Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 60
Filter
1.
Biochim Biophys Acta Rev Cancer ; 1877(1): 188668, 2022 01.
Article in English | MEDLINE | ID: mdl-34896436

ABSTRACT

Tamoxifen is a commonly used drug in the treatment of ER + ve breast cancers since 1970. However, development of resistance towards tamoxifen limits its remarkable clinical success. In this review, we have attempted to provide a brief overview of multiple mechanism that may lead to tamoxifen resistance, with a special emphasis on the roles played by the oncogenic kinase- PAK1. Analysing the genomic data sets available in the cBioPortal, we found that PAK1 gene amplification significantly affects the Relapse Free Survival of the ER + ve breast cancer patients. While PAK1 is known to promote tamoxifen resistance by phosphorylating ERα at Ser305, existing literature suggests that PAK1 can fuel up tamoxifen resistance obliquely by phosphorylating other substrates. We have summarised some of the approaches in the mass spectrometry based proteomics, which would enable us to study the tamoxifen resistance specific phosphoproteomic landscape of PAK1. We also propose that elucidating the multiple mechanisms by which PAK1 promotes tamoxifen resistance might help us discover druggable targets and biomarkers.


Subject(s)
Breast Neoplasms , Tamoxifen , Breast Neoplasms/drug therapy , Breast Neoplasms/genetics , Drug Resistance, Neoplasm/genetics , Female , Humans , Tamoxifen/pharmacology , Tamoxifen/therapeutic use , p21-Activated Kinases/genetics
2.
Oncogene ; 40(34): 5327-5341, 2021 08.
Article in English | MEDLINE | ID: mdl-34253860

ABSTRACT

P21 Activated Kinase 1 (PAK1) is an oncogenic serine/threonine kinase known to play a significant role in the regulation of cytoskeleton and cell morphology. Runt-related transcription factor 3 (RUNX3) was initially known for its tumor suppressor function, but recent studies have reported the oncogenic role of RUNX3 in various cancers. Previous findings from our laboratory provided evidence that Threonine 209 phosphorylation of RUNX3 acts as a molecular switch in dictating the tissue-specific dualistic functions of RUNX3 for the first time. Based on these proofs and to explore the translational significance of these findings, we designed a small peptide (RMR) from the protein sequence of RUNX3 flanking the Threonine 209 phosphorylation site. The selection of this specific peptide from multiple possible peptides was based on their binding energies, hydrogen bonding, docking efficiency with the active site of PAK1 and their ability to displace PAK1-RUNX3 interaction in our prediction models. We found that this peptide is stable both in in vitro and in vivo conditions, not toxic to normal cells and inhibits the Threonine 209 phosphorylation in RUNX3 by PAK1. We also tested the efficacy of this peptide to block the RUNX3 Threonine 209 phosphorylation mediated tumorigenic functions in in vitro cell culture models, patient-derived explant (PDE) models and in in vivo tumor xenograft models. These results proved that this peptide has the potential to be developed as an efficient therapeutic molecule for targeting RUNX3 Threonine 209 phosphorylation-dependent tumor phenotypes.


Subject(s)
p21-Activated Kinases , Carcinogenesis , Humans , Oncogenes , Phosphorylation , Protein Serine-Threonine Kinases , Threonine
3.
Gene ; 760: 144991, 2020 Nov 15.
Article in English | MEDLINE | ID: mdl-32717309

ABSTRACT

Breast cancer is the most frequently diagnosed cancer in women worldwide. Identifying reliable biomarkers and druggable molecular targets pose to be a significant quest in breast cancer research. p21-activated kinase 1 (PAK1) is a serine/threonine kinase that direct cell motility, cytoskeletal remodelling, and has been shown to function as a downstream regulator for various cancer signalling cascades that promote cell proliferation, apoptosis deregulation and hasten mitotic abnormalities, resulting in tumor formation and progression. The heterogeneity and acquired drug resistance are important factors that challenge the treatment of breast cancer. p21-activated kinase 1 signalling is crucial for activation of the Ras/RAF/MEK/ERK, PI3K/Akt/mTOR and Wnt signalling cascades which regulate cell survival, cell cycle progression, differentiation, and proliferation. A study involving proteogenomics analysis on breast cancer tissues showed the PAK1 as outlier kinase. In addition to this, few outlier molecules were identified specific to subtypes of breast cancer. A few substrates of PAK1 in breast cancer are already known. In this paper, we have discussed a similar approach called Kinase Interacting Substrate Screening (KISS) for the identification of novel oncogenic substrates of p21-activated kinase specific to subtypes of breast cancer. Such high throughput approaches are expected to accelerate the process of identifying novel drug targets and biomarkers.


Subject(s)
Breast Neoplasms/metabolism , p21-Activated Kinases/metabolism , Animals , Apoptosis/drug effects , Apoptosis/physiology , Breast Neoplasms/drug therapy , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Proliferation/physiology , Cell Survival/drug effects , Cell Survival/physiology , Female , Humans , Signal Transduction , p21-Activated Kinases/genetics
4.
Curr Top Med Chem ; 19(13): 1162-1172, 2019.
Article in English | MEDLINE | ID: mdl-31210110

ABSTRACT

BACKGROUND: Though virtual screening methods have proven to be potent in various instances, the technique is practically incomplete to quench the need of drug discovery process. Thus, the quest for novel designing approaches and chemotypes for improved efficacy of lead compounds has been intensified and logistic approaches such as scaffold hopping and hierarchical virtual screening methods were evolved. Till now, in all the previous attempts these two approaches were applied separately. OBJECTIVE: In the current work, we made a novel attempt in terms of blending scaffold hopping and hierarchical virtual screening. The prime objective is to assess the hybrid method for its efficacy in identifying active lead molecules for emerging PPI target Bcl-2 (B-cell Lymphoma 2). METHODS: We designed novel scaffolds from the reported cores and screened a set of 8270 compounds using both scaffold hopping and hierarchical virtual screening for Bcl-2 protein. Also, we enumerated the libraries using clustering, PAINS filtering, physicochemical characterization and SAR matching. RESULTS: We generated a focused library of compounds towards Bcl-2 interface, screened the 8270 compounds and identified top hits for seven families upon fine filtering with PAINS algorithm, features, SAR mapping, synthetic accessibility and similarity search. Our approach retrieved a set of 50 lead compounds. CONCLUSION: Finding rational approach meeting the needs of drug discovery process for PPI targets is the need of the hour which can be fulfilled by an extended scaffold hopping approach resulting in focused PPI targeting by providing novel leads with better potency.


Subject(s)
Proto-Oncogene Proteins c-bcl-2/antagonists & inhibitors , Small Molecule Libraries/pharmacology , Algorithms , Dose-Response Relationship, Drug , Drug Evaluation, Preclinical , Humans , Molecular Docking Simulation , Molecular Structure , Proto-Oncogene Proteins c-bcl-2/metabolism , Small Molecule Libraries/chemical synthesis , Small Molecule Libraries/chemistry , Structure-Activity Relationship
5.
Mol Cell Biol ; 39(7)2019 04 01.
Article in English | MEDLINE | ID: mdl-30670568

ABSTRACT

Breast cancer is a recurrent type of cancer among women worldwide. Despite remarkable progress in the prevention, detection, and treatment of breast cancer, it still remains a major chronic problem worldwide and poses significant challenges, like metastasis to distant organs, demanding the need for novel biomarkers and therapeutic targets. Focal adhesion kinase (FAK), a member of the protein tyrosine kinases, has been shown to be expressed in high levels in breast tumors. Of late, FAK has emerged as an impending curative target in breast carcinoma, with few of the small molecular inhibitors reaching the clinical trial stage. In the current study, we established that microRNA 551a (miR-551a) precisely regulates FAK by binding to the complementary sequences in the 3' untranslated region (UTR) of mRNAs of FAK and inhibits its expression in breast carcinoma cell lines. Further, results from human breast carcinoma samples illustrated that miR-551a levels were substantially downregulated in tumor samples, with a concurrent rise in the expression of FAK. Functional experimental studies using miR-551a-overexpressing breast cancer cells and nude mouse xenograft models revealed the tumor suppressor role of miR-551a. We also found that miR-551a expression decreased the invasion and migratory ability of breast carcinoma cells by inhibiting MMP-9 activity. Regulation studies performed utilizing promoter luciferase assays, chromatin immunoprecipitation (ChIP), and electrophoretic mobility shift assay (EMSA) revealed that c-Fos binds to the miR-551a promoter and activates it. Further, we observed a considerable increase in the amount of miR-551a levels upon c-Fos overexpression. All of these results showed that miR-551a can be of clinical relevance in understanding the regulation of FAK in breast tumorigenesis.


Subject(s)
Breast Neoplasms/metabolism , Focal Adhesion Kinase 1/metabolism , MicroRNAs/biosynthesis , Proto-Oncogene Proteins c-fos/metabolism , 3' Untranslated Regions , Animals , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Carcinogenesis/genetics , Carcinogenesis/metabolism , Cell Line, Tumor , Cell Movement/genetics , Cell Transformation, Neoplastic , Female , Focal Adhesion Kinase 1/antagonists & inhibitors , Focal Adhesion Kinase 1/genetics , Gene Expression Regulation, Neoplastic , Genes, fos , Heterografts , Humans , Mice , Mice, Nude , MicroRNAs/genetics , MicroRNAs/metabolism , Middle Aged , Proto-Oncogene Proteins c-fos/genetics , RNA, Messenger/genetics
6.
Small GTPases ; 10(4): 264-270, 2019 07.
Article in English | MEDLINE | ID: mdl-28628382

ABSTRACT

Post translational modifications of RUNX3 have been shown to play an important role in directing RUNX3 functions. In this review we highlight the phosphorylation dependent functions of RUNX3 as regulated by PAK1 and its implications on tumorigenesis.


Subject(s)
Core Binding Factor Alpha 3 Subunit/genetics , Core Binding Factor Alpha 3 Subunit/metabolism , Neoplasms/metabolism , p21-Activated Kinases/metabolism , Animals , Gene Expression Regulation, Neoplastic , Humans , Neoplasms/genetics , Phosphorylation , Protein Processing, Post-Translational
7.
J Cell Physiol ; 234(1): 969-975, 2018 01.
Article in English | MEDLINE | ID: mdl-30076705

ABSTRACT

Despite regular exposure of skin to solar UV-B irradiation, most individuals enjoy cancer-free existence, which is a testimony of the inherent capacity of human keratinocytes to either repair or restore cells damged by UV exposure. In this manuscript, we focus on delineating the mechanistic role of p21 activated kinase (Pak1) in UV-B provoked skin lesions. Molecular mechanistic studies revealed that Pak1 is triggered as a consequence to UV-B exposure via epidermal growth factor receptor (EGFR) and cyclobutane pyrimidine dimers (CPD) pathways, and both these membranous (EGFR) and nuclear (CPDs) events converge at Pak1 activation and contribute in a coordinated manner for yielding a complete response to UV-B via upregulating Ataxia-Telangiectasia and Rad3 related (ATR). This is the first study that evaluates the mechanistic role of a signaling molecule, Pak1, in premalignant skin lesions caused by sun exposure and designate that expression and instigation of Pak1 could operate as an alarming indicator of succession towards aggressive form of skin cancer, if neglected.


Subject(s)
Biomarkers, Tumor/genetics , Precancerous Conditions/genetics , Skin Neoplasms/genetics , p21-Activated Kinases/genetics , Ataxia Telangiectasia Mutated Proteins/genetics , DNA Damage/radiation effects , DNA Repair/radiation effects , ErbB Receptors/genetics , Humans , Keratinocytes/pathology , Keratinocytes/radiation effects , Precancerous Conditions/etiology , Precancerous Conditions/pathology , Pyrimidine Dimers/genetics , Signal Transduction/radiation effects , Skin/pathology , Skin/radiation effects , Skin Neoplasms/pathology , Ultraviolet Rays/adverse effects
8.
BMC Cancer ; 18(1): 589, 2018 May 24.
Article in English | MEDLINE | ID: mdl-29793439

ABSTRACT

BACKGROUND: This study was carried out to evaluate the prognostic value of KIBRA in breast cancer. METHODS: This retrospective study included breast cancer patients who sought the services of the immunohistochemistry laboratory of our unit from 2006 to 2015. Tissue microarrays were constructed and immunohistochemical staining was done to assess the KIBRA expression. The Kaplan-Meier model for univariate and Cox-regression model with backward stepwise factor retention method for multivariate analyses were used. Chi square test was used to find out the associations with the established prognostic features. RESULTS: A total of 1124 patients were included in the study and KIBRA staining of 909 breast cancers were available for analysis. Cytoplasmic KIBRA expression was seen in 39.5% and nuclear expression in 44.8%. Overall KIBRA-low breast cancers accounted for 41.5%. KIBRA nuclear expression was significantly associated with positive ER and PR expression. Luminal breast cancer patients who had endocrine therapy and KIBRA-low expression had a RFS disadvantage over those who were positive for KIBRA (p = 0.02). Similarly, patients who received chemotherapy and had overall KIBRA-low expression also demonstrated a RFS disadvantage compared to those who had overall positive KIBRA expression (p = 0.018). This effect of KIBRA was independent of the other factors considered for the model. CONCLUSION: Overall low-KIBRA expression has an independent effect on the RFS and predicts the RFS outcome of luminal breast cancer patients who received endocrine therapy and breast cancer patients who received chemotherapy.


Subject(s)
Biomarkers, Tumor/analysis , Breast Neoplasms/pathology , Intracellular Signaling Peptides and Proteins/analysis , Neoplasm Recurrence, Local/diagnosis , Phosphoproteins/analysis , Adult , Biomarkers, Tumor/metabolism , Breast/pathology , Breast Neoplasms/drug therapy , Breast Neoplasms/mortality , Cell Nucleus/metabolism , Chemotherapy, Adjuvant , Cross-Sectional Studies , Cytoplasm/metabolism , Disease-Free Survival , Female , Humans , Immunohistochemistry , Intracellular Signaling Peptides and Proteins/metabolism , Kaplan-Meier Estimate , Middle Aged , Phosphoproteins/metabolism , Prognosis , Receptors, Estrogen/metabolism , Receptors, Progesterone/metabolism , Retrospective Studies
9.
J Cell Physiol ; 233(3): 2613-2628, 2018 Mar.
Article in English | MEDLINE | ID: mdl-28782802

ABSTRACT

A series of 2, 3-dihydroquinazolinone derivatives were synthesized, characterized and their anticancer activity was determined. Among the compounds synthesized and screened, one compound (17) showed potent anticancer activity against human head and neck squamous cell carcinoma cell line, SCC131 and was non-toxic to normal cells. The compound inhibited the growth of SCC131 cells, with an IC50 of 1.75 µM, triggered apoptotic mode of cell death and caused tumor regression of SCC131 tumor xenografts in athymic mice. To decipher the target for the lead compound, a high throughput qPCR array was performed. Results showed that the compound 17, inhibited the expression of a vital transcription factor HNF4A, involved in regulation of metabolic pathways. Thus, the present work has identified a lead compound 17, with potent anticancer activity, minimal normal cell toxicity and a plausible target and hence definitely holds future prospects as an anticancer agent.


Subject(s)
Antineoplastic Agents/pharmacology , Carcinoma, Squamous Cell/drug therapy , Head and Neck Neoplasms/drug therapy , Hepatocyte Nuclear Factor 4/metabolism , Quinazolinones/pharmacology , Animals , Antineoplastic Agents/chemical synthesis , Apoptosis/drug effects , Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/metabolism , Carcinoma, Squamous Cell/pathology , Cell Cycle Checkpoints/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Dose-Response Relationship, Drug , Female , Head and Neck Neoplasms/genetics , Head and Neck Neoplasms/metabolism , Head and Neck Neoplasms/pathology , Hepatocyte Nuclear Factor 4/genetics , Humans , Inhibitory Concentration 50 , Mice, Nude , Quinazolinones/chemical synthesis , Squamous Cell Carcinoma of Head and Neck , Time Factors , Xenograft Model Antitumor Assays
10.
Gene ; 646: 120-128, 2018 Mar 10.
Article in English | MEDLINE | ID: mdl-29274909

ABSTRACT

P21-activated kinase 1 (Pak1) is known to be involved in a plethora of functions including cell growth, survival and can lead to cell transformation and tumor progression especially in breast tissue. Multiple studies have shown Pak1 dysregulation as a change in DNA copy number as well as gene expression levels, suggesting many regulatory mechanisms at transcriptional and translational level. However, very little is known about the transcriptional regulation of the human Pak1 promoter. Here, we focus on Pak1 promoter regulation by steroid hormones along with their respective receptors that are also crucial players in breast tissue function and tumorigenesis. Our results show high Pak1 expression in breast cancer cell lines and in breast tumor tissue. It also suggests that Pak1 is hormone responsive, whose expression can be modulated by steroid hormones namely, estrogen in the form of 17ß-estradiol (E2) and progesterone (P4). Sequence analysis of a 3.2kb Pak1 proximal promoter region shows the presence of PRE (progesterone response element) and ERE (estrogen response element) half sites, that were further cloned and characterized. Results from promoter analysis showed that Pak1 promoter activity is mediated by PR via its binding to PRE present on the Pak1 promoter that was further reaffirmed in vitro by electrophoretic mobility shift assay (EMSA) and chromatin immunoprecipitation assay (ChIP). Our results together suggest that it is the PR isoform B regulates Pak1 promoter. To our knowledge, this is the first study to report the detailed characterization and transcriptional regulation of the human Pak1 promoter by steroid hormones.


Subject(s)
Breast Neoplasms/genetics , Cloning, Molecular/methods , Estradiol/pharmacology , Progesterone/pharmacology , p21-Activated Kinases/chemistry , p21-Activated Kinases/genetics , Binding Sites , Cell Line, Tumor , Female , Gene Expression Regulation/drug effects , Humans , MCF-7 Cells , Promoter Regions, Genetic/drug effects , Transcription, Genetic , Up-Regulation , p21-Activated Kinases/metabolism
11.
RNA Biol ; 15(3): 320-326, 2018 03 04.
Article in English | MEDLINE | ID: mdl-28613101

ABSTRACT

Cancer of the head and neck are the most common cancers in India and account for 30% of all cancers. At molecular level, it could be attributed to the overexpression of growth factors like IGF1-R, EGFR, VEGF-R and deregulation of cell cycle regulators and tumor suppressors. IGF1-R is an emerging target in head and neck cancer treatment, because of its reported role in tumor development, progression and metastasis. IGF1R targeted agents are in advanced stages of clinical development. Nevertheless, these agents suffer from several disadvantages including acquired resistance and toxic side effects. Hence there is a need for developing newer agents targeting not only the receptor but also its downstream signaling. miRNAs are considered as master regulators of gene expression of multiple genes and has been widely reported to be a promising therapeutic strategy. This review discusses the present status of research in both these arenas and emphasizes the role of miRNA as a promising agent for biologic therapy.


Subject(s)
Antineoplastic Agents/pharmacology , MicroRNAs/genetics , Neoplasms/genetics , Receptors, Somatomedin/metabolism , Antineoplastic Agents/therapeutic use , Female , Gene Expression Regulation, Neoplastic/drug effects , Humans , Male , MicroRNAs/drug effects , Molecular Targeted Therapy , Receptor, IGF Type 1 , Receptors, Somatomedin/genetics , Signal Transduction/drug effects
12.
Br J Cancer ; 2017 Jun 29.
Article in English | MEDLINE | ID: mdl-28664913

ABSTRACT

BACKGROUND: KIBRA-initially identified as a neuronal associated protein is now shown to be functionally associated with other tissue types as well. KIBRA interacts with dyenin light chain 1 and this interaction is essential for oestrogen receptor transactivation in breast cancer cells. KIBRA as a substrate of Cdk1, Aurora kinase and ERK plays an important role in regulating cell cycle, cell proliferation and migration. Despite these evidences, the exact role of KIBRA in cancer progression is not known. METHODS: We studied the expression of KIBRA in breast tissues and breast cancer cell lines by western blotting, immunohistochemisry (IHC) and RT-PCR. Stable over expression and knockdown clones were generated to study the transforming properties of KIBRA by conventional assays. Xenograft studies were performed in nude mice to study the in vivo tumourigenic efficacy of KIBRA. qPCR array was performed to understand the molecular mechanism behind oncogenic activity of KIBRA. RESULTS: Our results showed that KIBRA is upregulated in breast cancer cells and in malignant human breast tumours by both western blotting and IHC. Interestingly, we found that KIBRA expression level goes up with increase in breast cancer progression in well-established MCF10A model system. Further, results from stable overexpression clones of KIBRA in fibroblasts (Rat-1) and epithelial breast cancer cells (ZR75) and lentiviral short hairpin RNA-mediated knockdown (KD) clones of KIBRA in ZR75 showed increase in transforming properties with KIBRA overexpression and vice-versa. Results also showed that fibroblasts stably overexpressing KIBRA showed increased tumourigenic potential in nude mice. By adopting a quantitative PCR array-based approach, we identified RASSF1A, a tumour suppressor, as a transcriptional target of KIBRA. CONCLUSIONS: This is the first study to demonstrate the in vivo tumourigenic property of KIBRA in a nude mouse model and also unravel the underlying molecular mechanism of KIBRA-mediated transformation via repression of RASSF1A.British Journal of Cancer advance online publication, 29 June 2017; doi:10.1038/bjc.2017.192 www.bjcancer.com.

13.
Cancer Invest ; 35(6): 431-442, 2017 Jul 03.
Article in English | MEDLINE | ID: mdl-28537455

ABSTRACT

Lung cancer is the leading cause of cancer deaths and the overall 5-year survival rate is less than 17%. Hyperthermia is an alternative approach for the treatment of lung cancer and is associated with fewer side effects. We employed ironoxide nanoparticles in inducing localized hyperthermia in lung cancer cells using a pulsed electromagnetic field (PEMF). We synthesized, characterized and determined the uptake of dipeptide-coated iron oxide nanoparticles. Further, their ability in inducing localized hyperthermia in PEMF on lung cancer cells was assessed. Results showed nanoparticles are non-cytotoxic and showed enhanced cellular uptake in lung cancer cells. In vivo studies in nude mice lung tumor xenografts confirmed the presence in the tumors. Lung cancer cells pretreated with dipeptide-coated magnetic nanoparticles upon PEMF exposure induced cell death.


Subject(s)
Electromagnetic Fields , Lung Neoplasms/genetics , Magnetite Nanoparticles/therapeutic use , Animals , Apoptosis , Cell Line, Tumor , Cell Proliferation , Disease Models, Animal , Humans , Male , Mice , Mice, Nude
15.
Curr Pharm Des ; 23(21): 3099-3104, 2017 08 30.
Article in English | MEDLINE | ID: mdl-28240171

ABSTRACT

Ovarian cancer, the worldwide leading cause of gynecological cancer-related death, is primarily treated by surgery followed by platinum chemotherapy. Though the tumor initially responds to the treatment, only 30% of 5 year survival period has been recorded and this is mainly attributed to the acquired chemo resistance and frequent recurrence of tumor. Combination chemotherapy as well, led to therapeutic failure due to non-specificity and subsequent side effects. However, polymer mediated drug delivery aids in overcoming these impediments. In particular, three dimensional macromolecule "Dendrimer" with its unique properties and numerous functionalities offer various advantages over the conventional approach and may improve the treatment outcome in patients with ovarian cancer. The present review highlights the various strategies employed using dendrimers to achieve targeted drug delivery and enhanced therapeutic efficacy in ovarian cancer.


Subject(s)
Dendrimers/therapeutic use , Ovarian Neoplasms/drug therapy , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Drug Resistance, Neoplasm , Female , Humans , Survival Analysis , Treatment Outcome
16.
Mol Cell Biol ; 37(6)2017 03 15.
Article in English | MEDLINE | ID: mdl-27956702

ABSTRACT

In this study, we have identified one microRNA, microRNA 493 (miR-493), which could simultaneously and directly regulate multiple genes downstream of the insulin-like growth factor 1 receptor (IGF1R) pathway, including IGF1R, by binding with complementary sequences in the 3' untranslated region (UTR) of mRNAs of IGF1R, insulin receptor substrate 1 (IRS1), and mitogen-activated protein kinase 1 (MAPK1), thereby potentiating their inhibitory function at multiple levels in development and progression of cancers. This binding was further confirmed by pulldown of miR with AGO-2 antibody. Further, results from head and neck samples showed that miR-493 levels were significantly downregulated in tumors, with a concomitant increase in the expression of IGF1R and key downstream effectors. Functional studies from miR-493 overexpression cells and nude-mouse models revealed the tumor suppressor functions of miR-493. Regulation studies revealed that Snail binds to the miR-493 promoter and represses it. We found the existence of a dynamic negative feedback loop in the regulation of IGF1R and miR-493 mediated via Snail. Our study showed that nicotine treatment significantly decreases the levels of miR-493-with a concomitant increase in the levels of Snail-an indication of progression of cells toward tumorigenesis, reestablishing the role of tobacco as a major risk factor for head and neck cancers and elucidating the mechanism behind nicotine-mediated tumorigenesis.


Subject(s)
Carcinogenesis/pathology , Feedback, Physiological , MicroRNAs/metabolism , Receptor, IGF Type 1/metabolism , Signal Transduction , Snail Family Transcription Factors/metabolism , Animals , Binding Sites , Carcinogenesis/drug effects , Carcinogenesis/genetics , Cell Line, Tumor , Cell Proliferation/drug effects , Down-Regulation/drug effects , Down-Regulation/genetics , Epithelial-Mesenchymal Transition/drug effects , Feedback, Physiological/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Genes, Tumor Suppressor , Head and Neck Neoplasms/genetics , Head and Neck Neoplasms/pathology , Humans , Kinetics , Mice, Nude , MicroRNAs/genetics , Models, Biological , Nicotine/pharmacology , Signal Transduction/drug effects , Snail Family Transcription Factors/genetics , Xenograft Model Antitumor Assays
18.
J Biol Chem ; 291(23): 12310-21, 2016 Jun 03.
Article in English | MEDLINE | ID: mdl-27044752

ABSTRACT

Parkinson disease (PD) is a neurodegenerative disorder with loss of dopaminergic neurons of the brain, which results in insufficient synthesis and action of dopamine. Metastasis-associated protein 1 (MTA1) is an upstream modulator of tyrosine hydroxylase (TH), the rate-limiting enzyme in dopamine synthesis, and hence MTA1 plays a significant role in PD pathogenesis. To impart functional and clinical significance to MTA1, we analyzed MTA1 and TH levels in the substantia nigra region of a large cohort of human brain tissue samples by Western blotting, quantitative PCR, and immunohistochemistry. Our results showed that MTA1 and TH levels were significantly down-regulated in PD samples as compared with normal brain tissue. Correspondingly, immunohistochemistry analysis for MTA1 in substantia nigra sections revealed that 74.1% of the samples had a staining intensity of <6 in the PD samples as compared with controls, 25.9%, with an odds ratio of 8.54. Because of the clinical importance of MTA1 established in PD, we looked at agents to modulate MTA1 expression in neuronal cells, and granulocyte colony-stimulating factor (G-CSF) was chosen, due to its clinically proven neurogenic effects. Treatment of the human neuronal cell line KELLY and acute 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine mouse model with G-CSF showed significant induction of MTA1 and TH with rescue of phenotype in the mouse model. Interestingly, the observed induction of TH was compromised on silencing of MTA1. The underlying molecular mechanism of MTA1 induction by G-CSF was proved to be through induction of c-Fos and its recruitment to the MTA1 promoter.


Subject(s)
Gene Expression/drug effects , Granulocyte Colony-Stimulating Factor/pharmacology , Histone Deacetylases/genetics , Neurons/drug effects , Repressor Proteins/genetics , 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine/pharmacology , Animals , Blotting, Western , Cell Line, Tumor , Dopamine/metabolism , Dopamine Agents/pharmacology , Histone Deacetylases/metabolism , Humans , Immunohistochemistry , Male , Mice, Inbred C57BL , Motor Activity/drug effects , Neurons/metabolism , Parkinson Disease/genetics , Parkinson Disease/metabolism , Parkinson Disease/physiopathology , Repressor Proteins/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Substantia Nigra/metabolism , Trans-Activators , Tyrosine 3-Monooxygenase/genetics , Tyrosine 3-Monooxygenase/metabolism
19.
Mol Cell Biol ; 36(9): 1354-65, 2016 May.
Article in English | MEDLINE | ID: mdl-26929199

ABSTRACT

Multifunctional adaptor proteins encompassing various protein-protein interaction domains play a central role in the DNA damage response pathway. In this report, we show that KIBRA is a physiologically interacting reversible substrate of ataxia telangiectasia mutated (ATM) kinase. We identified the site of phosphorylation in KIBRA as threonine 1006, which is embedded within the serine/threonine (S/T) Q consensus motif, by site-directed mutagenesis, and we further confirmed the same with a phospho-(S/T) Q motif-specific antibody. Results from DNA repair functional assays such as the γ-H2AX assay, pulsed-field gel electrophoresis (PFGE), Comet assay, terminal deoxynucleotidyltransferase-mediated dUTP-biotin nick end labeling (TUNEL) assay, and clonogenic cell survival assay using stable overexpression clones of wild-type (wt.) KIBRA and active (T1006E) and inactive (T1006A) KIBRA phosphorylation mutants showed that T1006 phosphorylation on KIBRA is essential for optimal DNA double-strand break repair in cancer cells. Further, results from stable retroviral short hairpin RNA-mediated knockdown (KD) clones of KIBRA and KIBRA knockout (KO) model cells generated by a clustered regularly interspaced short palindromic repeat (CRISPR)-Cas9 system showed that depleting KIBRA levels compromised the DNA repair functions in cancer cells upon inducing DNA damage. All these phenotypic events were reversed upon reconstitution of KIBRA into cells lacking KIBRA knock-in (KI) model cells. All these results point to the fact that phosphorylated KIBRA might be functioning as a scaffolding protein/adaptor protein facilitating the platform for further recruitment of other DNA damage response factors. In summary, these data demonstrate the imperative functional role of KIBRAper se(KIBRA phosphorylation at T1006 site as a molecular switch that regulates the DNA damage response, possibly via the nonhomologous end joining [NHEJ] pathway), suggesting that KIBRA could be a potential therapeutic target for modulating chemoresistance in cancer cells.


Subject(s)
Breast Neoplasms/genetics , DNA Repair/physiology , Intracellular Signaling Peptides and Proteins/metabolism , Phosphoproteins/metabolism , Ataxia Telangiectasia Mutated Proteins/genetics , Ataxia Telangiectasia Mutated Proteins/metabolism , Bleomycin/pharmacology , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Line, Tumor , DNA Breaks, Double-Stranded , DNA Damage/physiology , Drug Resistance, Neoplasm/drug effects , Drug Resistance, Neoplasm/genetics , Female , Gene Knockout Techniques , Humans , Intracellular Signaling Peptides and Proteins/genetics , Phosphoproteins/genetics , Phosphorylation
20.
Biochim Biophys Acta ; 1860(4): 636-47, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26723175

ABSTRACT

BACKGROUND: Further quest for new anti-fungal compounds with proven mechanisms of action arises due to resistance and dose limiting toxicity of existing agents. Among the human fungal pathogens C. albicans predominate by infecting several sites in the body and in particular oral cavity and root canals of human tooth. METHODS: In the present study, we screened a library of ß-lactam substituted polycyclic fused pyrrolidine/pyrrolizidine compounds against Candida sp. Detailed molecular studies were carried out with the active compound 3 on C. albicans. Morphological damage and antibiofilm activity of compound 3 on C. albicans was studied using scanning electron microscopy (SEM). Biochemical evidence for membrane damage was studied using flow cytometry. In silico docking studies were carried out to elucidate the mechanism of action of compound 3. Further, the antifungal activity of compound 3 was evaluated in an ex vivo dentinal tubule infection model. RESULTS: Screening data showed that several new compounds were active against Candida sp. Among them, Compound 3 was most potent and exerted time kill effect at 4h, post antifungal effect up to 6h. When used in combination with fluconazole or nystatin, compound 3 revealed an minimum inhibitory concentration (MIC) decrease by 4 fold for both drugs used. In-depth molecular studies with compound 3 on C. albicans showed that this compound inhibited yeast to hyphae (Y-H) conversion and this involved the cAMP pathway. Further, SEM images of C. albicans showed that compound 3 caused membrane damage and inhibited biofilm formation. Biochemical evidence for membrane damage was confirmed by increased propidium iodide (PI) uptake in flow cytometry. Further, in silico studies revealed that compound 3 docks with the active site of the key enzyme 14-α-demethylase and this might inhibit ergosterol synthesis. In support of this, ergosterol levels were found to be decreased by 32 fold in compound 3 treated samples as analyzed by high performance liquid chromatography (HPLC). Further, the antifungal activity of compound 3 was evaluated in an ex vivo dentinal tubule infection model, which mimics human tooth root canal infection. Confocal laser scanning microscopy studies showed 83% eradication of C. albicans and a 6 log reduction in colony forming unit (CFU) after 24h treatment in the infected tooth samples in this model. CONCLUSION: Compound 3 was found to be very effective in eradicating C. albicans by inhibiting cAMP pathway and ergosterol biosynthesis. GENERAL SIGNIFICANCE: The results of this study can pave the way for developing new antifungal agents with well deciphered mechanisms of action and can be a promising antifungal agent or medicament against root canal infection.


Subject(s)
14-alpha Demethylase Inhibitors/pharmacology , Antifungal Agents , Candida albicans/growth & development , Candidiasis/drug therapy , Cyclic AMP/metabolism , Dental Pulp Cavity/microbiology , Models, Biological , Second Messenger Systems , Sterol 14-Demethylase/metabolism , beta-Lactams , Antifungal Agents/chemistry , Antifungal Agents/pharmacology , Candida albicans/ultrastructure , Candidiasis/metabolism , Candidiasis/pathology , Dental Pulp Cavity/metabolism , Dental Pulp Cavity/ultrastructure , Humans , beta-Lactams/chemistry , beta-Lactams/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...