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










Publication year range
1.
Nat Commun ; 10(1): 1945, 2019 Apr 24.
Article in English | MEDLINE | ID: mdl-31019194

ABSTRACT

The original version of this Article contained an error in Fig. 4. In the left histogram of the right panel of Fig. 4d, several data points were inadvertently deleted from the histogram during the production process. This error has been corrected in both the PDF and HTML versions of the Article. The original, incorrect version of Fig. 4 is presented in the accompanying Publisher Correction.

2.
Nat Commun ; 10(1): 766, 2019 02 15.
Article in English | MEDLINE | ID: mdl-30770823

ABSTRACT

Primary triple negative breast cancers (TNBC) are prone to dissemination but sub-clonal relationships between tumors and resulting metastases are poorly understood. Here we use cellular barcoding of two treatment-naïve TNBC patient-derived xenografts (PDXs) to track the spatio-temporal fate of thousands of barcoded clones in primary tumors, and their metastases. Tumor resection had a major impact on reducing clonal diversity in secondary sites, indicating that most disseminated tumor cells lacked the capacity to 'seed', hence originated from 'shedders' that did not persist. The few clones that continued to grow after resection i.e. 'seeders', did not correlate in frequency with their parental clones in primary tumors. Cisplatin treatment of one BRCA1-mutated PDX model to non-palpable levels had a surprisingly minor impact on clonal diversity in the relapsed tumor yet purged 50% of distal clones. Therefore, clonal features of shedding, seeding and drug resistance are important factors to consider for the design of therapeutic strategies.


Subject(s)
Clone Cells , Triple Negative Breast Neoplasms/genetics , Animals , BRCA1 Protein/genetics , Cell Line, Tumor , Cisplatin/therapeutic use , Disease Models, Animal , Female , Gene Expression Regulation, Neoplastic/genetics , Humans , Mice , Mutation/genetics , Neoplasm Recurrence, Local/genetics , Triple Negative Breast Neoplasms/drug therapy , Xenograft Model Antitumor Assays
3.
Oncogene ; 35(15): 1877-87, 2016 Apr 14.
Article in English | MEDLINE | ID: mdl-26257067

ABSTRACT

The last three decades have seen significant progress in our understanding of the role of the pro-survival protein BCL-2 and its family members in apoptosis and cancer. BCL-2 and other pro-survival family members including Mcl-1 and BCL-XL have been shown to have a key role in keeping pro-apoptotic 'effector' proteins BAK and BAX in check. They also neutralize a group of 'sensor' proteins (such as BIM), which are triggered by cytotoxic stimuli such as chemotherapy. BCL-2 proteins therefore have a central role as guardians against apoptosis, helping cancer cells to evade cell death. More recently, an increasing number of BH3 mimetics, which bind and neutralize BCL-2 and/or its pro-survival relatives, have been developed. The utility of targeting BCL-2 in hematological malignancies has become evident in early-phase studies, with remarkable clinical responses seen in heavily pretreated patients. As BCL-2 is overexpressed in ~75% of breast cancer, there has been growing interest in determining whether this new class of drug could show similar promise in breast cancer. This review summarizes our current understanding of the role of BCL-2 and its family members in mammary gland development and breast cancer, recent progress in the development of new BH3 mimetics as well as their potential for targeting estrogen receptor-positive breast cancer.


Subject(s)
Breast Neoplasms/drug therapy , Carcinoma/drug therapy , Estrogens , Molecular Targeted Therapy , Neoplasm Proteins/antagonists & inhibitors , Neoplasms, Hormone-Dependent/drug therapy , Proto-Oncogene Proteins c-bcl-2/antagonists & inhibitors , Aniline Compounds/pharmacology , Aniline Compounds/therapeutic use , Animals , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Apoptosis/drug effects , Apoptosis Regulatory Proteins/physiology , Biomarkers, Tumor , Breast/growth & development , Breast/metabolism , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Bridged Bicyclo Compounds, Heterocyclic/pharmacology , Bridged Bicyclo Compounds, Heterocyclic/therapeutic use , Carcinoma/genetics , Carcinoma/pathology , Drug Design , Drug Synergism , Female , Gene Expression Regulation, Neoplastic , Genes, bcl-2 , Humans , Mice , Mitochondria/drug effects , Mitochondria/physiology , Multigene Family , Neoplasm Proteins/genetics , Neoplasms, Hormone-Dependent/genetics , Peptide Fragments/chemistry , Prognosis , Proto-Oncogene Proteins/chemistry , Proto-Oncogene Proteins c-bcl-2/biosynthesis , Proto-Oncogene Proteins c-bcl-2/physiology , Receptors, Estrogen/analysis , Signal Transduction/drug effects , Sulfonamides/pharmacology , Sulfonamides/therapeutic use , TOR Serine-Threonine Kinases/antagonists & inhibitors , Xenograft Model Antitumor Assays
4.
Oncogene ; 34(30): 3926-34, 2015 Jul 23.
Article in English | MEDLINE | ID: mdl-25263453

ABSTRACT

Evasion of cell death is fundamental to the development of cancer and its metastasis. The role of the BCL-2-mediated (intrinsic) apoptotic program in these processes remains poorly understood. Here we have investigated the relevance of the pro-apoptotic protein BIM to breast cancer progression using the MMTV-Polyoma middle-T (PyMT) transgenic model. BIM deficiency in PyMT females did not affect primary tumor growth, but substantially increased the survival of metastatic cells within the lung. These data reveal a role for BIM in the suppression of breast cancer metastasis. Intriguingly, we observed a striking correlation between the expression of BIM and the epithelial to mesenchymal transition transcription factor SNAI2 at the proliferative edge of the tumors. Overexpression and knockdown studies confirmed that these two genes were coordinately expressed, and chromatin immunoprecipitation analysis further revealed that Bim is a target of SNAI2. Taken together, our findings suggest that SNAI2-driven BIM-induced apoptosis may temper metastasis by governing the survival of disseminating breast tumor cells.


Subject(s)
Apoptosis Regulatory Proteins/genetics , Lung Neoplasms/metabolism , Mammary Neoplasms, Experimental/metabolism , Membrane Proteins/genetics , Proto-Oncogene Proteins/genetics , Transcription Factors/physiology , Animals , Apoptosis , Apoptosis Regulatory Proteins/metabolism , Bcl-2-Like Protein 11 , Cell Survival , Female , Gene Expression , Gene Expression Regulation, Neoplastic , Lung Neoplasms/secondary , Mammary Neoplasms, Experimental/pathology , Membrane Proteins/metabolism , Mice, Inbred C57BL , Mice, Knockout , Proto-Oncogene Proteins/metabolism , Snail Family Transcription Factors , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism
5.
J Mol Biol ; 425(7): 1101-10, 2013 Apr 12.
Article in English | MEDLINE | ID: mdl-23353824

ABSTRACT

LIM-only protein 4 (LMO4) is strongly linked to the progression of breast cancer. Although the mechanisms underlying this phenomenon are not well understood, a role is emerging for LMO4 in regulation of the cell cycle. We determined the solution structure of LMO4 in complex with CtIP (C-terminal binding protein interacting protein)/RBBP8, a tumour suppressor protein that is involved in cell cycle progression, DNA repair and transcriptional regulation. Our data reveal that CtIP and the essential LMO cofactor LDB1 (LIM-domain binding protein 1) bind to the same face on LMO4 and cannot simultaneously bind to LMO4. We hypothesise that overexpression of LMO4 may disrupt some of the normal tumour suppressor activities of CtIP, thereby contributing to breast cancer progression.


Subject(s)
Adaptor Proteins, Signal Transducing/chemistry , Carrier Proteins/chemistry , LIM Domain Proteins/chemistry , Nuclear Proteins/chemistry , Protein Structure, Tertiary , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Amino Acid Sequence , Binding Sites/genetics , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Carrier Proteins/genetics , Carrier Proteins/metabolism , Endodeoxyribonucleases , Female , Humans , LIM Domain Proteins/genetics , LIM Domain Proteins/metabolism , Magnetic Resonance Spectroscopy , Models, Molecular , Molecular Sequence Data , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Protein Binding , Sequence Homology, Amino Acid , Tumor Suppressor Proteins/chemistry , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism , Two-Hybrid System Techniques
7.
Oncogene ; 30(13): 1597-607, 2011 Mar 31.
Article in English | MEDLINE | ID: mdl-21132007

ABSTRACT

Disruption of the breast cancer susceptibility gene Brca1 results in defective lobular-alveolar development in the mammary gland and a predisposition to breast tumourigenesis in humans and in mice. Recent evidence suggests that BRCA1 loss in humans is associated with an expansion of the luminal progenitor cell compartment in the normal breast and tumours with a luminal progenitor-like expression profile. To further investigate the role of BRCA1 in the mammary gland, we examined the consequences of Brca1 loss in mouse mammary epithelial cells in vitro and in vivo. Here, we show that Brca1 loss is associated with defective morphogenesis of SCp2 and HC11 mouse mammary epithelial cell lines and that in the MMTV-Cre Brca1(Co/Co) mouse model of Brca1 loss, there is an accumulation of luminal progenitor (CD61(+)CD29(lo)CD24(+)) cells during pregnancy. By day 1 of lactation, there are marked differences in the expression of 1379 genes, with most significantly altered pathways and networks, including lactation, the immune response and cancer. One of the most differentially expressed genes was the luminal progenitor marker, c-kit. Immunohistochemical analysis revealed that the increase in c-kit levels is associated with an increase in c-kit positivity. Interestingly, an inverse association between Brca1 and c-kit expression was also observed during mammary epithelial differentiation, and small interfering RNA-mediated knockdown of Brca1 resulted in a significant increase in c-kit mRNA levels. We found no evidence that c-kit plays a direct role in regulating differentiation of HC11 cells, suggesting that Brca1-mediated induction of c-kit probably contributes to Brca1-associated tumourigenesis via another cellular process, and that c-kit is likely to be a marker rather than a mediator of defective lobular-alveolar development resulting from Brca1 loss.


Subject(s)
BRCA1 Protein/physiology , Breast Neoplasms/etiology , Mammary Glands, Animal/physiology , Proto-Oncogene Proteins c-kit/physiology , Animals , Cell Differentiation , Female , Gene Expression Profiling , Lactation , Mammary Glands, Animal/cytology , Mice , Pregnancy , Proto-Oncogene Proteins c-kit/analysis , Stem Cells/physiology
8.
Oncogene ; 28(41): 3608-18, 2009 Oct 15.
Article in English | MEDLINE | ID: mdl-19648968

ABSTRACT

ErbB2/HER2/Neu-overexpressing breast cancers are characterized by poor survival due to high proliferation and metastasis rates and identifying downstream targets of ErbB2 should facilitate developing novel therapies for this disease. Gene expression profiling revealed the transcriptional regulator LIM-only protein 4 (LMO4) is upregulated during ErbB2-induced mouse mammary gland tumorigenesis. Although LMO4 is frequently overexpressed in breast cancer and LMO4-overexpressing mice develop mammary epithelial tumors, the mechanisms involved are unknown. In this study, we report that LMO4 is a downstream target of ErbB2 and PI3K in ErbB2-dependent breast cancer cells. Furthermore, LMO4 silencing reduces proliferation of these cells, inducing a G2/M arrest that was associated with decreased cullin-3, an E3-ubiquitin ligase component important for mitosis. Loss of LMO4 subsequently results in reduced Cyclin D1 and Cyclin E. Further supporting a role for LMO4 in modulating proliferation by regulating cullin-3 expression, we found that LMO4 expression oscillates throughout the cell cycle with maximum expression occurring during G2/M and these changes precede oscillations in cullin-3 levels. LMO4 levels are also highest in high-grade/less differentiated breast cancers, which are characteristically highly proliferative. We conclude that LMO4 is a novel cell cycle regulator with a key role in mediating ErbB2-induced proliferation, a hallmark of ErbB2-positive disease.


Subject(s)
Breast Neoplasms/pathology , Cell Cycle , Homeodomain Proteins/metabolism , Receptor, ErbB-2/metabolism , Transcription Factors/metabolism , Adaptor Proteins, Signal Transducing , Animals , Breast Neoplasms/genetics , Cell Cycle/genetics , Cell Division/genetics , Cell Line, Tumor , Cell Proliferation , Cullin Proteins/metabolism , Cyclin D1/genetics , Cyclin D1/metabolism , G2 Phase/genetics , Gene Expression Regulation, Neoplastic , Homeodomain Proteins/genetics , Humans , LIM Domain Proteins , Mice , Neuregulin-1/metabolism , Phosphatidylinositol 3-Kinases/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Signal Transduction , Transcription Factors/genetics , Up-Regulation
9.
Article in English | MEDLINE | ID: mdl-19022771

ABSTRACT

Reconstitution assays have shown that mouse mammary stem cells reside within the mature mammary gland in vivo. Single cells could be prospectively isolated and shown to regenerate an entire mammary gland that exhibited full developmental capacity. The more recent identification of luminal progenitor populations has indicated that the mammary epithelium is organized in a hierarchical manner. Further definition of epithelial cell types in both mouse and human mammary glands will provide insight into the "cells of origin" in the different subtypes of breast cancer, as well as the nature of cancer-propagating cells. Here, we review the known characteristics of mammary stem and progenitor cells, their steroid receptor status, and the pathways that have thus far been implicated in regulating their self-renewal and differentiation.


Subject(s)
Mammary Glands, Animal/cytology , Stem Cells/cytology , Animals , Antigens, CD/metabolism , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Differentiation , Cell Proliferation , Cell Separation , Epithelial Cells/cytology , Epithelial Cells/metabolism , Estrogen Receptor alpha/metabolism , Female , Humans , Mammary Glands, Animal/metabolism , Mammary Glands, Human/cytology , Mammary Glands, Human/metabolism , Mice , Models, Biological , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Receptors, Progesterone/metabolism , Stem Cells/metabolism
10.
Br J Cancer ; 98(3): 537-41, 2008 Feb 12.
Article in English | MEDLINE | ID: mdl-18231110

ABSTRACT

Identification of a biomarker of prognosis and response to therapy that can be assessed preoperatively would significantly improve overall outcomes for patients with pancreatic cancer. In this study, patients whose tumours exhibited high LMO4 expression had a significant survival advantage following operative resection, whereas the survival of those patients whose tumours had low or no LMO4 expression was not significantly different when resection was compared with operative biopsy alone.


Subject(s)
Adenocarcinoma/metabolism , Carcinoma, Pancreatic Ductal/metabolism , Homeodomain Proteins/metabolism , Pancreatic Neoplasms/metabolism , Transcription Factors/metabolism , Adaptor Proteins, Signal Transducing , Adenocarcinoma/surgery , Adult , Aged , Aged, 80 and over , Biomarkers, Tumor/analysis , Carcinoma, Pancreatic Ductal/surgery , Cohort Studies , Female , Humans , LIM Domain Proteins , Male , Middle Aged , Pancreatectomy , Pancreatic Neoplasms/surgery , Prognosis , Survival Analysis , Treatment Outcome
11.
J Med Genet ; 43(1): 74-83, 2006 Jan.
Article in English | MEDLINE | ID: mdl-15923272

ABSTRACT

BACKGROUND: The vast majority of BRCA1 missense sequence variants remain uncharacterized for their possible effect on protein expression and function, and therefore are unclassified in terms of their pathogenicity. BRCA1 plays diverse cellular roles and it is unlikely that any single functional assay will accurately reflect the total cellular implications of missense mutations in this gene. OBJECTIVE: To elucidate the effect of two BRCA1 variants, 5236G>C (G1706A) and 5242C>A (A1708E) on BRCA1 function, and to survey the relative usefulness of several assays to direct the characterisation of other unclassified variants in BRCA genes. METHODS AND RESULTS: Data from a range of bioinformatic, genetic, and histopathological analyses, and in vitro functional assays indicated that the 1708E variant was associated with the disruption of different cellular functions of BRCA1. In transient transfection experiments in T47D and 293T cells, the 1708E product was mislocalised to the cytoplasm and induced centrosome amplification in 293T cells. The 1708E variant also failed to transactivate transcription of reporter constructs in mammalian transcriptional transactivation assays. In contrast, the 1706A variant displayed a phenotype comparable to wildtype BRCA1 in these assays. Consistent with functional data, tumours from 1708E carriers showed typical BRCA1 pathology, while tumour material from 1706A carriers displayed few histopathological features associated with BRCA1 related tumours. CONCLUSIONS: A comprehensive range of genetic, bioinformatic, and functional analyses have been combined for the characterisation of BRCA1 unclassified sequence variants. Consistent with the functional analyses, the combined odds of causality calculated for the 1706A variant after multifactorial likelihood analysis (1:142) indicates a definitive classification of this variant as "benign". In contrast, functional assays of the 1708E variant indicate that it is pathogenic, possibly through subcellular mislocalisation. However, the combined odds of 262:1 in favour of causality of this variant does not meet the minimal ratio of 1000:1 for classification as pathogenic, and A1708E remains formally designated as unclassified. Our findings highlight the importance of comprehensive genetic information, together with detailed functional analysis for the definitive categorisation of unclassified sequence variants. This combination of analyses may have direct application to the characterisation of other unclassified variants in BRCA1 and BRCA2.


Subject(s)
BRCA1 Protein/chemistry , BRCA1 Protein/genetics , Mutation, Missense/genetics , Adult , Aged , Australia , Breast Neoplasms/pathology , Centrosome/metabolism , Female , Genes, Reporter/genetics , Humans , Loss of Heterozygosity/genetics , Middle Aged , Models, Molecular , Protein Processing, Post-Translational , Protein Transport , RNA Splicing/genetics , RNA Stability/genetics , Transcription, Genetic , Transcriptional Activation/genetics
13.
Cell Death Differ ; 10(2): 185-92, 2003 Feb.
Article in English | MEDLINE | ID: mdl-12700646

ABSTRACT

Proteins of the Bcl-2 family are critical regulators of apoptosis. Proapoptotic members, like Bax, contain three of the four Bcl-2 homology regions (BH1-3), while BH3-only proteins, like Bim, possess only the short BH3 motif. Database searches revealed Bfk, an unusual novel member of the Bcl-2 family that contains a BH2 and BH3 region but not BH1 or BH4. Bfk is thus most closely related to Bcl-G(L). It lacks a C-terminal membrane anchor and is cytosolic. Enforced expression of Bfk weakly promoted apoptosis and antagonized Bcl-2's prosurvival function. Like Bcl-G(L), Bfk did not bind to any Bcl-2 family members, even though its BH3 motif can mediate association with prosurvival proteins. Low amounts of Bfk were found in stomach, ovary, bone marrow and spleen, but its level in the mammary gland rose markedly during pregnancy, suggesting that Bfk may play a role in mammary development.


Subject(s)
Apoptosis , Proto-Oncogene Proteins c-bcl-2/chemistry , Amino Acid Sequence , Animals , Conserved Sequence , Cytosol/chemistry , Female , Humans , Male , Mice , Molecular Sequence Data , NIH 3T3 Cells , Pregnancy , Sequence Homology, Amino Acid , Tissue Distribution
14.
Proc Natl Acad Sci U S A ; 98(25): 14452-7, 2001 Dec 04.
Article in English | MEDLINE | ID: mdl-11734645

ABSTRACT

LMO4 belongs to a family of LIM-only transcriptional regulators, the first two members of which are oncoproteins in acute T cell leukemia. We have explored a role for LMO4, initially described as a human breast tumor autoantigen, in developing mammary epithelium and breast oncogenesis. Lmo4 was expressed predominantly in the lobuloalveoli of the mammary gland during pregnancy. Consistent with a role in proliferation, forced expression of this gene inhibited differentiation of mammary epithelial cells. Overexpression of LMO4 mRNA was observed in 5 of 10 human breast cancer cell lines. Moreover, in situ hybridization analysis of 177 primary invasive breast carcinomas revealed overexpression of LMO4 in 56% of specimens. Immunohistochemistry confirmed overexpression in a high percentage (62%) of tumors. These studies imply a role for LMO4 in maintaining proliferation of mammary epithelium and suggest that deregulation of this gene may contribute to breast tumorigenesis.


Subject(s)
Breast Neoplasms/genetics , Homeodomain Proteins/genetics , Mammary Glands, Animal/cytology , Transcription Factors/genetics , Adaptor Proteins, Signal Transducing , Animals , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Caseins/biosynthesis , Cell Differentiation/genetics , DNA-Binding Proteins/genetics , Epithelial Cells/cytology , Epithelial Cells/metabolism , Female , Gene Expression Regulation, Developmental , Humans , In Situ Hybridization , LIM Domain Proteins , Mammary Glands, Animal/growth & development , Mammary Glands, Animal/metabolism , Mice , Pregnancy , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Neoplasm/genetics , RNA, Neoplasm/metabolism , Tumor Cells, Cultured
15.
Protein Eng ; 14(7): 493-9, 2001 Jul.
Article in English | MEDLINE | ID: mdl-11522923

ABSTRACT

The nuclear LIM-only (LMO) transcription factors LMO2 and LMO4 play important roles in both normal and leukemic T-cell development. LIM domains are cysteine/histidine-rich domains that contain two structural zinc ions and that function as protein-protein adaptors; members of the LMO family each contain two closely spaced LIM domains. These LMO proteins all bind with high affinity to the nuclear protein LIM domain binding protein 1 (ldb1). The LMO-ldb1 interaction is mediated through the N-terminal LIM domain (LIM1) of LMO proteins and a 38-residue region towards the C-terminus of ldb1 [ldb1(LID)]. Unfortunately, recombinant forms of LMO2 and LMO4 have limited solubility and stability, effectively preventing structural analysis. Therefore, we have designed and constructed a fusion protein in which ldb1(LID) and LIM1 of LMO2 can form an intramolecular complex. The engineered protein, FLIN2 (fusion of the LIM interacting domain of ldb1 and the N-terminal LIM domain of LMO2) has been expressed and purified in milligram quantities. FLIN2 is monomeric, contains significant levels of secondary structure and yields a sharp and well-dispersed one-dimensional (1)H NMR spectrum. The analogous LMO4 protein, FLIN4, has almost identical properties. These data suggest that we will be able to obtain high-resolution structural information about the LMO-ldb1 interactions.


Subject(s)
DNA-Binding Proteins/chemistry , Homeodomain Proteins/chemistry , Metalloproteins/chemistry , Protein Engineering , Transcription Factors/chemistry , Adaptor Proteins, Signal Transducing , Amino Acid Sequence , Animals , Binding Sites , Cloning, Molecular , DNA-Binding Proteins/isolation & purification , Escherichia coli/genetics , Homeodomain Proteins/isolation & purification , LIM Domain Proteins , Magnetic Resonance Spectroscopy , Metalloproteins/isolation & purification , Mice , Nuclear Proteins/chemistry , Protein Binding , Protein Folding , Protein Structure, Secondary , Recombinant Fusion Proteins , Transcription Factors/isolation & purification , Zinc/chemistry
16.
Genes Dev ; 15(13): 1631-6, 2001 Jul 01.
Article in English | MEDLINE | ID: mdl-11445538

ABSTRACT

Prolactin is essential for proliferation and differentiation of the developing mammary gland. We have explored a role for Suppressor of Cytokine Signaling 1 (SOCS1) as a modulator of the prolactin response using mice deficient in SOCS1, which were rescued from neonatal death by deletion of the Interferon gamma (IFN gamma) gene. SOCS1(-/-)/IFN gamma(-/-) mice exhibited accelerated lobuloalveolar development in the mammary gland during late pregnancy and precocious lactation. Significantly, the lactogenic defect in prolactin receptor heterozygous females could be rescued by deletion of a single SOCS1 allele. These findings establish a role for SOCS1 as a negative regulator of prolactin signaling and suggest that SOCS1 is required for the prevention of lactation prior to parturition.


Subject(s)
Breast/growth & development , Carrier Proteins/physiology , Lactation/physiology , Receptors, Prolactin/physiology , Repressor Proteins , Alleles , Animals , Carrier Proteins/genetics , Caseins/biosynthesis , Cell Line , Female , Gene Expression , Male , Mice , Mice, Knockout , Milk/metabolism , Pregnancy , Receptors, Prolactin/genetics , Suppressor of Cytokine Signaling 1 Protein , Suppressor of Cytokine Signaling Proteins
18.
J Biol Chem ; 276(1): 522-8, 2001 Jan 05.
Article in English | MEDLINE | ID: mdl-11016926

ABSTRACT

Multiple studies have shown that intracellular signal transduction by the protein kinase C (PKC) family participates in the initiation of megakaryocyte differentiation. In this study, multiple approaches addressed the functional contributions by specific PKC isozymes to megakaryocytic lineage commitment of two independent cell lines, K562 and human erythroleukemia (HEL). Pharmacologic profiles of induction and inhibition of megakaryocytic differentiation in both cell lines suggested a role for the calcium-independent novel PKCs, in particular PKC-epsilon. In transfection studies, the isolated variable domain of PKC-epsilon selectively blocked exogenous activation of the megakaryocyte-specific alpha IIb promoter. Constitutively active mutants of PKC-epsilon, but not of other PKC isozymes, cooperated with the transcription factor GATA-1 in the activation of the alpha IIb promoter. The functional cooperation between GATA-1 and PKC-epsilon displayed dependence on cellular milieu, as well as on the promoter context of GATA binding sites. In aggregate, the data suggest that PKC-epsilon specifically participates in megakaryocytic lineage commitment through functional cooperation with GATA-1 in the activation of megakaryocytic promoters.


Subject(s)
Cell Differentiation , Isoenzymes/metabolism , Megakaryocytes/cytology , Megakaryocytes/enzymology , Protein Kinase C/metabolism , Carbazoles/pharmacology , Cell Differentiation/drug effects , Cell Lineage/drug effects , Culture Media, Conditioned/pharmacology , DNA-Binding Proteins/metabolism , Diterpenes/pharmacology , Erythroid-Specific DNA-Binding Factors , Fluorescent Antibody Technique , GATA1 Transcription Factor , Gene Expression Regulation/drug effects , Genes, Reporter , Humans , Indoles/pharmacology , Isoenzymes/antagonists & inhibitors , Isoenzymes/chemistry , Isoenzymes/genetics , K562 Cells , Maleimides/pharmacology , Megakaryocytes/drug effects , Megakaryocytes/metabolism , Mutation , Promoter Regions, Genetic/genetics , Protein Kinase C/antagonists & inhibitors , Protein Kinase C/chemistry , Protein Kinase C/genetics , Protein Kinase C-epsilon , Protein Transport/drug effects , Response Elements/genetics , Signal Transduction/drug effects , Transcription Factors/metabolism , Transfection , Tumor Cells, Cultured
19.
Am J Pathol ; 157(2): 449-61, 2000 Aug.
Article in English | MEDLINE | ID: mdl-10934149

ABSTRACT

Proapoptotic Bcl-2 family members activate cell death by neutralizing their anti-apoptotic relatives, which in turn maintain cell viability by regulating the activation of the cell death effectors, the caspases. Bim belongs to a distinct subgroup of proapoptotic proteins that only resemble other Bcl-2 family members within the short BH3 domain. Gene targeting experiments in mice have shown that Bim is essential for the execution of some but not all apoptotic stimuli, for hematopoietic cell homeostasis, and as a barrier against autoimmunity. There are three Bim isoforms, Bim(S), Bim(L), and Bim(EL), which have different proapoptotic potencies due at least in part to differences in interaction with the dynein motor complex. The expression pattern of Bim was investigated by immunohistochemical staining, immunoprecipitation followed by Western blotting, and in situ hybridization. Bim was found in hematopoietic, epithelial, neuronal, and germ cells. Bim(L) and Bim(EL) were coexpressed at similar levels in many cell types, but Bim(S) was not detected. Microscopic examination revealed a punctate pattern of Bim(L) and Bim(EL) immunostaining, indicating association with cytoplasmic structures. These results are discussed in the context of the phenotype of Bim-deficient mice and the post-translational regulation of Bim's pro-apoptotic activity.


Subject(s)
Carrier Proteins/metabolism , Epithelial Cells/metabolism , Germ Cells/metabolism , Hematopoietic Stem Cells/metabolism , Membrane Proteins , Neurons/metabolism , Proto-Oncogene Proteins , 3T3 Cells , Animals , Apoptosis , Apoptosis Regulatory Proteins , Bcl-2-Like Protein 11 , Blotting, Western , Cardiovascular System/metabolism , Carrier Proteins/genetics , Cell Line , Central Nervous System/metabolism , Digestive System/metabolism , Endocrine System/metabolism , Epithelial Cells/cytology , Female , Genitalia/metabolism , Germ Cells/cytology , HeLa Cells , Hematopoietic Stem Cells/cytology , Humans , Immunohistochemistry , In Situ Hybridization , Kidney/metabolism , Male , Mice , Mice, Inbred C57BL , Muscle, Skeletal/metabolism , Neurons/cytology , Rats , Rats, Wistar , Respiratory System/metabolism , Skin/metabolism , Tissue Distribution , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL
...