Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 42
Filter
1.
IBRO Neurosci Rep ; 15: 218-224, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37822516

ABSTRACT

Rett Syndrome (RTT) is a neurodevelopmental disorder caused by pathogenic variants in the MECP2 gene. While the majority of RTT-causing variants are clustered in the methyl-CpG binding domain and NCoR/SMRT interaction domain, we report a female patient with a functionally uncharacterized MECP2 variant in the C-terminal domain, c.1030C>T (R344W). We functionally characterized MECP2-R344W in terms of protein stability, NCoR/SMRT complex interaction, and protein nuclear localization in vitro. MECP2-R344W cells showed an increased protein degradation rate without significant change in NCoR/SMRT complex interaction and nuclear localization pattern, suggesting that enhanced MECP2 degradation is sufficient to cause a Rett Syndrome-like phenotype. This study highlights the pathogenicity of the C-terminal domain in Rett Syndrome, and demonstrates the potential of targeting MECP2 protein stability as a therapeutic approach.

3.
J Biol Chem ; 299(4): 103023, 2023 04.
Article in English | MEDLINE | ID: mdl-36805338

ABSTRACT

Raf kinase inhibitor protein (RKIP) is a multifunctional modulator of intracellular signal transduction. Although most of its functions have been considered cytosolic, we show here that the localization of RKIP is primarily nuclear in both growing and quiescent Madin-Darby canine kidney epithelial cells and in Cal-51 and BT-20 human breast cancer cells. We have identified a putative bipartite nuclear localization signal (NLS) in RKIP that maps to the surface of the protein surrounding a known regulatory region. Like classical NLS sequences, the putative NLS of RKIP is rich in arginine and lysine residues. Deletion of and point mutations in the putative NLS lead to decreased nuclear localization. Point mutation of all the basic residues in the putative NLS of RKIP particularly strongly reduces nuclear localization. We found consistent results in reexpression experiments with wildtype or mutant RKIP in RKIP-silenced cells. A fusion construct of the putative NLS of RKIP alone to a heterologous reporter protein leads to nuclear localization of the fusion protein, demonstrating that this sequence alone is sufficient for import into the nucleus. We found that RKIP interacts with the nuclear transport factor importin α in BT-20 and MDA-MB-231 human breast cancer cells, suggesting importin-mediated active nuclear translocation. Evaluating the biological function of nuclear localization of RKIP, we found that the presence of the putative NLS is important for the role of RKIP in mitotic checkpoint regulation in MCF-7 human breast cancer cells. Taken together, these findings suggest that a bipartite NLS in RKIP interacts with importin α for active transport of RKIP into the nucleus and that this process may be involved in the regulation of mitotic progression.


Subject(s)
Nuclear Localization Signals , Phosphatidylethanolamine Binding Protein , alpha Karyopherins , Animals , Dogs , Humans , Active Transport, Cell Nucleus , alpha Karyopherins/genetics , alpha Karyopherins/metabolism , Cell Nucleus/metabolism , Nuclear Localization Signals/genetics , Nuclear Localization Signals/metabolism , Phosphatidylethanolamine Binding Protein/genetics , Phosphatidylethanolamine Binding Protein/metabolism , Madin Darby Canine Kidney Cells
4.
Cancers (Basel) ; 15(3)2023 Feb 02.
Article in English | MEDLINE | ID: mdl-36765912

ABSTRACT

Tumor microenvironment (TME) is the immediate environment where cancer cells reside in a tumor. It is composed of multiple cell types and extracellular matrix. Microenvironments can be restrictive or conducive to the progression of cancer cells. Initially, microenvironments are suppressive in nature. Stepwise accumulation of mutations in oncogenes and tumor suppressor genes enables cancer cells to acquire the ability to reshape the microenvironment to advance their growth and metastasis. Among the many genetic events, the loss-of-function mutations in tumor suppressor genes play a pivotal role. In this review, we will discuss the changes in TME and the ramifications on metastasis upon altered expression of tumor metastasis suppressor gene RKIP in breast cancer cells.

5.
Cancers (Basel) ; 14(15)2022 Jul 24.
Article in English | MEDLINE | ID: mdl-35892864

ABSTRACT

Raf-1 kinase inhibitor protein was first identified as a negative regulator of the Raf signaling pathway. Subsequently, it was shown to have a causal role in containing cancer progression and metastasis. Early studies suggested that RKIP blocks cancer progression by inhibiting the Raf-1 pathway. However, it is not clear if the RKIP tumor and metastasis suppression function involve other targets. In addition to the Raf signaling pathway, RKIP has been found to modulate several other signaling pathways, affecting diverse biological functions including immune response. Recent advances in medicine have identified both positive and negative roles of immune response in cancer initiation, progression and metastasis. It is possible that one way that RKIP exerts its effect on cancer is by targeting an immune response mechanism. Here, we provide evidence supporting the causal role of tumor and metastasis suppressor RKIP in downregulating signaling pathways involved with immune response in breast cancer cells and discuss its potential ramification on cancer therapy.

6.
Sci Rep ; 11(1): 17455, 2021 08 31.
Article in English | MEDLINE | ID: mdl-34465801

ABSTRACT

Raf-1 kinase inhibitor protein was initially discovered as a physiological kinase inhibitor of the MAPK signaling pathway and was later shown to suppress cancer cell invasion and metastasis. Yet, the molecular mechanism through which RKIP executes its effects is not completely defined. RhoA has both a pro- and anti-metastatic cell-context dependent functions. Given that Rho GTPases primarily function on actin cytoskeleton dynamics and cell movement regulation, it is possible that one way RKIP hinders cancer cell invasion/metastasis is by targeting these proteins. Here we show that RKIP inhibits cancer cell invasion and metastasis by stimulating RhoA anti-tumorigenic functions. Mechanistically, RKIP activates RhoA in an Erk2 and GEF-H1 dependent manner to enhance E-cadherin membrane localization and inhibit CCL5 expression.


Subject(s)
Biomarkers, Tumor/metabolism , Breast Neoplasms/prevention & control , Gene Expression Regulation, Neoplastic , Phosphatidylethanolamine Binding Protein/metabolism , rhoA GTP-Binding Protein/metabolism , Animals , Apoptosis , Biomarkers, Tumor/genetics , Breast Neoplasms/metabolism , Breast Neoplasms/secondary , Cell Proliferation , Female , Humans , Mice , Phosphatidylethanolamine Binding Protein/genetics , Tumor Cells, Cultured , Xenograft Model Antitumor Assays , rhoA GTP-Binding Protein/genetics
7.
Front Psychol ; 12: 659483, 2021.
Article in English | MEDLINE | ID: mdl-34248753

ABSTRACT

Policymakers are interested in programs that increase targeted pro-environmental behavior (PEB) and spill over to increase non-targeted PEBs. Theoretically, guilt should lead to negative spillover and identity to positive spillover, though this has rarely been tested empirically. Additionally, little is known about how reminders of past PEB behavior might also lead to downstream spillover effects. Across two studies, participants (Study 1: 377 MTurk workers; Study 2: 172 undergraduates) were randomly assigned to write about a prior PEB, an anti-environmental behavior, or to a control condition. Subsequently, respondents were given an opportunity to perform a PEB2 and completed measures of PEB3 intentions. Results showed some evidence of positive (through increasing identity) and negative (through decreasing guilt) indirect spillover pathways from prior PEB reminders to PEB2 performance and PEB3 curtailment intentions (but not efficiency upgrade intentions). However, there were no overall spillover effects from PEB reminders to PEB2 performance or PEB3 intentions, as the positive and negative indirect effects canceled each other out. Results also showed positive spillover from PEB2 performance to PEB3 curtailment intentions through increasing environmental guilt. The strength of the spillover effects depended on the comparison group for the experimental manipulation, whether environmental guilt or global guilt was measured, and the type of PEB. The results suggest that environmental communications that remind people of their prior PEB may not meaningfully spill over to further PEB performance or intentions.

8.
Sci Rep ; 9(1): 16351, 2019 11 08.
Article in English | MEDLINE | ID: mdl-31705019

ABSTRACT

The role of RhoA GTPases in breast cancer tumorigenesis and metastasis is unclear. Early studies within which mutations in RhoA were designed based on cancer-associated mutations in Ras supported an oncogene role for RhoA. However, recent whole-genome sequencing studies of cancers raised the possibility that RhoA may have a tumor suppression function. Here, using a syngeneic triple negative breast cancer murine model we investigated the physiological effects of reduced RhoA expression on breast cancer tumorigenesis and metastasis. RhoA knockdown had no effect on primary tumor formation and tumor proliferation, concurring with our in vitro findings where reduced RhoA had no effect on breast cancer cell proliferation and clonogenic growth. In contrast, primary tumors with RhoA knockdown efficiently invaded sentinel lymph nodes and significantly metastasized to lungs compared to control tumors. Mechanistically, the current study demonstrated that this is achieved by promoting a pro-tumor microenvironment, with increased cancer-associated fibroblasts and macrophage infiltration, and by modulating the CCL5-CCR5 and CXCL12-CXCR4 chemokine axes in the primary tumor. To our knowledge, this is the first such mechanistic study in breast cancer showing the ability of RhoA to suppress chemokine receptor expression in breast tumor cells. Our work suggests a physiological lung and lymph node metastasis suppressor role for RhoA GTPase in breast cancer.


Subject(s)
Biomarkers, Tumor/metabolism , Breast Neoplasms/pathology , Gene Expression Regulation, Neoplastic , Lung Neoplasms/secondary , Receptors, CCR5/metabolism , Receptors, CXCR4/metabolism , rhoA GTP-Binding Protein/metabolism , Animals , Apoptosis , Biomarkers, Tumor/genetics , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Cell Proliferation , Female , Humans , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Mice , Receptors, CCR5/genetics , Receptors, CXCR4/genetics , Signal Transduction , Tumor Cells, Cultured , Xenograft Model Antitumor Assays , rhoA GTP-Binding Protein/genetics
9.
Sci Rep ; 9(1): 6688, 2019 04 30.
Article in English | MEDLINE | ID: mdl-31040372

ABSTRACT

One third of newly diagnosed breast cancers in the US are early-stage lesions. The etiological understanding and treatment of these lesions have become major clinical challenges. Because breast cancer risk factors are often linked to aberrant nitric oxide (NO) production, we hypothesized that abnormal NO levels might contribute to the formation of early-stage breast lesions. We recently reported that the basal level of NO in the normal breast epithelia plays crucial roles in tissue homeostasis, whereas its reduction contributes to the malignant phenotype of cancer cells. Here, we show that the basal level of NO in breast cells plummets during cancer progression due to reduction of the NO synthase cofactor, BH4, under oxidative stress. Importantly, pharmacological deprivation of NO in prepubertal to pubertal animals stiffens the extracellular matrix and induces precancerous lesions in the mammary tissues. These lesions overexpress a fibrogenic cytokine, TGFß, and an oncogene, ERBB2, accompanied by the occurrence of senescence and stem cell-like phenotype. Consistently, normalization of NO levels in precancerous and cancerous breast cells downmodulates TGFß and ERBB2 and ameliorates their proliferative phenotype. This study sheds new light on the etiological basis of precancerous breast lesions and their potential prevention by manipulating the basal NO level.


Subject(s)
Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Nitric Oxide/biosynthesis , Precancerous Conditions/genetics , Precancerous Conditions/metabolism , Receptor, ErbB-2/genetics , Transforming Growth Factor beta/genetics , Animals , Biomarkers , Breast/metabolism , Breast/pathology , Breast Neoplasms/diagnostic imaging , Breast Neoplasms/pathology , Disease Susceptibility , Epithelial Cells/metabolism , Epithelial Cells/pathology , Female , Gene Expression , Humans , Mice , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Nitric Oxide Synthase Type I/metabolism , Nitric Oxide Synthase Type II/metabolism , Nitric Oxide Synthase Type III/metabolism , Oxidative Stress , Precancerous Conditions/pathology , Receptor, ErbB-2/metabolism , Transforming Growth Factor beta/metabolism
10.
PLoS One ; 14(4): e0204387, 2019.
Article in English | MEDLINE | ID: mdl-30995246

ABSTRACT

Recent high-throughput-sequencing of cancer genomes has identified oncogenic mutations in the B-Raf genetic locus as one of the critical events in melanomagenesis. B-Raf encodes a serine/threonine kinase that regulates the MAPK/ERK kinase (MEK) and extracellular signal-regulated kinase (ERK) protein kinase cascade. In normal cells, the activity of B-Raf is tightly regulated and is required for cell growth and survival. B-Raf gain-of-function mutations in melanoma frequently lead to unrestrained growth, enhanced cell invasion and increased viability of cancer cells. Although it is clear that the invasive phenotypes of B-Raf mutated melanoma cells are stringently dependent on B-Raf-MEK-ERK activation, the downstream effector targets that are required for oncogenic B-Raf-mediated melanomagenesis are not well defined. miRNAs have regulatory functions towards the expression of genes that are important in carcinogenesis. We observed that miR-10b expression correlates with the presence of the oncogenic B-Raf (B-RafV600E) mutation in melanoma cells. While expression of miR-10b enhances anchorage-independent growth of B-Raf wild-type melanoma cells, miR-10b silencing decreases B-RafV600E cancer cell invasion in vitro. Importantly, the expression of miR-10b is required for B-RafV600E-mediated anchorage independent growth and invasion of melanoma cells in vitro. Taken together our results suggest that miR-10b is an important mediator of oncogenic B-RafV600E activity in melanoma.


Subject(s)
Gain of Function Mutation , Gene Expression Regulation, Neoplastic , Melanoma/metabolism , MicroRNAs/biosynthesis , Proto-Oncogene Proteins B-raf/metabolism , RNA, Neoplasm/biosynthesis , Amino Acid Substitution , Cell Line, Tumor , Cell Survival , Humans , MAP Kinase Signaling System , Melanoma/genetics , Melanoma/pathology , MicroRNAs/genetics , Mutation, Missense , Neoplasm Invasiveness , Proto-Oncogene Proteins B-raf/genetics , RNA, Neoplasm/genetics
11.
Memory ; 26(10): 1379-1384, 2018 11.
Article in English | MEDLINE | ID: mdl-29781391

ABSTRACT

People often have erroneous knowledge about the world that is firmly entrenched in memory and endorsed with high confidence. Although strong errors in memory would seem difficult to "un-learn," evidence suggests that errors are more likely to be corrected through feedback when they are originally endorsed with high confidence compared to low confidence. This hypercorrection effect has been predominantly studied in laboratory settings with general knowledge (i.e., trivia) questions, however, and has not been systematically explored in authentic classroom contexts. In the current study, college students in an introductory horticulture class answered questions about the course content, rated their confidence in their answers, received feedback of the correct answers, and then later completed a posttest. Results revealed a significant hypercorrection effect, along with a tendency for students with higher prior knowledge of the material to express higher confidence in, and in turn more effective correction of, their error responses.


Subject(s)
Learning , Memory/physiology , Mental Recall/physiology , Adult , Feedback, Psychological , Female , Humans , Male , Students , Young Adult
12.
Oncotarget ; 7(18): 26925, 2016 May 03.
Article in English | MEDLINE | ID: mdl-27532884

ABSTRACT

Present: Due to an error made by the authors while submitting a revision, Dr. Tuan Zea Tan was omitted from the list of authors.Corrected: Correct author list can be found below. Authors sincerely apologize for this oversight. Ila Datar1, Xiaoliang Qiu1, Hong Zhi Ma1, Miranda Yeung1, Shweta Aras1, Ivana de la Serna1, Fahd Al-Mulla2, Tuan Zea Tan3, Jean Paul Thiery3, Robert Trumbly1, Xuan Fan4, Hongjuan Cui4 and Kam C. Yeung1 1 Department of Biochemistry and Cancer Biology, University of Toledo, College of Medicine, Health Science Campus, Toledo, OH, USA 2 Kuwait University, Faculty of Medicine. P.O. Box 24923, Safat, Kuwait 3 Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 4 State Key Laboratory Of Silkworm Genome Biology, Chongqing, China Original article: Oncotarget. 2015; 6(36): 39050-61. doi: 10.18632/oncotarget.5176.

13.
Oncotarget ; 6(36): 39050-61, 2015 Nov 17.
Article in English | MEDLINE | ID: mdl-26375811

ABSTRACT

Accumulating evidence suggests that presence of macrophages in the tumor microenvironment add to the invasive and tumor-promoting hallmarks of cancer cells by secreting angiogenic and growth factors. RKIP is a known metastasis suppressor and interferes with several steps of metastasis. However, the mechanistic underpinnings of its function as a broad metastasis suppressor remain poorly understood. Here, we establish a novel pathway for RKIP regulation of metastasis inhibition through the negative regulation of RANTES/CCL5 thereby limiting tumor macrophage infiltration and inhibition of angiogenesis. Using a combination of loss- and gain-of- function approaches, we show that RKIP hinders breast cancer cell invasion by inhibiting expression of the CC chemokine CCL5 in vitro. We also show that the expression levels of RKIP and CCL5 are inversely correlated among clinical human breast cancer samples. Using a mouse allograft breast cancer transplantation model, we highlight that ectopic expression of RKIP significantly decreases tumor vasculature, macrophage infiltration and lung metastases. Mechanistically, we demonstrate that the inhibition of the CCL5 expression is the cause of the observed effects resulting from RKIP expression. Taken together, our results underscore the significance of RKIP as important negative regulator of tumor microenvironment.


Subject(s)
Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Chemokine CCL5/biosynthesis , Macrophages/pathology , Phosphatidylethanolamine Binding Protein/metabolism , Animals , Breast Neoplasms/blood supply , Breast Neoplasms/genetics , Cell Line, Tumor , Chemokine CCL5/genetics , Chemokine CCL5/metabolism , Female , Heterografts , Humans , Mice , Mice, Inbred BALB C , Neoplasm Invasiveness , Neoplasm Metastasis , Neovascularization, Pathologic/metabolism , Neovascularization, Pathologic/pathology , Phosphatidylethanolamine Binding Protein/genetics , Tumor Microenvironment
14.
PLoS One ; 10(8): e0134494, 2015.
Article in English | MEDLINE | ID: mdl-26308852

ABSTRACT

Raf Kinase Inhibitory Protein or RKIP was initially identified as a Raf-1 binding protein using the yeast 2-hybrid screen. RKIP inhibits the activation phosphorylation of MEK by Raf-1 by competitively inhibiting the binding of MEK to Raf-1 and thus exerting an inhibitory effect on the Raf-MEK-Erk pathway. RKIP has been identified as a metastasis suppressor gene. Expression of RKIP is low in cancer metastases. Although primary tumor growth remains unaffected, re- expression of RKIP inhibits cancer metastasis. Mechanistically, RKIP constrains metastasis by inhibiting angiogenesis, local invasion, intravasation, and colonization. The molecular mechanism of how RKIP inhibits these individual steps remains undefined. In our present study, using an unbiased PCR based screening and by analyzing DNA microarray expression datasets we observe that the expression of multiple metalloproteases (MMPs) including MMP1, MMP3, MMP10 and MMP13 are negatively correlated with RKIP expression in breast cancer cell lines and clinical samples. Since expression of MMPs by cancer cells is important for cancer metastasis, we hypothesize that RKIP may mediate suppression of breast cancer metastasis by inhibiting multiple MMPs. We show that the expression signature of RKIP and MMPs is better at predicting high metastatic risk than the individual gene. Using a combination of loss- and gain-of-function approaches, we find that MMP13 is the cause of RKIP-mediated inhibition of local cancer invasion. Interestingly expression of MMP13 alone is not sufficient to reverse the inhibition of breast cancer cell metastasis to the lung due to the expression of RKIP. We find that RKIP negatively regulates MMP13 through the Erk2 signaling pathway and the repression of MMP13 by RKIP is transcription factor AP-1 independent. Together, our findings indicate that RKIP inhibits cancer cell invasion, in part, via MMP13 inhibition. These data also implicate RKIP in the regulation of MMP transcription, suggesting a potential mechanism by which RKIP inhibits tumor progression and metastasis.


Subject(s)
Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Matrix Metalloproteinase 13/genetics , Phosphatidylethanolamine Binding Protein/metabolism , Transcriptional Activation , Animals , Breast Neoplasms/genetics , Cell Line, Tumor , Cell Transformation, Neoplastic , Disease-Free Survival , Gene Expression Regulation, Neoplastic , Humans , Mice , Mitogen-Activated Protein Kinase 1/metabolism , Neoplasm Invasiveness , Neoplasm Metastasis , Signal Transduction
15.
J Aging Res ; 2015: 570410, 2015.
Article in English | MEDLINE | ID: mdl-26090227

ABSTRACT

Background. While behavioral and psychological symptoms are frequent in hospitalized older adults with dementia or delirium, data supporting the off-label use of intramuscular atypical antipsychotics remain scarce. We examined the use of short-acting intramuscular (IM) olanzapine in hospitalized older adults to manage behavioral and psychological symptoms. Methods. A retrospective observational study of inpatients 65 years or older with at least one order for olanzapine IM during admission in urban Ontario Canada was conducted. Patient demographics, prescriptions for olanzapine IM, reason for administration, perceived effectiveness, adverse events, concurrently prescribed psychotropics, comorbidities, and patient discharge destination were recorded. Results. Among 82 patients aged 65-96 years (mean ± SD 79.3 ± 7.7) 85 cases were identified. Cognitive impairment or dementia affected 63.5% and 50.6% had comorbidities. Olanzapine IM was ordered 102 times and 34 patients (41%) received at least one dose. The intended efficacy was achieved in 79.4% of 78 cases of 124 doses given (62.9%). Fourteen (41%) patients who received doses experienced adverse events, with sedation and hypotension being the most common. Conclusions. Olanzapine IM appears effective in hospitalized older adults but is associated with potential adverse events. Structured monitoring and documentation are needed to ensure safe use in this high-risk population.

16.
Prostate ; 75(3): 292-302, 2015 Feb 15.
Article in English | MEDLINE | ID: mdl-25327941

ABSTRACT

BACKGROUND: Raf kinase inhibitor protein (RKIP) has been shown to act as a metastasis suppressor gene in multiple models of cancer. Loss of RKIP expression promotes invasion and metastasis in cell transplantation animal models. However, it is unknown if RKIP expression can impact the progression of cancer in an autochthonous model of cancer. The goal of this study was to determine if loss of RKIP expression in a genetic mouse model of prostate cancer (PCa) impacts metastasis. METHODS: Endogenous RKIP expression was measured in the primary tumors and metastases of transgenic adenocarcinoma of the mouse prostate (TRAMP(+) ) mice. RKIP knockout mice (RKIP(-/-) ) were crossbred with (TRAMP(+) ) mice to create RKIP(-/-) TRAMP(+) mice. Mice were euthanized at 10, 20, and 30 weeks for evaluation of primary and metastatic tumor development. To determine if loss of RKIP alone promotes metastasis, RKIP was knocked down in the low metastatic LNCaP prostate cancer cell line. RESULTS: Endogenous RKIP expression decreased in TRAMP(+) mice as tumors progressed. Primary tumors developed earlier in RKIP(-/-) TRAMP(+) compared to TRAMP(+) mice. At 30 weeks of age, distant metastases were identified only the RKIP(-/-) TRAMP(+) mice. While prostate epithelial cell proliferation rates were higher at 10 and 20 weeks in RKIP(-/-) TRAMP(+) compared to TRAMP(+) mice, by 30 weeks there was no difference. Apoptosis rates in both groups were similar at all timepoints. Decreased RKIP expression did not impact the metastatic rate of LNCaP in an orthotopic PCa model. CONCLUSIONS: These results demonstrate that loss of RKIP decreases latency of tumor development and promotes distant metastasis in the TRAMP mouse model in the context of a pro-metastatic background; but loss of RKIP alone is insufficient to promote metastasis. These findings suggest that in addition to its known metastasis suppressor activity, RKIP may promote tumor progression through enhancing tumor initiation. Prostate 75:292-302, 2015. © 2014 Wiley Periodicals, Inc.


Subject(s)
Adenocarcinoma/pathology , Carcinogenesis/pathology , Neoplasm Metastasis/pathology , Phosphatidylethanolamine Binding Protein/genetics , Prostatic Neoplasms/pathology , Adenocarcinoma/genetics , Animals , Carcinogenesis/genetics , Cell Line, Tumor , Disease Models, Animal , Disease Progression , Humans , Male , Mice , Mice, Knockout , Neoplasm Metastasis/genetics , Prostatic Neoplasms/genetics
17.
PLoS One ; 9(3): e92478, 2014.
Article in English | MEDLINE | ID: mdl-24658061

ABSTRACT

Raf kinase inhibitor protein (RKIP) is a member of the phosphatidylethanolamine-binding-protein (PEBP) family that modulates the action of many kinases involved in cellular growth, apoptosis, epithelial to mesenchymal transition, motility, invasion and metastasis. Previously, we described an inverse association between RKIP and signal transducers and activators of transcription 3 (STAT3) expression in gastric adenocarcinoma patients. In this study, we elucidated the mechanism by which RKIP regulates STAT3 activity in breast and prostate cancer cell lines. RKIP over expression inhibited c-Src auto-phosphorylation and activation, as well as IL-6-, JAK1 and 2-, and activated Raf-mediated STAT3 tyrosine and serine phosphorylation and subsequent activation. In MDA-231 breast cancer cells that stably over express RKIP, IL-6 treatment blocked STAT3 phosphorylation and transcriptional activation. Conversely, in RKIP knockdown MDA-231 cells: STAT3 phosphorylation and activation increased in comparison to parental MDA-231 cells. RKIP over expression resulted in constitutive physical interaction with STAT3 and blocked c-Src and STAT3 association. The treatment of DU145 prostate, but not PC3 prostate or MDA-231 breast, cancer cell lines with ENMD-1198 or MKC-1 dramatically increased expression of RKIP. Overexpression of RKIP sensitized PC3 and MDA-231 cells to MTI-induced apoptosis. Moreover, MTI treatment resulted in a decrease in Src-mediated STAT3 tyrosine phosphorylation and activation, an effect that was significantly enhanced by RKIP over expression. In stable RKIP over expressing MDA-231 cells, tumor xenograft growth induced by activated STAT3 is inhibited. RKIP synergizes with MTIs to induce apoptosis and inhibit STAT3 activation of breast and prostate cancer cells. RKIP plays a critical role in opposing the effects of pro-oncogenic STAT3 activation.


Subject(s)
Breast Neoplasms/physiopathology , Phosphatidylethanolamine Binding Protein/metabolism , Prostatic Neoplasms/physiopathology , STAT3 Transcription Factor/antagonists & inhibitors , Animals , Cell Line, Tumor , Cell Proliferation/drug effects , Estrenes/pharmacology , Female , Humans , Interleukin-6/antagonists & inhibitors , Interleukin-6/pharmacology , Janus Kinase 1/antagonists & inhibitors , Male , Mice , Neoplasm Transplantation , Phosphatidylethanolamine Binding Protein/biosynthesis , STAT3 Transcription Factor/metabolism , Transfection , Tubulin Modulators/pharmacology
18.
Proc Natl Acad Sci U S A ; 111(3): E364-73, 2014 Jan 21.
Article in English | MEDLINE | ID: mdl-24395801

ABSTRACT

The sources and consequences of nongenetic variability in metastatic progression are largely unknown. To address these questions, we characterized a transcriptional regulatory network for the metastasis suppressor Raf kinase inhibitory protein (RKIP). We previously showed that the transcription factor BACH1 is negatively regulated by RKIP and promotes breast cancer metastasis. Here we demonstrate that BACH1 acts in a double-negative (overall positive) feedback loop to inhibit RKIP transcription in breast cancer cells. BACH1 also negatively regulates its own transcription. Analysis of the BACH1 network reveals the existence of an inverse relationship between BACH1 and RKIP involving both monostable and bistable transitions that can potentially give rise to nongenetic variability. Single-cell analysis confirmed monostable and bistable-like behavior. Treatment with histone deacetylase inhibitors or depletion of the polycomb repressor enhancer of zeste homolog 2 altered relative RKIP and BACH1 levels in a manner consistent with a prometastatic state. Together, our results suggest that the mutually repressive relationship between metastatic regulators such as RKIP and BACH1 can play a key role in determining metastatic progression in cancer.


Subject(s)
Basic-Leucine Zipper Transcription Factors/metabolism , Breast Neoplasms/metabolism , Cell Transformation, Neoplastic , Fanconi Anemia Complementation Group Proteins/metabolism , Gene Expression Regulation, Neoplastic , Phosphatidylethanolamine Binding Protein/metabolism , Amino Acid Motifs , Cell Line, Tumor , Chromatin Immunoprecipitation , Disease Progression , Feedback, Physiological , Female , Genetic Variation , Humans , MCF-7 Cells , Models, Theoretical , Neoplasm Metastasis , Oxidative Stress , Promoter Regions, Genetic , Time Factors , Transcription, Genetic
19.
Crit Rev Oncog ; 19(6): 417-30, 2014.
Article in English | MEDLINE | ID: mdl-25597352

ABSTRACT

Raf kinase inhibitory protein (RKIP) is known to modulate key signaling cascades and regulate normal physiological processes such as cellular proliferation, differentiation, and apoptosis. The expression of RKIP is found to be downregulated in several cancer metastases and the repressed RKIP expression can be reactivated on treatment with chemotherapeutic agents. RKIP is a proven tumor metastasis suppressor gene and investigating the mechanisms of transcriptional regulation of RKIP is therefore of immense clinical importance. In this review, we discuss the basal expression of RKIP in various tissues and the genetic aspects of the RKIP chromosomal locus including the structure of the RKIP promoter as well as gene regulatory elements such as enhancers. We also review the genetic and epigenetic modulation of RKIP transcription through EZH2, a component of the polycomb repressive complex 2 (PRC2) and sequence specific transcription factors (TFs) BACH1 and Snail. Emerging experimental evidence supports a unifying model in which both these TFs repress RKIP transcription in cancers by recruiting the EZH2 containing repressive complex to the proximal RKIP promoter. Finally, we review the known mechanisms employed by different types of chemotherapeutic agents to activate RKIP expression in cancer cells.


Subject(s)
Epigenesis, Genetic , Phosphatidylethanolamine Binding Protein/genetics , Animals , Enhancer Elements, Genetic , Gene Expression Regulation , Humans , Neoplasms/genetics , Neoplasms/pathology , Transcription, Genetic
20.
Am J Cancer Res ; 3(5): 446-64, 2013.
Article in English | MEDLINE | ID: mdl-24224123

ABSTRACT

Raf Kinase inhibitory protein (RKIP) is a well-established metastasis suppressor that is frequently downregulated in aggressive cancers. The impact of RKIP and its phosphorylated form on disease-free survival (DFS) and other clinicopathological parameters in breast cancer is yet to be discovered. To this end, we examined RKIP expression in 3 independent breast cancer cohorts. At the Protein level, loss or reduced total RKIP expression was associated with large-sized tumors characterized by high proliferative index, high-grade and diminished estrogen (ER) and progesterone receptor expression. Loss or diminution of RKIP expression was significantly associated with shorter DFS in all cohorts. Moreover, the complete loss of p-RKIP was an independent prognostic factor using multivariate analysis in operable invasive ductal breast cancer. We show for the first time that ER, partly, drives RKIP expression through MTA3-Snail axis. Consistent with this finding, we found that, at the mRNA level, RKIP expression varied significantly across the different molecular subtypes of breast cancer with the Luminal (ER+) subtype expressing high levels of RKIP and the more aggressive Claudin-low (ER-) subtype, which depicted the highest epithelial to mesenchymal transition (EMT) registered the lowest RKIP expression levels. In conclusion, loss of expression/diminution of RKIP or its phosphorylated form is associated with poor diseases-free survival in breast cancer. Determining the expression of RKIP and p-RKIP adds significant prognostic value to the management and subtyping of this disease.

SELECTION OF CITATIONS
SEARCH DETAIL
...