Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters











Publication year range
1.
BMC Med Genomics ; 15(1): 98, 2022 04 29.
Article in English | MEDLINE | ID: mdl-35488281

ABSTRACT

BACKGROUND: Founder populations that have recently undergone important genetic bottlenecks such as French-Canadians and Ashkenazi Jews can harbor some pathogenic variants at a higher carrier rate than the general population, putting them at a higher risk for certain genetic diseases. In these populations, there can be considerable benefit to performing ethnic-based or expanded preconception carrier screening, which can help in the prevention or early diagnosis and management of some genetic diseases. Acadians are descendants of French immigrants who settled in the Atlantic Coast of Canada in the seventeenth century. Yet, the Acadian population has never been investigated for the prevalence/frequency of disease-causing genetic variants. METHODS: An exome sequencing panel for 312 autosomal recessive and 30 X-linked diseases was designed and specimens from 60 healthy participants were sequenced to assess carrier frequency for the targeted diseases. RESULTS: In this study, we show that a sample population of Acadians in South-East New Brunswick harbor variants for 28 autosomal recessive and 1 X-linked diseases, some of which are significantly more frequent in comparison to reference populations. CONCLUSION: Results from this pilot study suggests a need for further investigation of genomic variation in this population and possibly implementation of targeted carrier and neonatal screening programs.


Subject(s)
Ethnicity , Canada , Humans , Infant, Newborn , New Brunswick , Pilot Projects , Exome Sequencing
2.
Carcinogenesis ; 40(8): 1010-1020, 2019 08 22.
Article in English | MEDLINE | ID: mdl-30605519

ABSTRACT

Recent studies have enabled the identification of important factors regulating cancer progression, such as paired box gene 5 (Pax-5). This transcription factor has consistently been associated to B-cell cancer lesions and more recently solid tumors including breast carcinoma. Although Pax-5 downstream activity is relatively well characterized, aberrant Pax-5 expression in a cancer-specific context is poorly understood. To investigate the regulation of Pax-5 expression, we turned to micro RNAs (miRNAs), small non-coding RNA molecules that regulate key biological processes. Extensive studies show that miRNA deregulation is prevalent in cancer lesions. In this study, we aim to elucidate a causal link between differentially expressed miRNAs in cancer cells and their putative targeting of Pax-5-dependent cancer processes. Bioinformatic prediction tools indicate that miRNAs 484 and 210 are aberrantly expressed in breast cancer and predicted to target Pax-5 messenger RNA (mRNA). Through conditional modulation of these miRNAs in breast cancer cells, we demonstrate that miRNAs 484 and 210 inhibit Pax-5 expression and regulate Pax-5-associated cancer processes. In validation, we show that these effects are probably caused by direct miRNA/mRNA interaction, which are reversible by Pax-5 recombinant expression. Interestingly, miRNAs 484 and 210, which are both overexpressed in clinical tumor samples, are also modulated during epithelial-mesenchymal transitioning and hypoxia that correlate inversely to Pax-5 expression. This is the first study demonstrating the regulation of Pax-5 expression and function by non-coding RNAs. These findings will help us better understand Pax-5 aberrant expression within cancer cells, creating the possibility for more efficient diagnosis and treatments for cancer patients.


Subject(s)
Breast Neoplasms/genetics , MicroRNAs/genetics , PAX5 Transcription Factor/genetics , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Movement/genetics , Cell Proliferation/genetics , Epithelial-Mesenchymal Transition/genetics , Female , Gene Expression Profiling/methods , Gene Expression Regulation, Neoplastic/genetics , Humans , Neoplasm Invasiveness/genetics , Neoplasm Invasiveness/pathology , RNA, Messenger/genetics , Transcription Factors/genetics , Tumor Hypoxia/genetics
3.
Anticancer Res ; 38(9): 5013-5026, 2018 Sep.
Article in English | MEDLINE | ID: mdl-30194145

ABSTRACT

BACKGROUND/AIM: In breast cancer, Pax-5 promotes pro-epithelial features and suppresses malignant cancer processes. However, the molecular mechanism of this antitumor activity remains largely unknown. This study aimed to identify the cellular roles of Pax-5-regulated miRNAs in breast cancer progression. MATERIALS AND METHODS: After transient transfection of Pax-5 in MDA-MB-231 breast cancer cells, Pax-5-regulated miRNA expression was examined by next-generation sequencing. The identified Pax-5-regulated miRNAs were then validated by qRT-PCR and examined for the roles they play in breast cancer cells. RESULTS: Pax-5 was shown to be an effective modulator of miR-215-5p and its target genes. MiR-215 inhibited cell proliferation and migration of breast cancer cells, but not cell invasion. More importantly, Pax-5-induced suppression of cancer cell proliferation and migration was found to be miR-215-dependent. Interestingly, miR-215 profiling in clinical tumor samples showed that miR-215 expression was lower in cancer tissues in comparison to healthy controls. CONCLUSION: Pax-5 reduces breast cancer proliferation and migration through up-regulation of the tumor suppressor miR-215. This result supports the use of miR-215 as a prognostic marker for breast cancer.


Subject(s)
Breast Neoplasms/genetics , MicroRNAs/genetics , PAX5 Transcription Factor/genetics , Up-Regulation , Cell Line, Tumor , Cell Movement , Cell Proliferation , Female , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , High-Throughput Nucleotide Sequencing , Humans , Sequence Analysis, RNA
4.
J Mammary Gland Biol Neoplasia ; 23(3): 177-187, 2018 09.
Article in English | MEDLINE | ID: mdl-30032344

ABSTRACT

Pax-5, an essential transcription factor in B cell development, is aberrantly expressed in various B cell cancer lesions and solid tumors such as breast carcinoma. We have recently shown that Pax-5 regulates NF-κB activity which lead to the modulation of breast cancer phenotypic features (EMT-MET). NF-κB is known as a central mediator in inflammation, stress response as well as being a gatekeeper of pro-tumorigenic activity. However, little is known as to how Pax-5 affects this modulation. We thus turned our attention to microRNAs as potential regulatory effectors. In this study, we set out to elucidate the regulatory network between differential Pax-5 expression and NF-κB activity which dictate breast cancer malignancy. Through next-generation sequencing (NGS) of breast cancer cells conditionally expressing Pax-5, we profile significantly upregulated microRNAs; including microRNA-155, a known regulator of pathological processes and suppressor of malignant growth. Through the conditional expression of microRNA-155 in breast cancer models, we identify and validate IKKε (IKBKE) as a downstream target and an essential effector of Pax-5-mediated suppression of NF-κB signaling. Using rescue experiments, we also confirm that Pax-5 modulates NF-κB activity via IKKε downregulation. Interestingly, we also show that microRNA-155, in turn, supresses Pax-5 expression, indicative of an auto-regulatory feedback loop. Altogether, we demonstrate that Pax-5 inhibits NF-κB signalling through the regulation of microRNA-155 and its downstream target IKKε. The elucidation of this signaling network is relevant as Pax-5 and NF-κB are potent transcriptional regulators of breast cancer aggressivity. In addition, IKKε is relevant oncogene aberrantly expressed in 30% of breast carcinomas. Further insight into the regulatory pathways of breast cancer progression will eventually identify strategic therapeutic and prognostic targets to improve cancer patient outcome.


Subject(s)
Breast Neoplasms/genetics , I-kappa B Kinase/genetics , MicroRNAs/genetics , NF-kappa B/genetics , PAX5 Transcription Factor/genetics , Breast/pathology , Breast Neoplasms/pathology , Carcinogenesis/genetics , Carcinogenesis/pathology , Cell Line, Tumor , Cell Movement/genetics , Cell Proliferation/genetics , Down-Regulation/genetics , Female , Gene Expression Regulation, Neoplastic/genetics , Humans , MCF-7 Cells , Signal Transduction/genetics
5.
J Cancer ; 7(14): 2035-2044, 2016.
Article in English | MEDLINE | ID: mdl-28070224

ABSTRACT

The study of genetic factors regulating breast cancer malignancy is a top priority to mitigate the morbidity and mortality associated with this disease. One of these factors, Pax-5, modulates cancer aggressiveness through the regulation of various components of the epithelial to mesenchymal transitioning (EMT) process. We have previously reported that Pax-5 expression profiles in cancer tissues inversely correlate with those of the Focal Adhesion Kinase (FAK), a potent activator of breast cancer malignancy. In this study, we set out to elucidate the molecular and regulatory relationship between Pax-5 and FAK in breast cancer processes. Interestingly, we found that Pax-5 mediated suppression of breast cancer cell migration is dependent of FAK activity. Our mechanistic examination revealed that Pax-5 inhibits FAK expression and activation. We also demonstrate that Pax-5 is a potent modulator of FAK repressors (p53 and miR-135b) and activator (NFκB) which results in the overall suppression of FAK-mediated signaling cascades. Altogether, our findings bring more insight to the molecular triggers regulating phenotypic transitioning process and signaling cascades leading to breast cancer progression.

6.
Mol Carcinog ; 55(7): 1150-62, 2016 07.
Article in English | MEDLINE | ID: mdl-26207726

ABSTRACT

Mammaglobin 1 (MGB1), a member of the secretoglobin family, is expressed in mammary epithelial tissues and is overexpressed in most mammary carcinomas. Despite the extensive research correlating MGB1 expression profiles to breast cancer pathogenesis and disease outcome, the biological significance of MGB1 in cancer processes is still unclear. We have thus set out to conduct a functional evaluation of the molecular and cellular roles of MGB1 in breast cancer processes leading to disease progression. Using a series of breast cancer cell models with conditional MGB1 expression, we demonstrate that MGB1 promotes cancer cell malignant features. More specifically, loss of MGB1 expression resulted in a decrease of cell proliferation, soft agar spheroid formation, migration, and invasion capacities of breast cancer cells. Concomitantly, we also observed that MGB1 expression activates signaling pathways mediated by MAPK members (p38, JNK, and ERK), the focal adhesion kinase (FAK), matrix metalloproteinases (MMPs) and NFκB. Moreover, MGB1 regulates epithelial to mesenchymal (EMT) features and modulates Snail, Twist and ZEB1 expression levels. Interestingly, we also observed that expression of MGB1 confers breast cancer cell sensitivity to anticancer drug-induced apoptosis. Together, our results support a role for MGB1 in tumor malignancy in exchange for chemosensitivity. These findings provide one of the first descriptive overview of the molecular and cellular roles of MGB1 in breast cancer processes and may offer new insight to the development of therapeutic and prognostic strategies in breast cancer patients. © 2015 Wiley Periodicals, Inc.


Subject(s)
Antineoplastic Agents/pharmacology , Breast Neoplasms/metabolism , Mammaglobin A/genetics , Mammaglobin A/metabolism , Breast Neoplasms/drug therapy , Breast Neoplasms/genetics , Cell Line, Tumor , Cell Proliferation , Cell Survival , Epithelial-Mesenchymal Transition , Female , Gene Expression Regulation, Neoplastic , Humans , MAP Kinase Signaling System , MCF-7 Cells , Neoplasm Invasiveness , Signal Transduction , Up-Regulation
7.
Molecules ; 20(7): 12576-89, 2015 Jul 10.
Article in English | MEDLINE | ID: mdl-26184141

ABSTRACT

Breast cancer is the second leading cause of death amongst women worldwide. As a result, many have turned their attention to new alternative approaches to treat this disease. Caffeic acid phenylethyl ester (CAPE), a well-known active compound from bee propolis, has been previously identified as a strong antioxidant, anti-inflammatory, antiviral and anticancer molecule. In fact, CAPE is well documented as inducing cell death by inhibiting NFκB and by inducing pro-apoptotic pathways (i.e., p53). With the objective of developing stronger anticancer compounds, we studied 18 recently described CAPE derivatives for their ability to induce apoptosis in breast cancer cell lines. Five of the said compounds, including CAPE, were selected and subsequently characterised for their anticancer mechanism of action. We validated that CAPE is a potent inducer of caspase-dependent apoptosis. Interestingly, some newly synthesized CAPE derivatives also showed greater cell death activity than the lead CAPE structure. Similarly to CAPE, analog compounds elicited p53 activation. Interestingly, one compound in particular, analog 10, induced apoptosis in a p53-mutated cell line. These results suggest that our new CAPE analog compounds may display the capacity to induce breast cancer apoptosis in a p53-dependent and/or independent manner. These CAPE analogs could thus provide new therapeutic approaches for patients with varying genotypic signatures (such as p53 mutations) in a more specific and targeted fashion.


Subject(s)
Apoptosis/drug effects , Caffeic Acids/pharmacology , Gene Expression Regulation, Neoplastic , Phenylethyl Alcohol/analogs & derivatives , Biphenyl Compounds/antagonists & inhibitors , Biphenyl Compounds/chemistry , Caffeic Acids/chemical synthesis , Caspase 3/genetics , Caspase 3/metabolism , Caspase 7/genetics , Caspase 7/metabolism , Cell Line, Tumor , Cell Survival/drug effects , Female , Free Radicals/antagonists & inhibitors , Free Radicals/chemistry , Genes, Reporter , Humans , Inhibitory Concentration 50 , Luciferases/genetics , Luciferases/metabolism , MCF-7 Cells , Mutation , Phenylethyl Alcohol/chemical synthesis , Phenylethyl Alcohol/pharmacology , Picrates/antagonists & inhibitors , Picrates/chemistry , Signal Transduction , Structure-Activity Relationship , Tumor Suppressor Protein p53/agonists , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism
8.
Anticancer Agents Med Chem ; 15(1): 79-88, 2015.
Article in English | MEDLINE | ID: mdl-24913660

ABSTRACT

The study of anticancer properties from natural products has regained popularity as natural molecules provide a high diversity of chemical structures with specific biological and medicinal activity. Based on a documented library of the most common medicinal plants used by the indigenous people of North America, we screened and isolated compounds with anti-breast cancer properties from Juniperus communis (common Juniper). Using bioassay-guided fractionation of a crude plant extract, we identified the diterpene isocupressic acid and the aryltetralin lignan deoxypodophyllotoxin (DPT) as potent inducers of caspase-dependent programmed cell death (apoptosis) in malignant MB231 breast cancer cells. Further elucidation revealed that DPT, in contrast to isocupressic acid, also concomitantly inhibited cell survival pathways mediated by the MAPK/ERK and NFκB signaling pathways within hours of treatment. Our findings emphasize the potential and importance of natural product screening for new chemical entities with novel anticancer activities. Natural products research complemented with the wealth of information available through the ethnobotanical and ethnopharmacological knowledge of the indigenous peoples of North America can provide new candidate entities with desirable bioactivities to develop new cancer therapies.


Subject(s)
Apoptosis/drug effects , Breast Neoplasms/drug therapy , Juniperus/chemistry , Podophyllotoxin/analogs & derivatives , Antineoplastic Agents, Phytogenic/chemistry , Antineoplastic Agents, Phytogenic/pharmacology , Biological Products/chemistry , Biological Products/pharmacology , Breast Neoplasms/metabolism , Carboxylic Acids/pharmacology , Caspases/metabolism , Cell Line, Tumor , Diterpenes/pharmacology , Drugs, Chinese Herbal , Female , Humans , Mitogen-Activated Protein Kinase Kinases/metabolism , NF-kappa B/metabolism , Plant Extracts/chemistry , Plant Extracts/pharmacology , Plants, Medicinal/chemistry , Podophyllotoxin/chemistry , Podophyllotoxin/pharmacology , Signal Transduction/drug effects , Tetrahydronaphthalenes/pharmacology
9.
Anticancer Agents Med Chem ; 14(1): 35-45, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24041228

ABSTRACT

S100 inflammatory proteins have been previously shown to modulate breast cancer processes. More specifically, genome-wide transcriptome studies associate S100A8 and S100A9 members to breast cancer progression and malignancy. Findings have shown that S100A8 and S100A9 can signal and regulate cancer cell behavior through both extracellular and intracellular-initiated cascades. However, functional studies exploring the effects of S100 proteins are often contradictory leaving ambiguity and a paucity of data relating to the specific function of S100A8 and S100A9 in breast cancer progression. In this study we sought to better define the functions of intracellular expressed S100A8 and S100A9 on key signaling and cellular processes driving breast cancer malignancy. We observed that extracellular treatments of the MCF7 breast cancer cell line with S100A8 and S100A9 proteins induces cell proliferation. In contrast, intracellular recombinant expression of S100A8 and S100A9 led to growth suppression. Furthermore our analysis revealed that intracellular-expressed S100A8 and S100A9 promote an epithelial-like phenotype through the induction of key markers, such as Ecadherin, integrin alpha-5 and Zona Occludens 1 (ZO-1). Concomitantly, S100A8 and S100A9 negatively regulate the activity of the promalignant Focal Adhesion Kinase-1 (FAK) signaling cascade leading to changes in cell adhesion and invasion properties. Our results uncover important differences in intracellular versus extracellular initiated S100A8 and S100A9 signaling cascades and their effects on mammary epithelial growth. Importantly, S100A8 and S100A9 appear to suppress breast cancer malignancy through an increase in mesenchymal to epithelial transitioning. Our findings shed insight into S100 protein involvement in breast cancer invasiveness and metastasis and clarify some of the controversies relating to these proteins in breast cancer processes.


Subject(s)
Breast Neoplasms/pathology , Calgranulin A/metabolism , Calgranulin B/metabolism , Epithelial-Mesenchymal Transition , Breast Neoplasms/metabolism , Cadherins/genetics , Cadherins/metabolism , Calgranulin A/genetics , Calgranulin B/genetics , Calgranulin B/pharmacology , Cell Adhesion/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Female , Focal Adhesion Protein-Tyrosine Kinases/genetics , Focal Adhesion Protein-Tyrosine Kinases/metabolism , Humans , Neoplasm Invasiveness , PAX5 Transcription Factor/metabolism , Recombinant Proteins/pharmacology , Zonula Occludens-1 Protein/genetics , Zonula Occludens-1 Protein/metabolism
10.
Cancer Biomark ; 11(6): 269-80, 2012.
Article in English | MEDLINE | ID: mdl-23248185

ABSTRACT

Cancer statistics show significant diagnosis numbers amongst men and women worldwide, where breast cancer is by far the most frequently diagnosed cancer in women. Multiple mechanisms and molecules have been shown to occupy major roles in cancer progression and aggressivity. Recently, small non-coding RNA molecules, called micro-RNAs, have become the subject of interest in many molecular pathways in relation to breast cancer, amongst many other pathologies. MiRNAs are capable of regulating gene expression in a sequence-specific manner and regulate diverse expression patterns which are dependent on the cell's state and identity. Studies have brought forward specific miRNAs that have the innate ability to govern unique gene expression profiles regulating cancer cell aggressivity. This review will outline recent findings of characterized miRNAs in relation to their molecular targets leading to cancer malignancy and progression. More specifically, we will focus on miRNAs associated with breast cancer metastatic processes including epithelial to mesenchymal and mesenchymal to epithelial transitioning (EMT/MET transition), migration, invasion and angiogenesis.


Subject(s)
Breast Neoplasms/genetics , Breast Neoplasms/pathology , MicroRNAs/genetics , Animals , Breast Neoplasms/metabolism , Female , Gene Expression Regulation, Neoplastic , Humans , MicroRNAs/metabolism , Neoplasm Invasiveness , Neoplasm Metastasis
SELECTION OF CITATIONS
SEARCH DETAIL