• Refine Query
  • Source
  • Publication year
  • to
  • Language
  • 66
  • 20
  • 7
  • 4
  • 3
  • 2
  • 1
  • 1
  • Tagged with
  • 112
  • 55
  • 19
  • 18
  • 16
  • 16
  • 14
  • 14
  • 13
  • 12
  • 9
  • 9
  • 8
  • 8
  • 7
  • About
  • The Global ETD Search service is a free service for researchers to find electronic theses and dissertations. This service is provided by the Networked Digital Library of Theses and Dissertations.
    Our metadata is collected from universities around the world. If you manage a university/consortium/country archive and want to be added, details can be found on the NDLTD website.
1

A 147L substitution in the HOXD13 homeodomain causes a novel human limb malformation by producing a selective loss of function

Caronia, Giuliana January 2002 (has links)
No description available.
2

Investigation of the Homeobox (HOX) gene expression in human gastric carcinogenesis

Huang, Jian-yuan 23 June 2009 (has links)
In the past study, we used a human gastric stem cell clone, KMU-GI2, isolated from endoscopically biopsied gastric mucosa. As we maintained this KMU-GI2 cell clone, we also found a subclone with spontaneous transformation, named KMU-CSN. We found this KMU-CSN cell line showed homogenous epithelial morphology, cell pile-up appearance due to contact insensitivity, enhanced anchorage independence (from 1.7% of KMU-GI2 to 22% of KMU-CSN) and cell immortalization (CPDL >100). The high tumorigenic potential of this KMU-CS12 cell line has been endorsed by the finding of the tumor formation in nude mice. By spectral karyotyping and SNP GeneChip® Mapping 500K Assay, we found chromosomal abnormalities in chromosome 12, which were derived from duplication of 7p15.1~15.3 and 7p22.1~22.3. In normal cells, HOXA genes were found to localize in chromosome 7. In this KMU-CS12 cell line, an increased expression of HOXA4¡B5¡B7¡B9¡B13 genes was also shown, as compared with KMU-CSN. With these findings, we hypothesized that HOXA genes may play a role in gastric carcinogenesis. Therefore, we can check the prevalence of HOXA genes abnormalities in human gastric cancer patients.
3

Developmental studies of the murine homeobox gene, Hoxa-9

Lincoln, Joy January 2002 (has links)
Cell patterning during embryogenesis is essential for establishing the identity of the developing body plan. Hox genes are fundamental regulators of tissue organisation along the anterior-posterior body axis of the developing embryo. These homeodomain-containing proteins act as transcription factors during normal development. The function of the homeodomain is to bind sequence-specific DNAmotifs which allows either activation or repression of downstream effector genes, which consequently results in the control of tissue-specific determination and differentiation. Aberrant expression of such Hox genes, including Hoxa-9 can result in homeotic transformations leading to phenotypic malformations and oncogenesis. However the normal function of Hoxa-9 is poorly understood. This study explored the potential role for Hoxa-9 in normal development and differentiation. An in situ hybridisation approach was taken to define the expression of Hoxa-9 in the developing mouse. Hoxa-9 was found to expressed in a temporarily and spatially regulated manner, in particular being detected in the developing cardiac atria, ventricles and cardiac vessels during E9.5-E12 stages of development. The expression of this homeotic gene during in vitro differentiation of embryonic stem cells into cardiomyocytes and haematopoietic cells demonstrated a profile that correlated with the emergence of these cell types. The functioning relationship between Hoxa-9 expression and lineage commitment was Airther explored using over-expression in embryonic stem cells. A potential role for Hoxa-9 in normal development is discussed.
4

Spécificité des protéines Hox : Rôle des motifs connus et découverte de nouveaux motifs

Litim-Mecheri, Isma 08 November 2011 (has links)
Les gènes Hox sont responsables de l’identité des segments le long de l’axe antéro-postérieur. Ils sont évolutivement conservés et codent des facteurs de transcription. In vitro, toutes les protéines Hox se lient à des séquences nucléotidiques très similaires via un domaine de liaison à l’ADN très conservé, l’homéodomaine (HD). Cette faible spécificité de liaison à l’ADN in vitro contraste avec leur spécificité d’action in vivo. Une manière d’expliquer ce paradoxe est que les protéines Hox agissent en interaction avec des protéines cofacteurs, dont le mieux caractérisé est Extradenticle (Exd) chez la drosophile, Pbx chez les mammifères (collectivement appelés PBC). L’interaction Hox-PBC s’appui sur un motif conservé chez la presque totalité des protéines Hox, situé en amont de l’HD, le motif Hexapeptide (HX). Des travaux récents au sein de notre équipe ont montré l’existence d’un nouveau mode d’interaction Hox-PBC, non-générique, médié par un motif spécifique à certains groupes de paralogies seulement, et situé en en C-terminal de l’HD. Ceci souligne que les interactions Hox-PBC, qui spécifient la fonction des protéines Hox, reposent sur de modes multiples d’interaction.Mes travaux de thèse ont porté sur le mode d’action des protéines Hox, en étudiant la fonction de trois motifs ayant un rôle démontré ou potentiel dans le recrutement du cofacteur Exd par la protéine Hox de drosophile AbdominalA (AbdA). L’approche empruntée visait à analyser de manière globale la manière dont chacun de ces motifs pris isolément, ou en interaction, définit la fonction d’AbdA. Les conclusions de ce travail soulignent l’absence de pléiotropie fonctionnelle et un haut degré d’interactivité entre ces motifs. Le second volet de ma thèse a été d’initier la découverte de nouveaux motifs fonctionnels au sein des protéines Hox. J’ai abordé cette question en sélectionnant des modules phylogénétiquement conservés. Afin d’évaluer leur fonction, ces motifs ont été mutés et l’impact de leur mutation a été analysé in vitro et in vivo. Les résultats obtenus ont permis l’identification d’au moins un domaine protéique qui contribue de manière prédominante à la fonction de la protéine Dfd. / Hox genes are responsible for the identity of segments along the antero-posterior axis. They are evolutionarily conserved and encode transcription factors. In vitro, all Hox proteins bind to a similar nucleotide sequence via a highly conserved DNA binding domain, the homeodomain (HD). This low specificity of DNA binding in vitro contrasts with their specificity in vivo. One way to explain this paradaox is that Hox protein function with protein cofactors, best represented by Extradenticle (Exd) in Drosophila, Pbx in mammals (collectivaly refered as PBC). Hox-PBC interaction relies on a motif located upstream of the HD, conserved in most Hox proteins, the Hexapeptide (HX). Recent work in our group identified a novel mode of Hox-PBC interaction, non-generic, specific to a subset only of Hox paralog groups, and relying on a motif located C-terminal to the HD. This highlight plasticity in Hox-PBC interaction.My PhD work aimed at investigating the mode of action of Hox protein, by studying the function of three protein motifs, with known or putative role in Exd recruitment by the Drosophila Hox protein AbdominalA (AbdA). The approach taken aimed at analyzing, using a large functional window, how these motifs, taken in isolation or collectively, define AbdA protein activity. Conclusions highlight the absence of pleitropy and a high degree of functional interaction for these protein motifs. The second part of my PhD work has been to initiate the search for novel functionally important protein motifs within Hox proteins. This was approached by selecting phylogenetically conserved motifs, and addressing their function in vitro and in vivo following motif mutations. At least one functional domain was isolated, that contributes predominantly to Dfd protein function.
5

Avaliação do perfil genômico dos genes da família HOX em tumores a partir de dados de bancos públicos / Genomic profile evaluation of HOX genes family in cancer using public databases

Plaça, Jessica Rodrigues 11 October 2017 (has links)
A família de genes HOX compreende um conjunto de fatores de transcrição altamente conservados evolutivamente. Em mamíferos, os genes HOX se subdividem em 4 clusters: HOXA, HOXB, HOXC e HOXD, atuando no desenvolvimento embrionário com a regulação de processos biológicos como proliferação, diferenciação, migração, angiogênese e apoptose que são reativados durante a carcinogênese. Estudos recentes apontam que os genes HOX podem exercer papel relevante na formação de diversos tumores sólidos, todavia ainda não foi possível caracterizar sistematicamente a expressão dos genes HOX em tumores bem como determinar seus alvos em tumores. Desta forma, o objetivo geral deste trabalho consistiu na caracterização in silico do modelo de atuação genes HOX na carcinogênese. Para cumprir este objetivo foi identificado o perfil diferencial dos genes HOX entre amostras normais e tumorais. Alvos de genes HOX foram identificados e, quando diferencialmente expressos, foram associados com os genes HOX, independentemente dos índices de metilação e CNA. Por fim, as associações finais entre os genes HOX e seus alvos foram enriquecidas com os bancos de dados KEGG e GO. Identificou-se diferentes assinaturas de expressão de genes HOX em diferentes tumores, associadas com o eixo ântero-posterior do corpo humano, bem como os folhetos embrionários originários aos tecidos tumorais, compatível com o padrão de expressão no desenvolvimento embrionário. Um número considerável de genes HOX atuam preferencialmente via enhancers na regulação de seus alvos. Como exemplo, os genes HOXB7 e HOXC11, que funcionam como moduladores anti tumorais. Finalmente, o estudo mostra que diante do número crescente de dados genômicos públicos, é possível viabilizar projetos de grande valor científico. / The HOX gene family comprises a set of evolutionarily highly conserved transcription factors. In mammals, HOX genes are subdivided into four clusters: HOXA, HOXB, HOXC and HOXD, acting on the embryonic development with regulation of biological processes such as proliferation, differentiation, migration, angiogenesis and apoptosis that are reactivated during carcinogenesis. Recent studies indicate that HOX genes may play a relevant role in the formation of several solid tumors, but it has not been possible to systematically characterize the expression of HOX genes in tumors as well as to determine their targets in tumors. Thus, the general aim of this project was to characterize the in vivo model of HOX genes in carcinogenesis. To accomplish this goal the differential profile of HOX genes was identified between normal and tumor samples. HOX gene targets were identified and, when differentially expressed, were associated with HOX genes regardless of methylation and CNA indices. Finally, the final associations between the HOX genes and their targets were enriched with the KEGG and GO databases. Different signatures of HOX gene expression were identified in different tumors, associated with the anteroposterior axis of the human body, as well as the embryonic leaflets originating from the tumor tissues, compatible with the expression pattern in the embryonic development. A considerable number of HOX genes preferentially act via enhancers in the regulation of their targets. As an example, the HOXB7 and HOXC11 genes, which function as pro-tumor modulators. Finally, the study shows that in view of the growing number of public genomic data, it is possible to make feasible projects of great scientific value.
6

Hox Transcription Factors: Their Involvement in Human Cancer Cells and In Vitro Functional Specificity

Svingen, Terje, n/a January 2005 (has links)
Hox genes are regulatory genes encoding small proteins containing a highly conserved 61-amino acid motif, the homeodomain, that enables Hox proteins to bind to DNA at specifically recognised binding sites and transcriptionally activate their target genes. In mammalian species there are 39 Hox genes and they are structural and functional homologs of the Drosophila homeotic complex (Horn-C). During embryogenesis and early development the Hox genes are expressed in a spatiotemporal fashion, where they operate as master transcriptional regulators. Hox genes are further expressed in fully differentiated adult cells, potentially in a tissue-specific manner involving maintenance of the normal phenotype. In selected oncogenic transformations, dysregulated Hox gene expression has been observed, indicating an involvement of these transcriptional regulators in carcinogenesis and metastasis. Utilising quantitative real-time PCR assays, these studies investigated the expression patterns of 20 Hox genes and two wellcharacterised Hox cofactors (Pbx and Meis) in malignant and non-malignant human breast and skin cancer cells. Dysregulated Hox expression was observed for all malignancies tested, of which some misexpressed Hox genes seemed random, whereas other Hox transcripts showed altered levels potentially corresponding with the invasive capacity of the cells. Also, the Hox cofactors Pbx and Meis showed no marked changes in expression levels from the non-malignant to the malignant phenotypes, indicating that it is dysregulated Hox gene expression rather than dysregulated gene expression of Hox cofactors that potentially commit the cell to redifferentiate and undergo oncogenic transformation. Although the Hox proteins are known to be key transcriptional regulators of development, the mechanisms by which they gain their in vivo functional specificity is still largely unknown. They all show strikingly similar transcriptional specificity in vitro, yet show unique specificity in their in vivo environment. This paradox has been the subject of intense scrutiny, however very few direct Hox target genes have been identified, making it a difficult task to decipher the exact manner in which Hox proteins exert their functional potential. Therefore, the studies presented herein were aimed at identifying further Hox target genes in the human system. Utilising differential display approaches, several potential downstream target genes were isolated. Substantiated with real-time PCR assays, one of these potential targets was selected as a likely direct Hox gene target, and as such subjected to further studies. By the combination of bioinformatic analyses, transfection protocols and luciferase assays, a gene encoding the SR-related protein SRrpl3O was shown to be trans-activated in vitro by HOXD4 via a putative Hox binding element within its promoter region. This is the first reported link between Hox transcription factors and the SR and SR-related family of pre-mRNA splicing proteins, offering a new and exciting insight into the complex nature of Hox functional specificity. Finally, this thesis also puts forward new ideas regarding how the Hox proteins gain their transcriptional and functional specificity. Utilising bioinformatic tools in conjunction with performing an extensive review of the disparate catalogue of Hox-related research reports, work herein offers the first comprehensive analysis of the mammalian Hox gene targets in relation to their promoter structures, as well as with respect to the expanded Hox DNA-binding elements. This work reports that identified Hox targets generally contain TATA-less core promoters, many of which have several GC-box elements. The Hox binding elements show no apparent preference regarding their location relative to the transcription start site (TSS), as they are found both upstream and downstream of the TSS, as well as being located close to proximal core promoter elements for some genes and at more distant positions in other gene promoters. Finally, the core Hox binding element TAAT/ATTA contains only part of the necessary recognition sequence involved in Hox-DNA binding, and the notion that flanking base pairs dictate trans-regulatory potential is further explored with the hypothesis that the immediate 3' base pair dictates an activator/repressor-switch of the Hox trans-regulatory effect.
7

Avaliação do perfil genômico dos genes da família HOX em tumores a partir de dados de bancos públicos / Genomic profile evaluation of HOX genes family in cancer using public databases

Jessica Rodrigues Plaça 11 October 2017 (has links)
A família de genes HOX compreende um conjunto de fatores de transcrição altamente conservados evolutivamente. Em mamíferos, os genes HOX se subdividem em 4 clusters: HOXA, HOXB, HOXC e HOXD, atuando no desenvolvimento embrionário com a regulação de processos biológicos como proliferação, diferenciação, migração, angiogênese e apoptose que são reativados durante a carcinogênese. Estudos recentes apontam que os genes HOX podem exercer papel relevante na formação de diversos tumores sólidos, todavia ainda não foi possível caracterizar sistematicamente a expressão dos genes HOX em tumores bem como determinar seus alvos em tumores. Desta forma, o objetivo geral deste trabalho consistiu na caracterização in silico do modelo de atuação genes HOX na carcinogênese. Para cumprir este objetivo foi identificado o perfil diferencial dos genes HOX entre amostras normais e tumorais. Alvos de genes HOX foram identificados e, quando diferencialmente expressos, foram associados com os genes HOX, independentemente dos índices de metilação e CNA. Por fim, as associações finais entre os genes HOX e seus alvos foram enriquecidas com os bancos de dados KEGG e GO. Identificou-se diferentes assinaturas de expressão de genes HOX em diferentes tumores, associadas com o eixo ântero-posterior do corpo humano, bem como os folhetos embrionários originários aos tecidos tumorais, compatível com o padrão de expressão no desenvolvimento embrionário. Um número considerável de genes HOX atuam preferencialmente via enhancers na regulação de seus alvos. Como exemplo, os genes HOXB7 e HOXC11, que funcionam como moduladores anti tumorais. Finalmente, o estudo mostra que diante do número crescente de dados genômicos públicos, é possível viabilizar projetos de grande valor científico. / The HOX gene family comprises a set of evolutionarily highly conserved transcription factors. In mammals, HOX genes are subdivided into four clusters: HOXA, HOXB, HOXC and HOXD, acting on the embryonic development with regulation of biological processes such as proliferation, differentiation, migration, angiogenesis and apoptosis that are reactivated during carcinogenesis. Recent studies indicate that HOX genes may play a relevant role in the formation of several solid tumors, but it has not been possible to systematically characterize the expression of HOX genes in tumors as well as to determine their targets in tumors. Thus, the general aim of this project was to characterize the in vivo model of HOX genes in carcinogenesis. To accomplish this goal the differential profile of HOX genes was identified between normal and tumor samples. HOX gene targets were identified and, when differentially expressed, were associated with HOX genes regardless of methylation and CNA indices. Finally, the final associations between the HOX genes and their targets were enriched with the KEGG and GO databases. Different signatures of HOX gene expression were identified in different tumors, associated with the anteroposterior axis of the human body, as well as the embryonic leaflets originating from the tumor tissues, compatible with the expression pattern in the embryonic development. A considerable number of HOX genes preferentially act via enhancers in the regulation of their targets. As an example, the HOXB7 and HOXC11 genes, which function as pro-tumor modulators. Finally, the study shows that in view of the growing number of public genomic data, it is possible to make feasible projects of great scientific value.
8

Genomic and phylogenetic assessment of sea lamprey (Petromyzon marinus) Hox genes and analysis of Hox genes in association with myomeres across multiple lamprey genera

Childs, Darcy 22 August 2013 (has links)
Lampreys are an important model for the study of early vertebrate development due to their unique evolutionary position as one of only two extant jawless vertebrates. In this study, 12 new putative Hox gene fragments were identified within the recently available Petromyzon marinus (sea lamprey) genome. These and the other previously-identified Hox genes were analyzed phylogenetically, which enabled the assignment of many of the new sequences to distinct paralogous gene clusters and showed distinctions between gnathostome and lamprey Hox sequences. An examination of Hox genes in other lamprey species was conducted using genomic PCR-based detection methods and identified 26 putative Hox gene homeobox fragments from multiple Hox genes across nine lamprey species. A study of Hox10 coding sequences in different lamprey species failed to find any correlation with variable numbers of trunk myomeres in lampreys, which suggests that other sequences or factors regulate the number of myomeres in different species.
9

Genomic and phylogenetic assessment of sea lamprey (Petromyzon marinus) Hox genes and analysis of Hox genes in association with myomeres across multiple lamprey genera

Childs, Darcy 22 August 2013 (has links)
Lampreys are an important model for the study of early vertebrate development due to their unique evolutionary position as one of only two extant jawless vertebrates. In this study, 12 new putative Hox gene fragments were identified within the recently available Petromyzon marinus (sea lamprey) genome. These and the other previously-identified Hox genes were analyzed phylogenetically, which enabled the assignment of many of the new sequences to distinct paralogous gene clusters and showed distinctions between gnathostome and lamprey Hox sequences. An examination of Hox genes in other lamprey species was conducted using genomic PCR-based detection methods and identified 26 putative Hox gene homeobox fragments from multiple Hox genes across nine lamprey species. A study of Hox10 coding sequences in different lamprey species failed to find any correlation with variable numbers of trunk myomeres in lampreys, which suggests that other sequences or factors regulate the number of myomeres in different species.
10

The Amphioxus Hox Cluster: Characterization, Comparative Genomics, and Evolution

Amemiya, Chris T., Prohaska, Sonja J., Hill-Force, Alicia, Cook, April, Wasserscheid, Jessica, Ferrier, David E.K., Anaya, Juan Pascal, Garcia-Fernández, Jordi, Dewar, Ken, Stadler, Peter F. 04 October 2018 (has links)
The amphioxus Hox cluster is often viewed as “archetypal” for the chordate lineage. Here we present a descriptive account of the 448kb region spanning the Hox cluster of the amphioxus Branchiostoma floridae from Hox14 to Hox1.We provide complete coding sequences of all 14 previously described amphioxus sequences and describe a detailed analysis of the conserved non-coding regulatory sequence elements. We find that the posterior part of the Hox cluster is so highly derived that even the complete genomic sequence is insufficient to decide whether the posterior Hox genes arose by independent duplications or whether they are true orthologs of the corresponding gnathostome paralog groups. In contrast, the anterior region is much better conserved. The amphioxus Hox cluster strongly excludes repetitive elements with the exception of two repeat islands in the posterior region. Repeat exclusion is also observed in gnathostomes, but not protostome Hox clusters. We thus hypothesize that the much shorter vertebrate Hox clusters are the result of extensive resolution of the redundancy of regulatory DNA following the genome duplications rather than the consequence of a selection pressure to remove non-functional sequence from the cluster.

Page generated in 0.0423 seconds