Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 48
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Med J Malaysia ; 75(2): 152-157, 2020 03.
Article in English | MEDLINE | ID: mdl-32281597

ABSTRACT

OBJECTIVES: To explore the epidemiological and histopathological patterns of glomerular diseases in Sabah. METHODS: A state-wide cross-sectional study was conducted. There were 336 native renal biopsies in 296 eligible patients from 1st January 2013 to 30th June 2016. All patients aged ≥12 years with sufficient sampling (≥8 glomeruli) for histopathological assessment were included. Graft kidney biopsies, protocol-based biopsies and patients with uncertain demographics were excluded. Demographics of patients, clinical data, laboratory parameters prior to biopsy, and histology findings of renal biopsies were collected from local unit database and recorded into a standardised data collection form. Descriptive statistical analyses were employed and factors associated with Lupus nephritis (LN) were explored using logistic regression. RESULTS: The mean age during biopsy was 34.53 years (Standard Deviation 0.759). Primary glomerulonephritis (PGN) accounted for 42.6% (126) of all native renal biopsies. The commonest cause of PGN was minimal change disease (38.9%, 49) followed by focal segmental glomerulosclerosis (33.3%, 42) and IgA nephropathy (14.3%, 18). LN is the leading cause for secondary glomerulonephritis (SGN) (87.2%, 136). Younger age (Odds Ratio, OR 0.978; 95% Confidence Interval, 95%CI 0.960, 0.996); female gender (OR 17.53; p<0.001); significant proteinuria (OR 132.0; p<0.001); creatinine level at biopsy (OR 11.26; p=0.004); positive antinuclear antibody (ANA) (OR 46.7; p<0.001); and ANA patterns (OR 8.038; p=0.018) were significant in predicting the odds of having LN. CONCLUSION: This is the first epidemiology study of glomerular diseases in Sabah. The predominance of LN suggests lower threshold for renal biopsy in patients with suspected glomerular disorders. We have identified significant predictors for early detection and treatment of LN.


Subject(s)
Biopsy , Kidney Diseases/diagnosis , Kidney Diseases/epidemiology , Adolescent , Adult , Child , Cross-Sectional Studies , Databases, Factual , Female , Glomerulonephritis/diagnosis , Glomerulonephritis/epidemiology , Humans , Logistic Models , Malaysia/epidemiology , Male , Middle Aged , Retrospective Studies , Young Adult
2.
Ann Oncol ; 30(1): 115-123, 2019 01 01.
Article in English | MEDLINE | ID: mdl-30423024

ABSTRACT

Background: Invasive lobular carcinoma (ILC) as a disease entity distinct from invasive ductal carcinoma (IDC) has merited focused studies of the genomic landscape, but those to date are largely limited to the assessment of early-stage cancers. Given that genomic alterations develop as acquired resistance to endocrine therapy, studies on refractory ILC are needed. Patients and methods: Tissue from 336 primary-enriched, breast-biopsied ILC and 485 estrogen receptor (ER)-positive IDC and metastatic biopsy specimens from 180 ILC and 191 ER-positive IDC patients was assayed with hybrid-capture-based comprehensive genomic profiling for short variant, indel, copy number variants, and rearrangements in up to 395 cancer-related genes. Results: Whereas ESR1 alterations are enriched in the metastases of both ILC and IDC compared with breast specimens, NF1 alterations are enriched only in ILC metastases (mILC). NF1 alterations are predominantly under loss of heterozygosity (11/14, 79%), are mutually exclusive with ESR1 mutations [odds ratio = 0.24, P < 0.027] and are frequently polyclonal in ctDNA assays. Assessment of paired specimens shows that NF1 alterations arise in the setting of acquired resistance. An in vitro model of CDH1 mutated ER-positive breast cancer demonstrates that NF1 knockdown confers a growth advantage in the presence of 4-hydroxy tamoxifen. Our study further identified a significant increase in tumor mutational burden (TMB) in mILCs relative to breast ILCs or metastatic IDCs (8.9% >20 mutations/mb; P < 0.001). Most TMB-high mILCs harbor an APOBEC trinucleotide signature (14/16; 88%). Conclusions: This study identifies alteration of NF1 as enriched specifically in mILC. Mutual exclusivity with ESR1 alterations, polyclonality in relapsed ctDNA, and de novo acquisition suggest a role for NF1 loss in endocrine therapy resistance. Since NF1 loss leads to RAS/RAF kinase activation, patients may benefit from a matched inhibitor. Moreover, for an independent subset of mILC, TMB was elevated relative to breast ILC, suggesting possible benefit from immune checkpoint inhibitors.


Subject(s)
Biomarkers, Tumor/genetics , Breast Neoplasms/pathology , Carcinoma, Ductal, Breast/secondary , Carcinoma, Lobular/secondary , Drug Resistance, Neoplasm/genetics , Neurofibromin 1/genetics , Adult , Aged , Aged, 80 and over , Antineoplastic Agents, Hormonal/therapeutic use , Breast Neoplasms/drug therapy , Breast Neoplasms/genetics , Carcinoma, Ductal, Breast/drug therapy , Carcinoma, Ductal, Breast/genetics , Carcinoma, Lobular/drug therapy , Carcinoma, Lobular/genetics , Female , Follow-Up Studies , Humans , Middle Aged , Neoplasm Metastasis , Prognosis , Receptor, ErbB-2/metabolism , Receptors, Estrogen/metabolism , Receptors, Progesterone/metabolism
3.
Ann Oncol ; 29(4): 872-880, 2018 04 01.
Article in English | MEDLINE | ID: mdl-29360925

ABSTRACT

Background: Estrogen receptor-positive (ER-positive) metastatic breast cancer is often intractable due to endocrine therapy resistance. Although ESR1 promoter switching events have been associated with endocrine-therapy resistance, recurrent ESR1 fusion proteins have yet to be identified in advanced breast cancer. Patients and methods: To identify genomic structural rearrangements (REs) including gene fusions in acquired resistance, we undertook a multimodal sequencing effort in three breast cancer patient cohorts: (i) mate-pair and/or RNAseq in 6 patient-matched primary-metastatic tumors and 51 metastases, (ii) high coverage (>500×) comprehensive genomic profiling of 287-395 cancer-related genes across 9542 solid tumors (5216 from metastatic disease), and (iii) ultra-high coverage (>5000×) genomic profiling of 62 cancer-related genes in 254 ctDNA samples. In addition to traditional gene fusion detection methods (i.e. discordant reads, split reads), ESR1 REs were detected from targeted sequencing data by applying a novel algorithm (copyshift) that identifies major copy number shifts at rearrangement hotspots. Results: We identify 88 ESR1 REs across 83 unique patients with direct confirmation of 9 ESR1 fusion proteins (including 2 via immunoblot). ESR1 REs are highly enriched in ER-positive, metastatic disease and co-occur with known ESR1 missense alterations, suggestive of polyclonal resistance. Importantly, all fusions result from a breakpoint in or near ESR1 intron 6 and therefore lack an intact ligand binding domain (LBD). In vitro characterization of three fusions reveals ligand-independence and hyperactivity dependent upon the 3' partner gene. Our lower-bound estimate of ESR1 fusions is at least 1% of metastatic solid breast cancers, the prevalence in ctDNA is at least 10× enriched. We postulate this enrichment may represent secondary resistance to more aggressive endocrine therapies applied to patients with ESR1 LBD missense alterations. Conclusions: Collectively, these data indicate that N-terminal ESR1 fusions involving exons 6-7 are a recurrent driver of endocrine therapy resistance and are impervious to ER-targeted therapies.


Subject(s)
Antineoplastic Agents, Hormonal/therapeutic use , Breast Neoplasms/drug therapy , Drug Resistance, Neoplasm/genetics , Estrogen Receptor alpha/metabolism , Recombinant Fusion Proteins/metabolism , Breast Neoplasms/pathology , Estrogen Receptor alpha/genetics , Female , High-Throughput Nucleotide Sequencing , Humans , Mutation , Neoplasm Metastasis , Recombinant Fusion Proteins/genetics
7.
Brain Behav Immun Health ; 4: 100058, 2020 Apr.
Article in English | MEDLINE | ID: mdl-34589844

ABSTRACT

Offspring adrenal function may be negatively affected in utero by maternal stressors such as microbial infection. Maternal supplementation with immunomodulatory compounds such as omega-3 polyunsaturated fatty acids (n-3 PUFA) may help minimize the adverse effects of maternal stress on fetal hypothalamic-pituitary-adrenal development and improve offspring health. Presently, n-3 PUFA sources are primarily fish-based, but n-3 PUFA microalgae (AL) may be an alternative. Previously, it was determined that maternal AL or fish oil (FO) supplementation to sows, in addition to maternal stress induced by Escherichia coli lipopolysaccharide (LPS) challenge appeared to have a greater influence on the stress response of male offspring compared to females. To further elaborate on these findings, this study assessed the effects of maternal AL or FO supplementation combined with a maternal LPS challenge on adrenal gene expression in male offspring fed a nursery diet containing low-quality protein sources. Forty-eight sows were fed gestation diets starting on gestation day (gd) 75 containing either 3.12% AL, 3.1% FO, or a control diet containing 1.89% corn oil. On gd 112, half the sows in each treatment were administered 10 â€‹µg/kg LPS i.m. Piglets were weaned at 21 days of age onto a common low-quality plant-based protein diet, and one week after weaning, four piglets per sow were administered 40 â€‹µg/kg LPS i.m. Two hours later, the piglets were euthanized to obtain adrenal tissue, and total RNA was extracted to carry out transcriptome analysis using the Affymetrix GeneChip WT Plus assay and subsequent validation by real-time PCR. Analysis revealed that adrenal steroidogenesis, fatty acid metabolism and immune function were significantly influenced by maternal diet and stress. Increased expression of immune-related genes including lymphocyte antigen 96, TLR-2 and NF-κB suggests that maternal AL supplementation may increase offspring sensitivity to inflammation after weaning. Decreased expression of lymphocyte antigen 96 in male offspring from sows receiving maternal LPS challenge also suggests a possible role of maternal stress in diminishing the offspring immune response to immune stress challenge. Increased expression of the genes encoding the 11BHSD2 enzyme in offspring from sows fed FO may also reduce the magnitude of the stress response. These data provide insight to the immune and metabolic mechanisms that may be influenced by maternal diet and stress.

9.
Vet Immunol Immunopathol ; 218: 109937, 2019 Dec.
Article in English | MEDLINE | ID: mdl-31522084

ABSTRACT

Maternal stress, such as a bacterial infection occurring in late gestation, may predispose offspring to a variety of diseases later in life. It may also alter programming of developing systems within the fetus, such as the hypothalamic-pituitary-adrenal (HPA) axis and immune system. Dietary supplementation during the last trimester of pregnancy with immune-modulating compounds may be a means of reducing potential adverse effects of maternal stress on the developing fetus. Essential omega-3 polyunsaturated fatty acids (n-3 PUFA) such as docosahexanoic acid (DHA) and eicosapentanoic acid are well-known for their immune-modulating and anti-inflammatory properties. Sources of these n-3 PUFA include fish products such as fish oil and microalgae, which may be a suitable alternative to fish-based products. The aim of this study was to determine the effect of supplementing gestating sow diets with n-3 PUFA and inducing an immune stress challenge in late gestation on piglet growth and immune responsiveness when placed on either a high- or low-quality protein diet after weaning. Forty-eight sows were fed gestation diets containing either 3.12% microalgae, 3.1% fish oil or a corn oil control diet containing 1.89% corn oil starting on gestation day (gd) 75. On gd112, half the sows in each treatment were immune stress challenged with bacterial lipopolysaccharide (LPS) endotoxin (10 µg/kg administered i.m). After farrowing, piglet BW gain was monitored weekly during lactation and pigs were weaned at 21 days of age. One week after weaning, four piglets per sow were immune stress challenged with LPS (40 µg/kg administered i.m.). At the same time, four piglets per sow were vaccinated with the novel antigens chicken ovalbumin (OVA) and Candida cellular antigen (CAA) and received booster vaccinations two weeks later. Four weeks after the initial vaccination, a transdermal hypersensitivity immune challenge was performed using the same antigens. Blood samples were also collected to quantify IgG antibody responses to both antigens. PUFA enrichment in sow blood and piglet brain was detected after sows were on feed for 40 days. Piglet growth was increased in pigs fed a high-quality diet in nursery phase 1. Concentrations of the cytokines IL-1ra, IL-6 and IL-10 were elevated in pigs fed a high-quality protein diet following LPS immune challenge. Overall, it appears that in the current study piglet nursery diet quality was more important for determining piglet health and growth than maternal diet and immune stress.


Subject(s)
Animal Feed/analysis , Diet/veterinary , Dietary Proteins/administration & dosage , Dietary Supplements/analysis , Microalgae , Sus scrofa/immunology , Age Factors , Animals , Dietary Proteins/analysis , Female , Fish Oils/administration & dosage , Male , Milk/chemistry , Pregnancy , Weaning
10.
Mol Cell Biol ; 20(5): 1489-96, 2000 Mar.
Article in English | MEDLINE | ID: mdl-10669726

ABSTRACT

Insulin receptor substrate 1 (IRS-1) is a critical adapter protein involved in both insulin and insulin-like growth factor (IGF) signaling. Due to the fact that alteration of IRS-1 levels can affect the sensitivity and response to both insulin and IGF-I, we examined the ability of each of these ligands to affect IRS-1 expression. IGF-I (10 nM) stimulation of MCF-7 breast cancer cells caused a transient tyrosine phosphorylation of IRS-1 that was maximal at 15 min and decreased thereafter. The decrease in tyrosine phosphorylation of IRS-1 was paralleled by an apparent decrease in IRS-1 levels. The IGF-mediated decrease in IRS-1 expression was posttranscriptional and due to a decrease in the half-life of the IRS-1 protein. Insulin (10 nM) caused tyrosine phosphorylation of IRS-1 but not degradation, whereas high concentrations of insulin (10 microM) resulted in degradation of IRS-1. IGF-I (10 nM) stimulation resulted in transient IRS-1 phosphorylation and extracellular signal-related kinase (ERK) activation. In contrast, insulin (10 nM) caused sustained IRS-1 phosphorylation and ERK activation. Inhibition of 26S proteasome activity by the use of lactacystin or MG132 completely blocked IGF-mediated degradation of IRS-1. Furthermore, coimmunoprecipitation experiments showed an association between ubiquitin and IRS-1 that was increased by treatment of cells with IGF-I. Finally, IGF-mediated degradation of IRS-1 was blocked by inhibition of phosphatidylinositol 3'-kinase activity but was not affected by inhibition of ERK, suggesting that this may represent a direct negative-feedback mechanism resulting from downstream IRS-1 signaling. We conclude that IGF-I can cause ligand-mediated degradation of IRS-1 via the ubiquitin-mediated 26S proteasome and a phosphatidylinositol 3'-kinase-dependent mechanism and that control of degradation may have profound effects on downstream activation of signaling pathways.


Subject(s)
Insulin-Like Growth Factor I/metabolism , Peptide Hydrolases/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Phosphoproteins/metabolism , Proteasome Endopeptidase Complex , Signal Transduction , Animals , Cell Line , Enzyme Inhibitors/pharmacology , Insulin Receptor Substrate Proteins , Phosphoinositide-3 Kinase Inhibitors , Phosphorylation , Signal Transduction/drug effects
11.
J Natl Cancer Inst ; 88(9): 601-6, 1996 May 01.
Article in English | MEDLINE | ID: mdl-8609661

ABSTRACT

BACKGROUND: The insulin-like growth factor (IGF)-binding proteins (IGFBPs) regulate the actions of the IGFs by influencing interactions between the IGFs and the IGF receptors. IGFBP-3, one of the six known species of IGFBPs, is the predominant IGFBP in serum and is expressed by breast cancer cells. Compared with estrogen receptor (ER)-positive samples, ER-negative breast cancer cell lines and tumors express higher levels of IGFBP-3. Therefore, expression of IGFBP-3 may be relevant in breast cancer biology, although it is unknown whether IGFBP-3 levels correlate with other breast cancer prognostic factors besides ER status. It is also not known how different methods used to measure IGFBP-3 in breast cancer correlate. PURPOSE: We measured IGFBP-3 messenger RNA (mRNA) and protein levels in breast tumors by different methods to test how these methods compare and to investigate the relationship between IGFBP-3 and breast cancer prognostic factors. METHODS: We analyzed 40 human breast tumors and examined IGFBP-3 expression by ligand blot analysis, immunoblot analysis, immunoradiometric assay (IRMA), and ribonuclease protection assay. Another set of 40 breast tumors, selected according to ER and progesterone receptor (PR) status, S phase, and ploidy, was analyzed by IRMA. RESULTS: In 26 (65%) of 40 samples in which RNA could be isolated, IGFBP-3 mRNA levels correlated with IGFBP-3 levels measured by IRMA (two-sided; P = .0001) but not with IGFBP-3 levels measured by ligand blot or immunoblot. Protein levels were highly correlated among all protein assays. Because the IRMA was more sensitive and accurate than the ligand blot and immunoblot assays, we used IRMA to examine IGFBP-3 levels in an additional 20 primary breast tumors with poor prognostic features (ER and PR negativity, high S phase, and aneuploidy) and in 20 tumors with good prognostic factors (opposite features). IGFBP-3 levels were threefold higher in tumors with poor prognostic features (mean +/- standard deviation = 32.8 +/- 25.2 versus 11.8 +/- 9.7 ng/mg; two-sided; P = .003). CONCLUSIONS: These findings suggest that in human breast cancer, IGFBP-3 mRNA and protein levels are correlated and higher levels of IGFBP-3 are detectable in tumors with poor prognostic features. IMPLICATIONS: IGFBP-3 may be involved in the regulation of breast cancer cell growth.


Subject(s)
Breast Neoplasms/chemistry , Insulin-Like Growth Factor Binding Protein 3/analysis , Aneuploidy , Autoradiography , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Densitometry/methods , Female , Gene Expression Regulation, Neoplastic , Hemoglobins/analysis , Humans , Immunoblotting , Immunoradiometric Assay , Insulin-Like Growth Factor Binding Protein 3/genetics , Neoplasm Proteins/analysis , Predictive Value of Tests , Prognosis , RNA, Messenger/analysis , RNA, Neoplasm/analysis , Receptors, Estrogen/analysis , Receptors, Progesterone/analysis , Regression Analysis , S Phase
12.
Cancer Res ; 58(18): 4199-205, 1998 Sep 15.
Article in English | MEDLINE | ID: mdl-9751635

ABSTRACT

Interleukin-4 (IL-4) is a pleiotropic cytokine produced by mast cells and T lymphocytes that promotes proliferation and immunoglobulin class-switching in B cells. IL-4 receptors (IL-4Rs) are also expressed by nonhematopoietic cells as well as some tumor cells. Unlike its mitogenic effect on B cells, IL-4 inhibits the growth of some cancer cells in vitro. In this study, we show that IL-4R is expressed by breast and ovarian cancer cell lines. Furthermore, anchorage-dependent and -independent growth of breast cancer cell lines MCF-7 and MDA-MB-231 is inhibited by IL-4 treatment, and this effect requires IL-4R. Interestingly, IL-4 only inhibited proliferating breast cancer cells and had no effect on basal, unstimulated growth. We therefore characterized the effect of IL-4 on breast cancer cell growth stimulated by either estradiol or insulin-like growth factor I (IGF-I). In both anchorage-dependent and -independent growth assays, IL-4 inhibited estradiol-stimulated growth. The antiestrogen effect of IL-4 was not due to IL-4 interference with the estrogen receptor, because IL-4 did not interfere with estrogen receptor-mediated reporter gene transactivation. In contrast, IL-4 had no effect on IGF-I-stimulated proliferation. Because IGF-I is known to inhibit programmed cell death, we examined apoptosis as a possible mechanism of IL-4 action. We established that IL-4 induced apoptosis in breast cancer cells by five independent criteria: (a) morphological indicators including pyknotic nuclei and cytoplasmic condensation; (b) DNA fragmentation; (c) the formation of DNA laddering; (d) the cleavage of poly(ADP-ribose) polymerase; and (e) the presence of cells with sub-G1 DNA content. IL-4 increased the percentage of apoptotic cells in MCF-7 and MDA-MB-231 cells 6.0- and 6.7-fold over that of the control, respectively. Finally, the addition of IGF-I reversed IL-4-induced apoptosis, suggesting that the mechanism of IL-4-induced growth inhibition in human breast cancer cells is the induction of programmed cell death.


Subject(s)
Apoptosis , Breast Neoplasms/pathology , Estradiol/pharmacology , Interleukin-4/pharmacology , Receptors, Estrogen/metabolism , Receptors, Interleukin-4/metabolism , Antineoplastic Agents, Phytogenic/pharmacology , Breast Neoplasms/metabolism , Cell Division/drug effects , Female , Humans , Insulin-Like Growth Factor I/pharmacology , Interleukin-4/metabolism , Ovarian Neoplasms/metabolism , Ovarian Neoplasms/pathology , Paclitaxel/pharmacology , Receptors, Estrogen/drug effects , Tumor Cells, Cultured , Tumor Stem Cell Assay
13.
Cancer Res ; 61(15): 5771-7, 2001 Aug 01.
Article in English | MEDLINE | ID: mdl-11479214

ABSTRACT

Estrogen can increase insulin-like growth factor-I receptor (IGF-IR) and insulin receptor substrate-1 (IRS-1) expression, two key components of IGF-I-mediated signaling. The result is sensitization of breast cancer cells to IGF-I and synergistic growth in the presence of estrogen and IGF-I. We hypothesized that loss of estrogen receptor alpha (ERalpha) would result in reduced IGF-mediated signaling and growth. To test this hypothesis, we examined IGF-I effects in MCF-7 breast cancer cell sublines that have been selected for loss of ERalpha (C4 and C4-12 cells are ERalpha-negative) by long-term estrogen withdrawal. C4 and C4-12 cells had reduced IGF-IR and IRS-1 mRNA and protein expression (compared with MCF-7 cells) that was not inducible by estrogen. Furthermore, C4 and C4-12 cells showed reduced IGF-I signaling and failed to show any growth response to either estrogen or IGF-I. To prove that loss of IGF and estrogen-mediated signaling and growth was a consequence of loss of ERalpha, we re-expressed ERalpha in C4-12 cells by stable transfection with HA-tagged ERalpha. Three independent C4-12 ERalpha-HA clones expressed a functional ERalpha that (a) was down-regulated by estrogen, (b) conferred estrogen-induction of cyclin D1 expression, and (c) caused estrogen-mediated increase in the number of cells in S phase. All of the effects were completely blocked by antiestrogens. Interestingly, ERalpha-HA expression in C4-12 cells did not restore estrogen induction of progesterone receptor expression. However, ERalpha-positive C4-12 cells now exhibited estrogen-induction of IGF-IR and IRS-1 levels and responded mitogenically to both estrogen and IGF-I. These data show that ERalpha is a critical requirement for IGF signaling, and to our knowledge this is the first report of functional ERalpha expression that confers estrogen-mediated growth of an ER-negative breast cancer cell line.


Subject(s)
Breast Neoplasms/metabolism , Estradiol/pharmacology , Insulin-Like Growth Factor I/pharmacology , Phosphoproteins/physiology , Receptor, IGF Type 1/physiology , Receptors, Estrogen/physiology , Signal Transduction/physiology , Breast Neoplasms/pathology , Cell Division/physiology , Cyclin D1/biosynthesis , Estrogen Receptor alpha , Hemagglutinins/genetics , Humans , Insulin Receptor Substrate Proteins , Phosphoproteins/biosynthesis , Phosphoproteins/metabolism , Phosphorylation/drug effects , Receptor, IGF Type 1/biosynthesis , Receptors, Estrogen/biosynthesis , Receptors, Estrogen/genetics , Transfection , Tumor Cells, Cultured
14.
Clin Cancer Res ; 7(12 Suppl): 4429s-4435s; discussion 4411s-4412s, 2001 Dec.
Article in English | MEDLINE | ID: mdl-11916236

ABSTRACT

Since the cloning of the estrogen receptor alpha (ERalpha) and subsequent identification of a second distinct form of ER, termed ERbeta, a large volume of research has begun to define the molecular mechanisms of ER action. However, although great progress has been made, ER action is still poorly understood. It is expected that a better understanding of ER action may lead to novel strategies and targets for breast cancer prevention and treatment. One of the early-realized functions of the ER was regulation of growth factor signaling, but the degree of interaction between these two mitogenic signaling pathways could not have been anticipated. Recent evidence suggests that the ER and the growth-factor-signaling pathways intersect and directly interact at every level of signal transduction. The resulting synergism between ER and growth factors has been documented both in normal breast development and, importantly, in breast cancer progression and antiestrogen resistance. In this review, we will highlight our current understanding of the molecular mechanisms of cross-talk between ER and growth-factor-signaling pathways.


Subject(s)
Breast Neoplasms/metabolism , Epidermal Growth Factor/metabolism , Insulin-Like Growth Factor I/metabolism , Receptor Cross-Talk/physiology , Receptors, Estrogen/metabolism , Drug Resistance, Neoplasm , Estrogen Receptor Modulators/therapeutic use , Female , Humans , Signal Transduction/physiology
15.
Clin Cancer Res ; 6(9): 3788-96, 2000 Sep.
Article in English | MEDLINE | ID: mdl-10999774

ABSTRACT

HET/SAF-B was originally cloned as a nuclear matrix protein that bound to matrix attachment regions and as a transcriptional repressor of the small heat shock protein hsp27. In addition, we have found recently that HET/SAF-B is also a corepressor of estrogen receptor activity. Estrogen receptor has a very well-described role in breast cancer, and aberrant expression of nuclear matrix and heat shock proteins has also been implicated in breast tumorigenesis. Therefore, we asked whether HET/SAF-B itself could be important in breast cancer. Toward this goal we examined its expression in breast cancer cell lines and asked whether HET/SAF-B can affect breast cancer cell proliferation. Finally, we studied HET/SAF-B expression in clinical breast cancer samples. HET/SAF-B protein and mRNA were detected at varying levels in all of the eight breast cancer cell lines examined. Using a number of different approaches to modulate the level of HET/SAF-B protein in the cell, we found that HET/SAF-B levels are inversely correlated with cell proliferation. In addition,transfection of HET/SAF-B fused to the green fluorescent protein led to the formation of multinucleated cells not observed in cells transfected with green fluorescent protein alone, suggesting that this effect is a direct result of HET/SAF-B overexpression. Western blot analysis of HET/SAF-B in 61 human breast tumors revealed widely varying levels of HET/SAF-B expression, with some tumors (16%) lacking any detectable HET/SAF-B. Statistical analysis showed that high HET/SAF-B expression in these tumors was associated with low S-phase fraction and with aneuploidy, consistent with our results from transfection experiments in tissue culture cells. We conclude that HET/SAF-B plays an important role in breast cancer, and we discuss possible mechanisms of the involvement of HET/SAF-B in cell proliferation and division.


Subject(s)
Aneuploidy , Breast Neoplasms/genetics , Breast Neoplasms/pathology , DNA-Binding Proteins/physiology , Matrix Attachment Region Binding Proteins , Nuclear Matrix-Associated Proteins , Nuclear Proteins/physiology , Receptors, Estrogen , 3T3 Cells/metabolism , Animals , Breast Neoplasms/metabolism , CHO Cells/metabolism , Cell Division/physiology , Cell Nucleus/physiology , Cricetinae , DNA-Binding Proteins/biosynthesis , DNA-Binding Proteins/genetics , Gene Expression , Green Fluorescent Proteins , Humans , Luminescent Proteins/biosynthesis , Luminescent Proteins/genetics , Mice , Nuclear Proteins/biosynthesis , Nuclear Proteins/genetics , Recombinant Fusion Proteins/biosynthesis , Recombinant Fusion Proteins/genetics , Transfection , Tumor Cells, Cultured
16.
Clin Cancer Res ; 3(1): 103-9, 1997 Jan.
Article in English | MEDLINE | ID: mdl-9815544

ABSTRACT

Insulin-like growth factors (IGFs) interact with specific cell surface receptors to mediate cell growth. Intracellular effects of the IGFs are mediated by activation of secondary messenger molecules. One of these proteins, insulin receptor substrate-1 (IRS-1), is phosphorylated after type I IGF receptor activation and has a major role in IGF signaling. Receptor activation also is influenced by high-affinity IGF binding proteins (IGFBPs). In serum, IGFBP-3 is the predominant species. The role of IGFBP-3 in the regulation of breast cancer cell growth is unclear; both growth inhibition and stimulation have been documented in tissue culture systems. To investigate the influence of IGFBP-3 and IRS-1 in breast cancer, we measured levels of these proteins by ELISA and immunoblotting in 195 node-negative primary human breast cancers and compared their levels with known prognostic factors and disease-free survival (DFS). IGFBP-3 levels correlated positively with tumor size (r = 0.27, P < 0.0001) and negatively with estrogen receptor (r = -0.35, P < 0. 0001) and progesterone receptor (r = -0.16, P = 0.021). In contrast, IRS-1 did not correlate with prognostic factors, but higher levels of IRS-1 predicted worse DFS for the subset of patients with tumors

Subject(s)
Breast Neoplasms/chemistry , Insulin-Like Growth Factor Binding Protein 3/analysis , Phosphoproteins/analysis , Breast Neoplasms/diagnosis , Breast Neoplasms/metabolism , Breast Neoplasms/mortality , Disease-Free Survival , Female , Humans , Insulin Receptor Substrate Proteins , Insulin-Like Growth Factor Binding Protein 3/metabolism , Middle Aged , Phosphoproteins/metabolism , Prognosis , Survival Analysis
17.
Mol Endocrinol ; 14(3): 369-81, 2000 Mar.
Article in English | MEDLINE | ID: mdl-10707955

ABSTRACT

The estrogen receptor (ER) is a ligand-dependent transcription factor that acts in a cell- and promoter-specific manner. Evidence suggests that the activity of the ER can be regulated by a number of other stimuli (e.g. growth factors) and that the effects of the ER are modulated by nuclear factors termed coregulators. While the interplay among these factors may in part explain the pleiotropic effects elicited by the ER, there are several other less well described mechanisms of control, such as interactions with the nuclear matrix. Here we report that the nuclear matrix protein/scaffold attachment factor HET/SAF-B is an ER-interacting protein. ER and HET/SAF-B interact in in vitro binding assays, with HET binding to both the ER DNA-binding domain and the hinge region. Coimmunoprecipitation experiments reveal that HET/SAF-B and ER associate in cell lines in the presence or absence of estradiol, but binding is increased by the antiestrogen tamoxifen. HET/SAF-B enhances tamoxifen antagonism of estrogen-induced ER-mediated transactivation, but at high concentrations can inhibit both estrogen and tamoxifen-induced ER activity. HET/SAF-B-mediated repression of ER activity is dependent upon interaction with the ER-DBD. While the existence of high-affinity binding sites for the ER in the nuclear matrix has been known for some time, we now provide evidence of a specific nuclear matrix protein binding to the ER. Furthermore, our data showing that HET/SAF-B binds to ER particularly strongly in the presence of tamoxifen suggests that it may be important for the antagonist effect of tamoxifen.


Subject(s)
DNA-Binding Proteins/metabolism , Estrogen Receptor Modulators/pharmacology , Matrix Attachment Region Binding Proteins , Nuclear Matrix-Associated Proteins , Nuclear Proteins/metabolism , Receptors, Estrogen/metabolism , Tamoxifen/pharmacology , Transcriptional Activation/drug effects , Adenocarcinoma/metabolism , Adenocarcinoma/pathology , Animals , Base Sequence , Bone Neoplasms/pathology , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , COS Cells , Carcinoma, Hepatocellular/pathology , Chlorocebus aethiops , DNA/metabolism , Depression, Chemical , Estradiol/pharmacology , Estrogen Receptor Modulators/metabolism , Female , Humans , Liver Neoplasms/pathology , Macromolecular Substances , Molecular Sequence Data , Neoplasm Proteins/metabolism , Neoplasms, Hormone-Dependent/metabolism , Neoplasms, Hormone-Dependent/pathology , Osteosarcoma/pathology , Protein Binding , Protein Structure, Tertiary , Receptors, Estrogen/drug effects , Recombinant Fusion Proteins/metabolism , Structure-Activity Relationship , Tamoxifen/metabolism , Tumor Cells, Cultured/drug effects
18.
Mol Endocrinol ; 13(5): 787-96, 1999 May.
Article in English | MEDLINE | ID: mdl-10319328

ABSTRACT

Cross-talk between insulin-like growth factor (IGF)- and estrogen receptor (ER)-signaling pathways results in synergistic growth. We show here that estrogen enhances IGF signaling by inducing expression of three key IGF-regulatory molecules, the type 1 IGF receptor (IGFR1) and its downstream signaling molecules, insulin receptor substrate (IRS)-1 and IRS-2. Estrogen induction of IGFR1 and IRS expression resulted in enhanced tyrosine phosphorylation of IRS-1 after IGF-I stimulation, followed by enhanced mitogen-activated protein kinase activation. To examine whether these pathways were similarly activated in vivo, we examined MCF-7 cells grown as xenografts in athymic mice. IRS-1 was expressed at high levels in estrogen-dependent growth of MCF-7 xenografts, but withdrawal of estrogen, which decreased tumor growth, resulted in a dramatic decrease in IRS-1 expression. Finally, we have shown that high IRS-1 expression is an indicator of early disease recurrence in ER-positive human primary breast tumors. Taken together, these data not only reinforce the concept of cross-talk between IGF- and ER-signaling pathways, but indicate that IGF molecules may be critical regulators of estrogen-mediated growth and breast cancer pathogenesis.


Subject(s)
Breast Neoplasms/metabolism , Estrogens/metabolism , Phosphoproteins/metabolism , Somatomedins/metabolism , Animals , Breast Neoplasms/drug therapy , Calcium-Calmodulin-Dependent Protein Kinases/drug effects , Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Estradiol/analogs & derivatives , Estradiol/pharmacology , Estrogen Antagonists/pharmacology , Female , Fulvestrant , Gene Expression Regulation, Neoplastic , Humans , Insulin Receptor Substrate Proteins , Insulin-Like Growth Factor I/metabolism , Insulin-Like Growth Factor I/pharmacology , Intracellular Signaling Peptides and Proteins , Mammary Neoplasms, Experimental/metabolism , Mice , Phosphoproteins/genetics , Phosphorylation , Receptor, Insulin/genetics , Receptor, Insulin/metabolism , Receptors, Estrogen/metabolism , Receptors, Somatomedin/genetics , Receptors, Somatomedin/metabolism , Signal Transduction , Survival Rate , Transplantation, Heterologous , Tumor Cells, Cultured
19.
Endocrinology ; 142(4): 1479-88, 2001 Apr.
Article in English | MEDLINE | ID: mdl-11250928

ABSTRACT

Overexpression of des(1-3) human insulin-like growth factor I (IGF-I) in the mammary glands of transgenic mice (WAP-DES) inhibits apoptosis during natural, but not forced, mammary involution. We hypothesized that this differential response would correlate with the expression of IGF signal transducers. Forced and natural involution were analyzed in nontransgenic and WAP-DES mice beginning on day 16 postpartum. During natural involution, mammary gland wet weight was higher and apoptosis was lower in WAP-DES than in nontransgenic mice. The WAP-DES transgene had no effect on these parameters during forced involution. Mammary tissue concentrations of the transgene protein were 2- to 10-fold higher than those of endogenous IGF-I. Western blot analysis of pooled mammary tissue extracts demonstrated only slightly higher phosphorylation of the IGF signal transducers insulin receptor substrate-1 (IRS-1) and Akt in the WAP-DES than in nontransgenic mice. Dramatic early reductions in phospho-IRS-1, phospho-Akt, IRS-1, IRS-2, and Akt proteins occurred during forced, but not natural, involution. The abundance of the IGF-I receptor and the messenger RNAs for the IGF-I receptors, IRS-1 and -2, were not affected by either genotype or involution. These findings support the conclusions that mammary cells lose their responsiveness to insulin-like signals during forced involution, and that posttranscriptional or posttranslational regulation of IRS-1 and IRS-2 may play a role in this loss.


Subject(s)
Breast/drug effects , Breast/metabolism , Insulin-Like Growth Factor I/pharmacology , Peptide Fragments/pharmacology , Protein Sorting Signals/physiology , Somatomedins/physiology , Animals , Apoptosis/drug effects , Blotting, Western , Breast/cytology , Female , Immunohistochemistry , Male , Mice , Organ Size/drug effects , Phosphorylation , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Signal Transduction/drug effects
20.
Endocrinology ; 144(6): 2683-94, 2003 Jun.
Article in English | MEDLINE | ID: mdl-12746333

ABSTRACT

Insulin receptor substrates (IRS) are central integrators of hormone, cytokine, and growth factor signaling. IRS proteins can be phosphorylated by a number of signaling pathways critical to normal mammary gland development. Studies in transgenic mice that overexpress IGF-I in the mammary gland suggested that IRS expression is important in the regulation of normal postlactational mammary involution. The goal of these studies was to examine IRS expression in the mouse mammary gland and determine the importance of IRS-1 to mammary development in the virgin mouse. IRS-1 and -2 show distinct patterns of protein expression in the virgin mouse mammary gland, and protein abundance is dramatically increased during pregnancy and lactation, but rapidly lost during involution. Consistent with hormone regulation, IRS-1 protein levels are reduced by ovariectomy, induced by combined treatment with estrogen and progesterone, and vary considerably throughout the estrous cycle. These changes occur without similar changes in mRNA levels, suggesting posttranscriptional control. Mammary glands from IRS-1 null mice have smaller fat pads than wild-type controls, but this reduction is proportional to the overall reduction in body size. Development of the mammary duct (terminal endbuds and branch points) is not altered by the loss of IRS-1, and pregnancy-induced proliferation is not changed. These data indicate that IRS undergo complex developmental and hormonal regulation in the mammary gland, and that IRS-1 is more likely to regulate mammary function in lactating mice than in virgin or pregnant mice.


Subject(s)
Estrogens/pharmacology , Mammary Glands, Animal/physiology , Phosphoproteins/genetics , Progesterone/pharmacology , Signal Transduction/physiology , Adipose Tissue/chemistry , Adipose Tissue/growth & development , Adipose Tissue/physiology , Animals , Estrous Cycle/physiology , Female , Gene Expression/drug effects , Gene Expression/physiology , Insulin Receptor Substrate Proteins , Intracellular Signaling Peptides and Proteins , Mammary Glands, Animal/chemistry , Mammary Glands, Animal/growth & development , Mice , Mice, Inbred Strains , Ovariectomy , Phosphoproteins/analysis , Pregnancy , Signal Transduction/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL