Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 27
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
J Allergy Clin Immunol ; 147(4): 1341-1353, 2021 04.
Artigo em Inglês | MEDLINE | ID: mdl-32781002

RESUMO

BACKGROUND: Chronic itch is a debilitating symptom of inflammatory skin diseases, but the underlying mechanism is poorly understood. We have recently demonstrated that astrocytes in the spinal dorsal horn become reactive in models of atopic and contact dermatitis via activation of the transcription factor signal transducer and activator of transcription 3 (STAT3) and critically contribute to chronic itch. In general, STAT3 is transiently activated; however, STAT3 activation in reactive astrocytes of chronic itch model mice persistently occurs via an unknown mechanism. OBJECTIVE: We aimed to determine the mechanisms of persistent activation of astrocytic STAT3 in chronic itch conditions. METHODS: To determine the factors that are required for persistent activation of astrocytic STAT3, Western blotting and calcium imaging with cultured astrocytes or spinal cord slices were performed. Thereafter, chronic itch model mice were used for genetic and behavioral experiments to confirm the role of the factors determined to mediate persistent STAT3 activation from in vitro and ex vivo experiments in chronic itch. RESULTS: IP3 receptor type 1 (IP3R1) knockdown in astrocytes suppressed IL-6-induced persistent STAT3 activation and expression of lipocalin-2 (LCN2), an astrocytic STAT3-dependent inflammatory factor that is required for chronic itch. IP3R1-dependent astrocytic Ca2+ responses involved Ca2+ influx through the cation channel transient receptor potential canonical (TRPC), which was required for persistent STAT3 activation evoked by IL-6. IL-6 expression was upregulated in dorsal root ganglion neurons in a mouse model of chronic itch. Dorsal root ganglion neuron-specific IL-6 knockdown, spinal astrocyte-specific IP3R1 knockdown, and pharmacologic spinal TRPC inhibition attenuated LCN2 expression and chronic itch. CONCLUSION: Our findings suggest that IP3R1/TRPC channel-mediated Ca2+ signals elicited by IL-6 in astrocytes are necessary for persistent STAT3 activation, LCN2 expression, and chronic itch, and they may also provide new targets for therapeutic intervention.


Assuntos
Astrócitos/imunologia , Receptores de Inositol 1,4,5-Trifosfato/imunologia , Interleucina-6/imunologia , Prurido/imunologia , Fator de Transcrição STAT3/imunologia , Canais de Cátion TRPC/imunologia , Animais , Sinalização do Cálcio , Células Cultivadas , Doença Crônica , Gânglios Espinais/imunologia , Receptores de Inositol 1,4,5-Trifosfato/genética , Camundongos Endogâmicos C57BL , Camundongos Knockout
2.
Front Immunol ; 11: 564, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32322252

RESUMO

Mast cells are a heterogeneous group of immune cells. The simplest and commonly accepted classification divides them in two groups according to their protease content. We have compared the action of diverse secretagogues on bone marrow derived (BMMC) and peritoneal (PMC) mast cells which represent classical models of mucosal and connective tissue type mast cells in mice. Whereas, antigen stimulation of the FcεRI receptors was similarly effective in triggering elevations of free intracellular Ca2+ concentration ([Ca2+]i) in both BMMC and PMC, robust [Ca2+]i rise following Endothelin-1 stimulation was observed only in a fraction of BMMC. Leukotriene C4 activating cysteinyl leukotriene type I receptors failed to evoke [Ca2+]i rise in either mast cell model. Stimulation of the recently identified target of many small-molecule drugs associated with systemic pseudo-allergic reactions, Mrgprb2, with compound 48/80, a mast cell activator with unknown receptor studied for many years, triggered Ca2+ oscillations in BMMC and robust [Ca2+]i rise in PMCs similarly to that evoked by FcεRI stimulation. [Ca2+]i rise in PMC could also be evoked by other Mrgprb2 agonists such as Tubocurarine, LL-37, and Substance P. The extent of [Ca2+]i rise correlated with mast cell degranulation. Expression analysis of TRPC channels as potential candidates mediating agonist evoked Ca2+ entry revealed the presence of transcripts of all members of the TRPC subfamily of TRP channels in PMCs. The amplitude and AUC of compound 48/80-evoked [Ca2+]i rise was reduced by ~20% in PMC from Trpc1/4/6-/- mice compared to Trpc1/4-/- littermatched control mice, whereas FcεRI-evoked [Ca2+]i rise was unaltered. Whole-cell patch clamp recordings showed that the reduction in compound 48/80-evoked [Ca2+]i rise in Trpc1/4/6-/- PMC was accompanied by a reduced amplitude of Compound 48/80-induced cation currents which exhibited typical features of TRPC currents. Together, this study demonstrates that PMC are an appropriate mast cell model to study mechanisms of Mrgprb2 receptor-mediated mast cell activation, and it reveals that TRPC channels contribute at least partially to Mrgprb2-mediated mast cellactivation but not following FcεRI stimulation. However, the channels conducting most of the Ca2+ entry in mast cells triggered by Mrgprb2 receptor stimulation remains to be identified.


Assuntos
Sinalização do Cálcio/imunologia , Degranulação Celular/imunologia , Mastócitos/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Canais de Cátion TRPC/deficiência , Animais , Células da Medula Óssea/imunologia , Masculino , Mastócitos/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Peritônio/citologia , Peritônio/imunologia , Canais de Cátion TRPC/imunologia
3.
J Immunother Cancer ; 7(1): 111, 2019 04 23.
Artigo em Inglês | MEDLINE | ID: mdl-31014395

RESUMO

BACKGROUND: Dysregulation in calcium (Ca2+) signaling is a hallmark of chronic lymphocytic leukemia (CLL). While the role of the B cell receptor (BCR) Ca2+ pathway has been associated with disease progression, the importance of the newly described constitutive Ca2+ entry (CE) pathway is less clear. In addition, we hypothesized that these differences reflect modifications of the CE pathway and Ca2+ actors such as Orai1, transient receptor potential canonical (TRPC) 1, and stromal interaction molecule 1 (STIM1), the latter being the focus of this study. METHODS: An extensive analysis of the Ca2+ entry (CE) pathway in CLL B cells was performed including constitutive Ca2+ entry, basal Ca2+ levels, and store operated Ca2+ entry (SOCE) activated following B cell receptor engagement or using Thapsigargin. The molecular characterization of the calcium channels Orai1 and TRPC1 and to their partner STIM1 was performed by flow cytometry and/or Western blotting. Specific siRNAs for Orai1, TRPC1 and STIM1 plus the Orai1 channel blocker Synta66 were used. CLL B cell viability was tested in the presence of an anti-STIM1 monoclonal antibody (mAb, clone GOK) coupled or not with an anti-CD20 mAb, rituximab. The Cox regression model was used to determine the optimal threshold and to stratify patients. RESULTS: Seeking to explore the CE pathway, we found in untreated CLL patients that an abnormal CE pathway was (i) highly associated with the disease outcome; (ii) positively correlated with basal Ca2+ concentrations; (iii) independent from the BCR-PLCγ2-InsP3R (SOCE) Ca2+ signaling pathway; (iv) supported by Orai1 and TRPC1 channels; (v) regulated by the pool of STIM1 located in the plasma membrane (STIM1PM); and (vi) blocked when using a mAb targeting STIM1PM. Next, we further established an association between an elevated expression of STIM1PM and clinical outcome. In addition, combining an anti-STIM1 mAb with rituximab significantly reduced in vitro CLL B cell viability within the high STIM1PM CLL subgroup. CONCLUSIONS: These data establish the critical role of a newly discovered BCR independent Ca2+ entry in CLL evolution, provide new insights into CLL pathophysiology, and support innovative therapeutic perspectives such as targeting STIM1 located at the plasma membrane.


Assuntos
Antineoplásicos Imunológicos/farmacologia , Linfócitos B/efeitos dos fármacos , Sinalização do Cálcio/genética , Leucemia Linfocítica Crônica de Células B/tratamento farmacológico , Proteínas de Neoplasias/antagonistas & inibidores , Molécula 1 de Interação Estromal/antagonistas & inibidores , Idoso , Anticorpos Monoclonais/farmacologia , Anticorpos Monoclonais/uso terapêutico , Antineoplásicos Imunológicos/uso terapêutico , Linfócitos B/citologia , Linfócitos B/imunologia , Linfócitos B/metabolismo , Cálcio/imunologia , Cálcio/metabolismo , Sinalização do Cálcio/imunologia , Membrana Celular/metabolismo , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/imunologia , Progressão da Doença , Feminino , Técnicas de Silenciamento de Genes , Humanos , Leucemia Linfocítica Crônica de Células B/sangue , Leucemia Linfocítica Crônica de Células B/imunologia , Leucemia Linfocítica Crônica de Células B/patologia , Masculino , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Proteína ORAI1/antagonistas & inibidores , Proteína ORAI1/genética , Proteína ORAI1/imunologia , Proteína ORAI1/metabolismo , Cultura Primária de Células , Estudos Prospectivos , RNA Interferente Pequeno/metabolismo , Molécula 1 de Interação Estromal/genética , Molécula 1 de Interação Estromal/metabolismo , Canais de Cátion TRPC/genética , Canais de Cátion TRPC/imunologia , Canais de Cátion TRPC/metabolismo , Resultado do Tratamento , Células Tumorais Cultivadas
5.
Curr Opin Immunol ; 38: 24-9, 2016 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-26584476

RESUMO

Transendothelial migration (TEM) of leukocytes is the step in leukocyte emigration in which the leukocyte actually leaves the blood vessel to carry out its role in the inflammatory response. It is therefore, arguably the most critical step in emigration. This review focuses on two of the many aspects of this process that have seen important recent developments. The adhesion molecules, PECAM (CD31) and CD99 that regulate two major steps in TEM, do so by regulating specific signals. PECAM initiates the signaling pathway responsible for the calcium flux that is required for TEM. Calcium enters through the cation channel TRPC6 and recruits the first wave of trafficking of membrane from the lateral border recycling compartment (LBRC). CD99 signals through soluble adenylate cyclase to activate protein kinase A to recruit a second wave of LBRC trafficking. Another process that is critical for TEM is transient removal of VE-cadherin from the site of TEM. However, the local signaling pathways that are responsible for this appear to be different from those that open the junctions to increase vascular permeability.


Assuntos
Regulação da Expressão Gênica/imunologia , Leucócitos/imunologia , Transdução de Sinais/imunologia , Migração Transendotelial e Transepitelial/imunologia , Antígeno 12E7 , Adenilil Ciclases/genética , Adenilil Ciclases/imunologia , Animais , Antígenos CD/genética , Antígenos CD/imunologia , Caderinas/genética , Caderinas/imunologia , Cálcio/imunologia , Cálcio/metabolismo , Moléculas de Adesão Celular/genética , Moléculas de Adesão Celular/imunologia , Proteínas Quinases Dependentes de AMP Cíclico/genética , Proteínas Quinases Dependentes de AMP Cíclico/imunologia , Humanos , Leucócitos/citologia , Molécula-1 de Adesão Celular Endotelial a Plaquetas/genética , Molécula-1 de Adesão Celular Endotelial a Plaquetas/imunologia , Canais de Cátion TRPC/genética , Canais de Cátion TRPC/imunologia , Canal de Cátion TRPC6
6.
Mol Cell Biol ; 35(16): 2729-39, 2015 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-26031335

RESUMO

Transient receptor potential channel 1 (TRPC1) is a nonselective cation channel that is required for Ca(2+) homeostasis necessary for cellular functions. However, whether TRPC1 is involved in infectious disease remains unknown. Here, we report a novel function for TRPC1 in host defense against Gram-negative bacteria. TRPC1(-/-) mice exhibited decreased survival, severe lung injury, and systemic bacterial dissemination upon infection. Furthermore, silencing of TRPC1 showed decreased Ca(2+) entry, reduced proinflammatory cytokines, and lowered bacterial clearance. Importantly, TRPC1 functioned as an endogenous Ca(2+) entry channel critical for proinflammatory cytokine production in both alveolar macrophages and epithelial cells. We further identified that bacterium-mediated activation of TRPC1 was dependent on Toll-like receptor 4 (TLR4), which induced endoplasmic reticulum (ER) store depletion. After activation of phospholipase Cγ (PLC-γ), TRPC1 mediated Ca(2+) entry and triggered protein kinase Cα (PKCα) activity to facilitate nuclear translocation of NF-κB/Jun N-terminal protein kinase (JNK) and augment the proinflammatory response, leading to tissue damage and eventually mortality. These findings reveal that TRPC1 is required for host defense against bacterial infections through the TLR4-TRPC1-PKCα signaling circuit.


Assuntos
Lesão Pulmonar/genética , Lesão Pulmonar/microbiologia , Proteína Quinase C-alfa/imunologia , Infecções por Pseudomonas/complicações , Infecções por Pseudomonas/genética , Pseudomonas aeruginosa/imunologia , Canais de Cátion TRPC/genética , Animais , Cálcio/imunologia , Células Cultivadas , Técnicas de Inativação de Genes , Pulmão/imunologia , Pulmão/metabolismo , Pulmão/microbiologia , Lesão Pulmonar/etiologia , Lesão Pulmonar/imunologia , Sistema de Sinalização das MAP Quinases , Camundongos , Camundongos Knockout , NF-kappa B/imunologia , Infecções por Pseudomonas/imunologia , Transdução de Sinais , Canais de Cátion TRPC/imunologia , Receptor 4 Toll-Like/imunologia
7.
PLoS One ; 10(4): e0122227, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25849346

RESUMO

Mechanical forces exerted on cells impose stress on the plasma membrane. Cells sense this stress and elicit a mechanoelectric transduction cascade that initiates compensatory mechanisms. Mechanosensitive ion channels in the plasma membrane are responsible for transducing the mechanical signals to electrical signals. However, the mechanisms underlying channel activation in response to mechanical stress remain incompletely understood. Transient Receptor Potential (TRP) channels serve essential functions in several sensory modalities. These channels can also participate in mechanotransduction by either being autonomously sensitive to mechanical perturbation or by coupling to other mechanosensory components of the cell. Here, we investigated the response of a TRP family member, TRPC5, to mechanical stress. Hypoosmolarity triggers Ca2+ influx and cationic conductance through TRPC5. Importantly, for the first time we were able to record the stretch-activated TRPC5 current at single-channel level. The activation threshold for TRPC5 was found to be 240 mOsm for hypoosmotic stress and between -20 and -40 mmHg for pressure applied to membrane patch. In addition, we found that disruption of actin filaments suppresses TRPC5 response to hypoosmotic stress and patch pipette pressure, but does not prevent the activation of TRPC5 by stretch-independent mechanisms, indicating that actin cytoskeleton is an essential transduction component that confers mechanosensitivity to TRPC5. In summary, our findings establish that TRPC5 can be activated at the single-channel level when mechanical stress on the cell reaches a certain threshold.


Assuntos
Membrana Celular/metabolismo , Estresse Mecânico , Canais de Cátion TRPC/metabolismo , Citoesqueleto de Actina/efeitos dos fármacos , Animais , Anticorpos/imunologia , Células CHO , Cálcio/metabolismo , Cricetinae , Cricetulus , Citocalasina D/farmacologia , Células HEK293 , Humanos , Mecanotransdução Celular/fisiologia , Camundongos , Pressão Osmótica , Técnicas de Patch-Clamp , Canais de Cátion TRPC/genética , Canais de Cátion TRPC/imunologia
8.
Cell Calcium ; 54(2): 120-5, 2013 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-23764169

RESUMO

Canonical transient receptor potential 1 (TRPC1) plasmalemmal cation channels mediate Ca2+ and Na+ fluxes and control respective cytoplasmic ion signals in rat cortical astrocytes. Mechanical stimulation of astrocytes results in an increase in the levels of cytosolic Ca2+ and Na+ that are in part due to entry of extracellular cations through TRPC1 containing channels. Inhibition of the TRPC1 pore with an antibody against its selective filter reduced cytosolic Ca2+ accumulation caused by mechanical stimulation. In contrast, this immunological treatment increased the cytosolic Na+ peak accumulation induced by mechanical stimulation. We propose that TRPC channels are amenable to changes in selective filtering, as mutations in previous studies and antibody binding in our present study differentially affect the flux of Ca2) and Na+. TRPC1 containing channels might represent focal points for co-ordination of Ca2+ and Na+ signalling in astroglia and this can have consequences on Ca(2+)- and Na(+)-dependent processes such as regulated exocytosis and lactate production, respectively, which in turn can modulate neuronal synaptic transmission.


Assuntos
Astrócitos/metabolismo , Sinalização do Cálcio/fisiologia , Cálcio/metabolismo , Sódio/metabolismo , Canais de Cátion TRPC/metabolismo , Animais , Anticorpos Anti-Idiotípicos/farmacologia , Astrócitos/citologia , Células Cultivadas , Exocitose/fisiologia , Técnicas In Vitro , Modelos Animais , Ratos , Ratos Sprague-Dawley , Estresse Mecânico , Transmissão Sináptica/fisiologia , Canais de Cátion TRPC/efeitos dos fármacos , Canais de Cátion TRPC/imunologia
9.
Am J Physiol Cell Physiol ; 303(11): C1156-72, 2012 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-23034388

RESUMO

Previous studies in pulmonary artery smooth muscle cells (PASMCs) showed that acute hypoxia activates capacitative Ca(2+) entry (CCE) but the molecular candidate(s) mediating CCE caused by acute hypoxia remain unclear. The present study aimed to determine if transient receptor potential canonical 1 (TRPC1) and Orai1 interact with stromal interacting molecule 1 (STIM1) and mediate CCE caused by acute hypoxia in mouse PASMCs. In primary cultured PASMCs loaded with fura-2, acute hypoxia caused a transient followed by a sustained rise in intracellular Ca(2+) concentration ([Ca(2+)](i)). The transient but not sustained rise in [Ca(2+)](i) was partially inhibited by nifedipine. Acute hypoxia also increased the rate of Mn(2+) quench of fura-2 fluorescence that was inhibited by SKF 96365, Ni(2+), La(3+), and Gd(3+), exhibiting pharmacological properties characteristic of CCE. The nifedipine-insensitive rise in [Ca(2+)](i) and the increase in Mn(2+) quench rate were both inhibited in cells treated with TRPC1 antibody or TRPC1 small interfering (si)RNA, in STIM1 siRNA-transfected cells and in Orai1 siRNA-transfected cells. Moreover, overexpression of STIM1 resulted in a marked increase in [Ca(2+)](i) and Mn(2+) quench rate caused by acute hypoxia, and they were reduced in cells treated with TRPC1 antibody and in cells transfected with Orai1 siRNA. Furthermore, TRPC1 and Orai1 coimmunoprecipitated with STIM1 and the precipitation levels of TRPC1 and Orai1 were increased in cells exposed to acute hypoxia. Immunostaining showed colocalizations of TRPC1-STIM1 and Orai1-STIM1, and the colocalizations of these proteins were more apparent in acute hypoxia. These data provide direct evidence that TRPC1 and Orai1 channels mediate CCE through activation of STIM1 in acute hypoxic mouse PASMCs.


Assuntos
Canais de Cálcio/fisiologia , Cálcio/fisiologia , Glicoproteínas de Membrana/fisiologia , Músculo Liso Vascular/fisiopatologia , Miócitos de Músculo Liso/fisiologia , Artéria Pulmonar/fisiopatologia , Canais de Cátion TRPC/fisiologia , Animais , Anticorpos Neutralizantes/farmacologia , Cálcio/análise , Bloqueadores dos Canais de Cálcio/farmacologia , Hipóxia Celular/efeitos dos fármacos , Hipóxia Celular/fisiologia , Células Cultivadas , Gadolínio/farmacologia , Inativação Gênica , Imidazóis/farmacologia , Lantânio/farmacologia , Masculino , Manganês/química , Camundongos , Camundongos Endogâmicos C57BL , Músculo Liso Vascular/citologia , Músculo Liso Vascular/efeitos dos fármacos , Miócitos de Músculo Liso/citologia , Miócitos de Músculo Liso/efeitos dos fármacos , Níquel/farmacologia , Nifedipino/farmacologia , Proteína ORAI1 , Artéria Pulmonar/efeitos dos fármacos , Molécula 1 de Interação Estromal , Canais de Cátion TRPC/genética , Canais de Cátion TRPC/imunologia
10.
Genes Immun ; 13(2): 175-83, 2012 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-21956657

RESUMO

Typhoid fever, which is caused by Salmonella typhi and paratyphi, is a severe systemic disease that remains a major public health issue in several areas of the world. We can model the human disease using mice infected with a related bacterium, Salmonella typhimurium. This model recapitulates several clinical aspects of the human disease and allows for the study of the host response to Salmonella typhimurium infection in vivo. Previous work in our laboratory has identified three Immunity to typhimurium loci (Ity, Ity2 and Ity3) in the wild-derived MOLF/Ei mice, influencing survival after infection with Salmonella typhimurium. The MOLF/Ei alleles at Ity and Ity2 are protective, while the MOLF/Ei allele at Ity3 confers susceptibility. In this paper, we have generated a novel cross combination between the highly susceptible strain, MOLF/Ei, and the resistant strain, 129S6, to better define the genetic architecture of susceptibility to infection in MOLF/Ei. Using this cross, we have replicated the locus on chr 11 (Ity2) and identified a novel locus on chr 13 (Ity13). Using microarrays and transcriptional profiling, we examined the response of uninfected and infected Ity2 congenic mice. These analyses demonstrate a role for both type-1-interferon (IFN) and TRP53 signaling in the pathogenesis of Salmonella infection.


Assuntos
Proteínas de Transporte de Cátions/imunologia , Infecções por Salmonella/genética , Salmonella typhimurium , Transdução de Sinais , Alelos , Animais , Proteínas de Transporte de Cátions/metabolismo , Feminino , Predisposição Genética para Doença , Interferon Tipo I/imunologia , Masculino , Camundongos , Análise de Sequência com Séries de Oligonucleotídeos , Infecções por Salmonella/imunologia , Canais de Cátion TRPC/imunologia , Canais de Cátion TRPC/metabolismo , Proteína Supressora de Tumor p53/imunologia , Proteína Supressora de Tumor p53/metabolismo
11.
Mol Ther ; 20(2): 432-42, 2012 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-21934655

RESUMO

In vivo dendritic cells (DC) targeting is an attractive approach with potential advantages in vaccine efficacy, cost, and availability. Identification of molecular adjuvants to in vivo "modulate " DC to coordinately render improved Th1 and CD8 T cell immunity, and attenuated deleterious Treg effects, is a critical challenge. Here, we report that in vivo genetic targeting of the active transcription factor XBP1s to DC (XBP1s/DC) potentiated vaccine-induced prophylactic and therapeutic antitumor immunity in multiple tumor models. This immunization strategy is based on a genetic vaccine encoding both cytomegalovirus (CMV)-driven vaccine Aghsp70 and DC-specific CD11c-driven XBP1s. The novel targeted vaccine induced durable Th1 and CD8 T cell responses to poorly immunogenic self/tumor antigen (Ag) and attenuated tumor-associated Treg suppressive function. Bone marrow (BM)-derived DC genetically modified to simultaneously overexpress XBP1s and express Aghsp70 upregulated CD40, CD70, CD86, interleukin (IL)-15, IL-15Rα, and CCR7 expression, and increased IL-6, IL-12, and tumor necrosis factor (TNF)-α production in vitro. XBP1s/DC elevated functional DEC205(+)CD8α(+)DC in the draining lymph nodes (DLN). The data suggest a novel role for XBP1s in modulating DC to potentiate tumor vaccine efficacy via overcoming two major obstacles to tumor vaccines (i.e., T cell hyporesponsiveness against poorly immunologic self/tumor Ag and tumor-associated Treg-mediated suppression) and improving DEC205(+)CD8α(+)DC.


Assuntos
Vacinas Anticâncer/genética , Vacinas Anticâncer/imunologia , Proteínas de Ligação a DNA/genética , Células Dendríticas/imunologia , Marcação de Genes , Neoplasias Experimentais/imunologia , Fatores de Transcrição/genética , Animais , Linfócitos T CD8-Positivos/imunologia , Vacinas Anticâncer/uso terapêutico , Linhagem Celular Tumoral , Sobrevivência Celular/genética , Proteínas de Ligação a DNA/metabolismo , Células Dendríticas/metabolismo , Feminino , Expressão Gênica , Ordem dos Genes , Interferon gama/biossíntese , Linfonodos/imunologia , Linfonodos/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Neoplasias Experimentais/tratamento farmacológico , Neoplasias Experimentais/mortalidade , Fatores de Transcrição de Fator Regulador X , Análise de Sobrevida , Linfócitos T Reguladores , Canais de Cátion TRPC/imunologia , Células Th1/imunologia , Fatores de Transcrição/metabolismo , Vacinas de DNA/genética , Vacinas de DNA/imunologia , Proteína 1 de Ligação a X-Box
12.
J Neurophysiol ; 106(3): 1515-24, 2011 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-21753024

RESUMO

Cholecystokinin (CCK) is one of the most abundant neuropeptides in the brain, where it interacts with two G protein-coupled receptors (CCK-1 and CCK-2). Activation of both CCK receptors increases the activity of PLC, resulting in increases in intracellular calcium ion (Ca(2+)) release and activation of PKC. Whereas high density of CCK receptors has been detected in the superficial layers of the entorhinal cortex (EC), the functions of CCK in this brain region have not been determined. Here, we studied the effects of CCK on neuronal excitability of layer III pyramidal neurons in the EC. Our results showed that CCK remarkably increased the firing frequency of action potentials (APs). The effects of CCK on neuronal excitability were mediated via activation of CCK-2 receptors and required the functions of G proteins and PLC. However, CCK-mediated facilitation of neuronal excitability was independent of inositol trisphosphate receptors and PKC. CCK facilitated neuronal excitability by activating a cationic channel to generate membrane depolarization. The effects of CCK were suppressed by the generic, nonselective cationic channel blockers, 2-aminoethyldiphenyl borate and flufenamic acid, but potentiated by gadolinium ion and lanthanum ion at 100 µM. Depletion of extracellular Ca(2+) also counteracted CCK-induced increases in AC firing frequency. Moreover, CCK-induced enhancement of neuronal excitability was inhibited significantly by intracellular application of the antibody to transient receptor potential channel 5 (TRPC5), suggesting the involvement of TRPC5 channels. Our results provide a cellular and molecular mechanism to help explain the functions of CCK in vivo.


Assuntos
Colecistocinina/fisiologia , Córtex Entorrinal/fisiologia , Neurônios/fisiologia , Canais de Cátion TRPC/fisiologia , Animais , Anticorpos/toxicidade , Colecistocinina/antagonistas & inibidores , Colecistocinina/deficiência , Camundongos , Camundongos Knockout , Neurônios/imunologia , Células Piramidais/imunologia , Células Piramidais/fisiologia , Ratos , Ratos Sprague-Dawley , Receptor de Colecistocinina B/deficiência , Receptor de Colecistocinina B/genética , Canais de Cátion TRPC/imunologia
13.
J Natl Cancer Inst ; 102(14): 1052-68, 2010 Jul 21.
Artigo em Inglês | MEDLINE | ID: mdl-20554944

RESUMO

BACKGROUND: Patients with glioblastoma multiforme, the most aggressive form of glioma, have a median survival of approximately 12 months. Calcium (Ca(2+)) signaling plays an important role in cell proliferation, and some members of the Ca(2+)-permeable transient receptor potential canonical (TRPC) family of channel proteins have demonstrated a role in the proliferation of many types of cancer cells. In this study, we investigated the role of TRPC6 in cell cycle progression and in the development of human glioma. METHODS: TRPC6 protein and mRNA expression were assessed in glioma (n = 33) and normal (n = 17) brain tissues from patients and in human glioma cell lines U251, U87, and T98G. Activation of TRPC6 channels was tested by platelet-derived growth factor-induced Ca(2+) imaging. The effect of inhibiting TRPC6 activity or expression using the dominant-negative mutant TRPC6 (DNC6) or RNA interference, respectively, was tested on cell growth, cell cycle progression, radiosensitization of glioma cells, and development of xenografted human gliomas in a mouse model. The green fluorescent protein (GFP) and wild-type TRPC6 (WTC6) were used as controls. Survival of mice bearing xenografted tumors in the GFP, DNC6, and WTC6 groups (n = 13, 15, and 13, respectively) was compared using Kaplan-Meier analysis. All statistical tests were two-sided. RESULTS: Functional TRPC6 was overexpressed in human glioma cells. Inhibition of TRPC6 activity or expression attenuated the increase in intracellular Ca(2+) by platelet-derived growth factor, suppressed cell growth and clonogenic ability, induced cell cycle arrest at the G2/M phase, and enhanced the antiproliferative effect of ionizing radiation. Cyclin-dependent kinase 1 activation and cell division cycle 25 homolog C expression regulated the cell cycle arrest. Inhibition of TRPC6 activity also reduced tumor volume in a subcutaneous mouse model of xenografted human tumors (P = .014 vs GFP; P < .001 vs WTC6) and increased mean survival in mice in an intracranial model (P < .001 vs GFP or WTC6). CONCLUSIONS: In this preclinical model, TRPC6 channels were essential for glioma development via regulation of G2/M phase transition. This study suggests that TRPC6 might be a new target for therapeutic intervention of human glioma.


Assuntos
Neoplasias Encefálicas/metabolismo , Divisão Celular , Transformação Celular Neoplásica/metabolismo , Fase G2 , Glioma/metabolismo , Canais de Cátion TRPC/metabolismo , Adenoviridae , Animais , Sequência de Bases , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/patologia , Linhagem Celular Tumoral , Transformação Celular Neoplásica/genética , Transformação Celular Neoplásica/imunologia , Modelos Animais de Doenças , Citometria de Fluxo , Técnica Indireta de Fluorescência para Anticorpo , Regulação Neoplásica da Expressão Gênica , Vetores Genéticos , Glioma/genética , Glioma/patologia , Humanos , Immunoblotting , Imuno-Histoquímica , Hibridização In Situ , Lentivirus , Camundongos , Dados de Sequência Molecular , Fenótipo , RNA Mensageiro/metabolismo , RNA Interferente Pequeno , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Canais de Cátion TRPC/antagonistas & inibidores , Canais de Cátion TRPC/genética , Canais de Cátion TRPC/imunologia , Canal de Cátion TRPC6 , Transplante Heterólogo , Regulação para Cima
14.
Rinsho Shinkeigaku ; 49(11): 789-93, 2009 Nov.
Artigo em Japonês | MEDLINE | ID: mdl-20030211

RESUMO

Autoantibodies impair acetylcholine receptor (AChR) in myasthenia gravis (MG) and P/Q-type voltage-gated calcium channel (VGCC) in Lambert-Eaton myasthenic syndrome (LEMS). (1) Some of MG and LEMS patients are "seronegative" for respective antibodies or modified by antibodies that recognize other proteins than AChR and VGCC such as MuSK, AChR allosteric site, membrane Na+ channel and ryanodine receptor-1 (RyR1) in MG, and synaptotagmin-1 in LEMS. (2) Autoimmune responses affect the proteins participating in the mechanisms to compensate for synaptic disorders on the basis of presynaptic Ca2+ homeostasis provided by VGCC and non-VGCC (receptor-operated TRPCs): they act as enhancers of Ca(2+) -mediated ACh release via phospholipase C signaling pathways including M1-type presynaptic muscarinic AChR, neurotrophin receptor (TrkB), and fast-mode of synaptic vesicle recycling. (3) The pathophysiology contributive to contractile fatigue in MG includes RyR1 and also TRPC3. The TRPC3 also forms a complex with STIM1 and Orail to make up for Ca2+ after sarcoplasmic Ca2+ release. The prevalent detection of anti-TRPC3 antibodies in MG with thymoma could affect muscle contractile machineries in addition to anti-RyR1-induced affection. (4) When one faces "seronegative" MG, one should be cautious to conformation-specific antibodies and also congenital myasthenic syndromes.


Assuntos
Autoanticorpos , Miastenia Gravis , Sinapses/imunologia , Cálcio/metabolismo , Cálcio/fisiologia , Canais de Cálcio/imunologia , Sinalização do Cálcio/fisiologia , Humanos , Síndrome Miastênica de Lambert-Eaton/genética , Síndrome Miastênica de Lambert-Eaton/imunologia , Miastenia Gravis/genética , Miastenia Gravis/imunologia , Receptores Proteína Tirosina Quinases/imunologia , Receptores Colinérgicos/imunologia , Receptores Muscarínicos/imunologia , Receptores Nicotínicos/imunologia , Canal de Liberação de Cálcio do Receptor de Rianodina/imunologia , Sinapses/fisiologia , Sinaptotagmina I/imunologia , Canais de Cátion TRPC/imunologia
15.
J Immunol ; 183(10): 6478-88, 2009 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-19864608

RESUMO

Ca(2+) mobilization is central to many cellular processes, including stimulated exocytosis and cytokine production in mast cells. Using single cell stimulation by IgE-specific Ag and high-speed imaging of conventional or genetically encoded Ca(2+) sensors in rat basophilic leukemia and bone marrow-derived rat mast cells, we observe Ca(2+) waves that originate most frequently from the tips of extended cell protrusions, as well as Ca(2+) oscillations throughout the cell that usually follow the initiating Ca(2+) wave. In contrast, Ag conjugated to the tip of a micropipette stimulates local, repetitive Ca(2+) puffs at the region of cell contact. Initiating Ca(2+) waves are observed in most rat basophilic leukemia cells stimulated with soluble Ag and are sensitive to inhibitors of Ca(2+) release from endoplasmic reticulum stores and to extracellular Ca(2+), but they do not depend on store-operated Ca(2+) entry. Knockdown of transient receptor potential channel (TRPC)1 and TRPC3 channel proteins by short hairpin RNA reduces the sensitivity of these cells to Ag and shifts the wave initiation site from protrusions to the cell body. Our results reveal spatially encoded Ca(2+) signaling in response to immunoreceptor activation that utilizes TRPC channels to specify the initiation site of the Ca(2+) response.


Assuntos
Basófilos/imunologia , Cálcio/imunologia , Mastócitos/imunologia , Animais , Basófilos/efeitos dos fármacos , Basófilos/metabolismo , Cálcio/antagonistas & inibidores , Cálcio/metabolismo , Bloqueadores dos Canais de Cálcio/farmacologia , Sinalização do Cálcio/efeitos dos fármacos , Sinalização do Cálcio/imunologia , Linhagem Celular Tumoral , Estrenos/farmacologia , Imunoglobulina E/imunologia , Imunoglobulina E/metabolismo , Mastócitos/citologia , Mastócitos/efeitos dos fármacos , Mastócitos/metabolismo , Nifedipino/farmacologia , Inibidores de Fosfodiesterase/farmacologia , Pirrolidinonas/farmacologia , Ratos , Esfingosina/farmacologia , Canais de Cátion TRPC/imunologia , Canais de Cátion TRPC/metabolismo
16.
Arch Biochem Biophys ; 490(2): 137-44, 2009 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-19695217

RESUMO

Ca(2+) entry, particularly store-operated Ca(2+) entry (SOCE), has been reported to be crucial for a variety of cellular functions. SOCE is a mechanism regulated by the Ca(2+) content of the stores, where the intraluminal Ca(2+) sensor STromal Interaction Molecule 1 (STIM1) has been reported to communicate the filling state of the intracellular Ca(2+) stores to the store-operated Ca(2+)-permeable channels in the plasma membrane, likely involving Orai1 and TRPC proteins, such as TRPC1. Here we have investigated the role of Orai1, STIM1 and TRPC1 in platelet aggregation, an event that occurs during the process of thrombosis and hemostasis. Electrotransjection of cells with anti-STIM1 (25-139) antibody, directed towards the Ca(2+)-binding motif, significantly reduced thrombin-induced aggregation and prevented ADP-evoked response. Extracellular application of the anti-STIM1 antibody, in order to block the function of plasma membrane-located STIM1, reduced thrombin- and ADP-stimulated platelet aggregation to a lesser extent. Introduction of an anti-Orai1 (288-301) antibody, which binds the STIM1-binding site located in the Orai1 C-terminus, or extracellular application of anti-hTRPC1 (557-571) antibody to impair hTRPC1 channel function, significantly reduced thrombin- and ADP-induced platelet aggregation. These findings suggest a role of STIM1, Orai1 and hTRPC1 in thrombin- and ADP-induced platelet aggregation probably through the regulation of Ca(2+) entry, which might become targets for the development of therapeutic strategies to treat platelet hyperactivity and thrombosis disorders.


Assuntos
Difosfato de Adenosina/farmacologia , Canais de Cálcio/sangue , Proteínas de Membrana/sangue , Proteínas de Neoplasias/sangue , Agregação Plaquetária/efeitos dos fármacos , Agregação Plaquetária/fisiologia , Canais de Cátion TRPC/sangue , Trombina/farmacologia , Animais , Anticorpos/administração & dosagem , Plaquetas/efeitos dos fármacos , Plaquetas/fisiologia , Cálcio/farmacologia , Canais de Cálcio/imunologia , Sinalização do Cálcio , Humanos , Técnicas In Vitro , Proteínas de Membrana/antagonistas & inibidores , Proteínas de Membrana/imunologia , Proteínas de Neoplasias/antagonistas & inibidores , Proteínas de Neoplasias/imunologia , Proteína ORAI1 , Molécula 1 de Interação Estromal , Canais de Cátion TRPC/antagonistas & inibidores , Canais de Cátion TRPC/imunologia
17.
Crit Rev Immunol ; 29(2): 155-86, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-19496745

RESUMO

Calcium signals mediate diverse cellular functions in immunological cells. Early studies with mast cells, then a preeminent model for studying Ca2+-dependent exocytosis, revealed several basic features of calcium signaling in non-electrically excitable cells. Subsequent studies in these and other cells further defined the basic processes such as inositol 1,4,5-trisphosphate-mediated release of Ca2+ from Ca2+ stores in the endoplasmic reticulum (ER); coupling of ER store depletion to influx of external Ca2+ through a calcium-release activated calcium (CRAC) channel now attributed to the interaction of the ER Ca2+ sensor, stromal interacting molecule-1 (STIM1), with a unique Ca2+-channel protein, Orai1/CRACM1, and subsequent uptake of excess Ca2+ into ER and mitochondria through ATP-dependent Ca2+ pumps. In addition, transient receptor potential channels and ion exchangers also contribute to the generation of calcium signals that may be global or have dynamic (e.g., waves and oscillations) and spatial resolution for specific functional readouts. This review discusses past and recent developments in this field of research, the pharmacologic agents that have assisted in these endeavors, and the mast cell as an exemplar for sorting out how calcium signals may regulate multiple outputs in a single cell.


Assuntos
Canais de Cálcio/metabolismo , Sinalização do Cálcio/fisiologia , Cálcio/metabolismo , Mastócitos/metabolismo , Mitocôndrias/metabolismo , Animais , Cálcio/imunologia , Canais de Cálcio/imunologia , ATPases Transportadoras de Cálcio/imunologia , ATPases Transportadoras de Cálcio/metabolismo , Humanos , Receptores de Inositol 1,4,5-Trifosfato/imunologia , Receptores de Inositol 1,4,5-Trifosfato/metabolismo , Mastócitos/imunologia , Potenciais da Membrana/fisiologia , Proteínas de Membrana/imunologia , Proteínas de Membrana/metabolismo , Mitocôndrias/imunologia , Transdução de Sinais/fisiologia , Canais de Cátion TRPC/imunologia , Canais de Cátion TRPC/metabolismo
18.
J Immunol ; 182(7): 4036-45, 2009 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-19299701

RESUMO

Several animal autoimmune disorders are suppressed by treatment with the GM1 cross-linking units of certain toxins such as B subunit of cholera toxin (CtxB). Due to the recent observation of GM1 being a binding partner for the endogenous lectin galectin-1 (Gal-1), which is known to ameliorate symptoms in certain animal models of autoimmune disorders, we tested the hypothesis that an operative Gal-1/GM1 interplay induces immunosuppression in a manner evidenced by both in vivo and in vitro systems. Our study of murine experimental autoimmune encephalomyelitis (EAE) indicated suppressive effects by both CtxB and Gal-1 and further highlighted the role of GM1 in demonstrating enhanced susceptibility to EAE in mice lacking this ganglioside. At the in vitro level, polyclonal activation of murine regulatory T (Treg) cells caused up-regulation of Gal-1 that was both cell bound and released to the medium. Similar activation of murine CD4(+) and CD8(+) effector T (Teff) cells resulted in significant elevation of GM1 and GD1a, the neuraminidase-reactive precursor to GM1. Activation of Teff cells also up-regulated TRPC5 channels which mediated Ca(2+) influx upon GM1 cross-linking by Gal-1 or CtxB. This involved co-cross-linking of heterodimeric integrin due to close association of these alpha(4)beta(1) and alpha(5)beta(1) glycoproteins with GM1. Short hairpin RNA (shRNA) knockdown of TRPC5 in Teff cells blocked contact-dependent proliferation inhibition by Treg cells as well as Gal-1/CtxB-triggered Ca(2+) influx. Our results thus indicate GM1 in Teff cells to be the primary target of Gal-1 expressed by Treg cells, the resulting co-cross-linking and TRPC5 channel activation contributing importantly to the mechanism of autoimmune suppression.


Assuntos
Encefalomielite Autoimune Experimental/imunologia , Gangliosídeo G(M1)/metabolismo , Galectina 1/metabolismo , Linfócitos T Reguladores/imunologia , Canais de Cátion TRPC/imunologia , Animais , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD4-Positivos/metabolismo , Linfócitos T CD8-Positivos/imunologia , Linfócitos T CD8-Positivos/metabolismo , Toxina da Cólera/imunologia , Toxina da Cólera/metabolismo , Reagentes de Ligações Cruzadas , Encefalomielite Autoimune Experimental/metabolismo , Encefalomielite Autoimune Experimental/patologia , Feminino , Citometria de Fluxo , Gangliosídeo G(M1)/imunologia , Galectina 1/imunologia , Imunoprecipitação , Ativação Linfocitária/imunologia , Camundongos , Camundongos Knockout , Medula Espinal/imunologia , Medula Espinal/metabolismo , Medula Espinal/patologia , Linfócitos T Reguladores/metabolismo
19.
Pain ; 140(2): 292-304, 2008 Nov 30.
Artigo em Inglês | MEDLINE | ID: mdl-18834667

RESUMO

The roles of ion channels in sensory neurons were examined in experimental models of muscle pain in the rat. Rats were injected with 50 microl of 4% carrageenan or subjected to an eccentric exercise (ECC) of the gastrocnemius muscle (GM). The Randall-Selitto and von Frey tests were performed on the calves to evaluate mechanical hyperalgesia of the muscle. The changes in expression of four genes and proteins of ion channels in dorsal root ganglia were examined using quantitative PCR and immunohistochemistry, respectively. Effects of antagonists to transient receptor potential (TRP) channels and acid sensing ion channels (ASICs) on the mechanical hyperalgesia induced by carrageenan injection or ECC were evaluated. The mechanical hyperalgesia was observed 6-24h after carrageenan injection and 1-3 days after ECC in the Randall-Selitto test. Infiltrations of the inflammatory cells in the GM were seen in carrageenan-injected animals but not in those subjected to ECC. Expressions of genes and proteins in sensory neurons showed no changes. Intramuscular injection of antagonists to TRPV1 showed an almost complete suppressive effect on ECC-induced muscle hyperalgesia but not a carrageenan-induced one. Antagonists to TRP channels and ASICs showed suppressive effects for both carrageenan- and ECC-induced muscle hyperalgesia. The carrageenan injection and ECC models are useful models of acute inflammatory pain and delayed onset muscle soreness (DOMS), respectively, and the time course and underlying etiology might be different. TRP channels and ASICs are closely related to the development of muscle mechanical hyperalgesia, and TRPV1 is involved in ECC-induced DOMS.


Assuntos
Modelos Animais de Doenças , Fibromialgia/imunologia , Gânglios Espinais/imunologia , Hiperalgesia/imunologia , Miosite/imunologia , Proteínas do Tecido Nervoso/imunologia , Canais de Sódio/imunologia , Canais de Cátion TRPC/imunologia , Canais Iônicos Sensíveis a Ácido , Animais , Masculino , Ratos , Ratos Sprague-Dawley , Tato
20.
J Neuroimmunol ; 200(1-2): 142-4, 2008 Aug 30.
Artigo em Inglês | MEDLINE | ID: mdl-18602703

RESUMO

The transient receptor potential canonical type-3 (TRPC3, receptor- and store-operated Ca(2+) influx channel) participates in skeletal muscle contraction; its functional interactions with ryanodine receptor-1 (RyR1) are independent of sarcoplasmic Ca(2+) content and dihydropyridine receptor. In 25 generalized myasthenia gravis (MG), we detected antibodies against human TRPC3 peptide in 9 patients (8 with thymoma and one with hyperplastic thymus) and those against human RyR1 peptides in 16 patients (15 with thymoma and one with hyperplastic thymus). Both antibodies were found in patients with more severe myasthenia and could contribute to the contractile abnormalities in MG.


Assuntos
Autoanticorpos/sangue , Miastenia Gravis/sangue , Canal de Liberação de Cálcio do Receptor de Rianodina/imunologia , Canais de Cátion TRPC/imunologia , Adolescente , Adulto , Idoso , Ensaio de Imunoadsorção Enzimática/métodos , Feminino , Humanos , Masculino , Pessoa de Meia-Idade
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA