Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
Transl Oncol ; 15(1): 101257, 2022 Jan.
Article in English | MEDLINE | ID: mdl-34715620

ABSTRACT

The involvement of cancer stem cells (CSCs) in driving tumor dormancy and drug resistance is well established. Most therapeutic regimens however are ineffective in targeting these regenerative populations. We report the development and evaluation of a monoclonal antibody, mAb150, which targets the metastasis associated antigen, Annexin A2 (AnxA2) through recognition of a N-terminal epitope. Treatment with mAb150 potentiated re-entry of CSCs into the cell cycle that perturbed tumor dormancy and facilitated targeting of CSCs as was validated by in vitro and in vivo assays. Epigenetic potentiation further improved mAb150 efficacy in achieving total tumor regression by targeting regenerative populations to achieve tumor regression, specifically in high-grade serous ovarian adenocarcinoma.

2.
Cell Stress Chaperones ; 25(3): 481-494, 2020 05.
Article in English | MEDLINE | ID: mdl-32221864

ABSTRACT

CARF (Collaborator of ARF) was discovered as an ARF-interacting protein that activated ARF-p53-p21WAF1 signaling involved in cellular response to a variety of stresses, including oxidative, genotoxic, oncogenic, or telomere deprotection stresses, leading to senescence, growth arrest, or apoptosis. Of note, whereas suppression of CARF was lethal, its enrichment was associated with increased proliferation and malignant transformation of cells. These reports have predicted that CARF could serve as a multi-stress marker with a predictive value for cell fates. Here, we recruited various in vitro stress models and examined their effect on CARF expression using human normal fibroblasts. We demonstrate that CARF levels in stress and post-stress conditions could predict the fate of cells towards either death or enhanced proliferation and malignant transformation. We provide extensive molecular evidence that (i) CARF expression changes in response to stress, (ii) it modulates cell death or survival signaling and determines the fate of cells, and (iii) it may serve as a predictive measure of cellular response to stress and an important marker for biosafety.


Subject(s)
Apoptosis Regulatory Proteins/metabolism , Cell Transformation, Neoplastic , RNA-Binding Proteins/metabolism , Stress, Physiological , Animals , Biomarkers/metabolism , Cell Death , Cell Line , Cell Proliferation , Cell Survival , Humans , Mice , NIH 3T3 Cells
3.
Sci Rep ; 9(1): 17344, 2019 11 22.
Article in English | MEDLINE | ID: mdl-31757995

ABSTRACT

Withaferin-A is a withanolide, predominantly present in Ashwagandha (Withania somnifera). It has been shown to possess anticancer activity in a variety of human cancer cells in vitro and in vivo. Molecular mechanism of such cytotoxicity has not yet been completely understood. Withaferin-A and Withanone were earlier shown to activate p53 tumor suppressor and oxidative stress pathways in cancer cells. 2,3-dihydro-3ß-methoxy analogue of Withaferin-A (3ßmWi-A) was shown to lack cytotoxicity and well tolerated at higher concentrations. It, on the other hand, protected normal cells against oxidative, chemical and UV stresses through induction of anti-stress and pro-survival signaling. We, in the present study, investigated the effect of Wi-A and 3ßmWi-A on cell migration and metastasis signaling. Whereas Wi-A binds to vimentin and heterogeneous nuclear ribonucleoprotein K (hnRNP-K) with high efficacy and downregulates its effector proteins, MMPs and VEGF, involved in cancer cell metastasis, 3ßmWi-A was ineffective. Consistently, Wi-A, and not 3ßmWi-A, caused reduction in cytoskeleton proteins (Vimentin, N-Cadherin) and active protease (u-PA) that are essential for three key steps of cancer cell metastasis (EMT, increase in cell migration and invasion).


Subject(s)
Heterogeneous-Nuclear Ribonucleoprotein K/metabolism , Withanolides/pharmacology , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Computational Biology , Epithelial-Mesenchymal Transition/drug effects , Heterogeneous-Nuclear Ribonucleoprotein K/chemistry , Humans , MCF-7 Cells , Models, Molecular , Molecular Docking Simulation , Withanolides/chemical synthesis , Withanolides/chemistry
4.
Methods Mol Biol ; 2019: 1-14, 2019.
Article in English | MEDLINE | ID: mdl-31359385

ABSTRACT

Retinoid and rexinoid receptors are known to regulate key processes during development, differentiation, and cell death in vertebrates. However, their contributions to progression of malignant disease remain largely elusive although it is realized that transformed cancer cells, which essentially evade apoptosis, may display altered molecular expressions or functions associated with retinoid signaling. Here, using a progression model of ovarian cancer, we describe a proteomics-based approach including experimental procedures toward identification and validation of altered protein profiles during transformation. Effectively, this specifies loss of RXR-γ during progression of epithelial ovarian cancer.


Subject(s)
Carcinoma, Ovarian Epithelial/pathology , Ovarian Neoplasms/pathology , Proteomics/methods , Retinoid X Receptor gamma/deficiency , Animals , Carcinoma, Ovarian Epithelial/metabolism , Cell Line, Tumor , Disease Progression , Electrophoresis, Gel, Two-Dimensional , Female , Gene Expression Regulation, Neoplastic , Humans , Mice , Neoplasm Transplantation , Ovarian Neoplasms/metabolism , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization
5.
Oncogenesis ; 7(5): 39, 2018 May 11.
Article in English | MEDLINE | ID: mdl-29748568

ABSTRACT

CARF (Collaborator of ARF)/CDKN2AIP was discovered as a novel ARF-binding protein. It has been established as an essential cell survival, p53-, and cell proliferation-regulatory protein. Although a moderate upregulation of CARF caused growth arrest and senescence, its excessively enriched levels were shown to facilitate aggressive proliferation and malignant transformation of cancer cells. Here, we examined the relevance of CARF levels in clinical tumors and found its amplification (both at gene and transcript levels) in a variety of invasive and metastatic malignancies. Consistent with the clinical readouts, enrichment of CARF in cancer cells promoted epithelial-mesenchymal transition (EMT). Cancer database and molecular analyses revealed that it activates Wnt/ß-catenin signaling axis, as evident by enhanced nuclear localization and function of ß-catenin marked by increased level of SNAIL1, SNAIL2, ZEB1, and TWIST1 and its downstream gene targets. Of note, targeted knockdown of CARF led to decrease in nuclear ß-catenin and its key downstream effectors, involved in EMT progression. Consistent with this, CARF targeting in vivo either by naked siRNA or CARF shRNA harboring adeno-oncolytic virus caused suppression of tumor progression and lung metastasis. Taken together, we report clinical and therapeutic relevance of CARF in EMT and cancer invasiveness/metastasis, and propose it as a potent therapeutic target of aggressive cancers.

6.
Sci Rep ; 8(1): 375, 2018 01 10.
Article in English | MEDLINE | ID: mdl-29321561

ABSTRACT

microRNAs (miRs) have recently emerged as small non-coding regulators of gene expression. We performed a loss-of-function screening by recruiting retrovirus mediated arbitrary manipulation of genome coupled with escape of cells from 5-Aza-2'-deoxycytidine (5-Aza-dC)-induced senescence. miRNA pool from cells that emerged from 5-Aza-dC-induced senescence was subjected to miR-microarray analysis with respect to the untreated control. We identified miR-451 as one of the upregulated miRs and characterized its functional relevance to drug resistance, cell growth, tumor suppressor proteins p53 and pRb, and stress response. We report that miR-451 caused growth arrest in cells leading to their resistance to 5-Aza-dC-induced senescence. Decrease in cyclin D1, CDK4 and phosphorylated pRB supported the growth arrest in miR-451 transfected cells. We demonstrate that Collaborator of ARF (CARF) protein is a new target of miR-451 that intermediates its function in tumor suppressor and stress signaling.


Subject(s)
Azacitidine/analogs & derivatives , Gene Expression Profiling/methods , MicroRNAs/genetics , Neoplasms/genetics , Transcription Factors/genetics , Up-Regulation , A549 Cells , Animals , Azacitidine/pharmacology , Cell Cycle Checkpoints , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , DNA-Binding Proteins , Decitabine , Drug Resistance, Neoplasm , Gene Expression Regulation, Neoplastic/drug effects , Humans , MCF-7 Cells , Mice , Neoplasm Transplantation , Neoplasms/pathology , Oligonucleotide Array Sequence Analysis/methods , Signal Transduction
7.
J Nat Prod ; 80(10): 2756-2760, 2017 10 27.
Article in English | MEDLINE | ID: mdl-29043807

ABSTRACT

2,3-Dihydro-3ß-methoxy withaferin-A (3ßmWi-A) is a natural withanolide that is structurally close to withaferin-A (Wi-A), is cytotoxic to human cancer cells, and is a candidate anticancer natural compound. Using cell-based biochemical, molecular, and imaging assays, we report that Wi-A and 3ßmWi-A possess contrasting activities. Whereas Wi-A caused oxidative stress to normal cells, 3ßmWi-A was well tolerated at even 10-fold higher concentrations. Furthermore, it promoted survival and protected normal cells against oxidative, UV radiation, and chemical stresses. We provide molecular evidence that 3ßmWi-A induces antistress and pro-survival signaling through activation of the pAkt/MAPK pathway. We demonstrate that 3ßmWi-A (i) contrary to Wi-A is safe and possesses stress-relieving activity, (ii) when given subsequent to a variety of stress factors including Wi-A, protects normal cells against their toxicity, and (iii) is a vital compound that may guard normal cells against the toxicity associated with various targeted therapeutic regimes in clinical practice.


Subject(s)
Cytoprotection/drug effects , Withanolides/pharmacology , Action Potentials , Humans , Mitogen-Activated Protein Kinase Kinases/metabolism , Molecular Structure , Oxidative Stress , Signal Transduction , Withanolides/chemistry
8.
Cancer Res ; 76(9): 2754-2765, 2016 05.
Article in English | MEDLINE | ID: mdl-26960973

ABSTRACT

Mortalin/mthsp70 (HSPA9) is a stress chaperone enriched in many cancers that has been implicated in carcinogenesis by promoting cell proliferation and survival. In the present study, we examined the clinical relevance of mortalin upregulation in carcinogenesis. Consistent with high mortalin expression in various human tumors and cell lines, we found that mortalin overexpression increased the migration and invasiveness of breast cancer cells. Expression analyses revealed that proteins involved in focal adhesion, PI3K-Akt and JAK-STAT signaling, all known to play key roles in cell migration and epithelial-to-mesenchymal transition (EMT), were upregulated in mortalin-expressing cancer cells. We further determined that expression levels of the mesenchymal markers vimentin (VIM), fibronectin (FN1), ß-catenin (CTNNB1), CK14 (KRT14) and hnRNP-K were also increased upon mortalin overexpression, whereas the epithelial markers E-cadherin (CDH1), CK8 (KRT8), and CK18 (KRT18) were downregulated. Furthermore, shRNA-mediated and pharmacological inhibition of mortalin suppressed the migration and invasive capacity of cancer cells and was associated with a diminished EMT gene signature. Taken together, these findings support a role for mortalin in the induction of EMT, prompting further investigation of its therapeutic value in metastatic disease models.

9.
Mol Oncol ; 9(9): 1877-89, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26278998

ABSTRACT

Collaborator of ARF (CARF), initially identified as a binding partner of ARF (Alternate Reading Frame), has been shown to activate ARF-p53 pathway by multiple ways including stabilization of ARF and p53 tumor suppressor proteins, and transcriptional repression of a p53 antagonist, HDM2. Level of CARF expression was shown to determine fate of cells. Whereas its knockdown caused apoptosis, its over- and super-expressions caused senescence and increase in malignant properties of cancer cells, respectively, and were closely linked to increase and decrease in p53 activity. Using p53-compromised cancer cells, we demonstrate that CARF induces growth arrest when wild type p53 is present and in p53-absence, it promotes carcinogenesis. Biochemical analyses on CARF-induced molecular signaling revealed that in p53-null cells, it caused transcriptional repression of p21(WAF1) leading to increase in CDK4, CDK6, pRb and E2F1 resulting in continued cell cycle progression. Furthermore, it instigated increase in migration and invasion of cancer cells that was marked by upregulation of MMP2, MMP3, MMP9, uPA, several interleukins and VEGF expression. Consistent with these findings, we found that human clinical samples of epithelial and glial cancers (frequently marked by loss of p53 function) possessed high level of CARF expression showing a relationship with cancer aggressiveness. The data demonstrated that CARF could be considered as a diagnostic marker and a therapeutic target in p53-compromised malignancies.


Subject(s)
Apoptosis Regulatory Proteins/genetics , Carcinogenesis/genetics , Neoplasms/genetics , RNA-Binding Proteins/genetics , Tumor Suppressor Protein p53/genetics , Animals , Carcinogenesis/pathology , Cell Cycle , Cell Line, Tumor , Gene Deletion , Gene Expression Regulation, Neoplastic , Humans , Mice, Nude , Neoplasms/pathology , Up-Regulation
10.
J Biol Chem ; 289(26): 18258-69, 2014 Jun 27.
Article in English | MEDLINE | ID: mdl-24825908

ABSTRACT

Collaborator of ARF (CARF) has been shown to directly bind to and regulate p53, a central protein that controls tumor suppression via cellular senescence and apoptosis. However, the cellular functions of CARF and the mechanisms governing its effect on senescence, apoptosis, or proliferation are still unknown. Our previous studies have shown that (i) CARF is up-regulated during replicative and stress-induced senescence, and its exogenous overexpression caused senescence-like growth arrest of cells, and (ii) suppression of CARF induces aneuploidy, DNA damage, and mitotic catastrophe, resulting in apoptosis via the ATR/CHK1 pathway. In the present study, we dissected the cellular role of CARF by investigating the molecular pathways triggered by its overexpression in vitro and in vivo. We found that the dosage of CARF is a critical factor in determining the proliferation potential of cancer cells. Most surprisingly, although a moderate level of CARF overexpression induced senescence, a very high level of CARF resulted in increased cell proliferation. We demonstrate that the level of CARF is crucial for DNA damage and checkpoint response of cells through ATM/CHK1/CHK2, p53, and ERK pathways that in turn determine the proliferative fate of cancer cells toward growth arrest or proproliferative and malignant phenotypes. To the best of our knowledge, this is the first report that demonstrates the capability of a fundamental protein, CARF, in controlling cell proliferation in two opposite directions and hence may play a key role in tumor biology and cancer therapeutics.


Subject(s)
Apoptosis Regulatory Proteins/metabolism , Cell Proliferation , DNA Damage , Neoplasms/metabolism , Neoplasms/physiopathology , RNA-Binding Proteins/metabolism , Apoptosis , Apoptosis Regulatory Proteins/genetics , Ataxia Telangiectasia Mutated Proteins/genetics , Ataxia Telangiectasia Mutated Proteins/metabolism , Cell Line, Tumor , Humans , Neoplasms/genetics , RNA-Binding Proteins/genetics , Signal Transduction , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism
11.
Exp Cell Res ; 322(2): 324-34, 2014 Apr 01.
Article in English | MEDLINE | ID: mdl-24485912

ABSTRACT

CARF is an ARF-binding protein that has been shown to regulate the p53-p21-HDM2 pathway. CARF overexpression was shown to cause growth arrest of human cancer cells and premature senescence of normal cells through activation of the p53 pathway. Because replicative senescence involves permanent withdrawal from the cell cycle in response to DNA damage response-mediated signaling, in the present study we investigated the relationship between CARF and the cell cycle and whether it is involved in the DNA damage response. We demonstrate that the half-life of CARF protein is less than 60 min, and that in cycling cells CARF levels are highest in G2 and early prophase. Serially passaged normal human skin and stromal fibroblasts showed upregulation of CARF during replicative senescence. Induction of G1 growth arrest and senescence by a variety of drugs was associated with increase in CARF expression at the transcriptional and translational level and was seen to correlate with increase in DNA damage response and checkpoint proteins, ATM, ATR, CHK1, CHK2, γH2AX, p53 and p21. Induction of growth arrest by oncogenic RAS and shRNA-mediated knockdown of TRF2 in cancer cells also caused upregulation of CARF. We conclude that CARF is associated with DNA damage response and checkpoint signaling pathways.


Subject(s)
Apoptosis Regulatory Proteins/metabolism , Cell Cycle Checkpoints/physiology , DNA Damage/physiology , Fibroblasts/metabolism , RNA-Binding Proteins/metabolism , Skin/metabolism , Stromal Cells/metabolism , Telomeric Repeat Binding Protein 2/metabolism , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Apoptosis Regulatory Proteins/genetics , Blotting, Western , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Cell Proliferation/drug effects , Cells, Cultured , Cellular Senescence/physiology , Fibroblasts/cytology , Fibroblasts/drug effects , Fluorescent Antibody Technique , Humans , Neoplasms/drug therapy , Neoplasms/metabolism , Neoplasms/pathology , Promoter Regions, Genetic/genetics , RNA, Messenger/genetics , RNA, Small Interfering/genetics , RNA-Binding Proteins/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Skin/cytology , Skin/drug effects , Stromal Cells/cytology , Stromal Cells/drug effects , Telomeric Repeat Binding Protein 2/antagonists & inhibitors , Telomeric Repeat Binding Protein 2/genetics
12.
PLoS One ; 8(8): e70398, 2013.
Article in English | MEDLINE | ID: mdl-23936423

ABSTRACT

The process of cellular transformation involves cascades of molecular changes that are modulated through altered epigenetic, transcription, post-translational and protein regulatory networks. Thus, identification of transformation-associated protein alterations can provide an insight into major regulatory pathways activated during disease progression. In the present protein expression profiling approach, we identified differential sets of proteins in a two-dimensional gel electrophoresis screen of a serous ovarian adenocarcinoma progression model. Function-based categorization of the proteins exclusively associated with pre-transformed cells identified four cellular processes of which RXR-γ is known to modulate cellular differentiation and apoptosis. We thus probed the functional relevance of RXR-γ expression and signaling in these two pathways during tumor progression. RXR-γ expression was observed to modulate cellular differentiation and apoptosis in steady-state pre-transformed cells. Interestingly, retinoid treatment was found to enhance RXR-γ expression in transformed cells and sensitize them towards apoptosis in vitro, and also reduce growth of xenografts derived from transformed cells. Our findings emphasize that loss of RXR-γ levels appears to provide mechanistic benefits to transformed cells towards the acquisition of resistance to apoptosis hallmark of cancer, while effective retinoid treatment may present a viable approach towards sensitization of tumor cells to apoptosis through induction of RXR-γ expression.


Subject(s)
Disease Progression , Gene Expression Profiling , Neoplasms, Glandular and Epithelial/metabolism , Ovarian Neoplasms/metabolism , Proteomics , Retinoid X Receptor gamma/metabolism , Animals , Apoptosis/drug effects , Carcinoma, Ovarian Epithelial , Cell Differentiation/drug effects , Cell Line, Tumor , Cell Transformation, Neoplastic/drug effects , Gene Ontology , Male , Mice , Neoplasms, Glandular and Epithelial/genetics , Neoplasms, Glandular and Epithelial/pathology , Ovarian Neoplasms/genetics , Ovarian Neoplasms/pathology , Retinoids/pharmacology , Signal Transduction/drug effects
13.
Cancer Res ; 70(12): 4809-19, 2010 Jun 15.
Article in English | MEDLINE | ID: mdl-20530682

ABSTRACT

Multiple, dissimilar genetic defects in cancers of the same origin contribute to heterogeneity in tumor phenotypes and therapeutic responses of patients, yet the associated molecular mechanisms remain elusive. Here, we show at the systems level that serous ovarian carcinoma is marked by the activation of interconnected modules associated with a specific gene set that was derived from three independent tumor-specific gene expression data sets. Network prediction algorithms combined with preestablished protein interaction networks and known functionalities affirmed the importance of genes associated with ovarian cancer as predictive biomarkers, besides "discovering" novel ones purely on the basis of interconnectivity, whose precise involvement remains to be investigated. Copy number alterations and aberrant epigenetic regulation were identified and validated as significant influences on gene expression. More importantly, three functional modules centering on c-Myc activation, altered retinoblastoma signaling, and p53/cell cycle/DNA damage repair pathways have been identified for their involvement in transformation-associated events. Further studies will assign significance to and aid the design of a panel of specific markers predictive of individual- and tumor-specific pathways. In the parlance of this emerging field, such networks of gene-hub interactions may define personalized therapeutic decisions.


Subject(s)
Biomarkers, Tumor/genetics , Cell Transformation, Neoplastic/genetics , Cystadenocarcinoma, Serous/genetics , Gene Regulatory Networks , Ovarian Neoplasms/genetics , Protein Interaction Mapping , Biomarkers, Tumor/metabolism , Cells, Cultured , Chromatin Immunoprecipitation , Cystadenocarcinoma, Serous/metabolism , Cystadenocarcinoma, Serous/pathology , Epigenesis, Genetic , Female , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Humans , Immunoblotting , Immunoprecipitation , In Situ Hybridization, Fluorescence , Oligonucleotide Array Sequence Analysis , Ovarian Neoplasms/metabolism , Ovarian Neoplasms/pathology , Prognosis , Signal Transduction
SELECTION OF CITATIONS
SEARCH DETAIL
...