Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
Más filtros

Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
Breast J ; 24(3): 343-349, 2018 05.
Artículo en Inglés | MEDLINE | ID: mdl-29139591

RESUMEN

To evaluate the MR appearance of noncalcified ductal carcinoma in situ (DCIS), with comparison to calcified DCIS. A retrospective, IRB-approved review of all DCIS diagnosed via MR biopsy between 2007 and 2011 was performed. DCIS was categorized as noncalcified based on the absence of calcifications on mammography and specimen radiography. MR morphology (focus, mass, nonmass enhancement [NME]) and enhancement kinetics (initial and delayed) for noncalcified DCIS were recorded and compared based on nuclear grade (1-3), size (<1.5 cm, 1.5-5 cm, >5 cm), and presence of necrosis. Imaging features of noncalcified and calcified DCIS were also compared. 115 cases of MR biopsy-proven DCIS were identified: 65 (56%) noncalcified and 50 (44%) calcified. For noncalcified DCIS, NME morphology was more common than mass or focus (60% vs 30.8% and 9.2%). There was a significant association between morphology and enhancement kinetics, with NME more likely demonstrating medium and persistent kinetics, and foci or masses demonstrating rapid and plateau or washout kinetics (P < .05). There was also a significant association between morphology and nuclear grade, with NME more likely seen with grade 3 DCIS (P = .024), and between size and initial enhancement, with lesions <1.5 cm more likely to have rapid initial enhancement (P = .0036). No significant difference was identified between calcified and noncalcified DCIS in terms of morphology, enhancement characteristics, nuclear grade, or presence of necrosis. The MR appearance of noncalcified DCIS closely mirrors that of calcified DCIS. Recognizing these imaging features may allow for improved identification of this MRI-detected abnormality, even in the absence of calcifications.


Asunto(s)
Neoplasias de la Mama/diagnóstico por imagen , Calcinosis/diagnóstico por imagen , Carcinoma Intraductal no Infiltrante/diagnóstico por imagen , Imagen por Resonancia Magnética/métodos , Neoplasias de la Mama/patología , Calcinosis/patología , Carcinoma Intraductal no Infiltrante/patología , Femenino , Humanos , Aumento de la Imagen , Biopsia Guiada por Imagen , Persona de Mediana Edad , Necrosis/diagnóstico por imagen , Necrosis/patología , Cuidados Preoperatorios
2.
Int J Cancer ; 140(11): 2484-2497, 2017 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-28263391

RESUMEN

Risk biomarkers for estrogen receptor (ER)-negative breast cancer have clear value for breast cancer prevention. We previously reported a set of lipid metabolism (LiMe) genes with high expression in the contralateral unaffected breasts (CUBs) of ER-negative cancer cases. We now further examine LiMe gene expression in both tumor and CUB, and investigate the role of Pre-B-cell leukemia homeobox-1 (PBX1) as a candidate common transcription factor for LiMe gene expression. mRNA was extracted from laser-capture microdissected epithelium from tumor and CUB of 84 subjects (28 ER-positive cases, 28 ER-negative cases, 28 healthy controls). Gene expression was quantitated by qRT-PCR. Logistic regression models were generated to predict ER status of the contralateral cancer. Protein expression of HMGCS2 and PBX1 was measured using immunohistochemistry. The effect of PBX1 on LiMe gene expression was examined by overexpressing PBX1 in MCF10A cells with or without ER, and by suppressing PBX1 in MDA-MB-453 cells. The expression of DHRS2, HMGCS2, UGT2B7, UGT2B11, ALOX15B, HPGD, UGT2B28 and GLYATL1 was significantly higher in ER-negative versus ER-positive CUBs, and predicted ER status of the tumor in test and validation sets. In contrast, LiMe gene expression was significantly lower in ER-negative than ER-positive tumors. PBX1 overexpression in MCF10A cells up-regulated most LiMe genes, but not in MCF10A cells overexpressing ER. Suppressing PBX1 in MDA-MB-453 cells resulted in decrease of LiMe gene expression. Four binding sites of PBX1 and cofactor were identified in three lipid metabolism genes using ChIP-qPCR. These data suggest a novel role for PBX1 in the regulation of lipid metabolism genes in benign breast, which may contribute to ER-negative tumorigenesis.


Asunto(s)
Neoplasias de la Mama/genética , Proteínas de Unión al ADN/genética , Metabolismo de los Lípidos/genética , Proteínas Proto-Oncogénicas/genética , Receptores de Estrógenos/genética , Adulto , Anciano , Línea Celular Tumoral , Femenino , Regulación Neoplásica de la Expresión Génica/genética , Humanos , Persona de Mediana Edad , Factor de Transcripción 1 de la Leucemia de Células Pre-B , ARN Mensajero/genética , Regulación hacia Arriba/genética
3.
Mol Cell Endocrinol ; 589: 112237, 2024 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-38599276

RESUMEN

The molecular basis of mullerian aplasia, also known as Mayer-Rokitansky-Kuster Hauser (MRKH) or congenital absence of the uterus and vagina, is largely unknown. We applied a multifaceted genetic approach to studying the pathogenesis of MRKH including exome sequencing of trios and duos, genome sequencing of families, qPCR, RT-PCR, and Sanger sequencing to detect intragenic deletions, insertions, splice variants, single nucleotide variants, and rearrangements in 132 persons with MRKH. We identified two heterozygous variants in ZNHIT3 localized to a commonly involved CNV region at chromosome 17q12 in two different families with MRKH. One is a frameshift, truncating variant that is predicted to interfere with steroid hormone binding of the LxxLL sequence of the C-terminal region. The second variant is a double missense/stopgain variant. Both variants impair protein expression in vitro. In addition, four more probands with MRKH harbored the stopgain variant without the nearby missense variant. In total, 6/132 (4.5%) of patients studied, including five with associated anomalies (type 2 MRKH), had ZNHIT3 variants that impair function in vitro. Our findings implicate ZNHIT3 as an important gene associated with MRKH within the 17q12 CNV region.


Asunto(s)
Trastornos del Desarrollo Sexual 46, XX , Cromosomas Humanos Par 17 , Anomalías Congénitas , Heterocigoto , Conductos Paramesonéfricos , Adolescente , Adulto , Femenino , Humanos , Trastornos del Desarrollo Sexual 46, XX/genética , Cromosomas Humanos Par 17/genética , Anomalías Congénitas/genética , Conductos Paramesonéfricos/anomalías , Linaje
4.
Histopathology ; 61(4): 660-6, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-22748158

RESUMEN

AIMS: Triple-negative breast cancers (TNBCs) are often poorly differentiated tumours that can present clinically as metastases of an unknown primary. Immunohistochemical panels are frequently used to determine the likelihood of a breast primary, but in this tumour subset cytokeratin (CK)7 may be the only positive finding. In this study we aimed to evaluate a commonly employed immunohistochemical panel using a large group of TNBCs (both basal-like and unclassified), and to analyse the CK7 staining patterns. METHODS AND RESULTS: Tissue microarrays containing 138 TNBCs were stained with antibodies against CK7, CK20, gross cystic disease fluid protein 15 (GCDFP-15), and mammaglobin. CK5/6 staining was used to identify basal-like tumours. CK7 staining was notably heterogeneous, with 14.5% of all cases demonstrating ≤20% tumour cell staining. A greater proportion of basal-like TNBCs than of unclassified TNBCs showed focal staining. GCDFP-15 and mammaglobin were not expressed in the majority of TNBCs, and were less frequently positive in basal-like than in unclassified TNBCs. CONCLUSION: TNBCs are commonly negative for most immunomarkers indicative of breast origin, with the exception of CK7. As about one in five TNBCs showed only focal CK7 positivity, use of this marker must be interpreted with caution, especially in small samples, so that the possibility of a breast primary is not overlooked.


Asunto(s)
Biomarcadores de Tumor/análisis , Queratina-7/biosíntesis , Neoplasias de la Mama Triple Negativas/metabolismo , Adulto , Anciano , Anciano de 80 o más Años , Femenino , Humanos , Inmunohistoquímica , Queratina-7/análisis , Persona de Mediana Edad , Análisis de Matrices Tisulares
5.
Acad Pathol ; 7: 2374289520912499, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32232128

RESUMEN

The following fictional case is intended as a learning tool within the Pathology Competencies for Medical Education (PCME), a set of national standards for teaching pathology. These are divided into three basic competencies: Disease Mechanisms and Processes, Organ System Pathology, and Diagnostic Medicine and Therapeutic Pathology. For additional information, and a full list of learning objectives for all three competencies, see http://journals.sagepub.com/doi/10.1177/2374289517715040.1.

6.
Clin Cancer Res ; 26(1): 25-34, 2020 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-31570566

RESUMEN

PURPOSE: Selective progesterone receptor modulators (SPRMs) show preclinical activity against hormone-sensitive breast cancer, but have not been tested in patients with early, treatment-naïve tumors. PATIENTS AND METHODS: In a double-blind presurgical window trial of oral telapristone acetate (TPA) 12 mg daily versus placebo, 70 patients with early-stage breast cancer were randomized 1:1 (stratified by menopause) and treated for 2 to 10 weeks. The primary endpoint was change in Ki67 between diagnostic biopsy and surgical specimens. Gene expression pre- and posttherapy was assessed using RNA-sequencing and gene set enrichment analysis was performed to determine pathways enriched in response to TPA and placebo treatments. RESULTS: Among 61 evaluable women (29 placebo and 32 telapristone acetate), 91% of tumors were ER/PR positive. The mean Ki67 declined by 5.5% in all women treated with telapristone acetate (P = 0.003), and by 4.2% in all women treated with placebo (P = 0.04). After menopausal stratification, the Ki67 decline remained significant in 22 telapristone acetate-treated premenopausal women (P = 0.03). Differential gene expression analysis showed no significant modulation overall. However, in a subset of tumors that demonstrated ≥30% relative reduction in Ki67 in the telapristone acetate group, genes related to cell-cycle progression, and those in the HER2 amplicon were significantly downregulated. In contrast, no significantly enriched pathways were identified in the placebo group. CONCLUSIONS: Patients treated with telapristone acetate whose Ki67 decreased by ≥30% demonstrated a selective antiproliferative signal, with a potentially important effect on HER2 amplicon genes. Evaluation of SPRMs in a neoadjuvant trial is merited, with attention to predictors of response to SPRM therapy, and inclusion of pre- and postmenopausal women.


Asunto(s)
Neoplasias de la Mama/tratamiento farmacológico , Antagonistas de Hormonas/uso terapéutico , Norpregnadienos/uso terapéutico , Receptores de Progesterona/antagonistas & inhibidores , Antineoplásicos/uso terapéutico , Biomarcadores de Tumor/genética , Biomarcadores de Tumor/metabolismo , Neoplasias de la Mama/genética , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Método Doble Ciego , Femenino , Perfilación de la Expresión Génica/métodos , Humanos , Antígeno Ki-67/metabolismo , Menopausia , Persona de Mediana Edad , Terapia Neoadyuvante/métodos , Estadificación de Neoplasias , Receptor ErbB-2/genética , Análisis de Secuencia de ARN/métodos , Resultado del Tratamiento
7.
Sci Rep ; 9(1): 20099, 2019 12 27.
Artículo en Inglés | MEDLINE | ID: mdl-31882810

RESUMEN

Approximately 0.5-1.4% of natal males and 0.2-0.3% of natal females meet DSM-5 criteria for gender dysphoria, with many of these individuals self-describing as transgender men or women. Despite recent improvements both in social acceptance of transgender individuals as well as access to gender affirming therapy, progress in both areas has been hampered by poor understanding of the etiology of gender dysphoria. Prior studies have suggested a genetic contribution to gender dysphoria, but previously proposed candidate genes have not yet been verified in follow-up investigation. In this study, we expand on the topic of gender identity genomics by identifying rare variants in genes associated with sexually dimorphic brain development and exploring how they could contribute to gender dysphoria. To accomplish this, we performed whole exome sequencing on the genomic DNA of 13 transgender males and 17 transgender females. Whole exome sequencing revealed 120,582 genetic variants. After filtering, 441 variants in 421 genes remained for further consideration, including 21 nonsense, 28 frameshift, 13 splice-region, and 225 missense variants. Of these, 21 variants in 19 genes were found to have associations with previously described estrogen receptor activated pathways of sexually dimorphic brain development. These variants were confirmed by Sanger Sequencing. Our findings suggest a new avenue for investigation of genes involved in estrogen signaling pathways related to sexually dimorphic brain development and their relationship to gender dysphoria.


Asunto(s)
Secuenciación del Exoma , Variación Genética , Estudio de Asociación del Genoma Completo , Personas Transgénero , Empalme Alternativo , Mapeo Cromosómico , Femenino , Mutación del Sistema de Lectura , Estudio de Asociación del Genoma Completo/métodos , Humanos , Masculino , Mutación Missense , Análisis de Secuencia de ADN , Procesos de Determinación del Sexo/genética
8.
Mol Cell Endocrinol ; 470: 151-159, 2018 07 15.
Artículo en Inglés | MEDLINE | ID: mdl-29050862

RESUMEN

Hypothalamic gonadotropin releasing hormone (GnRH) is crucial for the proper function of the hypothalamic-pituitary-gonadal (HPG) axis, subsequent puberty, and reproduction. When GnRH neuron migration or GnRH regulation is impaired, hypogonadotropic hypogonadism results. Mutations in the gene for nasal embryonic luteinizing hormone-releasing factor (NELF) have been identified in GnRH-deficient humans. NELF is a predominantly nuclear protein that may participate in gene transcription, but the genes NELF regulates are unknown. To address this question, RNA was extracted from NLT GnRH neuronal cells following either stable Nelf knockdown or scrambled control and subjected to cDNA arrays. Transcription factors and cell migration gene expression was altered most commonly. Members of the Janus kinase/signal transducers and activators of transcription (JAK/STAT) pathway, including Stat1, Stat2, Stat5a, Jak2, Irf7 and Irf9, were significantly down regulated as assessed by RT-qPCR. Protein levels of STAT1, phospho-STAT1, and JAK2 were reduced, but the protein level of phospho-JAK2 was not. These findings suggest a role for NELF in the regulation of the JAK/STAT signaling pathway, which have important functions in GnRH neurons.


Asunto(s)
Regulación de la Expresión Génica , Técnicas de Silenciamiento del Gen , Hormona Liberadora de Gonadotropina/metabolismo , Quinasas Janus/metabolismo , Neuronas/metabolismo , Factores de Transcripción STAT/metabolismo , Factores de Transcripción/metabolismo , Animales , Humanos , Ratones Transgénicos , Ratas , Reproducibilidad de los Resultados , Transducción de Señal
9.
Mol Cytogenet ; 11: 13, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29434669

RESUMEN

BACKGROUND: Little is known about the genetic contribution to Müllerian aplasia, better known to patients as Mayer-Rokitansky-Küster-Hauser (MRKH) syndrome. Mutations in two genes (WNT4 and HNF1B) account for a small number of patients, but heterozygous copy number variants (CNVs) have been described. However, the significance of these CNVs in the pathogenesis of MRKH is unknown, but suggests possible autosomal dominant inheritance. We are not aware of CNV studies in consanguineous patients, which could pinpoint genes important in autosomal recessive MRKH. We therefore utilized SNP/CGH microarrays to identify CNVs and define regions of homozygosity (ROH) in Anatolian Turkish MRKH patients. RESULTS: Five different CNVs were detected in 4/19 patients (21%), one of which is a previously reported 16p11.2 deletion containing 32 genes, while four involved smaller regions each containing only one gene. Fourteen of 19 (74%) of patients had parents that were third degree relatives or closer. There were 42 regions of homozygosity shared by at least two MRKH patients which was spread throughout most chromosomes. Of interest, eight candidate genes suggested by human or animal studies (RBM8A, CMTM7, CCR4, TRIM71, CNOT10, TP63, EMX2, and CFTR) reside within these ROH. CONCLUSIONS: CNVs were found in about 20% of Turkish MRKH patients, and as in other studies, proof of causation is lacking. The 16p11.2 deletion seen in mixed populations is also identified in Turkish MRKH patients. Turkish MRKH patients have a higher likelihood of being consanguineous than the general Anatolian Turkish population. Although identified single gene mutations and heterozygous CNVs suggest autosomal dominant inheritance for MRKH in much of the western world, regions of homozygosity, which could contain shared mutant alleles, make it more likely that autosomal recessively inherited causes will be manifested in Turkish women with MRKH.

10.
J Hematol Oncol ; 10(1): 89, 2017 04 17.
Artículo en Inglés | MEDLINE | ID: mdl-28412963

RESUMEN

BACKGROUND: Estrogen and progesterone are potent breast mitogens. In addition to steroid hormones, multiple signaling pathways input to estrogen receptor (ER) and progesterone receptor (PR) actions via posttranslational events. Protein kinases commonly activated in breast cancers phosphorylate steroid hormone receptors (SRs) and profoundly impact their activities. METHODS: To better understand the role of modified PRs in breast cancer, we measured total and phospho-Ser294 PRs in 209 human breast tumors represented on 2754 individual tissue spots within a tissue microarray and assayed the regulation of this site in human tumor explants cultured ex vivo. To complement this analysis, we assayed PR target gene regulation in T47D luminal breast cancer models following treatment with progestin (promegestone; R5020) and antiprogestins (mifepristone, onapristone, or aglepristone) in conditions under which the receptor is regulated by Lys388 SUMOylation (K388 intact) or is SUMO-deficient (via K388R mutation to mimic persistent Ser294 phosphorylation). Selected phospho-PR-driven target genes were validated by qRT-PCR and following RUNX2 shRNA knockdown in breast cancer cell lines. Primary and secondary mammosphere assays were performed to implicate phospho-Ser294 PRs, epidermal growth factor signaling, and RUNX2 in breast cancer stem cell biology. RESULTS: Phospho-Ser294 PR species were abundant in a majority (54%) of luminal breast tumors, and PR promoter selectivity was exquisitely sensitive to posttranslational modifications. Phospho-PR expression and target gene programs were significantly associated with invasive lobular carcinoma (ILC). Consistent with our finding that activated phospho-PRs undergo rapid ligand-dependent turnover, unique phospho-PR gene signatures were most prevalent in breast tumors clinically designated as PR-low to PR-null (luminal B) and included gene sets associated with cancer stem cell biology (HER2, PAX2, AHR, AR, RUNX). Validation studies demonstrated a requirement for RUNX2 in the regulation of selected phospho-PR target genes (SLC37A2). In vitro mammosphere formation assays support a role for phospho-Ser294-PRs via growth factor (EGF) signaling as well as RUNX2 as potent drivers of breast cancer stem cell fate. CONCLUSIONS: We conclude that PR Ser294 phosphorylation is a common event in breast cancer progression that is required to maintain breast cancer stem cell fate, in part via cooperation with growth factor-initiated signaling pathways and key phospho-PR target genes including SLC37A2 and RUNX2. Clinical measurement of phosphorylated PRs should be considered a useful marker of breast tumor stem cell potential. Alternatively, unique phospho-PR target gene sets may provide useful tools with which to identify patients likely to respond to selective PR modulators that block PR Ser294 phosphorylation as part of rational combination (i.e., with antiestrogens) endocrine therapies designed to durably block breast cancer recurrence.


Asunto(s)
Neoplasias de la Mama/patología , Genes Relacionados con las Neoplasias/genética , Células Madre Neoplásicas/patología , Procesamiento Proteico-Postraduccional , Receptores de Progesterona/metabolismo , Antiportadores/fisiología , Neoplasias de la Mama/metabolismo , Línea Celular Tumoral , Subunidad alfa 1 del Factor de Unión al Sitio Principal/fisiología , Femenino , Expresión Génica , Humanos , Ligandos , Células Madre Neoplásicas/metabolismo , Fosforilación , Serina/metabolismo , Análisis de Matrices Tisulares , Células Tumorales Cultivadas
11.
Cancer Prev Res (Phila) ; 10(1): 89-97, 2017 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-27789587

RESUMEN

miRNAs are noncoding RNAs with abnormal expression in breast cancer; their expression in high-risk benign breast tissue may relate to breast cancer risk. We examined miRNA profiles in contralateral unaffected breasts (CUB) of patients with breast cancer and validated resulting candidates in two additional sample sets. Expression profiles of 754 mature miRNAs were examined using TaqMan Low Density Arrays in 30 breast cancer samples [15 estrogen receptor (ER)-positive and 15 ER-negative] and paired CUBs and 15 reduction mammoplasty controls. Pairwise comparisons identified miRNAs with significantly differential expression. Seven candidate miRNAs were examined using qRT-PCR in a second CUB sample set (40 cases, 20 ER+, 20 ER-) and 20 reduction mammoplasty controls. Further validation was performed in 80 benign breast biopsy (BBB) samples; 40 from cases who subsequently developed breast cancer and 40 from controls who did not. Logistic regression, using tertiles of miRNA expression, was used to discriminate cases from controls. Seven miRNAs were differentially expressed in tumors and CUBs versus reduction mammoplasty samples. Among them, miR-18a and miR-210 were validated in the second CUB set, showing significantly higher expression in tumor and CUBs than in reduction mammoplasty controls. The expression of miR-18a and miR-210 was also significantly higher in BBB cases than in BBB controls. When both miR-18a and miR-210 were expressed in the upper tertiles in BBB, OR for subsequent cancer was 3.20, P = 0.023. miR-18a and miR-210 are expressed at higher levels in CUBs of patients with breast cancer, and in BBB prior to cancer development, and are therefore candidate breast cancer risk biomarkers. Cancer Prev Res; 10(1); 89-97. ©2016 AACR.


Asunto(s)
Neoplasias de la Mama/patología , Carcinogénesis/patología , MicroARNs/genética , Adulto , Anciano , Biomarcadores de Tumor/metabolismo , Biopsia , Mama , Progresión de la Enfermedad , Femenino , Perfilación de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Humanos , MicroARNs/metabolismo , Persona de Mediana Edad , Análisis de Secuencia por Matrices de Oligonucleótidos , Receptores de Estrógenos/metabolismo
12.
Fertil Steril ; 108(1): 145-151.e2, 2017 07.
Artículo en Inglés | MEDLINE | ID: mdl-28600106

RESUMEN

OBJECTIVE: To study the genetic cause of Mayer-Rokitansky-Kuster-Hauser syndrome (MRKH). Although a few candidate genes and genomic domains for have been reported for MRKH, the genetic underpinnings remain largely unknown. Some of the top candidate genes are WNT4, HNF1B, and LHX1. The goals of this study were to: 1) determine the prevalence of WNT4, HNF1B, and LHX1 point mutations, as well as new copy number variants (CNVs) in people with MRKH; and 2) identify and characterize MRKH cohorts. DESIGN: Laboratory- and community-based study. SETTING: Academic medical centers. PATIENT(S): A total of 147 MRKH probands and available family members. INTERVENTIONS(S): DNA sequencing of WNT4, HNF1B, and LHX1 in 100 MRKH patients, chromosomal microarray analysis in 31 North American MRKH patients, and characterization and sample collection of 147 North American and Turkish MRKH probands and their families. MAIN OUTCOME MEASURE(S): DNA sequence variants and CNVs; pedigree structural analysis. RESULT(S): We report finding CNVs in 6/31 people (∼19%) with MRKH, but no point mutations or small indels in WNT4, HNF1B, or LHX1 in 100 MRKH patients. Our MRKH families included 43 quads, 26 trios, and 30 duos. Of our MRKH probands, 87/147 (59%) had MRKH type 1 and 60/147 (41%) had type 2 with additional anomalies. CONCLUSION(S): Although the prevalence of WNT4, HNF1B, and LHX1 point mutations is low in people with MRKH, the prevalence of CNVs was ∼19%. Further analysis of our large familial cohort of patients will facilitate gene discovery to better understand the complex etiology of MRKH.


Asunto(s)
Trastornos del Desarrollo Sexual 46, XX/epidemiología , Trastornos del Desarrollo Sexual 46, XX/genética , Anomalías Congénitas/epidemiología , Anomalías Congénitas/genética , Factor Nuclear 1-beta del Hepatocito/genética , Proteínas con Homeodominio LIM/genética , Conductos Paramesonéfricos/anomalías , Polimorfismo de Nucleótido Simple/genética , Factores de Transcripción/genética , Proteína Wnt4/genética , Adulto , Estudios de Cohortes , Familia , Marcadores Genéticos/genética , Predisposición Genética a la Enfermedad/epidemiología , Predisposición Genética a la Enfermedad/genética , Humanos , Internacionalidad , Prevalencia , Factores de Riesgo , Adulto Joven
13.
Mol Cytogenet ; 9: 57, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27478502

RESUMEN

BACKGROUND: Mayer-Rokitansky-Kuster-Hauser (MRKH) syndrome, or the congenital absence of uterus and vagina, is the most severe anomaly of the female reproductive tract. It affects 1 in 5,000 females, and is the second most common cause of primary amenorrhea. The etiology remains unknown in most patients, although four single gene defects and some repetitive copy number variants (CNVs) have been identified. Translocations in MRKH patients are very rare, and reported only in three patients previously without breakpoint mapping. We have identified the fourth MRKH translocation patient and are the first to characterize the breakpoints mapped by molecular methods. RESULTS: The proband is a 17- year old white female with agenesis of the uterus and vagina who had a peripheral blood karyotype revealing a de novo balanced translocation 46,XX,t(3;16)(p22.3;p13.3)dn. There were no known related anomalies present in the proband or her family. No CNVs were found by chromosomal microarray analysis, and no genes were directly disrupted by the translocation. DNA sequencing of six nearby candidate genes-TRIM71, CNOT10, ZNF200, OR1F1, ZNF205, and ZNF213-did not reveal any mutations. RT-qPCR of proband lymphoblast RNA for 20 genes near the breakpoints of 3p22.3 and 16p13.3 showed significantly altered expression levels for four genes in the proband compared to three white female controls, after correction for multiple comparisons. Reduced expression was seen for CMTM7 and CCR4 on 3p22.3, while increased expression was observed for IL32 and MEFV on 16p13.3. CONCLUSION: We have mapped the breakpoints of our t(3;16)(p22.3;p13.3) translocation patient using molecular methods to within 13.6 kb at 3p22.3 and within 1.9 kb for 16p13.3 and have suggested 10 nearby genes that become plausible candidate genes for future study.

14.
Mol Cell Endocrinol ; 437: 86-96, 2016 12 05.
Artículo en Inglés | MEDLINE | ID: mdl-27502037

RESUMEN

The genetic basis is unknown for ∼60% of normosmic hypogonadotropic hypogonadism (nHH)/Kallmann syndrome (KS). DNAs from (17 male and 31 female) nHH/KS patients were analyzed by targeted next generation sequencing (NGS) of 261 genes involved in hypothalamic, pituitary, and/or olfactory pathways, or suggested by chromosome rearrangements. Selected variants were subjected to Sanger DNA sequencing, the gold standard. The frequency of Sanger-confirmed variants was determined using the ExAC database. Variants were classified as likely pathogenic (frameshift, nonsense, and splice site) or predicted pathogenic (nonsynonymous missense). Two novel FGFR1 mutations were identified, as were 18 new candidate genes including: AMN1, CCKBR, CRY1, CXCR4, FGF13, GAP43, GLI3, JAG1, NOS1, MASTL, NOTCH1, NRP2, PALM2, PDE3A, PLEKHA5, RD3, and TRAPPC9, and TSPAN11. Digenic and trigenic variants were found in 8/48 (16.7%) and 1/48 (2.1%) patients, respectively. NGS with confirmation by Sanger sequencing resulted in the identification of new causative FGFR1 gene mutations and suggested 18 new candidate genes in nHH/KS.


Asunto(s)
Estudios de Asociación Genética , Secuenciación de Nucleótidos de Alto Rendimiento/métodos , Hipogonadismo/genética , Síndrome de Kallmann/genética , Femenino , Humanos , Masculino , Mutación/genética , Linaje , Fenotipo
15.
Am J Clin Pathol ; 144(5): 722-6, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26486735

RESUMEN

OBJECTIVES: Bi- and multinucleated (B/M) cells are present in a variety of tumors. We evaluated lobular carcinoma in situ (classic and pleomorphic types) and ductal carcinoma in situ (DCIS) to determine if this objective morphologic feature aids the differential diagnosis. METHODS: The number of B/M cells was recorded in pleomorphic lobular carcinoma in situ (PLCIS) (n = 20), classic lobular carcinoma in situ (CLCIS) (n = 26), and DCIS (n = 37). RESULTS: Binucleated cells were significantly more frequent in PLCIS (100%) vs DCIS (43%; P < .0001) and CLCIS (54%; P = .0004). Multinucleated cells were present in 25% of PLCIS cases and 8% of DCIS cases, and they were absent in CLCIS. The quantity of B/M per high-power field (hpf) was less in DCIS (mean, 1.1) and CLCIS (mean, 2.5) compared with PLCIS (mean, 5.8). Thirty-five percent of PLCIS cases had more than five B/M per hpf. CONCLUSIONS: Binucleated cells are significantly more frequent in PLCIS vs CLCIS and DCIS. Multinucleated cells were never identified in CLCIS. PLCIS should be considered as a diagnosis when B/M is noted.


Asunto(s)
Neoplasias de la Mama/diagnóstico , Carcinoma Intraductal no Infiltrante/diagnóstico , Carcinoma Lobular/diagnóstico , Núcleo Celular/patología , Adulto , Anciano , Anciano de 80 o más Años , Biomarcadores de Tumor , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Cadherinas/metabolismo , Carcinoma Intraductal no Infiltrante/metabolismo , Carcinoma Intraductal no Infiltrante/patología , Carcinoma Lobular/metabolismo , Carcinoma Lobular/patología , Femenino , Humanos , Persona de Mediana Edad
16.
Mol Cell Endocrinol ; 407: 26-36, 2015 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-25731822

RESUMEN

Puberty and reproduction require proper signaling of the hypothalamic-pituitary-gonadal axis controlled by gonadotropin-releasing hormone (GnRH) neurons, which arise in the olfactory placode region and migrate along olfactory axons to the hypothalamus. Factors adversely affecting GnRH neuron specification, migration, and function lead to delayed puberty and infertility. Nasal embryonic luteinizing hormone-releasing factor (NELF) is a predominantly nuclear protein. NELF mutations have been demonstrated in patients with hypogonadotropic hypogonadism, but biallelic mutations are rare and heterozygous NELF mutations typically co-exist with mutations in another gene. Our previous studies in immortalized GnRH neurons supported a role for NELF in GnRH neuron migration. To better understand the physiology of NELF, a homozygous Nelf knockout (KO) mouse model was generated. Our findings indicate that female Nelf KO mice have delayed vaginal opening but no delay in time to first estrus, decreased uterine weight, and reduced GnRH neuron number. In contrast, male mice were normal at puberty. Both sexes of mice had impaired fertility manifested as reduced mean litter size. These data support that NELF has important reproductive functions. The milder than expected phenotype of KO mice also recapitulates the human phenotype since heterozygous NELF mutations usually require an additional mutation in a second gene to result in hypogonadotropic hypogonadism.


Asunto(s)
Sistema Hipotálamo-Hipofisario/metabolismo , Infertilidad/genética , Neuronas/metabolismo , Reproducción/genética , Factores de Transcripción/deficiencia , Útero/metabolismo , Animales , Recuento de Células , Movimiento Celular , Estro/genética , Femenino , Regulación de la Expresión Génica , Hormona Liberadora de Gonadotropina/biosíntesis , Hormona Liberadora de Gonadotropina/genética , Homocigoto , Humanos , Sistema Hipotálamo-Hipofisario/anomalías , Sistema Hipotálamo-Hipofisario/crecimiento & desarrollo , Infertilidad/fisiopatología , Tamaño de la Camada , Masculino , Ratones , Ratones Noqueados , Neuronas/patología , Maduración Sexual/genética , Transducción de Señal , Factores de Transcripción/genética , Útero/anomalías , Útero/crecimiento & desarrollo
17.
Cancer Chemother Pharmacol ; 76(6): 1235-46, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26560487

RESUMEN

PURPOSE: Women at high risk of breast cancer and those with carcinoma in situ need non-toxic, well-tolerated preventive interventions. One promising approach is drug delivery through the breast skin (local transdermal therapy, LTT). Our goal was to test novel drugs for LTT, to establish that LTT is applicable to non-steroidal drugs. METHODS: Athymic nude rats were treated with oral tamoxifen, transdermal 4-hydroxytamoxifen (4-OHT) or endoxifen gel applied daily to the axillary mammary gland for 6 weeks (Study 1). Study 2 was identical to Study 1, testing transdermal telapristone acetate (telapristone) gel versus subcutaneous implant. At euthanasia, mammary glands and blood were collected. In Study 3, consenting women requiring mastectomy were randomized to diclofenac patch applied to the abdomen or the breast for 3 days preoperatively. At surgery, eight tissue samples per breast were collected from predetermined locations, along with venous blood. Drug concentrations were measured using liquid chromatography-tandem mass spectroscopy. RESULTS: Mammary tissue concentrations of 4-OHT, endoxifen, and telapristone were significantly higher in the axillary glands of the gel-treated animals, compared to inguinal glands or to systemically treated animals. Plasma concentrations were similar in gel and systemically treated animals. The clinical trial showed significantly higher mammary concentrations when diclofenac was applied to the breast skin versus the abdominal skin, but concentrations were variable. CONCLUSIONS: These results demonstrate that lipophilic drugs can be developed for LTT; although the nude rat is suitable for testing drug permeability, delivery is systemic. In human, however, transdermal application to the breast skin provides local delivery.


Asunto(s)
Antineoplásicos/uso terapéutico , Neoplasias de la Mama/prevención & control , Mama/efectos de los fármacos , Carcinoma Intraductal no Infiltrante/tratamiento farmacológico , Administración Cutánea , Administración Oral , Adulto , Animales , Antineoplásicos/administración & dosificación , Mama/patología , Diclofenaco/administración & dosificación , Diclofenaco/uso terapéutico , Evaluación Preclínica de Medicamentos/métodos , Femenino , Geles , Humanos , Glándulas Mamarias Animales/efectos de los fármacos , Persona de Mediana Edad , Norpregnadienos/administración & dosificación , Norpregnadienos/farmacología , Evaluación de Resultado en la Atención de Salud , Proyectos Piloto , Periodo Preoperatorio , Distribución Aleatoria , Ratas Desnudas , Tamoxifeno/administración & dosificación , Tamoxifeno/análogos & derivados , Tamoxifeno/farmacología
18.
Nat Commun ; 6: 8094, 2015 Sep 08.
Artículo en Inglés | MEDLINE | ID: mdl-26348915

RESUMEN

Breast cancer is a leading cause of death for women, with mortality resulting from metastasis. Metastases are often detected once tumour cells affect the function of solid organs, with a high disease burden limiting effective treatment. Here we report a method for the early detection of metastasis using an implanted scaffold to recruit and capture metastatic cells in vivo, which achieves high cell densities and reduces the tumour burden within solid organs 10-fold. Recruitment is associated with infiltration of immune cells, which include Gr1(hi)CD11b(+) cells. We identify metastatic cells in the scaffold through a label-free detection system using inverse spectroscopic optical coherence tomography, which identifies changes to nanoscale tissue architecture associated with the presence of tumour cells. For patients at risk of recurrence, scaffold implantation following completion of primary therapy has the potential to identify metastatic disease at the earliest stage, enabling initiation of therapy while the disease burden is low.


Asunto(s)
Adenocarcinoma/diagnóstico , Materiales Biocompatibles , Neoplasias de la Mama/diagnóstico , Neoplasias Hepáticas/diagnóstico , Neoplasias Pulmonares/diagnóstico , Células Neoplásicas Circulantes , Andamios del Tejido , Adenocarcinoma/secundario , Animales , Neoplasias de la Mama/patología , Modelos Animales de Enfermedad , Detección Precoz del Cáncer , Femenino , Humanos , Neoplasias Hepáticas/secundario , Neoplasias Pulmonares/secundario , Ratones , Metástasis de la Neoplasia , Trasplante de Neoplasias , Prótesis e Implantes , Tomografía de Coherencia Óptica , Carga Tumoral
19.
Hum Pathol ; 45(3): 583-8, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24444467

RESUMEN

Intraductal papillomas (IDPs) of the breast can be associated with a variety of clinical symptoms and radiologic findings. Surgical excision is often recommended based on the possibility of an associated high-grade lesion. Although the rate of upgrades has been extensively evaluated for IDPs, many studies are hindered by broad inclusion criteria, a lack of pathologic-radiologic concordance, and no standard definition of what constitutes an upgrade. In the current study, we evaluate the risk of upgrade for a specific subset of IDPs: non-mass-associated IDPs. We identified all breast needle core biopsies with a diagnosis of IDP between 2003 and 2010. Patients with associated masses, architectural distortion, or ipsilateral breast cancer were excluded. All needle core biopsy slides and relevant imaging studies were reviewed to ensure pathologic-radiologic concordance. Excision pathology was also reviewed; an upgrade was defined as the presence of ductal carcinoma in situ or invasive carcinoma in the excision. Seventy-nine IDPs that met inclusion criteria were identified and were further divided into 3 histologic categories: micropapilloma, fragmented IDP, and atypical IDP. Micropapillomas and fragmented IDPs had no upgrades (0/37). In patients who did not undergo excision, none subsequently developed ipsilateral breast cancer (follow-up, 50-61 months). This is in contrast to atypical IDPs that had a 33% upgrade rate. One patient with an unexcised atypical IDP developed ipsilateral breast cancer within 2 years. Our data suggest that conservative follow-up is reasonable for non-mass-associated IDPs without atypia regardless of microscopic size, provided that careful pathologic-radiologic correlation is achieved.


Asunto(s)
Neoplasias de la Mama/cirugía , Mama/cirugía , Papiloma Intraductal/cirugía , Adulto , Anciano , Anciano de 80 o más Años , Biopsia , Mama/patología , Neoplasias de la Mama/patología , Progresión de la Enfermedad , Femenino , Humanos , Persona de Mediana Edad , Papiloma Intraductal/patología
20.
Clin Cancer Res ; 20(14): 3672-82, 2014 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-25028506

RESUMEN

PURPOSE: Local transdermal therapy to the breast may achieve effective target-organ drug delivery, while diminishing systemic effects. We conducted a randomized, double-blind, placebo-controlled phase II trial comparing transdermal 4-hydroxytamoxifen gel (4-OHT) to oral tamoxifen (oral-T) in women with ductal carcinoma in situ (DCIS). METHODS: Twenty-seven pre- and postmenopausal women were randomized to 4-OHT (4 mg/day) or oral-T (20 mg/day) for 6 to 10 weeks before surgery. Plasma, nipple aspirate fluid, and breast adipose tissue concentrations of tamoxifen and its major metabolites were determined by liquid chromatography/tandem mass spectrometry. The primary endpoint was Ki67 labeling in DCIS lesions, measured by immunohistochemistry. In plasma, insulin-like growth factor-1 (IGFI), sex hormone-binding globulin (SHBG), and coagulation protein concentrations were determined. RESULTS: Posttherapy Ki67 decreased by 3.4% in the 4-OHT and 5.1% in the oral-T group (P ≤ 0.03 in both, between-group P = 0. 99). Mean plasma 4-OHT was 0.2 and 1.1 ng/mL in 4-OHT and oral groups, respectively (P = 0.0003), whereas mean breast adipose tissue concentrations of 4-OHT were 5.8 ng/g in the 4-OHT group and 5.4 ng/g in the oral group (P = 0.88). There were significant increases in plasma SHBG, factor VIII, and von Willebrand factor and a significant decrease in plasma IGFI with oral-T, but not with 4-OHT. The incidence of hot flashes was similar in both groups. CONCLUSIONS: The antiproliferative effect of 4-OHT gel applied to breast skin was similar to that of oral-T, but effects on endocrine and coagulation parameters were reduced. These findings support the further evaluation of local transdermal therapy for DCIS and breast cancer prevention.


Asunto(s)
Antineoplásicos Hormonales/administración & dosificación , Neoplasias de la Mama/tratamiento farmacológico , Carcinoma Intraductal no Infiltrante/tratamiento farmacológico , Tamoxifeno/análogos & derivados , Administración Cutánea , Administración Oral , Anciano , Antineoplásicos Hormonales/farmacocinética , Biomarcadores de Tumor/sangre , Método Doble Ciego , Femenino , Humanos , Persona de Mediana Edad , Tamoxifeno/administración & dosificación , Tamoxifeno/farmacocinética , Resultado del Tratamiento
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA