Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
1.
J Clin Invest ; 117(12): 3979-87, 2007 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-17992261

RESUMO

The liver produces plasma sex hormone-binding globulin (SHBG), which transports sex steroids and regulates their access to tissues. In overweight children and adults, low plasma SHBG levels are a biomarker of the metabolic syndrome and its associated pathologies. Here, we showed in transgenic mice and HepG2 hepatoblastoma cells that monosaccharides (glucose and fructose) reduce human SHBG production by hepatocytes. This occurred via a downregulation of hepatocyte nuclear factor-4alpha (HNF-4alpha) and replacement of HNF-4alpha by the chicken OVA upstream promoter-transcription factor 1 at a cis-element within the human SHBG promoter, coincident with repression of its transcriptional activity. The dose-dependent reduction of HNF-4alpha levels in HepG2 cells after treatment with glucose or fructose occurred in concert with parallel increases in cellular palmitate levels and could be mimicked by treatment with palmitoyl-CoA. Moreover, inhibition of lipogenesis prevented monosaccharide-induced downregulation of HNF-4alpha and reduced SHBG expression in HepG2 cells. Thus, monosaccharide-induced lipogenesis reduced hepatic HNF-4alpha levels, which in turn attenuated SHBG expression. This provides a biological explanation for why SHBG is a sensitive biomarker of the metabolic syndrome and the metabolic disturbances associated with increased fructose consumption.


Assuntos
Frutose/farmacologia , Glucose/farmacologia , Fator 4 Nuclear de Hepatócito/metabolismo , Lipogênese/efeitos dos fármacos , Globulina de Ligação a Hormônio Sexual/biossíntese , Edulcorantes/farmacologia , Adulto , Animais , Biomarcadores/sangue , Linhagem Celular Tumoral , Galinhas/genética , Criança , Pré-Escolar , Frutose/efeitos adversos , Regulação da Expressão Gênica/efeitos dos fármacos , Regulação da Expressão Gênica/genética , Glucose/efeitos adversos , Hormônios Esteroides Gonadais/sangue , Fator 1-alfa Nuclear de Hepatócito/genética , Fator 1-alfa Nuclear de Hepatócito/metabolismo , Humanos , Lipogênese/genética , Síndrome Metabólica/sangue , Síndrome Metabólica/genética , Síndrome Metabólica/patologia , Camundongos , Camundongos Transgênicos , Sobrepeso/sangue , Sobrepeso/genética , Sobrepeso/patologia , Globulina de Ligação a Hormônio Sexual/genética , Edulcorantes/efeitos adversos
2.
J Clin Invest ; 109(7): 973-81, 2002 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-11927624

RESUMO

The access of testosterone and estradiol to target tissues is regulated by sex hormone-binding globulin (SHBG) in human blood. Serum SHBG levels are low in patients with hyperandrogenism, especially in association with polycystic ovarian syndrome (PCOS) and in individuals at risk for diabetes and heart disease. Here, we identify SHBG coding region variations from a compound heterozygous patient who presented with severe hyperandrogenism during pregnancy. Serum SHBG levels in this patient measured 2 years after her pregnancy were exceptionally low, and her non-protein-bound testosterone concentrations greatly exceeded the normal reference range. A single-nucleotide polymorphism within the proband's maternally derived SHBG allele encodes a missense mutation, P156L, which allows for normal steroid ligand binding but causes abnormal glycosylation and inefficient secretion of SHBG. This polymorphism was identified in four other patients with either PCOS, ioiopathic hirsutism, or ovarian failure. The proband's paternal SHBG allele carries a single-nucleotide deletion within exon 8, producing a reading-frame shift within the codon for E326 and a premature termination codon. CHO cells transfected with a SHBG cDNA carrying this mutation fail to secrete the predicted truncated form of SHBG. To our knowledge, these are the first examples of human SHBG variants linked to hyperandrogenism and ovarian dysfunction.


Assuntos
Variação Genética , Hiperandrogenismo/genética , Síndrome do Ovário Policístico/genética , Globulina de Ligação a Hormônio Sexual/genética , Adulto , Alelos , Animais , Sítios de Ligação , Células CHO , Cricetinae , Desoxirribonuclease HpaII , Éxons , Feminino , Deleção de Genes , Expressão Gênica , Testes Genéticos , Humanos , Ovário/fisiopatologia , Polimorfismo Genético , Gravidez , Testosterona/metabolismo
3.
Artigo em Inglês | MEDLINE | ID: mdl-27273980

RESUMO

With the growing numbers of nanomaterials (NMs), there is a great demand for rapid and reliable ways of testing NM safety-preferably using in vitro approaches, to avoid the ethical dilemmas associated with animal research. Data are needed for developing intelligent testing strategies for risk assessment of NMs, based on grouping and read-across approaches. The adoption of high throughput screening (HTS) and high content analysis (HCA) for NM toxicity testing allows the testing of numerous materials at different concentrations and on different types of cells, reduces the effect of inter-experimental variation, and makes substantial savings in time and cost. HTS/HCA approaches facilitate the classification of key biological indicators of NM-cell interactions. Validation of in vitro HTS tests is required, taking account of relevance to in vivo results. HTS/HCA approaches are needed to assess dose- and time-dependent toxicity, allowing prediction of in vivo adverse effects. Several HTS/HCA methods are being validated and applied for NM testing in the FP7 project NANoREG, including Label-free cellular screening of NM uptake, HCA, High throughput flow cytometry, Impedance-based monitoring, Multiplex analysis of secreted products, and genotoxicity methods-namely High throughput comet assay, High throughput in vitro micronucleus assay, and γH2AX assay. There are several technical challenges with HTS/HCA for NM testing, as toxicity screening needs to be coupled with characterization of NMs in exposure medium prior to the test; possible interference of NMs with HTS/HCA techniques is another concern. Advantages and challenges of HTS/HCA approaches in NM safety are discussed. WIREs Nanomed Nanobiotechnol 2017, 9:e1413. doi: 10.1002/wnan.1413 For further resources related to this article, please visit the WIREs website.


Assuntos
Ensaios de Triagem em Larga Escala/métodos , Nanoestruturas/toxicidade , Testes de Toxicidade/métodos , Animais , Linhagem Celular , Técnicas Citológicas , Humanos , Espaço Intracelular/química , Espaço Intracelular/metabolismo , Camundongos
4.
Hum Fertil (Camb) ; 9(2): 73-9, 2006 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-16825108

RESUMO

Spermatogenesis is a remarkably complex process in which diploid spermatogonial stem cells undergo a series of mitotic and meiotic cell divisions to give rise to haploid round spermatids. These haploid cells then go through a dramatic morphological remodelling involving extensive chromatin condensation, reduction in nuclear and cytoplasmic volume, formation of an acrosome system and tail, all of which contribute to the formation of a mature spermatozoon fully capable of fertilizing the oocyte and passing along its genetic information to the next generation. To accomplish such a complex program, an intricate and efficient mechanism is required to finely tune the levels of expression of specific genes necessary for this process. Accordingly, the regulation of gene expression in post-meiotic male germ cells is governed by specific mechanisms unique to these cells. The cyclic adenosine monophosphate (cAMP) response element modulator (CREM) is an essential component of this program, and its activity is regulated through interactions with a germ cell-specific, CREM phosphorylation-independent transcriptional co-activator, activator of CREM in testis (ACT). In turn, the ability of ACT to regulate CREM activity is controlled by a germ cell-specific kinesin, Kif17b, which regulates the subcellular distribution of ACT. Further, the mRNA from CREM target genes interacts with several germ cell-specific RNA-binding proteins, which function to transport and stabilize these mRNAs. This sophisticated and complex regulation of gene expression in post-meiotic germ cells is governed by unique mechanisms specific to these cells and is fundamental to male fertility.


Assuntos
Modulador de Elemento de Resposta do AMP Cíclico/fisiologia , Regulação da Expressão Gênica/fisiologia , Espermatogênese/fisiologia , Espermatozoides/fisiologia , Fatores de Transcrição/fisiologia , Modulador de Elemento de Resposta do AMP Cíclico/genética , Regulação da Expressão Gênica/genética , Humanos , Cinesinas/genética , Cinesinas/fisiologia , Proteínas com Domínio LIM , Masculino , Meiose/genética , Meiose/fisiologia , Proteínas Motores Moleculares/genética , Proteínas Motores Moleculares/fisiologia , Espermatogênese/genética , Fatores de Transcrição/genética , Transcrição Gênica
5.
J Neurosci Methods ; 192(2): 268-76, 2010 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-20709102

RESUMO

Analyses using antibodies directed against α-synuclein play a key role in the understanding of the pathologies associated with neurodegenerative disorders such as Parkinson's disease (PD), dementia with Lewy bodies (DLB) and multiple system atrophy (MSA). However, the generation of antibodies against immunogens with significant sequence similarity to host proteins such as α-synuclein is often hindered by host immunotolerance. In contrast to wild-type C57BL/6J and BALB/c mice immunized with recombinant human α-synuclein, C57BL/6S Δsnca mice presenting a natural deletion of the α-synuclein locus, bypassed the immunotolerance process which resulted in a much higher polyclonal antibody response. The native or fibrillized conformation of α-synuclein used as the immunogen did not have an impact on the amounts of specific antibodies in sera of the host. The immunization protocols resulted in the generation of the IgG AS11, raised against fibrillized recombinant human α-synuclein in C57BL/6S Δsnca mice. This monoclonal antibody, recognizing an N-terminal α-synuclein epitope, was selected for its specificity and significant reactivity in Western-blot, immunofluorescence and immunohistochemistry assays. The ability of AS11 to detect both soluble and aggregated forms of α-synuclein present in pathological cytoplasmic inclusions was further assessed using analysis of human brains with PD or MSA, transgenic mouse lines expressing A53T human α-synuclein, and cellular models expressing human α-synuclein. Taken together, our study indicates that novel antibodies helpful to characterize alterations of α-synuclein leading to neurodegeneration in PD and related disorders could be efficiently developed using this original immunization strategy.


Assuntos
Anticorpos Monoclonais/biossíntese , alfa-Sinucleína/imunologia , Animais , Anticorpos Monoclonais/imunologia , Western Blotting , Linhagem Celular , Células Cultivadas , Humanos , Imuno-Histoquímica , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , alfa-Sinucleína/genética
6.
Cell ; 120(3): 287-8, 2005 Feb 11.
Artigo em Inglês | MEDLINE | ID: mdl-15707884

RESUMO

Homeobox transcription factors exert essential roles in embryogenesis and are thought to govern regenerative cell differentiation. In this issue of Cell, MacLean and colleagues (MacLean et al., 2005) describe a new homeobox gene cluster composed of genes selectively expressed in reproductive tissues. Remarkably, the cluster is on the X chromosome, and the genes display a colinear pattern of expression.


Assuntos
Gametogênese/genética , Genes Homeobox/genética , Gônadas/embriologia , Organogênese/genética , Diferenciação Sexual/genética , Animais , Feminino , Regulação da Expressão Gênica no Desenvolvimento/genética , Gônadas/citologia , Gônadas/metabolismo , Humanos , Masculino , Família Multigênica/genética , Cromossomo X/genética
7.
J Biol Chem ; 280(6): 4462-8, 2005 Feb 11.
Artigo em Inglês | MEDLINE | ID: mdl-15574421

RESUMO

Expression of the sex hormone-binding globulin gene (SHBG) in the liver produces SHBG, which transports sex steroids in the blood. In rodents, the SHBG gene is also expressed in Sertoli cells giving rise to the testicular androgen-binding protein, which is secreted into the seminiferous tubule where it presumably controls testosterone action. Evidence that the SHBG gene functions in this way in the human testis is lacking, and mice containing a human SHBG transgene (shbg4) under the control of its own promoter sequence are characterized by SHBG gene expression in the liver but not in the testis. A potential cis-element, defined as footprint 4 (FP4) within the human SHBG promoter, is absent in SHBG promoters of mammals that produce the testicular androgen-binding protein, and we have produced mice harboring a shbg4 transgene in which FP4 was deleted to evaluate its functional significance. Remarkably, these mice express the modified human SHBG transgene in the testis as well as the liver. Human SHBG transcripts were found within their Sertoli cells, primary cultures of which secrete human SHBG, and this was increased by treatment with follicle-stimulating hormone, retinoic acid, and estradiol but not testosterone. We have also found that the upstream stimulatory factors (USF-1 and USF-2) bind FP4 in vitro by electromobility shift assay of Sertoli cell nuclear extracts and in vivo by chromatin immunoprecipitation assay and conclude that USF transcription factors repress human SHBG transcription in Sertoli cells through an interaction with FP4 within its proximal promoter.


Assuntos
Células de Sertoli/metabolismo , Globulina de Ligação a Hormônio Sexual/biossíntese , Globulina de Ligação a Hormônio Sexual/genética , Animais , Sequência de Bases , Imunoprecipitação da Cromatina , Proteínas de Ligação a DNA/metabolismo , Estradiol/metabolismo , Hormônio Foliculoestimulante/metabolismo , Regulação da Expressão Gênica , Humanos , Fígado/metabolismo , Masculino , Camundongos , Camundongos Transgênicos , Microscopia de Fluorescência , Dados de Sequência Molecular , Filogenia , Regiões Promotoras Genéticas , Ligação Proteica , RNA/metabolismo , Homologia de Sequência de Aminoácidos , Testículo/metabolismo , Fatores de Tempo , Fatores de Transcrição/metabolismo , Transgenes , Tretinoína/metabolismo , Fatores Estimuladores Upstream
8.
J Biol Chem ; 277(47): 45291-8, 2002 Nov 22.
Artigo em Inglês | MEDLINE | ID: mdl-12235141

RESUMO

Human sex hormone-binding globulin (SHBG) binds estradiol and testosterone with high affinity. Plasma SHBG is produced by hepatocytes, but the human SHBG gene is also expressed in the testis. Little is known about SHBG gene expression in the human testis, but human SHBG transcripts accumulate in a spermatogenic stage-dependent manner in the testes of mice containing an 11-kb human SHBG transgene. We have now found that human SHBG transcripts containing an alternative exon 1 sequence are located specifically in the testicular germ cells of these transgenic mice, whereas murine SHBG transcripts are confined to Sertoli cells. In addition, we have detected immunoreactive human SHBG in the acrosome during all stages of spermiogenesis in mice containing an 11-kb human SHBG transgene. Western blots of germ cell extracts from these transgenic mice and from human sperm indicate that the immunoreactive human SHBG in the acrosome composes electrophoretic variants, which are 3-5 kDa smaller than the major electrophoretic isoforms of human SHBG in the blood. This apparent size difference is due in part to differences in glycosylation of plasma and acrosomal SHBG isoforms. The function of the human SHBG isoform in the acrosome is unknown, but it binds steroid ligands with high affinity. This is the first demonstration that human SHBG transcripts encode an SHBG isoform that remains within a cellular compartment.


Assuntos
Acrossomo/metabolismo , Globulina de Ligação a Hormônio Sexual/metabolismo , Espermatogênese/fisiologia , Acrossomo/química , Animais , Éxons/genética , Células Germinativas/citologia , Células Germinativas/metabolismo , Humanos , Masculino , Camundongos , Camundongos Transgênicos , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Globulina de Ligação a Hormônio Sexual/genética , Espermatozoides/citologia , Espermatozoides/metabolismo , Testículo/fisiologia , Transgenes
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA