Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
1.
J Cell Biochem ; 116(11): 2541-51, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-25939245

RESUMO

Non-compensated dilated cardiomyopathy (DCM) leading to death from heart failure is rising rapidly in developed countries due to aging demographics, and there is a need for informative preclinical models to guide the development of effective therapeutic strategies to prevent or delay disease onset. In this study, we describe a novel model of heart failure based on cardiac-specific deletion of the prototypical mammalian BAR adapter-encoding gene Bin1, a modifier of age-associated disease. Bin1 deletion during embryonic development causes hypertrophic cardiomyopathy and neonatal lethality, but there is little information on how Bin1 affects cardiac function in adult animals. Here we report that cardiomyocyte-specific loss of Bin1 causes age-associated dilated cardiomyopathy (DCM) beginning by 8-10 months of age. Echocardiographic analysis showed that Bin1 loss caused a 45% reduction in ejection fraction during aging. Younger animals rapidly developed DCM if cardiac pressure overload was created by transverse aortic constriction. Heterozygotes exhibited an intermediate phenotype indicating Bin1 is haplo-insufficient to sustain normal heart function. Bin1 loss increased left ventricle (LV) volume and diameter during aging, but it did not alter LV volume or diameter in hearts from heterozygous mice nor did it affect LV mass. Bin1 loss increased interstitial fibrosis and mislocalization of the voltage-dependent calcium channel Cav 1.2, and the lipid raft scaffold protein caveolin-3, which normally complexes with Bin1 and Cav 1.2 in cardiomyocyte membranes. Our findings show how cardiac deficiency in Bin1 function causes age- and stress-associated heart failure, and they establish a new preclinical model of this terminal cardiac disease.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/deficiência , Envelhecimento/genética , Cardiomiopatia Dilatada/genética , Miócitos Cardíacos/patologia , Proteínas do Tecido Nervoso/deficiência , Proteínas Supressoras de Tumor/deficiência , Animais , Cardiomiopatia Dilatada/fisiopatologia , Modelos Animais de Doenças , Camundongos , Camundongos Knockout , Miócitos Cardíacos/metabolismo , Especificidade de Órgãos , Volume Sistólico
2.
J Neurol Neurosurg Psychiatry ; 83(9): 894-902, 2012 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-22791904

RESUMO

AIM: A first in human study to evaluate tolerability and pharmacokinetics followed by an early proof of mechanism (POM) study to determine whether the small orally, available molecule, Posiphen tartrate (Posiphen), lowers secreted (s) amyloid-ß precursor protein (APP) α and -ß, amyloid-ß peptide (Aß), tau (τ) and inflammatory markers in CSF of patients with mild cognitive impairment (MCI). STUDY DESIGN: Posiphen single and multiple ascending dose phase 1 randomised, double blind, placebo-controlled safety, tolerance, pharmacokinetic studies were undertaken in a total of 120 healthy volunteers to define a dose that was then used in a small non-randomised study of five MCI subjects, used as their own controls, to define target engagement. MAIN OUTCOME MEASURES: Pharmacodynamic: sAPPα, sAPPß, Aß(42), τ (total (t) and phosphorylated (p)) and inflammatory marker levels were time-dependently measured over 12 h and compared prior to and following 10 days of oral Posiphen treatment in four MCI subjects who completed the study. Pharmacokinetic: plasma and CSF drug and primary metabolite concentrations with estimated brain levels extrapolated from steady-state drug administration in rats. RESULTS: Posiphen proved well tolerated and significantly lowered CSF levels of sAPPα, sAPPß, t-τ, p-τ and specific inflammatory markers, and demonstrated a trend to lower CSF Aß(42). CONCLUSIONS: These results confirm preclinical POM studies, demonstrate that pharmacologically relevant drug/metabolite levels reach brain and support the continued clinical optimisation and evaluation of Posiphen for MCI and Alzheimer's disease.


Assuntos
Peptídeos beta-Amiloides/líquido cefalorraquidiano , Precursor de Proteína beta-Amiloide/líquido cefalorraquidiano , Disfunção Cognitiva/tratamento farmacológico , Fragmentos de Peptídeos/líquido cefalorraquidiano , Fisostigmina/análogos & derivados , Proteínas tau/líquido cefalorraquidiano , Animais , Disfunção Cognitiva/sangue , Disfunção Cognitiva/líquido cefalorraquidiano , Relação Dose-Resposta a Droga , Método Duplo-Cego , Feminino , Humanos , Mediadores da Inflamação/líquido cefalorraquidiano , Masculino , Fisostigmina/efeitos adversos , Fisostigmina/farmacocinética , Fisostigmina/farmacologia , Ratos , Ratos Endogâmicos F344
3.
Dig Dis Sci ; 57(7): 1813-21, 2012 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-22526583

RESUMO

BACKGROUND: Inflammatory bowel disease (IBD) is associated with defects in intestinal barriers that rely upon cellular tight junctions. Thus, identifying genes that could be targeted to enforce tight junctions and improve barrier function may lead to new treatment strategies for IBD. AIMS: This preclinical study aimed to evaluate an hypothesized role for the tumor suppressor gene Bin1 as a modifier of the severity of experimental colitis. METHODS: We ablated the Bin1 gene in a mosaic mouse model to evaluate its effects on experimental colitis and intestinal barrier function. Gross pathology, histology and inflammatory cytokine expression patterns were characterized and ex vivo physiology determinations were conducted to evaluate barrier function in intact colon tissue. RESULTS: Bin1 attenuation limited experimental colitis in a sexually dimorphic manner with stronger protection in female subjects. Colitis suppression was associated with an increase in basal transepithelial electrical resistance (TER) and a decrease in paracellular transepithelial flux, compared to control wild-type animals. In contrast, Bin1 attenuation did not affect short circuit current, nor did it alter the epithelial barrier response to non-inflammatory permeability enhancers in the absence of inflammatory stimuli. CONCLUSIONS: Bin1 is a genetic modifier of experimental colitis that controls the paracellular pathway of transcellular ion transport regulated by cellular tight junctions. Our findings offer a preclinical validation of Bin1 as a novel therapeutic target for IBD treatment.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/deficiência , Permeabilidade da Membrana Celular/fisiologia , Colite/prevenção & controle , Mucosa Intestinal/fisiologia , Proteínas do Tecido Nervoso/deficiência , Proteínas Supressoras de Tumor/deficiência , Proteínas Adaptadoras de Transdução de Sinal/genética , Proteínas Adaptadoras de Transdução de Sinal/fisiologia , Animais , Colite/induzido quimicamente , Colite/patologia , Sulfato de Dextrana/efeitos adversos , Modelos Animais de Doenças , Feminino , Mucosa Intestinal/patologia , Masculino , Camundongos , Camundongos Knockout , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/fisiologia , Índice de Gravidade de Doença , Caracteres Sexuais , Junções Íntimas/fisiologia , Proteínas Supressoras de Tumor/genética , Proteínas Supressoras de Tumor/fisiologia
4.
Biochim Biophys Acta ; 1795(1): 25-36, 2009 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-18930786

RESUMO

The evolutionarily conserved amphiphysin-like genes Bin1 and Bin3 function in membrane and actin dynamics, cell polarity, and stress signaling. Recent genetic studies in mice discriminate non-essential roles in endocytic processes commonly ascribed to amphiphysins from essential roles in cancer suppression. Bin1 acts in default pathways of apoptosis and senescence that are triggered by the Myc and Raf oncogenes in primary cells, and Bin1 gene products display a 'moonlighting function' in the nucleus where a variety of other 'endocytic' proteins are also found. Together, genetic investigations in yeast, flies, and mice suggest that amphiphysin-like adapter proteins may suppress cancer by helping integrate cell polarity signals generated by actin and vesicle dynamics with central regulators of cell cycle arrest, apoptosis, and immune surveillance.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/fisiologia , Genes Supressores de Tumor/fisiologia , Neoplasias/genética , Proteínas do Tecido Nervoso/fisiologia , Proteínas Adaptadoras de Transdução de Sinal/genética , Animais , Polaridade Celular/genética , Humanos , Proteínas dos Microfilamentos/genética , Proteínas dos Microfilamentos/fisiologia , Modelos Biológicos , Proteínas do Tecido Nervoso/genética , Proteínas Nucleares/genética , Proteínas Nucleares/fisiologia , Estresse Fisiológico/genética , Proteínas Supressoras de Tumor/genética , Proteínas Supressoras de Tumor/fisiologia
5.
Cancer Immunol Immunother ; 59(11): 1655-63, 2010 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-20640572

RESUMO

Indoleamine 2,3-dioxygenase (IDO) is generally considered to be immunosuppressive but recent findings suggest this characterization oversimplifies its role in disease pathogenesis. Recently, we showed that IDO is essential for tumor outgrowth in the classical two-stage model of inflammatory skin carcinogenesis. Here, we report that IDO loss did not exacerbate classical inflammatory responses. Rather, IDO induction could be elicited by environmental signals and tumor promoters as an integral component of the inflammatory tissue microenvironment even in the absence of cancer. IDO loss had limited impact on tumor outgrowth in carcinogenesis models that lacked an explicit inflammatory tumor promoter. In the context of inflammatory carcinogenesis where IDO was critical to tumor development, the most important source of IDO was radiation-resistant non-hematopoietic cells, consistent with evidence that loss of the IDO regulatory tumor suppressor gene Bin1 in transformed skin cells facilitates IDO-mediated immune escape by a cell autonomous mechanism. Taken together, our results identify IDO as an integral component of 'cancer-associated' inflammation that tilts the immune system toward tumor support. More generally, they promote the concept that mediators of immune escape and cancer-associated inflammation may be genetically synonymous.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/fisiologia , Tolerância Imunológica , Indolamina-Pirrol 2,3,-Dioxigenase/fisiologia , Inflamação/patologia , Proteínas do Tecido Nervoso/fisiologia , Neoplasias Cutâneas/imunologia , Neoplasias Cutâneas/patologia , Proteínas Supressoras de Tumor/fisiologia , 9,10-Dimetil-1,2-benzantraceno/toxicidade , Animais , Medula Óssea/efeitos dos fármacos , Medula Óssea/enzimologia , Medula Óssea/efeitos da radiação , Transplante de Medula Óssea , Carcinógenos/toxicidade , Sinergismo Farmacológico , Humanos , Inflamação/metabolismo , Interferon gama/farmacologia , Interleucina-1beta/farmacologia , Queratinócitos/citologia , Queratinócitos/efeitos dos fármacos , Queratinócitos/efeitos da radiação , Lipopolissacarídeos/farmacologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Knockout , Neoplasias Cutâneas/enzimologia , Acetato de Tetradecanoilforbol/farmacologia , Evasão Tumoral , Células U937 , Irradiação Corporal Total
6.
Cancer Res ; 67(1): 100-7, 2007 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-17210688

RESUMO

Genes that modify oncogenesis may influence dormancy versus progression in cancer, thereby affecting clinical outcomes. The Bin1 gene encodes a nucleocytosolic adapter protein that interacts with and suppresses the cell transforming activity of Myc. Bin1 is often attenuated in breast cancer but its ability to negatively modify oncogenesis or progression in this context has not been gauged directly. In this study, we investigated the effects of mammary gland-specific deletion of Bin1 on initiation and progression of breast cancer in mice. Bin1 loss delayed the outgrowth and involution of the glandular ductal network during pregnancy but had no effect on tumor susceptibility. In contrast, in mice where tumors were initiated by the ras-activating carcinogen 7,12-dimethylbenz(a)anthracene, Bin1 loss strongly accentuated the formation of poorly differentiated tumors characterized by increased proliferation, survival, and motility. This effect was specific as Bin1 loss did not accentuate progression of tumors initiated by an overexpressed mouse mammary tumor virus-c-myc transgene, which on its own produced poorly differentiated and aggressive tumors. These findings suggest that Bin1 loss cooperates with ras activation to drive progression, establishing a role for Bin1 as a negative modifier of oncogenicity and progression in breast cancer.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/deficiência , Proteínas Adaptadoras de Transdução de Sinal/genética , Transformação Celular Neoplásica/genética , Glândulas Mamárias Animais/patologia , Neoplasias Mamárias Experimentais/genética , Proteínas do Tecido Nervoso/deficiência , Proteínas do Tecido Nervoso/genética , Proteínas Supressoras de Tumor/deficiência , Proteínas Supressoras de Tumor/genética , 9,10-Dimetil-1,2-benzantraceno , Animais , Sequência de Bases , Carcinógenos , Transformação Celular Neoplásica/induzido quimicamente , Transformação Celular Neoplásica/patologia , Cocarcinogênese , Progressão da Doença , Feminino , Deleção de Genes , Genes ras , Glândulas Mamárias Animais/efeitos dos fármacos , Glândulas Mamárias Animais/fisiologia , Neoplasias Mamárias Experimentais/induzido quimicamente , Neoplasias Mamárias Experimentais/patologia , Camundongos , Camundongos Transgênicos , Dados de Sequência Molecular , Gravidez
7.
Cancer Res ; 67(16): 7605-12, 2007 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-17699764

RESUMO

Age is the major risk factor for cancer, but few genetic pathways that modify cancer incidence during aging have been described. Bin1 is a prototypic member of the BAR adapter gene family that functions in vesicle dynamics and nuclear processes. Bin1 limits oncogenesis and is often attenuated in human cancers, but its role in cancer suppression has yet to be evaluated fully in vivo. In the mouse, homozygous deletion of Bin1 causes developmental lethality, so to assess this role, we examined cancer incidence in mosaic null mice generated by a modified Cre-lox technology. During study of these animals, one notable phenotype was an extended period of female fecundity during aging, with mosaic null animals retaining reproductive capability until the age of 17.3 +/- 1.1 months. Through 1 year of age, cancer incidence was unaffected by Bin1 ablation; however, by 18 to 20 months of age, approximately 50% of mosaic mice presented with lung adenocarcinoma and approximately 10% with hepatocarcinoma. Aging mosaic mice also displayed a higher incidence of inflammation and/or premalignant lesions, especially in the heart and prostate. In mice where colon tumors were initiated by a ras-activating carcinogen, Bin1 ablation facilitated progression to more aggressive invasive status. In cases of human lung and colon cancers, immunohistochemical analyses evidenced frequent attenuation of Bin1 expression, paralleling observations in other solid tumors. Taken together, our findings highlight an important role for Bin1 as a negative modifier of inflammation and cancer susceptibility during aging.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/deficiência , Proteínas Adaptadoras de Transdução de Sinal/genética , Neoplasias Pulmonares/genética , Proteínas do Tecido Nervoso/deficiência , Proteínas do Tecido Nervoso/genética , Proteínas Supressoras de Tumor/deficiência , Proteínas Supressoras de Tumor/genética , Proteínas Adaptadoras de Transdução de Sinal/fisiologia , Adenocarcinoma/genética , Adenocarcinoma/patologia , Fatores Etários , Animais , Sequência de Bases , Neoplasias do Colo/genética , Neoplasias do Colo/patologia , Feminino , Neoplasias Hepáticas Experimentais/genética , Neoplasias Hepáticas Experimentais/patologia , Neoplasias Pulmonares/patologia , Masculino , Camundongos , Camundongos Knockout , Camundongos Transgênicos , Proteínas do Tecido Nervoso/fisiologia , Lesões Pré-Cancerosas/genética , Lesões Pré-Cancerosas/patologia , Proteínas Supressoras de Tumor/fisiologia
8.
Cancer Discov ; 2(8): 722-35, 2012 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-22822050

RESUMO

UNLABELLED: Indoleamine 2,3-dioxygenase (IDO) enzyme inhibitors have entered clinical trials for cancer treatment based on preclinical studies, indicating that they can defeat immune escape and broadly enhance other therapeutic modalities. However, clear genetic evidence of the impact of IDO on tumorigenesis in physiologic models of primary or metastatic disease is lacking. Investigating the impact of Ido1 gene disruption in mouse models of oncogenic KRAS-induced lung carcinoma and breast carcinoma-derived pulmonary metastasis, we have found that IDO deficiency resulted in reduced lung tumor burden and improved survival in both models. Micro-computed tomographic (CT) imaging further revealed that the density of the underlying pulmonary blood vessels was significantly reduced in Ido1-nullizygous mice. During lung tumor and metastasis outgrowth, interleukin (IL)-6 induction was greatly attenuated in conjunction with the loss of IDO. Biologically, this resulted in a consequential impairment of protumorigenic myeloid-derived suppressor cells (MDSC), as restoration of IL-6 recovered both MDSC suppressor function and metastasis susceptibility in Ido1-nullizygous mice. Together, our findings define IDO as a prototypical integrative modifier that bridges inflammation, vascularization, and immune escape to license primary and metastatic tumor outgrowth. SIGNIFICANCE: This study provides preclinical, genetic proof-of-concept that the immunoregulatory enzyme IDO contributes to autochthonous carcinoma progression and to the creation of a metastatic niche. IDO deficiency in vivo negatively impacted both vascularization and IL-6­dependent, MDSC-driven immune escape, establishing IDO as an overarching factor directing the establishment of a protumorigenic environment.


Assuntos
Indolamina-Pirrol 2,3,-Dioxigenase/antagonistas & inibidores , Neoplasias Pulmonares/enzimologia , Adenocarcinoma/irrigação sanguínea , Adenocarcinoma/enzimologia , Adenocarcinoma/genética , Adenocarcinoma/patologia , Adenocarcinoma de Pulmão , Animais , Transformação Celular Neoplásica/genética , Transformação Celular Neoplásica/metabolismo , Transformação Celular Neoplásica/patologia , Progressão da Doença , Genes ras , Células HL-60 , Humanos , Indolamina-Pirrol 2,3,-Dioxigenase/deficiência , Indolamina-Pirrol 2,3,-Dioxigenase/genética , Indolamina-Pirrol 2,3,-Dioxigenase/metabolismo , Inflamação/tratamento farmacológico , Inflamação/enzimologia , Interleucina-6/biossíntese , Estimativa de Kaplan-Meier , Neoplasias Pulmonares/irrigação sanguínea , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patologia , Neoplasias Pulmonares/secundário , Neoplasias Mamárias Experimentais/enzimologia , Neoplasias Mamárias Experimentais/patologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Metástase Neoplásica , Neovascularização Patológica/enzimologia , Análise de Sobrevida , Células U937
9.
Cancer Biol Ther ; 12(12): 1050-8, 2011 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-22157149

RESUMO

Indoleamine 2,3-dioxygenase (IDO) modifies adaptive immunity, in part by determining the character of inflammatory responses in the tissue microenvironment. Small molecule inhibitors of IDO are being developed to treat cancer, chronic infections and other diseases, so the systemic effects of IDO disruption on inflammatory phenomena may influence the design and conduct of early phase clinical investigations of this new class of therapeutic agents. Here, we report cardiac and gastrointestinal phenotypes observed in IDO deficient mice that warrant consideration in planned assessments of the safety risks involved in clinical development of IDO inhibitors. Calcification of the cardiac endometrium proximal to the right ventricle was a sexually dimorphic strain-specific phenotype with ~30% penetrance in BALB/c mice lacking IDO. Administration of complete Freund's adjuvant containing Toll-like receptor ligands known to induce IDO caused acute pancreatitis in IDO deficient mice, with implications for the design of planned combination studies of IDO inhibitors with cancer vaccines. In an established model of hyperlipidemia, IDO deficiency caused a dramatic elevation in levels of serum triglycerides. In the large intestine, IDO loss only slightly increased sensitivity to induction of acute colitis, but it markedly elevated tumor incidence, multiplicity and staging during inflammatory colon carcinogenesis. Together, our findings suggest potential cardiac and gastrointestinal risks of IDO inhibitors that should be monitored in patients as this new class of drugs enter early clinical development.


Assuntos
Gastroenteropatias/enzimologia , Cardiopatias/enzimologia , Indolamina-Pirrol 2,3,-Dioxigenase/deficiência , Animais , Cálcio/metabolismo , Transformação Celular Neoplásica/metabolismo , Colesterol/sangue , Neoplasias do Colo/enzimologia , Neoplasias do Colo/genética , Endométrio/metabolismo , Feminino , Adjuvante de Freund/efeitos adversos , Adjuvante de Freund/farmacologia , Gastroenteropatias/metabolismo , Gastroenteropatias/patologia , Cardiopatias/metabolismo , Cardiopatias/patologia , Hiperlipidemias/sangue , Hiperlipidemias/enzimologia , Hiperlipidemias/genética , Indolamina-Pirrol 2,3,-Dioxigenase/genética , Indolamina-Pirrol 2,3,-Dioxigenase/metabolismo , Inflamação/enzimologia , Inflamação/metabolismo , Inflamação/patologia , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Knockout , Pancreatite/induzido quimicamente , Pancreatite/enzimologia , Pancreatite/genética , Caracteres Sexuais
10.
Immunity ; 24(4): 405-15, 2006 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-16618599

RESUMO

Allelic exclusion ensures that individual B lymphocytes produce only one kind of antibody molecule. Previous studies have shown that allelic exclusion of the mouse Igkappa locus occurs by the combination of monoallelic silencing and a low level of monoallelic activation for rearrangement combined with a negative feedback loop blocking additional functional rearrangements. Using yeast artificial chromosome-based single-copy isotransgenic mice, we have identified a cis-acting element that negatively regulates rearrangement in this locus, specifically in B cells. The element, termed Sis, resides in the V-J intervening sequence. Sis specifies the targeting of Igkappa transgenes in pre-B and B cells to centromeric heterochromatin and associates with Ikaros, a repressor protein that also colocalizes with centromeric heterochromatin. Significantly, these are hallmarks of silenced endogenous germline Igkappa genes in B cells. These results lead us to propose that Sis participates in the monoallelic silencing aspect of allelic exclusion regulation.


Assuntos
Heterocromatina/genética , Fator de Transcrição Ikaros/genética , Cadeias kappa de Imunoglobulina/genética , Recombinação Genética/genética , Elementos Reguladores de Transcrição/genética , Animais , Rearranjo Gênico de Cadeia Leve de Linfócito B/genética , Inativação Gênica , Genes de Imunoglobulinas , Imageamento Tridimensional , Hibridização in Situ Fluorescente , Camundongos , Camundongos Transgênicos , Reação em Cadeia da Polimerase Via Transcriptase Reversa
11.
Microbiology (Reading) ; 146 ( Pt 7): 1707-1715, 2000 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-10878134

RESUMO

Comamonas testosteroni TA441 degrades phenol by a meta-cleavage pathway after the occurrence of a spontaneous mutation that derepresses the aphKLMNOPQB operon encoding phenol hydroxylase and catechol 2,3-dioxygenase, the enzymes for the initial two steps of the degradation pathway. A gene cluster, aphCEFGHJI, encoding the meta-pathway enzymes for degradation of 2-hydroxymuconic semialdehyde (HMS) to TCA cycle intermediates was found downstream of the aphK operon. The upstream operon and the downstream gene cluster were found to be separated by two open reading frames of unknown function and an oppositely oriented aphT gene, which is similar to regulatory genes for ortho-cleavage of catechol or chlorinated catechols. A promoter assay using an aphC::lacZ transcriptional fusion plasmid revealed that the aphC promoter activity is induced by both phenol and HMS. The phenol-dependent induction was mediated by AphR and the HMS-dependent induction was mediated by AphT. The aphC promoter in strain TA441 was not silenced, unlike the cases of the aphK and aphR promoters, and was highly induced by HMS.


Assuntos
Comamonas testosteroni/genética , Dioxigenases , Genes Bacterianos , Fenol/metabolismo , Biodegradação Ambiental , Catecol 2,3-Dioxigenase , Clonagem Molecular , Comamonas testosteroni/efeitos dos fármacos , Comamonas testosteroni/metabolismo , Regulação para Baixo , Inativação Gênica , Genes Reguladores , Oxigenases de Função Mista/biossíntese , Oxigenases de Função Mista/genética , Dados de Sequência Molecular , Família Multigênica , Mutação , Óperon , Oxigenases/biossíntese , Oxigenases/genética , Fenol/farmacologia , Filogenia
12.
Mol Microbiol ; 49(1): 235-40, 2003 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-12823824

RESUMO

Whereas diphtheria and the mechanism of action of diphtheria toxin, the bacterial molecule that induces the disease, have been studied and understood for some time, the receptor that allows animal cells to bind the toxin escaped identification until recently. The receptor was identified by its ability to confer toxin-sensitivity to mouse cells, which are normally toxin-resistant. Although mice are also naturally resistant, we now demonstrate that transgenic mice expressing the diphtheria toxin receptor are as sensitive to the toxin as are humans and other toxin-sensitive animals. These transgenic mice provide a suitable model for studying modern antidotes for diphtheria.


Assuntos
Toxina Diftérica/metabolismo , Toxina Diftérica/toxicidade , Receptores de Superfície Celular/genética , Receptores de Superfície Celular/metabolismo , Animais , Encéfalo/metabolismo , Fator de Crescimento Semelhante a EGF de Ligação à Heparina , Humanos , Peptídeos e Proteínas de Sinalização Intercelular , Camundongos , Camundongos Transgênicos , Miócitos Cardíacos/metabolismo , Proteínas Recombinantes/farmacologia , Recombinação Genética , Taxa de Sobrevida , Transgenes
13.
Infect Immun ; 70(5): 2344-50, 2002 May.
Artigo em Inglês | MEDLINE | ID: mdl-11953369

RESUMO

Although equine diphtheria antitoxin may be an effective therapy for human diphtheria, its use often induces serum sickness. We describe here a strategy for developing an alternative treatment based on the human diphtheria toxin (DT) receptor/heparin-binding epidermal growth factor-like growth factor (HB-EGF) precursor. Recombinant mature human HB-EGF acts as a soluble receptor analog, binding radioiodinated DT and preventing its binding to the cellular DT receptor/HB-EGF precursor. However, the possibility existed that radioiodinated DT-HB-EGF complexes associate with cells due to the binding of the heparin-binding domain of recombinant HB-EGF to cell surface heparan sulfate proteoglycans. This possibility was confirmed by performing DT binding studies in the presence of heparin. A recombinant truncated HB-EGF (residues 106 to 149), which lacks most of the heparin-binding domain, showed an essentially heparin-independent binding of radioiodinated DT to cells. Furthermore, it was a more effective inhibitor of DT binding than was recombinant mature HB-EGF. Since mature HB-EGF is a known ligand for the EGF receptor and is thus highly mitogenic (tumorigenic), we then changed amino acid residues in the EGF-like domain of the recombinant truncated HB-EGF and demonstrated that this DT receptor analog (I117A/L148A) displayed a low mitogenic effect. The truncated (I117A/L148A) HB-EGF protein retained high DT binding affinity, as confirmed by using surface plasmon resonance. Our results suggest that the truncated (I117A/L148A) HB-EGF protein could be an effective, safe antidote for human diphtheria.


Assuntos
Antídotos/uso terapêutico , Difteria/tratamento farmacológico , Fator de Crescimento Epidérmico/uso terapêutico , Receptores de Superfície Celular/metabolismo , Células 3T3 , Sequência de Aminoácidos , Animais , Toxina Diftérica/metabolismo , Fator de Crescimento Epidérmico/metabolismo , Fator de Crescimento Epidérmico/farmacologia , Heparina/farmacologia , Fator de Crescimento Semelhante a EGF de Ligação à Heparina , Peptídeos e Proteínas de Sinalização Intercelular , Camundongos , Mitógenos/farmacologia , Dados de Sequência Molecular
14.
J Biol Chem ; 277(36): 32640-9, 2002 Sep 06.
Artigo em Inglês | MEDLINE | ID: mdl-12080064

RESUMO

To identify new regulatory elements within the mouse Igkappa locus, we have mapped DNase I hypersensitive sites (HSs) in the chromatin of B cell lines arrested at different stages of differentiation. We have focused on two regions encompassing 50 kilobases suspected to contain new regulatory elements based on our previous high level expression results with yeast artificial chromosome-based mouse Igkappa transgenes. This approach has revealed a cluster of HSs within the 18-kilobase intervening sequence, which we cloned and sequenced in its entirety, between the Vkappa gene closest to the Jkappa region. These HSs exhibit pro/pre-B cell-specific transcriptional silencing of a Vkappa gene promoter in transient transfection assays. We also identified a plasmacytoma cell-specific HS in the far downstream region of the locus, which in analogous transient transfection assays proved to be a powerful transcriptional enhancer. Deletional analyses reveal that for each element multiple DNA segments cooperate to achieve either silencing or enhancement. The enhancer sequence is conserved in the human Igkappa gene locus, including NF-kappaB and E-box sites that are important for the activity. In summary, our results pinpoint the locations of presumptive regulatory elements for future knockout studies to define their functional roles in the native locus.


Assuntos
Cromatina/química , Elementos Facilitadores Genéticos , Inativação Gênica , Cadeias kappa de Imunoglobulina/genética , Transcrição Gênica , Animais , Sequência de Bases , Sítios de Ligação , Linhagem Celular , Cromatina/metabolismo , Cromossomos Artificiais de Levedura , DNA/metabolismo , Desoxirribonuclease I/metabolismo , Luciferases/metabolismo , Camundongos , Camundongos Transgênicos , Modelos Genéticos , Dados de Sequência Molecular , Mutagênese Sítio-Dirigida , NF-kappa B/metabolismo , Reação em Cadeia da Polimerase , Homologia de Sequência do Ácido Nucleico , Fatores de Tempo , Ativação Transcricional , Transfecção , Transgenes
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa