Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 52
Filter
1.
Br J Dermatol ; 2024 Apr 09.
Article in English | MEDLINE | ID: mdl-38591490

ABSTRACT

BACKGROUND: PRDM12 polyalanine tract expansions cause two different disorders; Midfacial Toddler Excoriation Syndrome (MiTES) - itch with normal pain sensation associated with homozygous 18 alanines (18A), and congenital insensitivity to pain (CIP) with normal itch with homozygous 19A. Knowledge of the phenotype, genotype, and disease mechanism of MiTES is incomplete. Why PRDM12 18A versus 19A can cause almost opposite phenotypes is unknown; no other poly-alanine or poly-glutamine tract expansion disease causes two such disparate phenotypes. METHODS: We assessed the genotype and phenotype of 9 new, 9 atypical, and 6 previously reported patients diagnosed with MiTES. Using cell lines with homozygous PRDM12 of 12A (normal), 18A (MiTES) and 19A (CIP) we examined PRDM12 aggregation and subcellular localisation by image separation confocal microscopy and sub-cellular fractionation western blotting. RESULTS: MiTES presents in the first year of life, and in all cases the condition regresses over the first decade leaving scarring. The MiTES phenotype is highly distinctive. Features overlapping with PRDM12-CIP are rarely found. The genotype-phenotype study of PRDM12 polyalanine tract shows that 7A -15A are normal; 16A -18A are associated with MiTES; 19A leads to CIP; and no clinically atypical MiTES cases had an expansion. PRDM12 aggregation and sub-cellular localisation differ significantly between 18A and normal 12A cell lines and between 18A and 19A cell lines. MiTES is a new protein aggregation disease. CONCLUSION: We provide diagnostic criteria for MiTES, and improved longitudinal data. MiTES and CIP are distinct phenotypes despite their genotypes varying by a single alanine in the PRDM12 polyalanine tract. We found clear distinctions between the cellular phenotypes of normal, MiTES and CIP cells.. We hypothesise that the developmental environment of the trigeminal ganglion is unique and critically sensitive to prenatal and postnatal levels of PRDM12.

2.
Hemoglobin ; 44(5): 371-375, 2020 Sep.
Article in English | MEDLINE | ID: mdl-33003974

ABSTRACT

The burden of ß-thalassemia (ß-thal) is largely underestimated in India with a carrier frequency of 3.0-4.0% in general, whereas highly stratified frequencies of up to 17.0% are reported in local endogamous subpopulations. We have no idea whether ß-thal carrier frequencies or ß-thal major (ß-TM) births are increasing or decreasing in the population. The cross-sectional nature of all carrier screening programs including large-scale task force and micro level, lack of registration of ß-TM births and mechanism to modulate knowledge, awareness programs in a long-term perspective, all preempt impact assessment of preventive programs. During the implementation of a Telangana State Government-sponsored program on 'Micro profiling of ß-thalassemia mutations in Telangana,' we documented extensive in-depth demographic information on each ß-TM child of the study sample that included age-sex distributions, parental and grand-parental ethnic affiliations (local endogamous group level), birth places, marital migrations, endogamy and consanguinity to identify high-risk districts as ethno-geographic regions. In Telangana State, we found ß-thal is widely prevalent in 31 districts and 48 local endogamous subpopulations. The present study provided a method of identification of four 'high-risk districts' and developed a district model for prevention on high priority in Telangana State. The model has the advantage of impact-assessment of all preventive programs in the district.


Subject(s)
beta-Thalassemia/etiology , beta-Thalassemia/prevention & control , Female , Humans , India/epidemiology , Male , Models, Theoretical , Public Health Surveillance , Risk Assessment , Risk Factors , beta-Thalassemia/epidemiology
3.
J Neuroendocrinol ; 30(2)2018 02.
Article in English | MEDLINE | ID: mdl-28485080

ABSTRACT

Proline-, glutamic acid- and leucine-rich protein 1 (PELP1) is an oestrogen receptor (ER) coregulator protein identified by our collaborative group. Work from our laboratory and others has shown that PELP1 is a scaffold protein that interacts with ERs and kinase signalling factors, as well as proteins involved in chromatin remodelling and DNA repair. Its role in mediating 17ß-oestradiol (E2 ) signalling and actions has been studied in detail in cancer cells, although only recently has attention turned to its role in the brain. In this review, we discuss the tissue, cellular and subcellular localisation of PELP1 in the brain. We also discuss recent evidence from PELP1 forebrain-specific knockout mice demonstrating a critical role of PELP1 in mediating both extranuclear and nuclear ER signalling in the brain, as well as E2 -induced neuroprotection, anti-inflammatory effects and regulation of cognitive function. Finally, the PELP1 interactome and unique gene network regulated by PELP1 in the brain is discussed, especially because it provides new insights into PELP1 biology, protein interactions and mechanisms of action in the brain. As a whole, the findings discussed in the present review indicate that PELP1 functions as a critical ER coregulator in the brain to mediate E2 signalling and actions.


Subject(s)
Brain/metabolism , Co-Repressor Proteins/metabolism , Estrogens/metabolism , Signal Transduction/physiology , Transcription Factors/metabolism , Animals , Co-Repressor Proteins/genetics , Mice , Mice, Knockout , Transcription Factors/genetics
4.
Oncogene ; 36(17): 2423-2434, 2017 04 27.
Article in English | MEDLINE | ID: mdl-27893719

ABSTRACT

Glioma stem cells (GSCs) have a central role in glioblastoma (GBM) development and chemo/radiation resistance, and their elimination is critical for the development of efficient therapeutic strategies. Recently, we showed that lysine demethylase KDM1A is overexpressed in GBM. In the present study, we determined whether KDM1A modulates GSCs stemness and differentiation and tested the utility of two novel KDM1A-specific inhibitors (NCL-1 and NCD-38) to promote differentiation and apoptosis of GSCs. The efficacy of KDM1A targeting drugs was tested on purified GSCs isolated from established and patient-derived GBMs using both in vitro assays and in vivo orthotopic preclinical models. Our results suggested that KDM1A is highly expressed in GSCs and knockdown of KDM1A using shRNA-reduced GSCs stemness and induced the differentiation. Pharmacological inhibition of KDM1A using NCL-1 and NCD-38 significantly reduced the cell viability, neurosphere formation and induced apoptosis of GSCs with little effect on differentiated cells. In preclinical studies using orthotopic models, NCL-1 and NCD-38 significantly reduced GSCs-driven tumor progression and improved mice survival. RNA-sequencing analysis showed that KDM1A inhibitors modulate several pathways related to stemness, differentiation and apoptosis. Mechanistic studies showed that KDM1A inhibitors induce activation of the unfolded protein response (UPR) pathway. These results strongly suggest that selective targeting of KDM1A using NCL-1 and NCD-38 is a promising therapeutic strategy for elimination of GSCs.


Subject(s)
Apoptosis/drug effects , Cell Differentiation/drug effects , Enzyme Inhibitors/pharmacology , Glioma/pathology , Histone Demethylases/antagonists & inhibitors , Neoplastic Stem Cells/drug effects , Unfolded Protein Response/drug effects , Animals , Cell Line, Tumor , Cell Survival/drug effects , Cell Transformation, Neoplastic , Disease Progression , Gene Expression Regulation, Neoplastic/drug effects , Mice , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Signal Transduction/drug effects , Survival Analysis , Transcription, Genetic/drug effects
5.
J Hum Genet ; 60(9): 485-92, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26016408

ABSTRACT

Little information is available regarding the molecular pathogenesis of Parkinson's disease (PD) among the Bengalee population in West Bengal, India. This study was undertaken to determine the contribution of Parkin variants in well-defined ethnically identical Bengalee population of India and further to describe the clinical spectrum associated with these mutations. A total of 150 unrelated PD patients and 150 controls were recruited for the study. The entire cohort was screened for mutations in all the 12 exons of the gene along with flanking splice junctions by polymerase chain reaction and DNA sequencing. Eleven nucleotide variants including two novel changes were detected. Cerebrospinal fluid (CSF) parkin protein expression of the novel mutation, Val186Ile (found in heterozygous condition in one patient only) was almost 2.7 folds lower than the controls and other PD patients. Molecular characterization of polymorphisms Ser167Asn and Val380Leu depicted that homozygous Ser167 and Val380 are significantly associated with the disease. We did not find any linkage disequilibrium among the SNPs, the low r(2) for every pair of single-nucleotide polymorphisms (SNPs) indicated that these SNPs cannot be tagged by each other. Another novel intronic change, IVS8+48C>T was present in almost equally in PD patients and controls. Among the ethnically defined Bengalee population of West Bengal, occurrence of Parkin mutation is 4% (6/150) of the PD patient pool supported with decreased folds of expression of CSF PARKIN protein. Parkin polymorphisms, Ser167 and Val380 are risk factors for the progression of the disease, and their frequency is greatly influenced by ethnic origin.


Subject(s)
Parkinson Disease/genetics , Polymorphism, Single Nucleotide , Ubiquitin-Protein Ligases/genetics , Adult , Aged , Aged, 80 and over , Case-Control Studies , Female , Gene Frequency , Genetic Association Studies , Genetic Predisposition to Disease/ethnology , Humans , India/epidemiology , Male , Middle Aged , Parkinson Disease/epidemiology
6.
Cell Death Differ ; 21(9): 1409-18, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24786831

ABSTRACT

Proline-, glutamic acid- and leucine-rich protein-1 (PELP1) is a scaffolding oncogenic protein that functions as a coregulator for a number of nuclear receptors. p53 is an important transcription factor and tumor suppressor that has a critical role in DNA damage response (DDR) including cell cycle arrest, repair or apoptosis. In this study, we found an unexpected role for PELP1 in modulating p53-mediated DDR. PELP1 is phosphorylated at Serine1033 by various DDR kinases like ataxia-telangiectasia mutated, ataxia telangiectasia and Rad3-related or DNAPKc and this phosphorylation of PELP1 is important for p53 coactivation functions. PELP1-depleted p53 (wild-type) breast cancer cells were less sensitive to various genotoxic agents including etoposide, camptothecin or γ-radiation. PELP1 interacts with p53, functions as p53-coactivator and is required for optimal activation of p53 target genes under genomic stress. Overall, these studies established a new role of PELP1 in DDRs and these findings will have future implications in our understanding of PELP1's role in cancer progression.


Subject(s)
Co-Repressor Proteins/metabolism , DNA Damage , Transcription Factors/metabolism , Tumor Suppressor Protein p53/metabolism , Cell Line, Tumor , Humans , MCF-7 Cells , Phosphorylation
7.
Oncogene ; 33(28): 3707-16, 2014 Jul 10.
Article in English | MEDLINE | ID: mdl-23975430

ABSTRACT

Tumor metastasis is the leading cause of death among breast cancer patients. PELP1 (proline, glutamic acid and leucine rich protein 1) is a nuclear receptor coregulator that is upregulated during breast cancer progression to metastasis and is an independent prognostic predictor of shorter survival of breast cancer patients. Here, we show that PELP1 modulates expression of metastasis-influencing microRNAs (miRs) to promote cancer metastasis. Whole genome miR array analysis using PELP1-overexpressing and PELP1-underexpressing model cells revealed that miR-200 and miR-141 levels inversely correlated with PELP1 expression. Consistent with this, PELP1 knockdown resulted in lower expression of miR-200a target genes ZEB1 and ZEB2. PELP1 knockdown significantly reduced tumor growth and metastasis compared with parental cells in an orthotopic xenograft tumor model. Furthermore, re-introduction of miR-200a and miR-141 mimetics into PELP1-overexpressing cells reversed PELP1 target gene expression, decreased PELP1-driven migration/invasion in vitro and significantly reduced in vivo metastatic potential in a preclinical model of experimental metastasis. Our results demonstrated that PELP1 binds to miR-200a and miR-141 promoters and regulates their expression by recruiting chromatin modifier histone deacetylase 2 (HDAC2) as revealed by chromatin immunoprecipitation, small interfering RNA and HDAC inhibitor assays. Taken together, our results suggest that PELP1 regulates tumor metastasis by controlling the expression and functions of the tumor metastasis suppressors miR-200a and miR-141.


Subject(s)
Breast Neoplasms/pathology , Co-Repressor Proteins/metabolism , Epithelial-Mesenchymal Transition , Histone Deacetylase 2/metabolism , MicroRNAs/metabolism , Transcription Factors/metabolism , Animals , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Cell Line, Tumor , Cell Proliferation , Co-Repressor Proteins/genetics , Epigenesis, Genetic , Female , Gene Expression Regulation, Neoplastic , Gene Knockdown Techniques , Humans , Mice , Mice, Nude , MicroRNAs/genetics , Neoplasm Metastasis , Transcription Factors/deficiency , Transcription Factors/genetics
8.
Minerva Ginecol ; 64(1): 75-88, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22334233

ABSTRACT

Estrogen receptor (ERα) plays an important role in the initiation and progression of breast cancer. Many breast cancer patients respond positively to hormonal therapy; however, initial or acquired resistance to endocrine therapies frequently occurs and tumors recur as metastases. Emerging evidence suggests that ERα action is complex and involves functional interactions with coregulators. The levels of ERα coregulator proteins are tightly regulated under normal conditions and ERα-coregulator proteins have the potential to be differentially expressed in malignant tumors and have altered functions leading to tumor progression and metastases. In this review, we summarize recent findings that relate to ERα coregulators and discuss implications of ERα coregulator deregulation in breast cancer metastasis.


Subject(s)
Breast Neoplasms/etiology , Estrogen Receptor alpha/physiology , Breast Neoplasms/drug therapy , Breast Neoplasms/pathology , Estrogen Receptor alpha/antagonists & inhibitors , Female , Humans , Neoplasm Metastasis
9.
J Neuroendocrinol ; 24(1): 34-47, 2012 Jan.
Article in English | MEDLINE | ID: mdl-21722216

ABSTRACT

17ß-Oestradiol (E(2)) is an important hormone signal that regulates multiple tissues and functions in the body. This review focuses on the neuroprotective actions of E(2) in the brain against cerebral ischaemia and the potential underlying mechanisms. A particular focus of the review will be on the role of E(2) to attenuate NADPH oxidase activation, superoxide and reactive oxygen species generation and reduce oxidative stress in the ischaemic brain as a potentially key neuroprotective mechanism. Evidence of a potential novel role of extranuclear oestrogen receptors in mediating E(2) signalling and neuroprotective actions is also discussed. An additional subject is the growing evidence indicating that periods of long-term oestrogen deprivation, such as those occurring after menopause or surgical menopause, may lead to loss or attenuation of E(2) signalling and neuroprotective actions in the brain, as well as enhanced sensitivity of the hippocampus to ischaemic stress damage. These findings have important implications with respect to the 'critical period hypothesis', which proposes that oestrogen replacement must be initiated at peri-menopause in humans to exert its beneficial cardiovascular and neural effects. The insights gained from these various studies will prove valuable for guiding future directions in the field.


Subject(s)
Brain Ischemia/metabolism , Brain/metabolism , Estrogens/metabolism , Neuroprotective Agents/metabolism , Stroke/metabolism , Animals , Brain Ischemia/drug therapy , Estrogens/therapeutic use , Humans , Reactive Oxygen Species/metabolism , Receptors, Estrogen/metabolism , Stroke/drug therapy
10.
Minerva Ginecol ; 62(6): 573-83, 2010 Dec.
Article in English | MEDLINE | ID: mdl-21079578

ABSTRACT

The estrogen receptor (ERa) is implicated in the progression of breast cancer. Hormonal therapies which block ER functions or local and systemic estrogen production are currently used to treat ERa positive breast cancer. Hormonal therapy shows beneficial effects, however, initial or acquired resistance to endocrine therapies frequently occurs, and tumors recur as metastasis. Emerging evidence suggests in addition to exerting its well-studied nuclear functions, ERa also participates in extranuclear signaling that involve growth factor signaling components, adaptor molecules and the stimulation of cytosolic kinases. ERa extranuclear pathways have the potential to activate gene transcription, modulate cytoskeleton, and promote tumor cell proliferation, survival, and metastasis. Cytoplasmic/membrane ERa is detected in a subset of breast tumors and expression of extranuclear components ERa is deregulated in tumors. The extranuclear actions of ER are emerging as important targets for tumorigenic and metastatic control. Inhibition of ERa extranuclear actions has the potential to prevent breast tumor progression and may be useful in preventing ERa positive metastasis. In this review, we summarize the results of recent research into the role of ERa mediated extranuclear actions in breast tumorigenesis and metastasis.


Subject(s)
Breast Neoplasms/genetics , Breast Neoplasms/secondary , Estrogen Receptor alpha/genetics , Signal Transduction/genetics , Antineoplastic Agents, Hormonal/therapeutic use , Biomarkers, Tumor/genetics , Breast Neoplasms/drug therapy , Breast Neoplasms/pathology , Disease Progression , Female , Humans , Neoplasm Invasiveness , Neoplasm Metastasis , Receptors, Estrogen/genetics , Treatment Outcome
11.
Histol Histopathol ; 22(1): 91-6, 2007 01.
Article in English | MEDLINE | ID: mdl-17128415

ABSTRACT

The estrogen receptors ERalpha and ERbeta have been implicated in the progression of a wide variety of cancers. The actions of ER are regulated by ER coregulator proteins, including proline-, glutamic acid- and leucine-rich-protein-1 (PELP1/MNAR). PELP1 has been shown to participate in both genomic and nongenomic functions of ER. The expression and localization of PELP1/MNAR are deregulated in a wide variety of tumors and have been implicated in the development of hormonal resistance in cancer cell lines. Emerging data suggest that PELP1/MNAR interacts with many proteins and activates several oncogenes, including Src kinase, phosphotidyl inositol 3 kinase (PI3K), and signal transducers and activators of transcription 3 (STAT3). These new results suggest that PELP1/MNAR may act as an oncogene as well as cooperating with other oncogenes. Thus, PELP1/MNAR may contribute to the tumorigenic potential of cancer cells by serving as a scaffolding protein that couples various signaling complexes with ER.


Subject(s)
Gene Expression Regulation, Neoplastic , Neoplasms/metabolism , Receptors, Estrogen/metabolism , Trans-Activators/physiology , Co-Repressor Proteins , Female , Humans , Male , Models, Biological , Neoplasms/genetics , Phosphatidylinositol 3-Kinases/metabolism , STAT3 Transcription Factor/metabolism , Signal Transduction , Transcription Factors
12.
Cell Mol Life Sci ; 61(3): 281-91, 2004 Feb.
Article in English | MEDLINE | ID: mdl-14770293

ABSTRACT

The steroid hormone estrogen and signaling from its receptors are increasingly recognized as critical mediators of a variety of organ-specific biological processes. Recent advances in the identification and functional characterization of novel estrogen receptor interacting proteins clearly show the complexity of hormonal signaling regulation, but may also contribute to our understanding of the roles of estrogen signaling in normal physiology and the pathobiology of human disease.


Subject(s)
Receptors, Estrogen/metabolism , Signal Transduction/physiology , Bone and Bones/physiology , Cardiovascular Physiological Phenomena , Chromatin/metabolism , Estrogens/metabolism , Humans , Neoplasms/metabolism , Neuroprotective Agents/metabolism , Nuclear Proteins/metabolism , Repressor Proteins/metabolism
13.
Insect Biochem Mol Biol ; 32(9): 1025-36, 2002 Sep.
Article in English | MEDLINE | ID: mdl-12213239

ABSTRACT

Many subspecies of the soil bacterium Bacillus thuringiensis produce various parasporal crystal proteins, also known as Cry toxins, that exhibit insecticidal activity upon binding to specific receptors in the midgut of susceptible insects. One such receptor, BT-R(1) (210 kDa), is a cadherin located in the midgut epithelium of the tobacco hornworm, Manduca sexta. It has a high binding affinity (K(d) approximately 1nM) for the Cry1A toxins of B. thuringiensis. Truncation analysis of BT-R(1) revealed that the only fragment capable of binding the Cry1A toxins of B. thuringiensis was a contiguous 169-amino acid sequence adjacent to the membrane-proximal extracellular domain. The purified toxin-binding fragment acted as an antagonist to Cry1Ab toxin by blocking the binding of toxin to the tobacco hornworm midgut and inhibiting insecticidal action. Exogenous Cry1Ab toxin bound to intact COS-7 cells expressing BT-R(1) cDNA, subsequently killing the cells. Recruitment of BT-R(1) by B. thuringiensis indicates that the bacterium interacts with a specific cell adhesion molecule during its pathogenesis. Apparently, Cry toxins, like other bacterial toxins, attack epithelial barriers by targeting cell adhesion molecules within susceptible insect hosts.


Subject(s)
Bacillus thuringiensis/metabolism , Bacterial Proteins/metabolism , Bacterial Toxins/metabolism , Cadherins/metabolism , Endotoxins/metabolism , Insect Proteins , Receptors, Cell Surface/metabolism , Amino Acid Sequence , Animals , Bacillus thuringiensis Toxins , Binding Sites , COS Cells , Chlorocebus aethiops , Hemolysin Proteins , Larva , Manduca , Molecular Sequence Data , Protein Binding , Protein Structure, Tertiary , Receptors, Cell Surface/isolation & purification , Transfection
14.
Biochem Biophys Res Commun ; 289(3): 725-32, 2001 Dec 07.
Article in English | MEDLINE | ID: mdl-11726208

ABSTRACT

Heregulin-beta1 (HRG) is a regulatory polypeptide having several distinct biological effects in mammary epithelial cells. To address the hypothesis that HRG selectively regulates gene expression, we performed differential display screening using cells grown in the presence or absence of HRG. One cDNA clone upregulated by HRG was identical to human calnexin, a protein with molecular chaperone function. This is the first demonstration of the regulation of calnexin mRNA and protein expression by a physiologically relevant polypeptide factor in human breast cancer cells. HRG stimulation also caused a rapid redistribution of calnexin from vesicle-like structures in the cell cytoplasm to the perinuclear area and to the cell membrane. Furthermore, HRG induced colocalization and physical interaction of calnexin with the HER2 growth factor receptor. Finally, calnexin protein levels were increased in progressive stages of human breast cancer. These findings suggest that stimulation of calnexin expression by HRG may constitute a mechanism of protein redistribution and facilitate downstream signaling events in growth-factor-activated cells.


Subject(s)
Breast Neoplasms/metabolism , Calcium-Binding Proteins/metabolism , Gene Expression Regulation, Neoplastic , Molecular Chaperones/metabolism , Neuregulin-1/pharmacology , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Calnexin , Cell Membrane/metabolism , Cycloheximide/pharmacology , Disease Progression , Female , Humans , Microscopy, Confocal , Oligonucleotide Array Sequence Analysis , Protein Synthesis Inhibitors/pharmacology , Protein Transport/drug effects , RNA, Messenger/biosynthesis , Receptor, ErbB-2/metabolism , Transcriptional Activation , Tumor Cells, Cultured
15.
J Biol Chem ; 276(41): 38272-9, 2001 Oct 12.
Article in English | MEDLINE | ID: mdl-11481323

ABSTRACT

Nuclear hormone receptors (NRs) are transcription factors whose activity is regulated by ligands and by coactivators or corepressors. We report the characterization of a new NR coregulator: proline-, glutamic acid-, leucine-rich protein 1 (PELP1), a novel human protein that comprises 1,282 amino acids and is localized on chromosome 17. The primary structure of PELP1 consists of several motifs present in most transcriptional regulators including nine NR-interacting boxes (LXXLL motifs), a zinc finger, and glutamic acid- and proline-rich regions. We demonstrate that PELP1 is a coactivator of estrogen receptor alpha (ERalpha). PELP1 enhances 17beta-estradiol-dependent transcriptional activation from the estrogen response element in a dose-dependent manner. PELP1 interacts with ERalpha and also with general transcriptional coactivators p300 and cAMP response element-binding protein-binding protein. PELP1 was differentially expressed in various human and murine tissues with the highest expression levels in the testes, mammary glands, and brain. We also provide evidence supporting the developmental regulation of PELP1 expression in murine mammary glands, the detectable expression of PELP1 in human mammary cancer cell lines, and the enhanced expression of PELP1 in human breast tumors. These findings suggest that PELP1 is a novel coregulator of ERalpha and may have a role in breast cancer tumorigenesis.


Subject(s)
Receptors, Estrogen/physiology , Trans-Activators/physiology , Amino Acid Sequence , Breast/metabolism , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cloning, Molecular , Co-Repressor Proteins , DNA, Complementary , Estrogen Receptor alpha , Humans , Molecular Sequence Data , Promoter Regions, Genetic , Protein Binding , Sequence Homology, Amino Acid , Trans-Activators/chemistry , Trans-Activators/genetics , Trans-Activators/metabolism , Transcription Factors , Tumor Cells, Cultured , Up-Regulation
16.
J Biol Chem ; 276(30): 28443-50, 2001 Jul 27.
Article in English | MEDLINE | ID: mdl-11328805

ABSTRACT

Heregulin-beta1 (HRG) promotes motility, scattering, and invasiveness of breast cancer cells. Tiam1, a newly identified guanine nucleotide exchange factor, has been shown to inhibit or promote cell migration in a cell type-dependent manner. In this study, we identified Tiam1 as a target of HRG signaling. HRG stimulation of breast cancer epithelial cells induced the phosphorylation and redistribution of Tiam1 to the membrane ruffles and the loosening of intercellular junctions. In addition, HRG-mediated scattering of breast epithelial cells was accompanied by stimulation of tyrosine phosphorylation and redistribution of beta-catenin from the cell junctions to the cytosol and, finally, entry into the nucleus. Decompaction of breast cancer epithelial cells by HRG was accompanied by a transient physical association of the tyrosine-phosphorylated beta-catenin with the activated human epidermal growth factor receptor 2 and subsequent nuclear translocation of beta-catenin, as well as beta-catenin-dependent transactivation of T-cell factor.lymphoid enhancer factor-1. All of these HRG-induced phenotypic changes were regulated in a phosphatidylinositol-3 kinase-sensitive manner. HRG-induced cellular ruffles, loss of intercellular adhesiveness, and increased cell migration could be mimicked by overexpression of a fully functional Tiam1 construct. Furthermore, ectopic expression of Tiam1 or of an active beta-catenin mutant led to potentiation of the beta-catenin-dependent T-cell factor.lymphoid enhancer factor-1 transactivation and invasiveness of HRG-treated cells. We also found preliminary evidence suggesting a close correlation between the status of Tiam1 expression and invasiveness of human breast tumor cells with the degree of progression of breast tumors. Together, these findings suggest that HRG regulate Tiam1 activation and lymphoid enhancer factor/beta-catenin nuclear signaling via phosphatidylinositol-3 kinase in breast cancer cells.


Subject(s)
Breast Neoplasms/metabolism , Cell Nucleus/metabolism , DNA-Binding Proteins/metabolism , Neuregulin-1/metabolism , Protein Biosynthesis , Proteins , Trans-Activators , Transcription Factors/metabolism , Active Transport, Cell Nucleus , Cell Adhesion , Cell Membrane/metabolism , Cell Movement , Cytoskeletal Proteins/metabolism , Cytosol/metabolism , Epithelial Cells/metabolism , ErbB Receptors/metabolism , Fluorescent Antibody Technique, Indirect , Guanine Nucleotide Exchange Factors , Humans , Lymphoid Enhancer-Binding Factor 1 , Microscopy, Fluorescence , Phenotype , Phosphatidylinositol 3-Kinases/metabolism , Phosphorylation , Precipitin Tests , Signal Transduction , T-Lymphoma Invasion and Metastasis-inducing Protein 1 , Transcriptional Activation , Tumor Cells, Cultured , Tyrosine/metabolism , beta Catenin
17.
J Biol Chem ; 276(31): 29403-9, 2001 Aug 03.
Article in English | MEDLINE | ID: mdl-11382770

ABSTRACT

Etk/Bmx, a member of the Tec family of nonreceptor protein-tyrosine kinases, is characterized by an N-terminal pleckstrin homology domain and has been shown to be a downstream effector of phosphatidylinositol 3-kinase. P21-activated kinase 1 (Pak1), another well characterized effector of phosphatidylinositol 3-kinase, has been implicated in the progression of breast cancer cells. In this study, we characterized the role of Etk in mammary development and tumorigenesis and explored the functional interactions between Etk and Pak1. We report that Etk expression is developmentally regulated in the mammary gland. Using transient transfection, coimmunoprecipitation and glutathione S-transferase-pull down assays, we showed that Etk directly associates with Pak1 via its N-terminal pleckstrin homology domain and also phosphorylates Pak1 on tyrosine residues. The expression of wild-type Etk in a non-invasive human breast cancer MCF-7 cells significantly increased proliferation and anchorage-independent growth of epithelial cancer cells. Conversely, expression of kinase-inactive mutant Etk-KQ suppressed the proliferation, anchorage-independent growth, and tumorigenicity of human breast cancer MDA-MB435 cells. These results indicate that Pak1 is a target of Etk and that Etk controls the proliferation as well as the anchorage-independent and tumorigenic growth of mammary epithelial cancer cells.


Subject(s)
Breast Neoplasms/pathology , Gene Expression Regulation, Developmental , Gene Expression Regulation, Enzymologic , Protein Serine-Threonine Kinases/metabolism , Protein-Tyrosine Kinases/genetics , Protein-Tyrosine Kinases/metabolism , Animals , Apoptosis , Breast Neoplasms/enzymology , Cell Division , Enzyme Activation , Female , Humans , Kinetics , Mammary Glands, Animal/embryology , Mammary Glands, Animal/enzymology , Mice , Mice, Nude , Organ Specificity , Phosphorylation , Phosphotyrosine/metabolism , Recombinant Proteins/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Transfection , Transplantation, Heterologous , Tumor Cells, Cultured , p21-Activated Kinases
18.
Gastroenterology ; 120(1): 71-8, 2001 Jan.
Article in English | MEDLINE | ID: mdl-11208715

ABSTRACT

BACKGROUND & AIMS: The colonic epithelial cells near the top of the crypt have been shown to undergo apoptosis. Because butyric acid (BA) is the major short-chain fatty acid produced by fermentation of dietary fiber in the large bowel, it may be an important regulator of apoptosis in colorectal cancer. We investigated which signaling pathway is triggered by BA to undergo apoptosis in human colorectal cancer cells. METHODS: Human DiFi and FET colorectal cells were treated with BA to undergo apoptosis and were assayed for activation of c-Jun N-terminal kinase (JNK), transcription factor activation protein 1 (AP1) and NF-kappaB, and the proapoptotic molecule Bax. The contribution of specific pathways was assessed by examining the effects of dominant-negative mutants of JNK/AP1 or NF-kappaB on BA-induced Bax expression and apoptosis. RESULTS: BA-mediated DNA fragmentation and Bax induction were preceded by early stimulation of JNK, and the DNA-binding activities of AP1 and NF-kappaB. BA-induced enhancement of DNA fragmentation and stimulation of Bax promoter activity were blocked by the expression of dominant-negative mutants of JNK1 or AP1 but not NF-kappaB. CONCLUSIONS: These findings suggest that apoptosis triggered by BA involves transcriptional stimulation of the Bax gene via activation of the JNK/AP1 pathway in colonic epithelial cells.


Subject(s)
Apoptosis/drug effects , Butyric Acid/pharmacology , Colorectal Neoplasms , Histamine Antagonists/pharmacology , Mitogen-Activated Protein Kinases/metabolism , Proto-Oncogene Proteins c-bcl-2 , Proto-Oncogene Proteins/genetics , Transcription Factor AP-1/metabolism , Apoptosis/physiology , DNA Fragmentation/drug effects , Gene Expression Regulation/drug effects , Humans , JNK Mitogen-Activated Protein Kinases , Mutagenesis/physiology , Promoter Regions, Genetic/physiology , Transcriptional Activation/physiology , Tumor Cells, Cultured/cytology , Tumor Cells, Cultured/drug effects , Tumor Cells, Cultured/physiology , bcl-2-Associated X Protein
19.
Nat Cell Biol ; 3(1): 30-7, 2001 Jan.
Article in English | MEDLINE | ID: mdl-11146623

ABSTRACT

Activation of the heregulin/HER2 pathway in oestrogen receptor (ER)-positive breast-cancer cells leads to suppression of oestrogen-receptor element (ERE)-driven transcription and disruption of oestradiol responsiveness, and thus contributes to progression of tumours to more invasive phenotypes. Here we report the identification of metastatic-associated protein 1 (MTA1), a component of histone deacetylase (HDAC) and nucleosome-remodelling complexes, as a gene product induced by heregulin-beta1 (HRG). Stimulation of cells with HRG is accompanied by suppression of histone acetylation and enhancement of deacetylase activity. MTA1 is also a potent corepressor of ERE transcription, as it blocks the ability of oestradiol to stimulate ER-mediated transcription. The histone-deacetylase inhibitor trichostatin A blocks MTA1-mediated repression of ERE transcription. Furthermore, MTA1 directly interacts with histone deacetylase-1 and -2 and with the activation domain of ER-alpha. Overexpression of MTA1 in breast-cancer cells is accompanied by enhancement of the ability of cells to invade and to grow in an anchorage-independent manner. HRG also promotes interaction of MTA1 with endogenous ER and association of MTA1 or HDAC with ERE-responsive target-gene promoters in vivo. These results identify ER-mediated transcription as a nuclear target of MTA1 and indicate that HDAC complexes associated with the MTA1 corepressor may mediate ER transcriptional repression by HRG.


Subject(s)
Breast Neoplasms/genetics , Cell Transformation, Neoplastic/genetics , Gene Expression Regulation, Neoplastic/physiology , Genes, Regulator/physiology , Neuregulin-1/genetics , Proteins/genetics , Receptors, Estrogen/genetics , Repressor Proteins , Transcription, Genetic/physiology , Acetylation/drug effects , Breast/drug effects , Breast/embryology , Breast/metabolism , Breast Neoplasms/metabolism , Breast Neoplasms/physiopathology , Cell Transformation, Neoplastic/drug effects , Female , Gene Expression Regulation, Neoplastic/drug effects , Genes, Regulator/drug effects , Histone Deacetylases/drug effects , Histone Deacetylases/metabolism , Histones/drug effects , Histones/metabolism , Humans , Neuregulin-1/metabolism , Neuregulin-1/pharmacology , Promoter Regions, Genetic/drug effects , Promoter Regions, Genetic/physiology , Proteins/drug effects , Proteins/metabolism , Receptors, Estrogen/drug effects , Receptors, Estrogen/metabolism , Trans-Activators , Transcription, Genetic/drug effects , Tumor Cells, Cultured/cytology , Tumor Cells, Cultured/drug effects , Tumor Cells, Cultured/metabolism
20.
J Biol Chem ; 276(8): 5636-42, 2001 Feb 23.
Article in English | MEDLINE | ID: mdl-11104760

ABSTRACT

The epidermal growth factor (EGF) family and its receptors regulate normal and cancerous epithelial cell proliferation, a process that could be suppressed by anti-receptor blocking antibodies. Polypeptide elongation factor-1alpha (EF-1alpha) is a multifunctional protein whose levels are positively correlated with the proliferative state of cells. To identify genes, whose expression may be modulated by anti-receptor blocking antibodies, we performed a differential display screening and isolated differentially expressed cDNAs. Isolates from one clone were 100% identical to human EF-1alpha. Both EGF and heregulin-beta1 (HRG) induced EF-1alpha promoter activity and mRNA and protein expression. Growth factor-mediated EF-1alpha expression was effectively blocked by pretreatment with humanized anti-EGF receptor antibody C225 or anti-human epidermal growth factor receptor-2 (HER2) antibody herceptin. Mutants and pharmacological inhibitors of p38(MAPK) and MEK, but not phosphatidylinositol 3-kinase, suppressed both constitutive and HRG-induced stimulation of EF-1alpha promoter activity in MCF-7 cells. Deletion analysis of the promoter suggested the requirement of the -393 to -204 region for growth factor-mediated transcription of EF-1alpha. Fine mapping and point mutation studies revealed a role of the SP1 site in the observed HRG-mediated regulation of the EF-1alpha promoter. In addition, we also provide new evidence to suggest that HRG stimulation of the EF-1alpha promoter involves increased physical interactions with acetylated histone H3 and histone H4. These results suggest that regulation of EF-1alpha expression by extracellular signals that function through human EGF receptor family members that are widely deregulated in human cancers and that growth factor regulation of EF-1alpha expression involve histone acetylation.


Subject(s)
Antibodies, Blocking/pharmacology , ErbB Receptors/metabolism , Neuregulin-1/metabolism , Peptide Elongation Factor 1/biosynthesis , Acetylation , Antibodies, Monoclonal/pharmacology , Antibodies, Monoclonal, Humanized , ErbB Receptors/immunology , Gene Expression Regulation , Histones/metabolism , Neuregulin-1/immunology , Promoter Regions, Genetic , Receptor Protein-Tyrosine Kinases/metabolism , Signal Transduction , Sp1 Transcription Factor/metabolism , Trastuzumab
SELECTION OF CITATIONS
SEARCH DETAIL
...