Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 37
Filtrar
Mais filtros












Base de dados
Intervalo de ano de publicação
1.
Nature ; 626(7997): 128-135, 2024 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-38233523

RESUMO

The assembly and specification of synapses in the brain is incompletely understood1-3. Latrophilin-3 (encoded by Adgrl3, also known as Lphn3)-a postsynaptic adhesion G-protein-coupled receptor-mediates synapse formation in the hippocampus4 but the mechanisms involved remain unclear. Here we show in mice that LPHN3 organizes synapses through a convergent dual-pathway mechanism: activation of Gαs signalling and recruitment of phase-separated postsynaptic protein scaffolds. We found that cell-type-specific alternative splicing of Lphn3 controls the LPHN3 G-protein-coupling mode, resulting in LPHN3 variants that predominantly signal through Gαs or Gα12/13. CRISPR-mediated manipulation of Lphn3 alternative splicing that shifts LPHN3 from a Gαs- to a Gα12/13-coupled mode impaired synaptic connectivity as severely as the overall deletion of Lphn3, suggesting that Gαs signalling by LPHN3 splice variants mediates synapse formation. Notably, Gαs-coupled, but not Gα12/13-coupled, splice variants of LPHN3 also recruit phase-transitioned postsynaptic protein scaffold condensates, such that these condensates are clustered by binding of presynaptic teneurin and FLRT ligands to LPHN3. Moreover, neuronal activity promotes alternative splicing of the synaptogenic Gαs-coupled variant of LPHN3. Together, these data suggest that activity-dependent alternative splicing of a key synaptic adhesion molecule controls synapse formation by parallel activation of two convergent pathways: Gαs signalling and clustered phase separation of postsynaptic protein scaffolds.


Assuntos
Processamento Alternativo , Receptores Acoplados a Proteínas G , Receptores de Peptídeos , Sinapses , Animais , Camundongos , Processamento Alternativo/genética , Subunidades alfa G12-G13 de Proteínas de Ligação ao GTP , Subunidades alfa Gs de Proteínas de Ligação ao GTP , Ligantes , Receptores Acoplados a Proteínas G/deficiência , Receptores Acoplados a Proteínas G/genética , Receptores Acoplados a Proteínas G/metabolismo , Receptores de Peptídeos/deficiência , Receptores de Peptídeos/genética , Receptores de Peptídeos/metabolismo , Sinapses/metabolismo , Transdução de Sinais
2.
FASEB J ; 35(5): e21515, 2021 05.
Artigo em Inglês | MEDLINE | ID: mdl-33811688

RESUMO

The conserved endoplasmic reticulum (ER) membrane protein TRAPα (translocon-associated protein, also known as signal sequence receptor 1, SSR1) has been reported to play a critical but unclear role in insulin biosynthesis. TRAPα/SSR1 is one component of a four-protein complex including TRAPß/SSR2, TRAPγ/SSR3, and TRAPδ/SSR4. The TRAP complex topologically has a small exposure on the cytosolic side of the ER via its TRAPγ/SSR3 subunit, whereas TRAPß/SSR2 and TRAPδ/SSR4 function along with TRAPα/SSR1 largely on the luminal side of the ER membrane. Here, we have examined pancreatic ß-cells with deficient expression of either TRAPß/SSR2 or TRAPδ/SSR4, which does not perturb mRNA expression levels of other TRAP subunits, or insulin mRNA. However, deficient protein expression of TRAPß/SSR2 and, to a lesser degree, TRAPδ/SSR4, diminishes the protein levels of other TRAP subunits, concomitant with deficient steady-state levels of proinsulin and insulin. Deficient TRAPß/SSR2 or TRAPδ/SSR4 is not associated with any apparent defect of exocytotic mechanism but rather by a decreased abundance of the proinsulin and insulin that accompanies glucose-stimulated secretion. Amino acid pulse labeling directly establishes that much of the steady-state deficiency of intracellular proinsulin can be accounted for by diminished proinsulin biosynthesis, observed in a pulse-labeling as short as 5 minutes. The proinsulin and insulin levels in TRAPß/SSR2 or TRAPδ/SSR4 null mutant ß-cells are notably recovered upon re-expression of the missing TRAP subunit, accompanying a rebound of proinsulin biosynthesis. Remarkably, overexpression of TRAPα/SSR1 can also suppress defects in ß-cells with diminished expression of TRAPß/SSR2, strongly suggesting that TRAPß/SSR2 is needed to support TRAPα/SSR1 function.


Assuntos
Proteínas de Ligação ao Cálcio/deficiência , Retículo Endoplasmático/metabolismo , Glucose/metabolismo , Insulina/biossíntese , Insulinoma/patologia , Glicoproteínas de Membrana/deficiência , Proinsulina/biossíntese , Receptores Citoplasmáticos e Nucleares/deficiência , Receptores de Peptídeos/deficiência , Animais , Células Cultivadas , Células Secretoras de Insulina/citologia , Insulinoma/metabolismo , Neoplasias Pancreáticas/metabolismo , Neoplasias Pancreáticas/patologia , Ratos
3.
Int J Mol Sci ; 21(21)2020 Nov 02.
Artigo em Inglês | MEDLINE | ID: mdl-33147779

RESUMO

Hyaline fibromatosis syndrome (HFS), resulting from ANTXR2 mutations, is an ultra-rare disease that causes intestinal lymphangiectasia and protein-losing enteropathy (PLE). The mechanisms leading to the gastrointestinal phenotype in these patients are not well defined. We present two patients with congenital diarrhea, severe PLE and unique clinical features resulting from deleterious ANTXR2 mutations. Intestinal organoids were generated from one of the patients, along with CRISPR-Cas9 ANTXR2 knockout, and compared with organoids from two healthy controls. The ANTXR2-deficient organoids displayed normal growth and polarity, compared to controls. Using an anthrax-toxin assay we showed that the c.155C>T mutation causes loss-of-function of ANTXR2 protein. An intrinsic defect of monolayer formation in patient-derived or ANTXR2KO organoids was not apparent, suggesting normal epithelial function. However, electron microscopy and second harmonic generation imaging showed abnormal collagen deposition in duodenal samples of these patients. Specifically, collagen VI, which is known to bind ANTXR2, was highly expressed in the duodenum of these patients. In conclusion, despite resistance to anthrax-toxin, epithelial cell function, and specifically monolayer formation, is intact in patients with HFS. Nevertheless, loss of ANTXR2-mediated signaling leads to collagen VI accumulation in the duodenum and abnormal extracellular matrix composition, which likely plays a role in development of PLE.


Assuntos
Colágeno/metabolismo , Duodeno/metabolismo , Síndrome da Fibromatose Hialina/metabolismo , Enteropatias Perdedoras de Proteínas/metabolismo , Receptores de Peptídeos/genética , Antígenos de Bactérias/química , Toxinas Bacterianas/química , Sistemas CRISPR-Cas , Consanguinidade , Diarreia/congênito , Matriz Extracelular/metabolismo , Humanos , Síndrome da Fibromatose Hialina/genética , Lactente , Masculino , Microscopia Eletrônica , Mutação , Fenótipo , Enteropatias Perdedoras de Proteínas/genética , Receptores de Peptídeos/deficiência , Transdução de Sinais
4.
J Clin Invest ; 128(7): 2927-2943, 2018 07 02.
Artigo em Inglês | MEDLINE | ID: mdl-29863500

RESUMO

Although nonmalignant stromal cells facilitate tumor growth and can occupy up to 90% of a solid tumor mass, better strategies to exploit these cells for improved cancer therapy are needed. Here, we describe a potent MMAE-linked antibody-drug conjugate (ADC) targeting tumor endothelial marker 8 (TEM8, also known as ANTXR1), a highly conserved transmembrane receptor broadly overexpressed on cancer-associated fibroblasts, endothelium, and pericytes. Anti-TEM8 ADC elicited potent anticancer activity through an unexpected killing mechanism we term DAaRTS (drug activation and release through stroma), whereby the tumor microenvironment localizes active drug at the tumor site. Following capture of ADC prodrug from the circulation, tumor-associated stromal cells release active MMAE free drug, killing nearby proliferating tumor cells in a target-independent manner. In preclinical studies, ADC treatment was well tolerated and induced regression and often eradication of multiple solid tumor types, blocked metastatic growth, and prolonged overall survival. By exploiting TEM8+ tumor stroma for targeted drug activation, these studies reveal a drug delivery strategy with potential to augment therapies against multiple cancer types.


Assuntos
Imunoconjugados/farmacologia , Proteínas de Neoplasias/antagonistas & inibidores , Neoplasias/tratamento farmacológico , Receptores de Superfície Celular/antagonistas & inibidores , Animais , Antineoplásicos/farmacocinética , Antineoplásicos/farmacologia , Biomarcadores Tumorais/antagonistas & inibidores , Biomarcadores Tumorais/deficiência , Biomarcadores Tumorais/genética , Brentuximab Vedotin , Linhagem Celular Tumoral , Feminino , Humanos , Imunoconjugados/farmacocinética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Nus , Camundongos SCID , Proteínas dos Microfilamentos , Neoplasias/metabolismo , Receptores de Peptídeos/antagonistas & inibidores , Receptores de Peptídeos/deficiência , Receptores de Peptídeos/genética , Células Estromais/efeitos dos fármacos , Microambiente Tumoral/efeitos dos fármacos , Ensaios Antitumorais Modelo de Xenoenxerto
5.
J Immunoassay Immunochem ; 38(1): 21-33, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-27379831

RESUMO

The failure of therapies targeting tumor angiogenesis may be caused by anti-angiogenic resistance mechanisms induced by VEGF and non-VEGF pathways alterations. Anti-angiogenic therapy failure is also attributed to immune system, acting by tumor-associated macrophages that release pro-angiogenic factors and a consequent increase of blood vessels. Recently, in a study by Rheal et al., a new angiogenic receptor, epidermal growth factor, latrophilin, and 7 trans-membrane domain-containing protein 1 on chromosome 1(ELTD1) has been identified as a promising glioma biomarker. In this study we aim to analyse whether this receptor may be used as a target molecule in glioblastoma therapy. Our results showed that small interfering RNA silencing ELTD1 caused cytotoxicity in glioblastoma cells. We also found that PDGFR, VEGFR, and their common PI3K/mTOR intracellular pathway inactivation-induced cytotoxicity in glioblastoma cells. Further, we found high percent of cytotoxicity in a low passage glioblastoma cell line after BEZ235 (a dual inhibitor of PI3K/mTOR pathway) treatment at nanomolar concentrations, compared to AG1433 (a PDGFR inhibitor) and SU1498 (a VEGFR inhibitor) that were only cytotoxic at micromolar ranges. In the future, these could prove as attractive therapeutic targets in single therapy or coupled with classic therapeutic approaches such as chemotherapy of radiotherapy.


Assuntos
Fator de Crescimento Epidérmico/deficiência , Inativação Gênica , Glioblastoma/tratamento farmacológico , Glioblastoma/patologia , RNA Interferente Pequeno/genética , Receptores Acoplados a Proteínas G/deficiência , Receptores de Peptídeos/deficiência , Biomarcadores Tumorais/biossíntese , Biomarcadores Tumorais/deficiência , Biomarcadores Tumorais/genética , Morte Celular/efeitos dos fármacos , Fator de Crescimento Epidérmico/biossíntese , Fator de Crescimento Epidérmico/genética , Inativação Gênica/efeitos dos fármacos , Glioblastoma/genética , Humanos , RNA Interferente Pequeno/farmacologia , Receptores Acoplados a Proteínas G/biossíntese , Receptores Acoplados a Proteínas G/genética , Receptores de Peptídeos/biossíntese , Receptores de Peptídeos/genética
6.
Proc Natl Acad Sci U S A ; 113(8): 2294-9, 2016 Feb 23.
Artigo em Inglês | MEDLINE | ID: mdl-26842836

RESUMO

Rupture of the ovarian follicle releases the oocyte at ovulation, a timed event that is critical for fertilization. It is not understood how the protease activity required for rupture is directed with precise timing and localization to the outer surface, or apex, of the follicle. We hypothesized that vasoconstriction at the apex is essential for rupture. The diameter and blood flow of individual vessels and the thickness of the apical follicle wall were examined over time to expected ovulation using intravital multiphoton microscopy. Vasoconstriction of apical vessels occurred within hours preceding follicle rupture in wild-type mice, but vasoconstriction and rupture were absent in Amhr2(cre/+)SmoM2 mice in which follicle vessels lack the normal association with vascular smooth muscle. Vasoconstriction is not simply a response to reduced thickness of the follicle wall; vasoconstriction persisted in wild-type mice when thinning of the follicle wall was prevented by infusion of protease inhibitors into the ovarian bursa. Ovulation was inhibited by preventing the periovulatory rise in the expression of the vasoconstrictor endothelin 2 by follicle cells of wild-type mice. In these mice, infusion of vasoconstrictors (either endothelin 2 or angiotensin 2) into the bursa restored the vasoconstriction of apical vessels and ovulation. Additionally, infusion of endothelin receptor antagonists into the bursa of wild-type mice prevented vasoconstriction and follicle rupture. Processing tissue to allow imaging at increased depth through the follicle and transabdominal ultrasonography in vivo showed that decreased blood flow is restricted to the apex. These results demonstrate that vasoconstriction at the apex of the follicle is essential for ovulation.


Assuntos
Folículo Ovariano/irrigação sanguínea , Folículo Ovariano/fisiologia , Ovulação/fisiologia , Vasoconstrição/fisiologia , Animais , Endotelina-2/deficiência , Endotelina-2/genética , Endotelina-2/fisiologia , Feminino , Microscopia Intravital , Camundongos , Camundongos da Linhagem 129 , Camundongos Transgênicos , Microscopia de Fluorescência por Excitação Multifotônica , Folículo Ovariano/diagnóstico por imagem , Ovulação/genética , Receptores Acoplados a Proteínas G/deficiência , Receptores Acoplados a Proteínas G/genética , Receptores Acoplados a Proteínas G/fisiologia , Receptores de Peptídeos/deficiência , Receptores de Peptídeos/genética , Receptores de Peptídeos/fisiologia , Receptores de Fatores de Crescimento Transformadores beta/deficiência , Receptores de Fatores de Crescimento Transformadores beta/genética , Receptores de Fatores de Crescimento Transformadores beta/fisiologia , Receptor Smoothened , Ultrassonografia , Vasoconstrição/genética
7.
PLoS One ; 10(7): e0133083, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26193363

RESUMO

Food intake and energy metabolism are tightly controlled to maintain stable energy homeostasis and healthy states. Thus, animals detect their stored energy levels, and based on this, they determine appropriate food intake and meal size. Drosophila melanogaster putative G protein-coupled receptor, Bride of sevenless (BOSS) is a highly evolutionarily conserved protein that responds to extracellular glucose levels in order to regulate energy homeostasis. To address how BOSS regulates energy homeostasis, we characterized a boss mutant by assessing its food intake and stored energy levels. Boss mutants exhibited increased food intake but decreased stored triacylglyceride levels. Using boss-GAL4 drivers, we found that boss is expressed in select tissues that are involved in nutrient sensing and food intake, in a subset of neurons in brain and chemosensory organs, in fat body, and in endocrine cells in gut (enteroendocrine cells). Flies with tissue-specific boss knockdowns in these tissues had abnormal stored energy levels and abnormal food intake. These results suggest that BOSS in either neurons or peripheral nutrient-sensing tissues affects energy homeostasis in ways that relate to the sensing of nutrients and regulation of food intake.


Assuntos
Proteínas de Drosophila/genética , Metabolismo Energético/fisiologia , Proteínas do Olho/genética , Comportamento Alimentar/fisiologia , Glicoproteínas de Membrana/genética , Receptores de Peptídeos/genética , Tecido Adiposo/metabolismo , Animais , Proteínas de Drosophila/deficiência , Drosophila melanogaster/metabolismo , Ingestão de Alimentos , Glicogênio/metabolismo , Insulina/metabolismo , Mucosa Intestinal/metabolismo , Glicoproteínas de Membrana/deficiência , Microscopia de Fluorescência , Neurônios/metabolismo , Receptores de Peptídeos/deficiência , Transdução de Sinais , Triglicerídeos/metabolismo
8.
Nature ; 501(7465): 63-8, 2013 Sep 05.
Artigo em Inglês | MEDLINE | ID: mdl-23995686

RESUMO

Bacillus anthracis, the causative agent of anthrax disease, is lethal owing to the actions of two exotoxins: anthrax lethal toxin (LT) and oedema toxin (ET). The key tissue targets responsible for the lethal effects of these toxins are unknown. Here we generated cell-type-specific anthrax toxin receptor capillary morphogenesis protein-2 (CMG2)-null mice and cell-type-specific CMG2-expressing mice and challenged them with the toxins. Our results show that lethality induced by LT and ET occurs through damage to distinct cell types; whereas targeting cardiomyocytes and vascular smooth muscle cells is required for LT-induced mortality, ET-induced lethality occurs mainly through its action in hepatocytes. Notably, and in contradiction to what has been previously postulated, targeting of endothelial cells by either toxin does not seem to contribute significantly to lethality. Our findings demonstrate that B. anthracis has evolved to use LT and ET to induce host lethality by coordinately damaging two distinct vital systems.


Assuntos
Antígenos de Bactérias/toxicidade , Bacillus anthracis/patogenicidade , Toxinas Bacterianas/toxicidade , Animais , Antraz/genética , Antraz/metabolismo , Antraz/microbiologia , Resistência à Doença/genética , Edema/induzido quimicamente , Células Endoteliais/efeitos dos fármacos , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/patologia , Feminino , Hepatócitos/efeitos dos fármacos , Hepatócitos/patologia , Mucosa Intestinal/metabolismo , Intestinos/efeitos dos fármacos , Intestinos/patologia , Fígado/citologia , Fígado/efeitos dos fármacos , Fígado/patologia , Masculino , Camundongos , Camundongos Transgênicos , Músculo Liso Vascular/citologia , Músculo Liso Vascular/efeitos dos fármacos , Músculo Liso Vascular/patologia , Miócitos Cardíacos/efeitos dos fármacos , Miócitos Cardíacos/patologia , Especificidade de Órgãos/efeitos dos fármacos , Receptores de Peptídeos/deficiência , Receptores de Peptídeos/genética , Receptores de Peptídeos/metabolismo , Especificidade por Substrato/efeitos dos fármacos , Análise de Sobrevida
9.
J Lipid Res ; 54(1): 85-96, 2013 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-23081987

RESUMO

A high-fat diet (HFD) is a well-known contributing factor in the development of obesity. Most rats fed HFDs become obese. Those that avoid obesity when fed HFDs are considered diet resistant (DR). We performed a microarray screen to identify genes specific to the mesenteric fat of DR rats and revealed high expression of guanylin and guanylyl cyclase C (GC-C) in some subjects. Our histologic studies revealed that the cellular source of guanylin and GC-C is macrophages. Therefore, we developed double-transgenic (Tg) rats overexpressing guanylin and GC-C in macrophages and found that they were resistant to the effects of HFDs. In the mesenteric fat of HFD-fed Tg rats, Fas and perilipin mRNAs were downregulated, and those of genes involved in fatty acid oxidation were upregulated, compared with the levels in HFD-fed wild-type rats. In vitro studies demonstrated that lipid accumulation was markedly inhibited in adipocytes cocultured with macrophages expressing guanylin and GC-C and that this inhibition was reduced after treatment with guanylin- and GC-C-specific siRNAs. Our results suggest that the macrophagic guanylin-GC-C system contributes to the altered expression of genes involved in lipid metabolism, leading to resistance to obesity.


Assuntos
Dieta Hiperlipídica/efeitos adversos , Hormônios Gastrointestinais/metabolismo , Macrófagos/metabolismo , Mesentério/citologia , Peptídeos Natriuréticos/metabolismo , Receptores Acoplados a Guanilato Ciclase/metabolismo , Receptores de Peptídeos/metabolismo , Adipócitos/metabolismo , Animais , Colesterol/sangue , Ácidos Graxos não Esterificados/sangue , Hormônios Gastrointestinais/deficiência , Hormônios Gastrointestinais/genética , Regulação da Expressão Gênica , Técnicas de Introdução de Genes , Insulina/sangue , Fígado/metabolismo , Macrófagos/enzimologia , Macrófagos Peritoneais/enzimologia , Macrófagos Peritoneais/metabolismo , Masculino , Peptídeos Natriuréticos/deficiência , Peptídeos Natriuréticos/genética , Oxirredução , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , RNA Interferente Pequeno/genética , Ratos , Ratos Transgênicos , Receptores de Enterotoxina , Receptores Acoplados a Guanilato Ciclase/deficiência , Receptores Acoplados a Guanilato Ciclase/genética , Receptores de Peptídeos/deficiência , Receptores de Peptídeos/genética , Triglicerídeos/sangue , Triglicerídeos/metabolismo
10.
Brain Res ; 1463: 85-92, 2012 Jun 29.
Artigo em Inglês | MEDLINE | ID: mdl-22575564

RESUMO

The LPHN3 gene has been associated with both attention deficit-hyperactivity disorder (ADHD) and addiction, suggesting that it may play a role in the etiology of these disorders. Unfortunately, almost nothing is known about the normal functions of this gene, which has hampered understanding of its potential pathogenic role. To begin to characterize such normal functions, we utilized a gene-trap embryonic stem cell line to generate mice mutant for the Lphn3 gene. We evaluated differential gene expression in whole mouse brain between mutant and wild type male littermates at postnatal day 0 using TaqMan gene expression assays. Most notably, we found changes in dopamine and serotonin receptors and transporters (Dat1, Drd4, 5Htt, 5Ht2a), changes in neurotransmitter metabolism genes (Th, Gad1), as well as changes in neural developmental genes (Nurr, Ncam). When mice were examined at 4-6 weeks of age, null mutants showed increased levels of dopamine and serotonin in the dorsal striatum. Finally, null mutant mice had a hyperactive phenotype in the open field test, independent of sex, and were more sensitive to the locomotor stimulant effects of cocaine. Considered together, these results suggest that Lphn3 plays a role in development and/or regulation of monoamine signaling. Given the central role for monoamines in ADHD and addiction, it seems likely that the influence of LPHN3 genotype on these disorders is mediated through alterations in monoamine signaling.


Assuntos
Transtorno do Deficit de Atenção com Hiperatividade/genética , Transtorno do Deficit de Atenção com Hiperatividade/metabolismo , Comportamento Aditivo/genética , Comportamento Aditivo/metabolismo , Receptores Acoplados a Proteínas G/deficiência , Receptores Acoplados a Proteínas G/genética , Receptores de Peptídeos/deficiência , Receptores de Peptídeos/genética , Animais , Transtorno do Deficit de Atenção com Hiperatividade/fisiopatologia , Comportamento Aditivo/fisiopatologia , Cocaína/administração & dosagem , Modelos Animais de Doenças , Feminino , Genótipo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Atividade Motora/genética , Mutação/genética , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/genética
11.
Cell Cycle ; 11(12): 2253-9, 2012 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-22617387

RESUMO

Blocking tumor angiogenesis is an important goal of cancer therapy, but clinically approved anti-angiogenic agents suffer from limited efficacy and adverse side effects, fueling the need to identify alternative angiogenesis regulators. Tumor endothelial marker 8 (TEM8) is a highly conserved cell surface receptor overexpressed on human tumor vasculature. Genetic disruption of Tem8 in mice revealed that TEM8 is important for promoting tumor angiogenesis and tumor growth but dispensable for normal development and wound healing. The induction of TEM8 in cultured endothelial cells by nutrient or growth factor deprivation suggests that TEM8 may be part of a survival response pathway that is activated by tumor microenvironmental stress. In preclinical studies, antibodies targeted against the extracellular domain of TEM8 inhibited tumor angiogenesis and blocked the growth of multiple human tumor xenografts. Anti-TEM8 antibodies augmented the activity of other anti-angiogenic agents, vascular targeting agents and conventional chemotherapeutic agents and displayed no detectable toxicity. Thus, anti-TEM8 antibodies provide a promising new tool for selective blockade of neovascularization associated with cancer and possibly other angiogenesis-dependent diseases.


Assuntos
Proteínas de Neoplasias/metabolismo , Neovascularização Patológica , Receptores de Superfície Celular/metabolismo , Animais , Anticorpos/imunologia , Anticorpos/uso terapêutico , Biomarcadores Tumorais/deficiência , Biomarcadores Tumorais/genética , Biomarcadores Tumorais/metabolismo , Células Cultivadas , Células Endoteliais/metabolismo , Células HEK293 , Humanos , Camundongos , Camundongos Knockout , Proteínas dos Microfilamentos , Proteínas de Neoplasias/genética , Neoplasias/tratamento farmacológico , Neoplasias/metabolismo , Neoplasias/patologia , Receptores de Superfície Celular/genética , Receptores de Peptídeos/deficiência , Receptores de Peptídeos/genética , Receptores de Peptídeos/metabolismo , Transfecção , Transplante Heterólogo
12.
Development ; 138(10): 1967-75, 2011 May.
Artigo em Inglês | MEDLINE | ID: mdl-21490063

RESUMO

During male sexual differentiation, the transforming growth factor-ß (TGF-ß) signaling molecule anti-Müllerian hormone (AMH; also known as Müllerian inhibiting substance, MIS) is secreted by the fetal testes and induces regression of the Müllerian ducts, the primordia of the female reproductive tract organs. Currently, the molecular identity of downstream events regulated by the AMH signaling pathway remains unclear. We found that male-specific Wnt4 expression in mouse Müllerian duct mesenchyme depends upon AMH signaling, implicating the WNT pathway as a downstream mediator of Müllerian duct regression. Inactivation of ß-catenin, a mediator of the canonical WNT pathway, did not affect AMH signaling activation in the Müllerian duct mesenchyme, but did block Müllerian duct regression. These data suggest that ß-catenin mediates AMH signaling for Müllerian duct regression during male sexual differentiation.


Assuntos
Ductos Paramesonéfricos/embriologia , Diferenciação Sexual/fisiologia , Testículo/embriologia , beta Catenina/fisiologia , Animais , Hormônio Antimülleriano/fisiologia , Sequência de Bases , Primers do DNA/genética , Feminino , Regulação da Expressão Gênica no Desenvolvimento , Genitália Feminina/embriologia , Genitália Feminina/metabolismo , Masculino , Mesoderma/embriologia , Mesoderma/metabolismo , Camundongos , Camundongos da Linhagem 129 , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Modelos Biológicos , Ductos Paramesonéfricos/metabolismo , Gravidez , Receptores de Peptídeos/deficiência , Receptores de Peptídeos/genética , Receptores de Peptídeos/fisiologia , Receptores de Fatores de Crescimento Transformadores beta/deficiência , Receptores de Fatores de Crescimento Transformadores beta/genética , Receptores de Fatores de Crescimento Transformadores beta/fisiologia , Diferenciação Sexual/genética , Transdução de Sinais , Testículo/metabolismo , Proteínas Wnt/deficiência , Proteínas Wnt/genética , Proteínas Wnt/fisiologia , Proteína Wnt4 , beta Catenina/deficiência , beta Catenina/genética
13.
PLoS One ; 6(1): e16139, 2011 Jan 31.
Artigo em Inglês | MEDLINE | ID: mdl-21305056

RESUMO

BACKGROUND: Guanylyl Cyclase C (GCC) signaling via uroguanylin (UGN) and guanylin activation is a critical mediator of intestinal fluid homeostasis, intestinal cell proliferation/apoptosis, and tumorigenesis. As a mechanism for some of these effects, we hypothesized that GCC signaling mediates regulation of intestinal barrier function. METHODOLOGY/PRINCIPAL FINDINGS: Paracellular permeability of intestinal segments was assessed in wild type (WT) and GCC deficient (GCC-/-) mice with and without lipopolysaccharide (LPS) challenge, as well as in UGN deficient (UGN-/-) mice. IFNγ and myosin light chain kinase (MLCK) levels were determined by real time PCR. Expression of tight junction proteins (TJPs), phosphorylation of myosin II regulatory light chain (MLC), and STAT1 activation were examined in intestinal epithelial cells (IECs) and intestinal mucosa. The permeability of Caco-2 and HT-29 IEC monolayers, grown on Transwell filters was determined in the absence and presence of GCC RNA interference (RNAi). We found that intestinal permeability was increased in GCC-/- and UGN-/- mice compared to WT, accompanied by increased IFNγ levels, MLCK and STAT1 activation in IECs. LPS challenge promotes greater IFNγ and STAT1 activation in IECs of GCC-/- mice compared to WT mice. Claudin-2 and JAM-A expression were reduced in GCC deficient intestine; the level of phosphorylated MLC in IECs was significantly increased in GCC-/- and UGN-/- mice compared to WT. GCC knockdown induced MLC phosphorylation, increased permeability in IEC monolayers under basal conditions, and enhanced TNFα and IFNγ-induced monolayer hyperpermeability. CONCLUSIONS/SIGNIFICANCE: GCC signaling plays a protective role in the integrity of the intestinal mucosal barrier by regulating MLCK activation and TJ disassembly. GCC signaling activation may therefore represent a novel mechanism in maintaining the small bowel barrier in response to injury.


Assuntos
Intestinos/fisiopatologia , Peptídeos/metabolismo , Receptores Acoplados a Guanilato Ciclase/deficiência , Receptores Acoplados a Guanilato Ciclase/metabolismo , Receptores de Peptídeos/deficiência , Receptores de Peptídeos/metabolismo , Transdução de Sinais , Animais , Células Cultivadas , Mucosa Intestinal/metabolismo , Intestinos/enzimologia , Camundongos , Camundongos Knockout , Permeabilidade , Receptores de Enterotoxina , Junções Íntimas
14.
J Neurochem ; 113(6): 1644-58, 2010 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-20374426

RESUMO

Lesioned axons do not regenerate in the adult mammalian CNS, owing to the over-expression of inhibitory molecules such as myelin-derived proteins or chondroitin sulphate proteoglycans. In order to overcome axon inhibition, strategies based on extrinsic and intrinsic treatments have been developed. For myelin-associated inhibition, blockage with NEP1-40, receptor bodies or IN-1 antibodies has been used. In addition, endogenous blockage of cell signalling mechanisms induced by myelin-associated proteins is a potential tool for overcoming axon inhibitory signals. We examined the participation of glycogen synthase kinase 3beta (GSK3beta) and extracellular-related kinase (ERK) 1/2 in axon regeneration failure in lesioned cortical neurons. We also investigated whether pharmacological blockage of GSK3beta and ERK1/2 activities facilitates regeneration after myelin-directed inhibition in two models: (i) cerebellar granule cells and (ii) lesioned entorhino-hippocampal pathway in slice cultures, and whether the regenerative effects are mediated by Nogo Receptor 1 (NgR1). We demonstrate that, in contrast to ERK1/2 inhibition, the pharmacological treatment of GSK3beta inhibition strongly facilitated regrowth of cerebellar granule neurons over myelin independently of NgR1. Finally, these regenerative effects were corroborated in the lesioned entorhino-hippocampal pathway in NgR1-/- mutant mice. These results provide new findings for the development of new assays and strategies to enhance axon regeneration in injured cortical connections.


Assuntos
Córtex Cerebral/citologia , Quinase 3 da Glicogênio Sintase/metabolismo , Neuritos/fisiologia , Neurônios/citologia , Aminofenóis/farmacologia , Animais , Animais Recém-Nascidos , Axotomia/métodos , Células Cultivadas , Técnicas de Cocultura/métodos , Relação Dose-Resposta a Droga , Inibidores Enzimáticos/farmacologia , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Feminino , Proteínas Ligadas por GPI , Regulação da Expressão Gênica/efeitos dos fármacos , Quinase 3 da Glicogênio Sintase/antagonistas & inibidores , Glicogênio Sintase Quinase 3 beta , Hipocampo/citologia , Humanos , Indóis/farmacologia , Lisina/análogos & derivados , Lisina/metabolismo , Maleimidas/farmacologia , Camundongos , Camundongos Knockout , Mutação/genética , Proteínas da Mielina/metabolismo , Proteínas da Mielina/farmacologia , Neuritos/efeitos dos fármacos , Proteínas Nogo , Receptor Nogo 1 , Gravidez , Ratos , Receptores de Superfície Celular , Receptores de Peptídeos/deficiência , Fatores de Tempo , Versicanas/metabolismo
15.
Oncogene ; 29(6): 789-801, 2010 Feb 11.
Artigo em Inglês | MEDLINE | ID: mdl-19901963

RESUMO

The Capillary Morphogenesis Gene 2 (CMG2) gene encodes an Anthrax toxin receptor (ANTXR2), but the normal physiological function is not known. ANTXR2/CMG2 was originally identified as a result of up-regulation during capillary morphogenesis of endothelial cells (ECs) cultured in vitro. We explored the hypothesis that key steps of the angiogenic process are either dependent or are influenced by ANTXR2/CMG2 activity. We describe the expression pattern of ANTXR2/CMG2 in several murine tissues and in normal breast and breast tumors. Endothelial expression was found in all of the tissues analyzed, in cultured ECs and in breast tumor vessels; however, ANTXR2/CMG2 expression was not restricted to this cell type. To assess potential angiogenic function, we used RNA interference to achieve significant reduction of ANTXR2/CMG2 expression in cultured human umbilical venous endothelial cells (HUVECs). Reduced ANTXR2/CMG2 expression resulted in significant inhibition of proliferation and reduced capacity of ECs to form capillary-like networks in vitro, whereas overexpression of ANTXR2/CMG2 in HUVEC increased proliferation and capillary-like network formation. Little change in migration of ECs was observed on knockdown or overexpression. We conclude that ANTXR2/CMG2 functions to promote endothelial proliferation and morphogenesis during sprouting angiogenesis, consistent with the endothelial expression of ANTXR2/CMG2 in several vascular beds.


Assuntos
Células Endoteliais/citologia , Células Endoteliais/patologia , Regulação da Expressão Gênica , Morfogênese , Neoplasias/irrigação sanguínea , Neoplasias/genética , Receptores de Peptídeos/metabolismo , Animais , Mama/irrigação sanguínea , Mama/citologia , Mama/metabolismo , Mama/patologia , Capilares/citologia , Capilares/crescimento & desenvolvimento , Capilares/patologia , Linhagem Celular , Movimento Celular/genética , Proliferação de Células , Células Endoteliais/metabolismo , Endotélio/crescimento & desenvolvimento , Técnicas de Silenciamento de Genes , Humanos , Imuno-Histoquímica , Camundongos , Camundongos Endogâmicos C57BL , Morfogênese/genética , Neoplasias/metabolismo , Neovascularização Fisiológica/genética , Receptores de Peptídeos/deficiência , Receptores de Peptídeos/genética
16.
Cancer Res ; 69(15): 6021-6, 2009 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-19622764

RESUMO

Tumor endothelial marker 8 (TEM8) was initially identified as a gene overexpressed in the vasculature of human tumors and was subsequently identified as an anthrax toxin receptor. To assess the functional role of TEM8, we disrupted the TEM8 gene in mice by targeted homologous recombination. TEM8(-/-) mice were viable and reached adulthood without defects in physiologic angiogenesis. However, histopathologic analysis revealed an excess of extracellular matrix in several tissues, including the ovaries, uterus, skin, and periodontal ligament of the incisors, the latter resulting in dental dysplasia. When challenged with B16 melanoma, tumor growth was delayed in TEM8(-/-) mice, whereas the growth of other tumors, such as Lewis lung carcinoma, was unaltered. These studies show that host-derived TEM8 promotes the growth of certain tumors and suggest that TEM8 antagonists may have utility in the development of new anticancer therapies.


Assuntos
Melanoma Experimental/patologia , Receptores de Peptídeos/fisiologia , Animais , Biomarcadores Tumorais , Carcinoma Pulmonar de Lewis/irrigação sanguínea , Carcinoma Pulmonar de Lewis/metabolismo , Carcinoma Pulmonar de Lewis/patologia , Feminino , Masculino , Melanoma Experimental/irrigação sanguínea , Melanoma Experimental/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteínas dos Microfilamentos , Neovascularização Patológica/patologia , Receptores de Superfície Celular , Receptores de Peptídeos/biossíntese , Receptores de Peptídeos/deficiência , Receptores de Peptídeos/genética
17.
Proc Natl Acad Sci U S A ; 106(17): 7203-8, 2009 Apr 28.
Artigo em Inglês | MEDLINE | ID: mdl-19359476

RESUMO

Many behavioral traits and most brain disorders are common to males and females but are more evident in one sex than the other. The control of these subtle sex-linked biases is largely unstudied and has been presumed to mirror that of the highly dimorphic reproductive nuclei. Sexual dimorphism in the reproductive tract is a product of Müllerian inhibiting substance (MIS), as well as the sex steroids. Males with a genetic deficiency in MIS signaling are sexually males, leading to the presumption that MIS is not a neural regulator. We challenge this presumption by reporting that most immature neurons in mice express the MIS-specific receptor (MISRII) and that male Mis(-/-) and Misrii(-/-) mice exhibit subtle feminization of their spinal motor neurons and of their exploratory behavior. Consequently, MIS may be a broad regulator of the subtle sex-linked biases in the nervous system.


Assuntos
Hormônio Antimülleriano/metabolismo , Comportamento Animal/fisiologia , Encéfalo/metabolismo , Caracteres Sexuais , Androgênios/metabolismo , Animais , Hormônio Antimülleriano/deficiência , Hormônio Antimülleriano/genética , Encéfalo/embriologia , Proliferação de Células , Feminino , Regulação da Expressão Gênica no Desenvolvimento , Masculino , Camundongos , Camundongos Knockout , Neurônios Motores/citologia , Neurônios Motores/metabolismo , Receptores de Peptídeos/deficiência , Receptores de Peptídeos/genética , Receptores de Peptídeos/metabolismo , Receptores de Fatores de Crescimento Transformadores beta/deficiência , Receptores de Fatores de Crescimento Transformadores beta/genética , Receptores de Fatores de Crescimento Transformadores beta/metabolismo , Coluna Vertebral/citologia , Coluna Vertebral/metabolismo
18.
Reprod Fertil Dev ; 21(3): 440-50, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-19261221

RESUMO

The peptide hormone relaxin is expressed in the prostate gland and secreted into the seminal plasma; however, its function within the prostate has not been established. Relaxin-mutant mice (Rln(-/-)) were reported to have abnormal prostate morphology, but there was no prostate phenotype in relaxin receptor-mutant (Rxfp1(-/-)) mice. The present study aimed to verify the phenotypes in the anterior, dorsal and lateral lobes of the prostate gland of Rln(-/-) and Rxfp1(-/-) mice at different adult ages. Rln(-/-) mice were also treated with relaxin to evaluate the effects of exogenously administered hormone on prostate morphology. Comparisons between these three lobes of the prostate demonstrated no obvious differences in duct morphology, epithelial height or collagen density between Rln(+/+) and Rln(-/-) mice at 2, 4, 6, 8 and 12 months of age. This was similar in Rxfp1(-/-) mice. Relaxin treatment did not affect morphology or epithelial cell height in the different lobes. Furthermore, prostate lobe morphology in transgenic mice overexpressing relaxin Tg(Rln) was not different from the wild-type controls. Rxfp1 was detected in the prostate throughout adult life, but there was no consistent expression of relaxin. In summary, the present study found no evidence to support a prostate phenotype in adult Rln- or Rxfp1-mutant mice.


Assuntos
Próstata/anatomia & histologia , Relaxina/deficiência , Envelhecimento , Animais , Células Epiteliais/citologia , Expressão Gênica , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Mutação , Próstata/química , RNA Mensageiro/análise , Receptores Acoplados a Proteínas G/deficiência , Receptores Acoplados a Proteínas G/genética , Receptores de Peptídeos/deficiência , Receptores de Peptídeos/genética , Relaxina/genética , Relaxina/farmacologia
19.
Eur J Neurosci ; 26(12): 3339-44, 2007 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-18052978

RESUMO

New neurons are added on a daily basis to the olfactory bulb (OB) of a mammal, and this phenomenon exists throughout its lifetime. These new cells are born in the subventricular zone and migrate to the OB via the rostral migratory stream (RMS). To examine the role of the prokineticin receptor 2 (Prokr2) in neurogenesis, we created a Prokr2 null mouse, and report a decrease in the volume of its OB and also a decrease in the number of bromodeoxyuridine (BrdU)-positive cells. There is disrupted architecture of the OB, with the glomerular layer containing terminal dUTP nick-end labeling (TUNEL) -positive nuclei and also a decrease in tyrosine hydroxylase-positive neurons in this layer. In addition, there are increased numbers of doublecortin-positive neuroblasts in the RMS and increased PSA-NCAM (polysialylated form of the neural cell adhesion molecule) -positive neuronal progenitors around the olfactory ventricle, indicating their detachment from homotypic chains is compromised. Finally, in support of this, Prokr2-deficient cells expanded in vitro as neurospheres are incapable of migrating towards a source of recombinant human prokineticin 2 (PROK2). Together, these findings suggest an important role for Prokr2 in OB neurogenesis.


Assuntos
Diferenciação Celular , Movimento Celular , Neurônios/patologia , Bulbo Olfatório/anormalidades , Bulbo Olfatório/patologia , Receptores Acoplados a Proteínas G/deficiência , Receptores de Peptídeos/deficiência , Células-Tronco/patologia , Animais , Animais Recém-Nascidos , Bromodesoxiuridina , Contagem de Células , Movimento Celular/efeitos dos fármacos , Proteínas do Domínio Duplacortina , Feminino , Hormônios Gastrointestinais/farmacologia , Homozigoto , Humanos , Lactação , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteínas Associadas aos Microtúbulos/metabolismo , Molécula L1 de Adesão de Célula Nervosa/metabolismo , Neurônios/metabolismo , Neuropeptídeos/metabolismo , Neuropeptídeos/farmacologia , Bulbo Olfatório/metabolismo , Bulbo Olfatório/fisiopatologia , Receptores Acoplados a Proteínas G/genética , Receptores de Peptídeos/genética , Proteínas Recombinantes/farmacologia , Ácidos Siálicos/metabolismo , Esferoides Celulares , Células-Tronco/metabolismo , Tirosina 3-Mono-Oxigenase/metabolismo
20.
Gastroenterology ; 133(2): 599-607, 2007 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-17681179

RESUMO

BACKGROUND AND AIMS: The most commonly lost gene products in colorectal carcinogenesis include guanylin and uroguanylin, endogenous ligands for guanylyl cyclase C (GCC). Beyond intestinal fluid balance, GCC mediates diarrhea induced by bacterial enterotoxins, and an inverse relationship exists between enterotoxigenic Escherichia coli infections producing the exogenous GCC ligand ST and colorectal cancer worldwide. However, the role of GCC in neoplasia remains obscure. METHODS: Intestinal tumorigenesis was examined in wild-type (Gcc(+/+)) and GCC-deficient (Gcc(-/-)) mice carrying mutations in Apc (Apc(Min/+)) or exposed to the carcinogen azoxymethane. Markers of DNA damage, loss of Apc heterozygosity, and beta-catenin mutations were used to assess genomic integrity. Hyperproliferation was explored using Ki67 and cell cycle markers. Apoptosis was quantified by transferase biotin-dUTP nick end labeling analysis. RESULTS: In colons of Apc(Min/+) mice, deletion of Gcc increased tumor incidence and multiplicity, reflecting uncoupling of loss of genomic integrity and compensatory apoptosis. Conversely, in the small intestine, elimination of Gcc increased tumorigenesis by enhancing proliferation without altering genomic integrity. Moreover, these distinct but mutually reinforcing mechanisms collaborate in azoxymethane-exposed mice, and deletion of Gcc increased tumor initiation and growth associated with hypermutation and hyperproliferation, respectively, in conjunction with attenuated apoptosis. CONCLUSIONS: GCC suppresses tumor initiation and growth by maintaining genomic integrity and restricting proliferation. This previously unrecognized role of GCC in inhibiting tumorigenesis, together with the invariant disruption in guanylin and uroguanylin expression early in carcinogenesis, and the uniform over-expression of GCC by tumors, underscores the potential of oral administration of GCC ligands for targeted prevention and therapy of colorectal cancer.


Assuntos
Proliferação de Células , Transformação Celular Neoplásica/metabolismo , Neoplasias do Colo/enzimologia , Regulação Neoplásica da Expressão Gênica , Genes APC , Guanilato Ciclase/metabolismo , Neoplasias Intestinais/enzimologia , Intestino Delgado/enzimologia , Receptores de Peptídeos/metabolismo , Animais , Apoptose , Azoximetano , Proteínas de Ciclo Celular/análise , Transformação Celular Neoplásica/genética , Transformação Celular Neoplásica/patologia , Neoplasias do Colo/induzido quimicamente , Neoplasias do Colo/genética , Neoplasias do Colo/patologia , Dano ao DNA , Modelos Animais de Doenças , Guanilato Ciclase/deficiência , Guanilato Ciclase/genética , Neoplasias Intestinais/induzido quimicamente , Neoplasias Intestinais/genética , Neoplasias Intestinais/patologia , Intestino Delgado/patologia , Antígeno Ki-67/análise , Perda de Heterozigosidade , Camundongos , Camundongos Knockout , Mutação , Receptores de Enterotoxina , Receptores Acoplados a Guanilato Ciclase , Receptores de Peptídeos/deficiência , Receptores de Peptídeos/genética , beta Catenina/genética , beta Catenina/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...