Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters










Publication year range
2.
Mol Med ; 28(1): 15, 2022 02 05.
Article in English | MEDLINE | ID: mdl-35123413

ABSTRACT

BACKGROUND: During embryogenesis lateral symmetry is broken, giving rise to Left/Right (L/R) breast tissues with distinct identity. L/R-sided breast tumors exhibit consistently-biased incidence, gene expression, and DNA methylation. We postulate that a differential L/R tumor-microenvironment crosstalk generates different tumorigenesis mechanisms. METHODS: We performed in-silico analyses on breast tumors of public datasets, developed xenografted tumors, and conditioned MDA-MB-231 cells with L/R mammary extracts. RESULTS: We found L/R differential DNA methylation involved in embryogenic and neuron-like functions. Focusing on ion-channels, we discovered significant L/R epigenetic and bioelectric differences. Specifically, L-sided cells presented increased methylation of hyperpolarizing ion channel genes and increased Ca2+ concentration and depolarized membrane potential, compared to R-ones. Functional consequences were associated with increased proliferation in left tumors, assessed by KI67 expression and mitotic count. CONCLUSIONS: Our findings reveal considerable L/R asymmetry in cancer processes, and suggest specific L/R epigenetic and bioelectric differences as future targets for cancer therapeutic approaches in the breast and many other paired organs.


Subject(s)
Electric Impedance , Epigenesis, Genetic , Unilateral Breast Neoplasms/genetics , Unilateral Breast Neoplasms/pathology , Animals , Cell Line, Tumor , Computational Biology , DNA Methylation , Female , Gene Expression Regulation, Neoplastic , Humans , Mice , Transcriptome , Tumor Microenvironment
3.
Oncotarget ; 11(28): 2774-2792, 2020 Jul 14.
Article in English | MEDLINE | ID: mdl-32733648

ABSTRACT

HER2 is a well-studied tyrosine kinase (TK) membrane receptor which functions as a therapeutic target in invasive ductal breast carcinomas (IDC). The standard of care for the treatment of HER2-positive breast is the antibody trastuzumab. Despite specific treatment unfortunately, 20% of primary and 70% of metastatic HER2 tumors develop resistance. HER2 belongs to a gene family, with four members (HER1-4) and these members could be involved in resistance to anti-HER2 therapies. In this study we designed a probemix to detect the amplification of the four HER oncogenes in a single reaction. In addition, we developed a protocol based on the combination of MLPA with ddPCR to detect the tumor proportion of co-amplified HERs. On 111 IDC, the HER2 MLPA results were validated by FISH (Adjusted r 2 = 0,91, p < 0,0001), CISH (Adjusted r 2 = 0,938, p < 0,0001) and IHC (Adjusted r 2 = 0,31, p < 0,0001). HER1-4 MLPA results were validated by RT-qPCR assays (Spearman Rank test p < 0,05). Of the 111 samples, 26% presented at least one HER amplified, of which 23% showed co-amplifications with other HERs. The percentage of cells with HER2 co-amplified varied among the tumors (from 2-72,6%). Independent in-silico findings show that the outcome of HER2+ patients is conditioned by the status of HER3 and HER4. Our results encourage further studies to investigate the relationship with patient's response to single or combined treatment. The approach could serve as proof of principle for other tumors in which the HER oncogenes are involved.

4.
BMC Cancer ; 19(1): 328, 2019 Apr 05.
Article in English | MEDLINE | ID: mdl-30953488

ABSTRACT

BACKGROUND: Cancer cells evolve and constitute heterogeneous populations that fluctuate in space and time and are subjected to selection generating intratumor heterogeneity. This phenomenon is determined by the acquisition of genetic/epigenetic alterations and their selection over time which has clinical implications on drug resistance. METHODS: DNA extracted from different tumor cell populations (breast carcinomas, cancer cell lines and cellular clones) were analyzed by MS-MLPA. Methylation profiles were used to generate a heterogeneity index to quantify the magnitude of epigenetic heterogeneity in these populations. Cellular clones were obtained from single cells derived of MDA-MB 231 cancer cell lines applying serial limiting dilution method and morphology was analyzed by optical microscopy and flow cytometry. Clones characteristics were examined through cellular proliferation, migration capacity and apoptosis. Heterogeneity index was also calculated from beta values derived from methylation profiles of TCGA tumors. RESULTS: The study of methylation profiles of 23 fresh breast carcinomas revealed heterogeneous allele populations in these tumor pieces. With the purpose to measure the magnitude of epigenetic heterogeneity, we developed an heterogeneity index based on methylation information and observed that all tumors present their own heterogeneity level. Applying the index calculation in pure cancer cell populations such as cancer cell lines (MDA-MB 231, MCF-7, T47D, HeLa and K-562), we also observed epigenetic heterogeneity. In addition, we detected that clones obtained from the MDA-MB 231 cancer cell line generated their own new heterogeneity over time. Using TCGA tumors, we determined that the heterogeneity index correlated with prognostic and predictive factors like tumor size (p = 0.0088), number of affected axillary nodes (p = 0.007), estrogen receptor expression (p < 0.0001) and HER2 positivity (p = 0.0007). When we analyzed molecular subtypes we found that they presented different heterogeneity levels. Interestingly, we also observed that all mentioned tumor cell populations shared a similar Heterogeneity index (HI) mean. CONCLUSIONS: Our results show that each tumor presents a unique epigenetic heterogeneity level, which is associated with prognostic and predictive factors. We also observe that breast tumor subtypes differ in terms of epigenetic heterogeneity, which could serve as a new contribution to understand the different prognosis of these groups.


Subject(s)
Breast Neoplasms/genetics , Carcinoma, Ductal, Breast/genetics , DNA Methylation/genetics , Epigenesis, Genetic , Adult , Apoptosis/genetics , Breast/pathology , Breast Neoplasms/mortality , Breast Neoplasms/pathology , Carcinoma, Ductal, Breast/mortality , Carcinoma, Ductal, Breast/pathology , Cell Line, Tumor , Cell Movement/genetics , Cell Proliferation/genetics , CpG Islands/genetics , Datasets as Topic , Female , Gene Expression Regulation, Neoplastic , Humans , Prognosis , Promoter Regions, Genetic/genetics
5.
Asian Pac J Cancer Prev ; 19(10): 2859-2866, 2018 Oct 26.
Article in English | MEDLINE | ID: mdl-30362313

ABSTRACT

Objective: Breast cancer is a heterogeneous disease characterized by an accumulation of genetic and epigenetic alterations that lead tumor cells to acquire characteristics like the capacity for invasion and metastasis. Metastasis remains a major challenge in cancer management and understanding of its molecular basis should result in improved prevention, diagnosis, and treatment of breast cancer patients. The aim of this study was to investigate how promoter DNA methylation regulates PAX6 gene expression and influences breast carcinoma cell migration. Methods: PAX6 promoter methylation was detected by Methyl Specific-Multiplex Ligation Probe Amplification (MS-MLPA). Gene expression was evaluated using qRT-PCR, while the effect of PAX6 on migration was ssessed by wound healing assay. In addition, MMP2 and MMP9 genes were studied using different bioinformatic tools. Results: The PAX6 promoter is methylated in breast cancer cell lines and methylation in this region impacts on its expression. Migration assays revealed that PAX6 overexpression promotes cell migration, while PAX6 inhibition decreases it. More importantly, we found that migration is affected by PAX6 methylation status. Employing bioinformatic analysis, binding sites for PAX6 on the regulatory regions of the MMP2 and MMP9 genes were established, PAX6 overexpression increasing MMP2 and MMP9 expression at the mRNA level. Conclusion: Our study provides novel insights into epigenetic events that regulate PAX6 expression and molecular mechanisms by which PAX6 modifies the migration capacity of breast cancer cells.


Subject(s)
Breast Neoplasms/genetics , DNA Methylation/genetics , Matrix Metalloproteinase 2/genetics , Matrix Metalloproteinase 9/genetics , PAX6 Transcription Factor/genetics , Promoter Regions, Genetic/genetics , Cell Line, Tumor , Cell Movement/genetics , Female , Gene Expression Regulation, Neoplastic/genetics , Humans , MCF-7 Cells
6.
Clin Epigenetics ; 10(1): 111, 2018 08 23.
Article in English | MEDLINE | ID: mdl-30139383

ABSTRACT

BACKGROUND: Inhibitor of differentiation protein 4 (ID4) is a dominant negative regulator of the basic helix-loop-helix (bHLH) family of transcription factors. During tumorigenesis, ID4 may act as a tumor suppressor or as an oncogene in different tumor types. However, the role of ID4 in breast cancer is not clear where both an oncogenic and a tumor suppressor function have been attributed. Here, we hypothesize that ID4 behaves as both, but its role in breast differs according to the estrogen receptor (ER) status of the tumor. METHODS: ID4 expression was retrieved from TCGA database using UCSC Xena. Association between overall survival (OS) and ID4 was assessed using Kaplan-Meier plotter. Correlation between methylation and expression was analyzed using the MEXPRESS tool. In vitro experiments involved ectopic expression of ID4 in MCF-7, T47D, and MDA-MB231 breast cancer cell lines. Migration and colony formation capacity were assessed after transfection treatments. Gene expression was analyzed by ddPCR and methylation by MSP, MS-MLPA, or ddMSP. RESULTS: Data mining analysis revealed that ID4 expression is significantly lower in ER+ tumors with respect to ER- tumors or normal tissue. We also demonstrate that ID4 is significantly methylated in ER+ tumors. Kaplan-Meier analysis indicated that low ID4 expression levels were associated with poor overall survival in patients with ER+ tumors. In silico expression analysis indicated that ID4 was associated with the expression of key genes of the ER pathway only in ER+ tumors. In vitro experiments revealed that ID4 overexpression in ER+ cell lines resulted in decreased migration capacity and reduced number of colonies. ID4 overexpression induced a reduction in ER levels in ER+ cell lines, while estrogen deprivation with fulvestrant did not induce changes neither in ID4 methylation nor in ID4 expression. CONCLUSIONS: We propose that ID4 is frequently silenced by promoter methylation in ER+ breast cancers and functions as a tumor suppressor gene in these tumors, probably due to its interaction with key genes of the ER pathway. Our present study contributes to the knowledge of the role of ID4 in breast cancer.


Subject(s)
Breast Neoplasms/genetics , DNA Methylation , Down-Regulation , Inhibitor of Differentiation Proteins/genetics , Breast Neoplasms/metabolism , Cell Line, Tumor , Cell Movement , Epigenesis, Genetic , Female , Gene Expression Regulation, Neoplastic , Humans , MCF-7 Cells , Prognosis , Receptors, Estrogen/metabolism , Signal Transduction , Survival Analysis
7.
PLoS One ; 11(7): e0157416, 2016.
Article in English | MEDLINE | ID: mdl-27383829

ABSTRACT

During the last decades it has been established that breast cancer arises through the accumulation of genetic and epigenetic alterations in different cancer related genes. These alterations confer the tumor oncogenic abilities, which can be resumed as cancer hallmarks (CH). The purpose of this study was to establish the methylation profile of CpG sites located in cancer genes in breast tumors so as to infer their potential impact on 6 CH: i.e. sustained proliferative signaling, evasion of growth suppressors, resistance to cell death, induction of angiogenesis, genome instability and invasion and metastasis. For 51 breast carcinomas, MS-MLPA derived-methylation profiles of 81 CpG sites were converted into 6 CH profiles. CH profiles distribution was tested by different statistical methods and correlated with clinical-pathological data. Unsupervised Hierarchical Cluster Analysis revealed that CH profiles segregate in two main groups (bootstrapping 90-100%), which correlate with breast laterality (p = 0.05). For validating these observations, gene expression data was obtained by RealTime-PCR in a different cohort of 25 tumors and converted into CH profiles. This analyses confirmed the same clustering and a tendency of association with breast laterality (p = 0.15). In silico analyses on gene expression data from TCGA Breast dataset from left and right breast tumors showed that they differed significantly when data was previously converted into CH profiles (p = 0.033). We show here for the first time, that breast carcinomas arising on different sides of the body present differential cancer traits inferred from methylation and expression profiles. Our results indicate that by converting methylation or expression profiles in terms of Cancer Hallmarks, it would allow to uncover veiled associations with clinical features. These results contribute with a new finding to the better understanding of breast tumor behavior, and can moreover serve as proof of principle for other bilateral cancers like lung, testes or kidney.


Subject(s)
Breast Neoplasms/genetics , Breast Neoplasms/physiopathology , Carcinoma/genetics , Carcinoma/physiopathology , CpG Islands , Adult , Aged , Cohort Studies , DNA Methylation , Epigenesis, Genetic , Female , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Humans , Middle Aged
8.
Clin Exp Metastasis ; 32(2): 99-110, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25628026

ABSTRACT

Breast cancer is a heterogeneous disease characterized by the accumulation of genetic and epigenetic alterations that contribute to the development of regional and distant metastases. Lymph node metastasis (LNM) status is the single most important prognostic factor. Metastatic cancer cells share common molecular alterations with those of the primary tumor, but in addition, they develop distinct changes that allow the cancer to progress. There is an urgent need for molecular studies which focus on identifying genomic and epigenomic markers that can predict the progression to metastasis. The objective of this study was to identify epigenetic similarities and differences between paired primary breast tumor (PBT) and LNM. We employed Methylation-Specific-MLPA (Multiplex ligation-dependent probe amplification) to assess the methylation status of 33 cancer-related genes in a cohort of 50 paired PBT and LNM specimens. We found that the methylation index, which represents the degree of aberrantly methylated genes in a specimen, was maintained during the progression to LNM. However, some genes presented differential methylation profiles. Interestingly, PAX6 presented a significant negative correlation between paired PBT and LNM (p = 0.03), which indicated a switch from methylated to unmethylated status in the progression from PBT to LNM. We further identified that the methylation status of PAX6 on the identified CpG site functionally affected the expression of PAX6 at the mRNA level. Our study unraveled significant epigenetic changes during the progression from PBT to LNM, which may contribute to improved prognosis, prediction and therapeutic management of metastatic breast cancer patients.


Subject(s)
Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Epigenesis, Genetic , Gene Expression Regulation, Neoplastic , Lymphatic Metastasis , Biomarkers, Tumor , Breast/pathology , Cohort Studies , CpG Islands , DNA Methylation , Disease Progression , Eye Proteins/metabolism , Female , Homeodomain Proteins/metabolism , Humans , Lymph Nodes/pathology , PAX6 Transcription Factor , Paired Box Transcription Factors/metabolism , Prognosis , Repressor Proteins/metabolism
9.
Cell Microbiol ; 15(6): 922-41, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23217169

ABSTRACT

Coxiella burnetii is a Gram-negative intracellular bacterium. As previously described, both the endocytic and the autophagic pathways contribute to the maturation of Coxiella replicative vacuoles (CRVs). The large CRVs share the properties of both phagolysosomal and autophagolysosomal compartments. Vamp3, Vamp7 and Vamp8 are v-SNAREs involved in the endocytic pathway which participate mainly in the fusion between endosomes and lysosomes. In the present study we observed that Vamp7 interacts with C. burnetii at different infection times (1 h-48 h p.i.). We have determined that a truncated mutant of Vamp7 (Vamp7 NT) and a siRNA against this SNARE protein affects the optimal development of CRVs, suggesting that Vamp7 mediates fusion events that are required for the biogenesis of CRVs. Indeed, we have observed that overexpression of Vamp7 NT inhibited the heterotypic fusion with lysosomes and the homotypic fusion between individual Coxiella phagosomes and CRVs. Moreover, we have detected in the vacuole membrane, at different infection times, the Vamp7 partners (Vti1a and Vti1b). Interestingly, treatment with chloramphenicol reduced the colocalization between C. burnetii and Vamp7, Vti1a or Vti1b, indicating that the recruitment of these SNAREs proteins is a bacteria-driven process that favours the CRV biogenesis, likely by facilitating the interaction with the endolysosomal compartment.


Subject(s)
Coxiella burnetii/pathogenicity , Endocytosis/physiology , SNARE Proteins/physiology , Vacuoles/microbiology , Animals , CHO Cells , Cell Line , Chloramphenicol/pharmacology , Chlorocebus aethiops , Coxiella burnetii/physiology , Cricetinae , Cricetulus , Disease Models, Animal , HeLa Cells , Humans , R-SNARE Proteins/drug effects , R-SNARE Proteins/physiology , RNA, Small Interfering/pharmacology , SNARE Proteins/drug effects , Vero Cells
10.
PLoS One ; 6(8): e24054, 2011.
Article in English | MEDLINE | ID: mdl-21901159

ABSTRACT

Serratia marcescens is an opportunistic human pathogen that represents a growing problem for public health, particularly in hospitalized or immunocompromised patients. However, little is known about factors and mechanisms that contribute to S. marcescens pathogenesis within its host. In this work, we explore the invasion process of this opportunistic pathogen to epithelial cells. We demonstrate that once internalized, Serratia is able not only to persist but also to multiply inside a large membrane-bound compartment. This structure displays autophagic-like features, acquiring LC3 and Rab7, markers described to be recruited throughout the progression of antibacterial autophagy. The majority of the autophagic-like vacuoles in which Serratia resides and proliferates are non-acidic and have no degradative properties, indicating that the bacteria are capable to either delay or prevent fusion with lysosomal compartments, altering the expected progression of autophagosome maturation. In addition, our results demonstrate that Serratia triggers a non-canonical autophagic process before internalization. These findings reveal that S. marcescens is able to manipulate the autophagic traffic, generating a suitable niche for survival and proliferation inside the host cell.


Subject(s)
Autophagy , Serratia marcescens/physiology , Vacuoles/microbiology , Ammonium Chloride/pharmacology , Androstadienes/pharmacology , Animals , CHO Cells , Cell Line , Cricetinae , Epithelial Cells/microbiology , Fluorescent Antibody Technique, Indirect , Gentamicins/pharmacology , Humans , Macrolides/pharmacology , Microscopy, Confocal , Serratia marcescens/drug effects , Wortmannin
11.
Infect Immun ; 79(1): 402-13, 2011 Jan.
Article in English | MEDLINE | ID: mdl-20937765

ABSTRACT

Coxiella burnetii is a Gram-negative obligate intracellular bacterium. After internalization, this bacterium replicates in a large parasitophorous vacuole that has features of both phagolysosomes and autophagosomal compartments. We have previously demonstrated that early after internalization Coxiella phagosomes interact with both the endocytic and the autophagic pathways. In this report, we present evidence that the Coxiella-replicative vacuoles (CRVs) also interact with the secretory pathway. Rab1b is a small GTPase responsible for the anterograde transport between the endoplasmic reticulum and the Golgi apparatus. We present evidence that Rab1b is recruited to the CRV at later infection times (i.e., after 6 h of infection). Interestingly, knockdown of Rab1b altered vacuole growth, indicating that this protein was required for the proper biogenesis of the CRV. In addition, overexpression of the active GTPase-defective mutant (GFP-Rab1b Q67L) affected the development of the Coxiella-replicative compartment inhibiting bacterial growth. On the other hand, disruption of the secretory pathway by brefeldin A treatment or by overexpression of Sar1 T39N, a defective dominant-negative mutant of Sar1, affected the typical spaciousness of the CRVs. Taken together, our results show for the first time that the Coxiella-replicative niche also intercepts the early secretory pathway.


Subject(s)
Bacterial Proteins/metabolism , Coxiella burnetii/physiology , Animals , Bacterial Proteins/genetics , Cell Division , Cell Line , Chlorocebus aethiops , Coxiella burnetii/cytology , Cricetinae , Gene Expression Regulation , Humans , Mice , RNA Interference , RNA, Small Interfering , Vacuoles/microbiology , rab1 GTP-Binding Proteins/metabolism
12.
Traffic ; 11(7): 1001-15, 2010 Jul 01.
Article in English | MEDLINE | ID: mdl-20374555

ABSTRACT

A fundamental feature of eukaryotic cells is the presence of distinct membrane-bound compartments having unique protein and lipid composition. These compartments are interconnected by active trafficking mechanisms that must direct macromolecules to defined locations, and at the same time maintain the protein and lipid composition of each organelle. It is well accepted that Rab proteins play a central role in intracellular transport regulating the recognition, fusion and fission of organelles. However, how the transport is achieved is not completely understood. We propose a model whereby a soluble component in the luminal compartment is transported along different Rab-containing organelles that interact according to the following simple principles: (i) only organelles with the same or compatible Rab membrane domains can fuse; (ii) after fusion, an asymmetric fission occurs producing a tubule and a round-shaped vesicle; and (iii) Rab membrane domains distribute asymmetrically between the two resulting organelles. When this model was tested in a simulation, efficient unidirectional transport was observed, while the compartment identity was preserved. All three principles were absolutely necessary for transport. The model is compatible with Rab association/dissociation dynamics and with Rab conversion. In simulations mimicking a simplified endocytic pathway, soluble and membrane-associated markers were efficiently transported preserving the identity of the interacting compartments.


Subject(s)
Intracellular Membranes/metabolism , Membrane Fusion/physiology , Models, Biological , Eukaryotic Cells/metabolism , Golgi Apparatus/metabolism , Organelles/metabolism , Protein Transport/physiology , rab GTP-Binding Proteins/metabolism , rab GTP-Binding Proteins/physiology
13.
Curr Top Microbiol Immunol ; 335: 227-50, 2009.
Article in English | MEDLINE | ID: mdl-19802568

ABSTRACT

Autophagy is an important cell survival process during nitrogen starvation conditions, and it also plays a housekeeping role, removing superfluous or aged organelles. Autophagy has also been linked to host cell control of several intracellular microorganisms. However, since it is an important host defense mechanism, some pathogens have also evolved strategies to exploit or subvert autophagy. Thus, certain pathogens harness autophagy, leading to persistent infection and pathogenesis. In this chapter we highlight our current understanding of those bacterial pathogens that transit through the autophagic pathway, efficiently replicating and surviving within the host cell. In addition, we discuss present knowledge of how autophagy modulation affects the infectious capacities and life cycles of several intracellular pathogens.


Subject(s)
Autophagy/immunology , Bacteria/immunology , Bacteria/pathogenicity , Bacterial Infections/immunology , Bacterial Infections/microbiology , Chronic Disease , Host-Pathogen Interactions/immunology , Humans , Immunity, Innate
14.
Biochim Biophys Acta ; 1793(9): 1465-77, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19303905

ABSTRACT

Numerous pathogens have developed the capacity to invade host cells to be protected from components of the systemic immune system. However, once in the host cells they utilize sophisticated strategies to avoid the powerful machinery built by the cells to kill invading pathogens. In the last few years cumulative evidence indicates that autophagy is one of the most remarkable tools of the intracellular host cell defense machinery that bacteria must confront upon cell invasion. However, several pathogens subvert the autophagic pathway and, manipulate this process at the molecular level, as a strategy to establish a persistent infection. In this review we have summarized the interaction between autophagy and different bacterial pathogens including those that take advantage of the host cell autophagy, allowing successful colonization, as well as those microorganisms which are controlled by autophagy as part of the innate surveillance mechanism.


Subject(s)
Autophagy , Bacterial Infections/pathology , Intracellular Space/microbiology , Intracellular Space/pathology , Animals , Bacterial Infections/immunology , Bacterial Infections/microbiology , Bacterial Toxins/immunology , Humans , Immunity , Intracellular Space/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...