Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters










Publication year range
1.
Front Med (Lausanne) ; 9: 906482, 2022.
Article in English | MEDLINE | ID: mdl-35911417

ABSTRACT

Successful sequencing of the human genome and evolving functional knowledge of gene products has taken genomic medicine to the forefront, soon combining broadly with traditional diagnostics, therapeutics, and prognostics in patients. Recent years have witnessed an extraordinary leap in our understanding of ocular diseases and their respective genetic underpinnings. As we are entering the age of genomic medicine, rapid advances in genome sequencing, gene delivery, genome surgery, and computational genomics enable an ever-increasing capacity to provide a precise and robust diagnosis of diseases and the development of targeted treatment strategies. Inherited retinal diseases are a major source of blindness around the world where a large number of causative genes have been identified, paving the way for personalized diagnostics in the clinic. Developments in functional genetics and gene transfer techniques has also led to the first FDA approval of gene therapy for LCA, a childhood blindness. Many such retinal diseases are the focus of various clinical trials, making clinical diagnoses of retinal diseases, their underlying genetics and the studies of natural history important. Here, we review methodologies for identifying new genes and variants associated with various ocular disorders and the complexities associated with them. Thereafter we discuss briefly, various retinal diseases and the application of genomic technologies in their diagnosis. We also discuss the strategies, challenges, and potential of gene therapy for the treatment of inherited and acquired retinal diseases. Additionally, we discuss the translational aspects of gene therapy, the important vector types and considerations for human trials that may help advance personalized therapeutics in ophthalmology. Retinal disease research has led the application of precision diagnostics and precision therapies; therefore, this review provides a general understanding of the current status of precision medicine in ophthalmology.

2.
Arch Oral Biol ; 122: 104981, 2021 Feb.
Article in English | MEDLINE | ID: mdl-33302041

ABSTRACT

OBJECTIVE: This study aims to investigate the role of p38 Mitogen-activated protein kinase (MAPK) in imparting cisplatin resistance in head and neck squamous cell carcinoma (HNSCC) cells. DESIGN: Laboratory generated cisplatin resistant HNSCC cells were treated with p38 inhibitor and were subjected to increasing dosage of cisplatin. Western blot, immunohistochemistry and RT PCR analysis were performed to investigate expression level of p-p38 and Cancer stem cell (CSC) markers in cisplatin resistant HNSCC cells with or without p38 inhibitor. Chemoresistance, wound healing capacity and Spheroids formation capacity were assessed following p38 inhibition in cisplatin resistant HNSCC cell lines. In addition, alkaline comet assay and γ-H2AX immunostaining were performed to evaluate the DNA damage response and repair abilities in cisplatin resistant HNSCC cells after p38 inhibition. RESULTS: It was observed that following p38 inhibition, cisplatin resistant HNSCC cells exhibited significant reduction in expression of CSC markers, ß-catenin, reduced migration potential and sphere forming ability along with increased apoptotic index demonstrating there was increased sensitivity towards Cisplatin. Molecular docking study identified several interface amino acid residues between p-p38 with CSC markers (Klf4 and CD44). p38 inhibited cisplatin resistant HNSCC cells also exhibited increased DNA damage as measured by Comet assay and γ-H2AX foci formation index. There was significant decrease in DNA repair as confirmed by reduced ERRC1 expression. CONCLUSIONS: Our study demonstrated that p38 MAPK inhibition can be a targeted approach to overcome resistance in HNSCC thereby escalating the effectiveness of chemotherapy in HNSCC.


Subject(s)
Cisplatin/pharmacology , Drug Resistance, Neoplasm , Head and Neck Neoplasms/enzymology , Squamous Cell Carcinoma of Head and Neck/enzymology , p38 Mitogen-Activated Protein Kinases/physiology , Cell Line, Tumor , DNA Damage , Head and Neck Neoplasms/drug therapy , Humans , Kruppel-Like Factor 4 , Molecular Docking Simulation , Neoplastic Stem Cells , Squamous Cell Carcinoma of Head and Neck/drug therapy , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors
3.
Cancers (Basel) ; 12(8)2020 Aug 12.
Article in English | MEDLINE | ID: mdl-32806555

ABSTRACT

The MDM2 gene is amplified in dedifferentiated liposarcoma (DDLPS). Treatment with MDM2 antagonists is a promising strategy to treat DDLPS; however, drug resistance is a major limitation when these drugs are used as a single agent. This study examined the impact of MDM2 antagonists on the mitogen-activated protein kinase (MAPK) pathway in DDLPS and investigated the potential synergistic activity of a MAPK kinase (MEK) inhibitor in combination with MDM2 antagonists. We identified a synergistic effect and identified the mechanism behind it. Combination effects of MDM2 antagonists and a MEK inhibitor were analyzed in a patient-derived xenograft mouse model and in DDLPS and leiomyosarcoma cell lines using different cell proliferation assays and immunoblot analysis. MDM2 antagonist (RG7388)-resistant IB115 [P4] cells and p53-silenced DDLPS cells were also established to understand the importance of functional p53. We found that MDM2 antagonists induced an upregulation of phosphorylated extracellular signal-regulated kinase (p-ERK) in DDLPS cells. The upregulation of p-ERK occurred due to mitochondrial translocation of p53, which resulted in increased production of reactive oxygen species, causing the activation of receptor tyrosine kinases (RTKs). Activated RTKs led to the activation of the downstream MEK/ERK signaling pathway. Treatment with a MEK inhibitor resulted in decreased expression of p-ERK, causing significant anti-tumor synergy when combined with MDM2 antagonists. Our results provide a framework for designing clinical studies of combination therapies in DDLPS patients.

4.
Int J Biol Macromol ; 149: 501-512, 2020 Apr 15.
Article in English | MEDLINE | ID: mdl-31953176

ABSTRACT

CD44 is one of the key cancer stem-like cell (CSC) marker and may have a potential role in tumorigenesis. In this study, we investigated the role of CD44 in prognosis of HNSCC patients, its possible crosstalk with Wnt/ß-catenin signaling and modulating cisplatin resistance. We observed increased expression of CD44 in the cut margin of recurrent HNSCC patients were associated with poor prognosis. We observed that inhibition of CD44 by using 1,2,3,4 tetrahydroisoquinoline (THIQ) modulates the expression of Wnt/ ß-catenin signaling proteins and further silencing of ß-catenin also decreases the expression of CD44. This led us to investigate the possible protein-protein interaction between CD44 and ß-catenin. Co-immunoprecipitation study illustrated possible interaction between CD44 and ß-catenin which was further confirmed by molecular docking and molecular dynamic (MD) simulation studies. Molecular docking study revealed that one interface amino acid residue Glu642 of ß -catenin interacts with Lys92 of CD44 which was also present for 20% of simulation time. Furthermore, we observed that inhibition of CD44 chemosensitizes cisplatin-resistant HNSCC cells towards cisplatin. In conclusion, this study investigated the possible role of CD44 along with Wnt/ ß-catenin signaling and their possible therapeutic role to abrogate cisplatin resistance.


Subject(s)
Carcinogenesis/genetics , Hyaluronan Receptors/genetics , Squamous Cell Carcinoma of Head and Neck/drug therapy , beta Catenin/genetics , Aged , Apoptosis/drug effects , Cell Line, Tumor/drug effects , Cell Proliferation/drug effects , Cisplatin/adverse effects , Cisplatin/pharmacology , Drug Resistance, Neoplasm/drug effects , Female , Gene Expression Regulation, Neoplastic/drug effects , Humans , Hyaluronan Receptors/antagonists & inhibitors , Male , Middle Aged , Molecular Docking Simulation , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/pathology , Squamous Cell Carcinoma of Head and Neck/genetics , Squamous Cell Carcinoma of Head and Neck/pathology , Tetrahydroisoquinolines/pharmacology , Wnt Signaling Pathway/drug effects , beta Catenin/antagonists & inhibitors
5.
Article in English | MEDLINE | ID: mdl-31585633

ABSTRACT

Cancer stem-like cells (CSCs) were reported to be linked with tumorigenesis, metastasis and resistant to chemo and radiotherapy in head and neck squamous cell carcinoma (HNSCC). In this study we investigated the role of CSCs in chemoresistance and abrogation of CSC mediated chemoresistance by combinatorial treatment with cisplatin and small molecule tankyrase inhibitor XAV-939. Two cisplatin-resistant HNSCC cells were generated by stepwise dose incremental strategy. We evaluated the chemoresistance, sphere forming capacity, extent of DNA damage and repair capacity in parental and cisplatin-resistant HNSCC cells. Furthermore, the abrogation of CSC mediated chemoresistance was evaluated in HNSCC cells with XAV-939 alone and in combination with cisplatin. It was observed that cisplatin-resistant HNSCC cell lines exhibited increase in chemoresistance, CSC phenotype and increased DNA repair capacity. We observed that combination of cisplatin and XAV-939 acts synergistically to abrogate chemoresistance by increasing DNA damage. Molecular docking study also revealed similar binding region that could contribute towards synergy predictions between cisplatin and XAV939. In conclusion, this study elucidated that combination of cisplatin and XAV-939 exerted cytotoxic and genotoxic effect to abrogate CSC mediated chemoresistance in HNSCC in synergistic manner.


Subject(s)
Antineoplastic Agents, Alkylating/pharmacology , Cisplatin/pharmacology , Head and Neck Neoplasms/pathology , Heterocyclic Compounds, 3-Ring/pharmacology , Neoplastic Stem Cells/drug effects , Squamous Cell Carcinoma of Head and Neck/pathology , Tankyrases/antagonists & inhibitors , Apoptosis/drug effects , Cell Cycle/drug effects , Cell Line, Tumor , Comet Assay , Cytokinesis , DNA Repair , Drug Resistance, Neoplasm/drug effects , Drug Synergism , Humans , Micronucleus Tests , Phenotype , RNA, Neoplasm/biosynthesis , Spheroids, Cellular/drug effects , Telomere/ultrastructure , beta Catenin/antagonists & inhibitors
6.
Article in English | MEDLINE | ID: mdl-31078500

ABSTRACT

OBJECTIVE: The presence of cancer stem-like cells (CSCs) in the majority of tumors is one of the factors responsible for disease relapse in oral squamous cell carcinoma (OSCC). In this study, we investigated the role of octamer-binding transcription factor 4 (OCT4) and Kruppel-like factor 4 (KLF4) in OSCC progression and disease relapse. STUDY DESIGN: In this study, 102 patients with OSCC were included. The expression of ß-catenin and CSC markers (KLF4 and OCT4) in surgical cut margin and tumor were investigated through Western blot analysis, immunohistochemistry, and quantitative polymerase chain reaction analysis. The χ2 test was used to evaluate the association of ß-catenin, OCT4, and KLF4 expression with clinicopathologic characteristics. Kaplan-Meier and Cox regression analyses were performed to correlate different clinical factors with the prognoses of patients with OSCC. RESULTS: We observed increased expression of OCT4, KLF4, and ß-catenin in the cut margins (CMs) in recurrent OSCC. The χ2 test exhibited recurrence as one of the key factors associated with high expression of these markers. Kaplan-Meier and COX regression analyses demonstrated that increased expression of KLF4 in the CM region of recurrent patients was independently associated with a poor prognosis. CONCLUSIONS: Our findings indicated that expression of KLF4 can be used for monitoring disease progression and may serve as prognostic marker to predict recurrence.


Subject(s)
Carcinoma, Squamous Cell , Kruppel-Like Transcription Factors/metabolism , Mouth Neoplasms , Biomarkers, Tumor , Humans , Kruppel-Like Factor 4 , Neoplasm Recurrence, Local , Prognosis
7.
Mutat Res Genet Toxicol Environ Mutagen ; 836(Pt A): 89-97, 2018 Dec.
Article in English | MEDLINE | ID: mdl-30389168

ABSTRACT

There is an intricate balance of DNA damage response and repair which determines the homeostasis of human genome function. p53 protein is widely known for its role in cell cycle regulation and tumor suppressor activity. In case of several cancers where function of p53 gene gets compromised either by mutation or partial inactivation, the role of p53 in response to DNA damage needs to be supplemented by another molecule or pathway. Due to sedentary lifestyle and exposure to genotoxic agents, genome is predisposed to chronic stress, which ultimately leads to unrepaired or background DNA damage. p38 MAPK signaling pathway is strongly activated in response to various environmental and cellular stresses. DNA damage response and the repair options have crucial links with chromosomal integrity. Telomere that regulates integrity of genome is protected by a six member shielding unit called shelterin complex which communicates with other pathways for functionality of telomeres. There are evidences that p38 gets activated through ATM in response to DNA damage. Dysfunctional telomere leads to activation of ATM which subsequently activates p38 suggesting a crosstalk between p38, ATM and shelterin complex. This review focuses on activation of p38 in response to genotoxic stress induced DNA damage in p53 mutated or compromised state and its possible cross talk with telomere shelterin proteins. Thus p38 may act as an important target to treat various diseases and in majority of cancers in p53 mutated state.


Subject(s)
DNA Damage , Signal Transduction , Telomere , Tumor Suppressor Protein p53/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism , Animals , Humans , Tumor Suppressor Protein p53/genetics , p38 Mitogen-Activated Protein Kinases/genetics
8.
Oncogenesis ; 7(7): 53, 2018 Jul 09.
Article in English | MEDLINE | ID: mdl-29983416

ABSTRACT

TRF2 is a telomere binding protein, a component of the shelterin complex that plays a major role in maintaining the integrity of the genome. TRF2 is over-expressed in a number of human cancers including Head and Neck cancer and might play a key role in tumor initiation and development. p38 MAPK signaling pathway is strongly activated in response to various environmental and cellular stresses and thus overexpressed in most of the Head and Neck cancer cases. In this study, we investigated potential interactions of TRF2 with p38 in HNSCC cells and patient samples. Using in silico experiments, we identified interface polar residue Asp-354 of p38 and Arg-492, Arg-496 of TRF2 as protein-protein interaction hotspots. In addition to these interactions, Arg-49 residue of p38 was also found to interact with Glu-456 of TRF2. A detailed understanding of how phosphorylated and unphosphorylated state of p38 protein can influence the stability, specificity and to some extent a conformational change of p38-TRF2 binding is presented. Silencing of TRF2 significantly decreased the phosphorylation of p38 in HNSCC cells which was confirmed by western blot, immunofluorescence and co-immunoprecipitation and alternatively inhibiting p38 using p38 inhibitor (SB 203580) decreased the expression of TRF2 in HNSCC cells. Furthermore, we checked the effect of TRF2 silencing and p38 inhibition in cisplatin induced chemosensitivity of SCC-131 cells. TRF2 silencing and p38 inhibition chemosensitize HNSCC cells to cisplatin. Thus, targeting TRF2 in combinatorial therapeutics can be a treatment modality for Head and Neck cancer which involves inhibition of p38 MAPK pathway.

9.
Stem Cell Rev Rep ; 14(6): 871-887, 2018 Dec.
Article in English | MEDLINE | ID: mdl-29872959

ABSTRACT

The major problem to effective treatment of oral cancer is the presence of therapy resistance. Presence of cancer stem cell in the bulk of tumor have been implicated in therapeutic resistance. In this study, we report a non-telomeric role of TRF2 in formation of oral cancer spheroids and CSC phenotype maintenance via an efficient DNA damage repair mechanism in the presence of chemotherapeutic insult. We report reduced sphere formation efficiency and reduced spheroid size in TRF2 silenced oral cancer cell lines. TRF2 silenced orospheres further reported reduced proliferative capacity as compared to non-silenced orospheres. Furthermore, TRF2 silencing hampered the migratory potential of oral cancer cell line and also reduced the expression of several CSC markers like CD44, Oct4, Sox2, KLF4 and c-Myc along with ß-catenin and hTERT molecules both in Cal27 cell line and generated orospheres. TRF2 silencing impaired efficient DNA damage repair capacity of non-orospheric and orospheric cells and repressed ERCC1 expression levels when treated with Cisplatin. TRF2 overexpression was also observed to correlate with poor overall survival and disease relapse of OSCC patients. In silico studies further identified several amino acid residues that show high binding affinity and strong protein-protein interactions among TRF2 and CSC marker KLF4. Hence, our report confirms a non-telomeric role of TRF2 in spheroid generation, maintenance of CSC phenotype and efficient DNA damage repair capacity contributing to chemotherapy resistance in oral cancer cell line. We further iterate the use of TRF2 as a prognostic marker in OSCC for faster detection and improved survival.


Subject(s)
Carcinoma, Squamous Cell/pathology , DNA Repair , Mouth Neoplasms/pathology , Neoplasm Recurrence, Local/pathology , Neoplastic Stem Cells/pathology , Spheroids, Cellular/metabolism , Telomere/metabolism , Telomeric Repeat Binding Protein 2/metabolism , Biomarkers, Tumor/metabolism , Carcinoma, Squamous Cell/metabolism , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Cisplatin/pharmacology , Computer Simulation , DNA Repair/drug effects , Down-Regulation/drug effects , Female , Gene Silencing/drug effects , Humans , Kruppel-Like Factor 4 , Male , Middle Aged , Mouth Neoplasms/metabolism , Neoplasm Recurrence, Local/metabolism , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/metabolism , Phenotype , Spheroids, Cellular/drug effects , Survival Analysis , Telomerase/metabolism
10.
J Oral Pathol Med ; 47(5): 492-501, 2018 May.
Article in English | MEDLINE | ID: mdl-29575240

ABSTRACT

AIM: This study aimed to investigate the role of p38 MAPK in maintenance of cancer stem cell (CSC) phenotype, therapy resistance, and DNA damage repair and response in head and neck squamous cell carcinoma (HNSCC). METHODS: In this study, 104 HNSCC patients were included. Western blot, immunohistochemistry, and qPCR analysis were performed to investigate the expression level of p-p38 and CSC markers in cut margin and tumor area of HNSCC patients. The expression level of p-p38 and CSC markers was also evaluated in HNSCC cells with or without p38 inhibitor. Chemoresistance, wound healing capacity, and multicellular tumor spheroids (MCTS) formation capacity were evaluated in HNSCC-derived cell lines with or without p38 inhibitor. In addition, DNA damage response and repair capacities were also evaluated in HNSCC cells after p38 inhibition using alkaline comet assay and γ-H2 AX immunostaining. RESULT: We observed that recurrence could be associated with upregulated status of p-p38 and p38α gene in cut margin area of HNSCC patients as compared to tumor region. p38-inhibited cells showed significantly reduced expression of CSC markers, chemosensitivity toward cisplatin, reduced migration potential, and sphere-forming ability along with increased apoptotic population after treatment with increasing concentration of cisplatin. p38-inhibited cells also exhibited significantly increased comet olive tail moment and accumulation of γ-H2 AX, demonstrating increased DNA damage. CONCLUSION: This study demonstrated that p38 MAPK activation may play a role in therapeutic resistance and disease relapse in HNSCC by maintenance of CSCs phenotype.


Subject(s)
Antineoplastic Agents/pharmacology , Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/pathology , Cisplatin/pharmacology , Gene Expression , Head and Neck Neoplasms/genetics , Head and Neck Neoplasms/pathology , Neoplastic Stem Cells/pathology , p38 Mitogen-Activated Protein Kinases/physiology , Apoptosis/drug effects , Cell Line, Tumor , DNA Damage/genetics , DNA Repair , Drug Resistance, Neoplasm , Histones/metabolism , Humans , Neoplasm Recurrence, Local , Phenotype , Squamous Cell Carcinoma of Head and Neck , p38 Mitogen-Activated Protein Kinases/metabolism
11.
Cell Oncol (Dordr) ; 41(2): 185-200, 2018 Apr.
Article in English | MEDLINE | ID: mdl-29243047

ABSTRACT

BACKGROUND: Head and neck squamous cell carcinoma (HNSCC) is one of the most common types of cancer in India with high incidence and rapid recurrence rates. Here, we aimed to investigate the role of ß-catenin, a developmental pathway gene, in HNSCC therapy resistance, DNA damage response, recurrence and prognosis. METHODS: In total 80 HNSCC samples were included. Western blot, immunohistochemistry and qRT-PCR analyses were performed to assess ß-catenin expression in the cut margin and tumor areas of each sample. Kaplan-Meier analyses were performed to correlate ß-catenin expression with the survival and prognosis of HNSCC patients. In addition, chemo-resistance, DNA damage response and DNA repair capacities were evaluated in HNSCC-derived cell lines through LiCl-mediated up-regulation and siRNA-mediated silencing of ß-catenin expression. RESULTS: We observed ß-catenin up-regulation in cut margin areas of recurrent patients compared to their corresponding tumor regions, which subsequently could be associated with poor prognosis. In addition, we found that LiCl-mediated up-regulation of ß-catenin in HNSCC-derived cells led to cisplatin resistance, evasion of apoptosis, enhanced DNA repair and enhanced migration. The effects of ß-catenin silencing correlated with its putative role in chemo-resistance and DNA damage response. CONCLUSION: From our results we conclude that ß-catenin may contribute to HNSCC therapy resistance and disease relapse. As such, ß-catenin may be explored as a therapeutic target along with conventional therapeutics.


Subject(s)
Cisplatin/pharmacology , Head and Neck Neoplasms/metabolism , beta Catenin/metabolism , Apoptosis/drug effects , Carcinoma, Squamous Cell/metabolism , Carcinoma, Squamous Cell/pathology , DNA Damage/drug effects , DNA Damage/genetics , HCT116 Cells , Head and Neck Neoplasms/pathology , Humans , Immunohistochemistry , Prognosis , Squamous Cell Carcinoma of Head and Neck
12.
Oral Oncol ; 73: 27-35, 2017 10.
Article in English | MEDLINE | ID: mdl-28939073

ABSTRACT

OBJECTIVE: CDKN2A/p16 is a known tumor suppressor gene with a homologous deletion in Oral Squamous cell carcinoma. CDKN2A/p16 is found to be inactivated in a broad spectrum of solid tumors and in more than 80% of OSCC. Molecular alteration of CDKN2A/p16 in progression of OSCC can pose an important tool for the prognosis of squamous cell carcinoma. MATERIAL AND METHOD: Systematic network analysis was carried out to obtain involvement of CDKN2A/p16 in oral cancer by polysearch and FunDO. In the present study we have screened 104 OSCC patients from eastern region of India for CDKN2A/p16 expression in recurrent and non-recurrent OSCC. The observation was validated by Comparative Genomic Hybridisation and Next generation sequencing in recurrent cases. RESULT: Systematic analysis revealed direct involvement of CDKN2A/p16 in oral cancer. There was a consistent downregulated expression of CDKN2A/p16 in the recurrent cases. The gene expression study confirmed a >5-fold downregulation of CDKN2A/p16 in recurrent tumors as compared to non-recurrent ones. Array CGH analysis revealed a copy number deletion in the recurrent case. Furthermore, next generation sequencing validated deletion of CDKN2A/p16 and reported it asa common variant with a nonsense mutation having stop /loss of function of the gene in recurrent cases. Recurrent cases with deleted CDKN2A/p16 expression had poor prognosis and low survival rate. CONCLUSION: CDKN2A/p16 frequently alters in oral cancer progression with a deletion/loss of function in the recurrent cases displaying its role in aiding several molecular events for the malignant transformations occurring throughout disease progression.


Subject(s)
Carcinoma, Squamous Cell/pathology , Genes, p16 , Mouth Neoplasms/pathology , Adult , Aged , Carcinoma, Squamous Cell/genetics , Comparative Genomic Hybridization , Disease Progression , Female , High-Throughput Nucleotide Sequencing , Humans , Male , Middle Aged , Mouth Neoplasms/genetics , Neoplasm Recurrence, Local , Prognosis
13.
J Stem Cells ; 9(4): 235-42, 2014.
Article in English | MEDLINE | ID: mdl-25942339

ABSTRACT

UNLABELLED: Wound healing is an inherent property of injured tissue or a group of cells. The healing front is always developed by new cells which are progenitor of differentiated parental cells. In cancer tissues we aim to study the healing front and observed an enriched population of stem cell like properties in the developing front when compared to the other areas of the cell matrix. METHOD: In vitro scratch assays with special focus on stem cell expression was used to analyze metastatic potential of the tumor cell, epithelial to mesenchymal transition and rate of cell migration to get an insight into the genes and the proteins getting expressed at the developing front. In this protocol we describe a fluorescence dependent method to document stem cell like enrichment at the developing front of a given wound in drug treated and untreated control cells under the same culture conditions in a time lag manner. We have tried to compare the rate of cell migration and the expression levels of stem cell markers between the treated and untreated cells. RESULTS: CD44 being a cell surface protein and being involved in cell migration and proliferation, higher intensity of CD44 was observed at the developing front with increasing time. The rate of cell migration differed with different treatments and so did the CD44 expression with expression being higher in 0.6mM concentration of bleomycin when compared to 0.4mM. Similar expression was observed for ALDH1 stem cell marker. This particular technique can not only be used for studying expression of CSC markers (like CD44, ALDH1) but also in assaying the expression profile of several proteins involved in cellular processes like EMT (Epithelial to Mesenchymal Transition), cell migration, tumorigeneisis and rate of proliferation. CONCLUSION: Would healing is an integral property of solid tissues and in solid tumors properties of solid tissue wound are important characteristics of tumor development. Therefore combining the properties of stem cell like enrichment in the development front would be an important and fast assay to study migratory and metastatic properties of an invitro culture.


Subject(s)
Cell Movement/genetics , Cell Proliferation/genetics , Epithelial-Mesenchymal Transition/genetics , Neoplastic Stem Cells/metabolism , Aldehyde Dehydrogenase 1 Family , Gene Expression Regulation, Neoplastic , HCT116 Cells , Humans , Hyaluronan Receptors/biosynthesis , Isoenzymes/biosynthesis , Neoplasm Metastasis , Neoplastic Stem Cells/pathology , Retinal Dehydrogenase/biosynthesis , Wound Healing/genetics
14.
J Stem Cells ; 9(4): 243-51, 2014.
Article in English | MEDLINE | ID: mdl-25942340

ABSTRACT

AIM: Clonospheres formed due to modified culture conditions are often studied for their stem cell like behaviour. The main objective of the current study is to compare the stem cell markers and link it to hTERT levels by monitoring their quantitative gene expression as they are potential targets for new generation combination therapeutics. METHOD: In the present study we created stable colonospheres of Human colon cancer cell line HCT-116 long term culture conditions of Serum deprivation. Clonospheres formed after 15 days were collected by gentle and enzymatic dissociation was performed. Single cell suspension was obtained by mechanically dissociating the cells through a 22G needle. Single cells were replanted at a density 1200 cells/ml in Serum Free Medium in the 6 well plates for further passage. Passaging of cells was done at an interval of 8 days. The spheres formed were cyto-spun in special slides for Immunocytochemistry (ICC) studies for ß-catenin protein and hTERT. The colonospheres were also processed for real time PCR expression studies for the same genes to confirm. RESULTS: In this present study, immunofluorescence studies revealed high ß-catenin expression in the nucleus in colonospheres as compared to that of differentiated cancer cell line HCT-116 where the signal was localized mostly in the membranous and non-nuclear regions. Also increased TRF2 signal in colonospheres indicated higher activity of hTERT gene as TRF2 is the direct activator of hTERT to protect the telomere. Quantitative PCR studies showed that there was a significant over expression (p<0.05) at the mRNA level of the hTERT, TRF2, Rap1 genes along with the ß-catenin over expression. Immunofluorescence analysis also revealed higher expression of CSC marker CD44 and ALDH1in colonospheres compared to the parental population. CONCLUSION: Clonospheres sub-population is showing higher degree of hTERT gene expression along with ß-catenin when compared to the parental HCT-116 cancer cells. We also checked the co expression of other telomere maintenance genes mainly TRF 2 and Rap1 which also showed similar results. Therefore, we conclude that not only hTERT but possibly other Sheltrin proteins are regulated by ß-catenin which is co expressed.


Subject(s)
Colonic Neoplasms/genetics , Neoplastic Stem Cells/metabolism , Telomerase/biosynthesis , beta Catenin/biosynthesis , Colonic Neoplasms/pathology , Gene Expression Regulation, Neoplastic , HCT116 Cells , Humans , Neoplastic Stem Cells/pathology
SELECTION OF CITATIONS
SEARCH DETAIL
...