suggesting TAGLN as potential prognostic marker associated with advanced CRC pathological stage.
- Published
- 2020
- Full Text
- View/download PDF
20. Silver Nanoparticles Alone or in Combination with Calcium Hydroxide Modulate the Viability, Attachment, Migration, and Osteogenic Differentiation of Human Mesenchymal Stem Cells.
- Author
-
Algazlan, Almaha S., Almuraikhi, Nihal, Muthurangan, Manikandan, Balto, Hanan, and Alsalleeh, Fahd
- Subjects
CALCIUM hydroxide ,MESENCHYMAL stem cell differentiation ,SILVER nanoparticles ,REVERSE transcriptase polymerase chain reaction ,RUNX proteins ,TRANSFORMING growth factors ,BIOMATERIALS - Abstract
This study aimed to evaluate the effect of silver nanoparticles (AgNPs) alone or in combination with calcium hydroxide (Ca(OH)
2 ) on the proliferation, viability, attachment, migration, and osteogenic differentiation of human mesenchymal stem cells (hMSCs). Different concentrations of AgNPs alone or mixed with Ca(OH)2 were prepared. Cell proliferation was measured using AlamarBlue, and hMSCs attachment to dentin disks was evaluated using scanning electron microscopy. Live–dead imaging was performed to assess apoptosis. Wound healing ability was determined using the scratch-migration assay. To evaluate osteogenic differentiation, the expression of Runt-related transcription factor (RUNX2), Transforming growth factor beta-1 (TGF-β1), Alkaline Phosphatase (ALP), and Osteocalcin (OCN) were measured using real-time reverse transcriptase polymerase chain reaction. ALP staining and activity were also performed as indicators of osteogenic differentiation. AgNPs alone seemed to favor cell attachment. Lower concentrations of AgNPs enhanced cell proliferation. AgNP groups showed markedly less apoptosis. None of the medicaments had adverse effects on wound closure. The expression of TGF-β1 was significantly upregulated in all groups, and OCN was highly expressed in the AgNP groups. AgNPs 0.06% showed the most enhanced ALP gene expression levels, activity, and marked cytochemical staining. In conclusion, AgNPs positively affect hMSCs, making them a potential biomaterial for various clinical applications. [ABSTRACT FROM AUTHOR]- Published
- 2023
- Full Text
- View/download PDF
21. Concurrent targeting of BMI1 and CDK4/6 abrogates tumor growth in vitro and in vivo
- Author
-
Radhakrishnan Vishnubalaji, Nehad M. Alajez, Ramesh Elango, Musaad Alfayez, Yasser A. Alshawakir, Abdullah Aldahmash, Sarah Binhamdan, Abdulaziz Siyal, and Muthurangan Manikandan
- Subjects
Cancer therapy ,endocrine system diseases ,Cell Survival ,Pyridines ,Molecular biology ,lcsh:Medicine ,Antineoplastic Agents ,Apoptosis ,Palbociclib ,Heterocyclic Compounds, 2-Ring ,Piperazines ,Article ,Transcriptome ,Cell Movement ,Cell Line, Tumor ,Humans ,E2F ,lcsh:Science ,Protein Kinase Inhibitors ,Mitogen-Activated Protein Kinase 7 ,Cell Proliferation ,Multidisciplinary ,biology ,Cell growth ,Cell Cycle ,lcsh:R ,Wnt signaling pathway ,Cyclin-Dependent Kinase 4 ,Cell migration ,Cyclin-Dependent Kinase 6 ,Thiazoles ,BMI1 ,Cancer research ,biology.protein ,lcsh:Q ,Cyclin-dependent kinase 6 - Abstract
Despite recent advances in cancer management and therapy, resistance to cytotoxic medications remains a major clinical challenge; hence, combination-based anti-cancer treatment regimens are currently gaining momentum. PTC-209 reduced BMI1 protein expression, while palbociclib inhibited CDK4, Rb, and pRbSer795 protein expression in MDA-MB-231 cells. PTC-209 and palbociclib exhibited dose-dependent cytotoxic effects against MDA-MB-231 (breast), HCT116 (colon), and PC-3 (prostate) models, which was more profound in the combination group. Transcriptome and pathway analyses revealed inhibition of insulin signaling, focal adhesion, DNA damage response, and Wnt/pluripotency signaling pathways as well as cell proliferation, and cellular movement functional categories by PTC-209. Transcriptome and pathway analyses revealed palbociclib to mainly affect cell cycle progression and survival. Upstream analysis identified several networks affected by PTC-209 (EZH2, IFNB1, TRIB3, EGFR, SREBF1, IL1A, ERG, TGFB1, MAX, MNT) and palbociclib (RABL6, MITF, RARA, TAL1, AREG, E2F3, FOXM1, ESR1, ERBB2, and E2F). PTC-209 and palbociclib reduced colony and sphere formation, cell migration, and cell viability, which was further enhanced in the combination group. Concordantly, combination of PTC-209 and palbociclib exhibited more profound effects on MDA-MB-231 tumor formation in vivo. Our data suggest concurrent targeting of BMI1 and CDK4/CDK6 might provide novel therapeutic opportunity for breast, colon, and prostate cancer.
- Published
- 2019
- Full Text
- View/download PDF
22. Convergence of TGFβ and BMP signaling in regulating human bone marrow stromal cell differentiation
- Author
-
Amer Mahmood, Nehad M. Alajez, Muthurangan Manikandan, Tasneem Shinwari, Sami Almalki, Mona Elsafadi, Moustapha Kassem, Musaad Alfayez, and Abdullah Aldahmash
- Subjects
Stromal cell ,Science ,Down-Regulation ,Bone Morphogenetic Protein 4 ,Biology ,Bone morphogenetic protein 2 ,Article ,Cell Line ,Transforming Growth Factor beta1 ,Downregulation and upregulation ,Osteogenesis ,Transforming Growth Factor beta ,Bone cell ,medicine ,Adipocytes ,Humans ,Gene Silencing ,Author Correction ,Serpins ,Multidisciplinary ,Adipogenesis ,Osteoblasts ,Mesenchymal stem cell ,Osteoblast ,Cell Differentiation ,Mesenchymal Stem Cells ,Cell biology ,Clone Cells ,medicine.anatomical_structure ,Medicine ,Signal transduction ,Stem cell ,Signal Transduction - Abstract
Targeting regulatory signaling pathways that control human bone marrow stromal (skeletal or mesenchymal) stem cell (hBMSC) differentiation and lineage fate determination is gaining momentum in the regenerative medicine field. Therefore, to identify the central regulatory mechanism of osteoblast differentiation of hBMSCs, the molecular phenotypes of two clonal hBMSC lines exhibiting opposite in vivo phenotypes, namely, bone forming (hBMSC+bone) and non-bone forming (hBMSC−Bone) cells, were studied. Global transcriptome analysis revealed significant downregulation of several TGFβ responsive genes, namely, TAGLN, TMP1, ACTA2, TGFβ2, SMAD6, SMAD9, BMP2, and BMP4 in hBMSC−Bone cells and upregulation on SERPINB2 and NOG. Transcriptomic data was associated with marked reduction in SMAD2 protein phosphorylation, which thereby implies the inactivation of TGFβ and BMP signaling in those cells. Concordantly, activation of TGFβ signaling in hBMSC−Bone cells using either recombinant TGFβ1 protein or knockdown of SERPINB2 TGFβ-responsive gene partially restored their osteoblastic differentiation potential. Similarly, the activation of BMP signaling using exogenous BMP4 or via siRNA-mediated knockdown of NOG partially restored the differentiation phenotype of hBMSC−Bone cells. Concordantly, recombinant NOG impaired ex vivo osteoblastic differentiation of hBMSC+Bone cells, which was associated with SERBINB2 upregulation. Our data suggests the existence of reciprocal relationship between TGFB and BMP signaling that regulates hBMSC lineage commitment and differentiation, whilst provide a plausible strategy for generating osteoblastic committed cells from hBMSCs for clinical applications.
- Published
- 2019
- Full Text
- View/download PDF
23. Author Correction: Tankyrase inhibitor XAV-939 enhances osteoblastogenesis and mineralization of human skeletal (mesenchymal) stem cells
- Author
-
Hanouf Alaskar, Nuha Almasoud, Ghaydaa Younis, Nihal AlMuraikhi, Sarah Binhamdan, Moustapha Kassem, Muthurangan Manikandan, Amal Alotaibi, and Musaad Alfayez
- Subjects
Tankyrases ,Multidisciplinary ,Osteoblasts ,Chemistry ,Science ,Mesenchymal stem cell ,Cell Differentiation ,Mesenchymal Stem Cells ,Mineralization (soil science) ,Alkaline Phosphatase ,Cell biology ,Osteogenesis ,Medicine ,Humans ,Author Correction ,Heterocyclic Compounds, 3-Ring ,Signal Transduction - Abstract
Tankyrase is part of poly (ADP-ribose) polymerase superfamily required for numerous cellular and molecular processes. Tankyrase inhibition negatively regulates Wnt pathway. Thus, Tankyrase inhibitors have been extensively investigated for the treatment of clinical conditions associated with activated Wnt signaling such as cancer and fibrotic diseases. Moreover, Tankyrase inhibition has been recently reported to upregulate osteogenesis through the accumulation of SH3 domain-binding protein 2, an adaptor protein required for bone metabolism. In this study, we investigated the effect of Tankyrase inhibition in osteoblast differentiation of human skeletal (mesenchymal) stem cells (hMSCs). A Tankyrase inhibitor, XAV-939, identified during a functional library screening of small molecules. Alkaline phosphatase activity and Alizarin red staining were employed as markers for osteoblastic differentiation and in vitro mineralized matrix formation, respectively. Global gene expression profiling was performed using the Agilent microarray platform. XAV-939, a Tankyrase inhibitor, enhanced osteoblast differentiation of hBMSCs as evidenced by increased ALP activity, in vitro mineralized matrix formation, and upregulation of osteoblast-related gene expression. Global gene expression profiling of XAV-939-treated cells identified 847 upregulated and 614 downregulated mRNA transcripts, compared to vehicle-treated control cells. It also points towards possible changes in multiple signaling pathways, including TGFβ, insulin signaling, focal adhesion, estrogen metabolism, oxidative stress, RANK-RANKL (receptor activator of nuclear factor κB ligand) signaling, Vitamin D synthesis, IL6, and cytokines and inflammatory responses. Further bioinformatic analysis, employing Ingenuity Pathway Analysis identified significant enrichment in XAV-939-treated cells of functional categories and networks involved in TNF, NFκB, and STAT signaling. We identified a Tankyrase inhibitor (XAV-939) as a powerful enhancer of osteoblastic differentiation of hBMSC that may be useful as a therapeutic option for treating conditions associated with low bone formation.
- Published
- 2021
24. MicroRNA-3148 acts as molecular switch promoting malignant transformation and adipocytic differentiation of immortalized human bone marrow stromal cells via direct targeting of the SMAD2/TGFβ pathway
- Author
-
Ibrahim O. Alanazi, Moustapha Kassem, Ammar C. Al-Rikabi, Dalia Ali, Nehad M. Alajez, Dana Hamam, Radhakrishnan Vishnubalaji, Rimi Hamam, Assim A. Alfadda, Hicham Benabdelkamel, Ramesh Elango, Musaad Alfayez, Abdul Aziz Siyal, Muthurangan Manikandan, Abdullah Aldahmash, and Afshan Masood
- Subjects
0301 basic medicine ,Cancer Research ,Stromal cell ,Immunology ,Biology ,medicine.disease_cause ,lcsh:RC254-282 ,Article ,Malignant transformation ,03 medical and health sciences ,Cellular and Molecular Neuroscience ,0302 clinical medicine ,microRNA ,medicine ,lcsh:QH573-671 ,Regulation of gene expression ,lcsh:Cytology ,Mesenchymal stem cell ,Cell Biology ,Oncogenes ,lcsh:Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,Gene expression profiling ,030104 developmental biology ,030220 oncology & carcinogenesis ,miRNAs ,Cancer research ,Stem cell ,Carcinogenesis - Abstract
MicroRNAs (miRs/miRNAs) play a key role in posttranscriptional regulation of gene expression and are implicated in a number of physiological and pathological conditions, including cellular malignant transformation. In the current study, we investigated the role of miR-3148 in regulating human stromal (mesenchymal) stem cell (hMSC) differentiation and transformation. Stable expression of miR-3148 in telomerized hMSC (hMSC-miR-3148) led to significant increase in in vitro adipocytic differentiation and suppression of osteoblastic differentiation. Concordantly, global gene expression profiling revealed significant enrichment in cholesterol biosynthesis pathway, and pathways related to enhanced cell movement and survival, whereas processes related to bone and connective tissue developments, cell death, apoptosis, and necrosis were downregulated. Global proteomic analysis using 2D-DIGE followed by mass spectrometry (MS) revealed significant changes in protein expression in hMSC-miR-3148 and enrichment in protein networks associated with carcinogenesis. Functional studies revealed that hMSC-miR-3148 exhibited enhanced in vitro cell proliferation, colony formation, migration, invasion, sphere formation, doxorubicin resistance, and increased active number of cells in S and G2/M cell cycle phases and formed sarcoma-like tumors with adipocyte infiltration when implanted into immunocompromised mice. SMAD2 was identified as bone fide gene target for miR-3148 using qRT-PCR, Western blotting, and UTR-based reporter assay. In agreement with our data, SMAD2 expression was downregulated in 47% of patients with soft tissue sarcoma. Bioinformatics analysis revealed that elevated miR-3148 expression correlates with poor prognosis in several human cancer types, including sarcoma. Our study identified miR-3148 as factor regulating hMSC differentiation and is involved in promoting malignant transformation of telomerized hMSC.
- Published
- 2020
- Full Text
- View/download PDF
25. Tankyrase inhibitor XAV-939 enhances osteoblastogenesis and mineralization of human skeletal (mesenchymal) stem cells
- Author
-
Nuha Almasoud, Musaad Alfayez, Nihal AlMuraikhi, Ghaydaa Younis, Moustapha Kassem, Sarah Binhamdan, Muthurangan Manikandan, Hanouf Alaskar, and Amal Alotaibi
- Subjects
Cell biology ,Multidisciplinary ,Drug discovery ,Molecular biology ,Chemistry ,lcsh:R ,Mesenchymal stem cell ,Wnt signaling pathway ,lcsh:Medicine ,Osteoblast ,Stem cells ,Article ,Gene expression profiling ,medicine.anatomical_structure ,Downregulation and upregulation ,Gene expression ,medicine ,lcsh:Q ,Stem cell ,Signal transduction ,lcsh:Science - Abstract
Tankyrase is part of poly (ADP-ribose) polymerase superfamily required for numerous cellular and molecular processes. Tankyrase inhibition negatively regulates Wnt pathway. Thus, Tankyrase inhibitors have been extensively investigated for the treatment of clinical conditions associated with activated Wnt signaling such as cancer and fibrotic diseases. Moreover, Tankyrase inhibition has been recently reported to upregulate osteogenesis through the accumulation of SH3 domain-binding protein 2, an adaptor protein required for bone metabolism. In this study, we investigated the effect of Tankyrase inhibition in osteoblast differentiation of human skeletal (mesenchymal) stem cells (hMSCs). A Tankyrase inhibitor, XAV-939, identified during a functional library screening of small molecules. Alkaline phosphatase activity and Alizarin red staining were employed as markers for osteoblastic differentiation and in vitro mineralized matrix formation, respectively. Global gene expression profiling was performed using the Agilent microarray platform. XAV-939, a Tankyrase inhibitor, enhanced osteoblast differentiation of hBMSCs as evidenced by increased ALP activity, in vitro mineralized matrix formation, and upregulation of osteoblast-related gene expression. Global gene expression profiling of XAV-939-treated cells identified 847 upregulated and 614 downregulated mRNA transcripts, compared to vehicle-treated control cells. It also points towards possible changes in multiple signaling pathways, including TGFβ, insulin signaling, focal adhesion, estrogen metabolism, oxidative stress, RANK-RANKL (receptor activator of nuclear factor κB ligand) signaling, Vitamin D synthesis, IL6, and cytokines and inflammatory responses. Further bioinformatic analysis, employing Ingenuity Pathway Analysis identified significant enrichment in XAV-939-treated cells of functional categories and networks involved in TNF, NFκB, and STAT signaling. We identified a Tankyrase inhibitor (XAV-939) as a powerful enhancer of osteoblastic differentiation of hBMSC that may be useful as a therapeutic option for treating conditions associated with low bone formation.
- Published
- 2020
- Full Text
- View/download PDF
26. Molecular profiling of ALDH1+ colorectal cancer stem cells reveals preferential activation of MAPK, FAK, and oxidative stress pro-survival signalling pathways
- Author
-
Radhakrishnan Vishnubalaji, Muthurangan Manikandan, Moustapha Kassem, Abdullah Aldahmash, Musaad Alfayez, Rimi Hamam, Mohamed Fahad, and Nehad M. Alajez
- Subjects
cancer stem cells ,0301 basic medicine ,Tumour heterogeneity ,Colorectal cancer ,Resistance ,ALDH ,colorectal cancer ,Microarray ,Metastasis ,resistance ,03 medical and health sciences ,0302 clinical medicine ,Cancer stem cell ,medicine ,biology ,Cancer stem cells ,business.industry ,medicine.disease ,Molecular medicine ,ALDH1A1 ,Gene expression profiling ,030104 developmental biology ,Oncology ,030220 oncology & carcinogenesis ,biology.protein ,Cancer research ,Stem cell ,business ,microarray ,Research Paper - Abstract
// Radhakrishnan Vishnubalaji 1, 2 , Muthurangan Manikandan 1 , Mohamed Fahad 1 , Rimi Hamam 1, 3 , Musaad Alfayez 1 , Moustapha Kassem 1, 2, 4 , Abdullah Aldahmash 1, 5 and Nehad M. Alajez 1 1 Stem Cell Unit, Department of Anatomy, College of Medicine, King Saud University, Riyadh, Saudi Arabia 2 Molecular Endocrinology Unit (KMEB), Department of Endocrinology, University Hospital of Odense and University of Southern Denmark, Odense, Denmark 3 Departement de Medecine, Universite de Montreal, Montreal, Canada 4 Department of Cellular and Molecular Medicine, Danish Stem Cell Center (DanStem), University of Copenhagen, Copenhagen, Denmark 5 Prince Naif Health Research Center, King Saud University, Riyadh, Saudi Arabia Correspondence to: Nehad M. Alajez, email: nalajez@ksu.edu.sa Keywords: colorectal cancer; cancer stem cells; ALDH; microarray; resistance Received: August 02, 2017 Accepted: February 01, 2018 Published: February 05, 2018 ABSTRACT Tumour heterogeneity leads to variable clinical response and inaccurate diagnostic and prognostic assessment. Cancer stem cells (CSCs) represent a subpopulation responsible for invasion, metastasis, therapeutic resistance, and recurrence in many human cancer types. However, the true identity of colorectal cancer (CRC) SCs remains elusive. Here, we aimed to characterize and define the gene expression portrait of CSCs in CRC-model SW403 cells. We found that ALDH + positive cells are clonogenic and highly proliferative; their global gene expression profiling-based molecular signature revealed gene enrichment related to DNA damage, MAPK, FAK, oxidative stress response, and Wnt signalling. ALDH + cells showed enhanced ROS stress resistance, whereas MAPK/FAK pathway pharmacologic inhibition limited their survival. Conversely, 5-fluorouracil increased the ALDH + cell fraction among the SW403, HCT116 and SW620 CRC models. Notably, analysis of ALDH1A1 and POU5F1 expression levels in cohorts of 462 or 420 patients for overall (OS) or disease-free (DFS) survival, respectively, obtained from the Cancer Genome Atlas CRC dataset, revealed strong association between elevated expression and poor OS ( p = 0.006) and poor DFS ( p = 0.05), thus implicating ALDH1A1 and POU5F1 in CRC prognosis. Our data reveal distinct molecular signature of ALDH + CSCs in CRC and suggest pathways relevant for successful targeted therapies and management of CRC.
- Published
- 2018
- Full Text
- View/download PDF
27. ACI/EG eutectic mixture mediated synthesis, characterization and in vitro osteoblast differentiation assessment of spiropyrrolo[1,2-b]isoquinoline analogues
- Author
-
Muthurangan Manikandan, Natarajan Arumugam, Ali Aldalbahi, Dhanaraj Premnath, Mostafizur Rahaman, Raju Suresh Kumar, Govindasami Periyasami, and Musaad Alfayez
- Subjects
0301 basic medicine ,chemistry.chemical_classification ,Molecular model ,010405 organic chemistry ,Stereochemistry ,Chemistry ,General Chemical Engineering ,Osteoblast ,General Chemistry ,01 natural sciences ,In vitro ,0104 chemical sciences ,03 medical and health sciences ,chemistry.chemical_compound ,030104 developmental biology ,medicine.anatomical_structure ,Cell culture ,medicine ,Isoquinoline ,Acetylcholine ,medicine.drug ,Eutectic system ,Tricyclic - Abstract
An eco-friendly acetylcholine iodide–ethylene glycol (ACI/EG) deep eutectic mixture mediated green protocol has been developed for the synthesis of hitherto unexplored multi-functionalized linear tricyclic spiropyrrolo[1,2-b]isoquinoline analogues. The effects of the synthesized compounds on the osteoblast differentiation of hBMSC-TERT cell lines were investigated and promising results were observed with significant IC50 values. In addition, molecular modeling simulations were also performed with the 3D structure of BMP-2 to reveal binding interactions and orientations of highly potent spiropyrrolo[1,2-b]isoquinoline analogues.
- Published
- 2018
- Full Text
- View/download PDF
28. Author Correction: Convergence of TGFβ and BMP signaling in regulating human bone marrow stromal cell differentiation
- Author
-
Muthurangan Manikandan, Mona Elsafadi, Amer Mahmood, Nehad M. Alajez, Moustapha Kassem, Abdullah Aldahmash, Sami Almalki, Musaad Alfayez, and Tasneem Shinwari
- Subjects
Multidisciplinary ,Stromal cell ,Bmp signaling ,lcsh:R ,lcsh:Medicine ,Human bone ,lcsh:Q ,Convergence (relationship) ,Biology ,lcsh:Science ,Cell biology - Abstract
An amendment to this paper has been published and can be accessed via a link at the top of the paper.
- Published
- 2019
- Full Text
- View/download PDF
29. Notch Signaling Inhibition by LY411575 Attenuates Osteoblast Differentiation and Decreased Ectopic Bone Formation Capacity of Human Skeletal (Mesenchymal) Stem Cells
- Author
-
Dalia Ali, Radhakrishnan Vishnubalaji, Muhammad Atteya, Abdullah Aldahmash, Muthurangan Manikandan, Moustapha Kassem, Nihal AlMuraikhi, Musaad Alfayez, Abdulaziz Siyal, and Nehad M. Alajez
- Subjects
lcsh:Internal medicine ,Article Subject ,Cellular differentiation ,Mesenchymal stem cell ,Notch signaling pathway ,Osteoblast ,Cell Biology ,Cell biology ,Gene expression profiling ,chemistry.chemical_compound ,medicine.anatomical_structure ,chemistry ,medicine ,Stem cell ,Signal transduction ,lcsh:RC31-1245 ,Molecular Biology ,Sirius Red ,Research Article - Abstract
Background. Chemical biology approaches using small molecule inhibitors targeting specific signaling pathways are useful tools to dissect the molecular mechanisms governing stem cell differentiation and for their possible use in therapeutic interventions. Methods. Stem cell signaling small molecule library functional screen was performed employing human bone marrow skeletal (mesenchymal) stem cells (hBMSCs). Alkaline phosphatase (ALP) activity and formation of mineralized matrix visualized by Alizarin red staining were employed as markers for osteoblastic differentiation. Global gene expression profiling was conducted using the Agilent microarray platform, and data normalization and bioinformatics were performed using GeneSpring software. Pathway analyses were conducted using the Ingenuity Pathway Analysis (IPA) tool. In vivo ectopic bone formation was performed using hBMSC mixed with hydroxyapatite–tricalcium phosphate granules that were implanted subcutaneously in 8-week-old female nude mice. Hematoxylin and eosin staining and Sirius red staining were performed to identify bone formation in vivo. Results. Among the tested molecules, LY411575, a potent γ-secretase and Notch signaling inhibitor, exhibited significant inhibitory effects on osteoblastic differentiation of hBMSCs manifested by reduced ALP activity, mineralized matrix formation, and decreased osteoblast-specific gene expression as well as in vivo ectopic bone formation. Global gene expression profiling of LY411575-treated cells revealed changes in multiple signaling pathways, including focal adhesion, insulin, TGFβ, IL6, and Notch signaling, and decreased the expression of genes associated with functional categories of tissue development. Among the affected signaling networks were TGFβ1, SPP1, and ERK regulatory networks. Conclusions. We identified γ-secretase inhibitor (LY411575) as a potent regulator of osteoblastic differentiation of hBMSC that may be useful as a therapeutic option for treating conditions associated with ectopic bone formation.
- Published
- 2019
- Full Text
- View/download PDF
30. Hedgehog Signaling Inhibition by Smoothened Antagonist BMS-833923 Reduces Osteoblast Differentiation and Ectopic Bone Formation of Human Skeletal (Mesenchymal) Stem Cells
- Author
-
Nehad M. Alajez, Radhakrishnan Vishnubalaji, Nuha Almasoud, Nihal AlMuraikhi, Dalia Ali, Muthurangan Manikandan, Musaad Alfayez, Ghaydaa Younis, Muhammad Atteya, Moustapha Kassem, Sarah Binhamdan, Abdulaziz Siyal, and Abdullah Aldahmash
- Subjects
lcsh:Internal medicine ,Article Subject ,Chemistry ,Osteoblast ,Cell Biology ,Hedgehog signaling pathway ,Bone remodeling ,Cell biology ,medicine.anatomical_structure ,medicine ,Stem cell ,lcsh:RC31-1245 ,Smoothened ,Molecular Biology ,Endochondral ossification ,Hedgehog ,Research Article ,Smoothened Antagonist BMS-833923 - Abstract
Background. Hedgehog (Hh) signaling is essential for osteoblast differentiation of mesenchymal progenitors during endochondral bone formation. However, the critical role of Hh signaling during adult bone remodeling remains to be elucidated. Methods. A Smoothened (SMO) antagonist/Hedgehog inhibitor, BMS-833923, identified during a functional screening of a stem cell signaling small molecule library, was investigated for its effects on the osteoblast differentiation of human skeletal (mesenchymal) stem cells (hMSC). Alkaline phosphatase (ALP) activity and Alizarin red staining were employed as markers for osteoblast differentiation and in vitro mineralization capacity, respectively. Global gene expression profiling was performed using the Agilent® microarray platform. Effects on in vivo ectopic bone formation were assessed by implanting hMSC mixed with hydroxyapatite-tricalcium phosphate granules subcutaneously in 8-week-old female nude mice, and the amount of bone formed was assessed using quantitative histology. Results. BMS-833923, a SMO antagonist/Hedgehog inhibitor, exhibited significant inhibitory effects on osteoblast differentiation of hMSCs reflected by decreased ALP activity, in vitro mineralization, and downregulation of osteoblast-related gene expression. Similarly, we observed decreased in vivo ectopic bone formation. Global gene expression profiling of BMS-833923-treated compared to vehicle-treated control cells, identified 348 upregulated and 540 downregulated genes with significant effects on multiple signaling pathways, including GPCR, endochondral ossification, RANK-RANKL, insulin, TNF alpha, IL6, and inflammatory response. Further bioinformatic analysis employing Ingenuity Pathway Analysis revealed significant enrichment in BMS-833923-treated cells for a number of functional categories and networks involved in connective and skeletal tissue development and disorders, e.g., NFκB and STAT signaling. Conclusions. We identified SMO/Hedgehog antagonist (BMS-833923) as a powerful inhibitor of osteoblastic differentiation of hMSC that may be useful as a therapeutic option for treating conditions associated with high heterotopic bone formation and mineralization.
- Published
- 2019
- Full Text
- View/download PDF
31. Whole genome mRNA expression profiling revealed multiple deregulated pathways in stromal vascular fraction from erectile dysfunction patients
- Author
-
Radhakrishnan Vishnubalaji, Raed Almannie, Mohamad Habous, Abdullah Aldahmash, Muthurangan Manikandan, Nehad M. Alajez, Dulaim Alhajeri, Musaad Alfayez, Abdullah Aljarbou, and Saleh Binsaleh
- Subjects
0301 basic medicine ,Adult ,Male ,Microarray ,Biophysics ,Receptors, Antigen, T-Cell ,Adipose tissue ,Biochemistry ,03 medical and health sciences ,Erectile Dysfunction ,Transforming Growth Factor beta ,Gene expression ,Medicine ,Humans ,RNA, Messenger ,Molecular Biology ,Research Articles ,Cellular Senescence ,Aged ,Epidermal Growth Factor ,business.industry ,Genome, Human ,Gene Expression Regulation, Developmental ,Cell Differentiation ,Mesenchymal Stem Cells ,Cell Biology ,Stromal vascular fraction ,Middle Aged ,Androgen receptor ,stem cell ,ErbB Receptors ,030104 developmental biology ,Adipose Tissue ,Adipogenesis ,Receptors, Androgen ,Tissue Array Analysis ,Cancer research ,gene expression ,Blood Vessels ,Signal transduction ,Stem cell ,Stromal Cells ,business ,Research Article ,Penis ,Signal Transduction - Abstract
Background: Stem-cell-based therapies have recently been explored in the field of erectile dysfunction (ED). However, the cellular and molecular phenotype of adipose derived stem cells (ADSCs) stromal vascular fraction (SVF) from ED patients remains largely unknown. Herein we compared the global gene expression profile in the SVF from ED patients and healthy individuals and identified altered signaling pathways between the two groups. Methods: Samples (2–5 g) of abdominal adipose tissue from ED patients (n = 6) and healthy individual controls (n = 3) undergoing elective cosmetic liposuction were collected. Immediately after removal, SVF was separated using Collagenase type I and type IV protocol. RNA was isolated and microarray experiments were conducted using the Agilent platform. Data were normalized and pathway analyses were performed using GeneSpring software. Results: Our data revealed multiple differentially expressed genes between the ED and control group. Hierarchical clustering based on differentially expressed mRNAs revealed clear separation of the two groups. The distribution of the top enriched pathways for the up-regulated genes indicated enrichment in inflammatory response and T-cell receptor signaling, while pathway analysis performed on the down-regulated genes revealed enrichment in mitogen-activated protein kinase, TGF-β, senescence, FAK, adipogenesis, androgen receptor, and EGF–EGFR signaling pathways in SVF from ED patient. Conclusion: Our data revealed the existence of multiple altered signaling pathways in the SVF from ED patients, which could potentially play a role in the etiology of this disease. Therefore, therapeutic strategies targeting these pathways might provide novel therapeutic opportunity for ED patients.
- Published
- 2018
32. Stem cell library screen identified ruxolitinib as regulator of osteoblastic differentiation of human skeletal stem cells
- Author
-
Aliah R Alshanwani, Radhakrishnan Vishnubalaji, Muhammad Atteya, Abdulaziz Siyal, Musaad Alfayez, Nihal AlMuraikhi, Muthurangan Manikandan, Dalia Ali, Nehad M. Alajez, Abdullah Aldahmash, and Moustapha Kassem
- Subjects
0301 basic medicine ,Stromal cell ,Cellular differentiation ,Primary Cell Culture ,Medicine (miscellaneous) ,Mice, Nude ,Biochemistry, Genetics and Molecular Biology (miscellaneous) ,lcsh:Biochemistry ,03 medical and health sciences ,chemistry.chemical_compound ,Mice ,Osteogenesis ,Nitriles ,medicine ,Animals ,Humans ,lcsh:QD415-436 ,Endochondral ossification ,Sirius Red ,Janus Kinases ,lcsh:R5-920 ,Osteoblasts ,Chemistry ,Osteoblast ,Cell Differentiation ,Mesenchymal Stem Cells ,Cell Biology ,Cell biology ,Gene expression profiling ,030104 developmental biology ,medicine.anatomical_structure ,Pyrimidines ,Molecular Medicine ,Alkaline phosphatase ,Heterografts ,Pyrazoles ,Female ,Hydroxyapatites ,Stem cell ,lcsh:Medicine (General) ,Research Article - Abstract
Background Better understanding of the signaling pathways that regulate human bone marrow stromal stem cell (hBMSC) differentiation into bone-forming osteoblasts is crucial for their clinical use in regenerative medicine. Chemical biology approaches using small molecules targeting specific signaling pathways are increasingly employed to manipulate stem cell differentiation fate. Methods We employed alkaline phosphatase activity and staining assays to assess osteoblast differentiation and Alizarin R staining to assess mineralized matrix formation of cultured hBMSCs. Changes in gene expression were assessed using an Agilent microarray platform, and data normalization and bioinformatics were performed using GeneSpring software. For in vivo ectopic bone formation experiments, hMSCs were mixed with hydroxyapatite–tricalcium phosphate granules and implanted subcutaneously into the dorsal surface of 8-week-old female nude mice. Hematoxylin and eosin staining and Sirius Red staining were used to detect bone formation in vivo. Results We identified several compounds which inhibited osteoblastic differentiation of hMSCs. In particular, we identified ruxolitinib (INCB018424) (3 μM), an inhibitor of JAK-STAT signaling that inhibited osteoblastic differentiation and matrix mineralization of hMSCs in vitro and reduced ectopic bone formation in vivo. Global gene expression profiling of ruxolitinib-treated cells identified 847 upregulated and 822 downregulated mRNA transcripts, compared to vehicle-treated control cells. Bioinformatic analysis revealed differential regulation of multiple genetic pathways, including TGFβ and insulin signaling, endochondral ossification, and focal adhesion. Conclusions We identified ruxolitinib as an important regulator of osteoblast differentiation of hMSCs. It is plausible that inhibition of osteoblast differentiation by ruxolitinib may represent a novel therapeutic strategy for the treatment of pathological conditions caused by accelerated osteoblast differentiation and mineralization. Electronic supplementary material The online version of this article (10.1186/s13287-018-1068-x) contains supplementary material, which is available to authorized users.
- Published
- 2018
- Full Text
- View/download PDF
33. NR2F1 mediated down-regulation of osteoblast differentiation was rescued by bone morphogenetic protein-2 (BMP-2) in human MSC
- Author
-
Jamil A. Hashmi, Hassan Mohammed Almalak, Hussain AlSalman, Abdullah Aldahmash, Mona Elsafadi, Abdulaziz Siyal, Muthurangan Manikandan, Sarah Abuelreich, Musaad Alfayez, Moustapha Kassem, and Amer Mahmood
- Subjects
0301 basic medicine ,Cancer Research ,Cellular differentiation ,TGFb1 signaling ,Bone Morphogenetic Protein 2 ,Bone Marrow Cells ,Biology ,Transfection ,Bone morphogenetic protein 2 ,Transforming Growth Factor beta1 ,03 medical and health sciences ,Human bone marrow MSCs ,Osteogenesis ,medicine ,Humans ,RNA, Small Interfering ,Molecular Biology ,Transcription factor ,Endochondral ossification ,Bone Development ,COUP Transcription Factor I ,Osteoblasts ,Gene Expression Regulation, Developmental ,Osteoblast ,Cell Differentiation ,Mesenchymal Stem Cells ,Cell Biology ,BMP2 signaling ,NR2F1 ,Cell biology ,RUNX2 ,030104 developmental biology ,medicine.anatomical_structure ,DDIT3 ,Nuclear receptor ,Signal transduction ,Developmental Biology ,Signal Transduction - Abstract
Endochondral ossification is the process by which long bones are formed; the process of long bone formation is regulated by numerous factors such as transcription factors, cytokines, and extracellular matrix molecules. Human hormone Nuclear receptors (hHNR) are a family of ligand-regulated transcription factors that are activated by steroid hormones, such as estrogen and progesterone, and various lipid-soluble signals, including retinoic acid, oxysterols, and thyroid hormone. Whole genome microarray data from our previous study revealed that most hHNR's are up-regulated during osteoblast differentiation in hMSCS. NR2F1 was among the highest expressed hHNR during osteogenesis, NR2F1 belongs to the steroid/thyroid hormone nuclear receptor superfamily. NR2F1 is designated as an orphan nuclear receptor because its ligands are unknown. NR2F1 plays a wide range of roles, including cell differentiation, cancer progression, and central and peripheral neurogenesis. Identifying signaling networks involved in osteoblast differentiation is important in orchestrating new therapeutic and clinical applications in bone biology. This study aimed to identify alterations in signaling networks mediated by NR2F1 in osteoblast differentiation. siRNA-mediated down-regulation of NR2F1 leads to impairment in the differentiation of hBMSC-TERT to osteoblast; gene-expression results confirmed the down-regulation of osteoblast markers such as RUNX2, ALPL, OSC, and BSP. Global whole gene expression analysis revealed that most down-regulated genes were associated with osteoblast differentiation (DDIT3, BMP2). Pathway analysis revealed prominent signaling pathways that were down-regulated, including the TGFβ pathway and MAPK pathway. Functional studies on NR2F1 transfected cells, during osteoblast differentiation in combination with TGFβ1 and BMP-2, showed that TGFβ1 does not recover osteoblast differentiation, whereas BMP-2 rescues osteoblast differentiation in NR2F1 siRNA transfected cells. Thus, our results showed that BMP-2 could intervene in NR2F1 down-regulated signaling pathways to recover osteoblast differentiation.
- Published
- 2018
- Full Text
- View/download PDF
34. Romidepsin Promotes Osteogenic and Adipocytic Differentiation of Human Mesenchymal Stem Cells through Inhibition of Histondeacetylase Activity
- Author
-
Muthurangan Manikandan, Rimi Hamam, Elna P. Chalisserry, Dalia Ali, Abdullah Aldahmash, Musaad Alfayez, Moustapha Kassem, and Nehad M. Alajez
- Subjects
0301 basic medicine ,lcsh:Internal medicine ,Article Subject ,Chemistry ,Mesenchymal stem cell ,Cell Biology ,KLF15 ,Romidepsin ,Cell biology ,03 medical and health sciences ,030104 developmental biology ,stomatognathic system ,Multipotent Stem Cell ,Adipogenesis ,medicine ,Epigenetics ,Signal transduction ,lcsh:RC31-1245 ,Molecular Biology ,Chromatin immunoprecipitation ,Research Article ,medicine.drug - Abstract
Bone marrow mesenchymal stem cells (BMSCs) are adult multipotent stem cells that can differentiate into mesodermal lineage cells, including adipocytes and osteoblasts. However, the epigenetic mechanisms governing the lineage-specific commitment of BMSCs into adipocytes or osteoblasts are under investigation. Herein, we investigated the epigenetic effect of romidepsin, a small molecule dual inhibitor targeting HDAC1 and HDAC2 identified through an epigenetic library functional screen. BMSCs exposed to romidepsin (5 nM) exhibited enhanced adipocytic and osteoblastic differentiation. Global gene expression and signaling pathway analyses of differentially expressed genes revealed a strong enrichment of genes involved in adipogenesis and osteogenesis in romidepsin-treated BMSCs during induction into adipocytes or osteoblasts, respectively. Pharmacological inhibition of FAK signaling during adipogenesis or inhibition of FAK or TGFβsignaling during osteogenesis diminished the biological effects of romidepsin on BMSCs. The results of chromatin immunoprecipitation combined with quantitative polymerase chain reaction indicated a significant increase in H3K9Ac epigenetic markers in the promoter regions of peroxisome proliferator-activated receptor gamma (PPARγ) and KLF15 (related to adipogenesis) or SP7 (Osterix) and alkaline phosphatase (ALP) (related to osteogenesis) in romidepsin-treated BMSCs. Our data indicated that romidepsin is a novel in vitro modulator of adipocytic and osteoblastic differentiation of BMSCs.
- Published
- 2018
- Full Text
- View/download PDF
35. ACI/EG eutectic mixture mediated synthesis, characterization and
- Author
-
Govindasami, Periyasami, Natarajan, Arumugam, Mostafizur, Rahaman, Raju Suresh, Kumar, Muthurangan, Manikandan, Musaad A, Alfayez, Dhanaraj, Premnath, and Ali, Aldalbahi
- Abstract
An eco-friendly acetylcholine iodide-ethylene glycol (ACI/EG) deep eutectic mixture mediated green protocol has been developed for the synthesis of hitherto unexplored multi-functionalized linear tricyclic spiropyrrolo[1,2
- Published
- 2018
36. SERPINB2 is a novel TGFβ-responsive lineage fate determinant of human bone marrow stromal cells
- Author
-
Muthurangan Manikandan, Moustapha Kassem, Mona Elsafadi, Amer Mahmood, Nehad M. Alajez, Muhammad Atteya, Musaad Alfayez, Zahid Ali Kaimkhani, Abdullah Aldahmash, Sami Almalki, and Raed Abu Dawud
- Subjects
0301 basic medicine ,medicine.medical_treatment ,Cellular differentiation ,lcsh:Medicine ,600 Technik, Medizin, angewandte Wissenschaften::610 Medizin und Gesundheit ,0302 clinical medicine ,Transforming Growth Factor beta ,Adipocytes ,RNA, Small Interfering ,lcsh:Science ,Regulation of gene expression ,Multidisciplinary ,biology ,Gene Expression Regulation, Developmental ,Cell Differentiation ,Osteoblast ,Cell biology ,Phenotype ,medicine.anatomical_structure ,medicine.medical_specialty ,Stromal cell ,Stem-cell differentiation ,Article ,Transforming Growth Factor beta1 ,03 medical and health sciences ,Gene expression analysis ,Cell Line, Tumor ,Internal medicine ,Journal Article ,medicine ,Humans ,Cell Lineage ,Serpins ,Osteoblasts ,Gene Expression Profiling ,Growth factor ,lcsh:R ,Laboratory techniques and procedures ,Computational Biology ,Fatty acid derivative metabolic process ,Mesenchymal Stem Cells ,Transforming growth factor beta ,Fat cell differentiation ,Gene Ontology ,030104 developmental biology ,Endocrinology ,biology.protein ,lcsh:Q ,030217 neurology & neurosurgery - Abstract
TGF-β1, a multifunctional regulator of cell growth and differentiation, is the most abundant bone matrix growth factor. During differentiation of human bone stromal cells (hBMSCs), which constitute bone marrow osteoblast (OS) and adipocyte (AD) progenitor cells, continuous TGF-β1 (10 ng/ml) treatment enhanced OS differentiation as evidenced by increased mineralised matrix production. Conversely, pulsed TGF-β1 administration during the commitment phase increased mature lipid-filled adipocyte numbers. Global gene expression analysis using DNA microarrays in hBMSCs treated with TGF-β1 identified 1587 up- and 1716 down-regulated genes in OS-induced, TGF-β1-treated compared to OS-induced hBMSCs (2.0 fold change (FC), p SERPINB2) was down-regulated 3-fold in TGF-β1-treated hBMSCs. siRNA-mediated SERPINB2 inhibition enhanced OS and AD differentiation. Thus, TGF-β signalling is important for hBMSC OS and AD differentiation and SERPINB2 is a TGF-β-responsive gene that plays a negative regulatory role in hBMSC differentiation.
- Published
- 2017
- Full Text
- View/download PDF
37. Ultrastructural Characteristics of Three Undifferentiated Mouse Embryonic Stem Cell Lines and Their Differentiated Three-Dimensional Derivatives: A Comparative Study
- Author
-
Muthurangan Manikandan, Radhakrishnan Vishnubalaji, Saleh Karim, Mohammed Mobarak, Suzan Al-Harbi, May Salem Alnabaheen, Ali Alrwili, Amer Mahmood, Fatma Al-Qudsi, Abdullah Aldahmash, and Mona Elsafadi
- Subjects
Cytoplasm ,Embryoid body ,Germ layer ,Biology ,Cell Line ,Mice ,symbols.namesake ,Species Specificity ,Animals ,Embryonic Stem Cells ,Cell Nucleus ,Mice, Inbred BALB C ,Endoplasmic reticulum ,Original Articles ,Cell Biology ,Golgi apparatus ,Molecular biology ,Embryonic stem cell ,Mitochondria ,Cell biology ,Cell culture ,symbols ,Ultrastructure ,Developmental Biology ,Biotechnology - Abstract
The fine structures of mouse embryonic stem cells (mESCs) grown as colonies and differentiated in three-dimensional (3D) culture as embryoid bodies (EBs) were analyzed by transmission electron microscopy. Undifferentiated mESCs expressed markers that proved their pluripotency. Differentiated EBs expressed different differentiation marker proteins from the three germ layers. The ultrastructure of mESCs revealed the presence of microvilli on the cell surfaces, large and deep infolded nuclei, low cytoplasm-to-nuclear ratios, frequent lipid droplets, nonprominent Golgi apparatus, and smooth endoplasmic reticulum. In addition, we found prominent juvenile mitochondria and free ribosomes-rich cytoplasm in mESCs. Ultrastructure of the differentiated mESCs as EBs showed different cell arrangements, which indicate the different stages of EB development and differentiation. The morphologies of BALB/c and 129 W9.5 EBs were very similar at day 4, whereas C57BL/6 EBs were distinct from the others at day 4. This finding suggested that differentiation of EBs from different cell lines occurs in the same pattern but not at the same rate. Conversely, the ultrastructure results of BALB/c and 129 W9.5 ESCs revealed differentiating features, such as the dilated profile of a rough endoplasmic reticulum. In addition, we found low expression levels of undifferentiated markers on the outer cells of BALB/c and 129 W9.5 mESC colonies, which suggests a faster differentiation potential.
- Published
- 2014
- Full Text
- View/download PDF
38. Cytotoxicity of Different Concentrations of Three Root Canal Sealers on Human Mesenchymal Stem Cells
- Author
-
Alsubait, Sara, primary, Al Ajlan, Reem, additional, Mitwalli, Hala, additional, Aburaisi, Nour, additional, Mahmood, Amer, additional, Muthurangan, Manikandan, additional, Almadhri, Randa, additional, Alfayez, Musaad, additional, and Anil, Sukumaran, additional
- Published
- 2018
- Full Text
- View/download PDF
39. Teratoma Formation in Immunocompetent Mice After Syngeneic and Allogeneic Implantation of Germline Capable Mouse Embryonic Stem Cells
- Author
-
Radhakrishnan Vishnubalaji, Muhammad Atteya, Amer Mahmood, Suzan Al-Harbi, Muthurangan Manikandan, Husain A Al-Mubarak, Mona Elsafadi, Abdullah Aldahmash, Klaus Ingo Matthaei, May Al-Nbaheen, and Ali H. Al-Roalle
- Subjects
Cancer Research ,Mesoderm ,Pathology ,medicine.medical_specialty ,Epidemiology ,Stratified squamous epithelium ,Ectoderm ,Biology ,Germline ,Cell Line ,Mice ,medicine ,Animals ,Embryonic Stem Cells ,Mice, Inbred BALB C ,Teratoma ,Public Health, Environmental and Occupational Health ,Cell Differentiation ,medicine.disease ,Embryonic stem cell ,Mice, Inbred C57BL ,medicine.anatomical_structure ,Oncology ,Cell culture ,Immunology ,Endoderm - Abstract
Background Embryonic stem cells (ESCs) have the potential to form teratomas when implanted into immunodeficient mice, but data in immunocompetent mice are limited. We therefore investigated teratoma formation after implantation of three different mouse ESC (mESC) lines into immunocompetent mice. Materials and methods BALB/c mice were injected with three highly germline competent mESCs (129Sv, BALB/c and C57BL/6) subcutaneously or under the kidney capsule. After 4 weeks, mice were euthanized and examined histologically for teratoma development. The incidence, size and composition of teratomas were compared using Pearson Chi-square, t-test for dependent variables, one-way analysis of variance and the nonparametric Kruskal- Wallis analysis of variance and median test. Results Teratomas developed from all three cell lines. The incidence of formation was significantly higher under the kidney capsule compared to subcutaneous site and occurred in both allogeneic and syngeneic mice. Overall, the size of teratoma was largest with the 129Sv cell line and under the kidney capsule. Diverse embryonic stem cell-derived tissues, belonging to the three embryonic germ layers, were encountered, reflecting the pluripotency of embryonic stem cells. Most commonly represented tissues were nervous tissue, keratinizing stratified squamous epithelium (ectoderm), smooth muscle, striated muscle, cartilage, bone (mesoderm), and glandular tissue in the form of gut- and respiratory-like epithelia (endoderm). Conclusions ESCs can form teratomas in immunocompetent mice and, therefore, removal of undifferentiated ESC is a pre-requisite for a safe use of ESC in cell-based therapies. In addition the genetic relationship of the origin of the cell lines to the ability to transplant plays a major role.
- Published
- 2013
- Full Text
- View/download PDF
40. Characterization of Cellular and Molecular Heterogeneity of Bone Marrow Stromal Cells
- Author
-
Muthurangan Manikandan, Muhammad Atteya, Amer Mahmood, Moustapha Kassem, Mona Elsafadi, Jamil A. Hashmi, Musaad Alfayez, Zafar Iqbal, and Abdullah Aldahmash
- Subjects
0301 basic medicine ,musculoskeletal diseases ,lcsh:Internal medicine ,Stromal cell ,Article Subject ,Osteoblast ,Cell Biology ,Biology ,Molecular biology ,Transcriptome ,03 medical and health sciences ,Haematopoiesis ,030104 developmental biology ,medicine.anatomical_structure ,medicine ,CD146 ,Bone marrow ,Progenitor cell ,Stem cell ,lcsh:RC31-1245 ,Molecular Biology ,Research Article - Abstract
Human bone marrow-derived stromal stem cells (hBMSC) exhibit multiple functions, including differentiation into skeletal cells (progenitor function), hematopoiesis support, and immune regulation (nonprogenitor function). We have previously demonstrated the presence of morphological and functional heterogeneity of hBMSC cultures. In the present study, we characterized in detail two hTERT-BMSC clonal cell populations termed here CL1 and CL2 that represent an opposing phenotype with respect to morphology, markers expression: alkaline phosphatase (ALP) and CD146, andex vivodifferentiation potential. CL1 differentiated readily to osteoblasts, adipocytes, and chondrocytes as shown by expression of lineage specific genes and proteins. Whole genome transcriptome profiling of CL1 versus CL2 revealed enrichment in CL1 of bone-, mineralization-, and skeletal muscle-related genes, for example,ALP,POSTN,IGFBP5 BMP4, andCXCL12. On the other hand, CL2 transcriptome was enriched in immune modulatory genes, for example,CD14, CD99, NOTCH3, CXCL6, CFB, andCFI. Furthermore, gene expression microarray analysis of osteoblast differentiated CL1 versus CL2 showed significant upregulation in CL1 of bone development and osteoblast differentiation genes which included several homeobox genes:TBX15, HOXA2andHOXA10, andIGF1, FGFR3, BMP6, MCAM, ITGA10, IGFBP5, andALP. siRNA-based downregulation of theALPgene in CL1 impaired osteoblastic and adipocytic differentiation. Our studies demonstrate the existence of molecular and functional heterogeneity in cultured hBMSC. ALP can be employed to identify osteoblastic and adipocytic progenitor cells in the heterogeneous hBMSC cultures.
- Published
- 2016
- Full Text
- View/download PDF
41. Transgelin is a TGFβ-inducible gene that regulates osteoblastic and adipogenic differentiation of human skeletal stem cells through actin cytoskeleston organization
- Author
-
Musaed Alfayez, Rimi Hamam, Mona Elsafadi, Nehad M. Alajez, Muthurangan Manikandan, Abdullah Aldahmash, Amer Mahmood, Moustapha Kassem, and Raed Abu Dawud
- Subjects
0301 basic medicine ,Male ,Cancer Research ,Cytoskeleton organization ,Cellular differentiation ,Muscle Proteins ,600 Technik, Medizin, angewandte Wissenschaften::610 Medizin und Gesundheit ,RNA, Small Interfering/metabolism ,0302 clinical medicine ,Cell Movement ,Transforming Growth Factor beta ,Adipocytes ,RNA, Small Interfering ,Actin Cytoskeleton/metabolism ,Muscle Proteins/genetics ,Adipogenesis ,Adipocytes/cytology ,Microfilament Proteins ,Cell migration ,Cell biology ,Actin Cytoskeleton ,030220 oncology & carcinogenesis ,Original Article ,Stem cell ,Adipogenesis/genetics ,Transforming Growth Factor beta/metabolism ,Signal Transduction ,Stromal cell ,Immunology ,Foreskin ,Biology ,03 medical and health sciences ,Cellular and Molecular Neuroscience ,Cell Proliferation/genetics ,Microfilament Proteins/genetics ,Cell Movement/genetics ,Humans ,Progenitor cell ,Muscle, Skeletal ,Cell Proliferation ,Cell Nucleus ,Osteoblasts ,Cell growth ,Mesenchymal Stem Cells/cytology ,Gene Expression Profiling ,Osteoblasts/cytology ,Mesenchymal Stem Cells ,Cell Biology ,Fibroblasts ,Actin cytoskeleton ,equipment and supplies ,030104 developmental biology ,Fibroblasts/cytology ,Muscle, Skeletal/cytology ,Foreskin/cytology ,Cell Nucleus/metabolism ,Cancer research ,Signal Transduction/genetics - Abstract
Regenerative medicine is a novel approach for treating conditions in which enhanced bone regeneration is required. We identified transgelin (TAGLN), a transforming growth factor beta (TGFβ)-inducible gene, as an upregulated gene during in vitro osteoblastic and adipocytic differentiation of human bone marrow-derived stromal (skeletal) stem cells (hMSC). siRNA-mediated gene silencing of TAGLN impaired lineage differentiation into osteoblasts and adipocytes but enhanced cell proliferation. Additional functional studies revealed that TAGLN deficiency impaired hMSC cell motility and in vitro transwell cell migration. On the other hand, TAGLN overexpression reduced hMSC cell proliferation, but enhanced cell migration, osteoblastic and adipocytic differentiation, and in vivo bone formation. In addition, deficiency or overexpression of TAGLN in hMSC was associated with significant changes in cellular and nuclear morphology and cytoplasmic organelle composition as demonstrated by high content imaging and transmission electron microscopy that revealed pronounced alterations in the distribution of the actin filament and changes in cytoskeletal organization. Molecular signature of TAGLN-deficient hMSC showed that several genes and genetic pathways associated with cell differentiation, including regulation of actin cytoskeleton and focal adhesion pathways, were downregulated. Our data demonstrate that TAGLN has a role in generating committed progenitor cells from undifferentiated hMSC by regulating cytoskeleton organization. Targeting TAGLN is a plausible approach to enrich for committed hMSC cells needed for regenerative medicine application.
- Published
- 2016
- Full Text
- View/download PDF
42. In vitro differentiation of human skin-derived multipotent stromal cells into putative endothelial-like cells
- Author
-
Radhakrishnan Vishnubalaji, Muthurangan Manikandan, May Al-Nbaheen, Abdullah Aldahmash, Balamuthu Kadalmani, and Nehad M. Alajez
- Subjects
Adult ,Male ,Stromal cell ,Cellular differentiation ,Foreskin ,Cell Culture Techniques ,Biology ,Real-Time Polymerase Chain Reaction ,Regenerative Medicine ,Antigens, CD ,Human Umbilical Vein Endothelial Cells ,medicine ,Humans ,lcsh:QH301-705.5 ,Cell Shape ,Cells, Cultured ,Skin ,Stem cell transplantation for articular cartilage repair ,integumentary system ,Tissue Engineering ,Multipotent Stem Cells ,Mesenchymal stem cell ,Infant, Newborn ,Endothelial Cells ,Cell Differentiation ,Stromal vascular fraction ,Antigens, Differentiation ,Culture Media ,Cell biology ,Endothelial stem cell ,Phenotype ,medicine.anatomical_structure ,lcsh:Biology (General) ,Adipose Tissue ,Multipotent Stem Cell ,Immunology ,Bone marrow ,Research Article ,Developmental Biology - Abstract
Background Multipotent stem cells have been successfully isolated from various tissues and are currently utilized for tissue-engineering and cell-based therapies. Among the many sources, skin has recently emerged as an attractive source for multipotent cells because of its abundance. Recent literature showed that skin stromal cells (SSCs) possess mesoderm lineage differentiation potential; however, the endothelial differentiation and angiogenic potential of SSC remains elusive. In our study, SSCs were isolated from human neonatal foreskin (hNFSSCs) and adult dermal skin (hADSSCs) using explants cultures and were compared with bone marrow (hMSC-TERT) and adipose tissue-derived mesenchymal stem cells (hADMSCs) for their potential differentiation into osteoblasts, adipocytes, and endothelial cells. Results Concordant with previous studies, both MSCs and SSCs showed similar morphology, surface protein expression, and were able to differentiate into osteoblasts and adipocytes. Using an endothelial induction culture system combined with an in vitro matrigel angiogenesis assay, hNFSSCs and hADSSCs exhibited the highest tube-forming capability, which was similar to those formed by human umbilical vein endothelial cells (HUVEC), with hNFSSCs forming the most tightly packed, longest, and largest diameter tubules among the three cell types. CD146 was highly expressed on hNFSSCs and HUVEC followed by hADSSCs, and hMSC-TERT, while its expression was almost absent on hADMSCs. Similarly, higher vascular density (based on the expression of CD31, CD34, vWF, CD146 and SMA) was observed in neonatal skin, followed by adult dermal skin and adipose tissue. Thus, our preliminary data indicated a plausible relationship between vascular densities, and the expression of CD146 on multipotent cells derived from those tissues. Conclusions Our data is the first to demonstrate that human dermal skin stromal cells can be differentiated into endothelial lineage. Hence, SSCs represents a novel source of stem/stromal cells for tissue regeneration and the vascularization of engineered tissues. Moreover, the CD146 investigations suggested that the microenvironmental niche might contribute to direct stromal cells multipotency toward certain lineages, which warrants further investigation.
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.