Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
1.
Int J Cancer ; 131(7): 1528-36, 2012 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-22223266

RESUMEN

Recurrent chromosomal alterations are found in roughly 20% of all uterine fibroids but in the majority cytogenetic changes are lacking. Recently, mutations of the gene mediator subcomplex 12 (MED12) have been detected in a majority of fibroids but no information is available whether or not they co-occur with cytogenetic subtypes as, e.g., rearrangements of the genes encoding high mobility group AT-hook (HMGA) proteins. In a total of 80 cytogenetically characterized fibroids from 50 patients, we were not only able to confirm the frequent occurrence of MED12 mutations but also to stratify two mutually exclusive pathways of leiomyomagenesis with either rearrangements of HMGA2 reflected by clonal chromosome abnormalities affecting 12q14~15 or by mutations affecting exon 2 of MED12. On average the latter mutations were associated with a significantly smaller tumor size. However, G>A transitions of nucleotides c.130 or c.131 correlate with a significantly larger size of the fibroids compared to other MED12 mutations thus explaining the high prevalence of the former mutations among clinically detectable fibroids. Interestingly, fibroids with MED12 mutations expressed significantly higher levels of the gene encoding wingless-type MMTV integration site family, member 4 (WNT4). Based on these findings and data from the literature, we hypothesize that estrogen and the mutated MED12 cooperate in activating the Wnt pathway which in turn activates ß-catenin known to cause leiomyoma-like lesions in a mouse model. The occurrence of a "fibroid-type mutation" in a rare histologic subtype of endometrial polyps suggests that this mechanism is not confined to uterine leiomyomas.


Asunto(s)
Leiomioma/genética , Complejo Mediador/genética , Mutación , Neoplasias Uterinas/genética , Adenomioma/genética , Anciano , Secuencia de Bases , Células Cultivadas , Bandeo Cromosómico , Neoplasias Endometriales/genética , Neoplasias Endometriales/patología , Femenino , Genotipo , Proteína HMGA2/genética , Humanos , Cariotipo , Lipoma/genética , Pólipos/genética , Recombinación Genética , Proteína Wnt4/genética
2.
Biochem Biophys Res Commun ; 422(3): 411-6, 2012 Jun 08.
Artículo en Inglés | MEDLINE | ID: mdl-22575509

RESUMEN

MicroRNAs of the chromosome 19 microRNA cluster (C19MC) are known to be abundantly expressed in the placenta. Their genes are located on the long arm of chromosome 19 and seem to be part of a large imprinted region. Although the data available so far suggest important functions in the placenta, no data are available on their general expression patterns in cultures of placenta-derived mesenchymal stromal cells (PDMSC). Surprisingly, qRT-PCR on tissue cultures from first-trimester and term placenta mesenchymal stromal cells showed an abundant expression of the cluster members miR-517a-3p, miR-519a-3p, and miR-520c-3p. Accordingly, analyses of methylation patterns suggested that these cells had escaped methylation and epigenetic silencing, respectively, of the paternal allele. This was confirmed by the results of treatment of chorionic villous stromal cells by the demethylating agent 5-Aza-2'-deoxycytidine. Our results offer clear evidence that, in contrast to what is suggested in previous papers, members of C19MC are highly expressed in PDMSC indicating that their placenta-specific functions are not restricted to the trophoblast.


Asunto(s)
Cromosomas Humanos Par 19/genética , Metilación de ADN , Regulación de la Expresión Génica , MicroARNs/genética , Placenta/metabolismo , Secuencia de Bases , Línea Celular Tumoral , Células Cultivadas , Vellosidades Coriónicas/metabolismo , Islas de CpG/genética , Femenino , Humanos , Datos de Secuencia Molecular , Placenta/citología , Embarazo , Células del Estroma/metabolismo
3.
Mol Hum Reprod ; 17(2): 135-42, 2011 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-20926602

RESUMEN

In human fibroids genes encoding the high-mobility proteins containing the 'AT-hook' DNA-binding motif (HMGA) are frequently affected by non-random chromosomal rearrangements. Thus, the different proteins and their derivatives resulting from these genomic rearrangements can be assumed to be involved in the genesis of these tumors by activation of largely identical downstream pathways. Constructs encoding HMGA proteins and their relevant derivatives were overexpressed in human myometrial cells, and RNA isolated from these cells was hybridized to filter arrays. Four genes were either up- or down-regulated at least 2-fold after overexpression of either of the HMGA genes and their derivatives. FGF2 (fibroblast growth factor 2) was one of these genes, and we were then able to show by microarray analyses that tumors with rearrangements of the HMGA2 locus (n = 8) expressed significantly higher levels of FGF2 than those with an apparently normal karyotype (n = 47). Accordingly, by quantitative real-time PCR uterine leiomyomas with rearrangements of the HMGA2 locus were found to express significantly higher levels of FGF2 than those with an apparently normal karyotype with a linear relationship between the expression of FGF2 and the level of HMGA2 overexpression as well as the tumor size. The results of western blot analyses confirmed these findings. Moreover, stimulation of myometrial tissue by FGF1, a strong inducer of HMGA2, leads to an increase of HMGA2 as well as FGF2 expression. In conclusion, the results contribute to the understanding of the association between the overexpression of HMGA proteins, the regulation of FGF2 expression and the size of fibroids.


Asunto(s)
Factor 2 de Crecimiento de Fibroblastos/genética , Proteínas HMGA/genética , Proteínas HMGA/metabolismo , Proteína HMGA2/genética , Leiomioma/genética , Miometrio/metabolismo , Neoplasias Uterinas/genética , Western Blotting , Células Cultivadas , Femenino , Factor 1 de Crecimiento de Fibroblastos/farmacología , Factor 2 de Crecimiento de Fibroblastos/metabolismo , Regulación Neoplásica de la Expresión Génica , Proteína HMGA2/metabolismo , Humanos , Leiomioma/metabolismo , Leiomioma/patología , Análisis por Micromatrices , Mutación , Reacción en Cadena de la Polimerasa , Neoplasias Uterinas/metabolismo , Neoplasias Uterinas/patología
4.
Cytokine ; 56(3): 811-6, 2011 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-22019704

RESUMEN

BMP4 has been linked to early steps of adipocyte lineage differentiation but only little is known about its corresponding downstream pathways. Herein, we have investigated whether or not the expression of high mobility group protein HMGA2, another protein linked to proliferation and differentiation within the process of adipogenesis, may be influenced by BMP4 signaling in adipose tissue derived stem cells. Compared to FGF1, a strong inducer of HMGA2 in immortalized pre-adipocytes, BMP4 was found moderately to induce the HMGA2 mRNA expression in serum starved adipose tissue derived stem cells and myometrial cells. In contrast, no such activity was noted in canine bone marrow derived mesenchymal stem cells. As to adipocyte lineage differentiation the functions of BMP4 and HMGA2 mechanistically overlap. Thus, we propose that in adipose tissue BMP4 acts in part by activating HMGA2 making this architectural transcription factor one of the major downstream players in that system.


Asunto(s)
Proteína Morfogenética Ósea 4/farmacología , Proteína HMGA2/genética , Células Madre Mesenquimatosas/efectos de los fármacos , Células Madre Mesenquimatosas/metabolismo , Tejido Adiposo/citología , Animales , Western Blotting , Células de la Médula Ósea/citología , Células de la Médula Ósea/efectos de los fármacos , Células de la Médula Ósea/metabolismo , Proliferación Celular/efectos de los fármacos , Perros , Femenino , Factor 1 de Crecimiento de Fibroblastos/farmacología , Regulación de la Expresión Génica/efectos de los fármacos , Proteína HMGA2/metabolismo , Humanos , Células Madre Mesenquimatosas/citología , Miometrio/citología , Miometrio/efectos de los fármacos , Miometrio/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , ARN Interferente Pequeño/metabolismo
5.
Genes Chromosomes Cancer ; 49(8): 661-8, 2010 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-20544840

RESUMEN

Pathogenetically, uterine leiomyomas (ULs) can be interpreted as the result of a monoclonal abnormal proliferation of myometrial cells. Oncogene-induced senescence (OIS) is a frequent phenomenon in premalignant lesions that leads to a growth arrest mainly by the activation of two potent growth-inhibitory pathways as represented by p16(Ink4a) and p19(Arf). The relevance of OIS for the development of UL has not been addressed, but HMGA2, encoded by a major target gene of recurrent chromosomal abnormalities in UL, has been implicated in the repression of the Ink4a/Arf (CDKN2A) locus. This prompted us to examine if HMGA2 contributes to the growth of leiomyomas by repressing this locus. Contrary to the expectations, we were able to show that generally ULs express significantly higher levels of p19(Arf) mRNA than myometrium and that UL with 12q14 approximately 15 rearrangements showed higher expression levels than UL with other cytogenetic aberrations. Furthermore, the finding of a significant correlation between the expressions of p19(Arf) and CDKN1A shows that p19(Arf) triggers senescence rather than apoptosis in UL. Furthermore, the expression levels of HMGA2, p19(Arf), and CDKN1A were found to be correlated with the size of the tumors, indicating that an enhanced growth potential is counterbalanced by the p19(Arf) pathway. Mechanistically, the UL may thus execute a program already present in their cell of origin, where it is activated to protect the genome, for example, in the case of enhanced proliferation. In summary, the results identify the p19(Arf)-TP53-CDKN1A pathway as a major player in the growth control and genomic stability of uterine fibroids.


Asunto(s)
Inhibidor p16 de la Quinasa Dependiente de Ciclina/genética , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/genética , Proteína HMGA2/genética , Leiomioma/genética , Proteína p53 Supresora de Tumor/genética , Neoplasias Uterinas/genética , Apoptosis , Senescencia Celular , Cromosomas Humanos Par 12/genética , Femenino , Reordenamiento Génico , Humanos , Leiomioma/patología , Leiomioma/cirugía , Miometrio/metabolismo , Miometrio/patología , ARN Mensajero/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Neoplasias Uterinas/patología , Neoplasias Uterinas/cirugía
6.
Histopathology ; 54(2): 233-40, 2009 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-19207948

RESUMEN

AIMS: Anorectal melanoma (AM) forms a rare but highly malignant subset of mucosal melanoma with an extremely poor prognosis. Although AMs display histological and immunohistochemical features very similar to cutaneous melanoma (CM), no association exists either with exposure to ultraviolet light or with melanocytic naevi. While AMs are clearly distinguished from CM by displaying few BRAF mutations, they are commonly indistinguishable from CM at the level of gene expression. The aim was to carry out expression analyses of classical immunohistochemical markers and of the protein deleted in malignant brain tumours 1 (DMBT1) in cases of primary anorectal malignant melanoma and CM. METHODS AND RESULTS: Expression analyses of classical immunohistochemical markers (S100, HMB45, Melan A and MiTF) and of the protein DMBT1 were carried out in 27 cases of primary anorectal malignant melanoma and 26 cases of CM. All AM cases analysed showed expression of at least three of the classical markers for melanoma. However, immunohistochemistry showed 19 out of 27 AM to be positive for DMBT1, which represented a statistically significant difference (P = 0.0009) compared with CM (six out of 26), which more commonly are negative for DMBT1 expression. CONCLUSION: These results identify DMBT1 as a molecular feature that may allow distinction between AM and CM and support the notion that AM represents an entity molecularly distinct from CM.


Asunto(s)
Neoplasias del Ano/diagnóstico , Biomarcadores de Tumor/análisis , Melanoma/diagnóstico , Receptores de Superficie Celular/biosíntesis , Neoplasias Cutáneas/diagnóstico , Adulto , Anciano , Anciano de 80 o más Años , Neoplasias del Ano/metabolismo , Proteínas de Unión al Calcio , Proteínas de Unión al ADN , Femenino , Humanos , Inmunohistoquímica , Masculino , Melanoma/metabolismo , Persona de Mediana Edad , Neoplasias Cutáneas/metabolismo , Proteínas Supresoras de Tumor
7.
Int J Cancer ; 122(3): 547-57, 2008 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-17918156

RESUMEN

Recent studies have shown that inhibition of cyclooxygenases (e.g. COX-2) exerts antitumorigenic effects on hepatocellular carcinomas (HCCs), which are to a significant extent due to the abrogation of PGE(2) synthesis. PGE(2) acts via differentially regulated prostaglandin receptors (EP(1-4)). Our study was designed to investigate the expression pattern of EP-receptors in HCCs and to evaluate the therapeutic potential of selective EP-receptor antagonists. Using tissue microarrays including a total of 14 control livers, 17 liver cirrhoses, 22 premalignant dysplastic nodules (DNs) and 162 HCCs with different histological grades, the expression of COX-2, mPGES-1 and -2 and EP(1-4)-receptors was analyzed. Western immunoblot analyses were performed to confirm the expression in HCC cell lines. The effects of EP(1-4)-receptor antagonism on cell viability and apoptosis were investigated using MTT-assays and FACS-analyses, respectively. COX-2, mPGES-1 and -2 and EP(1-4)-receptors were expressed in all HCC tissues. COX-2 expression was highest in DNs and declined with loss of HCC-differentiation. With respect to COX-2 expression, a converse expression of EP(1-3) -receptors and mPGES-1 and -2 was found in DNs compared to HCCs. Selectively antagonizing EP(1)- and EP(3)-receptors reduced the viability of HCC cells in a dose-dependent manner, which was associated with apoptosis induction. Our results suggest a differential regulation of EP-receptor subtype expression with dedifferentiation of HCCs in which a converse expression pattern for COX-2 in comparison to EP(1-3)-receptors occurs. Of clinical interest, selectively antagonizing EP(1)- and EP(3)-receptors may provide a novel systemic therapeutic approach to the treatment of HCCs.


Asunto(s)
Apoptosis/efectos de los fármacos , Carcinoma Hepatocelular/metabolismo , Ciclooxigenasa 2/metabolismo , Receptores de Prostaglandina E/metabolismo , Western Blotting , Carcinoma Hepatocelular/etiología , Carcinoma Hepatocelular/patología , Supervivencia Celular/efectos de los fármacos , Femenino , Humanos , Técnicas para Inmunoenzimas , Oxidorreductasas Intramoleculares/metabolismo , Hígado/efectos de los fármacos , Hígado/metabolismo , Hígado/patología , Cirrosis Hepática/etiología , Cirrosis Hepática/metabolismo , Cirrosis Hepática/patología , Neoplasias Hepáticas/etiología , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/patología , Masculino , Prostaglandina-E Sintasas , Análisis de Matrices Tisulares
8.
Oncotarget ; 8(21): 34762-34772, 2017 May 23.
Artículo en Inglés | MEDLINE | ID: mdl-28410233

RESUMEN

Uterine leiomyomas (UL) are the most prevalent symptomatic human tumors at all and somatic mutations of the gene encoding mediator subcomplex 12 (MED12) constitute the most frequent driver mutations in UL. Recently, a rapid loss of mutated cells during in vitro growth of UL-derived cell cultures was reported, resulting in doubts about the benefits of UL-derived cell cultures. To evaluate if the rapid loss of MED12-mutated cells in UL cell cultures depends on in vitro passaging, we set up cell cultures from nine UL from 40-50 year old Caucasian patients with at least one UL. Cultured UL cells were investigated for loss of MED12-mutated cells. Genetic characterization of native tumor samples and adjacent myometrium was done by array analysis. "Aged" primary cultures without passaging were compared to cells of three subsequent passages. Comparative analyses of the mutated/non-mutated ratios between native tissue, primary cells, and cultured tumor cells revealed a clear decrease of MED12-mutated cells. None of the tumors showed gross alterations of the array profiles, excluding the presence of gross genomic imbalances besides the MED12 mutations as a reason for the intertumoral variation in the loss of MED12-mutated cells. Albeit at a lesser rate, loss of MED12-mutated cells from cell cultures of UL occurs even without passaging thus indicating the requirement of soluble factors or matrix components lacking in vitro. Identification of these factors can help to understand the mechanisms of the growth of the most frequent type of uterine leiomyomas and to decipher novel drug targets.


Asunto(s)
Técnicas de Cultivo de Célula/métodos , Leiomioma/genética , Complejo Mediador/genética , Mutación , Neoplasias Uterinas/genética , Proliferación Celular , Femenino , Inestabilidad Genómica , Humanos , Técnicas In Vitro , Células Tumorales Cultivadas
9.
Mol Cytogenet ; 8: 76, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26468330

RESUMEN

Mutations of mediator subcomplex 12 (MED12) and of high mobility group protein AT-hook 2 (HMGA2) are driver mutations in uterine leiomyomas (UL) that have not been observed to coexist in one tumor and even rarely coexist in different UL tumors of one patient. Here we describe a patient who underwent hysterectomy because of multiple leiomyomas which were studied by cytogenetics, MED12 hotspot sequencing, and copy number variation arrays. Two of the UL tumors had different HMGA2 rearrangements not detected by G-banding. Two UL tumors had deletions of the long arm of chromosome 3, in one case associated with a MED12 mutation. Both deletions lead to the loss of MED12L showing strong similarity with MED12. It remains to be determined if this gene can play a role in leiomyomagenesis independent of MED12. In summary, the patient presented exhibits an unusual coincidence of different driver mutations among her leiomyomas.

10.
Mol Cytogenet ; 7(1): 19, 2014 Mar 03.
Artículo en Inglés | MEDLINE | ID: mdl-24593849

RESUMEN

BACKGROUND: Epitheloid leiomyoma is a rare subtype of benign smooth muscle tumors. RESULTS: Herein, we present the results of classical cytogenetics, MED12 mutation analysis, and copy number variation array evaluation in one such case. Whereas cytogenetic did not show evidence for clonal chromosome abnormalities and no MED12 mutation in the "fibroid hot spot" region was detected, array hybridization revealed multiple abnormalities. Most noteworthy, almost all chromosomes showed copy-number neutral loss of heterozygosity. As examples of further abnormalities, trisomies of chromosomes 8, 12, 20, and X were noted. DISCUSSION: The data presented suggest a near-haploid karyotype of the tumor as the initial genetic alteration followed by secondary duplications of large parts of the genome. The absence of any clonal karyotypic alterations after performing classical cytogenetics is likely explained by a reduced ability of the tumor cells to proliferate in vitro. However, to the best of our knowledge this is the first report of an uterine leiomyoma showing extended uniparental disomy. It remains to be determined if this is a more common phenomenon in epithelioid leiomyomas or even subsets of "ordinary" leiomyomas.

11.
Mol Cytogenet ; 7(1): 88, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25506394

RESUMEN

BACKGROUND: Recent findings on genetic changes in uterine leiomyomas suggest these benign tumors being a heterogeneous group of diseases in terms of molecular pathogenesis with those showing karyotype alterations as well as those characterized only by cytogenetically invisible mutations of mediator subcomplex 12 (MED12). Herein, five uterine leiomyomas (UL) with an apparently normal karyotype that lacked MED12-mutations were investigated by copy number variation arrays along with their matching myometrium to search for small genomic imbalances. RESULTS: Of five tumors one showed chromothripsis-like phenomena with numerous gains and losses of small segments mainly clustered to five chromosomal regions i.e. 2p14-2pter, 2q33.1-2q37.3, 5q31.3-5qter,11q14.1-11qter, and 18p11.21-18q2.3. Apparently, these cells had escaped detection by classical cytogenetics. Histologically, the tumor presented as a cellular leiomyoma with extended hyalinization. Of the remaining four tumors, one had a small intragenic deletion of the HMGA2 gene that was lacking in the corresponding myometrium. The other three tumors did not show relevant copy number alterations at all. CONCLUSIONS: Overall, the results suggest that leiomyomas with an apparently normal karyotype based on classical cytogenetics and lacking MED12 mutations represent a heterogeneous group of diseases. While the HMGA2 deletion detected in one of the tumors likely represents the driver mutation and, due to its size, has escaped detection by classical cytogenetics, the extended genomic imbalances detected in one of the other cases cannot be overlooked by this method suggesting an inability of the affected cells to divide in vitro. Of particular interest in that case is the occurrence of so-called "chromothripsis" or "firestorms" without involvement of the loci of common chromosomal rearrangements in UL, as e.g. 12q14 ~ 15 and 6p21. While chromothripsis was initially described as a hallmark of malignancy, the etiology and significance of this phenomenon in benign tumors still remain obscure. In uterine smooth muscle tumors, these changes per se do not indicate malignancy.

12.
PLoS One ; 9(2): e88126, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24516594

RESUMEN

In pleomorphic adenomas of the salivary glands (PASG) recurrent chromosomal rearrangements affecting either 8q12 or 12q14∼15 lead to an overexpression of the genes of the genuine transcription factor PLAG1 or the architectural transcription factor HMGA2, respectively. Both genes are also affected by recurrent chromosomal rearrangements in benign adipocytic tumors as e. g. lipomas and lipoblastomas. Herein, we observed a strong correlation between the expression of HMGA2 and PLAG1 in 14 benign and 23 malignant thyroid tumors. To address the question if PLAG1 can be activated by HMGA2, the expression of both genes was quantified in 32 uterine leiomyomas 17 of which exhibited an overexpression of HMGA2. All leiomyomas with HMGA2 overexpression also revealed an activation of PLAG1 in the absence of detectable chromosome 8 abnormalities affecting the PLAG1 locus. To further investigate if the overexpression of PLAG1 is inducible by HMGA2 alone, HMGA2 was transiently overexpressed in MCF-7 cells. An increased PLAG1 expression was observed 24 and 48 h after transfection. Likewise, stimulation of HMGA2 by FGF1 in adipose tissue-derived stem cells led to a simultaneous increase of PLAG1 mRNA. Altogether, these data suggest that HMGA2 is an upstream activator of PLAG1. Accordingly, this may explain the formation of tumors as similar as lipomas and lipoblastomas resulting from an activation of either of both genes by chromosomal rearrangements.


Asunto(s)
Adenoma/metabolismo , Carcinoma Papilar/metabolismo , Proteínas de Unión al ADN/metabolismo , Proteína HMGA2/metabolismo , Leiomioma/metabolismo , Neoplasias de la Tiroides/metabolismo , Neoplasias Uterinas/metabolismo , Adenoma/genética , Adenoma/patología , Carcinoma Papilar/genética , Carcinoma Papilar/patología , Línea Celular Tumoral , Proteínas de Unión al ADN/genética , Femenino , Proteína HMGA2/genética , Humanos , Leiomioma/genética , Leiomioma/patología , Neoplasias de la Tiroides/genética , Neoplasias de la Tiroides/patología , Neoplasias Uterinas/genética , Neoplasias Uterinas/patología
13.
Mol Cytogenet ; 6(1): 23, 2013 Jun 05.
Artículo en Inglés | MEDLINE | ID: mdl-23738817

RESUMEN

BACKGROUND: Deletions of the gene encoding mediator subcomplex 12 (MED12) in human smooth muscle tumors rank among the most frequent genomic alterations in human tumors at all. In a minority of these cases, small deletions are found. In an attempt to delineate key features of the deletions aimed at a better understanding of the molecular pathogenesis of uterine smooth muscle tumors we have analyzed 70 MED12 deletions including 46 cases from the literature and 24 own unpublished cases. RESULTS: The average length of the deletions was 18.7 bp ranging between 2 bp and 43 bp. While in general multitudes of 3 clearly dominated leaving the transcript in frame, deletions of 21, 24, 30, and 33 nucleotides were clearly underrepresented. Within the DNA segment affected deletion breakpoints were not randomly distributed. Most breakpoints clustered within the center of the segment where two peaks of breakpoint clusters could be distinguished. Interestingly, one of these clusters coincides with the loop of a putative folded non-B DNA structure whereas a much lower number of breaks noted in the 5' and 3' stem of the structure forming an intramolecular B-helix. The second cluster mainly consisting of 3' breaks was located in a region downstream adjacent to the stem. CONCLUSION: The present study describes for the first time main characteristics of MED12 deletions occurring in smooth muscle tumors. Interestingly, the non-random distribution of breakpoints within the deletion hotspot region may point to a role of non-canonical DNA structures for the occurrence of these mutations and the molecular pathogenesis of uterine smooth muscle tumors, respectively.

14.
Anticancer Res ; 32(5): 1589-93, 2012 May.
Artículo en Inglés | MEDLINE | ID: mdl-22593436

RESUMEN

BACKGROUND: The expression of high mobility group protein AT-hook2 (HMGA2) indicates a worse prognosis in many epithelial malignancies, such as colon cancer. The present study addresses methodological aspects, as well as the genetic background, of the HMGA2 expression in colon cancer. MATERIALS AND METHODS: Samples of 38 colon carcinomas were studied for the expression of HMGA2 by quantitative Real-Time PCR (qRT-PCR). In selected cases, immunohistochemistry (IHC) was also performed. RESULTS: The overexpression of HMGA2, compared to adjacent mucosa, is not consistent among colon carcinomas: Only a minority of carcinomas strongly overexpressed HMGA2, but in no more than 50% of the tumors did the expression exceed the average value in mucosa samples. qRT-PCR clearly reveals a continuum between cases with high and low expression. CONCLUSION: For HMGA2-based risk assessment, continuous rather than discontinuous models seem to be most appropriate. However, in daily practice, IHC seems to be a suitable method to stratify for high-risk patients.


Asunto(s)
Neoplasias del Colon/química , Regulación Neoplásica de la Expresión Génica , Proteína HMGA2/genética , Adulto , Anciano , Anciano de 80 o más Años , Femenino , Proteína HMGA2/análisis , Humanos , Inmunohistoquímica , Masculino , Persona de Mediana Edad , ARN Mensajero/análisis , Reacción en Cadena en Tiempo Real de la Polimerasa
15.
Anticancer Res ; 31(3): 753-61, 2011 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-21498692

RESUMEN

AIM: To address the influence of genes involved in stem cell self-renewal and senescence on the growth of leiomyoma cells in vitro and to explore possible therapeutic implications of a targeted disruption of the p53-murine double minute 2 (MDM2) interaction. MATERIALS AND METHODS: Gene expression studies (qRT-PCR) of fibroid tissue and cells; ß-galactosidase stain and qRT-PCR after antagonizing MDM2. RESULTS: In fibroid cells, expression of HMGA2 decreased with passaging while that of p14(Arf) increased. Expression of these markers significantly positively, and negatively, respectively, influenced proliferation. Administration of nutlin-3, an MDM2 antagonist, induced cellular senescence and increased the expression of BAX. This, along with a significant correlation between p14(Arf) and BAX expression in native fibroids, suggests that p14(Arf) triggers senescence as well as apoptosis. CONCLUSION: p14(Arf) and HMGA2 seem to play a pivotal role in controlling the growth of fibroid cells. Antagonizing MDM2 induces senescence, as well as apoptosis, and may offer a chance to treat fibroids.


Asunto(s)
Senescencia Celular , Proteína HMGA2/metabolismo , Leiomioma/patología , Leiomioma/terapia , Proteína p14ARF Supresora de Tumor/metabolismo , Neoplasias Uterinas/patología , Neoplasias Uterinas/terapia , Apoptosis/efectos de los fármacos , Biomarcadores de Tumor/metabolismo , Línea Celular Transformada , Proliferación Celular/efectos de los fármacos , Senescencia Celular/efectos de los fármacos , Femenino , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Imidazoles/farmacología , Leiomioma/genética , Leiomioma/metabolismo , Piperazinas/farmacología , Proteínas Proto-Oncogénicas c-mdm2/antagonistas & inhibidores , Proteínas Proto-Oncogénicas c-mdm2/metabolismo , ARN Interferente Pequeño/metabolismo , Neoplasias Uterinas/genética , beta-Galactosidasa/metabolismo
16.
Cancer Genet Cytogenet ; 202(1): 53-7, 2010 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-20804922

RESUMEN

The in vitro growth of cells from uterine fibroids is characterized by an early onset of senescence. Often, an even lower growth potential than that of matching myometrial cells is noted. Also, the tremendous differences in the expression of the high mobility group protein HMGA2 seen when comparing fibroids of different genetic subtypes are surprisingly not reflected by significant differences in their growth potential in vitro. We aimed to evaluate possible changes of the HMGA2 expression level between the native tissue and cell cultures, so we performed quantitative real-time polymerase chain reaction studies that revealed a marked decrease of the HMGA2 mRNA in culture in those cases with overexpression of HMGA2. In the two cases initially showing the highest expression, it decreased by approximately 97%. Associated with the decrease of HMGA2 was a clearly increased expression of the senescence-associated p19(Arf). Together, these findings explain the similar behavior of cell cultures from fibroids of different genetic subgroups and may also offer an explanation for the early onset of in vitro senescence in these cell cultures.


Asunto(s)
Senescencia Celular/fisiología , Proteína HMGA2/genética , Leiomioma/patología , Adenocarcinoma/genética , Carcinoma de Células Escamosas/genética , Técnicas de Cultivo de Célula/métodos , División Celular , Bandeo Cromosómico , Cromosomas Humanos Par 12 , Cartilla de ADN , ADN Complementario/genética , Femenino , Regulación Neoplásica de la Expresión Génica , Reordenamiento Génico , Humanos , Leiomioma/genética , Leiomioma/cirugía , ARN Neoplásico/genética , ARN Neoplásico/aislamiento & purificación , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transcripción Genética
17.
Melanoma Res ; 20(5): 431-4, 2010 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-20562656

RESUMEN

Anorectal melanomas (AMs) are very rare and highly malignant tumors that are often diagnosed in advanced stages. After the differentiation between cutaneous melanoma (CM) and AM on the molecular level based on the presence of BRAF mutations, further modes of differentiation opened up, such as the recently discovered immunohistologically relevant protein deleted in malignant brain tumors 1 (DMBT1). Over the past several years, increasingly specific therapies have been developed on the basis of new therapy principles. Tyrosin kinase receptors such as Her2 and EGFR have been awarded a large role in this context. The goal of this study was to examine AMs for a possible expression or overexpression of these markers. Expression analyses of Her2 and EGFR were performed immunohistologically on 25 primary AMs. An overexpression of Her2 (score: 3+) was found in one AM from a 68-year-old female patient among these samples. In contrast, EGFR expression was not found in any of the AMs. The results presented here show that isolated cases of AM may benefit from an additive Her2-directed therapy, as the overexpression of Her2 was found in one of our AM patients.


Asunto(s)
Neoplasias del Ano/metabolismo , Melanoma/metabolismo , Receptor ErbB-2/metabolismo , Neoplasias del Recto/metabolismo , Adulto , Anciano , Anciano de 80 o más Años , Neoplasias del Ano/diagnóstico , Neoplasias del Ano/epidemiología , Diagnóstico Diferencial , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Incidencia , Masculino , Melanoma/diagnóstico , Melanoma/epidemiología , Persona de Mediana Edad , Técnicas de Diagnóstico Molecular , Neoplasias del Recto/diagnóstico , Neoplasias del Recto/epidemiología , Neoplasias Cutáneas/diagnóstico , Neoplasias Cutáneas/epidemiología , Neoplasias Cutáneas/metabolismo , Regulación hacia Arriba
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA