Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
J Clin Diagn Res ; 11(4): ZC64-ZC67, 2017 Apr.
Article in English | MEDLINE | ID: mdl-28571265

ABSTRACT

INTRODUCTION: A clinician's path to success is a clean root canal system with three dimensional seal. Mechanical instrumentation of root canals alone leaves behind a smear layer covering the dentinal walls. Instrumentation must always be supported by use of irrigants which are considered as an essential prerequisite for root canal debridement. AIM: The aim of this in vitro study was to compare the efficacy of four irrigating solutions in removing the smear layer in primary root canals after hand instrumentation. MATERIALS AND METHODS: A total number of 40 human primary incisors were decoronated and split longitudinally. The specimens were divided randomly into four groups (n=10): Group I: 5.25% Sodium Hypochloride (NaOCl), Group II: 6% citric acid solution, Group III: smear clear and Group IV: 0.2% chitosan. Scanning electron microscopic analysis was performed to assess the presence or absence of smear layer at the coronal, middle and the apical portion of each canal. The data was analysed using Stastical Package For Social Sciences (SPSS) version 19.0 Armonk, NY IBM Corp soft ware. RESULTS: The pictures from the scanning electron microscopy showed that Group II exhibited better efficacy in removing smear layer without altering the normal dentinal structures with lowest mean scores (p<0.001) followed by Group III, Group IV and Group I. The presence of debris was more evident in the apical third rather than in the middle and the coronal part of the root canal. CONCLUSION: A 6% citric acid removed the smear layer more efficiently than other test irrigants in primary root canals.

2.
Oncotarget ; 8(66): 109861-109876, 2017 Dec 15.
Article in English | MEDLINE | ID: mdl-29299114

ABSTRACT

Mesenchymal stromal cells (hMSCs) have been used to understand the stromal cell properties in solid tumors because of their ablity to differentiate into most cell types. We investigated the role of EVs from hMSCs (hMSC-EVs) in breast cancer metastasis using MDA-MB-231 parental cell line and organotropic sub-lines. We demonstrated that hMSC-EVs significantly suppressed the metastatic potential of the parental cell line when compared to their organotropic sublines. hMSC-EVs induce dormancy in the parental cell line but not in their organotropic sub-lines and miR-205 and miR-31 from EV cargo played a role. Further, Ubiquitin Conjugating Enzyme E2 N (UBE2N/Ubc13) - metastasis-regulating gene, is a target of these miRNAs and silencing of UBE2N/Ubc13 expression significantly suppressed migration, invasion, and proliferation of breast cancer cells. To summarize, hMSC-EVs support primary breast tumor progression but suppress the metastasis of breast cancer cells that are not organ-committed through the UBE2N/Ubc13 pathway and play a role in premetastic niche formation.

3.
Front Biosci (Elite Ed) ; 9(1): 162-173, 2017 01 01.
Article in English | MEDLINE | ID: mdl-27814597

ABSTRACT

It is well recognized that one of the major drawbacks of using traditional two dimensional cultures to model the living systems is inaccurately reflecting the physiological manner in which modulators, nutrients, oxygen, and metabolites are applied and removed. Moreover, the two dimensional culture system poorly reflects how different cell types interact with each other in the same microenvironment. Since the first global development of three dimensional (3D) cell culture techniques in the late 1960s, this last decade has seen an explosion of studies to promote 3D models in the fields of regenerative medicine and cancer. The recent surge of interest in 3D cell culture in cancer research is attributable to the interest in developing closer to real life models. The ability to include various cell types and extracellular components reflect more the physiological conditions of tumor microenvironment. In this short review, we will discuss different approaches of 3D culture system models and techniques with a focus on the 3D interactions of cancer cells with stromal cells in the goal to reevaluate old and develop new therapeutics.


Subject(s)
Cell Culture Techniques , Neoplasms/pathology , Drug Evaluation, Preclinical/methods , Humans , Lab-On-A-Chip Devices , Neoplasms, Connective Tissue/pathology , Neoplasms, Glandular and Epithelial/pathology , Research Design , Spheroids, Cellular , Tissue Scaffolds , Tumor Cells, Cultured , Tumor Microenvironment
4.
PLoS One ; 11(11): e0166027, 2016.
Article in English | MEDLINE | ID: mdl-27812189

ABSTRACT

Studies have shown that mesenchymal stem/stromal cells (MSCs) from bone marrow are involved in the growth and metastasis of solid tumors but the mechanism remains unclear in osteosarcoma (OS). Previous studies have raised the possibility that OS cells may receive support from associated MSCs in the nutrient deprived core of the tumors through the release of supportive macromolecules and growth factors either in vesicular or non-vesicular forms. In the present study, we used stressed mesenchymal stem cells (SD-MSCs), control MSCs and OS cells to examine the hypothesis that tumor-associated MSCs in nutrient deprived core provide pro-proliferative, anti-apoptotic, and metastatic support to nearby tumor cells. Assays to study of the effects of SD-MSC conditioned media revealed that OS cells maintained proliferation when compared to OS cells grown under serum-starved conditions alone. Furthermore, OS cells in MSCs and SD-MSC conditioned media were significantly resistant to apoptosis and an increased wound healing rate was observed in cells exposed to either conditioned media or EVs from MSCs and SD-MSCs. RT-PCR assays of OS cells incubated with extracellular vesicles (EVs) from SD-MSCs revealed microRNAs that could potentially target metabolism and metastasis associated genes as predicted by in silico algorithms, including monocarboxylate transporters, bone morphogenic receptor type 2, fibroblast growth factor 7, matrix metalloproteinase-1, and focal adhesion kinase-1. Changes in the expression levels of focal adhesion kinase, STK11 were confirmed by quantitative PCR assays. Together, these data indicate a tumor supportive role of MSCs in osteosarcoma growth that is strongly associated with the miRNA content of the EVs released from MSCs under conditions that mimic the nutrient deprived core of solid tumors.


Subject(s)
Apoptosis , Cell Communication , Cell Movement , Extracellular Vesicles/pathology , Mesenchymal Stem Cells/cytology , Osteosarcoma/pathology , Oxidative Stress , Cell Line, Tumor , Cell Proliferation , Cell Survival , Culture Media, Conditioned , Gene Expression Regulation, Neoplastic , Humans , Intercellular Signaling Peptides and Proteins/genetics , MicroRNAs/genetics , Tumor Microenvironment
5.
J Indian Soc Pedod Prev Dent ; 34(3): 233-7, 2016.
Article in English | MEDLINE | ID: mdl-27461806

ABSTRACT

BACKGROUND: Children consume foods that are colorful which contain food additives that stain not only the tooth structure but also the restorations. As esthetics is of prime concern for both parents and children nowadays, long-term color stability of restorative materials is of utmost importance. AIM: To evaluate the color stability of two tooth-colored restorative materials (conventional glass ionomer cement [GIC] and giomer) when immersed in various consumable drinks and food (aerated beverage, ice candy, and health drink) at different immersion periods (low, moderate, and high). MATERIALS AND METHODS: A total of 100 specimens were made with each restorative material. Ten were used as a control and remaining (n = 90) as experimental. The experimental specimens were divided into three groups based on media of immersion (n = 30 each) and were further divided into three subgroups based on immersion time (n = 10 each). The color changes (ΔE values) were measured using spectrophotometer. RESULTS: Both the tested materials showed color change; however, conventional GIC showed greater ΔE values when compared to giomer and the samples exposed to aerated beverage resulted in highest color change. It is also noticed that greater the exposure time, higher are the ΔE values. CONCLUSION: Giomer showed more resistance to color change than conventional GIC with all the tested media and immersion regimes.


Subject(s)
Dental Materials/standards , Dental Restoration, Permanent/methods , Esthetics, Dental , Pediatric Dentistry/methods , Child , Color , Dental Restoration, Permanent/standards , Glass Ionomer Cements/standards , Humans , In Vitro Techniques , Pediatric Dentistry/standards , Spectrophotometry
6.
Stem Cells Int ; 2016: 1073140, 2016.
Article in English | MEDLINE | ID: mdl-26649044

ABSTRACT

Stem cells are proposed to continuously secrete trophic factors that potentially serve as mediators of autocrine and paracrine activities, associated with reprogramming of the tumor microenvironment, tissue regeneration, and repair. Hitherto, significant efforts have been made to understand the level of underlying paracrine activities influenced by stem cell secreted trophic factors, as little is known about these interactions. Recent findings, however, elucidate this role by reporting the effects of stem cell derived extracellular vesicles (EVs) that mimic the phenotypes of the cells from which they originate. Exchange of genetic information utilizing persistent bidirectional communication mediated by stem cell-EVs could regulate stemness, self-renewal, and differentiation in stem cells and their subpopulations. This review therefore discusses stem cell-EVs as evolving communication factors in stem cell biology, focusing on how they regulate cell fates by inducing persistent and prolonged genetic reprogramming of resident cells in a paracrine fashion. In addition, we address the role of stem cell-secreted vesicles in shaping the tumor microenvironment and immunomodulation and in their ability to stimulate endogenous repair processes during tissue damage. Collectively, these functions ensure an enormous potential for future therapies.

7.
Int J Cancer ; 138(1): 14-21, 2016 Jan 01.
Article in English | MEDLINE | ID: mdl-25559768

ABSTRACT

In recent years, the knowledge about the control of tumor microenvironment has increased and emerged as an important player in tumorigenesis. The role of normal stromal cells in the tumor initiation and progression has brought our vision in to the forefront of cell-to-cell communication. In this review, we focus on the mechanism of communication between stromal and tumor cells, which is based on the exchange of extracellular vesicles (EVs). We describe several, evergrowing, pieces of evidence that EVs transfer messages through their miRNA, lipid, protein and nucleic acid contents. A better understanding of this sophisticated method of communication between normal cancer cells may lead to developing novel approaches for personalized diagnostics and therapeutics.


Subject(s)
Extracellular Vesicles/metabolism , Neoplasms/metabolism , Neoplasms/pathology , Tumor Microenvironment , Animals , Biological Transport , Cell Communication , Extracellular Vesicles/genetics , Gene Expression Regulation, Neoplastic , Humans , Lipids , Mesenchymal Stem Cells/metabolism , MicroRNAs , Neoplasm Metastasis , Neoplasms/genetics , Proteins , Signal Transduction
8.
Curr Stem Cell Res Ther ; 11(2): 141-8, 2016.
Article in English | MEDLINE | ID: mdl-26423301

ABSTRACT

Mesenchymal Stem/stromal cell (MSCs) transplantation procedures have been used since the 1960's to treat leukemia and other diseases, but due to the risks involved only patients with life threatening illnesses were typically subjected to the transplantation procedure until the last decade. Recent advancements in transplantation techniques have made it more feasible to use it for non-life-threatening diseases. However, the potential uses for stem cells are still limited by their rarity, and, in the case of allogeneic transplants, graft-vs.-host complications. An evolving alternative to conventional stem cell therapies is induced pluripotent stem-cell derived mesenchymal stem/stromal cells (iPSC- MSCs), which have a multi-lineage potential comparable to conventionally acquired MSCs with the added benefit of being less immunoreactive. However there are still many hurdles left to be overcome before they can be used regularly for personalized therapies. This review will focus on recent advancements that have been made regarding the role MSCs play in tumor development and the potential uses iPSC-MSCs may have in future cancer treatment.


Subject(s)
Induced Pluripotent Stem Cells/transplantation , Mesenchymal Stem Cell Transplantation/trends , Mesenchymal Stem Cells , Precision Medicine , Humans
9.
Nat Commun ; 6: 7389, 2015 Jun 12.
Article in English | MEDLINE | ID: mdl-26067754

ABSTRACT

Mutant p53 (mtp53) is an oncogene that drives cancer cell proliferation. Here we report that mtp53 associates with the promoters of numerous nucleotide metabolism genes (NMG). Mtp53 knockdown reduces NMG expression and substantially depletes nucleotide pools, which attenuates GTP-dependent protein activity and cell invasion. Addition of exogenous guanosine or GTP restores the invasiveness of mtp53 knockdown cells, suggesting that mtp53 promotes invasion by increasing GTP. In addition, mtp53 creates a dependency on the nucleoside salvage pathway enzyme deoxycytidine kinase for the maintenance of a proper balance in dNTP pools required for proliferation. These data indicate that mtp53-harbouring cells have acquired a synthetic sick or lethal phenotype relationship with the nucleoside salvage pathway. Finally, elevated expression of NMG correlates with mutant p53 status and poor prognosis in breast cancer patients. Thus, mtp53's control of nucleotide biosynthesis has both a driving and sustaining role in cancer development.


Subject(s)
Brain Neoplasms/genetics , Breast Neoplasms/genetics , Gene Expression Regulation, Neoplastic/genetics , Nucleotides/metabolism , Tumor Suppressor Protein p53/genetics , Animals , Blotting, Western , Brain Neoplasms/secondary , Breast Neoplasms/metabolism , Cell Cycle , Cell Line, Tumor , Cell Proliferation/genetics , Deoxycytidine Kinase , Female , Gene Knockdown Techniques , Guanosine Triphosphate , Humans , Immunoprecipitation , Kaplan-Meier Estimate , Mice , Mutation , Neoplasm Invasiveness/genetics , Neoplasm Transplantation , Nucleosides/metabolism , Prognosis , Promoter Regions, Genetic , Proportional Hazards Models , Tumor Stem Cell Assay , Tumor Suppressor Protein p53/metabolism
10.
Oncotarget ; 6(7): 4953-67, 2015 Mar 10.
Article in English | MEDLINE | ID: mdl-25669974

ABSTRACT

Human mesenchymal stem/stromal cells (hMSCs) have been shown to support breast cancer cell proliferation and metastasis, partly through their secretome. hMSCs have a remarkable ability to survive for long periods under stress, and their secretome is tumor supportive. In this study, we have characterized the cargo of extracellular vesicular (EV) fraction (that is in the size range of 40-150nm) of serum deprived hMSCs (SD-MSCs). Next Generation Sequencing assays were used to identify small RNA secreted in the EVs, which indicated presence of tumor supportive miRNA. Further assays demonstrated the role of miRNA-21 and 34a as tumor supportive miRNAs. Next, proteomic assays revealed the presence of ≈150 different proteins, most of which are known tumor supportive factors such as PDGFR-ß, TIMP-1, and TIMP-2. Lipidomic assays verified presence of bioactive lipids such as sphingomyelin. Furthermore, metabolite assays identified the presence of lactic acid and glutamic acid in EVs. The co-injection xenograft assays using MCF-7 breast cancer cells demonstrated the tumor supportive function of these EVs. To our knowledge this is the first comprehensive -omics based study that characterized the complex cargo of extracellular vesicles secreted by hMSCs and their role in supporting breast cancers.


Subject(s)
Breast Neoplasms/metabolism , Extracellular Vesicles/metabolism , Mesenchymal Stem Cells/metabolism , MicroRNAs/metabolism , Animals , Bone Neoplasms/metabolism , Bone Neoplasms/pathology , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Proliferation/physiology , Extracellular Vesicles/pathology , Female , Heterografts , Humans , Lipid Metabolism , MCF-7 Cells , Mice , Mice, Nude , Osteosarcoma/metabolism , Osteosarcoma/pathology , Proteome/metabolism , Tumor Microenvironment , Xenograft Model Antitumor Assays
11.
Stem Cells Dev ; 21(17): 3104-13, 2012 Nov 20.
Article in English | MEDLINE | ID: mdl-22676452

ABSTRACT

Multipotent mesenchymal stem cells (MSCs) are promising candidates for regenerative cell-based therapy. The mechanisms underlying MSC differentiation and other functions relevant to therapeutic avenues remain however a matter of debate. Recent reports imply a critical role for intercellular contacts in MSC differentiation. We studied MSC differentiation to vascular smooth muscle cells (VSMCs) in a coculture model using human primary MSCs and VSMCs. We observed that under these conditions, MSCs did not undergo the expected differentiation process. Instead, they revealed an increased proliferation rate. The upregulated MSC proliferation was initiated by direct contacts of MSCs with VSMCs; indirect coculture of both cell types in transwells was ineffective. Intercellular contacts affected cell growth in a unidirectional fashion, since VSMC proliferation was not changed. We observed formation of so-called tunneling nanotubes (TNTs) between MSCs and VSMCs that revealed an intercellular exchange of a fluorescent cell tracker dye. Disruption of TNTs using cytochalasin D or latrunculin B abolished increased proliferation of MSCs initiated by contacts with VSMCs. Using specific fluorescent markers, we identified exchange of mitochondria via TNTs. By generation of VSMCs with mitochondrial dysfunction, we show that mitochondrial transfer from VSMCs to MSCs was required to regulate MSC proliferation in coculture. Our data suggest that MSC interaction with other cell types does not necessarily result in the differentiation process, but rather may initiate a proliferative response. They further point to complex machinery of intercellular communications at the place of vascular injury and to an unrecognized role of mitochondria in these processes.


Subject(s)
Cell Communication/drug effects , Cell Proliferation , Mesenchymal Stem Cells/cytology , Mitochondria/metabolism , Myocytes, Smooth Muscle/metabolism , Nanotubes , Bridged Bicyclo Compounds, Heterocyclic/pharmacology , Cell Differentiation , Cells, Cultured , Coculture Techniques , Cytochalasin D/pharmacology , Endocytosis , Flow Cytometry , Fluorescent Dyes , Humans , Intercellular Junctions/drug effects , Intercellular Junctions/metabolism , Mesenchymal Stem Cells/metabolism , Microscopy, Confocal/methods , Muscle, Smooth, Vascular/cytology , Muscle, Smooth, Vascular/metabolism , Myocytes, Smooth Muscle/cytology , Thiazolidines/pharmacology
12.
Cardiovasc Res ; 90(1): 113-21, 2011 Apr 01.
Article in English | MEDLINE | ID: mdl-21088115

ABSTRACT

AIMS: Multipotent mesenchymal stem cells (MSCs) have regenerative properties and are recognized as putative players in the pathogenesis of cardiovascular diseases. The underlying molecular mechanisms remain, however, sparsely explored. Our study was designed to elucidate a probable role for the multifunctional urokinase (uPA)/urokinase receptor (uPAR) system in MSC regulation. Though uPAR has been implicated in a broad spectrum of pathophysiological processes, nothing is known about uPAR in MSCs. METHODS AND RESULTS: uPAR was required to mobilize MSCs from the bone marrow (BM) of mice stimulated with granulocyte colony-stimulating factor (G-CSF) in vivo. An insignificant amount of MSCs was mobilized in uPAR(-/-) C57BL/6J mice, whereas in wild-type animals G-CSF induced an eight-fold increase of mobilized MSCs. uPAR(-/-) mice revealed up-regulated expression of G-CSF and stromal cell-derived factor 1 (CXCR4) receptors in BM. uPAR down-regulation leads to inhibition of human MSC migration, as shown in different migration assays. uPAR down- or up-regulation resulted in inhibition or stimulation of MSC differentiation into vascular smooth muscle cells (VSMCs) correspondingly, as monitored by changes in cell morphology and expression of specific marker proteins. Injection of fluorescently labelled MSCs in non-obese diabetic/severe combined immunodeficiency (NOD/SCID) mice after femoral artery wire injury demonstrated impaired engraftment of uPAR-deficient MSCs at the place of injury. CONCLUSIONS: These data suggest a multifaceted function of uPAR in MSC biology contributing to vascular repair. uPAR might guide and control the trafficking of MSCs to the vascular wall in response to injury or ischaemia and their differentiation towards functional VSMCs at the site of arterial injury.


Subject(s)
Cell Differentiation , Cell Movement , Mesenchymal Stem Cells/metabolism , Muscle, Smooth, Vascular/metabolism , Myocytes, Smooth Muscle/metabolism , Receptors, Urokinase Plasminogen Activator/metabolism , Vascular System Injuries/metabolism , Animals , Biomarkers/metabolism , Cell Shape , Disease Models, Animal , Femoral Artery/metabolism , Femoral Artery/pathology , Granulocyte Colony-Stimulating Factor/administration & dosage , HEK293 Cells , Hematopoietic Stem Cell Mobilization/methods , Humans , Male , Mesenchymal Stem Cell Transplantation , Mice , Mice, Inbred C57BL , Mice, Inbred NOD , Mice, Knockout , Mice, SCID , Muscle, Smooth, Vascular/injuries , Muscle, Smooth, Vascular/pathology , Myocytes, Smooth Muscle/pathology , Myocytes, Smooth Muscle/transplantation , RNA Interference , Receptors, CXCR4/metabolism , Receptors, Urokinase Plasminogen Activator/deficiency , Receptors, Urokinase Plasminogen Activator/genetics , Time Factors , Transfection , Vascular System Injuries/genetics , Vascular System Injuries/pathology , Vascular System Injuries/surgery
SELECTION OF CITATIONS
SEARCH DETAIL
...