Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Int J Mol Sci ; 24(21)2023 Oct 30.
Artigo em Inglês | MEDLINE | ID: mdl-37958763

RESUMO

A modified version of the PGDx elioTM Plasma Resolve assay was validated as a laboratory-developed test (LDT) for clinical use in the Molecular Diagnostics Laboratory at Fox Chase Cancer Center. The test detects single nucleotide variants (SNVs) and small insertions and deletions (indels) in 33 target genes using fragmented genomic DNA extracted from plasma. The analytical performance of this assay was assessed with reference standard DNA and 29 samples from cancer patients and detected 66 SNVs and 23 indels. Using 50 ng of input DNA, the sensitivity was 95.5% to detect SNVs at 0.5% allele frequency, and the specificity was 92.3%. The sensitivity to detect indels at 1% allele frequency was 70.4%. A cutoff of 0.25% variant allele frequency (VAF) was set up for diagnostic reporting. An inter-laboratory study of concordance with an orthologous test resulted in a positive percent agreement (PPA) of 91.7%.


Assuntos
DNA Tumoral Circulante , Neoplasias , Humanos , DNA Tumoral Circulante/genética , Patologia Molecular , Neoplasias/diagnóstico , Neoplasias/genética , Mutação INDEL , Técnicas de Diagnóstico Molecular , Sequenciamento de Nucleotídeos em Larga Escala/métodos , Mutação , Biomarcadores Tumorais/genética
2.
Am J Blood Res ; 11(4): 438-445, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34540353

RESUMO

Anaemia among non-pregnant females of reproductive age remains a common public health problem globally, as well as in Sri Lanka. The objective of the study was to determine the prevalence of anaemia, asses the knowledge and the associated factors of anaemia among non-pregnant females of reproductive age in a tea estate community in Hantana, Kandy district, Sri Lanka. A descriptive cross-sectional study was conducted among 236 randomly selected non-pregnant females of reproductive age within the tea estate community belonging to two MOH (Medical Officers of Health) areas. The proportion of anaemia was determined by measuring haemoglobin (Hb) concentration using Mindray five-part automated blood analyser. The cut-off value to determine anaemia was set at 12.0 g/dL and respondents were categorized into three anaemia categories based on their Hb value. The common risk factors and knowledge regarding anaemia were assessed using a pre-tested interviewer administered questionnaire. Data was analysed with SPSS version 25. Chi-square test was used to conduct a bi-variate analysis. Prevalence of anaemia was 33.1%, among whom 53.8% had mild anaemia, 39.7% had moderate anaemia and 6.4% had severe anaemia. Anaemia was significantly associated with being employed, delivery of a baby within the past 4 years, advancing age, low income (less than 20,000 LKR) and prolonged menstrual bleeding for more than 3 days. Majority (58.5%) had poor knowledge regarding anaemia with a mean score of 5.69 (SD ± 2.42) out of 12. Since anaemia is a multifactorial condition it requires a combination of interventions such as health education and promotion activities. This study aids in assessing the prevalence of anaemia among estate workers identify the significant factors contributing to anaemia.

3.
Breast Cancer Res ; 21(1): 46, 2019 03 28.
Artigo em Inglês | MEDLINE | ID: mdl-30922380

RESUMO

BACKGROUND: Full-term pregnancy (FTP) at an early age confers long-term protection against breast cancer. Previously, we reported that a FTP imprints a specific gene expression profile in the breast of postmenopausal women. Herein, we evaluated gene expression changes induced by parity in the breast of premenopausal women. METHODS: Gene expression profiling of normal breast tissue from 30 nulliparous (NP) and 79 parous (P) premenopausal volunteers was performed using Affymetrix microarrays. In addition to a discovery/validation analysis, we conducted an analysis of gene expression differences in P vs. NP women as a function of time since last FTP. Finally, a laser capture microdissection substudy was performed to compare the gene expression profile in the whole breast biopsy with that in the epithelial and stromal tissues. RESULTS: Discovery/validation analysis identified 43 differentially expressed genes in P vs. NP breast. Analysis of expression as a function of time since FTP revealed 286 differentially expressed genes (238 up- and 48 downregulated) comparing all P vs. all NP, and/or P women whose last FTP was less than 5 years before biopsy vs. all NP women. The upregulated genes showed three expression patterns: (1) transient: genes upregulated after FTP but whose expression levels returned to NP levels. These genes were mainly related to immune response, specifically activation of T cells. (2) Long-term changing: genes upregulated following FTP, whose expression levels decreased with increasing time since FTP but did not return to NP levels. These were related to immune response and development. (3) Long-term constant: genes that remained upregulated in parous compared to nulliparous breast, independently of time since FTP. These were mainly involved in development/cell differentiation processes, and also chromatin remodeling. Lastly, we found that the gene expression in whole tissue was a weighted average of the expression in epithelial and stromal tissues. CONCLUSIONS: Genes transiently activated by FTP may have a role in protecting the mammary gland against neoplastically transformed cells through activation of T cells. Furthermore, chromatin remodeling and cell differentiation, represented by the genes that are maintained upregulated long after the FTP, may be responsible for the lasting preventive effect against breast cancer.


Assuntos
Perfilação da Expressão Gênica , Genômica , Glândulas Mamárias Humanas/metabolismo , Paridade , Pré-Menopausa , Transcriptoma , Biomarcadores , Biologia Computacional/métodos , Feminino , Perfilação da Expressão Gênica/métodos , Regulação da Expressão Gênica , Ontologia Genética , Genômica/métodos , Humanos , Imuno-Histoquímica , Reprodutibilidade dos Testes , Transdução de Sinais
4.
Br J Cancer ; 115(5): 564-70, 2016 08 23.
Artigo em Inglês | MEDLINE | ID: mdl-27482646

RESUMO

BACKGROUND: The rarity of neuroendocrine malignancies limits the ability to develop new therapies and thus a better understanding of the underlying biology is critical. METHODS: Through a prospective, IRB-approved protocol, patients with neuroendocrine malignancies underwent next-generation sequencing of their tumours to detect somatic mutations (SMs) in 50 cancer-related genes. Clinicopathologic correlation was made among poorly differentiated neuroendocrine carcinomas (NECs/poorly differentiated histology and Ki-67 >20%) and pancreatic neuroendocrine tumours (PanNETs/Ki67 ⩽20%) and non-pancreatic neuroendocrine tumours (NP-NETs/Ki67 ⩽20%). RESULTS: A total of 77 patients were enrolled, with next-generation sequencing results available on 63 patients. Incidence of SMs was 83% (19 out of 23) in poorly differentiated NECs, 45% (5 out of 11) in PanNETs and 14% (4 out of 29) in NP-NETs. TP53 was the most prevalent mutation in poorly differentiated NECs (57%), and KRAS (30%), PIK3CA/PTEN (22%) and BRAF (13%) mutations were also found. Small intestinal neuroendocrine tumours (Ki67 <2%/n=9) did not harbour any mutations. Prevalence of mutations correlated with higher risk of progression within the previous year (32% (low risk) vs 11% (high risk), P=0.01) and TP53 mutation correlated with worse survival (2-year survival 66% vs 97%, P=0.003). CONCLUSIONS: Poorly differentiated NECs have a high mutation burden with potentially targetable mutations. The TP53 mutations are associated with poor survival in neuroendocrine malignancies. These findings have clinical trial implications for choice of therapy and prognostic stratification and warrant confirmation.


Assuntos
Biomarcadores Tumorais/metabolismo , Tumores Neuroendócrinos/patologia , Adulto , Idoso , Idoso de 80 Anos ou mais , Feminino , Perfilação da Expressão Gênica , Humanos , Masculino , Pessoa de Meia-Idade , Tumores Neuroendócrinos/genética , Projetos Piloto , Prognóstico
5.
Breast Cancer Manag ; 2(4): 283-294, 2013 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-24738009

RESUMO

Pregnancy and its effects on breast cancer risk have been widely investigated; there is consensus among researchers that early pregnancy confers protection against breast cancer later in life, whereas nulliparity and late-age parity have been associated with increased risk of developing breast cancer. The answer to the question of how pregnancy reduces breast cancer risk has been elusive; however, pregnancy, like breast cancer, is a similar hormone-dependent entity under direct control of estrogen, progesterone and, of particular importance, human chorionic gonadotropin (hCG). In this report, we emphasize the main changes, previously described by our laboratory, in morphology and gene expression levels of the mammary gland of Sprague-Dawley rats exposed to known cancer-preventative conditions (pregnancy, hCG and progesterone + estrogen). In addition, we postulate a protective mechanism induced by hCG that could reduce the cell's potential to be transformed by carcinogens.

6.
BMC Med Genomics ; 5: 46, 2012 Oct 11.
Artigo em Inglês | MEDLINE | ID: mdl-23057841

RESUMO

BACKGROUND: It is accepted that a woman's lifetime risk of developing breast cancer after menopause is reduced by early full term pregnancy and multiparity. This phenomenon is thought to be associated with the development and differentiation of the breast during pregnancy. METHODS: In order to understand the underlying molecular mechanisms of pregnancy induced breast cancer protection, we profiled and compared the transcriptomes of normal breast tissue biopsies from 71 parous (P) and 42 nulliparous (NP) healthy postmenopausal women using Affymetrix Human Genome U133 Plus 2.0 arrays. To validate the results, we performed real time PCR and immunohistochemistry. RESULTS: We identified 305 differentially expressed probesets (208 distinct genes). Of these, 267 probesets were up- and 38 down-regulated in parous breast samples; bioinformatics analysis using gene ontology enrichment revealed that up-regulated genes in the parous breast represented biological processes involving differentiation and development, anchoring of epithelial cells to the basement membrane, hemidesmosome and cell-substrate junction assembly, mRNA and RNA metabolic processes and RNA splicing machinery. The down-regulated genes represented biological processes that comprised cell proliferation, regulation of IGF-like growth factor receptor signaling, somatic stem cell maintenance, muscle cell differentiation and apoptosis. CONCLUSIONS: This study suggests that the differentiation of the breast imprints a genomic signature that is centered in the mRNA processing reactome. These findings indicate that pregnancy may induce a safeguard mechanism at post-transcriptional level that maintains the fidelity of the transcriptional process.


Assuntos
Mama/metabolismo , Perfilação da Expressão Gênica , Genoma Humano/genética , Paridade/genética , Idoso , Análise por Conglomerados , Ciclinas/genética , Ciclinas/metabolismo , Regulação para Baixo/genética , Feminino , Humanos , Imuno-Histoquímica , Pessoa de Meia-Idade , Análise de Sequência com Séries de Oligonucleotídeos , Inclusão em Parafina , Gravidez , Reprodutibilidade dos Testes , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Transcriptoma/genética , Regulação para Cima/genética
7.
Int J Cancer ; 131(5): 1059-70, 2012 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-22025034

RESUMO

Early pregnancy and multiparity are known to reduce the risk of women to develop breast cancer at menopause. Based on the knowledge that the differentiation of the breast induced by the hormones of pregnancy plays a major role in this protection, this work was performed with the purpose of identifying what differentiation-associated molecular changes persist in the breast until menopause. Core needle biopsies (CNB) obtained from the breast of 42 nulliparous (NP) and 71 parous (P) postmenopausal women were analyzed in morphology, immunocytochemistry and gene expression. Whereas in the NP breast, nuclei of epithelial cells were large and euchromatic, in the P breast they were small and hyperchromatic, showing strong methylation of histone 3 at lysine 9 and 27. Transcriptomic analysis performed using Affymetrix HG_U133 oligonucleotide arrays revealed that in CNB of the P breast, there were 267 upregulated probesets that comprised genes controlling chromatin organization, transcription regulation, splicing machinery, mRNA processing and noncoding elements including XIST. We concluded that the differentiation process induced by pregnancy is centered in chromatin remodeling and in the mRNA processing reactome, both of which emerge as important regulatory pathways. These are indicative of a safeguard step that maintains the fidelity of the transcription process, becoming the ultimate mechanism mediating the protection of the breast conferred by full-term pregnancy.


Assuntos
Biomarcadores/metabolismo , Mama/citologia , Mama/metabolismo , Diferenciação Celular , Montagem e Desmontagem da Cromatina/genética , Células Epiteliais/metabolismo , Pós-Menopausa/genética , Idoso , Feminino , Perfilação da Expressão Gênica , Humanos , Pessoa de Meia-Idade , Análise de Sequência com Séries de Oligonucleotídeos , Paridade/genética , Gravidez , RNA Mensageiro/genética , Reação em Cadeia da Polimerase em Tempo Real , Reação em Cadeia da Polimerase Via Transcriptase Reversa
8.
Cancer Prev Res (Phila) ; 4(9): 1457-64, 2011 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-21622728

RESUMO

The objective of this study was to comprehensively compare the genomic profiles in the breast of parous and nulliparous postmenopausal women to identify genes that permanently change their expression following pregnancy. The study was designed as a two-phase approach. In the discovery phase, we compared breast genomic profiles of 37 parous with 18 nulliparous postmenopausal women. In the validation phase, confirmation of the genomic patterns observed in the discovery phase was sought in an independent set of 30 parous and 22 nulliparous postmenopausal women. RNA was hybridized to Affymetrix HG_U133 Plus 2.0 oligonucleotide arrays containing probes to 54,675 transcripts, scanned and the images analyzed using Affymetrix GCOS software. Surrogate variable analysis, logistic regression, and significance analysis of microarrays were used to identify statistically significant differences in expression of genes. The false discovery rate (FDR) approach was used to control for multiple comparisons. We found that 208 genes (305 probe sets) were differentially expressed between parous and nulliparous women in both discovery and validation phases of the study at an FDR of 10% and with at least a 1.25-fold change. These genes are involved in regulation of transcription, centrosome organization, RNA splicing, cell-cycle control, adhesion, and differentiation. The results provide initial evidence that full-term pregnancy induces long-term genomic changes in the breast. The genomic signature of pregnancy could be used as an intermediate marker to assess potential chemopreventive interventions with hormones mimicking the effects of pregnancy for prevention of breast cancer.


Assuntos
Regulação da Expressão Gênica , Genômica , Adulto , Idoso , Adesão Celular , Ciclo Celular , Feminino , Humanos , Pessoa de Meia-Idade , Análise de Sequência com Séries de Oligonucleotídeos , Paridade , Pós-Menopausa , Gravidez , Reprodutibilidade dos Testes
9.
Menopause ; 15(1): 67-73, 2008.
Artigo em Inglês | MEDLINE | ID: mdl-17558338

RESUMO

OBJECTIVE: This retrospective study systematically compared mammographic density with histology in women receiving or not receiving menopausal hormone therapy (HT). DESIGN: This study was approved by the institutional review board. Twenty-eight postmenopausal women using HT were matched with 28 postmenopausal women not using HT at the time of breast cancer diagnosis. Noncancerous tissue from mastectomy specimens was examined histologically to quantitate the content of fibrous stroma, ducts, and lobule types 1, 2, and 3. Tissue samples were also evaluated for estrogen receptor, progesterone receptor, and Ki67 activity in the ducts and lobules. Breast density was quantified by digitizing the contralateral mammogram and computer-assisted interactive thresholding. RESULTS: High breast density in women using HT was correlated with greater fibrous stroma (P = 0.020) and lobule type 1 (P = 0.016). Breast density also correlated with Ki67 activity in the ducts (P = 0.031) and lobules (P= 0.023) for both groups combined. Estrogen and progesterone receptors did not correlate with either breast density or HT use. CONCLUSIONS: Increased fibrous stroma and lobule type 1 are associated with increasing mammographic density in women using HT, independent of estrogen and progesterone receptor up-regulation. These findings suggest that increased breast density may be mediated through a paracrine effect. The increase in breast cancer risk with HT use may be due to an increase in target lobule type 1 cells.


Assuntos
Mama/efeitos dos fármacos , Mama/patologia , Receptor alfa de Estrogênio/metabolismo , Terapia de Reposição de Estrogênios/efeitos adversos , Receptores de Progesterona/metabolismo , Neoplasias da Mama/etiologia , Neoplasias da Mama/patologia , Estudos de Casos e Controles , Receptor alfa de Estrogênio/efeitos dos fármacos , Terapia de Reposição de Estrogênios/métodos , Feminino , Humanos , Interpretação de Imagem Assistida por Computador , Antígeno Ki-67/análise , Mamografia , Pessoa de Meia-Idade , Pós-Menopausa , Receptores de Progesterona/efeitos dos fármacos
10.
Int J Oncol ; 31(5): 1165-75, 2007 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-17912444

RESUMO

Our studies are aimed at determining whether pregnancy induces a specific genomic signature in the postmenopausal breast that is responsible for the protective effect elicited by this physiological process. For this purpose we designed a study to compare the gene expression profiles in normal breast tissue from parous postmenopausal women with (case) and without (control) breast cancer. We have used breast samples from 18 parous controls and 41 parous cases. The epithelium and the interlobular stroma were dissected using laser capture microdissection and the RNA of each compartment and each sample was isolated, amplified using PCR methodology, and hybridized to cDNA glass-microarrays containing 40,000 genes, placing the human reference RNA in the green channel (Cy3) and the breast tissue samples in the red channel (Cy5). The normalization and statistical analysis of the expression data were carried out by using the LIMMA software package for the R programming environment which provides functions to summarize the results using the linear model perform hypothesis tests and adjust the p-values for multiple testing. We were able to identify 126 genes that were upregulated and 103 that were downregulated in the parous control group. There were only 56 genes differentially expressed in the interlobular stroma in the parous control group in relation to the other group of women under study. The gene categories that were overrepresented in the breast epithelium of the parous control breast are related to apoptosis, DNA repair, response to exogenous agents and transcription regulation. In the present study we demonstrate that full-term pregnancy imprints a specific genomic signature in the breast epithelium of postmenopausal parous control women that is significantly different from women who have developed cancer. This genomic signature induced by pregnancy could help to predict in which women parity is protective.


Assuntos
Neoplasias da Mama/genética , Mama/metabolismo , Perfilação da Expressão Gênica , Paridade , Idoso , Apoptose , Reparo do DNA , Epitélio/metabolismo , Feminino , Humanos , Pessoa de Meia-Idade , Pós-Menopausa , Gravidez , Xenobióticos/metabolismo
11.
Int J Cancer ; 120(8): 1821-4, 2007 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-17230531

RESUMO

Metabolic conversion of endogenous estrogens, estradiol (E2) and estrone (E1), to the catechol estrogens 4-hydroxyE1(E2) [4-OHE1(E2)] has been implicated in the initiation of cancer in rodents and humans. Evidence collected in our laboratories has shown that 4-OHE1(E2) are enzymatically oxidized to E1(E2)-3,4-quinones [E1(E2)-3,4-Q], which have the potential to damage DNA by forming predominantly depurinating adducts, 4-OHE1(E2)-1-N3Ade and 4-OHE1(E2)-1-N7Gua, leading to the accumulation of mutations and probably cell transformation. The human breast epithelial cell line MCF-10F has been transformed by treatment with E2 or 4-OHE2. We have used MCF-10F cells to study the presence of adducts and conjugates after treatment with 4-OHE2. To mimic the intermittent exposure of breast cells to endogenous estrogens, MCF-10F cells were treated with 1 microM 4-OHE2 for a 24-h period at 72, 120, 192 and 240 h postplating. Culture media were collected at each point, extracted by solid-phase extraction and analyzed by HPLC connected with a multichannel electrochemical detector and/or ultraperformance liquid chromatography/tandem mass spectrometry. Media from successive treatments with 4-OHE2 showed the formation of methoxy and cysteine conjugates, and the depurinating adducts 4-OHE1(E2)-1-N3Ade. The amount of 4-OHE1(E2)-1-N3Ade adducts was higher after the third treatment; smaller amounts of the 4-OHE1(E2)-1-N7Gua adducts were detected after the second and third treatments. These results demonstrate that MCF-10F cells oxidize 4-OHE2 to E1(E2)-3,4-Q, which react with DNA to form the depurinating N3Ade and N7Gua adducts. This DNA damage can play an important role in the 4-OHE2-induced mutations and transformation of MCF-10F cells to malignant cells.


Assuntos
Adenina/química , Adutos de DNA/metabolismo , DNA/metabolismo , Estradiol/análogos & derivados , Estrogênios de Catecol/metabolismo , Guanina/química , Mama/citologia , Mama/metabolismo , Células Cultivadas , Cromatografia Líquida de Alta Pressão , DNA/efeitos dos fármacos , Dano ao DNA , Estradiol/química , Estradiol/metabolismo , Estradiol/farmacologia , Humanos , Estrutura Molecular , Extração em Fase Sólida , Espectrometria de Massas em Tandem
12.
Eur J Cancer Prev ; 15(4): 306-42, 2006 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-16835503

RESUMO

We have postulated that the lifetime protective effect of an early pregnancy against breast cancer is due to the complete differentiation of the mammary gland characterized by a specific genomic signature imprinted by the physiological process of pregnancy. In the present work, we show evidence that the breast tissue of postmenopausal parous women has had a shifting of stem cell 1 to stem cell 2 with a genomic signature different from similar structures derived from postmenopausal nulliparous women that have stem cell 1. Those genes that are significantly different are grouped in major categories on the basis of their putative functional significance. Among them are those gene transcripts related to immune surveillance, DNA repair, transcription, chromatin structure/activators/co-activators, growth factor and signal transduction pathway, transport and cell trafficking, cell proliferation, differentiation, cell adhesion, protein synthesis and cell metabolism. From these data, it was concluded that during pregnancy there are significant genomic changes that reflect profound alterations in the basic physiology of the mammary gland that explain the protective effect against carcinogenesis. The implication of this knowledge is that when the genomic signature of protection or refractoriness to carcinogenesis is acquired by the shifting of stem cell 1 to stem cell 2, the hormonal milieu induced by pregnancy or pregnancy-like conditions is no longer required. This is a novel concept that challenges the current knowledge that a chemopreventive agent needs to be given for a long period to suppress a metabolic pathway or abrogate the function of an organ.


Assuntos
Neoplasias da Mama/prevenção & controle , Mama/metabolismo , Expressão Gênica , Paridade/genética , Gravidez/genética , Apoptose/genética , Mama/citologia , Neoplasias da Mama/imunologia , Diferenciação Celular , Proliferação de Células , Reparo do DNA , Feminino , Perfilação da Expressão Gênica , Humanos , Modelos Biológicos , Paridade/fisiologia
13.
FASEB J ; 20(10): 1622-34, 2006 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-16873885

RESUMO

Breast cancer is a malignancy whose dependence on estrogen exposure has long been recognized even though the mechanisms whereby estrogens cause cancer are not clearly understood. This work was performed to determine whether 17beta-estradiol (E2), the predominant circulating ovarian steroid, is carcinogenic in human breast epithelial cells and whether nonreceptor mechanisms are involved in the initiation of breast cancer. For this purpose, the effect of four 24 h alternate periods of 70 nM E2 treatment of the estrogen receptor alpha (ER-alpha) negative MCF-10F cell line on the in vitro expression of neoplastic transformation was evaluated. E2 treatment induced the expression of anchorage-independent growth, loss of ductulogenesis in collagen, invasiveness in Matrigel, and loss of 9p11-13. Only invasive cells that exhibited a 4p15.3-16 deletion were tumorigenic. Tumors were poorly differentiated ER-alpha and progesterone receptor-negative adenocarcinomas that expressed keratins, EMA, and E-cadherin. Tumors and tumor-derived cell lines exhibited loss of chromosome 4, deletions in chromosomes 3p12.3-13, 8p11.1-21, 9p21-qter, and 18q, and gains in 1p, and 5q15-qter. The induction of complete transformation of MCF-10F cells in vitro confirms the carcinogenicity of E2, supporting the concept that this hormone could act as an initiator of breast cancer in women. This model provides a unique system for understanding the genomic changes that intervene for leading normal cells to tumorigenesis and for testing the functional role of specific genomic events taking place during neoplastic transformation.


Assuntos
Neoplasias da Mama/etiologia , Neoplasias da Mama/patologia , Transformação Celular Neoplásica/induzido quimicamente , Estradiol/efeitos adversos , Invasividade Neoplásica/patologia , Neoplasias da Mama/genética , Linhagem Celular , Aberrações Cromossômicas , Células Epiteliais/patologia , Receptor alfa de Estrogênio/deficiência , Feminino , Humanos , Receptores de Progesterona/deficiência
14.
Int J Oncol ; 28(2): 399-410, 2006 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-16391795

RESUMO

We have postulated that the lifetime protective effect of an early pregnancy against breast cancer is due to the complete differentiation of the mammary gland characterized by a specific genomic signature imprinted by the physiological process of pregnancy. For demonstrating this hypothesis we compared the genomic profile of the epithelium and the stroma of normal breast tissues from reduction mammoplasties performed in postmenopausal parous and nulliparous women. The epithelium and the stroma were separately dissected using laser capture microdissection (LCM) and the RNA of each compartment and each sample was isolated, amplified using PCR methodology, and hybridized to cDNA glass-microarrays containing 40,000 human cDNA features. The separation of the epithelial compartment from the interlobular stroma of Lob 1 using LCM allowed us to determine that the epithelial component contained 4,828 genes that were equally expressed in both nulliparous and parous women. There were 73 known genes that included immune-modulation-, DNA repair-, programmed cell death-, chromatin remodeling- and transcription-related genes, whereas in the breast of nulliparous women there were 20 different known genes that were upregulated. Our data provide evidence that breast tissues of postmenopausal parous women express in both the epithelial and the stromal compartments numerous genes that differ significantly from those present in breast tissues of post-menopausal nulliparous women, which could be important contributors to the genomic signature induced by an early full term pregnancy.


Assuntos
Mama/metabolismo , Regulação da Expressão Gênica , Paridade/genética , Mama/crescimento & desenvolvimento , Diferenciação Celular , Células Epiteliais/metabolismo , Feminino , Perfilação da Expressão Gênica , Humanos , Pessoa de Meia-Idade , Reação em Cadeia da Polimerase , Pós-Menopausa , Gravidez , Células Estromais/metabolismo , Regulação para Cima
15.
Front Biosci ; 11: 151-72, 2006 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-16146722

RESUMO

The breast attains its maximum development during pregnancy and lactation. After menopause the breast regresses in both nulliparous and parous women containing lobular structures that have been designated lobules type 1. Despite the similarity in the lobular composition of the breast at menopause, the fact that nulliparous women are at higher risk of developing breast cancer than parous women, indicates that Lobules type 1 in these two groups of women might be biologically different, or exhibit different susceptibility to carcinogenesis. Based on these observations it was postulated that the Lobule type 1 found in the breast of nulliparous women and of parous women with breast cancer never went through the process of differentiation, retaining a high concentration of epithelial cells that are targets for carcinogens and therefore susceptible to undergo neoplastic transformation, these cell are called Stem cells 1, whereas Lobules type 1 structures found in the breast of early parous postmenopausal women free of mammary pathology, on the other hand, are composed of an epithelial cell population that is refractory to transformation called Stem cells 2. It was further postulated that the degree of differentiation acquired through early pregnancy has changed the "genomic signature" that differentiates the Lobule type 1 from the early parous women from that of the nulliparous women by shifting the Stem cell 1 to a Stem cell 2 that is refractory to carcinogenesis, making this the postulated mechanism of protection conferred by early full term pregnancy. The identification of a putative breast stem cell (Stem cell 1) has reached in the last decade a significant impulse and several markers also reported for other tissues have been found in the mammary epithelial cells of both rodents and humans. Although still more work needs to be done in order to better understand the role of the Stem cell 2 and its interaction with the genes that confer it a specific signature, collectively, the data presently available provides evidence that pregnancy, through the process of cell differentiation, shifts the Stem cell 1 to Stem cell 2, cells that exhibit a specific genomic signature that could be responsible for the refractoriness of the mammary gland to carcinogenesis.


Assuntos
Glândulas Mamárias Animais/patologia , Glândulas Mamárias Humanas/patologia , Neoplasias/metabolismo , Células-Tronco/metabolismo , Animais , Neoplasias da Mama/metabolismo , Carcinoma Ductal de Mama/patologia , Diferenciação Celular , Linhagem Celular Tumoral , Transformação Celular Neoplásica , Células Epiteliais/metabolismo , Receptor alfa de Estrogênio/biossíntese , Receptor beta de Estrogênio/biossíntese , Feminino , Regulação da Expressão Gênica , Humanos , Modelos Biológicos , Gravidez , RNA , Ratos , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Fatores de Tempo
16.
Clin Cancer Res ; 11(2 Pt 2): 931s-6s, 2005 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-15701889

RESUMO

Sporadic breast cancer is a fatal disease most frequently diagnosed in American women from all ethnic groups, suggesting that primary prevention should be the ultimate goal for breast cancer control. We have developed a novel paradigm for breast cancer prevention arising from the well-established knowledge that an early first full-term pregnancy protects the breast against neoplastic transformation, as well as from our studies of the biological principle underlying this protection. We have shown experimentally that the first pregnancy induces the expression of a specific genomic signature in the breast that results from the completion of a cycle in this organ's differentiation driven by the reproductive process. This signature, in turn, is a biomarker associated with a possible overall lifetime decrease in breast cancer risk. We have shown in an experimental model that a short treatment with human chorionic gonadotropin, a placental hormone secreted during pregnancy, induces the same genomic signature that occurs in pregnancy, inhibiting not only the initiation but also the progression of mammary carcinomas, and stopping the development of early lesions such as intraductal proliferations and carcinoma in situ. These observations indicate that human chorionic gonadotropin given for a very short period, only until this genomic signature is acquired, has significant potential as a chemopreventive agent, protecting the normal cell from becoming malignant. This is a novel concept which challenges the current knowledge that a chemopreventive agent needs to be given for a long period of time to suppress a metabolic pathway or abrogate the function of an organ.


Assuntos
Neoplasias da Mama/prevenção & controle , Mama/crescimento & desenvolvimento , Diferenciação Celular/fisiologia , Transformação Celular Neoplásica , Animais , Mama/citologia , Suscetibilidade a Doenças , Células Epiteliais/citologia , Feminino , Substâncias de Crescimento , Humanos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...