Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 59
Filter
2.
Oncogene ; 38(25): 5111-5112, 2019 Jun.
Article in English | MEDLINE | ID: mdl-30867566

ABSTRACT

In the published version of this article, the images for cytoplasmic and nuclear FGF7 in MDA-MB-231 cells were duplicated and mistaken for total FGF7 in SKBR-3 and MDA-MB-231 cells.

3.
Oncogenesis ; 5(8): e252, 2016 Aug 15.
Article in English | MEDLINE | ID: mdl-27526106

ABSTRACT

The forkhead box M1 (FOXM1) transcription factor has a central role in genotoxic agent response in breast cancer. FOXM1 is regulated at the post-translational level upon DNA damage, but the key mechanism involved remained enigmatic. RNF168 is a ubiquitination E3-ligase involved in DNA damage response. Western blot and gene promoter-reporter analyses showed that the expression level and transcriptional activity of FOXM1 reduced upon RNF168 overexpression and increased with RNF168 depletion by siRNA, suggesting that RNF168 negatively regulates FOXM1 expression. Co-immunoprecipitation studies in MCF-7 cells revealed that RNF168 interacted with FOXM1 and that upon epirubicin treatment FOXM1 downregulation was associated with an increase in RNF168 binding and conjugation to the protein degradation-associated K48-linked polyubiquitin chains. Consistently, RNF168 overexpression resulted in an increase in turnover of FOXM1 in MCF-7 cells treated with the protein synthesis inhibitor cycloheximide. Conversely, RNF168, knockdown significantly enhanced the half-life of FOXM1 in both absence and presence of epirubicin. Using a SUMOylation-defective FOXM1-5x(K>R) mutant, we demonstrated that SUMOylation is required for the recruitment of RNF168 to mediate FOXM1 degradation. In addition, clonogenic assays also showed that RNF168 mediates epirubicin action through targeting FOXM1, as RNF168 could synergise with epirubicin to repress clonal formation in wild-type but not in FOXM1-deficient mouse embryo fibroblasts (MEFs). The physiological relevance of RNF168-mediated FOXM1 repression is further emphasized by the significant inverse correlation between FOXM1 and RNF168 expression in breast cancer patient samples. Moreover, we also obtained evidence that RNF8 recruits RNF168 to FOXM1 upon epirubicin treatment and cooperates with RNF168 to catalyse FOXM1 ubiquitination and degradation. Collectively, these data suggest that RNF168 cooperates with RNF8 to mediate the ubiquitination and degradation of SUMOylated FOXM1 in breast cancer genotoxic response.

4.
Oncogenesis ; 5: e214, 2016 Apr 04.
Article in English | MEDLINE | ID: mdl-27043660

ABSTRACT

BRCA1 mutation or depletion correlates with basal-like phenotype and poor prognosis in breast cancer but the underlying reason remains elusive. RNA and protein analysis of a panel of breast cancer cell lines revealed that BRCA1 deficiency is associated with downregulation of the expression of the pleiotropic tumour suppressor FOXO3. Knockdown of BRCA1 by small interfering RNA (siRNA) resulted in downregulation of FOXO3 expression in the BRCA1-competent MCF-7, whereas expression of BRCA1 restored FOXO3 expression in BRCA1-defective HCC70 and MDA-MB-468 cells, suggesting a role of BRCA1 in the control of FOXO3 expression. Treatment of HCC70 and MDA-MB-468 cells with either the DNA methylation inhibitor 5-aza-2'-deoxycitydine, the N-methyltransferase enhancer of zeste homologue 2 (EZH2) inhibitor GSK126 or EZH2 siRNA induced FOXO3 mRNA and protein expression, but had no effect on the BRCA1-competent MCF-7 cells. Chromatin immunoprecipitation (ChIP) analysis demonstrated that BRCA1, EZH2, DNMT1/3a/b and histone H3 lysine 27 trimethylation (H3K27me3) are recruited to the endogenous FOXO3 promoter, further advocating that these proteins interact to modulate FOXO3 methylation and expression. In addition, ChIP results also revealed that BRCA1 depletion promoted the recruitment of the DNA methyltransferases DNMT1/3a/3b and the enrichment of the EZH2-mediated transcriptional repressive epigenetic marks H3K27me3 on the FOXO3 promoter. Methylated DNA immunoprecipitation assays also confirmed increased CpG methylation of the FOXO3 gene on BRCA1 depletion. Analysis of the global gene methylation profiles of a cohort of 33 familial breast tumours revealed that FOXO3 promoter methylation is significantly associated with BRCA1 mutation. Furthermore, immunohistochemistry further suggested that FOXO3 expression was significantly associated with BRCA1 status in EZH2-positive breast cancer. Consistently, high FOXO3 and EZH2 mRNA levels were significantly associated with good and poor prognosis in breast cancer, respectively. Together, these data suggest that BRCA1 can prevent and reverse FOXO3 suppression via inhibiting EZH2 and, consequently, its ability to recruit the transcriptional repressive H3K27me3 histone marks and the DNA methylases DNMT1/3a/3b, to induce DNA methylation and gene silencing on the FOXO3 promoter.

5.
Hong Kong Med J ; 22(2): 144-51, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26823385

ABSTRACT

OBJECTIVES: To compare the PathVysion fluorescence in-situ hybridisation assay with the INFORM HER2 Dual in-situ hybridisation assay on 104 invasive breast cancers with a broad spectrum of immunohistochemistry scores. METHODS: This case series involved consecutive patients diagnosed with invasive breast carcinoma with equivocal immunohistochemistry score and referred for further HER2 assessment from the departments of Surgery and/or Clinical Oncology of the two hospitals between January 2013 and February 2014. An additional 10 cases with negative HER2 immunohistochemistry and 11 cases with positive HER2 immunohistochemistry were further included. RESULTS: The results of both fluorescence in-situ hybridisation and dual in-situ hybridisation were available in 99 of 104 cases, respectively. Student'st test showed no statistically significant difference in the mean number of HER2 count, CEP17 copies, or HER2/CEP17 ratio between that obtained by fluorescence in-situ hybridisation and that obtained by dual in-situ hybridisation. Pearson's correlation of results for the two assays was strong for HER2/CEP17 signal ratio (R=0.963, P<0.001) and mean HER2 copies per nucleus (R=0.897, P<0.001). Overall agreement was 96.0% (95 out of 99 cases, ĸ0.882). Three of the four discordant cases were equivocal for either fluorescence in-situ hybridisation or dual in-situ hybridisation. The results of immunohistochemistry 0/1+ and 3+ cases showed 100% concordance between the two assays. The failure rate was 0.96% for fluorescence in-situ hybridisation and 3.85% for dual in-situ hybridisation. Cases that failed for fluorescence in-situ hybridisation were successful for dual in-situ hybridisation and vice versa. CONCLUSIONS: Our study showed that dual in-situ hybridisation is a reliable and useful option for HER2 testing in breast cancer.


Subject(s)
Breast Neoplasms/pathology , In Situ Hybridization, Fluorescence/methods , In Situ Hybridization/methods , Receptor, ErbB-2/genetics , Breast Neoplasms/genetics , Female , Gene Amplification , Hong Kong , Humans , Immunohistochemistry , Neoplasm Invasiveness , Reproducibility of Results , Retrospective Studies
6.
Oncogene ; 35(8): 990-1002, 2016 Feb 25.
Article in English | MEDLINE | ID: mdl-25961928

ABSTRACT

FOXM1 has been implicated in taxane resistance, but the molecular mechanism involved remains elusive. In here, we show that FOXM1 depletion can sensitize breast cancer cells and mouse embryonic fibroblasts into entering paclitaxel-induced senescence, with the loss of clonogenic ability, and the induction of senescence-associated ß-galactosidase activity and flat cell morphology. We also demonstrate that FOXM1 regulates the expression of the microtubulin-associated kinesin KIF20A at the transcriptional level directly through a Forkhead response element (FHRE) in its promoter. Similar to FOXM1, KIF20A expression is downregulated by paclitaxel in the sensitive MCF-7 breast cancer cells and deregulated in the paclitaxel-resistant MCF-7Tax(R) cells. KIF20A depletion also renders MCF-7 and MCF-7Tax(R) cells more sensitive to paclitaxel-induced cellular senescence. Crucially, resembling paclitaxel treatment, silencing of FOXM1 and KIF20A similarly promotes abnormal mitotic spindle morphology and chromosome alignment, which have been shown to induce mitotic catastrophe-dependent senescence. The physiological relevance of the regulation of KIF20A by FOXM1 is further highlighted by the strong and significant correlations between FOXM1 and KIF20A expression in breast cancer patient samples. Statistical analysis reveals that both FOXM1 and KIF20A protein and mRNA expression significantly associates with poor survival, consistent with a role of FOXM1 and KIF20A in paclitaxel action and resistance. Collectively, our findings suggest that paclitaxel targets the FOXM1-KIF20A axis to drive abnormal mitotic spindle formation and mitotic catastrophe and that deregulated FOXM1 and KIF20A expression may confer paclitaxel resistance. These findings provide insights into the underlying mechanisms of paclitaxel resistance and have implications for the development of predictive biomarkers and novel chemotherapeutic strategies for paclitaxel resistance.


Subject(s)
Antineoplastic Agents/pharmacology , Breast Neoplasms/drug therapy , Breast Neoplasms/genetics , Down-Regulation , Drug Resistance, Neoplasm , Forkhead Transcription Factors/physiology , Kinesins/genetics , Mitosis , Paclitaxel/pharmacology , Animals , Cell Survival/drug effects , Cellular Senescence/drug effects , Female , Forkhead Box Protein M1 , Forkhead Transcription Factors/drug effects , Gene Expression Regulation, Neoplastic , Humans , Kinesins/metabolism , Mice , Mitosis/drug effects , Promoter Regions, Genetic , Spindle Apparatus/physiology , Tumor Cells, Cultured
7.
Oncogene ; 35(11): 1433-44, 2016 Mar 17.
Article in English | MEDLINE | ID: mdl-26148240

ABSTRACT

The forkhead transcription factor FOXM1 has a key role in DNA damage response, and its deregulated overexpression is associated with genotoxic drug resistance in breast cancer. However, little is known about the posttranslational mechanisms by which FOXM1 expression is regulated by genotoxic agents and how they are deregulated in resistant cells. Initial co-immunoprecipitation studies verified previous proteomic analysis finding that the OTUB1 is a novel FOXM1-interacting protein. Western blot analysis showed that both OTUB1 and FOXM1 expression reduced upon genotoxic agent treatment in MCF-7 cells, but remained relatively constant in resistant cells. FOXM1 expression reduced upon OTUB1 depletion by siRNA and increased with OTUB1 overexpression in MCF-7 cells, arguing that OTUB1 positively regulates FOXM1 expression. In agreement, co-immunoprecipitation experiments demonstrated that FOXM1 expression is associated with OTUB1 binding but inversely correlates with conjugation to the protein degradation-associated Lys-48-linked ubiquitin-chains. Overexpression of wild-type (WT) OTUB1, but not the OTUB1(C91S) mutant, disrupted the formation of Lys48-linked ubiquitin-conjugates on FOXM1. Importantly, knockdown of OTUB1 by siRNA resulted in an increase in turnover of FOXM1 in MCF-7 cells treated with the protein synthesis inhibitor cycloheximide, whereas overexpression of WT OTUB1, but not the OTUB1(C91S) mutant, significantly enhances the half-life of FOXM1. In addition, proliferative and clonogenic assays also show that OTUB1 can enhance the proliferative rate and epirubicin resistance through targeting FOXM1, as OTUB1 has little effect on FOXM1-deficient cells. The physiological relevance of the regulation of FOXM1 by OTUB1 is further underscored by the significant correlations between FOXM1 and OTUB1 expression in breast cancer patient samples. Cox-regression survival analysis indicates that OTUB1 overexpression is linked to poorer outcome in particular in patients treated with chemotherapy. Collectively, these data suggest that OTUB1 limits the ubiquitination and degradation of FOXM1 in breast cancer and has a key role in genotoxic agent resistance.


Subject(s)
Antibiotics, Antineoplastic/pharmacology , Breast Neoplasms/genetics , Cysteine Endopeptidases/genetics , Drug Resistance, Neoplasm/genetics , Epirubicin/pharmacology , Forkhead Transcription Factors/metabolism , Breast Neoplasms/drug therapy , Cell Line, Tumor , Cell Proliferation/drug effects , Cycloheximide/pharmacology , DNA Damage/genetics , DNA Repair/genetics , Deubiquitinating Enzymes , Female , Forkhead Box Protein M1 , Forkhead Transcription Factors/genetics , Gene Expression Regulation, Neoplastic/genetics , Humans , MCF-7 Cells , Protein Synthesis Inhibitors/pharmacology , RNA Interference , RNA, Small Interfering/genetics , Ubiquitination/genetics
8.
Oncogenesis ; 4: e167, 2015 Sep 07.
Article in English | MEDLINE | ID: mdl-26344694

ABSTRACT

The forkhead transcription factor FOXK2 has recently been implicated in cancer cell proliferation and survival, but a role in cancer chemotherapeutic drug resistance has hitherto not been explored. Here we demonstrate that FOXK2 has a central role in mediating the cytotoxic drug response in breast cancer. Clonogenic and cell viability assays showed that enhanced FOXK2 expression sensitizes MCF-7 breast cancer cells to paclitaxel or epirubicin treatment, whereas FOXK2 depletion by small interfering RNAs (siRNAs) confers drug resistance. Our data also showed that the activation of the tumour suppressor FOXO3a by paclitaxel and epirubicin is mediated through the induction of FOXK2, as depletion of FOXK2 by siRNA limits the induction of FOXO3a by these drugs in MCF-7 cells. Chromatin immunoprecipitation (ChIP) analysis showed that in response to drug treatment, FOXK2 accumulates and binds to the proximal FOXO3a promoter region in MCF-7 cells. Furthermore, we also uncovered that FOXK2 is deregulated and, therefore, can express at high levels in the nucleus of both the paclitaxel and epirubicin drug-resistant MCF-7 cells. Our results showed that ectopically overexpressed FOXK2 accumulates in the nuclei of drug-resistant MCF-7 cells but failed to be recruited to target genes, including FOXO3a. Crucially, we found that FOXO3a is required for the anti-proliferative and epirubicin-induced cytotoxic function of FOXK2 in MCF-7 cells by sulphorhodamine and clonogenic assays. The physiological importance of the regulation of FOXO3a by FOXK2 is further confirmed by the significant correlations between FOXO3a and FOXK2 expression in breast carcinoma patient samples. Further survival analysis also reveals that high nuclear FOXK2 expression significantly associates with poorer clinical outcome, particularly in patients who have received conventional chemotherapy, consistent with our finding that FOXK2 is deregulated in drug-resistant cells. In summary, our results suggest that paclitaxel and epirubicin target the FOXK2 to modulate their cytotoxicity and deregulated FOXK2 confers drug resistance.

9.
Oncogene ; 34(39): 5012-24, 2015 Sep 24.
Article in English | MEDLINE | ID: mdl-25531315

ABSTRACT

FOXA1 expression correlates with the breast cancer luminal subtype and patient survival. RNA and protein analysis of a panel of breast cancer cell lines revealed that BRCA1 deficiency is associated with the downregulation of FOXA1 expression. Knockdown of BRCA1 resulted in the downregulation of FOXA1 expression and enhancement of FOXA1 promoter methylation in MCF-7 breast cancer cells, whereas the reconstitution of BRCA1 in Brca1-deficent mouse mammary epithelial cells (MMECs) promoted Foxa1 expression and methylation. These data suggest that BRCA1 suppresses FOXA1 hypermethylation and silencing. Consistently, the treatment of MMECs with the DNA methylation inhibitor 5-aza-2'-deoxycitydine induced Foxa1 mRNA expression. Furthermore, treatment with GSK126, an inhibitor of EZH2 methyltransferase activity, induced FOXA1 expression in BRCA1-deficient but not in BRCA1-reconstituted MMECs. Likewise, the depletion of EZH2 by small interfering RNA enhanced FOXA1 mRNA expression. Chromatin immunoprecipitation (ChIP) analysis demonstrated that BRCA1, EZH2, DNA methyltransferases (DNMT)1/3a/3b and H3K27me3 are recruited to the endogenous FOXA1 promoter, further supporting the hypothesis that these proteins interact to modulate FOXA1 methylation and repression. Further co-immunoprecipitation and ChIP analysis showed that both BRCA1 and DNMT3b form complexes with EZH2 but not with each other, consistent with the notion that BRCA1 binds to EZH2 and negatively regulates its methyltransferase activity. We also found that EZH2 promotes and BRCA1 impairs the deposit of the gene silencing histone mark H3K27me3 on the FOXA1 promoter. These associations were validated in a familial breast cancer patient cohort. Integrated analysis of the global gene methylation and expression profiles of a set of 33 familial breast tumours revealed that FOXA1 promoter methylation is inversely correlated with the transcriptional expression of FOXA1 and that BRCA1 mutation breast cancer is significantly associated with FOXA1 methylation and downregulation of FOXA1 expression, providing physiological evidence to our findings that FOXA1 expression is regulated by methylation and chromatin silencing and that BRCA1 maintains FOXA1 expression through suppressing FOXA1 gene methylation in breast cancer.


Subject(s)
Breast Neoplasms/genetics , Chromatin/genetics , DNA Methylation , Gene Silencing , Genes, BRCA1 , Hepatocyte Nuclear Factor 3-alpha/genetics , Promoter Regions, Genetic , Animals , Breast Neoplasms/pathology , Cell Line, Tumor , DNA (Cytosine-5-)-Methyltransferases/metabolism , Enhancer of Zeste Homolog 2 Protein , Female , Gene Knockdown Techniques , Humans , Mice , Polycomb Repressive Complex 2/metabolism , DNA Methyltransferase 3B
10.
Oncogene ; 33(32): 4144-55, 2014 Aug 07.
Article in English | MEDLINE | ID: mdl-24141789

ABSTRACT

FOXM1 is implicated in genotoxic drug resistance but its mechanism of action remains elusive. We show here that FOXM1-depletion can sensitize breast cancer cells and mouse embryonic fibroblasts (MEFs) into entering epirubicin-induced senescence, with the loss of long-term cell proliferation ability, the accumulation of γH2AX foci, and the induction of senescence-associated ß-galactosidase activity and cell morphology. Conversely, reconstitution of FOXM1 in FOXM1-deficient MEFs alleviates the accumulation of senescence-associated γH2AX foci. We also demonstrate that FOXM1 regulates NBS1 at the transcriptional level through an forkhead response element on its promoter. Like FOXM1, NBS1 is overexpressed in the epirubicin-resistant MCF-7Epi(R) cells and its expression level is low but inducible by epirubicin in MCF-7 cells. Consistently, overexpression of FOXM1 augmented and FOXM1 depletion reduced NBS1 expression and epirubicin-induced ataxia-telangiectasia mutated (ATM)phosphorylation in breast cancer cells. Together these findings suggest that FOXM1 increases NBS1 expression and ATM phosphorylation, possibly through increasing the levels of the MRN(MRE11/RAD50/NBS1) complex. Consistent with this idea, the loss of P-ATM induction by epirubicin in the NBS1-deficient NBS1-LBI fibroblasts can be rescued by NBS1 reconstitution. Resembling FOXM1, NBS1 depletion also rendered MCF-7 and MCF-7Epi(R) cells more sensitive to epirubicin-induced cellular senescence. In agreement, the DNA repair-defective and senescence phenotypes in FOXM1-deficent cells can be effectively rescued by overexpression of NBS1. Moreover, overexpression of NBS1 and FOXM1 similarly enhanced and their depletion downregulated homologous recombination (HR) DNA repair activity. Crucially, overexpression of FOXM1 failed to augment HR activity in the background of NBS1 depletion, demonstrating that NBS1 is indispensable for the HR function of FOXM1. The physiological relevance of the regulation of NBS1 expression by FOXM1 is further underscored by the strong and significant correlation between nuclear FOXM1 and total NBS1 expression in breast cancer patient samples, further suggesting that NBS1 as a key FOXM1 target gene involved in DNA damage response, genotoxic drug resistance and DNA damage-induced senescence.


Subject(s)
Cell Cycle Proteins/physiology , Cellular Senescence , DNA Damage , Drug Resistance, Neoplasm , Epirubicin/chemistry , Forkhead Transcription Factors/physiology , Nuclear Proteins/physiology , Animals , Antibiotics, Antineoplastic/chemistry , Cell Cycle Proteins/genetics , DNA Repair , DNA-Binding Proteins , Fibroblasts/cytology , Forkhead Box Protein M1 , Forkhead Transcription Factors/genetics , Gene Deletion , Gene Expression Regulation, Neoplastic , Gene Silencing , Humans , MCF-7 Cells , Mice , Nuclear Proteins/genetics , Phenotype , Phosphorylation , Promoter Regions, Genetic , Signal Transduction
11.
Curr Mol Med ; 13(3): 410-6, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23331013

ABSTRACT

CXC chemokine receptor type 4 (CXCR4) plays a prominent role in cancer progression and metastasis. However, its association with breast cancer subtypes is still unknown. In the current study, we analyzed the expression level and the cellular location of CXCR4 in 175 cases of human breast tumors, including 75 cases of triple-negative breast cancers (TNBCs), 41 cases of luminal-subtypes and 60 cases of HER2-positive breast cancers by using immmunohistochemistry (IHC). We found that CXCR4 was expressed more frequently in the TNBCs than in other subtypes (71% for TNBC vs 44% for HER2-positive and 37% for luminal subtype, p < 0.001). In the TNBC group, CXCR4 positive patients have a significantly higher rate of visceral metastasis (liver, lung and brain). The expression level of CXCR4 is also significantly related to tumor size, advanced TNM stage, shorter overall- and disease-free survival. However, in luminal or HER2-positive breast cancer groups, CXCR4 is not correlated with such clinico-pathological characteristics and survival. Taken together, our data indicate that CXCR4 might exert its function exclusively in TNBC patients, suggesting that targeting CXCR4 might be an effective therapy for TNBC patients.


Subject(s)
Breast Neoplasms/metabolism , Breast Neoplasms/mortality , Neoplasm Metastasis , Receptors, CXCR4/metabolism , Adult , Aged , Aged, 80 and over , Brain Neoplasms/secondary , Breast Neoplasms/pathology , Disease Progression , Female , Gene Expression , Humans , Liver Neoplasms/secondary , Lung Neoplasms/secondary , Middle Aged , Prognosis , Receptor, ErbB-2/metabolism , Receptors, CXCR4/biosynthesis , Receptors, Estrogen/metabolism , Receptors, Progesterone/metabolism , Survival Rate
12.
Oncogene ; 31(14): 1845-58, 2012 Apr 05.
Article in English | MEDLINE | ID: mdl-21860419

ABSTRACT

Vascular endothelial growth factor (VEGF) has a central role in breast cancer development and progression, but the mechanisms that control its expression are poorly understood. Breast cancer tissue microarrays revealed an inverse correlation between the Forkhead transcription factor Forkhead box class O (FOXO)3a and VEGF expression. Using the lapatinib-sensitive breast cancer cell lines BT474 and SKBR3 as model systems, we tested the possibility that VEGF expression is negatively regulated by FOXO3a. Lapatinib treatment of BT474 or SKBR3 cells resulted in nuclear translocation and activation of FOXO3a, followed by a reduction in VEGF expression. Transient transfection and inducible expression experiments showed that FOXO3a represses the proximal VEGF promoter, whereas another Forkhead member, FOXM1, induces VEGF expression. Chromatin immunoprecipitation and oligonucleotide pull-down assays showed that both FOXO3a and FOXM1 bind a consensus Forkhead response element (FHRE) in the VEGF promoter. Upon lapatinib stimulation, activated FOXO3a displaces FOXM1 bound to the FHRE before recruiting histone deacetylase 2 (HDAC2) to the promoter, leading to decreased histones H3 and H4 acetylation, and concomitant transcriptional inhibition of VEGF. These results show that FOXO3a-dependent repression of target genes in breast cancer cells, such as VEGF, involves competitive displacement of DNA-bound FOXM1 and active recruitment of transcriptional repressor complexes.


Subject(s)
Breast Neoplasms/metabolism , Forkhead Transcription Factors/metabolism , Vascular Endothelial Growth Factor A/metabolism , Cell Line, Tumor , Female , Forkhead Box Protein M1 , Forkhead Box Protein O3 , Gene Expression Regulation, Neoplastic , Histone Deacetylase 2/metabolism , Humans , Lapatinib , Quinazolines/pharmacology
16.
J Med Genet ; 46(1): 32-9, 2009 Jan.
Article in English | MEDLINE | ID: mdl-18782836

ABSTRACT

BACKGROUND: The BRCA1 gene is an important breast-cancer susceptibility gene. Promoter polymorphisms can alter the binding affinity of transcription factors, changing transcriptional activity and may affect susceptibility to disease. METHODS AND RESULTS: Using direct sequencing of the BRCA1 promoter region, we identified four polymorphisms c.-2804T-->C (rs799908:T-->C), c.-2265C-->T (rs11655505:C-->T), c.-2004A-->G (rs799906:A-->G) and c.-1896(ACA)(1)-->(ACA)(2) (rs8176071:(ACA)(1)-->(ACA)(2)) present in Hong Kong Chinese. Each polymorphism was studied independently and in combination by functional assays. Although all four variants significantly altered promoter activity, the c.-2265T allele had stronger binding than the C allele, and the most common mutant haplotype, which contains the c.-2265T allele, increased promoter activity by 70%. Risk association first tested in Hong Kong Chinese women with breast cancer and age-matched controls and replicated in a large population-based study of Shanghai Chinese, together totalling >3000 participants, showed that carriers of the c.-2265T allele had a reduced risk for breast cancer (combined odd ratio (OR) = 0.80, 95% CI 0.69 to 0.93; p = 0.003) which was more evident among women aged >or=45 years at first diagnosis of breast cancer and without a family history of breast cancer (combined OR = 0.75, 95% CI 0.61 to 0.91; p = 0.004). The most common haplotype containing the c.-2265T allele also showed significant risk association for women aged >or=45 years without a family history of breast cancer (OR = 0.64, 95% CI 0.46 to 0.89; p = 0.008). CONCLUSION: This comprehensive study of BRCA1 promoter polymorphisms found four variants that altered promoter activity and with the most significant contribution from c.-2265C-->T, which could affect susceptibility to breast cancer in the Chinese population. Its significance in other populations remains to be investigated.


Subject(s)
BRCA1 Protein/genetics , Breast Neoplasms/genetics , Polymorphism, Genetic/genetics , Promoter Regions, Genetic/genetics , Transcription, Genetic , Asian People/genetics , Binding Sites , Breast Neoplasms/epidemiology , Case-Control Studies , China/epidemiology , Cohort Studies , Electrophoretic Mobility Shift Assay , Female , Genetic Predisposition to Disease , Genotype , Hong Kong/epidemiology , Humans , Risk Factors , Transcription Factors/genetics , Transcription Factors/metabolism
17.
Hong Kong Med J ; 14 Suppl 4: 31-5, 2008 Aug.
Article in English | MEDLINE | ID: mdl-18708672

ABSTRACT

1. A genetic risk-association study involving more than 1200 subjects showed individuals homozygous for L-SIGN tandem repeats are less susceptible to SARS infection. 2. This was supported by in vitro binding studies that demonstrated homozygous L-SIGN, compared to heterozygous, had higher binding capacity for SARS coronavirus (SARS-CoV), with higher proteasome-dependent viral degradation. In contrast, homozygous L-SIGN demonstrated lower binding capacity for HIV1-gp120.3. Genetic-association studies for single nucleotide polymorphisms of the inflammatory response genes, namely TNF-alpha, INF-alpha, INF-beta, INF-gamma, IL1-alpha, IL1-beta, IL-4, IL-6 and iNOS, failed to show a significant association with SARS clinical outcomes or susceptibility.


Subject(s)
Cell Adhesion Molecules/genetics , Genetic Predisposition to Disease , Lectins, C-Type/genetics , Polymorphism, Genetic , Receptors, Cell Surface/genetics , Severe Acute Respiratory Syndrome/genetics , Severe acute respiratory syndrome-related coronavirus/genetics , Adult , Alleles , Analysis of Variance , Case-Control Studies , Communicable Diseases/genetics , Communicable Diseases/physiopathology , Confidence Intervals , Cytokines/genetics , Cytokines/metabolism , Female , Gene Frequency , Humans , Male , Middle Aged , Odds Ratio , Probability , Severe acute respiratory syndrome-related coronavirus/metabolism , Severe Acute Respiratory Syndrome/physiopathology , Tandem Repeat Sequences , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/metabolism
18.
BMC Cancer ; 8: 196, 2008 Jul 11.
Article in English | MEDLINE | ID: mdl-18616832

ABSTRACT

BACKGROUND: hRad9 is a cell cycle checkpoint gene that is up-regulated in breast cancer. We have previously shown that the mRNA up-regulation correlated with tumor size and local recurrence. Immunohistochemical studies were made to better define the role of hRad9 in breast carcinogenesis. METHODS: Localisation of hRad9 protein were performed on paired tumor and normal breast tissues. Immunoblotting with and without dephosphorylation was used to define the protein isolated from breast cancer cells. RESULTS: Increased hRad9 protein was observed in breast cancer cells nucleus compared to non-tumor epithelium. This nuclear protein existed in hyperphosphorylated forms which may be those of the hRad9-hRad1-hHus1 complex. CONCLUSION: Finding of hyperphosphorylated forms of hRad9 in the nucleus of cancer cells is in keeping with its function in ameliorating DNA instability, whereby it inadvertently assists tumor growth.


Subject(s)
Breast Neoplasms/genetics , Carcinoma/genetics , Cell Cycle Proteins/biosynthesis , Breast Neoplasms/pathology , Carcinoma/pathology , Cell Cycle Proteins/genetics , Cell Transformation, Neoplastic/genetics , DNA Repair/genetics , Female , Gene Expression Regulation, Neoplastic , Humans , Immunohistochemistry , Mammary Glands, Human/metabolism , Organ Specificity , Phosphorylation , Phosphotransferases/genetics , Phosphotransferases/metabolism , Protein Isoforms/biosynthesis , Transcription Factors/biosynthesis , Transcription Factors/genetics , Transcriptional Activation
19.
Hong Kong Med J ; 14(2): 130-5, 2008 Apr.
Article in English | MEDLINE | ID: mdl-18382020

ABSTRACT

OBJECTIVES: To evaluate the prevalence of human epidermal growth factor receptor 2 (HER2) gene overexpression in breast cancer patients encountered in Hong Kong and the concordance of HER2 findings from primary immunohistochemistry assays and confirmatory in-situ hybridisation assays. DESIGN: Retrospective study. SETTING: Department of Clinical Oncology in a public hospital in Hong Kong. PATIENTS: All patient referrals between July 2006 and June 2007 with newly diagnosed invasive breast cancer (for prevalence evaluation), and all patients treated at our unit with confirmatory in-situ hybridisation tests performed within the study period (for concordance evaluation). RESULTS: There were 272 consecutive breast cancer patients eligible for prevalence evaluation. The distribution for immunohistochemistry staining in 249 cases for scores 0, 1+, 2+, and 3+ were 99 (40%), 40 (16%), 58 (23%), and 52 (21%) respectively. In the remaining 23 patients, four and 19 breast cancers were unscored and reported by immunohistochemistry to be HER2-positive and -negative, respectively. The overall HER2 overexpression rate (3+ or reported as positive) was 21%. HER2 overexpression was associated with grade 3 histology (P<0.001) and negative hormonal receptor status (P<0.001). However, it was not associated with age (P=0.525), T-classification (P=0.740), N-classification (P=0.691), nor group stages (P=0.433). Of the 37 patients with confirmatory in-situ hybridisation tests performed, 10 (71%) of 14 with immunohistochemistry staining of 3+ and 1 (4%) of 23 with immunohistochemistry staining of 2+ were found to have HER2 gene amplification. CONCLUSIONS: More than 25% of HER2 overexpression identified by immunohistochemistry assays in this Hong Kong cohort could not be verified by confirmatory in-situ hybridisation assays. Compliance with the latest guidelines for HER2 testing should improve the future accuracy and concordance.


Subject(s)
Breast Neoplasms/genetics , Carcinoma, Ductal/genetics , Carcinoma, Lobular/genetics , Immunohistochemistry , In Situ Hybridization, Fluorescence , In Situ Hybridization/methods , Receptor, ErbB-2/genetics , Adult , Breast Neoplasms/epidemiology , Breast Neoplasms/pathology , Carcinoma, Ductal/epidemiology , Carcinoma, Ductal/pathology , Carcinoma, Lobular/epidemiology , Carcinoma, Lobular/pathology , Cross-Sectional Studies , Female , Gene Amplification/genetics , Gene Expression Regulation, Neoplastic/genetics , Hong Kong , Humans , Middle Aged , Neoplasm Staging , Predictive Value of Tests , Retrospective Studies
20.
Fam Cancer ; 7(2): 125-33, 2008.
Article in English | MEDLINE | ID: mdl-17657584

ABSTRACT

INTRODUCTION: Germline mutations of BRCA1 and BRCA2 account for the majority of hereditary breast cancers, many of which are classified as variants of unknown significance (VUS). We report the identification of a novel BRCA2 variant (c.7806-9T > G) in a Chinese family with multiple breast cancers and document it as a pathogenic mutation. METHODS: The proband in this family was diagnosed with breast cancer at age 50 with a strong family history of breast cancer. DNA and RNA were extracted from the blood of the proband and her family, and was used for BRCA gene mutation/deletion screening and RNA splicing analysis. RESULTS: BRCA2 c.7806-9T > G was identified in the proband, which was suggestive of a variant. This change was also found in two sisters of the proband with a history of breast cancer, as well as from the proband's maternal gastric cancer. The only sibling free of breast cancer did not carry the BRCA2 variant, thus demonstrating that the mutation segregates with the clinical phenotype in this family. RNA analysis on the proband blood sample revealed three aberrant splicing variants: c.7806_7874del, c.7806_7976del, and c.7806-8_7806-1ins. The latter causes a frameshift and creates a truncated protein, whilst the other two splicing variants resulted in shorter forms of the protein. CONCLUSIONS: The identified BRCA2 c.7806-9T > G [Genbank: DQ889340] was found to be pathogenic, based on aberrant splicing events resulting in the formation of truncated protein products. Thus, better understanding and classification of BRCA variants as neutral or disease causing has important implications for genetic counseling so that appropriate management can be given.


Subject(s)
Asian People/genetics , Breast Neoplasms/genetics , Genes, BRCA2 , Mutation , Adult , Base Sequence , Breast Neoplasms/pathology , Female , Genes, BRCA1 , Germ-Line Mutation , Humans , Middle Aged , Pedigree , Point Mutation , Risk Factors , Sequence Analysis, DNA
SELECTION OF CITATIONS
SEARCH DETAIL
...