963 resultados para Wilms-tumor Gene
Resumo:
Context: To assess the efficacy of preoperative chemotherapy in Wilms’ tumor patients and explore its true value for specific subgroups. Objectives: In the presence of these controversies, a meta-analysis that examines the efficacy of preoperative chemotherapy in Wilms’ tumor patients and specific subgroups is needed to clarify these issues. The objective of this meta-analysis is to assess the efficacy of preoperative chemotherapy in Wilms’ tumor patients and explore its true value for specific subgroups. Data Sources: Computer-based systematic search with “preoperative chemotherapy”, “Neoadjuvant Therapy” and “Wilms’ tumor” as search terms till January 2013 was performed. Study Selection: No language restrictions were applied. Searches were limited to randomized clinical trials (RCTs) or retrospective studies in human participants under 18 years. A manual examination of references in selected articles was also performed. Data Extraction: Relative Risk (RR) and their 95% Confidence Interval (CI) for Tumor Shrinkage (TS), total Tumor Resection (TR), Event-Free Survival (EFS) and details of subgroup analysis were extracted. Meta-analysis was carried out with the help of the software STATA 11.0. Finally, four original Randomized Clinical Trials (RCTs) and 28 retrospective studies with 2375 patients were included. Results: For preoperative chemotherapy vs. up-front surgery (PC vs. SU) group, the pooled RR was 9.109 for TS (95% CI: 5.109 - 16.241; P < 0.001), 1.291 for TR (95% CI: 1.124 - 1.483; P < 0.001) and 1.101 for EFS (95% CI: 0.980 - 1.238; P = 0.106). For subgroup short course vs. long course (SC vs. LC), the pooled RR was 1.097 for TS (95% CI: 0.784 - 1.563; P = 0.587), 1.197 for TR (95% CI: 0.960 - 1.493; P = 0.110) and 1.006 for EFS (95% CI: 0.910 - 1.250; P = 0.430). Conclusions: Short course preoperative chemotherapy is as effective as long course and preoperative chemotherapy only benefits Wilms’ tumor patients in tumor shrinkage and resection but not event-free survival.
Resumo:
variety of transcription factors including Wilms tumor gene (Wt-1), steroidogenic factor 1 (Sf-1), dosage-sensitive sex reversal, adrenal hypoplasia congenita on the X-chromosome, Gene 1 (Dax-1), and pre-B-cell transcription factor 1 (Pbx1) have been defined as necessary for regular adrenocortical development. However, the role of Pbx1 for adrenal growth and function in the adult organism together with the molecular relationship between Pbx1 and these other transcription factors have not been characterized. We demonstrate that Pbx haploinsufficiency (Pbx1(+/-)) in mice is accompanied by a significant lower adrenal weight in adult animals compared with wild-type controls. Accordingly, baseline proliferating cell nuclear antigen levels are lower in Pbx1(+/-) mice, and unilateral adrenalectomy results in impaired contralateral compensatory adrenal growth, indicating a lower proliferative potential in the context of Pbx1 haploinsufficiency. In accordance with the key role of IGFs in adrenocortical proliferation and development, real-time RT-PCR demonstrates significant lower expression levels of the IGF-I receptor, and up-regulation of IGF binding protein-2. Functionally, Pbx1(+/-) mice display a blunted corticosterone response after ACTH stimulation coincident with lower adrenal expression of the ACTH receptor (melanocortin 2 receptor, Mc2-r). Mechanistically, in vitro studies reveal that Pbx1 and Sf-1 synergistically stimulates Mc2-r promoter activity. Moreover, Sf-1 directly activates the Pbx1 promoter activity in vitro and in vivo. Taken together, these studies provide evidence for a role of Pbx1 in the maintenance of a functional adrenal cortex mediated by synergistic actions of Pbx1 and Sf-1 in the transcriptional regulation of the critical effector of adrenocortical differentiation, the ACTH receptor.
Resumo:
Wilms tumor is a childhood tumor of the kidney arising from the undifferentiated metanephric mesenchyme. Tumorigenesis is attributed to a number of genetic and epigenetic alterations. In 20% of Wilms tumors, Wilms tumor gene 1 (WT1) undergoes inactivating homozygous mutations causing loss of function of the zinc finger transcription factor it encodes. It is hypothesized that mutations in WT1 result in dysregulation of downstream target genes, leading to aberrant kidney development and/or Wilms tumor. These downstream target genes are largely unknown, and identification is important for further understanding Wilms tumor development. Heatmap data of human Wilms tumor protein expression, generated by reverse phase protein assay analysis (RPPA), show significant correlation between WT1 mutation status and low PRKCα expression (p= 0.00013); additionally, p-PRKCα (S657) also shows decreased expression in these samples (p= 0.00373). These data suggest that the WT1 transcription factor regulates PRKCα expression, and that PRKCα plays a potential role in Wilms tumor tumorigenesis. We hypothesize that the WT1 transcription factor directly/indirectly regulates PRKCα and mutations occurring in WT1 lead to decreased expression of PRKCα. Prkcα and Wt1 have been shown to co-localize in E14.5 mesenchymal cells of the developing kidney. siRNA knockdown, in-vivo ablation, and tet-inducible expression of Wt1 each independently confirm regulation of Prkcα expression by Wt1 at both RNA and protein levels, and investigation into possible WT1 binding sites in PRKCα regulatory regions has identified multiple sites to be confirmed by luciferase reporter constructs. With the goal of identifying WT1 and PRKCα downstream targets, RPPA analysis of protein expression in mesenchymal cell culture, following lentiviral delivered shRNA knockdown of Wt1 and shRNA knockdown of Prkcα, will be carried out. Apart from Wilms tumor, WT1 also plays an important role in Acute Myeloid Leukemia (AML). WT1 mutation status has been implicated, controversially, as an independent poor-prognosis factor in leukemia, leading to decreased probability of overall survival, complete remission, and disease free survival. RPPA analysis of AML patient samples showed significant decreases in PRKCα/p-PRKCα protein expression in a subset of patients (Kornblau, personal communication); therefore, the possible role of WT1 and PRKCα in leukemia disease progression is an additional focus of this study. WT1 mutation analysis of diploid leukemia patient samples revealed two patients with mutations predicted to affect WT1 activity; of these two samples, only one corresponded to the low PRKCα expression cohort. Further characterization of the role of WT1 in AML, and further understanding of WT1 regulated PRKCα expression, will be gained following RPPA analysis of protein expression in HL60 leukemia cell lines with lentiviral delivered shRNA knockdown of WT1 and shRNA knockdown of PRKCα.
Resumo:
Most studies on kidney development have considered the interaction of the metanephric mesenchyme and the ureteric bud to be the major inductive event that maintains tubular differentiation and branching morphogenesis. The mesenchyme produces Gdnf, which stimulates branching, and the ureteric bud stimulates continued growth of the mesenchyme and differentiation of nephrons from the induced mesenchyme. Null mutation of the Wt1 gene eliminates outgrowth of the ureteric bud, but Gdnf has been identified as a target of Pax2, but not of Wt1. Using a novel system for microinjecting and electroporating plasmid expression constructs into murine organ cultures, it has been demonstrated that Vegfa expression in the mesenchyme is regulated by Wt1. Previous studies had identified a population of Flk1-expressing cells in the periphery of the induced mesenchyme, and adjacent to the stalk of the ureteric bud, and that Vegfa was able to stimulate growth of kidneys in organ culture. Here it is demonstrated that signaling through Flk1 is required to maintain expression of Pax2 in the mesenchyme of the early kidney, and for Pax2 to stimulate expression of Gdnf. However, once Gdnf stimulates branching of the ureteric bud, the Flk1-dependent angioblast signal is no longer required to maintain branching morphogenesis and induction of nephrons. Thus, this work demonstrates the presence of a second set of inductive events, involving the mesenchymal and angioblast populations, whereby Wt1-stimulated expression of Vegfa elicits an as-yet-unidentified signal from the angioblasts, which is required to stimulate the expression of Pax2 and Gdnf, which in turn elicits an inductive signal from the ureteric bud.
Resumo:
Gene-chip technology was employed to study the effect of dietary vitamin E (VE) on gene expression in rat testes. Male albino rats were fed with either a diet deficient in VE or a standard diet containing VE. Differential gene expression was monitored at five individual time-points over a period of 14 months with all animals individually pro. led. Low VE intake resulted in the consistent upregulation of 7-dehydrocholesterol reductase and GATA binding protein 4, both involved in testosterone synthesis. Cyclin D3, important in cell cycle progression and Wilms tumor 1, related to cancer development, were also up-regulated in the vitamin E deficient animals. This study demonstrates that low dietary VE intake has long-term effects on gene expression in the testes. Our data provides insights into the possible molecular mechanisms underlying the beneficial effects of vitamin E on the male reproductive organ.
Resumo:
Wilms tumor (WT) is an embryonal renal tumor with a heterogeneous genetic etiology that serves as a valuable model for studying tumorigenesis. Biallelic inactivation of the tumor suppressor gene WT1, a zinc-finger transcriptional regulator located at 11p13, is critical for the development of some Wilms tumors. Interestingly, WT1 genomic analysis has demonstrated mutations in less than 20% of WT cases. This suggests either other genes play a more major role in Wilms tumorigenesis or WT1 is functionally altered by mechanisms other than DNA mutation. Previous observations in rat and in WT xenograft cell lines have suggested that abnormal WT1 RNA processing (exon 6 RNA editing and aberrant exon 2 splicing, respectively) is a potential mechanism of altering WT1 function in the absence of a WT1 DNA mutation. However, the role of this abnormal RNA processing has not previously been assessed in primary Wilms tumors. ^ To test the hypothesis that abnormal WT1 RNA processing is a mechanism of WT1alteration during tumor development, WT1 RNA from 85 primary tumors was analyzed using reverse transcription and polymerase chain reaction amplification (RT-PCR). Although no evidence for WT1 RNA editing was observed, variable levels (5% to 50%) of aberrant WT1 exon 2 splicing were detected for 11 tumors in the absence of a detectable WT1 DNA mutation. Also, alteration of normal WT1 alternative splicing, observed as RNA isoform loss, was detected in five tumors with no apparent WT1 genomic alteration, although no consistent pattern of RNA isoform loss was detected. This abnormal WT1 splicing, detected by either loss of exon 2 from some of the transcripts or loss of RNA isoforms, is statistically correlated with relapse (p = 0.005). These studies demonstrate that abnormal WT1 RNA processing is not a common mechanism of abrogating normal WT1 function in primary tumors. However, in those cases in which abnormal WTI splicing is present, these data indicate that it may serve as a useful prognostic marker for relapse in WT patients. ^
Resumo:
Elevated expression levels of the bcl-2 proto-oncogene have been correlated with the appearance of androgen independence in prostate cancer. Although bcl-2 was first cloned as the t (14:18) translocation breakpoint from human follicular B cell lymphoma, the mechanism of overexpression of bcl-2 is largely undefined for advanced prostate cancer, there being no gross alterations in the gene structure. We investigated the role of the product of the prostate apoptosis response gene-4 (Par-4) and the product of the Wilms' tumor 1 gene (WT1) in the regulation of Bcl-2 expression in prostate cancer cell lines. We observed growth arrest and apoptosis, upon decreasing Bcl-2 protein and transcript in the high Bcl-2 expressing, androgen-independent prostate cancer cell lines, by all trans-retinoic acid treatment but this did not occur in the androgen-dependent cell lines expressing low levels of Bcl-2. Changes in localization of Par-4, and an induction in the expression of WT1 protein accompanied the decrease in the Bcl-2 protein and transcript following all trans-retinoic acid treatment, in the androgen-independent prostate cancer cell line. In stable clones expressing ectopic Par-4 we observed decreased Bcl-2 protein and transcript. This was accompanied by an induction in WT1 expression. Finally, we detected Par-4 and WT1 proteins binding to a previously identified WT1 binding site on the bcl-2 promoter both in vitro and in vivo leading to a decrease in transcription from the bcl-2 promoter. We conclude that Par-4 regulates Bcl-2 through a WT1 binding site on the bcl-2 promoter. ^
Resumo:
Among the human diseases that result from chromosomal aberrations, a de novo deletion in chromosome 11p13 is clinically associated with a syndrome characterized by Wilms' tumor, aniridia, genitourinary anomalies, and mental retardation (WAGR). Not all genes in the deleted region have been characterized biochemically or functionally. We have recently identified the first Class III cyclic nucleotide phosphodiesterase, Rv0805, from Mycobacterium tuberculosis, which biochemically and structurally belongs to the superfamily of metallophosphoesterases. We performed a large scale bioinformatic analysis to identify orthologs of the Rv0805 protein and identified many eukaryotic genes that included the human 239FB gene present in the region deleted in the WAGR syndrome. We report here the first detailed biochemical characterization of the rat 239FB protein and show that it possesses metallophosphodiesterase activity. Extensive mutational analysis identified residues that are involved in metal interaction at the binuclear metal center. Generation of a rat 239FB protein with a mutation corresponding to a single nucleotide polymorphism seen in human 239FB led to complete inactivation of the protein. A close ortholog of 239FB is found in adult tissues, and biochemical characterization of the 239AB protein demonstrated significant hydrolytic activity against 2',3'-cAMP, thus representing the first evidence for a Class III cyclic nucleotide phosphodiesterase in mammals. Highly conserved orthologs of the 239FB protein are found in Caenorhabditis elegans and Drosophila and, coupled with available evidence suggesting that 239FB is a tumor suppressor, indicate the important role this protein must play in diverse cellular events.
Resumo:
Metallophosphoesterase-domain-containing protein 2 (MPPED2) is a highly evolutionarily conserved protein with orthologs found from worms to humans. The human MPPED2 gene is found in a region of chromosome 11 that is deleted in patients with WAGR (Wilms tumor, aniridia, genitourinary anomalies, and mental retardation) syndrome, and MPPED2 may function as a tumor suppressor. However, the precise cellular roles of MPPED2 are unknown, and its low phosphodiesterase activity suggests that substrate hydrolysis may not be its prime function. We present here the structures of MPPED2 and two mutants, which show that the poor activity of MPPED2 is not only a consequence of the substitution of an active-site histidine residue by glycine but also due to binding of AMP or GMP to the active site. This feature, enhanced by structural elements of the protein, allows MPPED2 to utilize the conserved phosphoprotein-phosphatase-like fold in a unique manner, ensuring that its enzymatic activity can be combined with a possible role as a scaffolding or adaptor protein. (C) 2011 Elsevier Ltd. All rights reserved.
Resumo:
Purpose: To detect the occurrence and expression of the suppressor gene p53 and of the oncogene c-Myc in eyelid tumors of dogs using the PCR, RT-PCR, PCR-ELISA and RT-PCR-ELISA techniques. These genes have not been described in dog eyelid tumors before. Methods: Nine samples of eyelid or third eyelid epithelial tumors were obtained from the archives of the Department of Veterinary Pathology. Tumor diagnosis was confirmed by evaluation of hematoxylin-eosin stained sections, and immunohistochemistry for cytokeratin AE1/AE3 and vimentin V9. A canine mammary tumor was used for positive control. Agarose gel electrophoresis, PCR-ELISA and RT-PCR-ELISA were used to detect p53 and c-Myc genes. Results: The occurrence of p53 was detected in most of the eyelid tumors and third eyelid tumors studied (88.8%, n = 8) and was expressed in 75% of the positive samples, as indicated by ELISA. The c-Myc gene was found in 77.7% (n = 7) of the samples and was expressed in eight samples. Conclusions: Eyelid and third eyelid tumors of dogs express both the p53 and the c-Myc genes as shown by PCR and RT-PCR. However, PCR ELISA and RT-PCR ELISA were more efficient in assessing occurrence and expression of these genes because they identified amplified products that were not detected by agarose gel electrophoresis. © 2010 American College of Veterinary Ophthalmologists.
Resumo:
The search for molecular markers to improve diagnosis, individualize treatment and predict behavior of tumors has been the focus of several studies. This study aimed to analyze homeobox gene expression profile in oral squamous cell carcinoma (OSCC) as well as to investigate whether some of these genes are relevant molecular markers of prognosis and/or tumor aggressiveness. Homeobox gene expression levels were assessed by microarrays and qRT-PCR in OSCC tissues and adjacent non-cancerous matched tissues (margin), as well as in OSCC cell lines. Analysis of microarray data revealed the expression of 147 homeobox genes, including one set of six at least 2-fold up-regulated, and another set of 34 at least 2-fold down-regulated homeobox genes in OSCC. After qRT-PCR assays, the three most up-regulated homeobox genes (HOXA5, HOXD10 and HOXD11) revealed higher and statistically significant expression levels in OSCC samples when compared to margins. Patients presenting lower expression of HOXA5 had poorer prognosis compared to those with higher expression (P=0.03). Additionally, the status of HOXA5, HOXD10 and HOXD11 expression levels in OSCC cell lines also showed a significant up-regulation when compared to normal oral keratinocytes. Results confirm the presence of three significantly upregulated (>4-fold) homeobox genes (HOXA5, HOXD10 and HOXD11) in OSCC that may play a significant role in the pathogenesis of these tumors. Moreover, since lower levels of HOXA5 predict poor prognosis, this gene may be a novel candidate for development of therapeutic strategies in OSCC.
Resumo:
In der vorliegenden Dissertation wurden verschiedene Kandidatengene für den Wilmstumor (WT), eine Tumorerkrankung der Niere, identifiziert und charakterisiert. Da dieses frühkindliche Malignom aus einer inkorrekt ablaufenden Metanephrogenese resultiert, wurden die Genexpressionsmuster verschiedener humaner Wilmstumor- und Normalnierengewebe (adulte sowie fetale Niere) mit Hilfe der Technik des differential display verglichen und die als differenziell exprimiert identifizierten Gene kloniert und charakterisiert. Bei TM7SF1 handelt es sich um ein neues Gen, dessen Transkription im Zuge der Metanephrogenese angeschaltet wird. Das von ihm codierte putative Protein kann aufgrund von Strukturvorhersagen vermutlich zur Familie G Protein-gekoppelter Rezeptoren gezählt werden. Die ableitbare Funktion als Signalmolekül der Nierenentwicklung, sowie seine Lokalisation in einem WT-Lokus (1q42-q43) machen TM7SF1 zu einem aussichtsreichen Kandidatengen für den WT. Darüber hinaus konnten die Voraussetzungen für funktionelle Tests, die eine weitere Charakterisierung von TM7SF1 erlauben, geschaffen werden (Identifikation und Klonierung des murinen Homologen, stabil überexprimierende WT-Zelllinien, Antikörper gegen den Aminoterminus des putativen Proteins). Mit TCF2 wurde ein weiteres Gen identifiziert, dessen Produkt in Prozessen der Metanephrogenese eine Rolle spielt. Die signifikante Herunterregulation der TCF2-Expression in der großen Mehrzahl der untersuchten WTs, die innerhalb der vorliegenden Arbeit gezeigte Regulation durch das WT1-Genprodukt, sowie seine genomische Lokalisation in einem Intervall für die familiäre Form des WT (FWT1 in 17q12-q21) zeigen das Potenzial von TCF2, als Kandidatengen für den FWT zu gelten. Darüber hinaus wurde mit GLI3 ein in verschiedenen WTs stark exprimiertes Gen identifiziert. Sein Produkt ist eine Komponente des entwicklungsbiologisch relevanten und in verschiedene Tumorerkrankungen involvierten sonic hedgehog-Signaltransduktionsweges. Mit FE7A3 und CDT151 konnten zwei differenziell exprimierte cDNAs identifiziert werden, die Teile neuer Gene darstellen und die in WT-Loci kartiert werden konnten. Aufgrund von Homologievergleichen im Bereich der identifizierten offenen Leserahmen konnte eine mögliche Bedeutung der putativen Genprodukte für die WT-Pathogenese als Zelladhäsionsmolekül (FE7A3) bzw. als mit der Proliferation assoziiertem Transkriptionsfaktor (CDT151) herausgearbeitet werden. Neben den komparativen Genexpressionsuntersuchungen wurde in einem zweiten Ansatz die transkriptionelle Regulation des einzigen bisher klonierten Wilmstumorgens (WT1) analysiert. Mit Hilfe vergleichender Reportergenanalysen in WT1-exprimierenden und nicht-exprimierenden Zelllinien konnten neue für die transkriptionelle Regulation von WT1 relevante Bereiche identifiziert werden. Darüber hinaus wurde der für die Transkriptionsfaktoren SP1 und SP3 an anderen Promotoren beschriebene funktionelle Antagonismus für die WT1-Expression untersucht und in Gelretardationsanalysen mit dem WT1-Expressionsstatus oben genannter Zelllinien korreliert.
Resumo:
Ansatz zur Generierung einer konditionalen, reversiblen Wt1 k.o.-Maus Der Wilms-Tumor (WT, Nephroblastom) ist ein embryonaler Nierentumor, der durch die maligne Transformation von undifferenziertem Nierengewebe, sog. nephrogenen Resten, entsteht. WT treten mit einer Inzidenz von 1 in 10.000 Lebendgeburten auf. Das Hauptmanifestationsalter, der normalerweise einseitig und sporadisch auftretenden Tumore, liegt zwischen dem 3. und 4. Lebensjahr. Etwa 10 % der Patienten entwickeln jedoch bilaterale Tumore. In diesen Fällen ist eine Assoziation mit komplexen genetischen Krankheitsbildern (u. a. WAGR-, Denys-Drash-, Frasier- und Beckwith-Wiedemann-Syndrom) festzustellen. In 15 % der sporadischen WT sind Mutationen im WT1 (Wilms-Tumor 1)-Gen beschrieben. WT1 besteht aus zehn Exons und weist typische Merkmale von Transkriptionsfaktoren (z. B. vier Zinkfinger) auf. Zwei alternative Spleißereignisse betreffen Exon 5 (+/−Exon 5) und Exon 9 (Transkripte mit bzw. ohne die codierenden Sequenzen für die AS Lysin-Threonin-Serin; +/−KTS). Die Lage der drei alternativ vorhandenen AS zwischen den Zinkfingern 3 und 4 bestimmt die verschiedenen Funktionen der WT1-Proteine (4 Isoformen) als Transkriptionsfaktor (−KTS) bzw. als RNA-bindendes Protein (+KTS). Das zunächst im Zusammenhang mit WT als Tumorsuppressorgen identifizierte WT1 ist ein Entwicklungsgen mit einem sehr komplexen Expressionsmuster in der Embryonalentwicklung. Dabei ist v. a. die Bedeutung in der Urogenitalentwicklung entscheidend. Konstitutive, homozygote Wt1−/− k.o.-Mäuse sind embryonal (~ E12,5 dpc) letal und bilden u. a. keine Gonaden und keine Nieren. Aus diesem Grund existiert bisher kein Wilms-Tumormodell. Die Herstellung eines konditionalen murinen Tiermodells auf Basis des Tet on/off-Systems zur Untersuchung der Nierenentwicklung bzw. zur Analyse der Wilms-Tumorpathogenese war Ziel dieser Arbeit. Hierfür wurden drei Mauslinien generiert: Zwei transgene sog. Responder-Linien, die eine chimäre spleißbare Wt1-cDNA der Variante musWt1+Exon 5;+/−KTS unter der Kontrolle eines Tet-responsiven Promotors im Genom tragen. Dieses tTA/Dox-abhängig regulierbare Wt1-Transgen (tgWt1) sollte (exogen regulierbar) die Expression des endogenen Wt1-Lokus ausreichend nachahmen, um die kritischen Phasen der Embryogenese zu überwinden und lebensfähige Tiere zu erhalten. Parallel dazu wurde die Wt1-Effektor-Mauslinie (WE2) generiert. Diese trägt einen tetrazyklinabhängigen Transaktivator (tTA) zur Steuerung Tet-regulierbarer Transgene unter der Kontrolle des endogenen Wt1-Promotors. Die durch homologe Rekombination in ES-Zellen erreichte Integration des tTA direkt am Translationsstartpunkt des Wt1-Lokus hat in den Tieren einen heterozygoten Wt1 knock out/tTA knock in zur Folge. Die bisher vorgenommenen Verpaarungen doppelt transgener Wt1-tTA+/−/Resp-Mäuse ergaben keinen Rescue des letalen Wt1 k.o. und es konnten bislang keine Wilms-Tumore induziert werden. Alle im Verlauf der Arbeit generierten Mauslinien wurden umfassend charakterisiert. So konnte für die Tiere der Responder-Linien Wt1-Resp1 (mit zusätzlichen Isolator-Sequenzen zum Schutz des Transgens vor Positionseffekten) und Wt1-Resp2 (ohne Isolatoren) konnte die Tet-induzierbare Expression und die Spleißbarkeit des tgWt1 in MEF-Assays und mittels Effektor-Mäusen auf RNA-Ebene nachgewiesen werden. Die genomische Charakterisierung der WE2-Linie ergab eine ungeklärte etwa 120 kb große Inversion am Wt1-Lokus, die alle 5'-regulatorischen Sequenzen mitsamt des tTA vom Rest von Wt1 trennt. Tiere dieser Linie weisen aber dennoch einen funktionalen Wt1 k.o. auf: Unter den Nachkommen aus Intercross-Verpaarungen von Wt1-tTA+/−-Mäusen lassen sich auf Grund der Letalität keine Wt1−/−-Genotypen nachweisen. Die Charakterisierung der Effektor-Linie auf RNA-Ebene und mittels Reporter-Mäusen liefert ein Wt1-analoges tTA-Expressionsmuster: So findet man eine deutliche tTA-Expression u. a. in Niere (Glomeruli), Uterus, Ovar und Testis. Die hier vorgestellten Experimente ergeben darüber hinaus eindeutige Hinweise einer Beteiligung von Wt1 in der Entstehung der glatten Muskulatur bzw. in der Vaskulogenese.
Resumo:
Introduction: Desmoplastic small round cell tumor (DSRCT) is an uncommon, embryonic-type neoplasm, typically presenting as an abdominal mass in young men. A single case of DSRCT arising in the peripheral nervous system has been reported. Methods: The clinical course, imaging, electrophysiological, intraoperative, histopathological, molecular findings, and postoperative follow-up are reported. Results: A 43-year-old man presented with slowly progressive right brachial plexopathy. Magnetic resonance imaging revealed an enlarged medial cord with heterogeneous contrast enhancement. Histology showed a "small round cell" neoplasm with a polyphenotypic immunoprofile, including epithelial and mesenchymal markers. A pathognomonic fusion of Ewing sarcoma breakpoint region 1 and Wilms tumor 1 genes (EWSR1/WT1) was present. Treatment involved gross total excision and local radiotherapy. Conclusion: Our findings confirm the occurrence of DSRCT as a primary peripheral nerve tumor. Despite its usually very aggressive clinical course, prolonged recurrence-free survival may be reached. Histomorphology and immunoprofile of DSRCT may lead to misdiagnosis as small cell carcinoma. © 2013 Wiley Periodicals, Inc.