Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 118
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Oncogene ; 27(7): 1013-8, 2008 Feb 07.
Artigo em Inglês | MEDLINE | ID: mdl-17700532

RESUMO

The ataxia telangiectasia-mutated (ATM) gene has been implicated as an early barrier to the growth and progression of incipient solid tumors. Here, we show that germ-line nullizygosity for the mouse Atm gene significantly increases the proliferative index, net growth rate and multiplicity of intestinal adenomas in two distinct models of familial colon cancer: Apc(Min/+) and Apc(1638N/+). These effects of Atm deficiency are quantitatively different from deficiency for either of the genomic stability genes Bloom's syndrome helicase or DNA ligase 4, and the effect of Atm loss on tumor multiplicity is largely independent of the effect of ionizing radiation. Furthermore, the loss of heterozygosity rates at the adenomatous polyposis coli (Apc) locus are unaffected by Atm loss. Taken together, these data implicate the Atm gene product as a barrier to dysplastic growth in the early stages of intestinal tumor progression, independent of its effects on genomic stability.


Assuntos
Adenoma/metabolismo , Proteína da Polipose Adenomatosa do Colo/fisiologia , Proteínas de Ciclo Celular/fisiologia , Proteínas de Ligação a DNA/fisiologia , Mucosa Intestinal/metabolismo , Neoplasias Intestinais/metabolismo , Proteínas Serina-Treonina Quinases/fisiologia , Proteínas Supressoras de Tumor/fisiologia , Proteína da Polipose Adenomatosa do Colo/genética , Adenosina Trifosfatases/genética , Adenosina Trifosfatases/metabolismo , Animais , Proteínas Mutadas de Ataxia Telangiectasia , Neoplasias do Colo/genética , Neoplasias do Colo/patologia , DNA Helicases/genética , DNA Helicases/metabolismo , DNA Ligase Dependente de ATP , DNA Ligases/genética , DNA Ligases/metabolismo , Perda de Heterozigosidade , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Radiação Ionizante , RecQ Helicases
2.
Cancer Res ; 60(14): 3965-70, 2000 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-10919675

RESUMO

Altered patterns of the 5-cytosine methylation of genomic DNA are associated with the development of a wide range of human cancers. We have studied the mechanisms and genetic pathways by which a targeted heterozygous deficiency in the murine 5-cytosine DNA methyltransferase gene (Dnmt1(N/+)) diminishes intestinal tumorigenesis in C57BL/6-multiple intestinal neoplasia (Min)/+ mice. We found that Dnmt1(N/+) retards the net growth rate of intestinal adenomas and reduces tumor multiplicity by approximately 50%. This tumor resistance affects the entire intestinal tract and is independent of the status of modifier of Min 1 and p53, two loci that have been found to confer strong resistance to Min-induced neoplasia Interestingly, Dnmt/(N/+) and modifier of Min 1 resistance interact synergistically, together virtually eliminating tumor incidence. This finding may provide an insight into potential combinatorial therapeutic approaches for treating human colon cancer.


Assuntos
Adenoma/tratamento farmacológico , DNA (Citosina-5-)-Metiltransferases/genética , Genes p53/genética , Neoplasias Intestinais/tratamento farmacológico , Adenoma/genética , Adenoma/patologia , Fatores Etários , Alelos , Animais , Apoptose/genética , Bromodesoxiuridina/metabolismo , Divisão Celular/efeitos dos fármacos , DNA (Citosina-5-)-Metiltransferase 1 , DNA (Citosina-5-)-Metiltransferases/fisiologia , Metilação de DNA , Replicação do DNA/genética , Feminino , Genótipo , Mutação em Linhagem Germinativa , Neoplasias Intestinais/genética , Neoplasias Intestinais/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Mutantes , Mitose/genética , Mutagênese Sítio-Dirigida , Neoplasias Experimentais/tratamento farmacológico , Neoplasias Experimentais/genética , Neoplasias Experimentais/patologia
3.
Oncogene ; 19(28): 3182-92, 2000 Jun 29.
Artigo em Inglês | MEDLINE | ID: mdl-10918573

RESUMO

The Mom1 (Modifier of Min-1) region of distal chromosome 4 was identified during a screen for polymorphic modifiers of intestinal tumorigenesis in ApcMin/+ mice. Here, we demonstrate that the Mom1AKR allele consists of two genetic components. These include the secretory phospholipase Pla2g2a, whose candidacy as a Mom1 resistance modifier has now been tested with several transgenic lines. A second region, distal to Pla2g2a, has also been identified using fine structure recombinants. Pla2g2aAKR transgenic mice demonstrate a modest resistance to tumorigenesis in the small intestine and a very robust resistance in the large intestine. Moreover, the tumor resistance in the colon of Pla2g2aAKR animals is dosage-dependent, a finding that is consistent with our observation that Pla2g2a is expressed in goblet cells. By contrast, mice carrying the distal Mom1 modifier demonstrate a modest tumor resistance that is confined to the small intestine. Thus, the phenotypes of these two modifier loci are complementary, both in their quantitative and regional effects. The additive effects and tight linkage of these modifiers may have been necessary for the initial identification of the Mom1 region.


Assuntos
Polipose Adenomatosa do Colo/genética , Proteínas do Citoesqueleto/genética , Fosfolipases A/genética , Proteína da Polipose Adenomatosa do Colo , Animais , Feminino , Células Caliciformes/patologia , Humanos , Imunidade Inata/genética , Intestinos/patologia , Masculino , Camundongos , Camundongos Endogâmicos AKR , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Fosfolipases A/biossíntese
4.
Oncogene ; 19(23): 2774-9, 2000 May 25.
Artigo em Inglês | MEDLINE | ID: mdl-10851078

RESUMO

Defects in APC and DNA mismatch repair genes are associated with a strong predisposition to colon cancer in humans, and numerous mouse strains with mutations in these genes have been generated. In this report we describe the phenotype of Min/+ Mlh1-/- mice. We find that these doubly mutant mice develop more than three times the number of intestinal adenomas compared to Min/+ Mlh1+/+ or +/- mice but that these tumors do not show advanced progression in terms of tumor size or histological appearance. Full length Apc protein was not detected in the tumor cells from Min/+ Mlh1-/- mice. Molecular analyses indicated that in many tumors from Min/+ Mlh1-/- mice, Apc was inactivated by intragenic mutation. Mlh1 deficiency in Min/+ mice also led to an increase in cystic intestinal crypt multiplicity as well as enhancing desmoid tumorigenesis and epidermoid cyst development. Thus, Mlh1 deficiency influences the somatic events involved in the development of most of the phenotypes associated with the Min mutation. Oncogene (2000).


Assuntos
Proteínas do Citoesqueleto/genética , Neoplasias Intestinais/genética , Proteínas de Neoplasias/genética , Proteínas Adaptadoras de Transdução de Sinal , Proteína da Polipose Adenomatosa do Colo , Animais , Pareamento Incorreto de Bases , Proteínas de Transporte , Imuno-Histoquímica , Neoplasias Intestinais/patologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Mutantes , Proteína 1 Homóloga a MutL , Mutação , Proteínas de Neoplasias/deficiência , Proteínas Nucleares , Fenótipo
5.
Proc Natl Acad Sci U S A ; 97(7): 3461-6, 2000 Mar 28.
Artigo em Inglês | MEDLINE | ID: mdl-10716720

RESUMO

The interaction between mutations in the tumor-suppressor genes Apc and p53 was studied in congenic mouse strains to minimize the influence of polymorphic modifiers. The multiplicity and invasiveness of intestinal adenomas of Apc(Min/+) (Min) mice was enhanced by deficiency for p53. In addition, the occurrence of desmoid fibromas was strongly enhanced by p53 deficiency. The genetic modifier Mom1 and the pharmacological agents piroxicam and difluoromethylornithine each reduced intestinal adenoma multiplicity in the absence of p53 function. Mom1 showed no influence on the development of desmoid fibromas, whereas the combination of piroxicam and difluoromethylornithine exerted a moderate effect. The ensemble of tumor suppressors and modifiers of a neoplastic process can be usefully analyzed in respect to tissue specificity and synergy.


Assuntos
Adenoma/genética , Neoplasias Intestinais/genética , Neoplasias Primárias Múltiplas/genética , Adenoma/patologia , Animais , Eflornitina/farmacologia , Feminino , Fibromatose Agressiva/genética , Genes APC , Genes p53 , Neoplasias Intestinais/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Neoplasias Primárias Múltiplas/patologia , Piroxicam/farmacologia
6.
Arch Microbiol ; 172(6): 364-76, 1999 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-10591846

RESUMO

During the life cycle of Physarum polycephalum, uninucleate amoebae develop into multinucleate syncytial plasmodia. These two cell types differ greatly in cellular organisation, behaviour and gene expression. Classical genetic analysis has identified the mating-type gene, matA, as the key gene controlling the initiation of plasmodium development, but nothing is known about the molecular events controlled by matA. In order to identify genes involved in regulating plasmodium formation, we constructed a subtracted cDNA library from cells undergoing development. Three genes that have their highest levels of expression during plasmodium development were identified: redA, redB (regulated in development) and mynD (myosin). Both redA and redB are single-copy genes and are not members of gene families. Although redA has no significant sequence similarities to known genes, redB has sequence similarity to invertebrate sarcoplasmic calcium-binding proteins. The mynD gene is closely related to type II myosin heavy-chain genes from many organisms and is one of a family of type II myosin genes in P. polycephalum. Our results indicate that many more red genes remain to be identified, some of which may play key roles in controlling plasmodium formation.


Assuntos
Genes de Protozoários , Physarum polycephalum/crescimento & desenvolvimento , Physarum polycephalum/genética , Sequência de Aminoácidos , Animais , DNA Complementar/genética , DNA de Protozoário/genética , Regulação da Expressão Gênica no Desenvolvimento , Biblioteca Gênica , Dados de Sequência Molecular , Homologia de Sequência de Aminoácidos
7.
Genomics ; 57(3): 333-41, 1999 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-10328999

RESUMO

Multiple alleles of the quaking (qk) gene have a variety of phenotypes ranging in severity from early embryonic death to viable dysmyelination. A previous study identified a candidate gene, QKI, that contains an RNA-binding domain and encodes at least three protein isoforms (QKI-5, -6 and -7). We have determined the genomic structure of QKI, identifying an additional alternative end in cDNAs. Further we have examined the exons and splice sites for mutations in the lethal alleles qkl-1, qkkt1, qkk2, and qkkt3. The mutation in qkl-1 creates a splice site in the terminal exon of the QKI-6 isoform. Missense mutations in the KH domain and the QUA1 domains in qkk2 and qkkt3, respectively, indicate that these domains are of critical functional importance. Although homozygotes for each ENU induced allele die as embryos, their phenotypes as viable compound heterozygotes with qkv differ. Compound heterozygous qkv animals carrying qkkt1, qkk2, and qkkt3 all exhibit a permanent quaking phenotype similar to that of qkv/qkv animals, whereas qkv/qkl-1 animals exhibit only a transient quaking phenotype. The qkl-1 mutation eliminates the QKI-5 isoform, showing that this isoform plays a crucial role in embryonic survival. The transient quaking phenotype observed in qkv/qkl-1 mice indicates that the QKI-6 and QKI-7 isoforms function primarily during myelination, but that QKI-5 may have a concentration-dependent role in early myelination. This mutational analysis demonstrates the power of series of alleles to examine the function of complex loci and suggests that additional mutant alleles of quaking could reveal additional functions of this complex gene.


Assuntos
Etilnitrosoureia/farmacologia , Genes Letais , Mutagênicos/farmacologia , Proteínas de Ligação a RNA/genética , Animais , Sequência de Bases , Mapeamento Cromossômico , DNA Complementar , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Quaking , Dados de Sequência Molecular , Mutagênese , Proteínas de Ligação a RNA/efeitos dos fármacos
8.
Proc Natl Acad Sci U S A ; 95(18): 10826-31, 1998 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-9724789

RESUMO

Previous studies of Min/+ (multiple intestinal neoplasia) mice on a sensitive genetic background, C57BL/6 (B6), showed that adenomas have lost heterozygosity for the germ-line ApcMin mutation in the Apc (adenomatous polyposis coli) gene. We now report that on a strongly resistant genetic background, AKR/J (AKR), Min-induced adenoma multiplicity is reduced by about two orders of magnitude compared with that observed on the B6 background. Somatic treatment with a strong mutagen increases tumor number in AKR Min/+ mice in an age-dependent manner, similar to results previously reported for B6 Min/+ mice. Immunohistochemical analyses indicate that Apc expression is suppressed in all intestinal tumors from both untreated and treated AKR Min/+ mice. However, the mechanism of Apc inactivation in AKR Min/+ mice often differs from that observed for B6 Min/+ mice. Although loss of heterozygosity is observed in some tumors, a significant percentage of tumors showed neither loss of heterozygosity nor Apc truncation mutations. These results extend our understanding of the effects of genetic background on Min-induced tumorigenesis in several ways. First, the AKR strain carries modifiers of Min in addition to Mom1. This combination of AKR modifiers can almost completely suppress spontaneous intestinal tumorigenesis associated with the Min mutation. Second, even on such a highly resistant genetic background, tumor formation continues to involve an absence of Apc function. The means by which Apc function is inactivated is affected by genetic background. Possible scenarios are discussed.


Assuntos
Adenoma/patologia , Genes APC , Neoplasias Intestinais/patologia , Perda de Heterozigosidade , Adenoma/induzido quimicamente , Adenoma/genética , Alelos , Animais , Carcinógenos , Etilnitrosoureia , Mutação em Linhagem Germinativa , Neoplasias Intestinais/induzido quimicamente , Neoplasias Intestinais/genética , Camundongos
9.
Exp Lung Res ; 24(4): 437-53, 1998.
Artigo em Inglês | MEDLINE | ID: mdl-9659576

RESUMO

Although the methodology for mapping genes controlling susceptibility to tumor development in mice is becoming well established, it remains a formidable challenge to move from linkage to locus. Positional cloning, now commonly used in the identification of loci affecting a qualitative phenotype, has yet to be successfully applied to quantitative trait loci. This study describes the application of candidate gene testing, a method complementary to positional cloning. The method has been applied to evaluate candidates for the quantitative trait locus, Mom1, which modifies the susceptibility of ApcMin/+ mice to spontaneous intestinal tumor development. The authors also discuss the further testing of one candidate, the phospholipase gene Pla2g2a, by transgenesis. Finally, studies on the mode of action of Mom1 are discussed in light of the evidence that Mom1 encodes this secretory phospholipase.


Assuntos
Polipose Adenomatosa do Colo/genética , Regulação Neoplásica da Expressão Gênica/genética , Genes APC , Neoplasias Intestinais/genética , Polipose Adenomatosa do Colo/patologia , Animais , Mapeamento Cromossômico , Genes Supressores de Tumor , Mutação em Linhagem Germinativa , Neoplasias Intestinais/patologia , Camundongos , Camundongos Endogâmicos , Camundongos Mutantes , Fosfolipases A/genética , Característica Quantitativa Herdável
10.
Philos Trans R Soc Lond B Biol Sci ; 353(1370): 915-23, 1998 Jun 29.
Artigo em Inglês | MEDLINE | ID: mdl-9684289

RESUMO

The Min (multiple intestinal neoplasia) strain of the laboratory mouse and its derivatives permit the fundamental study of factors that regulate the transition between normal and neoplastic growth. A gene of central importance in mediating these alternative patterns of growth is Apc, the mouse homologue of the human adenomatous polyposis coli (APC) gene. When adenomas form in the Min mouse, both copies of the Apc gene must be inactivated. One copy is mutated by the nonsense Apc allele carried in heterozygous form in this strain. The other copy can be silenced by any of several mechanisms. These range from loss of the homologue bearing the wild-type Apc allele; to interstitial deletions surrounding the wild-type allele; to intragenic mutation, including nonsense alleles; and finally, to a reduction in expression of the locus, perhaps owing to mutation in a regulatory locus. Each of these proposed mechanisms may constitute a two-hit genetic process as initially posited by Knudson; however, apparently the two hits could involve either a single locus or two loci. The kinetic order for the transition to adenoma may be still higher than two, if polyclonal adenomas require stronger interactions than passive fusion. The severity of the intestinal neoplastic phenotype of the Min mouse is strongly dependent upon loci other than Apc. One of these, Mom1, has now been rigorously identified at the molecular level as encoding an active resistance conferred by a secretory phospholipase. Mom1 acts locally within a crypt lineage, not systemically. Within the crypt lineage, however, its action seems to be non-autonomous: when tumours arise in Mom1 heterozygotes, the active resistance allele is maintained in the tumour (MOH or maintenance of heterozygosity). Indeed, the secretory phospholipase is synthesized by post-mitotic Paneth cells, not by the proliferative cells that presumably generate the tumour. An analysis of autonomy of modifier gene action in chimeric mice deserves detailed attention both to the number of genetic factors for which an animal is chimeric and to the clonal structure of the tissue in question. Beyond Mom1, other loci can strongly modify the severity of the Min phenotype. An emergent challenge is to find ways to identify the full set of genes that interact with the intestinal cancer predisposition of the Min mouse strain. With such a set, one can then work, using contemporary mouse genetics, to identify the molecular, cellular and organismal strategies that integrate their functions. Finally, with appropriately phenotyped human families, one can investigate by a candidate approach which modifying factors influence the epidemiology of human colon cancer. Even if a candidate modifier does not explain any of the genetic epidemiology of colon cancer in human populations, modifier activities discovered by mouse genetics provide candidates for chemopreventive and/or therapeutic modalities in the human.


Assuntos
Mucosa Intestinal/fisiologia , Neoplasias Intestinais/genética , Neoplasias Intestinais/fisiopatologia , Adenoma/genética , Adenoma/patologia , Polipose Adenomatosa do Colo/genética , Animais , Deleção Cromossômica , Genes APC , Genes Reguladores , Humanos , Mucosa Intestinal/citologia , Neoplasias Intestinais/patologia , Camundongos
12.
Nat Genet ; 17(1): 88-91, 1997 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-9288104

RESUMO

Individuals inheriting the same mutation predisposing to cancer may show very different outcomes, ranging from early aggressive cancer to disease-free survival. Experimental mouse models can provide a powerful tool to identify factors in the environment and genetic background that account for such modifications. The Min mouse strain, in which the ApcMin mutation disrupts the mouse homologue of the human familial polyposis gene, develops intestinal neoplasms whose multiplicity is strongly affected by genetic background. We previously mapped a strong modifier locus, Mom1 (modifier of Min-1), to a 4-cM region on mouse chromosome 4 containing a candidate gene Pla2g2a encoding a secretory phospholipase. Here, we report that a cosmid transgene overexpressing Pla2g2a caused a reduction in tumour multiplicity and size, comparable to that conferred by a single copy of the resistance allele of Mom1. These results offer strong evidence that this secretory phospholipase can provide active tumour resistance. The association of Pla2g2a with Mom1 thus withstands a strong functional test and is likely to represent the successful identification of a polymorphic quantitative trait locus in mammals.


Assuntos
Mapeamento Cromossômico , Genes Supressores de Tumor , Neoplasias Intestinais/genética , Fosfolipases A/genética , Polipose Adenomatosa do Colo/genética , Animais , Sequência de Bases , Primers do DNA , Genes APC , Genótipo , Humanos , Imunidade Inata , Camundongos , Camundongos Endogâmicos AKR , Camundongos Transgênicos , Dados de Sequência Molecular , Fosfolipases A/análise , Fosfolipases A/biossíntese , Reação em Cadeia da Polimerase
13.
Genetics ; 146(3): 1049-60, 1997 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-9215907

RESUMO

Clock is a semidominant mutation identified from an N-ethyl-N-nitrosourea mutagenesis screen in mice. Mice carrying the Clock mutation exhibit abnormalities of circadian behavior, including lengthening of endogenous period and loss of rhythmicity. To identify the gene affected by this mutation, we have generated a high-resolution genetic map (> 1800 meioses) of the Clock locus. We report that Clock is 0.7 cM distal of Kit on mouse chromosome 5. Mapping shows that Clock lies within the W19H deletion. Complementation analysis of different Clock and W19H compound genotypes indicates that the Clock mutation behaves as an antimorph. This antimorphic behavior of Clock strongly argues that Clock defines a gene centrally involved in the mammalian circadian system.


Assuntos
Mapeamento Cromossômico , Transativadores/genética , Animais , Proteínas CLOCK , Ritmo Circadiano , Feminino , Deleção de Genes , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C3H , Camundongos Endogâmicos C57BL , Mutação , Fenótipo , Proteínas Proto-Oncogênicas c-kit/genética
14.
Cancer Res ; 57(10): 1999-2006, 1997 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-9157997

RESUMO

We have demonstrated previously that intestinal tumor formation in B6 Min/+ mice is always accompanied by loss of the wild-type adenomatous polyoposis coli (Apc) allele and that intestinal tumor multiplicity in B6 Min/+ mice can be significantly increased by treatment with a single dose of N-ethyl-N-nitrosourea (ENU). Here, we show that some tumors from ENU-treated Min/+ mice can form without complete elimination of Apc+. At least 25% of these tumors acquired somatic Apc truncation mutations. Interestingly, some ENU-induced tumors demonstrated loss of the Apc+ allelic marker examined by the quantitative PCR assay used here. Using two methods of mutation detection, we identified no Apc mutations in at least 12% of the tumors from ENU-treated B6 Min/+ mice. Finally, no H- or K-ras-activating mutations were detected in intestinal tumors from either untreated or ENU-treated Min/+ mice. The majority of somatic human APC mutations in intestinal tumors lead to APC truncation. Our results demonstrate that somatic Apc truncation mutations also frequently occur in ENU-induced intestinal tumors in Min mice.


Assuntos
Neoplasias Intestinais/genética , Mutação , Alelos , Animais , Carcinógenos , Etilnitrosoureia , Feminino , Deleção de Genes , Regulação Neoplásica da Expressão Gênica , Genes APC , Genes ras , Neoplasias Intestinais/induzido quimicamente , Masculino , Camundongos , Camundongos Endogâmicos
15.
Proc Natl Acad Sci U S A ; 94(11): 5848-53, 1997 May 27.
Artigo em Inglês | MEDLINE | ID: mdl-9159163

RESUMO

Mice heterozygous for the ApcMin (Min) mutation develop adenomas throughout the intestinal tract. Apc is believed to be involved in cell migration, adhesion, and polarity. Adenoma multiplicity and growth rate are modulated by an unlinked modifier locus, Mom1. The secretory phospholipase Pla2g2a is a candidate for Mom1. Here, we investigate the range of action of Apc and Mom1. Analysis of chimeric Min mice indicates that the actions of both Apc and Mom1 are localized within the cell lineage that gives rise to intestinal tumors.


Assuntos
Adenoma/genética , Genes APC , Neoplasias Intestinais/genética , Mutação , Adenoma/patologia , Animais , Quimera , Cruzamentos Genéticos , Feminino , Genótipo , Heterozigoto , Neoplasias Intestinais/patologia , Intestinos/transplante , Masculino , Camundongos , Camundongos Endogâmicos , Camundongos Mutantes , Camundongos Transgênicos , Mórula/fisiologia , Fenótipo , Transplante Isogênico , beta-Galactosidase/análise , beta-Galactosidase/biossíntese , beta-Galactosidase/genética
17.
Cancer Res ; 57(5): 812-4, 1997 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-9041176

RESUMO

We have tested the hypothesis that enteric bacteria are necessary for formation of intestinal adenomas in C57BL/6-ApcMin/+ mouse. Germ-free mice developed 2-fold fewer adenomas than conventional controls in the medial small intestine (7.3 versus 14.9; P < 0.003), but there were no significant differences in the rest of the intestinal tract. We conclude that microbial status does not strongly alter the adenoma phenotype in this mouse model of familial adenomatous polyposis. In parallel, we have found that C57BL/6-ApcMin/+ mice mutated at the beige locus, which controls natural killer activity, are also unaltered in adenoma multiplicity.


Assuntos
Polipose Adenomatosa do Colo/genética , Neoplasias Intestinais/etiologia , Intestinos/microbiologia , Células Matadoras Naturais/imunologia , Animais , Vida Livre de Germes , Intestino Delgado/microbiologia , Intestino Delgado/patologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Mutantes
18.
Mamm Genome ; 8(2): 98-101, 1997 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-9060407

RESUMO

We describe the genetic mapping of hyperphenylal-aninemia 2 (hph2), a recessive mutation in the mouse that causes deficient amino acid transport similar to Hartnup disorder, a human genetic amino acid transport disorder. The hph2 locus was mapped in three separate crosses to identify candidate genes for hph2 and a region of homology in the human genome where we propose the Hartnup Disorder gene might lie. The mutation maps to mouse Chromosome (Chr) 7 distal of the simple sequence length polymorphism (SSLP) marker D7Mit140 and does not recombine with D7Nds4, an SSLP marker in the fibroblast growth factor 3 (Fgf3) gene. Unexpectedly, the mutant chromosome affects recombination frequency in the D7Mit12 to D7Nds4 interval.


Assuntos
Mapeamento Cromossômico , Doença de Hartnup/genética , Fenilalanina/metabolismo , Animais , Transporte Biológico , Cruzamentos Genéticos , Modelos Animais de Doenças , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Mutação
19.
Mamm Genome ; 8(2): 102-7, 1997 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-9060408

RESUMO

The mutant mouse strain HPH2 (hyperphenylalaninemia) was isolated after N-ethyl-N-nitrosourea (ENU) mutagenesis on the basis of delayed plasma clearance of an injected load of phenylalanine. Animals homozygous for the recessive hph2 mutation excrete elevated concentrations of many of the neutral amino acids in the urine, while plasma concentrations of these amino acids are normal. In contrast, mutant homozygotes excrete normal levels of glucose and phosphorus. These data suggest an amino acid transport defect in the mutant, confirmed in a small reduction in normalized values of 14C-labeled glutamine uptake by kidney cortex brush border membrane vesicles (BBMV). The hyperaminoaciduria pattern is very similar to that of Hartnup Disorder cases also show niacin deficiency symptoms, of Hartnup Disorder cases also show niacin deficiency symptoms, which are thought to be multifactorially determined. Similarly, the HPH2 mouse exhibits a niacin-reversible syndrome that is modified by diet and by genetic background. Thus, HPH2 provides a candidate mouse model for the study of Hartnup Disorder, an amino acid transport deficiency and a multifactorial disease in the human.


Assuntos
Modelos Animais de Doenças , Doença de Hartnup/metabolismo , Fenilalanina/metabolismo , Animais , Transporte Biológico , Glicemia/análise , Glicosúria , Doença de Hartnup/genética , Humanos , Rim/metabolismo , Camundongos
20.
Proc Natl Acad Sci U S A ; 94(25): 13927-31, 1997 Dec 09.
Artigo em Inglês | MEDLINE | ID: mdl-9391129

RESUMO

When tumors form in intestinal epithelia, it is important to know whether they involve single initiated somatic clones. Advanced carcinomas in humans and mice are known to be monoclonal. However, earlier stages of tumorigenesis may instead involve an interaction between cells that belong to separate somatic clones within the epithelium. The clonality of early tumors has been investigated in mice with an inherited predisposition to intestinal tumors. Analysis of Min (multiple intestinal neoplasia) mice chimeric for a ubiquitously expressed cell lineage marker revealed that normal intestinal crypts are monoclonal, but intestinal adenomas frequently have a polyclonal structure, presenting even when very small as single, focal adenomas composed of at least two somatic lineages. Furthermore, within these polyclonal adenomas, all tumor lineages frequently lose the wild-type Apc allele. These observations can be interpreted by several models for clonal interaction within the epithelium, ranging from passive fusion within regions of high neoplastic potential to a requirement for active clonal cooperation.


Assuntos
Polipose Adenomatosa do Colo/genética , Polipose Adenomatosa do Colo/patologia , Genes APC , Proteína da Polipose Adenomatosa do Colo , Animais , Quimera/genética , Células Clonais/patologia , Proteínas do Citoesqueleto/genética , Proteínas do Citoesqueleto/metabolismo , Feminino , Genótipo , Humanos , Imuno-Histoquímica , Perda de Heterozigosidade , Masculino , Camundongos , Camundongos Mutantes , Modelos Genéticos , Mosaicismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...