962 resultados para Cell Signaling
Resumo:
ABSTRACT: BACKGROUND: Many studies have been published outlining the global effects of 17 beta-estradiol (E2) on gene expression in human epithelial breast cancer derived MCF-7 cells. These studies show large variation in results, reporting between ~100 and ~1500 genes regulated by E2, with poor overlap. RESULTS: We performed a meta-analysis of these expression studies, using the Rank product method to obtain a more accurate and stable list of the differentially expressed genes, and of pathways regulated by E2. We analyzed 9 time-series data sets, concentrating on response at 3-4 hrs (early) and at 24 hrs (late). We found >1000 statistically significant probe sets after correction for multiple testing at 3-4 hrs, and >2000 significant probe sets at 24 hrs. Differentially expressed genes were examined by pathway analysis. This revealed 15 early response pathways, mostly related to cell signaling and proliferation, and 20 late response pathways, mostly related to breast cancer, cell division, DNA repair and recombination. CONCLUSIONS: Our results show that meta-analysis identified more differentially expressed genes than the individual studies, and that these genes act together in networks. These results provide new insight into E2 regulated mechanisms, especially in the context of breast cancer.
Resumo:
Antigenic recognition by naive CD4+ T cells induces their proliferation and differentiation into functionally distinct T helper (Th) cell. Each CD4+ Th cell subset expresses specific transcription factors and produces signature cytokines that coordinate immune responses against encountered pathogens. Among the factors influencing CD4+ Th cell differentiation, Notch signaling pathway has been reported to play a role in the differentiation and function of multiple CD4+Thcell subsets. Notch signaling is an evolutionarily conserved cell-to-cell signaling cascade involved in many cell fate decision processes. How Notch signaling modulates the differentiation of CD4+ Th cell subsets and whether Notch signaling alone is sufficient or not for the differentiation of CD4+ Th cells is still a matter of debate. Th17 cells are a distinct subset of CD4+ Th cells. They play a role in the control of extracellular bacterial and fungal infections and may lead to inflammatory and autoimmune diseases if not properly regulated. Th17 cells are defined by the expression of RAR-related orphan receptor (ROR)a and RORyT transcription factors and their secretion of IL-17A, IL-17F cytokines. The involvement of Notch signaling in Th17 cell differentiation has mostly been studied in vitro. However, neither the experimental conditions when Notch signaling might be involved in Th17 cell differentiation in vitro and in vivo nor the precise role of Notch in this process remain clear. To better define how Notch signaling impacts Th17 differentiation, we used mice with T cell specific ablation of Notchl and Notch2 (N1 N2ACD4Cre) or of Notch transcriptional repressor RBP- JK (RBP-J ACD4Cre). We show that impaired Notch signaling in T cells, when TCR activating signal were reduced, increased RORyT and IL-17 mRNA levels during in vitro Th17 cell differentiation. Following immunization with OVA in CFA, an adjuvant that induces mostly Th17 cell response, increased IL-17A mRNA and intracellular IL-17A levels were observed in draining lymph nodes of Notch-deficient CD4+T cells. Our data suggest that Notch limited Th17 cell differentiation. Despite high levels of IL-17 mRNA and intracellular IL-17 proteins observed in Notch-deficient T cells, their release of Th17 cytokines ex vivo was markedly decreased, indicating a role for Notch signaling. During the second part of this thesis, we observed that the impact of Notch on Th17 cell differentiation and effector functions was context-dependent using different in vivo experimental models, in which Th17 cells and IL-17A were reported to contribute in the disease development. Collectively, our data reveal that Notch signaling controls the fine-tuning of Th17 cell differentiation and effector functions by limiting their differentiation but promoting selectively cytokine release through Notch-dependent mechanisms that still need to be defined. -- Lors d'une réponse immunitaire et grâce à la reconnaissance antigénique, les lymphocytes CD4+ T naïfs prolifèrent, puis se différencient en CD4+ T auxiliaires ("T helper" ou Th) fonctionnellement distincts. Chaque sous-population de lymphocytes CD4+ T auxiliaires exprime des facteurs de transcription et des cytokines spécifiques qui coordonnent la réponse immunitaire contre les pathogènes rencontrés. Parmi les facteurs influençant la différenciation des lymphocytes CD4+ T auxiliaires, la voie de signalisation Notch a été identifiée comme ayant un rôle dans la différenciation et la fonction des différents sous-types de cellules CD4+ T auxiliaires. La voie de signalisation Notch est une voie évolutivement conservée, qui est impliquée dans la signalisation entre les cellules et dans de nombreux processus de décisions cellulaires. La manière dont la voie de signalisation Notch régule la différenciation des lymphocytes CD4+ T en sous-types de cellules CD4+ auxiliaires, mais également la question de savoir si la voie de signalisation Notch est capable ou non d'induire la différenciation des cellules CD4+T auxiliaires, restent à débattre. Les cellules T auxiliaires 17 (Th17) sont un sous-type distinct de cellules CD4+T. Elles jouent un rôle important dans la défense immunitaire contre des pathogènes tels que les bactéries extracellulaires et les champignons. Une dérégulation de la réponse des cellules Th17 peut conduire à des inflammations mais également à des maladies auto-immunes. Les cellules Th17 sont définies par l'expression de leurs facteurs de transcription RAR-related orphan receptor (ROR)a, RORyT et par la sécrétion de cytokines comme IL-17A, IL-17F. Le rôle de la voie de signalisation Notch dans la différenciation des cellules Th17 a principalement été démontré in vitro. Malgré tout, ni les conditions expérimentales dans lesquelles cette voie pourrait être impliquée dans la différenciation des cellules Th17 in vitro et in vivo, mais également ni la fonction exacte de Notch dans ces processus, ne sont des questions résolues. Afin de mieux définir comment la voie de signalisation Notch est impliquée dans la différenciation des cellules Th17, nous avons utilisé des souris avec une déficience spécifique dans les cellules T des récepteurs Notchl et Notch2 (N1N2ACD4Cre) ou du répresseur transcriptionnel de Notch RBP-JK (RBP-J ACD4Cre). Nous avons montré que lorsque la voie de signalisation Notch est déficiente, les niveaux d'ARN messager (ARNm) de RORyT et de IL-17A sont augmentés dans les cellules Th17 pendant la différenciation in vitro, en présence de niveaux réduits des signaux activant les cellules T CD4+. Une augmentation dans les niveaux d'ARNm de IL-17A et de IL-17A intracellulaire au niveau protéinique a été observée dans les cellules T CD4+ Notch déficientes, au niveau des ganglions drainants après immunisation avec l'OVA dans le CFA, un adjuvant induisant une réponse des cellules Th17. Nos résultats suggèrent que Notch pourrait réguler négativement l'expression de IL-17A au niveau transcriptionnel mais également protéinique. Malgré une augmentation de IL-17A au niveau de l'ARNm et protéinique dans les cellules CD4+ T Notch déficientes, paradoxalement la sécrétion de IL-17A mais également de cytokines associées aux fonctions effectrices des cellules Th17 sont profondément diminuées 6X vivo, suggérant un rôle de la voie de signalisation Notch dans ce processus. Dans la deuxième partie de ce travail de thèse, nous avons observé que l'impact de Notch dans la différenciation des cellules Th17 et dans leurs fonctions effectrices était dépendant du contexte dans d'autres modèles expérimentaux in vivo, où les cellules Th17 et l'IL-17A ont été identifiées comme ar-.riCociêSM dans le développement ds la pathologie. En résumé, nous avons montré que la voie de la signalisation Notch contrôle la régulation précise de la différenciation des cellules Th17 en limitant leur différenciation, mais en promouvant sélectivement leur relâchement en cytokines associés aux cellules Th17 par l'intermédiaire de mécanismes dépendant de Notch, qui restent toujours à déterminer. -- Lors d'une réponse immunitaire et grâce à la reconnaissance antigénique, les lymphocytes CD4+ T naïfs prolifèrent, puis se différencient en CD4+ T auxiliaires ("T helper" ou Th) fonctionnellement distincts. Chaque sous-population de lymphocytes T auxiliaires exprime des facteurs de transcription et des cytokines spécifiques qui coordonnent une réponse immunitaire contre différents pathogènes. Les mécanismes liés à la différenciation des lymphocytes CD4+ T auxiliaires sont complexes et régulés. Une mauvaise régulation de la différenciation des lymphocytes CD4+ T auxiliaires peut conduire à des maladies auto-immunes, mais également à des processus inflammatoires. Parmi les facteurs influençant la différenciation des lymphocytes T auxiliaires, la voie de signalisation Notch a été identifiée comme ayant un rôle dans la différenciation et la fonction des différents sous-types de cellules CD4+ T auxiliaires. La voie de signalisation Notch est une voie évolutivement conservée, qui est impliquée dans la signalisation entre les cellules, mais également dans de nombreux processus de décisions cellulaires. Quelle est l'implication de la voie de signalisation Notch dans la différenciation des lymphocytes CD4+ en sous-types de cellules CD4+T auxiliaires et comment cette voie agit dans ce processus, sont des questions débattues. Les cellules T auxiliaires 17 (Th17) sont une sous-population distincte de lymphocytes CD4+. Elles jouent un rôle important dans la défense immunitaire contre les bactéries extracellulaires et les champignons. Une dérégulation de la réponse des cellules Th17 a été associée à des maladies auto-immunes et à l'inflammation. Les cellules Th17 sont définies par l'expression du facteur de transcription RAR-related orphan receptor (ROR)yT et des cytokines comme IL-17A, IL-17F. Le rôle de la voie de signalisation Notch dans la différenciation des cellules Th17 a été principalement démontré dans des études expérimentales in vitro. Malgré tout, les conditions expérimentales exactes dans lesquelles la voie de signalisation de Notch pourrait être impliquée dans la différenciation des cellules Th17, mais également le rôle de Notch dans ce processus ne sont pas encore clairement élucidés. Afin de mieux définir comment la voie de signalisation Notch est impliquée dans la différenciation des cellules Th17, nous avons utilisé des souris avec une déficience spécifique dans les cellules T des récepteurs Notchl et Notch2 (N1 N2ACD4Cre) ou du répresseur transcriptionnel de Notch RBP-JK (RBP-JACD4CRE). Nous avons montré que lorsque la voie de signalisation Notch est déficiente, les niveaux d'ARN messager (ARNm) de RORyT et de IL-17 sont augmentés dans les cellules Th17 pendant leur différenciation in vitro. Cet effet de Notch sur la transcription apparaît être facultatif lorsque les conditions environnementales sont en excès in vitro. Après immunisation avec un adjuvant qui induit principalement une réponse des cellules Th17, nous avons observé que les niveaux de ARNm de IL-17A et aussi de IL-17A intracellulaire au niveau protéinique étaient augmentés dans les ganglions drainants dans les cellules CD4+ Notch déficientes. Ces résultats suggèrent que Notch pourrait réguler négativement l'expression de IL- 17 au niveau transcriptionnel mais également protéinique. Malgré des niveaux plus élevés de IL- 17 ARNm et aussi IL-17A intracellulaire dans les cellules T Notch déficientes, le relâchement en cytokines Th17 est profondément diminué indiquant un rôle de la voie de signalisation Notch dans ces processus de sécrétion. Dans la deuxième partie de cette thèse, nous avons observé que le rôle de Notch dans ia différenciation dss cellules Ti,17 et dans leurs fonctions effectrices était dépendant du contexte dans d'autres modèles expérimentaux, qui ont été rapportés comme une réponse induisant des cellules Th17. En résumé, nos données montrent que la voie de la signalisation Notch contrôle la régulation précise de la différenciation des cellules Th17 en limitant leur différenciation mais en promouvant sélectivement le relâchement en cytokines associées aux cellules Th17 par des mécanismes dépendant de Notch qui restent toujours à déterminer. Par conséquent, l'inhibition de la voie de signalisation Notch pourrait être utilisée dans des situations inflammatoires ou d'auto-immunité où la réponse des cellules Th17 est exacerbée.
Resumo:
Gomesin is an antimicrobial peptide isolated from hemocytes of a common Brazilian tarantula spider named Acanthoscurriagomesiana. This peptide exerts antitumor activity in vitro and in vivo by an unknown mechanism. In this study, the cytotoxic mechanism of gomesin in human neuroblastoma SH-SY5Y and rat pheochromocytoma PC12 cells was investigated. Gomesin induced necrotic cell death and was cytotoxic to SH-SY5Y and PC12 cells. The peptide evoked a rapid and transient elevation of intracellular calcium levels in Fluo-4-AM loaded PC12 cells, which was inhibited by nimodipine, an L-type calcium channel blocker. Preincubation with nimodipine also inhibited cell death induced by gomesin in SH-SY5Y and PC12 cells. Gomesin-induced cell death was prevented by the pretreatment with MAPK/ERK, PKC or PI3K inhibitors, but not with PKA inhibitor. In addition, gomesin generated reactive oxygen species (ROS) in SH-SY5Y cells, which were blocked with nimodipine and MAPK/ERK, PKC or PI3K inhibitors. Taken together, these results suggest that gomesin could be a useful anticancer agent, which mechanism of cytotoxicity implicates calcium entry through L-type calcium channels, activation of MAPK/ERK, PKC and PI3K signaling as well as the generation of reactive oxygen species. (C) 2010 Elsevier Ireland Ltd. All rights reserved.
Resumo:
Small changes in the complex between a peptide and a molecule of the major histocompatibility complex generate ligands able to partially activate (partial agonist) or even inhibit (antagonist) T-cell functions. T-cell receptor engagement of antagonist complex results in a partial zeta chain phosphorylation without activation of the associated ZAP-70 kinase. Herein we show that, despite a strong inhibition of both inositol phospholipid hydrolysis and extracellular increasing antagonist concentrations increased the activity of the CD4-Lck kinase. Addition of anti-CD4 antibody to culture medium prevented inhibitory effects induced by antagonist ligand. We propose that CD4-Lck activation triggered by antagonist complexes may act in a dominant negative mode, thus overriding stimulatory signals coming from agonist ligand. These findings identify a new T-cell signaling profile that may explain the ability of some T-cell receptor variant ligands to inhibit specific biological activities or trigger alternative activation programs.
Resumo:
Antigen-specific activation of T lymphocytes, via stimulation of the T-cell antigen receptor (TCR) complex, is marked by a rapid and sustained increase in the concentration of cytoplasmic free Ca2+ ([Ca2+]i). It has been suggested that the second messenger inositol 1,4,5-trisphosphate (IP3) produced after TCR stimulation binds to the IP3 receptor (IP3R), an intracellular Ca(2+)-release channel, and triggers the increase in [Ca2+]i that activates transcription of the gene for T-cell growth factor interleukin 2 (IL-2). However, the role of the IP3R in T-cell signaling and possibly in plasma membrane Ca2+ influx in T cells remains unproven. Stable transfection of T cells (Jurkat) with antisense type 1 IP3R cDNA prevented type 1 IP3R expression, providing a tool for dissecting the role of IP3 signaling during T-cell activation. T cells lacking type 1 IP3R failed to increase [Ca2+]i or produce IL-2 after TCR stimulation. Moreover, depletion of intracellular Ca2+ stores without TCR activation stimulated Ca2+ influx in cells lacking the type 1 IP3R. These results establish that the type 1 IP3R is required for intracellular Ca2+ release that triggers antigen-specific T-cell proliferation but not for plasma membrane Ca2+ influx.
Resumo:
The efficacy of antioxidant supplementation in the prevention of cardiovascular disease appears equivocal, however the use of more potent antioxidant combinations than those traditionally used may exert a more positive effect. We have shown previously that supplementation of vitamin E and α-lipoic acid increases cardiac performance during post-ischemia reperfusion in older rats and increases Bcl-2 levels in endothelial cells. The purpose of this study was to examine the effects of vitamin E and α-lipoic acid supplementation on myocardial gene expression with a view to determine their mechanism of action. Young male rats received either a control (n=7) or vitamin E and α-lipoic acid supplemented diet (n=8) for 14 weeks. RNA from myocardial tissue was then amplified and samples were pooled within groups and competitively hybridized to 5K oligonucleotide rat microarrays. The relative expression of each gene was then compared to the control sample. Animals that received the antioxidant-supplemented diet exhibited upregulation (>1.5×) of 13 genes in the myocardium with 2 genes downregulated.� �Upregulated genes include those involved in cell growth and maintenance (LynB, Csf1r, Akt2, Tp53), cell signaling (LynB, Csf1r) and signal transduction (Pacsin2, Csf1r). Downregulated genes encode thyroid (Thrsp) and F-actin binding proteins (Nexilin).
Resumo:
Placental tissue injury is concomitant with tumor development. We investigated tumor-driven placental damage by tracing certain steps of the protein synthesis and degradation pathways under leucine-rich diet supplementation in MAC16 tumor-bearing mice. Cell signaling and ubiquitin-proteasome pathways were assessed in the placental tissues of pregnant mice, which were distributed into three groups on a control diet (pregnant control, tumor-bearing pregnant, and pregnant injected with MAC-ascitic fluid) and three other groups on a leucine-rich diet (pregnant, tumor-bearing pregnant, and pregnant injected with MAC-ascitic fluid). MAC tumor growth down-regulated the cell-signaling pathways of the placental tissue and decreased the levels of IRS-1, Akt/PKB, Erk/MAPK, mTOR, p70S6K, STAT3, and STAT6 phosphorylated proteins, as assessed by the multiplex Millipore Luminex assay. Leucine supplementation maintained the levels of these proteins within the established cell-signaling pathways. In the tumor-bearing group (MAC) only, the placental tissue showed increased PC5 mRNA expression, as assessed by quantitative RT-PCR, decreased 19S and 20S protein expression, as assessed by Western blot analysis, and decreased placental tyrosine levels, likely reflecting up-regulation of the ubiquitin-proteasome pathway. Similar effects were found in the pregnant injected with MAC-ascitic fluid group, confirming that the effects of the tumor were mimicked by MAC-ascitic fluid injection. Although tumor progression occurred, the degradation pathway-related protein levels were modulated under leucine-supplementation conditions. In conclusion, tumor evolution reduced the protein expression of the cell-signaling pathway associated with elevated protein degradation, thereby jeopardizing placental activity. Under the leucine-rich diet, the impact of cancer on placental function could be minimized by improving the cell-signaling activity and reducing the proteolytic process.
Resumo:
The cell signaling cascades that mediate pigment movements in crustacean chromatophores are not yet well established, although Ca(2+) and cyclic nucleotide second messengers are involved. Here, we examine the participation of cyclic guanosine monophosphate (cGMP) in pigment aggregation triggered by red pigment concentrating hormone (RPCH) in the red ovarian chromatophores of freshwater shrimp. In Ca(2+)-containing (5.5 mmol l(-1)) saline, 10 mu mol l(-1) dibutyryl cGMP alone produced complete pigment aggregation with the same time course (approximate to 20 min) and peak velocity (approximate to 17 mu m/min) as 10(-8) mol l(-1) RPCH; however, in Ca(2+)-free saline (9 X 10(-11) mol l(-1) Ca(2+)), db-cGMP was without effect. The soluble guanylyl cyclase (GC-S) activators sodium nitroprusside (SNP, 0.5 mu mol l(-1)) and 3-morpholinosydnonimine (SIN-1, 100 mu mol l(-1)) induced moderate aggregation by themselves (approximate to 35%-40%) but did not affect RPCH-triggered aggregation. The GC-S inhibitors zinc protoporphyrin IX (ZnPP-XI, 30 mu mol l(-1)) and 6-anilino-5,8-quinolinedione (LY83583, 10 mu mol l(-1)) partially inhibited RPCH-triggered aggregation by approximate to 35%. Escherichia coli heat-stable enterotoxin (STa, 1 mu mol l(-1)), a membrane-receptor guanylyl cyclase stimulator, did not induce or affect RPCH-triggered aggregation. We propose that the binding of RPCH to an unknown membrane-receptor type activates a Ca(2+)-dependent signaling cascade coupled via cytosolic guanylyl cyclase and cGMP to protein kinase G-phosphorylated proteins that regulate aggregation-associated, cytoskeletal molecular motor activity. This is a further example of a cGMP signaling cascade mediating the effect of a crustacean X-organ neurosecretory peptide.
Resumo:
Neutrophilic granulocytes play a major role in the initiation and resolution of the inflammatory response, and demonstrate significant transcriptional and translational activity. Although much was known about neutrophils prior to the introduction of proteomics, the use of MS-based methodologies has provided an unprecedented tool to confirm and extend previous findings. In the present study, we performed a Gel-LC-MS/MS analysis of neutrophil detergent insoluble and whole cell lysate fractions of resting neutrophils. We achieved a set of identifications through the use of high-resolution mass spectrometry and validation of its data. We identified a total of 1249 proteins with a wide range of intensities from both detergent-insoluble and whole cell lysate fractions, allowing a mapping of proteins such as those involved in intracellular transport (Rab and Sec family proteins) and cell signaling (S100 proteins). These results represent the most comprehensive proteomic characterization of resting human neutrophils to date, and provide important information relevant for further studies of the immune system in health and disease. The methods applied here can be employed to help us understand how neutrophils respond to various physiologic and pathophysiologic conditions and could be extended to protein quantitation after cell activation.
Resumo:
Histoplasma capsulatum (Hc) is a facultative, intracellular parasite of worldwide significance. Infection with Hc produces a broad spectrum of diseases and may progress to a life-threatening systemic disease, particularly in individuals with HIV infection. Resolution of histoplasmosis is associated with the activation of cell-mediated immunity, and leukotriene B(4) plays an important role in this event. Lipid bodies (LBs) are increasingly being recognized as multifunctional organelles with roles in inflammation and infection. In this study, we investigated LB formation in histoplasmosis and its putative function in innate immunity. LB formation in leukocytes harvested from Hc-infected C57BL/6 mice peaks on day 2 postinfection and correlates with enhanced generation of lipid mediators, including leukotriene B(4) and PGE(2). Pretreatment of leukocytes with platelet-activating factor and BLT1 receptor antagonists showed that both lipid mediators are involved in cell signaling for LB formation. Alveolar leukocytes cultured with live or dead Hc also presented an increase in LB numbers. The yeast alkali-insoluble fraction 1, which contains mainly beta-glucan isolated from the Hc cell wall, induced a dose- and time-dependent increase in LB numbers, indicating that beta-glucan plays a signaling role in LB formation. In agreement with this hypothesis, beta-glucan-elicited LB formation was inhibited in leukocytes from 5-LO(-/-), CD18(low) and TLR2(-/-) mice, as well as in leukocytes pretreated with anti-Dectin-1 Ab. Interestingly, human monocytes from HIV-1-infected patients failed to produce LBs after beta-glucan stimulation. These results demonstrate that Hc induces LB formation, an event correlated with eicosanoid production, and suggest a role for these lipid-enriched organelles in host defense during fungal infection. The Journal of Immunology, 2009, 182: 4025-4035.
Resumo:
RESUMO: As células eucarióticas evoluíram um sistema de sinalização complexo que lhes permite responder aos sinais extracelulares e intracelulares. Desta forma, as vias de sinalização são essenciais para a sobrevivência da célula e do organismo, uma vez que regulam processos fundamentais, tais como o desenvolvimento, o crescimento, a imunidade, e a homeostase dos tecidos. A via de transdução de sinal Hedgehog (Hh) envolve o receptor Patched1 (Ptch1), que tem um efeito inibidor sobre a proteína Smoothened (Smo) na ausência dos seus ligandos, as proteínas Sonic hedgehog (Shh). Estas proteínas são reguladores fundamentais do desenvolvimento embrionário, como ilustrado pelas malformações drásticas observadas em embriões humanos e de murganho com perturbações da transdução de sinal da via Hh e que incluem polidactilia, defeitos craniofaciais e malformações ósseas. Igualmente importantes são as consequências da ativação inapropriada da via de sinalização Hh na formação de tumores. Curiosamente, os componentes desta via localizam-se nos cílios primários. Além disso, demonstrou-se que esta localização é crucial para a sinalização através da via Hh. Na presença dos ligandos, Ptch1 é internalizado e destinado a degradação ou sequestrado num compartimento da célula de onde não pode desempenhar o seu papel inibitório. A proteína Arl13b é uma pequena GTPase pertencente à família Arf/Arl da superfamília Ras de pequenas GTPases e foi implicada no síndrome de Joubert, uma ciliopatia caracterizada por ataxia congénita cerebelar, hipotonia, atrso mental e cardiopatia congénita. Murganhos deficientes para Arl13b, chamado hennin (hnn) morrem morrem prematuramente ao dia 13,5 de gestação (E13,5) e exibem anomalias morfológicas nos cílios que levam à interrupção da sinalização Hh. Além disso, a Arl13b está diretamente envolvida na regulação da via Hh, controlando a localização de vários componentes desta via nos cílios primários. Neste trabalho, mostramos que a Arl13b se localiza em circular dorsal ruffles (CDRs), que são estruturas de actina envolvidas em macropinocitose e internalização de recetores, e que regula a sua formação. Além disso, aprofundámos o conhecimento do processo de ativação da via de sinalização Hh, mostrando que as CDRs sequestram seletivamente e internalizam o recetor Ptch1. As CDRs formam-se minutos após ativação da via por ligandos Shh ou pelo agonista de Smo SAG e continuam a ser formadas a partir daí, sugerindo uma indução contínua da reorganização do citoesqueleto de actina quando a via está ativada. Observámos ainda que a inibição da formação de CDRs através do silenciamento de WAVE1, uma proteína necessária para a formação destas estruturas, resulta na diminuição da ativação da via de sinalização Hh. Além disso, o bloqueio da macropinocitose, que se segue ao fecho das CDRs, através do silenciamento de uma proteína necessária para a cisão de macropinossomas, nomeadamente a proteína BARS, tem um efeito semelhante. Estes resultados sugerem que as CDRs e a macropinocitose são necessárias para a ativação da via de sinalização Hh e indicam que esta via de internalização controla os níveis de sinal Hh. Durante o desenvolvimento, as células proliferativas dependem do cílio primário para a transdução de várias vias de sinalização. A via Hh induz a diferenciação do músculo cardíaco. Por conseguinte, os murganhos deficientes na via de sinalização Hh exibem uma variedade de defeitos de lateralidade, incluindo alteração do looping do coração, como pode ser visto em murganhos deficientes para Arl13b. Por conseguinte, investigámos o papel da Arl13b no desenvolvimento do coração. Mostramos que a Arl13b é altamente expressa no coração de embriões de murganho e de murganhos adultos ao nível do mRNA e da proteína. Além disso, o perfil de distribuição da Arl13b no coração segue o dos cílios primários, que são essenciais para o desenvolvimento cardíaco. Corações de murganhos hnn no estadio E12,5 mostram um canal átrio-ventricular aberto, espessamento da camada compacta ventricular e aumento do índice mitótico no ventrículo esquerdo. Além disso, um atraso de 1 a 2 dias no desenvolvimento é observado em corações de murganhos hnn, quando comparados com controlos selvagens no estadio E13,5. Assim, estes resultados sugerem que a Arl13b é necessária para o desenvolvimento embrionário do coração e que defeitos cardíacos podem contribuir para a letalidade embrionária de murganhos hnn. Em suma, foi estabelecido um novo mecanismo para a regulação dos níveis de superfície do recetor Ptch1, que envolve a remodelação do citoesqueleto de actina e a formação de CDRs após a ativação da via de sinalização Hh. Este mecanismo permite um feedback negativo que evita a repressão excessiva da via através da remoção de Ptch1 da superfície da célula. Além disso, determinou-se que uma mutação de perda de função na Arl13b causa defeitos cardíacos durante o desenvolvimento, possivelmente relacionados com a associação dos defeitos em cílios primários e na sinalização Hh, existentes em murganhos deficientes para Arl13b. A via de sinalização Hh tem tido um papel central entre as vias de sinalização, uma vez que a sua regulação é crucial para o funcionamento apropriada da célula. Assim, a descoberta de um novo mecanismo de tráfego através de macropinocitose e CDRs que controla a ativação e repressão da via de sinalização Hh traz novas perspetivas de como esta via pode ser regulada e pode ainda conduzir à identificação de novos alvos e estratégias terapêuticas. --------------------ABSTRACT: Eukaryotic cells have evolved a complex signaling system that allows them to respond to extracellular and intracellular cues. Signaling pathways are essential for cell and organism survival, since they regulate fundamental processes such as development, growth, immunity, and tissue homeostasis. The Hedgehog (Hh) pathway of signal transduction involves the receptor Patched1 (Ptch1), which has an inhibitory effect on Smoothened (Smo) in the absence of its ligands, the Sonic hedgehog (Shh) proteins. These proteins are fundamental regulators of embryonic development, as illustrated by the dramatic malformations seen in human and mouse embryos with perturbed Hh signal transduction that include polydactyly, craniofacial defects and skeletal malformations. Equally important are the consequences of inappropriate activation of the Hh signaling response in tumor formation. Interestingly, the components of this pathway localize to primary cilia. Moreover, it has been shown that this localization is crucial for Hh signaling. However, in the presence of the ligands, Ptch1 is internalized and destined for degradation or sequestered in a cell compartment where it no longer can play its inhibitory role. ADP-ribosylation factor-like (Arl) 13b, a small GTPase belonging to Arf/Arl family of the Ras superfamily of small GTPases has been implicated in Joubert syndrome, a ciliopathy characterized by congenital cerebellar ataxia, hypotonia, intellectual disability and congenital heart disease. Arl13b-deficient mice, called hennin (hnn) die at embryonic day 13.5 (E13.5) and display morphological abnormalities in primary cilia that lead to the disruption of Hh signaling. Furthermore, Arl13b is directly involved in the regulation of Hh signaling by controlling the localization of several components of this pathway to primary cilia. Here, we show that Arl13b localizes to and regulates the formation of circular dorsal rufles (CDRs), which are actin-basedstructures known to be involved in macropinocytosis and receptor internalization. Additionally, we extended the knowledge of the Hh signaling activation process by showing that CDRs selectively sequester and internalize Ptch1 receptors. CDRs are formed minutes after Hh activation by Shh ligands or the Smo agonist SAG and keep being formed thereafter, suggesting a continuous induction of actin reorganization when the pathway is switched on. Importantly, we observed that disruption of CDRs by silencing WAVE1, a protein required for CDR formation, results in down-regulation of Hh signaling activation. Moreover, the blockade of macropinocytosis, which follows CDR closure, through silencing of a protein necessary for the fission of macropinosomes, namely BARS has a similar effect. These results suggest that CDRs and macropinocytosis are necessary for activation of Hh signaling and indicate that this pathway of internalization controls Hh signal levels. During development, proliferating cells rely on the primary cilium for the transduction of several signaling pathways. Hh induces the differentiation of cardiac muscle. Accordingly, Hh-deficient mice display a variety of laterality defects, including alteration of heart looping, as seen in Arl13b-deficient mice. Therefore, we investigated the role of Arl13b in heart development. We show that Arl13b is highly expressed in the heart of both embryonic and adult mice at mRNA and protein levels. Also, Arl13b localization profile mimics that of primary cilia, which have been shown to be essential to early heart development. E12.5 hnn hearts show an open atrioventricular channel, increased thickening of the ventricular compact layer and increased mitotic index in the left ventricle. Moreover, a delay of 1 to 2 days in development is observed in hnn hearts, when compared to wild-type controls at E13.5. Hence, these results suggest that Arl13b is necessary for embryonic heart development and that cardiac defects might contribute to the embryonic lethality of hnn mice. Altogether, we established a novel mechanism for the regulation of Ptch1 surface levels, involving cytoskeleton remodeling and CDR formation upon Hh signaling activation. This mechanism allows a negative feedback loop that prevents excessive repression of the pathway by removing Ptch1 from the cell surface. Additionally, we determined that the Arl13b loss-offunction mutation causes cardiac defects during development, possibly related to the associated ciliary and Hh signaling defects found in Arl13b-deficient mice. Hh signaling has taken a center stage among the signaling pathways since its regulation is crucial for the appropriate output and function of the cell. Hence, the finding of a novel trafficking mechanism through CDRs and macropinocytosis that controls Hh signaling activation and repression brings new insights to how this pathway can be regulated and can lead to the discovery of novel therapeutic targets and strategies.
Resumo:
T-cell anergy, adaptor proteins, T-cell signaling
Resumo:
Mast cells (MC) are important in the numerous physiological processes of homeostasis and disease. Most notably, MC are critical effectors in the development and exacerbation of allergic disorders. Nitric oxide (NO) is a diatomic radical produced by nitric oxide synthase (NOS), and has pluripotent cell signaling and cytotoxic properties. NO can influence many MC functions. Recent evidence shows the source of this NO can be from the mast cell itself. Governing the production of this endogenous NO, through alterations in the expression of tetrahydrobiopterin (BH4), a NOS cofactor, has stabilizing effects on MC degranulation. Furthermore, NO regulates the synthesis and secretion of de novo generated mediators, including leukotrienes and chemokines. These novel observations add to the growing body of knowledge surrounding the role of NO in the MC.
Resumo:
The tumor microenvironment mediates induction of the immunosuppressive programmed cell death-1 (PD-1) pathway, and targeted interventions against this pathway can help restore antitumor immunity. To gain insight into these responses, we studied the interaction between PD-1 expressed on T cells and its ligands (PD-1:PD-L1, PD-1:PD-L2, and PD-L1:B7.1), expressed on other cells in the tumor microenvironment, using a syngeneic orthotopic mouse model of epithelial ovarian cancer (ID8). Exhaustion of tumor-infiltrating lymphocytes (TIL) correlated with expression of PD-1 ligands by tumor cells and tumor-derived myeloid cells, including tumor-associated macrophages (TAM), dendritic cells, and myeloid-derived suppressor cells (MDSC). When combined with GVAX or FVAX vaccination (consisting of irradiated ID8 cells expressing granulocyte macrophage colony-stimulating factor or FLT3 ligand) and costimulation by agonistic α-4-1BB or TLR 9 ligand, antibody-mediated blockade of PD-1 or PD-L1 triggered rejection of ID8 tumors in 75% of tumor-bearing mice. This therapeutic effect was associated with increased proliferation and function of tumor antigen-specific effector CD8(+) T cells, inhibition of suppressive regulatory T cells (Treg) and MDSC, upregulation of effector T-cell signaling molecules, and generation of T memory precursor cells. Overall, PD-1/PD-L1 blockade enhanced the amplitude of tumor immunity by reprogramming suppressive and stimulatory signals that yielded more powerful cancer control.
Resumo:
The mechanical properties of the living cell are intimately related to cell signaling biology through cytoskeletal tension. The tension borne by the cytoskeleton (CSK) is in part generated internally by the actomyosin machinery and externally by stretch. Here we studied how cytoskeletal tension is modified during stretch and the tensional changes undergone by the sites of cell-matrix interaction. To this end we developed a novel technique to map cell-matrix stresses during application of stretch. We found that cell-matrix stresses increased with imposition of stretch but dropped below baseline levels on stretch release. Inhibition of the actomyosin machinery resulted in a larger relative increase in CSK tension with stretch and in a smaller drop in tension after stretch release. Cell-matrix stress maps showed that the loci of cell adhesion initially bearing greater stress also exhibited larger drops in traction forces after stretch removal. Our results suggest that stretch partially disrupts the actin-myosin apparatus and the cytoskeletal structures that support the largest CSK tension. These findings indicate that cells use the mechanical energy injected by stretch to rapidly reorganize their structure and redistribute tension.